Authors: Boyini Sirisha1&2 and Ch. Venkataramana Devi2
Journal Name: Microbiology Archives, an International Journal
DOI: https://doi.org/10.51470/MA.2023.5.2.07
Keywords: Smart antibacterial agents; antimicrobial resistance; nanotechnology precision medicine; bacterial infections
Abstract
The escalating threat of antimicrobial resistance (AMR) has necessitated the development of innovative strategies beyond conventional antibiotics. Smart antibacterial agents—engineered systems capable of targeted, responsive, and selective antimicrobial action—have emerged as promising tools in modern infection control. These include nanomaterials, stimuli-responsive drug delivery platforms, quorum sensing inhibitors, and gene-editing technologies such as CRISPR-Cas systems. This review explores the diverse mechanisms by which smart antibacterial agents combat bacterial pathogens, emphasizing their ability to enhance therapeutic precision while minimizing off-target effects. It also addresses the major challenges in clinical translation, including safety, scalability, and resistance development. Finally, the review outlines future directions, highlighting the integration of artificial intelligence, synthetic biology, and personalized medicine in advancing next-generation antibacterial therapies. Smart agents represent a paradigm shift in the management of bacterial infections, offering a versatile and adaptable approach to overcoming the limitations of traditional antibiotics.
Introduction
The rise of antibiotic resistance poses a profound threat to global public health, with implications that span across clinical, economic, and societal domains. Since the discovery of penicillin in the early 20th century, antibiotics have revolutionized modern medicine, enabling the treatment of infectious diseases, facilitating surgical procedures, and supporting immunocompromised patients [1-2]. However, the extensive and often indiscriminate use of antibiotics in both healthcare and agriculture has accelerated the natural process of microbial evolution, fostering the emergence and spread of multidrug-resistant (MDR) bacterial strains. According to the World Health Organization (WHO), antimicrobial resistance (AMR) is one of the top ten global public health threats facing humanity, responsible for an estimated 1.27 million deaths annually and expected to cause 10 million deaths per year by 2050 if left unaddressed. Conventional antibiotics typically function through generalized mechanisms such as inhibition of cell wall synthesis, protein translation, or DNA replication. While effective, these broad-spectrum agents exert considerable selective pressure on bacterial populations, inadvertently promoting the survival of resistant variants [3-4]. Moreover, traditional antibiotics lack the specificity to differentiate between pathogenic and commensal microbes, often leading to collateral damage in the host microbiome, which can have lasting effects on human health. The urgency of the resistance crisis, combined with a stagnating antibiotic development pipeline, has catalyzed a paradigm shift in antimicrobial research—toward the design and deployment of “smart” antibacterial agents.
Smart antibacterial agents represent a new generation of antimicrobial technologies that integrate advances in nanotechnology, molecular biology, synthetic biology, bioengineering, and data science to achieve high levels of precision, responsiveness, and adaptability in targeting bacterial infections. These agents are designed not only to kill or neutralize pathogens but also to intelligently interact with their environment—releasing therapeutic payloads only under specific physiological conditions, recognizing bacterial biomarkers, or even modulating immune responses to enhance treatment efficacy [5-6]. This level of control significantly reduces off-target effects and minimizes the development of resistance. Several categories of smart antibacterial strategies have been developed or are under investigation. Nanoparticle-based agents can be engineered for site-specific delivery, often triggered by environmental cues such as pH, redox status, or enzymatic activity. These nanoparticles may carry traditional antibiotics, antimicrobial peptides, or photothermal/photodynamic agents that activate under external stimuli like light or heat. Other strategies involve the use of CRISPR-Cas systems for sequence-specific targeting of bacterial genomes, allowing for precise elimination of resistant genes or pathogenic strains without disturbing the broader microbial ecosystem. Additionally, quorum-sensing inhibitors have emerged as an attractive avenue for disarming bacteria rather than killing them outright [7-8]. By interfering with the communication systems that regulate virulence factor expression and biofilm formation, these agents reduce bacterial pathogenicity and may lower the likelihood of resistance selection. Smart drug delivery systems that respond to infection-specific signals—such as local inflammation, low oxygen levels, or bacterial enzyme activity—can further enhance therapeutic specificity and reduce systemic toxicity.
