Open Access Article
This Open Access Article is licensed under a Creative Commons Attribution-Non Commercial 3.0 Unported Licence

Nanoparticle-mediated approaches to combat antibiotic resistance: a comprehensive review on current progress, mechanisms, and future perspectives

Sumreen Hayata, Asma Ashrafb, Muhammad Hussnain Siddiquec, Bilal Aslamd, Hamna Shafaqata, Saba Javeda, Zeeshan Taja, Muhammad Hassan Sarfraza, Hafsa Rafiqa and Saima Muzammil*a
aInstitute of Microbiology, Government College University, Faisalabad, Pakistan. E-mail: saimamuzammil83@gmail.com
bDepartment of Zoology, Government College University, Faisalabad, Pakistan
cDepartment of Bioinformatics and Biotechnology, Government College University, Faisalabad, Pakistan
dDepartment of Veterinary Preventive Medicine, College of Veterinary Medicine, Qassim University, Buraydah, Kingdom of Saudi Arabia

Received 13th June 2025 , Accepted 29th October 2025

First published on 4th November 2025


Abstract

Antibiotic resistance has become a serious global health issue that is responsible for millions of deaths each year globally. Multidrug resistant bacteria (MDR) are difficult to treat and pose a formidable health challenge to clinicians. The misuse of antibiotics has augmented the rise of resistant bacteria like ESKAPE (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species), thus highlighting the urgent need for innovative strategies. The use of nanoparticles for disturbing bacterial growth, inhibiting biofilm formation and targeting antibiotic delivery could be a promising solution to MDR bacteria. This comprehensive review illustrates how nanoparticles cope with MDR infections due to antibacterial photodynamic therapy and use as carriers for targeted drug delivery systems. Though the applications of nanoparticles in the field of medicine to treat multidrug resistant infections is a promising solution, however, the challenges persist in translating nanoparticle-based systems into clinical settings. The main hurdles include biocompatibility, minimizing the cytotoxicity, overcoming scalability problems, and addressing regulatory and environmental concerns. This review explains the recent progress in metallic and non-metallic nanoparticles that help to combat antibiotic resistance, highlighting their therapeutic applications, mechanisms of action, and integration into existing antibacterial strategies. Future directions highlight research to enhance efficient, safe, and sustainable nanoparticle-based therapeutics that address the growing antibiotic resistance crisis.


Introduction

Over the past 50 years, antimicrobial agents, particularly antibiotics, have been vital to human health. In 1928, when looking back to the pre-antibiotic age, Alexander Fleming's accidentally discovered penicillin as a miracle medication that transformed traditional treatment approaches. This was because of its remarkable capacity to treat or avoid dangerous infections, which used to be the primary cause of long-term sickness or death. Fleming did caution against the negative effects of incorrect and excessive penicillin usage.1 Despite the caution, antimicrobial resistance has emerged as a result of the widespread usage of antibiotics in agricultural, veterinary and clinical settings.2 Due to this, many pathogens are becoming MDR (multidrug-resistant) pathogens and increasing the global health crisis, rendering many standard treatments ineffective and leading to increased morbidity, mortality, and healthcare costs.3

Antimicrobial resistance is one of the world's public health issues, according to WHO it causes at least 4.95 million deaths worldwide each year. According to the CDC, Antibiotic Resistance (AR) Threats Report 2019, approximately 2.8 million illnesses each year in the US is caused only by antibiotic-resistant bacteria, leading to over 35[thin space (1/6-em)]000 deaths. One of major cause of antibiotic resistance are the biofilm production, which is connected to 65–80% of human illnesses.4 Drug resistance in biofilms can result from several processes, such as the transfer of resistant genes, decreased intracellular drug levels, lower drug absorption throughout the extracellular polymeric matrix, and slow down the bacterial metabolism.5

MDR prevents the organism's innate defenses against infections or reduces the efficacy of treatment. There are many risk factors associated with MDR bacteria including treatment failure and high mortality rate as shown in Fig. 1. Patients with impaired immune systems, such as those undergoing organ transplantation, chemotherapy for cancer, immunosuppressive medication, or chronic illness, are more susceptible to MDR infection. Furthermore, the cost of treatment has gone up due to MDR since microbes have become resistant to commonly available antibiotics, necessitating the use of a more costly one.6 According to recent report, current annual healthcare costs estimated at around USD 66 billion globally against MDR infections and this would be going to increase USD 2 trillion by 2050.7


image file: d5ra04206b-f1.tif
Fig. 1 Risk factors associated with multidrug resistance.

According to recent reports, the development of antibiotic resistant genes (ARG) among infectious microbes has been made through HGT (horizontal gene transfer), a serious common health problem.8 Genes resistant to antibiotics can spread through mobile genetic elements like transposomes and plasmids.9 Through various processes, including changes in targeted drug, decreased antibiotic absorption, and antibiotic degradation in bacteria could evolve resistance.10

As the number of diseases caused by various infectious bacteria rises, pharmaceutical firms and researchers are attempting to find new antibacterial agents that could chemically replace current antibiotics.11 Since these bacteria are constantly evolving, therefore present antibiotics offer no therapeutic advantage. As a result, this challenging scenario has motivated researchers to look for longer-term treatment approaches to stop the emergence of bacterial resistance.12 To address the issue of drug resistance, current studies have focused on using nanoparticles as antimicrobial agents against various infections caused by MDR, as well as serving as antimicrobial delivery vectors targeted at certain tissues.13,14 Nanotechnology and nanoparticles offer a promising solution to combat bacterial resistance, MDR, and microbial.15 Nanoparticles are biomaterials that range in size from one to one hundred nanometers (nm). Nanomaterials have drawn a lot of interest because of their extensive use in cosmetics, medicines, drug delivery system, agriculture, and most importantly as antibacterial components. They are now thought to be effective additions to or replacements for the antimicrobials that are currently in use.16 Nanoparticles are formed of three layers since they are not simple molecules. (a) The first layer is the surface, that functions with a range of metal ions, small molecules, polymers and surfactants. (b) The shell layer, that is entirely distinct chemically from the core; and (c) the core, that is the main part of the nanoparticles and is typically used to refer to the nanoparticles itself.17 Nanoparticles could be used directly for treatment (e.g., zinc oxide, titanium dioxide, silver nanoparticles) and as vehicle for antibacterial agents (e.g., dendrimers, liposomes, polymeric nanoparticles). There are two common applications of nanoparticles: (1) as anti-microbial agents themselves, or (2) in conjunction with clinically related antibiotics that are currently on the market to improve and alter their physiochemical characteristics to overcome antibacterial resistance property. The main areas of action for antibiotics are the inhibition of the synthesis of nucleic acids, translation or transcription during protein production, and the inhibition of production or rupture of the outer wall of cell.18 However, it is found that the use of nanoparticle technologies has an impact on the respiration system of bacteria, affecting the antioxidant system and causing the system of ROS (reactive oxygen species). This offers a new treatment strategy to overcome antimicrobial resistance.19

This study review explores the potential of nanoparticles in overcoming antibiotic resistance by targeting resistant pathogens and enhancing drug efficacy while addressing challenges such as toxicity, scalability, and regulatory hurdles. It highlights recent advances and future directions for integrating nanoparticles into antimicrobial strategies. Unlike previous studies, this review focus mainly the multiple mechanisms of nanoparticles to mediate antibiotic resistance and it has been divided into different sections systematically covering from antibiotic resistance mechanism to nanoparticles mode of action and then therapeutic discussing challenges and future directions.

Mechanisms of antibiotic resistance

Antibiotic resistance is a natural and old phenomenon found in bacteria that live in various biological niches, such as forest soil, isolated deep cave networks and marine sediments.20 Even though bacteria naturally develop resistance to antibiotics, human factors like excessive antibiotic use, improper prescription practices, and widespread agricultural use have enabled the development and spread of MDR bacteria, which exhibit resistance to numerous therapeutically significant drug.21 The WHO recently released the initial list of 12 MDR infections that are now biggest threats to human wellness for which new medications must be developed immediately.22 Bacteria develop antibiotic resistance through different mechanisms like efflux pumps, biofilm formation, enzyme production, antibiotic destruction, antibiotic modification, modifications of antibiotic-activating enzymes, target site alteration, target site protection, and lower the permeability of bacterial cell membrane.23 Fig. 2 shows the different mechanisms of antibiotic resistance, while some of them are shortly discussed here.
image file: d5ra04206b-f2.tif
Fig. 2 Various mechanisms of antibiotic resistance, including drug efflux with the help of efflux pump, enzymatic modifications of the antibiotic, enzymatic breakdown of the antibiotics, and modification in the target sites.

Efflux pump

Bacteria have chromosome-encoded efflux pump genes. Few are constitutively produced, while some may induced or overexpressed in response to specific environmental stimuli or when a suitable substrate is present.24 Efflux pumps are complicated bacterial systems found on the cytoplasmic membrane that require energy to function and could propel harmful substances from the cell. Escherichia coli was the first bacterium to be described with a plasmid-encoded efflux pump that pumped tetracycline outside the cell.25 Since then, few bacteria have been discovered that contain many efflux mechanisms implicated in antibiotic resistance. Most efflux systems can transfer several unrelated compounds, can result in resistance to multiple drugs.26

Biofilm formation

The development of biofilms is another tactic used by bacteria to increase their resistance to antimicrobials. Associations of microorganisms trapped in matrix of extracellular cells that they manufacture themselves are known as biofilms. Through a variety of processes that rely on variables like biofilm composition, architecture, the stage of biofilm development, and growth circumstances, they produce specific habitats that provide bacteria antibiotic tolerance and resistance.27 The structure of the biofilm prevents antibiotics from penetrating and may also stop bactericidal concentrations from building up throughout the biofilm. Furthermore, the biofilm's gradients in the spreading of oxygen and nutrients produce distinct metabolic states for each cell and promote the growth of bacterial persistence and antibiotic tolerance.5 Moreover, there are several ways in which biofilms might become resistant to antibiotics. For example, Staphylococcus aureus biofilms can colonize within medical instruments like pacemakers, and Pseudomonas aeruginosa biofilms cause severe lung problems in people with cystic fibrosis.28

Enzymes degradation

Certain enzymes that the bacterium generates precisely render the antibiotic inactive, depriving it of its biological activity. This happens, for example, when beta-lactamases break down beta-lactam medications.29 Some bacteria develop ESBLs (extended-spectrum-beta-lactamases), that have the same neutralizing action and are difficult to eradicate. Additional enzymes that have the ability to render some antibiotics ineffective include adenyl transferase, phosphotransferase, and acetyl transferase.4,30

