Next Article in Journal
Unexplained High Prevalence of ESBL-Escherichia coli Among Cattle and Pigs in Peru
Previous Article in Journal
Evaluation of Ceftaroline Use in Pediatric Patients: A Retrospective Case Series
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Biofilms Exposed: Innovative Imaging and Therapeutic Platforms for Persistent Infections

by
Manasi Haval
1,
Chandrashekhar Unakal
2,
Shridhar C. Ghagane
3,
Bijay Raj Pandit
2,
Esther Daniel
4,
Parbatee Siewdass
4,
Kingsley Ekimeri
5,
Vijayanandh Rajamanickam
6,
Angel Justiz-Vaillant
2,
Kathy-Ann A. Lootawan
5,
Fabio Muniz De Oliveira
7,
Nivedita Bashetti
8,
Tatheer Alam Naqvi
9,
Arun Shettar
1 and
Pramod Bhasme
1,*
1
Department of Pre-Clinical Research and Drug Development, Cytxon Biosolutions Pvt. Ltd., Hubballi 580031, Karnataka, India
2
Department of Pathology/Microbiology & Pharmacology, The University of the West Indies, St. Augustine Campus, St. Augustine 330912, Trinidad and Tobago
3
KAHER’s Dr. Prabhakar Kore Basic Science Research Centre, KLE Academy of Higher Education and Research, JNMC Campus, Belagavi 590010, Karnataka, India
4
School of Nursing, Faculty of Medical Sciences, The University of the West Indies, St. Augustine Campus, St. Augustine 330912, Trinidad and Tobago
5
Optometry Unit-Clinical Surgical Science, The University of the West Indies, St. Augustine Campus, St. Augustine 330912, Trinidad and Tobago
6
Department of Computing and Information Technology, The University of the West Indies, St. Augustine Campus, St. Augustine 330912, Trinidad and Tobago
7
Department of Biomedical Sciences, Estácio University Center of Goiás, Goiânia 74063-010, GO, Brazil
8
Department of Biotechnology and Microbiology, Karnataka University, Dharwad 580003, Karnataka, India
9
Department of Biotechnology, COMSATS University Islamabad, Abbottabad Campus, Abbottabad 22060, Pakistan
*
Author to whom correspondence should be addressed.
Antibiotics 2025, 14(9), 865; https://doi.org/10.3390/antibiotics14090865
Submission received: 8 July 2025 / Revised: 18 August 2025 / Accepted: 19 August 2025 / Published: 28 August 2025

Abstract

Biofilms constitute a significant challenge in the therapy of infectious diseases, offering remarkable resistance to both pharmacological treatments and immunological elimination. This resilience is orchestrated through the regulation of extracellular polymeric molecules, metabolic dormancy, and quorum sensing, enabling biofilms to persist in both clinical and industrial environments. The resulting resistance exacerbates chronic infections and contributes to mounting economic burdens. This review examines the molecular and structural complexities that drive biofilm persistence and critically outlines the limitations of conventional diagnostic and therapeutic approaches. We emphasize advanced technologies such as super-resolution microscopy, microfluidics, and AI-driven modeling that are reshaping our understanding of biofilm dynamics and heterogeneity. Further, we highlight recent progress in biofilm-targeted therapies, including CRISPR-Cas-modified bacteriophages, quorum-sensing antagonists, enzyme-functionalized nanocarriers, and intelligent drug-delivery systems responsive to biofilm-specific cues. We also explore the utility of in vivo and ex vivo models that replicate clinical biofilm complexity and promote translational applicability. Finally, we discuss emerging interventions grounded in synthetic biology, such as engineered probiotic gene circuits and self-regulating microbial consortia, which offer innovative alternatives to conventional antimicrobials. Collectively, these interdisciplinary strategies mark a paradigm shift from reactive antibiotic therapy to precision-guided biofilm management. By integrating cutting-edge technologies with systems biology principles, this review proposes a comprehensive framework for disrupting biofilm architecture and redefining infection treatment in the post-antibiotic era.

1. Introduction

Biofilm-associated infections are formidable challenges to manage across clinical and industrial domains, due to their resilience and intrinsic resistance to standard antimicrobial therapies. Despite substantial advancements in medicine, current interventions frequently fall short of completely eradicating biofilms in persistent infections, which often result in prolonged hospitalization, increased morbidity, and escalating healthcare expenditures. In clinical contexts, biofilms are implicated in up to 80% chronic infections, particularly those involving indwelling medical devices, non-healing wounds, for example, diabetic foot ulcers (DFUs), and respiratory complications in cystic fibrosis patients [1].
Structurally, biofilms are sophisticated microbial consortia encased in a self-produced extracellular polymeric matrix composed of polysaccharides, proteins, lipids, and extracellular DNA [2,3]. This matrix provides structural integrity and environmental protection while facilitating the adhesion to biotic and abiotic surfaces, therefore enhancing microbial persistence in adverse conditions. Outside of clinical settings, biofilms significantly contribute to biofouling in water-treatment systems, food-processing facilities, and energy infrastructure, with global economic losses estimated in the billions annually [4].
A defining attribute of bacteria-forming biofilms is their extraordinary resistance to antimicrobial agents, often up to 1000-fold higher than that of planktonic bacteria attributable to interrelated defense mechanisms. These defenses combine the EPS-mediated diffusion barrier, metabolic dormancy within subpopulations of persister cells, quorum sensing, release of surfactants, enhanced efflux-pump expression, and horizontal gene transfer facilitating rapid accumulation of resistance determinants [5]. Simultaneously, bacterial biofilms undermine host immune responses by hindering phagocytosis, degrading antimicrobial peptides, and resisting complement activation [6]. These multifaceted resistance strategies render traditional antibiotic regimens and mechanical removal approaches largely ineffective, often resulting in recurrent infections like DFU patients and heightened antimicrobial resistance [7].
Given these limitations, there is an urgent need to develop and deploy advanced therapeutic strategies tailored specifically to the unique biology of bacterial biofilms. Promising avenues encompass the application of antimicrobial nanoparticles that are capable of penetrating the biofilm matrix [8], CRISPR-Cas-modified bacteriophages engineered for targeted lysis [9], quorum-sensing inhibitors that interfere with microbial communication [10], targeting efflux-pump-producing genes and artificial intelligence-driven platforms for accelerated discovery of novel antibiofilm agents [11]. Simultaneously, innovations in biofilm imaging (e.g., confocal microscopy, microfluidics) and systems biology approaches (e.g., metagenomics, transcriptomics, and proteomics) are redefining our understanding of biofilm dynamics and therapeutic vulnerabilities [12,13].
This review critically examines the limitations of conventional biofilm therapies, clarifying the biological and structural complexities that underlie their resistance. Additionally, this discussion will provide an evaluation of the most recent and impactful technological advances poised to transform biofilm management, from nanotechnology and phage therapy to enzyme-based disruption and targeted drug delivery. By integrating insights from interdisciplinary research, this article aims to provide a forward-looking perspective on combating biofilms, paving the way toward more effective, durable, and precision-guided therapeutic interventions.

Objectives of This Review

The following are the objectives of this review, considering the growing clinical burden of infections linked to biofilms and the growing interest in novel treatment approaches:
  • Provide an overview of the current understanding of biology, structural organization, and clinical significance of biofilms in relation to different types of infections.
  • Examine the shortcomings of traditional antimicrobial methods in terms of successfully eliminating biofilms.
  • Describe the latest developments in biofilm-targeted treatment approaches, such as CRISPR-Cas-based interventions, bacteriophage therapy, enzyme-mediated methods, nanotechnology-based systems, and quorum-sensing inhibition.
  • Highlight the translational significance of preclinical in vitro, ex vivo, and in vivo models that resemble clinical biofilms.
  • Examine intelligent drug-delivery systems that respond to cues unique to biofilms.
  • Determine the present obstacles, unmet research needs, and possible avenues for the creation of clinically feasible anti-biofilm treatments.

2. Classical Techniques in Biofilm Research: Historical Foundations and Limitations

Traditional techniques have been crucial in developing the understanding of bacterial biofilm biology. Early methods such as crystal violet staining, colony-forming unit (CFU) enumeration, motility assay, and light microscopy were among the first tools employed to identify and characterize bacterial biofilms as structured microbial communities [14,15]. These methodologies have allowed researchers to assess surface adherence, structural dynamics, estimate total biomass, and evaluate antimicrobial tolerance in vitro, which provides key insights into biofilm formation and resilience [16]. Despite the lack of quality in the resolution, sensitivity, and dynamic monitoring compared to the capabilities of modern technologies, they remain widely used due to their simplicity, reproducibility, and cost-effectiveness. Traditional assays still continue to serve as reliable benchmarks in comparative studies and high-throughput screening applications [17,18,19]. The use of these techniques has been critical in defining the behavioral and physiological differences between biofilm-associated and planktonic bacterial cells [20]. These methods helped shape early discoveries and lay the groundwork for the development of the advanced imaging, molecular, and computational tools that now dominate biofilm research [21,22] (Figure 1).

2.1. Crystal Violet Assay

The crystal violet assay is a colorimetric assay used to measure the total biomass of biofilm developed on solid surfaces (Figure 1a). Biofilms are stained with crystal violet, which adheres to both microbial cells and extracellular matrix constituents [23,24]. Post-staining, the dye is solubilized with ethanol or acetic acid, and absorbance is quantified spectrophotometrically (generally at 570–600 nm). The assay is straightforward, cost-effective, and highly compatible with high-throughput screening. Nonetheless, it fails to distinguish between viable and non-viable cells, nor does it furnish details on biofilm structure or composition and dynamics [25].

2.2. Colony-Forming Unit (CFU) Counts

CFUs are used to quantify the number of viable and culturable bacteria present in biofilms (Figure 1b). Biofilms are subjected to physical or enzymatic disruption, followed by dilution and plating on agar [26,27]. Although CFUs on selective media indicate the presence of culturable bacteria, this does not necessarily represent the states of infection of the bacteria that were viable or metabolically active at the original tissue site. Furthermore, due to the low metabolic activity of bacteria within biofilms, the bacterial cells are involved in the infection. Environmental stress, sampling procedures, or biofilm-associated dormancy may allow previously non-viable cells to become culturable during in vitro incubation, leading to potential overestimation of in situ viability. This method is also subject to being influenced by partial biofilm recovery or sampling variability [28,29].

2.3. Tube Method

The tube method evaluates biofilm growth on the inner surfaces of test tubes (Figure 1c) [30,31]. Following incubation, the tubes are rinsed, dyed with crystal violet, and examined visually for the presence of biofilm. Although this method requires minimal setup, it presents limitations in quantification, reproducibility, and sensitivity. Furthermore, it does not allow for the identification of microcolonies of biofilm released during the dispersion phase. It is currently obsolete but is historically significant for early biofilm detection [32,33].

2.4. Congo Red Agar Assay

The Congo red agar assay is a common qualitative test used to check for the production of extracellular polysaccharides (EPSs) and the formation of biofilms (Figure 1d) [34]. In this method, bacteria are grown on agar medium with Congo red dye and sometimes sucrose to help make the matrix. Strains that make biofilms usually have black, dry, crystalline, or wrinkled colonies, while strains that do not make biofilms usually have smooth red or pink colonies. This easy and cheap method makes it easy to quickly tell the difference between matrix producers and non-producers. But the method is still semi-quantitative and is affected by the type of media, the length of time it is incubated, and the conditions of the culture [35,36]. Also, it does not give any information about cell viability, total biomass, or the structure of biofilms. Therefore, Congo red agar is very much suited as a preliminary screening assay and is often complemented by more quantitative methods such as crystal violet staining or CFU enumeration [37].

2.5. Drip-Flow and Early Flow-Cell Systems

The drip-flow and early flow-cell systems are used to replicate dynamic biofilm formation under shear forces (Figure 1e). Drip-flow reactors expose surfaces to continuous medium flow, while flow cells allow for real-time microscopy during biofilm growth [38]. These early models introduced physiologically relevant growth conditions but required complex setup and lacked standardization, paving the way for modern microfluidic platforms [39].

2.6. Biomass Dry-Weight Measurement

Biomass dry-weight measurements quantify total biofilm mass by drying and weighing collected material (Figure 1f). It is straightforward and does not require staining or labeling [40,41]. It is not used to distinguish between live cells, dead cells, and extracellular matrix, and offers no information on structure or function. It is often used to complement other quantitative or visual techniques [42].

2.7. Light Microscopy

Light microscopy uses a variation between bright-field, phase-contrast, and fluorescence imaging of stained or live biofilms [43,44]. This rapid visualization technique allows for a look into morphology and coverage, often in conjunction with Gram staining or fluorescent dyes. However, light microscopy has limited resolution and penetration, making it suboptimal for analyzing thick or complex biofilms (Figure 1g).

2.8. Scanning Electron Microscopy (SEM)

SEM provides high-resolution surface images of biofilms, revealing structural organization, cellular arrangement, and extracellular matrix features [45,46]. Samples are fixed, dehydrated, and coated with conductive material before imaging (Figure 1h). This technique is valuable for visualizing biofilm morphology on medical or industrial surfaces. Limitations include no live-cell imaging, and require specialized preparation [47,48].

2.9. Transmission Electron Microscopy (TEM)

TEM enables the observation of the interior ultrastructure of biofilms via ultrathin sectioning of fixed specimens (Figure 1i) [49,50]. It offers nanometer-scale resolution, which allows for the visualization of intracellular features and matrix density [51,52]. Nonetheless, it is technically challenging, time-intensive, and inappropriate for high-throughput investigations or live imaging. TEM is predominantly utilized for comprehensive structural study [53]. Owing to the constraints of classical methodologies, such as their destructive characteristics, restricted resolution, and incapacity to observe live or dynamic biofilm activities, researchers have innovated advanced technologies to enhance biofilm study. These contemporary instruments provide enhanced sensitivity, spatial resolution, and temporal dynamics, facilitating real-time, comprehensive analysis of biofilm structure, physiology, and interactions. The following are some of the most revolutionary techniques utilized in biofilm research.

3. Cutting-Edge Technologies and Models in Biofilm Research

3.1. Advanced Microscopy and Imaging Techniques in Biofilm Research

To comprehend the intricate spatial arrangement, metabolic function, and therapeutic resistance of biofilms there is a necessity for high-resolution, dynamic imaging techniques [13,54]. In the last ten years, a range of sophisticated microscopy and imaging technologies has enhanced the capacity to visualize biofilms at micro- to nanoscale levels, in both in vitro and in vivo settings [55,56]. Here, we outline ten essential strategies commonly employed in biofilm research, detailing their principles, advantages, and specific applications (Figure 2). A summary of advanced imaging techniques used in biofilm research is presented in Table 1.

3.1.1. Stimulated Emission Depletion (STED) Microscopy

STED microscopy surpasses the diffraction limit of traditional optics by employing a depletion laser to restrict the release of fluorescence to a nanoscale volume [57,58]. This approach attains a resolution beneath 50 nm, rendering it very useful for revealing the ultrastructure of extracellular polymeric substances (EPSs) and intercellular spacing within biofilms [45,59]. STED is crucial for examining how the biofilm matrix obstructs antibiotic penetration and facilitates structural mapping at the nanoscale scale (Figure 2a). Although predominantly applied to fixed samples, its negligible phototoxicity through pulsed lasers renders it an effective instrument for structural biofilm characterization [60,61].

3.1.2. Lattice Light-Sheet Microscopy (LLSM)

Lattice light-sheet microscopy integrates temporal resolution with minimal phototoxicity, rendering it optimal for the live-cell imaging of dense, hydrated biofilms [62,63]. LLSM facilitates optical sectioning and real-time monitoring of nutrient flow, cellular motility, and structural remodeling through the generation of thin sheets of light (Figure 2b). This method is particularly effective for modeling host–microbe interactions and monitoring biofilm growth, detachment, and antibiotic responses over prolonged durations [64].

3.1.3. Stochastic Optical Reconstruction Microscopy (STORM)

The STORM technique achieves super-resolution imaging through the stochastic activation of fluorophores and through the reconstruction of molecular locations with nanoscale precision [65]. This technique details the peptidoglycan deposition, matrix-associated enzymes, and cellular changes throughout biofilm formation (Figure 2c). When utilized alongside fluorescent D-amino acids or alternative labels, STORM serves as an essential instrument for delineating drug–biofilm interactions and regional heterogeneity [51,66].

3.1.4. Confocal Laser Scanning Microscopy (CLSM)

CLSM is the benchmark for three-dimensional imaging of biofilms, especially for evaluating thickness, vitality, and spatial architecture [46]. It facilitates multi-channel fluorescence imaging and live-cell analysis within microfluidic chambers or infection models. CLSM is extensively utilized for assessing antibiotic penetration, monitoring biofilm growth dynamics, (Figure 2d) and integrating structural data with viability markers in both in vitro and in vivo investigations [67,68].

3.1.5. Atomic Force Microscopy (AFM)

AFM offers high-resolution surface topography and mechanical characterization of biofilms without requiring staining [69,70]. It measures characteristics like surface roughness, elasticity, and adhesion, providing distinctive insights on matrix cohesion and mechanical durability (Figure 2e). AFM enhances optical techniques by elucidating biomechanical responses to pharmacological interventions and biofilm–substrate interactions at the nanoscale [71,72].

3.1.6. Cryo-Electron Microscopy (Cryo-EM)

Cryo-EM preserves biofilm ultrastructure in a vitrified state, enabling near-atomic resolution of embedded biomolecules such as pili, ribosomes, and matrix proteins [50,73]. Though not suited for live imaging, it plays a crucial role in structural biology, mapping biofilm phage interactions, or antibiotic target engagement at molecular resolution (Figure 2f). Its integration with other super-resolution techniques supports multiscale biofilm analysis [74,75].

3.1.7. Optical Coherence Tomography (OCT)

OCT provides a label-free, non-invasive image of biofilms utilizing near-infrared light to capture cross-sectional structures [76,77]. It infiltrates several millimeters into turbid samples, rendering it especially advantageous in oral biofilms or those linked to medical equipment (Figure 2g). OCT facilitates real-time assessment of biofilm thickness, stratification, and fluid channels, making it suitable for dynamic flow investigations [38,78].

3.1.8. Raman Spectroscopy Imaging (RSI)

The RSI technique offers chemical composition maps of biofilms derived from molecular vibrational signatures (Figure 2h) [51]. This label-free method differentiates proteins, nucleic acids, and EPS in situ, uncovering metabolic heterogeneity and resistance mechanisms [79]. RSI is particularly effective in monitoring biochemical reactions to drugs and environmental stressors in complex or polymicrobial biofilms [80].

3.1.9. Bioluminescence Imaging (BLI)

BLI uses genetically engineered bacteria that emit light through luciferase activity, enabling real-time, non-invasive imaging of biofilm growth and infection dynamics in live animals [81,82]. Despite the limit in spatial resolution, BLI excels in longitudinal studies of metabolic activity, antimicrobial response, and disease progression in murine (Figure 2i) or catheter-based infection models [83].

3.1.10. Digital Holographic Microscopy (DHM)

DHM collects phase-shift data from unstained specimens, facilitating the reconstruction of 3D pictures and the real-time quantification of biomass, thickness, and motility [84,85]. It is an economical, non-invasive method suitable for prolonged imaging in microfluidic devices (Figure 2j). DHM is especially appropriate for kinetic investigations of biofilm development, antibiotic impacts, and high-throughput screening in microbiology [86].
Table 1. Summary of advanced imaging techniques in biofilm research.
Table 1. Summary of advanced imaging techniques in biofilm research.
TechniqueResolution
nm
Live ImagingLabel-FreeKey FeaturesTypical ApplicationsReference
Stochastic Optical Reconstruction
(STED) Microscopy
~30–50NoNoSuper-resolution; EPS structure; nanoscale matrix mappingAntibiotic penetration, EPS-targeted therapies[87]
Lattice Light-Sheet Microscopy~300 lateral, ~500 axialYesNoLow phototoxicity; fast volumetric imaging; dynamic events in 3D biofilmsHost–microbe interaction, live biofilm dynamics[63]
Stochastic Optical Reconstruction Microscopy (STORM)~20–30NoNoSingle-molecule localization; super-resolution of matrix, enzymesPeptidoglycan mapping, maturation tracking[88]
Confocal laser scanning electron microscopy (CLSM)~200–300YesNo3D reconstructions; multichannel imaging; live/dead discriminationBiofilm architecture, viability, drug diffusion studies[89]
Atomic Force Microscopy (AFM)~1 (surface)Yes (partial)YesNanoscale topography; mechanical properties; elasticity and adhesionMatrix cohesion, drug-induced biomechanical changes[90]
Cryo-Electron Microscopy (Cryo-EM)~0.3–1NoYesNear-atomic resolution; native-state imaging of molecular complexesStructural biology, biofilm–phage/protein interactions[91]
Optical Coherence Tomography (OCT)~1–15 µmYesYesDeep penetration; real-time label-free cross-sectionsMedical/industrial biofilms, flow-cell monitoring[92]
Raman Spectroscopy Imaging~300–500Yes (slow)YesMolecular composition; metabolic fingerprinting; antibiotic mappingChemical analysis, resistance zones, EPS structure[93]
Bioluminescence Imaging (BLI)~1–5 mm (low)YesYesReal-time in vivo imaging; metabolic activity monitoringInfection progression, therapeutic efficacy[83]
Digital Holographic Microscopy (DHM)~500 lateralYesYesLabel-free 3D imaging; real-time biomass/motility trackingHigh-throughput screening, early-stage studies[94]

3.2. AI-Driven Biofilm Modeling: Transforming Research and Therapeutic Development

Recent advancements in artificial intelligence (AI) are transforming biofilm research by equipping scientists with tools to anticipate the formation, behavior, and therapeutic responses of biofilms (Figure 3).
Ten significant AI modeling methodologies have been established for this emerging domain [95,96]. Convolutional Neural Networks (CNNs) facilitate the analysis of microscopic pictures by delineating biofilm structures and quantifying their thickness, EPS distribution, and heterogeneity (Figure 3a). Reinforcement Learning (RL) simulates the alterations of biofilms in reaction to treatments. It employs adaptive feedback to identify the optimal method for regulating nutrients or antibiotics [97,98]. Graph Neural Networks (GNNs) accurately depict quorum sensing and intercellular signaling by effectively capturing spatial correlations within three-dimensional biofilm matrices [99]. Generative Adversarial Networks (GANs) produce synthetic biofilm images. They add to datasets for unusual traits and model how biofilms grow in different settings [100,101]. Random Forests (RF) utilize genetic, proteomic, or metabolic data to forecast biofilm growth and identify critical characteristics that enhance its resistance. Support Vector Machines (SVMs) categorize bacterial phenotypes based on their stress responses or gene-expression profiles (Figure 3b). This aids in diagnostics and prediction modeling [12,102]. Autoencoders compress omics or imaging data to uncover latent patterns associated with the proliferation or dissemination of biofilms. This enables the elimination of noise and the identification of abnormalities [103]. Long Short-Term Memory (LSTM) networks elucidate the temporal dynamics of biofilms, predicting their evolution, dissemination, and therapeutic responses [104]. Bayesian Networks utilize clinical and omics data to ascertain the risk and resistance of biofilm by delineating the probability relationships with genes and phenotypes (Figure 3c). Ultimately, Physics-Informed Neural Networks (PINNs) integrate data-driven learning with physical concepts, such as fluid dynamics, to elucidate the behavior of biofilms on surfaces or inside flow. This links computational models to biological constraints [105,106]. These AI tools effectively complement experimental procedures to expedite the discovery of novel therapeutics and facilitate customized, systems-oriented strategies for biofilm management (Figure 3d).

