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

Beyond antibiotics: novel solutions to address antibacterial resistance

Afsaneh Arshadi Edlo a, Kamran Akhbari *a and David J. Henry b
aSchool of Chemistry, College of Science, University of Tehran, P.O. Box 14155-6455, Tehran, Iran. E-mail: akhbari.k@ut.ac.ir; Fax: +98 21 66495291; Tel: +98 21 61113734
bSchool of Mathematics, Statistics, Chemistry and Physics, Murdoch University, Murdoch, Western Australia 6150, Australia. E-mail: d.henry@murdoch.edu.au; Tel: +61 8 9360 2681

Received 27th May 2025 , Accepted 13th September 2025

First published on 15th September 2025


Abstract

In recent years, there has been a significant increase in antibacterial resistance, leading to a decline in the effectiveness of antibiotic drugs. This situation underscores the urgent need to explore suitable alternatives to antibiotics. To address this global challenge, it is crucial to understand new approaches, including their mechanisms, advantages, and limitations, which can help in the design of effective substitutes for antibiotics. Extensive research in this field has yielded notable progress. This review article aims to summarize innovative strategies for combating antibacterial resistance, such as metal–organic frameworks (MOFs), metal nanoparticles, photodynamic therapy (PDT), and antibacterial peptides. Additionally, the article discusses examples of their effectiveness and applications. Further research has also focused on combining these methods to enhance their efficiency, with some relevant studies highlighted. It is hoped that in the future, these materials will serve as replacements for current drugs, ultimately resolving the issue of antibacterial resistance.


image file: d5ma00552c-p1.tif

Afsaneh Arshadi Edlo

Afsaneh Arshadi Edlo is an Inorganic Chemistry researcher specializing in metal–organic frameworks (MOFs) and metal nanoparticles (MNPs). Her research focuses on designing and functionalizing nanomaterials for biomedical applications, particularly in the areas of antibacterial activity, drug delivery, cancer treatment, and environmental catalysis. Afsaneh completed her Master's degree in Inorganic Chemistry at the University of Tehran in 2022, following her Bachelor's degree in Applied Chemistry from Shahid Beheshti University. Her scientific contributions include publications in peer-reviewed journals and presentations at international conferences, where she highlights innovative strategies to combat antimicrobial resistance.

image file: d5ma00552c-p2.tif

Kamran Akhbari

Kamran Akhbari joined University of Tehran, Department of Chemistry in 2013. He earned a Bachelor's Degree in Chemistry from Shahid Beheshti University in 2005, a Master of Science Degree in 2008 and PhD Degree in 2012 in Inorganic Chemistry from Tarbiat Modares University. He completed a post-doctoral fellowship under the supervision of Prof. Ali Morsali at Tarbiat Modares University. He has published more than 150 papers, and his current research focuses on the crystal engineering of coordination polymers and supramolecular compounds, metal–organic frameworks (MOFs) and their potential applications in energy storage materials, separation, drug delivery and catalysis. He also conducts research into the synthesis and characterization of nano-scale materials. In 2013, he won second place in the Khwarizmi Young Award for his work in the field of modulating methane storage in nanoporous anionic MOFs via post synthetic cation exchange.

image file: d5ma00552c-p3.tif

David J. Henry

Professor David J. Henry is a Professor in the Department of Chemistry and Physics at Murdoch University, Australia. He earned his PhD in Biological and Chemical Sciences from Deakin University in 1998. Following this, he undertook a Postdoctoral Fellowship at the Research School of Chemistry, Australian National University. He later joined Murdoch University in 2015, where he has since advanced to his current role. Professor Henry's research is interdisciplinary, spanning inorganic and computational chemistry, nanomaterials, and environmental chemistry. His work employs advanced computational methods, including density functional theory (DFT), quantum chemical modeling, and molecular dynamics simulations, to investigate problems in soil water repellency, bio-inorganic systems, and the design of nanostructured materials. He has published nearly 100 peer-reviewed articles, with over 2500 citations, and continues to contribute actively to both fundamental and applied aspects of chemistry.


1. Introduction

One of the biggest challenges to humanity today is antibacterial resistance, which has significantly grown globally in recent years and presents a serious medical problem in healthcare settings.1 Excessive antibiotic use and misuse have contributed to the emergence of superbugs – pathogens that can withstand even the most potent medications, exacerbating antibiotic resistance.2 This resistance is a natural phenomenon, driven by the prevalence of resistance genes and their interaction with complex ecosystems. Furthermore, the use of antibiotics in agriculture and animal husbandry, as well as their role as growth promoters, compounds the problem, while global factors like travel, trade, and immigration accelerate its spread. These combined issues put millions of lives at risk annually and threaten to undo decades of medical progress. Additionally, the unsupervised release of antibacterial chemicals into the environment increases selection pressure on microbes, further intensifying the crisis.3 The World Health Organization (WHO) cautions that by 2050, antibiotic-resistant diseases may account for more deaths than cancer if nothing is done.4 In order to address this rising global health concern, it is critical that techniques for fighting bacterial infections be reconsidered, with a focus on innovation beyond conventional antibiotics.

Metal–organic frameworks (MOFs) demonstrate significant promise in antibacterial applications owing to their tunable porosity, high surface area, and structural adaptability.5,6 These materials facilitate controlled encapsulation and release of antimicrobial agents while providing direct bactericidal activity via sustained metal ion delivery (e.g., Ag+, Zn2+, Cu2+).7–10 Their structural versatility, achieved through tailored ligand design and synthesis strategies, permits precise modulation of pore geometry, functionality, and biocompatibility.11,12 Enhanced antimicrobial efficacy is realized through advanced mechanisms such as photocatalytic reactive oxygen species (ROS) generation and stimuli-responsive agent release.13 Engineered defects in MOFs further optimize cargo loading capacity and enable targeted delivery under environmental triggers.14–16 When integrated with nanotechnology, MOFs exhibit synergistic antibacterial effects, reduced off-target toxicity, and precision in infection control, positioning them as innovative solutions for combating resistant pathogens.17–21

Another highly successful method for combating antibacterial resistance is the use of metal nanoparticles (MNPs).22 These materials have shown excellent results both independently and in combination with other approaches, playing a very significant role in this field.23 These particles can physically disrupt bacterial membranes, produce reactive oxygen species (ROS) that damage cellular components, and inhibit critical processes like RNA and protein synthesis, biofilm formation, and membrane potential.24–26 Advances in technology have enabled precise control over nanoparticle size, shape, surface charges, and functionalization, enhancing their efficacy and safety in combating multi-drug-resistant bacterial infections.27,28 MNPs also exhibit synergistic effects when combined with conventional antibiotics, reducing resistance and improving therapeutic outcomes by lowering dosage requirements and minimizing side effects.29 Intermetallic nanoparticles, formed by combining different metals, have demonstrated superior antibacterial properties compared to monometallic forms, further broadening their biomedical applications.30,31 With their stability, cost-effectiveness, and ability to enhance drug solubility and efficacy, MNPs represent a promising alternative to traditional antibiotics, offering versatile and multi-faceted approaches to tackle antibiotic-resistant pathogens.32

In recent years, photodynamic therapy (PDT) has emerged as a promising and innovative approach to combat resistant bacteria by using light to activate photosensitizers (PS), which produce reactive oxygen species (ROS) to selectively destroy target cells without inducing bacterial resistance.33–35 Widely utilized in fields such as dermatology, oncology, and infectious disease, PDT combines the localized or systemic application of PS compounds with light irradiation to achieve precise microbial inactivation. Advances in nanomedicine have significantly improved PDT's efficacy by enhancing biocompatibility, safety, and site-specific enrichment.36–38

Beyond these metal-based strategies, a host of other innovations is emerging in the fight against resistant bacteria.39 Antimicrobial peptides (AMPs), for instance, inspired by natural immune defenses, offer a promising solution to combat bacterial resistance through their unique ability to selectively target bacterial membranes. These diverse small proteins, also known as cationic host defense peptides, are found in animals, plants, bacteria, and yeast, and can also be synthesized in laboratories.40,41 AMPs exhibit broad-spectrum antimicrobial, antiviral, antifungal, and anti-mitogenic properties. Alongside their roles as immune modulators and anti-inflammatory agents, AMPs have potential as alternatives to conventional drugs.42 Their mechanism of action, based on membrane destruction, minimizes the risk of inducing bacterial resistance, and they have been effectively demonstrated in both in vitro and in vivo models.43–45 AMPs hold significant promise for applications in clinical antibacterial treatments, animal and plant disease resistance, and as food preservatives.46–49 However, challenges such as high production costs, susceptibility to clearance, and potential adverse reactions currently limit their widespread use. Strategies like combining AMPs with metal ions, self-assembly to enhance stability, and developing responsive and synergized forms aim to overcome these hurdles and expand their therapeutic potential.50,51

Addressing antibacterial resistance requires a multifaceted approach that combines diverse strategies and leverages complementary mechanisms for more effective and sustainable outcomes. The focus of antimicrobial research has shifted from merely killing bacteria to combating antibiotic-resistant pathogens. Combination therapies are also emerging as a key solution.52,53 Using multiple drugs together reduces individual drug dosages, minimizes side effects, lowers the risk of resistance development, and achieves synergistic effects with enhanced antibacterial efficacy.54 Additionally, combination therapies offer wide-spectrum action and the ability to target multiple sites simultaneously, making them a powerful tool in the fight against resistant infections.55

This review critically examines four key non-antibiotic strategies: metal–organic frameworks (MOFs), metal nanoparticles (MNPs), photodynamic therapy (PDT), and antimicrobial peptides (AMPs). Rather than simply summarizing the existing literature, we provide a novel integrative analysis of these strategies. Our unique contribution lies in synthesizing these diverse fields by connecting fundamental molecular mechanisms to their potential applications and the challenges of translating these findings into practice. We emphasize how innovative synthesis methods, such as the modulated synthesis of MOF nanocomposites, significantly influence key parameters, including antibacterial efficacy, metal ion release kinetics, and biocompatibility. Additionally, this review critically evaluates the synergistic potential of combining these approaches, an important frontier for overcoming the limitations of single-mode therapies. By integrating perspectives from materials science, nanotechnology, photochemistry, and peptide engineering, we propose a comprehensive framework for developing the next generation of antimicrobials. We conclude with concrete and innovative integrated systems for future research, aiming to stimulate the interdisciplinary collaboration necessary to translate these promising materials from the laboratory to clinical practice.

2. Mechanisms of antibacterial resistance

When bacteria learn to withstand exposure to antibiotics that would typically kill them or stop their growth, antibacterial resistance develops.56,57 This process arises from the development of resistance genes through horizontal gene transfer and genetic alterations.58 Overcoming bacterial resistance requires an understanding of the mechanisms by which resistance is achieved.59 Bacteria can be broadly classified as either Gram-positive or Gram-negative, according to the structure of their cell walls. Gram-negative bacteria have a thinner peptidoglycan layer than Gram-positive bacteria because they have a different outer membrane.60 Examples of common multidrug-resistant (MDR) pathogens include Gram-negative bacteria such as Escherichia coli, Pseudomonas aeruginosa, Acinetobacter baumannii, Klebsiella pneumoniae, and Serratia marcescens, as well as Gram-positive bacteria such as Staphylococcus aureus, Streptococcus pneumoniae, and Staphylococcus epidermidis.61 Table 1 includes some examples of commonly encountered multidrug-resistant pathogens associated with healthcare. The antibacterial activity of conventional antibiotics, such as penicillin, rifampicin, and tetracycline, stems from their ability to inhibit cell wall synthesis or interfere with DNA, RNA, or protein synthesis. However, bacteria have developed a range of resistance mechanisms (Fig. 1).57,62 Many bacteria produce enzymes that deactivate antibiotics. For example, Enterobacteriaceae, such as E. coli and K. pneumoniae, produce extended-spectrum beta-lactamases (ESBLs) that hydrolyze beta-lactam antibiotics.63 Aminoglycoside-modifying enzymes, chemically alter antibiotics, diminishing their ability to bind to bacterial targets.64 In addition, mutations in bacterial proteins can also prevent antibiotics from binding effectively. For instance, modifications in penicillin-binding proteins (PBPs) reduce the efficacy of beta-lactam antibiotics,65 while mutations in DNA gyrase or topoisomerase IV confer resistance to fluoroquinolones.66 Efflux pumps actively expel antibiotics from bacterial cells, reducing intracellular concentrations. The AcrAB-TolC efflux system in Gram-negative bacteria is an example that confers resistance to multiple drug classes.67 In another way, bacteria acquire resistance genes through conjugation, transformation, or transduction. Mobile genetic elements like plasmids and integrons facilitate the rapid spread of resistance within and across bacterial species.68 Moreover, a lot of studies have indicated that bacteria in biofilms are embedded in a self-produced matrix that shields them from antibiotics. Biofilms contribute to 80% of bacterial infections and are a key factor in the persistence of infections.69 They reduce drug penetration and provide a protective environment for dormant bacterial cells, making them highly resistant to treatment.70
Table 1 Monitoring list for healthcare-associated multidrug-resistant and extensively drug-resistant pathogens (key indicator organisms)
Bacteria Common resistance to Ref.
a ESBL, extended-spectrum b-lactamase.b MRSA, meticillin-resistant S. aureus.
Acinetobacter Ceftazidime, aminoglycosides, fluoroquinolones, carbapenems 72
Enterobacteriaceae (e.g. Escherichia coli, Klebsiella pneumoniae) Cephalosporins (aESBL-producers), fluoroquinolones, aminoglycosides 73
Cephalosporins (aESBL-producers), fluoroquinolones, aminoglycosides, carbapenems
Enterococcus spp. (especially E. faecium) Ampicillin, aminoglycosides (high-level) 74
Ampicillin, aminoglycosides (high-level), glycopeptides
Pseudomonas aeruginosa Piperacillin/tazobactam, ceftazidime, ciprofloxacin, aminoglycosides 75
Piperacillin/tazobactam, ceftazidime, ciprofloxacin, aminoglycosides, carbapenems
Staphylococcus aureus (healthcare-associated) b-Lactam antibiotics (except new anti-bMRSA cephalosporins), macrolides, fluoroquinolones, aminoglycosides 76



image file: d5ma00552c-f1.tif
Fig. 1 Main antibiotic targets and associated mechanisms of resistance.77 Reproduced under the terms of the Creative Commons Attribution 2.0 International Public License (CC BY 2.0). Copyright 2010, the Authors. Published by BioMed Central Ltd.

Understanding these diverse mechanisms is critical for addressing the resistance crisis and guiding the development of next-generation antimicrobial strategies.71 Effective solutions must consider not only the biological intricacies of resistance but also the environmental and societal factors that contribute to its rise.