MDR bacteria and EDR (extensively drug-resistant bacteria) pose a different challenge to public health and the global healthcare community etc. They increase the morbidity and mortality rate, create diagnostic challenges, limit the development of new antibiotics, compromise the medical and surgical interventions, limit the treatment options, cause longer hospital stays, and also cause economic burdens.4 At least 700[thin space (1/6-em)]000 deaths worldwide are attributed to MDR per annum, with 23[thin space (1/6-em)]000 occurring in the US and 25[thin space (1/6-em)]000 in the EU, according to reports.31 The WHO says that misuse and overuse of antibiotics are responsible for about 80% of MDR or XDR (extensively drug resistant) bacteria and that these infections have serious side effects.32 By 2050, 10 million people worldwide are expected to die of bacterial diseases if nothing is done to stop bacterial resistance or develop new medications.33

Mechanism of action of nanoparticles

For nanoparticles to have an antibacterial effect, they must encounter bacterial cells. Hydrophobic interactions, electrostatic attraction, receptor–ligand interactions and van der Waals forces are among the recognized kinds of contact.34 The nanoparticles penetrate the cell membrane, gather within the metabolic route, and alter the structure and functionality of the cell membrane.35 Subsequently, nanoparticles associate with the fundamental elements of the bacterial cell, including enzymes, DNA, ribosomes and lysosomes. This association results in oxidative stress, heterogeneous alterations, modifications to cell membrane permeability, inhibition of enzymes, imbalances in electrolyte levels, changes in gene expression and deactivation of proteins.36 According to the latest research, oxidative stress, dissolved metal ion release, and non-oxidative processes are the most often suggested mode of action as in Fig. 3.
image file: d5ra04206b-f3.tif
Fig. 3 Primary modes of action for nanoparticles. In addition to activating metal ion release, adhesion, accumulation, and damage to the cell wall and membrane, nanoparticles also show DNA damage, enzyme inactivation, protein oxidation and denaturation, increased reactive oxygen species (ROS) production, and inhibition of electron transport with resulting decrease in ATP level.

The following are the different modes of action of nanoparticles against antibiotic resistance.

Generation of reactive oxygen species (ROS)

Nanoparticles can disrupt the normal metabolic pathways of disease-causing agents through oxidative stress that is due to ROS reactive oxygen species (ROS). The adverse effects of nanoparticles are associated with the generation of reactive oxygen species, such as hydroxyl radicals, superoxide anions and hydrogen peroxide. These ROS hinder DNA replication and protein production, as well as damage cell membranes through lipid peroxidation, affecting membrane permeability and inhibiting oxidative phosphorylation.37 The quantity of ROS produced by nanoparticles depends on the chemical composition of the nanoparticles.38

Disruption of cell membranes

The potential of nanoparticles involves direct contact with the outer wall of bacteria e.g. Ag-NPs or ZnO. can permeate the cell wall, leading to alterations in the cell membrane of bacteria. This results in loss of membrane integrity, structural damage, and finally lead to cell die.39 Nanoparticles also induce the formation of pits in the outer wall of bacteria; for example, Ag-NPs aggregate on the cell surface create small pores in the cell wall move in the cell, and come in contact with fundamental molecules and organelles like DNA and enzyme.40 On the other hand, TiO2 produces bactericidal effects by triggering photocatalytic reactions on the membrane of the cell.41 Anuj et al. claim that Ag-NPs have degraded Pseudomonas aeruginosa membrane.42 The perforation of bacterial membranes caused by nanoparticle adhesion permits the nanoparticles to move in to the cell and interact with vital components and organelles including DNA and enzymes.38

Destruction of biofilm

Adhesion to the cell surfaces initiates the production of biofilms. However, when cells undergo treatment with nanoparticles, they cannot adhere and establish such a community. This is crucial, particularly when combating pathogenic organisms that generate biofilms.43 Due to their high surface area-to-volume ratio, nanoparticles interact more effectively with the components of biofilms. This larger surface area makes it possible to contact microbial cells more successfully. Nanoparticles could damage microbial cells by invasion of the biofilm matrix (Fig. 3). Due to their small size, they can more easily penetrate the EPS (extracellular polymeric substance) that envelops biofilm cells and get to the microbial cells that are embedded within ref. 44 Many methods of inhibiting the formation of biofilms include targeting and interfering with quorum-sensing molecules; ZnO-NPs destroy biofilms by releasing Zn+ ions.45 Nanomaterials can damage bacterial membranes and prevent the formation of biofilms, which reduces the microorganism's capacity for life, as numerous studies have shown ref. 46.

Nanoparticles as efflux pump inhibitor

It has recently been discovered that metallic nanoparticles may be able to obstruct bacterial efflux pumps. Nanoparticles attach directly to the pump of the cell membrane, stopping drugs from being whisked away.18,47 Metal nanoparticles may here act as a competitive inhibitor of antibiotics for the binding site of efflux pumps. Another possible mechanism is through the disruption of efflux kinetics. For Example, the effect of silver nanoparticles for disruption of the efflux kinetics of MDR efflux pump, has already been examined in Pseudomonas aeruginosa.48

Recent advances in nanoparticle research

Rise of antibacterial resistance has led to significant advancement in nanoparticle-based tactics to combat antibiotic resistance. Here are some recent advances in nanoparticle research for combating antibiotic resistance.

Biomimetic nanoparticle

The application of biomimetic nanoparticles that are altered by artificial or natural cell membranes has improved medication delivery and drawn a lot of interest recently against MDR bacteria.49 These nanoparticles mimic cell membranes or antimicrobial peptides to evade immune responses and target resistant bacteria. Some of the greatest characteristics of artificial and host nanoparticles are combined in nanoparticles that are coated with cell membrane (CM-NPs).50 Due to their distinct characteristics, including improved targeting ability and immune invasion, CM-NPs offer substantial therapeutic and diagnostic use. Preserving cell membranes' inherent qualities and functions is another advantage of CM-NPs. Because the body views CM-NPs as an integral part of itself, their biocompatibility is quite good. For examples, erythrocyte membrane-coated nanoparticles (RBC-NPs) have been used to improve circulation time and biocompatibility by mimicking red blood cells.48 It has been reported that Platelet membrane-coated nanoparticles (PNPs) having platelet adhesion properties could target bacteria at sites of injury and interfere the biofilm formation.51 Consequently, many membrane-coated nanoplatforms have been developed for biomedical devices, primarily focused on cancer therapy.52 Additionally, it has been documented that CM-NPs can target pathogenic bacteria, neutralize toxins, avoid immune recognition, and deliver medications to treat microbial infections.53 Currently, CM-NPs can be made using the membranes of erythrocytes, macrophages, platelets, neutrophils, bacteria epithelial cells, and hybrid membranes.51

Enzymes mimicking nanoparticles (nanozymes)

Nanozymes also known as enzymes mimicking nanoparticles were first reported in 2007 and are a novel class of synthetic enzymes characterized by distinct physicochemical characteristics and enzymes like properties to disturb bacterial processes.53 Scientists used the term “artificial enzyme” verbally to describe mimic enzyme models.54 Compared to natural enzymes, nanozymes offer certain advantages such as low cost and high stability, making them suitable against bacterial pathogens.55 Medical science has benefited from the use of these nanozymes as many nanozymes have now been used for cancer treatment. As shown in Fig. 4, nanozymes have been known for catalytic activity, ROS production, biofilm disruption and antibacterial activities.54
image file: d5ra04206b-f4.tif
Fig. 4 Catalytic properties of enzyme-mimicking nanoparticles (nanozymes) and their biomedical applications.

The antimicrobial mode of nanozymes is also based on the production of ROS.POD (peroxidase)-like nanozymes may break down extremely low concentrations of H2O2 to form highly oxidative ˙OH, which increases antibacterial activity without endangering healthy tissues.56 Certain nanomaterials can mimic OXD (oxidase), which is the direct activation of oxygen to produce ROS such as H2O2, superoxide anions, and single oxygen on their own. These ROS can kill bacteria and also destroy their biofilm.57 CuO nanozymes have been found to degrade biofilms formed by Pseudomonas aeruginosa through oxidative stress.58

Moreover, a variety of nanomaterials demonstrate multi-enzyme-like properties as shown in Fig. 4. For instance, Cu2WS4 nanocrystals have outstanding OXD-like and POD-like actions to produce ROS and work as effective antibacterial agents to treat skin infections caused by Staphylococcus aureus in mice.59 MnO2 and Chitosan nanoparticles could disrupt Gram-negative bacterial membranes like Staphlococus, Acinetobacter etc by peroxidase-like action.60

Nanoparticle-based antibacterial photodynamic therapy (aPDT)

The merging of nanoparticles in antibacterial photodynamic therapy has improved the efficiency of aPDT, particularly concerning MDR and infections that occur due to biofilms are hard to treat with traditional antibiotics. Nanoparticles enhance delivery, stability, and photodynamic efficiency of photosensitizers, resulting in an increased application of aPDT against bacterial infections. Nanoparticles enhance delivery, stability, and photodynamic efficiency of photosensitizers (PS), resulting in an increased application of aPDT against bacterial infections (Fig. 5). The efficient use of nanoparticles with photosensitizers against wound healing and dental treatment has also been reported.61 For example, Ag-NPs enhanced the ROS production when combined with rose Bengal (PS) during aPDT against E. coli.62 aPDT is a non-invasive treatment that uses light, photosensitizer, and molecular oxygen together.63 Meanwhile, it has been found that nanoparticles augment photosensitizers delivery by penetrating into biofilms like chitosan nanoparticles with curcumin (PS) targeted the Pseudomonas aeruginosa.64 ZnO-NPs displayed efficacy against Methicilline resistant Pseudomonas aeruginosa.65 Au and Ag-NPs enhanced ROS production with precise bacterial membrane target in aPDT.66
image file: d5ra04206b-f5.tif
Fig. 5 An overview of nanoparticle-based antibacterial photodynamic therapy (aPDT), illustrating general mechanisms of ROS generation and bacterial inactivation.