Importantly, the integration of smart technologies into antimicrobial therapy is not limited to the molecular or cellular level. The use of artificial intelligence (AI) and machine learning is increasingly being explored to optimize the design, function, and application of smart agents. AI-driven models can predict bacterial susceptibility, simulate drug interactions, and identify novel therapeutic targets, thereby accelerating the drug discovery process and informing precision treatment strategies. Despite their promise, smart antibacterial agents are not without challenges. Issues related to safety, biocompatibility, large-scale manufacturing, cost-effectiveness, and regulatory approval remain significant hurdles to clinical translation. Additionally, bacteria continue to exhibit remarkable adaptability, necessitating continual innovation and vigilant monitoring of resistance trends [9-10]. Nevertheless, the multifaceted capabilities of smart agents position them as a critical component in the next generation of antimicrobial strategies, the advent of smart antibacterial agents marks a significant evolution in the field of infectious disease management. These intelligent systems offer a means to overcome the limitations of conventional antibiotics by leveraging targeted mechanisms, responsive behavior, and interdisciplinary design. As bacterial resistance continues to escalate, the development and implementation of smart antibacterial therapies will be essential to safeguard public health and ensure the sustainability of modern medicine. This review explores the various mechanisms, current challenges, and future directions in the field of smart antibacterial agents, with the goal of illuminating their potential and guiding their responsible development [11].
2. Mechanisms of Smart Antibacterial Agents
Smart antibacterial agents utilize innovative biochemical and biophysical strategies to enhance specificity, efficacy, and adaptability in treating bacterial infections. Unlike conventional antibiotics, which often function via broad-spectrum and static mechanisms, smart agents are engineered to respond dynamically to bacterial signals or host environmental cues [12]. This section outlines four principal mechanisms: targeted nanomaterials, stimuli-responsive drug delivery, quorum-sensing inhibition, and gene-editing technologies.
2.1 Targeted Nanomaterials
Nanotechnology has revolutionized antimicrobial research by enabling the development of nanoscale platforms with intrinsic or carrier-based antibacterial functions. Targeted nanomaterials, such as silver nanoparticles (AgNPs), zinc oxide nanoparticles, graphene oxide, mesoporous silica, and polymer-based nanostructures, exhibit unique physicochemical properties that allow for precise interactions with bacterial cells. These materials can be functionalized with ligands—such as antibodies, peptides, or aptamers—that recognize specific bacterial surface markers, enabling selective targeting of pathogenic bacteria while sparing commensal flora. Upon binding, nanoparticles exert their antimicrobial effects via multiple mechanisms: membrane disruption, generation of reactive oxygen species (ROS), interference with cellular respiration, or induction of intracellular stress responses. Some formulations are designed to carry traditional antibiotics or antimicrobial peptides, which are released in a controlled or site-specific manner. For instance, functionalized nanoparticles that respond to pH or bacterial enzymes can deliver antibiotics preferentially to infection sites, minimizing systemic exposure and toxicity [13]. In addition to direct antibacterial action, these nanomaterials can penetrate biofilms—dense, extracellular matrices that shield bacteria from conventional antibiotics—and facilitate the eradication of chronic infections.
2.2 Stimuli-Responsive Drug Delivery
Stimuli-responsive or “smart” drug delivery systems are engineered to release therapeutic agents in response to specific physiological or biochemical triggers found at infection sites. These stimuli include variations in pH, redox gradients, temperature, enzymatic activity, or external signals such as light and magnetic fields. Infections often create localized environments that differ markedly from healthy tissue. For example, inflamed or infected tissues exhibit acidic pH, elevated levels of bacterial enzymes (e.g., lipases or β-lactamases), or hypoxic and oxidative stress conditions. Stimuli-responsive carriers—such as liposomes, dendrimers, hydrogels, and micelles—can be constructed to remain inert under normal physiological conditions and activate drug release only in the presence of these infection-specific cues. This targeted release reduces the likelihood of off-target effects, improves the local concentration of antibiotics at the site of infection, and enhances overall treatment efficiency [14-15]. Notably, this strategy is particularly advantageous for treating intracellular infections or infections in difficult-to-access tissues such as bone, where drug penetration is often a limiting factor.
