Translate this page into:
Antibiotic resistance: The growing threats and potential solutions
*Corresponding author: Olisaemeka Zikora Akunne, Department of Pharmacy, ASK Medical and Diagnostic Center, Abuja, Nigeria. olisaemeka.akunne.181526@unn.edu.ng
-
Received: ,
Accepted: ,
How to cite this article: Akunne OZ, Emmanuel BN, Saidu UF, et al. Antibiotic resistance: The growing threats and potential solutions. Am J Pharmacother Pharm Sci 2025:013
Abstract
Antibiotic resistance (AR) has become a critical global health crisis, threatening the efficacy of antibiotics and increasing healthcare costs, morbidity, and mortality. The One Health approach, which emphasizes the interconnectedness of human, animal, and environmental health, is increasingly recognized as an essential framework for addressing the complex drivers of AR. This review explores the mechanisms of resistance, such as genetic mutations, horizontal gene transfer, and biofilm formation, along with contributing factors such as the overuse of antibiotics in healthcare and agriculture. We assess both traditional and emerging strategies to combat AR, including antimicrobial stewardship, the development of new antibiotics, and alternative therapies such as bacteriophages, probiotics, and metal complexes. In addition, innovative approaches such as nanotechnology, combination therapies, and precision medicine are highlighted as promising tools for AR mitigation. Through case studies, this paper illustrates the effectiveness of the One Health approach in managing zoonotic diseases and antimicrobial resistance, while also underscoring the need for transdisciplinary collaboration to secure a sustainable and effective future for antibiotic use.
Keywords
Alternative therapies
Antibiotic stewardship
Antimicrobial resistance
One health approach
Precision medicine
Public health
INTRODUCTION
Antibiotics are drugs designed to prevent and treat bacterial infections, and their discovery in the 20th century marked a transformative period in modern medicine, significantly reducing mortality from infectious diseases.[1-3] However, just 70 years after their introduction, antibiotic resistance (AR) has emerged as a major global health threat.[1] AR develops when bacteria adapt to evade the effects of antibiotics – often due to misuse, overuse, and selective pressure – through various mechanisms [Table 1].[4,5] This resistance has been exacerbated by limited innovation in drug development, profit-driven healthcare practices, and the absence of diagnostic testing before prescriptions.[6] The World Health Organization (WHO) has identified this trend as a critical issue, categorizing certain antimicrobials – including fluoroquinolones and third- and fourth-generation cephalosporins – as highest priority drugs for treating multidrug-resistant (MDR) infections.[7,8] Antibiotics, like all potent medications, offer considerable benefits when used appropriately but can cause adverse effects when misused, with no clinical advantage.[9] As health systems advance globally, the silent but widespread rise of antimicrobial resistance (AMR) threatens to reverse decades of medical progress.[10,11] This includes the proliferation of MDR pathogens such as Mycobacterium tuberculosis, Escherichia coli, Acinetobacter baumannii, Klebsiella pneumoniae, Staphylococcus aureus, and Streptococcus pneumoniae.[12]
| Method of resistance | Description | Examples |
|---|---|---|
| Mutation | Genetic mutations in bacteria can lead to changes in protein structure, preventing antibiotic binding. | Rifampicin resistance through mutations in RNA polymerase gene. |
| Efflux pumps | Bacteria use efflux pumps to expel antibiotics out of the cell, reducing drug concentration. | Tetracycline efflux in Gram-negative bacteria. |
| Enzymatic degradation | Bacteria produce enzymes that chemically modify or destroy the antibiotic. | Beta-lactamase enzymes breaking down penicillin. |
| Alteration of target sites | Bacteria alter antibiotic target sites, reducing the drug’s ability to bind and inhibit functions. | MRSA alters penicillin-binding proteins. |
| Reduced permeability | Modifications in the cell wall or membrane reduce antibiotic uptake. | Porin mutations in Pseudomonas aeruginosa. |
| Horizontal gene transfer | Bacteria acquire resistance genes from other bacteria through transformation, transduction, or conjugation. |
Streptococcus pneumoniaeacquiring genes for capsular polysaccharides, which can increase its virulence and ability to cause disease. Salmonella acquiring genes for AR through phage-mediated transduction The transfer of plasmids carrying AR genes, such as the R-plasmid, between Escherichia colicells. |
| Biofilm formation | Bacteria form biofilms that protect them from antibiotic penetration and host defenses. | Pseudomonas aeruginosain cystic fibrosis patients. |
MRSA: Methicillin-resistant Staphylococcus aureus, AR: Antibiotic resistance
According to the WHO, AR was responsible for 700,000 deaths annually in 2019, with projections estimating that this figure could climb to 20 million by 2050. In addition, AMR incurs a financial burden exceeding $2.9 trillion.[10,13] This rise in resistance jeopardizes the efficacy of commonly used antibiotics in treating bacterial infections globally.[14] The 2022 Global AMR and Use Surveillance System report highlights concerning levels of resistance among prevalent bacterial pathogens. For instance, resistance rates for third-generation cephalosporin-resistant E. coli reached 42%, while methicillin-resistant S. aureus (MRSA) stood at 35% across 76 countries. Resistance to standard antibiotics such as ampicillin, cotrimoxazole, and fluoroquinolones was observed in 20% of E. coli-related urinary tract infections in 2020, complicating the treatment of these common infections.[14] Furthermore, the WHO noted rising AR in bloodstream infections with Klebsiella pneumoniae, being the third most common pathogen in such infections, showing increased resistance to third-generation cephalosporins.[15] This trend has led to greater reliance on carbapenems, which has in turn fueled the global spread of carbapenem-resistant Enterobacterales (CREs) and high resistance rates to carbapenems and aminoglycosides in Acinetobacter spp. Such high levels of resistance are of great concern due to their association with MDR strains and treatment failures.[16] These resistance patterns were closely associated with antimicrobial use, influenced by various factors across different countries and regions.[12] A 2019 modeling study attributed 1.27 million deaths to AMR, with sub-Saharan Africa particularly affected by the challenge.[17,18] Limited access to microbiology resources and expertise in these regions further complicates the detection and management of antibiotic-resistant infections.[18]
Moreover, despite extensive research and development efforts to combat AR, significant gaps remain that hinder the effective management and mitigation of this global health threat. Existing studies have provided valuable insights into the mechanisms underlying AR, such as genetic mutations, horizontal gene transfer (HGT), and biofilm formation.[19] In addition, research has identified major contributors to the growing prevalence of resistance, including the overuse and misuse of antibiotics, agricultural practices, and environmental factors.[19]
Although strategies such as antibiotic stewardship programs, the development of new antibiotics, and alternative treatments (e.g., phage therapy and probiotics) have shown potential, there remains a pressing need for more robust exploration of innovative and sustainable solutions. One promising area involves the application of metal complexes as therapeutic agents against antibiotic-resistant bacteria.[20] However, the current studies in this domain are often constrained by concerns related to toxicity, stability, and regulatory challenges, limiting their broader applicability.[20]
In addition, the One Health approach, which highlights the interconnectedness of human, animal, and environmental health,[21,22] has not been fully integrated into the research and development of solutions for AR. By adopting this integrative framework, researchers could gain a deeper understanding of the complex nature of resistance and develop more holistic strategies. Moreover, emerging technologies and methodologies such as nanotechnology, combination therapies, and precision medicine, hold significant potential for advancing the fight against AR.[4] However, these innovative strategies are still in the early stage of research, so their practical applications remain underexplored. Therefore, addressing these research gaps is crucial for advancing our understanding of AR and developing sustainable solutions to this pressing global health issue. Collaboration among researchers, industry professionals, and policymakers will be crucial to foster interdisciplinary research, address regulatory barriers, and encourage international cooperation.[23] Hence, the aim of this review is to provide a comprehensive analysis of the current state of AR, its mechanisms and contributing factors, as well as to explore existing and emerging strategies to combat this global health threat. Specifically, this review will:
Examine the historical significance and current challenges associated with AR
Analyze the mechanisms of AR, including genetic mutations, HGT, and biofilm formation
Highlight major factors driving the increase in AR, including inappropriate antibiotic use, agricultural practices, and environmental pressures
Assess health implications, economic consequences, and global public health concerns related to AR
Evaluate current strategies to address AR, such as stewardship programs, new antibiotic development, and alternative therapies (e.g., phage therapy and probiotics)
Investigate the potential of metal complexes as therapeutic agents against antibiotic-resistant bacteria, focusing on their mechanisms of action, examples of their use, challenges, and the limitations associated with their development and application
Discuss the One Health approach, its importance, and its integration into the fight against AR, highlighting relevant case studies and examples
Finally, explore future directions and innovations in the field, including nanotechnology, combination therapies, and precision medicine approaches.
ANTIBIOTIC RESISTANCE: THE GROWING THREATS AND POTENTIAL SOLUTIONS
Understanding the mechanisms of antibiotic resistance
AR presents a major challenge to modern medicine, as nearly all approved antibiotics face resistance from one or more microorganisms. This resistance undermines antibiotic efficacy by enabling bacteria to evade or neutralize drug action through mechanisms generally classified as either intrinsic or acquired [Figure 1].[24-26] While antibiotics typically target vital bacterial functions such as protein synthesis, DNA replication, and cell wall biosynthesis, bacteria have evolved diverse molecular strategies to counter them. These include target site modification, efflux pump activation, enzymatic inactivation, reduced membrane permeability, metabolic bypass, and biofilm formation.[26-29] Understanding these mechanisms is critical for guiding antibiotic design, optimizing clinical application, and developing new drugs that can circumvent or suppress resistance.[29-30]

- Mechanism of antibiotic resistances.
MDR in bacteria may result from chromosomal mutations or the acquisition of mobile genetic elements such as plasmids, integrons, and transposons, many of which carry multiple resistance genes.[31-33] The enzyme β-lactamase, for example, hydrolyzes β-lactam antibiotics, rendering them ineffective. Resistance to such enzymes can be managed through the use of β-lactamase-stable drugs or inhibitors.[28] In addition, efflux pumps actively expel antibiotics from bacterial cells and are often regulated in response to environmental triggers.[34-36] Their contribution to resistance can be countered using efflux pump inhibitors.[37] Alarmingly, bacteria have evolved to exploit antibiotics as nutrient sources rather than therapeutic threats.[38,39] The emergence of resistance is thus driven by a combination of genetic adaptability and environmental pressure, making it a formidable challenge in clinical settings.[38,40]
Factors contributing to the rise of resistance
Numerous biological and environmental factors contribute to the emergence and spread of resistance. These include spontaneous gene mutations, HGT, phenotypic adaptation, and biofilm formation.[41] The following subsections expand on the primary molecular mechanisms involved in resistance development.
Genetic mutations
Genetic mutations play a fundamental role in the emergence of AR. These mutations can occur spontaneously and lead to structural or functional changes in bacterial targets that antibiotics typically act upon.[42] For example, mutations in genes encoding ribosomal subunits can reduce antibiotic binding, while changes in DNA gyrase or topoisomerase IV can confer resistance to fluoroquinolones.[43] Similarly, point mutations in penicillin-binding proteins can lower the affinity of β-lactam antibiotics.[44] The genomic flexibility of bacteria enables rapid adaptation to environmental stressors, including antibiotic exposure, allowing them to evade lethal effects through mutational events.
Biofilm-forming bacteria such as Pseudomonas aeruginosa and S. aureus exhibit an even higher mutation rate due to their heterogeneous and stressed microenvironment, which promotes genetic variability.[45] These mutations can enhance resistance by producing enzymes such as β-lactamases that degrade antibiotics or by upregulating efflux pumps that expel drugs from the cell.[45,46] Over time, these adaptations accumulate, creating highly resistant bacterial populations that are increasingly difficult to treat.