3. Metal–organic frameworks (MOFs) as antibacterial agents

Metal–organic frameworks (MOFs) represent a groundbreaking class of low-density, crystalline porous materials constructed from metal nodes (metal ions or clusters) and organic linkers.78–80 Their high surface area, adjustable porosity, and functional versatility make them promising candidates for a variety of applications, including catalysis, sensing, gas storage, and biomedical uses.14,79,81–87 In the fight against antibacterial resistance, MOFs present unique advantages: they can release bactericidal ions in a controlled way, generate reactive oxygen species (ROS), or serve as carriers for antimicrobial drugs, providing multifunctional strategies to combat pathogens.88–91 Table 2 provides a summary of various metal–organic frameworks (MOFs) assessed for antibacterial activity. It includes details about their metal ions or cores, ligands, target bacterial strains, and activity indicators, as well as the experimental conditions used. Commonly used metal nodes in MOFs include silver, zinc, copper, and cobalt, all of which exhibit intrinsic antibacterial properties against both Gram-positive and Gram-negative bacteria, such as E. coli and S. aureus.92–94 Compared to traditional antibiotics, MOFs offer several distinct benefits: (i) they can be constructed with bactericidal ions and antimicrobial ligands;95,96 (ii) their chelation effect enhances lipophilicity and improves membrane penetration;97,98 (iii) their high porosity allows for significant drug loading;99 and (iv) their electronic tunability enables efficient photocatalytic ROS generation.100–102
Table 2 Summarizes representative MOFs evaluated for antibacterial activity
MOF/composite Metal ion Ligand Target bacteria Activity indicators Experimental conditions Ref.
Cu-MOF NPs Cu2+ Trimesic acid E. coli, S. aureus, Pseudomonas, Klebsiella, Candida 100 μg mL−1; outperformed ampicillin/ciprofloxacin Nutrient broth, 106 CFU per mL, 24 h, 37 °C 106
Zn-MOFs Zn2+ Terephthalate S. aureus, E. coli Zn-MOF strongest via Zn2+ release Broth culture 104
Ni-MOFs Ni2+ Organic linker ESBL E. coli, P. aeruginosa, MRSA IC50 = 15–25 μg mL−1; biofilm inhibition Standard broth 107
ZIF-4, ZIF-7, ZIF-8 Zn2+ Imidazolate S. aureus, E. coli ZIF-8 strongest; ZnO derived from ZIF-4 enhanced activity Normal broth 91
Zr-TCPP (PCN-224, Cu2+-doped) Zr4+ (Cu2+-doped) Porphyrin (TCPP) S. aureus 99.71% inhibition Light irradiation (660 nm, 20 min) 109
ZIF-8-PAA-MB@AgNPs@Van-PEG Zn2+ Imidazolate E. coli, S. aureus, MRSA Synergistic killing under laser irradiation Laser irradiation 120
UIO-66-NH2@PEG Zr4+ Amino-terephthalate VRSA Superior antibacterial & anti-biofilm vs. free drugs Standard broth 114
Zr-MOF-1/2 (CIP delivery) Zr4+ Organic linker E. coli (CIP delivery) Sustained release over 7 days, pH-responsive pH 9.2 > neutral/acid; 7-day release study 113


MOFs exhibit their antibacterial properties through multiple mechanisms:

(1) Release of metal ions: many MOFs are designed to release bioactive metal ions, such as zinc, copper, or silver, which can interfere with bacterial cell walls, membranes, and intracellular components. These ions can disrupt enzymatic processes and promote the production of reactive oxygen species (ROS), which cause oxidative damage to bacterial cells.103–105 In 2018, Sheta et al. prepared Cu-MOF nanoparticles (Cu-MOF-NPs) and tested them against E. coli, S. aureus, Pseudomonas, Klebsiella, and Candida spp. at 100 μg mL−1. Experiments were conducted in nutrient broth with an initial inoculum of ∼106 CFU per mL, incubated for 24 h at 37 °C, and compared to standard antimicrobial agents (ampicillin, ciprofloxacin, amphotericin B). Under these conditions, Cu-MOF-NPs matched or outperformed the reference drugs.106 In 2021, Nakhaei et al. demonstrated that three zinc-terephthalate MOFs (MOF-5, Zn-MOF, TMU-3) exhibited antibacterial activity against S. aureus and E. coli, with Zn-MOF showing the highest efficacy due to its enhanced Zn2+ ion release, particularly in its activated form, which disrupts bacterial cell integrity through sustained metal ion release.104 Prabhu et al. later developed Ni-MOFs, proving their potent antimicrobial effectiveness against extended-spectrum beta-lactamase (ESBL) strains such as ESBL-1 and P. aeruginosa. These materials also inhibited biofilm formation by the MRSA strain ATCC 33591 and clinical strain N7, with IC50 values of 15.19 ± 1.41 μg mL−1 and 25.14 ± 0.75 μg mL−1, respectively. The antimicrobial effect was attributed to the positively charged Ni2+ ions interacting with the negatively charged bacterial cell walls, generating ROS to kill the bacteria, combined with a synergistic effect from the organic linker in the Ni-MOF.107 In 2024, Khatami et al. demonstrated that three zinc-based zeolitic-imidazolate frameworks (ZIF-4, ZIF-7, ZIF-8) exhibited antibacterial activity against S. aureus and E. coli, with ZIF-8 showing the strongest effect due to its high Zn2+ ion release. ZnO nanoparticles derived from ZIF-4 further enhanced antibacterial performance, attributed to their uniform nanostructure and controlled ion release.91 Despite their significant antibacterial potential, many MOFs face limitations due to the inherent toxicity and the low biocompatibility of certain metal ions and organic ligands. To expand their bio-applications, it is crucial to develop strategies for controlled release and efficient removal of excess metal ions and ligands, as well as to explore the use of biologically derived ligands to enhance their safety.

(2) ROS generation: MOFs can catalyze the production of ROS under specific conditions, such as light activation. These ROS can damage bacterial DNA, proteins, and lipids, leading to cell death.108 Han et al. introduced a Cu2+-doped Zr-based porphyrinic MOF (Zr-TCPP, PCN-224; TCPP = 5,10,15,20-tetrakis(4-carboxyphenyl) porphyrin) with remarkable bacteriostatic efficiency, achieving 99.71% inhibition of S. aureus within 20 minutes under 660 nm light irradiation. The doped Cu2+ ions played a key role by trapping electrons, enhancing carrier transfer, reducing electron–hole recombination, and converting absorbed light energy into heat. This process amplified ROS generation and photothermal effects, contributing to its strong antimicrobial activity.109 In recent years, MOFs have increasingly been employed as photocatalysts for in vitro photocatalytic disinfection. This method relies on in situ ROS generation to degrade bacteria into CO2 and H2O. Li et al. compared five MOFs (MIL-100, NH2-MIL-125, NH2-UiO-66, ZIF-11, and ZIF-8) for photocatalytic inactivation of E. coli; ZIF-8 (Zn2+/Hmim) achieved >99.9999% reduction within 2 h in saline under simulated solar light (Xe lamp, AM 1.5, 100 mW cm−2). Blank controls (light only, ZIF-8 in dark) showed negligible killing, while ZnO and TiO2 achieved only ∼2–3 log reductions, confirming ZIF-8's superior photocatalytic activity.110 Importantly, ROS generation is not limited to light-activated mechanisms. Hamarawf et al. demonstrated that Zn- and Co-MOFs could exert potent antibacterial and antibiofilm effects through metal-ion-mediated ROS generation without the need for external light activation. The study proposed that the electrostatic attachment of the MOFs to bacterial cells initiates a process of lipid peroxidation, ultimately leading to lethal ROS production within the bacteria. This mechanism highlights an intrinsic reactive property of certain MOFs that can be harnessed for antimicrobial purposes, independent of photocatalysis.111 Inspired by these results, ZIF-8 was used to create a MOF-based filter mask for integrated pollution control. Experimental findings demonstrated that the antibacterial performance of this MOF-based mask surpassed that of commercial masks, offering innovative prospects for using porous materials in public health protection.111 (3) Drug and antibacterial delivery vehicles: MOFs serve as carriers for antibiotics or other antimicrobial agents, enhancing their solubility, stability, and targeted delivery.112 This approach not only increases the effectiveness of the drugs but also reduces the required dosage and potential side effects. Ishfaq et al. investigated zirconium-based MOFs, Zr-MOF-1 and Zr-MOF-2, as pH-responsive carriers for the antibiotic ciprofloxacin (CIP). They found that CIP was released more quickly in a basic medium (pH = 9.2) compared to neutral or acidic conditions. Zr-MOF-2 exhibited sustained release over seven days, suggesting its potential for long-acting formulations in alkaline infections.113 Also, Rahmanian et al. developed a PEG-coated UIO-66-NH2 nanoparticle for the co-delivery of vancomycin and amikacin against vancomycin-resistant Staphylococcus aureus (VRSA). The dual-drug-loaded nano-system (VAN/AMK-UIO-66-NH2@PEG) exhibited superior antibacterial and anti-biofilm activity compared to the free drugs. Crucially, it also significantly downregulated the expression of key resistance (vanA, mecA) and biofilm-forming (icaA, icaD) genes in VRSA isolates. This study highlights the potential of engineered MOFs to overcome resistant pathogens by simultaneously delivering multiple antibiotics and suppressing resistance mechanisms at a genetic level.114

Silver (Ag) nanoparticles (Ag NPs) have garnered significant attention for their broad-spectrum antimicrobial properties against bacteria, viruses, and fungi. Their effectiveness arises from direct interaction with microorganisms, the release of Ag+ ions, and reactive oxygen species (ROS) generation. However, excessive Ag+ release can harm normal tissues, making controlled release a crucial strategy for their use as antimicrobial agents. For instance, Guo et al. developed Ag-CuTCPP by synthesizing CuTCPP and encapsulating Ag NPs. This MOF material demonstrated superior antibacterial performance compared to penicillin against S. aureus, B. subtilis, and E. coli, with lower cytotoxicity than Ag+ or standalone Ag NPs.112,115 Similarly, Salam et al. synthesized Ag NPs@Ni-MOF, a Ni-MOF nanosheet loaded with Ag NPs. This composite showed enhanced antimicrobial effects compared to Ni-MOF alone, achieving inhibition rates of 93.85%, 92.15%, 87.43%, and 84.07% against B. subtilis, E. coli, P. aeruginosa, and C. albicans, respectively, due to the additional release of Ag NPs.116 Soltani et al. developed a Cu-BTC MOF loaded with chlorhexidine (CHX@Cu-BTC), which exhibited enhanced antibacterial activity against S. aureus and E. coli through synergistic effects from controlled release of Cu2+ ions and CHX, achieving lower MIC values compared to individual components.117 In another work, they embedded gemifloxacin (GEM) into ZIF-8 via a one-step aqueous synthesis, achieving remarkable drug loading (DLC = 69.82%, DLE = 89.03%). The GEM@ZIF-8 system exhibited H2O2-responsive release (47.7% cumulative release in infected tissue conditions) and maintained potent antibacterial activity (MIC < 0.6 μg mL−1) comparable to free GEM, while enabling sustained, targeted delivery to reduce off-target effects.118 Arenas-Vivo et al. proposed a photoactive composite coating, Ag-encapsulated MIL-125(Ti)–NH2, which achieved an impressive 99.9999% inhibition rate against S. aureus biofilms. This efficacy was attributed to the combined antibacterial effects of the MOF, Ag NPs, and photoactivity under UVA light. In recent years, MOFs have gained interest for their ability to dynamically target antibacterial agents to specific microenvironments.119 These systems respond to endogenous stimuli such as light, acidity, or oxidative stress associated with bacterial infections. For example, Chen et al. developed a composite nanomaterial, ZIF-8-PAA-MB@AgNPs@Van-PEG. The encapsulation of methylene blue (MB) into ZIF-8-PAA imparted light and pH responsiveness. Ag NPs were then formed on the ZIF-8-PAA-MB via in situ reduction of AgNO3, followed by the addition of Van-PEG to load antibacterial agents.120,121 This composite exhibited potent antibacterial activity against E. coli, S. aureus, and MRSA due to the synergistic effects of vancomycin and ROS generation under laser irradiation.121

Antibacterial performance depends strongly on MOF composition. Zn-based frameworks often show strong activity due to sustained Zn2+ release; Cu-based frameworks are effective via both ion release and photothermal contributions; Ni-based MOFs excel in biofilm inhibition due to strong electrostatic interactions; and Ag-based MOFs provide broad-spectrum efficacy but raise cytotoxicity concerns. Understanding these relationships provides guidance for designing safer, more effective MOFs. Despite their significant promise, challenges in stability, cytotoxicity, and scalable production must be addressed for the successful clinical translation of metal–organic frameworks (MOFs). To overcome these hurdles, future efforts could focus on integrating biologically derived ligands (e.g., amino acid- or peptide-based linkers) to reduce toxicity, and combining experimental microbiology with computational modeling to predict ion release and reactive oxygen species (ROS) efficiency. By leveraging their inherent biodegradability, diverse functionality, and molecular adaptability, MOFs can be rationally designed to become a cornerstone of next-generation antimicrobial strategies, offering sustainable solutions to the growing crisis of antibacterial resistance.

4. Metal nanoparticles and their antibacterial mechanisms

The antibacterial mechanisms of metal nanoparticles (MNPs) are multifaceted and complex, involving physical interactions, chemical properties, and reactive processes that contribute to their bactericidal activity.122 The precise mechanisms of their activity are not yet fully understood, although significant research efforts have been directed at elucidating these processes (Table 3).123 MNPs exhibit antibacterial effects through various pathways, including physical damage to the bacterial cell wall and membrane, ion leaching, and reactive oxygen species (ROS) production.124 Physical interactions involve the adsorption and penetration of NPs into bacterial cell walls, which disrupts membrane integrity, depolarizes the cell wall, and leads to leakage of intracellular components.125 Positively charged NPs show enhanced bactericidal activity due to electrostatic interactions with the negatively charged bacterial cell surface.126 Additionally, the size and shape of NPs are critical factors; smaller NPs with higher surface-area-to-volume ratios penetrate cells more effectively and produce more ROS, which induces oxidative stress and damages essential biomolecules like lipids, proteins, and DNA.127 Zare et al. synthesized ZnO NPs in various sizes and shapes, and their antibacterial and antioxidant activities were found to be dependent on both size and morphology.128 Similarly, Korshed et al. discovered an inverse relationship between the size of NPs and their bactericidal effects.129 They also found that smaller Ag NPs generated more reactive oxygen species (ROS) than larger ones. In another study, it was shown that 18 nm Ag NPs were more toxic than 80 nm Ag NPs in water, though their toxicity became similar when tested in PBS buffer.130
Table 3 The main mechanisms of bacterial resistance to NPs
Bacteria Nanoparticles (size in nm) Resistance mechanisms Year Ref.
NS = not specified.
B. subtilis, S. aureus, E. coli Nanoscale zero-valent iron (NS) ROS response 2013 157
E. coli ZnO nanorod (diameter ∼ 45 and length ∼ 250) Changes in plasma membrane 2013 158
E. coli Ag NP (10–30) Production of extracellular substance 2014 159
Mycobacterium smegmatis Ag NP (21.7) Genetic changes 2014 160
B. subtilis Ag NP (8.3) ROS response, production of extracellular substances, quorum sensing, stress response 2015 161
E. coli SiO2 NP (15) Production of extracellular substances 2016 162
P. aeruginosa PAO1 CuO NP (<50) Genetic changes, regulation of porins, metal efflux transporters 2017 163
E. coli ZnO NP (18) Adaptive morphogenesis, regulation of porins 2018 164
Pseudomonas CeO2 NP (50) Biofilm formation, stress response 2019 165
S. aureus Ag NP (18) ROS response, genetic changes 2020 166
E. coli Ag NP (NS) Changes in plasma membrane, genetic changes 2021 167
E. faecalis Ag NP (10) Production of extracellular substances, ROS response, 2022 168


In recent studies, surface functionalization and charge modulation have been shown to significantly affect NP biocompatibility and antimicrobial properties.131 For example, positively charged NPs demonstrate higher affinity for bacterial cells, enhancing their bactericidal potential.132 Conversely, bacterial cells can adapt by modifying their surface charges or employing efflux systems to reduce NP toxicity.133 El Badawy et al. investigated the toxicity of four types of Ag NPs with various surface charges, from highly negative to highly positive, and concluded that the toxicity of Ag NPs was dependent on the surface charge when tested against different bacterial species.134

ROS production remains a central mechanism in NP-induced antibacterial activity. These reactive species disrupt bacterial membranes, degrade proteins and nucleic acids, and inhibit metabolic functions, leading to cell death.135 Certain metal NPs, such as silver, zinc oxide, and titanium dioxide, are particularly effective at ROS generation.136 Additionally, nanocomposites like silver-copper NPs have demonstrated enhanced ROS-mediated antibacterial activity.137 Wang et al. reported that the bactericidal effect of Ag/CeO2 nanoparticles on E. coli was predominantly attributed to intracellular ROS generation and the disruption of the cell wall and membrane, rather than the release of silver ions.138 Similarly, Mujeeb et al. demonstrated that silver–copper nanocomposites (Ag–Cu NCs) synthesized using Olax scandens leaf extract exhibited superior antimicrobial activity compared to monometallic Ag NPs, primarily due to enhanced ROS production.139

The chemical properties of NPs also play a vital role in their antibacterial activity. Metal ions released from NPs interact with bacterial phospholipid layers and interfere with intracellular biomacromolecules such as DNA and enzymes.140 Transition metals (e.g., Ag, Zn, Cu) and metalloids (e.g., Se, Te) are particularly effective due to their ability to release ions that disrupt cellular processes. The dissolution of NPs in acidic conditions can enhance ion release and bactericidal activity, while ROS production—induced by NPs under various conditions—leads to cell wall damage, membrane permeability interference, and metabolic pathway disruption.141 For instance, Moreau et al. observed that ZnO NPs dissolved more readily under acidic conditions, leading to an increased release of Zn2+ ions.142 Similarly, Saliani et al. found that the antibacterial effect of ZnO NPs was stronger when the pH dropped from 7 to more acidic levels.143 Peretyazhko et al. noted that when Ag NPs are released into aquatic environments, they undergo oxidative dissolution, which results in the release of Ag+ ions and triggers antibacterial activity.144

