Next Article in Journal
Co-Expression of Podoplanin and CD44 in Proliferative Vitreoretinopathy Epiretinal Membranes
Next Article in Special Issue
Research Progress of Benzothiazole and Benzoxazole Derivatives in the Discovery of Agricultural Chemicals
Previous Article in Journal
Bacterial Therapy of Cancer: A Way to the Dustbin of History or to the Medicine of the Future?
Previous Article in Special Issue
Detection of Antibiotic Resistance in Feline-Origin ESBL Escherichia coli from Different Areas of China and the Resistance Elimination of Garlic Oil to Cefquinome on ESBL E. coli
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

Current State of Knowledge Regarding WHO High Priority Pathogens—Resistance Mechanisms and Proposed Solutions through Candidates Such as Essential Oils: A Systematic Review

1
Doctoral School, “Victor Babeş” University of Medicine and Pharmacy, 2 Eftimie Murgu Street, 300041 Timisoara, Romania
2
Faculty of Medicine, “Victor Babeş” University of Medicine and Pharmacy, 2 Eftimie Murgu Street, 300041 Timisoara, Romania
3
Faculty of Pharmacy, “Victor Babeş” University of Medicine and Pharmacy, 2 Eftimie Murgu Street, 300041 Timisoara, Romania
4
OncoGen Centre, County Hospital ‘Pius Branzeu’, Blvd. Liviu Rebreanu 156, 300723 Timisoara, Romania
5
Multidisciplinary Research Center on Antimicrobial Resistance, “Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Square, 300041 Timisoara, Romania
6
Research Center for Pharmaco-Toxicological Evaluation, “Victor Babes” University of Medicine and Pharmacy, 2 Eftimie Murgu Square, 300041 Timisoara, Romania
7
Louvain Drug Research Institute, Université Catholique de Louvain, Avenue Emmanuel Mounier 73, 1200 Brussels, Belgium
8
Faculty of Agriculture, University of Life Sciences “King Mihai I” from Timisoara, Calea Aradului 119, 300645 Timisoara, Romania
9
Ineu City Hospital, 2 Republicii Street, 315300 Ineu, Romania
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2023, 24(11), 9727; https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24119727
Submission received: 5 April 2023 / Revised: 31 May 2023 / Accepted: 2 June 2023 / Published: 4 June 2023
(This article belongs to the Special Issue Antibacterial Activity of Drug-Resistant Strains)

Abstract

:
Combating antimicrobial resistance (AMR) is among the 10 global health issues identified by the World Health Organization (WHO) in 2021. While AMR is a naturally occurring process, the inappropriate use of antibiotics in different settings and legislative gaps has led to its rapid progression. As a result, AMR has grown into a serious global menace that impacts not only humans but also animals and, ultimately, the entire environment. Thus, effective prophylactic measures, as well as more potent and non-toxic antimicrobial agents, are pressingly needed. The antimicrobial activity of essential oils (EOs) is supported by consistent research in the field. Although EOs have been used for centuries, they are newcomers when it comes to managing infections in clinical settings; it is mainly because methodological settings are largely non-overlapping and there are insufficient data regarding EOs’ in vivo activity and toxicity. This review considers the concept of AMR and its main determinants, the modality by which the issue has been globally addressed and the potential of EOs as alternative or auxiliary therapy. The focus is shifted towards the pathogenesis, mechanism of resistance and activity of several EOs against the six high priority pathogens listed by WHO in 2017, for which new therapeutic solutions are pressingly required.

1. Introduction

1.1. Antibiotics and Antimicrobial Resistance

Antibiotics (ABs) are common agents used in healthcare to treat and prevent potentially fatal bacterial infections [1]. Their introduction into clinical practice was arguably the greatest discovery of modern medicine in the twentieth century [2]. Yet, ABs have been around for millennia, as many of them are compounds synthetized by microorganisms to protect themselves and dominate different habitats [3]. While humans started exploiting the power of ABs as early as 1550 BC, it was not until circa 100 years ago that we managed to understand, synthesize and purify ABs, salvarsan and penicillin being the promoters of the AB era [1]. Mishandling ABs has resulted in the rapid expansion of antimicrobial resistance (AMR). Even though AMR is a naturally occurring process, it was first identified 50 years ago, when Staphylococcus aureus began to develop penicillin resistance [4]. AMR is commonly associated with the presence of AB-resistant genes (ARGs) in the bacterial genome [5]. Given that bacteria are able to pass on ARGs through vertical or horizontal gene transfer [6], there is clear evidence that prolonged exposure to ABs can easily turn non-resistant bacteria into resistant ones [7].

