Next Article in Journal
Supragingival Plaque Microbiomes in a Diverse South Florida Population
Previous Article in Journal
Infective Endocarditis—Predictors of In-Hospital Mortality, 17 Years, Single-Center Experience in Bulgaria
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

An Overview of the Recent Advances in Antimicrobial Resistance

by
Manuela Oliveira
1,2,*,
Wilson Antunes
3,
Salete Mota
4,
Áurea Madureira-Carvalho
1,5,6,
Ricardo Jorge Dinis-Oliveira
1,2,6,7 and
Diana Dias da Silva
1,5,8,9,10
1
Associate Laboratory i4HB—Institute for Health and Bioeconomy, University Institute of Health Sciences—CESPU, Avenida Central de Gandra 1317, 4585-116 Gandra, Portugal
2
UCIBIO—Research Unit on Applied Molecular Biosciences, Translational Toxicology Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), Avenida Central de Gandra 1317, 4585-116 Gandra, Portugal
3
Instituto Universitário Militar, CINAMIL, Unidade Militar Laboratorial de Defesa Biológica e Química, Avenida Doutor Alfredo Bensaúde, 4 piso, do LNM, 1849-012 Lisbon, Portugal
4
ULSEDV—Unidade Local De Saúde De Entre Douro Vouga, Unidade de Santa Maria da Feira e Hospital S. Sebastião, Rua Dr. Cândido Pinho, 4520-211 Santa Maria da Feira, Portugal
5
UCIBIO—Applied Molecular Biosciences Unit, Forensics and Biomedical Sciences Research Laboratory, University Institute of Health Sciences (1H-TOXRUN, IUCS-CESPU), Avenida Central de Gandra 1317, 4585-116 Gandra, Portugal
6
Department of Public Health and Forensic Sciences and Medical Education, Faculty of Medicine, University of Porto, Alameda Prof. Hernâni Monteiro, 4200-319 Porto, Portugal
7
FOREN—Forensic Science Experts, Avenida Dr. Mário Moutinho 33-A, 1400-136 Lisbon, Portugal
8
REQUIMTE/LAQV, ESS, Polytechnic of Porto, Rua Dr. António Bernardino de Almeida, 4200-072 Porto, Portugal
9
Associate Laboratory i4HB—Institute for Health and Bioeconomy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
10
UCIBIO—Applied Molecular Biosciences Unit, Laboratory of Toxicology, Faculty of Pharmacy, University of Porto, Rua de Jorge Viterbo Ferreira 228, 4050-313 Porto, Portugal
*
Author to whom correspondence should be addressed.
Microorganisms 2024, 12(9), 1920; https://doi.org/10.3390/microorganisms12091920
Submission received: 3 September 2024 / Revised: 15 September 2024 / Accepted: 18 September 2024 / Published: 21 September 2024
(This article belongs to the Special Issue Progress and Challenges in Antimicrobial Resistance)

Abstract

:
Antimicrobial resistance (AMR), frequently considered a major global public health threat, requires a comprehensive understanding of its emergence, mechanisms, advances, and implications. AMR’s epidemiological landscape is characterized by its widespread prevalence and constantly evolving patterns, with multidrug-resistant organisms (MDROs) creating new challenges every day. The most common mechanisms underlying AMR (i.e., genetic mutations, horizontal gene transfer, and selective pressure) contribute to the emergence and dissemination of new resistant strains. Therefore, mitigation strategies (e.g., antibiotic stewardship programs—ASPs—and infection prevention and control strategies—IPCs) emphasize the importance of responsible antimicrobial use and surveillance. A One Health approach (i.e., the interconnectedness of human, animal, and environmental health) highlights the necessity for interdisciplinary collaboration and holistic strategies in combating AMR. Advancements in novel therapeutics (e.g., alternative antimicrobial agents and vaccines) offer promising avenues in addressing AMR challenges. Policy interventions at the international and national levels also promote ASPs aiming to regulate antimicrobial use. Despite all of the observed progress, AMR remains a pressing concern, demanding sustained efforts to address emerging threats and promote antimicrobial sustainability. Future research must prioritize innovative approaches and address the complex socioecological dynamics underlying AMR. This manuscript is a comprehensive resource for researchers, policymakers, and healthcare professionals seeking to navigate the complex AMR landscape and develop effective strategies for its mitigation.

1. Introduction

Antimicrobial resistance (AMR) is an outstanding challenge for contemporary medicine and public health, in which microorganisms adapt to withstand the effects of antimicrobial agents, rendering previous treatments ineffective [1,2]. AMR is responsible for increased morbidity, mortality, and healthcare costs, corresponding to nearly 700,000 deaths worldwide every year, which is estimated to increase to up to 10 million deaths per year by 2050. In 2014, the World Health Organization (WHO) included AMR among the top ten global health threats, a position that remains the same almost ten years later [3].
AMR’s emergence and dissemination is accelerated by the inappropriate use of antimicrobial agents (agriculture, veterinary and human health), poor infection control practices, inadequate sanitation, and improper food handling (Figure 1) [4,5,6]. At its core, AMR is driven by genetic changes within microbial populations, enabling them to survive and proliferate in the presence of antimicrobial drugs [7,8].
Infections caused by resistant organisms are often associated with augmented mortality, morbidity, lengths of hospitalization, and healthcare costs (i.e., an additional cost of USD 6000–30,000 when compared to those with susceptible infections or no infection) [9,10,11]. Moreover, the increase in AMR also threatens the effectiveness of medical procedures and limits the available therapies [9,12]. Therefore, addressing AMR requires a multifaceted approach comprising various sectors and stakeholders [12,13]. Effective strategies include the following:
  • Promoting ASPs to optimize the use of antimicrobial agents;
  • Implementing robust IPC strategies;
  • Investing in the research and development (R&D) of new antimicrobial agents and alternative therapies;
  • Strengthening surveillance and monitoring systems to track the emergence and dissemination of resistant pathogens.
Curiously, AMR’s appearance preceded the discovery of the first antibiotic. Salvarsan, belonging to arsphenamines, was used to treat syphilis (1910–1940s), and resistance to it emerged in the early 1920s [14]. Later, in 1928, penicillin’s introduction revolutionized medicine, offering a potent weapon against bacterial infections and saving millions of lives during the Second World War [15]. However, in less than ten years, as foreseen by Alexander Flemming himself, the extensive use of antibiotics and the selective pressure exerted led to the first reports of penicillin-resistant bacteria and, by the 1950s, the appearance of the first “hard-to-treat” infections [15,16,17]. The following decades saw rapid expansion in the discovery and development of new classes of antibiotics. However, with each new antibiotic introduced, resistance soon followed, underscoring the evolutionary arms race between microbes and medicine [18,19]. The 21st century has witnessed alarming trends in antimicrobial resistance, with the emergence of multidrug-resistant pathogens such as methicillin-resistant Staphylococcus aureus (MRSA), extended-spectrum β-lactamase-producing Enterobacteriaceae (ESBL), multidrug-resistant Mycobacterium tuberculosis (MDR-TB), and MDR Pseudomonas aeruginosa [20,21,22].
In agriculture, the widespread use of antibiotics for growth promotion and disease prevention significantly contributes to the emergence of resistant strains transmitted through food chains, posing risks to human health through the consumption of contaminated meat products [23,24]. Furthermore, agricultural runoff contaminates water bodies and soil with antibiotic residues, facilitating antimicrobial resistance genes’ (ARGs) dissemination among environmental bacteria [25]. Sustainable agricultural practices must prioritize judicious antimicrobial use and explore alternative strategies to mitigate the environmental dissemination of AMR (Figure 1) [26,27].
The environmental dimension of AMR accentuates its complexity and global ramifications. Resistant bacteria, genes, and antibiotics persist in diverse environmental reservoirs (e.g., soil, water, and air) that serve as hotspots for genetic exchange between microbial communities, fostering the evolution and dissemination of resistance traits. Anthropogenic activities (e.g., pharmaceutical manufacturing and improper medical waste disposal) further exacerbate environmental contamination with antimicrobial agents. Effective AMR management requires holistic strategies encompassing environmental monitoring, mitigation measures, and sustainable practices to mitigate ecological perturbations and safeguard biodiversity (Figure 1) [28,29,30].
Addressing AMR demands concerted efforts from diverse healthcare, agriculture, and environmental stakeholders [31,32]. Multidisciplinary collaboration fosters knowledge exchange, innovation, and the implementation of comprehensive strategies for AMR mitigation [31,32,33]. Surveillance systems are pivotal in monitoring resistance patterns, guiding antimicrobial stewardship programs, and informing policy interventions. Furthermore, research must prioritize developing novel antimicrobial agents, vaccines, and alternative therapies to counteract resistance mechanisms. Educational initiatives aimed at healthcare professionals, farmers, and the public are essential in raising awareness and fostering responsible antimicrobial use practices [32].
This manuscript provides a comprehensive exploration of AMR, structured into eight sections covering its development, dissemination, impact, and strategies for mitigation. Section 1 explains the genetic basis of AMR emergence in diverse microorganisms. Section 2 examines AMR’s global prevalence and distribution and elucidates the factors contributing to AMR dissemination. Section 3 debates the impact of AMR on human health, presents the strategies to address AMR, and highlights the role of surveillance programs in monitoring resistance and guiding interventions. Section 4 emphasizes the One Health approach by recognizing the interconnectedness of human, animal, and environmental health in addressing AMR. Section 5 highlights the AMR impacts on health and healthcare systems, challenges in treatment, economic costs, global implications, the need for coordinated action, and the social and ethical dimensions of this problem. Section 6 explores ASPs and IPC strategies in combating AMR, addressing responsible antibiotic use, infection prevention, global surveillance, and emerging novel approaches. Section 7 emphasizes the need for cross-sector collaboration to address AMR through the interconnectedness of human, animal, and environmental health. Section 8 reviews policies and regulations promoting responsible antimicrobial use and highlights challenges in their global implementation. Section 9: explores emerging AMR threats, the need for new treatments, and the importance of global collaboration in combating AMR. Finally, Section 10 presents the main conclusions from this work including the urgent need for a coordinated global response to AMR, integrating APS, IPC, R&D, and cross-sector collaboration to protect public health and ensure sustainable treatment solutions.

2. Materials and Methods

The manuscript selection process followed a structured approach with precise inclusion and exclusion criteria. Nearly 21,500 manuscripts were retrieved from public article databases such as PubMed (U.S. National Library of Medicine) and Google Scholar, using the keywords “Antimicrobial Resistance”, “Genetic Mechanisms”, “Epidemiology”, “Impact”, “Antibiotic Stewardship Program”, “One Health Approach”, “Policy and Regulation”, “Future Directions”, and “Challenges”. After duplicate removal, a date limit was applied (2014 and 2024). Only peer-reviewed articles with robust methods and high-quality data were considered for inclusion (journals belonging to Q1 and Q2). Manuscripts that demonstrated practical relevance, showing clear applications to casework scenarios, were also included. As exclusion criteria, manuscripts that did not directly address the core topics, lacked scientific rigor, or were outdated were removed from the analysis. The research team analyzed more than 500 manuscripts at the end of this selection process. This process ensured that the revised manuscript incorporated the most pertinent and high-quality research, providing a comprehensive update on AMR (Figure 2).

3. Mechanisms of Antimicrobial Resistance

3.1. Development of Antimicrobial Resistance

AMR, a significant global health challenge, poses a threat to the effectiveness of drugs that combat microorganisms (i.e., bacteria, viruses, fungi, and parasites), which have developed diverse biological mechanisms to evade the effects of antimicrobial agents (Table 1).

3.1.1. Antimicrobial Resistance Mechanisms: Bacteria

In the AMR context, bacteria are some the most extensively studied microorganisms, mainly due to their capacity to develop several mechanisms that enable them to survive antibiotic exposure. In one of these mechanisms, known as target modification, bacteria alter their molecular targets for antibiotics, preventing the drugs from effectively binding [34]. For instance, mutations in bacterial ribosomal RNA (rRNA) hinder the binding of aminoglycosides, enabling the bacteria to continue protein synthesis despite the antibiotic’s presence [35]. The recent study by Babosan and co-workers demonstrated the importance of nonessential transfer RNA (tRNA) and rRNA modifications in the bacterial response to antibiotic stress, while it proposes that differential translation and codon decoding play an essential role in the stress response [36].
In some bacteria, efflux pumps actively transport a broad range of drugs outside the cytoplasm, reducing their intracellular concentrations and allowing the bacteria to survive at higher antibiotic concentrations [37,38]. These membrane proteins are particularly significant in antibiotic resistance to tetracyclines, fluoroquinolones, and macrolides [39].
Bacteria produce enzymes, often involved in the biosynthesis of the cell wall, nucleic acids, and metabolites, which serve as a direct target for antibiotics. Structural changes in these enzymes also change the bacteria’s resistance profiles [40]. For instance, β-lactamases hydrolyze the β-lactam ring in penicillin, cephalosporin, and carbapenems (relatively resistant to β-lactamase), making these antibiotics ineffective [41,42]. Therefore, β-lactam antibiotics are frequently used in association with beta-lactamase inhibitors [41,43]. Nevertheless, continuous bacterial evolution and mutation lead to the appearance of new β-lactamases that can resist these inhibitors [42].
Some bacteria, especially Gram-negative ones, possess a mechanism that reduces the cell membrane permeability, preventing antibiotics from entering the cell. Therefore, the bacterial outer membrane acts as a selective diffusion barrier, limiting antibiotic influx [44] by two main pathways: diffusion using protein channels (hydrophilic antibiotics) and a lipid-mediated pathway (hydrophobic antibiotics). Modifications in the lipid and protein compositions of the outer membrane can significantly impact the bacterial sensitivity to antibiotics [45,46]. Better knowledge of the molecular basis of outer membrane permeability and the physicochemical parameters influencing antibiotic translocation through porins is essential in combating antibiotic resistance [44].
Finally, some bacterial strains form biofilms, becoming highly resistant to antibiotics due to the presence of a self-produced protective extracellular matrix that limits drug influx, while the bacteria within, often presenting lower metabolic activity and longer doubling rates, are less susceptible to antibiotics targeting actively dividing cells [47,48]. The biofilm structure presents two advantages concerning bacterial survival: it creates gradients of nutrients and oxygen, leading to diverse metabolic states and promoting antibiotic tolerance, while facilitating HGT and inducing higher mutation frequencies [49]. These mechanisms provide critical knowledge for the development of new strategies to combat biofilm-associated infections and address antibiotic resistance [49,50].

3.1.2. Antimicrobial Resistance Mechanisms: Viruses

Antiviral resistance arises through mechanisms that are distinct from those in bacteria. These differences are because viruses rely on host cells for replication and do not possess the same cellular targets as bacteria [51,52].
Traditional therapeutical approaches use antiviral drugs targeting viral enzymes (e.g., reverse transcriptase, protease, or RNA polymerase). Mutations in the genes encoding these enzymes reduce the drug’s binding affinity, allowing the virus to continue replicating. For instance, HIV resistance is due to reverse transcriptase and protease inhibitors [53]. To solve this problem, new targets based on components that are essential for the virus but not for the host (e.g., small interfering RNA (siRNA), CRISPR, and small-molecule inhibitors targeting cellular kinases) are being studied and present promising results [54]. These new therapeutical developments also include proteolysis-targeting chimeras (PROTACs), which leverage the ubiquitin–proteasome system for protein degradation. Compounds targeting host factors (e.g., human dihydroorotate dehydrogenase) have also shown good results as potential broad-spectrum antivirals [76].
Some antivirals block viral entry into host cells by targeting viral surface proteins. Mutations in these proteins can prevent the drug from binding, allowing the virus to infect cells despite treatment. This mechanism is relevant in resistance to entry inhibitors used in treating HIV [77].
Finally, some antiviral drugs require activation by viral or host enzymes to become effective [54]. Therefore, mutations that reduce the efficiency of this activation result in resistance to these drugs. For instance, acyclovir resistance in herpes simplex virus (HSV) is due to mutations in the viral thymidine kinase gene (TK), resulting in complete activity loss, reduced expression, or altered affinity [78].

3.1.3. Antimicrobial Resistance Mechanisms: Fungi

Fungal pathogens develop resistance to antifungal agents through mechanisms analogous to those in bacteria and viruses but are characterized by unique aspects associated with their specific biology [79]. The emergence of multidrug-resistant fungi (e.g., Candida auris and Aspergillus spp.) has significantly complicated the treatment options [80]. Better knowledge of these resistance mechanisms would assist in developing alternative therapeutic strategies to combat refractory mycoses [80,81].
Fungi can alter the targets of antifungal drugs. For instance, mutations in the lanosterol 14-alpha-demethylase gene (ERG11), the target for antifungals, result in several aminoacidic changes that change the drug’s capacity to bind and inhibit the gene, resulting in decreased susceptibility to azoles (fluconazole, itraconazole, and voriconazole) [55].
Fungi also possess efflux pumps, mainly belonging to the ATP-binding cassette (ABC) or major facilitator superfamily (MFS), that expel antifungal drugs from fungal cells [56,57]. The overexpression of these pumps is a common resistance mechanism to azoles and other antifungals in Candida species and other fungi [58,59,60]. Efflux-mediated resistance can be surpassed by alternative antifungals that are not pump substrates, target transcriptional regulators and stress response pathways, block the energy supply, and directly inhibit these pumps [61].
Fungi can modify their cell wall compositions to reduce the binding or penetration of antifungal agents. For instance, resistance to echinocandins primarily arises from mutations in the FKS gene, which encodes the catalytic subunit of glucan synthase and is associated with reduced sensitivity to the enzyme [62]. By modifying echinocandin structures with some dehydroxylated derivatives, it is possible to surpass this resistance mechanism [63].
Fungi can also form biofilms, which are particularly problematic in infections involving medical devices (e.g., catheters, pacemakers, and prosthetic joints) [64]. Due to their structure—mainly the extracellular matrix—biofilms are intrinsically resistant to conventional antifungal therapeutics [65]. Developing novel strategies to prevent and treat biofilm formation is crucial in managing these difficult-to-treat infections [66].

3.1.4. Antimicrobial Resistance Mechanisms: Parasites

Parasites (e.g., protozoa and helminths) are becoming more resistant to antiparasitic drugs [82,83]. Resistance is induced by frequent treatment, underdosing, non-compliance with the duration of the dosing regimen, poor drug quality, drug interactions, poor or erratic absorption and misdiagnosis, and parasite genetics [82]. Several mechanisms for antiparasitic drug resistance have been proposed [82].
Parasites can accumulate mutations in the genes encoding the molecular targets of antiparasitic drugs, leading to reduced drug efficacy. The genes involved differ from species to species, and the same species can exhibit the involvement of several genes. For instance, in Plasmodium falciparum (malaria parasites), antifolate drug resistance involves point mutations in dihydropteroate synthase (dhps) and dihydrofolate reductase (dhfr) [84]; mutations in the Kelch13 protein (K13), involved in multiple intracellular processes including hemoglobin catabolism, are associated with artemisinin resistance [67,68]; and overexpression or mutations in the digestive vacuole membrane-bound ABC transporter PfMDR1 are associated with resistance to chloroquine and other 4-aminoquinolines [69,70]. In Toxoplasma gondii (toxoplasmosis), resistance to auranofin is associated with mutations in redox-relevant genes, enhancing the parasite’s ability to manage oxidative stress [71].
Another mechanism is associated with the efflux pumps that actively remove antiparasitic drugs from their cells. This mechanism has been observed in P. falciparum, where these pumps enable the efflux of drugs (e.g., chloroquine, amodiaquine, and piperaquine) from the digestive vacuole, blocking their antimalarial action [70].
Parasites can alter their metabolic pathways to bypass the effects of drugs. For instance, mutations in the genes encoding enzymes in the glycolytic pathway confer resistance to drugs targeting this pathway in trypanosomes (sleeping sickness) [72].
Finally, some antiparasitic drugs require activation within the parasite to exhibit a therapeutical effect—a mechanism known as a “magic bomb” [73]. Modifications in this activation process can lead to resistance. For instance, nifurtimox and benznidazole, often used to control Trypanosoma cruzi (Chagas disease), are activated by a mitochondrial nitroreductase (NTR) that, when downregulated, results in cross-resistance [74]. Artemisinin-based drugs for malaria require heme-mediated activation, and resistance is associated with changes in the parasite’s life cycle and enhanced stress responses [75].

3.2. Genetic Mechanisms of Antimicrobial Resistance

The development of AMR is mediated by several genetic mechanisms that enable microorganisms to adapt and survive in the presence of these agents [85,86]. These mechanisms will be covered in the following sections.

3.2.1. Mutations

Mutations, a fundamental genetic process, introduce random changes in an organism’s DNA sequence. These can occur spontaneously or be induced by external factors, such as antimicrobial exposure [87]. Spontaneous mutations naturally occur during DNA replication, resulting in the emergence of resistant strains if the mutation affects a gene involved in drug susceptibility [88]. Despite the action of replication error repair systems, some errors escape repair due to temporal constraints [89]. For example, a point mutation in the gene encoding bacterial DNA gyrase confers resistance to quinolones by reducing the drug’s binding affinity [90]. The rate of spontaneous mutation during replication varies among microorganisms and even between those that are closely related. For instance, herpes simplex virus type 2 (HSV-2) presents a higher mutation rate than HSV-1 (laboratory strains: 9–16-fold; clinical isolates: nearly 30-fold) [91]. However, the emergence of resistance often results from mutations in several genes. For instance, in M. tuberculosis, resistance to PA-824 is associated with mutations in genes related to prodrug activation and the F420 biosynthetic pathway (genes: ddn, fgd1, fbiA, fbiB, and fbiC), resulting in reduced sensitivity [92].
Exposure to sub-lethal concentrations of antimicrobial agents induces DNA damage and genomic instability, leading to a higher mutation rate in microorganisms and, potentially, the emergence of new AMR—antibiotic-induced mutagenesis [93,94]. This phenomenon occurs due to the production of reactive oxygen species (ROS) and the consequent oxidative stress, which induces error-prone polymerases, misbalancing nucleotide metabolism or directly affecting DNA [95]. As a result, a wide range of genetic perturbations can be observed, from single-nucleotide polymorphisms (SNPs) to severe chromosomal rearrangements [94]. It has been suggested that heavy metals also increase the mutation rates and allow the emergence of mutants with resistance to multiple antibiotics, highlighting the potential role of environmental contaminants in promoting antibiotic resistance [96]. Besides these two, other factors may contribute to the increased mutation rate in bacteria and the potential emergence of antimicrobial resistance, including chemical pollutants (e.g., biocides, nanoparticles, and disinfection byproducts) and non-chemical stressors (e.g., ultraviolet radiation, electrical stimulation, and starvation) [97].

3.2.2. Horizontal Gene Transfer

Horizontal gene transfer (HGT), an essential process for the evolution of various organisms, allows for genetic material to be transferred between distinct species, bypassing traditional modes of inheritance [98,99]. This process contributes to genome plasticity and adaptive evolution in prokaryotes and eukaryotes by introducing new genes and functions or serving as a source of phenotypic innovation and niche adaptation [100,101]. HGT has been implicated in the emergence and dissemination of pathogenic traits in eukaryotes, such as antibiotic resistance and virulence factors [102,103]. There are three classical modes of HGT: conjugation, transformation, and transduction. Recently, a fourth mode of DNA transfer through extracellular vesicles—vesiduction—has been proposed [104].
Conjugation involves the unidirectional transfer of genetic material between a donor and a recipient bacterium through direct cell-to-cell contact [105]. Despite small differences in Gram-negative and -positive bacteria, the transfer involves conjugative plasmids and integrating conjugative elements (ICEs, including transposons) that resort to complex machinery that includes a type IV secretion system (T4SS), relaxosome, and pilus [106,107]. This process is regulated by plasmid- and host-encoded factors responding to environmental and physiological signals [108,109]. For instance, in Bacillus subtilis, the conjugative plasmid pLS20 uses several control levels (e.g., a quorum-sensing system and strict repression of the main conjugation promoter Pc), ensuring that conjugation genes are only expressed under favorable conditions [110]. However, phage plasmids, which carry numerous clinically important resistance genes usually included in integrons, achieve resistance through infection and lysogenic conversion, potentially transferring genes to distant bacteria without cell-to-cell contact [111]. Plasmids often carry multiple resistance genes, enabling the recipient bacterium to simultaneously acquire resistance to several antibiotics in a single conjugation event [112]. Conjugative plasmids have developed mechanisms to disable host defenses and compete with other plasmids, facilitating their transmission and establishment in new bacterial hosts [113]. The mecA gene, responsible for the antibiotic resistance observed in MRSA, encodes for penicillin-binding protein 2a (PBP2a) with low affinity for β-lactam antibiotics (e.g., methicillin and other penicillin derivatives). Normally, β-lactam antibiotics bind to penicillin-binding proteins (PBPs) and inhibit bacterial cell wall synthesis. In MRSA, PBP2a’s altered structure allows the cell wall to be biosynthesized, even in the presence of these antibiotics, rendering them ineffective. The gene is located on a mobile genetic element known as the staphylococcal cassette chromosome mec (SCCmec), which can be horizontally transferred between Staphylococcus species, further disseminating resistance [114]. Carbapenemases, enzymes produced by Gram-negative bacteria, are responsible for carbapenem resistance. These enzymes belong to different classes, with the most notable being the Klebsiella pneumoniae carbapenemase (KPC), New Delhi metallo-beta-lactamase (NDM), Verona integron-encoded metallo-beta-lactamase (VIM), and oxacillinase (OXA). Carbapenemases break down the β-lactam rings of carbapenems, preventing PBPs’ inhibition and neutralizing their antibacterial effects. Plasmids encoding carbapenemases, such as KPC, NDM, and OXA-48-like, found in Enterobacteriaceae (CRE), Acinetobacter, and Pseudomonas, facilitate the rapid dissemination of carbapenem resistance [115]. Plasmids carrying aminoglycoside-modifying enzyme (AME) genes, such as aac(6′)-Ib and aph(3′)-VI, which often co-occur with other resistance genes, enable bacteria to resist aminoglycosides and β-lactams [116]. The co-occurrence of AME and carbapenemase genes, such as blaNDM-1 and blaKPC-2, observed in Enterobacterales, further limits the available treatment options [117]. Colistin resistance, due to plasmids with mcr genes (mcr-1 and mcr-10) in E. coli and other species, further complicates the treatment of infections caused by multidrug-resistant Gram-negative bacteria, especially in co-occurrence with other antibiotic resistance genes [118]. Plasmids carrying extended-spectrum β-lactamase (ESBL) genes, such as TEM, SHV, and CTX-M, are frequently found in bacteria such as E. coli and K. pneumoniae and hydrolyze β-lactam antibiotics and often carry additional genes, resulting in multidrug resistance to aminoglycosides and fluoroquinolones [119]. It has been proven that when several bacterial species are present in co-cultures, interspecies plasmid transfer may occur, facilitating ARGs’ dissemination among bacteria and allowing the emergence of multidrug-resistant bacteria [120].
Natural transformation, where bacteria uptake and incorporate environmental DNA, is an important mechanism for AMR acquisition, influenced by biotic and abiotic factors, (e.g., environmental stresses and antimicrobials’ presence) [121,122]. This process facilitates the transfer of large antibiotic resistance islands between bacterial species, particularly in pathogens such as Acinetobacter baumannii [123]. Moreover, bacteria can incorporate these mobile genetic elements carrying ARGs and then remove them, better adjusting to intermittent stress exposure and maximizing their fitness [124].
Transduction involves phages (bacteriophages) that carry bacterial DNA, including ARG and virulence genes, between bacterial hosts [125]. This process plays an important role in acquiring, maintaining, and disseminating antibiotic resistance [126]. For instance, it contributes to the dissemination of resistance genes among foodborne pathogens, especially in Enterobacteriaceae [127]. Transduction can occur through three different mechanisms: generalized (i.e., phages package chromosomal and/or bacterial DNA and transfer it to another bacterium), specialized (i.e., limited to the transfer of specific sets of genes when viral and bacterial host DNA are encapsidated as a hybrid molecule), and lateral transduction (i.e., a natural part of the phage life cycle that transfers bacterial chromosomal DNA at frequencies at least 1000-fold greater than previous mechanisms) [128].
Vesiduction, a HGT mechanism that involves membrane vesicles (MVs), has been recently proposed as a fourth mode of DNA transfer [104]. MVs—particularly outer membrane vesicles (OMVs) in Gram-negative bacteria—play important roles in bacterial communication, secretion, and self-defense, including protection against antibiotics [129]. For instance, in Enterococcus faecium, vancomycin exposure induces the increased production of MVs containing vanA and vanB resistance genes [130].

3.2.3. Selection Pressure and Its Role in AMR Development

Selection pressure is the environmental force that drives AMR evolution. When a given microorganism is exposed to an antimicrobial agent, those with mutations conferring either resistance or acquired resistance genes have a survival advantage and proliferate; those that are susceptible are eliminated, leading to the dominance of resistant strains within the population [131]. The environment acts as an AMR reservoir, with hospital wastewater and agricultural runoff contributing to the dissemination of resistant microorganisms and its ARGs in the soil, water, and even air [29]. Diverse factors (e.g., infection control standards, sanitation, and travel) affect the role of selection pressure in AMR development, being essential to understand the ecological and evolutionary processes leading to ARGs’ mobilization and dissemination in order to prevent it [132].

Antimicrobial Use

Antimicrobials’ widespread and often inappropriate use in healthcare, agriculture, and animal husbandry creates strong selection pressure for the development and dissemination of resistance [133]. Agriculture, including livestock and aquaculture, significantly contributes to AMR development due to extensive antimicrobial use, often at subtherapeutic levels [134,135], inducing strong selection pressure for resistant bacteria, which can potentially be transmitted to humans through the food chain, direct animal contact, and environmental contamination [133]. This problem is exacerbated by the large, interconnected human population and the frequent misuse of antimicrobials in healthcare [136]. Addressing AMR requires an integrated and multifaced approach, comprising improved surveillance, reduced antimicrobial usage in agriculture and healthcare, the development of new drugs, and education programs to change public perceptions of antimicrobial use [133,136].

Suboptimal Dosing

Inadequate antibiotic dosing or incomplete courses are also responsible for AMR emergence, and resistant infections often occur during or after antibiotic therapy [137]. Protocols developed to optimize pharmacokinetic/pharmacodynamic principles in antibiotic dosing maximize their efficacy and suppress resistance [138]. On the other hand, antibiotic exposure also induces hypervirulence in pathogens, potentially worsening disease outcomes [139]. ASPs promote prudent antimicrobial use, including appropriate diagnostics and IPC strategies, limiting AMR emergence in clinical settings by applying interventions such as guidelines, pre-authorization, reviews, audits, feedback, and education [140].