The shape of NPs influences their antibacterial activity as well. Spherical, rod-shaped, and cubic NPs each exhibit different levels of effectiveness, with nanocubes and nanorods often showing higher bactericidal properties due to their exposed crystal planes and oxidation capabilities.145 Structural features like corners, edges, and defects further enhance the interaction of NPs with bacterial cells, increasing their toxicity.146 Huynh et al. found that gold nanostars could replace antibiotics in acne treatment due to their strong bactericidal effect against propionibacterium acne.135 Additionally, Hong et al. discovered that Ag nanowires had lower antibacterial effectiveness compared to Ag nanocubes and nanospheres, likely due to reduced interaction with bacterial cells.147 The same study also showed that silver nanocubes outperformed nanospheres in antimicrobial activity, as they provided larger contact areas and had more reactive facets.145

MNPs' ability to disrupt bacterial biofilms and prevent bacterial adhesion on surfaces has inspired the development of artificial antimicrobial surfaces.148 By leveraging the natural bactericidal properties of nanostructures, researchers have created advanced materials with enhanced antimicrobial activity for medical and commercial applications.149 Incorporating gold nanoparticles (Au NPs) into PMMA-based bone cement enhances the polymer matrix's mechanical properties and reduces Staphylococcus aureus biofilm formation. Reducing Au NPs to nanoclusters (NCs) within the 1–2 nm range significantly improves antimicrobial activity against Gram-positive and Gram-negative bacteria by increasing ROS levels while maintaining low cytotoxicity and genotoxicity in host cells.150

Pathogenic bacteria have evolved mechanisms to evade the immune system, making intracellular infections difficult to treat with conventional antibiotics due to limitations such as poor cellular permeability, low retention, and instability in mammalian cells.151 Nanotechnology offers an innovative solution by enabling targeted delivery of antimicrobial agents to both extracellular and intracellular pathogens.152 Metallic nanoparticles (MNPs) can act as carriers for antibiotics, improving pharmacokinetics, targeting infection sites, and enabling controlled drug release. Studies highlight the synergistic antibacterial effects of MNP–antibiotic composites.148 Fe3O4 nanoparticles (NPs) can enter Gram-negative bacteria via siderophore channels located in their outer membrane. These nanoparticles can act as “Trojan horses,” facilitating the delivery of antibiotics attached to them, which are typically obstructed by the bacterial outer membrane.153 Remarkable efficacy has also been observed against biofilms. Research by Ali et al. demonstrated that α-Fe2O3 nanoparticles interact with the extracellular polymeric substances (EPS) of biofilms and penetrate bacterial cells, inhibiting their growth by generating reactive oxygen species (ROS) within the cells.154

Nanoparticles can be functionalized in various ways to design advanced drug delivery systems. Drugs can be loaded via noncovalent binding, allowing efficient release without specific bond cleavage. Alternatively, covalent binding can link therapeutic, targeting, and functionalization agents to nanoparticles, functioning like prodrugs that release their payload upon specific stimuli. The use of metallic nanoparticles as drug carriers have been shown to significantly enhance the antibacterial activity of antibiotics, providing a promising approach to combat resistant pathogens and improve therapeutic outcomes.155 Turki Al Hagbani and his team incorporated vancomycin into gold nanoparticles (AuNPs) using a simple one-pot method to create V-GNPs. In vitro antibacterial tests revealed that V-GNPs exhibited significantly stronger antibacterial activity compared to vancomycin alone against various bacterial strains.156 Specifically, the inhibitory effectiveness of V-GNPs was 1.4 times greater against Escherichia coli, 1.6 times higher against Klebsiella oxytoca, 1.8 times more effective against Pseudomonas aeruginosa, and 1.6 times more potent against Staphylococcus aureus.

Despite their potential, the long-term stability and resistance mechanisms of bacteria against NPs pose challenges. Environmental factors, such as pH and medium composition, influence NP dissolution and ion release. Moreover, bacterial resistance strategies, including efflux systems and surface modifications, can diminish NP effectiveness over time. Further research is required to optimize their use and address potential resistance and cytotoxicity concerns.

Future advances should focus on developing biodegradable or self-degrading MNPs to minimize long-term environmental accumulation. Combining nanomaterial science with microbiology and toxicology can provide a clearer understanding of how MNPs interact with microbial membranes versus human cells, enabling selective antibacterial action while ensuring biosafety.

5. Photodynamic therapy-based synergistic antibacterial approach

Antimicrobial photodynamic therapy (APDT) represents an innovative approach for combating bacterial infections, particularly multidrug-resistant (MDR) strains.169,170 The mechanism of APDT relies on the interaction of light, photosensitizer (PS) molecules, and molecular oxygen, producing reactive oxygen species (ROS) or reactive molecular species (RMS) that exert a bactericidal or bacteriostatic effect through two primary pathways: type I and type II mechanisms.171 In the type I mechanism, light-activated PS molecules transition from a ground state to an excited singlet state and subsequently to a triplet state. From the triplet state, they transfer electrons or hydrogen atoms directly to surrounding substrates, forming free radicals such as hydroxyl radicals (˙OH) and superoxide anions (O2˙).172 These radicals disrupt bacterial cell membranes by initiating lipid peroxidation, leading to structural damage and increased ion permeability.173 The type II mechanism involves the transfer of energy from the triplet state PS molecules to oxygen, generating singlet oxygen (1O2), a highly reactive form of oxygen. This singlet oxygen oxidatively damages key bacterial biomolecules, including unsaturated lipids, proteins, and enzymes, effectively killing bacteria and weakening the structural integrity of biofilms (Fig. 2).172,174
image file: d5ma00552c-f2.tif
Fig. 2 Jablonski diagram to depict the type I and II mechanisms of APDT.177 Reproduced under the terms of Creative Commons Attribution 4.0 International (CC BY 4.0) license. Copyright 2022, the Authors. Published in MDPI.

The multitargeted nature of ROS generated by APDT enables the therapy to attack various cellular structures, including cell membranes, cell walls, and internal biomolecules such as DNA and proteins.175 This broad-spectrum activity ensures effectiveness against bacteria regardless of antibiotic resistance mechanisms, with a lower risk of inducing further resistance compared to conventional antibiotics.176 Additionally, ROS production within biofilms weakens the extracellular polymeric substance (EPS) matrix, reduces biofilm adhesion, and compromises pathogen metabolic activity, facilitating bacterial eradication.70

Photodynamic therapy (PDT) has been combined with various non-invasive treatment methods, demonstrating additive and synergistic effects that enhance outcomes in numerous in vitro studies, as well as in preclinical and clinical applications.178 The approaches that photodynamic antibacterial therapy can integrate with can be categorized into six types: antibiotics, antibacterial agents, chemotherapy, photothermal therapy (PTT), nitric oxide (NO), and enhanced photosensitizers (PS). Table 4 provides a summary of the advantages and disadvantages of these methods.179–183 These represent non-nanomaterial-based APDT synergistic antibacterial strategies. Among these, photodynamic antibiotic therapy (PACT) has emerged as a promising synergistic approach for bacterial inactivation.180,184,185 In a study investigating the synergistic antibacterial effects of methylene blue (MB)-mediated photodynamic therapy (PDT) combined with antibiotics, Shih et al. assessed its efficacy against Mycobacterium avium keratitis. Using the micro-broth dilution method, they evaluated the bactericidal impact of combining MB-mediated PDT with antibiotics such as ciprofloxacin, moxifloxacin, and amikacin. The findings revealed that phototoxicity initially targets the cytoplasm during sterilization, followed by cell wall lysis, ultimately leading to the destruction of M. avium.186 Almeida et al. assessed the antimicrobial effectiveness of APDT combined with antibiotics (ampicillin and chloramphenicol) and the surfactant SDS, utilizing cationic porphyrins as photosensitizers (PSs). The study was conducted in synergy with either phosphate buffer or filtered hospital wastewater containing multidrug-resistant bacteria. The results showed that at both subinhibitory and inhibitory concentrations, the combination of APDT and antibiotics led to a faster reduction in bacterial survival.187 Hou et al. developed a polymeric antimicrobial agent designed to synergize chemotherapeutic and photodynamic therapy for combating drug-resistant bacterial infections. In this approach, amphiphilic polyaspartic acid-block-polycaprolactone polymeric micelles were used as carriers, with the photosensitizer protoporphyrin IX (PPIX) encapsulated in the micelle core. The micelle shell was then decorated with silver via in situ reduction. This polymeric antimicrobial agent demonstrated chemophotodynamic activity, effectively combining therapeutic strategies to eradicate drug-resistant bacterial infections.188 Non-nanomaterials also present certain limitations.189 Due to their lack of adjustable properties, it is difficult to effectively control the interaction of their physical and chemical characteristics and targeting capabilities.190 As a result, non-nanomaterial-based photodynamic antibacterial strategies need to be optimized by adopting features of nanomaterials.

Table 4 Comparison of combined PDT strategies
Strategy Advantages Disadvantages Ref.
PDT + antibiotics Lowers antibiotic dose; synergistic killing Phototoxicity, limited penetration 180
PDT + antibacterial agents Broadens antibacterial spectrum; multiple modes of action Possible drug interactions, risk of toxicity 178
PDT + NO donors Enhances ROS & NO synergy Limited NO delivery efficiency 179
PDT + PTT (photothermal therapy) Dual-mode killing; strong biofilm disruption Heat damage to normal tissue 182
PDT + chemotherapy Dual antibacterial–anticancer effect; versatile in infected tumors Systemic side effects; limited selectivity 195
PDT + advanced PS nanocarriers Targeted delivery, lower dark toxicity Complexity of nanomaterial design 179


On the other hand, subcategories of the nanomaterials-based APDT synergistic antibacterial strategy include nanoparticles (NPs)-mediated approaches, nanomaterials-based PDT/PTT, and composite nanofiber membrane-based methods.17,191,192 Bagchi et al. utilized squaraine (SQ) dye as a photosensitizer, covalently adsorbing it onto the surface of ZnO nanoparticles (ZnO NPs) to create ZnO-SQ nanohybrids. The photo-induced interfacial electron transfer (ET) process from the excited state of SQ to the ZnO conduction band enhanced the nanohybrids' reactive oxygen species (ROS) production, leading to a significant antibacterial effect against S. aureus. This synergistic mechanism, involving cell membrane disruption, nanoparticle internalization, and subsequent photo-induced intracellular ROS generation, enabled the nanohybrids to achieve 95% bacterial killing efficiency.193 Teng et al. incorporated iodine into ZIF-8 and immobilized it on microarc titanium oxide. The combination of NIR light-induced iodine release and ZIF-8-mediated ROS oxidative stress significantly boosted the antimicrobial effectiveness of this approach both in vitro and in vivo. Furthermore, this composite coating promotes the osteogenic differentiation of bone marrow stromal cells without compromising the osteogenic potential of the implant, alongside the enhanced antimicrobial effect. The immobilization of iodine on orthopedic implants using MOFs provides a synergistic antimicrobial effect against bacterial infections.194 Cai et al. developed a composite membrane of PCL/Cur@ZIF-8 with enhanced antimicrobial properties. The membrane incorporates the natural photosensitizer curcumin into the highly porous nanocrystals of ZIF-8 to improve curcumin's water solubility and stability. Upon release of zinc ions and curcumin, and under blue light irradiation, curcumin molecules generate singlet-state oxygen. The synergistic effect of zinc ions and singlet-state oxygen resulted in 99.9% inhibition of E. coli and S. aureus, as well as a 99.9% reduction in adherent flora when the Cur@ZIF-8 loading exceeded 15%. This composite membrane shows significant potential as an antimicrobial packaging material to extend the shelf life of fruits, meat, and other products.183

Antimicrobial photodynamic therapy is a promising strategy for combating infectious diseases caused by drug-resistant bacteria. Its key advantages include: (1) modifiability of light-controlled photosensitizers (PS), with high phototoxicity and low dark toxicity to reduce side effects;196 (2) broad-spectrum antibacterial properties due to nonspecific targeting, preferentially binding to bacteria at infection sites;197 (3) minimal damage to host cells, ensuring treatment safety;198 (4) reliability, convenience, and reusability; and (5) compatibility with other therapies like radiotherapy, chemotherapy, and photothermal therapy (PTT).199–201 However, PDT faces challenges in deep tissue application due to limited light penetration, which is affected by tissue thickness. Longer wavelengths have better tissue penetration but may not effectively promote ROS production due to low energy.182 Additionally, the short lifespan and limited reach of reactive oxygen species (ROS) limit PDT's effectiveness, while some PSs suffer from dark toxicity, poor stability, and low bacterial targeting.202–204 To improve PDT's practical application, strategies are needed to enhance its efficacy and address these limitations. The integration of PDT with immunotherapy and nanocarrier engineering represents a promising future direction. By combining light-activated antibacterial activity with immune stimulation and targeted delivery systems, interdisciplinary approaches may overcome the current limitations of tissue penetration and off-target phototoxicity.

6. Antimicrobial peptide-based multifunctional antibacterial strategies

Antimicrobial peptides (AMPs) are a diverse group of molecules characterized by their variation in chemical structures and amino acid compositions (Table 5).205–207 Typically, AMPs are concise, with lengths ranging from 12 to 50 amino acids. Around 50% of these amino acids are hydrophobic, contributing to interactions with microbial membranes. Another key feature of AMPs is their amphiphilic nature and a net positive charge, generally ranging from +2 to +11, although some naturally occurring AMPs are negatively charged, such as dermcidin and histatin. AMPs are classified based on their source, structure, activity, and amino acid composition.208,209 They possess advantages like low molecular weight, high solubility, thermal stability, low cytotoxicity, and environmental degradability, making them promising candidates for antimicrobial therapies. Unlike traditional antibiotics, AMPs degrade easily, reducing environmental pollution and resistance issues while serving as immune mediators.210–213
Table 5 A list of some antimicrobial peptides based on various classification criteria
AMP Sequence Origin Structure Activity Ref.
Piscidin FFHHIFRGIVHVGKTIHRLVTG Fish morone chrysops α-Helix Gram-negative bacteria 214
LL-37 LLGDFFRKSKEKIGKEFKRIVQRIKDFLRNLVPRTES Human (cathelicidin) α-Helix Broad-spectrum, including bacteria, fungi, and viruses 215
Nisin ILLSKFLRNWAILAILKWRNA Bacteria Lactococcus lactis Polycyclic β-sheet with loops Gram-positive bacteria 216
Defensin (HNP-1) ACYCRIPACIAGERRYGTCIYQGRLWAFCC Human (neutrophil) β-Sheet Broad-spectrum, including bacteria, fungi, viruses (s HIV, influenza) 217
Gramicidin A VGALAVVVWLWLWLWG Soil bacterium Brevibacillus brevis Linear Gram-positive bacteria 218
Melittin GIGAVLKVLTTGLPALISWIKRKRQQ Bee venom α-Helix Broad-spectrum, including bacteria, fungi, cancer 219


AMPs employ two primary mechanisms to kill bacteria: membrane targeting and intracellular activity.220 Membrane-targeting mechanisms include: (1) toroidal pore model: AMPs interact with lipids and water to form transient pores, maintaining the lipid bilayer's integrity while allowing ion and molecule passage, ultimately leading to cell death.221 This mechanism is displayed by magainin 2, and human cathelicidin LL-37.222 (2) Barrel-Stave model: AMPs assemble into oligomeric structures, forming transmembrane channels that disrupt the membrane's barrier function. Alamethicin, and ceratotoxins are examples of peptides acting by this mechanism.223,224 (3) Carpet model where AMPs cover the membrane surface like a detergent, disrupting lipid packing and causing membrane destabilization and lysis. Examples of peptides that likely operate in a detergent-like manner include dermaseptin S. aurein 1.2, and cecropin.225 AMPs also target bacterial cell walls, disrupting peptidoglycan synthesis in Gram-positive bacteria and lipopolysaccharide layers in Gram-negative bacteria.226 For instance, nisin binds lipid II to inhibit cell wall synthesis. In addition to membrane-targeting, AMPs exhibit intracellular activity by entering bacterial cells and interfering with vital processes. They damage nucleic acids (e.g., indolicidin unwinds bacterial DNA), inhibit protein synthesis (e.g., PrAMPs block elongation or termination during translation), and affect organelles (e.g., periplanetasin-4 disrupts mitochondria).227–230