Synergistic effects of nanoparticles with antibiotics

Nanoparticles have capacity to fight bacterial infections by acting as Nano weapons. A promising tactic to fight MDR bacteria is the integration of nanoparticles along with antibiotics to augment their activity. The effects of antibiotics enhance due to nanoparticles beyond their individual action. In fact, against different microbes that are resistant to many drugs, metal oxides and metal nanoparticles have shown encouraging bactericidal activity.37 When antibiotics are coupled with nanoparticles, antibacterial activity has been enhanced and the adverse results which belong to long-term use of wide-range antibiotics lessened.67 One of the most promising approaches to address the growing danger of antibacterial resistance is to use nanoparticles in conjunction with antibiotics, as evidenced by their synergistic effects.40 For example, Synergistic interactions between therapeutically relevant antibiotics and silver nanoparticles, which target the bacterial cell wall, will increase the antibacterial efficacy of the medications. Ag-NPs considerably enhanced the synergistic action of antibiotics (azithromycin, cefuroxime, fosfomycin, cefotaxime, and chloramphenicol) against Escherichia coli as compared to antibiotics alone. Nanoparticles can disturb and degrade biofilms that enable the antibiotics to kill bacteria more efficiently. It has been studied that Ag-NPs with penicillin showed increased antibiotic effect against a variety of bacteria.68 Similarly, Au-NPs with vancomycin, ZnO-NPs with tetracycline, and GO-NPs with ciprofloxacin have showed synergistic antimicrobial activity against MDR bacterial strains.34

Nanoparticle-based antibiotic delivery systems

Encasing antibiotics into nanoparticles has emerged, one of the most innovative ways for targeted antibiotic delivery. Nano antibiotics are antibacterial nanoparticles that can enhance the effectiveness of antibiotics.69 Nanoparticles which are loaded with antibacterial agents also help to combat antibacterial resistance by enhancing (intracellular) uptake, lowering drug-efflux, and inhibiting biofilm development.70 Nanoparticles-coated antibiotics deliver medications to the target site and simultaneously trigger several antibacterial actions. Because of this combined approach, nanoparticles are superior to single medication or combinations of medication in the treatment of MDR strains.71 Drug delivery issues are being addressed by nano systems because they can be used to create nano systems that combine the best qualities of the mixture of biomaterials used to create them with the avoidance of their drawbacks.71 A main aspect of delivery system is its capacity to persist for long time in the circulatory system.72 Nanoparticles used in delivery systems include a wide variety of types. These include liposomes, organic and inorganic nanoparticles, solid-lipid nanoparticles, and non-toxic biodegradable polymers.73

Formulation types of nanoparticles

Here we discuss the nanoparticle types used as antibacterial against MDR (multi drug resistance) bacteria based on metallic and non-metallic forms.

Metallic nanoparticles

Silver nanoparticles. Metallic nanoparticles are either metals or metal oxides.74 Among metallic nanoparticles, due to its minimal cytotoxicity, silver (Ag) can have antiseptic and antimicrobial properties against various Gram-negative and Gram-positive bacteria.75,76 Ag-NPs have proven to be successful in treating infectious disorders by inhibiting MDR microorganisms.8,77 Ag-NPs can destroy plasma membranes and change the structure and metabolism of bacteria because they are small in size and have high ratio of surface area to volume78 Various techniques based on biological processes has been used to synthesize silver nanoparticles.79 Because of their nanoscale size (1–10 nm), they can enter bacteria and interfere with intracellular functions, which can hinder the formation of proteins, translation, and transcription.80 Ag-NPs can cause bacteria's cell membranes to get damaged and produce ROS.78 Ag-NPs effectively obstruct biofilm development by Staphylococcus aureus.81,82 Ag-NPs have proved to hinder the progress of many microbes, including Staphylococcus aureus, Salmonella typhii, Citrobacter koseri, Bacillus cereus, Escherichia coli, Klebsiella pneumoniae, Pseudomonas aeruginosa and Vibrio parahaemolyticus via attaching to the molecules present within bacterial cells.37,38,83
Gold nanoparticles. Gold nanoparticles block transcription, create pores in cell wall, and bound to Deoxyribonucleic acid.66 When compared to other metallic nanoparticles, gold (Au-NPs) has not so strong antibacterial effectiveness against both Gram-positive and Gram-negative microbial species. However, Au-NPs smaller than two nanometers exhibit enhanced antibacterial activity, which arises not only from their reduced size but also from the presence of concomitant substances introduced during synthesis. Compounds such as polyvinylpyrrolidone and ascorbic acid can modify the nanoparticles' reactivity, surface charge, and interactions with bacterial membranes, thereby contributing to the observed antibacterial effects.84 When used alone, Au-NPs antimicrobial properties are primarily derived from their capacity to disrupt protein formation by blocking capability of transfer RNA to bind ribosome,85 and to alter the trans-membrane potential.86

The suppression of ATPase synthesis, which lowers cell metabolism, and inhibiting the ribosome binding component to transfer RNA are associated with the antibacterial activity of AuNPs but they better associate with vaccines, antibodies, and antibiotics.85 Nicotinamide might be destroyed by Au-NPs, which would then upset the microbial electron transport chain.84 Au-NPs target microorganisms like Streptococcus Bovis, Staphylococcus epidermidis, Enterococcus aerogenes, Escherichia coli and Pseudomonas aeroginosa.78,87,88

Copper nanoparticles. Copper-containing nanoparticles are efficient against bacteria, decrease MDR biofilms from growing, and function as antimicrobial coating agents. Copper oxide nanoparticles release metallic ions, which can form (ROS) reactive oxygen species and damage the DNA.38 When CuO-NPs enter bacteria, they impact metabolic activities such as active transport and metabolism.89 Copper nanoparticles can prevent biofilm formation, inhibit ATP production by interacting with bacterial cells.90 The combination of Cu2O nanoparticles with aminoglycoside antibiotics resulted in significant synergistic antibacterial action against Escherichia coli.78 Copper ions exhibit antibacterial activity against several organisms, including Escherichia coli, Clostridium jejuni, Salmonella enterica, Listeria monocytogenes, Staphylococcus aureus and Bacillus subtilis91,92
Zinc nanoparticles. Strong, broad-spectrum bactericidal action is demonstrated by ZnO-NPs. They are affordable, biocompatible, and belong to the class of inorganic antibacterial agents. It demonstrates amplification, sensitization, and a synergistic bactericidal activity.93 ZnO-NPs, are tiny, high surface-area, biosafe, and environmentally benign materials. These NPs have antimicrobial characteristics due to the formation of ROS (hydroxyl radicals and H2O2), which enhance cell membrane permeability, internalization of nanoparticles due to force loss, and dissolved zinc ion uptake, which results in a decrease in mitochondrial activity and cell death. They may also cause leakage and bacterial mortality by damaging the bacterial cell membrane's integrity.38
Iron nanoparticles. Iron nanoparticles (Fe-NPs) constitute an additional family of antimicrobial compounds. According to several research, altering surfaces activates their antibacterial qualities, which eliminates bacterial biofilms of both Gram-negative and Gram-positive bacteria94 These have demonstrated as less expensive substitutes in medication solutions, antibacterial coatings, and other applications that aim to limit microbial invasion or eradication since they also hinder the formation of biofilms.95
Titanium dioxide nanoparticles. TiO2 (titanium dioxide) nanoparticles are able to degrade the common food-borne bacteria, Listeria monocytogenes biofilms, which can lead to health issues when food is being prepared. Conversely, TiO2 nanoparticles suppress the proliferation of microbes and reduce biofilms.96 TiO2 nanoparticles have a biocidal impact even though it is inert and non-toxic to humans. According to a number of research on the antibacterial properties of TiO2, when exposed to sunlight or ultraviolet radiation, TiO2 also produces reactive oxygen species particularly –OH free radicals such as free radicals superoxide ions and hydrogen peroxide which induce tangible harm to the cell walls of microorganisms.97,98 TiO2 nanoparticles are also used in hospital environments in antibacterial coatings.

Generally, antibacterial efficiency tends to decrease with increasing particle size, probably because smaller nanoparticles might be capable of passing through the plasma membrane with greater efficacy. In the view of metal nanoparticles, smaller nanoparticles release their metal ions faster if their volume to surface area ratio is larger.62 These nanoparticles have several special qualities, including increased bioactivity, improved bioavailability, high reactivity with molecules, and novel surface characteristics99 The antimicrobial activity of various metallic nanoparticles has been summarized in Table 1. The proper incorporation of nanoparticles led to effective interaction with MDR bacteria, leads to the generation of ROS and plasma membrane, which damage and cause the death of the bacterial species.100