2.3 Quorum-Sensing Inhibitors
Quorum sensing (QS) is a bacterial communication system that enables population-wide coordination of gene expression in response to cell density. Through the release and detection of small signaling molecules—such as acyl-homoserine lactones (AHLs) in Gram-negative bacteria or autoinducing peptides (AIPs) in Gram-positive species—QS regulates key pathogenic behaviors including virulence factor production, toxin release, motility, and biofilm formation. Smart antibacterial strategies that target quorum sensing aim to disarm rather than kill bacteria. Quorum-sensing inhibitors (QSIs) interfere with signal synthesis, signal reception, or downstream regulatory pathways, effectively silencing bacterial cooperation without exerting strong selective pressure for resistance. Because QSIs do not inhibit growth directly, they are less likely to drive the rapid evolution of resistance mechanisms [16-17]. QSIs can be used synergistically with conventional antibiotics to enhance penetration through biofilms or reduce the minimum inhibitory concentration (MIC) required for effective bacterial clearance. Some QSIs are derived from natural sources (e.g., furanones from marine algae), while others are synthetically engineered to mimic or antagonize native signaling molecules.
2.4 Gene-Editing Antibacterial
Recent advances in gene-editing technologies—particularly CRISPR-Cas systems—have opened new avenues for targeted bacterial eradication. CRISPR (Clustered Regularly Interspaced Short Palindromic Repeats) systems, originally part of bacterial adaptive immunity, can be repurposed to identify and cleave specific DNA sequences within bacterial genomes. Smart CRISPR-based antimicrobials utilize sequence-specific guide RNAs to target resistance genes, virulence factors, or essential bacterial genes. Delivery vectors such as bacteriophages, conjugative plasmids, or nanoparticle carriers transport the CRISPR payload into bacterial cells, where Cas nucleases introduce double-strand breaks, leading to cell death or functional inactivation [18-19]. This precision allows for selective removal of pathogenic strains while leaving beneficial microbiota intact—a major advantage over traditional antibiotics. Furthermore, CRISPR antimicrobials can be designed to silence horizontal gene transfer pathways, preventing the spread of resistance genes across microbial communities. Challenges remain in optimizing delivery efficiency, avoiding off-target effects, and ensuring stability within host environments. Nevertheless, gene-editing approaches hold immense promise for the future of personalized antibacterial therapy, particularly in combating MDR pathogens.
3. Challenges and Limitations
While smart antibacterial agents offer transformative potential in the fight against bacterial infections and antimicrobial resistance, their clinical translation remains fraught with critical challenges [21-22]. These include concerns regarding safety, bacterial adaptability, manufacturing complexity, and regulatory readiness. A comprehensive understanding of these limitations is essential for guiding future research and policy development.
3.1 Safety and Biocompatibility
A major barrier to the clinical deployment of smart antibacterial agents is ensuring their safety and biocompatibility. Many nanomaterials—such as metallic nanoparticles (e.g., silver, gold, zinc oxide), carbon-based nanostructures (e.g., graphene oxide), and polymeric carriers—may induce cytotoxicity, oxidative stress, inflammatory responses, or immunogenicity in human cells. These adverse effects are often dose-dependent and influenced by particle size, shape, surface chemistry, and route of administration. Furthermore, long-term accumulation and biodistribution of nanomaterials raise concerns about chronic toxicity and environmental persistence [23-24]. While in vitro assays provide initial insights, they may not accurately predict complex in vivo interactions. Therefore, rigorous preclinical evaluations—including pharmacokinetic studies, histopathological assessments, and immune profiling in multiple animal models—are necessary to assess the full toxicological profile of smart therapeutics. Ensuring safety across diverse patient populations, including immunocompromised individuals, pregnant women, and pediatric patients, presents additional layers of complexity.
3.2 Bacterial Adaptability and Resistance Evolution
Despite their novel modes of action, smart antibacterial agents are not immune to the fundamental issue of bacterial adaptability. Microorganisms possess remarkable genetic plasticity and can rapidly evolve resistance through mutations, efflux pumps, enzyme production, or horizontal gene transfer. While some smart strategies, such as quorum-sensing inhibition or CRISPR-mediated targeting, are designed to exert lower selective pressure than conventional antibiotics, the potential for resistance evolution remains [25-26]. For example, bacteria may alter surface markers to evade targeted nanoparticles or develop compensatory quorum-sensing pathways. Similarly, delivery systems for gene-editing tools may become ineffective if receptors mutate or entry mechanisms are blocked. As a result, it is imperative to design these agents with built-in flexibility—such as modular targeting domains or adaptive delivery systems—and to use them in combination with other antimicrobial strategies. Long-term surveillance and resistance-monitoring protocols will be crucial to detect emerging resistance patterns early and adjust therapeutic strategies accordingly. Incorporating ecological and evolutionary principles into the design of smart agents may help delay or circumvent resistance development.