Environmental Factors

Water, soil, and sediments act as reservoirs and vectors for resistant microorganisms and ARGs [29]. Antibiotic residues disseminated in the environment, particularly near intensive animal husbandry farms, create selective pressure for resistant organisms at concentrations below the minimum inhibitory concentration [141]. Moreover, anthropogenic activities (e.g., hospital and municipal discharges) disseminate resistant bacteria and their genes in aquatic environments [142]. To address this issue, further research is required, focusing on the relationship between environmental antibiotic concentrations and resistance prevalence, improved wastewater treatment, and management practices [143,144].

3.3. Interplay between Genetic Mechanisms and Selection Pressure

The interplay between genetic mechanisms and selection pressure in AMR is complex and multifaceted. On the one hand, bacteria adapt to host defense systems and antimicrobial agents through diverse mechanisms (e.g., structural changes, enzymatic processes, and gene regulation) [145]. On the other hand, mobile genetic elements (e.g., plasmids and transposons) disseminate ARGs, and non-genetic factors (e.g., transcriptional heterogeneity) facilitate the development of genetic drug resistance [146]. Additionally, epigenetic modifications (e.g., DNA methylation and non-coding RNA regulation) contribute to bacterial adaptation and antibiotic resistance [147]. Finally, rapid AMR dissemination is further intensified by processes of genetic recombination and selective evolutionary pressure induced by the inappropriate use of antimicrobial drugs [148].
Recently, bioinformatics approaches, including whole-genome sequencing (WGS) and other high-throughput technologies, have become valuable tools for the study of AMR mechanisms and the discovery of new resistance signatures [149].

3.4. Clinical and Public Health Implications

In low- and middle-income countries (LMICs), AMR is a major public health concern [150]. Understanding the mechanisms of AMR is essential in developing effective strategies to combat this global health threat. For instance, the ESKAPE pathogens (E. faecium, S. aureus, K. pneumoniae, A. baumannii, P. aeruginosa, and Enterobacter species), along with Clostridioides difficile, are responsible for most nosocomial infections worldwide. This not only demonstrates the challenges posed by multidrug-resistant organisms but also highlights the importance of understanding their specific resistance mechanisms for the development of novel antimicrobial agents [151,152,153].
To reduce AMR emergence, clinical practitioners should use antibiotics judiciously. Nearly 50% of antibiotic prescriptions are considered unnecessary or inadequate (e.g., overprescribing and switching to same-class agents for therapy failure) [154] due to factors such as uncertain diagnosis and patient influences [152]. To address the AMR crisis, clinicians must implement adequate strategies, such as rapid microbiological diagnostics, the use of inflammation markers, shorter antibiotic courses, and individualized therapies [153]. Healthcare institutions should develop and enforce guidelines for appropriate antimicrobial use [152]. The U.S. CDC’s 2019 Antibiotic Resistance Threats Report provides valuable insights for frontline providers on the national AMR crisis [154].
Public health initiatives, such as education and awareness campaigns, should focus on improving the use of antimicrobial drugs, both in human and animal health [155]. Moreover, implementing effective IPC strategies, such as improved hygiene, sanitation, and facility decontamination, is critical to prevent the dissemination of resistant microbes [150]. Complementary surveillance programs have been developed to monitor AMR trends and guide interventions to reduce AMR dissemination [156]. The One Health approach, which recognizes the interconnectedness of human, animal, and environmental health, addresses AMR comprehensively [157].
Novel and innovative vaccine technologies (e.g., reverse vaccinology) and adjuvants have been proven effective in targeting multidrug-resistant bacteria [158], with their wider application being limited only by the need to better understand immunological mechanisms and identify correlates of protection [159]. Moreover, new approaches for antibiotic delivery and alternative modalities, such as targeting host factors and blocking bacterial virulence, are also being explored [160]. Rapid, accurate, point-of-care diagnostics not only allow for improved prescribing practices but also enable more efficient clinical trials [161].

4. Epidemiology of Antimicrobial Resistance

AMR affects human and veterinary medicine and threatens to reverse decades of progress in the treatment of infectious diseases [162,163]. Therefore, population-based studies are required to understand the burden, distribution, and determinants of AMR, as well as to develop effective prevention and management strategies [164]. These studies will provide unbiased prevalence estimates and overcome the limitations of laboratory-based surveillance (e.g., weak laboratory capacity, poor health systems, and limited resources) common in LMICs [165,166]. To address this global challenge, the WHO has developed action plans to promote new antibiotics, reduce antibiotic misuse, and improve surveillance [167]. The metagenomic analysis of urban sewage has been proposed as an economically feasible approach for continuous global AMR surveillance [168].

4.1. Global Prevalence and Distribution of Antimicrobial Resistance

AMR’s global distribution significantly varies across worldwide locations (Figure 3) [168]. Multidrug-resistant organisms (MDROs) (e.g., MRSA; vancomycin-resistant Enterococci—VRE; CRE; hospital-associated C. difficile infection—HA-CDI; and community-associated C. difficile infection—CA-CDI) have become gradually more common in healthcare settings and the community [169,170,171].
MRSA is globally widespread, particularly in North America, Europe, and East Asia, mainly due to acquiring virulence factors and resistance to multiple antibiotics [172]. It has been proposed that international travel has contributed to MRSA dissemination across continents, potentially replacing the endemic strains with these more transmissible variants [173].
The SENTRY Antimicrobial Surveillance Program reported that VRE is responsible for more than 8% of enterococcal isolates globally, with North America having the highest prevalence at 21.6% [174].
CRE presents a critical issue in healthcare settings, especially in South and Southeast Asia, where prolonged hospital stays are associated with high mortality rates, mainly due to limited access to effective treatments [175]. The frequent treatment failures and the increased economic burden on patients and nations has forced the WHO to prioritize CRE as a top pathogen of public health importance to contain its dissemination [176].
Another important emerging MDRO is C. difficile, a nosocomial pathogen with a global distribution, primarily affecting individuals with disrupted gut microbiota due to antibiotic use [177]. Epidemiological data indicate its presence in various environmental reservoirs, including healthcare settings, where its transmission is facilitated [178]. The emergence of hypervirulent strains like ribotype 027 has exacerbated this trend, driving higher morbidity and mortality rates worldwide [178].
Viral AMR is also a growing concern since several viruses (e.g., HIV, hepatitis B and C, influenza, herpes simplex, varicella-zoster) have become resistant to antiviral therapies [179,180,181]. This is particularly problematic in regions such as sub-Saharan Africa, where the expansion of antiretroviral therapy (ART) is associated with poor staff capacities and infrastructure to support consistent treatment adherence, leading to the emergence of resistant strains [182,183].
Although less discussed, fungal AMR also poses a significant threat to immunocompromised patients, mainly those with hematologic malignancies and organ transplants [184,185]. Some C. auris strains are resistant to multiple antifungal agents (e.g., azoles, amphotericin B, and echinocandins) [186]. This pathogen, first identified in Japan in 2009, has rapidly disseminated across Asia, Europe, and the Americas [187]. Its ability to persist on surfaces, colonize patients for extended periods, and be efficiently transmitted from person to person makes this pathogen difficult to control [188], being often associated with high mortality rates [189]. Azole resistance in Aspergillus species, mainly A. fumigatus, is a growing concern worldwide, and the transboundary spread of this microorganism in many countries has been suggested [190].
AMR in parasites, notably in malaria-causing P. falciparum, is also a major concern, significantly threatening efforts to control and eliminate this disease [191]. Resistance to artemisinins has been disseminated from Southeast Asia, in the Greater Mekong Subregion, to Africa and South America [192]. Chloroquine resistance, first reported in South America and Southeast Asia (early 1960s), was disseminated to East Africa (late 1970s) and then westward [193]. Finally, the emergence and dissemination of resistance to sulfadoxine–pyrimethamine occurred in Southeast Asia and South America, subsequently spreading to Africa, similar to that observed for chloroquine [194].

4.2. Trends in Antimicrobial Resistance over Time and across Different Regions

Although with significant regional variations, AMR’s prevalence is steadily increasing worldwide. Monitoring data from various surveillance networks (e.g., European Antimicrobial Resistance Surveillance Network, EARS-Net) reveal that the incidence of resistant infections has significantly risen over recent decades [195]. For instance, while the prevalence of MRSA has decreased in some European countries due to rigorous IPC strategies, it remains high in others [196]. Moreover, there has been an alarming rise in resistance to last-resort antibiotics, such as colistin, especially in Gram-negative bacteria [197].
The distribution of AMR is heavily influenced by regional factors, including the misuse of antibiotics, inadequate living conditions, poor healthcare infrastructure, and deficient public health policies [198]. For instance, in many African countries, weak healthcare systems, limited access to proper diagnosis, and non-prescription antibiotic dispensing have contributed to high levels of AMR [199,200]. Resistant infections, such as those caused by Salmonella typhi and K. pneumoniae, are particularly prevalent, complicating treatment protocols. International initiatives, such as the Global Antimicrobial Resistance Surveillance System (GLASS; WHO) and AMRSNET (Africa CDC), aim to enhance AMR surveillance and control in this continent [200].
South and Southeast Asia are recognized as hotspots for AMR, mainly driven by the high usage of antibiotics in healthcare and agriculture [201]. This region has experienced rapid economic growth associated with increased animal product consumption, contributing to AMR emergence [202]. Antibiotic use for growth promotion and treatment is prevalent, with a significant overlap between critically important antibiotics used in animals and humans [203]. Easy access to antibiotics, coupled with limited veterinary care, drives inappropriate use [203]. While some countries have policies to address AMR, most lack detailed implementation plans [204]. Developing locally relevant strategies based on situation analysis is critical, as one-size-fits-all policies may be inappropriate for the region’s unique context [204]. In contrast, Western European countries have reduced their MRSA rates by implementing stringent IPC strategies (e.g., targeted screening, decolonization, hand hygiene, and ASPs). However, this region continues to face challenges with Gram-negative bacteria, such as CRE, especially in refugee patients [205,206,207]. On the other hand, countries from Eastern Europe continue to struggle with high levels of AMR, mainly due to overuse and weaker healthcare systems. It has been reported that the drivers of irrational antibiotic use in some of these countries include a lack of public awareness, access to antibiotics without a prescription, and inadequate medical training [208]. For instance, Poland has one of the highest rates of antibiotic consumption among all European countries [209].
The AMR trends have fluctuated in the United States, with recent increases in resistant gonorrhea and CRE. While the MDR gonorrhea rates have remained relatively low over the past three decades, a peak was observed in 2011 and it settled at lower values five years later [210]; in recent years, there has been a concerning increase in azithromycin resistance [211]. Concerning CRE, the rates slightly increased between 2015 and 2019 [212], with the highest infection rates observed in long-term acute-care hospitals [213]. Latin America is experiencing an emerging AMR problem, with increasing reports of resistant infections associated with healthcare settings, such as carbapenem resistance in A. baumannii and P. aeruginosa [214]. Healthcare professionals recognize the importance of antibiotic stewardship; however, they do not perceive AMR as a problem in their facilities [215]. Furthermore, rural settings also contribute to AMR through intensive animal production and agricultural practices [216].
In viral infections, the widespread use of ART has led to a significant decline in HIV-related deaths globally, avoiding a total of 9.5 million deaths between 1995 and 2015 [217]. However, the emergence of drug-resistant HIV strains is beginning to undermine these positive results, threatening the effectiveness of ART and the control of the HIV epidemic [218]. These alarming results are mainly due to non-compliance, insufficient dosages, drug interactions, and viral mutations [218]. Similarly, resistance to influenza antivirals, such as oseltamivir, has been documented, with its prevalence varying across different influenza seasons [219].

4.3. Factors Contributing to the Dissemination of Antimicrobial Resistance

AMR dissemination, regulated by an interconnected network of factors, includes healthcare practices, antimicrobial use in agriculture, international travel, medical tourism, and environmental factors.
In healthcare settings, the inadequate use of antibiotics is the main contributor to AMR, being commonly influenced by other factors such as a lack of public knowledge, access to antibiotics without a prescription, inadequate medical training, and insufficient diagnostic tests [208]. In many regions, antibiotics are frequently overprescribed, often for conditions where they are ineffective, such as viral infections. For instance, in the United States, at least 30% of outpatient antibiotic prescriptions are considered unnecessary, rising to 50% for respiratory tract infections [220]. This overuse contributes to antimicrobial resistance, adverse drug events, and increased healthcare costs. Additionally, inadequate infection control practices in hospitals contribute to the dissemination of resistant organisms, mainly Gram-negative bacteria [221]. For instance, the overuse of broad-spectrum antibiotics in intensive care units (ICUs) has been linked to the emergence of resistant strains such as CRE [222].
Healthcare-associated infections (HAIs) caused by resistant bacteria (e.g., MRSA, VRE, the “ESKAPE” group, and HA-CDI) are particularly problematic in hospitals, where they rapidly disseminate among vulnerable patients [223]. During the COVID-19 pandemic, shortages in personnel, personal protective equipment (PPE—e.g., gloves, gowns, and masks), and medical supplies led to deviations from standard IPC strategies, resulting in outbreaks of carbapenem-resistant A. baumannii [224]. Coordinated approaches, including IPC and ASPs, could prevent an estimated 619,000 HAIs over five years [225]. Moreover, the lack of rapid diagnostic tools in many healthcare settings means that antibiotics are often prescribed empirically, further contributing to the problem [226].
Antibiotic use in agriculture also plays a significant role in AMR emergence and dissemination [227]. In many countries, particularly the United States, Brazil, and China, antibiotics are widely used in livestock for the treatment and prevention of infections and to promote growth in food animals [25]. Antibiotics at sub-therapeutic doses often promote resistant bacteria’s emergence and selection, as well as their transmission to humans through the food chain, direct contact with animals, or environmental contamination [228].
In aquaculture, antibiotics are used to prevent and treat diseases [229], accumulating antibiotic residues in cultured aquatic products and developing bacterial resistance [230]. In crop production, especially in LMICs, antibiotic use is being recommended more frequently and for an increasing variety of crops [231]. The widespread use of antibiotics, coupled with the application of animal waste to agricultural land, contributes to antibiotic-related environmental contamination [232]. The environmental concentrations are generally below the therapeutic doses, but their long-term impacts on ecosystems, animals, and human health remain uncertain, requiring further research and stricter regulations [230,232].
Globalization and international travel have facilitated rapid AMR dissemination across borders [233]. Recent studies demonstrate that nearly 30% of international travelers acquire AMR bacteria during their trips to regions where resistant strains are prevalent and bring them back to their home countries, contributing to global AMR dissemination [234,235]. This is particularly concerning in pathogens such as Neisseria gonorrhoeae, where resistant strains have been globally disseminated and complicated treatment protocols [236]. Healthcare facilities should consider patients’ travel histories to implement appropriate treatment protocols and prevent further dissemination [233]. International surveillance and policy efforts are crucial to monitor and respond to this emerging global health crisis [233,234].
Medical tourism (i.e., individuals that travel to other countries for medical treatment) also contributes to AMR dissemination. Patients acquire resistant infections in healthcare settings abroad, particularly in countries with high AMR rates, and carry them back to their home countries [237]. It has been demonstrated that high-income countries (HICs) are more likely to receive AMR from LMICs, with Asia being a common origin for resistant bacteria—mainly enteric bacteria resistant to β-lactams and quinolones [238].
The environment acts as a reservoir for MDROs and ARGs due to the discharge of untreated or inadequately treated waste from hospitals, pharmaceutical factories, and agricultural operations into the environment [29]. These environmental MDROs can then enter human and animal populations, with anthropogenic activities contributing to their dissemination [239]. In high-income countries, human-to-human transmission is the primary route of AMR acquisition, while, in LMICs, inter-reservoir transmission is more common, mainly due to poor sanitation, a lack of clean water, and inadequate farming practices [239].

4.4. Comprehensive Strategies for Combating Antimicrobial Resistance

One of the most significant strategies to limit AMR emergence and dissemination is the implementation of ASPs in healthcare settings to promote rational antibiotic use [240]. These programs optimize clinical outcomes and healthcare costs and minimize AMR’s emergence and incidence by implementing several strategies (e.g., prospective audits with feedback, formulary restrictions, educational interventions, and rapid diagnostic tools) [241].
Strengthening the IPC strategies in hospitals and other healthcare facilities limits resistant organisms’ dissemination. Improving hand hygiene (HH), active screening for high-risk patients, isolating patients with resistant infections, and enhancing the use of PPEs are key components of this strategy [242]. Additionally, facility decontamination plays a crucial role in this multi-approach strategy. Decontaminating surfaces and equipment in healthcare settings can significantly reduce the environmental reservoirs of resistant pathogens, such as C. difficile spores, thereby lowering the risk of transmission and contributing to the overall effectiveness of antimicrobial resistance (AMR) control efforts. This measure complements other IPC strategies, ensuring a more comprehensive approach to combating AMR [243].
By monitoring AMR trends, global surveillance networks (e.g., GLASS and EARS-Net) guide public health interventions, control antibiotic use, and help to control AMR dissemination [244] by collecting antibiotic susceptibility data from clinical laboratories, assisting in developing diagnostics, vaccines, and novel antibiotics [244]. These programs face challenges in delivering consistent, high-quality data, especially in LMICs [245]. Alternative approaches are being explored to surpass these limitations. For instance, the use of sewage for AMR surveillance through metagenomic sequencing [245] provides affordable community-level surveillance in resource-poor settings, as revealed during the SARS-CoV-2 epidemic, when an integrated global AMR surveillance in wastewater environments was established [246,247]. As a result, several recommendations have been proposed for these programs. Reducing antibiotic use in agriculture, particularly their use as growth promoters, is vital in restraining AMR’s dissemination [133].
Another recommendation is to increase the public awareness of AMR’s dangers and educate healthcare professionals, farmers, and the public about responsible antibiotic use. Public health campaigns can change behavior by reducing unnecessary antibiotic use, a critical step to control AMR’s emergence and dissemination [133]. Investing in developing new antibiotics, alternative therapies, and rapid diagnostic tools allows us to stay ahead of this continuously evolving resistance. The development of vaccines and other preventive measures can reduce the need for antibiotics and help to control infection dissemination [133]. Finally, fostering global collaboration in research and innovation is essential to address AMR’s challenges [133].

5. Impact of Antimicrobial Resistance

AMR is one of the most pressing global health challenges of the 21st century, with profound consequences for human health, the effectiveness of medical treatments, and the economic stability of healthcare systems and societies worldwide [136]. According to an estimation, by 2050, AMR will be responsible for 10 million deaths annually, associated with a global economic burden of USD 100 trillion [240]. The ability of microorganisms to resist the effects of drugs intended to eliminate them leads to increased morbidity, mortality, and healthcare costs, posing significant challenges for public health [133].

5.1. Consequences of Antimicrobial Resistance for Human Health

The increase in AMR has severe implications for human health, primarily through increased morbidity and mortality [248]. Infections that were once easily treated with standard antibiotics have become more difficult to manage, leading to prolonged illnesses and higher risks of complications [249]. As a result, common infections (e.g., pneumonia, urinary tract infections, and bloodstream infections) are becoming increasingly resistant to first-line treatments, contributing to treatment failures and relapsing infections, further exacerbating health outcomes [250]. In France, an estimated 158,000 infections due to MDROs occurred in 2012, resulting in approximately 12,500 deaths [251]. Moreover, MDRO-caused HAIs are associated with higher mortality risks and readmission rates when compared to susceptible strains [252], as confirmed by a meta-analysis of studies in developing countries, particularly for the “ESKAPE” group pathogens [253]. The mortality rate for systemic infections with ESBL and MRSA is almost twice that of infections caused by susceptible pathogens [254]. As a result, AMR infections lead to increased healthcare costs, emphasizing the need for effective surveillance and prevention strategies to combat this growing global challenge [253].
Finally, AMR further complicates the management of chronic diseases and medical procedures. Patients undergoing chemotherapy, organ transplants, or major surgeries are at a higher risk of acquiring resistant infections due to their weakened immune systems [255]. In surgical settings, antibiotic prophylaxis prevents surgical site infections (SSIs), but increasing AMR raises the risk of SSIs complicated by resistant bacteria, leading to poorer outcomes [256].

5.2. Challenges in Treating Resistant Infections

Treating MDRO-caused infections is clinically challenging. The limited availability of effective antibiotics forces healthcare professionals to resort to less commonly used or more toxic alternatives, which may not be as effective and can lead to adverse side effects. In some cases, no effective treatment options may be available, leading to treatment failure and increased mortality [257].
One of the major challenges in treating resistant infections is the need for timely and accurate diagnosis [226]. Identifying the causative pathogen and its resistance profile is useful in selecting the appropriate treatment. The current diagnostic methods, primarily culture-based, are time-consuming and often lead to inappropriate antibiotic use [258]. Emerging technologies offer promising solutions (e.g., sample-in-answer-out PCR tests, rapid susceptibility tests, and sequencing-based approaches) and can guide targeted therapy, improve patient outcomes, and reduce AMR emergence [258,259]. Nevertheless, their implementation faces challenges, such as limited accessibility, high costs, and a lack of trained personnel [226]. Diagnostic stewardship, involving proper test selection and result interpretation, is essential for the optimal use of these tools [226]. Multidisciplinary collaboration and an increased awareness of the impact of good microbiology diagnostics are necessary to address these challenges and improve antibiotic use [226].
The emergence of MDR and extensively drug-resistant (XDR) organisms exacerbates the challenge of treatment. MDR pathogens, such as CRE, A. baumannii, and P. aeruginosa, are considered the highest-priority pathogens due to their ability to form biofilms and acquire multiple resistance genes [260]. The rise of XDR or “superbugs” (i.e., strains with resistance to most or all available antibiotics, resulting in extremely or totally drug-resistant phenotypes), such as MRSA, has led to an estimated 10,000 annual cases of nosocomial MRSA bloodstream infections in the United States [261,262,263].
The potential for treatment failure is particularly concerning in vulnerable populations (e.g., elderly or immunocompromised patients and those with underlying health conditions) [264], due to the body’s compromised ability to fight infections and the presence of resistant pathogens, which further diminishes the chances of recovery [265], impacting patients and placing a significant burden on healthcare systems. C. difficile exacerbates this issue, particularly among elderly and immunocompromised patients, leading to high morbidity and mortality and substantial economic burdens on healthcare systems worldwide. In the United States alone, CDI affects nearly 500,000 patients annually and results in around 30,000 deaths [265].

5.3. Economic Impact on Healthcare Systems and Society

The costs for healthcare systems are primarily attributed to longer hospital stays, intensive care expenditures, and more costly treatments for patients with resistant infections [266,267,268]. In Ireland, the AMR-associated excess length-of-stay costs were estimated at EUR 12 million in 2019 [269]. In France, the AMR costs exceed EUR 109 million, potentially reaching EUR 287 million if all cases were identified [270]. In the United States, the AMR infection treatment cost has doubled since 2002, reaching USD 2.2 billion annually (around USD 1400/patient) [270]. AMR infections increase mortality, with odds ratios of 1.844 (95% CI: 1.187–2.865) and higher readmission rates [271]. In France, the mean excess length of stay has been estimated at 1.6 days and 7.4 days (95% CI: 3.4–11.4) in a meta-analysis [269].
Beyond the direct healthcare costs, AMR presents other economic implications for society, mainly associated with productivity losses due to prolonged illness or premature death [272]. In Thailand, the estimated economic burden of AMR is USD 0.5 billion, and it is USD 2.9 billion in the US for five key pathogens [273]. LMICs bear a disproportionate burden, with AMR potentially reducing the global gross domestic product (GDP) by 1.1–3.8% annually by 2050.
The impact of AMR on the pharmaceutical industry is also noteworthy. The high cost of new antibiotic development, associated with the relatively short period of efficacy before resistance develops, has led to a decline in investment in antibiotic R&D, resulting in a dwindling pipeline of new antimicrobial agents and further exacerbating the AMR problem [274]. The high costs of clinical trials and the complexity of regulatory requirements further discourage companies from pursuing antibiotic development [161]. Governments and health agencies have attempted to incentivize reinvestment in antibiotic development, but the funding remains insufficient [275]. To address this crisis, there is a need for a broader scientific agenda, the better integration of industry and academia, streamlined regulatory paths, and new commercial models for antibacterial agents [275].

5.4. Global Implications and the Need for Coordinated Action

Addressing AMR requires collaboration among various stakeholders (e.g., healthcare professionals, policymakers, and the public), implementing strategies such as antimicrobial stewardship programs, controlling over-the-counter (OTC) sales, raising public awareness, establishing national guidelines for prudent antimicrobial use, sanitation improvements, and agricultural practices [13,276,277]. The fight against AMR requires collective responsibility and coordinated efforts at the local, national, regional, and international levels to ensure the sustained effectiveness of antimicrobials [278].
According to the WHO’s Global Action Plan (GAP), national action plans (NAPs) focusing on improving ASPs, enhancing IPC strategies, and increasing access to diagnostic tools and effective treatments are essential to combat AMR [279]. Many countries have developed NAPs that are fully aligned with the GAP’s objectives. Nonetheless, their implementation remains inconsistent, particularly in LMICs, due to limited resources, policy fragmentation, and “isomorphic mimicry”, where countries adopt policies without effective implementation [280]. To strengthen NAP implementation, it has been recommended that governance quality be improved, stakeholders be engaged, and actions across the human, animal, and environmental health sectors be coordinated [281]. A governance framework for NAPs has been proposed, focusing on policy design, implementation tools, and monitoring and evaluation [281]. International cooperation and legally binding responsibilities enhance global AMR governance and minimize policy fragmentation [280].

5.5. The Social and Ethical Dimensions of AMR

AMR extends beyond the clinical and economic impacts, creating important social and ethical issues. Social issues include balancing individual liberty with public health protection, addressing global distributive justice issues, and considering intergenerational responsibilities [282]. Recent studies have revealed a significant gap in understanding AMR’s social and structural drivers, with a limited focus on equity, gender, and social determinants of health [283]. These factors influence exposure, vulnerability, and the consequences of AMR, while also affecting the effectiveness of interventions [284]. A comprehensive approach is needed to ensure equitable access to antibiotics, improve living conditions, and challenge socioeconomic inequalities [285]. International cooperation is essential, as individual countries cannot combat AMR alone [286].
Ethical tensions arise between ensuring access to antibiotics in low-resource settings, curbing excessive use, and balancing personal interests against broader societal needs to combat AMR [287]. These issues (e.g., equity concerns, environmental impacts, and the role of pharmaceutical industries in drug development and promotion [288]) underscore the importance of a holistic, globally coordinated approach to combat AMR, involving multiple stakeholders and prioritizing equitable access to effective antimicrobials. Ethical issues also include antimicrobials’ use in agriculture and animal husbandry, which contributes to the emergence of resistant bacteria that can be transmitted to humans through the food chain. Balancing the need for food security and preserving the effectiveness of antibiotics for human health presents a significant ethical challenge [289].

6. Antimicrobial Stewardship and Infection Prevention Control

ASPs and IPC strategies are two key components of the global effort to combat AMR. While ASPs promote rational antibiotic use and preserve treatment effectiveness [290], improving patient outcomes and reducing healthcare costs [291], IPC strategies protect patients, healthcare professionals, and the broader community from infections and contribute to better patient outcomes, reduce healthcare costs, combat AMR, and ensure healthcare systems’ overall safety and trustworthiness [292].

6.1. Strategies to Promote Responsible Antimicrobial Use in Healthcare Settings

ASPs involve a systematic effort to optimize the use of antibiotics, ensuring that they are only used when necessary, in the correct dosage, and for the appropriate duration. Effective stewardship programs are essential in reducing the emergence of MDROs and improving patient outcomes. ASPs’ success relies on the intervention of multidisciplinary teams composed of infectious disease physicians and clinical pharmacists [241]. Several key strategies are employed in healthcare settings to promote responsible antimicrobial use.
One of the primary approaches is the implementation of guidelines with clear protocols for antimicrobials’ prescription. Often based on evidence-based practices, these guidelines are tailored to specific healthcare environments, such as hospitals, outpatient clinics, or long-term care facilities. By adhering to these guidelines, healthcare professionals can reduce the unnecessary use of antibiotics, which is a major driver of resistance [293].
Another critical aspect of ASPs is the role of antimicrobial stewardship teams (ASTs), including infectious disease specialists, pharmacists, microbiologists, and infection control experts [244], who are responsible for reviewing antibiotic prescriptions, providing feedback to prescribers, making recommendations for alternative treatments when appropriate, and educating healthcare staff about the risks of AMR and the importance of responsible antibiotic use [294]. Successful ASP implementation requires leadership, administrative support, and adaptation to the available resources in each healthcare facility [294].
Diagnostic stewardship is another essential component of responsible antimicrobial use [226]. By promoting the use of diagnostic tests and other technologies to quickly and accurately identify the causative pathogen, healthcare providers can tailor antibiotic therapies to the specific infection, reducing the use of broad-spectrum antibiotics and minimizing the risk of AMR development [295]. Molecular diagnostic tests (e.g., GeneXpert and GenoType MTBDRplus) identify the presence of specific ARGs (e.g., rifampicin and isoniazid), allowing the targeted treatment of infections, such as tuberculosis, within hours instead of days or weeks, as required for traditional culture methods [296].
Education and training programs for healthcare providers promote responsible antibiotic use and AMR management. These programs aim to improve prescribing practices and encourage the use of non-antibiotic alternatives when appropriate [297]. Moreover, patient education also helps to reduce the demand for antibiotics for viral infections, such as colds and the flu, where antibiotics are ineffective [298].