Brevinin-1, a peptide obtained from frog skin secretions, was modified to improve its therapeutic efficacy by altering its net charge, structural conformation, and hydrophobicity. Both the peptide and its derivatives effectively inhibited biofilm formation by methicillin-resistant S. aureus and Enterococcus faecalis.231 Another example is the synthetic antimicrobial peptide P5, which has demonstrated activity against carbapenem-resistant Pseudomonas aeruginosa (CRPA). Its mode of action involves disrupting bacterial cell membranes, with experimental findings highlighting its significant role in biofilm eradication, making it a promising candidate for treating multi-resistant infections.232 The human cathelicidin peptide LL-37 has demonstrated effectiveness against biofilms formed by Staphylococcus aureus in both meticillin-sensitive Staphylococcus aureus (MSSA) and meticillin resistant Staphylococcus aureus (MRSA) strains and can serve as an adjunct therapy for wound infections where biofilm development plays a significant role.233 Cbf-K16, a cathelicidin-like antimicrobial peptide, shows strong antimicrobial effects against both Gram-positive and Gram-negative bacteria, along with notable anti-biofilm properties. It has demonstrated a promising synergistic interaction with ceftazidime or ampicillin against MRSA.234

Beyond conventional AMPs, novel approaches and strategies are emerging for the treatment of drug-resistant bacterial infections. The combination of antibiotics and AMPs is emerging as a potential therapeutic strategy to combat antibiotic resistance, enhance bacterial killing, and reduce toxicity and side effects.235 This approach aims to minimize adverse effects, increase compound selectivity, improve bacterial membrane permeability, and decrease the efflux of antibiotics, thereby inhibiting bacterial survival.236 Peng et al. developed antibiotic-conjugated antimicrobial lipopeptides from paenipeptin C′ and ciprofloxacin. This design operates through a dual mechanism: the AMP disrupts the bacterial membrane, allowing the antibiotic to enter and inhibit targets like DNA gyrase.237 Similarly, combining conventional antibiotics with new synthetic peptides inspired by human cationic peptides such as LL-37 and thrombocidin-1 (TC-1) has shown synergistic antibacterial and anti-biofilm activity against S. aureus.238 Li et al. demonstrated that the combination of the tetracycline antibiotic demeclocycline hydrochloride (DMCT) and the AMP SAAP-148 exhibited synergistic antimicrobial activity against multidrug-resistant Pseudomonas aeruginosa strains PAO1 and ATCC27853.239 Additionally, Tarvirdipour et al. developed a powerful surface-coating technology based on rifampicin-loaded peptide multicompartment micelles (RIF-MCMs). Immobilized on a surface, these micelles provide a dual-function antimicrobial defense: they enable sustained, temperature-responsive release of antibiotics while simultaneously altering surface topography to passively inhibit bacterial adhesion and biofilm formation.240

The use of nanoparticle (NP)-conjugated systems for delivering AMPs has recently attracted attention. NPs offer a large surface area for AMP adsorption and help prevent AMP self-aggregation.241 Nanostructures are emerging as potential drug delivery carriers. As effective carriers, they must possess two key properties: non-cytotoxicity and non-immunogenicity. These nanostructures can be internalized into the cytoplasm without the need for transfectants, utilizing endocytosis and exocytosis pathways that are independent of multidrug efflux pumps. Nanotechnology-based approaches can enhance the stability and efficacy of AMPs while reducing toxicity to host tissue cells. Encapsulating AMPs in nanomaterials holds significant potential due to their small size, high surface area, and strong targeting capabilities.242,243 For instance, the proline-rich AMP dimer A3-APO and its single-chain metabolite (APO monomer) were tested in mice with burn wounds infected with MDR Acinetobacter baumannii, a strain isolated from an injured soldier. A dose of 5 mg kg−1 A3-APO significantly improved survival and reduced bacterial counts in the blood and wounds compared to other antibiotic treatments, including colistin and imipenem. This approach not only enhances the industrial utility and commercial viability of the product but also offers added value in smart biomedical applications.244

Despite the high potential of antimicrobial peptides (AMPs), only a few have been FDA-approved for clinical use. The development of AMPs involves a lengthy and complex process, including discovery, optimization, and clinical trials. However, several challenges hinder AMP advancement. These include unclear mechanisms of action, instability, and weak antibacterial activity.245 AMPs are vulnerable to degradation by proteases, sensitivity to pH, salt, and serum components, which can alter their structure and function. For example, salt ions can affect the antimicrobial activity by altering peptide conformation. Additionally, extreme pH conditions and serum proteins can impact AMP stability and efficacy.246,247 To address these challenges, various strategies, such as modifying peptide sequences, encapsulating AMPs in nanoparticles, or using dimerization, are being explored to enhance their stability, bioavailability, and therapeutic potential.248

AMPs are generally low in cytotoxicity, but their potential toxicity can vary depending on factors like peptide sequence, concentration, and the route of administration. Some AMPs, such as pore-forming peptides, can be toxic to human cells, especially at high concentrations, which can lead to secondary diseases.249 Hydrophobicity also plays a crucial role in antimicrobial activity, but excessive hydrophobicity may result in mammalian cell toxicity. Using drug carriers such as nanoparticles can reduce toxicity by targeting infected sites specifically. The administration route also impacts toxicity; systemic delivery may lead to more toxicity than topical applications.250

Lastly, the cost of producing AMPs is high due to the complex synthesis and purification processes required for peptides with long sequences. These peptides are more effective against a broader range of pathogens but come with challenges in terms of cost and scalability.251 The application of artificial intelligence and synthetic biology offers new opportunities to design next-generation AMPs with enhanced stability, reduced cytotoxicity, and lower production costs. By bridging computational peptide design with industrial biotechnology, more clinically viable and scalable AMP-based therapies can be achieved.

7. Conclusion

The increasing global threat of antibiotic resistance underscores the urgent need for alternatives to traditional antibiotics. Emerging technologies, particularly metal–organic frameworks (MOFs), metal nanoparticles (MNPs), photodynamic therapy (PDT), and antimicrobial peptides (AMPs), show significant promise as complementary strategies (Fig. 3). When used together, these approaches could potentially overcome the limitations of any single method, creating new pathways for sustainable antibacterial treatment.
image file: d5ma00552c-f3.tif
Fig. 3 Overview of Four Strategies to Combat Antimicrobial Resistanc.

Each strategy has distinct advantages but also faces critical challenges that hinder clinical application (Table 6). MOFs are tunable and multifunctional, yet concerns about in vivo degradation and potential immunotoxicity persist. MNPs offer broad-spectrum activity and strong synergy with antibiotics but raise issues regarding long-term environmental accumulation. PDT provides non-invasive, resistance-free bacterial inactivation; however, limited light penetration and phototoxicity restrict its use. AMPs uniquely disrupt bacterial membranes and evade conventional resistance mechanisms, but their high production costs and limited scalability pose challenges for industrial applications. Overcoming these hurdles requires an interdisciplinary approach integrating chemistry, biology, toxicology, and engineering.

Table 6 Comparison of the four methods: an overview
Strategy Main mechanism Strengths Weaknesses
MOFs Controlled metal ion release + ROS + drug delivery Tunable, multifunctional, strong in vitro activity Cytotoxicity, in vivo stability
MNPs Membrane disruption, ion release, ROS Broad-spectrum, synergize with antibiotics Environmental accumulation, bacterial adaptation
PDT Light-activated ROS Resistance-free, precise, broad-spectrum Poor penetration, phototoxicity
AMPs Membrane targeting + intracellular action Low resistance potential, multifunctional High cost, instability, potential cytotoxicity


Social and environmental considerations are essential for future strategies. As the use of nanotechnology in antimicrobial research grows, so do public concerns over the safety and environmental impact of nanomaterials. Future research should prioritize the design of nanomaterials that emphasize biocompatibility, biodegradability, and safe degradation pathways to minimize risks to human health and ecosystems while maximizing therapeutic potential.

Future research directions should focus on the following: (1) designing hybrid systems that integrate MOFs with AMPs or photosensitizers to enable multifunctional activity while reducing toxicity. (2) Developing biodegradable or self-degrading MNPs to mitigate long-term environmental accumulation. (3) Enhancing PDT through advanced light delivery systems, nanocarrier engineering, and combinations with immunotherapies. (4) Utilizing computational modeling, peptide engineering, and synthetic biology to create cost-effective and stable AMPs suitable for clinical use. By embracing these interdisciplinary and sustainability-focused directions, the antibacterial field can progress towards the safe and effective integration of innovative technologies. These combined strategies not only hold promise for reducing reliance on conventional antibiotics but also for responsibly addressing the growing crisis of antibiotic resistance.

Author contributions

Afsaneh Arshadi Edlo: conceptualization, resources, investigation, writing – original draft and visualization. Kamran Akhbari: validation, data curation, writing – review & editing, supervision, project administration. David J. Henry: review & editing.

Conflicts of interest

The authors declare no competing financial interests for this article.

Data availability

All data and materials supporting this review article are publicly available in the references listed. No original datasets were generated for this study.

Acknowledgements

The authors would like to acknowledge the support from the University of Tehran.