3.3 Manufacturing and Scalability
The fabrication of smart antibacterial agents often involves sophisticated engineering processes, including nanostructure synthesis, surface functionalization, multi-step drug loading, and sterility assurance. These processes, while effective at laboratory scale, frequently pose challenges when translated to commercial-scale production. Variability in size distribution, surface charge, or encapsulation efficiency can impact therapeutic performance and reproducibility [27-28]. Moreover, large-scale manufacturing must comply with Good Manufacturing Practice (GMP) standards, requiring robust quality control, cleanroom facilities, and specialized equipment. These requirements contribute to high production costs, which can limit accessibility and adoption, particularly in low- and middle-income countries where the burden of bacterial infections is often greatest. To overcome these challenges, scalable and cost-effective production methods—such as microfluidic synthesis, self-assembly processes, and green chemistry approaches—are being explored. Establishing standardized protocols for batch consistency, storage stability, and product shelf life will be essential for transitioning smart agents from bench to bedside.
3.4 Regulatory and Policy Barriers
Current regulatory frameworks are primarily designed for conventional small-molecule drugs and biologics, and may not be well-equipped to evaluate the complex architectures and multifunctional properties of smart antibacterial agents. For instance, the pharmacodynamics of stimuli-responsive systems or CRISPR-based therapeutics may not fit neatly into existing evaluation criteria for safety, efficacy, and mechanism of action. Additionally, regulatory agencies such as the U.S. Food and Drug Administration (FDA) and European Medicines Agency (EMA) lack standardized guidelines for assessing the long-term effects, environmental impact, and post-market surveillance of nanomedicines or synthetic gene-editing constructs. This creates uncertainty and delays in clinical translation, especially for novel combination products or hybrid platforms [29-30]. To address these gaps, regulatory bodies must collaborate with academic researchers, industry stakeholders, and international health organizations to develop tailored frameworks. These should include clear definitions, risk-benefit assessment tools, and adaptive approval pathways for emerging smart antibacterial technologies. Public engagement and ethical considerations—especially for gene-editing applications—should also be integrated into regulatory planning.
4. Future Directions
The next generation of smart antibacterial agents will be shaped by the convergence of interdisciplinary advances across biotechnology, computational science, materials engineering, and clinical medicine. These developments promise to not only overcome current limitations but also establish new paradigms in the prevention and treatment of bacterial infections. This section outlines four major trajectories likely to define the future of smart antibacterial therapy.
4.1 Integration with Artificial Intelligence and Machine Learning
Artificial intelligence (AI) and machine learning (ML) are emerging as powerful tools for accelerating the development of smart antibacterial agents. By leveraging large-scale biological and chemical datasets, AI algorithms can predict bacterial resistance patterns, optimize drug delivery systems, and model host-pathogen interactions with unprecedented speed and accuracy.
Machine learning frameworks can be employed to identify novel antimicrobial compounds through virtual screening and de novo molecular design, reducing the time and cost associated with traditional drug discovery pipelines [30]. Furthermore, predictive analytics can aid in the design of stimuli-responsive materials and nanocarriers by modeling their interactions under physiological conditions. AI-driven approaches also hold promise in forecasting treatment outcomes based on pathogen genotype, infection site characteristics, and patient-specific variables, thereby contributing to more adaptive and informed therapeutic decisions. In the clinical setting, AI tools integrated with real-time diagnostics and electronic health records could support personalized treatment recommendations, monitor therapeutic response, and even anticipate microbial evolution, paving the way for proactive infection control.