3.3. In Vivo Models for Biofilm Research

In vivo models are necessary for connecting biofilm research results from the lab to the clinic [107,108]. To date, around 1100 in vivo models have successfully replicated clinical biofilms. These models help us understand how biofilms form, stay alive, affect the immune system, and how well treatments work in different parts of the body [109,110]. To study biofilms that form on devices, researchers put biomaterials like catheters, titanium disks, or mesh under the skin of mice and rats (Figure 3e). These models are great for testing colonization, immunological infiltration, and drug-delivery systems in a controlled setting [111,112]. Orthopedic implant models, usually in mice or rabbits, mimic infections in prosthetic joints by surgically putting pins or screws into the femur or tibia. A lot of people utilize them to test local antimicrobial treatments and biofilm-induced osteomyelitis [113,114]. Chronic wound biofilm models that use diabetic mice or pigs with weak immune systems mimic human wounds that take a long time to heal [115,116]. These are necessary for testing topical antibiofilm drugs and regenerative techniques [117]. Intra-tracheal instillation of P. aeruginosa is used in pulmonary infection models (Figure 3g), especially in cystic fibrosis (CF), zebrafish (Figure 3f) and pig models (Figure 3h), to replicate the thick mucus biofilms that are present in CF airways [118].
Models of urinary catheter-associated infections in rats and rabbits facilitate the examination of uropathogen biofilm development, encrustation, and inflammation within a therapeutically pertinent urological framework [119,120]. Conversely, rodent oral biofilm models emphasize the development of dental plaque and periodontitis caused by organisms such as S. mutans and P. gingivalis, facilitating the assessment of dental treatments [121,122]. Rabbit models of endocarditis, employing catheter-induced valve injury, provide a highly accurate simulation of heart valve infections characterized by biofilm formation under significant shear stress [123,124]. Gastrointestinal models employing gnotobiotic mice populated with specific microbial communities are crucial for analyzing mucosal biofilm–host interactions and dysbiosis [125]. Models of ear and sinus infections, created by introducing pathogens into the middle ear or nasal cavity of mice or rats, are utilized to investigate biofilms associated with otitis media and chronic rhinosinusitis [126,127]. Ex vivo human xenograft models entail the implantation of human tissues, such as skin or lung, into immunodeficient SCID mice. So far, approximately 162 ex vivo models have been reported to replicate clinical biofilms. These models provide high-resolution examinations of human-specific biofilm dynamics and individualized therapy evaluation [128,129]. Collectively, these models offer comprehensive frameworks for assessing antimicrobial tactics, host-pathogen interactions, and translational biofilm therapies across many clinical scenarios. The zebrafish embryo model emerged as a valuable tool for studying the in vivo biofilm formation of an opportunistic bacteria, P. aeruginosa. The P. aeruginosa was injected into the bloodstream as well as into the egg yolk of zebrafish to investigate the role of various virulence factors, including quorum sensing and type III secretion system, that participate in the development of biofilm and other virulence factors [130].
C. elegans is considered a model organism for studying quorum-sensing-regulated virulence factors, including biofilm formation, which can explain the interaction between bacteria and invertebrates [131]. Yersinia pseudotuberculosis forms biofilms on C. elegans that block the feeding in nematodes. C. elegans is considered a model organism for studying the virulence factors under the control of quorum sensing, including the biofilm [132].

4. Biofilm Control Approach

Effectively managing biofilms requires a multifaceted strategy that surpasses conventional antimicrobial treatments (Figure 4). Traditional therapies often fall short, prompting the development of innovative approaches targeting biofilm structure, genetics, and regulation [133,134]. Advances in synthetic biology, nanotechnology, quorum-sensing inhibition, engineered bacteriophages, and host immune modulation underpin these emerging interventions. Each method offers a unique mechanism ranging from gene silencing and matrix disruption to enhanced drug delivery and immune system reprogramming. Collectively, they represent a paradigm shift toward precise, adaptive, and sustainable biofilm control.

4.1. Synthetic Biology and Genetic Engineering Approaches for Biofilm Control

Synthetic biology is transforming the way we fight biofilms from blunt-force antibiotic regimens to precision-guided, genetically programmed solutions [135]. At the center of this revolution are CRISPR-Cas systems, powerful tools that allow scientists to target and silence specific biofilm-associated genes with high accuracy. CRISPR interference (CRISPRi) enables tunable, high-throughput repression of gene activity, helping identify the molecular circuits that drive biofilm resilience [136,137]. Even more compelling, engineered bacteriophages armed with CRISPR-Cas9 have demonstrated the ability to selectively destroy drug-resistant pathogens inside biofilms without harming beneficial microbes. Thus far, fewer than 10 researchers have explored this area, as delivering CRISPR-Cas9 machinery into bacteria within mature biofilms poses considerable difficulty. Meanwhile, CRISPR-based biosensors now allow researchers to watch gene expression unfold in real time within live biofilms, uncovering new insights into their hidden dynamics (Figure 4b) [138,139].
In parallel, synthetic ecology is introducing programmable microbial communities that can self-regulate, compete, or cooperate to dismantle biofilms from within. Designer E. coli strains have been engineered to secrete enzymes like dispersin B, which actively degrade S. aureus biofilms and prevent their return [140,141]. Other microbes have been modified to act as quorum quenchers, intercepting and degrading the chemical signals bacteria use to organize into structured communities [10,142]. These living interventions are particularly promising in chronic wounds, catheter infections, and even industrial settings where standard disinfectants fail to reach embedded bacteria. Probiotic engineering is also breaking new ground. Scientists are equipping Lactobacillus strains with genes that produce antimicrobial peptides and biofilm-disrupting enzymes. These customized probiotics have shown strong results against P. aeruginosa in cystic fibrosis airway models often outperforming conventional antibiotics [143]. Even more futuristic are “smart” probiotics that only activate under specific conditions, like low pH or inflammation, making them both targeted and safe [144]. Taken together, synthetic biology offers a powerful arsenal of precision, adaptive, and microbiome-sparing tools to combat biofilms. From gene-editing platforms to engineered living therapies, these innovations are shifting the paradigm in both clinical treatment and industrial biofouling control [145,146].

4.2. Nanotechnology Approaches for Biofilm Control

Nanotechnology is unlocking a new frontier in biofilm treatment, one where precision, responsiveness, and control replace the brute force of traditional antibiotics [134,147]. Engineered nanoparticles (NPs), tailored at the nanoscale, are now being designed to penetrate biofilm barriers, deliver drugs with pinpoint accuracy, and even adapt to the infection microenvironment. These platforms offer a versatile solution to the long-standing challenge of biofilm resistance (Figure 4a) [148,149,150].
Among the most effective innovations are metal–organic framework (MOF) nanoparticles with high surface area, tunable porosity, and customizable surface charge. MOFs have been shown to deliver antibiotics deep into biofilms, achieving penetration rates up to 100 times greater than free drugs [151,152]. Similarly, lipid-polymer hybrid nanoparticles encapsulating tobramycin have drastically reduced the MBEC (minimum biofilm eradication concentration) in P. aeruginosa biofilms, outperforming standard treatments [153,154]. Even more sophisticated are enzyme-functionalized nanoparticles such as DNase I-coated gold NPs that actively degrade extracellular DNA within the matrix, restoring antibiotic efficacy [155].
What sets nanotechnology apart is the emergence of “smart” nanoparticles systems that respond to the unique biochemical cues of the biofilm environment. For example, pH-sensitive polymeric nanoparticles release vancomycin selectively in acidic zones, reaching drug concentrations up to 50-fold higher than systemic delivery [156]. Others are designed to react to quorum-sensing molecules, activating only in the presence of mature biofilms. A particularly exciting innovation is the use of Janus nanoparticles that propel themselves using hydrogen peroxide produced by the biofilm itself, enabling directional delivery to deep matrix zones [80,157,158].
Beyond delivery, nanotechnology is fueling synergistic combinations that break through resistance mechanisms. Chitosan silver nanoparticles disrupt bacterial membranes, enhancing fluoroquinolone uptake [159,160], while graphene oxide particles inhibit efflux pumps, restoring ciprofloxacin effectiveness in resistant E. coli strains [161]. A novel strategy combining cerium oxide nanoparticles with β-lactam antibiotics induces redox-mediated stress responses, triggering cell death in K. pneumoniae biofilms through non-classical mechanisms [162]. These advances mark a fundamental shift from passive diffusion therapies to dynamic, programmable nanoplatforms that interact directly with biofilm architecture and behavior. With their ability to adapt, self-navigate, and act selectively, nanoparticles are redefining the future of biofilm control particularly in hard-to-treat infections such as chronic wounds, implant-associated biofilms, and drug-resistant lung infections [163]. A comprehensive overview of the therapeutic targeting of biofilm forming bacteria is summarized in Table 2.
Table 2. Therapeutic nanomaterials targeting biofilm-forming bacteria.
Table 2. Therapeutic nanomaterials targeting biofilm-forming bacteria.
NanomaterialPathogen NameMechanismMaterial BaseActive Concentration AppliedMBICReference
Chitosan-based nanogelsS. aureus,
K. pneumoniae,
P. aeruginosa
Chitosan’s cationic nature allows it to interact with negatively charged bacterial cell membranes, leading to increased permeability and disruption of biofilm formation. Additionally, chitosan can be functionalized with antimicrobial agents to enhance its efficacy.Chitosan25 μg/mL200 μg/mL[164]
Hyaluronic acid-based nanogelsMethicillin-resistant S. aureus (MRSA),
P. aeruginosa
These nanogels can deliver nitric oxide (NO) and antimicrobial peptides (AMPs) directly into biofilms. NO disrupts biofilm structure, while AMPs kill the bacteria, resulting in synergistic antibiofilm activity.Hyaluronic acid128 mg/mL512 mg/mL[165]
Alginate-based nanogelsS. aureus, E. coliAlginate nanogels can be loaded with agents like tannic acid and iron ions to induce photothermal therapy (PTT) and chemodynamic therapy (CDT), generating reactive oxygen species (ROS) that disrupt biofilms.Alginate0.625 mg/L25 μg/mL[166]
Polymeric nanoparticlesS. aureus,
P. aeruginosa,
E. coli
These nanoparticles can encapsulate antibiotics, enhancing their stability and penetration into biofilms. They can also be designed to release drugs in response to specific stimuli within the biofilm environment.Caged guanidine groupsAqueous dispersions of CGNs, GNs, and ICGNs (0.1 mg mL−1, 0.2 mL)0.4 mg mL−1, 0.2 mL[167]
LiposomesS. aureus, P. aeruginosaLiposomes can fuse with bacterial membranes, facilitating the delivery of encapsulated antibiotics directly into the biofilm matrix, thereby increasing local drug concentration and efficacy.PhospholipidsNo lipid concentrations givenBiofilm inhibition inferred from relative viability assays[168]
DendrimersS. aureus,
P. aeruginosa
Dendrimers possess a highly branched structure with functional groups that can be tailored for antimicrobial activity. They can disrupt bacterial membranes and inhibit biofilm formation.Quaternary ammonium-functionalized, metal ion complexed)No quantitative biofilm inhibition thresholds providedNo quantitative biofilm inhibition thresholds provided[169]
Peptide-based nanoparticlesS. aureus,
P. aeruginosa
These nanoparticles utilize antimicrobial peptides that can insert into bacterial membranes, causing disruption and cell death, effectively preventing biofilm development.Peptide dendrimer (Trp/Arg)20 μM, 40 μM, 80 μM16–32 mg/L[170]
Gold nanoparticles (Au NPs)Methicillin- Resistant S. aureus.Photothermal activity of Au NPs induce the antimicrobial activity against drug-resistant bacteria species.Gold nanoparticles (AuNPs)109.5 μg/mL165 μg/mL
Silver nanoparticles
(Ag NPs)
P. aeruginosaAg NPs generate ROS inside the cell membrane which led to cell wall rupture and cell death.Silver nanoparticles (various types)1–200 µg/mL against P. aeruginosaNot reported as a distinct value[171]
Silver nanoparticles
(Ag NPs)
P. aeruginosa, E. coli, K. pneumonae and S. aureusSmall size and spherical shape of silver nanoparticles responsible for antibacterial activity.Silver nanoparticles (AgNPs)0.3 mg1.0 mg[172]
Green synthesized Ag NPs.Ampicillin-resistant K. pneumoniae, E. coliGreen Ag NPs interrupt the metabolic activities of bacteria by interacting with enzymes.Iron oxide nanoparticles (Fe3O4 NPs)64 μg/mL64 μg/mL[173]
Methionine capped-Au NPsGram-negative-
A. baumannii and
S. enterica Gram-positive- Methicillin- Resistant S. aureus and M. luteus.
Photothermal activity of Au NPs induce the antimicrobial activity against drug-resistant bacteria species.Ultra-small gold nanoparticles (Au0/Au+)30 mg L70 mg L−1[174]
Gold nanorodsMethicillin- Resistant S. aureusThe presence of single-valent gold on the surface of nanoparticles induced the antimicrobial activity against both Gram-positive and gram- negative drug-resistant bacteria.Gold nanorods (AuNRs)2× MIC8 μg mL−1[175]
Metallosurfactant-based cobalt oxide/hydroxide nanoparticles.S. aureusPresence of metallosurfactants enhanced the antibacterial activity of Co NPs by rupturing the cell wall of bacteria.Chitosan-based nanoparticles (CSNPs)10 μg mL−110 μg mL−1[176]
Polyhydroxybutyrate-Co3O4 bio nanocomposites.E. coli, S. aureusBiosynthesized Co NPs have high thermal stability and improved structural properties which enhanced the bactericidal properties of Co NPs.Zinc oxide nanoparticles (ZnO NPs)50 μg/mL50 μg/mL[177]
Silver nanoparticles (AgNPs) synthesized using chemical reduction methodA. baumanniiFe3O4 NPs developed antibacterial activity by interacting with the ATP associated mechanisms.Silver nanoparticles (AgNPs)0.5 mg/mL0.5 mg/mL[178]
Copper oxide nanoparticles (CuO NPs) synthesized using Azadirachta indica (neem) leaf extractS. aureusMagnetic nanoparticles generate artificial magnetic field inside the biofilm and enhance the drug penetration and antimicrobial activity inside bacterial cell.Copper oxide nanoparticles (CuO NPs)32 μg/mL32 μg/mL[179]
Aluminum oxide nanoparticles
(Al2O3 NPs)
A. baumanniiAl2O3 NPs inhibit the EPS and reduce the formation of biofilm.Al2O3 NPs120 µg mL−1120 µg mL−1[180]
AgNPs_mPEG, AgNPs_mPEG_AK, and AKP. aeruginosaThe antimicrobial activity of AgNPs relies on their ability to release Ag1 to the bacteria, as determined by the corrosion rate, which depends on the morphology (size and shape) of the NPs. Accordingly, it has also been reported that smaller AgNPs have more powerful bactericidal activity as the area of surface contact of AgNPs with microorganisms is increased in relation to larger AgNPs.Selenium nanoparticles (SeNPs)50 μg/mL50 μg/mL[181]
Liquid crystal nanoparticles (LCNPs)P. aeruginosa
S. aureus
The nanostructure enables the antibiotic to be released in a controlled and sustained manner, ensuring therapeutic concentrations for extended periods of time. By delivering the antibiotic directly into the biofilm, LCNPs raise the local concentration of the medication at the site of infection, improving its bactericidal activity.Liquid crystal nanoparticles (LCNPs)2 μg/mL2 μg/mL[182]
Nitric oxide (NO) releasing nanocomposite
Activation: near-infrared (NIR) light irradiation
P. gingivalis
F. nucleatum
S. mutans
Photodynamic therapy (PDT): When exposed to near-infrared light, ICG produces reactive oxygen species (ROS), which destroy bacteria. Photothermal therapy (PTT): NIR irradiation causes localized heating, breaking biofilm structure and hastening bacterial elimination.
Nitric oxide release: Heat from PTT causes the release of NO from SNO, which has antibacterial characteristics and aids in biofilm breakdown.
Bismuth sulfide (Bi2S3) nanoparticles and bis-N-nitroso compounds (BNN)128 μg/mL128 μg/mL[183]
Silver nanoparticles (AgNPs)S. aureus
P. aeruginosa
Disrupt bacterial membranes. Generate reactive oxygen species (ROS). Interfere with DNA replication
Penetrate biofilms due to their small size
silver nitrate (AgNO3)0.25 μg/mL to 2.0 μg/mL0.25 μg/mL to 2.0 μg/mL[184]
Zinc oxide nanoparticles (ZnO NPs)S. aureus, E. coliROS generation. Disruption of membrane integrity. Photocatalytic activity (especially under UV light)Zinc oxide nanoparticles50 µg/mL50 µg/mL[185]
Chitosan nanoparticlesS. aureus,
P. aeruginosa
Electrostatic interaction with negatively charged bacterial membranes. Disruption of cell wall and membrane. Interference with nutrient transport and metabolism.Chitosan–silver nanocomposite (CS–AgNPs)64 μg/mL64 μg/mL[186]
Polymeric nanoparticles (e.g., PLGA-based)P. aeruginosa,
S. aureus
Sustained drug release. Targeted delivery to biofilm. Potential surface modification for improved interaction with biofilm.PLGA (polylactic-co-glycolic acid).10–12.5 mg/mL102.5 mg/mL[187]
Chitosan nanoparticles (CS NPs)C. albicansChitosan and bCTL are positively charged, allowing for strong electrostatic interaction with the negatively charged fungal cell walls and biofilm matrix. Nanoparticles penetrate the extracellular matrix of the biofilm, destabilizing its structure. bCTL disrupts fungal cell membranes, increasing permeability and leading to cell death.Chitosan and the antifungal crosslinker phytic acid140 ± 2.2 µg/mL140 ± 2.2 µg/mL[188]
Copper nanoparticles (CuNPs)E. coli, S. aureus, Membrane damage.
Oxidative stress via ROS.
Metal ion toxicity
Green synthesized copper nanoparticles (CuNPs)30 µg/mLMIC—89–91% inhibition[189]
Titanium dioxide nanoparticles (TiO2 NPs)Broad-spectrum (Gram-positive and Gram-negative bacteria)Photocatalytic ROS generation under UV light. Oxidative damage to cellular structuresTitanium dioxide nanoparticles (TiO2 NPs)100 μg/mL100 μg/mL against P. aeruginosa[190]
Graphene oxide (GO) and reduced graphene oxide (rGO)E. coli, S. aureus, K. pneumoniaePhysical disruption (sharp edges pierce membranes).
Oxidative stress, electron-transfer interactions.
Graphene oxide silver nanocomposite (GO-Ag)16 μg/mL16 μg/mL[191]
Glycopeptide dendrimersP. aeruginosaGlycopeptide dendrimers bind exclusively to the P. aeruginosa lectin LecA via multivalent carbohydrate–lectin interactions, affecting biofilm attachment mechanisms. This binding disrupts the appropriate cell-cell and cell-surface interactions required for biofilm development. The multivalency considerably improves biofilm inhibition while also promoting the dispersal of produced biofilms without killing bacteria.Tetravalent G2 dendrimers (GalAG2 and GalBG2) and Octavalent G3 dendrimers (GalAxG3 and GalBxG3).G2—20 µM
G3—≈9–13 µM
13 µM[192]
Nitric oxide (NO)-releasing alkyl-modified poly(amidoamin) (PAMAM) dendrimersS. mutansDisrupts bacterial signaling and weakens the biofilm matrix. The hydrophobic alkyl chains enhance bacterial membrane permeability, promoting antimicrobial effects.Poly(amidoamine) (PAMAM) dendrimersNot specifiedNot reported[193]
Biodegradable nanoemulsions (benzoyl peroxide (BPO) and ciprofloxacin (CIP))S. aureus
P. aeruginosa
The nanoemulsions carry both benzoyl peroxide, which produces reactive oxygen species (ROS), and ciprofloxacin, a broad-spectrum antibiotic, straight to biofilms. The lipid-based nanoemulsion improves penetration into the biofilm matrix, resulting in sustained and localized drug release. This synergistic combination breaks bacterial membranes, lowers biofilm biomass, and efficiently kills bacteria in wound-associated biofilms.BPO nanoparticles in a lemongrass oil–based nanoemulgel20 µM20 µM[194]
Crosslinked nanoemulsionsS. aureus
P. aeruginosa
The crosslinked nanoemulsions penetrate biofilms and deliver antibacterial essential oils in a regulated manner. Their crosslinked structure promotes wound stability and retention, hence increasing antibacterial activity. This disrupts bacterial membranes, reduces biofilm biomass, and accelerates wound healing without the use of synthetic medications.Flavonoids, tannins31–125 µg/mL31–250 µg/mL[195]
ZIF-8 (Zn-based MOF)E. coli, S. aureus, MRSAReleases Zn2+ ions, disrupts membranes, generates ROSZinc (Zn)~10–100 µg/mL~10–100 µg/mL[196]
UiO-66 (Zr-based MOF)S. aureus, P. aeruginosaHigh-stability MOF; used for drug delivery and ROS generationUiO-66-NH2 (and UiO-66) nanoparticles loaded with cefazolinMIC measured but not numerically detailedOnly noted as improved[197]
MOF-antibiotic hybridsDrug-resistant E. coli, S. aureus, MRSAControlled antibiotic release enhances efficacy and reduces resistance developmentSynthetic antimicrobial peptide (AMP)1000 µg/mL2000 µg/mL[198]
Ag-loaded nano-metal–organic framework (Ag@nanoMOF)S. aureus
E. coli
The nanoMOF emits silver ions (Ag+), disrupting bacterial membranes and biological functions. The MOF produces reactive oxygen species (ROS), which improves antibacterial and anti-biofilm properties. This dual approach effectively inhibits and eliminates biofilms.Silver nanoparticles (chemically reduced)0.5 mg/mL0.5 mg/mL[151]
Cu-MOF (IITI-3)M. tuberculosisThe anti-mycobacterial activity of INH@IITI-3 demonstrated significant bacterial killing and altered the structural morphology of the bacteria. Copper (Cu)50–500 µg/mL50–500 µg/mL[199]
Outer-Membrane Vesicles (OMVs)P. aeruginosaOMVs carry β-lactamases (antibiotic resistance), deliver quorum-sensing signals (PQS), and neutralize AMPs.Amoxicillin (AMX)100 µg/mL100 µg/mL[200]
Outer-Membrane Vesicles (OMVs)N. meningitidisOMVs deliver lipooligosaccharides and porins; modulate immune response and mediate adhesion/invasion.Zn-MOF (carrier alone)10 µg/mL10 µg/mL[201]
Outer-Membrane Vesicles (OMVs)H. pyloriOMVs contain VacA cytotoxin, promote inflammation, and facilitate biofilm formation.Am-Zn-MOF (Amoxicillin-loaded)10 µg/mL10 µg/mL[202]
Outer-Membrane Vesicles (OMVs)B. ovatusOMVs deliver inulin-degrading enzymes, aiding nutrient sharing and mutualistic gut ecologyLPS, adhesins, OmpA10 µg (spot)10 µg [203]
Outer-Membrane Vesicles (OMVs)B. fragilisOMVs package glycosidases and proteases; aid in polysaccharide digestion and microbial cross-feeding.Polysaccharide A, OmpA-like proteins, LPS variants0.2, 1, 2 µg/mL0.2, 1, 2 µg/mL[204]
Polymeric CO-releasing micellesP. aeruginosaCO inhibits bacterial respiration and biofilm construction, increasing amikacin’s penetration and potency against bacteria.Amikacin-loaded PLGA nanoparticles5 µg/mL5 µg/mL[205]
P(PEGMA-b-DEAEMA) polymeric micellesC. albicans and C. tropicalisPromote ~70% biofilm removal and significantly reduce cell viability of both strains.Ultra-small solid lipid nanoparticles (us-SLNs)32 µg/mL>256 µg/mL[206]
P(PEGMA-b-DEAEMA) polymeric micelles co-delivered with fluconazole (Flu)C. albicansExhibit synergistic effects, leading to a 2.2-log reduction in cell viability.Poly(ε-caprolactone)-based nanofiber mats loaded with chlorhexidine4 µg/mL8 µg/mL[206]
Gentamicin-conjugated magnetic nanoparticles (MNPs-G)S. aureusUnder optimized magnetic field exposure, MNPs-G achieve homogenous distribution over the biofilm, resulting in increased bacterial death.Silica–gentamicin nanohybrids~6.26 µg/mL~6.26 µg/mL[207]
Gentamicin-conjugated magnetic nanoparticles (MNPs-G)E. coli and other ESKAPE pathogensMNPs-G exhibit bactericidal activity comparable to free gentamicin in solution, effectively killing planktonic cells of various ESKAPE pathogens.Gentamicin-loaded PLGA nanoparticles5 µg/mL10 µg/mL[208]
Magnetic iron oxide nanoparticles (MIONPs)S. aureusMagnetically propagated MIONPs create artificial pathways within the biofilm matrix, allowing gentamicin to penetrate more deeply. Iron oxide nanoparticles64 µg/mL64 µg/mL[209]
TiO2 films (Sol–gel method)D. geothermalisExposure to 360 nm UV light, TiO2’s photocatalytic activity produces reactive oxygen species (ROS), resulting in considerable biofilm reduction (>107 to <106 cells/cm2).Titanium dioxide nanoparticles100 µg/mL100 µg/mL[210]
TiO2 films (ALD method)D. geothermalisALD-prepared TiO2 films generate ROS under UV light, disrupting adhesion structures and reducing biofilm density.Titanium dioxide (TiO2) photocatalytic coatingsUV irradiation of the TiO2: 360 nm light, 20 Wh m−2not applicable[211]
Sulfur-doped TiO2 filmsD. geothermalisDoping TiO2 with sulfur did not enhance the biofilm-destroying capacity under UV light exposure.Photocatalytic TiO2 coatings, prepared via sol–gel and atomic layer deposition (ALD)UV light exposure-20 W·h m−2 of 360 nm radiationNot applicable[212]
H2S-releasing polymeric nanoparticlesSulfate-reducing bacteria (e.g., Desulfovibrio)Controlled release of H2S modulates bacterial growth by altering redox balance; can inhibit overgrowth of SRB, impacting gut microbial homeostasis.hydrogen sulfide (H2S)Not reportedNot reported[213]
Metal sulfide nanoparticles (FeS, MoS2)F. nucleatumMay release H2S or interact with sulfur metabolism, influencing bacterial survival and virulence factors related to gut dysbiosis and colorectal cancer risk.Multilayered magnetic nanoparticles (ML-MNPs) composed of an iron oxide core (Fe3O4), followed by inner silver (Ag) and ultrasmall MoS2Not reportedNot reported[214]
Sulfur-doped TiO2 nanoparticlesA. baumanniiEnhanced photocatalytic activity produces reactive sulfur species affecting biofilm integrity and bacterial survival through oxidative stress mechanisms.Green-synthesized anatase TiO2 nanoparticles (~47 nm)~7.81 µg/mL~7.81 µg/mL[215]
AgPd0.38 nanocagesE. coli, S. aureus,
P. aeruginosa
Generates surface-bound ROS via oxidase-like activity, leading to bacterial cell wall disruption and death.AgPd0.38 62.5 µg/mL62.5 µg/mL[216]
Mesoporous silica nanoparticles (nMS) loaded with nano-silver (nAg) and chlorhexidine (Chx)S. mutans (primary cariogenic bacterium) and other oral biofilm bacteriapH-responsive release of nano-silver and chlorhexidine in acidic biofilm microenvironments; suppresses acid production and biofilm formation.nMS-nAg-Chx18.75 µg/mL18.75 µg/mL[217]
Micro-nano hybrid multifunctional motor (MnO2-based)S. aureus, P. aeruginosa, E. coliSelf-propulsion via oxygen microbubbles (H2O2 + MnO2 catalyst) enables deep biofilm penetration; generates hydroxyl radicals (OH) that disrupt biofilm matrix and kill bacteria.Manganese dioxide (MnO2)1, 2, and 4 mg/mL2, 4, and 8 mg/mL[218]