References

  1. W. H. Organization, Interagency Coordination Group on Antimicrobial Resistance, No time to wait: securing the future from drug-resistant infections, Report to the secretary-general of the united nations, 2019, vol. 1, p. 28.
  2. A. Catalano, D. Iacopetta, J. Ceramella, M. Pellegrino, F. Giuzio, M. Marra, C. Rosano, C. Saturnino, M. S. Sinicropi and S. Aquaro, Antibiotic-resistant ESKAPE pathogens and COVID-19: the pandemic beyond the pandemic, Viruses, 2023, 15, 1843 CrossRef CAS PubMed.
  3. J. Davies and D. Davies, Origins and evolution of antibiotic resistance, Microbiol. Mol. Biol. Rev., 2010, 74, 417–433 Search PubMed.
  4. W. H. Organization, Thirteenth general programme of work, 2019–2023: promote health, keep the world safe, serve the vulnerable, World Health Organization, 2019.
  5. Y. Yang, X. Wu, C. He, J. Huang, S. Yin, M. Zhou, L. Ma, W. Zhao, L. Qiu and C. Cheng, Metal–organic framework/Ag-based hybrid nanoagents for rapid and synergistic bacterial eradication, ACS Appl. Mater. Interfaces, 2020, 12, 13698–13708 CrossRef CAS PubMed.
  6. R. Karimi Alavijeh and K. Akhbari, Biocompatible MIL-101 (Fe) as a smart carrier with high loading potential and sustained release of curcumin, Inorg. Chem., 2020, 59, 3570–3578 CrossRef CAS PubMed.
  7. T. Yao, X. Zeng, X. Tao and H. Xu, Recent progress of MOF-based antibacterial hydrogels, Chem. Eng. J., 2024, 487, 150641 CrossRef CAS.
  8. A. A. Edlo and K. Akhbari, Modulating the antibacterial activity of a CuO@ HKUST-1 nanocomposite by optimizing its synthesis procedure, New J. Chem., 2023, 47, 20770–20776 Search PubMed.
  9. M. Kalati and K. Akhbari, Optimizing the metal ion release and antibacterial activity of ZnO@ ZIF-8 by modulating its synthesis method, New J. Chem., 2021, 45, 22924–22931 Search PubMed.
  10. M. Kalati and K. Akhbari, Copper (II) nitrate and Copper (II) oxide loading on ZIF-8; synthesis, characterization and antibacterial activity, J. Porous Mater., 2022, 29, 1909–1917 CrossRef CAS.
  11. Z. Zhang, D. Xing, Q. Liang, D. Yong and X. Han, Size controllable synthesis and antimicrobial activity of poly-N,N′-[(4,5-dihydroxy-1,2-phenylene) bis (methylene)] bisacrylamide microspheres, RSC Adv., 2014, 4, 57891–57898 RSC.
  12. R. K. Alavijeh, S. Beheshti, K. Akhbari and A. Morsali, Investigation of reasons for metal–organic framework's antibacterial activities, Polyhedron, 2018, 156, 257–278 Search PubMed.
  13. Y. Yang, Y. Deng, J. Huang, X. Fan, C. Cheng, C. Nie, L. Ma, W. Zhao and C. Zhao, Size-transformable metal–organic framework–derived nanocarbons for localized chemo-photothermal bacterial ablation and wound disinfection, Adv. Funct. Mater., 2019, 29, 1900143 CrossRef.
  14. M. Parsaei and K. Akhbari, MOF-801 as a nanoporous water-based carrier system for in situ encapsulation and sustained release of 5-FU for effective cancer therapy, Inorg. Chem., 2022, 61, 5912–5925 Search PubMed.
  15. M. Parsaei and K. Akhbari, Smart multifunctional UiO-66 metal–organic framework nanoparticles with outstanding drug-loading/release potential for the targeted delivery of quercetin, Inorg. Chem., 2022, 61, 14528–14543 Search PubMed.
  16. M. Nakhaei, K. Akhbari and A. Davoodi, Biocompatible MOF-808 as an iodophor antimicrobial agent with controlled and sustained release of iodine, CrystEngComm, 2021, 23, 8538–8545 RSC.
  17. H. Wang, L. Song, R. Jiang, Y. Fan, J. Zhao and L. Ren, Super-repellent photodynamic bactericidal hybrid membrane, J. Membr. Sci., 2020, 614, 118482 CrossRef CAS.
  18. A. Terzopoulou, J. D. Nicholas, X.-Z. Chen, B. J. Nelson, S. Pane and J. Puigmarti-Luis, Metal–organic frameworks in motion, Chem. Rev., 2020, 120, 11175–11193 CrossRef CAS PubMed.
  19. D. Wang, D. Jana and Y. Zhao, Metal–organic framework derived nanozymes in biomedicine, Acc. Chem. Res., 2020, 53, 1389–1400 CrossRef CAS PubMed.
  20. A. Davoodi, K. Akhbari and M. Alirezvani, Prolonged release of silver and iodine from ZIF-7 carrier with great antibacterial activity, CrystEngComm, 2023, 25, 3931–3942 Search PubMed.
  21. S. Soltani, K. Akhbari and A. Phuruangrat, Improved antibacterial activity by incorporation of silver sulfadiazine on Nanoporous Cu-BTC Metal-organic-framework, Inorg. Chim. Acta, 2022, 543, 121182 Search PubMed.
  22. J. W. Lim, D. Ha, J. Lee, S. K. Lee and T. Kim, Review of micro/nanotechnologies for microbial biosensors, Front. Bioeng. Biotechnol., 2015, 3, 61 Search PubMed.
  23. T. Mocan, C. T. Matea, T. Pop, O. Mosteanu, A. D. Buzoianu, C. Puia, C. Iancu and L. Mocan, Development of nanoparticle-based optical sensors for pathogenic bacterial detection, J. Nanobiotechnol., 2017, 15, 1–14 CrossRef PubMed.
  24. J. S. Möhler, W. Sim, M. A. Blaskovich, M. A. Cooper and Z. M. Ziora, Silver bullets: A new lustre on an old antimicrobial agent, Biotechnol. Adv., 2018, 36, 1391–1411 Search PubMed.
  25. C. Mutalik, G. Okoro, D. I. Krisnawati, A. Jazidie, E. Q. Rahmawati, D. Rahayu, W.-T. Hsu and T.-R. Kuo, Copper sulfide with morphology-dependent photodynamic and photothermal antibacterial activities, J. Colloid Interface Sci., 2022, 607, 1825–1835 Search PubMed.
  26. E. Sánchez-López, D. Gomes, G. Esteruelas, L. Bonilla, A. L. Lopez-Machado, R. Galindo, A. Cano, M. Espina, M. Ettcheto and A. Camins, Metal-based nanoparticles as antimicrobial agents: an overview, Nanomaterials, 2020, 10, 292 Search PubMed.
  27. T. Nguyen, R. Selvanayagam, K. Ho, R. Chen, S. Kutty, S. Rice, N. Kumar, N. Barraud, H. Duong and C. Boyer, Co-delivery of nitric oxide and antibiotic using polymeric nanoparticles, Chem. Sci., 2016, 7, 1016–1027 RSC.
  28. L. Tian, W. Zhou, X. Wu, Z. Hu, L. Qiu, H. Zhang, X. Chen, S. Zhang and Z. Lu, CTLs: killers of intracellular bacteria, Front. Cell. Infect. Microbiol., 2022, 12, 967679 CrossRef CAS PubMed.
  29. P. A. Hernández-Venegas, R. E. Martínez-Martínez, E. A. Zaragoza-Contreras, R. A. Domínguez-Pérez, S. Y. Reyes-López, A. Donohue-Cornejo, J. C. Cuevas-González, N. Molina-Frechero and L. F. Espinosa-Cristóbal, Bactericidal activity of silver nanoparticles on oral biofilms related to patients with and without periodontal disease, J. Funct. Biomater., 2023, 14, 311 Search PubMed.
  30. M. F. Salas-Orozco, A. C. Lorenzo-Leal, I. de Alba Montero, N. P. Marín, M. A. C. Santana and H. Bach, Mechanism of escape from the antibacterial activity of metal-based nanoparticles in clinically relevant bacteria: A systematic review, Nanomedicine, 2024, 55, 102715 CrossRef CAS PubMed.
  31. Y. Song, Q. Sun, J. Luo, Y. Kong, B. Pan, J. Zhao, Y. Wang and C. Yu, Cationic and anionic antimicrobial agents co-templated mesostructured silica nanocomposites with a spiky nanotopology and enhanced biofilm inhibition performance, Nano-Micro Lett., 2022, 14, 83 CrossRef CAS PubMed.
  32. B. Essghaier, H. Hannachi, R. Nouir, F. Mottola and L. Rocco, Green Synthesis and characterization of Novel Silver nanoparticles using Achillea maritima subsp. maritima Aqueous Extract: antioxidant and antidiabetic potential and effect on virulence mechanisms of bacterial and fungal pathogens, Nanomaterials, 2023, 13, 1964 CrossRef CAS PubMed.
  33. J. H. M. Sin, L. J. Walsh, C. M. Figueredo and R. George, Evaluation of effectiveness of photosensitizers used in laser endodontics disinfection: A systematic review, Transl. Biophotonics, 2021, 3, e202000007 CrossRef CAS.
  34. S. Bera, Nanotechnology Based Strategies for Combating Antimicrobial Resistance, Springer, 2024, pp. 351–391 Search PubMed.
  35. A. Kiang, S. Benson, C. Lochenie, S. Duncan, S. M. P. Mohanan, G. O. Williams, K. Dhaliwal, M. Vendrell and B. Mills, Novel dual antimicrobial-imaging photodynamic therapy agent labels and kills AMR gram-positive bacteria, SPIE, 2024, PC12822, PC1282207 Search PubMed.
  36. Q. Yu, C. Wang, X. Zhang, H. Chen, M. X. Wu and M. Lu, Photochemical Strategies toward Precision Targeting against Multidrug-Resistant Bacterial Infections, ACS Nano, 2024, 18, 14085–14122 Search PubMed.
  37. M. Lu, S. Li, Y. Liu, B. Xu, S. Liu, J. Zhang, D. Zhou and H. Liu, Advances in phototherapy for infectious diseases, Nano Today, 2024, 57, 102327 Search PubMed.
  38. T. W. Rees, P. Y. Ho and J. Hess, Recent advances in metal complexes for antimicrobial photodynamic therapy, ChemBioChem, 2023, 24, e202200796 Search PubMed.
  39. X. Wang, L. He, Z. Huang, Q. Zhao, J. Fan, Y. Tian and A. Huang, Isolation, identification and characterization of a novel antimicrobial peptide from Moringa oleifera seeds based on affinity adsorption, Food Chem., 2023, 398, 133923 CrossRef CAS PubMed.
  40. M. Mahlapuu, J. Håkansson, L. Ringstad and C. Björn, Antimicrobial peptides: an emerging category of therapeutic agents, Front. Cell. Infect. Microbiol., 2016, 6, 235805 Search PubMed.
  41. J. Mwangi, X. Hao, R. Lai and Z.-Y. Zhang, Antimicrobial peptides: new hope in the war against multidrug resistance, Zool. Res., 2019, 40, 488 Search PubMed.
  42. S. Ji, F. An, T. Zhang, M. Lou, J. Guo, K. Liu, Y. Zhu, J. Wu and R. Wu, Antimicrobial peptides: An alternative to traditional antibiotics, Eur. J. Med. Chem., 2024, 265, 116072 Search PubMed.
  43. M. Erdem Büyükkiraz and Z. Kesmen, Antimicrobial peptides (AMPs): A promising class of antimicrobial compounds, J. Appl. Microbiol., 2022, 132, 1573–1596 CrossRef PubMed.
  44. T. Takahashi and K. Yamasaki, Psoriasis and antimicrobial peptides, Int. J. Mol. Sci., 2020, 21, 6791 CrossRef CAS PubMed.
  45. K.-T. Cheng, C.-L. Wu, B.-S. Yip, H.-Y. Yu, H.-T. Cheng, Y.-H. Chih and J.-W. Cheng, High level expression and purification of the clinically active antimicrobial peptide P-113 in Escherichia coli, Molecules, 2018, 23, 800 CrossRef PubMed.
  46. H. Chen, C. Liu, D. Chen, K. Madrid, S. Peng, X. Dong, M. Zhang and Y. Gu, Bacteria-targeting conjugates based on antimicrobial peptide for bacteria diagnosis and therapy, Mol. Pharmaceutics, 2015, 12, 2505–2516 CrossRef CAS PubMed.
  47. E. Finkina, D. Melnikova, I. Bogdanov and T. Ovchinnikova, Peptides of the innate immune system of plants. Part I. Structure, biological activity, and mechanisms of action, Russ. J. Bioorg. Chem., 2018, 44, 573–585 CrossRef CAS.
  48. Y. Qin, Z. D. Qin, J. Chen, C. G. Cai, L. Li, L. Y. Feng, Z. Wang, G. J. Duns, N. Y. He and Z. S. Chen, From antimicrobial to anticancer peptides: the transformation of peptides, Recent Pat. Anti-Cancer Drug Discovery, 2019, 14, 70–84 CrossRef CAS PubMed.
  49. T.-J. Sun, H.-L. Bu, X. Yan, Z.-H. Sun, M.-S. Zha and G.-F. Dong, LABAMPsGCN: A framework for identifying lactic acid bacteria antimicrobial peptides based on graph convolutional neural network, Front. Genet., 2022, 13, 1062576 CrossRef CAS PubMed.
  50. U. L. Urmi, A. K. Vijay, R. Kuppusamy, S. Islam and M. D. Willcox, A review of the antiviral activity of cationic antimicrobial peptides, Peptides, 2023, 166, 171024 CrossRef CAS PubMed.
  51. H. Li, J. Niu, X. Wang, M. Niu and C. Liao, The Contribution of Antimicrobial Peptides to Immune Cell Function: A Review of Recent Advances, Pharmaceutics, 2023, 15, 2278 CrossRef CAS PubMed.
  52. G. Cottarel and J. Wierzbowski, Combination drugs, an emerging option for antibacterial therapy, Trends Biotechnol., 2007, 25, 547–555 CrossRef CAS PubMed.
  53. A. R. Coates, Y. Hu, J. Holt and P. Yeh, Antibiotic combination therapy against resistant bacterial infections: synergy, rejuvenation and resistance reduction, Expert Rev. Anti-Infect. Ther., 2020, 18, 5–15 CrossRef CAS PubMed.
  54. N. Wang, J. Luo, F. Deng, Y. Huang and H. Zhou, Antibiotic combination therapy: a strategy to overcome bacterial resistance to aminoglycoside antibiotics, Front. Pharmacol., 2022, 13, 839808 CrossRef CAS PubMed.
  55. Z. Si, K. Pethe and M. B. Chan-Park, Chemical basis of combination therapy to combat antibiotic resistance, JACS Au, 2023, 3, 276–292 CrossRef CAS PubMed.
  56. R. J. Fair and Y. Tor, Antibiotics and bacterial resistance in the 21st century, Perspect. Med. Chem., 2014, 6, PMC.S14459 CrossRef PubMed.
  57. Z. Alexander, Interference plasmids and their use in combating bacterial resistance, Microbiol. Indep. Res. J., 2019, 6, 37–42 Search PubMed.
  58. W. Y. Belay, M. Getachew, B. A. Tegegne, Z. H. Teffera, A. Dagne, T. K. Zeleke, R. B. Abebe, A. A. Gedif, A. Fenta and G. Yirdaw, Mechanism of antibacterial resistance, strategies and next-generation antimicrobials to contain antimicrobial resistance: A review, Front. Pharmacol., 2024, 15, 1444781 CrossRef CAS PubMed.
  59. M. Alavi and M. Rai, Recent advances in antibacterial applications of metal nanoparticles (MNPs) and metal nanocomposites (MNCs) against multidrug-resistant (MDR) bacteria, Expert Rev. Anti-Infect. Ther., 2019, 17, 419–428 CrossRef CAS PubMed.
  60. C. K. Lai, R. W. Ng, S. S. Leung, M. Hui and M. Ip, Overcoming the rising incidence and evolving mechanisms of antibiotic resistance by novel drug delivery approaches–an overview, Adv. Drug Delivery Rev., 2022, 181, 114078 CrossRef CAS PubMed.
  61. A. Gupta, S. Mumtaz, C.-H. Li, I. Hussain and V. M. Rotello, Combatting antibiotic-resistant bacteria using nanomaterials, Chem. Soc. Rev., 2019, 48, 415–427 RSC.
  62. H. C. Neu, The crisis in antibiotic resistance, Science, 1992, 257, 1064–1073 CrossRef CAS PubMed.
  63. U. Theuretzbacher, Global antibacterial resistance: The never-ending story, J. Global Antimicrob. Resist., 2013, 1, 63–69 CrossRef PubMed.
  64. S. Jana and J. Deb, Molecular understanding of aminoglycoside action and resistance, Appl. Microbiol. Biotechnol., 2006, 70, 140–150 CrossRef CAS PubMed.
  65. A. Zapun, C. Contreras-Martel and T. Vernet, Penicillin-binding proteins and β-lactam resistance, FEMS Microbiol. Rev., 2008, 32, 361–385 CrossRef CAS PubMed.
  66. R. Azargun, M. H. S. Barhaghi, H. S. Kafil, M. A. Oskouee, V. Sadeghi, M. Y. Memar and R. Ghotaslou, Frequency of DNA gyrase and topoisomerase IV mutations and plasmid-mediated quinolone resistance genes among Escherichia coli and Klebsiella pneumoniae isolated from urinary tract infections in Azerbaijan, Iran, J. Global Antimicrob. Resist., 2019, 17, 39–43 CrossRef PubMed.
  67. R. Alenazy, Drug efflux pump inhibitors: a promising approach to counter multidrug resistance in Gram-negative pathogens by targeting AcrB protein from AcrAB-TolC multidrug efflux pump from Escherichia coli, Biology, 2022, 11, 1328 Search PubMed.
  68. S. R. Partridge, S. M. Kwong, N. Firth and S. O. Jensen, Mobile genetic elements associated with antimicrobial resistance, Clin. Microbiol. Rev., 2018, 31, e00088-17 CrossRef PubMed.