4.2 Personalized Antimicrobial Therapy
The future of antibacterial therapy is increasingly moving toward personalization. Advances in next-generation sequencing (NGS), transcriptomics, proteomics, and microbiome profiling now allow clinicians to characterize individual infection profiles with high resolution. These technologies enable the identification of pathogen species, resistance genes, virulence factors, and the composition of the surrounding microbial community [31]. Smart antibacterial agents tailored to these patient-specific signatures can improve therapeutic outcomes by enhancing selectivity, reducing toxicity, and minimizing disruption to beneficial microbiota. For example, CRISPR-based antimicrobials can be programmed to selectively target resistance genes or strain-specific DNA sequences. Similarly, nanocarriers can be functionalized based on the phenotypic traits of the infecting organism or the immune status of the patient [32]. Incorporating patient genomics, immune profiling, and microbiota composition into treatment design will not only improve efficacy but also reduce the risk of adverse reactions, antimicrobial overuse, and emergence of secondary infections.
4.3 Bioinspired and Biodegradable Platforms
Nature provides a wealth of inspiration for the development of next-generation smart antibacterial agents. Bioinspired systems—such as antimicrobial peptides (AMPs), bacteriophage-derived enzymes, and quorum-quenching molecules—offer mechanisms that have evolved over millennia to target pathogens with high specificity and minimal resistance induction. Future research is expected to increasingly integrate such biologically derived components into smart delivery systems or synthetic constructs. For instance, AMPs can be conjugated to nanoparticles to confer both targeting and bactericidal capabilities [33]. Bacteriophages and phage-derived lysins can be engineered to deliver therapeutic payloads or directly lyse bacterial cells, offering alternatives for multidrug-resistant (MDR) infections. Moreover, the use of biodegradable polymers and naturally derived materials in constructing smart delivery systems can significantly enhance safety and reduce environmental impact. Materials such as chitosan, alginate, and poly(lactic-co-glycolic acid) (PLGA) not only degrade safely within the body but also offer tunable release kinetics and functional versatility. Such eco-conscious design is especially important in mitigating the ecological burden of large-scale therapeutic deployment.
4.4 Synergistic Combinations and Multifunctional Platforms
Combining smart antibacterial agents with conventional antibiotics, immunotherapies, anti-virulence agents, or host-directed therapies offers a robust strategy to tackle complex infections. Multifunctional systems can be engineered to simultaneously attack different bacterial targets—such as membranes, metabolic pathways, and communication systems—thereby reducing the likelihood of resistance development and improving therapeutic efficacy. For example, nanoparticles can be co-loaded with antibiotics and quorum-sensing inhibitors to disrupt biofilms while killing planktonic bacteria. Alternatively, CRISPR systems can be co-administered with antibiotics to first silence resistance genes, rendering bacteria susceptible to standard treatments. Smart platforms may also deliver immunomodulatory agents to enhance host defenses, particularly in immunocompromised patients. Future formulations are likely to adopt modular designs, allowing for customizable combinations tailored to specific infections and patient needs [34]. Such integrative approaches hold promise not only for more effective treatments but also for reducing dosing frequency, shortening therapy duration, and improving patient compliance.
5. Conclusion
The advent of smart antibacterial agents marks a paradigm shift in the ongoing fight against bacterial infections and the global threat of antibiotic resistance. These next-generation therapeutics leverage advancements in nanotechnology, synthetic biology, and computational modeling to enable targeted, adaptive, and context-responsive interventions. Unlike conventional antibiotics, smart agents can selectively identify pathogens, respond to environmental cues, and disrupt resistance mechanisms—offering a more precise and personalized therapeutic strategy. Yet, despite their promising potential, the translation of these technologies from the laboratory to the clinic is not without significant hurdles. Issues related to safety, scalability, regulatory compliance, and long-term efficacy must be addressed through rigorous multidisciplinary research and robust preclinical testing. Moreover, the adaptability of bacterial pathogens continues to challenge even the most advanced strategies, underscoring the need for dynamic and resilient solutions. Equally important are the ethical and societal dimensions surrounding the use of gene-editing tools, nanomaterials, and AI-driven diagnostics in human health. Developing comprehensive regulatory frameworks, public engagement strategies, and equitable access models will be crucial to ensure that the benefits of smart antibacterial agents are widely and responsibly shared. Ultimately, the successful integration of smart antibacterial agents into clinical practice will depend on sustained collaboration across microbiology, materials science, engineering, data science, and medicine. With coordinated effort, these innovative tools have the potential not only to improve patient outcomes but also to reshape the future of infectious disease management in a more sustainable, effective, and intelligent manner.