4.3. Quorum-Sensing Inhibitors and Anti-Virulence Strategies: Disrupting Bacterial Communication

One of the most promising approaches in biofilm control involves disarming bacteria instead of killing them [10]. Quorum sensing (QS), the system bacteria use to communicate and coordinate biofilm formation, is now a prime target for precision anti-virulence therapy. By interfering with this communication, quorum-sensing inhibitors (QSIs) can suppress pathogenic behaviors without applying the selective pressure that often leads to resistance [219,220]. In Gram-negative pathogens such as P. aeruginosa, small chemicals known as N-acyl homoserine lactones (AHLs) modulate gene expression related to virulence and biofilm formation. LuxR-type receptor antagonists have been demonstrated to inhibit AHL binding and diminish biofilm biomass by over 70% in chronic wound models [10,221]. In Gram-positive bacteria like S. aureus, RNAIII-decreasing peptides (RIPs) have interfered with the Agr system, inhibiting toxin production without eliminating the bacteria, hence maintaining the healthy microbiome while diminishing pathogenicity. Artificial intelligence is currently expediting the identification of new quorum-sensing inhibitors. Researchers have been using Generative Adversarial Networks (GANs) and Reinforcement Learning to create synthetic analogs that exhibit enhanced potency and stability relative to natural QS molecules [222,223]. AI-optimized furanone compounds block the P. aeruginosa LasR receptor with an effectiveness tenfold greater. Multi-target inhibitors are being developed to simultaneously inhibit the Las transcriptional regulator, which initiates the quorum sensing (QS) cascade; Rhl, which controls secondary QS signals; and PqsR, which regulates PQSs that influence virulence and biofilm matrix formation. Consequently, this approach aims to prevent compensatory mechanisms that bacteria might employ to circumvent quorum-sensing disruption [10,224]. Besides pharmacological inhibitors, quorum quenching agents provide a biological remedy by reducing or blocking quorum-sensing signals. Natural chemicals such as halogenated furanones derived from marine algae and ajoene sourced from garlic disrupt the synthesis or stability of quorum-sensing molecules. Engineered enzymes, including AHL lactonases and acylases, enzymatically destroy quorum-sensing molecules before their attainment of quorum thresholds [225,226]. An alternative new strategy utilizes synthetic QS-mimicking nanoparticles that function as decoys, binding to bacterial receptors and obstructing authentic signals from activating virulence programs. These techniques signify a paradigm shift in antimicrobial therapy from indiscriminate bacterial eradication to specific virulence mitigation. By focusing on bacterial activity instead of viability, QS inhibitors can compromise biofilm integrity, diminish resistance pressure, and maintain beneficial microbiota (Figure 4b). To date, approximately 724 papers have been published on QS inhibition. With numerous medicines progressing to clinical trials, particularly for respiratory and urinary tract infections, the future of biofilm management depends on inhibiting bacterial communication rather than employing chemical warfare [227]. Therapeutic anti-quorum-sensing materials for treating biofilm-forming bacteria are summarized in Table 3.
Table 3. Anti-quorum-sensing materials to treat biofilm-producing bacteria.
Table 3. Anti-quorum-sensing materials to treat biofilm-producing bacteria.
Inhibitor (Quorum Sensing)Target MechanismBacteriaVirulence SuppressionSourceActive Inhibitory ConcentrationReference
CRISPR-Cas9Anti-QS
Anti-biofilm
E. coli SE15Reduced biofilm formation.
Downregulation of mqsR, pgaB, pgaC, csgE, and csgF
CRISPR–Cas9 genome editing (donor DNA for homologous recombination; generated ΔluxS mutants from clinical E. coli SE15).none reported[228]
CRISPR interferenceAnti-QS Anti-biofilmE. coli AK-117Reduced biofilm formationCRISPRi (dCas9 + sgRNAs targeting luxS in E. coli AK-117)not applicable for CRISPRi studies[229]
ZingeroneAnti-QS Anti-biofilmP. aeruginosa PAO1 P. aeruginosa clinical isolates.Reduced biofilm, pyocyanin, hemolysin, elastase, proteases, rhamno lipid production. Reduced swarming, swimming, and twitching motilityZingerone (phytochemical from ginger)Not provided in this study[230]
ZeaxanthinAnti-QS Anti-biofilmP. aeruginosa PAO1Reduced biofilm formation. Downregulated rhlA and lasB expressionPurified from the green alga Chlorella ellipsoidea53.5 µM[231]
Solonamides analogsAnti-QS Anti-toxinS. aureusReduced RNAIII and hla expression. Marginally enhanced biofilm formationCyclodepsipeptide isolated from the marine bacterium Photobacterium halotolerans5 µg/mL[232]
FlavonoidsAnti-QSP. aeruginosa PA14Reduced pyocyanin production and swarming motility.
rhlA transcription inhibition
Naturally produced plant metabolitesQuercetin (100 µM): LasR activity ≈ 20–25% of control.[233]
AIP analogsAnti-QS
Anti-biofilm
S. epidermidis RP62AReduced biofilm formation (using non-native agonist of AgrC-type I)Analogs were created via solid-phase peptide synthesisAIP-I (native)-99 nM
AIP-I D1AS6A-18 nM
[234]
CoumarinAnti-QS Anti-biofilmP. aeruginosa PAO1 and clinical isolates1. Reduced biofilm production, 2. Down-regulation of lasI, rhlI, rhlR, pqsB, pqsC, pqsH, ambBCDE, 3. Reduced protease and pyocyanin production
4. Reduced expression of T3SS secretion system-associated genes
Coumarin–hydroxamic acid conjugate3.6 µM[235]
Fluoro-substituted IsothiocyanatesAnti-QSP. aeruginosa1. Reduced pyocyanin production. 2. Reduced swarming motility. 3. Attenuated in vivo virulence of P. aeruginosa PAO1-UW toward C.elegans;
4. Attenuated P. aeruginosa P14 virulence in an ex vivo human skin burn wound model
Synthetic isothiocyanate- and maleimide-containing HSL-like small molecules3.6 ± 1.9 µM[236]
Pyridoxal lactohydrazoneAnti-QSP. aeruginosa PAO1Reduced swarming and twitching motilityCondensation of pyridoxal (vitamin B6) with lactic acid hydrazideSub-MIC concentrations (8 µg/mL and 32 µg/mL)[237]
Glyceryl trinitrateAnti-QS Anti-biofilmP. aeruginosa PAO1 and clinical isolatesReduced biofilm, pyocyanin and proteases productionAntivirulence compound0.25 mg/mL (¼ MIC; sub-inhibitory)[238]
TerreinAnti-QS Anti-biofilmP. aeruginosa PAO11. Reduced elastase, pyocyanin, rhamnolipid, and biofilm production; 2. Attenuated in vivo virulence of P. aeruginosa PAO1 toward C. elegans and miceSecondary metabolite isolated from the fungus Aspergillus terreusNo half-maximal inhibitory concentration calculated[239]
1,5-dihydropyrrol-2-ones analogsAnti-QSE. coli JB357 gfp reporter strainQS inhibitionThioether-linked dihydropyrrol-2-one analogs32 µM[240]
ParthenolideAnti-QS Anti-biofilmP. aeruginosa PAO1Reduced pyocyanin, proteases, and biofilm productionNatural phytochemicalNo[241]
DiketopiperazineAnti-QS Anti-biofilmB. cenocepacia1. Reduced biofilm formation. 2. Reduced protease and siderophore productionSynthetic diketopiperazine analogs7.2 ± 0.2 µM[242]
LysionotinAnti-toxin Anti-QSS. aureus1. Downregulate fhla and gr expression. 2. Reducedα-hemolysin productionNatural flavonoidNo[243]
Methyl Eugenol-AInterferes with AHL-regulated functionsC. violaceumInhibits EPS production, biofilm formation, and flagellar movementNatural phenolic found in clove oil0.2 mg/mL[244]
Curcumin/10-undecenoic acidInhibit LuxS/AI-2 and LasI/LasR QS systemsP. aeruginosa,
B. subtilis
Counteracts bacterial pathogenicity and virulenceNatural polyphenolic compoundMIC values: 62.5 μg/mL[245]
AjoeneDownregulates QS genesP. aeruginosaInhibits virulence factors and biofilm formationBioassay-guided fractionation of garlic oil15 µM[246]
Tanreqing (TRQ) formulaInhibits upstream QS regulators (e.g., GacS/GacA, PprA/PprB)P. aeruginosaInhibits virulence factorsLab-prepared extract combining all five herbs (Huang Qin, Jin Yin Hua, Lian Qiao, Xiong Dan, Shan Yang Jiao)Results are shown across TRQ dilutions (1/4, 1/8, 1/16 TRQ), not as IC50 values[247]
PaecilomyconeInhibits PQS and HHQ synthesis in PQS systemP. aeruginosaInhibits virulence factorsnatural compound originally isolated from a fungal source-specifically Paecilomyces species96.5 µM[248]
Synthetic quorum-sensing inhibitorsMimicking or blocking natural quorum-sensing signals to disrupt communication pathways.Ralstonia solanacearum species complexImpaired coordination of virulence factor expression, leading to diminished disease symptoms in host plants.N-sulfonyl homoserine lactone analogs1.66-4.91 µM[249]
RalfuranonesInterference with quorum-sensing signaling molecules, affecting the regulation of virulence factors.Ralstonia solanacearum species complexDecreased expression of virulence genes, resulting in reduced pathogenicity.Derived from the 3-hydroxymethyl-2-methyl-4(1H)-quinolone23.7 µM[250]
DSF analogsCompete with natural DSF molecules, disrupting normal QS signaling.S. maltophilia,
P. aeruginosa
Altered biofilm dynamics and reduced antibiotic resistance, diminishing virulence.Natural DSF comes from Xanthomonas, Burkholderia, Stenotrophomonas, etc. via biosynthesis.0.5 µM[222]
Enzymatic degradation (e.g., DSF hydrolases)Degrade DSF molecules, lowering their concentration and interfering with QS.E. carotovora
P. aeruginosa
Broad-spectrum reduction in QS-mediated virulence traits, including toxin production and biofilm formation.Stenotrophomonas maltophiliaNot reported[251]
Essential oils (e.g., clove, cinnamon, oregano)Disrupt AHL-mediated signaling pathways, inhibiting signal synthesis and reception.P. fluorescens,
S. putrefaciens
Reduction in biofilm formation and extracellular enzyme production, leading to decreased spoilage activity.Eugenol, β-caryophylleneMIC: 0.2 mg/mL[252]
Phenolic compounds (e.g., vanillin, eugenol)Interfere with QS signal molecules and inhibit biofilm formation.A. hydrophila,
L. monocytogenes
Suppression of biofilm development and virulence factor expression, enhancing food safety.Vanilla bean
Clove, cinnamon, basil
MIC: 250 µg/mL
MIC: 700 mg/L
[253]
Plant extracts (e.g., garlic, ginger)Contain compounds that mimic or degrade QS signals, disrupting communication.E. coli,
S. enterica
Inhibition of QS-regulated behaviors, including toxin production and motility, reducing pathogenicity.Allicin, ajoene and
S-allyl cysteine
MIC: 0.325-0.625 mg/mL[254]
Lactic acid bacteria metabolitesProduce bacteriocins and acids that interfere with QS systems.S. thermophiles,
L. bulgaricus
Decrease in QS activity, leading to reduced biofilm formation and spoilage potential. Lactic acid, formic acid, folic acid, pyruvic acid, glutathioneNot reported[255]
AHL analogsCompete with natural AHLs for receptor binding, disrupting QS signaling.P. aeruginosa,
E. coli
Inhibition of biofilm formation and suppression of virulence gene expression. Natural phenolic compound100 µM[10]
FuranonesInterfere with QS signal reception and stability.Vibrio spp.,
P. aeruginosa
Reduction in biofilm development and attenuation of virulence factors.Synthetic brominated furanone derivative50 µM[256]
Plant-derived compounds (e.g., garlic extract, cinnamaldehyde)Inhibit QS signal synthesis and reception pathways.E. coli,
S. enterica
Decreased biofilm formation and prevention of VBNC state induction.Allium sativum0.325–0.625 mg/mL[257]
Enzymatic degraders (e.g., AHL lactonases, acylases)Degrade QS signaling molecules, disrupting communication.E. coli,
S. enterica
Impaired biofilm maturation and reduced virulence expression.AHL lactonase (AiiA)
Bacillus sps
Not reported[258]
AdenosineModulates QS pathways, potentially interfering with signal molecule synthesis or reception.CylosporaMay reduce expression of virulence factors and biofilm formation, enhancing host immune response.Endogenous nucleoside; found in all living cellsNot reported[259]
AI-2, DSF family signalsBiofilm formation, motility, and antibiotic resistanceS. aureus, E. coliDecrease in QS-regulated behaviors, including toxin production and biofilm formation.Universally produced by many Gram-negative and Gram-positive bacteriaNot applicable[80]
Probiotic-produced AutoinducersInterfere with pathogenic QS signals by competitive binding or signal degradation, disrupting bacterial communication.E. coli, S. enterica, C. difficileReduced biofilm formation and downregulation of virulence gene expression.Lactobacillus, Bifidobacterium, E. coli Nissle 1917Not reported[260]
Probiotic biofilm formationColonizes intestinal mucosa to outcompete pathogens for adhesion, indirectly suppressing QS-mediated virulence.E. faecalis, S. aureusLimits pathogen colonization and strengthens mucosal barrier functionEnterococcus faecalis, Lactobacillus spp., Bifidobacterium spp.Not reported[261]
Plant-derived QS inhibitors (e.g., flavonoids, phenolics)Block QS signal synthesis or reception, degrade signaling molecules.P. aeruginosa, Vibrio spp.Inhibits biofilm development and virulence factor production.Various plant sources50 µM[262]
Immune modulation by probioticsStimulates host immune responses, producing cytokines that counteract infection and inflammation.S. enterica,
C. difficile
Supports intestinal barrier integrity and suppresses pathogen-induced inflammation.Citrus fruits, green tea, various plants50–100 µM[263]
Synthetic QS inhibitorsEngineered molecules or circuits designed to interfere with QS signaling pathways.P. aeruginosaDisruption of QS-mediated behaviors such as biofilm formation, virulence factor production, and antibiotic resistance.Engineered small molecules or genetic circuits designed via synthetic biology10–100 µM for synthetic analogs[264]
Probiotic interferenceProbiotic strains engineered to produce QS inhibitors or compete with pathogens for QS signals.E. coli, S. enterica,
C. difficile
Reduction in pathogen colonization and virulence through competitive inhibition and signal interference.Engineered or natural probiotic strains (Lactobacillus, Bifidobacterium, E. coli Nissle 1917)Rarely specified[265]
Phage-mediated QS disruptionBacteriophages engineered to degrade QS signals or produce QS inhibitors.P. aeruginosaDecreased biofilm formation and virulence factor expression through targeted QS disruption.Engineered bacteriophages designed to degrade QS signals or produce QS inhibitorsNot reported[266]
Sulfonamide-based DSF analogsInterfere with DSF receptor binding, disrupting DSF-mediated QS.S. maltophilia,
B. cepacia
Reduced biofilm formation and virulence factor production.Chemically synthesized small molecules mimicking DSF structure5–50 µM[267]
Retinoic acid (RA)Modulates DSF-mediated QS, affecting biofilm formation and motility.S. maltophiliaDecreased biofilm formation and motility, enhancing antimicrobial efficacy.Naturally occurring metabolite of Vitamin A50 µM[268]
Endogenous DSF analogMimics DSF, disrupting DSF-mediated QS.S. maltophiliaInhibition of biofilm formation and motility.Naturally produced by bacteria or chemically synthesized to mimic DSFNot reported[269]
Coumarin–chalcone conjugateAnti-QS
Anti-biofilm
P. eruginosaDecreased biofilm formation and virulence factor expression through targeted QS disruption.Chemically synthesized conjugate combining coumarin and chalcone scaffolds20–50 µM[270]
QteESequesters LasR, increasing the threshold for QS activation.P. aeruginosaDelayed expression of QS controlled genes, reducing early activation of virulence factors.Protein from Pseudomonas aeruginosaNot reported[271]
QscRBinds to LasR and RhlR, preventing premature activation of QS.P. aeruginosaSuppressed expression of QS controlled genes, mitigating early virulence factor production.Transcriptional regulator from Pseudomonas aeruginosa56–62 nM[221]
QslAInhibits LasR and RhlR activity, modulating QS response.P. aeruginosaReduced biofilm formation and virulence factor production.Transcriptional regulator from Pseudomonas aeruginosaNot reported[272]
TyramineQuorum-sensing inhibition via interference with the CepIR systemB. cenocepacia.Decreased biofilm formation, reduced motility, attenuated virulence in Caenorhabditis elegans model.Naturally occurring biogenic amineNot reported[273]
QStatinSpecifically targets LuxO, a central response regulator in the quorum-sensing pathway of Vibrio species. QStatin binds selectively to phosphorylated LuxO, blocking its ability to activate small regulatory RNAs (sRNAs), which are essential for downstream quorum-sensing responses.V. cholerae
V. harveyi
Disrupts quorum-sensing-controlled gene expression
Reduces virulence-related phenotypes
Selective: QStatin does not inhibit general bacterial growth or non-target bacteria significantly
Small-molecule inhibitor designed to selectively target LuxO2–5 µM[274]
FuranonesCompetitive inhibition of AHL receptor proteins (LuxR-type regulators)P. aeruginosa,
E. coli, V. spp.
Reduces biofilm formation, motility, and production of virulence factors (e.g., toxins)Synthetic halogenated furanone3–9 µM[275]
BaicaleinDownregulates quorum-sensing-regulated gene expressionP. aeruginosa,
S. aureus
Inhibits biofilm formation and reduces toxin productionFlavonoid from Scutellaria baicalensis and other plantsNot specified[276]
QuercetinInterferes with AI-2 signaling pathwaysE. coli, S. spp.Inhibits quorum-sensing gene expression, reduces swarming, and suppresses virulenceNaturally occurring flavonoid281 µM for biofilm inhibition[227]
VanillinBlocks AHL signal molecule activityC. violaceum,
P. aeruginosa
Inhibits violacein pigment production and biofilm formationNatural compound from vanilla beansIC50 = 0.81 mM; MIC = 16 mM[277]
Furanone C-30Inhibits LasR receptor in the las quorum-sensing systemP. aeruginosaReduces biofilm formation, elastase activity, and enhances tobramycin susceptibility (initially)Synthetic brominated furanoneIC50: 3–9 µM [278]
QS Inhibitor-loaded biomimetic nanoparticlesInhibits AI-2 quorum-sensing system, disrupts cell communication
+ induces ferroptosis-like bacterial death via lipid peroxidation
Methicillin-Resistant S. aureus (MRSA)Suppresses virulence gene expression, reduces biofilm formation, and promotes bacterial death in infected lung tissueNatural compounds or synthetic analogs2–5 µM[279]
L-mimosineInhibits quorum-sensing-regulatory small RNAs (sRNAs) specifically, qrr sRNAs in Vibrio speciesV. harveyi
E. coli
P. aeruginosa
Reduces bioluminescence (in Vibrio), biofilm formation, motility, and toxin productionMimosa pudica and Leucaena speciesNot well reported[280]
2-(4-(acridin 9yl amino)phenyl)isoindoline-1,3-dione derivativesInhibits the Pseudomonas Quinolone Signal (PQS) quorum-sensing systemP. aeruginosaReduces virulence-related phenotypesAcridine-based isoindoline derivativesNot well reported[281]