HGT
HGT is one of the most efficient and concerning mechanisms for the dissemination of AR genes (ARGs) among bacteria.[47] Through HGT, bacteria can acquire resistance not only from closely related species but also from distant ones, significantly broadening the scope of resistance spread.[48] This gene exchange often involves mobile genetic elements (MGEs) such as plasmids, transposons, and integrons, which facilitate the transfer of multiple resistance genes in a single event.[49,50]
Plasmids, in particular, play a dominant role due to their high stability and the abundance of ARGs they carry. They can replicate independently and are often conjugative, enabling direct transfer through bacterial mating processes.[50] Furthermore, plasmids frequently harbor integrons – genetic platforms that can capture and express gene cassettes, including ARGs – enhancing bacterial adaptability.[50,51]
Transposons, or “jumping genes,” can move between chromosomal and plasmid DNA, enabling the reshuffling of resistance determinants within and across genomes.[52] These elements not only increase genetic variability but also contribute to bacterial clonal expansion, particularly under selective pressure from antibiotics.[53] The efficiency of HGT is further amplified in dense bacterial communities such as biofilms, where proximity facilitates frequent gene exchange.[54,55] This horizontal acquisition, combined with the mobility and stability of MGEs, allows for the rapid and widespread dissemination of resistance traits.
Biofilm formation
Biofilm, as illustrated in Figure 2, is a group of microorganisms attached to a surface, which can form on both inert and living surfaces. Biofilm formation is an important factor in protecting bacterial cells from antibiotic treatment.[56] During biofilm formation, bacterial strains and species cooperate, using cell-to-cell communication, known as quorum sensing, facilitated by signaling molecules.[54] The biofilm formation practically involves these dynamical processes, which comprise:

- Steps in biofilm formation. EPS (extracellular polymeric substances) are secreted to promote attachment and maturation. Source (Saucer et al., 2022).
Conditioned surface attachment by bacteria (this is an initial stage, which is reversible)
Irreversible attachment (the cell adheres and accumulates, forming clusters)
Development of early biofilm structure (micro-colonies are formed from the coordinated community)
Biofilm maturation at this stage, a mature three-dimensional biofilm structure is formed
Matrix detachment of cells to free form (at this final stage, the mature biofilm detaches into individual bacterial cells again).[57,58]
Biofilm-associated bacteria are intrinsically more resistant to antibiotics than their planktonic counterparts due to a combination of interrelated mechanisms [Figure 3]. First, the extracellular polymeric substance matrix of the biofilm acts as a physical barrier that impedes the penetration of antibiotics, limiting their access to the bacterial cells embedded within the biofilm structure.[59,60] In addition, biofilms create a highly heterogeneous microenvironment, characterized by gradients in nutrients and oxygen. These gradients lead to the development of metabolic variability among the bacterial population, with some cells entering a dormant or slow-growing state in which antibiotics are significantly less effective.[59,61] Quorum sensing further contributes to this resilience by facilitating coordinated gene expression, thereby enhancing bacterial survival strategies and promoting resistance traits.[61,62] Moreover, phenotypic diversity within the biofilm contributes to differential susceptibility, where certain subpopulations remain sensitive to antibiotics while others exhibit full resistance, further complicating eradication efforts.[59,60]

- Mechanism of antibiotic resistance in biofilm. Arrows indicate decreased antibiotic penetration through the biofilm matrix and the resulting slow diffusion. Source (Abebe, 2020).
In addition, biofilms promote HGT due to the close proximity of cells, enabling frequent plasmid and integron exchange.[54,55] The matrix itself, often described as a biological “superglue,” supports cell adhesion and shields the community from immune responses and environmental stressors.[63,64] Biofilm formation also upregulates genes involved in resistance mechanisms, such as efflux pumps and antibiotic-degrading enzymes, further compounding treatment difficulties.[65]
Importantly, cells released from mature biofilms can retain acquired resistance traits, making them potent disseminators of AMR in the environment or host.[65] AR is further reinforced by structural modifications in cellular organelles targeted by antibiotics – such as the cell wall, ribosomes, nucleic acid polymerases, and folate synthesis pathways – which reduce drug susceptibility.[66,67] Therefore, disrupting biofilm development at early stages is a critical target for emerging antibiofilm and antimicrobial strategies.[68]
Overuse and misuse of antibiotics
Aside from genetic mutations, the overuse and misuse of antibiotics contribute greatly to AR in microorganisms, posing one of the highest risk factors.[38,69] As a major driver of AR, inappropriate antibiotic use is widespread and rampant,[70] with an estimated 20% of all antibiotics prescriptions deemed unnecessary. Aside from repeated clinician prescriptions, the purchase of over-the-counter medicine and self-medication have contributed greatly to the increase in the misuse or overuse of antibiotics. In addition, another study indicated that some patients use antibiotics for non-bacterial infections.[71] Research has shown that factors leading to antibiotic misuse include insufficient funds for health professional consultation, inability to afford laboratory tests, unavailability of qualified health personnel, delay in getting medical attention, and personal indulgence and counsel from inexperienced individuals.[72]
Agricultural use
The extensive use of antibiotics in agriculture has significantly contributed to the rise in AR, as observed in recent studies.[47] One major factor is the application of organic manure derived from the waste and remains of humans and animals, which exposes plants to antibiotics. In agricultural practices, antibiotics are often used to promote growth and improve feed efficiency by altering gut microbiota – such as by enhancing vitamin synthesis, reducing microbial competition, and modifying rumen metabolism.[73] However, this non-therapeutic use, particularly in livestock and poultry, contributes significantly to the selection and spread of AR bacteria within agricultural ecosystems.[74]
The increase in anthropogenic activities, including manure from antibiotic-treated animals and the application of fertilizers, has turned the soil into a reservoir for AMR.[75] These activities contribute to the persistence and spread of antibiotic-resistant genes and microbial contaminants in the soil through evolutionary processes and multiplication within hosts[74] [Figure 4]. The mechanisms of AR in animals are similar to those in humans, as resistance genes can be transferred horizontally through mobile genetic elements.[76] As a result, agriculture has become a critical platform for the development and dissemination of AR.

- Antibiotic resistance in agricultural production. WWTP (wastewater treatment plant) effluent contributes to contamination of aquatic sources. Source (Iwu et al., 2020).
Environmental factors
Both the internal and external environments within an organism’s microbiota play a crucial role in the rate at which AR develops. The interaction between bacterial, human, and animal environmental microbiota is essential in understanding AR mechanisms, as genes can easily cross environmental boundaries.[77] Mutated genes are transferred between bacteria, making it easier for new strains to develop resistance to antibiotics in a short period.
External environments are considered to contribute less to mutation-based pathogens’ resistance evolution; however, environments with ecological niches that are highly variable, such as the soil, water, and other environments, provide a greatly diverse gene pool exceeding domestic animals and human microbiota.[78,79] Humans and animals are exposed to the environmental microbiota through food, water, and most especially through the raw vegetables route.[80] The distribution of ARGs among bacterial communities in these environments plays a major role in the spread of ARGs.[81] Environmental pollutants like heavy metal contamination in areas of high anthropogenic activities, like the industrial area, are great contributors to antibiotic resistance, with the ability of bacteria to thrive successfully and distribute ARGs.[82]
IMPACT OF ANTIBIOTIC RESISTANCE
AR presents a growing challenge impacting health, economies, and the overall public health system. This phenomenon occurs as bacteria evolve mechanisms to withstand drugs, diminishing the effectiveness of treatments.[14]
Health implications
The emergence of resistance carries health implications affecting patient outcomes and healthcare practices at large. A primary consequence is the heightened morbidity and mortality linked to infections that were easily managed. Infections caused by antibiotic-resistant bacteria such as MRSA and carbapenem-resistant Enterobacteriaceae (CRE) lead to more complications, extended illnesses, and higher death rates. According to the WHO, drug-resistant infections caused approximately 4.95 million deaths in 2019, with 1.27 million of those deaths directly attributed to AMR.[83]
Patients suffering from antibiotic-resistant infections often experience extended hospital stays, higher healthcare costs, and an increased likelihood of further medical complications.[84-87] These infections can lead to severe conditions, such as sepsis or organ failure, requiring intensive medical care and prolonged recovery periods.[77] In addition, the use of more potent antibiotics to treat resistant infections can have additional side effects, further complicating patient recovery, as highlighted by both the WHO and Nature.[87]
Cancer patients are particularly vulnerable to AR. Those undergoing chemotherapy have weakened immune systems, making them more susceptible to infections. The increased frequency of infections undermines preventive treatments’ effectiveness, often necessitating stronger and more toxic medications. This not only raises the risk of side effects but also complicates cancer treatment plans, potentially affecting the overall success of therapies and increasing mortality rates.[88]
In healthcare settings, AR complicates infection control. Hospitals and clinics encounter additional difficulties in preventing the spread of bacteria that resist treatment. This involves implementing stricter infection control measures, such as isolating patients with infections, improving cleaning and disinfection practices, and ensuring hand hygiene. The increased use of equipment (personal protective equipment) and other infection control strategies adds to the workload and cost in healthcare facilities.[89] Persistent outbreaks of bacteria can result in transmission within healthcare environments, posing a continuous risk to patient safety. The emotional toll on patients and healthcare workers is another aspect of resistance. Patients dealing with infections that resist treatment often face heightened anxiety and stress because of uncertainty about treatment outcomes and the potential for complications.[90] On the other hand, healthcare professionals experience the pressure of handling infections that are hard to treat, limited treatment choices, and the emotional weight of negative patient outcomes. This mental strain can impact the wellness and job satisfaction of healthcare workers, making the management of antibiotic infections even more complex.[89,90]
Economic and public health impact of AR
AR imposes substantial burdens on both the economy and global public health. Patients with resistant infections often require longer hospital stays, more complex treatments, and higher healthcare expenditures.[91] The WHO has reported that treating resistant infections is significantly more expensive than managing non-resistant cases, with one study estimating that AMR adds approximately $1,383– $30,093 per patient due to longer hospital stays and the need for second- or third-line treatments, underscoring the substantial economic burden.[84,92,93]
On a macroeconomic scale, the World Economic Forum warns that AR could reduce global GDP by 1.1–3.8% by 2050, potentially resulting in trillions of dollars in losses due to decreased productivity, increased mortality, and healthcare strain.[94] Sectors such as agriculture are also affected – resistant infections in animals reduce productivity, increase veterinary costs, and pose trade and food security risks.
Public health systems face immense pressure. Resistant infections complicate the treatment of diseases such as tuberculosis and gonorrhea, especially in vulnerable populations such as the elderly, immunocompromised individuals, and patients undergoing chemotherapy or organ transplants.[95] These challenges are further magnified in low- and middle-income countries, where limited access to diagnostics, quality healthcare, and effective antibiotics widens health disparities.[83]
The growing threat also undermines confidence in healthcare systems and poses challenges for modern medicine, which relies heavily on antibiotics for infection control in surgeries, cancer treatment, and intensive care. Combating AR thus requires a coordinated global response – emphasizing responsible antibiotic use, robust surveillance, investment in research and development, and equitable access to healthcare and public education.[96]
ANTIMICROBIAL STEWARDSHIP PROGRAM
Antimicrobial stewardship is a comprehensive strategy employed by healthcare institutions to combat AMR, incorporating policies, guidelines, surveillance, prevalence data, education, and practice audits.[97] It essentially promotes using the best antibiotic for the patient and the current clinical condition. Coordination and teamwork among healthcare professionals are essential for the success of antimicrobial stewardship programs because they guarantee shared information, uniformity in approach, and broad practice dissemination.[98]
The implementation of antimicrobial stewardship programs to optimize antibiotic use in acute care settings is on the rise. These programs primarily focus on guidelines and policies for prescribers.[98] There is a recognized correlation between antibiotic use and the development of AR.[99] A major challenge is effectively educating healthcare providers about the indirect costs and collateral effects of antibiotic use. Furthermore, systems that increase and maintain healthcare practitioners’ knowledge of the fundamentals of responsible antibiotic use must be created. Prescribers’ decisions are often influenced by outdated clinical guidelines and poorly designed medication charts, which can contribute to inappropriate or incorrect prescriptions.[100]
This results in less-than-ideal options, such as prescribing broad-spectrum antibiotics unnecessarily or prolonging antibiotic therapy beyond what is clinically required. To overcome these barriers and enhance prescribing practices, it is imperative to tackle the entire environment and processes around antibiotic medication decision-making.[98]
Developing new antibiotics
The primary strategies for developing new antibiotics to combat resistance include modifying existing antibiotics, discovering new antibiotics from unique sources, and identifying new antibiotics from small-molecule libraries.