References
- Li, X., Wu, B., Chen, H., Nan, K., Jin, Y., Sun, L., & Wang, B. (2018). Recent developments in smart antibacterial surfaces to inhibit biofilm formation and bacterial infections. Journal of materials chemistry b, 6(26), 4274-4292.
- Wei, T., Yu, Q., & Chen, H. (2019). Responsive and synergistic antibacterial coatings: Fighting against bacteria in a smart and effective way. Advanced healthcare materials, 8(3), 1801381.
- Li, W., Thian, E. S., Wang, M., Wang, Z., & Ren, L. (2021). Surface design for antibacterial materials: from fundamentals to advanced strategies. Advanced Science, 8(19), 2100368.
- León-Buitimea, A., Garza-Cárdenas, C. R., Garza-Cervantes, J. A., Lerma-Escalera, J. A., & Morones-Ramírez, J. R. (2020). The demand for new antibiotics: antimicrobial peptides, nanoparticles, and combinatorial therapies as future strategies in antibacterial agent design. Frontiers in microbiology, 11, 1669.
- Milad, S. M. A. B. (2022). Antimycotic sensitivity of fungi isolated from patients with Allergic Bronchopulmonary Aspergillosis (ABPA). In Acta Biology Forum (Vol. 1, No. 02, pp. 10-13).
- Wang, Y., Yang, Y., Shi, Y., Song, H., & Yu, C. (2020). Antibiotic‐free antibacterial strategies enabled by nanomaterials: progress and perspectives. Advanced Materials, 32(18), 1904106.
- Wang, L., Hu, C., & Shao, L. (2017). The antimicrobial activity of nanoparticles: present situation and prospects for the future. International journal of nanomedicine, 1227-1249.
- Wang, X., Shan, M., Zhang, S., Chen, X., Liu, W., Chen, J., & Liu, X. (2022). Stimuli‐responsive antibacterial materials: molecular structures, design principles, and biomedical applications. Advanced Science, 9(13), 2104843.
- Eleraky, N. E., Allam, A., Hassan, S. B., & Omar, M. M. (2020). Nanomedicine fight against antibacterial resistance: an overview of the recent pharmaceutical innovations. Pharmaceutics, 12(2), 142.
- Bhakta, S., Sipra, B. S., Dutta, P., Sahu, E., Panda, S. K., & Bas-tia, A. K. (2022). Water silk (Spirogyra bichromatophora) as a natural resource for antimicrobial phycochemicals. Acta Botanica Plantae. V01i03, 08-14.
- Seriki, S. A., & Otoikhila, O. C. (2022). Phytochemical screening and medicinal properties of ethanolic leaf extract of Harungana madagascariensis. Acta Botanica Plantae, 11-17.
- Ogori, A. F., Eke, M. O., Girgih, T. A., & Abu, J. O. (2022). Nitrate Fertilizer Improves the Physiology of Taro (Colocasia esculenta (L.) Schott var esculenta) Plants. Acta Botanica Plantae. V01i03, 36-42.
- Cavallaro, A., Taheri, S., & Vasilev, K. (2014). Responsive and “smart” antibacterial surfaces: Common approaches and new developments. Biointerphases, 9(2).
- Cloutier, M., Mantovani, D., & Rosei, F. (2015). Antibacterial coatings: challenges, perspectives, and opportunities. Trends in biotechnology, 33(11), 637-652.
- VijayKumar, R. (2019). Biochar in soil restoration: A comprehensive review on enhancing soil health and carbon sequestration. Environmental Reports.
- Mantravadi, P. K., Kalesh, K. A., Dobson, R. C., Hudson, A. O., & Parthasarathy, A. (2019). The quest for novel antimicrobial compounds: emerging trends in research, development, and technologies. Antibiotics, 8(1), 8.
- Nweze, Chibuzo Carole, Titilayo Oluwayemisi Bamidele, Eneh William Nebechukwu, Wandoo Tseaa, Rahima Yunusa, Happy Abimiku Manasseh, and Adedikpe Lateefat Bisola. “Biochemical effects of some preservatives on Vigna unguiculata in Adult Male Wistar Rats.” In Acta Biology Forum, vol. 1, pp. 14-23. 2022.