6.2. Importance of Infection Prevention and Control Strategies

IPC strategies not only reduce HAIs but also combat AMR. By preventing infections from occurring in the first place, the need for antibiotics is reduced, thereby limiting the opportunities for resistance to emerge and dissemination (Figure 4).
Compliance with HH, one of the most effective IPC strategies and considered as the cornerstone of infection control, remains suboptimal, especially among doctors [299]. Healthcare providers, patients, and visitors are encouraged to practice proper HH, using alcohol or chlorhexidine gluconate-based hand sanitizers or soap and water to prevent the transmission of pathogens [300]. Regular HH compliance audits and feedback are necessary to ensure adherence to this critical practice [301].
Another key IPC strategy is the use of PPE, particularly in situations where there is a high risk of infection transmission. Nevertheless, the use of PPE is often limited by several factors, such as discomfort, supply shortages, inadequate infrastructure, and a lack of training. To improve PPE adherence and effectiveness, healthcare facilities should develop context-driven implementation strategies, address healthcare personnel wellbeing, reorganize work processes, and adopt structural changes [302].
Water-free patient care aims to minimize or even eliminate water use in healthcare. In personal hygiene, pre-moistened disposable wipes replace traditional bed baths, ensuring patients’ cleanliness and comfort. Water-free dressings and sprays replace conventional saline solutions in wound management, reducing infection risks. Alcohol-based hand sanitizers replace soap and water, improving HH and infection control. The benefits of this innovative approach include environmental sustainability through reduced water usage, coupled with improved infection control and enhanced patient comfort [303].
Sink removal, mainly in intensive care units (ICUs), has been proven efficient in healthcare settings. Sinks constitute reservoirs for harmful bacteria, including MDROs, which are easily disseminated to the surrounding environment. By eliminating sinks, the contamination risk decreases, reducing HAIs. Moreover, in the absence of sinks, healthcare professionals rely on alternative HH practices that have been proven to be more effective, leading to better HH compliance among staff and reducing the likelihood of infection transmission [304].
Environmental cleaning, disinfection, and decontamination are critical components of IPC. The regular cleaning and disinfection of high-contact surfaces, medical equipment, and patient care areas are essential to eliminate pathogens and reduce the risk of infection [305]. Traditional manual cleaning methods are often insufficient, highlighting the need for advanced solutions such as gas generation systems and fogging equipment capable of producing a homogeneous cloud of decontaminants, such as peroxide-based disinfectants. These methods can reach difficult-to-access areas where pathogens may proliferate, contributing to environmental reservoirs in healthcare facilities and other settings. Additional approaches, such as electrolyzed water and “no-touch” automated systems such as UV-C light devices, are valuable in a multifaceted strategy to control AMR [306]. These procedures are particularly important since several pathogens of concern for IPC can persist on inanimate surfaces for periods of time that range from weeks to almost two years (K. pneumoniae: 600 days; S. aureus: 318 days; C. difficile: 140 days; Acinetobacter spp. 90 days; E. coli: 56 days; Coronavirus: 20 days; C. auris: 14 days) [307].
The isolation and cohorting of patients with known or suspected resistant infections are additional strategies to prevent MDRO dissemination [308]. Nevertheless, isolation may negatively affect patient outcomes, including longer hospital stays, higher costs, and increased readmission rates. Moreover, isolation implementation may be challenging in LMICs due to resource constraints [309].
Vaccines prevent targeted pathogen infections, protect against resistant subtypes, and decrease bystander selection [310]. The impact of vaccination on AMR has been demonstrated for vaccines against Streptococcus pneumoniae, Haemophilus influenzae type b, and influenza [303]. Despite the GAP recommendation on vaccination, its adoption in NAPs is not universal, particularly in LMICs [311], due to challenges such as the National Immunization Technical Advisory Group (NITAG) capacity, access to evidence, and resources for systematic vaccine policy decision making [312]. The development of vaccines against resistant pathogens faces challenges due to pathogens’ complexity and technical difficulties [313]. Mathematical models suggest that the impact of vaccination on antibiotic resistance depends on the relative strength and directionality of the competition between drug-resistant and drug-sensitive strains, emphasizing the need for tailored policies [314].

6.3. Global Surveillance Systems for Antimicrobial Resistance

Surveillance is essential to the global response to AMR, guiding interventions, informing policies, and tracking stewardship and IPC strategies’ effectiveness [315,316]. Different surveillance approaches exist, from traditional clinical sample analysis to sewage-based surveillance using metagenomic sequencing [245]. Surveillance systems operate at different levels, from local to international, providing data on AMR prevalence and distribution. Effective surveillance requires timely feedback to stakeholders and correlation with demographic and clinical data to understand the epidemiology and formulate interventions [317].
National surveillance programs, such as the United States Centers for Disease Control and Prevention’s (CDC) National Healthcare Safety Network (NHSN) and the European Antimicrobial Resistance Surveillance Network (EARS-Net), collect data from healthcare facilities across their respective regions to monitor resistance trends, identify emerging threats, and provide benchmarks to compare resistance rates between different locations [315]. The Study for Monitoring Antimicrobial Resistance Trends (SMART) is a long-running (i.e., 20 years), industry-sponsored program that has collected data from over 60 countries, providing valuable information for treatment guidelines and research [317]. In the Netherlands, a successful national AMR surveillance system based on routine data from medical microbiological laboratories has been supporting policymaking and guideline development since 2008 [318].
At the international level, GLASS coordinates efforts to monitor AMR by collecting and sharing data from participating countries, providing a global picture of resistance patterns and trends [319]. Despite the challenging implementation of these programs in LMICs, guidelines have been developed to facilitate their participation [320]. In Thailand, data collection is challenging and specimen contamination often occurs, highlighting the need for additional training [321]. A recent analysis of GLASS data demonstrated significant correlations between antimicrobial consumption and resistance rates for certain pathogens and antibiotics across participating countries [322]. This information can be used to identify global AMR hotspots, assess intervention impacts, and guide international efforts to combat AMR [319,322].
Surveillance data help in developing APSs across healthcare settings. By identifying the most prevalent resistant pathogens and their antibiotic resistance profiles, healthcare professionals can make informed decisions on the most appropriate treatment options, avoiding inappropriate antibiotic use and not only preserving the existing antibiotics’ effectiveness but also reducing AMR dissemination [323].

6.4. Novel Approaches to Combat Antimicrobial Resistance

There is an urgent need for novel approaches to combat AMR and preserve antimicrobial therapies’ effectiveness. Several promising strategies are currently being explored, including the development of new antibiotics, alternative therapies, and innovative technologies (Table 2).
One of the most critical areas of research is the development of new antibiotics with novel mechanisms of action. Traditional antibiotics often target essential bacterial processes (e.g., cell wall synthesis and protein production). For instance, teixobactin, a novel antibiotic isolated from uncultured bacteria with potent activity against Gram-positive pathogens, destroys bacteria through a two-pronged attack on the cell envelope by targeting cell wall precursors (Lipids II and III) and disrupting the membrane [324].
Bacteriophages are emerging as an alternative to traditional antibiotics due to their high specificity, ecological safety, and potential applications beyond clinical settings [325,326]. Nevertheless, this alternative presents some challenges, mainly associated with large-scale production, formulation, stability, and bacterial resistance to phages [326]. Another phage-related innovative approach involves exploiting phage–bacteria interactions to discover new antibacterial targets and develop small molecules that mimic inhibitory phage proteins [327].
Antimicrobial peptides (AMPs) constitute another promising candidate in combating AMR due to their broad-spectrum activity and lower propensity for resistance development [328]. These naturally occurring molecules, found in most multicellular organisms, exhibit potent antibacterial, antiviral, and antifungal properties and are capable of disrupting cell membranes [329]. Despite their ancient origin, AMPs represent a promising therapeutic strategy against antimicrobial resistance, with several peptide-based drugs already approved by the U.S. Food and Drug Administration (FDA) [330]. However, further research is required to understand AMPs’ action mechanisms and optimize their clinical applications.
Alternative therapies, such as immunotherapies and probiotics, are also being explored as potential strategies to combat AMR. Monoclonal antibodies are promising due to their high specificity and bacteria’s inability to develop resistance [331]. These antibodies neutralize bacterial toxins and virulence factors [332]. Probiotics and fecal transplant therapy are being studied to enhance the human microbiota and prevent infections [333]. Pathogen-oriented therapy (POT) is emerging as a targeted approach to circumvent AMR, utilizing antibiotic–antibiotic conjugates [334], nanotechnologies, and CRISPR-Cas systems [335]. Combination therapies and the repurposing of existing drugs are also being explored [336].
Nanoparticle-based technologies have been shown to be promising to overcome resistance in planktonic and biofilm phenotypes, by combining novel nanomaterials with classic antimicrobial therapies [337]. Nanoparticles enhance antibiotic activity, preserve existing antibiotics’ efficacy, and provide novel antibacterial action modes [338].
AI applications include predictive modeling, rational antibiotic use, and the development of antimicrobial peptides and antibiotic combinations [339]. Machine learning techniques are accelerating new antibiotic discovery, optimizing treatment regimens, and predicting resistance patterns [340]. These AI-driven approaches focus on predicting antimicrobial activity and drug-likeness traits and enabling de novo molecular design. The integration of these technologies offers promising avenues to address the current AMR crisis.
Finally, there is a growing emphasis on the One Health approach to combat AMR. International organizations, such as the WHO, ECDC, and World Organisation for Animal Health (OIE), play a significant role in promoting the One Health approach to AMR, preserving the effectiveness of existing antimicrobials, and limiting infection dissemination across the human, animal, and environmental domains [341,342,343,344].

7. The One Health Approach to Antimicrobial Resistance

The One Health approach has emerged as a comprehensive strategy to combat AMR by recognizing the interconnectedness of human, animal, and environmental health. This approach emphasizes the need for collaborative efforts across various sectors to address the complex and multifaceted challenges posed by AMR [341,342,343,344].

7.1. Recognition of the Interconnectedness of Human, Animal, and Environmental Health

The interconnectedness between the health of humans, animals, and ecosystems, supported by the One Health approach, is particularly evident in the context of AMR, where the use of antimicrobials in one domain profoundly affects others. The overuse or misuse of antibiotics in agriculture, human medicine, and veterinary care contributes to MDROs’ emergence and dissemination among these different system parts [341,342,343,344].
Many countries have implemented NAPs based on the One Health approach, following guidelines from international organizations (e.g., WHO, OIE, and Food and Agriculture Organization of the United Nations—FAO), aiming to mitigate AMR dissemination, raise awareness about antibiotic use, and promote antimicrobial stewardship [345].

7.1.1. The Human Health Sector

In hospitals and outpatient clinics, antibiotics are frequently prescribed for conditions where they are not needed, such as viral infections [220], leading to the selection of resistant strains of bacteria, which can disseminate within the community and healthcare facilities, causing difficult-to-treat infections [346]. Patient expectations and misconceptions contribute to this issue, as 53% of patients incorrectly believe that antibiotics are effective against viral infections [347]. Clinicians often prescribe antibiotics due to perceived patient demands, concerns about patient satisfaction, and a fear of negative repercussions after denying antibiotics [348]. Effective clinician–patient communication is essential to provide clear explanations, use specific diagnoses, and offer alternative treatments to reduce unnecessary antibiotic prescription while maintaining patient satisfaction [349].

7.1.2. The Animal Health Sector

The widespread use of antibiotics in animal husbandry, for therapeutic purposes or as growth promoters, creates a reservoir of resistant bacteria in animals that can be transmitted to humans through the food chain or direct contact, such as Salmonella, Campylobacter, E. coli, S. aureus, and C. difficile [350]. Some countries have banned the use of antibiotics as growth promoters and are focusing on developing alternatives (e.g., vaccines, probiotics, and bacteriophages), including new biosafety–biosecurity and management approaches, to reduce AMR dissemination among animals in farms [351,352].

7.1.3. The Environmental Health Sector

Antibiotics and antibiotic-resistant bacteria may enter the environment through various pathways (e.g., agricultural runoff, wastewater discharge, and improper disposal of pharmaceuticals) [239], and, once in the environment, they can persist and disseminate, affecting wildlife and entering the human food chain. The presence of antibiotics in water bodies, soil, and sediments promotes the selection of resistant organisms in these ecosystems, creating reservoirs of resistance that can be transmitted back to humans and animals [29]. The agricultural ecosystem facilitates AMR emergence through the indiscriminate use of antibiotics in agricultural and veterinary practices [353]. While HICs have more controlled discharges, LMICs face greater challenges due to poor sanitation and deficient farming practices [239]. Addressing environmental AMR requires improved waste treatment, reduced antibiotic use, and better pharmaceutical waste control [239].

7.2. Importance of Collaborative Efforts between Healthcare, Veterinary, and Environmental Sectors

The One Health approach emphasizes the importance of collaboration and coordination among various sectors to effectively combat AMR. By working together, stakeholders from healthcare, veterinary medicine, agriculture, and environmental management can develop and implement strategies that address the causes of resistance and mitigate its impact on public health [341].

7.2.1. Assessing One Health Integration in Antimicrobial Resistance Surveillance Systems

One of the key components of the One Health approach is the establishment of integrated surveillance systems that monitor the use of antimicrobials and resistance dissemination across the environmental, animal, and human domains. Unfortunately, many of these programs are often fragmented and lack full integration [354]. A review of electronic information systems (EIS) for AMR surveillance found no systems that fully integrated data across all three sectors, highlighting a significant gap in global efforts to address AMR [355]. A conceptual framework has been developed to evaluate the integration of the One Health approach in AMR surveillance systems, focusing on five key components: the capacity to integrate a One Health approach, produce One Health information, generate actionable knowledge, influence decision making, and impact outcomes [356].

7.2.2. Collaborative Policy Approaches to Combat Antimicrobial Resistance

Effective policies to combat AMR require collaboration between environmental, veterinary, and healthcare policymakers to develop regulations promoting several measures [33], such as ASPs in veterinary and human medicine [357], environmental regulations limiting antibiotic release in ecosystems [33], and the harmonization of regulatory frameworks across countries [13]. Challenges include balancing individual ministry priorities with cross-cutting issues and building mutual trust [357]. Immediate actions required include establishing ASPs, controlling OTC sales, and promoting public awareness [13]. A global fund and international collaboration integrating social, economic, and behavioral factors are recommended to address AMR effectively [13].

7.2.3. Joint Research and Innovation

The One Health interdisciplinary approach encourages collaborative research efforts to develop innovative solutions to combat AMR [358,359]. The exploration of bacteriophages as potential alternatives to antibiotics in human and veterinary medicine has been shown to be a promising alternative, due to their highly specific and potent antimicrobial activity against resistant species [326], although challenges remain in their large-scale production and implementation [326].

7.2.4. Education and Awareness

Various educational activities, including games, roleplay, and e-learning, can effectively engage diverse audiences in understanding this phenomenon [360]. While healthcare professionals across the animal and human sectors often demonstrate high AMR awareness, this knowledge does not necessarily translate into reduced antibiotic prescribing [361].

7.2.5. Strengthening Infection Prevention and Control

IPC strategies are essential in reducing resistant pathogens’ dissemination across environmental, animal, and human interfaces. In healthcare settings, IPC practices (e.g., HH, PPE use, environmental cleaning, and routine decontamination) prevent the transmission of resistant bacteria [341]. In veterinary medicine, biosafety and biosecurity measures, vaccination, and proper animal husbandry practices reduce the incidence of infections in livestock, thereby decreasing the need for antibiotics [362]. Environmental management practices (e.g., proper waste disposal and the treatment of agricultural runoff) limit resistant organisms’ dissemination in the environment [363].

7.2.6. Global Cooperation and Capacity Building

LMICs often face significant challenges in addressing AMR, such as limited healthcare access, weak regulatory frameworks, and inadequate infrastructure for surveillance and laboratory testing [364]. International cooperation among the WHO, OIE, and FAO builds the capacity in these countries by providing technical assistance, funding, and access to expertise [365]. Global cooperation also involves sharing best practices, data, and resources and coordinating efforts to address AMR in transboundary contexts, such as controlling resistant pathogens’ dissemination through international travel and trade [365].

8. Policy and Regulatory Initiatives

In response to the growing threat of AMR, international and national policymakers have developed a range of strategies and regulatory frameworks to combat AMR, promote responsible antimicrobial use, and preserve the effectiveness of existing treatments.

8.1. Overview of National and International Policies

The recognition of AMR as a transboundary issue that requires a global response has led to the formulation of several AMR-oriented international and national policies focusing on several key areas, such as surveillance, ASPs, R&D, and public awareness [126].

8.1.1. International Policies

International organizations (e.g., WHO, OIE, FAO) have taken the lead in coordinating global efforts to combat AMR [366]. In 2015, the WHO launched the GAP on AMR, which encouraged more than 100 countries to develop their own NAPs to address this issue [280]. The GAP outlines five strategic objectives that should be implemented in NAPs: improving the awareness and understanding of AMR, strengthening surveillance and research, reducing the incidence of infection, optimizing the use of antimicrobial agents, and fostering sustainable investment in new medicines and diagnostic tools, vaccines, and other interventions [280]. The WHO also oversees the GLASS, which collects and analyzes AMR data from member states, aiming to strengthen national surveillance systems and facilitate data sharing among countries [319]. Despite the guidelines developed to ensure data validity and comparability, the implementation of this system in LMICs faces several challenges, as previously described [320,321].
The OIE has developed standards for the responsible use of antimicrobials in veterinary medicine, which are integrated into the national regulations of its member countries [367]. The OIE has also created the List of Antimicrobial Agents of Veterinary Importance, which collects data on antimicrobial use and supports member countries in implementing standards through various programs [367].
The FAO has focused on reducing antibiotic use in agrifood systems through programs to strengthen governance, increase awareness, enhance AMR surveillance, and implement best practices in food production [368]. Developing countries face challenges in controlling AMR in the food chain, necessitating comprehensive measures from farm to fork [253]. Environmental factors (e.g., soil and water contamination) also contribute to ARG dissemination in food production systems [369].

8.1.2. National Policies

Many countries have developed NAPs in line with the WHO’s GAP, and these typically include measures to enhance surveillance, promote antimicrobial stewardship, support R&D, and raise the public awareness of AMR [279,280]. While strong vertical alignment exists between the GAP and NAPs, particularly in LMICs, horizontal alignment within regions is weaker [280]. NAPs generally align well with the GAP objectives but less with recommended actions [279]. Therefore, gaps exist in monitoring and evaluation, financing plans, and targets for antimicrobial use reduction [370]. The United Kingdom’s “UK Five Year Antimicrobial Resistance Strategy” emphasizes the importance of improving IPC strategies, optimizing the use of antimicrobials in animal and human medicine, and investing in new treatments and diagnostics [371]. Similarly, the United States’ “National Action Plan for Combating Antibiotic-Resistant Bacteria” aimed to establish ASPs in all acute care hospitals by 2020 [372]. In China, an NAP significantly reduced the AMR rates and culture positivity in a teaching hospital [373]. In Tanzania, a year after launching their NAP, the implementation of AMR surveillance and ASPs was found to be insufficient and inconsistent across healthcare facilities, highlighting areas for improvement in addressing AMR [374].
In many LMICs, NAPs often focus on strengthening the healthcare infrastructure, improving access to point-of-care treatment [375], and ensuring the availability of quality medicines (i.e., avoiding substandard or falsified medicines). However, challenges such as limited resources, weak regulatory frameworks, and inadequate surveillance systems can hinder the effective implementation of these policies [376].

8.2. Regulatory Frameworks to Promote Responsible Antimicrobial Use

Regulatory frameworks ensure the responsible use of antimicrobials and preserve their effectiveness, and they typically encompass regulations on antibiotics’ sale and use, guidelines for healthcare professionals, and measures to control antimicrobial use in agriculture and veterinary medicine.

8.2.1. Regulatory Frameworks and Antimicrobial Stewardship in Combating Antibiotic Misuse in Human Health

In the healthcare sector, regulatory frameworks often include guidelines for the prescription of antibiotics, OTC sales restrictions, and measures to prevent the misuse of antimicrobials. In countries such as India [377], Nepal [378], Thailand [379], Ghana [380], and Zambia [380], OTC sales remain as a common practice, with amoxicillin, azithromycin, and ciprofloxacin commonly sold for respiratory issues and diarrhea [381,382]. In contrast, in other countries, such as Portugal, antibiotics are prescription-only medicines, meaning that they can only be dispensed with a valid prescription from a licensed clinician, preventing antibiotic overuse and misuse, considered major drivers of AMR [383].
Regulatory bodies (e.g., the FDA and the European Medicines Agency—EMA) ensure the safety and efficacy of antimicrobial drugs by evaluating new agents, monitoring their use, and publishing guidelines for healthcare professionals on appropriate prescribing practices [234]. These agencies also oversee ASP development and implementation, which is designed to optimize the use of antibiotics in healthcare settings [384]. ASPs often include educational initiatives to inform healthcare professionals about the AMR risks and the importance of judicious antibiotic use. They may also involve establishing multidisciplinary teams (e.g., pharmacists, infectious disease specialists, and microbiologists) to guide antibiotic prescribing and monitor patient outcomes [294].

8.2.2. Regulatory Frameworks and Antimicrobial Stewardship in Combating Antibiotic Misuse in Animal Health and Agriculture

Regulatory frameworks in the agriculture and veterinary sectors typically focus on reducing the use of antibiotics as growth promoters, limiting their use for prophylactic purposes, and promoting best practices in animal husbandry to prevent infections. In the European Union (EU), the use of antibiotics as growth promoters in livestock was banned in 2006, while countries such as Denmark and the Netherlands have successfully reduced veterinary antibiotic usage [385]. Alternative approaches to antibiotics in livestock (e.g., bacteriophages, engineered peptides, probiotics, and quorum-quenching molecules) have shown promising results [386]. Improved biosecurity measures, animal husbandry practices, and farm management have also reduced antibiotic use in many countries [387].
In 2014, the FDA curbed the use of medically important antibiotics as growth promoters in livestock [388] and implemented guidelines for the judicious use of antibiotics (e.g., banning growth promotion applications and requiring veterinary oversight for feed and water administration) [389]. The FDA’s approach also includes a qualitative risk assessment for new animal drugs to evaluate their potential for the development of resistance in bacteria harmful to humans [390]. However, challenges remain in surveillance and regulatory oversight; while the poultry industry has significantly reduced its antibiotic use, their usage in the pork and beef industries remains considerable [389].

8.2.3. Regulatory Frameworks and Antimicrobial Stewardship in Combating Antibiotic Misuse in Environmental Health

Various drivers contribute to AMR dissemination, including antibiotic use in healthcare and agriculture and the release of antibiotic residues into soil, water, and air [391]. Pharmaceutical effluents, particularly in LMICs, pose significant risks to ecosystems and public health [392]. Critical knowledge gaps exist regarding the sources of antibiotics and ARGs in the environment, the role of anthropogenic inputs in resistance evolution, the health impacts of environmental exposure, and the efficacy of mitigation strategies [28]. Addressing AMR requires a collaborative One Health approach involving policymakers, regulators, manufacturers, and researchers to develop global standards for pharmaceutical effluent management, improve treatment methods, and implement comprehensive national action plans [391,392]. The EU’s Water Framework Directive (WFD) addresses the problem of AMR in aquatic environments by setting environmental quality standards for antibiotic concentrations [393]. To guide regulatory efforts, predicted no-effect concentrations for resistance selection have been proposed for common antibiotics, ranging from 8 ng/L to 64 μg/L [132]. These concentrations are often lower than ecotoxicological thresholds, highlighting the need for specific regulations addressing antibiotic resistance. The presence of antibiotics in aquatic ecosystems negatively impacts microbial flora and promotes ARG dissemination [394]. The implementation of the WFD to control antibiotic pollution could drive other legislation to consider AMR-associated risks.

8.3. Challenges and Opportunities for Implementation of Policy Interventions

While significant progress has been made in developing policies and regulatory frameworks, several challenges remain in their implementation. These challenges include limited resources, a lack of political will, a weak regulatory capacity, and the need for global coordination.

8.3.1. Resource Limitations

One of the primary challenges in implementing AMR policies is the lack of resources and infrastructure, particularly in LMICs [379,395]. Many countries face significant barriers in establishing robust surveillance systems, implementing ASPs, and enforcing regulations. Limited access to healthcare, inadequate laboratory infrastructure, and insufficient funding for public health initiatives hinder the effective implementation of AMR control policies [364]. To address these challenges, countries must strengthen their health systems, improve their technical capacities, and enhance their governance for effective multi-sectoral action [379]. AMR surveillance systems with integrated models combining clinical, laboratory, and demographic data are more informative than laboratory surveillance alone [323].

8.3.2. Political Will and Regulatory Capacity

The successful implementation of AMR policies depends on political will and the regulatory capacity. In some countries, there may be resistance in adopting strict regulations on antibiotic use, particularly in agriculture, due to concerns about the economic impact and industry opposition. Weak regulatory frameworks and a lack of enforcement capacity can further undermine efforts to control AMR [396].
Building political will requires advocacy and public awareness campaigns highlighting AMR’s long-term effects and economic costs [13]. Engaging stakeholders from various sectors, including healthcare, agriculture, and the pharmaceutical industry, can help to build a consensus on the need for action [33]. Strengthening regulatory agencies and ensuring adequate funding for enforcement activities ensures the successful implementation of AMR policies [397].
Public awareness campaigns about the long-term effects and economic costs of AMR are necessary [398]. However, challenges such as insufficient public awareness, limited community engagement, and inadequate human resources hinder this progress [397]. Opportunities for improvement include developing coordinated surveillance systems and capitalizing on existing structures and practices to manage the antimicrobial supply and prescription [33,397].

8.3.3. Global Coordination and Harmonization

The disparities in regulatory frameworks, surveillance systems, and enforcement capacities can create challenges for global coordination. For instance, while some countries have banned the use of antibiotics as growth promoters in livestock, others continue to allow this practice, creating a potential source of resistant bacteria that can be disseminated across borders.
International organizations (e.g., WHO, OIE, and FAO) promote harmonization and facilitate information exchange [399]. However, a comprehensive “whole United Nations approach” to AMR is still lacking, with only 21 out of 78 assessed organizations having AMR-specific activities [366]. The FAO supports nations in improving ASPs through various programs, including developing a platform to collect data on AMR in animals and antimicrobial use in plants [368]. While there is strong vertical alignment between NAPs and the GAP, particularly among LMICs, the horizontal alignment within regions is weaker. The implementation of NAPs often lacks policy alignment, suggesting a need for legally binding global governance initiatives [280].

8.3.4. Opportunities for Innovation and Collaboration

Despite the challenges, there are significant opportunities for innovation and collaboration in AMR management. Public–private partnerships, such as CARB-X, are accelerating the development of new antibiotics, vaccines, and diagnostics [400]. Advances in understanding bacterial physiology, clinical trial designs, and regulatory pathways enable the development of pathogen-targeted antibiotics responsible for patient outcome improvements and ASPs [401]. To address the declining antibacterial pipeline, a broader scientific agenda, better industry–academia integration, and a new commercial model are needed [154]. Horizon scanning methods have identified more than 3000 innovative AMR prevention, detection, and monitoring technologies, focusing on diagnostic innovations for the rapid and accurate detection of drug-resistant infections [402]. These technological advancements present opportunities to strengthen national AMR strategies and support appropriate antimicrobial prescribing.

9. Future Directions and Challenges

AMR has rapidly evolved from a manageable health concern into a threatening global crisis. The ability of pathogens to develop antimicrobial resistance undermines decades of progress in medicine, rendering common infections potentially lethal and complicating medical procedures. As the world struggles with this escalating threat, it is essential to explore the emerging challenges, identify research priorities, and consider the broader societal and ethical implications of the fight against AMR.

9.1. Emerging Threats in Antimicrobial Resistance

9.1.1. Multidrug-Resistant Pathogens

One of the most alarming developments in the realm of AMR is the rise of MDROs (e.g., MRSA, CRE, MDR-TB, and multidrug-resistant C. difficile) capable of resisting multiple classes of antimicrobials, severely limiting the treatment options and leading to higher mortality rates [393]. MDROs’ dissemination in healthcare settings and the community highlights the urgent need for robust IPC strategies and the development of new treatment options.

9.1.2. Novel Resistance Mechanisms

The emergence of novel antibiotic resistance mechanisms also poses significant challenges to public health. The mcr-1 gene, conferring resistance to colistin, has rapidly disseminated worldwide, mainly by its capacity to be transferred by HGT [403,404]. ESBLs contribute to resistance against a wide range of β-lactam antibiotics, while metallo-β-lactamases (MBLs) are a major concern, particularly in carbapenem-resistant Gram-negative bacteria, such as K. pneumoniae, A. baumannii, P. aeruginosa, E. coli, and Enterobacter species [405]. The MCR-1 protein, a phosphoethanolamine transferase, has been extensively studied regarding the potential development of MCR-1 inhibitors for use in combination therapies [406].

9.2. Research Priorities for New Antimicrobial Agents and Alternative Strategies

9.2.1. Development of Novel Antimicrobials

Given the declining efficacy of existing antibiotics, there is an urgent need to develop novel antimicrobial agents. Research priorities include identifying new drug targets and mechanisms of action [407]. The discovery of teixobactin, a novel antibiotic with a unique mechanism of action, exemplifies the potential for breakthroughs in this area [408]. However, economic challenges have delayed progress in antibiotic development, requiring innovative financial models to incentivize pharmaceutical companies [409]. Alternative explored strategies include combination therapy, the targeting of resistance mechanisms, and novel drug delivery systems [407]. According to the WHO’s reports, only 60 antibacterial drugs are currently under clinical development, with only six meeting this novel criterion [410]. Other promising approaches include nanotechnology, computational methods, and drug repurposing [411]. Non-traditional alternatives (e.g., antimicrobial peptides, bacteriophages, and prodrugs) are also being investigated [411]. Continued research, public investment, and regulatory reforms are essential to address the antibiotic development crisis [410].

9.2.2. Alternative Treatment Strategies

There is growing interest in alternative treatment strategies that can enhance the effectiveness of existing drugs or provide entirely new approaches to combat AMR. These include phage therapy, AMPs, immunotherapy, and microbiome modulation.
Bacteriophage therapy has re-emerged as a promising alternative to antibiotics [412]. Phages are viruses that specifically target and kill bacteria, offering an environmentally friendly approach to controlling pathogens in agriculture and aquaculture [413]. Their unique mechanism of action and cost-effective production make them ideal in addressing AMR infections, especially in LMICs [412]. Phages have also demonstrated effectiveness against various foodborne and zoonotic pathogens (e.g., Salmonella, E. coli, and Listeria) [414]. Historically, phage therapy has been used since 1919, with successful treatments for dysentery and other bacterial infections [415]. Despite these challenges, ongoing research focuses on expanding clinical trials, standardizing the production and storage of phage cocktails, and promoting international collaboration to advance phage therapy as a solution to AMR [412].
AMPs, essential innate immune system components, exhibit broad-spectrum antimicrobial activity against several microorganisms [416], being promising therapeutic agents due to their low resistance induction and ability to modulate the immune response [417]. However, their clinical use is limited by their toxicity to mammalian cells, reduced activity under physiological conditions, and susceptibility to proteolytic degradation [418]. Research should focus on optimizing AMPs through modifications and multimerization to improve their antimicrobial activity, specificity, and biocompatibility [417]. Nanomedicine approaches are being explored to develop targeted delivery systems for AMPs, potentially increasing their efficacy and reducing their toxicity [419].
Recent research considers boosting host immune responses to combat infections, particularly in the context of respiratory diseases. Innate immunity, including antimicrobial peptides and ROS, defends against pathogens and regulates the microbiota [420]. Host-directed therapies aim to enhance the innate defenses, especially against ARB. Toll-like receptor (TLR) agonists have shown promise in triggering trained immunity, enhancing antimicrobials’ functions through metabolic and epigenetic modifications [421]. Modulating the host response has emerged as a potential strategy for severe lung infections to mitigate damage and prepare for future pandemics [422]. However, some bacteria can exploit the anti-inflammatory responses in the lungs to evade elimination. Targeting these bacterial-induced immunosuppressive mechanisms could improve the treatment options for chronic lung infections [423]. These approaches offer pathogen-agnostic strategies to enhance the host defenses against various infections.
The human microbiome plays a role in health and disease, while its modulation offers potential therapeutic benefits [424]. Strategies to manipulate the microbiome include probiotics, prebiotics, postbiotics, and fecal microbiota transplantation (FMT) [425]. These interventions are being investigated for various conditions, including infections and complications associated with allogeneic hematopoietic cell transplantation [426]. Probiotics, prebiotics, and postbiotics have shown promise in preventing and treating diseases, although their use is currently limited to dietary supplements [424]. The rise of AMR pathogens has led to increased interest in microbiota-based treatments as alternatives to traditional antibiotics [427]. However, challenges remain in defining precise microbiome targets, refining interventions, and standardizing the outcome measures across studies [425]. Further research is needed to fully understand the microbiome’s role in various diseases and optimize its modulation for therapeutic purposes.