Table 1 Summarize the various metallic nanoparticles with their mechanism of action and their applications
Nanoparticles used Targeted pathogens Nanoparticles mode of action Applications Refrences
Silver nanoparticles Staphylococcus epidermidis, Streptococcus viridans, Staphylococcus aureus, Klebsiella pneumoniae, Moraxella catarrhalis, Proteus mirabilis Damage bacterial membranes and trigger the release of (reactive oxygen species) ROS, producing radicals with a strong bactericidal activity Antiseptic and antimicrobial properties against Gram negative and Gram-positive microbes 101
Pseudomonas aeruginosa, Escherichia coli Inhibits growth of pathogens by ROS Drug delivery and have antibacterial activity 102
Staphylococcus aureus, Escherichia coli Silver ions cause death of bacteria Used to control growth of bacteria, inhibit cell division, damage the cell envelope 103
Staphylococcus aureus, Escherichia coli, Citrobacter koseri, Pseudomonas aeruginosa, Salmonella typhii, Bacillus cereus Microbicidal effect by producing reactive oxygen Microbicidal effect against various infections and diseases, and antiseptic efficacy against microbes 90
Enterococcus faecalis Staphylococcus epidermidis, Pseudomonas aeruginosa, Staphylococcus aureus Generation of ROS peroxidation of lipid cytochrome inhibition in electron transport chain Used in drug design and drug delivery 104
Escherichia coli Reactive oxygen species (ROS) generation Direct effect on bacterial cell membranes, affect respiration, proliferation and metabolism 38
Escherichia coli, Staphylococcus aureus, Streptococcus Pneumoniae Ribosomal destabilization intercalation of DNA bases Magnetics, optics, catalysis, mechanics, nanobiotechnology, and nanomedicine, antimicrobial activity, anticancer effects 105
Klebsiella pneumoniae, Bacillus cereus Proton gradient dissipation leads to lysis Antimicrobial activities, against MRSA synergistic effect 37
Klebsiella pneumonia, Vibrio parahaemolyticus Bactericidal activity, damage the membrane of bacteria Therapeutics, diagnostics, and photovoltaics, as well as catalysts 106
Proteus sp Inhibit replication of DNA thus causes death of bacteria Inhibit DNA replication, act as antibacterial agents, disrupt the cell membrane 107
Gold nanoparticles Escherichia coli, Pseudomonas aeruginosa Antibacterial activity, decrease the level of ATP Control of diseases, against the cancer cells photothermal therapy, photoimaging 66
Streptococcus bovis, Staphylococcus epidermidis, Enterobacter aerogenes, Escherichia coli Bacterial membrane rupture. Bacterial cell wall disruption damage the DNA Antimicrobial activities against MDR Escherichia coli 38
Staphylococcus aureus, Bacillus subtilis, Kleblesilla pneumoniae, Escherichia coli Bind to DNA to suppress transcription Drug & gene delivery systems in cancer therapy, diabetes mellitus, cardiovascular, antibacterial activity 90
Vibrio cholerae, Bacillus subtilis Loss of membrane potential, inhibit the protein synthesis Biocidal nano weapons, disrupt the membrane of bacteria, denature the 30 S subunit of ribosome and penetrate inside the cell 37
Streptococcus bovis, Staphylococcus epidermidis, Enterobacter aerogenes, Pseudomonas aeruginosa Antimicrobial activities, attach to the cell surface and damage membrane of microorganisms Penetrate the biological membranes, bactericidal activity, cancer therapy 108
Escherichia coli, Klebsiella pneumoniae Inhibition of ATPase production, disrupt the cytoplasmic membrane Antimicrobial activity, diagnosis of cancer 109
Escherichia coli Antibacterial mechanism, attach to the cell wall and penetrate the plasma membrane Effective against MRSA, antibiofilm and antibacterial activity 110
Enterococcus faecalis, Staphylococcus epidermidis, Streptococcus pyogenes, Enterococcus faecalis and Bacillus, Staphylococcus aureus (MRSA) Antibacterial and immunological action, generate reactive oxygen species, protein phosphorylation inhibition Early detection systems, imaging diagnosis, and therapy for diseases specially against drug-resistant microorganisms, immunological characters 111
Vibrio cholerae, Escherichia coli, Staphylococcus aureus, Pseudonomas aeroginosa, Enterococcus species Antibacterial agents, leads to decrease in the levels of ATP Biocompatible, non-cytotoxic and used as therapeutic drug delivery vehicles 88
Pseudomonas aeruginosa, Escherichia coli, and MRSA Antimicrobial activities, attach to the cell surface and damage membrane of microorganisms Localized surface plasmon resonance (LSPR), role in many applications such as biosensors 112
Pseudmonas aeruginosa Interact with the surface of cell, antimicrobial activity Enhance activity of immune cells against microbes 113
Escherichia coli, Staphylococcus aureus, Listeris monocytogenes, Pseudomonas aeruginosa Anti-biofilm and antibacterial activity, breakdown the structures of bacteria, denature the proteins Prevent biofilm formation, break mature biofilms, and kill many types of Gram-positive 114
Copper oxide nanoparticles Escherichia coli, Pseudomonas aeruginosa Antibacterial effect destruction of cell membrane potential Gas sensing, hydrogen production, CO (catalytic oxidation), photocatalysis 78
Bacillus subtilis, Campylobacter jejuni, Listeria monocytogenes Inhibit the growth of different pathogenic microbes Solar cells, gas sensors, conductors and in preservation of polishes and wood 90
Salmonella enteric, Klebsiella pneumonia, Enterobacter aerogenes, Salmonella typhimurium ROS production, antimicrobial agents Biomedical and pharmaceutical sciences 115
Escherichia coli Reactive oxygen generation, intracellular content of cell leakage thus causes cell death High toxicity causes oxidative lesions 37
Staphylococcus aureus, Staphylococcus epidermidis Binding to DNA disrupts the helix structure, localize into organelles thus disrups the normal functions of cells To reduce infections in hospitals, burn treatment, prevent microbes and fungi colonization on catheters, vascular grafts, dental materials, eliminate microbes on textile 116
Staphylococcus aureus, Pseudomonas aeruginosa, Clostridium difficile, Escherichia coli Result in malfunctions and ultimately kills the bacterial cells, damages the cell and result in the death of cell Neuropeptide production, cell signaling pathway regulation, antioxidant defence, and immune cell function 117
Pseudomonas aeruginosa, Staphylococcus aureus Antibacterial activity, decrease the level of ATP, generates the ROS Semiconductors and heat transfer nanofluids, electronic chips 118
Staphylococcus aureus, Escherichia coli, Bacillus subtilis Disrupt chemical processes within bacterial cells, disrupts the plasma membrane Field of catalysis, plant pathology therapy, and electrical sensors 92
Pseudomonas aeruginosa, Staphylococcus aureus, Escherichia coli Effect metabolic functions of bacteria Act as antimicrobial coating agents 38
Zinc oxide nanoparticles Staphylococcus aureus, Mycobacterium tuberculosis, Proteus mirabilis, Streptococcus pyogenes Anti-bacterial activity, generates the reactive oxygen species, damages the membrane of cell Preservation of food items, cosmetics, wound dressing for fast healing,as an antiseptic ointment, disinfecting agent 119
Klebsiella aerogenes, Escherichia coli Antimicrobial mechanism, disrupts the plasma membrane, penetrates to the lipid membrane and causes the death of cell Can be used as potential antibacterial in medicines, biomedical applications, has wide role in addressing drug resistivity issue 115
Salmonella enterica, Campylobacter jejuni, Listeria monocytogenes Enter the bacterial cells and damage them Act as antibiotic agent for the reduction of coliform bacteria 120
Listeria monocytogenes, Salmonella Toxic for pathogenic strains along with antibacterial efficacy, generates the reactive oxygen species, damages the DNA Enhances cell permeability, used as drug delivery vehicle, Act as antibacterial agent 90
Escherichia coli Disrupt the microbial plasma membrane, produces the ROS (reactive oxygen species), disrupts the membrane and leads to programmed death of cell Can be used in photocatalysis, gas sensing, hydrogen production 78
Staphylococcus aureus, Pseudomonas aeruginosa Inhibition of enzyme lipid and protein damage Can be used as therapeutic agents, transport of drug to specific site 108
Escherichia coli, Staphylococcus aureus Inhibit growth of bacterial cells, causes disruption of cell membrane, generates ROS Antibacterial activity 121
Staphylococcus aureus, Streptococcus pyogenes, Escherichia coli, Klebsiella aerogenes, Pseudomonas aeruginosa, Proteus mirabilis, Mycobacterium tuberculosis, Bacillus subtilis Antimicrobial activity, ROS production causes oxidative stress, cell death by apoptosis Anti-bacterial agent, antiseptic ointment, disinfecting agent and for coating of medical devices, water treatment 119
Pseudomonas aeruginosa, Vibrio parahaemolyticus, Bacillus licheniformis Inhibit growth of bacteria, penetrates the bacterial cell, produces ROS Antibacterial and antibiofilm activity 122
Iron-containing nanoparticles Pseudomonas aeruginosa, Staphylococcus aureus, Bacillus subtilis, Escherichia coli Bactericidal activity, damage the membrane of bacteria, causes structural damage to cell Elimination of harmful textile dyes in an environmentally friendly way, effective materials for therapies and have biological applications 115
Staphylococcus epidermidis, Staphylococcus aureus, Escherichia coli, Streptococcus pneumoniae Eliminate bacterial biofilms produces oxidative stress, damge the components of cell Drug solutions and antibacterial coatings and other applications in which microbial elimination occur 90
Staphylococcus aureus, Salmonella enterica, Pseudomonas mirabilis, Escherichia coli Antibacterial activity, interferes with the processes of cell, causes death of bacteria Kill the growth of microorganisms 123
Bacillus subtilis, Escherichia coli, Staphylococcus aureus, Staphylococcus epidermidis Generation of ROS, bactericidal activity, effects the transport of ions and molecules Analytical chemistry, antigen diagnosis, pathogen detection, tissue repair and hyperthermia 124
Salmonella typhimurium, Staphylococcus aureus Bactericidal and antibiofilm activity, act as drug delivery systems, damage the plasma membrane of bacteria Agriculture, biomedical, and engineering, drug delivery, magnetic resonance imaging, hypothermia therapy in biomedicine 125
Escherichia coli Dysfunction of microbial membrane, produces ROS, have antibacterial characteristics Magnetic resonance imaging 126
Escherichia coli, Pseudomonas aeruginosa Antibacterial activity, interacts with the iron levels of cell, destroys the cell wall, cause destruction of DNA Effective against gram positive and negative bacteria 127
Titanium dioxide nanoparticles Staphylococcus aureus, Escherichia coli Inhibit the growth of bacteria, produces hyrdoxyl radicals, superoxide anions Control the spread and infection of a variety of microbial strains 128
Enterococcus faecium, Klebsiella pneumoniae, Pseudomonas aeruginosa Produce oxidixing agents and have antibacterial activity Utilized in food items such as in chewing gum and sweets, photocatalysts, oxidizing agents 38
Escherichia coli Decomposes the membrane, causes the mutuations in DNA, damages the proteins Photocatalyst, solar cell material, self-cleaning coating material, anti-fogging 129
Staphylococcus aureus Reaction with (thioL) group of amino acids (proteins) that occur on bacterial exterior surface and release ions Antibacterial activity 130
Pseudomonas aeruginosa, Enterococcus faecium, Klebsiella pneumoniae, Staphylococcus aureus Damages the DNA thus generates ROS, degrades the proteins and lipids Semiconductors, and heat transfer nanofluids, electronic chips 114
Listeria monocytogenes, staphylococcus aureus Minimizing the production of biofilms, damages the DNA, leads to the oxidative stress Can be used on the surface of medical implants to reduce the rate of contamination by microbes 96


Non-metallic nanoparticles. Dendrimers are an attractive nano-platform for microbicidal activity and drug delivery because of their large surface area, high (in vivo) reactivity, and ability to load both non-polar and polar molecules.131 Dendrimers have a huge surface region that makes them useful including drug and gene delivery.132–134 The first and most studied dendrimer for antimicrobial medication delivery is polyamidation, or PAMAM.135 Certain properties of dendrimers include: its highly branched structure gives a large surface contact-to-volume ratio, leading to exceptional response to microorganisms in vivo.136 Their capacity to load polar and non-polar molecules render them an excellent nano-platform for the delivery of drugs.131

Antibacterial activity of dendrimers with a high proportion of (positively) charged sites was greater than that of free antibiotics. This is because the polycationic nature of quaternary ammonium compounds allows them to bind to the (negatively) charged bacterial plasma membrane. This increases membrane permeability, allowing more dendrimers to enter the bacteria, leaking potassium (K+) ions, and eventually destroying the bacterial cell membrane.137 In literature, Dendrimers are particularly more efficient against MDR S. aureus and P. aeruginosa.138

Liposomes are spherical nano vesicles that include aqueous compartments or units accompanied by one or more phospholipid bilayers.139 They have ability to encapsulate medications that are hydrophilic within the aqueous phase or hydrophobic drugs within lipid bilayer distinguishes them from other nanoparticles and significantly expands the range of drugs that may be integrated Fig. 6.140 According to several studies, liposomal encapsulation increases the safety and stability of antibiotics, extending their bloodstream life and enabling more accurate targeting of infection sites through a variety of delivery methods. This leads to more suitable pharmacokinetic and pharmacodynamics profiles.141 Aerosolized liposome antimicrobials like ciprofloxacin, tobramycin, amphotericin B and amikacin have been used to treat chronic respiratory tract infections.


image file: d5ra04206b-f6.tif
Fig. 6 Liposomal drug delivery systems with entrapped molecules.