Modifying the chemical structure of existing antibiotics can help bypass resistance mechanisms. For example, omadacycline, a modified tetracycline, overcomes common resistance mechanisms such as efflux pumps and ribosomal protection.[101] It is structurally related to older tetracyclines such as tetracycline, doxycycline, and minocycline but exhibits improved activity against resistant pathogens that have developed mechanisms limiting the efficacy of these earlier agents.[102,103] Similarly, modifications to vancomycin have led to the creation of synthetic analogs that are effective against vancomycin-resistant bacteria.[104-106] New antibiotics have also been developed by attaching a catechol-type siderophore to a cephalosporin core, enhancing its stability against β-lactamases. A prominent example is cefiderocol, a siderophore-cephalosporin conjugate that utilizes iron transport systems to enter Gram-negative bacteria and remains stable against most β-lactamases, including metallo-β-lactamases.[107]
The traditional method of discovering antibiotics often involves screening natural sources such as plants, soil actinomycetes, and marine environments. For instance, catechols from plants are known to inhibit microbial enzymes, and compounds derived from marine fungi and bacteria have shown activity against resistant strains like MRSA.[108,109] In addition, insect-derived antimicrobial peptides, like thanatin, exhibit potent activity against drug-resistant bacteria.[110]
New antibiotics can also be discovered through screening small-molecule libraries, which contain thousands of synthetic compounds with different appendages and molecular skeletons.[111] While the chances of a new compound becoming an approved drug are low (1 in 10,000), large libraries increase the likelihood of finding promising candidates. For example, a compound screened from a library of 167,405 molecules inhibited the growth of antibiotic-resistant Gram-positive bacteria by targeting an enzyme critical for their growth.[78] This compound targets lipoteichoic acid synthase, a key enzyme required for the growth of Gram-positive bacteria.[112] In addition, small-molecule libraries, which contain thousands of synthetic compounds with diverse structures, can be used to discover new antibiotics. While the chances of any given compound becoming an approved drug are low, the use of large libraries increases the possibility of finding promising new leads.[78,112]
Alternative therapies
Aside from the use of drugs developed from natural microbiomes, small drug libraries, and modification of existing drugs, other alternative strategies to combating AR are the use of bacteriophage and probiotic therapy.
Bacteriophage therapy
Bacteriophages, which are the most abundant biomass on Earth, outnumber prokaryotes by a factor of approximately ten.[113] These viruses specifically target bacterial cells to replicate. Their capsid protein heads protect and carry the viral genome, which can vary in size, structure (circular, linear, or segmented), and type (single-stranded DNA, double-stranded DNA, single-stranded RNA, or double-stranded RNA).[114] Bacteriophages are highly host-specific, attacking only particular bacterial strains based on the bacteriophage type and the bacterial surface proteins they target for adhesion. After attaching to their host through specific receptors on the bacterial cell, the bacteriophage injects its genetic material.[115] Once inside, the viral genome either integrates into the host genome (lysogenic cycle) or remains separate as extragenomic material (lytic cycle). Virulent phages follow the lytic cycle, hijacking the host’s replication mechanisms to produce multiple copies of the virus, ultimately causing the host cell to lyse. Temperate phages, on the other hand, enter a lysogenic phase and only switch to a lytic cycle under certain stress conditions. These phages are more host-specific than virulent ones, as they are confined to the host cell where they are integrated.[116] Unlike antibiotics, bacteriophage therapy does not disrupt the natural microbiota of humans or animals, as the virus can only replicate in the presence of susceptible bacteria, destroying their cells. This self-limiting characteristic makes bacteriophages especially effective against antibiotic-resistant bacteria. Bacteriophages, particularly lytic ones, are safe and well-tolerated by immunocompromised patients and those allergic to antibiotics, likely due to their composition of proteins and nucleic acids, which are nontoxic.[117,118] Although their use remains largely experimental in many parts of the world, phage therapy is actively being explored in clinical trials, with some compassionate-use cases reported, particularly in Europe and the United States. Several bacteriophage-based therapeutics are currently in early-phase clinical development, with growing interest in regulatory approval pathways and integration into personalized infection management strategies.[119]
Probiotic therapy
Probiotics are microorganisms that, when administered in adequate amounts, provide health benefits to the host.[120] The most common probiotic species include Lactobacilli and Bifidobacteria, although other genera such as E. coli, Saccharomyces cerevisiae var. boulardii, and Bacillus coagulans are also widely used.[120] New species such as Akkermansia muciniphila, Eubacterium hallii, and Faecalibacterium prausnitzii are being investigated for their potential probiotic benefits.
Probiotic bacteria offer several health benefits by enhancing the ability to control pathogenic bacteria. These benefits include improving intestinal barrier function, reducing bacterial adherence to cells, promoting co-aggregation, and producing organic acids that inhibit pathogenic bacteria.[121] Many probiotics also produce antimicrobial substances [Figure 5] such as short-chain fatty acids, hydrogen peroxide, nitric oxide, and bacteriocins, which help them compete with other microorganisms in the gastrointestinal tract and may inhibit pathogenic bacteria.[122]

- Effect of bacteriophage and probiotic on pathogenic bacteria.
The WHO and Food and Agriculture Organization (FAO) recommend probiotics as a complementary strategy to reduce antibiotic use and support gut health, particularly in infection prevention and recovery. Probiotics may lower the risk of antibiotic-associated diarrhea and secondary infections, helping reduce antibiotic consumption and slow resistance development.[123,124]
They also restore microbial balance after antibiotic use, limiting the spread of resistant bacteria. In agriculture, probiotic use is emerging as an alternative to prophylactic antibiotics, particularly in livestock, aligning with broader efforts to reduce reliance on antibiotics and combat AMR.[125]
INTRODUCTION TO METAL COMPLEXES IN MEDICINE
Metal complexes – structures composed of a central metal ion bonded to surrounding ligands – exhibit diverse physicochemical properties including magnetism, catalytic activity, biological function, and color.[126] These complexes play vital roles in physiological processes such as oxygen transport (e.g., hemoglobin), enzymatic catalysis, and metabolic regulation.[127] In medicinal chemistry, metal ions such as copper (Cu) ion, zinc (Zn) ion, and ferrous ion are fundamental to various therapeutic mechanisms, including antimicrobial, antidepressant, and diuretic effects. Deficiencies in these ions have been linked to several health conditions: Iron deficiency causes pernicious anemia, Cu deficiency may result in cardiac complications in infants, and Zn deficiency is associated with impaired growth and immune function.[128,129]
The significance of metal ions in disease pathology and treatment has made them a central focus in bioinorganic and medicinal chemistry. Abnormal metal ion concentrations are often implicated in diseases such as cancer, highlighting the therapeutic relevance of metal complexes either as active drugs or prodrugs.[130] Advances in coordination chemistry have led to the development of transition metal complexes with demonstrated anti-inflammatory, anti-infective, and anti-diabetic properties. Although some metal-based drugs are associated with side effects, they remain widely used in chemotherapeutic regimens, underscoring their ongoing importance in pharmaceutical research and clinical applications.[131,132]
Mechanisms of action of metal complexes against bacteria
Metal complexes have emerged as a promising strategy in addressing AR due to their diverse and unique mechanisms of action. Unlike traditional organic antibiotics that often target a single pathway, metal-based compounds can attack bacteria through multiple modes. These include releasing active molecules inside the cell, disrupting key biological processes through ligand exchange, or producing reactive oxygen species (ROS) through photoactivation.[133]
Research has shown that many metal complexes function similarly to catalytic drugs, either by generating toxic compounds within bacterial cells or by depleting vital cellular components.[134,135] Their three-dimensional structures also enable them to interact with biological targets in ways that flat, organic molecules often cannot – an advantage linked to greater effectiveness in past studies.[136] Furthermore, metal fragments are now being explored in drug discovery due to their ability to cover more diverse chemical space.[137]
Some metal complexes can bind directly to bacterial DNA – such as cisplatin, which forms stable adducts – interfering with replication and function.[138] Others act by mimicking essential metal ions that bacteria use, disrupting enzymes or cell signaling. Additional mechanisms include redox-active complexes that damage cells by generating ROS, and photoactivatable metal compounds used in therapies like photodynamic therapy (PDT).[138]
While certain metal-based antibiotics have shown similar resistance patterns to traditional drugs, several studies have reported that bacteria do not readily develop resistance to metal complexes, even after repeated exposure.[139-141] For example, antimicrobial PDT (aPDT) works by generating ROS that damages multiple cellular targets simultaneously, making it very difficult for bacteria to adapt or resist. So far, no conclusive evidence suggests that bacteria can develop resistance to Apdt.[120,142,143]
Although further research is needed, the current findings suggest that metal complexes may offer a powerful alternative to conventional antibiotics, with a lower risk of resistance development. Several metal-based agents – such as silver (Ag) nanoparticles, gallium, Cu(II), ruthenium, and gold complexes – are under early investigation for their broad-spectrum activity and unique mechanisms, including DNA binding and enzyme inhibition.[144,145] Some have shown efficacy against MDR bacteria such as P. aeruginosa, S. aureus, and E. coli, with ongoing studies aiming to improve their safety and selectivity.[144,146]
Examples of metal complexes used in combating antibiotic-resistant bacteria
Metal-based compounds have significantly contributed to medicinal chemistry through the development of therapies targeting a range of pathogens, including antibiotic-resistant bacteria.[133] In recent years, research into the antimicrobial properties of metal complexes has accelerated, revealing their potential as alternative or adjunct therapies in the fight against AMR.[147] A growing body of research has demonstrated that metal ions such as Cu, Zn, Ag, cobalt (Co), and nickel (Ni) possess intrinsic antimicrobial properties. When these metals are formulated into coordination complexes, they can not only exhibit standalone antibacterial activity but also enhance the pharmacological efficacy of existing antimicrobial agents.
Silver-based complexes, notably Ag(I)-N-heterocyclic carbene complexes, have shown potent activity against resistant bacterial strains such as MRSA, P. aeruginosa, and E. coli, in addition to antifungal activity against Candida albicans.[148-151] Similarly, Cu(II) complexes, particularly those incorporating Schiff bases and other chelating ligands, have demonstrated significant bactericidal effects. These complexes are known to act by inducing oxidative stress and disrupting bacterial membranes, with documented efficacy against E. coli, Klebsiella pneumoniae, and Bacillus subtilis strains.[152-155]
Zn(II) and Co(III) complexes have also garnered attention due to their ability to inhibit bacterial β-lactamase enzymes – key contributors to AR in many pathogens.[156] Furthermore, recent findings by Olagboye et al. highlight the dual antibacterial and antifungal properties of Ni(II) and manganese (II) barbitone complexes, which exhibit robust activity against both Gram-positive and Gram-negative bacterial strains.[157] Although some metal-based compounds, such as Cu(II)-TBZH, have been primarily investigated for their antifungal potential,[158] the broader literature increasingly supports their integration into antibacterial drug development pipelines.
Collectively, these metal-based agents offer unique, multi-targeted mechanisms of action that not only enhance antimicrobial potency but also reduce the likelihood of resistance development. As such, they represent a valuable addition to the therapeutic arsenal against MDR organisms.