9.3. Societal and Ethical Implications of Antimicrobial Resistance Interventions

9.3.1. Access and Equity

Antimicrobial resistance disproportionately affects LMICs, exacerbating health inequalities [288]. Therefore, addressing AMR requires a holistic approach that considers its ethical components, such as equitable access to antibiotics and the role of social determinants of health [288]. Current approaches often focus on individual behavior change, but AMR is deeply embedded in societal values and culture, necessitating a shift toward collective responsibility [428]. The One Health approach is essential in AMR studies but often remains anthropocentric in practice [429]. Recently, a more-than-human ethical framework based on relationality and care ethics has been proposed to address these challenges [429]. Successful AMR mitigation will require collaboration at the individual, community, and national levels to ensure the continued effectiveness of antimicrobials [398].

9.3.2. Stewardship and Overuse

Although ASPs aim to optimize antibiotic use, they fail to balance patient care with resistance prevention [165]. The drivers of broad-spectrum antibiotic overuse include individual, social, and structural factors, which vary across healthcare contexts [430]. Ethical considerations in antibiotic use and distribution encompass issues of equity, access, and environmental impacts [288]. Solutions to improve stewardship include increasing practitioner responsibility, enhancing caregivers’ roles as diagnosticians, improving patient communication, reducing economic influences on prescribing, and identifying antibiotic alternatives [431]. The WHO has developed a toolkit for the implementation of ASPs, particularly in LMICs, where the AMR burden is high and resources are limited [156].

9.3.3. Environmental Impact

AMR requires a One Health approach, recognizing the interconnectedness of human, animal, and environmental health [344]. The environment, especially water resources, constitutes the primary reservoir and dissemination pathway for AMR [432]. Inadequate antibiotic use in the agriculture, veterinary, and medical sectors contributes to the spread of ARB and ARGs in soil, air, water, and sediments [13]. Agricultural soils are a major sink for antibiotics and AMR from livestock farming, with manure application augmenting soil resistomes [25]. The current global and national ASPs primarily focus on mitigating antibiotic use in the human and animal sectors, overlooking environmental drivers [432]. Addressing AMR requires the identification of high-risk environments; improvements in infection control, sanitation, and access to clean water; and the regulation of antimicrobial use in agriculture [13].

9.3.4. Ethical Dilemmas in Research and Innovation

The development of new antimicrobial treatments, mainly phage therapy, presents significant ethical dilemmas. Despite showing promising results, phage therapy faces challenges related to regulatory frameworks and clinical trial designs [433]. Ethical concerns arise from the manipulation of viruses that may evolve unpredictably and the complexities of informed consent in vulnerable populations [434]. These ethical implications of AMR also include issues of equity in access to treatments and the responsibilities of stakeholders in antibiotic stewardship [283]. Balancing the urgent need for new therapies against the necessity for rigorous testing and ethical considerations is crucial for the responsible advancement of phage therapy and other innovative interventions in the fight against AMR.

9.4. Strategic Roadmap and Challenges Ahead

9.4.1. Strengthening Global Surveillance and Data Sharing

AMR surveillance is crucial for global health, particularly in LMICs, where the burden of bacterial infections is high [364]. However, LMICs face significant challenges in implementing effective AMR surveillance due to limited resources, a weak laboratory capacity, and poor health systems [166]. The GAP has prompted countries to improve their AMR surveillance capacities, but data from these efforts often lack representativeness and standardization [364]. Estimating the global burden of AMR is challenging due to insufficient high-quality, patient-level data, especially in resource-limited settings [435]. To address these issues, a multidisciplinary approach involving various stakeholders is necessary [136]. Strengthening global surveillance networks, improving data sharing, and investing in infrastructure and capacity building are essential for informed decision making and timely interventions against AMR [136].

9.4.2. Integrating AMR into National and Global Health Agendas

Integrating AMR into broader health agendas and the Sustainable Development Goals (SDGs) framework is essential in addressing this challenge [436]. However, current AMR initiatives have had limited impacts, compelling a shift towards emphasizing AMR as being interlinked with other global challenges and incorporating upstream interventions into developmental agendas [437]. While global and national stewardship efforts primarily focus on mitigating antibiotic use in the human and animal sectors, the environmental drivers are often overlooked, particularly water-related factors [432]. Addressing AMR requires a multidisciplinary One Health approach acknowledging the connections between humans, animals, and their shared environment [398]. The successful mitigation of AMR will involve collaboration at the individual, community, and national levels to ensure the continued effectiveness of antimicrobials [398].

9.4.3. Fostering Collaboration across Sectors

Addressing AMR requires collaboration across multiple sectors, including healthcare, agriculture, the environment, and industry. However, implementing this approach is challenging, particularly in LMICs [438]. Barriers include the limited policy visibility of zoonotic diseases, conflicting departmental priorities, and insufficient institutional capacities [438]. Effective multisectoral coordination requires the strengthening of national policy frameworks, establishment of governance structures, and improvement of technical working groups [438]. For instance, poor awareness, inadequate regulations, and limited IPC strategies in Nigeria contribute to inappropriate antibiotic use across sectors [439]. Malaysia also faces similar challenges, with gaps in research assessing AMR’s coexistence across sectors and a limited understanding of antimicrobial use behaviors [430]. To operationalize One Health, countries must invest in governance structures, foster community participation, and promote cross-sectoral collaboration while addressing the socioeconomic and behavioral factors influencing antimicrobial use [439,440].

9.4.4. Promoting Public Awareness and Behavioral Change

Public awareness and education are crucial components in AMR management. Recent studies have explored various interventions to improve the AMR awareness and promote behavioral change. Interactive elements, such as games and videos, have shown higher engagement in public education campaigns [441]. However, there is a lack of evidence on the long-term behavioral impacts and sustained change [442]. In LMICs, multi-faceted interventions targeting healthcare professionals have been most effective when combining education with supportive measures [442]. Researchers emphasize the need for a multidisciplinary approach, incorporating insights from the social sciences, such as education and psychology, to develop more effective AMR interventions [31]. While some interventions have demonstrated cost-effectiveness, more economic evaluations are needed to support decision making in resource-constrained environments [442].

10. Conclusions

Antimicrobial resistance arises through genetic changes and environmental pressures, enabling microorganisms to survive exposure to drugs designed to inhibit or kill them. This complexity underscores the need for comprehensive strategies, including prudent antimicrobial use, robust infection control, and ongoing research on novel therapies. The epidemiology of AMR reveals its widespread and increasing prevalence across different pathogens, regions, and sectors, highlighting the urgent need for a coordinated global response. Without concerted action, the world risks a future where common infections become untreatable, severely undermining modern medicine’s achievements.
AMR has grave consequences for global health, leading to higher morbidity, mortality, and healthcare costs and straining economic resources. Addressing this crisis demands a multifaceted effort involving governments, healthcare providers, the pharmaceutical industry, and the public. By promoting ASPs, enhancing IPC, and investing in research and transboundary collaboration, the global community can mitigate the impact of AMR and preserve life-saving treatments for future generations.
The One Health approach is responsible for a paradigm shift in addressing AMR by recognizing the interconnectedness of human, animal, and environmental health. This comprehensive framework supports effective surveillance, coordinated policies, innovative research, and educational initiatives.
Policy and regulatory initiatives are essential in guiding the global AMR response. National and international policies provide frameworks for coordinated action, while regulatory frameworks ensure the responsible use of antimicrobials. Successfully implementing these initiatives requires the overcoming of challenges such as resource limitations, political will, and global coordination. However, with innovation and collaboration, significant progress can be made in combating AMR.
Antimicrobial resistance presents a complex, multifaceted challenge that requires a coordinated global response. The emergence of multidrug-resistant pathogens and novel resistance mechanisms highlights the need for new antimicrobial agents and alternative treatment strategies. The societal and ethical implications of AMR interventions must be carefully considered to ensure the equitable distribution of the benefits. Strengthening global surveillance, integrating AMR into broader health agendas, fostering cross-sectoral collaboration, and promoting public awareness are all vital components of the fight against AMR. By embracing a comprehensive and collaborative approach, we can safeguard the efficacy of antimicrobials and protect global public health for future generations.
As previously stated, the fight against AMR is a defining challenge of our time, requiring a unified global effort that spans disciplines, sectors, and borders. By understanding the mechanisms of resistance, addressing its epidemiological complexities, and embracing innovative approaches through the One Health framework, we can develop sustainable solutions to this growing threat. A collaborative approach that integrates policy, regulation, research, and public engagement is essential to preserving antimicrobials’ effectiveness and ensuring future generations’ health and safety. The time to act is now, and the stakes could not be higher.

Author Contributions

M.O. conceived of the presented manuscript and was responsible for the methodology, formal analysis, investigation, resources, writing—original draft preparation, writing—review and editing, and visualization. W.A., S.M., Á.M.-C., R.J.D.-O. and D.D.d.S. were responsible for the writing, reviewing, and editing. All authors have read and agreed to the published version of the manuscript.

Funding

This work was financed by FCT/MCTES within the scope of the projects UIDP/04378/2020 and UIDB/04378/2020 of the Research Unit on Applied Molecular Biosciences—UCIBIO; the project LA/P/0140/2020 of the Associate Laboratory Institute for Health and Bioeconomy—i4HB; and LAQV (LA/P/0008/2020 DOI 10.54499/LA/P/0008/2020, UIDP/50006/2020 DOI 10.54499/UIDP/50006/2020 and UIDB/50006/2020 DOI 10.54499/UIDB/50006/2020).

Conflicts of Interest

The authors declare no conflicts of interest.