1.2. Main Determinants of AMR

Given that AMR is linked to the emergence of multidrug resistant (MDR) and extensive drug resistant (XDR) pathogens [8], it represents a serious global threat of growing concern that affects humans, animals and the environment [9]. According to the World Health Organization (WHO), AMR is annually responsible for the deaths of at least 700,000 people worldwide and the death toll could reach 10 million by 2050 [10]. There are several reasons why AMR occurs: (1) inappropriate prescribing of ABs; (2) dispensing ABs without a prescription; (3) poor AB regulations and lack of surveillance of resistance development; (4) excessive use of ABs in food-producing animals; (5) limited decontamination of wastewater; and (6) lack of research on new ABs [8,11].
Initial, therapy errors such as misdiagnosing the infection aetiology, choosing an incorrect dosage, overly extending the duration of treatment, overlooking the recommended guidelines regarding first line ABs or prescribing ABs without a clear clinical indication all raise the risk of developing AMR [11,12]. These mistakes might be due to gaps in knowledge; fear of complications from infections; concerns of not meeting perceived patient expectation; financial benefits or incentives, and misleading advertising from the industry [13]. Poor regulations in AB use contribute to medication abuse and the occurrence of AMR. Dispensing ABs without a medical prescription is associated with inappropriate drug choice, wrong dosage, shorter treatment course and increased risk of adverse drug reactions [14]. Unregulated overuse of ABs in hospitals has increased the rates of resistance in nosocomial infections and thus cross-transmission [8]. Confronting MDR bacteria has led many hospitals to introduce antimicrobial stewardship programmes to monitor antimicrobial use and identify ways to reduce development and transmission of AMR [15]. However, just reducing the consumption of ABs and not working on socio-economic factors will not have the desired impact on the prevalence of AMR [16].
Antibiotic use in the animal industry has been found to be a key element in the development of AMR. Given that ABs might stimulate the intestinal synthesis of vitamins and lower the competition for nutrients between host and bacteria, sub-lethal doses of ABs are commonly used as growth promoters for food-producing animals [17]. In developed countries, 60–80% of ABs are also given to animals, especially poultry, pigs and cattle [18], with penicillins, tetracyclines and sulphonamides having the highest AMR rates [19]. In addition, the use of ABs as preservatives in meat [20] or heavy metals (such as copper and zinc) as growth promoters led to a massive increase in AMR [17]. Moreover, ABs are able to reach the environment via human and animal excretions, improper disposal of unused drugs and waste streams from the production line [21]. If micropollutants—including ABs and microorganisms—are not successfully removed, they represent an important source of soil and water contamination with ARGs [22].
Researchers are unable to keep the pace of finding new ABs in the face of emerging MDR strains. Starting with the discovery of penicillin, many ABs discoveries were serendipitous, made by empirical screening [23]. Nevertheless, despite technological progress, the last 25 years have not seen a breakthrough in the development of novel antibacterial drug classes [24] and there is a critical need for drugs targeting Gram-negative ESKAPE pathogens [25].

1.3. How AMR Is Addressed Globally and at European Level

In 2015, WHO developed an action plan to combat AMR which includes, among other objectives, the reduction of AB use in humans and animals [26], followed in 2017 by the publication of a list of bacteria that urgently need the development of new ABs, dividing them into three classes based on their healthcare burden. The following bacteria were classified as having critical priority (priority 1): Acinetobacter baumannii, Pseudomonas aeruginosa and Enterobacteriaceae. High priority pathogens (HPPs) were categorized as priority 2: Enterococcus faecium, Staphylococcus aureus, Helicobacter pylori, Campylobacter spp., Salmonellae spp. and Neisseria gonorrhoeae [27]. In May 2022, WHO issued a review of ABs in development worldwide. Since their first analysis in 2017, 12 new ABs have been approved; however, most were derivatives of existing classes, where resistance mechanisms have been established. At the moment, 27 ABs targeting WHO priority pathogens are in clinical development with 13 having confirmed activity against at least one of the critical Gram-negative bacteria [28]. The incoming United Nations General Assembly in 2024 aims to identify clear and practical steps to address AMR [29].
The global AMR situation is briefly presented by One Health Trust’s Resistance Map [30]. However, this database only covers the resistance of a few HPPs and some ABs (Table 1), whereas the reality might be more severe. On the other hand, the AMR picture in Europe is presented by the Surveillance Atlas of Infectious Diseases [31,32]. Here, although the situation varies broadly from country to country, the highest prevalence seems to be in the south-eastern region (Appendix A, Table A1). Given that both the European and the global databases lack reports from several countries, the real magnitude of AMR is difficult to grasp.