4.4. Phage Therapy and Bacteriophage-Derived Enzymes: Precision Tools Against Biofilms

Bacteriophage therapy, once regarded as a last option, is currently experiencing a resurgence, driven by advancements in synthetic biology and precise engineering. Virus-based therapies that selectively infect and eliminate bacteria are demonstrating exceptional efficacy in addressing biofilms, particularly those resistant to antibiotics (Figure 4c) [282,283]. Contemporary advancements have converted phages into programmable precision instruments. CRISPR-Cas-modified bacteriophages may now selectively cleave antibiotic-resistance genes within biofilm communities. This method eradicates resistant bacteria while safeguarding commensal species, presenting a significant benefit over broad-spectrum antibiotics [284]. Through the engineering of phage capsids and tail fibers, researchers have broadened host ranges and improved matrix penetration, facilitating the efficient eradication of polymicrobial biofilms [285]. The generation of lysis-deficient phages is one of the most significant advancements. In contrast to conventional phages that lyse host cells, these engineered viruses eliminate bacteria by inducing intracellular toxin production, hence preventing the discharge of inflammatory debris. In prosthetic-joint infection models, these phages attained 3 to 5 log decreases in biofilm load without inducing immune over activation [286]. This invention is particularly pertinent in delicate locations where inflammation must be minimized. In addition to phage therapy, phage-derived enzymes are becoming acknowledged as effective antibiofilm agents. These encompass polysaccharide depolymerases that particularly degrade essential constituents of the biofilm matrix, including alginate in P. aeruginosa, PNAG in S. aureus, and cellulose in E. coli [287]. Fusion enzymes, which integrate matrix-degrading activity with antimicrobial peptides, are even more potent as they concurrently dismantle structure and eliminate embedded cells [288]. These enzymes are currently being encapsulated in nanoparticles for prolonged release at difficult-to-access infection locations such as implants.
The most promising method may be phage antibiotic synergy (PAS). This method utilizes phages to disrupt biofilms and inhibit resistance mechanisms, while sub-lethal antibiotics promote phage replication by inducing stress in the bacteria. In clinical research, PAS has elevated cure rates in K. pneumoniae catheter biofilms from 31% with antibiotics alone to 78% when administered in conjunction [289]. AI-driven algorithms are currently being created to identify appropriate phage antibiotic combinations based on bacterial genetics and behaviors, facilitating tailored PAS regimens [290]. Phage treatment is fundamentally transforming the principles of antimicrobial intervention. Bacteriophages offer a physiologically selective, self-replicating, and adaptable solution to the biofilm issue, utilizing tailored lytic precision, matrix-targeting enzymes, or synergistic combinations [291]. With the increasing clinical interest, these developments are set to become essential elements of next-generation infectious disease management [292].

4.5. Host-Directed Therapies and Immune Modulation: Rewiring Defenses Against Biofilms

Conventional biofilm medicines directly target bacteria; however, a powerful alternative is emerging that enhances the human immune system to fight more effectively. Biofilms are known for forming an immunosuppressive environment that inhibits phagocytosis, modifies cytokine profiles, and shields infectious pathogens from being eliminated. Host-directed treatments (HDTs) seek to counteract immunological infectiveness [293] and rescue the body’s innate immune defense mechanisms [294]. Recent findings in immunometabolism have identified critical sites for immunological reprogramming. In the context of chronic wounds, bacteria biofilm-secreted proteins prompt macrophages to assume an M2 (anti-inflammatory) phenotype, thereby suppressing antimicrobial responses and hindering the migration of other immune cells to control the infection [295]. Nonetheless, low-dose everolimus, a mTOR pathway inhibitor, has demonstrated the capacity to revert macrophages to the pro-inflammatory M1 phenotype [296], thus enhancing their ability to produce reactive oxygen species and eradicate biofilm infections by as much as fivefold [297]. Cytokine treatments, including IL-12 in conjunction with GM-CSF, have reinstated Th1 immune responses, augmented neutrophil extracellular trap (NET) production, and significantly diminished S. aureus biofilms in implant-associated infections (Figure 4d) [298]. Apart from small compounds, an emerging array of targeted biologics and modified immune cells is broadening the potential for host regulation. Monoclonal antibody F598 inhibits P. aeruginosa’s LecA-mediated immune suppression, demonstrating the first therapeutic efficacy in ventilator-associated pneumonia. Simultaneously, nanovesicle-based decoys are being engineered to bind and neutralize immunosuppressive substances released by biofilms, thereby restoring neutrophil activity in cystic fibrosis airway models. CRISPR-edited neutrophils, which have been modified to exhibit enhanced surface markers such as CD11b, demonstrate improved motility and penetration into biofilm matrices by up to 40% [299].
The development of vaccines is also garnering interest. Researchers are developing next-generation vaccines that provoke protective, biofilm-specific immune responses by targeting matrix components and stress-induced biofilm proteins. These may be especially beneficial for high-risk individuals experiencing repeated device-related or respiratory infections [22]. Precision medicine enhances the efficacy of host-directed interventions. Patient-derived three-dimensional organoids and autologous immune cells now provide individualized ex vivo evaluation of immunomodulators [295]. Machine learning methods are utilized to simulate immune responses and inform cytokine therapy choices based on individual immunoprofiles. In a clinical investigation, ex vivo primed macrophage therapy resulted in a 75% increase in biofilm clearance in diabetic foot infections vs. usual treatment [300]. Collectively, these host-centric developments are transforming the paradigm of biofilm therapy from antibiotic eradication to immunological rescue. HDTs provide sustainable, microbiome-conserving, and highly individualized therapies by addressing the fundamental immune malfunction that enables the persistence of biofilms [147]. As these techniques advance, they are set to become essential adjuncts to direct-acting antimicrobials in the management of complex, chronic illnesses.

5. Translational Challenges and Future Directions in Biofilm Therapeutics

The transition from laboratory research to clinical use in biofilm therapies presents not only a technical obstacle but also regulatory, ethical, and societal challenges. Next-generation therapies, such as modified phages, CRISPR-based antimicrobials, and biofilm-responsive nanotechnologies, exhibit significant potential; yet their trajectory toward clinical implementation is intricate and disjointed [133]. This section delineates the principal problems and forthcoming priorities influencing the domain of biofilm medicine.

5.1. Regulatory Bottlenecks and Standardization Gaps

Existing regulatory frameworks mostly focus on traditional small-molecule antibiotics, resulting in a gap in the assessment of intricate biologics and synthetic structures. Engineered phages, CRISPR nanocarriers, and probiotic therapies frequently lack definitive approval processes owing to their multifunctional and dynamic characteristics. Although attempts like the expedited approval of CTX-2024, a CRISPR-enhanced phage treatment for cystic fibrosis biofilms, indicate advancement, these regulatory frameworks are used inconsistently across different areas and therapeutic categories [301]. There is an urgent requirement for standardized global protocols that can address the complexities of biologics targeting biofilms. This entails the delineation of biofilm-specific clinical objectives (e.g., biomass reduction, matrix degradation, recurrence suppression) and the formulation of rigorous protocols for post-market surveillance, especially for treatments exhibiting environmental persistence [302].

5.2. Ethical and Biosafety Considerations

The introduction of genetically modified organisms presents numerous biosafety and ethical challenges. Issues related to horizontal gene transfer, off-target impacts, and unforeseen ecological repercussions necessitate the incorporation of fail-safe design elements, including kill-switch circuits, auxotrophic dependencies, and non-replicative vectors [303]. The WHO’s 2024 consensus framework has advanced by excluding heritable gene modifications and requiring temporary, self-limiting structures for clinical applications. In addition to biosafety, public perception and trust are essential. Surveys reveal more endorsement for engineered probiotics in therapeutic settings compared to environmental uses, highlighting the necessity for clear scientific communication and inclusive stakeholder involvement [304]. Ethical success will rely on responsible innovation and the establishment of societal consensus via proactive discussion.

5.3. Microbiome-Preserving Strategies and Precision Design

A significant progression in next-generation therapeutics is the shift towards microbiome-sparing approaches. In contrast to broad-spectrum antibiotics, these approaches specifically target pathogenic biofilms while safeguarding commensal microbial communities. For instance, PslG depolymerase specifically targets the matrix polysaccharides of P. aeruginosa with little collateral effects [305], whereas tailored probiotics and metabolite-guided therapy have shown an over 85% decrease in pathogens without altering microbiota composition [306]. The integration of artificial intelligence, nanotechnology, and synthetic biology is facilitating enhanced precision. Examples comprise machine learning-optimized antimicrobial peptides designed for matrix penetration, and CRISPR nanocarriers that administer both gene editing and medicinal payloads with spatial precision [301].

5.4. Towards Real-Time, Autonomous Therapies

Looking forward, biofilm management may be revolutionized by autonomous, closed-loop therapeutic platforms. These systems integrate biosensors, responsive delivery mechanisms, and adaptive algorithms to continuously monitor and modulate therapy in real time. Early models show promise in eradicating biofilms through on-demand activation triggered by environmental signals [307]. However, significant technical, safety, and validation hurdles remain before clinical integration becomes feasible.

5.5. Pathways to Clinical Translation

Bridging the translational gap in biofilm therapeutics demands a unified, interdisciplinary approach. Developing standardized preclinical models that replicate human tissues, immune responses, and microbiota is critical for predicting clinical efficacy [308]. Collaboration among researchers, clinicians, regulators, and ethicists will ensure that novel interventions are both effective and ethically sound. Robust evaluation metrics must be refined to assess not only bacterial clearance but also long-term safety, resistance suppression, and tissue recovery [309]. Cross-sector alignment is essential to streamline regulatory pathways for emerging biologics. Public–private partnerships can facilitate resource-sharing and accelerate innovation. Equally, iterative clinical feedback loops must inform early-stage design. Transparency in biosafety practices will build public trust. Investment in education and policy will support broader adoption. Ultimately, success hinges on translating precision science into safe, scalable, and societally accepted biofilm solutions.

6. Conclusions

Biofilms present a major challenge in modern microbiology, tightly linked to antibiotic resistance, persistent infections, and therapeutic failure. Our thorough investigation indicates that conventional antibiotic strategies are inadequate to infiltrate the protective matrix, metabolic dormancy, and collaborative behaviors inherent in biofilm ecosystems. The integration of synthetic biology, nanomedicine, artificial intelligence, and host-directed therapeutics has initiated a novel era of precision antibiofilm methods.
We emphasize the future interventions that are driven by machine learning-based drug discovery, CRISPR-modified phages, and responsive nanoparticles, all of which can offer support in providing tailored interventions that affect biofilm architecture, communication, and metabolic control. These methods are not only more efficacious but are also engineered to safeguard the microbiome, thereby reducing off-target harm and the emergence of resistance. The integration of host-directed therapies, including cytokine modulation and modified immune cells, signifies a transformative shift towards enhancing the host immune response, thus mitigating the immunosuppressive microenvironments established by biofilms. Progress relies not just on technological advancements but also on system-level integration via standardized preclinical models, ethical biosafety protocols, and iterative clinical feedback. The creation of real-time, autonomous treatment systems with environmental sensing and adaptive response capabilities will expedite the clinical application of these sophisticated techniques. In conclusion, eliminating the biofilm stronghold necessitates a multifaceted strategy that integrates microbial precision, host resilience, and regulatory foresight. As we progress towards a post-antibiotic era, the integration of cross-disciplinary innovation presents a viable approach to sustainable, scalable, and socially accepted biofilm treatments.

Author Contributions

The manuscript was conceptualized by M.H., P.B., T.A.N. and A.S.; methodology, S.C.G. and C.U.; resources, B.R.P., E.D. and P.S.; writing-original draft preparation, M.H.; writing-review and editing, K.E., V.R., A.J.-V., K.-A.A.L., N.B. and F.M.D.O.; supervision P.B.; All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

The dataset supporting the findings of this study is included within the manuscript and its referenced sources, ensuring comprehensive access to the relevant data for further examination and analysis.

Conflicts of Interest

Authors Manasi Haval, Arun Shettar and Pramod Bhasme were employed by the company Cytxon Biosolutions Pvt. Ltd. The remaining authors declare that the research was conducted in the absence of any commercial or financial relationships that could be construed as a potential conflict of interest.

Abbreviations

The following abbreviations are used in this manuscript:
AFMAtomic Force Microscopy
AHLsN-acyl homoserine lactones
BLIBioluminescence Imaging
CFcystic fibrosis
CFUColony-forming unit
CLSMConfocal Laser Scanning Microscopy
CNNsConvolutional Neural Networks
Cryo-EMCryo-Electron Microscopy
DFUDiabetic foot ulcers
DHMDigital Holographic Microscopy
EPSExtracellular Polymeric Substance
GANsGenerative Adversarial Networks
GNNsGraph Neural Networks
HDTsHost-directed therapies
LLSMLattice Light-Sheet Microscopy
LSTMLong Short-Term Memory
MBECMinimum biofilm eradication concentration
MOFsMetal–organic frameworks
NETNeutrophil extracellular trap
NPsEngineered nanoparticles
OCTOptical Coherence Tomography
PASPhage antibiotic synergy
PINNsPhysics-Informed Neural Networks
QSQuorum sensing
QSIsQuorum-sensing inhibitors
RFRandom Forests
RIPsRNAIII-inhibiting peptides
RLReinforcement Learning
RSIRaman Spectroscopy Imaging
SEMScanning Electron Microscopy
STEDStimulated Emission Depletion Microscopy
STORMStochastic Optical Reconstruction Microscopy
SVMsSupport Vector Machines
TEMTransmission Electron Microscopy
A. baumanniiAcinetobacter baumannii
A. hydrophilaAeromonas hydrophila
B. cenocepaciaBurkholderia cenocepacia
B. cepacianBurkholderia cepacian
B. fragilisBacteroides fragilis
B. ovatusBacteroides ovatus
C. albicansCandida albicans
C. difficileClostridium difficile
C. glabrataCandida glabrata
C. violaceumChromobacterium violaceum
E. carotovoraErwinia carotovora
E. coliEscherichia coli
F. nucleatumFusobacterium nucleatum
H. pyloriHelicobacter pylori
K. pneumoniaeKlebsiella pneumoniae
L. monocytogenesListeria monocytogenes
M. luteusMicrococcus luteus
M. tuberculosisMycobacterium tuberculosis
N. meningitidisNeisseria meningitidis
P. aeruginosaPseudomonas aeruginosa
P. fluorescensPseudomonas fluorescens
P. gingivalisPorphyromonas gingivalis
S. aureusStaphylococcus aureus
S. entericaSalmonella enterica
S. fonticolaSerratia fonticola
S. maltophiliaStenotrophomonas maltophilia
S. mutanStreptococcus mutans
S. putrefaciensShewanella putrefaciens
S. suisStreptococcus suis
V. choleraeVibrio cholerae
V. harveyiVibrio harveyi
X. campestrisXanthomonas campestris