References

  1. Merlino, J. Antimicrobial Resistance a Threat to Public Health. Microbiol. Aust. 2017, 38, 165–167. [Google Scholar] [CrossRef]
  2. Capozzi, C.; Maurici, M.; Panà, A. Antimicrobial Resistance: It Is a Global Crisis, a Slow Tsunami. Ig. Sanita Pubbl. 2019, 75, 429–450. [Google Scholar] [PubMed]
  3. Aijaz, M.; Ahmad, M.; Ansari, M.A.; Ahmad, S. Antimicrobial Resistance in a Globalized World: Current Challenges and Future Perspectives. Int. J. Pharm. Drug Des. 2023, 1, 7–22. [Google Scholar]
  4. Jindal, A.K.; Pandya, K.; Khan, I.D. Antimicrobial Resistance: A Public Health Challenge. Med. J. Armed Forces India 2015, 71, 178–181. [Google Scholar] [CrossRef] [PubMed]
  5. Larson, E. Community Factors in the Development of Antibiotic Resistance. Annu. Rev. Public Health 2015, 28, 435–447. [Google Scholar] [CrossRef]
  6. Holmes, A.H.; Moore, L.S.; Sundsfjord, A.; Steinbakk, M.; Regmi, S.; Karkey, A.; Piddock, L.J. Understanding the Mechanisms and Drivers of Antimicrobial Resistance. Lancet 2016, 387, 176–187. [Google Scholar] [CrossRef]
  7. Tenover, F.C. Mechanisms of Antimicrobial Resistance in Bacteria. Am. J. Med. 2006, 119, S3–S10. [Google Scholar] [CrossRef] [PubMed]
  8. Foster, T.J. Antibiotic Resistance in Staphylococcus aureus. Current Status and Future Prospects. FEMS Microbiol. Rev. 2017, 41, 430–449. [Google Scholar] [CrossRef] [PubMed]
  9. Eliopoulos, G.M.; Cosgrove, S.E.; Carmeli, Y. The Impact of Antimicrobial Resistance on Health and Economic Outcomes. Clin. Infect. Dis. 2003, 36, 1433–1437. [Google Scholar] [CrossRef]
  10. Cosgrove, S.E. The Relationship between Antimicrobial Resistance and Patient Outcomes: Mortality, Length of Hospital Stay, and Health Care Costs. Clin. Infect. Dis. 2006, 42 (Suppl. S2), S82–S89. [Google Scholar] [CrossRef]
  11. Maragakis, L.L.; Perencevich, E.N.; Cosgrove, S.E. Clinical and Economic Burden of Antimicrobial Resistance. Expert Rev. Anti-Infect. Ther. 2008, 6, 751–763. [Google Scholar] [CrossRef] [PubMed]
  12. Marston, H.D.; Dixon, D.M.; Knisely, J.M.; Palmore, T.N.; Fauci, A.S. Antimicrobial Resistance. JAMA 2016, 316, 1193–1204. [Google Scholar] [CrossRef] [PubMed]
  13. Premanandh, J.; Samara, B.S.; Mazen, A.N. Race against Antimicrobial Resistance Requires Coordinated Action–An Overview. Front. Microbiol. 2016, 6, 1536. [Google Scholar] [CrossRef] [PubMed]
  14. Stekel, D. First Report of Antimicrobial Resistance Pre-Dates Penicillin. Nature 2018, 562, 192. [Google Scholar] [CrossRef] [PubMed]
  15. Ramalingam, A.J. History of Antibiotics and Evolution of Resistance. Res. J. Pharm. Technol. 2015, 8, 1719–1724. [Google Scholar] [CrossRef]
  16. Rosenblatt-Farrell, N. The Landscape of Antibiotic Resistance. Environ. Health Perspect. 2009, 117, A244–A250. [Google Scholar] [CrossRef] [PubMed]
  17. Acar, J.F.; Moulin, G. Antimicrobial Resistance: A Complex Issue. Rev. Sci. Tech. 2012, 31, 23–31. [Google Scholar] [CrossRef] [PubMed]
  18. Hutchings, M.I.; Truman, A.W.; Wilkinson, B. Antibiotics: Past, Present, and Future. Curr. Opin. Microbiol. 2019, 51, 72–80. [Google Scholar] [CrossRef]
  19. Vanegas-Múnera, J.M.; Jiménez-Quiceno, J.N. Resistencia Antimicrobiana en el Siglo XXI: ¿Hacia una Era Postantibiótica? Rev. Fac. Nac. Salud Pública 2020, 38, 1–6. [Google Scholar] [CrossRef]
  20. Medina, E.; Pieper, D.H. Tackling Threats and Future Problems of Multidrug-Resistant Bacteria. In How to Overcome the Antibiotic Crisis; Springer: Singapore, 2016; pp. 3–33. [Google Scholar]
  21. Donkor, E.S.; Codjoe, F.S. Methicillin-Resistant Staphylococcus aureus and Extended-Spectrum Beta-Lactamase Producing Enterobacteriaceae: A Therapeutic Challenge in the 21st Century. Open Microbiol. J. 2019, 13, 94–100. [Google Scholar] [CrossRef]
  22. Ynion, G.P.L.; Rosal, C.J.; Ordanel, Z.A.A.; Caipang, C.M. Challenges and Emerging Molecular Approaches in Combating Antimicrobial Resistance. J. Bacteriol. Virol. 2024, 54, 12–39. [Google Scholar] [CrossRef]
  23. Kumar, S.B.; Arnipalli, S.R.; Ziouzenkova, O. Antibiotics in Food Chain: The Consequences for Antibiotic Resistance. Antibiotics 2020, 9, 688. [Google Scholar] [CrossRef] [PubMed]
  24. Silbergeld, E.K.; Graham, J.; Price, L.B. Industrial Food Animal Production, Antimicrobial Resistance, and Human Health. Annu. Rev. Public Health 2008, 29, 151–169. [Google Scholar] [CrossRef] [PubMed]
  25. Jadeja, N.B.; Worrich, A. From Gut to Mud: Dissemination of Antimicrobial Resistance between Animal and Agricultural Niches. Environ. Microbiol. 2022, 28, 3290–3306. [Google Scholar] [CrossRef] [PubMed]
  26. Khachatourians, G.G. Agricultural Use of Antibiotics and the Evolution and Transfer of Antibiotic-Resistant Bacteria. Can. Med. Assoc. J. 1998, 159, 1129–1136. [Google Scholar]
  27. Freeland, G.; Atungulu, H.N.G.; Apple, J.; Mukherjee, S. Strategies to Combat Antimicrobial Resistance from Farm to Table. Food Rev. Int. 2023, 39, 27–40. [Google Scholar] [CrossRef]
  28. Larsson, D.J.; Bengtsson-Palme, A.A.; Brandt, K.K.; de Roda Husman, A.M.; Fagerstedt, P.; Wernersson, A.S. Critical Knowledge Gaps and Research Needs Related to the Environmental Dimensions of Antibiotic Resistance. Environ. Int. 2018, 117, 132–138. [Google Scholar] [CrossRef] [PubMed]
  29. Ahmad, I.; Malak, H.A.; Abulreesh, H.H. Environmental Antimicrobial Resistance and Its Drivers: A Potential Threat to Public Health. J. Glob. Antimicrob. Resist. 2021, 27, 101–111. [Google Scholar]
  30. Wang, W.; Weng, Y.; Luo, T.; Wang, Q.; Yang, G.; Jin, Y. Antimicrobial and the Resistances in the Environment: Ecological and Health Risks, Influencing Factors, and Mitigation Strategies. Toxics 2023, 11, 185. [Google Scholar] [CrossRef]
  31. Calvo-Villamañán, A.; Álvarez, S.M.; Carrilero, L. Tackling AMR from a Multidisciplinary Perspective: A Primer from Education and Psychology. Int. Microbiol. 2023, 26, 1–9. [Google Scholar] [CrossRef]
  32. Hayes, J.F. Fighting Back against Antimicrobial Resistance with Comprehensive Policy and Education: A Narrative Review. Antibiotics 2022, 11, 644. [Google Scholar] [CrossRef] [PubMed]
  33. Williams, S.J.; Wall, S. The AMR Emergency: Multi-Sector Collaboration and Collective Global Policy Action Is Needed Now. Glob. Health Action 2019, 12 (Suppl. S1), 1855831. [Google Scholar] [CrossRef] [PubMed]
  34. Martínez-Trejo, A.; Ruiz-Ruiz, J.M.; Gonzalez-Avila, L.U.; Saldaña-Padilla, A.; Hernández-Cortez, C.; Loyola-Cruz, M.A.; Castro-Escarpulli, G. Evasion of Antimicrobial Activity in Acinetobacter baumannii by Target Site Modifications: An Effective Resistance Mechanism. Int. J. Mol. Sci. 2022, 23, 6582. [Google Scholar] [CrossRef] [PubMed]
  35. Arenz, S.; Wilson, D.N. Bacterial Protein Synthesis as a Target for Antibiotic Inhibition. Cold Spring Harb. Perspect. Med. 2016, 6, a025361. [Google Scholar] [CrossRef]
  36. Babosan, A.; Fruchard, L.; Carvalho, K.E.A.; Mazel, D.; Baharoglu, Z. Nonessential tRNA and rRNA Modifications Impact the Bacterial Response to Sub-MIC Antibiotic Pressure. Nucleic Acids Res. 2023, 51, 4878–4893. [Google Scholar]
  37. Sun, J.; Deng, Z.; Yan, A. Bacterial Multidrug Efflux Pumps: Mechanisms, Physiology and Pharmacological Exploitations. Biochem. Biophys. Res. Commun. 2014, 453, 254–267. [Google Scholar] [CrossRef]
  38. Huang, L.; Wu, C.; Gao, H.; Xu, C.; Dai, M.; Huang, L.; Cheng, G. Bacterial Multidrug Efflux Pumps at the Frontline of Antimicrobial Resistance: An Overview. Antibiotics 2022, 11, 520. [Google Scholar] [CrossRef]
  39. Zgurskaya, H.I.; Nikaido, H. Multidrug Resistance Mechanisms: Drug Efflux across Two Membranes. Mol. Microbiol. 2000, 37, 219–225. [Google Scholar] [CrossRef]
  40. Egorov, A.M.; Ulyashova, M.M.; Rubtsova, M.Y. Bacterial Enzymes and Antibiotic Resistance. Acta Naturae 2018, 10, 33–48. [Google Scholar] [CrossRef] [PubMed]
  41. Lakshmi, R.; Nusrin, K.S. Role of Beta-Lactamases in Antibiotic Resistance: A Review. Int. Res. J. Pharm 2014, 5, 37–41. [Google Scholar] [CrossRef]
  42. Agarwal, V.; Tiwari, A.; Varadwaj, P. An Extensive Review on β-Lactamase Enzymes and Their Inhibitors. Curr. Med. Chem. 2023, 30, 783–808. [Google Scholar] [CrossRef] [PubMed]
  43. Bush, K.; Bradford, P.A. β-Lactams and β-Lactamase Inhibitors: An Overview. Cold Spring Harb. Perspect. Med. 2016, 6, a025247. [Google Scholar] [CrossRef] [PubMed]
  44. Ghai, I.; Ghai, S. Understanding Antibiotic Resistance via Outer Membrane Permeability. Infect. Drug Resist. 2018, 11, 523–530. [Google Scholar] [CrossRef] [PubMed]
  45. Ghai, I.; Ghai, S. Exploring Bacterial Outer Membrane Barrier to Combat Bad Bugs. Infect. Drug Resist. 2017, 10, 261–273. [Google Scholar] [CrossRef]
  46. Horne, J.E.; Brockwell, D.J.; Radford, S.E. Role of the Lipid Bilayer in Outer Membrane Protein Folding in Gram-Negative Bacteria. J. Biol. Chem. 2020, 295, 10340–10367. [Google Scholar] [CrossRef]
  47. Sahoo, J.P.; Mishra, A.P.; Samal, K.C.; Dash, A.K. Insights into the Antibiotic Resistance in Biofilms—A Review. Environ. Conserv. J. 2021, 22, 59–67. [Google Scholar] [CrossRef]
  48. Bano, S.; Hassan, N.; Rafiq, M.; Hassan, F.; Rehman, M.; Iqbal, N.; Kang, Y.Q. Biofilms as Battlefield Armor for Bacteria against Antibiotics: Challenges and Combating Strategies. Microorganisms 2023, 11, 2595. [Google Scholar] [CrossRef]
  49. Uruén, C.; Chopo-Escuin, G.; Tommassen, J.; Romero, M.-J.R.C.; Arenas, J. Biofilms as Promoters of Bacterial Antibiotic Resistance and Tolerance. Antibiotics 2020, 10, 3. [Google Scholar] [CrossRef] [PubMed]
  50. Hathroubi, S.; Mekni, M.A.; Domenico, P.; Nguyen, D.; Jacques, M. Biofilms: Microbial Shelters against Antibiotics. Microb. Drug Resist. 2017, 23, 147–156. [Google Scholar] [CrossRef]
  51. Lou, Z.; Sun, Y.; Rao, Z. Current Progress in Antiviral Strategies. Trends Pharmacol. Sci. 2014, 35, 86–102. [Google Scholar] [CrossRef] [PubMed]
  52. Irwin, K.K.; Renzette, N.; Kowalik, T.F.; Jensen, J.D. Antiviral Drug Resistance as an Adaptive Process. Virus Evol. 2016, 2, vew014. [Google Scholar] [CrossRef] [PubMed]
  53. Weber, I.T. Can We Design Drugs for HIV/AIDS That Are Less Susceptible to Resistance? Future Med. Chem. 2015, 7, 2301–2304. [Google Scholar] [CrossRef] [PubMed]
  54. Kumar, N.; Sharma, S.; Kumar, R.; Tripathi, B.N.; Barua, S.; Ly, H.; Rouse, B.T. Host-Directed Antiviral Therapy. Clin. Microbiol. Rev. 2020, 33, 10–1128. [Google Scholar] [CrossRef] [PubMed]
  55. Flowers, S.A.; Colón, B.; Whaley, S.G.; Schuler, M.A.; Rogers, P.D. Contribution of Clinically Derived Mutations in ERG11 to Azole Resistance in Candida albicans. Antimicrob. Agents Chemother. 2015, 59, 450–460. [Google Scholar] [CrossRef] [PubMed]
  56. Engle, K.; Kumar, G. Tackling Multi-Drug Resistant Fungi by Efflux Pump Inhibitors. Biochem. Pharmacol. 2024, 226, 116400. [Google Scholar] [CrossRef]
  57. Garcia, Í.R.; de Oliveira Garcia, F.A.; Pereira, P.S.; Coutinho, H.D.M.; Siyadatpanah, A.; Norouzi, R.; Rodrigues, F.F.G. Microbial Resistance: The Role of Efflux Pump Superfamilies and Their Respective Substrates. Life Sci. 2022, 295, 120391. [Google Scholar] [CrossRef] [PubMed]
  58. Prasad, R.; Nair, R.; Banerjee, A. Multidrug Transporters of Candida Species in Clinical Azole Resistance. Fungal Genet. Biol. 2019, 132, 103252. [Google Scholar] [CrossRef] [PubMed]
  59. Cordeiro, R.D.; Portela, F.V.; Pereira, L.M.; de Andrade, A.R.; de Sousa, J.K.; Aguiar, A.L.; Sidrim, J.J. Efflux Pump Inhibition Controls Growth and Enhances Antifungal Susceptibility of Fusarium solani Species Complex. Future Microbiol. 2020, 15, 9–20. [Google Scholar] [CrossRef]
  60. Aneke, C.I.; Rhimi, W.; Otranto, D.; Cafarchia, C. Synergistic Effects of Efflux Pump Modulators on the Azole Antifungal Susceptibility of Microsporum canis. Mycopathologia 2020, 185, 279–288. [Google Scholar] [CrossRef]
  61. Cannon, R.D.; Lamping, E.; Holmes, A.R.; Niimi, K.; Baret, P.V.; Keniya, M.V.; Monk, B.C. Efflux-Mediated Antifungal Drug Resistance. Clin. Microbiol. Rev. 2009, 22, 291–321. [Google Scholar] [CrossRef]
  62. Hori, Y.; Shibuya, K. Role of FKS Gene in the Susceptibility of Pathogenic Fungi to Echinocandins. Med. Mycol. J. 2018, 59, E31–E40. [Google Scholar] [CrossRef] [PubMed]
  63. Jospe-Kaufman, M.; Ben-Zeev, E.; Mottola, A.; Dukhovny, A.; Berman, J.; Carmeli, S.; Fridman, M. Reshaping Echinocandin Antifungal Drugs to Circumvent Glucan Synthase Point-Mutation-Mediated Resistance. Angew. Chem. Int. Ed. 2024, 63, e202314728. [Google Scholar] [CrossRef] [PubMed]
  64. Costa, P.D.S.; Prado, A.; Bagon, N.P.; Negri, M.; Svidzinski, T.I.E. Mixed Fungal Biofilms: From Mycobiota to Devices, a New Challenge on Clinical Practice. Microorganisms 2022, 10, 1721. [Google Scholar] [CrossRef] [PubMed]
  65. de Barros, P.P.; Rossoni, L.; Scorzoni, L.; Fenley, J.D.C.; Junqueira, J.C. Candida Biofilms: An Update on Developmental Mechanisms and Therapeutic Challenges. Mycopathologia 2020, 185, 415–424. [Google Scholar] [CrossRef] [PubMed]
  66. Khatoon, Z.; McTiernan, C.D.; Suuronen, E.J.; Mah, T.F.; Alarcon, E.I. Bacterial Biofilm Formation on Implantable Devices and Approaches to Its Treatment and Prevention. Heliyon 2018, 4, e01067. [Google Scholar] [CrossRef]
  67. Birnbaum, J.; Scharf, S.; Schmidt, S.; Jonscher, E.; Hoeijmakers, W.A.M.; Flemming, S.; Spielmann, T. A Kelch13-Defined Endocytosis Pathway Mediates Artemisinin Resistance in Malaria Parasites. Science 2020, 367, 51–59. [Google Scholar] [CrossRef]
  68. Yang, T.; Yeoh, L.M.; Tutor, M.V.; Dixon, M.W.; McMillan, P.J.; Xie, S.C.; Cobbold, S.A. Decreased K13 Abundance Reduces Hemoglobin Catabolism and Proteotoxic Stress, Underpinning Artemisinin Resistance. Cell Rep. 2019, 29, 2917–2928. [Google Scholar] [CrossRef]
  69. Ward, K.E.; Fidock, D.A.; Bridgford, J.L. Plasmodium falciparum Resistance to Artemisinin-Based Combination Therapies. Curr. Opin. Microbiol. 2022, 69, 102–193. [Google Scholar] [CrossRef]
  70. Wicht, K.J.; Mok, S.F.D.A. Molecular Mechanisms of Drug Resistance in Plasmodium falciparum Malaria. Annu. Rev. Microbiol. 2020, 74, 431–454. [Google Scholar] [CrossRef]
  71. Ma, C.I.; Tirtorahardjo, J.A.; Jan, S.; Schweizer, S.S.; Rosario, S.A.; Du, Y.; Andrade, R.M. Auranofin Resistance in Toxoplasma gondii Decreases the Accumulation of Reactive Oxygen Species but Does Not Target Parasite Thioredoxin Reductase. Front. Cell. Infect. Microbiol. 2021, 11, 618994. [Google Scholar] [CrossRef]
  72. Tekwani, B.L.; Shukla, O.P.; Ghatak, S. Altered Drug Metabolism in Parasitic Diseases. Parasitol. Today 1988, 4, 4–10. [Google Scholar] [CrossRef] [PubMed]
  73. Wittlin, S.; Mäser, P. From Magic Bullet to Magic Bomb: Reductive Bioactivation of Antiparasitic Agents. ACS Infect. Dis. 2021, 7, 2777–2786. [Google Scholar] [CrossRef] [PubMed]
  74. Wilkinson, S.R.; Taylor, M.C.; Horn, D.; Kelly, J.M.; Cheeseman, I. A Mechanism for Cross-Resistance to Nifurtimox and Benznidazole in Trypanosomes. Proc. Natl. Acad. Sci. USA 2008, 105, 5022–5027. [Google Scholar] [CrossRef] [PubMed]
  75. Wang, J.; Xu, C.; Lun, Z.R.; Meshnick, S.R. Unpacking “Artemisinin Resistance”. Trends Pharmacol. Sci. 2017, 38, 506–511. [Google Scholar] [CrossRef]
  76. Ma, Y.; Frutos-Beltrán, E.; Kang, D.; Pannecouque, C.; De Clercq, E.; Menéndez-Arias, L.; Zhan, P. Medicinal Chemistry Strategies for Discovering Antivirals Effective against Drug-Resistant Viruses. Chem. Soc. Rev. 2021, 45, 4514–4540. [Google Scholar] [CrossRef] [PubMed]
  77. Nastri, B.M.; Pagliano, P.; Zannella, C.; Folliero, V.; Masullo, A.; Rinaldi, L.; Franci, G. HIV and Drug-Resistant Subtypes. Microorganisms 2023, 11, 221. [Google Scholar] [CrossRef]
  78. Lefaan, Y.F.M.; Setiadhi, R. Thymidine Kinase as a Causative Factor for Type 1 Herpes Simplex Virus Resistance against Acyclovir. Dentino J. Kedokt. Gigi 2020, 5, 159–164. [Google Scholar] [CrossRef]
  79. Revie, N.M.; Iyer, K.R.; Robbins, N.; Cowen, L.E. Antifungal Drug Resistance: Evolution, Mechanisms and Impact. Curr. Opin. Microbiol. 2018, 45, 70–76. [Google Scholar] [CrossRef]
  80. Arastehfar, A.; Lass-Flörl, C.; Garcia-Rubio, R.; Daneshnia, F.; Ilkit, M.; Boekhout, T.; Perlin, D.S. The Quiet and Underappreciated Rise of Drug-Resistant Invasive Fungal Pathogens. J. Fungi 2020, 6, 138. [Google Scholar] [CrossRef]
  81. Ben-Ami, R.; Kontoyiannis, D.P. Resistance to Antifungal Drugs. Infect. Dis. Clin. N. Am. 2021, 35, 279–311. [Google Scholar] [CrossRef]
  82. Fissiha, W.; Kinde, M.Z. Anthelmintic Resistance and Its Mechanism: A Review. Heliyon 2021, 7, e08487. [Google Scholar] [CrossRef] [PubMed]
  83. Alvi, M.A.; Athar Ali, R.M.; Asghar, F.; Saqib, M.; Ashfaq, K.; Rashid, I.; Ahmad, F. Drug Resistance in Parasitic Infections. In Parasitism and Parasitic Control in Animals: Strategies for the Developing World; CABI: Boston, MA, USA, 2023; pp. 111–123. [Google Scholar]
  84. Heinberg, A.; Kirkman, L. The Molecular Basis of Antifolate Resistance in Plasmodium falciparum: Looking Beyond Point Mutations. Ann. N. Y. Acad. Sci. 2015, 1342, 10–18. [Google Scholar] [CrossRef] [PubMed]
  85. Reygaert, W.C. An Overview of the Antimicrobial Resistance Mechanisms of Bacteria. AIMS Microbiol. 2018, 4, 482. [Google Scholar] [CrossRef] [PubMed]
  86. Lee, Y.; Robbins, N.; Cowen, L.E. Molecular Mechanisms Governing Antifungal Drug Resistance. NPJ Antimicrob. Resist. 2023, 1, 5. [Google Scholar] [CrossRef]
  87. Schroeder, J.W.; Yeesin, P.; Simmons, L.A.; Wang, J.D. Sources of Spontaneous Mutagenesis in Bacteria. Crit. Rev. Biochem. Mol. Biol. 2018, 53, 29–48. [Google Scholar] [CrossRef]
  88. Fukuda, H.; Hiramatsu, K. Mechanisms of Endogenous Drug Resistance Acquisition by Spontaneous Chromosomal Gene Mutation. Nihon Rinsho 1997, 55, 1185–1190. [Google Scholar] [PubMed]
  89. Bena, C.E.; Ollion, J.; De Paepe, M.; Ventroux, M.; Robert, L.; Elez, M. Real-Time Monitoring of Replication Errors’ Fate Reveals the Origin and Dynamics of Spontaneous Mutations. Nat. Commun. 2024, 15, 2702. [Google Scholar] [CrossRef]
  90. Willmott, C.J.; Maxwell, A. A Single Point Mutation in the DNA Gyrase A Protein Greatly Reduces Binding of Fluoroquinolones to the Gyrase-DNA Complex. Antimicrob. Agents Chemother. 1993, 37, 126–127. [Google Scholar] [CrossRef] [PubMed]
  91. Sarisky, R.T.; Nguyen, T.T.; Duffy, K.E.; Wittrock, R.J.; Leary, J.J. Difference in Incidence of Spontaneous Mutations Between Herpes Simplex Virus Types 1 and 2. Antimicrob. Agents Chemother. 2000, 44, 1524–1529. [Google Scholar] [CrossRef]
  92. Haver, H.L.; Chua, A.; Ghode, P.; Lakshminarayana, S.B.; Singhal, A.; Mathema, B.; Bifani, P. Mutations in Genes for the F420 Biosynthetic Pathway and a Nitroreductase Enzyme Are the Primary Resistance Determinants in Spontaneous in Vitro-Selected PA-824-Resistant Mutants of Mycobacterium tuberculosis. Antimicrob. Agents Chemother. 2015, 59, 5316–5323. [Google Scholar] [CrossRef]
  93. Revitt-Mills, S.A.; Robinson, A. Antibiotic-Induced Mutagenesis: Under the Microscope. Front. Microbiol. 2020, 11, 585175. [Google Scholar] [CrossRef] [PubMed]
  94. Shapiro, R.S. Antimicrobial-Induced DNA Damage and Genomic Instability in Microbial Pathogens. PLoS Pathog. 2015, 11, e1004678. [Google Scholar] [CrossRef] [PubMed]
  95. Blázquez, J.; Couce, A.; Rodríguez-Beltrán, J.; Rodríguez-Rojas, A. Antimicrobials as Promoters of Genetic Variation. Curr. Opin. Microbiol. 2012, 15, 561–569. [Google Scholar] [CrossRef] [PubMed]
  96. Li, X.G.A.Z.; Xie, Z.Y.B.; Li, D.; Chen, J. Sub-Lethal Concentrations of Heavy Metals Induce Antibiotic Resistance via Mutagenesis. J. Hazard. Mater. 2019, 369, 9–16. [Google Scholar] [CrossRef] [PubMed]
  97. Wang, D.; Ning, Q.; Deng, Z.; Zhang, M.; You, J. Role of Environmental Stresses in Elevating Resistance Mutations in Bacteria: Phenomena and Mechanisms. Environ. Pollut. 2022, 317, 119603. [Google Scholar] [CrossRef]
  98. Sevillya, G.; Adato, O.; Snir, S. Detecting Horizontal Gene Transfer: A Probabilistic Approach. BMC Genom. 2020, 21, 106. [Google Scholar] [CrossRef]
  99. Zhou, H.; Beltrán, J.F.; Brito, I.L. Functions Predict Horizontal Gene Transfer and the Emergence of Antibiotic Resistance. Sci. Adv. 2021, 7, eabj5056. [Google Scholar] [CrossRef]
  100. Arnold, B.J.; Huang, I.T.; Hanage, W.P. Horizontal Gene Transfer and Adaptive Evolution in Bacteria. Nat. Rev. Microbiol. 2022, 20, 206–218. [Google Scholar] [CrossRef]
  101. Van Etten, J.; Bhattacharya, D. Horizontal Gene Transfer in Eukaryotes: Not If, but How Much? Trends Genet. 2020, 36, 915–925. [Google Scholar] [CrossRef] [PubMed]
  102. Fang, G.Y.; Liu, X.Q.; Jiang, Y.J.; Mu, X.J.; Huang, B.W. Horizontal Gene Transfer in Activated Sludge Enhances Microbial Antimicrobial Resistance and Virulence. Sci. Total Environ. 2024, 912, 168908. [Google Scholar] [CrossRef]
  103. Liu, G.; Thomsen, L.E.; Olsen, J.E. Antimicrobial-Induced Horizontal Transfer of Antimicrobial Resistance Genes in Bacteria: A Mini-Review. J. Antimicrob. Chemother. 2022, 77, 556–567. [Google Scholar] [CrossRef] [PubMed]
  104. Soler, N.; Forterre, P. Vesiduction: The Fourth Way of HGT. Environ. Microbiol. 2020, 22, 2457–2460. [Google Scholar] [CrossRef] [PubMed]
  105. Virolle, C.; Goldlust, K.; Djermoun, S.; Bigot, S.; Lesterlin, C. Plasmid Transfer by Conjugation in Gram-Negative Bacteria: From the Cellular to the Community Level. Genes 2020, 11, 1239. [Google Scholar] [CrossRef] [PubMed]
  106. Waksman, G. From Conjugation to T4S Systems in Gram-Negative Bacteria: A Mechanistic Biology Perspective. EMBO Rep. 2019, 20, e47012. [Google Scholar] [CrossRef] [PubMed]
  107. Goessweiner-Mohr, N.; Arends, K.; Keller, W.; Grohmann, E. Conjugation in Gram-Positive Bacteria. In Plasmids: Biology and Impact in Biotechnology and Discovery; American Society for Microbiology Press: Washington, DC, USA, 2015; pp. 237–256. [Google Scholar]
  108. Bañuelos-Vazquez, L.A.; Tejerizo, G.T.; Brom, S. Regulation of Conjugative Transfer of Plasmids and Integrative Conjugative Elements. Plasmid 2017, 91, 82–89. [Google Scholar] [CrossRef]
  109. Kohler, V.; Keller, W.; Grohmann, E. Regulation of Gram-Positive Conjugation. Front. Microbiol. 2019, 19, 1134. [Google Scholar] [CrossRef]
  110. Meijer, W.J.; Boer, D.R.; Ares, S.; Alfonso, C.; Rojo, F.; Luque-Ortega, J.R.; Wu, L.J. Multiple Layered Control of the Conjugation Process of the Bacillus subtilis Plasmid pLS20. Front. Mol. Biosci. 2021, 8, 648468. [Google Scholar] [CrossRef]
  111. Pfeifer, E.; Bonnin, R.A.; Rocha, E.P. Phage-Plasmids Spread Antibiotic Resistance Genes Through Infection and Lysogenic Conversion. mBio 2022, 13, e01851-22. [Google Scholar] [CrossRef]
  112. Povolo, V.R.; Ackermann, M. Disseminating Antibiotic Resistance During Treatment. Science 2019, 364, 737–738. [Google Scholar] [CrossRef]
  113. Shen, Z.; Tang, C.M.; Liu, G.Y. Towards a Better Understanding of Antimicrobial Resistance Dissemination: What Can Be Learnt from Studying Model Conjugative Plasmids? Mil. Med. Res. 2022, 9, 3. [Google Scholar] [CrossRef] [PubMed]
  114. Lade, H.; Kim, J.S. Molecular determinants of β-lactam resistance in methicillin-resistant Staphylococcus aureus (MRSA): An updated review. Antibiotics 2023, 12, 1362. [Google Scholar] [CrossRef] [PubMed]
  115. Manohar, P.; Leptihn, S.; Lopes, B.S.; Nachimuthu, R. Dissemination of carbapenem resistance and plasmids encoding carbapenemases in Gram-negative bacteria isolated in India. JAC-Antimicrob. Resist. 2021, 3, dlab015. [Google Scholar] [CrossRef] [PubMed]
  116. Bodendoerfer, E.; Marchesi, M.; Imkamp, F.; Courvalin, P.; Böttger, E.C.; Mancini, S. Co-occurrence of aminoglycoside and β-lactam resistance mechanisms in aminoglycoside-non-susceptible Escherichia coli isolated in the Zurich area, Switzerland. Int. J. Antimicrob. Agents 2020, 56, 106019. [Google Scholar] [CrossRef] [PubMed]
  117. Firmo, E.F.; Beltrão, E.M.B.; da Silva, F.R.F.; Alves, L.C.; Brayner, F.A.; Veras, D.L.; Lopes, A.C.S. Association of blaNDM-1 with blaKPC-2 and aminoglycoside-modifying enzyme genes among Klebsiella pneumoniae, Proteus mirabilis and Serratia marcescens clinical isolates in Brazil. J. Glob. Antimicrob. Resist. 2020, 21, 255–261. [Google Scholar] [CrossRef]
  118. Chopjitt, P.; Boueroy, P.; Morita, M.; Iida, T.; Akeda, Y.; Hamada, S.; Kerdsin, A. Genetic characterization of multidrug-resistant Escherichia coli harboring colistin-resistant gene isolated from food animals in food supply chain. Front. Cel. Infect. Microbiol. 2024, 14, 1289134. [Google Scholar] [CrossRef] [PubMed]
  119. Ghenea, A.E.; Zlatian, O.M.; Cristea, O.M.; Ungureanu, A.; Mititelu, R.R.; Balasoiu, A.T.; Salan, A.; Iliuta, D.; Popescu, M.; Udriștoiu, A.-L.; et al. TEM, CTX-M, SHV genes in ESBL-producing Escherichia coli and Klebsiella pneumoniae isolated from clinical samples in a county clinical emergency hospital Romania-predominance of CTX-M-15. Antibiotics 2022, 11, 503. [Google Scholar] [CrossRef] [PubMed]
  120. Cheng, Y.Y.; Zhou, Z.; Papadopoulos, J.M.; Zuke, J.D.; Falbel, T.G.; Anantharaman, K.; Burton, B.M.; Venturelli, O.S. Efficient plasmid transfer via natural competence in a microbial co-culture. Mol. Syst. Biol. 2023, 19, e11406. [Google Scholar] [CrossRef] [PubMed]
  121. Winter, M.; Buckling, A.; Harms, K.; Johnsen, P.J.; Vos, M. Antimicrobial Resistance Acquisition via Natural Transformation: Context Is Everything. Curr. Opin. Microbiol. 2021, 64, 133–138. [Google Scholar] [CrossRef]
  122. Huang, M.; Liu, M.; Huang, L.; Wang, M.; Jia, R.; Zhu, D.; Chen, S.; Zhao, X.; Zhang, S.; Gaio, Q.; et al. The Activation and Limitation of the Bacterial Natural Transformation System: The Function in Genome Evolution and Stability. Microbiol. Res. 2021, 252, 126856. [Google Scholar] [CrossRef]
  123. Perez, F.; Stiefel, U. The Impact of Natural Transformation on the Acquisition of Antibiotic Resistance Determinants. mBio 2022, 13, e00336-22. [Google Scholar] [CrossRef]
  124. Carvalho, G.; Fouchet, D.; Danesh, G.; Godeux, A.S.; Laaberki, M.H.; Pontier, D.; Charpentier, X.; Venner, S. Bacterial Transformation Buffers Environmental Fluctuations Through the Reversible Integration of Mobile Genetic Elements. mBio 2020, 11, 10–1128. [Google Scholar] [CrossRef] [PubMed]
  125. Schneider, C.L. Bacteriophage-Mediated Horizontal Gene Transfer: Transduction. In Bacteriophage; Springer: Singapore, 2021; pp. 151–192. [Google Scholar]
  126. Calero-Cáceres, W.; Ye, M.; Balcázar, J.L. Bacteriophages as Environmental Reservoirs of Antibiotic Resistance. Trends Microbiol. 2019, 27, 570–577. [Google Scholar] [CrossRef] [PubMed]
  127. Colavecchio, A.; Cadieux, B.; Lo, A.; Goodridge, L.D. Bacteriophages Contribute to the Spread of Antibiotic Resistance Genes Among Foodborne Pathogens of the Enterobacteriaceae Family—A Review. Front. Microbiol. 2017, 8, 1108. [Google Scholar] [CrossRef] [PubMed]
  128. Chiang, Y.N.; Penadés, J.R.; Chen, J. Genetic Transduction by Phages and Chromosomal Islands: The New and Noncanonical. PLoS Pathog. 2019, 15, e1007878. [Google Scholar] [CrossRef] [PubMed]
  129. Chattopadhyay, M.K.; Jagannadham, M.V. Vesicles-Mediated Resistance to Antibiotics in Bacteria. Front. Microbiol. 2015, 6, 758. [Google Scholar] [CrossRef] [PubMed]
  130. Lehmkuhl, J.; Schneider, J.S.; Werth, K.L.V.; Scherff, N.; Mellmann, A.; Kampmeier, S. Role of Membrane Vesicles in the Transmission of Vancomycin Resistance in Enterococcus faecium. Sci. Rep. 2024, 14, 1895. [Google Scholar] [CrossRef]
  131. Marrec, L.; Bitbol, A.F. Resist or Perish: Fate of a Microbial Population Subjected to a Periodic Presence of Antimicrobial. PLoS Comput. Biol. 2020, 16, e1007798. [Google Scholar] [CrossRef]
  132. Bengtsson-Palme, J.; Kristiansson, E.; Larsson, D.J. Environmental Factors Influencing the Development and Spread of Antibiotic Resistance. FEMS Microbiol. Rev. 2018, 42, fux053. [Google Scholar] [CrossRef]
  133. Salam, M.A.; Al-Amin, M.Y.; Salam, M.T.; Pawar, J.S.; Akhter, N.; Rabaan, A.A.; Alqumber, M.A. Antimicrobial Resistance: A Growing Serious Threat for Global Public Health. Healthcare 2023, 11, 1946. [Google Scholar] [CrossRef]
  134. Kasimanickam, V.; Kasimanickam, M.; Kasimanickam, R. Antibiotics Use in Food Animal Production: Escalation of Antimicrobial Resistance: Where Are We Now in Combating AMR? Med. Sci. 2021, 9, 14. [Google Scholar] [CrossRef]