Liposomes have a bilayer membrane that mimics cell membranes and can fuse directly with microbes. This allows the liposome's pharmacological payload to be released into the microorganism. Furthermore, liposome surfaces can be easily changed to improve their in vivo stability, or to target ligands to enable more selective drug delivery. The use of liposomes as antimicrobial carriers to combat microorganism resistance has grown in research.131,142

Liposomes can have antibacterial capabilities, either alone or when encapsulated, which can improve medicine's antimicrobial efficacy. Gram-negative bacteria's outer membrane is a complex barrier that can alter antibiotic interactions with the bacterial wall or block internalization, which makes it a major source of new resistances.143 However, liposomes may encourage the fusing of bacterial membranes, disrupting the structure and perhaps reversing poor permeability.143,144 Liposomes have demonstrated activity against MDR bacteria like Methicillin-resistant Staphylococcus aureus (MRSA) and Pseudomonas aeruginosa.145

Another type is polymeric nanoparticles like chitosan, PLGA (Polylactic-Co-Glycolic Acid). They are used as antibacterial against MDR strains and targeted drug delivery.146 Chitosan having antibacterial properties has been widely used against different bacteria such as Acinetobacter, Escherichia coli, Staphylococcus aures.147 PLGA nanoparticles were employed to deliver antibiotics (ciprofloxacin and ampicillin) in controlled mannered improved the efficacy against MDR Klebsiella pneumoniae.148 Lipid based nanoparticles (SLNs) are used for sustained release of antibiotics against Staphylococcus and Pseudomonas.149 Meso-porous silica nanoparticles (MSN) pores allow the encapsulation of antibiotics (tetracycline and gentamicin) and release them at target to control infection caused by Pseudomonas aeruginosa150 (Table 2).

Table 2 Summarize the role of non-metallic/organic nanoparticles as antibacterial and in drug delivery system
Nanoparticles used Targeted pathogens Nanoparticles mode of action Applications References
Liposomes Bacillus subtilis Liposomes containing antibacterial agents like streptomycin or kanamycin can stop synthesis of protein, thus leads to death of cell Drug delivery for bacterial infections and antibacterial formulations, also used in cancer therapy and vaccination 151
Pseudomonas aeruginosa Bacterial cell membrane disruption can cause lysis of cell when liposomes containing antibacterial drugs such as colistin or tobramycin are present Antibacterial nanocarriers for controlled release and in cancer therapy 152
Staphylococcus aureus, Pseudomonas aeruginosa Saprophyticus Act as antibiotic delivery nano-systems, decreasing the selection of resistant strains prolonging the duration of antibiotics in the circulation, enabling more accurate administration of the drugs through different routes to the infection locations Used in antibacterials that includes antibiotics 145
Staphylococcus aureus Vancomycin- or daptomycin-containing liposomes have the ability to stop the formation of cell walls, which kills the cell Can be delivered directly into microbial membranes, act as drug delivery systems, enhance antibiotic concentration at the site of infection 153
Dendrimers Proteus hauseri, Escherichia coli, Pseudomonas aeruginosa, Staphylococcus saprophyticus, Staphylococcus aureus, Staphylococcus epidermidis Antibacterial activity is greater than that of antibiotics, bind to the microbial cell wall, increases the permeability of the membrane, resulting in (k) potassium ion leakage, that eventually leads to the destruction of the microbial plasma membrane Gene therapy, biomedical applications, biocompatibility and pharmacokinetics, antibacterial agents 136
Escherichia coli Antibacterial compounds such as silver or copper found in dendrimers plasma damage bacterial cell membranes, results in lysis and death of cell Encapsulates the drugs, helps in controlled release of medicines(drugs), deliver the drug safely act as drug delivery systems 146
Staphylococcus aureus Cell death may result from dendrimers that contain vancomycin or daptomycin, which block the formation of cell walls Protects the drugs from degradation, function as non-viral vectors, target specific tissues in the body, promotes differentiation and growth of cell 154
Bacillus subtilis Protein synthesis can be inhibited by dendrimers containing antibacterial drugs such as streptomycin or kanamycin, which can ultimately result in cell death Antbiofilm and antibacterial characteristics 155
SWCNTs (single walled nanotubes) Staphylococcus aureus, Escherichia coli k12 The attachment or depositing of bacteria on a bacterial surface. Intracellular fluid leakage and impairment of the cell membrane Mechanical, thermal and electrical characteristics used in biosensors and drug delivery 156
Escherichia coli, Bacillus subtilis Injuries to the cell wall, seepage of internal fluid, reduction in cell height and volume, and increased roughness on the surface of bacteria Used in photovoltaics, and nanoelectronics to help in integration into complexed materials 157
f-SWNTs (functionalized single walled nanotubes) with functional groups (–OH, –COOH, –NH2) Staphylococcus aureus, Bacillus subtilis, and Salmonella typhimurium While SWNTs functionalized with –NH2 only exhibited antibacterial activity at high concentrations, those functionalized with –OH and –COOH functional groups showed greater microbial inhibition rate (7[thin space (1/6-em)]log decrease) against certain pathogens Facilitates the safe delivery of medicines, enhances the specificity such as in MRI and imaging, prevent the growth of bacteria, helps in the repair of tissues 158
SWNTs bound with polyamide WCNmembranes Escherichia coli Sixty percent of the microbial cells were rendered inactive by the nanocomposite complex after one hour of interaction Helps in the process of de-salination, importance in separation of gas technologies, helps in development of membranes which can bear high pressure 159
Chitosan nanoparticles Escherichia coli, Pseudomonas aeruginosa Disruption of bacterial membranes due to cationic nature interaction with bacterial cell wall components (e.g., peptidoglycan) ROS generation and inhibition of bacterial enzymes Drug delivery systems (e.g., antibiotics). Wound healing (chitosan dressings). Antibacterial coatings for medical devices 160
Polymeric micelles Escherichia coli, Staphylococcus aureus Controlled release of encapsulated drugs at target sites enhanced antibacterial efficacy through surface modification Targeted drug delivery (e.g., cancer, bacterial infections) and coatings for implants and devices 161
Poly (lactic-co-glycolic acid) (PLGA) nanoparticles Klebsiella pneumoniae, Streptococcus pneumoniae Controlled release of antimicrobial agents biodegradability and biocompatibility enhancing therapeutic effects Targeted drug delivery (e.g., to infected tissues) antibacterial and anticancer therapy 162
Polyethylene glycol (PEG)-based nanoparticles Escherichia coli, Staphylococcus aureus PEGylation provides steric stabilization and prevents rapid immune recognition encapsulation of antibiotics for controlled release Targeted delivery for infections and prolonged circulation time in the bloodstream 163
Polycaprolactone (PCL) nanoparticles Staphylococcus epidermidis, Pseudomonas aeruginosa Encapsulation of hydrophobic antimicrobial agents – slow release over extended periods due to PCL's biodegradability Controlled release of antibiotics and antibacterial wound dressings 164


Challenges, limitations and future directions

In the expanding planet, health-related challenges are getting worse. To battle them adequately and sufficiently, the most recent methods are required. The use of nanotechnology and its connection to antibiotic resistance is one of the effective strategies.165 Though, dealing with nanoparticles has certain limitations. Because of their distinctive size and physical characteristics, nanoparticles provide significant advantages over conventional antibiotics; however, due to this small size characteristic, nanoparticles can breach the physiological barriers of living things and trigger undesirable biological processes.37 It is evident that nanoparticles can also enter the body via skin, intestines, or lungs etc. So, long-term exposure could be responsible for threat to public health because of growing worries about the toxicological consequences of these particles.166 Bioaccumulation of nanomaterials in different tissues leads to toxicity in lungs or kidney. They can also cause cardiac issues, inflammation in the lungs and brain damage.167

Sometime, use of organic nanomaterials like liposomes for specific drug delivery leads to premature drug absorption. Also, to develop functionalized organic nanomaterials for targeted delivery is costly and labor-intensive, Scalability and consistent quality in large-scale production remain significant hurdles for organic nanomaterials.66,168 The development of oxidative stress, DNA damage, and apoptosis by nanoparticles, toxic effects may also result in morphological abnormalities, reproductive problems, and malformations in several non-mammalian animal models.169

To date, despite lots of research work on nanomaterials, there is limited possibility of their use against MDR bacteria in clinical settings due to various challenges. The potential toxicity of nanomaterials, which can cause oxidative stress, inflammation, and DNA damage, raises safety issues for human uses.78,170 Nanomaterials, normally also a subject of great concern for the regulatory authorities. To understand the interactions, between nanomaterials and antibacterial drug is great challenge. Pharmacological assessments are also a concern for nanoparticles due to diverse size range of these nanoparticles. So, uniform pharmacokinetic properties are difficult to achieve for their therapeutic applications. Meanwhile, reproducibility of these nanomaterials at large-scale production is still a limitation factor.48,141 FDA and European Medicines Agency (EMA) have authorized several nanoparticles for cancer therapy, but guidelines for use these products in soft tissues are still lacking. For the efficient and safe use of nanomaterials, advanced evaluation and detailed regulatory advice is needed. For this, coordination between agencies such as the FDA, WHO, and other health agencies is crucial to develop harmonized risk-assessment protocols, environmental impact guidelines, and post-market surveillance systems for nano-based antibacterial agents.

In the future research should be focused to address the problems faced by the clinicians to apply nanoparticle based system in real life. Therefore, focus on enhancing biocompatibility and reducing cytotoxicity should be preferred. Furthermore, addressing the environmental and regulatory dimensions alongside the development of scalable, eco-friendly synthesis methods will be essential for realizing the full clinical and industrial potential of nanoparticle-based therapeutics against multidrug-resistant pathogens.

Author contributions

Sumreen Hayat: writing – original draft, visualization, investigation, data curation, conceptualization. Asma Ashraf: writing review & editing, Muhammad Hussnain Siddique: writing– review & editing, resources, conceptualization. Bilal Aslam: writing– review & editing, conceptualization, Hamna Shafaqat: writing – original draft, Saba Javed: writing – original draft, resources, Zeeshan Taj: writing– review & editing, Muhammad Hassan Sarfraz: visualization, writing– review & editing, Hafsa Rafiq: visualization, Saima Muzammil: supervision, writing – review & editing.

Conflicts of interest

The authors declare no conflicts of interest.

Data availability

No primary research results and no new data were generated or analysed as part of this review.