References

  1. Alam, K.; Farraj, D.A.A.; Mah-e-Fatima, S.; Yameen, M.A.; Elshikh, M.S.; Alkufeidy, R.M.; Mustafa, A.E.-Z.M.A.; Bhasme, P.; Alshammari, M.K.; Alkubaisi, N.A.; et al. Anti-biofilm activity of plant derived extracts against infectious pathogen-Pseudomonas aeruginosa PAO1. J. Infect. Public Health 2020, 13, 1734–1741. [Google Scholar] [CrossRef] [PubMed]
  2. Li, P.; Yin, R.; Cheng, J.; Lin, J. Bacterial biofilm formation on biomaterials and approaches to its treatment and prevention. Int. J. Mol. Sci. 2023, 24, 11680. [Google Scholar] [CrossRef]
  3. Bhasme, P.; Wei, Q.; Xu, A.M.; Naqvi, S.T.A.; Wang, D.; Ma, L.Y.Z. Evaluation and characterization of the predicted diguanylate cyclase-encoding genes in. Microbiologyopen 2020, 9, e975. (In English) [Google Scholar] [CrossRef]
  4. Selim, H.M.R.M.; Gomaa, F.A.M.; Alshahrani, M.Y.; Morgan, R.N.; Aboshanab, K.M. Phage therapeutic delivery methods and clinical trials for combating clinically relevant pathogens. Ther. Deliv. 2025, 16, 247–269. [Google Scholar]
  5. Xu, A.; Wang, D.; Wang, Y.; Zhang, L.; Xie, Z.; Cui, Y.; Bhamse, P.; Yu, H.; Zhang, X.; Li, D.; et al. Mutations in surface-sensing receptor WspA lock the Wsp signal transduction system into a constitutively active state. Environ. Microbiol. 2024, 24, 247–269. [Google Scholar] [CrossRef]
  6. Hanke, M.L.; Kielian, T. Deciphering mechanisms of staphylococcal biofilm evasion of host immunity. Front. Cell. Infect. Microbiol. 2012, 2, 62. [Google Scholar] [CrossRef]
  7. Silva, A.; Silva, V.; López, M.; Rojo-Bezares, B.; Carvalho, J.A.; Castro, A.P.; Sáenz, Y.; Igrejas, G.; Poeta, P. Antimicrobial resistance, genetic lineages, and biofilm formation in Pseudomonas aeruginosa isolated from human infections: An emerging one health concern. Antibiotics 2023, 12, 1248. [Google Scholar] [CrossRef]
  8. Afrasiabi, S.; Partoazar, A. Targeting bacterial biofilm-related genes with nanoparticle-based strategies. Front. Microbiol. 2024, 15, 1387114. [Google Scholar]
  9. Azeredo, J.; García, P.; Drulis-Kawa, Z. Targeting biofilms using phages and their enzymes. Curr. Opin. Biotechnol. 2021, 68, 251–261. [Google Scholar] [CrossRef] [PubMed]
  10. Juszczuk-Kubiak, E. Molecular aspects of the functioning of pathogenic bacteria biofilm based on quorum sensing (QS) signal-response system and innovative non-antibiotic strategies for their elimination. Int. J. Mol. Sci. 2024, 25, 2655. [Google Scholar] [PubMed]
  11. Ding, Y.; Chen, S. A bibliographic outlook: Machine learning on biofilm. Res. Dir. Biotechnol. Des. 2025, 3, e2. [Google Scholar] [CrossRef]
  12. Seneviratne, C.J.; Suriyanarayanan, T.; Widyarman, A.S.; Lee, L.S.; Lau, M.; Ching, J.; Delaney, C.; Ramage, G. Multi-omics tools for studying microbial biofilms: Current perspectives and future directions. Crit. Rev. Microbiol. 2020, 46, 759–778. [Google Scholar] [CrossRef]
  13. Almatroudi, A. Investigating biofilms: Advanced methods for comprehending microbial behavior and antibiotic resistance. Front. Biosci. Landmark 2024, 29, 133. [Google Scholar]
  14. Grudlewska-Buda, K.; Skowron, K.; Gospodarek-Komkowska, E. Comparison of the intensity of biofilm formation by Listeria monocytogenes using classical culture-based method and digital droplet PCR. AMB Express 2020, 10, 75. [Google Scholar] [CrossRef] [PubMed]
  15. Haney, E.F.; Trimble, M.J.; Hancock, R.E. Microtiter plate assays to assess antibiofilm activity against bacteria. Nat. Protoc. 2021, 16, 2615–2632. [Google Scholar] [CrossRef]
  16. Walter, B.M.; Cartman, S.T.; Minton, N.P.; Butala, M.; Rupnik, M. The SOS response master regulator LexA is associated with sporulation, motility and biofilm formation in Clostridium difficile. PLoS ONE 2015, 10, e0144763. [Google Scholar]
  17. Rajamani, S.; Sandy, R.; Kota, K.; Lundh, L.; Gomba, G.; Recabo, K.; Duplantier, A.; Panchal, R.G. Robust biofilm assay for quantification and high throughput screening applications. J. Microbiol. Methods 2019, 159, 179–185. [Google Scholar] [CrossRef] [PubMed]
  18. Thieme, L.; Hartung, A.; Tramm, K.; Graf, J.; Spott, R.; Makarewicz, O.; Pletz, M.W. Adaptation of the start-growth-time method for high-throughput biofilm quantification. Front. Microbiol. 2021, 12, 631248. [Google Scholar]
  19. Ayon, N.J. High-throughput screening of natural product and synthetic molecule libraries for antibacterial drug discovery. Metabolites 2023, 13, 625. [Google Scholar] [CrossRef]
  20. Jia, J.; Xue, X.; Guan, Y.; Fan, X.; Wang, Z. Biofilm characteristics and transcriptomic profiling of Acinetobacter johnsonii defines signatures for planktonic and biofilm cells. Environ. Res. 2022, 213, 113714. [Google Scholar] [CrossRef]
  21. Gilbert, P.; Maira-Litran, T.; McBain, A.J.; Rickard, A.H.; Whyte, F.W. The physiology and collective recalcitrance of microbial biofilm communities. Adv. Microb. Physiol. 2002, 46, 203–256. [Google Scholar]
  22. Wu, Y.; Li, X.; Fu, X.; Huang, X.; Zhang, S.; Zhao, N.; Ma, X.; Saiding, Q.; Yang, M.; Tao, W. Innovative nanotechnology in drug delivery systems for advanced treatment of posterior segment ocular diseases. Adv. Sci. 2024, 11, 2403399. [Google Scholar]
  23. Kragh, K.N.; Alhede, M.; Kvich, L.; Bjarnsholt, T. Into the well—A close look at the complex structures of a microtiter biofilm and the crystal violet assay. Biofilm 2019, 1, 100006. [Google Scholar] [CrossRef] [PubMed]
  24. Amran, S.S.D.; Syaida, A.A.R.; Jalil, M.T.M.; Nor, N.H.M.; Yahya, M.F.Z.R. Preparation of Biofilm Assay Using 96-Well and 6-Well Microplates for Quantitative Measurement and Structural Characterization: A Review. Sci. Lett. 2024, 18, 121–134. [Google Scholar]
  25. Salcedo, F.; Pereyra, C.M.; Arruebarrena Di Palma, A.; Lamattina, L.; Creus, C.M. Methods for studying biofilms in Azospirillum and other PGPRs. In Handbook for Azospirillum: Technical Issues and Protocols; Springer: Berlin/Heidelberg, Germany, 2015; pp. 199–229. [Google Scholar]
  26. Cullimore, R. Monitoring Methodologies for Biofouling Events. In Microbiology of Well Biofouling; CRC Press: Boca Raton, FL, USA, 2018; pp. 77–136. [Google Scholar]
  27. Kırmusaoğlu, S. Biofilm and Screening Antibiofilm Activity of Agents. In Antimicrobials, Antibiotic Resistance, Antibiofilm Strategies and Activity Methods; IntechOpen: London, UK, 2019; p. 99. [Google Scholar]
  28. Nwaiwu, J. Development of Alternative Protocols for Antimicrobial Efficacy Testing of Antimicrobial Surfaces. Master’s Thesis, University of Waterloo, Waterloo, CN, Canada, 2020. [Google Scholar]
  29. Ogundero, A. The Theoretical and Experimental Exploration of the Use of Predatory Bacteria to Control Biofilms. Ph.D. Thesis, University of Glasgow, Glasgow, UK, 2023. [Google Scholar]
  30. Schmutzler, K.; Schmid, A.; Buehler, K. A three-step method for analysing bacterial biofilm formation under continuous medium flow. Appl. Microbiol. Biotechnol. 2015, 99, 6035–6047. [Google Scholar] [CrossRef] [PubMed]
  31. Goeres, D.M.; Walker, D.K.; Buckingham-Meyer, K.; Lorenz, L.; Summers, J.; Fritz, B.; Goveia, D.; Dickerman, G.; Schultz, J.; Parker, A.E. Development, standardization, and validation of a biofilm efficacy test: The single tube method. J. Microbiol. Methods 2019, 165, 105694. [Google Scholar] [CrossRef]
  32. Wilson, M.-M. In Vitro Study of Effectivity of Bacteriophages in the Prevention and Degradation of Biofilms Formed by Uropathogenic E. coli in Urinary Chatheters. Master’s Thesis, University of the Witwatersrand, Johannesburg, South Africa, 2018. [Google Scholar]
  33. Allkja, J. Development of Minimum Information Guidelines and Standardized in Vitro Methods for Biofilm Experiments. Ph.D. Thesis, Universidade do Porto, Porto, Portugal, 2021. [Google Scholar]
  34. Xiong, Z.; Tian, X.; Wang, G.; Song, X.; Xia, Y.; Zhang, H.; Ai, L. Development of a high-throughput screening method for exopolysaccharide-producing Streptococcus thermophilus based on Congo red. Food Res. Int. 2022, 162, 112094. [Google Scholar] [PubMed]
  35. Oladoye, P.O.; Bamigboye, M.O.; Ogunbiyi, O.D.; Akano, M.T. Toxicity and decontamination strategies of Congo red dye. Groundw. Sustain. Dev. 2022, 19, 100844. [Google Scholar] [CrossRef]
  36. Harja, M.; Buema, G.; Bucur, D. Recent advances in removal of Congo Red dye by adsorption using an industrial waste. Sci. Rep. 2022, 12, 6087. [Google Scholar] [CrossRef]
  37. Sudha, S.S.; Aranganathan, V. Antibiofilm analysis, synergistic potential and biocompatibility evaluation of a bacteriocin from Bacillus subtilis (MK733983). Indian J. Microbiol. 2024, 64, 1646–1663. [Google Scholar] [CrossRef] [PubMed]
  38. Papadopoulos, C. A Novel Experimental Platform for Monitoring and Imaging Bacterial Biofilm Growth in Porous Media Flows. Ph.D. Thesis, Institut National Polytechnique de Toulouse-INPT, Toulouse, France, 2023. [Google Scholar]
  39. Rabbitt, D.; Villapún, V.M.; Carter, L.N.; Man, K.; Lowther, M.; O’Kelly, P.; Knowles, A.J.; Mottura, A.; Tang, Y.T.; Luerti, L. Rethinking biomedical titanium alloy design: A review of challenges from biological and manufacturing perspectives. Adv. Healthc. Mater. 2025, 14, 2403129. [Google Scholar]
  40. Dhanasinghe, C.; Kao, C.M.; Liu, P.F.; Surampalli, R.Y.; Zhang, T.C.; Al-Hashimi, B.M. Advanced Techniques for Sampling, Quantification, and Characterization of Microplastics. In Microplastics in the Environment: Fate, Impacts, Removal, and Management; Wiley: Hoboken, NJ, USA, 2025; pp. 69–106. [Google Scholar]
  41. Jones, L.M. Microbiologically Influenced Corrosion: Development of a Model System to Investigate the Role of Biofilm Communities Within MIC and Their Control Using Industrial Biocides. Ph.D. Thesis, University of Southampton, Southampton, UK, 2025. [Google Scholar]
  42. Bourn, M.D.; Daly, L.F.; Huggett, J.F.; Braybrook, J.; Rivera, J.F. Evaluation of image analysis tools for the measurement of cellular morphology. Front. Cell Dev. Biol. 2025, 13, 1572212. [Google Scholar] [CrossRef] [PubMed]
  43. Rodrigues, C.F.; Allkja, J.; Mendes, L.; Azevedo, A.S. Methods for the visualization of multispecies biofilms. In Multispecies Biofilms: Technologically Advanced Methods to Study Microbial Communities; Springer: Berlin/Heidelberg, Germany, 2022; pp. 35–78. [Google Scholar]
  44. Lee, D.; Lee, H.-S.; Lee, M.; Kang, M.; Kim, G.; Kim, T.Y.; Lee, N.Y.; Park, Y. Advances in imaging techniques for the study of individual bacteria and their pathophysiology. arXiv 2025, arXiv:2501.01592. [Google Scholar] [CrossRef]
  45. Relucenti, M.; Familiari, G.; Donfrancesco, O.; Taurino, M.; Li, X.; Chen, R.; Artini, M.; Papa, R.; Selan, L. Microscopy methods for biofilm imaging: Focus on SEM and VP-SEM pros and cons. Biology 2021, 10, 51. [Google Scholar] [CrossRef]
  46. Gerardi, D.; Bernardi, S.; Bruni, A.; Falisi, G.; Botticelli, G. Characterization and morphological methods for oral biofilm visualization: Where are we nowadays? AIMS Microbiol. 2024, 10, 391. [Google Scholar] [CrossRef] [PubMed]
  47. Choubey, S.; Deshpande, J. A Study on Exploring the Composition, Structure and Innovative Analytical Techniques Developed in Biomembrane Research. Biosci. Biotechnol. Res. Asia 2025, 22, 63. [Google Scholar] [CrossRef]
  48. Samuel, H.S.; Ekpan, F.-D.M. The use of Scanning Electron Microscopy (SEM) for medical application: A mini review. Eurasian J. Sci. Technol. 2024, 4, 289–294. [Google Scholar]
  49. McCutcheon, J.; Southam, G. Advanced biofilm staining techniques for TEM and SEM in geomicrobiology: Implications for visualizing EPS architecture, mineral nucleation, and microfossil generation. Chem. Geol. 2018, 498, 115–127. [Google Scholar] [CrossRef]
  50. Rohde, M. Bacterial ultrastructure. In Molecular Medical Microbiology; Elsevier: Amsterdam, The Netherlands, 2024; pp. 23–43. [Google Scholar]
  51. Van Hoogstraten, S.; Kuik, C.; Arts, J.; Cillero-Pastor, B. Molecular imaging of bacterial biofilms—A systematic review. Crit. Rev. Microbiol. 2024, 50, 971–992. [Google Scholar] [CrossRef]
  52. Millan-Solsona, R.; Brown, S.R.; Zhang, L.; Madugula, S.S.; Zhao, H.; Dumerer, B.; Bible, A.N.; Lavrik, N.V.; Vasudevan, R.K.; Biswas, A. Analysis of Biofilm Assembly by Large Area Automated AFM. NPJ Biofilms Microbiomes 2025, 11, 75. [Google Scholar]
  53. Das, M.; Khamaru, D.; Santra, S.; Gupta, R.; Das, S.; Banerjee, R. The Enigmatic World of Biofilm. In Biofilm Applications to Revolutionize Food Technology; Springer: Berlin/Heidelberg, Germany, 2025; pp. 1–26. [Google Scholar]
  54. Albalawi, T. Strategies for Detecting Bacterial Biofilms: Unveiling the Hidden World of Microbial Aggregates. Egypt. J. Soil Sci. 2024, 64, 1069–1096. [Google Scholar] [CrossRef]
  55. Zhang, Y.; Young, P.; Traini, D.; Li, M.; Ong, H.X.; Cheng, S. Challenges and current advances in in vitro biofilm characterization. Biotechnol. J. 2023, 18, 2300074. [Google Scholar] [CrossRef]
  56. Coppola, F.; Fratianni, F.; Bianco, V.; Wang, Z.; Pellegrini, M.; Coppola, R.; Nazzaro, F. New Methodologies as Opportunities in the Study of Bacterial Biofilms, Including Food-Related Applications. Microorganisms 2025, 13, 1062. [Google Scholar] [CrossRef]
  57. Müller, T.; Schumann, C.; Kraegeloh, A. STED microscopy and its applications: New insights into cellular processes on the nanoscale. ChemPhysChem 2012, 13, 1986–2000. [Google Scholar] [CrossRef]
  58. Blom, H.; Widengren, J. Stimulated emission depletion microscopy. Chem. Rev. 2017, 117, 7377–7427. [Google Scholar] [CrossRef] [PubMed]
  59. Di Martino, P. Extracellular polymeric substances, a key element in understanding biofilm phenotype. AIMS Microbiol. 2018, 4, 274. [Google Scholar] [CrossRef]
  60. Thomsen, H. Biophotonics Targeting Pharmaceutical Challenges. Ph.D. Thesis, University of Gothenburg, Gothenburg, Sweden, 2018. [Google Scholar]
  61. Kromer, C. Nanoscale Tools for Biosensing and Treatment of Bacterial Biofilms and Eukaryotic Cells. Ph.D. Thesis, Freie Universität Berlin, Berlin, Germany, 2024. [Google Scholar]
  62. Bharadwaj, A.; Kumar, A.; Mathew, S.P.; Mitra, R.; Bhattacharyya, J.; Jaganathan, B.G.; Boruah, B.R. Advancing cellular insights: Super-resolution STORM imaging of cytoskeletal structures in human stem and cancer cells. Biochem. Biophys. Rep. 2024, 39, 101798. [Google Scholar] [CrossRef] [PubMed]
  63. Zhang, J.; Zhang, M.; Wang, Y.; Donarski, E.; Gahlmann, A. Optically accessible microfluidic flow channels for noninvasive high-resolution biofilm imaging using lattice light sheet microscopy. J. Phys. Chem. B 2021, 125, 12187–12196. [Google Scholar] [CrossRef]
  64. Hassan, L.; Rukan, M.; Mohammad, E.; Syed, M.A. Recent Advances in Microbiology Techniques for Imaging Microbes and Their Components. In Microbiology in the Era of Artificial Intelligence; CRC Press: Boca Raton, FL, USA, 2025; pp. 103–135. [Google Scholar]
  65. Xu, J.; Ma, H.; Liu, Y. Stochastic optical reconstruction microscopy (STORM). Curr. Protoc. Cytom. 2017, 81, 12.46.1–12.46.27. [Google Scholar] [CrossRef]
  66. Bharadwaj, A.; Kumar, A.; Kalita, R.; Mitra, R.; Sharma, A.; Jaganathan, B.G.; Bhattacharyya, J.; Sarma, A.; Garcia, E.; Kumar, S. Super-resolved fluorescence imaging utilising accessible stochastic optical reconstruction microscopy (easySTORM) implemented on a low-cost, modular open-source (openFrame) microscope. Meas. Sci. Technol. 2024, 35, 125402. [Google Scholar]
  67. Liu, H.; Wang, X.; Wang, Z.; Shen, Y. Evaluation of bacterial biofilm, smear layer, and debris removal efficacy of a hydro-dynamic cavitation system with physiological saline using a new ex vivo model: A CLSM and SEM study. BMC Oral Health 2025, 25, 95. [Google Scholar] [CrossRef]
  68. Tartari, T.; Estrela, C.; de Araújo, L.B.B.; Graeff, M.S.Z.; de Andrade, F.B.; Duarte, M.A.H. Use of confocal laser scanning microscopy to evaluate the metal ion removal and destabilization of Enterococcus faecalis biofilms by EDTA and etidronic acid. Odontology 2025, 1–9. [Google Scholar] [CrossRef]
  69. Dhahri, S.; Marlière, C. Atomic Force Microsocopy: Key Unconventional Approach for Bacterial Nanotubes Characterization In Vivo. ACS Omega 2024, 9, 46950–46959. [Google Scholar] [CrossRef] [PubMed]
  70. Wang, X.; Xue, J.; Zhu, H.; Chen, S.; Wang, Y.; Xiao, Z.; Luo, Y. Advances in biofilm characterization: Utilizing rheology and atomic force microscopy in foods and related fields. Adv. Compos. Hybrid Mater. 2024, 7, 143. [Google Scholar] [CrossRef]
  71. Medina-Ramirez, I.E.; Macías-Díaz, J.E.; Masuoka-Ito, D.; Zapien, J.A. Holotomography and atomic force microscopy: A powerful combination to enhance cancer, microbiology and nanotoxicology research. Discov. Nano 2024, 19, 64. [Google Scholar]
  72. Francius, G. Correlation between nanomechanical and chemical features of crosslinked hydrogels combining AFM with chemometric methods. In Proceedings of the AFM BioMed Conference, Barcelona, Spain, 8–11 April 2025; Volume 469, pp. 100–133. [Google Scholar]
  73. Heydari, S.; Liu, J. High-throughput cryo-electron tomography enables multiscale visualization of the inner life of microbes. Curr. Opin. Struct. Biol. 2025, 93, 103065. [Google Scholar]
  74. Viljoen, A.; Mathelié-Guinlet, M.; Ray, A.; Strohmeyer, N.; Oh, Y.J.; Hinterdorfer, P.; Müller, D.J.; Alsteens, D.; Dufrêne, Y.F. Force spectroscopy of single cells using atomic force microscopy. Nat. Rev. Methods Primers 2021, 1, 63. [Google Scholar]
  75. Mohammadi, A.T.; Naderi, N.; Kenari, P.B.; Mir, A.; Sabouri, F.; Amin, M.; Fatahi, G. Advancements in Pharmacology: Integrating Molecular Mechanisms, Therapeutics, and Drug Development; Nobel Sciences: Stockholm, Sweden, 2025. [Google Scholar]
  76. Li, K.; Li, D.; Gao, L.; Weng, Z.; Zheng, M.; He, Y.; Li, Z. Application of optical coherence tomography for morphological change of Staphylococcus during reproduction. In Proceedings of the 10th International Symposium on Advanced Optical Manufacturing and Testing Technologies: Intelligent Sensing Technologies and Applications, Chengdu, China, 20–24 June 2021; Volume 12075, SPIE. pp. 124–133. [Google Scholar]
  77. Lin, W.C. Development and Applications of Near-and Mid-infrared OCT. Ph.D. Thesis, University of Sheffield, Sheffield, UK, 2021. [Google Scholar]
  78. Qamar, A.; Kerdi, S.; Amin, N.; Zhang, X.; Vrouwenvelder, J.; Ghaffour, N. A deep neural networks framework for in-situ biofilm thickness detection and hydrodynamics tracing for filtration systems. Sep. Purif. Technol. 2022, 301, 121959. [Google Scholar] [CrossRef]
  79. Lima, C.; Ahmed, S.; Xu, Y.; Muhamadali, H.; Parry, C.; McGalliard, R.J.; Carrol, E.D.; Goodacre, R. Simultaneous Raman and infrared spectroscopy: A novel combination for studying bacterial infections at the single cell level. Chem. Sci. 2022, 13, 8171–8179. [Google Scholar] [CrossRef] [PubMed]
  80. Wang, Y.; Bian, Z.; Wang, Y. Biofilm formation and inhibition mediated by bacterial quorum sensing. Appl. Microbiol. Biotechnol. 2022, 106, 6365–6381. [Google Scholar] [CrossRef]
  81. Syed, A.J.; Anderson, J.C. Applications of bioluminescence in biotechnology and beyond. Chem. Soc. Rev. 2021, 50, 5668–5705. [Google Scholar] [CrossRef]
  82. Resendiz-Sharpe, A.; Vanhoffelen, E.; Velde, G.V. Bioluminescence Imaging, a Powerful Tool to Assess Fungal Burden in Live Mouse Models of Infection. In Antifungal Immunity: Methods and Protocols; Springer: Berlin/Heidelberg, Germany, 2023; pp. 197–210. [Google Scholar]
  83. Jiang, T.; Bai, X.; Li, M. Advances in the development of bacterial bioluminescence imaging. Annu. Rev. Anal. Chem. 2024, 17, 265–288. [Google Scholar] [CrossRef]
  84. Mihaylova, E.M. Imaging of Live Cells by Digital Holographic Microscopy. Photonics 2024, 11, 980. [Google Scholar] [CrossRef]
  85. Riekeles, M.L.I. Microbial Motility as a Universal Biosignature. Ph.D. Thesis, Technische Universitaet Berlin, Berlin, Germany, 2025. [Google Scholar]
  86. Buzalewicz, I.; Kaczorowska, A.; Fijałkowski, W.; Pietrowska, A.; Matczuk, A.K.; Podbielska, H.; Wieliczko, A.; Witkiewicz, W.; Jędruchniewicz, N. Quantifying the dynamics of bacterial biofilm formation on the surface of soft contact lens materials using digital holographic tomography to advance biofilm research. Int. J. Mol. Sci. 2024, 25, 2653. [Google Scholar] [CrossRef] [PubMed]
  87. Parthasarathi, S.; Chaudhury, A.; Swain, A.; Basu, J.K. Microscopy Insights as an Invaluable Tool for Studying Antimicrobial Interactions with Bacterial Membranes. ChemistrySelect 2025, 10, e02345. [Google Scholar] [CrossRef]
  88. Jeong, D.; Kim, M.J.; Park, Y.; Chung, J.; Kweon, H.-S.; Kang, N.-G.; Hwang, S.J.; Youn, S.H.; Hwang, B.K.; Kim, D. Visualizing extracellular vesicle biogenesis in gram-positive bacteria using super-resolution microscopy. BMC Biol. 2022, 20, 270. [Google Scholar] [CrossRef]
  89. Reichhardt, C.; Parsek, M.R. Confocal laser scanning microscopy for analysis of Pseudomonas aeruginosa biofilm architecture and matrix localization. Front. Microbiol. 2019, 10, 677. [Google Scholar] [CrossRef]
  90. Rotsch, C.; Radmacher, M. Drug-induced changes of cytoskeletal structure and mechanics in fibroblasts: An atomic force microscopy study. Biophys. J. 2000, 78, 520–535. [Google Scholar] [CrossRef]
  91. Liedtke, J.; Depelteau, J.S.; Briegel, A. How advances in cryo-electron tomography have contributed to our current view of bacterial cell biology. J. Struct. Biol. X 2022, 6, 100065. [Google Scholar] [CrossRef] [PubMed]
  92. Kore, S.M. Impact of Shear Flow on Marine Biofilms Using Hyperspectral Imaging. Ph.D. Thesis, Clemson University, Clemson, SC, USA, 2025. [Google Scholar]
  93. Zhang, P.; Feng, B.; Chen, Y.-P.; Dai, Y.-Z.; Guo, J.-S. In situ characterizations for EPS-involved microprocesses in biological wastewater treatment systems. Crit. Rev. Environ. Sci. Technol. 2019, 49, 917–946. [Google Scholar] [CrossRef]
  94. Pan, J.; Peng, Q.; Zhang, G.; Xie, Q.; Gong, X.; Qian, P.-Y.; Ma, C.; Zhang, G. Antifouling mechanism of natural product-based coatings investigated by digital holographic microscopy. J. Mater. Sci. Technol. 2021, 84, 200–207. [Google Scholar] [CrossRef]
  95. Bankar, N.; Bankar, M.; Shelke, Y.; Kale, Y.; Khedkar, S. The role of artificial intelligence in biofilm detection. Proc. AIP Conf. Proc. 2024, 3188, 080038. [Google Scholar]
  96. Sahana, M. AI-Driven Discovery of Nanostructures That Disrupt Antibiotic-Resistant Biofilms. Master’s Thesis, University of Mysore, Mysore, Indian, 2025. [Google Scholar]
  97. Chen, H.; Xia, A.; Yan, H.; Huang, Y.; Zhu, X.; Zhu, X.; Liao, Q. Mass transfer in heterogeneous biofilms: Key issues in biofilm reactors and AI-driven performance prediction. Environ. Sci. Ecotechnol. 2024, 22, 100480. [Google Scholar] [CrossRef]
  98. Zou, H.; Sopasakis, A.; Maillard, F.; Karlsson, E.; Duljas, J.; Silwer, S.; Ohlsson, P.; Hammer, E.C. Bacterial community characterization by deep learning aided image analysis in soil chips. Ecol. Inform. 2024, 81, 102562. [Google Scholar] [CrossRef]
  99. Liao, Y.; Zhao, J.; Bian, J.; Zhang, Z.; Xu, S.; Qin, Y.; Miao, S.; Li, R.; Liu, R.; Zhang, M. From mechanism to application: Decrypting light-regulated denitrifying microbiome through geometric deep learning. Imeta 2024, 3, e162. [Google Scholar] [CrossRef] [PubMed]
  100. Wang, J.; Tabassum, N.; Toma, T.T.; Wang, Y.; Gahlmann, A.; Acton, S.T. 3D GAN image synthesis and dataset quality assessment for bacterial biofilm. Bioinformatics 2022, 38, 4598–4604. [Google Scholar] [CrossRef]
  101. Holicheva, A.A.; Kozlov, K.S.; Boiko, D.A.; Kamanin, M.S.; Provotorova, D.V.; Kolomoets, N.I.; Ananikov, V.P. Deep generative modeling of annotated bacterial biofilm images. NPJ Biofilms Microbiomes 2025, 11, 16. [Google Scholar] [CrossRef]
  102. Huang, Y. Machine Learning for Actionable Knowledge Discovery in Synthetic Biology. Doctoral Dissertation, Newcastle University, Newcastle upon Tyne, UK, 2023. [Google Scholar]
  103. Samue, F. Machine Learning Approaches to Anomaly Detection in Biomedical Data and Medical Imaging. Available online: https://www.researchgate.net/publication/385078064_Machine_Learning_Approaches_to_Anomaly_Detection_in_Biomedical_Data_and_Medical_Imaging?enrichId=rgreq-a8bbaa738c8ee17c0e6c5b61c3ab4afc-XXX&enrichSource=Y292ZXJQYWdlOzM4NTA3ODA2NDtBUzoxMTQzMTI4MTI4NDcyMj (accessed on 18 August 2025).
  104. Shojaee, A. Bacteria Growth Modeling Using Long-Short-Term-Memory Networks. Master’s Thesis, University of Cincinnati, Cincinnati, OH, USA, 2021. [Google Scholar]
  105. Franco, N.R. Machine Learning for Precision Medicine: A Combination of Data-Driven and Physics Based Models. 2022. Available online: https://hdl.handle.net/10589/196604 (accessed on 18 August 2025).
  106. Millevoi, C. Physics-Informed Neural Network in Porous Media and Epidemiological Applications. Ph.D. Thesis, Università degli Studi di Padova, Padova, Italy, 2024. [Google Scholar]
  107. Guzmán-Soto, I.; McTiernan, C.; Gonzalez-Gomez, M.; Ross, A.; Gupta, K.; Suuronen, E.J.; Mah, T.-F.; Griffith, M.; Alarcon, E.I. Mimicking biofilm formation and development: Recent progress in in vitro and in vivo biofilm models. Iscience 2021, 24, 102443. [Google Scholar] [CrossRef]
  108. Highmore, C.; Melaugh, G.; Morris, R.; Parker, J.; Direito, S.; Romero, M.; Soukarieh, F.; Robertson, S.; Bamford, N. Translational challenges and opportunities in biofilm science: A BRIEF for the future. NPJ Biofilms Microbiomes 2022, 8, 68. [Google Scholar] [CrossRef]
  109. Jandl, B.; Dighe, S.; Gasche, C.; Makristathis, A.; Muttenthaler, M. Intestinal biofilms: Pathophysiological relevance, host defense, and therapeutic opportunities. Clin. Microbiol. Rev. 2024, 37, e0013323. [Google Scholar] [CrossRef]
  110. Sahoo, K.; Meshram, S.; Sahoo Jr, K. Biofilm Formation in Chronic Infections: A Comprehensive Review of Pathogenesis, Clinical Implications, and Novel Therapeutic Approaches. Cureus 2024, 16. [Google Scholar] [CrossRef]
  111. Bhattacharjee, B.; Ghosh, S.; Patra, D.; Haldar, J. Advancements in release-active antimicrobial biomaterials: A journey from release to relief. Wiley Interdiscip. Rev. Nanomed. Nanobiotechnol. 2022, 14, e1745. [Google Scholar] [CrossRef]