  135. Magouras, I.; Carmo, L.P.; Stärk, K.D.; Schüpbach-Regula, G. Antimicrobial Usage and-Resistance in Livestock: Where Should We Focus? Front. Vet. Sci. 2017, 4, 148. [Google Scholar] [CrossRef] [PubMed]
  136. Coque, T.M.; Cantón, R.; Pérez-Cobas, A.E.; Fernández-de-Bobadilla, M.D.; Baquero, F. Antimicrobial Resistance in the Global Health Network: Known Unknowns and Challenges for Efficient Responses in the 21st Century. Microorganisms 2023, 11, 105. [Google Scholar] [CrossRef] [PubMed]
  137. Guidry, C.A.; Davies, S.W.; Metzger, R.; Swenson, B.R.; Sawyer, R.G. Whence Resistance? Surg. Infect. 2015, 16, 716–720. [Google Scholar] [CrossRef] [PubMed]
  138. Heffernan, A.J.; Lim, S.M.S.; Lipman, J.; Roberts, J.A. A Personalised Approach to Antibiotic Pharmacokinetics and Pharmacodynamics in Critically Ill Patients. Anaesth. Crit. Care Pain Med. 2021, 40, 100970. [Google Scholar] [CrossRef]
  139. Goneau, L.W.; Delport, J.; Langlois, L.; Poutanen, S.M.; Razvi, H.; Reid, G.; Burton, J.P. Issues Beyond Resistance: Inadequate Antibiotic Therapy and Bacterial Hypervirulence. FEMS Microbes 2020, 1, xtaa004. [Google Scholar] [CrossRef]
  140. Zhou, S.; Nagel, J.L.K.K.S.; LaPlante, K.L.; Albin, O.R.; Pogue, J.M. Antimicrobial Stewardship and the Infection Control Practitioner: A Natural Alliance. Infect. Dis. Clin. N. Am. 2021, 35, 771–787. [Google Scholar] [CrossRef]
  141. Blanco, G.; López-Hernández, I.; Morinha, F.; López-Cerero, L. Intensive Farming as a Source of Bacterial Resistance to Antimicrobial Agents in Sedentary and Migratory Vultures: Implications for Local and Transboundary Spread. Sci. Total Environ. 2020, 739, 140356. [Google Scholar] [CrossRef]
  142. Liguori, K.; Keenum, I.; Davis, B.C.; Calarco, J.; Milligan, E.; Harwood, V.J.; Pruden, A. Antimicrobial Resistance Monitoring of Water Environments: A Framework for Standardized Methods and Quality Control. Environ. Sci. Technol. 2022, 56, 9149–9160. [Google Scholar] [CrossRef] [PubMed]
  143. Li, S.; Zhang, C.; Li, F.; Hua, T.; Zhou, Q.; Ho, S.H. Technologies Towards Antibiotic Resistance Genes (ARGs) Removal from Aquatic Environment: A Critical Review. J. Hazard. Mater. 2021, 411, 125148. [Google Scholar] [CrossRef] [PubMed]
  144. Aljeldah, M.M. Antimicrobial Resistance and Its Spread Is a Global Threat. Antibiotics 2022, 11, 1082. [Google Scholar] [CrossRef] [PubMed]
  145. Ramamurthy, T.; Ghosh, A.; Chowdhury, G.; Mukhopadhyay, A.K.; Dutta, S.; Miyoshi, S.I. Deciphering the Genetic Network and Programmed Regulation of Antimicrobial Resistance in Bacterial Pathogens. Front. Cell. Infect. Microbiol. 2022, 12, 952491. [Google Scholar] [CrossRef] [PubMed]
  146. Farquhar, K.S.; Rasouli Koohi, S.; Charlebois, D.A. Does Transcriptional Heterogeneity Facilitate the Development of Genetic Drug Resistance? BioEssays 2021, 43, 2100043. [Google Scholar] [CrossRef] [PubMed]
  147. Wang, X.; Yu, D.; Chen, L. Antimicrobial Resistance and Mechanisms of Epigenetic Regulation. Front. Cell. Infect. Microbiol. 2023, 13, 1199646. [Google Scholar] [CrossRef] [PubMed]
  148. Morrison, L.; Zembower, T.R. Antimicrobial Resistance. Gastrointest. Endosc. Clin. N. Am. 2020, 30, 619–635. [Google Scholar] [CrossRef] [PubMed]
  149. Van Camp, P.J.; Haslam, D.B.; Porollo, A. Bioinformatics Approaches to the Understanding of Molecular Mechanisms in Antimicrobial Resistance. Int. J. Mol. Sci. 2020, 21, 1363. [Google Scholar] [CrossRef] [PubMed]
  150. Sharma, A.; Singh, A.; Dar, M.A.; Kaur, R.J.; Charan, J.; Iskandar, K.; Haque, M.; Murti, K.; Ravichandiran, V.; Dhingra, S. Menace of Antimicrobial Resistance in LMICs: Current Surveillance Practices and Control Measures to Tackle Hostility. J. Infect. Public Health 2021, 15, 172–181. [Google Scholar] [CrossRef]
  151. Santajit, S.; Indrawattana, N. Mechanisms of Antimicrobial Resistance in ESKAPE Pathogens. Biomed. Res. Int. 2016, 2016, 2475067. [Google Scholar] [CrossRef]
  152. Baraka, M.A.; Alboghdadly, A.; Alshawwa, S.; Elnour, A.A.; Alsultan, H.; Alsalman, T.; Alaithan, H.; Islam, A.; El-Fass, K.A.; Mohamed, Y.; et al. Perspectives of Healthcare Professionals Regarding Factors Associated with Antimicrobial Resistance (AMR) and Their Consequences: A Cross Sectional Study in Eastern Province of Saudi Arabia. Antibiotics 2021, 10, 878. [Google Scholar] [CrossRef]
  153. Bassetti, S.; Tschudin-Sutter, S.; Egli, A.; Osthoff, M. Optimizing Antibiotic Therapies to Reduce the Risk of Bacterial Resistance. Eur. J. Intern. Med. 2022, 99, 7–12. [Google Scholar] [CrossRef]
  154. Kadri, S.S. Key Takeaways from the US CDC’s 2019 Antibiotic Resistance Threats Report for Frontline Providers. Crit. Care Med. 2020, 48, 939–945. [Google Scholar] [CrossRef]
  155. Godman, B.; Egwuenu, A.; Haque, M.; Malande, O.O.; Schellack, N.; Kumar, S.; Saleem, Z.; Sneddon, J.; Hoxha, I.; Islam, S.; et al. Strategies to Improve Antimicrobial Utilization with a Special Focus on Developing Countries. Life 2021, 11, 528. [Google Scholar] [CrossRef] [PubMed]
  156. Pierce, J.; Apisarnthanarak, A.; Schellack, N.; Cornistein, W.; Al Maani, A.; Adnan, S.; Stevens, M.P. Global Antimicrobial Stewardship with a Focus on Low-and Middle-Income Countries: A Position Statement for the International Society for Infectious Diseases. Int. J. Infect. Dis. 2020, 96, 621–629. [Google Scholar] [CrossRef] [PubMed]
  157. Pokharel, S.; Shrestha, P.; Adhikari, B. Antimicrobial Use in Food Animals and Human Health: Time to Implement ‘One Health’ Approach. Antimicrob. Resist. Infect. Control 2020, 9, 181. [Google Scholar] [CrossRef] [PubMed]
  158. Micoli, F.; Bagnoli, F.; Rappuoli, R.; Serruto, D. The Role of Vaccines in Combatting Antimicrobial Resistance. Nat. Rev. Microbiol. 2018, 19, 287–302. [Google Scholar] [CrossRef] [PubMed]
  159. Brazzoli, M.; Piccioli, D.; Marchetti, F. Challenges in Development of Vaccines Directed Toward Antimicrobial Resistant Bacterial Species. Hum. Vaccines Immunother. 2023, 19, 2228669. [Google Scholar] [CrossRef] [PubMed]
  160. Baker, S.J.; Payne, D.J.; Rappuoli, R.; De Gregorio, E. Technologies to Address Antimicrobial Resistance. Proc. Natl. Acad. Sci. USA 2018, 115, 12887–12895. [Google Scholar] [CrossRef]
  161. Payne, D.J.; Miller, L.F.; Findlay, D.; Anderson, J.; Marks, L. Time for a Change: Addressing R&D and Commercialization Challenges for Antibacterials. Philos. Trans. R. Soc. B 2015, 370, 20140086. [Google Scholar]
  162. Watkins, R.R.; Bonomo, R.A. Overview: The Ongoing Threat of Antimicrobial Resistance. Infect. Dis. Clin. N. Am. 2020, 34, 649–658. [Google Scholar] [CrossRef] [PubMed]
  163. Vercelli, C.; Amadori, M.; Gambino, G.; Re, G. Implications of Veterinary Medicine in the Comprehension and Stewardship of Antimicrobial Resistance Phenomenon. From the Origin Till Nowadays. Vet. Anim. Sci. 2022, 16, 100249. [Google Scholar] [CrossRef]
  164. Allcock, S.; Gurdasani, Y.E.H.H.M.D.; Dougan, G.; Sandhu, M.S.; Solomon, L.; Török, M.E. Antimicrobial Resistance in Human Populations: Challenges and Opportunities. Glob. Health Epidemiol. Genom. 2017, 2, e4. [Google Scholar] [CrossRef]
  165. van Leth, F.; Schultsz, C. Unbiased Antimicrobial Resistance Prevalence Estimates Through Population-Based Surveillance. Clin. Microbiol. Infect. 2023, 29, 429–433. [Google Scholar] [CrossRef] [PubMed]
  166. Iskandar, K.; Molinier, L.; Hallit, S.; Sartelli, M.; Hardcastle, T.C.; Lugova, H.; Dhindra, S.; Sharma, P.; Islam, S.; Mohammed, I.; et al. Surveillance of Antimicrobial Resistance in Low-and Middle-Income Countries: A Scattered Picture. Antimicrob. Resist. Infect. Control 2021, 10, 63. [Google Scholar] [CrossRef] [PubMed]
  167. Ranjbar, R.; Alam, M. Global Burden of Bacterial Antimicrobial Resistance in 2019: A Systematic Analysis. Evid.-Based Nurs. 2023, 26, ebnurs-2022. [Google Scholar]
  168. Hendriksen, R.S.; Munk, P.; Njage, P.; Van Bunnik, B.; McNally, L.; Lukjancenko, O.; Röder, T.; Nieuwenhuijse, D.; Pedersen, S.K.; Kjeldgaard, J.; et al. Global Monitoring of Antimicrobial Resistance Based on Metagenomics Analyses of Urban Sewage. Nat. Commun. 2019, 10, 1124. [Google Scholar] [CrossRef] [PubMed]
  169. Jernigan, J.A.; Hatfield, K.M.; Wolford, H.; Nelson, R.E.; Olubajo, B.; Reddy, S.C.; McCarthy, N.; Prabasaj, P.; McDonald, L.C.; Kallen, A.; et al. Multidrug-Resistant Bacterial Infections in US Hospitalized Patients, 2012–2017. N. Engl. J. Med. 2020, 382, 1309–1319. [Google Scholar] [CrossRef] [PubMed]
  170. Friedman, N.D.; Carmeli, Y.; Walton, A.L.; Schwaber, M.J. Carbapenem-Resistant Enterobacteriaceae: A Strategic Roadmap for Infection Control. Infect. Control Hosp. Epidemiol. 2017, 38, 580–594. [Google Scholar] [CrossRef]
  171. Vehreschild, M.J.; Haverkamp, M.; Biehl, L.M.; Lemmen, S.; Fätkenheuer, G. Vancomycin-Resistant Enterococci (VRE): A Reason to Isolate? Infection 2019, 47, 7–11. [Google Scholar] [CrossRef]
  172. Lee, A.S.; De Lencastre, H.; Garau, J.; Kluytmans, J.; Malhotra-Kumar, S.; Peschel, A.; Harbarth, S. Methicillin-Resistant Staphylococcus aureus. Nat. Rev. Dis. Primers 2018, 4, 18033. [Google Scholar] [CrossRef] [PubMed]
  173. Nurjadi, D.; Fleck, R.; Lindner, A.; Schäfer, J.; Gertler, M.; Mueller, A.; Lagler, H.; van Genderen, P.J.J.; Caumes, E.; Boutin, S.; et al. Import of Community-Associated, Methicillin-Resistant Staphylococcus aureus to Europe Through Skin and Soft-Tissue Infection in Intercontinental Travellers, 2011–2016. Clin. Microbiol. Infect. 2019, 25, 739–746. [Google Scholar] [CrossRef]
  174. Pfaller, M.A.; Cormican, M.; Flamm, R.K.; Mendes, R.E.; Jones, R.N. Temporal and Geographic Variation in Antimicrobial Susceptibility and Resistance Patterns of Enterococci: Results from the SENTRY Antimicrobial Surveillance Program, 1997–2016. Open Forum Infect. Dis. 2019, 6 (Suppl. S1), S54–S62. [Google Scholar] [CrossRef]
  175. Ansari, W.; Quintana, A.; Mohamed, N.; Patino, N.; Irani, P.; Coyle, K.; Chang, H.; Uyei, J. Impact of CRE Infections on Hospital LOS and Mortality in Asia. Value Health Reg. Issues 2020, 22, S56. [Google Scholar] [CrossRef]
  176. Sannathimmappa, M.B. Global Escalation in Carbapenem-Resistant Enterobacterales and Carbapenem-Resistant Acinetobacter baumannii Infections: Serious Threat to Human Health from the Pink Corner. Biomed. Biotechnol. Res. J. 2023, 7, 9–16. [Google Scholar] [CrossRef]
  177. Markovska, R.; Dimitrov, G.; Gergova, R.; Boyanova, L. Clostridioides difficile, a New “Superbug”. Microorganisms 2023, 11, 845. [Google Scholar] [CrossRef] [PubMed]
  178. Liu, C.; Monaghan, T.; Yadegar, A.; Louie, T.; Kao, D. Insights into the Evolving Epidemiology of Clostridioides difficile Infection and Treatment: A Global Perspective. Antibiotics 2023, 12, 1141. [Google Scholar] [CrossRef] [PubMed]
  179. Piret, J.; Boivin, G. Antiviral Resistance in Herpes Simplex Virus and Varicella-Zoster Virus Infections: Diagnosis and Management. Curr. Opin. Infect. Dis. 2016, 29, 654–662. [Google Scholar] [CrossRef] [PubMed]
  180. Menéndez-Arias, L.; Álvarez, M. Antiretroviral Therapy and Drug Resistance in Human Immunodeficiency Virus Type 2 Infection. Antivir. Res. 2014, 102, 70–86. [Google Scholar] [CrossRef]
  181. Rehermann, B.; Bertoletti, A. Immunological Aspects of Antiviral Therapy of Chronic Hepatitis B Virus and Hepatitis C Virus Infections. Hepatology 2015, 61, 712–721. [Google Scholar] [CrossRef] [PubMed]
  182. Heestermans, T.; Browne, J.L.; Aitken, S.C.; Vervoort, S.C.A.; Klipstein-Grobusch, K. Determinants of Adherence to Antiretroviral Therapy Among HIV-Positive Adults in Sub-Saharan Africa: A Systematic Review. BMJ Glob. Health 2016, 1, e000125. [Google Scholar] [CrossRef] [PubMed]
  183. Kadia, B.M.; Dimala, C.A.; Fongwen, N.T.; Smith, A.D. Barriers to and Enablers of Uptake of Antiretroviral Therapy in Integrated HIV and Tuberculosis Treatment Programmes in Sub-Saharan Africa: A Systematic Review and Meta-Analysis. AIDS Res. Ther. 2021, 18, 85. [Google Scholar] [CrossRef]
  184. Bennett, J. Concerning Features of Emerging Fungal Infections. Phys. Assist. Clin. 2023, 8, 433–452. [Google Scholar] [CrossRef]
  185. Vitiello, A.; Ferrara, F.; Boccellino, M.; Ponzo, A.; Cimmino, C.; Comberiati, E.; Zovi, A.; Clemente, S.; Sabbatucci, M. Antifungal Drug Resistance: An Emergent Health Threat. Biomedicines 2023, 11, 1063. [Google Scholar] [CrossRef] [PubMed]
  186. Ademe, M.; Girma, F. Candida auris: From Multidrug Resistance to Pan-Resistant Strains. Infect. Drug Resist. 2020, 13, 1287–1294. [Google Scholar] [PubMed]
  187. Nett, J.E. Candida auris: An Emerging Pathogen “Incognito”? PLoS Pathog. 2019, 15, e1007638. [Google Scholar] [CrossRef] [PubMed]
  188. Geremia, N.; Brugnaro, P.; Solinas, M.; Scarparo, C.; Panese, S. Candida auris as an Emergent Public Health Problem: A Current Update on European Outbreaks and Cases. Healthcare 2023, 11, 425. [Google Scholar] [CrossRef] [PubMed]
  189. Thatchanamoorthy, N.; Devi, V.R.; Chandramathi, S.; Tay, S.T. Candida auris: A Mini Review on Epidemiology in Healthcare Facilities in Asia. J. Fungi 2022, 8, 1126. [Google Scholar] [CrossRef]
  190. Wiederhold, N.P.; Patterson, T.F. Emergence of Azole Resistance in Aspergillus. Semin. Respir. Crit. Care Med. 2015, 36, 673–680. [Google Scholar] [CrossRef]
  191. Barrett, M.P.; Kyle, D.E.; Sibley, L.D.; Radke, J.B.; Tarleton, R.L. Protozoan Persister-Like Cells and Drug Treatment Failure. Nat. Rev. Microbiol. 2019, 17, 607–620. [Google Scholar] [CrossRef] [PubMed]
  192. Dhorda, M.; Amaratunga, C.; Dondorp, A.M. Artemisinin and Multidrug-Resistant Plasmodium falciparum—A Threat for Malaria Control and Elimination. Curr. Opin. Infect. Dis. 2021, 34, 432–439. [Google Scholar] [CrossRef]
  193. Cheema, H.S.; Singh, M.P. Drug Resistance in Plasmodium, Future Malaria Management Strategies and Importance of Medicinal Plants. J. Ayurvedic Herb Med. 2022, 8, 107–112. [Google Scholar] [CrossRef]
  194. Mita, T.; Venkatesan, M.; Ohashi, J.; Culleton, R.; Takahashi, N.; Tsukahara, T.; Ndounga, M.; Dysoley, L.; Endo, H.; Hombhanje, F.; et al. Limited Geographical Origin and Global Spread of Sulfadoxine-Resistant dhps Alleles in Plasmodium falciparum Populations. J. Infect. Dis. 2011, 204, 1980–1988. [Google Scholar] [CrossRef]
  195. Anand, S.; Kaur, S.; Khuda, P.; Dudhraj, V.; Rajesh, K.; Bahl, A.; Singh, S.K. A Review of Antimicrobial Surveillance Networks Across the Globe. Epidemiol. Int. 2022, 7, 13–20. [Google Scholar]
  196. Lee, A.S.; Huttner, B.D.; Catho, G.; Harbarth, S. Methicillin-Resistant Staphylococcus aureus: An Update on Prevention and Control in Acute Care Settings. Infect. Dis. Clin. N. Am. 2021, 35, 931–952. [Google Scholar] [CrossRef] [PubMed]
  197. Mondal, A.H.; Khare, K.; Saxena, P.; Debnath, P.; Mukhopadhyay, K.; Yadav, D. A Review on Colistin Resistance: An Antibiotic of Last Resort. Microorganisms 2024, 12, 772. [Google Scholar] [CrossRef] [PubMed]
  198. Allel, K.; Carvajal, L.J.C.; Garcia, P.; Cifuentes, M.; Silva, F.; Munita, J.M.; Undurraga, E.A. Trends and Socioeconomic, Demographic, and Environmental Factors Associated with Antimicrobial Resistance: A Longitudinal Analysis in 39 Hospitals in Chile 2008–2017. Lancet Reg. Health Am. 2023, 21, 100484. [Google Scholar] [CrossRef] [PubMed]
  199. Sono, T.M.; Yeika, E.; Cook, A.; Kalungia, A.; Opanga, S.A.; Acolatse, J.E.E.; Sefah, I.A.; Jelic, A.G.; Campbell, S.; Lorenzetti, G.; et al. Current Rates of Purchasing of Antibiotics Without a Prescription Across Sub-Saharan Africa; Rationale and Potential Programmes to Reduce Inappropriate Dispensing and Resistance. Expert Rev. Anti Infect. Ther. 2023, 21, 1025–1055. [Google Scholar] [CrossRef] [PubMed]
  200. Musa, K.; Okoliegbe, I.; Abdalaziz, T.; Aboushady, A.T.; Stelling, J.; Gould, I.M. Laboratory Surveillance, Quality Management, and Its Role in Addressing Antimicrobial Resistance in Africa: A Narrative Review. Antibiotics 2023, 12, 1313. [Google Scholar] [CrossRef] [PubMed]
  201. Zellweger, R.M.; Carrique-Mas, J.; Limmathurotsakul, D.; Day, N.P.J.; Thwaites, G.E.; Baker, S.; Southeast Asia Antimicrobial Resistance Network. A Current Perspective on Antimicrobial Resistance in Southeast Asia. J. Antimicrob. Chemother. 2017, 72, 2963–2972. [Google Scholar] [CrossRef]
  202. Nhung, N.T.; Cuong, N.V.; Thwaites, G.; Carrique-Mas, J. Antimicrobial Usage and Antimicrobial Resistance in Animal Production in Southeast Asia: A Review. Antibiotics 2016, 5, 37. [Google Scholar] [CrossRef]
  203. Malijan, G.M.; Howteerakul, N.; Ali, N.; Siri, S.; Kengganpanich, M.; ODSET Group; Nascimento, R.; Booton, R.D.; Turner, K.M.E.; Cooper, B.S.; et al. A Scoping Review of Antibiotic Use Practices and Drivers of Inappropriate Antibiotic Use in Animal Farms in WHO Southeast Asia Region. One Health 2022, 15, 100412. [Google Scholar] [CrossRef]
  204. Kakkar, M.; Chatterjee, P.; Chauhan, A.S.; Grace, D.; Lindahl, J.; Beeche, A.; Jing, F.; Chiu, S. Antimicrobial Resistance in South East Asia: Time to Ask the Right Questions. Glob. Health Action 2018, 11, 1483637. [Google Scholar] [CrossRef]
  205. Tacconelli, E.; Buhl, M.; Humphreys, H.; Malek, V.; Presterl, E.; Rodriguez-Baño, J.; Vos, M.C.; Zingg, W.; Mutters, N.T. Analysis of the Challenges in Implementing Guidelines to Prevent the Spread of Multidrug-Resistant Gram-Negatives in Europe. BMJ Open 2019, 9, e027683. [Google Scholar] [CrossRef] [PubMed]
  206. Kossow, A.; Stühmer, B.; Schaumburg, F.; Becker, K.; Glatz, B.; Möllers, M.; Kampmeier, S.; Mellmann, A. High Prevalence of MRSA and Multi-Resistant Gram-Negative Bacteria in Refugees Admitted to the Hospital—But No Hint of Transmission. PLoS ONE 2018, 13, e0198103. [Google Scholar] [CrossRef] [PubMed]
  207. Henderson, A.; Nimmo, G.R. Control of Healthcare- and Community-Associated MRSA: Recent Progress and Persisting Challenges. Br. Med. Bull. 2018, 125, 25–41. [Google Scholar] [CrossRef] [PubMed]
  208. Machowska, A.; Lundborg, C.S. Drivers of Irrational Use of Antibiotics in Europe. Int. J. Environ. Res. Public Health 2019, 16, 27. [Google Scholar] [CrossRef] [PubMed]
  209. Wojkowska-Mach, J.; Godman, B.; Glassman, A.; Kurdi, A.; Pilc, A.; Rozanska, A.; Skoczyński, S.; Wałaszek, M.; Bochenek, T. Antibiotic Consumption and Antimicrobial Resistance in Poland; Findings and Implications. Antimicrob. Resist. Infect. Control 2018, 7, 136. [Google Scholar] [CrossRef] [PubMed]
  210. St Cyr, S.; Kersh, E.; Weinstock, H.; Torrone, E. Trends in Multi-Drug-Resistant Gonorrhea, Gonococcal Isolate Surveillance Project, United States, 1987–2016. Open Forum Infect. Dis. 2018, 5 (Suppl. S1), S352. [Google Scholar] [CrossRef]
  211. Schlanger, K. Perspectives from North America. Sex. Transm. Infect. 2019, 95 (Suppl. S1), A30. [Google Scholar]
  212. Marcella, S.; Kanakamedala, H.; Zhou, Y.; Cai, B.; Pogue, J.M. Trends of Carbapenem Resistance in Enterobacterales in the US Between 2015 and 2019. Open Forum Infect. Dis. 2020, 7 (Suppl. S1), S644. [Google Scholar] [CrossRef]
  213. Livorsi, D.J.; Chorazy, M.L.; Schweizer, M.L.; Balkenende, E.C.; Blevins, A.E.; Nair, R.; Samore, M.H.; Nelson, R.E.; Khander, K.; Perencevich, E.N. A Systematic Review of the Epidemiology of Carbapenem-Resistant Enterobacteriaceae in the United States. Antimicrob. Resist. Infect. Control 2018, 7, 55. [Google Scholar] [CrossRef]
  214. Labarca, J.A.; Salles, M.J.C.; Seas, C.; Guzmán-Blanco, M. Carbapenem Resistance in Pseudomonas aeruginosa and Acinetobacter baumannii in the Nosocomial Setting in Latin America. Crit. Rev. Microbiol. 2016, 42, 276–292. [Google Scholar]
  215. Fabre, V.; Cosgrove, S.E.; Lessa, F.C.; Patel, T.S.; Reyes-Morales, G.; Aleman, W.R.; Alvarez, A.A.; Aquiles, B.; Arauz, A.B.; Arguello, F.; et al. Knowledge, Attitudes and Perceptions of Latin American Healthcare Workers Relating to Antibiotic Stewardship and Antibiotic Use: A Cross-Sectional Multi-Country Study. Antimicrob. Resist. Infect. Control 2024, 13, 47. [Google Scholar] [CrossRef] [PubMed]
  216. Medina-Pizzali, M.L.; Hartinger, S.M.; Salmon-Mulanovich, G.; Larson, A.; Riveros, M.; Mäusezahl, D. Antimicrobial Resistance in Rural Settings in Latin America: A Scoping Review with a One Health Lens. Int. J. Environ. Res. Public Health 2021, 18, 9837. [Google Scholar] [CrossRef] [PubMed]
  217. Forsythe, S.S.; McGreevey, W.; Whiteside, A.; Shah, M.; Cohen, J.; Hecht, R.; Bollinger, L.A.; Kinghorn, A. Twenty Years of Antiretroviral Therapy for People Living with HIV: Global Costs, Health Achievements, Economic Benefits. Health Aff. 2019, 38, 1163–1172. [Google Scholar] [CrossRef] [PubMed]
  218. Apetroaei, M.M.; Velescu, B.Ș.; Nedea, M.I.; Dinu-Pîrvu, C.E.; Drăgănescu, D.; Fâcă, A.I.; Udeanu, D.I.; Arsene, A.L. The Phenomenon of Antiretroviral Drug Resistance in the Context of Human Immunodeficiency Virus Treatment: Dynamic and Ever Evolving Subject Matter. Biomedicines 2024, 12, 915. [Google Scholar] [CrossRef] [PubMed]
  219. Toledo-Rueda, W.; Rosas-Murrieta, N.H.; Muñoz-Medina, J.E.; González-Bonilla, C.R.; Reyes-Leyva, J.; Santos-López, G. Antiviral Resistance Markers in Influenza Virus Sequences in Mexico, 2000–2017. Infect. Drug Resist. 2018, 11, 1751–1756. [Google Scholar] [CrossRef] [PubMed]
  220. Hersh, A.L.; Kronman, M.P. Inappropriate Antibiotic Prescribing: Wind at Our Backs or Flapping in the Breeze? Pediatrics 2017, 139, e20170027. [Google Scholar] [CrossRef]
  221. Weinberg, S.E.; Villedieu, A.; Bagdasarian, N.; Karah, N.; Teare, L.; Elamin, W.F. Control and Management of Multidrug-Resistant Acinetobacter baumannii: A Review of the Evidence and Proposal of Novel Approaches. Infect. Prev. Pract. 2020, 2, 100077. [Google Scholar] [CrossRef] [PubMed]
  222. Karam, G.; Chastre, J.; Wilcox, M.H.; Vincent, J.L. Antibiotic Strategies in the Era of Multidrug Resistance. Crit. Care 2016, 20, 136. [Google Scholar] [CrossRef]
  223. Fernando, S.A.; Gray, T.J.; Gottlieb, T. Healthcare-Acquired Infections: Prevention Strategies. Intern. Med. J. 2017, 47, 1341–1351. [Google Scholar] [CrossRef] [PubMed]
  224. Perez, S.; Innes, G.K.; Walters, M.S.; Mehr, J.; Arias, J.; Greeley, R.; Chew, D. Increase in Hospital-Acquired Carbapenem-Resistant Acinetobacter baumannii Infection and Colonization in an Acute Care Hospital During a Surge in COVID-19 Admissions—New Jersey, February–July 2020. Morb. Mortal. Wkly. Rep. 2020, 69, 1827–1831. [Google Scholar] [CrossRef]
  225. Slayton, R.B.; Toth, D.; Lee, B.Y.; Tanner, W.; Bartsch, S.M.; Khader, K.; Wonh, K.; Brown, K.; McKinnell, J.A.; Ray, W.; et al. Vital Signs: Estimated Effects of a Coordinated Approach for Action to Reduce Antibiotic-Resistant Infections in Health Care Facilities—United States. Am. J. Transplant. 2015, 15, 3002–3007. [Google Scholar] [CrossRef]
  226. Zakhour, J.; Haddad, S.F.; Kerbage, A.; Wertheim, H.; Tattevin, P.V.A.; Ünal, S.; Ouedraago, A.S.; Kanj, S.S. Diagnostic Stewardship in Infectious Diseases: A Continuum of Antimicrobial Stewardship in the Fight Against Antimicrobial Resistance. Int. J. Antimicrob. Agents 2023, 62, 106816. [Google Scholar] [CrossRef] [PubMed]
  227. Cheng, G.; Ning, J.; Ahmed, S.; Huang, J.; Ullah, R.; An, H.H.; Dai, M.; Wang, X.; Yuan, Z. Selection and Dissemination of Antimicrobial Resistance in Agri-Food Production. Antimicrob. Resist. Infect. Control 2019, 8, 158. [Google Scholar] [CrossRef] [PubMed]
  228. Economou, V.; Gousia, P. Agriculture and Food Animals as a Source of Antimicrobial-Resistant Bacteria. Infect. Drug Resist. 2015, 8, 49–61. [Google Scholar] [CrossRef] [PubMed]
  229. Liu, X.; Steele, J.C.; Meng, X.Z. Usage, Residue, and Human Health Risk of Antibiotics in Chinese Aquaculture: A Review. Environ. Pollut. 2017, 223, 161–169. [Google Scholar] [CrossRef]
  230. Chen, J.; Sun, R.; Pan, C.; Sun, Y.; Mai, B.; Li, Q.X. Antibiotics and Food Safety in Aquaculture. J. Agric. Food Chem. 2020, 68, 11908–11919. [Google Scholar] [CrossRef]
  231. Taylor, P.; Reeder, R. Antibiotic Use on Crops in Low and Middle-Income Countries Based on Recommendations Made by Agricultural Advisors. CABI Agric. Biosci. 2020, 1, 1. [Google Scholar] [CrossRef]
  232. Franklin, A.M.; Aga, D.S.; Cytryn, E.; Durso, L.M.; McLain, J.E.; Pruden, A.; Roberts, M.C.; Rothrock, M.J.; Snow, D.D.; Dungan, R.S. Antibiotics in Agroecosystems: Introduction to the Special Section. J. Environ. Qual. 2016, 45, 377–393. [Google Scholar] [CrossRef]
  233. Frost, I.; Van Boeckel, T.P.; Pires, J.; Craig, J.; Laxminarayan, R. Global Geographic Trends in Antimicrobial Resistance: The Role of International Travel. J. Travel Med. 2019, 26, taz036. [Google Scholar] [CrossRef] [PubMed]
  234. Sridhar, S.; Turbett, S.E.; Harris, J.B.; LaRocque, R.C. Antimicrobial-Resistant Bacteria in International Travelers. Curr. Opin. Infect. Dis. 2021, 34, 423–431. [Google Scholar] [CrossRef]
  235. Thomas, C.M.; Morkeberg, O.H.; Walker, P.F.; Stauffer, W.M. The Cost of Global Connectivity: Faster and More Efficient Spread of Antimicrobial Resistance by International Travelers–A Controversial Commentary. Travel Med. Infect. Dis. 2021, 41, 102045. [Google Scholar] [CrossRef] [PubMed]
  236. Młynarczyk-Bonikowska, B.; Majewska, A.; Malejczyk, M.; Młynarczyk, G.; Majewski, S. Multiresistant Neisseria gonorrhoeae: A New Threat in Second Decade of the XXI Century. Med. Microbiol. Immunol. 2020, 209, 95–108. [Google Scholar] [CrossRef] [PubMed]
  237. Pavli, A.; Maltezou, H.C. Infectious Complications Related to Medical Tourism. J. Travel Med. 2021, 28, taaa210. [Google Scholar] [CrossRef] [PubMed]
  238. Bokhary, H.; Pangesti, K.N.; Rashid, H.; Abd El Ghany, M.; Hill-Cawthorne, G.A. Travel-Related Antimicrobial Resistance: A Systematic Review. Trop. Med. Infect. Dis. 2021, 6, 11. [Google Scholar] [CrossRef]
  239. Martak, D.; Henriot, C.P.; Hocquet, D. Environment, Animals, and Food as Reservoirs of Antibiotic-Resistant Bacteria for Humans: One Health or More? Infect. Dis. Now 2024, 54, 104895. [Google Scholar] [CrossRef] [PubMed]
  240. Ha, D.R.; Haste, N.M.; Gluckstein, D.P. The Role of Antibiotic Stewardship in Promoting Appropriate Antibiotic Use. Am. J. Lifestyle Med. 2019, 13, 376–383. [Google Scholar] [CrossRef]
  241. Giamarellou, H.; Galani, L.; Karavasilis, T.; Ioannidis, K.; Karaiskos, I. Antimicrobial Stewardship in the Hospital Setting: A Narrative Review. Antibiotics 2023, 12, 1557. [Google Scholar] [CrossRef]
  242. Magiorakos, A.P.; Burns, K.; Rodríguez Baño, J.; Borg, M.; Daikos, G.; Dumpis, U.; Lucet, J.C.; Moro, M.L.; Tacconelli, E.; Simonse, G.S.; et al. Infection Prevention and Control Measures and Tools for the Prevention of Entry of Carbapenem-Resistant Enterobacteriaceae into Healthcare Settings: Guidance from the European Centre for Disease Prevention and Control. Antimicrob. Resist. Infect. Control 2017, 6, 113. [Google Scholar] [CrossRef]
  243. Carling, P.C.; Parry, M.F.; Olmstead, R. Environmental Approaches to Controlling Clostridioides difficile Infection in Healthcare Settings. Antimicrob. Resist. Infect. Control 2023, 12, 94. [Google Scholar]
  244. Pitout, J.D. Transmission Surveillance for Antimicrobial-Resistant Organisms in the Health System. In Microbial Transmission; ASM Press: Washington, DC, USA, 2019; pp. 215–227. [Google Scholar]
  245. Aarestrup, F.M.; Woolhouse, M.E. Using Sewage for Surveillance of Antimicrobial Resistance. Science 2020, 367, 630–632. [Google Scholar] [CrossRef]
  246. Pruden, A.; Vikesland, P.J.; Davis, B.C.; de Roda Husman, A.M. Seizing the Moment: Now Is the Time for Integrated Global Surveillance of Antimicrobial Resistance in Wastewater Environments. Curr. Opin. Microbiol. 2021, 64, 91–99. [Google Scholar] [CrossRef] [PubMed]
  247. Bhattacharya, R.; Bose, D.; Gulia, K.; Jaiswal, A. Impact of antimicrobial resistance on sustainable development goals and the integrated strategies for meeting environmental and socio-economic targets. Environ. Prog. Sustain. Energy 2024, 43, e14320. [Google Scholar] [CrossRef]
  248. Hay, S.I.; Rao, P.C.; Dolecek, C.; Day, N.P.; Stergachis, A.; Lopez, A.D.; Murray, C.J. Measuring and Mapping the Global Burden of Antimicrobial Resistance. BMC Med. 2018, 16, 78. [Google Scholar] [CrossRef]
  249. Van Hecke, O.; Wang, K.; Lee, J.J.; Roberts, N.W.; Butler, C.C. Implications of Antibiotic Resistance for Patients’ Recovery from Common Infections in the Community: A Systematic Review and Meta-Analysis. Clin. Infect. Dis. 2017, 65, 371–382. [Google Scholar] [CrossRef] [PubMed]