LIMITATIONS AND CHALLENGES IN THE DEVELOPMENT OF NEW ANTIBIOTICS
AMR has emerged as a major global health crisis, with the shrinking pool of effective antibiotics posing a significant threat to modern medicine.[132] The development of new antibiotics is hindered by a complex interplay of scientific, regulatory, and economic obstacles [Table 2]. Pharmaceutical companies have increasingly withdrawn from this arena due to significant challenges and limited financial returns, with the average cost of developing a new antibiotic estimated to exceed USD 1 billion and requiring over a decade of research and trials.[159-161] Consequently, the pipeline of novel antibiotics is alarmingly depleted. The rise of antibiotic-resistant bacteria underscores the urgent need for innovative solutions, yet the path to developing new antibiotics is fraught with difficulties.[162-164] Overcoming these hurdles necessitates a collaborative effort involving researchers, policymakers, and the pharmaceutical industry.
| Challenge | Description |
|---|---|
| Scientific challenges | Difficulty in discovering novel antibiotic targets, complex resistance mechanisms, and translation of research findings into effective drugs. |
| Economic challenges | High costs of research and development, limited financial returns, and disincentives for pharmaceutical companies to invest. |
| Regulatory challenges | Lengthy and costly approval processes, limited patient enrollment in clinical trials, and difficulty in demonstrating efficacy without widespread use. |
Historically, pharmaceutical companies have been essential in developing potent new antibiotics to combat resistant pathogens. However, reliance on large companies is no longer a viable solution, as they have increasingly shifted their focus toward therapies for chronic conditions that offer better financial returns.[165] The already limited number of novel antibiotic classes in the research and development pipeline exacerbates the situation.[164]
The discovery and development of new antibiotics are hindered by a combination of scientific, economic, and regulatory obstacles. Traditional antibiotic targets – such as DNA replication, protein synthesis, and cell wall formation – have been heavily exploited, and bacteria have developed diverse resistance mechanisms, including enzymatic inactivation, efflux pump overexpression, target modification, and HGT.[162,166-168] Despite advancements in molecular biology and genomics, identifying novel bacterial vulnerabilities has proven increasingly difficult.[167,169] Ideally, new antibiotics should exploit uncharted pathways to circumvent existing resistance mechanisms, but translating these insights into clinically viable therapies remains a formidable challenge.[168]
In parallel, the antibiotic development pipeline is stifled by high costs and extended timelines. The process of bringing a new antibiotic to market can exceed a decade and cost billions of dollars, discouraging investment from the pharmaceutical industry. Between 2003 and 2013, antimicrobial research received only a fraction of the venture capital allocated to other therapeutic areas.[166] As of 2021, only six out of 32 antibiotics in clinical development were both novel and aimed at infections categorized as critical by the WHO.[162,166,170-172] This shortfall reflects the poor commercial incentives: Antibiotics are typically prescribed for short durations and often reserved for severe or resistant infections, making them less financially attractive than drugs for chronic diseases.[173]
Regulatory frameworks further exacerbate these challenges. Agencies such as the European Medicines Agency and the U.S. Food and Drug Administration require rigorous evidence of safety and efficacy before granting approval for new antibiotics.[174] However, conducting large-scale clinical trials for serious bacterial infections is inherently difficult due to the complexity of identifying and enrolling suitable patients,[174,175] demonstrating efficacy against resistant pathogens while limiting widespread early use – so as not to accelerate resistance – poses a paradox for both regulators and developers.[176]
These hurdles are reflected in the declining rate of approvals: Of 506 drugs in development by major pharmaceutical and biotechnology firms, only six were new antibiotics.[177] Addressing this crisis will require coordinated efforts, including novel scientific approaches, economic incentives for research investment, and regulatory innovation to streamline approvals while safeguarding public health.
THE ONE HEALTH APPROACH
The One Health approach is a comprehensive framework that acknowledges the interconnectedness of human, animal, and environmental health. This paradigm is increasingly vital, as 75% of new human infectious diseases in the last three decades have originated from animals.[178] Environmental degradation, contamination, and pollution can disrupt ecosystems in ways that facilitate the spread of infectious diseases across species.[176]
The core tenets of the One Health approach involve integrating the various disciplines and institutions that work at the intersection of human, animal, and ecosystem health. This necessitates collaboration between public health professionals, veterinarians, environmental scientists, and other stakeholders to address complex health issues that transcend traditional boundaries [Figure 6].[179] For example, efforts to monitor and control zoonotic diseases, such as influenza, Ebola, and COVID-19, have highlighted the need for coordinated surveillance and response across human and animal populations.[180] Similarly, addressing the environmental drivers of disease emergence, such as habitat loss and climate change, requires an integrated One Health approach.

- Integrating disciplines for a One Health approach.
Despite growing awareness of the One Health concept, its implementation remains inconsistent. Some studies have noted that much of the existing literature on One Health has primarily focused on zoonotic diseases, with insufficient attention paid to the role of ecosystems and environmental health determinants.[181] A more comprehensive One Health approach should consider a broader range of stakeholders and perspectives, including economics, equity, and cultural context.[182] By adopting a One Health perspective, stakeholders can develop more effective prevention and control strategies that consider all three domains of health. This holistic approach is crucial for addressing global health threats, such as pandemics, AMR, and food safety concerns.[183]
Concept and importance of the One Health approach
The growing impact of zoonotic (transmitted from animals to humans) infections on human and animal health has been brought to light by recent worldwide illness occurrences.[184] It additionally seems clear that environmental changes, such as population expansion, climate change, agricultural intensification, and human encroachment into wildlife habitats, are what cause the formation of these zoonotic diseases[185,186] and that human and animal populations are at risk due to environmental contamination with harmful chemicals and other dangers.[187,188] International organizations, including the WHO, the World Organization for Animal Health (OIE), and the FAO of the United Nations, have recognized One Health as a critical component of disease control and prevention strategies.[189]
One Health strategies aimed at combating AMR primarily focus on reducing antibiotic use in food animals. While establishing a direct causal link is complex, systematic reviews and meta-analyses have shown associations between reduced antibiotic use in food animals and decreased AMR, particularly in animals, with some evidence of limited human impact. These findings highlight significant research gaps that need further exploration.[190]
Case studies and examples of One Health initiatives
Several case studies illustrate the effectiveness of the One Health approach in tackling emerging infectious diseases, promoting global collaboration, and addressing pressing public health challenges.
One of the earliest and most significant examples is the outbreak of Severe Acute Respiratory Syndrome (SARS), which emerged in 2002 as the first major transmissible novel disease of the 21st century. SARS underscored the urgency of a globally coordinated response to emerging health threats and revealed the potential for unidentified zoonotic pathogens to arise from wildlife at any time and place. The epidemic exposed vulnerabilities in national and international preparedness systems and highlighted the critical need for improved alert mechanisms, transparent information sharing, and rapid response capacities. It epitomized the foundational principles of the One Health approach by demonstrating that large-scale outbreaks require multidisciplinary and international cooperation.[191]
The global response to the emergence of H5N1 avian influenza further reinforced the value of the One Health framework. Concerns about its pandemic potential led the United Nations Secretary-General to appoint a UN Systems Coordinator for Avian and Animal Influenza, facilitating collaboration between major international organizations, including the WHO, FAO, OIE, UNICEF, the World Bank, and numerous national health ministries. This coalition launched the International Ministerial Conferences on Avian and Pandemic Influenza, which played a vital role in improving global surveillance, risk communication, and pandemic preparedness efforts.[192]
The Global Health Security Agenda (GHSA) provides another compelling example of One Health in action. Launched to strengthen global capacity to prevent, detect, and respond to infectious disease threats, the GHSA focuses on the interconnectedness of human, animal, and environmental health. It has fostered cross-sectoral collaboration and led to the development of more robust surveillance systems and response mechanisms, ultimately enhancing global public health security.[192,193]
The 2009 H1N1 influenza pandemic again demonstrated the importance of integrated, cross-sector responses. Public health officials, veterinarians, and environmental scientists worked collaboratively to monitor the outbreak and implement effective control strategies. This One Health-oriented coordination facilitated a better understanding of the virus’s transmission patterns and enabled timely interventions, thereby limiting the pandemic’s overall impact.[194]
At the national level, Kenya has implemented a One Health initiative to combat the growing threat of AMR. The country developed a national AMR training curriculum for healthcare workers, which has been widely adopted and implemented across 19 countries.[195] Kenya’s National Action Plan reflects a One Health strategy, although implementation faces challenges due to decentralized healthcare and funding gaps.[196] The country has established multiple coordination mechanisms for zoonoses, AMR, aflatoxicosis, and pesticide-related health threats but lacks an overarching coordination structure.[197] To address this, experts recommend expanding the Zoonotic Disease Unit into a broader One Health Office. In addition, the Fleming Fund consortium in Kenya has supported AMR surveillance efforts across animal and human health sectors, including antimicrobial stewardship training and capacity building.[198]
Together, these case studies illustrate the transformative power of the One Health approach in addressing a wide range of global health challenges. From emerging infectious diseases to the escalating problem of AMR, these examples underscore the importance of interdisciplinary collaboration, strengthened surveillance, and coordinated response strategies. The success of One Health initiatives across diverse contexts confirms its critical role in improving public health outcomes, enhancing global preparedness, and delivering sustainable solutions to complex health threats. This highlights why, despite increased efforts, the pipeline for new antibiotics from major pharmaceutical and biotechnology companies remains limited.[177]
FUTURE DIRECTIONS AND INNOVATIONS
Nanotechnology
Nanotechnology offers transformative potential in combating AR through the development of nanoscale materials and systems with novel antimicrobial capabilities. One key application involves antimicrobial nanoparticles – such as those composed of Ag, gold, or Zn oxide – which inherently exhibit bactericidal activity. These nanoparticles can disrupt bacterial membranes, generate ROS, and interfere with essential metabolic functions. Owing to their small size, they can efficiently penetrate bacterial cells, thereby enhancing efficacy against resistant strains.[199] Beyond intrinsic antimicrobial action, nanotechnology also enables the development of nano-carriers for antibiotic delivery. These include liposomes, dendrimers, and polymeric nanoparticles, which can encapsulate antibiotics, protect them from enzymatic degradation, and deliver them specifically to infection sites. Such targeted delivery enhances drug bioavailability, reduces off-target effects, and minimizes the likelihood of resistance development.[200] Another critical advantage is the ability of nanomaterials to disrupt biofilms – protective matrices that render bacteria particularly resilient to antibiotics. By penetrating these biofilms and releasing antimicrobials directly at the infection site, nanoparticles can dismantle biofilm structures, thereby restoring or enhancing antibiotic effectiveness.[201]
Combination therapies
Combination therapies represent another innovative avenue for addressing AR. These strategies involve the use of multiple therapeutic agents – either multiple antibiotics or antibiotics paired with non-antibiotic compounds – to achieve greater antimicrobial efficacy and limit resistance emergence. For instance, synergistic antibiotic combinations, which involve drugs with complementary mechanisms of action, can yield effects that exceed the sum of their individual activities. This not only improves therapeutic outcomes but also reduces the selection pressure for resistance.[202] In addition, antibiotics can be co-administered with adjuvants – non-antibiotic compounds that enhance their effectiveness. Common examples include beta-lactamase inhibitors, which protect beta-lactam antibiotics from enzymatic degradation and thereby restore their potency against resistant organisms.[203] A further promising approach is the integration of bacteriophages – viruses that specifically infect and kill bacteria – into treatment regimens. Phage therapy, especially when used in conjunction with antibiotics, offers the advantage of precision targeting of resistant bacterial strains and improved biofilm disruption, thereby enhancing antibiotic penetration and efficacy.[204]
Precision medicine approaches
Precision medicine introduces a paradigm shift in infectious disease treatment by tailoring interventions to individual patients based on genetic, microbial, and pharmacological factors. One application is genomic profiling of pathogens, where advanced sequencing technologies are used to detect resistance genes. This allows clinicians to select antibiotics with the highest likelihood of success, thereby reducing reliance on broad-spectrum agents and limiting resistance selection.[205] Another emerging strategy involves microbiome analysis. By understanding the composition and dynamics of the patient’s microbiota, clinicians can implement interventions – such as the use of probiotics or prebiotics – to preserve microbial balance and prevent colonization by resistant organisms.[206] Finally, personalized antibiotic dosing, guided by pharmacokinetic and pharmacodynamic modeling, ensures that each patient receives the optimal drug concentration to maximize bacterial eradication while minimizing toxicity and the development of resistance. This approach is especially valuable for populations with variable drug metabolism, including children, the elderly, and patients with co-morbidities.[207]
CONCLUSION
The rise of AR represents a profound global health challenge, exacerbated by the overuse and misuse of antibiotics across human, animal, and environmental sectors. This escalating issue, driven by genetic mutations, HGT, biofilm formation, and environmental factors, threatens the effectiveness of essential treatments; endangers public health; and imposes a heavy burden on healthcare systems and economies worldwide. MDR pathogens continue to complicate routine medical procedures and treatment, making it crucial to implement coordinated strategies that target the molecular mechanisms of resistance, promote responsible antibiotic use, and foster interdisciplinary collaboration.