References

  1. J. O'neill, Review on Antimicrobial Resistance, 2014 Search PubMed.
  2. M. Osman, H. Al Mir, R. Rafei, F. Dabboussi, J.-Y. Madec, M. Haenni and M. Hamze, J. Glob. Antimicrob. Resist., 2019, 17, 123–129 CrossRef.
  3. H. Chen, R. Chen, L. Jing, X. Bai and Y. Teng, Environ. Pollut., 2019, 245, 398–407 CrossRef CAS.
  4. H. F. Hetta, Y. N. Ramadan, A. I. Al-Harbi, E. A. Ahmed, B. Battah, N. H. Abd Ellah, S. Zanetti and M. G. Donadu, Biomedicines, 2023, 11, 413 CrossRef CAS.
  5. C. W. Hall and T.-F. Mah, FEMS Microbiol. Rev., 2017, 41, 276–301 CrossRef CAS.
  6. J. Tanwar, S. Das, Z. Fatima and S. Hameed, Interdiscip. Perspect. Infect. Dis., 2014, 2014, 541340 Search PubMed.
  7. CDC, Antibiotic Resistance Threats In The United States, https://stacks.cdc.gov/view/cdc/82532/cdc_82532_DS1.pdf, accessed 5 September, 2025 Search PubMed.
  8. J. Lu, Y. Wang, M. Jin, Z. Yuan, P. Bond and J. Guo, Water Res., 2020, 169, 115229 CrossRef CAS.
  9. E. Y. Klein, T. P. Van Boeckel, E. M. Martinez, S. Pant, S. Gandra, S. A. Levin, H. Goossens and R. Laxminarayan, Proc. Natl. Acad. Sci. U. S. A., 2018, 115, E3463–E3470 CAS.
  10. E. Peterson and P. Kaur, Front. Microbiol., 2018, 9, 2928 CrossRef PubMed.
  11. I. M. Hamouda, J. Biomed. Res., 2012, 26, 143–151 CrossRef CAS.
  12. I. Khan, K. Saeed and I. Khan, Arab. J. Chem., 2019, 12, 908–931 CrossRef CAS.
  13. Y. Wang, Y. Jin, W. Chen, J. Wang, H. Chen, L. Sun, X. Li, J. Ji, Q. Yu and L. Shen, Chem. Eng. J., 2019, 358, 74–90 CrossRef CAS.
  14. I. E. Mba and E. I. Nweze, World J. Microbiol. Biotechnol., 2021, 37, 1–30 CrossRef.
  15. M. H. Siddique, S. Hayat, S. Muzammil, A. Ashraf, A. M. Khan, M. U. Ijaz, M. Khurshid and M. Afzal, Drug Dev. Ind. Pharm., 2022, 48, 502–509 CrossRef CAS PubMed.
  16. S. Li, T. Zhu, J. Huang, Q. Guo, G. Chen and Y. Lai, Int. J. Nanomed., 2017, 2593–2606 CrossRef CAS PubMed.
  17. W.-K. Shin, J. Cho, A. G. Kannan, Y.-S. Lee and D.-W. Kim, Sci. Rep., 2016, 6, 26332 CrossRef CAS.
  18. C. Ashajyothi, K. H. Harish, N. Dubey and R. K. Chandrakanth, J. Nanostruct. Chem., 2016, 6, 329–341 CrossRef CAS.
  19. M. H. Sarfraz, S. Hayat, M. H. Siddique, B. Aslam, A. Ashraf, M. Saqalein, M. Khurshid, M. F. Sarfraz, M. Afzal and S. Muzammil, Prog. Org. Coat., 2024, 188, 108235 CrossRef CAS.
  20. J. Davies and D. Davies, Microbiol. Mol. Biol. Rev., 2010, 74, 417–433 CrossRef CAS PubMed.
  21. S. Hayat, S. Muzammil, M. H. Rasool, Z. Nisar, S. Z. Hussain, A. N. Sabri and S. Jamil, Microbiol. Immunol., 2018, 62, 211–220 CrossRef CAS PubMed.
  22. I. Alav, J. M. Sutton and K. M. Rahman, J. Antimicrob. Chemother., 2018, 73, 2003–2020 CrossRef CAS PubMed.
  23. D. Versluis, M. M. D'Andrea, J. Ramiro Garcia, M. M. Leimena, F. Hugenholtz, J. Zhang, B. Öztürk, L. Nylund, D. Sipkema and W. v. Schaik, Sci. Rep., 2015, 5, 11981 CrossRef PubMed.
  24. W. C. Reygaert, AIMS Microbiol., 2018, 4, 482 CAS.
  25. J. M. Blair, G. E. Richmond and L. J. Piddock, Future Microbiol., 2014, 9, 1165–1177 CrossRef CAS.
  26. N. A. Villagra, J. A. Fuentes, M. R. Jofré, A. A. Hidalgo, P. García and G. C. Mora, J. Antimicrob. Chemother., 2012, 67, 921–927 CrossRef CAS.
  27. C. Uruén, G. Chopo-Escuin, J. Tommassen, R. C. Mainar-Jaime and J. Arenas, Antibiotics, 2021, 10, 3 CrossRef.
  28. P. K. Singh, A. L. Schaefer, M. R. Parsek, T. O. Moninger, M. J. Welsh and E. Greenberg, Nature, 2000, 407, 762–764 CrossRef CAS.
  29. K. S. Rizi, K. Ghazvini and M. K. Noghondar, J. Infect. Dis. Ther., 2018, 6, 2332–0877 Search PubMed.
  30. R. M. Abd El-Baky, S. M. Farhan, R. A. Ibrahim, K. M. Mahran and H. F. Hetta, Alexandria J. Med., 2020, 56, 4–13 CrossRef.
  31. M. Yin, M. Yang, D. Yan, L. Yang, X. Wan, J. Xiao, Y. Yao and J. Luo, ACS Appl. Mater. Interfaces, 2022, 14, 8847–8864 CrossRef CAS PubMed.
  32. W. H. Organization, Antibiotic Resistance: Multi-Country Public Awareness Survey, World Health Organization, 2015 Search PubMed.
  33. T. J. Abdullah, M. Atif, S. Khalid, K. Metwally, G. Yahya, M. Moisa and D. S. Cavalu, Int. J. Mol. Sci., 2024, 25, 8915 CrossRef CAS.
  34. L. Wang, C. Hu and L. Shao, Int. J. Nanomed., 2017, 1227–1249 CrossRef PubMed.
  35. A. Ullah, F. A. Al-Saeed, A. M. Abduallah, A. E. Ahmed, A. Shahzad, N. Amjad, A. Ali, M. S. Mostafa and R. Hussain, Pak. Vet. J., 2023, 43, 241–247 CAS.
  36. Y. Xu, M.-T. Wei, H. D. Ou-Yang, S. G. Walker, H. Z. Wang, C. R. Gordon, S. Guterman, E. Zawacki, E. Applebaum and P. R. Brink, J. Nanobiotechnol., 2016, 14, 1–16 CrossRef PubMed.
  37. H. A. Hemeg, Int. J. Nanomed., 2017, 8211–8225 CrossRef CAS.
  38. T. Bekele and G. Alamnie, Ann. Adv. Chem., 2022, 6, 001–009 CrossRef.
  39. R. Brayner, R. Ferrari-Iliou, N. Brivois, S. Djediat, M. F. Benedetti and F. Fiévet, Nano Lett., 2006, 6, 866–870 CrossRef CAS.
  40. S. Muzammil, S. Hayat, M. Fakhar-E-Alam, B. Aslam, M. H. Siddique, M. A. Nisar, M. Saqalein, M. Atif, A. Sarwar and A. Khurshid, Front. Biosci., 2018, 10, 352–374 CrossRef.
  41. J. Kiwi and V. Nadtochenko, Langmuir, 2005, 21, 4631–4641 CrossRef CAS PubMed.
  42. S. A. Anuj, H. P. Gajera, D. G. Hirpara and B. A. Golakiya, J. Trace Elem. Med. Biol., 2019, 51, 219–225 CrossRef CAS.
  43. S. M. Gamboa, E. Rojas, V. Martínez and J. Vega-Baudrit, Int. J. Biosens. Bioelectron., 2019, 5, 166–173 Search PubMed.
  44. R. M. Pinto, F. A. Soares, S. Reis, C. Nunes and P. Van Dijck, Front. Microbiol., 2020, 11, 952 CrossRef PubMed.
  45. M. Rai, K. Kon, A. Gade, A. Ingle, D. Nagaonkar, P. Paralikar and S. da Silva, Antibiot. Resist., 2016, 121–143 Search PubMed.
  46. P. P. Mahamuni-Badiger, P. M. Patil, M. V. Badiger, P. R. Patel, B. S. Thorat-Gadgil, A. Pandit and R. A. Bohara, Mater. Sci. Eng., C, 2020, 108, 110319 CrossRef CAS.
  47. H. Kotrange, A. Najda, A. Bains, R. Gruszecki, P. Chawla and M. M. Tosif, Int. J. Mol. Sci., 2021, 22, 9596 CrossRef CAS.
  48. D. Gupta, A. Singh and A. U. Khan, Nanoscale Res. Lett., 2017, 12, 1–6 CrossRef CAS.
  49. R. H. Fang, A. V. Kroll, W. Gao and L. Zhang, Adv. Mater., 2018, 30, 1706759 CrossRef.
  50. X. Zhen, P. Cheng and K. Pu, Small, 2019, 15, 1804105 CrossRef PubMed.
  51. J. Ma, L. Jiang and G. Liu, Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol., 2022, 14, e1825 CrossRef CAS PubMed.
  52. M. Imran, K. R. Paudel, S. K. Jha, P. M. Hansbro, K. Dua and Y. Mohammed, Nanomedicine, 2022, 17, 665–670 CrossRef CAS PubMed.
  53. L. Rao, R. Tian and X. Chen, ACS Nano, 2020, 14, 2569–2574 CrossRef CAS PubMed.
  54. G. Saleem, X. Chen, R. Gu, M. Qasim, M. Usama and N. Rajput, Nanotechnol. Rev., 2022, 11, 2575–2583 CrossRef CAS.
  55. J. Wu, X. Wang, Q. Wang, Z. Lou, S. Li, Y. Zhu, L. Qin and H. Wei, Chem. Soc. Rev., 2019, 48, 1004–1076 RSC.
  56. L. Tonoyan, D. Montagner, R. Friel and V. O'Flaherty, Biochem. Pharmacol., 2020, 182, 114281 CrossRef CAS PubMed.
  57. Y. Zhang, X. Hu, J. Shang, W. Shao, L. Jin, C. Quan and J. Li, Theranostics, 2022, 12, 5995 CrossRef CAS.
  58. I. Liaqat, S. Noor, A. S. Qureshi, S. Ali, N. Al-Arifa, S. Alam, A. Ajmal, T. Zia and M. Munawar, Pak. Vet. J., 2023, 43, 118–124 CAS.
  59. J. Shan, X. Li, K. Yang, W. Xiu, Q. Wen, Y. Zhang, L. Yuwen, L. Weng, Z. Teng and L. Wang, ACS Nano, 2019, 13, 13797–13808 CrossRef CAS PubMed.