  250. Huemer, M.; Mairpady Shambat, S.; Brugger, S.D.; Zinkernagel, A.S. Antibiotic Resistance and Persistence—Implications for Human Health and Treatment Perspectives. EMBO Rep. 2020, 21, e51034. [Google Scholar] [CrossRef]
  251. Colomb-Cotinat, M.; Lacoste, J.; Brun-Buisson, C.; Jarlier, V.; Coignard, B.; Vaux, S. Estimating the Morbidity and Mortality Associated with Infections Due to Multidrug-Resistant Bacteria (MDRB), France, 2012. Antimicrob. Resist. Infect. Control 2016, 5, 56. [Google Scholar] [CrossRef] [PubMed]
  252. Barrasa-Villar, J.I.; Aibar-Remón, C.; Prieto-Andrés, P.; Mareca-Doñate, R.; Moliner-Lahoz, J. Impact on Morbidity, Mortality, and Length of Stay of Hospital-Acquired Infections by Resistant Microorganisms. Clin. Infect. Dis. 2017, 65, 644–652. [Google Scholar] [CrossRef]
  253. Founou, R.C.; Founou, L.L.; Essack, S.Y. Clinical and Economic Impact of Antibiotic Resistance in Developing Countries: A Systematic Review and Meta-Analysis. PLoS ONE 2017, 12, e0189621. [Google Scholar] [CrossRef] [PubMed]
  254. Simonsen, G.S. Antimicrobial Resistance Surveillance in Europe and Beyond. Eurosurveillance 2018, 23, 1800560. [Google Scholar] [CrossRef]
  255. So, M.; Walti, L. Challenges of Antimicrobial Resistance and Stewardship in Solid Organ Transplant Patients. Curr. Infect. Dis. Rep. 2022, 24, 63–75. [Google Scholar] [CrossRef]
  256. Menz, B.D.; Charani, E.; Gordon, D.L.L.A.J.; Moonesinghe, S.R.; Phillips, C.J. Surgical Antibiotic Prophylaxis in an Era of Antibiotic Resistance: Common Resistant Bacteria and Wider Considerations for Practice. Infect. Drug Resist. 2021, 10, 5235–5252. [Google Scholar] [CrossRef] [PubMed]
  257. Cerceo, E.; Deitelzweig, S.B.; Sherman, B.M.; Amin, A.N. Multidrug-Resistant Gram-Negative Bacterial Infections in the Hospital Setting: Overview, Implications for Clinical Practice, and Emerging Treatment Options. Microb. Drug Resist. 2016, 22, 412–431. [Google Scholar] [CrossRef] [PubMed]
  258. Trotter, A.J.; Aydin, A.; Strinden, M.J.; O’grady, J. Recent and Emerging Technologies for the Rapid Diagnosis of Infection and Antimicrobial Resistance. Curr. Opin. Microbiol. 2019, 51, 39–45. [Google Scholar] [CrossRef] [PubMed]
  259. Rentschler, S.; Kaiser, L.; Deigner, H.P. Emerging Options for the Diagnosis of Bacterial Infections and the Characterization of Antimicrobial Resistance. Int. J. Mol. Sci. 2021, 22, 456. [Google Scholar] [CrossRef] [PubMed]
  260. Mancuso, G.; De Gaetano, S.; Midiri, A.; Zummo, S.; Biondo, C. The Challenge of Overcoming Antibiotic Resistance in Carbapenem-Resistant Gram-Negative Bacteria: “Attack on Titan”. Microorganisms 2023, 11, 1912. [Google Scholar] [CrossRef]
  261. Paterson, D.L. The Challenge of Treating Superbugs. Semin. Respir. Crit. Care Med. 2015, 36, 001–002. [Google Scholar] [CrossRef] [PubMed]
  262. Rossolini, G.M.; Arena, F.; Pecile, P.; Pollini, S. Update on the Antibiotic Resistance Crisis. Curr. Opin. Pharmacol. 2014, 18, 56–60. [Google Scholar] [CrossRef]
  263. Morris, S.; Cerceo, E. Trends, Epidemiology, and Management of Multi-Drug Resistant Gram-Negative Bacterial Infections in the Hospitalized Setting. Antibiotics 2020, 9, 196. [Google Scholar] [CrossRef]
  264. Margolis, E.; Rosch, J.W. Fitness Landscape of the Immune Compromised Favors the Emergence of Antibiotic Resistance. ACS Infect. Dis. 2018, 4, 1275–1277. [Google Scholar] [CrossRef]
  265. Feuerstadt, P.; Theriault, N.; Tillotson, G. The Burden of CDI in the United States: A Multifactorial Challenge. BMC Infect. Dis. 2023, 23, 132. [Google Scholar] [CrossRef]
  266. Dadgostar, P. Antimicrobial Resistance: Implications and Costs. Infect. Drug Resist. 2019, 12, 3903–3910. [Google Scholar] [CrossRef] [PubMed]
  267. Broderick, N.; Walsh, K.A.; O’Brien, K.K.; Smith, S.S.; Harrington, P.; O’Neill, M.; Ryan, M.; Fawsitt, C.G. POSC124 Economic Burden of Antimicrobial Resistance: An Analysis of the Additional Bed Day Costs Associated with Treating Resistant Infections in Ireland. Value Health 2022, 25, S111. [Google Scholar] [CrossRef]
  268. Touat, M.; Opatowski, M.; Brun-Buisson, C.; Cosker, K.; Guillemot, D.; Salomon, J.; Tuppin, P.; de Lagasnerie, G.; Watier, L. A Payer Perspective of the Hospital Inpatient Additional Care Costs of Antimicrobial Resistance in France: A Matched Case–Control Study. Appl. Health Econ. Health Policy 2019, 17, 381–389. [Google Scholar] [CrossRef] [PubMed]
  269. Thorpe, K.E.; Joski, P.; Johnston, K.J. Antibiotic-Resistant Infection Treatment Costs Have Doubled Since 2002, Now Exceeding $2 Billion Annually. Health Aff. 2018, 37, 662–669. [Google Scholar] [CrossRef] [PubMed]
  270. Poudel, A.N.; Zhu, S.; Cooper, N.; Little, P.; Tarrant, C.; Hickman, M.; Yao, G. The Economic Burden of Antibiotic Resistance: A Systematic Review and Meta-Analysis. PLoS ONE 2023, 18, e0285170. [Google Scholar] [CrossRef]
  271. Ait Ouakrim, D.; Cassini, A.; Cecchini, M.; Plauchoras, D. The Health and Economic Burden of Antimicrobial Resistance. Eur. J. Public Health 2020, 30 (Suppl. S5), ckaa165-1201. [Google Scholar] [CrossRef]
  272. Shrestha, P.; Cooper, B.S.; Coast, J.; Oppong, R.; Do Thi Thuy, N.; Phodha, T.; Celhay, O.; Guerin, P.J.; Wertheim, H.; Lubell, Y. 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. [Google Scholar] [CrossRef]
  273. Cole, S. Who Will Develop New Antibacterial Agents? Philos. Trans. R. Soc. B 2014, 368, 20130430. [Google Scholar] [CrossRef] [PubMed]
  274. Tomaras, A.P.; Dunman, P.M. In the Midst of the Antimicrobial Discovery Conundrum: An Overview. Curr. Opin. Microbiol. 2015, 27, 103–107. [Google Scholar] [CrossRef]
  275. Mattar, C.; Edwards, S.; Baraldi, E.; Hood, J. An Overview of the Global Antimicrobial Resistance Research and Development Hub and the Current Landscape. Curr. Opin. Microbiol. 2020, 57, 56–61. [Google Scholar] [CrossRef]
  276. Guardabassi, L. Antimicrobial Resistance: A Global Threat with Remarkable Geographical Differences. N. Z. Vet. J. 2017, 65, 57–59. [Google Scholar] [CrossRef] [PubMed]
  277. Keown, O.P.; Warburton, W.; Davies, S.C.; Darzi, A. Antimicrobial Resistance: Addressing the Global Threat through Greater Awareness and Transformative Action. Health Aff. 2014, 33, 1620–1626. [Google Scholar] [CrossRef] [PubMed]
  278. Howard, S.J.; Hopwood, S.; Davies, S.C. Antimicrobial Resistance: A Global Challenge. Sci. Transl. Med. 2014, 6, 236ed10. [Google Scholar] [CrossRef]
  279. Willemsen, A.; Reid, S.; Assefa, Y. A Review of National Action Plans on Antimicrobial Resistance: Strengths and Weaknesses. Antimicrob. Resist. Infect. Control 2022, 11, 90. [Google Scholar] [CrossRef]
  280. Munkholm, L.; Rubin, O. The Global Governance of Antimicrobial Resistance: A Cross-Country Study of Alignment between the Global Action Plan and National Action Plans. Glob. Health 2020, 16, 109. [Google Scholar] [CrossRef] [PubMed]
  281. Anderson, M.; Schulze, K.; Cassini, A.; Plauchoras, D.; Mossialos, E. Strengthening Implementation of Antimicrobial Resistance National Action Plans. Eur. J. Public Health 2020, 30 (Suppl. S5), ckaa165-1200. [Google Scholar] [CrossRef]
  282. Littmann, J.; Buyx, A.; Cars, O. Antibiotic Resistance: An Ethical Challenge. Int. J. Antimicrob. Agents 2015, 46, 359–361. [Google Scholar] [CrossRef] [PubMed]
  283. Saint, V.; Mohsenpour, A.; Mühling, C.; Bozorgmehr, K. Exploring Equity, Gender and Social Determinants Aspects of Antimicrobial Resistance: Scoping Review. Eur. J. Public Health 2020, 30 (Suppl. S5), ckaa166-707. [Google Scholar] [CrossRef]
  284. Saint, V. Exploring Equity, Social Determinants of Health and Gender Considerations for Antimicrobial Resistance. Eur. J. Public Health 2019, 29 (Suppl. S4), ckz185-799. [Google Scholar] [CrossRef]
  285. Adebisi, Y.A.; Ogunkola, I.O. The Global Antimicrobial Resistance Response Effort Must Not Exclude Marginalized Populations. Trop. Med. Health 2023, 51, 33. [Google Scholar] [CrossRef]
  286. Chioro, A.; Coll-Seck, A.M.; Høie, B.; Moeloek, N.; Motsoaledi, A.; Rajatanavin, R.; Touraine, M. Antimicrobial Resistance: A Priority for Global Health Action. Bull. World Health Organ. 2015, 93, 439. [Google Scholar] [CrossRef] [PubMed]
  287. Pokharel, S.; Adhikari, B.; Johnson, T.; Cheah, P.Y. Interventions to Address Antimicrobial Resistance: An Ethical Analysis of Key Tensions and How They Apply in Low-Income and Middle-Income Countries. BMJ Glob. Health 2024, 9, e012874. [Google Scholar] [CrossRef] [PubMed]
  288. Adebisi, Y.A. Balancing the Risks and Benefits of Antibiotic Use in a Globalized World: The Ethics of Antimicrobial Resistance. Glob. Health 2023, 19, 27. [Google Scholar] [CrossRef] [PubMed]
  289. Parsonage, B.; Hagglund, P.K.; Keogh, L.; Wheelhouse, N.; Brown, R.E.; Dancer, S.J. Control of Antimicrobial Resistance Requires an Ethical Approach. Front. Microbiol. 2017, 8, 2124. [Google Scholar] [CrossRef] [PubMed]
  290. Majumder, M.A.A.; Rahman, S.; Cohall, D.; Bharatha, A.; Singh, K.; Haque, M.; Gittens-St Hilaire, M. Antimicrobial Stewardship: Fighting Antimicrobial Resistance and Protecting Global Public Health. Infect. Drug Resist. 2020, 13, 4713–4738. [Google Scholar] [CrossRef]
  291. Aryee, A.; Price, N. Antimicrobial Stewardship—Can We Afford to Do without It? Br. J. Clin. Pharmacol. 2015, 79, 173–181. [Google Scholar] [CrossRef] [PubMed]
  292. Büchler, H.S. Challenges and Success Stories of the Implementation of Infection Control and Antimicrobial Stewardship Strategies. In Proceedings of the 5th Global Ministerial Summit on Patient Safety, Montreux, Switzerland, 17–18 April 2024. [Google Scholar]
  293. Gitaka, J.; Kamita, M.; Mureithi, D.; Ndegwa, D.; Masika, M.; Omuse, G.; Ngari, M.; Makokha, F.; Mwaura, P.; Mathai, R.; et al. Combating Antibiotic Resistance Using Guidelines and Enhanced Stewardship in Kenya: A Protocol for an Implementation Science Approach. BMJ Open 2020, 10, e030823. [Google Scholar] [CrossRef] [PubMed]
  294. Hara, G.L. The Role of the ‘A’ Team in the Hospital near You. Int. J. Infect. Dis. 2018, 72, 45. [Google Scholar] [CrossRef]
  295. Minejima, E.; Wong-Beringer, A. Implementation of Rapid Diagnostics with Antimicrobial Stewardship. Expert Rev. Anti-Infect. Ther. 2016, 14, 1065–1075. [Google Scholar] [CrossRef]
  296. Banerjee, R.; Patel, R. Molecular Diagnostics for Genotypic Detection of Antibiotic Resistance: Current Landscape and Future Directions. JAC Antimicrob. Resist. 2023, 5, dlad018. [Google Scholar] [CrossRef]
  297. Lee, C.R.; Lee, J.H.; Kang, L.W.; Jeong, B.C.; Lee, S.H. Educational Effectiveness, Target, and Content for Prudent Antibiotic Use. Biomed. Res. Int. 2015, 2015, 214021. [Google Scholar] [CrossRef] [PubMed]
  298. McNicholas, M.; Hooper, G. Effects of Patient Education to Reduce Antibiotic Prescribing Rates for Upper Respiratory Infections in Primary Care. Fam. Pract. 2022, 39, 1–5. [Google Scholar] [CrossRef] [PubMed]
  299. Smiddy, M.P.; Murphy, O.M.; Savage, E.; Fitzgerald, A.P.; O’Sullivan, B.; Murphy, C.; Bernard, M.; Browne, J.P. Efficacy of Observational Hand Hygiene Audit with Targeted Feedback on Doctors’ Hand Hygiene Compliance: A Retrospective Time Series Analysis. J. Infect. Prev. 2019, 20, 164–170. [Google Scholar] [CrossRef] [PubMed]
  300. Glowicz, J.B.; Landon, E.; Sickbert-Bennett, E.E.; Aiello, A.E.; Dekay, K.; Hoffmann, K.K.; Maragakis, L.; Olmsted, R.N.; Polgreen, P.M.; Trexler, P.A.; et al. SHEA/IDSA/APIC Practice Recommendation: Strategies to Prevent Healthcare-Associated Infections through Hand Hygiene: 2022 Update. Infect. Control Hosp. Epidemiol. 2023, 44, 355–376. [Google Scholar] [CrossRef] [PubMed]
  301. Donati, D.; Miccoli, G.A.; Cianfrocca, C.; Di Stasio, E.; De Marinis, M.G.; Tartaglini, D. Effectiveness of Implementing Link Nurses and Audits and Feedback to Improve Nurses’ Compliance with Standard Precautions: A Cluster Randomized Controlled Trial. Am. J. Infect. Control 2020, 48, 1204–1210. [Google Scholar] [CrossRef]
  302. Cordeiro, L.; Gnatta, J.R.; Ciofi-Silva, C.L.; Price, A.; de Oliveira, N.A.; Almeida, R.M.; Mainardi, G.M.; Srinivas, S.; Chan, W.; Levin, A.S.S.; et al. Personal Protective Equipment Implementation in Healthcare: A Scoping Review. Am. J. Infect. Control 2022, 50, 898–905. [Google Scholar] [CrossRef]
  303. Inkster, T.; Walker, J.; Weinbren, M. Water-Free Patient Care, a Narrative Review of the Literature and Discussion of the Pressing Need for a Way Forward. J. Hosp. Infect. 2024, 152, 36–41. [Google Scholar] [CrossRef] [PubMed]
  304. Low, J.M.; Chan, M.; Low, J.L.; Chua, M.C.W.; Lee, J.H. The Impact of Sink Removal and Other Water-Free Interventions in Intensive Care Units on Water-Borne Healthcare-Associated Infections: A Systematic Review. J. Hosp. Infect. 2024, 150, 61–71. [Google Scholar] [CrossRef]
  305. Rutala, W.A.; Boyce, J.M.; Weber, D.J. Disinfection, Sterilization and Antisepsis: An Overview. Am. J. Infect. Control 2023, 51, A3–A12. [Google Scholar] [CrossRef]
  306. Rutala, W.A.; Donskey, C.J.; Weber, D.J. Disinfection and Sterilization: New Technologies. Am. J. Infect. Control 2023, 51, A13–A21. [Google Scholar] [CrossRef]
  307. Porter, L.; Sultan, O.; Mitchell, B.G.; Jenney, A.; Kiernan, M.; Brewster, D.J.; Russo, P.L. How Long Do Nosocomial Pathogens Persist on Inanimate Surfaces? J. Hosp. Infect. 2024, 147, 25–31. [Google Scholar] [CrossRef] [PubMed]
  308. Santos-Marques, C.; Ferreira, H.; Pereira, S.G. Infection Prevention and Control Strategies against Carbapenem Resistant Enterobacteriaceae–A Systematic Review. J. Infect. Prev. 2022, 23, 167–185. [Google Scholar] [CrossRef] [PubMed]
  309. Saliba, R.; Zahar, J.R.; Dabar, G.; Riachy, M.; Karam-Sarkis, D.; Husni, R. Limiting the Spread of Multidrug-Resistant Bacteria in Low-to-Middle-Income Countries: One Size Does Not Fit All. Pathogens 2023, 12, 144. [Google Scholar] [CrossRef] [PubMed]
  310. Jit, M.; Cooper, B. The Role of Vaccines in Combating Antimicrobial Resistance. Eur. J. Public Health 2020, 30 (Suppl. S5), ckaa165-1204. [Google Scholar] [CrossRef]
  311. Van Heuvel, L.; Caini, S.; Dückers, M.L.; Paget, J. Assessment of the Inclusion of Vaccination as an Intervention to Reduce Antimicrobial Resistance in AMR National Action Plans: A Global Review. Glob. Health 2022, 18, 85. [Google Scholar] [CrossRef]
  312. Ba-Nguz, A.; Shah, A.; Bresee, J.S.; Lafond, C.K.E.; Donadel, M.; Seward, J.F. Supporting National Immunization Technical Advisory Groups (NITAGs) in Resource-Constrained Settings: New Strategies and Lessons Learned from the Task Force for Global Health’s Partnership for Influenza Vaccine Introduction. Vaccine 2019, 37, 3446–3653. [Google Scholar] [CrossRef] [PubMed]
  313. Costanzo, V.; Roviello, G.N. The Potential Role of Vaccines in Preventing Antimicrobial Resistance (AMR): An Update and Future Perspectives. Vaccines 2023, 11, 333. [Google Scholar] [CrossRef]
  314. Davies, N.G.; Jit, F.S.M.; Atkins, K.E. Modeling the Effect of Vaccination on Selection for Antibiotic Resistance in Streptococcus pneumoniae. Sci. Transl. Med. 2021, 13, eaaz8690. [Google Scholar] [CrossRef] [PubMed]
  315. Perez, F.; Villegas, M.V. The Role of Surveillance Systems in Confronting the Global Crisis of Antibiotic-Resistant Bacteria. Curr. Opin. Infect. Dis. 2015, 18, 375–383. [Google Scholar] [CrossRef]
  316. Johnson, A.P. Surveillance of Antibiotic Resistance. Philos. Trans. R. Soc. B 2015, 370, 20140080. [Google Scholar] [CrossRef]
  317. Cantón, R.; Gottlieb, T.; Coombs, G.W.; Woo, P.C.; Korman, T.M.; Garcia-Castillo, M.; Daley, D.; Bauer, K.A.; Wong, M.; Wolf, D.J.; et al. Antimicrobial Surveillance: A 20-Year History of the SMART Approach to Addressing Global Antimicrobial Resistance into the Future. Int. J. Antimicrob. Agents 2023, 62, 107014. [Google Scholar] [CrossRef] [PubMed]
  318. Altorf-van der Kuil, W.; Schoffelen, A.F.; de Greeff, S.C.; Thijsen, S.F.; Alblas, H.J.; Notermans, D.W.; Vlek, A.L.M.; van der Sande, M.A.B.; Leenstra, T.; National AMR Surveillance Study Group. National Laboratory-Based Surveillance System for Antimicrobial Resistance: A Successful Tool to Support the Control of Antimicrobial Resistance in the Netherlands. Eurosurveillance 2017, 22, 17–00062. [Google Scholar] [CrossRef] [PubMed]
  319. Lee, H.; Yoon, E.J.; Kim, D.; Jeong, S.H.; Shin, J.H.; Shin, J.H.; Kim, Y.A.; Uh, Y.; Park, C.; Lee, K.J. Establishment of the South Korean National Antimicrobial Resistance Surveillance System, Kor-GLASS, in 2016. Eurosurveillance 2018, 23, 1700734. [Google Scholar] [CrossRef]
  320. Seale, A.C.; Gordon, N.C.I.J.; Peacock, S.J.; Scott, J.A.G. AMR Surveillance in Low and Middle-Income Settings—A Roadmap for Participation in the Global Antimicrobial Surveillance System (GLASS). Wellcome Open Res. 2017, 2, 92. [Google Scholar] [CrossRef] [PubMed]
  321. Sirijatuphat, R.; Chayangsu, S.; Srisompong, J.; Ruangkriengsin, D.; Thamlikitkul, V.; Tiengrim, S.; Wangchinda, W.; Koomanachai, P.; Rattanaumpawan, P. Feasibility, Challenges, and Benefits of Global Antimicrobial Resistance Surveillance System Implementation: Results from a Multicenter Quasi-Experimental Study. Antibiotics 2022, 11, 348. [Google Scholar] [CrossRef] [PubMed]
  322. Ajulo, S.; Awosile, B. 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. [Google Scholar] [CrossRef] [PubMed]
  323. Sibani, M.; Mazzaferri, F.; Carrara, E.; Pezzani, M.D.; Arieti, F.; Göpel, S.; Paul, M.; Tacconelli, E.; Mutters, N.T.; Vos, A.; et al. White Paper: Bridging the Gap between Surveillance Data and Antimicrobial Stewardship in Long-Term Care Facilities—Practical Guidance from the JPIAMR ARCH and COMBACTE-MAGNET EPI-Net Networks. J. Antimicrob. Chemother. 2020, 75 (Suppl. S2), ii33–ii41. [Google Scholar] [CrossRef]
  324. Shukla, R.; Lavore, F.; Maity, S.; Derks, M.G.N.; Jones, C.R.; Vermeulen, B.J.A.; Melcrová, A.; Morris, M.A.; Becker, L.M.; Wang, X.; et al. Teixobactin kills bacteria by a two-pronged attack on the cell envelope. Nature 2022, 608, 390–396. [Google Scholar] [CrossRef]
  325. Domingo-Calap, P.; Delgado-Martínez, J. Bacteriophages: Protagonists of a Post-Antibiotic Era. Antibiotics 2018, 7, 66. [Google Scholar] [CrossRef]
  326. Garvey, M. Bacteriophages and the One Health Approach to Combat Multidrug Resistance: Is This the Way? Antibiotics 2020, 9, 414. [Google Scholar] [CrossRef]
  327. Wan, X.; Hendrix, H.; Skurnik, M.; Lavigne, R. Phage-Based Target Discovery and Its Exploitation towards Novel Antibacterial Molecules. Curr. Opin. Biotechnol. 2021, 68, 1–7. [Google Scholar] [CrossRef] [PubMed]
  328. Lazzaro, B.P.; Zasloff, M.; Rolff, J. Antimicrobial Peptides: Application Informed by Evolution. Science 2020, 368, eaau5480. [Google Scholar] [CrossRef] [PubMed]
  329. Ageitos, J.M.; Sánchez-Pérez, A.; Calo-Mata, P.; Villa, T.G. Antimicrobial Peptides (AMPs): Ancient Compounds That Represent Novel Weapons in the Fight against Bacteria. Biochem. Pharmacol. 2017, 133, 117–138. [Google Scholar] [CrossRef] [PubMed]
  330. Nuti, R.; Goud, N.S.; Saraswati, A.P.; Alvala, R.; Alvala, M. Antimicrobial Peptides: A Promising Therapeutic Strategy in Tackling Antimicrobial Resistance. Curr. Med. Chem. 2017, 24, 4303–4314. [Google Scholar] [CrossRef] [PubMed]
  331. Streicher, L.M. Exploring the Future of Infectious Disease Treatment in a Post-Antibiotic Era: A Comparative Review of Alternative Therapeutics. J. Glob. Antimicrob. Resist. 2021, 24, 285–295. [Google Scholar] [CrossRef]
  332. Ghosh, C.; Sarkar, P.; Issa, R.; Haldar, J. Alternatives to Conventional Antibiotics in the Era of Antimicrobial Resistance. Trends Microbiol. 2019, 27, 323–338. [Google Scholar] [CrossRef] [PubMed]
  333. Shang, Z.; Chan, S.Y.; Song, Q.; Li, P.; Huang, W. The Strategies of Pathogen-Oriented Therapy on Circumventing Antimicrobial Resistance. Research 2020, 2020, 2016201. [Google Scholar] [CrossRef] [PubMed]
  334. Sheikh, B.A.; Bhat, B.A.; Mir, M.A. Antimicrobial resistance: New insights and therapeutic implications. App. Microbiol. Biotechnol. 2022, 106, 6427–6440. [Google Scholar] [CrossRef]
  335. Wang, C.H.; Hsieh, Y.H.; Powers, Z.M.; Kao, C.Y. Defeating Antibiotic-Resistant Bacteria: Exploring Alternative Therapies for a Post-Antibiotic Era. Int. J. Mol. Sci. 2020, 21, 1061. [Google Scholar] [CrossRef]
  336. Weldick, P.J.; Wang, A.; Halbus, A.F.; Paunov, V.N. Emerging Nanotechnologies for Targeting Antimicrobial Resistance. Nanoscale 2022, 14, 4018–4041. [Google Scholar] [CrossRef]
  337. Kumar, M.; Curtis, A.; Hoskins, C. Application of Nanoparticle Technologies in the Combat against Antimicrobial Resistance. Pharmaceutics 2018, 10, 11. [Google Scholar] [CrossRef] [PubMed]
  338. Lv, J.; Deng, S.; Zhang, L. A Review of Artificial Intelligence Applications for Antimicrobial Resistance. Biosaf. Health 2021, 3, 22–31. [Google Scholar] [CrossRef]
  339. Melo, M.C.; Maasch, J.R.; de la Fuente-Nunez, C. Accelerating Antibiotic Discovery through Artificial Intelligence. Commun. Biol. 2021, 4, 1050. [Google Scholar] [CrossRef]
  340. Cella, E.; Giovanetti, M.; Benedetti, F.; Scarpa, F.; Johnston, C.; Borsetti, A.; Azarian, T.; Zella, D.; Ciccozzi, M. Joining Forces against Antibiotic Resistance: The One Health Solution. Pathogens 2023, 12, 1074. [Google Scholar] [CrossRef] [PubMed]
  341. Nardulli, P.; Ballini, A.; Zamparella, M.; De Vito, D. The Role of Stakeholders’ Understandings in Emerging Antimicrobial Resistance: A One Health Approach. Microorganisms 2023, 11, 2797. [Google Scholar] [CrossRef]
  342. Kamenshchikova, A.; Wolffs, P.F.; Hoebe, C.J.; Horstman, K. Anthropocentric Framings of One Health: An Analysis of International Antimicrobial Resistance Policy Documents. Crit. Public Health 2021, 31, 306–315. [Google Scholar] [CrossRef]
  343. Velazquez-Meza, M.E.; Galarde-López, M.; Carrillo-Quiróz, B.; Alpuche-Aranda, C.M. Antimicrobial Resistance: One Health Approach. Vet. World 2022, 15, 743. [Google Scholar] [CrossRef]
  344. Aslam, B.; Khurshid, M.; Arshad, M.I.; Muzammil, S.; Rasool, M.; Yasmeen, N.; Shah, T.; Chaudhry, T.H.; Rasool, M.H.; Shahid, A.; et al. Antibiotic Resistance: One Health One World Outlook. Front. Cell. Infect. Microbiol. 2021, 11, 771510. [Google Scholar] [CrossRef]
  345. Van Duin, D.; Paterson, D.L. Multidrug-Resistant Bacteria in the Community: Trends and Lessons Learned. Infect. Dis. Clin. N. Am. 2016, 30, 377–390. [Google Scholar] [CrossRef]
  346. Davis, M.E.; Liu, T.L.; Taylor, Y.J.; Davidson, L.; Schmid, M.; Yates, T.; Scotton, J.; Spencer, M.D. Exploring Patient Awareness and Perceptions of the Appropriate Use of Antibiotics: A Mixed-Methods Study. Antibiotics 2017, 6, 23. [Google Scholar] [CrossRef]
  347. Kohut, M.R.; Keller, S.C.; Linder, J.A.; Tamma, P.D.; Cosgrove, S.E.; Speck, K.; Ahn, R.; Dullabh, P.; Miller, M.A.; Szymczak, J.E. The Inconvincible Patient: How Clinicians Perceive Demand for Antibiotics in the Outpatient Setting. Fam. Pract. 2019, 37, 276–282. [Google Scholar] [CrossRef] [PubMed]
  348. Magin, P.; Davey, A.R.; Davis, J. Evidence-Based Strategies for Better Antibiotic Prescribing. Aust. J. Gen. Pract. 2022, 51, 21–24. [Google Scholar] [CrossRef] [PubMed]
  349. Lekshmi, M.; Ammini, P.; Kumar, S.; Varela, M.F. The Food Production Environment and the Development of Antimicrobial Resistance in Human Pathogens of Animal Origin. Microorganisms 2017, 5, 11. [Google Scholar] [CrossRef]
  350. Kim, J.; Ahn, J. Emergence and Spread of Antibiotic-Resistant Foodborne Pathogens from Farm to Table. Food Sci. Biotechnol. 2022, 31, 1481–1499. [Google Scholar] [CrossRef] [PubMed]
  351. Vora, N.M.; Hannah, L.; Walzer, C.; Vale, M.M.; Lieberman, S.; Emerson, A.; Jennings, J.; Bonds, M.H.; Evans, J.; Chilukuri, B.; et al. Interventions to Reduce Risk for Pathogen Spillover and Early Disease Spread to Prevent Outbreaks, Epidemics, and Pandemics. Emerg. Infect. Dis. 2023, 3, e221079. [Google Scholar] [CrossRef]
  352. Iwu, C.D.; Korsten, L.; Okoh, A.I. The Incidence of Antibiotic Resistance within and Beyond the Agricultural Ecosystem: A Concern for Public Health. Microbiol. Open 2020, 9, e1035. [Google Scholar] [CrossRef] [PubMed]
  353. Collineau, L.; Bourély, C.; Rousset, L.; Berger-Carbonne, A.; Ploy, M.C.; Pulcini, C.; Colomb-Cotinat, M. Towards One Health Surveillance of Antibiotic Resistance: Characterisation and Mapping of Existing Programmes in Humans, Animals, Food and the Environment in France, 2021. Eurosurveillance 2023, 28, 2200804. [Google Scholar] [CrossRef]
  354. Oberin, M.; Badger, S.; Faverjon, C.; Cameron, A.; Bannister-Tyrrell, M. Electronic Information Systems for One Health Surveillance of Antimicrobial Resistance: A Systematic Scoping Review. BMJ Glob. Health 2022, 7, e007388. [Google Scholar] [CrossRef] [PubMed]
  355. Aenishaenslin, C.; Häsler, B.; Ravel, A.; Parmley, E.J.; Mediouni, S.; Bennani, H.; Stärk, K.D.C.; Buckeridge, D.L. Evaluating the Integration of One Health in Surveillance Systems for Antimicrobial Use and Resistance: A Conceptual Framework. Front. Vet. Sci. 2021, 8, 611931. [Google Scholar] [CrossRef]
  356. Joshi, M.P.; Hafner, T.; Twesigye, G.; Ndiaye, A.; Kiggundu, R.; Mekonnen, N.; Kusu, N.; Berthé, S.; Lusaya, E.P.; Acho, A. Strengthening Multisectoral Coordination on Antimicrobial Resistance: A Landscape Analysis of Efforts in 11 Countries. J. Pharm. Policy Pract. 2021, 21, 27. [Google Scholar] [CrossRef]
  357. Falkenberg, T. Applying a One Health Approach to Inter- and Trans-Disciplinary Research on Antimicrobial Resistance. Eur. J. Public Health 2019, 29 (Suppl. S4), ckz185-798. [Google Scholar] [CrossRef]
  358. Rhouma, M.; Archambault, M.; Butaye, P. Antimicrobial Use and Resistance in Animals from a One Health Perspective. Vet. Sci. 2023, 10, 319. [Google Scholar] [CrossRef] [PubMed]
  359. Marvasi, M.; Casillas, L.; Vassallo, A.; Purchase, D. Educational Activities for Students and Citizens Supporting the One Health Approach on Antimicrobial Resistance. Antibiotics 2021, 10, 1519. [Google Scholar] [CrossRef] [PubMed]
  360. Pearson, M.; Chandler, C. Knowing Antimicrobial Resistance in Practice: A Multi-Country Qualitative Study with Human and Animal Healthcare Professionals. Glob. Health Action 2019, 12 (Suppl. S1), 1599560. [Google Scholar] [CrossRef] [PubMed]
  361. McEwen, S.A.; Collignon, P.J. Antimicrobial Resistance: A One Health Perspective. In Antimicrobial Resistance in Bacteria from Livestock and Companion Animals; ASM Press: Washington, DC, USA, 2018; pp. 521–547. [Google Scholar]
  362. Pieri, A.; Aschbacher, R.; Fasani, G.; Mariella, J.; Brusetti, L.; Pagani, E.; Sartelli, M.; Pagani, L. Country Income Is Only One of the Tiles: The Global Journey of Antimicrobial Resistance among Humans, Animals, and Environment. Antibiotics 2020, 9, 473. [Google Scholar] [CrossRef] [PubMed]
  363. Gandra, S.; Alvarez-Uria, G.; Turner, P.; Joshi, J.; Limmathurotsakul, D.; van Doorn, H.R. Antimicrobial Resistance Surveillance in Low- and Middle-Income Countries: Progress and Challenges in Eight South Asian and Southeast Asian Countries. Clin. Microbiol. Rev. 2020, 33, 10–1128. [Google Scholar] [CrossRef] [PubMed]
  364. Nadimpalli, M.; Delarocque-Astagneau, E.; Love, D.C.; Price, L.B.; Huynh, B.T.; Collard, J.M.; Lay, K.S.; Borand, L.; Ndir, A.; Walsh, T.R.; et al. Combating Global Antibiotic Resistance: Emerging One Health Concerns in Lower- and Middle-Income Countries. Clin. Infect. Dis. 2018, 66, 963–969. [Google Scholar] [CrossRef]
  365. Wernli, D.; Harbarth, S.; Levrat, N.; Pittet, D. A ‘Whole of United Nations Approach’ to Tackle Antimicrobial Resistance? A Mapping of the Mandate and Activities of International Organisations. BMJ Glob. Health 2022, 7, e008181. [Google Scholar] [CrossRef]
  366. Pinto Ferreira, J.; Gochez, D.; Jeannin, M.; Magongo, M.W.; Loi, C.; Bucher, K.; Moulin, G.; Erlacher-Vindel, E. From OIE Standards to Responsible and Prudent Use of Antimicrobials: Supporting Stewardship for the Use of Antimicrobial Agents in Animals. JAC-Antimicrob. Resist. 2022, 4, dlac017. [Google Scholar] [CrossRef]
  367. Pinto Ferreira, J.; Battaglia, D.; Dorado García, A.; Tempelman, K.; Bullon, C.; Motriuc, N.; Caudell, M.; Cahill, S.; Song, J.; LeJeune, J. Achieving Antimicrobial Stewardship on the Global Scale: Challenges and Opportunities. Microorganisms 2022, 10, 1599. [Google Scholar] [CrossRef]