  112. Billings, C.J. Implantable Medical Devices for Local Drug Delivery and Tissue Regeneration to Combat Chronic Bacterial Infection. Doctoral Dissertation, University of Tennessee College of Veterinary, Knoxville, TN, USA, 2022. [Google Scholar]
  113. Mannering, N.; Narulla, R.; Lenane, B. In-Vitro and In-Vivo Models for the Study of Prosthetic Joint Infections. In Infection in Knee Replacement; Springer: Berlin/Heidelberg, Germany, 2021; pp. 41–47. [Google Scholar]
  114. van Agtmaal, J.L.; van Hoogstraten, S.W.; Arts, J.J. Prosthetic Joint Infection Research Models in NZW Rabbits: Opportunities for Standardization—A Systematic Review. J. Funct. Biomater. 2024, 15, 307. [Google Scholar] [CrossRef] [PubMed]
  115. Chen, V.; Burgess, J.L.; Verpile, R.; Tomic-Canic, M.; Pastar, I. Novel diagnostic technologies and therapeutic approaches targeting chronic wound biofilms and microbiota. Curr. Dermatol. Rep. 2022, 11, 60–72. [Google Scholar] [CrossRef]
  116. Saeed, S.; Martins-Green, M. Assessing animal models to study impaired and chronic wounds. Int. J. Mol. Sci. 2024, 25, 3837. [Google Scholar] [CrossRef]
  117. Miron, A.; Giurcaneanu, C.; Mihai, M.M.; Beiu, C.; Voiculescu, V.M.; Popescu, M.N.; Soare, E.; Popa, L.G. Antimicrobial biomaterials for chronic wound care. Pharmaceutics 2023, 15, 1606. [Google Scholar] [CrossRef] [PubMed]
  118. Reyne, N.; McCarron, A.; Cmielewski, P.; Parsons, D.; Donnelley, M. To bead or not to bead: A review of Pseudomonas aeruginosa lung infection models for cystic fibrosis. Front. Physiol. 2023, 14, 1104856. [Google Scholar] [CrossRef]
  119. Werneburg, G.T. Catheter-associated urinary tract infections: Current challenges and future prospects. Res. Rep. Urol. 2022, 14, 109–133. [Google Scholar] [CrossRef] [PubMed]
  120. Nissanka, M.C.; Dilhari, A.; Wijesinghe, G.K.; Weerasekera, M.M. Advances in experimental bladder models: Bridging the gap between in vitro and in vivo approaches for investigating urinary tract infections. BMC Urol. 2024, 24, 206. [Google Scholar] [CrossRef]
  121. Jakubovics, N.S.; Goodman, S.D.; Mashburn-Warren, L.; Stafford, G.P.; Cieplik, F. The dental plaque biofilm matrix. Periodontol. 2000 2021, 86, 32–56. [Google Scholar] [CrossRef]
  122. Haque, M.M.; Yerex, K.; Kelekis-Cholakis, A.; Duan, K. Advances in novel therapeutic approaches for periodontal diseases. BMC Oral Health 2022, 22, 492. [Google Scholar] [CrossRef]
  123. Barnes, A.M.; Frank, K.L.; Dale, J.L.; Manias, D.A.; Powers, J.L.; Dunny, G.M. Enterococcus faecalis colonizes and forms persistent biofilm microcolonies on undamaged endothelial surfaces in a rabbit endovascular infection model. FEMS Microbes 2021, 2, xtab014. [Google Scholar] [CrossRef]
  124. Schwarz, C.; Töre, Y.; Hoesker, V.; Ameling, S.; Grün, K.; Völker, U.; Schulze, P.C.; Franz, M.; Faber, C.; Schaumburg, F. Host-pathogen interactions of clinical S. aureus isolates to induce infective endocarditis. Virulence 2021, 12, 2073–2087. [Google Scholar] [CrossRef]
  125. Motta, J.-P.; Wallace, J.L.; Buret, A.G.; Deraison, C.; Vergnolle, N. Gastrointestinal biofilms in health and disease. Nat. Rev. Gastroenterol. Hepatol. 2021, 18, 314–334. [Google Scholar] [CrossRef]
  126. Duff, A.F.; Jurcisek, J.A.; Kurbatfinski, N.; Chiang, T.; Goodman, S.D.; Bakaletz, L.O.; Bailey, M.T. Oral and middle ear delivery of otitis media standard of care antibiotics, but not biofilm-targeted antibodies, alter chinchilla nasopharyngeal and fecal microbiomes. Npj Biofilms Microbiomes 2024, 10, 10. [Google Scholar] [CrossRef] [PubMed]
  127. Empitu, M.A.; Kadariswantiningsih, I.N.; Shakri, N.M. Pharmacological strategies for targeting biofilms in otorhinolaryngologic infections and overcoming antimicrobial resistance. Biomed. Rep. 2025, 22, 95. [Google Scholar] [CrossRef] [PubMed]
  128. Gill, R.P.K.; Gantchev, J.; Martínez Villarreal, A.; Ramchatesingh, B.; Netchiporouk, E.; Akilov, O.E.; Ødum, N.; Gniadecki, R.; Koralov, S.B.; Litvinov, I.V. Understanding cell lines, patient-derived xenograft and genetically engineered mouse models used to study cutaneous T-cell lymphoma. Cells 2022, 11, 593. [Google Scholar] [CrossRef] [PubMed]
  129. Lloyd, M.G.; Moffat, J.F. Humanized severe combined immunodeficient (SCID) mouse models for varicella-zoster virus pathogenesis. In Varicella-zoster Virus: Genetics, Pathogenesis and Immunity; Springer: Berlin/Heidelberg, Germany, 2022; pp. 135–161. [Google Scholar]
  130. Mohamed, H.S.; Houmed Aboubaker, M.; Dumont, Y.; Didelot, M.-N.; Michon, A.-L.; Galal, L.; Jean-Pierre, H.; Godreuil, S. Multidrug-Resistant Enterobacterales in Community-Acquired Urinary Tract Infections in Djibouti, Republic of Djibouti. Antibiotics 2022, 11, 1740. [Google Scholar] [CrossRef]
  131. Meneely, P.M.; Dahlberg, C.L.; Rose, J.K. Working with Worms: Caenorhabditis elegans as a Model Organism. Curr. Protoc. Essent. Lab. Tech. 2019, 19, e35. [Google Scholar] [CrossRef]
  132. Atkinson, S.; Goldstone, R.J.; Joshua, G.W.P.; Chang, C.Y.; Patrick, H.L.; Cámara, M.; Wren, B.W.; Williams, P. Biofilm Development on by Is Facilitated by Quorum Sensing-Dependent Repression of Type III Secretion. PLoS Pathog. 2011, 7, e1001250. (In English) [Google Scholar] [CrossRef]
  133. Javanmard, Z.; Pourhajibagher, M.; Bahador, A. Advancing anti-biofilm strategies: Innovations to combat biofilm-related challenges and enhance efficacy. J. Basic Microbiol. 2024, 64, e2400271. [Google Scholar] [CrossRef]
  134. Singh, R. Revolutionizing Antimicrobial Therapies Through Biofilm-Targeted Nanomedicine. Curr. Pharm. Res. 2025, 1, 78–97. [Google Scholar] [CrossRef]
  135. Martínez-García, E.; de Lorenzo, V. Pseudomonas putida as a synthetic biology chassis and a metabolic engineering platform. Curr. Opin. Biotechnol. 2024, 85, 103025. [Google Scholar] [CrossRef]
  136. Arora, A. Crispri-mediated gene silencing in biofilm cycle and quorum sensing. In Gene Editing in Plants: Crispr-Cas and Its Applications; Springer: Berlin/Heidelberg, Germany, 2024; pp. 139–178. [Google Scholar]
  137. Zhang, Y.; Zhang, T.; Xiao, X.; Kawalek, A.; Ou, J.; Ren, A.; Sun, W.; de Bakker, V.; Liu, Y.; Li, Y. Genome-wide CRISPRi-seq identified ferredoxin-NADP reductase FprB as a synergistic target for gallium therapy in Pseudomonas aeruginosa. bioRxiv 2024. [Google Scholar]
  138. Zuberi, A.; Ahmad, N.; Ahmad, H.; Saeed, M.; Ahmad, I. Beyond antibiotics: CRISPR/Cas9 triumph over biofilm-associated antibiotic resistance infections. Front. Cell. Infect. Microbiol. 2024, 14, 1408569. [Google Scholar] [CrossRef] [PubMed]
  139. Ghaznavi, G.; Vosough, P.; Ghasemian, A.; Tabar, M.M.M.; Tayebi, L.; Taghizadeh, S.; Savardashtaki, A. Engineering bacteriophages for targeted superbug eradication. Mol. Biol. Rep. 2025, 52, 221. [Google Scholar] [CrossRef]
  140. Chen, Y.C.; Destouches, L.; Cook, A.; Fedorec, A.J. Synthetic microbial ecology: Engineering habitats for modular consortia. J. Appl. Microbiol. 2024, 135, lxae158. [Google Scholar] [CrossRef]
  141. Kozaeva, E.; Eida, A.A.; Gunady, E.F.; Dangl, J.L.; Conway, J.M.; Brophy, J.A. Roots of synthetic ecology: Microbes that foster plant resilience in the changing climate. Curr. Opin. Biotechnol. 2024, 88, 103172. [Google Scholar] [CrossRef]
  142. Xu, B.; Su, Q.; Yang, Y.; Huang, S.; Yang, Y.; Shi, X.; Choo, K.-H.; Ng, H.Y.; Lee, C.-H. Quorum quenching in membrane bioreactors for fouling retardation: Complexity provides opportunities. Environ. Sci. Technol. 2024, 58, 13171–13193. [Google Scholar] [CrossRef] [PubMed]
  143. Ghosh, S.; Ray, R.R. Nanomaterials, Phytocompounds. In Biofilm Associated Livestock Diseases and their Management; Springer Nature: Berlin/Heidelberg, Germany, 2025; p. 233. [Google Scholar]
  144. Stojanov, S.; Berlec, A. Smart bionanomaterials for treatment and diagnosis of inflammatory bowel disease. Nanotechnol. Rev. 2024, 13, 20240057. [Google Scholar] [CrossRef]
  145. Chang, X.; Liu, X.; Wang, X.; Ma, L.; Liang, J.; Li, Y. Recent Advances in Spatiotemporal Manipulation of Engineered Bacteria for Precision Cancer Therapy. Int. J. Nanomed. 2025, 20, 5859–5872. [Google Scholar] [CrossRef]
  146. Elazzazy, A.M.; Baeshen, M.N.; Alasmi, K.M.; Alqurashi, S.I.; Desouky, S.E.; Khattab, S.M. Where Biology Meets Engineering: Scaling up microbial nutraceuticals to bridge nutrition, therapeutics, and global impact. Microorganisms 2025, 13, 566. [Google Scholar] [CrossRef]
  147. Dash, A.; Ragavendran, C. Innovative Approaches to Combating Dental Biofilms: Nanotechnology and its Applications. Biocatal. Agric. Biotechnol. 2024, 61, 103406. [Google Scholar] [CrossRef]
  148. Waheed, S.; Li, Z.; Zhang, F.; Chiarini, A.; Armato, U.; Wu, J. Engineering nano-drug biointerface to overcome biological barriers toward precision drug delivery. J. Nanobiotechnol. 2022, 20, 395. [Google Scholar] [CrossRef]
  149. Mohanta, Y.K.; Chakrabartty, I.; Mishra, A.K.; Chopra, H.; Mahanta, S.; Avula, S.K.; Patowary, K.; Ahmed, R.; Mishra, B.; Mohanta, T.K. Nanotechnology in combating biofilm: A smart and promising therapeutic strategy. Front. Microbiol. 2023, 13, 1028086. [Google Scholar] [CrossRef]
  150. Bharti, S.; Kumar, A. Nanotechnology in Targeted Delivery of Antimicrobials and Overcoming Resistance. BioNanoScience 2025, 15, 1–22. [Google Scholar] [CrossRef]
  151. Arenas-Vivo, A.; Amariei, G.; Aguado, S.; Rosal, R.; Horcajada, P. An Ag-loaded photoactive nano-metal organic framework as a promising biofilm treatment. Acta Biomater. 2019, 97, 490–500. [Google Scholar] [CrossRef] [PubMed]
  152. Arenas-Vivo, A.; Celis Arias, V.; Amariei, G.; Rosal, R.; Izquierdo-Barba, I.; Hidalgo, T.; Vallet-Regí, M.; Beltrán, H.I.; Loera-Serna, S.; Horcajada, P. Antiadherent AgBDC metal–organic framework coating for escherichia coli biofilm inhibition. Pharmaceutics 2023, 15, 301. [Google Scholar] [CrossRef]
  153. Sadeghi Mohammadi, S.; Vaezi, Z.; Naderi-Manesh, H. Improvement of anti-biofilm activities via co-delivery of curcumin and gentamicin in lipid-polymer hybrid nanoparticle. J. Biomater. Sci. Polym. Ed. 2022, 33, 174–196. [Google Scholar] [CrossRef] [PubMed]
  154. Ahsan, A.; Thomas, N.; Barnes, T.J.; Subramaniam, S.; Loh, T.C.; Joyce, P.; Prestidge, C.A. Lipid nanocarriers-enabled delivery of antibiotics and antimicrobial adjuvants to overcome bacterial biofilms. Pharmaceutics 2024, 16, 396. [Google Scholar] [CrossRef]
  155. Rubio-Canalejas, A.; Baelo, A.; Herbera, S.; Blanco-Cabra, N.; Vukomanovic, M.; Torrents, E. 3D spatial organization and improved antibiotic treatment of a Pseudomonas aeruginosaStaphylococcus aureus wound biofilm by nanoparticle enzyme delivery. Front. Microbiol. 2022, 13, 959156. [Google Scholar] [CrossRef]
  156. Das, D.; Lin, C.-W.; Chuang, H.-S. LAMP-based point-of-care biosensors for rapid pathogen detection. Biosensors 2022, 12, 1068. [Google Scholar] [CrossRef]
  157. Kameswaran, S.; Gujjala, S.; Zhang, S.; Kondeti, S.; Mahalingam, S.; Bangeppagari, M.; Bellemkonda, R. Quenching and quorum sensing in bacterial bio-films. Res. Microbiol. 2024, 175, 104085. [Google Scholar] [CrossRef] [PubMed]
  158. Almatroudi, A. Biofilm resilience: Molecular mechanisms driving antibiotic resistance in clinical contexts. Biology 2025, 14, 165. [Google Scholar] [CrossRef]
  159. Polinarski, M.A.; Beal, A.L.; Silva, F.E.; Bernardi-Wenzel, J.; Burin, G.R.; de Muniz, G.I.; Alves, H.J. New perspectives of using chitosan, silver, and chitosan–silver nanoparticles against multidrug-resistant bacteria. Part. Part. Syst. Charact. 2021, 38, 2100009. [Google Scholar] [CrossRef]
  160. Laib, I.; Mohammed, H.A.; Laouini, S.E.; Bouafia, A.; Abdullah, M.M.; Al-Lohedan, H.A.; Al-Essa, Q.; Trzepieciński, T. Cutting-edge Nano-therapeutics: Silver nanoparticles loaded with ciprofloxacin for powerful antidiabetic, antioxidant, anti-inflammatory, and antibiotic action against resistant pathogenic bacteria. Int. J. Food Sci. Technol. 2025, 60, vvaf024. [Google Scholar] [CrossRef]
  161. Bhattacharya, P.; Sengupta, I.; Dey, A.; Chakraborty, S.; Neogi, S. Antibacterial effect of ciprofloxacin loaded reduced graphene oxide nanosheets against Pseudomonas aeruginosa strain. Colloid Interface Sci. Commun. 2021, 40, 100344. [Google Scholar] [CrossRef]
  162. Yun, J.; Kang, J.; Sharipov, M.; Ryu, W. Recent progress in hydrogel microneedle sensors based on electrochemical and optical sensing. Appl. Spectrosc. Rev. 2025, 1–41. [Google Scholar] [CrossRef]
  163. Li, B.; Mao, J.; Wu, J.; Mao, K.; Jia, Y.; Chen, F.; Liu, J. Nano–Bio Interactions: Biofilm-Targeted Antibacterial Nanomaterials. Small 2024, 20, 2306135. [Google Scholar] [CrossRef]
  164. Ma, S.; Moser, D.; Han, F.; Leonhard, M.; Schneider-Stickler, B.; Tan, Y. Preparation and antibiofilm studies of curcumin loaded chitosan nanoparticles against polymicrobial biofilms of Candida albicans and Staphylococcus aureus. Carbohydr. Polym. 2020, 241, 116254. [Google Scholar] [CrossRef]
  165. Kłodzińska, S.N.; Wan, F.; Jumaa, H.; Sternberg, C.; Rades, T.; Nielsen, H.M. Utilizing nanoparticles for improving anti-biofilm effects of azithromycin: A head-to-head comparison of modified hyaluronic acid nanogels and coated poly (lactic-co-glycolic acid) nanoparticles. J. Colloid Interface Sci. 2019, 555, 595–606. [Google Scholar] [CrossRef]
  166. Deacon, J.; Abdelghany, S.M.; Quinn, D.J.; Schmid, D.; Megaw, J.; Donnelly, R.F.; Jones, D.S.; Kissenpfennig, A.; Elborn, J.S.; Gilmore, B.F. Antimicrobial efficacy of tobramycin polymeric nanoparticles for Pseudomonas aeruginosa infections in cystic fibrosis: Formulation, characterisation and functionalisation with dornase alfa (DNase). J. Control. Release 2015, 198, 55–61. [Google Scholar] [CrossRef]
  167. Wang, C.; Zhao, W.; Cao, B.; Wang, Z.; Zhou, Q.; Lu, S.; Lu, L.; Zhan, M.; Hu, X. Biofilm-responsive polymeric nanoparticles with self-adaptive deep penetration for in vivo photothermal treatment of implant infection. Chem. Mater. 2020, 32, 7725–7738. [Google Scholar] [CrossRef]
  168. Ferreira, M.; Pinto, S.N.; Aires-da-Silva, F.; Bettencourt, A.; Aguiar, S.I.; Gaspar, M.M. Liposomes as a Nanoplatform to Improve the Delivery of Antibiotics into Staphylococcus aureus Biofilms. Pharmaceutics 2021, 13, 321. [Google Scholar] [CrossRef] [PubMed]
  169. Alfei, S.; Caviglia, D. Prevention and eradication of biofilm by dendrimers: A possibility still little explored. Pharmaceutics 2022, 14, 2016. [Google Scholar] [CrossRef] [PubMed]
  170. Osiro, K.O.; Hashemi, N.; Brango-Vanegas, J.; Oliveira, S.M.; Franco, O.L. Emerging peptide-based technology for biofilm control. Expert Opin. Biol. Ther. 2024, 24, 1311–1315. [Google Scholar] [CrossRef]
  171. de Lacerda Coriolano, D.; de Souza, J.B.; Bueno, E.V.; Medeiros, S.M.d.F.R.d.S.; Cavalcanti, I.D.L.; Cavalcanti, I.M.F. Antibacterial and antibiofilm potential of silver nanoparticles against antibiotic-sensitive and multidrug-resistant Pseudomonas aeruginosa strains. Braz. J. Microbiol. 2021, 52, 267–278. [Google Scholar] [CrossRef]
  172. Neihaya, H.; Zaman, H. Investigating the effect of biosynthesized silver nanoparticles as antibiofilm on bacterial clinical isolates. Microb. Pathog. 2018, 116, 200–208. [Google Scholar] [CrossRef]
  173. Hamida, R.S.; Ali, M.A.; Goda, D.A.; Khalil, M.I.; Redhwan, A. Cytotoxic effect of green silver nanoparticles against ampicillin-resistant Klebsiella pneumoniae. RSC Adv. 2020, 10, 21136–21146. [Google Scholar] [CrossRef]
  174. Žalnėravičius, R.; Mikalauskaitė, A.; Niaura, G.; Paškevičius, A.; Jagminas, A. Ultra-small methionine-capped Au0/Au+ nanoparticles as efficient drug against the antibiotic-resistant bacteria. Mater. Sci. Eng. C 2019, 102, 646–652. [Google Scholar] [CrossRef]
  175. Chen, J.; Dai, T.; Yu, J.; Dai, X.; Chen, R.; Wu, J.; Li, N.; Fan, L.; Mao, Z.; Sheng, G. Integration of antimicrobial peptides and gold nanorods for bimodal antibacterial applications. Biomater. Sci. 2020, 8, 4447–4457. [Google Scholar] [CrossRef]
  176. Dogra, V.; Kaur, G.; Jindal, S.; Kumar, R.; Kumar, S.; Singhal, N.K. Bactericidal effects of metallosurfactants based cobalt oxide/hydroxide nanoparticles against Staphylococcus aureus. Sci. Total Environ. 2019, 681, 350–364. [Google Scholar] [CrossRef]
  177. Safaei, M.; Taran, M.; Jamshidy, L.; Imani, M.M.; Mozaffari, H.R.; Sharifi, R.; Golshah, A.; Moradpoor, H. Optimum synthesis of polyhydroxybutyrate-Co3O4 bionanocomposite with the highest antibacterial activity against multidrug resistant bacteria. Int. J. Biol. Macromol. 2020, 158, 477–485. [Google Scholar] [CrossRef] [PubMed]
  178. Gabrielyan, L.; Hakobyan, L.; Hovhannisyan, A.; Trchounian, A. Effects of iron oxide (Fe3O4) nanoparticles on Escherichia coli antibiotic-resistant strains. J. Appl. Microbiol. 2019, 126, 1108–1116. [Google Scholar] [CrossRef] [PubMed]
  179. Arias, L.S.; Pessan, J.P.; de Souza Neto, F.N.; Lima, B.H.R.; de Camargo, E.R.; Ramage, G.; Delbem, A.C.B.; Monteiro, D.R. Novel nanocarrier of miconazole based on chitosan-coated iron oxide nanoparticles as a nanotherapy to fight Candida biofilms. Colloids Surf. B Biointerfaces 2020, 192, 111080. [Google Scholar] [CrossRef] [PubMed]
  180. Muzammil, S.; Khurshid, M.; Nawaz, I.; Siddique, M.H.; Zubair, M.; Nisar, M.A.; Imran, M.; Hayat, S. Aluminium oxide nanoparticles inhibit EPS production, adhesion and biofilm formation by multidrug resistant Acinetobacter baumannii. Biofouling 2020, 36, 492–504. [Google Scholar] [CrossRef]
  181. Palau Gauthier, M.; Muñoz, E.; Gustà, M.F.; Larrosa, M.N.; Gomis, X.; Gilabert, J.; Almirante Gragera, B.; Puntes, V.; Texidó, R.; Gavaldà, J. In Vitro Antibacterial Activity of Silver Nanoparticles Conjugated with Amikacin and Combined with Hyperthermia against Drug-Resistant and Biofilm-Producing Strains. Microbiol. Spectr. 2023, 11, e0028023. [Google Scholar]
  182. Thorn, C.R.; Kopecki, Z.; Wignall, A.; Kral, A.; Prestidge, C.A.; Thomas, N. Liquid crystal nanoparticle platform for increased efficacy of cationic antimicrobials against biofilm infections. Nanomed. Nanotechnol. Biol. Med. 2022, 42, 102536. [Google Scholar] [CrossRef]
  183. Wu, X.; Qi, M.; Liu, C.; Yang, Q.; Li, S.; Shi, F.; Sun, X.; Wang, L.; Li, C.; Dong, B. Near-infrared light-triggered nitric oxide nanocomposites for photodynamic/photothermal complementary therapy against periodontal biofilm in an animal model. Theranostics 2023, 13, 2350. [Google Scholar] [CrossRef]
  184. Kim, M.-H. Nanoparticle-based therapies for wound biofilm infection: Opportunities and challenges. IEEE Trans. Nanobiosci. 2016, 15, 294–304. [Google Scholar] [CrossRef]
  185. Elabbasy, M.T.; El Bayomi, R.M.; Abdelkarim, E.A.; Hafez, A.E.-S.E.; Othman, M.S.; Ghoniem, M.E.; Samak, M.A.; Alshammari, M.H.; Almarshadi, F.A.; Elsamahy, T. Antibacterial and Antibiofilm Activity of Green-Synthesized Zinc Oxide Nanoparticles Against Multidrug-Resistant Escherichia coli Isolated from Retail Fish. Molecules 2025, 30, 768. [Google Scholar] [CrossRef] [PubMed]
  186. Wang, P.; Zeng, Y.; Liu, J.; Wang, L.; Yang, M.; Zhou, J. Antimicrobial and anti-biofilm effects of dihydroartemisinin-loaded chitosan nanoparticles against methicillin-resistant Staphylococcus aureus. Microb. Pathog. 2025, 199, 107208. [Google Scholar] [CrossRef]
  187. Rostamnejad, D.; Esnaashari, F.; Zahmatkesh, H.; Rasti, B.; Zamani, H. Diclofenac-loaded PLGA nanoparticles downregulate LasI/R quorum sensing genes in pathogenic P. aeruginosa isolates. Arch. Microbiol. 2024, 206, 112. [Google Scholar] [CrossRef]
  188. Liu, X.; Xu, S.; Chen, X.; Kang, S.; Liu, J.; Jiang, Q.; Zhang, S.; Hao, L.; Ran, H.; Huang, W. Synergistic effect of bovine cateslytin-loaded nanoparticles combined with ultrasound against Candida albicans biofilm. Future Med. Chem. 2023, 15, 43–55. [Google Scholar] [CrossRef] [PubMed]
  189. Ali, A.; Ovais, M.; Cui, X.; Rui, Y.; Chen, C. Safety assessment of nanomaterials for antimicrobial applications. Chem. Res. Toxicol. 2020, 33, 1082–1109. [Google Scholar] [CrossRef]
  190. Bereanu, A.-S.; Vintilă, B.I.; Bereanu, R.; Codru, I.R.; Hașegan, A.; Olteanu, C.; Săceleanu, V.; Sava, M. TiO2 Nanocomposite Coatings and Inactivation of Carbapenemase-Producing Klebsiella Pneumoniae Biofilm—Opportunities and Challenges. Microorganisms 2024, 12, 684. [Google Scholar] [CrossRef]
  191. Maksimova, Y.; Pyankova, E.; Nesterova, L.; Maksimov, A. Effects of Graphene Derivatives and Near-Infrared Laser Irradiation on E. coli Biofilms and Stress Response Gene Expression. Int. J. Mol. Sci. 2025, 26, 4728. [Google Scholar] [CrossRef]
  192. Reymond, J.-L.; Bergmann, M.; Darbre, T. Glycopeptide dendrimers as Pseudomonas aeruginosa biofilm inhibitors. Chem. Soc. Rev. 2013, 42, 4814–4822. [Google Scholar] [CrossRef]
  193. Backlund, C.J.; Worley, B.V.; Schoenfisch, M.H. Anti-biofilm action of nitric oxide-releasing alkyl-modified poly (amidoamine) dendrimers against Streptococcus mutans. Acta Biomater. 2016, 29, 198–205. [Google Scholar] [CrossRef]
  194. Nabawy, A.; Makabenta, J.M.; Schmidt-Malan, S.; Park, J.; Li, C.-H.; Huang, R.; Fedeli, S.; Chattopadhyay, A.N.; Patel, R.; Rotello, V.M. Dual antimicrobial-loaded biodegradable nanoemulsions for synergistic treatment of wound biofilms. J. Control. Release 2022, 347, 379–388. [Google Scholar] [CrossRef] [PubMed]
  195. Li, C.-H.; Landis, R.F.; Makabenta, J.M.; Nabawy, A.; Tronchet, T.; Archambault, D.; Liu, Y.; Huang, R.; Golan, M.; Cui, W. Nanotherapeutics using all-natural materials. Effective treatment of wound biofilm infections using crosslinked nanoemulsions. Mater. Horiz. 2021, 8, 1776–1782. [Google Scholar] [CrossRef]
  196. Hu, W.; Ouyang, Q.; Jiang, C.; Huang, S.; Alireza, N.-E.; Guo, D.; Liu, J.; Peng, Y. Biomedical Metal–Organic framework materials on antimicrobial therapy: Perspectives and challenges. Mater. Today Chem. 2024, 41, 102300. [Google Scholar] [CrossRef]
  197. Dastneshan, A.; Rahiminezhad, S.; Mezajin, M.N.; Jevinani, H.N.; Akbarzadeh, I.; Abdihaji, M.; Qahremani, R.; Jahanbakhshi, M.; Lalami, Z.A.; Heydari, H. 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. [Google Scholar] [CrossRef]
  198. Hamza, A. Sensing Platform based on Metal Organic Framework for the Quantitative Determination of Antibiotic Remains in Animal Derived Food. Chemistry Department COMSATS university Islamabad Lahore Campus. Doctoral Dissertation, COMSATS University Islamabad, Islamabad, Pakistan, 2024. [Google Scholar]
  199. Kumar, P.; Behera, A.; Tiwari, P.; Karthik, S.; Biswas, M.; Sonawane, A.; Mobin, S.M. Exploring the antimicrobial potential of isoniazid loaded Cu-based metal–organic frameworks as a novel strategy for effective killing of Mycobacterium tuberculosis. J. Mater. Chem. B 2023, 11, 10929–10940. [Google Scholar] [CrossRef] [PubMed]
  200. Gurunathan, S.; Thangaraj, P.; Das, J.; Kim, J.-H. Antibacterial and antibiofilm effects of Pseudomonas aeruginosa derived outer membrane vesicles against Streptococcus mutans. Heliyon 2023, 9, e22606. [Google Scholar] [CrossRef]
  201. Martins, P.; Machado, D.; Theizen, T.H.; Guarnieri, J.P.O.; Bernardes, B.G.; Gomide, G.P.; Corat, M.A.F.; Abbehausen, C.; Módena, J.L.P.; Melo, C.F.O.R. Outer membrane vesicles from Neisseria meningitidis (Proteossome) used for nanostructured Zika virus vaccine production. Sci. Rep. 2018, 8, 8290. [Google Scholar] [CrossRef] [PubMed]
  202. Ahmed, A.A.Q.; Besio, R.; Xiao, L.; Forlino, A. Outer membrane vesicles (OMVs) as biomedical tools and their relevance as immune-modulating agents against H. pylori infections: Current status and future prospects. Int. J. Mol. Sci. 2023, 24, 8542. [Google Scholar] [CrossRef]
  203. Xerri, N.L.; Payne, S.M. Bacteroides thetaiotaomicron outer membrane vesicles modulate virulence of Shigella flexneri. MBio 2022, 13, e02360-22. [Google Scholar] [CrossRef]
  204. de Freitas, M.C.R.; de Almeida, P.E.; Vieira, W.V.; Ferreira-Machado, A.B.; Resende, J.A.; da Silva, V.L.; Diniz, C.G. Inflammatory modulation and outer membrane vesicles (OMV) production associated to Bacteroides fragilis response to subinhibitory concentrations of metronidazole during experimental infection. Anaerobe 2022, 73, 102504. [Google Scholar] [CrossRef]
  205. Zhou, Q.; Wang, T.; Li, K.; Zhang, S.; Wang, K.; Hong, W.; Liu, R.; Li, P. Biofilm microenvironment-responsive polymeric CO releasing micelles for enhanced amikacin efficacy. J. Control. Release 2023, 357, 561–571. [Google Scholar] [CrossRef]
  206. Albayaty, Y.N.; Thomas, N.; Ramirez-Garcia, P.D.; Davis, T.P.; Quinn, J.F.; Whittaker, M.R.; Prestidge, C.A. Polymeric micelles with anti-virulence activity against Candida albicans in a single-and dual-species biofilm. Drug Deliv. Transl. Res. 2021, 11, 1586–1597. [Google Scholar] [CrossRef] [PubMed]
  207. Quan, K.; Zhang, Z.; Ren, Y.; Busscher, H.J.; Van Der Mei, H.C.; Peterson, B.W. Homogeneous distribution of magnetic, antimicrobial-carrying nanoparticles through an infectious biofilm enhances biofilm-killing efficacy. ACS Biomater. Sci. Eng. 2019, 6, 205–212. [Google Scholar] [CrossRef]
  208. El-Khawaga, A.M.; Ayman, M.; Hafez, O.; Shalaby, R.E. Photocatalytic, antimicrobial and antibiofilm activities of MgFe2O4 magnetic nanoparticles. Sci. Rep. 2024, 14, 12877. [Google Scholar] [CrossRef] [PubMed]
  209. Esfahani, M.B.; Khodavandi, A.; Alizadeh, F.; Bahador, N. Antibacterial and anti-biofilm activities of microbial synthesized silver and magnetic iron oxide nanoparticles against Pseudomonas aeruginosa. IEEE Trans. NanoBioscience 2023, 22, 956–966. [Google Scholar] [CrossRef]
  210. Raulio, M.; Pore, V.; Areva, S.; Ritala, M.; Leskelä, M.; Lindén, M.; Rosenholm, J.B.; Lounatmaa, K.; Salkinoja-Salonen, M. Destruction of Deinococcus geothermalis biofilm by photocatalytic ALD and sol-gel TiO2 surfaces. J. Ind. Microbiol. Biotechnol. 2006, 33, 261–268. [Google Scholar] [CrossRef]
  211. Tao, L. Synthesis and Characterizations of Antimicrobial Coating by Atomic Layer Deposition Technique for Personal Protective Equipment Applications. Doctoral Dissertation, University of British Columbia, Vancouver, BC, Canada, 2022. [Google Scholar]
  212. Xin, H.; Liu, Y.; Xiao, Y.; Wen, M.; Sheng, L.; Jia, Z. Design and nanoengineering of photoactive antimicrobials for bioapplications: From fundamentals to advanced strategies. Adv. Funct. Mater. 2024, 34, 2402607. [Google Scholar] [CrossRef]