Innovative solutions, such as phage therapy, metal complexes, and the One Health framework, offer promising potential to address this complex issue. The One Health approach, which integrates human, animal, and environmental health, has already demonstrated its value in managing emerging infectious diseases, zoonotic outbreaks, and AMR. Case studies, such as those involving SARS, H5N1 influenza, and AMR initiatives in Kenya, highlight the importance of a coordinated, global response to health threats that transcend traditional boundaries. However, for One Health to reach its full potential, a broader approach is needed – one that incorporates environmental, economic, cultural, and equity perspectives.
Looking ahead, technological advancements in nanotechnology, combination therapies, and precision medicine offer exciting new avenues to combat AR. Nonetheless, substantial research gaps remain, and effective long-term mitigation requires not only innovations in therapeutic development but also global cooperation, policy regulation, and public education. Addressing AR through interdisciplinary research, international collaboration, and the integration of new technologies will be essential in safeguarding public health and ensuring the continued effectiveness of antibiotics for future generations. Ultimately, a multifaceted approach, with a focus on sustainability and global health security, will be a key to overcoming the looming threat of AMR.
Ethical approval:
Institutional review board approval is not required.
Declaration of patient consent:
Patient’s consent is not required, as there are no patients in this study.
Conflicts of interest:
There are no conflicts of interest
Use of artificial intelligence (AI)-assisted technology for manuscript preparation:
The authors confirm that there was no use of artificial intelligence (AI)-assisted technology for assisting in the writing or editing of the manuscript, and no images were manipulated using AI.
Financial support and sponsorship: None.
References
- The bacterial challenge: Time to react. 2009. European Centre for Disease Prevention and Control/European Medicines Agency. Available from: https://www.ecdc.europa.eu/en/publications-data/ecdcemeajoint-technical-report-bacterial-challenge-time-react Accessed 2025 Mar 26
- [Google Scholar]
- Antibiotics on the farm: Agriculture's role in drug resistance. 2015. CGD policy paper 59. Washington, DC: Center for Global Development; Available from: https://www.cgdev.org/publication/antibiotics-farm-agricultures-role-drugresistance Accessed 26 Mar 2025
- [Google Scholar]
- A review on antibiotic resistance and way of combating antimicrobial resistance. GSC Biol Pharm Sci. 2021;14:87-97. doi:10.30574/gscbps.2021.14.2.0037
- [CrossRef] [Google Scholar]
- Antibiotic resistance: The challenges and some emerging strategies for tackling a global menace. J Clin Lab Anal. 2022;36:e24655. doi:10.1002/jcla.24655
- [CrossRef] [PubMed] [Google Scholar]
- Antibiotic resistance In: StatPearls. Treasure Island, FL: StatPearls Publishing; 2023. Available from: https://www.ncbi.nlm.nih.gov/books/nbk513277 Accessed 26 Mar 2025
- [Google Scholar]
- Emergent crisis of antibiotic resistance: A silent pandemic threat to 21st century. Microb Pathog. 2023;174:105923. doi:10.1016/j.micpath.2022.105923
- [CrossRef] [PubMed] [Google Scholar]
- Critically important antimicrobials for human medicine Geneva: World Health Organization; 2019.
- [Google Scholar]
- World Health Organization ranking of antimicrobials according to their importance in human medicine: A critical step for developing risk management strategies to control antimicrobial resistance from food animal production. Clin Infect Dis. 2016;63(8):1087-1093. doi:10.1093/cid/ciw475
- [CrossRef] [PubMed] [Google Scholar]
- Association of adverse events with antibiotic use in hospitalized patients. JAMA Internal Med. 2017;177(9):1308. doi:10.1001/jamainternmed.2017.1938
- [CrossRef] [PubMed] [Google Scholar]
- Antibiotic resistance in microbes: History, mechanisms, therapeutic strategies and future prospects. J Infect Public Health. 2021;14:1750-1766. doi:10.1016/j.jiph.2021.10.020
- [CrossRef] [PubMed] [Google Scholar]
- Antibiotic resistance: From the bench to patients. Antibiotics (Basel). 2019;8:129. doi:10.3390/antibiotics8030129
- [CrossRef] [PubMed] [Google Scholar]
- Global antimicrobial resistance and use surveillance system (GLASS 2022): Investigating the relationship between antimicrobial resistance and antimicrobial consumption data across the participating countries. PLoS One. 2024;19:e0297921. doi:10.1371/journal.pone.0297921
- [CrossRef] [PubMed] [Google Scholar]
- Overview: Global local impact of antibiotic resistance. Infect Dis Clin North Am. 2016;30:313-322. doi:10.1016/j.idc.2016.02.001
- [CrossRef] [PubMed] [Google Scholar]
- Antimicrobial resistance. November 21 2023. Accessed September 20 2025. Available from: https://www.who.int/news-room/fact-sheets/detail/antimicrobial-resistance
- [Google Scholar]
- Global Antimicrobial Resistance and Use Surveillance System (GLASS) Report: 2022 Geneva, Switzerland: World Health Organization; 2022. ISBN: 978-924-006270-2. Accessed September 20, 2025
- [Google Scholar]
- Klebsiella pneumonia and Its Antibiotic Resistance: A bibliometric analysis. BioMed Research International. 2022;2022:1-10. doi:10.1155/2022/1668789
- [CrossRef] [PubMed] [Google Scholar]
- Global burden of bacterial antimicrobial resistance in 2019: A systematic analysis. Lancet. 2022;399:629-655. doi:10.1016/S0140-6736(21)02724-0
- [CrossRef] [PubMed] [Google Scholar]
- Mortality associated with third generation cephalosporin resistance in Enterobacteriaceae bloodstream infections at one South African hospital. J Glob Antimicrob Resist. 2022;3:176-184. doi:10.1016/j.jgar.2022.03.001
- [CrossRef] [PubMed] [Google Scholar]
- Mechanisms and implications of antibiotic resistance: A global public health challenge. J Popul Ther Clin Pharmacol. 2024;31:45-59. doi:10.53555/ztnsyy41
- [CrossRef] [Google Scholar]
- Metals to combat antimicrobial resistance. Nat Rev Chem. 2023;7:202-224. doi:10.1038/s41570-023-00463-4
- [CrossRef] [PubMed] [Google Scholar]
- Antimicrobial resistance in human health: a comprehensive review of One Health approach. Bengal Physician J. 2024;11:18-23. doi:10.5005/jp-journals-10070-8039
- [CrossRef] [Google Scholar]
- Emerging non-traditional approaches to combat antibiotic resistance. Curr Microbiol. 2022;79:330. doi:10.1007/s00284-022-03029-7
- [CrossRef] [PubMed] [Google Scholar]
- Ensuring antibiotic development, equitable availability, and responsible use of effective antibiotics: Recommendations for multisectoral action. Clin Infect Dis. 2018;68:1952-1959. doi:10.1093/cid/ciy824
- [CrossRef] [PubMed] [Google Scholar]
- Fluoroquinolones In: Giguère S, Prescott JF, Dowling PM, eds. Antimicrobial therapy in veterinary medicine (5th ed). New Jersey: John Wiley and Sons. Inc.; 2013. p. :295-314. doi:10.1002/9781118675014.ch18
- [CrossRef] [Google Scholar]
- Effect of subtherapeutic administration of antibiotics on the prevalence of antibiotic-resistant Escherichia coli bacteria in feedlot cattle. Appl Environ Microbiol. 2008;74:4405-4416. doi:10.1128/aem.00489-08
- [CrossRef] [PubMed] [Google Scholar]
- A review on molecular mechanisms of bacterial resistance to antibiotics. Eur J Appl Sci. 2016;5:301. doi:10.5829/idosi.ejas.2016.8.5.1143
- [CrossRef] [Google Scholar]
- Molecular mechanisms of antibiotic resistance. Chem Commun (Camb). 2011;47:4055-4061. doi:10.1039/C0CC05111J
- [CrossRef] [PubMed] [Google Scholar]
- Mechanism of bacterial resistance to antibiotics. Arch Intern Med. 1991;151:886-895.
- [CrossRef] [Google Scholar]
- Molecular mechanisms of antibiotic resistance revisited. Nat Rev Microbiol. 2023;21:280-295. doi:10.1038/s41575-022-00820-y
- [CrossRef] [PubMed] [Google Scholar]
- Action and resistance mechanism of antibiotic: A guide for clinicians. J Anaesthesiol Clin Pharmacol. 2017;33:300-305. doi:10.4103/joacp.JOACP_349_15
- [CrossRef] [PubMed] [Google Scholar]
- Molecular mechanisms of antibacterial multidrug resistance. Cell. 2007;128:1037-1050. doi:10.1016/j.cell.2007.30.004
- [CrossRef] [PubMed] [Google Scholar]
- Antibiotic resistance mechanisms of clinically important bacteria. Medicina (Kaunas). 2011;47:137-146.
- [CrossRef] [PubMed] [Google Scholar]
- Antimicrobials: Modes of action and mechanisms of resistance. Int J Toxicol. 2003;22:135-143. doi:10.1080/10915810305089
- [CrossRef] [PubMed] [Google Scholar]
- Acinetobacter baumannii efflux pumps and antibiotic resistance. Infect Drug Resist. 2020;13:423-434. doi:10.2147/idr.s228089
- [CrossRef] [PubMed] [Google Scholar]
- Phylogeny of multidrug transporters. Semin Cell Dev Biol. 2001;12:205-213. doi:10.1006/scdb.2000.0246
- [CrossRef] [PubMed] [Google Scholar]
- Histidine kinases and response regulator proteins in two-component signaling systems. Trends Biochem Sci. 2001;26:369-376. doi:10.1016/s0968-0004(01)01852-7
- [CrossRef] [PubMed] [Google Scholar]
- Bacterial efflux pump inhibitors from natural sources. J Antimicrob Chemother. 2006;59:1247-1260. doi:10.1093/jac/dkl460
- [CrossRef] [PubMed] [Google Scholar]
- Antimicrobial resistance, mechanisms and its clinical significance. Dis Mon. 2020;66:100971. doi:10.1016/j.disamonth.2020.100971
- [CrossRef] [PubMed] [Google Scholar]
- Antibiotic resistance: An overview of mechanisms and a paradigm shift. Curr Sci. 2009;96:11.
- [Google Scholar]
- Antibiotic resistance mechanisms in bacteria: Biochemical and genetic aspects. Food Technol Biotechnol. 2008;46:11-21.