  69. L. K. Vestby, T. Grønseth, R. Simm and L. L. Nesse, Bacterial biofilm and its role in the pathogenesis of disease, Antibiotics, 2020, 9, 59 CrossRef CAS PubMed.
  70. M. Jamal, W. Ahmad, S. Andleeb, F. Jalil, M. Imran, M. A. Nawaz, T. Hussain, M. Ali, M. Rafiq and M. A. Kamil, Bacterial biofilm and associated infections, J. Chin. Med. Assoc., 2018, 81, 7–11 CrossRef PubMed.
  71. B. Ribeiro da Cunha, L. P. Fonseca and C. R. Calado, Antibiotic discovery: where have we come from, where do we go?, Antibiotics, 2019, 8, 45 CrossRef PubMed.
  72. I. Kyriakidis, E. Vasileiou, Z. D. Pana and A. Tragiannidis, Acinetobacter baumannii antibiotic resistance mechanisms, Pathogens, 2021, 10, 373 CrossRef CAS PubMed.
  73. J. Iredell, J. Brown and K. Tagg, Antibiotic resistance in Enterobacteriaceae: mechanisms and clinical implications, BMJ, 2016, 352, h6420 Search PubMed.
  74. J. R. Hayes, L. L. English, P. J. Carter, T. Proescholdt, K. Y. Lee, D. D. Wagner and D. G. White, Prevalence and antimicrobial resistance of Enterococcus species isolated from retail meats, Appl. Environ. Microbiol., 2003, 69, 7153–7160 Search PubMed.
  75. D. Subedi, A. K. Vijay and M. Willcox, Overview of mechanisms of antibiotic resistance in Pseudomonas aeruginosa: an ocular perspective, Clin. Exp. Optom., 2018, 101, 162–171 Search PubMed.
  76. L. M. Weiner-Lastinger, S. Abner, J. R. Edwards, A. J. Kallen, M. Karlsson, S. S. Magill, D. Pollock, I. See, M. M. Soe and M. S. Walters, Antimicrobial-resistant pathogens associated with adult healthcare-associated infections: summary of data reported to the National Healthcare Safety Network, 2015–2017, Infect. Control Hosp. Epidemiol., 2020, 41, 1–18 Search PubMed.
  77. G. D. Wright, Q&A: Antibiotic resistance: where does it come from and what can we do about it?, BMC Biol., 2010, 8, 1–6 CrossRef PubMed.
  78. M. Parsaei, K. Akhbari, E. Tylianakis, G. E. Froudakis and J. M. White, Efficient Gas Adsorption in MUT-11: Insights from Theoretical Calculations and GCMC Simulations, Cryst. Growth Des., 2024, 24, 10299–10313 CrossRef CAS.
  79. M. Parsaei, K. Akhbari and S. Kawata, Computational Simulation of CO2/CH4 Separation on a Three-Dimensional Cd-Based Metal–Organic Framework, Cryst. Growth Des., 2023, 23, 5705–5718 CrossRef CAS.
  80. M. Parsaei and K. Akhbari, Synthesis and application of MOF-808 decorated with folic acid-conjugated chitosan as a strong nanocarrier for the targeted drug delivery of quercetin, Inorg. Chem., 2022, 61, 19354–19368 CrossRef CAS PubMed.
  81. D. M. Amidi and K. Akhbari, Iodine-loaded ZIF-7-coated cotton substrates show sustained iodine release as effective antibacterial textiles, New J. Chem., 2024, 48, 2016–2027 Search PubMed.
  82. M. Parsaei, K. Akhbari, E. Tylianakis and G. E. Froudakis, Computational Simulation of a Three-Dimensional Mg-based Metal–Organic Framework as Nanoporous Anticancer Drug Carrier, Cryst. Growth Des., 2023, 23, 8396–8406 Search PubMed.
  83. R. K. Alavijeh, K. Akhbari and J. White, Solid–liquid conversion and carbon dioxide storage in a calcium-based metal–organic framework with micro-and nanoporous channels, Cryst. Growth Des., 2019, 19, 7290–7297 CrossRef CAS.
  84. S. Salimi, K. Akhbari, S. M. F. Farnia and J. M. White, Multiple Construction of a Hierarchical Nanoporous Manganese (II)-Based Metal–Organic Framework with Active Sites for Regulating N2 and CO2 Trapping, Cryst. Growth Des., 2022, 22, 1654–1664 CrossRef CAS.
  85. M. Parsaei, K. Akhbari and J. White, Modulating carbon dioxide storage by facile synthesis of nanoporous pillared-layered metal–organic framework with different synthetic routes, Inorg. Chem., 2022, 61, 3893–3902 CrossRef CAS PubMed.
  86. R. Ghasemzadeh, K. Akhbari and S. Kawata, Ag@ MUT-16 nanocomposite as a Fenton-like and plasmonic photocatalyst for degradation of Quinoline Yellow under visible light, Dalton Trans., 2024, 53, 11094–11111 RSC.
  87. R. Ghasemzadeh, K. Akhbari and S. Kawata, rGO/MUT-15 nanocomposite as a Fenton-like photocatalyst for the degradation of Acid Yellow 73 under visible light, Dalton Trans., 2024, 53, 18268–18282 RSC.
  88. P. K. Kermanshahi, K. Akhbari, M. T. HesarAmiri, R. Karimi, E. Zivari and M. Madadi, The effect of layering order of ZIF-8@ PCL/PLA composites in electrospinning and electrospraying process on antibacterial wound healing, Mater. Chem. Phys., 2024, 328, 129878 CrossRef.
  89. M. Parsaei and K. Akhbari, Magnetic UiO-66-NH2 Core–Shell Nanohybrid as a Promising Carrier for Quercetin Targeted Delivery toward Human Breast Cancer Cells, ACS Omega, 2023, 8, 41321–41338 CrossRef CAS PubMed.
  90. R. K. Alavijeh and K. Akhbari, Cancer therapy by nano MIL-n series of metal-organic frameworks, Coord. Chem. Rev., 2024, 503, 215643 CrossRef.
  91. P. K. Kermanshahi and K. Akhbari, The antibacterial activity of three zeolitic-imidazolate frameworks and zinc oxide nanoparticles derived from them, RSC Adv., 2024, 14, 5601–5608 RSC.
  92. A. Arshadi Edlo and K. Akhbari, Modulated antibacterial activity in ZnO@ MIL-53 (Fe) and CuO@ MIL-53 (Fe) nanocomposites prepared by simple thermal treatment process, Appl. Organomet. Chem., 2024, 38, e7326 CrossRef CAS.
  93. R. Li, T. Chen and X. Pan, Metal–organic-framework-based materials for antimicrobial applications, ACS Nano, 2021, 15, 3808–3848 CrossRef CAS PubMed.
  94. D. Mohammadi Amidi, K. Akhbari and S. Soltani, Loading of ZIF-67 on silk with sustained release of iodine as biocompatible antibacterial fibers, Appl. Organomet. Chem., 2023, 37, e6913 CrossRef CAS.
  95. Z. Song, Y. Wu, Q. Cao, H. Wang, X. Wang and H. Han, pH-responsive, light-triggered on-demand antibiotic release from functional metal–organic framework for bacterial infection combination therapy, Adv. Funct. Mater., 2018, 28, 1800011 CrossRef.
  96. G. Huang, Y. Li, Z. Qin, Q. Liang, C. Xu and B. Lin, Hybridization of carboxymethyl chitosan with MOFs to construct recyclable, long-acting and intelligent antibacterial agent carrier, Carbohydr. Polym., 2020, 233, 115848 CrossRef CAS PubMed.
  97. H. Ghasempour, K.-Y. Wang, J. A. Powell, F. ZareKarizi, X.-L. Lv, A. Morsali and H.-C. Zhou, Metal–organic frameworks based on multicarboxylate linkers, Coord. Chem. Rev., 2021, 426, 213542 CrossRef CAS.
  98. B. Sun, M. Bilal, S. Jia, Y. Jiang and J. Cui, Design and bio-applications of biological metal-organic frameworks, Korean J. Chem. Eng., 2019, 36, 1949–1964 CrossRef CAS.
  99. S. Soltani and K. Akhbari, Facile and single-step entrapment of chloramphenicol in ZIF-8 and evaluation of its performance in killing infectious bacteria with high loading content and controlled release of the drug, CrystEngComm, 2022, 24, 1934–1941 RSC.
  100. Y. Zhang, P. Sun, L. Zhang, Z. Wang, F. Wang, K. Dong, Z. Liu, J. Ren and X. Qu, Silver-infused porphyrinic metal–organic framework: Surface-adaptive, on-demand nanoplatform for synergistic bacteria killing and wound disinfection, Adv. Funct. Mater., 2019, 29, 1808594 CrossRef.
  101. K. Wang, Y. Yin, C. Li, Z. Geng and Z. Wang, Facile synthesis of zinc (II)-carboxylate coordination polymer particles and their luminescent, biocompatible and antibacterial properties, CrystEngComm, 2011, 13, 6231–6236 RSC.
  102. A. Dastneshan, S. Rahiminezhad, M. N. Mezajin, H. N. Jevinani, I. Akbarzadeh, M. Abdihaji, R. Qahremani, M. Jahanbakhshi, Z. A. Lalami and H. Heydari, Cefazolin encapsulated UIO-66-NH2 nanoparticles enhance the antibacterial activity and biofilm inhibition against drug-resistant S. aureus: In vitro and in vivo studies, Chem. Eng. J., 2023, 455, 140544 CrossRef CAS.
  103. H. Qiu, F. Pu, Z. Liu, Q. Deng, P. Sun, J. Ren and X. Qu, Depriving bacterial adhesion-related molecule to inhibit biofilm formation using CeO2-decorated metal-organic frameworks, Small, 2019, 15, 1902522 CrossRef PubMed.
  104. M. Nakhaei, K. Akhbari, M. Kalati and A. Phuruangrat, Antibacterial activity of three zinc-terephthalate MOFs and its relation to their structural features, Inorg. Chim. Acta, 2021, 522, 120353 CrossRef CAS.
  105. M. Z. Isfahani, K. Akhbari, S. Soltani and A. Phuruangrat, Be safe against bacteria with nano CuBDC metal-organic framework loaded on silk fibers, Mater. Chem. Phys., 2022, 290, 126582 CrossRef.
  106. S. M. Sheta, S. M. El-Sheikh and M. M. Abd-Elzaher, Simple synthesis of novel copper metal–organic framework nanoparticles: biosensing and biological applications, Dalton Trans., 2018, 47, 4847–4855 RSC.
  107. P. Raju, T. Ramalingam, T. Nooruddin and S. Natarajan, In vitro assessment of antimicrobial, antibiofilm and larvicidal activities of bioactive nickel metal organic framework, J. Drug Delivery Sci. Technol., 2020, 56, 101560 CrossRef CAS.
  108. K. Zhang, X. Meng, Y. Cao, Z. Yang, H. Dong, Y. Zhang, H. Lu, Z. Shi and X. Zhang, Metal–organic framework nanoshuttle for synergistic photodynamic and low-temperature photothermal therapy, Adv. Funct. Mater., 2018, 28, 1804634 CrossRef.
  109. D. Han, Y. Han, J. Li, X. Liu, K. W. K. Yeung, Y. Zheng, Z. Cui, X. Yang, Y. Liang and Z. Li, Enhanced photocatalytic activity and photothermal effects of cu-doped metal-organic frameworks for rapid treatment of bacteria-infected wounds, Appl. Catal., B, 2020, 261, 118248 CrossRef CAS.
  110. P. Li, J. Li, X. Feng, J. Li, Y. Hao, J. Zhang, H. Wang, A. Yin, J. Zhou and X. Ma, Metal-organic frameworks with photocatalytic bactericidal activity for integrated air cleaning, Nat. Commun., 2019, 10, 2177 CrossRef PubMed.
  111. D. Ma, P. Li, X. Duan, J. Li, P. Shao, Z. Lang, L. Bao, Y. Zhang, Z. Lin and B. Wang, A hydrolytically stable Vanadium (IV) metal–organic framework with photocatalytic bacteriostatic activity for autonomous indoor humidity control, Angew. Chem., 2020, 132, 3933–3937 CrossRef.
  112. Y.-F. Guo, W.-J. Fang, J.-R. Fu, Y. Wu, J. Zheng, G.-Q. Gao, C. Chen, R.-W. Yan, S.-G. Huang and C.-C. Wang, Facile synthesis of Ag@ ZIF-8 core-shell heterostructure nanowires for improved antibacterial activities, Appl. Surf. Sci., 2018, 435, 149–155 CrossRef CAS.
  113. M. Ishfaq, D. Lateef, Z. Ashraf, M. Sajjad, M. Owais, W. Shoukat, M. Mohsin, M. Ibrahim, F. Verpoort and A. H. Chughtai, Zirconium-based MOFs as pH-responsive drug delivery systems: encapsulation and release profiles of ciprofloxacin, RSC Adv., 2025, 15, 26647–26659 RSC.
  114. N. Rahmanian, P. Moulavi, F. Ashrafi, A. Sharifi and S. Asadi, Surface-functionalized UIO-66-NH2 for dual-drug delivery of vancomycin and amikacin against vancomycin-resistant Staphylococcus aureus, BMC Microbiol., 2024, 24, 462 CrossRef CAS PubMed.
  115. G. Ximing, G. Bin, W. Yuanlin and G. Shuanghong, Preparation of spherical metal–organic frameworks encapsulating Ag nanoparticles and study on its antibacterial activity, Mater. Sci. Eng., C, 2017, 80, 698–707 CrossRef PubMed.
  116. H. Abd El Salam, H. N. Nassar, A. S. Khidr and T. Zaki, Antimicrobial activities of green synthesized Ag nanoparticles@ Ni-MOF nanosheets, J. Inorg. Organomet. Polym. Mater., 2018, 28, 2791–2798 CrossRef CAS.
  117. S. Soltani and K. Akhbari, Cu-BTC metal–organic framework as a biocompatible nanoporous carrier for chlorhexidine antibacterial agent, JBIC, J. Biol. Inorg. Chem., 2022, 1–7 Search PubMed.
  118. S. Soltani and K. Akhbari, Embedding an extraordinary amount of gemifloxacin antibiotic in ZIF-8 framework with one-step synthesis and measurement of its H 2 O 2-sensitive release and potency against infectious bacteria, New J. Chem., 2022, 46, 19432–19441 RSC.
  119. A. Arenas-Vivo, G. Amariei, S. Aguado, R. Rosal and P. Horcajada, An Ag-loaded photoactive nano-metal organic framework as a promising biofilm treatment, Acta Biomater., 2019, 97, 490–500 CrossRef CAS PubMed.
  120. H. Chen, J. Yang, L. Sun, H. Zhang, Y. Guo, J. Qu, W. Jiang, W. Chen, J. Ji and Y. W. Yang, Synergistic chemotherapy and photodynamic therapy of endophthalmitis mediated by zeolitic imidazolate framework-based drug delivery systems, Small, 2019, 15, 1903880 CrossRef CAS PubMed.
  121. S. Lin, X. Liu, L. Tan, Z. Cui, X. Yang, K. W. Yeung, H. Pan and S. Wu, Porous iron-carboxylate metal–organic framework: a novel bioplatform with sustained antibacterial efficacy and nontoxicity, ACS Appl. Mater. Interfaces, 2017, 9, 19248–19257 CrossRef CAS PubMed.
  122. A. Frei, A. D. Verderosa, A. G. Elliott, J. Zuegg and M. A. Blaskovich, Metals to combat antimicrobial resistance, Nat. Rev. Chem., 2023, 7, 202–224 CrossRef CAS PubMed.
  123. A. Girma, Alternative mechanisms of action of metallic nanoparticles to mitigate the global spread of antibiotic-resistant bacteria, Cell Surf., 2023, 10, 100112 CrossRef CAS PubMed.
  124. S. T. Khan, J. Musarrat and A. A. Al-Khedhairy, Countering drug resistance, infectious diseases, and sepsis using metal and metal oxides nanoparticles: current status, Colloids Surf., B, 2016, 146, 70–83 Search PubMed.
  125. M. Rai, K. Kon, A. Gade, A. Ingle, D. Nagaonkar, P. Paralikar and S. da Silva, Antibiotic resistance: can nanoparticles tackle the problem, 2016, ch. 6, pp. 121–143 Search PubMed.
  126. F. Amaro, Á. Morón, S. Díaz, A. Martín-González and J. C. Gutiérrez, Metallic nanoparticles—friends or foes in the battle against antibiotic-resistant bacteria?, Microorganisms, 2021, 9, 364 CrossRef CAS PubMed.
  127. A. Panáček, L. Kvítek, M. Smékalová, R. Večeřová, M. Kolář, M. Röderová, F. Dyčka, M. Šebela, R. Prucek and O. Tomanec, Bacterial resistance to silver nanoparticles and how to overcome it, Nat. Nanotechnol., 2018, 13, 65–71 CrossRef PubMed.
  128. M. Zare, K. Namratha, K. Byrappa, D. Surendra, S. Yallappa and B. Hungund, Surfactant assisted solvothermal synthesis of ZnO nanoparticles and study of their antimicrobial and antioxidant properties, J. Mater. Sci. Technol., 2018, 34, 1035–1043 CrossRef CAS.
  129. P. Korshed, L. Li, Z. Liu, A. Mironov and T. Wang, Size-dependent antibacterial activity for laser-generated silver nanoparticles, J. Interdiscip. Nanomed., 2019, 4, 24–33 CrossRef CAS.
  130. L. Cui, P. Chen, S. Chen, Z. Yuan, C. Yu, B. Ren and K. Zhang, In situ study of the antibacterial activity and mechanism of action of silver nanoparticles by surface-enhanced Raman spectroscopy, Anal. Chem., 2013, 85, 5436–5443 CrossRef CAS PubMed.
  131. A. Abbaszadegan, Y. Ghahramani, A. Gholami, B. Hemmateenejad, S. Dorostkar, M. Nabavizadeh and H. Sharghi, The effect of charge at the surface of silver nanoparticles on antimicrobial activity against Gram-positive and Gram-negative bacteria: a preliminary study, J. Nanomater., 2015, 2015, 720654 CrossRef.