  60. M. H. Sarfraz, S. Muzammil, S. Hayat, M. Khurshid and A. H. Sayyid, Int. J. Biol. Macromol., 2023, 242, 124954 CrossRef CAS.
  61. B. A. Thomas-Moore, C. A. Del Valle, R. A. Field and M. J. Marín, Photochem. Photobiol. Sci., 2022, 21, 1111–1131 CrossRef CAS PubMed.
  62. Y. Chen, G. Fu, F. Liang, J. Wei, J. He and J. Bai, J. Transcult. Nurs., 2020, 31, 284–293 CrossRef PubMed.
  63. Z. Zhuang, J. Dai, M. Yu, J. Li, P. Shen, R. Hu, X. Lou, Z. Zhao and B. Z. Tang, Chem. Sci., 2020, 11, 3405–3417 RSC.
  64. N. Saad, M. A. El-Abasy, F. El-Khayat, N. G. Ali and M. M. Ismail, Vet. J., 2023, 43, 573–578 CAS.
  65. M. Li, X. Jin, T. Liu, F. Fan, F. Gao, S. Chai and L. Yang, Nat. Commun., 2022, 13, 4137 CrossRef CAS PubMed.
  66. P. Singh, S. Pandit, J. Garnæs, S. Tunjic, V. R. Mokkapati, A. Sultan, A. Thygesen, A. Mackevica, R. V. Mateiu and A. E. Daugaard, Int. J. Nanomed., 2018, 3571–3591 CrossRef CAS.
  67. U. H. Abo-Shama, H. El-Gendy, W. S. Mousa, R. A. Hamouda, W. E. Yousuf, H. F. Hetta and E. E. Abdeen, Infect. Drug Resist., 2020, 351–362 CrossRef CAS.
  68. B. M. ALRashdi, M. O. Germoush, S. S. Sani, I. Ayub, W. Bashir, B. Hussain, M. Mazhar, S. Ali, Z. Zahid and S. Ayesha, Pak. Vet. J., 2023, 43, 282–289 Search PubMed.
  69. R. Labruère, A. Sona and E. Turos, Front. Pharmacol., 2019, 10, 1121 CrossRef PubMed.
  70. S. Hadiya, R. A. Ibrahem, R. M. Abd El-Baky, M. Elsabahy and S. A. Aly, Microb. Drug Resist., 2022, 28, 972–979 CrossRef CAS.
  71. M. M. Elhassan Taha, S. I. Abdelwahab, S. S. Moni, A. Farasani, I. A. Aljahdali, B. Oraibi, H. A. Alfaifi, A. H. Alzahrani and A. Ali Jerah, Hum. Vaccines Immunother., 2024, 20, 2427464 CrossRef.
  72. X. Yang, W. Ye, Y. Qi, Y. Ying and Z. Xia, Front. Bioeng. Biotechnol., 2021, 9, 696514 CrossRef PubMed.
  73. J. K. Patra, G. Das, L. F. Fraceto, E. V. R. Campos, M. d. P. Rodriguez-Torres, L. S. Acosta-Torres, L. A. Diaz-Torres, R. Grillo, M. K. Swamy and S. Sharma, J. Nanobiotechnol., 2018, 16, 1–33 CrossRef.
  74. J. W. Alexander, Surg. Infect., 2009, 10, 289–292 CrossRef PubMed.
  75. A. Fouda, M. A. Awad, Z. E. Al-Faifi, M. E. Gad, A. A. Al-Khalaf, R. Yahya and M. F. Hamza, Catalysts, 2022, 12, 462 CrossRef CAS.
  76. F. Baghbani-Arani, R. Movagharnia, A. Sharifian, S. Salehi and S. A. S. Shandiz, J. Photochem. Photobiol., B, 2017, 173, 640–649 CrossRef CAS PubMed.
  77. J. S. Möhler, W. Sim, M. A. Blaskovich, M. A. Cooper and Z. M. Ziora, Biotechnol. Adv., 2018, 36, 1391–1411 CrossRef.
  78. Y. Zhang, Y. Yuan, W. Chen, J. Fan, H. Lv and Q. Wu, Colloids Surf., B, 2019, 183, 110371 CrossRef CAS PubMed.
  79. J. Y. Song and B. S. Kim, Bioproc. Biosyst. Eng., 2009, 32, 79–84 CrossRef CAS.
  80. M. Rai, A. Yadav and A. Gade, Biotechnol. Adv., 2009, 27, 76–83 CrossRef CAS.
  81. R. Singh, M. Smitha and S. P. Singh, J. Nanosci. Nanotechnol., 2014, 14, 4745–4756 CrossRef CAS PubMed.
  82. T. C. Dakal, A. Kumar, R. S. Majumdar and V. Yadav, Front. Microbiol., 2016, 7, 1831 CrossRef.
  83. K. S. Siddiqi, A. Husen and R. A. Rao, J. Nanobiotechnol., 2018, 16, 1–28 CrossRef.
  84. S. Shamaila, N. Zafar, S. Riaz, R. Sharif, J. Nazir and S. Naseem, Nanomaterials, 2016, 6, 71 CrossRef.
  85. Y. Cui, Y. Zhao, Y. Tian, W. Zhang, X. Lü and X. Jiang, Biomaterials, 2012, 33, 2327–2333 CrossRef CAS PubMed.
  86. S. Gharpure, A. Akash and B. Ankamwar, J. Nanosci. Nanotechnol., 2020, 20, 3303–3339 CrossRef.
  87. J. N. Payne, H. K. Waghwani, M. G. Connor, W. Hamilton, S. Tockstein, H. Moolani, F. Chavda, V. Badwaik, M. B. Lawrenz and R. Dakshinamurthy, Front. Microbiol., 2016, 7, 607 CrossRef.
  88. A. N. Brown, K. Smith, T. A. Samuels, J. Lu, S. O. Obare and M. E. Scott, Appl. Environ. Microbiol., 2012, 78, 2768–2774 CrossRef CAS PubMed.
  89. Y. Su, X. Zheng, Y. Chen, M. Li and K. Liu, Sci. Rep., 2015, 5, 15824 CrossRef CAS.
  90. F. Fatima, S. Siddiqui and W. A. Khan, Biol. Trace Elem. Res., 2021, 199, 2552–2564 CrossRef CAS.
  91. D. Longano, N. Ditaranto, L. Sabbatini, L. Torsi and N. Cioffi, Nano-antimicrobials: Progress and Prospects, 2012, pp. 85–117 Search PubMed.
  92. A. I. El-Batal, N. M. Balabel, M. S. Attia and G. S. El-Sayyad, J. Cluster Sci., 2020, 31, 1021–1040 CrossRef CAS.
  93. M. Germec, A. Demirci and I. Turhan, Biocatal. Agric. Biotechnol., 2020, 27, 101662 CrossRef.
  94. N. Beyth, Y. Houri-Haddad, A. Domb, W. Khan and R. Hazan, Evid. base Compl. Alternative Med., 2015, 2015, 246012 Search PubMed.
  95. L. M. Armijo, S. J. Wawrzyniec, M. Kopciuch, Y. I. Brandt, A. C. Rivera, N. J. Withers, N. C. Cook, D. L. Huber, T. C. Monson and H. D. Smyth, J. Nanobiotechnol., 2020, 18, 1–27 CrossRef.
  96. Y. Cao, M. Naseri, Y. He, C. Xu, L. J. Walsh and Z. M. Ziora, J. Glob. Antimicrob. Resist., 2020, 21, 445–451 CrossRef.
  97. Y. Xing, X. Li, L. Zhang, Q. Xu, Z. Che, W. Li, Y. Bai and K. Li, Prog. Org. Coat., 2012, 73, 219–224 CrossRef CAS.
  98. M. Azizi-Lalabadi, A. Ehsani, B. Divband and M. Alizadeh-Sani, Sci. Rep., 2019, 9, 17439 CrossRef.
  99. P. Chaudhary, F. Fatima and A. Kumar, J. Inorg. Organomet. Polym. Mater., 2020, 30, 5180–5192 CrossRef CAS.
  100. P. J. P. Espitia, N. d. F. F. Soares, J. S. d. R. Coimbra, N. J. de Andrade, R. S. Cruz and E. A. A. Medeiros, Food Bioprocess Technol., 2012, 5, 1447–1464 CrossRef CAS.
  101. G. Franci, A. Falanga, S. Galdiero, L. Palomba, M. Rai, G. Morelli and M. Galdiero, Molecules, 2015, 20, 8856–8874 CrossRef CAS.
  102. D. Bose and S. Chatterjee, Indian J. Microbiol., 2015, 55, 163–167 CrossRef CAS.
  103. W. K. Jung, H. C. Koo, K. W. Kim, S. Shin, S. H. Kim and Y. H. Park, Appl. Environ. Microbiol., 2008, 74, 2171–2178 CrossRef CAS PubMed.
  104. M. Esmaeillou, G. Zarrini, M. A. Rezaee and A. Bahadori, Adv. Pharmaceut. Bull., 2017, 7, 479 CrossRef CAS PubMed.
  105. R. Thapa, C. Bhagat, P. Shrestha, S. Awal and P. Dudhagara, Ann. Clin. Microbiol. Antimicrob., 2017, 16, 1–10 CrossRef.
  106. M. J. Sweet, A. Chessher and I. Singleton, Adv. Appl. Microbiol., 2012, 80, 113–142 CAS.
  107. K. Chandraker, R. Nagwanshi, S. Jadhav, K. K. Ghosh and M. L. Satnami, Spectrochim. Acta, Part A, 2017, 181, 47–54 CrossRef CAS.
  108. R. Huang, L. Zhang, X. Li, F. Liu, X. Cheng, H. Ran, Z. Wang, Y. Li, Y. Feng and L. Liang, J. Controlled Release, 2023, 356, 610–622 CrossRef CAS.
  109. S. Shaikh, S. M. D. Rizvi, S. Shakil, T. Hussain, T. M. Alshammari, W. Ahmad, S. Tabrez, M. H. Al-Qahtani and A. M. Abuzenadah, J. Cell. Biochem., 2017, 118, 2802–2808 CrossRef CAS PubMed.
  110. P. V. Baptista, M. P. McCusker, A. Carvalho, D. A. Ferreira, N. M. Mohan, M. Martins and A. R. Fernandes, Front. Microbiol., 2018, 9, 1441 CrossRef PubMed.
  111. T. Cherian, D. Maity, R. T. Rajendra Kumar, G. Balasubramani, C. Ragavendran, S. Yalla, R. Mohanraju and W. J. Peijnenburg, Nanomaterials, 2022, 12, 2940 CrossRef CAS.
  112. N. Rabiee, S. Ahmadi, O. Akhavan and R. Luque, Materials, 2022, 15, 1799 CrossRef CAS.
  113. Q. Yu, J. Li, Y. Zhang, Y. Wang, L. Liu and M. Li, Sci. Rep., 2016, 6, 26667 CrossRef CAS PubMed.
  114. F. Khan, D. T. N. Pham, S. F. Oloketuyi, P. Manivasagan, J. Oh and Y.-M. Kim, Colloids Surf., B, 2020, 185, 110627 CrossRef CAS.
  115. S. Gautam, D. K. Das, J. Kaur, A. Kumar, M. Ubaidullah, M. Hasan, K. K. Yadav and R. K. Gupta, Discover Nano, 2023, 18, 84 CrossRef CAS PubMed.