- Nagpal, N., Tomar, P. C., & Baisla, N. (2022). Allium cepa: A treasure trove of therapeutic components and an asset for well-being. Acta Botanica Plantae, 18-30.
- Bal‐Öztürk, A., Özkahraman, B., Özbaş, Z., Yaşayan, G., Tamahkar, E., & Alarçin, E. (2021). Advancements and future directions in the antibacterial wound dressings–A review. Journal of Biomedical Materials Research Part B: Applied Biomaterials, 109(5), 703-716.
- Karahan, H. E., Wiraja, C., Xu, C., Wei, J., Wang, Y., Wang, L., … & Chen, Y. (2018). Graphene materials in antimicrobial nanomedicine: current status and future perspectives. Advanced healthcare materials, 7(13), 1701406.
- Mei, L., Zhu, S., Yin, W., Chen, C., Nie, G., Gu, Z., & Zhao, Y. (2020). Two-dimensional nanomaterials beyond graphene for antibacterial applications: current progress and future perspectives. Theranostics, 10(2), 757.
- VijayKumar, R. (2019). Microbial Interactions in Silviculture for Resilient Forest Ecosystems. Microbiology Archives, an International Journal.
- Vardhan, H., Sharma, Y., Sharma, A., Paras, M., Agrahari, R., & Sharma, K. (2022). Exploring Heterosis Utilization in Cucurbitaceous Vegetable Crops. Agriculture Archives. DOI: https://doi. org/10.51470/AGRI, 1.
- Han, D., Liu, X., & Wu, S. (2022). Metal organic framework-based antibacterial agents and their underlying mechanisms. Chemical Society Reviews, 51(16), 7138-7169.
- Zhang, W., Hu, E., Wang, Y., Miao, S., Liu, Y., Hu III, Y., … & Shen, Y. (2021). Emerging antibacterial strategies with application of targeting drug delivery system and combined treatment. International journal of nanomedicine, 6141-6156.
- Pranantyo, D., Zhang, K., Si, Z., Hou, Z., & Chan-Park, M. B. (2022). Smart multifunctional polymer systems as alternatives or supplements of antibiotics to overcome bacterial resistance. Biomacromolecules, 23(5), 1873-1891.
- Nweze, C. C., Bamidele, T. O., Nebechukwu, E. W., Tseaa, W., Yunusa, R., Manasseh, H. A., & Bisola, A. L. (2022). Biochemical effects of some preservatives on Vigna unguiculata in Adult Male Wistar Rats. In Acta Biology Forum (Vol. 1, pp. 14-23).
- Kavanagh, N., Ryan, E. J., Widaa, A., Sexton, G., Fennell, J., O’Rourke, S., & Kerrigan, S. W. (2018). Staphylococcal osteomyelitis: disease progression, treatment challenges, and future directions. Clinical microbiology reviews, 31(2), 10-1128.
- Raza, S., Matuła, K., Karoń, S., & Paczesny, J. (2021). Resistance and adaptation of bacteria to non-antibiotic antibacterial agents: Physical stressors, nanoparticles, and bacteriophages. Antibiotics, 10(4), 435.
- Thentu, T. L., Pradeep, M., Nagarjuna, D., Jyothi, G. S., & Mallikarjuna, M. (2021). The im-pact of Weather-Based Agricultural Advisories on Rice Production and Income of Farmers in Nellore District, Andhra Pradesh, India, Under a Changing Climate Scenario. Acta Botanica (G), 46-52.
- Kim, I., Viswanathan, K., Kasi, G., Thanakkasaranee, S., Sadeghi, K., & Seo, J. (2022). ZnO nanostructures in active antibacterial food packaging: Preparation methods, antimicrobial mechanisms, safety issues, future prospects, and challenges. Food Reviews International, 38(4), 537-565.
- Chen, J., Wang, F., Liu, Q., & Du, J. (2014). Antibacterial polymeric nanostructures for biomedical applications. Chemical communications, 50(93), 14482-14493.
- Shoji, M. M., & Chen, A. F. (2020). Biofilms in periprosthetic joint infections: a review of diagnostic modalities, current treatments, and future directions. The journal of knee surgery, 33(02), 119-131.
- VijayKumar, R. (2020). Impact of Microbial Inoculants on Rice Growth and Yield in a Drumstick-Based Agroforestry System. Microbiology Archives, an International Journal.