  213. Buret, A.G.; Allain, T.; Motta, J.-P.; Wallace, J.L. Effects of hydrogen sulfide on the microbiome: From toxicity to therapy. Antioxid. Redox Signal. 2022, 36, 211–219. [Google Scholar] [CrossRef]
  214. Sethulekshmi, A.; Saritha, A.; Joseph, K.; Aprem, A.S.; Sisupal, S.B. MoS2 based nanomaterials: Advanced antibacterial agents for future. J. Control. Release 2022, 348, 158–185. [Google Scholar] [CrossRef]
  215. Tariq, F.; Hussain, R.; Noreen, Z.; Javed, A.; Shah, A.; Mahmood, A.; Sajjad, M.; Bokhari, H.; ur Rahman, S. Enhanced antibacterial activity of visible light activated sulfur-doped TiO2 nanoparticles against Vibrio cholerae. Mater. Sci. Semicond. Process. 2022, 147, 106731. [Google Scholar] [CrossRef]
  216. Gao, F.; Shao, T.; Yu, Y.; Xiong, Y.; Yang, L. Surface-bound reactive oxygen species generating nanozymes for selective antibacterial action. Nat. Commun. 2021, 12, 745. [Google Scholar] [CrossRef] [PubMed]
  217. Wang, S.; Fang, L.; Zhou, H.; Wang, M.; Zheng, H.; Wang, Y.; Weir, M.D.; Masri, R.; Oates, T.W.; Cheng, L. Silica nanoparticles containing nano-silver and chlorhexidine respond to pH to suppress biofilm acids and modulate biofilms toward a non-cariogenic composition. Dent. Mater. 2024, 40, 179–189. [Google Scholar] [CrossRef]
  218. Ji, H.; Hu, H.; Tang, Q.; Kang, X.; Liu, X.; Zhao, L.; Jing, R.; Wu, M.; Li, G.; Zhou, X. Precisely controlled and deeply penetrated micro-nano hybrid multifunctional motors with enhanced antibacterial activity against refractory biofilm infections. J. Hazard. Mater. 2022, 436, 129210. [Google Scholar] [CrossRef]
  219. Hemmati, J.; Nazari, M.; Abolhasani, F.S.; Ahmadi, A.; Asghari, B. In vitro investigation of relationship between quorum-sensing system genes, biofilm forming ability, and drug resistance in clinical isolates of Pseudomonas aeruginosa. BMC Microbiol. 2024, 24, 99. [Google Scholar] [CrossRef] [PubMed]
  220. Neil, B.; Cheney, G.L.; Rosenzweig, J.A.; Sha, J.; Chopra, A.K. Antimicrobial resistance in aeromonads and new therapies targeting quorum sensing. Appl. Microbiol. Biotechnol. 2024, 108, 205. [Google Scholar] [CrossRef]
  221. Kachhadiya, D.K.; Georrge, J.J. Quorum Sensing and its Inhibition in Pseudomonas aeruginosa: Molecular Targets and Mode of Action. Curr. Drug Targets 2025, 26, 613–631. [Google Scholar] [CrossRef] [PubMed]
  222. He, Y.-W.; Deng, Y.; Miao, Y.; Chatterjee, S.; Tran, T.M.; Tian, J.; Lindow, S. DSF-family quorum sensing signal-mediated intraspecies, interspecies, and inter-kingdom communication. Trends Microbiol. 2023, 31, 36–50. [Google Scholar] [CrossRef] [PubMed]
  223. Tuan, D.A.; Uyen, P.V.N.; Masak, J. Hybrid quorum sensing and machine learning systems for adaptive synthetic biology: Toward autonomous gene regulation and precision therapies. Preprint 2024. [Google Scholar] [CrossRef]
  224. Tu, Y.; Li, H.; Huo, J.; Gou, L.; Wen, X.; Yu, X.; Zhang, X.; Zeng, J.; Li, Y. Disrupting the bacterial language: Quorum quenching and its applications. Crit. Rev. Microbiol. 2025, 1–15. [Google Scholar] [CrossRef]
  225. Efremenko, E.; Aslanli, A.; Domnin, M.; Stepanov, N.; Senko, O. Enzymes with lactonase activity against fungal quorum molecules as effective antifungals. Biomolecules 2024, 14, 383. [Google Scholar] [CrossRef]
  226. Bravo, M.; Conchillo-Solé, Ò.; Coves, X.; García-Navarro, A.; Gómez, A.-C.; Márquez-Martínez, M.; Ferrer-Miralles, N.; Daura, X.; Gibert, I.; Yero, D. An acyl-homoserine lactone acylase found in Stenotrophomonas maltophilia exhibits both quorum quenching activity and the ability to degrade penicillin antibiotics. Sci. Rep. 2025, 15, 8557. [Google Scholar] [CrossRef]
  227. Zhang, C.; Li, C.; Aziz, T.; Alharbi, M.; Cui, H.; Lin, L. Screening of E. coli O157: H7 AI-2 QS inhibitors and their inhibitory effect on biofilm formation in combination with disinfectants. Food Biosci. 2024, 58, 103821. [Google Scholar] [CrossRef]
  228. Kang, S.; Kim, J.; Hur, J.K.; Lee, S.-S. CRISPR-based genome editing of clinically important Escherichia coli SE15 isolated from indwelling urinary catheters of patients. J. Med. Microbiol. 2017, 66, 18–25. [Google Scholar] [CrossRef]
  229. Arruda-Silva, F.; Bianchetto-Aguilera, F.; Gasperini, S.; Polletti, S.; Cosentino, E.; Tamassia, N.; Cassatella, M.A. Human neutrophils produce CCL23 in response to various TLR-agonists and TNFα. Front. Cell. Infect. Microbiol. 2017, 7, 176. [Google Scholar] [CrossRef]
  230. Kumar, L.; Chhibber, S.; Kumar, R.; Kumar, M.; Harjai, K. Zingerone silences quorum sensing and attenuates virulence of Pseudomonas aeruginosa. Fitoterapia 2015, 102, 84–95. [Google Scholar] [CrossRef]
  231. Gökalsın, B.; Aksoydan, B.; Erman, B.; Sesal, N.C. Reducing virulence and biofilm of Pseudomonas aeruginosa by potential quorum sensing inhibitor carotenoid: Zeaxanthin. Microb. Ecol. 2017, 74, 466–473. [Google Scholar] [CrossRef]
  232. Baldry, M.; Kitir, B.; Frøkiær, H.; Christensen, S.B.; Taverne, N.; Meijerink, M.; Franzyk, H.; Olsen, C.A.; Wells, J.M.; Ingmer, H. The agr inhibitors solonamide B and analogues alter immune responses to Staphylococccus aureus but do not exhibit adverse effects on immune cell functions. PLoS ONE 2016, 11, e0145618. [Google Scholar] [CrossRef] [PubMed]
  233. Paczkowski, J.E.; Mukherjee, S.; McCready, A.R.; Cong, J.-P.; Aquino, C.J.; Kim, H.; Henke, B.R.; Smith, C.D.; Bassler, B.L. Flavonoids suppress Pseudomonas aeruginosa virulence through allosteric inhibition of quorum-sensing receptors. J. Biol. Chem. 2017, 292, 4064–4076. [Google Scholar] [CrossRef] [PubMed]
  234. Yang, T.; Tal-Gan, Y.; Paharik, A.E.; Horswill, A.R.; Blackwell, H.E. Structure–function analyses of a Staphylococcus epidermidis autoinducing peptide reveals motifs critical for AgrC-type receptor modulation. ACS Chem. Biol. 2016, 11, 1982–1991. [Google Scholar] [CrossRef]
  235. Zhang, Y.; Sass, A.; Van Acker, H.; Wille, J.; Verhasselt, B.; Van Nieuwerburgh, F.; Kaever, V.; Crabbé, A.; Coenye, T. Coumarin reduces virulence and biofilm formation in Pseudomonas aeruginosa by affecting quorum sensing, type III secretion and C-di-GMP levels. Front. Microbiol. 2018, 9, 1952. [Google Scholar] [CrossRef] [PubMed]
  236. Amara, N.; Gregor, R.; Rayo, J.; Dandela, R.; Daniel, E.; Liubin, N.; Willems, H.M.E.; Ben-Zvi, A.; Krom, B.P.; Meijler, M.M. Fine-Tuning Covalent Inhibition of Bacterial Quorum Sensing. ChemBioChem 2016, 17, 825–835. [Google Scholar] [CrossRef]
  237. Heidari, A.; Noshiranzadeh, N.; Haghi, F.; Bikas, R. Inhibition of quorum sensing related virulence factors of Pseudomonas aeruginosa by pyridoxal lactohydrazone. Microb. Pathog. 2017, 112, 103–110. [Google Scholar] [CrossRef] [PubMed]
  238. Abbas, H.A.; Shaldam, M.A. Glyceryl trinitrate is a novel inhibitor of quorum sensing in Pseudomonas aeruginosa. Afr. Health Sci. 2016, 16, 1109–1117. [Google Scholar] [CrossRef] [PubMed]
  239. Kim, B.; Park, J.-S.; Choi, H.-Y.; Yoon, S.S.; Kim, W.-G. Terrein is an inhibitor of quorum sensing and c-di-GMP in Pseudomonas aeruginosa: A connection between quorum sensing and c-di-GMP. Sci. Rep. 2018, 8, 8617. [Google Scholar] [CrossRef]
  240. Goh, W.-K.; Gardner, C.R.; Sekhar, K.V.C.; Biswas, N.N.; Nizalapur, S.; Rice, S.A.; Willcox, M.; Black, D.S.; Kumar, N. Synthesis, quorum sensing inhibition and docking studies of 1, 5-dihydropyrrol-2-ones. Bioorganic Med. Chem. 2015, 23, 7366–7377. [Google Scholar] [CrossRef]
  241. Kalia, M.; Yadav, V.K.; Singh, P.K.; Sharma, D.; Narvi, S.S.; Agarwal, V. Exploring the impact of parthenolide as anti-quorum sensing and anti-biofilm agent against Pseudomonas aeruginosa. Life Sci. 2018, 199, 96–103. [Google Scholar] [CrossRef]
  242. Scoffone, V.C.; Chiarelli, L.R.; Makarov, V.; Brackman, G.; Israyilova, A.; Azzalin, A.; Forneris, F.; Riabova, O.; Savina, S.; Coenye, T. Discovery of new diketopiperazines inhibiting Burkholderia cenocepacia quorum sensing in vitro and in vivo. Sci. Rep. 2016, 6, 32487. [Google Scholar] [CrossRef]
  243. Teng, Z.; Shi, D.; Liu, H.; Shen, Z.; Zha, Y.; Li, W.; Deng, X.; Wang, J. Lysionotin attenuates Staphylococcus aureus pathogenicity by inhibiting α-toxin expression. Appl. Microbiol. Biotechnol. 2017, 101, 6697–6703. [Google Scholar] [CrossRef]
  244. Ren, S.; Wang, B.; Qiu, X.; Wang, S.; Huang, G.; Wang, Y. Unveiling Antibacterial and Antibiofilm Mechanisms of Methyleugenol: Implications for Ecomaterial Functionalization. ACS EST Eng. 2024, 4, 1206–1217. [Google Scholar] [CrossRef]
  245. Vieira, T.F.; Leitão, M.M.; Cerqueira, N.M.; Sousa, S.F.; Borges, A.; Simões, M. Montelukast and cefoperazone act as antiquorum sensing and antibiofilm agents against Pseudomonas aeruginosa. J. Appl. Microbiol. 2024, 135, lxae088. [Google Scholar] [CrossRef]
  246. Jakobsen, T.H.; van Gennip, M.; Phipps, R.K.; Shanmugham, M.S.; Christensen, L.D.; Alhede, M.; Skindersoe, M.E.; Rasmussen, T.B.; Friedrich, K.; Uthe, F. Ajoene, a sulfur-rich molecule from garlic, inhibits genes controlled by quorum sensing. Antimicrob. Agents Chemother. 2012, 56, 2314–2325. [Google Scholar] [CrossRef] [PubMed]
  247. Yang, W.; Wei, Q.; Tong, Q.; Cui, K.; He, G.; Lin, L.; Ma, L.Z.; Cornelis, P.; Wang, Y. Traditional Chinese medicine Tanreqing inhibits quorum sensing systems in Pseudomonas aeruginosa. Front. Microbiol. 2020, 11, 517462. [Google Scholar] [CrossRef] [PubMed]
  248. Whiteley, M.; Diggle, S.P.; Greenberg, E.P. Bacterial quorum sensing: The progress and promise of an emerging research area. Nature 2017, 551, 313. [Google Scholar] [CrossRef]
  249. Kai, K. The phc quorum-sensing system in Ralstonia solanacearum species complex. Annu. Rev. Microbiol. 2023, 77, 213–231. [Google Scholar] [CrossRef]
  250. Yoshihara, A.; Shimatani, M.; Sakata, M.; Takemura, C.; Senuma, W.; Hikichi, Y.; Kai, K. Quorum sensing inhibition attenuates the virulence of the plant pathogen Ralstonia solanacearum species complex. ACS Chem. Biol. 2020, 15, 3050–3059. [Google Scholar] [CrossRef]
  251. Wang, H.; Liao, L.; Chen, S.; Zhang, L.-H. A quorum quenching bacterial isolate contains multiple substrate-inducible genes conferring degradation of diffusible signal factor. Appl. Environ. Microbiol. 2020, 86, e02930-19. [Google Scholar] [CrossRef]
  252. Nazzaro, F.; Fratianni, F.; d’Acierno, A.; Coppola, R.; Ayala-Zavala, F.J.; da Cruz, A.G.; De Feo, V. Essential oils and microbial communication. In Essential Oils-Oils of Nature; IntechOpen: Rijeka, Croatia, 2019. [Google Scholar]
  253. Ribeiro, T.A.N.; Dos Santos, G.A.; Dos Santos, C.T.; Soares, D.C.F.; Saraiva, M.F.; Leal, D.H.S.; Sachs, D. Eugenol as a promising antibiofilm and anti-quorum sensing agent: A systematic review. Microb. Pathog. 2024, 196, 106937. [Google Scholar] [CrossRef]
  254. Kameswaran, S.; Ramesh, B. Natural QSIs for Biofilm Control in Pathogenic Bacteria. In Quorum Quenching; Elsevier: Amsterdam, The Netherlands, 2023. [Google Scholar]
  255. Mokoena, M.P.; Omatola, C.A.; Olaniran, A.O. Applications of lactic acid bacteria and their bacteriocins against food spoilage microorganisms and foodborne pathogens. Molecules 2021, 26, 7055. [Google Scholar] [CrossRef]
  256. Sadik, A.; Viswaswar, J.P.; Rajamoney, A.; Rekha, A.; Raj, D.M.; Prakashan, D.; Vasudevan, M.; Visakh, J.; Renuka, D.; Hely, S. Mitigation of quorum sensing mediated virulence factors of Pseudomonas aeruginosa: The role of Meldrum’s acid activated furan. Front. Microbiol. 2024, 14, 1272240. [Google Scholar] [CrossRef] [PubMed]
  257. Angelini, P. Plant-derived antimicrobials and their crucial role in combating antimicrobial resistance. Antibiotics 2024, 13, 746. [Google Scholar] [CrossRef]
  258. Davares, A.K.; Arsene, M.M.; Viktorovna, P.I.; Vyacheslavovna, Y.N.; Vladimirovna, Z.A.; Aleksandrovna, V.E.; Nikolayevich, S.A.; Nadezhda, S.; Anatolievna, G.O.; Nikolaevna, S.I. Quorum-sensing inhibitors from probiotics as a strategy to combat bacterial cell-to-cell communication involved in food spoilage and food safety. Fermentation 2022, 8, 711. [Google Scholar] [CrossRef]
  259. Antonioli, L.; Haskó, G. May be adenosine an immuno-quorum-sensing signal? Purinergic Signal. 2022, 18, 205–209. [Google Scholar] [CrossRef] [PubMed]
  260. Deng, Z.; Luo, X.M.; Liu, J.; Wang, H. Quorum sensing, biofilm, and intestinal mucosal barrier: Involvement the role of probiotic. Front. Cell. Infect. Microbiol. 2020, 10, 538077. [Google Scholar] [CrossRef] [PubMed]
  261. Zhang, H.; Zhang, Z.; Li, J.; Qin, G. New strategies for biocontrol of bacterial toxins and virulence: Focusing on quorum-sensing interference and biofilm inhibition. Toxins 2023, 15, 570. [Google Scholar] [CrossRef]
  262. Arya, S.; Usha, R. Bioprospecting and Exploration of Phytochemicals as Quorum Sensing Inhibitors against Cariogenic Dental Biofilm. J. Pure Appl. Microbiol. 2024, 18, 100–117. [Google Scholar] [CrossRef]
  263. Raheem, A.; Liang, L.; Zhang, G.; Cui, S. Modulatory effects of probiotics during pathogenic infections with emphasis on immune regulation. Front. Immunol. 2021, 12, 616713. [Google Scholar] [CrossRef]
  264. Stephens, K.; Bentley, W.E. Synthetic biology for manipulating quorum sensing in microbial consortia. Trends Microbiol. 2020, 28, 633–643. [Google Scholar] [CrossRef]
  265. Gunaratnam, S.; Millette, M.; McFarland, L.V.; DuPont, H.L.; Lacroix, M. Potential role of probiotics in reducing Clostridioides difficile virulence: Interference with quorum sensing systems. Microb. Pathog. 2021, 153, 104798. [Google Scholar] [CrossRef]
  266. Wang, Y.; Dai, J.; Wang, X.; Wang, Y.; Tang, F. Mechanisms of interactions between bacteria and bacteriophage mediate by quorum sensing systems. Appl. Microbiol. Biotechnol. 2022, 106, 2299–2310. [Google Scholar] [CrossRef] [PubMed]
  267. Huedo, P.; Coves, X.; Daura, X.; Gibert, I.; Yero, D. Quorum sensing signaling and quenching in the multidrug-resistant pathogen Stenotrophomonas maltophilia. Front. Cell. Infect. Microbiol. 2018, 8, 122. [Google Scholar] [CrossRef]
  268. Wijesinghe, G.; Nobbs, A.; Bandara, H. The diffusible signaling factor family in microbial signaling: A current perspective. Crit. Rev. Microbiol. 2025, 1–16. [Google Scholar] [CrossRef]
  269. Horgan, C.; Baccari, C.; O’Driscoll, M.; Lindow, S.E.; O’Sullivan, T.P. BDSF Analogues Inhibit Quorum Sensing-Regulated Biofilm Production in Xylella fastidiosa. Microorganisms 2024, 12, 2496. [Google Scholar] [CrossRef]
  270. Zhang, Y.; Bhasme, P.; Reddy, D.S.; Liu, D.J.; Yu, Z.X.; Zhao, T.H.; Zheng, Y.Q.; Kumar, A.; Yu, H.Y.; Ma, L.Z. Dual functions: A coumarin-chalcone conjugate inhibits cyclic-di-GMP and quorum-sensing signaling to reduce biofilm formation and virulence of pathogens. MLife 2023, 2, 283–294. (In English) [Google Scholar] [CrossRef]
  271. Smith, P.; Schuster, M. Antiactivators prevent self-sensing in Pseudomonas aeruginosa quorum sensing. Proc. Natl. Acad. Sci. USA 2022, 119, e2201242119. [Google Scholar] [CrossRef]
  272. Ren, Y.; Zhu, R.; You, X.; Li, D.; Guo, M.; Fei, B.; Liu, Y.; Yang, X.; Liu, X.; Li, Y. Quercetin: A promising virulence inhibitor of Pseudomonas aeruginosa LasB in vitro. Appl. Microbiol. Biotechnol. 2024, 108, 57. [Google Scholar] [CrossRef]
  273. Yin, L.; Wang, Y.; Xiang, S.; Xu, K.; Wang, B.; Jia, A.-Q. Tyramine, one quorum sensing inhibitor, reduces pathogenicity and restores tetracycline susceptibility in Burkholderia cenocepacia. Biochem. Pharmacol. 2023, 218, 115906. [Google Scholar] [CrossRef] [PubMed]
  274. Kim, B.S.; Jang, S.Y.; Bang, Y.-j.; Hwang, J.; Koo, Y.; Jang, K.K.; Lim, D.; Kim, M.H.; Choi, S.H. QStatin, a selective inhibitor of quorum sensing in Vibrio species. MBio 2018, 9, e02262-17. [Google Scholar] [CrossRef]
  275. Kalia, V.C.; Patel, S.K.; Kang, Y.C.; Lee, J.-K. Quorum sensing inhibitors as antipathogens: Biotechnological applications. Biotechnol. Adv. 2019, 37, 68–90. [Google Scholar] [CrossRef]
  276. Zeng, L.; Lin, F.; Ling, B. Effect of traditional Chinese medicine monomers interfering with quorum-sensing on virulence factors of extensively drug-resistant Acinetobacter baumannii. Front. Pharmacol. 2023, 14, 1135180. [Google Scholar] [CrossRef] [PubMed]
  277. Woods, K.E.; Akhter, S.; Rodriguez, B.; Townsend, K.A.; Smith, N.; Smith, B.; Wambua, A.; Craddock, V.; Abisado-Duque, R.G.; Santa, E.E. Characterization of natural product inhibitors of quorum sensing reveals competitive inhibition of Pseudomonas aeruginosa RhlR by ortho-vanillin. Microbiol. Spectr. 2024, 12, e0068124. [Google Scholar] [CrossRef] [PubMed]
  278. Bové, M.; Bao, X.; Sass, A.; Crabbé, A.; Coenye, T. The quorum-sensing inhibitor furanone C-30 rapidly loses its tobramycin-potentiating activity against Pseudomonas aeruginosa biofilms during experimental evolution. Antimicrob. Agents Chemother. 2021, 65, e0041321. [Google Scholar] [CrossRef]
  279. Hu, H.; Hua, S.Y.; Lin, X.; Lu, F.; Zhang, W.; Zhou, L.; Cui, J.; Wang, R.; Xia, J.; Xu, F. Hybrid biomimetic membrane coated particles-mediated bacterial ferroptosis for acute MRSA pneumonia. ACS Nano 2023, 17, 11692–11712. [Google Scholar] [CrossRef]
  280. Jakobsen, T.H.; Warming, A.N.; Vejborg, R.M.; Moscoso, J.A.; Stegger, M.; Lorenzen, F.; Rybtke, M.; Andersen, J.B.; Petersen, R.; Andersen, P.S.; et al. A Broad Range Quorum Sensing Inhibitor Working Through sRNA Inhibition. Sci. Rep. 2017, 7, 9857. [Google Scholar] [CrossRef]
  281. Mane, S.G.; Katagi, K.S.; Bhasme, P.; Pattar, S.; Wei, Q.; Joshi, S.D. Design, synthesis, antibiofilm, quorum sensing inhibition, anticancer and docking studies of novel 2-(4-acridine-9-ylamino)isoindoline-1,3-dione. Chem. Data Collect. 2019, 20, 100198. [Google Scholar] [CrossRef]
  282. São-José, C.; Costa, A.R.; Melo, L.D. Bacteriophages and their lytic enzymes as alternative antibacterial therapies in the age of antibiotic resistance. Front. Microbiol. 2022, 13, 884176. [Google Scholar] [CrossRef] [PubMed]
  283. Hassannia, M.; Naderifar, M.; Salamy, S.; Akbarizadeh, M.R.; Mohebi, S.; Moghadam, M.T. Engineered phage enzymes against drug-resistant pathogens: A review on advances and applications. Bioprocess Biosyst. Eng. 2024, 47, 301–312. [Google Scholar] [CrossRef]
  284. Olatunji, A.O.; Olaboye, J.A.; Maha, C.C.; Kolawole, T.O.; Abdul, S. Next-Generation strategies to combat antimicrobial resistance: Integrating genomics, CRISPR, and novel therapeutics for effective treatment. Eng. Sci. Technol. J. 2024, 5, 2284–2303. [Google Scholar] [CrossRef]
  285. Gliźniewicz, M.; Miłek, D.; Olszewska, P.; Czajkowski, A.; Serwin, N.; Cecerska-Heryć, E.; Dołęgowska, B.; Grygorcewicz, B. Advances in bacteriophage-mediated strategies for combating polymicrobial biofilms. Front. Microbiol. 2024, 14, 1320345. [Google Scholar] [CrossRef]
  286. Peng, J.; Guo, C.; Yang, C.; Zhang, L.; Yang, F.; Huang, X.; Yu, Y.; Zhang, T.; Peng, J. Phage therapy for bone and joint infections: A comprehensive exploration of challenges, dynamics, and therapeutic prospects. J. Glob. Antimicrob. Resist. 2024, 39, 12–21. [Google Scholar] [CrossRef]
  287. Srivastava, A.; Verma, N.; Kumar, V.; Apoorva, P.; Agarwal, V. Biofilm inhibition/eradication: Exploring strategies and confronting challenges in combatting biofilm. Arch. Microbiol. 2024, 206, 212. [Google Scholar] [CrossRef]
  288. Panthi, V.K.; Fairfull-Smith, K.E.; Islam, N. Liposomal drug delivery strategies to eradicate bacterial biofilms: Challenges, recent advances, and future perspectives. Int. J. Pharm. 2024, 655, 124046. [Google Scholar] [CrossRef]
  289. Lordelo, R.; Branco, R.; Gama, F.; Morais, P.V. Assessment of antimicrobial resistance, biofilm formation, and surface modification potential in hospital strains of Pseudomonas aeruginosa and Klebsiella pneumoniae. Heliyon 2024, 10, e30464. [Google Scholar] [CrossRef] [PubMed]
  290. Aerts, E. AI-Guided Detection of Antibiotic-Resistantbacteria Using Resistance Genes. Master’s Thesis, Chalmers University of Technology, Gothenburg, Sweden, 2024. [Google Scholar]
  291. Ikpe, F.; Williams, T.; Orok, E.; Ikpe, A. Antimicrobial resistance: Use of phage therapy in the management of resistant infections. Mol. Biol. Rep. 2024, 51, 925. [Google Scholar] [CrossRef]
  292. Wilson, C.; Thomson, E.C. Resilience to emerging infectious diseases and the importance of scientific innovation. Future Healthc. J. 2024, 11, 100023. [Google Scholar] [CrossRef]
  293. Gholap, A.D.; Khuspe, P.R.; Pardeshi, S.R.; Uddin, M.J.; Das, U.; Hatvate, N.T.; Rojekar, S.; Giram, P.; Khalid, M.; Choonara, Y.E. Achieving Optimal Health With Host-Directed Therapies (HDTs) in Infectious Diseases—A New Horizon. Adv. Ther. 2025, 8, 2400169. [Google Scholar] [CrossRef]
  294. Mahajan, S.; Rana, P.S.; Ferguson, D.L.; Wozniak, D.J. Unraveling the Complexity: Biofilms and Their Interactions with Myeloid Immune Cells. In Biofilm Matrix; Springer: Berlin/Heidelberg, Germany, 2024; pp. 311–336. [Google Scholar]
  295. Sun, D.; Chang, Q.; Lu, F. Immunomodulation in diabetic wounds healing: The intersection of macrophage reprogramming and immunotherapeutic hydrogels. J. Tissue Eng. 2024, 15, 20417314241265202. [Google Scholar] [CrossRef]
  296. do Nascimento Júnior, J.X.; Sola-Penna, M.; Zancan, P. Clotrimazole reverses macrophage M2 polarization by disrupting the PI3K/AKT/mTOR pathway. Biochem. Biophys. Res. Commun. 2024, 696, 149455. [Google Scholar] [CrossRef]
  297. Genito, C.J.; Darwitz, B.P.; Reber, C.P.; Moorman, N.J.; Graves, C.L.; Monteith, A.J.; Thurlow, L.R. mTOR signaling is required for phagocyte free radical production, GLUT1 expression, and control of Staphylococcus aureus infection. Mbio 2024, 15, e00862-24. [Google Scholar] [CrossRef]
  298. Rasquel-Oliveira, F.S.; Ribeiro, J.M.; Martelossi-Cebinelli, G.; Costa, F.B.; Nakazato, G.; Casagrande, R.; Verri, W.A. Staphylococcus aureus in inflammation and pain: Update on pathologic mechanisms. Pathogens 2025, 14, 185. [Google Scholar] [CrossRef] [PubMed]
  299. Nasri, M.; Ritter, M.U.; Mir, P.; Dannenmann, B.; Kaufmann, M.M.; Arreba-Tutusaus, P.; Xu, Y.; Borbaran-Bravo, N.; Klimiankou, M.; Lengerke, C. CRISPR-Cas9n-mediated ELANE promoter editing for gene therapy of severe congenital neutropenia. Mol. Ther. 2024, 32, 1628–1642. [Google Scholar] [CrossRef] [PubMed]
  300. Liu, Y.; Long, S.; Wang, H.; Wang, Y. Biofilm therapy for chronic wounds. Int. Wound J. 2024, 21, e14667. [Google Scholar] [CrossRef]
  301. Qu, S.; Ji, Y.; Fan, L.; Yan, T.; Zhu, G.; Song, H.; Yang, K.; Han, X. Light-Enhanced Hypoxia-Responsive Gene Editing for Hypoxia-Resistant Photodynamic and Immunotherapy. Adv. Healthc. Mater. 2024, 13, 2302615. [Google Scholar] [CrossRef]
  302. Tran, H.H.; Watkins, A.; Oh, M.J.; Babeer, A.; Schaer, T.P.; Steager, E.; Koo, H. Targeting biofilm infections in humans using small scale robotics. Trends Biotechnol. 2024, 42, 479–495. [Google Scholar] [CrossRef]
  303. Resnik, D.B. Biosafety, biosecurity, and bioethics. Monash Bioeth. Rev. 2024, 42, 137–167. [Google Scholar] [CrossRef]
  304. Singh, V.; Rastogi, M. Future and Challenges of Microbiome Engineering. In Microbiome Engineering; CRC Press: Boca Raton, FL, USA, 2024; pp. 263–280. [Google Scholar]
  305. Wiersma, B. Assessment of Biofilm Matrix Disruption as Treatment for Pseudomonas aeruginosa Lung Infections. Master’s Thesis, Utrecht University, Utrecht, The Netherlands, 2024. [Google Scholar]
  306. Fang, X.; Li, P.; Luo, C.; Wu, C.; Shi, X.; Lan, J.; Zhao, F.; Wang, F.; Zhang, K.; Yang, C. Epiphytic bacterial consortia drive growth regulation in potato under methyl jasmonate elicitation: A leaf surface multi-omics perspective. Plant Physiol. Biochem. 2025, 222, 109737. [Google Scholar] [CrossRef] [PubMed]
  307. Ramachandran, G.; Palanisamy, B.; Chackaravarthy, G.; Chelliah, C.K.; Rajivgandhi, G.; Quero, F.; Natesan, M. Diagnosis of Physical Stimuli Response Enhances the Anti-Quorum Sensing Agents in Controlling Bacterial Biofilm Formation. In Nanoscience and Nanotechnology for Smart Prevention, Diagnostics and Therapeutics: Fundamentals to Applications; Wiley: Hoboken, NJ, USA, 2024; pp. 91–115. [Google Scholar]
  308. Ma, Z.; Zuo, T.; Frey, N.; Rangrez, A.Y. A systematic framework for understanding the microbiome in human health and disease: From basic principles to clinical translation. Signal Transduct. Target. Ther. 2024, 9, 237. [Google Scholar] [CrossRef] [PubMed]
  309. Odoom, A.; Osman, A.-H.; Dzuvor, C.K. Recent advances in immunotherapeutic and vaccine-based approaches for the treatment of drug-resistant bacterial infections. ACS Infect. Dis. 2025, 11, 1366–1402. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Classical techniques in biofilm research: Created in Biorender. Bhasme, P. (2025). https://BioRender.com/b67h2c1.
Figure 1. Classical techniques in biofilm research: Created in Biorender. Bhasme, P. (2025). https://BioRender.com/b67h2c1.
Antibiotics 14 00865 g001
Figure 2. Advanced microscopy and imaging techniques in biofilm research: Created in Biorender. Bhasme, P. (2025). https://BioRender.com/b67h2c1.
Figure 2. Advanced microscopy and imaging techniques in biofilm research: Created in Biorender. Bhasme, P. (2025). https://BioRender.com/b67h2c1.
Antibiotics 14 00865 g002
Figure 3. In vivo and AI technology in biofilm research: Created in Biorender. Bhasme, P. (2025). https://BioRender.com/b67h2c1.
Figure 3. In vivo and AI technology in biofilm research: Created in Biorender. Bhasme, P. (2025). https://BioRender.com/b67h2c1.
Antibiotics 14 00865 g003
Figure 4. Synthetic biology and genetic engineering approaches for biofilm control: Created in Biorender. Bhasme, P. (2025). https://BioRender.com/b67h2c1.
Figure 4. Synthetic biology and genetic engineering approaches for biofilm control: Created in Biorender. Bhasme, P. (2025). https://BioRender.com/b67h2c1.
Antibiotics 14 00865 g004
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Haval, M.; Unakal, C.; Ghagane, S.C.; Pandit, B.R.; Daniel, E.; Siewdass, P.; Ekimeri, K.; Rajamanickam, V.; Justiz-Vaillant, A.; Lootawan, K.-A.A.; et al. Biofilms Exposed: Innovative Imaging and Therapeutic Platforms for Persistent Infections. Antibiotics 2025, 14, 865. https://doi.org/10.3390/antibiotics14090865