  368. Skandalis, N.; Maeusli, M.; Papafotis, D.; Miller, S.; Lee, B.; Theologidis, I.; Luna, B. Environmental Spread of Antibiotic Resistance. Antibiotics 2021, 10, 640. [Google Scholar] [CrossRef] [PubMed]
  369. Orubu, E.S.F.; Sutradhar, I.; Zaman, M.H.; Wirtz, V.J. Benchmarking National Action Plans on Antimicrobial Resistance in Eight Selected LMICs: Focus on the Veterinary Sector Strategies. J. Glob. Health 2020, 10, 020414. [Google Scholar] [CrossRef] [PubMed]
  370. Johnson, A.P.; Ashiru-Oredope, D.; Beech, E. Antibiotic Stewardship Initiatives as Part of the UK 5-Year Antimicrobial Resistance Strategy. Antibiotics 2015, 4, 467–479. [Google Scholar] [CrossRef] [PubMed]
  371. Manning, M.L.; Pfeiffer, J.; Larson, E.L. Combating Antibiotic Resistance: The Role of Nursing in Antibiotic Stewardship. Am. J. Infect. Control 2016, 44, 1454–1457. [Google Scholar] [CrossRef]
  372. Wang, S.; Hu, Y.J.; Little, P.; Wang, Y.; Chang, Q.; Zhou, X.; Moore, M.; Harwell, J.I. The Impact of the National Action Plan on the Epidemiology of Antibiotic Resistance among 352,238 Isolates in a Teaching Hospital in China from 2015 to 2018. Antimicrob. Resist. Infect. Control 2019, 8, 22. [Google Scholar] [CrossRef]
  373. Sangeda, R.Z.; Kibona, J.; Munishi, C.; Arabi, F.; Manyanga, V.P.; Mwambete, K.D.; Horumpende, P.G. Assessment of Implementation of Antimicrobial Resistance Surveillance and Antimicrobial Stewardship Programs in Tanzanian Health Facilities a Year after Launch of the National Action Plan. Front. Public Health 2020, 8, 454. [Google Scholar] [CrossRef]
  374. Kuupiel, D.; Bawontuo, V.; Mashamba-Thompson, T.P. Improving the Accessibility and Efficiency of Point-of-Care Diagnostics Services in Low- and Middle-Income Countries: Lean and Agile Supply Chain Management. Diagnostics 2017, 7, 58. [Google Scholar] [CrossRef]
  375. Roth, L.; Bempong, D.; Babigumira, J.B.; Banoo, S.; Cooke, E.; Jeffreys, D.; Kasonde, L.H.G.M.; Lin, J.C.W.; Lumpkin, M. Expanding Global Access to Essential Medicines: Investment Priorities for Sustainably Strengthening Medical Product Regulatory Systems. Glob. Health 2018, 14, 102. [Google Scholar] [CrossRef] [PubMed]
  376. Porter, G.; Kotwani, A.; Bhullar, L.; Joshi, J. Over-the-Counter Sales of Antibiotics for Human Use in India: The Challenges and Opportunities for Regulation. Med. Law Int. 2012, 21, 147–173. [Google Scholar] [CrossRef]
  377. Adhikari, B.; Pokharel, S.; Raut, S.; Adhikari, J.; Thapa, S.; Paudel, K.; Narayan, G.C.; Neupane, S.; Neupane, R.; Yadav, R. Why Do People Purchase Antibiotics Over-the-Counter? A Qualitative Study with Patients, Clinicians and Dispensers in Central, Eastern and Western Nepal. BMJ Glob. Health 2021, 6, e005829. [Google Scholar] [CrossRef]
  378. Sommanustweechai, A.; Chanvatik, S.; Sermsinsiri, V.; Sivilaikul, S.; Patcharanarumol, W.; Yeung, S.; Tangcharoensathien, V. Antibiotic Distribution Channels in Thailand: Results of Key-Informant Interviews, Reviews of Drug Regulations and Database Searches. Bull. World Health Organ. 2018, 96, 101. [Google Scholar] [CrossRef] [PubMed]
  379. Yevutsey, S.K.; Buabeng, K.O.; Aikins, M.; Anto, B.P.; Biritwum, R.B.; Frimodt-Møller, N.; Gyansa-Lutterodt, M. Situational Analysis of Antibiotic Use and Resistance in Ghana: Policy and Regulation. BMC Public Health 2017, 17, 896. [Google Scholar] [CrossRef]
  380. Kalungia, A.C.; Burger, J.; Godman, B.; Costa, J.D.O.; Simuwelu, C. Non-Prescription Sale and Dispensing of Antibiotics in Community Pharmacies in Zambia. Expert Rev. Anti-Infect. Ther. 2016, 14, 1215–1223. [Google Scholar] [CrossRef] [PubMed]
  381. Batista, A.D.; Rodrigues, D.; Figueiras, A.; Zapata-Cachafeiro, M.; Roque, F.; Herdeiro, M.T. Antibiotic Dispensation Without a Prescription Worldwide: A Systematic Review. Antibiotics 2020, 9, 786. [Google Scholar] [CrossRef] [PubMed]
  382. Simões, A.S.; Alves, D.A.; Gregório, J.; Couto, I.; Dias, S.; Póvoa, P.; Viveiros, M.; Gonçalves, L.; Lapão, L.V. Fighting Antibiotic Resistance in Portuguese Hospitals: Understanding Antibiotic Prescription Behaviours to Better Design Antibiotic Stewardship Programmes. J. Glob. Antimicrob. Resist. 2018, 13, 226–230. [Google Scholar] [CrossRef]
  383. Bauer, K.A.; Kullar, R.; Gilchrist, M.; File, T.M., Jr. Antibiotics and Adverse Events: The Role of Antimicrobial Stewardship Programs in ‘Doing No Harm’. Curr. Opin. Infect. Dis. 2019, 32, 553–558. [Google Scholar] [CrossRef]
  384. Wagenaar, J.A.; Scott, H.M. Reducing Antimicrobial Usage in Agriculture and Aquaculture: Beyond Regulatory Policy. Zoonoses Public Health 2015, 62, 1–2. [Google Scholar] [CrossRef] [PubMed]
  385. Low, C.X.; Tan, L.T.H.; Ab Mutalib, N.S.; Pusparajah, P.; Goh, B.H.; Chan, K.G.; Letchumanan, V.; Lee, L.H. Unveiling the Impact of Antibiotics and Alternative Methods for Animal Husbandry: A Review. Antibiotics 2021, 10, 578. [Google Scholar] [CrossRef]
  386. Dewulf, J.; Sternberg-Lewerin, S.; Ryan, M. Tackling Antimicrobial Resistance in the Food and Livestock Sector. In Challenges to Tackling Antimicrobial Resistance: Economic and Policy Responses; Anderson, M., Cecchini, M., Mossialos, E., Eds.; Cambridge University Press: Cambridge, UK, 2020; pp. 99–124, (European Observatory on Health Systems and Policies). [Google Scholar]
  387. Kuehn, B.M. FDA Moves to Curb Antibiotic Use in Livestock. JAMA 2014, 311, 347–348. [Google Scholar] [CrossRef]
  388. Patel, S.J.; Wellington, M.; Shah, R.M.; Ferreira, M.J. Antibiotic Stewardship in Food-Producing Animals: Challenges, Progress, and Opportunities. Clin. Ther. 2020, 42, 1649–1658. [Google Scholar] [CrossRef]
  389. Miller, R.A.; Salmon, P.; Sharkey, M. Approaches to Developing Judicious Uses of Veterinary Antibacterial Drugs. J. Vet. Pharmacol. Ther. 2021, 44, 201–206. [Google Scholar] [CrossRef] [PubMed]
  390. Singer, A.C.; Shaw, H.; Rhodes, V.; Hart, A. Review of Antimicrobial Resistance in the Environment and Its Relevance to Environmental Regulators. Front. Microbiol. 2016, 17, 1728. [Google Scholar] [CrossRef] [PubMed]
  391. Rayan, R.A. Pharmaceutical Effluent Evokes Superbugs in the Environment: A Call to Action. Biosaf. Health 2023, 50, 363–371. [Google Scholar] [CrossRef]
  392. Ågerstrand, M.; Josefsson, H.; Wernersson, A.S.; Larsson, D.J. Opportunities to Tackle Antibiotic Resistance Development in the Aquatic Environment through the Water Framework Directive. Ambio 2023, 52, 941–951. [Google Scholar] [CrossRef] [PubMed]
  393. Okoye, C.O.; Nyaruaba, R.; Ita, R.E.; Okon, S.U.; Addey, C.I.; Ebido, C.C.; Opabunmi, A.O.; Okeke, E.S.; Chukwudozie, K.I. Antibiotic Resistance in the Aquatic Environment: Analytical Techniques and Interactive Impact of Emerging Contaminants. Environ. Toxicol. Pharmacol. 2022, 96, 103995. [Google Scholar] [CrossRef] [PubMed]
  394. Hijazi, K.; Joshi, C.; Gould, I.M. Challenges and Opportunities for Antimicrobial Stewardship in Resource-Rich and Resource-Limited Countries. Expert Rev. Anti-Infect. Ther. 2019, 17, 621–634. [Google Scholar] [CrossRef]
  395. Khan, M.S.; Durrance-Bagale, A.; Mateus, A.; Sultana, Z.; Hasan, R.; Hanefeld, J. What Are the Barriers to Implementing National Antimicrobial Resistance Action Plans? A Novel Mixed-Methods Policy Analysis in Pakistan. Health Policy Plan. 2020, 35, 973–982. [Google Scholar] [CrossRef]
  396. Frumence, G.; Mboera, L.E.; Katale, B.Z.; Sindato, C.; Kimera, S.; Durrance-Bagale, A.; Mshana, S.E.; Clark, T.G.; Rweyemamu, M.M.; Legido-Quigley, H.; et al. Policy Actors and Human and Animal Health Practitioners’ Perceptions of Antimicrobial Use and Resistance in Tanzania: A Qualitative Study. J. Glob. Antimicrob. Resist. 2021, 25, 40–47. [Google Scholar] [CrossRef]
  397. Tang, K.; Millar, B.; Moore, J. Antimicrobial Resistance (AMR). Br. J. Biomed. Sci. 2023, 80, 11387. [Google Scholar] [CrossRef]
  398. Umair, M.; Mohsin, M.; Sönksen, U.W.; Walsh, T.R.; Kreienbrock, L.; Laxminarayan, R. Measuring Antimicrobial Use Needs Global Harmonization. Glob. Chall. 2021, 5, 2100017. [Google Scholar] [CrossRef]
  399. Alm, R.A.; Gallant, K. Innovation in Antimicrobial Resistance: The CARB-X Perspective. ACS Infect. Dis. 2020, 6, 1317–1322. [Google Scholar] [CrossRef] [PubMed]
  400. Altarac, D.; Gutch, M.; Mueller, J.; Ronsheim, M.; Tommasi, R.; Perros, M. Challenges and Opportunities in the Discovery, Development, and Commercialization of Pathogen-Targeted Antibiotics. Drug Discov. Today 2021, 26, 2084–2089. [Google Scholar] [CrossRef] [PubMed]
  401. Barrass, L.; Roberts, A.; Arisa, O.; Robertson, E.; Oliver, A.; Sharma, O.; Kinnersley, J.; Craig, D.; Oyewole, A. OP57: The Identification of Technological Innovations to Address the Challenge of Antimicrobial Resistance Using Horizon Scanning Approaches. Int. J. Technol. Assess. Health Care 2022, 38 (Suppl. S1), S22. [Google Scholar] [CrossRef]
  402. Sharma, S.; Chauhan, A.; Ranjan, A.; Mathkor, D.M.H.S.; Ramniwas, S.; Tuli, H.S.; Jindal, T.; Yadav, V. Emerging Challenges in Antimicrobial Resistance: Implications for Pathogenic Microorganisms, Novel Antibiotics, and Their Impact on Sustainability. Front. Microbiol. 2024, 15, 1403168. [Google Scholar] [CrossRef]
  403. Feng, Y. Transferability of MCR-1/2 Polymyxin Resistance: Complex Dissemination and Genetic Mechanism. ACS Infect. Dis. 2018, 4, 291–300. [Google Scholar] [CrossRef] [PubMed]
  404. Talebi, G.; Hashemi, A. Metallo-β-Lactamase, Extended Spectrum β-Lactamase, and mcr-1 Gene as Major Therapeutic Challenges. Rev. Res. Med. Microbiol. 2022, 33, e40–e47. [Google Scholar] [CrossRef]
  405. Son, S.J.; Huang, R.; Squire, C.J.; Leung, I.K. MCR-1: A Promising Target for Structure-Based Design of Inhibitors to Tackle Polymyxin Resistance. Drug Discov. Today 2019, 24, 206–216. [Google Scholar] [CrossRef] [PubMed]
  406. Murugaiyan, J.; Kumar, P.A.; Rao, G.S.; Iskandar, K.; Hawser, S.H.J.P.; Mohsen, Y.; Awual, W.A.; Alvarez, R.; Nanono, S. Progress in Alternative Strategies to Combat Antimicrobial Resistance: Focus on Antibiotics. Antibiotics 2022, 11, 200. [Google Scholar] [CrossRef]
  407. Karas, J.A.; Chen, F.; Schneider-Futschik, E.K.; Kang, Z.; Hussein, M.S.J.; Hoyer, D.; Giltrap, A.; Payne, R.J.; Li, J.; Velkov, T. Synthesis and Structure−Activity Relationships of Teixobactin. Ann. N. Y. Acad. Sci. 2020, 1459, 86–105. [Google Scholar] [CrossRef] [PubMed]
  408. Bhavnani, S.M.; Krause, K.M.; Ambrose, P.G. A Broken Antibiotic Market: Review of Strategies to Incentivize Drug Development. Open Forum Infect. Dis. 2020, 7, ofaa083. [Google Scholar] [CrossRef]
  409. Beyer, P.; Paulin, S. Priority Pathogens and the Antibiotic Pipeline: An Update. Bull. World Health Organ. 2020, 98, 151. [Google Scholar] [CrossRef] [PubMed]
  410. Breijyeh, Z.; Karaman, R. Design and Synthesis of Novel Antimicrobial Agents. Antibiotics 2023, 12, 628. [Google Scholar] [CrossRef] [PubMed]
  411. Hitchcock, N.M.; Gomes Nunes, D.D.; Shiach, J.; Saraiva Hodel, K.V.; Barbosa, J.D.V.; Rodrigues, L.A.P.; Colerm, S.; Soares, M.B.P.; Badaró, R. Current Clinical Landscape and Global Potential of Bacteriophage Therapy. Microorganisms 2023, 15, 1020. [Google Scholar] [CrossRef] [PubMed]
  412. Sieiro, C.; Areal-Hermida, L.; Pichardo-Gallardo, Á.; Almuiña-González, R.; De Miguel, T.; Sánchez, S.; Sánchez-Pérez, A.; Villa, T.G. A Hundred Years of Bacteriophages: Can Phages Replace Antibiotics in Agriculture and Aquaculture? Antibiotics 2020, 9, 493. [Google Scholar] [CrossRef]
  413. Alomari, M.M.M.; Dec, M.; Urban-Chmiel, R. Bacteriophages as an Alternative Method for Control of Zoonotic and Foodborne Pathogens. Viruses 2021, 13, 2348. [Google Scholar] [CrossRef] [PubMed]
  414. Adesanya, O.; Oduselu, T.; Akin-Ajani, O.; Adewumi, O.M.; Ademowo, O.G. An Exegesis of Bacteriophage Therapy: An Emerging Player in the Fight against Anti-Microbial Resistance. AIMS Microbiol. 2020, 6, 204. [Google Scholar] [CrossRef] [PubMed]
  415. Huan, Y.; Kong, Q.; Mou, H.; Yi, H. Antimicrobial Peptides: Classification, Design, Application and Research Progress in Multiple Fields. Front. Microbiol. 2020, 11, 582770. [Google Scholar] [CrossRef]
  416. Matthyssen, T.; Li, W.; Holden, J.A.; Lenzo, J.C.; Hadjigol, S.; O’Brien-Simpson, N.M. The Potential of Modified and Multimeric Antimicrobial Peptide Materials as Superbug Killers. Front. Chem. 2022, 9, 795433. [Google Scholar] [CrossRef]
  417. Bellotti, D.; Remelli, M. Lights and Shadows on the Therapeutic Use of Antimicrobial Peptides. Molecules 2022, 27, 4584. [Google Scholar] [CrossRef]
  418. Teixeira, M.C.; Carbone, C.; Sousa, M.C.; Espina, M.; Garcia, M.L.; Sanchez-Lopez, E.; Souto, E.B. Nanomedicines for the Delivery of Antimicrobial Peptides (AMPs). Nanomaterials 2020, 10, 560. [Google Scholar] [CrossRef]
  419. Bergman, P.; Raqib, R.; Rekha, R.S.; Agerberth, B.; Gudmundsson, G.H. Host Directed Therapy against Infection by Boosting Innate Immunity. Front. Immunol. 2020, 11, 1209. [Google Scholar] [CrossRef] [PubMed]
  420. Owen, A.M.; Fults, J.B.; Patil, N.K.; Hernandez, A.; Bohannon, J.K. TLR Agonists as Mediators of Trained Immunity: Mechanistic Insight and Immunotherapeutic Potential to Combat Infection. Front. Immunol. 2021, 11, 622614. [Google Scholar] [CrossRef] [PubMed]
  421. Latreille, E.; Lee, W.L. Modulation of the Host Response as a Therapeutic Strategy in Severe Lung Infections. Viruses 2023, 15, 1462. [Google Scholar] [CrossRef] [PubMed]
  422. Kelly, A.M.; McLoughlin, R.M. Target the Host, Kill the Bug; Targeting Host Respiratory Immunosuppressive Responses as a Novel Strategy to Improve Bacterial Clearance during Lung Infection. Front. Immunol. 2020, 11, 767. [Google Scholar] [CrossRef] [PubMed]
  423. Ji, J.; Jin, W.; Liu, S.J.; Jiao, Z.; Li, X. Probiotics, Prebiotics, and Postbiotics in Health and Disease. MedComm 2023, 4, e420. [Google Scholar] [CrossRef]
  424. Quigley, E.M.; Gajula, P. Recent Advances in Modulating the Microbiome. F1000Research 2020, 9, F1000 Faculty Rev-46. [Google Scholar] [CrossRef]
  425. Schwabkey, Z.I.; Jenq, R.R. Microbiome Anomalies in Allogeneic Hematopoietic Cell Transplantation. Annu. Rev. Med. 2020, 71, 137–148. [Google Scholar] [CrossRef]
  426. Wuethrich, I.; Pelzer, B.W.; Khodamoradi, Y.; Vehreschild, M.J. The Role of the Human Gut Microbiota in Colonization and Infection with Multidrug-Resistant Bacteria. Gut Microbes 2021, 13, 1911279. [Google Scholar] [CrossRef] [PubMed]
  427. Broom, A.; Kenny, K.; Prainsack, B.; Broom, J. Antimicrobial Resistance as a Problem of Values? Views from Three Continents. Crit. Public Health 2021, 31, 451–463. [Google Scholar] [CrossRef]
  428. Sariola, S.; Butcher, A. In Critique of Anthropocentrism: A More-than-Human Ethical Framework for Antimicrobial Resistance. Med. Humanit. 2022, 48, e16. [Google Scholar]
  429. Tarrant, C.; Colman, A.M.; Jenkins, D.R.; Chattoe-Brown, E.; Perera, N.; Mehtar, S.; Nakkawita, W.M.I.D.; Bolscher, M.; Krockow, E. Drivers of Broad-Spectrum Antibiotic Overuse across Diverse Hospital Contexts—A Qualitative Study of Prescribers in the UK, Sri Lanka, and South Africa. Antibiotics 2021, 19, 94. [Google Scholar] [CrossRef] [PubMed]
  430. Blaser, M.J.; Melby, M.K.; Lock, M.; Nichter, M. Accounting for Variation in and Overuse of Antibiotics among Humans. BioEssays 2021, 43, 2000163. [Google Scholar] [CrossRef] [PubMed]
  431. Kaiser, R.A.; Taing, L.; Bhatia, H. Antimicrobial Resistance and Environmental Health: A Water Stewardship Framework for Global and National Action. Antibiotics 2022, 11, 63. [Google Scholar] [CrossRef] [PubMed]
  432. Anyaegbunam, N.J.; Anekpo, C.C.; Anyaegbunam, Z.K.G.; Doowuese, Y.; Chinaka, C.B.; Odo, O.J.; Sharndama, H.C.; Mba, I.E. The Resurgence of Phage-Based Therapy in the Era of Increasing Antibiotic Resistance: From Research Progress to Challenges and Prospects. Microbiol. Res. 2022, 264, 127155. [Google Scholar] [CrossRef] [PubMed]
  433. Luong, T.; Salabarria, A.C.; Roach, D.R. Phage Therapy in the Resistance Era: Where Do We Stand and Where Are We Going? Clin. Ther. 2020, 42, 1659–1680. [Google Scholar] [CrossRef]
  434. Dunachie, S.J.; Day, N.P.; Dolecek, C. The Challenges of Estimating the Human Global Burden of Disease of Antimicrobial Resistant Bacteria. Curr. Opin. Microbiol. 2020, 57, 95–101. [Google Scholar] [CrossRef]
  435. Gajdács, M.; Urbán, E.; Stájer, A.; Baráth, Z. Antimicrobial Resistance in the Context of the Sustainable Development Goals: A Brief Review. Eur. J. Investig. Health Psychol. Educ. 2021, 11, 71–82. [Google Scholar] [CrossRef] [PubMed]
  436. Thornber, K.; Kirchhelle, C. Hardwiring Antimicrobial Resistance Mitigation into Global Policy. JAC-Antimicrob. Resist. 2022, 4, dlac083. [Google Scholar] [CrossRef]
  437. Asaaga, F.A.; Young, J.C.; Oommen, M.A.; Chandarana, R.; August, J.; Joshi, J.; Chanda, M.M.; Vanak, A.T.; Srinivas, P.N.; Hoti, S.L.; et al. Operationalising the “One Health” Approach in India: Facilitators of and Barriers to Effective Cross-Sector Convergence for Zoonoses Prevention and Control. BMC Public Health 2021, 21, 1517. [Google Scholar] [CrossRef]
  438. Achi, C.R.; Ayobami, O.; Mark, G.; Egwuenu, A.; Ogbolu, D.; Kabir, J. Operationalising One Health in Nigeria: Reflections from a High-Level Expert Panel Discussion Commemorating the 2020 World Antibiotics Awareness Week. Front. Public Health 2021, 9, 673504. [Google Scholar] [CrossRef]
  439. Mariappan, V.; Vellasamy, K.M.; Mohamad, N.A.; Subramaniam, S.; Vadivelu, J. OneHealth Approaches Contribute Towards Antimicrobial Resistance: Malaysian Perspective. Front. Microbiol. 2021, 12, 718774. [Google Scholar] [CrossRef] [PubMed]
  440. Parveen, S.; Garzon-Orjuela, N.; Amin, D.; McHugh, P.; Vellinga, A. Public Health Interventions to Improve Antimicrobial Resistance Awareness and Behavioural Change Associated with Antimicrobial Use: A Systematic Review Exploring the Use of Social Media. Antibiotics 2022, 11, 669. [Google Scholar] [CrossRef] [PubMed]
  441. Fletcher-Miles, H.; Gammon, J.; Williams, S.; Hunt, J. A Scoping Review to Assess the Impact of Public Education Campaigns to Affect Behavior Change Pertaining to Antimicrobial Resistance. Am. J. Infect. Control 2020, 48, 433–442. [Google Scholar] [CrossRef] [PubMed]
  442. Cuevas, C.; Batura, N.; Wulandari, L.P.L.; Khan, M.; Wiseman, V. Improving Antibiotic Use through Behaviour Change: A Systematic Review of Interventions Evaluated in Low-and Middle-Income Countries. Health Policy Plan. 2021, 36, 754–773. [Google Scholar] [CrossRef] [PubMed]
Figure 1. Schematic representation of antimicrobial use in agriculture and animal and human health and the flux of these drugs among all counterparts.
Figure 1. Schematic representation of antimicrobial use in agriculture and animal and human health and the flux of these drugs among all counterparts.
Microorganisms 12 01920 g001
Figure 2. Schematic representation of the flowchart of the searched articles and the inclusion and exclusion criteria.
Figure 2. Schematic representation of the flowchart of the searched articles and the inclusion and exclusion criteria.
Microorganisms 12 01920 g002
Figure 3. Global distribution of the main multi-resistant strains and antimicrobial resistance (CD: Clostridium difficile; CRE: carbapenem-resistant Enterobacterales; ESBL: extended-spectrum beta-lactamase; MRSA: methicillin-resistant Staphylococcus aureus; VRE: vancomycin-resistant Enterococcus; TB: multidrug-resistant Mycobacterium tuberculosis).
Figure 3. Global distribution of the main multi-resistant strains and antimicrobial resistance (CD: Clostridium difficile; CRE: carbapenem-resistant Enterobacterales; ESBL: extended-spectrum beta-lactamase; MRSA: methicillin-resistant Staphylococcus aureus; VRE: vancomycin-resistant Enterococcus; TB: multidrug-resistant Mycobacterium tuberculosis).
Microorganisms 12 01920 g003
Figure 4. Effective IPC strategies to prevent MDRO emergence and dissemination.
Figure 4. Effective IPC strategies to prevent MDRO emergence and dissemination.
Microorganisms 12 01920 g004
Table 1. Overview of the main antimicrobial resistance mechanisms found in bacteria, viruses, fungi, and parasites.
Table 1. Overview of the main antimicrobial resistance mechanisms found in bacteria, viruses, fungi, and parasites.
OrganismMechanismDetailsExampleRef.
BacteriaTarget
Modification
Antibiotic’s molecular target alteration inhibits drug bindingrRNA mutations hinder aminoglycoside binding[34,35,36]
Efflux PumpsMembrane proteins expel antibiotics and reduce their intracellular concentrationsEfflux of tetracyclines, fluoroquinolones, and macrolides by specific bacterial pumps[37,38,39]
Enzymatic
Degradation
Enzymes chemically modify or degrade antibiotics and neutralize their effectsβ-lactamases hydrolyze the β-lactam ring (penicillin, cephalosporin)[40,41,42,43]
Membrane
Permeability
Outer membrane permeability reduction limits antibiotic entryGram-negative bacteria alter outer membrane proteins to resist drug influx[44,45,46]
Biofilm
Formation
Biofilms limit antibiotic penetration and enhance resistanceBiofilm-associated resistance in P. aeruginosa infections[47,48,49,50]
VirusesMutationMutations in viral genes encoding enzymes reduce the binding affinity of antiviral drugsHIV resistance due to mutations in reverse transcriptase and protease[51,52]
Changes in Viral Entry ProteinsSurface protein mutations prevent antiviral drugs from achieving host cell entryResistance to entry inhibitors in HIV treatment[53]
Altered Drug
Activation
Mutations impair the activation of antiviral drugs within the virus or host cellsAcyclovir resistance in herpes simplex virus due to TK gene mutations[54]
FungiTarget
Modification
Antifungal drugs’ molecular target alteration reduces drug efficacyAzole resistance in Candida spp. due to ERG11 gene mutations[55]
Efflux PumpsMembrane proteins expel antifungal drugs and reduce their intracellular concentrationsAzole resistance in C. albicans due to ABC transporter overexpression[56,57,58,59,60,61]
Cell Wall
Alteration
Cell wall composition modifications decrease drug binding or penetrationEchinocandin resistance in Candida spp. due to FKS gene mutations [62,63]
Biofilm FormationBiofilms protect cells from antifungal drugs and difficult infection treatmentBiofilm-related resistance in Candida infections associated with medical devices[64,65,66]
ParasitesTarget
Modification
Parasites acquire mutations in genes encoding drug targets, leading to reduced drug efficacyAntifolate resistance in P. falciparum due to DHFR gene mutations [67,68,69,70,71]
Efflux pumpsMembrane proteins actively remove drugs from cells and reduce their intracellular concentrationsChloroquine resistance in P. falciparum due to transporter protein expression[70]
Metabolic Pathway AlterationMetabolic pathway alterations to bypass drug effectsGlycolytic pathway alterations confer resistance to drugs in trypanosomes[72]
Reduced Drug
Activation
Mutations reduce the activation of drugs within parasites, leading to resistanceMetronidazole resistance in G. lamblia due to NTR mutations[73,74,75]
Table 2. Novel approaches to combat antimicrobial resistance.
Table 2. Novel approaches to combat antimicrobial resistance.
ApproachDescriptionAdvantagesChallengesExamplesRef.
New Antibiotics with Novel
Mechanisms
Development of antibiotics targeting non-traditional bacterial processes.Target novel aspects of bacterial physiology, useful for resistant bacteria.Difficulty in the discovery and development of novel classes.Teixobactin—targets bacterial cell wall synthesis in a new way, with no observed resistance yet.[324]
Bacteriophage
Therapy
Utilizes viruses that specifically infect and kill bacteria.High specificity, ecological safety, and potential beyond clinical applications.Large-scale production, stability, and bacterial resistance to phages.Phage therapy for P. aeruginosa infections in cystic fibrosis patients.[325,326]
Phage-Based
Small Molecule
Development
Exploits phage–bacteria interactions to identify new antibacterial targets and inhibitory proteins.Novel targets for drug development, potentially more specific and effective treatments.Development and scaling challenges.Phage-derived enzymes (endolysins) are used to treat S. aureus infections.[327]
Antimicrobial
Peptides
(AMPs)
Naturally occurring peptides with broad-spectrum antibacterial, antiviral, and antifungal properties.Broad-spectrum activity, lower risk of resistance development, several FDA-approved drugs.Requires more research on mechanisms of action and clinical optimization.Daptomycin is used to treat infections by resistant Gram-positive bacteria (e.g., MRSA).[328,329,330]
ImmunotherapiesMonoclonal antibodies target and neutralize bacterial toxins and virulence factors.High specificity, lower risk of resistance development.High production costs and the need for targeted pathogen identification.Bezlotoxumab used to neutralize C. difficile toxins.[331,332]
Probiotics and
Fecal Transplant Therapy
Enhances human microbiota to prevent infections.Restores healthy microbiomes, potentially reducing antibiotic use.Efficacy varies and needs more standardized research.FMT used to treat recurrent C. difficile infections.[333]
Pathogen-Oriented Therapy (POT)Targeted approach using antibiotic conjugates, nanotechnologies, and CRISPR-Cas systems.Highly specific to pathogens, avoids non-targeted antimicrobial use.Complex development, regulatory challenges, and scaling issues.CRISPR-Cas antimicrobials targeting resistant bacteria, antibiotic–antibody conjugates.[334,335]
Combination
Therapies
Use of multiple drugs to prevent or overcome resistance.Enhances efficacy and prevents resistance development.Risk of drug interactions and complex treatment regimens.Ceftazidime-avibactam is used to treat multidrug-resistant Gram-negative bacteria.[336]
Repurposing
Existing Drugs
Using known drugs for new antibacterial purposes.Cost-effective, faster clinical adoption due to existing safety profiles.Limited applicability, potential to develop resistance to repurposed drugs.Colistin is used to treat MDR Acinetobacter and Pseudomonas species.[336]
NanotechnologyUse of nanoparticles to enhance antibiotic efficacy or act as antimicrobial agents.Enhances antibiotic activity and overcomes biofilm-related resistance.Safety, bioavailability, and large-scale production.Silver nanoparticles used in combination with antibiotics to enhance their efficacy.[337,338]
Artificial
Intelligence
AI-driven approaches for drug discovery, resistance prediction, and treatment optimization.Accelerates drug discovery, predicts resistance, optimizes antimicrobial regimens.Data quality, model accuracy, and integration into clinical workflows.IBM’s AI-driven drug discovery used to identify new antibiotics like halicin.[339,340]
One Health
Approach
Multisectoral approach linking human, animal, and environmental health to combat AMR.Promotes cross-sector collaboration and addresses complex AMR drivers globally.Requires significant international cooperation and systemic change across multiple industries.WHO Global Action Plan on AMR, One Health AMR Surveillance Programs (WHO, FAO, OIE).[341,342,343,344]
Disclaimer/Publisher’s Note: The statements, opinions and data contained in all publications are solely those of the individual author(s) and contributor(s) and not of MDPI and/or the editor(s). MDPI and/or the editor(s) disclaim responsibility for any injury to people or property resulting from any ideas, methods, instructions or products referred to in the content.

Share and Cite

MDPI and ACS Style

Oliveira, M.; Antunes, W.; Mota, S.; Madureira-Carvalho, Á.; Dinis-Oliveira, R.J.; Dias da Silva, D. An Overview of the Recent Advances in Antimicrobial Resistance. Microorganisms 2024, 12, 1920. https://doi.org/10.3390/microorganisms12091920

AMA Style

Oliveira M, Antunes W, Mota S, Madureira-Carvalho Á, Dinis-Oliveira RJ, Dias da Silva D. An Overview of the Recent Advances in Antimicrobial Resistance. Microorganisms. 2024; 12(9):1920. https://doi.org/10.3390/microorganisms12091920

Chicago/Turabian Style

Oliveira, Manuela, Wilson Antunes, Salete Mota, Áurea Madureira-Carvalho, Ricardo Jorge Dinis-Oliveira, and Diana Dias da Silva. 2024. "An Overview of the Recent Advances in Antimicrobial Resistance" Microorganisms 12, no. 9: 1920. https://doi.org/10.3390/microorganisms12091920

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

Article Metrics

Back to TopTop