- [Google Scholar]
- Antibiotics resistance mechanism In: Antibiotics and antimicrobial resistance genes in the environment. Amsterdam: Elsevier; 2020. p. :292-312. doi:10.1016/b978-0-12-818882-8.00019-x
- [CrossRef] [Google Scholar]
- Mechanisms of antibiotic resistance. Microbiol Spectr. 2016;4:2. doi:10.1128/microbiolspec.vmbf-0016-2015
- [CrossRef] [PubMed] [Google Scholar]
- Mechanisms of drug resistance: Quinolone resistance. Ann N Y Acad Sci. 2015;1354:12-31. doi:10.1111/nyas.12830
- [CrossRef] [PubMed] [Google Scholar]
- Resistance to ß-lactam antibiotics conferred by point mutations in penicillin-binding proteins PBP3, PBP4 and PBP6 in Salmonella enterica. PLoS One. 2014;9:e97202. doi:10.1371/journal.pone.0097202
- [CrossRef] [PubMed] [Google Scholar]
- Tolerance and resistance of Pseudomonas aeruginosa biofilms to antimicrobial agents-how P. aeruginosa can escape antibiotics. Front Microbiol. 2019;10:913. doi:10.3389/fmicb.2019.00913
- [CrossRef] [PubMed] [Google Scholar]
- Antibiotic resistance and biofilm tolerance: A combined threat in the treatment of chronic infections. J Wound Care. 2018;27:273-277. doi:10.12968/jowc.2018.27.5.273
- [CrossRef] [PubMed] [Google Scholar]
- Antibiotics and antibiotic resistance genes in animal manure-consequences of its application in agriculture. Front Microbiol. 2021;12:610656. doi:10.3389/fmicb.2021.610656
- [CrossRef] [PubMed] [Google Scholar]
- Mechanisms of antimicrobial resistance in bacteria. Am J Infect Control. 2006;34:S3-S10. doi:10.1016/j.amjmed.2006.03.011
- [CrossRef] [PubMed] [Google Scholar]
- Antibiotics and antibiotic resistance genes in natural environments. Science. 2008;321:365-367. doi:10.1126/science.1159483
- [CrossRef] [PubMed] [Google Scholar]
- Antibiotics, resistome and resistance mechanisms: A bacterial perspective. Front Microbiol. 2018;9:2066. doi:10.3389/fmicb.2018.02066
- [CrossRef] [PubMed] [Google Scholar]
- Integrons. Annu Rev Genet. 2010;44:141-166. doi:10.1146/annurev-genet-102209-163504
- [CrossRef] [PubMed] [Google Scholar]
- Transposons: The agents of antibiotic resistance in bacteria. J Basic Microbiol. 2018;58:905-917. doi:10.1002/jobm.201800204
- [CrossRef] [PubMed] [Google Scholar]
- Mobile DNA and evolution in the 21st century. Mob DNA. 2010;1:4. doi:10.1186/1759-8753-1-4
- [CrossRef] [PubMed] [Google Scholar]
- The role of bacterial biofilm in antibiotic resistance and food contamination. Int J Microbiol. 2020;2020:1705814. doi:10.1155/2020/1705814
- [CrossRef] [PubMed] [Google Scholar]
- The role of biofilms as environmental reservoirs of antibiotic resistance. Front Microbiol. 2015;6:1216. doi:10.3389/fmicb.2015.01216
- [CrossRef] [PubMed] [Google Scholar]
- The biofilm life cycle: Expanding the conceptual model of biofilm formation. Nat Rev Microbiol. 2022;20:608-620. doi:10.1038/s41579-022-00767-0
- [CrossRef] [PubMed] [Google Scholar]
- Matrix exopolysaccharides; the sticky side of biofilm formation. FEMS Microbiol Lett. 2017;364:fnx120. doi:10.1093/femsle/fnx120
- [CrossRef] [PubMed] [Google Scholar]
- Bacterial biofilm: Structure, function, and antimicrobial resistance. Endod Topics. 2012;22:2-16. doi:10.1111/j.1601-1546.2012.00277.x
- [CrossRef] [Google Scholar]
- Peering into the matrix: A look at biofilms and their inherent antibiotic resistance. Surg J (NY). 2013;6:71-77. doi:10.21083/surg.v6i2.2201
- [CrossRef] [Google Scholar]
- Biofilm-specific antibiotic resistance. Future Microbiol. 2012;7:1061-1072. doi:10.2217/fmb.12.76
- [CrossRef] [PubMed] [Google Scholar]
- Innate and induced resistance mechanisms of bacterial biofilms. Curr Top Microbiol Immunol. 2008;322:85-105. doi:10.1007/978-3-540-75418-3_5
- [CrossRef] [PubMed] [Google Scholar]
- A review on resistance mechanisms in bacterial biofilm formations. Recent Dev Med Res. 2021;3:7-18. doi:10.9734/bpi/rdmmr/v3/4469f
- [CrossRef] [Google Scholar]
- Microbial biofilm detection on food contact surfaces by macro-scale fluorescence imaging. J Food Eng. 2010;99:314-322. doi:10.1016/j.jfoodeng.2010.03.005
- [CrossRef] [Google Scholar]
- Living in a microbial world. Abingdon, UK: Garland Science, Taylor and Francis Group; 2011.
- [CrossRef] [Google Scholar]
- Znaczenie biofilmu w kontekscie narastania opornosci bakterii na antybiotyki [The importance of biofilm in the context of increasing bacterial resistance to antibiotics] Pol Merkur Lekarski. 2019;47:197-202.
- [Google Scholar]
- A review of the mechanisms of action and resistance of antimicrobial agents. Can Respir J. 1999;6(Suppl A):20A-22A.
- [Google Scholar]
- Antibiotics classification and visual target sites for bacterial inhibition. Adv Pharmacol Clin Trials. 2018;3:3. doi:10.23880/apct-16000137
- [CrossRef] [Google Scholar]
- Bacterial biofilm eradication agents: A current review. Front Chem. 2019;7:824. doi:10.3389/fchem.2019.00824
- [CrossRef] [PubMed] [Google Scholar]
- Antimicrobials misuse/overuse adverse effect, mechanism, challenges and strategies to combat resistance. Open Biotechnol J. 2020;14:107. doi:10.2174/1874070702014010107
- [CrossRef] [Google Scholar]
- Overuse and misuse of antibiotics. J Biomed Pharm Res. 2022;11:38-47. doi:10.32553/jbpr.v11i1.899
- [CrossRef] [Google Scholar]
- Accounting for variation in and overuse of antibiotics among humans. Bioessays. 2021;43:2000163. doi:10.1002/bies.202000163
- [CrossRef] [PubMed] [Google Scholar]
- Antimicrobial stewardship: Knowledge, perceptions, and factors associated with antibiotics misuse among consumers visiting community pharmacies in a Nigeria southwestern state. J Pharm Policy Pract. 2023;16:120. doi:10.1186/s40545-023-00629-x
- [CrossRef] [PubMed] [Google Scholar]
- Agriculture and food animals as a source of antimicrobial-resistant bacteria. Infect Drug Resist. 2015;8:49-61. doi:10.2147/IDR.S55778
- [CrossRef] [PubMed] [Google Scholar]
- The incidence of antibiotic resistance within and beyond the agricultural ecosystem: A concern for public health. Microbiol Open. 2020;9:e1035. doi:10.1002/mbo3.1035
- [CrossRef] [PubMed] [Google Scholar]
- Antibiotic resistance genes (ARGs) in agricultural soils from the Yangtze River Delta, China. Sci Total Environ. 2020;740:140001. doi:10.1016/j.scitotenv.2020.140001
- [CrossRef] [PubMed] [Google Scholar]
- Antibiotic resistance gene spread due to manure application on agricultural fields. Curr Opin Microbiol. 2011;14:236-243. doi:10.1016/j.mib.2011.04.009
- [CrossRef] [PubMed] [Google Scholar]
- Antibiotic resistance in the environment. Nat Rev Microbiol. 2022;20:257-269. doi:10.1038/s41579-021-00649-x
- [CrossRef] [PubMed] [Google Scholar]
- Small molecule inhibitor of lipoteichoic acid synthesis is an antibiotic for gram-positive bacteria. Proc Natl Acad Sci USA. 2013;110:3531-3536. doi:10.1073/pnas.1217337110
- [CrossRef] [PubMed] [Google Scholar]
- Towards a balanced view of the bacterial tree of life. Microbiome. 2017;5:140. doi:10.1186/s40168-017-0360-9
- [CrossRef] [PubMed] [Google Scholar]
- Prevalence of antibiotic resistome in ready-to-eat salad. Front Public Health. 2020;8:92. doi:10.3389/fpubh.2020.00092
- [CrossRef] [PubMed] [Google Scholar]
- Contribution of environmental factors on the distribution of antibiotic resistance genes in agricultural soil. Eur J Soil Biol. 2020;102:103269. doi:10.1016/j.ejsobi.2020.103269
- [CrossRef] [Google Scholar]
- Health risk assessment of heavy metal exposure to street dust in the zinc smelting district, northeast of China. Sci Total Environ. 2010;408:726-733. doi:10.1016/j.scitotenv.2009.10.075
- [CrossRef] [PubMed] [Google Scholar]
- Bacteria and antibiotic resistance genes (ARGs) in PM2.5 from China: Implications for human exposure. Environ Sci Technol. 2019;53:963-972. doi:10.1021/acs.est.8b04630
- [CrossRef] [PubMed] [Google Scholar]
- Hospital and societal costs of antimicrobial-resistant infections in a Chicago teaching hospital: Implications for antibiotic stewardship. Clin Infect Dis. 2009;49:1175-1184. doi:10.1086/605630
- [CrossRef] [PubMed] [Google Scholar]
- The relationship between antimicrobial resistance and patient outcomes: Mortality, length of hospital stay, and health care costs. Clin Infect Dis. 2005;42(Suppl 2):S82-S89. doi:10.1086/499406
- [CrossRef] [PubMed] [Google Scholar]
- High cost burden and health consequences of antibiotic resistance: The price to pay. J Infect Dev Ctries. 2014;8:1096-1102. doi:10.3855/jidc.4745
- [CrossRef] [PubMed] [Google Scholar]
- Clinical and economic burden of antimicrobial resistance. Expert Rev Anti Infect Ther. 2008;6:751-763. doi:10.1586/14787210.6.5.751
- [CrossRef] [PubMed] [Google Scholar]
- Food animals and antimicrobials: Impacts on human health. Clin Microbiol Rev. 2011;24:718-733. doi:10.1128/CMR.00002-11
- [CrossRef] [PubMed] [Google Scholar]
- The hidden mental health crisis of antimicrobial resistance. Lancet Psychiatry. 2023;10:e21-e22. doi:10.1016/S2215-0366(23)00192-X
- [CrossRef] [PubMed] [Google Scholar]
- Interprofessional perceptions and emotional impact of multidrug-resistant organisms: A qualitative study. Am J Infect Control. 2019;47:876-882. doi:10.1016/j.ajic.2019.01.019
- [CrossRef] [PubMed] [Google Scholar]
- Clinical and economic impact of antibiotic resistance in developing countries: A systematic review and meta-analysis. PLoS One. 2017;12:e0189621. doi:10.1371/journal.pone.0189621
- [CrossRef] [PubMed] [Google Scholar]
- Antibiotic-resistant infection treatment costs have doubled since 2002, now exceeding $2 billion annually. Health Aff (Millwood). 2018;37:662-669. doi:10.1377/hlthaff.2017.1153
- [CrossRef] [PubMed] [Google Scholar]
- Impact of antimicrobial multidrug resistance on inpatient care cost: An evaluation of the evidence. Expert Rev Anti Infect Ther. 2013;11:321-331. doi:10.1586/eri.13.4
- [CrossRef] [PubMed] [Google Scholar]
- Enumerating the economic cost of antimicrobial resistance per antibiotic consumed to inform the evaluation of interventions affecting their use. Antimicrob Resist Infect Control. 2018;7:98. doi:10.1186/s13756-018-0384-3
- [CrossRef] [PubMed] [Google Scholar]
- Africa centres for disease control and Prevention's framework for antimicrobial resistance control in Africa. Afr J Lab Med. 2018;7:830. doi:10.4102/ajlm.v7i2.830
- [CrossRef] [PubMed] [Google Scholar]
- Assessment of global health risk of antibiotic resistance genes. Nat Commun. 2022;13:1553. doi:10.1038/s41467-022-29283-8
- [CrossRef] [PubMed] [Google Scholar]
- Antimicrobial stewardship programs in health care systems. Clin Microbiol Rev. 2005;18:638-656. doi:10.1128/CMR.18.4.638-656.2005
- [CrossRef] [PubMed] [Google Scholar]
- Antibiotic stewardship programmes-what's missing? J Antimicrob Chemother. 2010;65:2275-2277.