  132. Z. Li, J. Ma, J. Ruan and X. Zhuang, Using positively charged magnetic nanoparticles to capture bacteria at ultralow concentration, Nanoscale Res. Lett., 2019, 14, 1–8 CrossRef PubMed.
  133. N. Mammari, E. Lamouroux, A. Boudier and R. E. Duval, Current knowledge on the oxidative-stress-mediated antimicrobial properties of metal-based nanoparticles, Microorganisms, 2022, 10, 437 CrossRef CAS PubMed.
  134. A. El Badawy, Assessment of the fate and transport of silver nanoparticles in porous media, University of Cincinnati, 2011 Search PubMed.
  135. P. T. Huynh, G. D. Nguyen, K. T. L. Tran, T. M. Ho, B. T. Duong, V. Q. Lam and T. V. K. Ngo, One-Pot, Surfactant-Free Synthesis of Gold Nanostars and Evaluation of Their Antibacterial Effects against Propionibacterium acnes, J. Nanomater., 2021, 2021, 6650661 Search PubMed.
  136. S. Ranjan and C. Ramalingam, Titanium dioxide nanoparticles induce bacterial membrane rupture by reactive oxygen species generation, Environ. Chem. Lett., 2016, 14, 487–494 CrossRef CAS.
  137. H. Xu, F. Qu, H. Xu, W. Lai, Y. Andrew Wang, Z. P. Aguilar and H. Wei, Role of reactive oxygen species in the antibacterial mechanism of silver nanoparticles on Escherichia coli O157: H7, Biometals, 2012, 25, 45–53 CrossRef CAS PubMed.
  138. L. Wang, H. He, Y. Yu, L. Sun, S. Liu, C. Zhang and L. He, Morphology-dependent bactericidal activities of Ag/CeO2 catalysts against Escherichia coli, J. Inorg. Biochem., 2014, 135, 45–53 Search PubMed.
  139. A. A. Mujeeb, N. A. Khan, F. Jamal, K. F. Badre Alam, H. Saeed, S. Kazmi, A. W. F. Alshameri, M. Kashif, I. Ghazi and M. Owais, Olax scandens mediated biogenic synthesis of Ag-Cu nanocomposites: Potential against inhibition of drug-resistant microbes, Front. Chem., 2020, 8, 103 Search PubMed.
  140. S. Mohapatra, T. A. Nguyen and P. Nguyen-Tri, Noble metal-metal oxide hybrid nanoparticles: Fundamentals and applications, Elsevier, 2018 Search PubMed.
  141. N. Watanabe, K. Kryukov, S. Nakagawa, J. S. Takeuchi, M. Takeshita, Y. Kirimura, S. Mitsuhashi, T. Ishihara, H. Aoki and S. Inokuchi, Detection of pathogenic bacteria in the blood from sepsis patients using 16S rRNA gene amplicon sequencing analysis, PLoS One, 2018, 13, e0202049 Search PubMed.
  142. J. W. Moreau, P. K. Weber, M. C. Martin, B. Gilbert, I. D. Hutcheon and J. F. Banfield, Extracellular proteins limit the dispersal of biogenic nanoparticles, Science, 2007, 316, 1600–1603 Search PubMed.
  143. M. Saliani, R. Jalal and E. K. Goharshadi, Effects of pH and temperature on antibacterial activity of zinc oxide nanofluid against Escherichia coli O157: H7 and Staphylococcus aureus, Jundishapur J. Microbiol., 2015, 8, e17115 Search PubMed.
  144. T. S. Peretyazhko, Q. Zhang and V. L. Colvin, Size-controlled dissolution of silver nanoparticles at neutral and acidic pH conditions: kinetics and size changes, Environ. Sci. Technol., 2014, 48, 11954–11961 Search PubMed.
  145. D. Franco, G. Calabrese, S. Petralia, G. Neri, C. Corsaro, L. Forte, S. Squarzoni, S. Guglielmino, F. Traina and E. Fazio, Antimicrobial effect and cytotoxic evaluation of Mg-doped hydroxyapatite functionalized with Au-nano rods, Molecules, 2021, 26, 1099 Search PubMed.
  146. X. Li, K. Z. Ahmad, J. He, H. Li, X. Wang, Z. Feng, X. Wang, G. Shen and X. Ding, Silver nanoflowers coupled with low dose antibiotics enable the highly effective eradication of drug-resistant bacteria, J. Mater. Chem. B, 2021, 9, 9839–9851 RSC.
  147. X. Hong, J. Wen, X. Xiong and Y. Hu, Shape effect on the antibacterial activity of silver nanoparticles synthesized via a microwave-assisted method, Environ. Sci. Pollut. Res., 2016, 23, 4489–4497 Search PubMed.
  148. E. Altun, M. O. Aydogdu, E. Chung, G. Ren, S. Homer-Vanniasinkam and M. Edirisinghe, Metal-based nanoparticles for combating antibiotic resistance, Appl. Phys. Rev., 2021, 8, e041303 Search PubMed.
  149. P. Makvandi, M. Ashrafizadeh, M. Ghomi, M. Najafi, H. H. S. Hossein, A. Zarrabi, V. Mattoli and R. S. Varma, Injectable hyaluronic acid-based antibacterial hydrogel adorned with biogenically synthesized AgNPs-decorated multi-walled carbon nanotubes, Prog. Biomater., 2021, 10, 77–89 CrossRef CAS PubMed.
  150. T. Russo, A. Gloria, R. De Santis, U. D’amora, G. Balato, A. Vollaro, O. Oliviero, G. Improta, M. Triassi and L. Ambrosio, Preliminary focus on the mechanical and antibacterial activity of a PMMA-based bone cement loaded with gold nanoparticles, Bioact. Mater., 2017, 2, 156–161 CAS.
  151. A. Barui, R. Kotcherlakota and C. Patra, Inorganic Frameworks as Smart Nanomedicines, William Publ, 2018, 1, 239–278 Search PubMed.
  152. S. Hussain, J. Joo, J. Kang, B. Kim, G. B. Braun, Z.-G. She, D. Kim, A. P. Mann, T. Mölder and T. Teesalu, Antibiotic-loaded nanoparticles targeted to the site of infection enhance antibacterial efficacy, Nat. Biomed. Eng., 2018, 2, 95–103 Search PubMed.
  153. U. A. Hasanova, M. A. Ramazanov, A. M. Maharramov, Q. M. Eyvazova, Z. A. Agamaliyev, Y. V. Parfyonova, S. F. Hajiyeva, F. V. Hajiyeva and S. B. Veliyeva, Nano-Coupling of Cephalosporin Antibiotics with Fe3O4 Nanoparticles: Trojan Horse Approach in Antimicrobial Chemotherapy of Infections Caused by Klebsiella spp, J. Biomater. Nanobiotechnol., 2015, 6, 225–235 Search PubMed.
  154. K. Ali, B. Ahmed, M. S. Khan and J. Musarrat, Differential surface contact killing of pristine and low EPS Pseudomonas aeruginosa with Aloe vera capped hematite (α-Fe2O3) nanoparticles, J. Photochem. Photobiol., B, 2018, 188, 146–158 CrossRef CAS PubMed.
  155. S. J. Amina and B. Guo, A review on the synthesis and functionalization of gold nanoparticles as a drug delivery vehicle, Int. J. Nanomed., 2020, 9823–9857 CrossRef CAS PubMed.
  156. T. A. Hagbani, H. Yadav, A. Moin, A. S. A. Lila, K. Mehmood, F. Alshammari, S. Khan, E.-S. Khafagy, T. Hussain and S. M. D. Rizvi, Enhancement of vancomycin potential against pathogenic bacterial strains via gold nano-formulations: a nano-antibiotic approach, Materials, 2022, 15, 1108 CrossRef CAS PubMed.
  157. Q. Chen, J. Li, Y. Wu, F. Shen and M. Yao, Biological responses of Gram-positive and Gram-negative bacteria to nZVI (Fe 0), Fe2+ and Fe3+, RSC Adv., 2013, 3, 13835–13842 Search PubMed.
  158. A. Jain, R. Bhargava and P. Poddar, Probing interaction of Gram-positive and Gram-negative bacterial cells with ZnO nanorods, Mater. Sci. Eng., C, 2013, 33, 1247–1253 CrossRef CAS PubMed.
  159. F. Kang, P. J. Alvarez and D. Zhu, Microbial extracellular polymeric substances reduce Ag+ to silver nanoparticles and antagonize bactericidal activity, Environ. Sci. Technol., 2014, 48, 316–322 CrossRef CAS PubMed.
  160. C. Larimer, M. S. Islam, A. Ojha and I. Nettleship, Mutation of environmental mycobacteria to resist silver nanoparticles also confers resistance to a common antibiotic, Biometals, 2014, 27, 695–702 CrossRef CAS PubMed.
  161. M. Gambino, V. Marzano, F. Villa, A. Vitali, C. Vannini, P. Landini and F. Cappitelli, Effects of sublethal doses of silver nanoparticles on Bacillus subtilis planktonic and sessile cells, J. Appl. Microbiol., 2015, 118, 1103–1115 CrossRef CAS PubMed.
  162. Q. Wang, F. Kang, Y. Gao, X. Mao and X. Hu, Sequestration of nanoparticles by an EPS matrix reduces the particle-specific bactericidal activity, Sci. Rep., 2016, 6, 21379 CrossRef CAS PubMed.
  163. J. Guo, S.-H. Gao, J. Lu, P. L. Bond, W. Verstraete and Z. Yuan, Copper oxide nanoparticles induce lysogenic bacteriophage and metal-resistance genes in Pseudomonas aeruginosa PAO1, ACS Appl. Mater. Interfaces, 2017, 9, 22298–22307 CrossRef CAS PubMed.
  164. R. Zhang, F. Carlsson, M. Edman, M. Hummelgård, B. G. Jonsson, D. Bylund and H. Olin, Escherichia coli bacteria develop adaptive resistance to antibacterial ZnO nanoparticles, Adv. Biosyst., 2018, 2, 1800019 CrossRef PubMed.
  165. Y. Xu, C. Wang, J. Hou, P. Wang, G. You and L. Miao, Effects of cerium oxide nanoparticles on bacterial growth and behaviors: induction of biofilm formation and stress response, Environ. Sci. Pollut. Res., 2019, 26, 9293–9304 CrossRef CAS PubMed.
  166. A. Kędziora, M. Wernecki, K. Korzekwa, M. Speruda, Y. Gerasymchuk, A. Łukowiak and G. Bugla-Płoskońska, Consequences of long-term bacteria's exposure to silver nanoformulations with different physicochemical properties, Int. J. Nanomed., 2020, 199–213 CrossRef PubMed.
  167. A. Kędziora, M. Speruda, M. Wernecki, B. Dudek, K. Kapczynska, E. Krzyżewska, J. Rybka and G. Bugla-Płoskońska, How bacteria change after exposure to silver nanoformulations: analysis of the genome and outer membrane proteome, Pathogens, 2021, 10, 817 CrossRef PubMed.
  168. M. F. Salas-Orozco, N. Niño-Martínez, G. A. Martínez-Castañón, F. T. Méndez, G. M. M. Morán, A. E. Bendaña-Piñeiro, F. Ruiz and H. Bach, Proteomic analysis of an Enterococcus faecalis mutant generated against the exposure to silver nanoparticles, J. Appl. Microbiol., 2022, 132, 244–255 CrossRef CAS PubMed.
  169. D. Muehler, C. M. Rupp, S. Keceli, C. Brochhausen, H. Siegmund, T. Maisch, K.-A. Hiller, W. Buchalla and F. Cieplik, Insights into mechanisms of antimicrobial photodynamic action toward biofilms using phenalen-1-one derivatives as photosensitizers, Front. Microbiol., 2020, 11, 589364 CrossRef PubMed.
  170. L. Sharab, R. E. Baier, S. Ciancio and T. Mang, Influence of photodynamic therapy on bacterial attachment to titanium surface, J. Oral Implantol., 2021, 47, 427–435 Search PubMed.
  171. N. Iluz, Y. Maor, N. Keller and Z. Malik, The synergistic effect of PDT and oxacillin on clinical isolates of Staphylococcus aureus, Lasers Surg. Med., 2018, 50, 535–551 CrossRef PubMed.
  172. F. Vatansever, W. C. de Melo, P. Avci, D. Vecchio, M. Sadasivam, A. Gupta, R. Chandran, M. Karimi, N. A. Parizotto and R. Yin, Antimicrobial strategies centered around reactive oxygen species–bactericidal antibiotics, photodynamic therapy, and beyond, FEMS Microbiol. Rev., 2013, 37, 955–989 CrossRef CAS PubMed.
  173. F. Heinemann, J. Karges and G. Gasser, Critical overview of the use of Ru (II) polypyridyl complexes as photosensitizers in one-photon and two-photon photodynamic therapy, Acc. Chem. Res., 2017, 50, 2727–2736 CrossRef CAS PubMed.
  174. A. F. Silva, A. Borges, E. Giaouris, J. M. Graton Mikcha and M. Simões, Photodynamic inactivation as an emergent strategy against foodborne pathogenic bacteria in planktonic and sessile states, Crit. Rev. Microbiol., 2018, 44, 667–684 CrossRef CAS PubMed.
  175. N. R. Luke-Marshall, L. A. Hansen, G. Shafirstein and A. A. Campagnari, Antimicrobial photodynamic therapy with chlorin e6 is bactericidal against biofilms of the primary human otopathogens, mSphere, 2020, 5, e00492-20 CrossRef PubMed.
  176. Y. A. Anane, T. Apalata, S. Vasaikar, G. E. Okuthe and S. P. Songca, In vitro antimicrobial photodynamic inactivation of multidrug-resistant Acinetobacter baumannii biofilm using Protoporphyrin IX and Methylene blue, Photodiagn. Photodyn. Ther., 2020, 30, 101752 CrossRef CAS PubMed.
  177. S. P. Songca and Y. Adjei, Applications of antimicrobial photodynamic therapy against bacterial biofilms, Int. J. Mol. Sci., 2022, 23, 3209 CrossRef CAS PubMed.
  178. R. Youf, M. Müller, A. Balasini, F. Thétiot, M. Müller, A. Hascoët, U. Jonas, H. Schönherr, G. Lemercier and T. Montier, Antimicrobial photodynamic therapy: Latest developments with a focus on combinatory strategies, Pharmaceutics, 2021, 13, 1995 CrossRef CAS PubMed.
  179. A. Fraix and S. Sortino, Combination of PDT photosensitizers with NO photodononors, Photochem. Photobiol. Sci., 2018, 17, 1709–1727 CrossRef CAS PubMed.
  180. V. Pérez-Laguna, I. García-Luque, S. Ballesta, A. Rezusta and Y. Gilaberte, Photodynamic therapy combined with antibiotics or antifungals against microorganisms that cause skin and soft tissue infections: A planktonic and biofilm approach to overcome resistances, Pharmaceuticals, 2021, 14, 603 CrossRef PubMed.
  181. L. Beytollahi, M. Pourhajibagher, N. Chiniforush, R. Ghorbanzadeh, R. Raoofian, B. Pourakbari and A. Bahador, The efficacy of photodynamic and photothermal therapy on biofilm formation of Streptococcus mutans: An in vitro study, Photodiagn. Photodyn. Ther., 2017, 17, 56–60 CrossRef CAS PubMed.
  182. Q. Gao, D. Huang, Y. Deng, W. Yu, Q. Jin, J. Ji and G. Fu, Chlorin e6 (Ce6)-loaded supramolecular polypeptide micelles with enhanced photodynamic therapy effect against Pseudomonas aeruginosa, Chem. Eng. J., 2021, 417, 129334 CrossRef CAS.
  183. X. Cai, J. Tian, J. Zhu, J. Chen, L. Li, C. Yang, J. Chen and D. Chen, Photodynamic and photothermal co-driven CO-enhanced multi-mode synergistic antibacterial nanoplatform to effectively fight against biofilm infections, Chem. Eng. J., 2021, 426, 131919 CrossRef CAS.
  184. L. Mei, Z. Xu, Z. Miao, M. Yun, Y. Luan, D. Yang and L. Xia, Polymyxin B-functionalized phthalocyanine for chemo-photodynamic antibacterial therapy in enhanced wound healing, New J. Chem., 2021, 45, 6450–6457 RSC.
  185. A. Woźniak, B. Kruszewska, M. K. Pierański, M. Rychłowski and M. Grinholc, Antimicrobial photodynamic inactivation affects the antibiotic susceptibility of Enterococcus spp. clinical isolates in biofilm and planktonic cultures, Biomolecules, 2021, 11, 693 CrossRef PubMed.
  186. M.-H. Shih and F.-C. Huang, Effects of photodynamic therapy on rapidly growing nontuberculous mycobacteria keratitis, Invest Ophthalmol. Visual Sci., 2011, 52, 223–229 CrossRef PubMed.
  187. J. Almeida, J. P. Tomé, M. G. Neves, A. C. Tomé, J. A. Cavaleiro, Â. Cunha, L. Costa, M. A. Faustino and A. Almeida, Photodynamic inactivation of multidrug-resistant bacteria in hospital wastewaters: influence of residual antibiotics, Photochem. Photobiol. Sci., 2014, 13, 626–633 CrossRef CAS PubMed.
  188. X. Hou, L. Yang, J. Liu, Y. Zhang, L. Chu, C. Ren, F. Huang and J. Liu, Silver-decorated, light-activatable polymeric antimicrobials for combined chemo-photodynamic therapy of drug-resistant bacterial infection, Biomater. Sci., 2020, 8, 6350–6361 RSC.
  189. J. Dolansky, P. Henke, P. Kubat, A. Fraix, S. Sortino and J. Mosinger, Polystyrene nanofiber materials for visible-light-driven dual antibacterial action via simultaneous photogeneration of NO and O2 (1Δg), ACS Appl. Mater. Interfaces, 2015, 7, 22980–22989 CrossRef CAS PubMed.