AMA Style

Haval M, Unakal C, Ghagane SC, Pandit BR, Daniel E, Siewdass P, Ekimeri K, Rajamanickam V, Justiz-Vaillant A, Lootawan K-AA, et al. Biofilms Exposed: Innovative Imaging and Therapeutic Platforms for Persistent Infections. Antibiotics. 2025; 14(9):865. https://doi.org/10.3390/antibiotics14090865

Chicago/Turabian Style

Haval, Manasi, Chandrashekhar Unakal, Shridhar C. Ghagane, Bijay Raj Pandit, Esther Daniel, Parbatee Siewdass, Kingsley Ekimeri, Vijayanandh Rajamanickam, Angel Justiz-Vaillant, Kathy-Ann A. Lootawan, and et al. 2025. "Biofilms Exposed: Innovative Imaging and Therapeutic Platforms for Persistent Infections" Antibiotics 14, no. 9: 865. https://doi.org/10.3390/antibiotics14090865

APA Style

Haval, M., Unakal, C., Ghagane, S. C., Pandit, B. R., Daniel, E., Siewdass, P., Ekimeri, K., Rajamanickam, V., Justiz-Vaillant, A., Lootawan, K.-A. A., Oliveira, F. M. D., Bashetti, N., Naqvi, T. A., Shettar, A., & Bhasme, P. (2025). Biofilms Exposed: Innovative Imaging and Therapeutic Platforms for Persistent Infections. Antibiotics, 14(9), 865. https://doi.org/10.3390/antibiotics14090865

Note that from the first issue of 2016, this journal uses article numbers instead of page numbers. See further details here.

Article Metrics

Back to TopTop