- [CrossRef] [PubMed] [Google Scholar]
- The zeitgeist of resistance. J Antimicrob Chemother. 2007;60(Suppl 1):i59-i61. doi:10.1093/jac/dkm160
- [CrossRef] [PubMed] [Google Scholar]
- Decision support systems for antibiotic prescribing. Curr Opin Infect Dis. 2008;21:573-579. doi:10.1097/QCO.0b013e3283118932
- [CrossRef] [PubMed] [Google Scholar]
- Microbiology and preclinical review of omadacycline. Clin Infect Dis. 2019;69(Suppl 1):S6-15. doi:10.1093/cid/ciz395
- [CrossRef] [PubMed] [Google Scholar]
- Structure-activity relationship of the aminomethylcyclines and the discovery of omadacycline. Antimicrob Agents Chemother. 2015;59:7044-7053.
- [CrossRef] [PubMed] [Google Scholar]
- Mechanism of action of the novel aminomethylcycline antibiotic omadacycline. Antimicrob Agents Chemother. 2013;58:1279-1283. doi:10.1128/AAC.01066-13
- [CrossRef] [PubMed] [Google Scholar]
- Peripheral modifications of [ΨDCH2NH]Tpg4]vancomycin with added synergistic mechanisms of action provide durable and potent antibiotics. Proc Natl Acad Sci USA. 2017;114:E5052-E5061.
- [CrossRef] [PubMed] [Google Scholar]
- Maxamycins: Durable antibiotics derived by rational redesign of vancomycin. Acc Chem Res. 2020;53:2587-2599. doi:10.1021/acs.accounts.0c00569
- [CrossRef] [PubMed] [Google Scholar]
- Vancomycin C-terminus guanidine modifications and further insights into an added mechanism of action imparted by a peripheral structural modification. ACS Infect Dis. 2020;6:2169-2180. doi:10.1021/acsinfecdis.0c00258
- [CrossRef] [PubMed] [Google Scholar]
- Cefiderocol: Discovery, chemistry, and in vivo profiles of a novel siderophore cephalosporin. Clin Infect Dis. 2019;69(Suppl 7):S538-S543. doi:10.1093/cid/ciz826
- [CrossRef] [PubMed] [Google Scholar]
- Antimicrobial compounds from plants In: Fighting antimicrobial resistance. Zagreb: IAPC-OBP; 2018. p. :243-271.
- [CrossRef] [Google Scholar]
- Potential of marine natural products against drug-resistant bacterial infections. Lancet Infect Dis. 2019;19:e237-e245. doi:10.1016/S1473-3099(18)30711-4
- [CrossRef] [PubMed] [Google Scholar]
- Structure-activity analysis of thanatin, a 21-residue inducible insect defense peptide with sequence homology to frog skin antimicrobial peptides. Proc Natl Acad Sci USA. 1996;93:1221-1225. doi:10.1073/pnas.93.3.1221
- [CrossRef] [PubMed] [Google Scholar]
- Small molecule drug discovery. Netherlands: Elsevier; 2020 doi:10.1016/C2018-0-03933-X
- [CrossRef] [Google Scholar]
- Current promising strategies against antibiotic-resistant bacterial infections. Antibiotics (Basel). 2022;12:67. doi:10.3390/antibiotics12010067
- [CrossRef] [PubMed] [Google Scholar]
- Phage genomics: Small is beautiful. Cell. 2002;108:13-16. doi:10.1016/S0092-8674(01)00637-7
- [CrossRef] [PubMed] [Google Scholar]
- 5500 Phages examined in the electron microscope. Arch Virol. 2007;152:227-243. doi:10.1007/s00705-006-0849-1
- [CrossRef] [PubMed] [Google Scholar]
- Bacteriophage therapy. Antimicrob Agents Chemother. 2001;45:649-659. doi:10.1128/AAC.45.3.649-659.2001
- [CrossRef] [PubMed] [Google Scholar]
- Bacteriophages as potential treatment option for antibiotic resistant bacteria. Adv Exp Med Biol. 2014;807:97-110. doi:10.1007/978-81-322-1777-0_7
- [CrossRef] [PubMed] [Google Scholar]
- Pros and cons of phage therapy. Bacteriophage. 2011;1:111-114. doi:10.4161/bact.1.2.14590
- [CrossRef] [PubMed] [Google Scholar]
- Bacteriophage translocation. FEMS Immunol Med Microbiol. 2006;46:313-319. doi:10.1111/j.1574-695X.2006.00044.x
- [CrossRef] [PubMed] [Google Scholar]
- The role of regulated clinical trials in the development of bacteriophage therapeutics. J Mol Genet Med. 2012;6:279-286. doi:10.4172/1747-0862.1000050
- [CrossRef] [PubMed] [Google Scholar]
- Health and nutritional properties of probiotics in food including powder milk with live lactic acid bacteria. Prevention. 2001;5:1-10.
- [Google Scholar]
- Probiotic approach to prevent antibiotic resistance. Ann Med. 2016;48:246-255. doi:10.3109/07853890.2016.1161232
- [CrossRef] [PubMed] [Google Scholar]
- Class IIa bacteriocins: Biosynthesis, structure and activity. FEMS Microbiol Rev. 2000;24:85-106. doi:10.1111/j.1574-6976.2000.tb00534.x
- [CrossRef] [PubMed] [Google Scholar]
- FAO/WHO guidelines on probiotics. J Clin Gastroenterol. 2012;46(Suppl):S1-S2. doi:10.1097/MCG.0b013e318269fdd5
- [CrossRef] [PubMed] [Google Scholar]
- Role of probiotics in health improvement, infection control and disease treatment and management. Saudi Pharm J. 2015;23:107-114. doi:10.1016/j.jsps.2013.07.001
- [CrossRef] [PubMed] [Google Scholar]
- Probiotics as antibiotic alternatives for human and animal applications. Encyclopedia. 2023;3:561-581. doi:10.3390/encyclopedia3020040
- [CrossRef] [Google Scholar]
- Special issue: Practical applications of metal complexes. Molecules. 2015;20:7951-7956. doi:10.3390/molecules20057951
- [CrossRef] [PubMed] [Google Scholar]
- Metal complexes in medicine: An overview and update from drug design perspective. Cancer Ther Oncol Int J. 2019;14:555883. doi:10.19080/CTOIJ.2019.14.555883
- [CrossRef] [Google Scholar]
- Transition metal complexes as potential therapeutic agents. Biotechnol Mol Biol Rev. 2010;5:38-45. doi:10.5897/BMBR2010.0003
- [CrossRef] [Google Scholar]
- Medicinal inorganic chemistry: Introduction. Chem Rev. 1999;99:2201-2204. doi:10.1021/cr980419w
- [CrossRef] [PubMed] [Google Scholar]
- Transition metal complexes and their application in drugs and cosmetics-A review. J Chem Pharm Res. 2011;3:951-958.
- [Google Scholar]
- Metal complexes, an untapped source of antibiotic potential? Antibiotics (Basel). 2020;9(2):90. doi:10.3390/antibiotics9020090
- [CrossRef] [PubMed] [Google Scholar]
- Catalytic organometallic anticancer complexes. Proc Natl Acad Sci U S A. 2008;105(33):11628-11633. doi:10.1073/pnas.0800076105
- [CrossRef] [PubMed] [Google Scholar]
- Transfer hydrogenation catalysis in cells as a new approach to anticancer drug design. Nat Commun. 2015;6:6582. doi:10.1038/ncomms7582
- [CrossRef] [PubMed] [Google Scholar]
- Escape from Flatland 2: Complexity and promiscuity. Medchemcomm (Camb). 2013;4(3):515-519. doi:10.1039/c2md20347b
- [CrossRef] [Google Scholar]
- Expanding medicinal chemistry into 3D space: Metallofragments as 3D scaffolds for fragment-based drug discovery. Chem Sci. 2020;11(5):1216-1225. doi:10.1039/c9sc05586j
- [CrossRef] [PubMed] [Google Scholar]
- Classification of metal-based drugs according to their mechanisms of action. Chem. 2020;6(1):41-60. doi:10.1016/j.chempr.2019.10.013
- [CrossRef] [PubMed] [Google Scholar]
- Biological activity of complex ions. Nature. 1952;170(4320):190-191. doi:10.1038/170190a0
- [CrossRef] [PubMed] [Google Scholar]
- Integrative approach for the analysis of the proteome-wide response to bismuth drugs in Helicobacter pylori. Chem Sci. 2017;8(7):4626-4633. doi:10.1039/c7sc00766c
- [CrossRef] [PubMed] [Google Scholar]
- Auranofin is an effective agent against clinical isolates of Staphylococcus aureus. Future Med Chem. 2019;11(18):1417-1425. doi:10.4155/fmc-2018-0544
- [CrossRef] [PubMed] [Google Scholar]
- Antimicrobial photodynamic therapy-what we know and what we don theCrit Rev Microbiol. . 2018;44(5):571-589. doi:10.1080/1040841x.2018.1467876
- [CrossRef] [PubMed] [Google Scholar]
- Can microbial cells develop resistance to oxidative stress in antimicrobial photodynamic inactivation? Drug Resist Updat. 2017;31:31-42. doi:10.1016/j.drup.2017.07.003
- [CrossRef] [PubMed] [Google Scholar]
- Metal nanoparticles against multi-drug-resistance bacteria. J Inorg Biochem. 2022;237:111938. doi:10.1016/j.jinorgbio.2022.111938
- [CrossRef] [PubMed] [Google Scholar]
- Metal-based nanoparticles as antimicrobial agents: An overview. Nanomaterials (Basel). 2020;10(2):292. doi:10.3390/nano10020292
- [CrossRef] [PubMed] [Google Scholar]
- Specificity in the susceptibilities of Escherichia coli, Pseudomonas aeruginosa, and Staphylococcus aureus clinical isolates to six metal antimicrobials. Antibiotics (Basel). 2019;8(2):51. doi:10.3390/antibiotics8020051
- [CrossRef] [PubMed] [Google Scholar]
- Copper In: Merck manual home health handbook. Whitehouse Station, NJ: Merck Sharp & Dohme Corp.; 2008.
- [Google Scholar]
- Synthesis, characterization and antimicrobial activities of novel silver(I) complexes with coumarin substituted N-heterocyclic carbene ligands. Bioorg Med Chem. 2016;24(3):643-650. doi:10.1016/j.bmc.2015.12.032
- [CrossRef] [PubMed] [Google Scholar]
- Synthesis, characterization, and antimicrobial activity of silver carbene complexes derived from 4,5,6,7-tetrachlorobenzimidazole against antibiotic resistant bacteria. Dalton Trans. 2012;41(21):6500-6512. doi:10.1039/c2dt00055e
- [CrossRef] [PubMed] [Google Scholar]
- Silver-N-heterocyclic carbene complexes: Synthesis, characterization, and antimicrobial properties. J Chin Chem Soc. 2017;64(4):420-426. doi:10.1002/jccs.201700024
- [CrossRef] [Google Scholar]
- Synthesis and antimicrobial activity of novel Ag-N-heterocyclic carbene complexes. Molecules. 2010;15(4):2499-2508. doi:10.3390/molecules15042499
- [CrossRef] [PubMed] [Google Scholar]
- Spectroscopic, thermal and antimicrobial properties of the copper(II) complex of Schiff base derived from 2-(salicylidene) aminopyridine. Appl Organomet Chem. 2012;26(5):356-361. doi:10.1002/aoc.2872
- [CrossRef] [Google Scholar]
- Mixed ligand Cu(II) complexes of an unsymmetrical Schiff base ligand and N-donor heterocyclic co-ligands: investigation of the effect of co-ligand on the antibacterial properties. Inorg Chim Acta. 2019;499:119185. doi:10.1016/j.ica.2019.119185
- [CrossRef] [Google Scholar]
- Synthesis, structural characterization and antimicrobial studies of novel Schiff base copper(II) complexes. J Chil Chem Soc. 2009;54(4):354-357. doi:10.4067/s0717-97072009000400006
- [CrossRef] [Google Scholar]
- Dinuclear Cu(II), Co(II), Ni(II), and Mn(II) complexes framework based on 1-(2-hydroxyphenyl) ethanone ligand: Synthesis, structural investigation and biological properties. Int J Pharm Sci Rev Res. 2013;21:270-280.