  190. M. Pourhajibagher, G. Plotino, N. Chiniforush and A. Bahador, Dual wavelength irradiation antimicrobial photodynamic therapy using indocyanine green and metformin doped with nano-curcumin as an efficient adjunctive endodontic treatment modality, Photodiagn. Photodyn. Ther., 2020, 29, 101628 CrossRef CAS PubMed.
  191. A. A. Al-Kheraif, A. A. Khan, N. A. AlMufareh, D. D. Divakar, H. Dewan, S. Al-Jadani, J. Alrahimi, S. Hassoubah and K. S. Allemailem, Photodynamic antimicrobial chemotherapy through photosensitizers loaded poly-l-glycolic acid on Candida albicans in denture lining material: Release, biological and hardness study, Photodiagn. Photodyn. Ther., 2021, 33, 102134 CrossRef CAS PubMed.
  192. Q. Yu, X. Huang, T. Zhang, W. Wang, D. Yang, J. Shao and X. Dong, Near-infrared Aza-BODIPY dyes through molecular surgery for enhanced photothermal and photodynamic antibacterial therapy, Chem. Res. Chin. Univ., 2021, 37, 951–959 CrossRef CAS.
  193. D. Bagchi, V. S. Rathnam, P. Lemmens, I. Banerjee and S. K. Pal, NIR-light-active ZnO-based nanohybrids for bacterial biofilm treatment, ACS Omega, 2018, 3, 10877–10885 Search PubMed.
  194. W. Teng, Z. Zhang, Y. Wang, Y. Ye, E. Yinwang, A. Liu, X. Zhou, J. Xu, C. Zhou and H. Sun, Iodine Immobilized Metal–Organic Framework for NIR-Triggered Antibacterial Therapy on Orthopedic Implants, Small, 2021, 17, 2102315 CrossRef CAS PubMed.
  195. H. Lv, Y. Zhang, P. Chen, J. Xue, X. Jia and J. Chen, Enhanced synergistic antibacterial activity through a smart platform based on UiO-66 combined with photodynamic therapy and chemotherapy, Langmuir, 2020, 36, 4025–4032 Search PubMed.
  196. Y.-Y. Xie, Y.-W. Zhang, X.-Z. Liu, X.-F. Ma, X.-T. Qin, S.-R. Jia and C. Zhong, Aggregation-induced emission-active amino acid/berberine hydrogels with enhanced photodynamic antibacterial and anti-biofilm activity, Chem. Eng. J., 2021, 413, 127542 CrossRef CAS.
  197. J. Sun, Y. Fan, W. Ye, L. Tian, S. Niu, W. Ming, J. Zhao and L. Ren, Near-infrared light triggered photodynamic and nitric oxide synergistic antibacterial nanocomposite membrane, Chem. Eng. J., 2021, 417, 128049 Search PubMed.
  198. Q. Yao, Z. Ye, L. Sun, Y. Jin, Q. Xu, M. Yang, Y. Wang, Y. Zhou, J. Ji and H. Chen, Bacterial infection microenvironment-responsive enzymatically degradable multilayer films for multifunctional antibacterial properties, J. Mater. Chem. B, 2017, 5, 8532–8541 RSC.
  199. Y. Wang, Y. Jin, W. Chen, J. Wang, H. Chen, L. Sun, X. Li, J. Ji, Q. Yu and L. Shen, Construction of nanomaterials with targeting phototherapy properties to inhibit resistant bacteria and biofilm infections, Chem. Eng. J., 2019, 358, 74–90 Search PubMed.
  200. M. A. Castriciano, R. Zagami, M. P. Casaletto, B. Martel, M. Trapani, A. Romeo, V. Villari, M. T. Sciortino, L. Grasso and S. Guglielmino, Poly (carboxylic acid)-cyclodextrin/anionic porphyrin finished fabrics as photosensitizer releasers for antimicrobial photodynamic therapy, Biomacromolecules, 2017, 18, 1134–1144 CrossRef CAS PubMed.
  201. F. Nederberg, Y. Zhang, J. P. Tan, K. Xu, H. Wang, C. Yang, S. Gao, X. D. Guo, K. Fukushima and L. Li, Biodegradable nanostructures with selective lysis of microbial membranes, Nat. Chem., 2011, 3, 409–414 CrossRef CAS PubMed.
  202. Y. Zhao, M. Hu, Y. Zhang, J. Liu, C. Liu, S. K. Choi, Z. Zhang and L. Song, Multifunctional therapeutic strategy of Ag-synergized dual-modality upconversion nanoparticles to achieve the rapid and sustained cidality of methicillin-resistant Staphylococcus aureus, Chem. Eng. J., 2020, 385, 123980 CrossRef.
  203. J. Dolanský, P. Henke, Z. Malá, L. Žárská, P. Kubát and J. Mosinger, Antibacterial nitric oxide-and singlet oxygen-releasing polystyrene nanoparticles responsive to light and temperature triggers, Nanoscale, 2018, 10, 2639–2648 RSC.
  204. E. J. Son, J. H. Kim, K. Kim and C. B. Park, Quinone and its derivatives for energy harvesting and storage materials, J. Mater. Chem. A, 2016, 4, 11179–11202 RSC.
  205. J. K. Boparai and P. K. Sharma, Mini review on antimicrobial peptides, sources, mechanism and recent applications, Protein Pept. Lett., 2020, 27, 4–16 CrossRef CAS PubMed.
  206. A. de Breij, M. Riool, R. A. Cordfunke, N. Malanovic, L. de Boer, R. I. Koning, E. Ravensbergen, M. Franken, T. van der Heijde and B. K. Boekema, The antimicrobial peptide SAAP-148 combats drug-resistant bacteria and biofilms, Sci. Transl. Med., 2018, 10, eaan4044 Search PubMed.
  207. N. G. Oliveira Júnior, C. M. Souza, D. F. Buccini, M. H. Cardoso and O. L. Franco, Antimicrobial peptides: structure, functions and translational applications, Nat. Rev. Microbiol., 2025, 1–14 Search PubMed.
  208. G. Buda De Cesare, S. A. Cristy, D. A. Garsin and M. C. Lorenz, Antimicrobial peptides: a new frontier in antifungal therapy, mBio, 2020, 11, e02123-20 CrossRef PubMed.
  209. Q.-Y. Zhang, Z.-B. Yan, Y.-M. Meng, X.-Y. Hong, G. Shao, J.-J. Ma, X.-R. Cheng, J. Liu, J. Kang and C.-Y. Fu, Antimicrobial peptides: mechanism of action, activity and clinical potential, Mil. Med. Res., 2021, 8, 1–25 Search PubMed.
  210. H.-Q. Zhang, C. Sun, N. Xu and W. Liu, The current landscape of the antimicrobial peptide melittin and its therapeutic potential, Front. Immunol., 2024, 15, 1326033 Search PubMed.
  211. I. Nagaoka, H. Tamura and J. Reich, Therapeutic potential of cathelicidin peptide LL-37, an antimicrobial agent, in a murine sepsis model, Int. J. Mol. Sci., 2020, 21, 5973 CrossRef CAS PubMed.
  212. S. Zhang, M. Ma, Z. Shao, J. Zhang, L. Fu, X. Li, W. Fang and L. Gao, Structure and formation mechanism of antimicrobial peptides temporin b-and l-induced tubular membrane protrusion, Int. J. Mol. Sci., 2021, 22, 11015 CrossRef CAS PubMed.
  213. C. A. Roque-Borda, L. M. D. G. Primo, K. P. Medina-Alarcón, I. C. Campos, C. D. F. Nascimento, M. M. Saraiva, A. Berchieri Junior, A. M. Fusco-Almeida, M. J. S. Mendes-Giannini and J. Perdigão, Antimicrobial peptides: a promising alternative to conventional antimicrobials for combating polymicrobial biofilms, Adv. Sci., 2025, 12, 2410893 CrossRef CAS PubMed.
  214. U. Silphaduang and E. J. Noga, Peptide antibiotics in mast cells of fish, Nature, 2001, 414, 268–269 CrossRef CAS PubMed.
  215. K. A. Henzler Wildman, D.-K. Lee and A. Ramamoorthy, Mechanism of lipid bilayer disruption by the human antimicrobial peptide, LL-37, Biochemistry, 2003, 42, 6545–6558 Search PubMed.
  216. K. Scherer, I. Wiedemann, C. Ciobanasu, H.-G. Sahl and U. Kubitscheck, Aggregates of nisin with various bactoprenol-containing cell wall precursors differ in size and membrane permeation capacity, Biochim. Biophys. Acta, Biomembr., 2013, 1828, 2628–2636 Search PubMed.
  217. K. R. Parducho, B. Beadell, T. K. Ybarra, M. Bush, E. Escalera, A. T. Trejos, A. Chieng, M. Mendez, C. Anderson and H. Park, The antimicrobial peptide human beta-defensin 2 inhibits biofilm production of Pseudomonas aeruginosa without compromising metabolic activity, Front. Immunol., 2020, 11, 805 Search PubMed.
  218. Y. Takada, H. Itoh, A. Paudel, S. Panthee, H. Hamamoto, K. Sekimizu and M. Inoue, Discovery of gramicidin A analogues with altered activities by multidimensional screening of a one-bead-one-compound library, Nat. Commun., 2020, 11, 4935 CrossRef CAS PubMed.
  219. X. Yu, S. Jia, S. Yu, Y. Chen, C. Zhang, H. Chen and Y. Dai, Recent advances in melittin-based nanoparticles for antitumor treatment: from mechanisms to targeted delivery strategies, J. Nanobiotechnol., 2023, 21, 454 CrossRef CAS PubMed.
  220. E. V. Ledger, A. Sabnis and A. M. Edwards, Polymyxin and lipopeptide antibiotics: membrane-targeting drugs of last resort, Microbiology, 2022, 168, 001136 CrossRef CAS PubMed.
  221. O. V. Kondrashov, M. V. Volovik, Z. G. Denieva, P. K. Gifer, T. R. Galimzyanov, P. I. Kuzmin, O. V. Batishchev and S. A. Akimov, Dialectics of antimicrobial peptides II: Theoretical models of pore formation and membrane protection, Langmuir, 2025, 41, 19003–19022 Search PubMed.
  222. D. W. Juhl, E. Glattard, C. Aisenbrey and B. Bechinger, Antimicrobial peptides: mechanism of action and lipid-mediated synergistic interactions within membranes, Faraday Discuss., 2021, 232, 419–434 RSC.
  223. G. Satchanska, S. Davidova and A. Gergova, Diversity and mechanisms of action of plant, animal, and human antimicrobial peptides, Antibiotics, 2024, 13, 202 CrossRef CAS PubMed.
  224. D. Balleza, The Role of Flexibility in the Bioactivity of Short α-Helical Antimicrobial Peptides, Antibiotics, 2025, 14, 422 Search PubMed.
  225. H. Haddad, R. Mejri and A. Zaïri, Evaluation of the Antibacterial Activity of New Dermaseptin Derivatives against Acinetobacter baumannii, Pharmaceuticals, 2024, 17, 171 Search PubMed.
  226. A. J. Clarke, Another brick in the wall, Nat. Chem. Biol., 2017, 13, 695–696 Search PubMed.
  227. K. N. Allen and B. Imperiali, Structural and mechanistic themes in glycoconjugate biosynthesis at membrane interfaces, Curr. Opin. Struct. Biol., 2019, 59, 81–90 Search PubMed.
  228. M. Graf and D. N. Wilson, Intracellular antimicrobial peptides targeting the protein synthesis machinery, Antimicrobial Peptides: Basics for Clinical Application, 2019, pp. 73–89 Search PubMed.
  229. Y. Zhang, S. Liu, S. Li, Y. Cheng, L. Nie, G. Wang, C. Lv, W. Wei, C. Cheng and F. Hou, Novel short antimicrobial peptide isolated from Xenopus laevis skin, J. Pept. Sci., 2017, 23, 403–409 CrossRef CAS PubMed.
  230. H. Lee, J. S. Hwang and D. G. Lee, Periplanetasin-4, a novel antimicrobial peptide from the cockroach, inhibits communications between mitochondria and vacuoles, Biochem. J., 2019, 476, 1267–1284 CrossRef CAS PubMed.
  231. Y. Luo and Y. Song, Mechanism of antimicrobial peptides: antimicrobial, anti-inflammatory and antibiofilm activities, Int. J. Mol. Sci., 2021, 22, 11401 CrossRef CAS PubMed.
  232. M. Martinez, S. Gonçalves, M. R. Felício, P. Maturana, N. C. Santos, L. Semorile, A. Hollmann and P. C. Maffía, Synergistic and antibiofilm activity of the antimicrobial peptide P5 against carbapenem-resistant Pseudomonas aeruginosa, Biochim. Biophys. Acta, Biomembr., 2019, 1861, 1329–1337 CrossRef CAS PubMed.
  233. M. Demirci, A. Yigin and C. Demir, Efficacy of antimicrobial peptide LL-37 against biofilm forming Staphylococcus aureus strains obtained from chronic wound infections, Microb. Pathog., 2022, 162, 105368 CrossRef CAS PubMed.
  234. B. Li, W. Kang, H. Liu, Y. Wang, C. Yu, X. Zhu, J. Dou, H. Cai and C. Zhou, The antimicrobial activity of Cbf-K 16 against MRSA was enhanced by β-lactamantibiotics through cell wall non-integrity, Arch. Pharmacal Res., 2016, 39, 978–988 CrossRef CAS PubMed.
  235. Z. Si, H. W. Lim, M. Y. Tay, Y. Du, L. Ruan, H. Qiu, R. Zamudio-Vazquez, S. Reghu, Y. Chen and W. S. Tiong, A glycosylated cationic block poly (β-peptide) reverses intrinsic antibiotic resistance in all ESKAPE Gram-negative bacteria, Angew. Chem., Int. Ed., 2020, 59, 6819–6826 CrossRef CAS PubMed.
  236. H. Mathur, D. Field, M. C. Rea, P. D. Cotter, C. Hill and R. P. Ross, Bacteriocin-antimicrobial synergy: a medical and food perspective, Front. Microbiol., 2017, 8, 1205 CrossRef PubMed.
  237. X. Peng, Y. Luo, T. Xu, Z. Chen, P. Chen, C. Hu and S. Liu, Antibiotic-conjugated antimicrobial peptides for enhanced bacterial inhibition, RSC Adv., 2025, 15, 19751–19761 RSC.
  238. B. C. Koppen, P. P. Mulder, L. de Boer, M. Riool, J. W. Drijfhout and S. A. Zaat, Synergistic microbicidal effect of cationic antimicrobial peptides and teicoplanin against planktonic and biofilm-encased Staphylococcus aureus, Int. J. Antimicrob. Agents, 2019, 53, 143–151 CrossRef CAS PubMed.
  239. S. Li, P. She, L. Zhou, X. Zeng, L. Xu, Y. Liu, L. Chen and Y. Wu, High-throughput identification of antibacterials against Pseudomonas aeruginosa, Front. Microbiol., 2020, 11, 591426 CrossRef PubMed.
  240. S. Tarvirdipour, S. N. Abdollahi, J. Köser, M. Bina, C.-A. Schoenenberger and C. G. Palivan, Enhanced antimicrobial protection through surface immobilization of antibiotic-loaded peptide multicompartment micelles, J. Mater. Chem. B, 2025, 13, 5365–5379 RSC.
  241. H. Sun, Y. Hong, Y. Xi, Y. Zou, J. Gao and J. Du, Synthesis, self-assembly, and biomedical applications of antimicrobial peptide–polymer conjugates, Biomacromolecules, 2018, 19, 1701–1720 CrossRef CAS PubMed.
  242. E. Lepeltier, P. Rijo, F. Rizzolio, R. Popovtzer, V. Petrikaite, Y. G. Assaraf and C. Passirani, Nanomedicine to target multidrug resistant tumors, Drug Resist. Updates, 2020, 52, 100704 CrossRef PubMed.
  243. A. O. Fadaka, N. R. S. Sibuyi, A. M. Madiehe and M. Meyer, Nanotechnology-based delivery systems for antimicrobial peptides, Pharmaceutics, 2021, 13, 1795 CrossRef CAS PubMed.
  244. S. Gera, E. Kankuri and K. Kogermann, Antimicrobial peptides–unleashing their therapeutic potential using nanotechnology, Pharmacol. Ther., 2022, 232, 107990 CrossRef CAS PubMed.
  245. X. Luo, H. Chen, Y. Song, Z. Qin, L. Xu, N. He, Y. Tan and W. Dessie, Advancements, challenges and future perspectives on peptide-based drugs: Focus on antimicrobial peptides, Eur. J. Pharm. Sci., 2023, 181, 106363 CrossRef CAS PubMed.
  246. B. Lin, A. Hung, W. Singleton, K. K. Darmawan, R. Moses, B. Yao, H. Wu, A. Barlow, M. A. Sani and A. J. Sloan, The effect of tailing lipidation on the bioactivity of antimicrobial peptides and their aggregation tendency: Special issue: Emerging investigators, Aggregate, 2023, 4, e329 CrossRef CAS.
  247. E. Malik, S. R. Dennison, F. Harris and D. A. Phoenix, pH dependent antimicrobial peptides and proteins, their mechanisms of action and potential as therapeutic agents, Pharmaceuticals, 2016, 9, 67 Search PubMed.
  248. N. Chen and C. Jiang, Antimicrobial peptides: Structure, mechanism, and modification, Eur. J. Med. Chem., 2023, 255, 115377 CrossRef CAS PubMed.
  249. C. H. Chen and T. K. Lu, Development and challenges of antimicrobial peptides for therapeutic applications, Antibiotics, 2020, 9, 24 CrossRef CAS PubMed.
  250. A. Mazurkiewicz-Pisarek, J. Baran and T. Ciach, Antimicrobial peptides: challenging journey to the pharmaceutical, biomedical, and cosmeceutical use, Int. J. Mol. Sci., 2023, 24, 9031 CrossRef CAS PubMed.
  251. J. Sun, Y. Xia, D. Li, Q. Du and D. Liang, Relationship between peptide structure and antimicrobial activity as studied by de novo designed peptides, Biochim. Biophys. Acta, Biomembr., 2014, 1838, 2985–2993 CrossRef CAS PubMed.

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