  116. T. Kruk, K. Szczepanowicz, J. Stefańska, R. P. Socha and P. Warszyński, Colloids Surf., B, 2015, 128, 17–22 CrossRef CAS.
  117. R. Chakraborty, R. K. Sarkar, A. K. Chatterjee, U. Manju, A. P. Chattopadhyay and T. Basu, Biochim. Biophys. Acta Gen. Subj., 2015, 1850, 845–856 CrossRef CAS PubMed.
  118. A. L. Ulloa-Ogaz, H. A. Piñón-Castillo, L. N. Muñoz-Castellanos, M. S. Athie-García, M. D. L. Ballinas-Casarrubias, J. G. Murillo-Ramirez, L. Á. Flores-Ongay, R. Duran and E. Orrantia-Borunda, Environ. Sci. Pollut. Res., 2017, 24, 22048–22060 CrossRef CAS PubMed.
  119. H. Agarwal, S. Menon, S. V. Kumar and S. Rajeshkumar, Chem. Biol. Interact., 2018, 286, 60–70 CrossRef CAS.
  120. P. Horky, S. Skalickova, L. Urbankova, D. Baholet, S. Kociova, Z. Bytesnikova, E. Kabourkova, Z. Lackova, N. Cernei and M. Gagic, J. Anim. Sci. Biotechnol., 2019, 10, 1–12 CrossRef PubMed.
  121. K. Lingaraju, H. Raja Naika, K. Manjunath, R. Basavaraj, H. Nagabhushana, G. Nagaraju and D. Suresh, Appl. Nanosci., 2016, 6, 703–710 CrossRef CAS.
  122. B. Malaikozhundan, B. Vaseeharan, S. Vijayakumar, K. Pandiselvi, M. A. R. Kalanjiam, K. Murugan and G. Benelli, Microb. Pathog., 2017, 104, 268–277 CrossRef CAS PubMed.
  123. T. Naseem and M. A. Farrukh, J. Chem., 2015, 2015, 912342 Search PubMed.
  124. M. Arakha, S. Pal, D. Samantarrai, T. K. Panigrahi, B. C. Mallick, K. Pramanik, B. Mallick and S. Jha, Sci. Rep., 2015, 5, 14813 CrossRef CAS.
  125. F. Erci and R. Cakir-Koc, Inorg. Nano-Met. Chem., 2020, 51, 683–692 CrossRef.
  126. A. K. Chaurasia, N. D. Thorat, A. Tandon, J.-H. Kim, S. H. Park and K. K. Kim, Sci. Rep., 2016, 6, 33662 CrossRef CAS PubMed.
  127. F. Farouk, M. Abdelmageed, M. Azam Ansari and H. M. Azzazy, Biotechnol. Lett., 2020, 42, 231–240 CrossRef CAS PubMed.
  128. N. Jones, B. Ray, K. T. Ranjit and A. C. Manna, FEMS Microbiol. Lett., 2008, 279, 71–76 CrossRef CAS.
  129. U. Joost, K. Juganson, M. Visnapuu, M. Mortimer, A. Kahru, E. Nõmmiste, U. Joost, V. Kisand and A. Ivask, J. Photochem. Photobiol., B, 2015, 142, 178–185 CrossRef CAS PubMed.
  130. A. S. Roy, A. Parveen, A. R. Koppalkar and M. A. Prasad, J. Biomaterials Nanobiotechnol., 2010, 1, 37 CrossRef CAS.
  131. L. Zhang, D. Pornpattananangkul, C.-M. Hu and C.-M. Huang, Curr. Med. Chem., 2010, 17, 585–594 CrossRef CAS PubMed.
  132. A. R. Menjoge, R. M. Kannan and D. A. Tomalia, Drug discovery today, 2010, 15, 171–185 CrossRef CAS PubMed.
  133. S. H. Medina, V. Tekumalla, M. V. Chevliakov, D. S. Shewach, W. D. Ensminger and M. E. El-Sayed, Biomaterials, 2011, 32, 4118–4129 CrossRef CAS PubMed.
  134. Y.-b. Lim, T. Kim, J. W. Lee, S.-m. Kim, H.-J. Kim, K. Kim and J.-s. Park, Bioconjug. Chem., 2002, 13, 1181–1185 CrossRef CAS PubMed.
  135. R. V. d. Araújo, S. d. S. Santos, E. Igne Ferreira and J. Giarolla, Molecules, 2018, 23, 2849 CrossRef PubMed.
  136. A. A. Chis, C. Dobrea, C. Morgovan, A. M. Arseniu, L. L. Rus, A. Butuca, A. M. Juncan, M. Totan, A. L. Vonica-Tincu and G. Cormos, Molecules, 2020, 25, 3982 CrossRef CAS PubMed.
  137. J. Kwasniewska and A. W. Bara, Int. J. Mol. Sci., 2022, 23, 1306 CrossRef CAS PubMed.
  138. A. Falanga, V. Del Genio and S. Galdiero, Pharmaceutics, 2021, 13, 101 CrossRef CAS.
  139. M. Ferreira, M. Ogren, J. N. Dias, M. Silva, S. Gil, L. Tavares, F. Aires-da-Silva, M. M. Gaspar and S. I. Aguiar, Molecules, 2021, 26, 2047 CrossRef CAS.
  140. M. Eugénia, M. Cruz, M. M. Gaspar, M. Bárbara, F. Martins and M. L. Corvo, in Methods in Enzymology, Elsevier, 2005, vol. 391, pp. 395–413 Search PubMed.
  141. N. E. Eleraky, A. Allam, S. B. Hassan and M. M. Omar, Pharmaceutics, 2020, 12, 142 CrossRef CAS.
  142. R. Lakshminarayanan, E. Ye, D. J. Young, Z. Li and X. J. Loh, Adv. Healthcare Mater., 2018, 7, 1701400 CrossRef PubMed.
  143. Z. Drulis-Kawa and A. Dorotkiewicz-Jach, Int. J. Pharm., 2010, 387, 187–198 CrossRef CAS.
  144. R. Nisini, N. Poerio, S. Mariotti, F. De Santis and M. Fraziano, Front. Immunol., 2018, 9, 155 CrossRef PubMed.
  145. Z. Makhlouf, A. A. Ali and M. H. Al-Sayah, Antibiotics, 2023, 12, 875 CrossRef CAS PubMed.
  146. Y. Wang and H. Sun, Pharmaceutics, 2021, 13, 2108 CrossRef CAS PubMed.
  147. M. H. Sarfraz, M. Zubair, B. Aslam, A. Ashraf, M. H. Siddique, S. Hayat, J. N. Cruz, S. Muzammil, M. Khurshid and M. F. Sarfraz, Front. Microbiol., 2023, 14, 1188743 CrossRef.
  148. S. Paliwal, J. Sharma, V. Dave, S. Sharma, K. Verma, K. Tak, R. R. Kakarla, V. Sadhu, P. Walvekar and T. M. Aminabhavi, Polym. Bull., 2024, 81, 535–547 CrossRef CAS.
  149. M. B. Zewail, A. S. Doghish, H. M. El-Husseiny, E. A. Mady, O. A. Mohammed, A. M. Elbadry, A. S. Elbokhomy, A. Bhnsawy and W. A. El-Dakroury, Biomater. Sci., 2024, 12, 6163–6195 RSC.
  150. A. Sharma, D. Kumar, V. Kumar, S. P. Singh, A. R. Sharma and S. K. Sharma, Hybrid Adv., 2024, 5, 100133 CrossRef.
  151. S. Gupta and G. J. Schneider, Soft Matter, 2020, 16, 3245–3256 RSC.
  152. X. Cheng, H. Yan, S. Pang, M. Ya, F. Qiu, P. Qin, C. Zeng and Y. Lu, Front. Chem., 2022, 10, 963004 CrossRef CAS PubMed.
  153. F. Hajiahmadi, M. Y. Alikhani, H. Shariatifar, M. R. Arabestani and D. Ahmadvand, Int. J. Nanomed., 2019, 5943–5955 CrossRef CAS PubMed.
  154. C. K. Ezeh and M. E. Dibua, ADMET and DMPK, 2024, 12, 239–267 Search PubMed.
  155. H. An, X. Deng, F. Wang, P. Xu and N. Wang, Polymers, 2023, 15, 2292 CrossRef CAS.
  156. M. Azizi-Lalabadi, H. Hashemi, J. Feng and S. M. Jafari, Adv. Colloid Interface Sci., 2020, 284, 102250 CrossRef CAS PubMed.
  157. N. Hadidi and M. Mohebbi, Microorganisms, 2022, 10, 2439 CrossRef CAS.
  158. M. A. Saleemi, Y. L. Kong, P. V. C. Yong and E. H. Wong, Adv. Pharmaceut. Bull., 2022, 12, 449 CrossRef CAS.
  159. S. F. Nitodas, M. Das and R. Shah, Membranes, 2022, 12, 454 CrossRef CAS PubMed.
  160. Y. A. R. Gaviria, W. D. C. Chacon, K. Cesca, G. C. Leandro, G. A. Valencia and C. da Costa, Int. J. Biol. Macromol., 2024, 263, 130513 CrossRef CAS.
  161. Z. Wan, R. Zheng, P. Moharil, Y. Liu, J. Chen, R. Sun, X. Song and Q. Ao, Molecules, 2021, 26, 1220 CrossRef CAS PubMed.
  162. A. Shariati, Z. Chegini, E. Ghaznavi-Rad, E. N. Zare and S. M. Hosseini, Front. Cell. Infect. Microbiol., 2022, 12, 926363 CrossRef CAS.
  163. I. C. Pereira, A. S. Duarte, A. S. Neto and J. Ferreira, Mater. Sci. Eng., C, 2019, 96, 606–615 CrossRef CAS.
  164. S. Javaid, N. M. Ahmad, A. Mahmood, H. Nasir, M. Iqbal, N. Ahmad and S. Irshad, Polymers, 2021, 13, 2180 CrossRef CAS PubMed.
  165. G. Muteeb, Microorganisms, 2023, 11, 1489 CrossRef CAS PubMed.
  166. R. A. Yokel and R. C. MacPhail, J. Occup. Med. Toxicol., 2011, 6, 1–27 CrossRef PubMed.
  167. G. Oberdörster, E. Oberdörster and J. Oberdörster, Environ. Health Perspect., 2005, 113, 823–839 CrossRef PubMed.
  168. L. Wei, J. Lu, H. Xu, A. Patel, Z.-S. Chen and G. Chen, Drug discovery today, 2015, 20, 595–601 CrossRef CAS.
  169. Q. H. Tran and A.-T. Le, Adv. Nat. Sci. Nanosci. Nanotechnol., 2013, 4, 033001 CrossRef CAS.
  170. H. Wang, P. Li, D. Yu, Y. Zhang, Z. Wang, C. Liu, H. Qiu, Z. Liu, J. Ren and X. Qu, Nano Lett., 2018, 18, 3344–3351 CrossRef CAS.

This journal is © The Royal Society of Chemistry 2025
Click here to see how this site uses Cookies. View our privacy policy here.