- [Google Scholar]
- Antibiotic use in some Nigerian communities: Knowledge and attitudes of consumers. Trop J Pharm Res. 2013;12(6):1137-1142. doi:10.4314/tjpr.v12i6.33
- [CrossRef] [Google Scholar]
- Synthesis, characterization, and antimicrobial activities of Ni(II) and Mn(II) of barbitone in water medium. J Emerg Technol Innov Res. 2023;10:2349-5162.
- [Google Scholar]
- Acute oral toxicity evaluations of some zinc(II) complexes derived from 1-(2-salicylaldiminoethyl)piperazine Schiff bases in rats. Int J Mol Sci. 2012;13(2):1393-1404. doi:10.3390/ijms13021393
- [CrossRef] [PubMed] [Google Scholar]
- Antibiotics and bacterial resistance in the 21st century. Perspect Med Chem. 2014;6:25-64. doi:10.4137/PMC.S14459
- [CrossRef] [Google Scholar]
- Time for a change: addressing R&D and commercialization challenges for antibacterials. Philos Trans R Soc Lond B Biol Sci. 2015;370(1670):20140086. doi:10.1098/rstb.2014.0086
- [CrossRef] [PubMed] [Google Scholar]
- Antibiotics in late clinical development. Biochem Pharmacol. 2017;133:152-163. doi:10.1016/j.bcp.2016.09.025
- [CrossRef] [PubMed] [Google Scholar]
- Antibiotics for emerging pathogens. Science. 2009;325(5944):1089-1093. doi:10.1126/science.1176667
- [CrossRef] [PubMed] [Google Scholar]
- Antibiotics: Natural products essential to human health. Med Res Rev. 2009;29(6):821-842. doi:10.1002/med.20154
- [CrossRef] [PubMed] [Google Scholar]
- An assessment of the future impact of alternative technologies on antibiotics markets. J Pharm Policy Pract. 2016;9:34. doi:10.1186/s40545-016-0085-3
- [CrossRef] [PubMed] [Google Scholar]
- When the bug cannot be killed: The rising challenge of antimicrobial resistance. Medicines (Basel). 2017;4:40. doi:10.3390/medicines4020040
- [CrossRef] [PubMed] [Google Scholar]
- Origin of antibiotics and antibiotic resistance, and their impacts on drug development: A narrative review. Pharmaceuticals (Basel). 2023;16(11):1615. doi:10.3390/ph16111615
- [CrossRef] [PubMed] [Google Scholar]
- Discovery and preclinical development of new antibiotics. Ups J Med Sci. 2014;119(2):162-169. doi:10.3109/03009734.2014.896437
- [CrossRef] [PubMed] [Google Scholar]
- The quest for novel antimicrobial compounds: Emerging trends in research, development, and technologies. Antibiotics (Basel). 2019;8(1):8. doi:10.3390/antibiotics8010008
- [CrossRef] [PubMed] [Google Scholar]
- Challenges and opportunities in the discovery, development, and commercialization of pathogen-targeted antibiotics. Drug Discov Today. 2021;26(9):2084-2089. doi:10.1016/j.drudis.2021.02.014
- [CrossRef] [PubMed] [Google Scholar]
- Antibiotics special issue: Challenges and opportunities in antibiotic discovery and development. ACS Infect Dis. 2020;6(6):1286-1288. doi:10.1021/acsinfecdis.0c00331
- [CrossRef] [PubMed] [Google Scholar]
- Antibiotic discovery and resistance: the chase and the race. Antibiotics (Basel). 2022;11(2):182. doi:10.3390/antibiotics11020182
- [CrossRef] [PubMed] [Google Scholar]
- An emptying quiver: Antimicrobial drugs and resistance. Emerg Infect Dis. 2005;11(6):791-793. doi:10.3201/eid1106.050471
- [CrossRef] [PubMed] [Google Scholar]
- Encouraging the development of new antibiotics: Are financial incentives the right way forward? A systematic review and case study. Infect Drug Resist. 2021;14:415-434. doi:10.2147/IDR.S287792
- [CrossRef] [PubMed] [Google Scholar]
- Antibiotics: The changing regulatory and pharmaceutical industry paradigm. J Antimicrob Chemother. 2015;70(5):1281-1284. doi:10.1093/jac/dku572
- [CrossRef] [PubMed] [Google Scholar]
- Addressing the regulatory hurdles facing antibiotic innovation. 2014. Available from: https://www.pewtrusts.org/en/research-and-analysis/articles/2014/12/addressing-the-regulatory-hurdles-facing-antibiotic-innovation Accessed 16 Jan 2015
- [Google Scholar]
- Antibiotic development-economic, regulatory and societal challenges. Nat Rev Microbiol. 2020;18(5):267-274. doi:10.1038/s41579-019-0293-3
- [CrossRef] [PubMed] [Google Scholar]
- Trends in antimicrobial drug development: Implications for the future. Clin Infect Dis. 2004;38(9):1279-1286. doi:10.1086/420937
- [CrossRef] [PubMed] [Google Scholar]
- The science behind one health: At the interface of humans, animals, and the environment. Ann N Y Acad Sci. 2017;1395(1):12-32. doi:10.1111/nyas.13355
- [CrossRef] [PubMed] [Google Scholar]
- One health: A holistic approach to tackling global health issues. Indian J Community Med. 2024;49(2):260-263. doi:10.4103/ijcm.ijcm_521_23
- [CrossRef] [PubMed] [Google Scholar]
- One health -WOAH - World Organisation for animal health. Available from: https://www.woah.org/en/what-we-do/global-initiatives/one-health Accessed 26 Mar 2025
- [Google Scholar]
- A review of the one health concept and its application as a tool for policy-makers. Int J One Health. 2020;6:83-89. doi:10.14202/ijoh.2020.83-89
- [CrossRef] [Google Scholar]
- One health for neglected tropical diseases. Trans R Soc Trop Med Hyg. 2021;115(2):182-184.
- [CrossRef] [PubMed] [Google Scholar]
- One health: The theory and practice of integrated health approaches. New Delhi: CABI eBooks; 2020 doi:10.1079/9781789242577.0000
- [CrossRef] [Google Scholar]
- Risk factors for human disease emergence. Philos Trans R Soc Lond B Biol Sci. 2001;356(1411):983-989. doi:10.1098/rstb.2001.0888
- [CrossRef] [PubMed] [Google Scholar]
- Interdisciplinary approaches to understanding disease emergence: The past, present, and future drivers of Nipah virus emergence. Proc Natl Acad Sci U S A. 2013;110(1):3681-3688. doi:10.1073/pnas.1201243109
- [CrossRef] [PubMed] [Google Scholar]
- Host range and emerging and reemerging pathogens. Emerg Infect Dis. 2005;11(12):1842-1847. doi:10.3201/eid1112.050997
- [CrossRef] [PubMed] [Google Scholar]
- Toward proof of concept of a one health approach to disease prediction and control. Emerg Infect Dis. 2013;19(12):e130265. doi:10.3201/eid1912.130265
- [CrossRef] [PubMed] [Google Scholar]
- The need for one health degree programs. Infect Ecol Epidemiol. 2011;1:7919. doi:10.3402/iee.v1i0.7919
- [CrossRef] [PubMed] [Google Scholar]
- One health: Questions and answers. Available from: https://www.who.int/news-room/questions-and-answers/item/one-health Accessed 12 Nov 2024
- [Google Scholar]
- One health: From concept to practice In: Yamada A, Kahn LH, Kaplan B, eds. Confronting emerging zoonoses. Berlin: Springer Japan; 2014. p. :163-189. doi:10.1007/978-4-431-55120-1_8
- [CrossRef] [Google Scholar]
- The one health approach: Why is it so important? Trop Med Infect Dis. 2019;4(2):88. doi:10.3390/tropicalmed4020088
- [CrossRef] [PubMed] [Google Scholar]
- One health security: An important component of the global health security agenda. Biosecur Bioterror. 2014;12(5):221-224. doi:10.1089/bsp.2014.0044
- [CrossRef] [PubMed] [Google Scholar]
- Strengthening global health security at the human-animal interface. Available from: https://www.who.int/activities/strengthening-global-health-security-at-the-human-animal-interface Accessed 26 Mar 2025
- [Google Scholar]
- One health approach to address zoonotic diseases. Indian J Community Med. 2020;45(Suppl S1):S6-S8. doi:10.4103/ijcm.IJCM_398_19
- [CrossRef] [PubMed] [Google Scholar]
- Development, roll-out and implementation of an antimicrobial resistance training curriculum harmonizes delivery of in-service training to healthcare workers in Kenya. Front Microbiol. 2023;14:1142622. doi:10.3389/fmicb.2023.1142622
- [CrossRef] [PubMed] [Google Scholar]
- The fragmented picture of antimicrobial resistance in Kenya: A situational analysis of antimicrobial consumption and the imperative for antimicrobial stewardship. Antibiotics (Basel). 2024;13(3):197. doi:10.3390/antibiotics13030197
- [CrossRef] [PubMed] [Google Scholar]
- Expanding beyond zoonoses: The benefits of a national One Health coordination mechanism to address antimicrobial resistance and other shared health threats at the human-animal-environment interface in Kenya. Rev Sci Tech. 2019;38(1):155-171. doi:10.20506/rst.38.1.2950
- [CrossRef] [PubMed] [Google Scholar]
- Scaling antimicrobial resistance surveillance nationally: The case of the Fleming Fund in Kenya. One Health Cases. 2024;2024 doi:10.1079/onehealthcases.2024.0009
- [CrossRef] [Google Scholar]
- Nanotechnology as a therapeutic tool to combat microbial resistance. Adv Drug Deliv Rev. 2013;65(13-14):1803-1815. doi:10.1016/j.addr.2013.07.011
- [CrossRef] [PubMed] [Google Scholar]
- Nanotechnology-based strategies for treatment of oral biofilms and infectious diseases. Microorganisms. 2018;6(2):60.
- [Google Scholar]
- Inorganic nanoparticles engineered to attack bacteria. Chem Soc Rev. 2015;44(21):7787-7807. doi:10.1039/c5cs00041f
- [CrossRef] [PubMed] [Google Scholar]
- Antimicrobial interactions: mechanisms and implications for drug discovery and resistance evolution. Curr Opin Microbiol. 2015;27:1-9. doi:10.1016/j.mib.2015.05.008
- [CrossRef] [PubMed] [Google Scholar]
- Combination approaches to combat multidrug-resistant bacteria. Trends Biotechnol. 2013;31(3):177-184. doi:10.1016/j.tibtech.2012.12.006
- [CrossRef] [PubMed] [Google Scholar]
- Phage treatment of human infections. Bacteriophage. 2011;1(2):66-85. doi:10.4161/bact.1.2.15845
- [CrossRef] [PubMed] [Google Scholar]
- Transforming clinical microbiology with bacterial genome sequencing. Nat Rev Genet. 2012;13(9):601-612. doi:10.1038/nrg3226
- [CrossRef] [PubMed] [Google Scholar]
- Antibiotic use and its consequences for the normal microbiome. Science. 2016;352(6285):544-545. doi:10.1126/science.aad9358
- [CrossRef] [PubMed] [Google Scholar]
- Personalized antibiotic dosing in the treatment of multidrug-resistant Gram-negative infections: Challenges and potential solutions. Curr Opin Infect Dis. 2020;33(4):395-400.
- [Google Scholar]