1.4. Essential Oils as Potential Tools against AMR

Essential oils (EOs), also called volatile oils, are complex mixtures of tens of lipophilic, volatile compounds, at different concentrations [33]. Generally, two or three major components are present in rather high concentrations (20–70%), while other compounds are present in trace amounts [34]. The main constituents of EOs are terpenes—compounds derived from isoprene units that often have several chemical functionalities, such as alcohol, phenol, aldehyde, ketone, ether and hydrocarbon groups [35,36]. EOs represent secondary metabolites produced by aromatic plants as a protective mechanism against predators, microorganisms or austere weather conditions [37]. Many parts of the plant are able to produce EOs, which can be then extracted using methods such as solvent extraction (solvent, subcritical water, supercritical CO2), distillation (hydrodistillation, steam distillation, hydrodiffusion), solvent-free microwave extraction and combined methods [38]. The applications of EOs range from aromatherapy and perfume production to food industry and animal nutrition [39].
In medical practice, EOs have been reported to have antimicrobial, antioxidant, anti-inflammatory, analgesic, antiemetic and cancer chemo-protective activities [40,41]. Some EOs also exhibit cytotoxic (against bacteria, viruses, fungi, protozoa, parasites and mites), allelopathic and insect repellent and insecticidal activities, thus they could be exploited as alternative strategies in a variety of industries [40]. Certain EOs are effective against pathogens of public health interest. In a previous review paper, we have presented that several EOs exhibit in vitro antimicrobial activity against the WHO priority 1 list of pathogens [42]. Notably, it is generally considered that EOs are more active against Gram-positive than Gram-negative bacteria [40]. Interestingly, unlike classic ABs, EOs exhibit good activity against pathogenic bacteria, while showing a lower effect on beneficial bacteria in the gut, such as Lactobacillus or Bifidobacterium [43]. Thanks to developments in pharmaceutical formulations, EOs can be loaded in carriers such as nanoparticles which significantly improve their bioavailability and stability [44]. However, the general population can also benefit from the effects of antimicrobial EOs through the use of spices [45].
The chemical functionalities responsible for antimicrobial activity are generally phenols and aldehydes, while a high proportion of esters, ketones and terpene hydrocarbons result in weak or no effect [46]. Thanks to their hydrophobicity, EOs can inhibit bacterial growth by degrading membrane proteins and increasing cell permeability [46,47]. They can interfere with the expression of genes encoding efflux pumps (tetA, tetK, pmrA, norA, blaTEM, blaOXA-23) in a variety of microorganisms [48]. Proton pumps can also be affected by EOs, resulting in reduced membrane potential and ATP depletion [49]. Moreover, EOs can hinder biofilm formation and disrupt quorum sensing. Thus, they alter cell-to-cell communication and interfere with gene expression regulation—crucial adaptive measurement in hostile environments [50].
The two most common in vitro techniques used to assess the antimicrobial activity of EOs are the agar diffusion method (paper disc or well) and the dilution method (agar or liquid broth) [51]. The agar diffusion methods are one of the most convenient techniques, in terms of price and methodology. In the agar well diffusion method, an agar plate is inoculated with a pathogenic microorganism through the spreading plate approach (an exact volume of the microbial solution is spread over the surface of agar, through a glass diffuser). A well or hole is aseptically made with a sterile cork borer of diameter 6–8mm, flooded with the tested solution (e.g., extract) and incubated at optimal temperature and conditions. The tested solution will diffuse progressively through the agar medium, thus inhibiting the microorganism growth. Later, the diameter of the inhibition zone will be measured. In the agar disc diffusion method, a filter paper disc containing the test solution is placed on the agar medium and then inoculated with the tested strains [51]. Overall, the agar diffusion techniques make it possible to easily test several extracts/substances against various microorganisms, although they are unable to highlight the minimum inhibitory concentration (MIC) or the ability of a substance/extract to inhibit or kill a microorganism [51].
In the dilution method a series of successive dilutions of concentrated solutions of microbial strains are prepared to accurately count the viable cells within a culture (bacterial, fungus or viruses). Each diluted sample is added to a liquefied agar medium, poured into a petri dish and solidifies, holding the microorganisms within its matrix. It is possible to count the microorganisms with precision as they disperse across the agar plate. This method is used to determine the MIC of a substance, as well as its ability to kill or inhibit the development of the tested strains. Moreover, it is the reference for antimicrobial susceptibility testing [51].
Bacterial resistance is generally defined using the minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) of the selected AB. However, the reported values for MIC and MBC are extremely divergent, possibly as a result of the wide diversity of bacterial strains, methodological dissimilarities and study design variations [52]. Notably, a number of variables related to the variability of the extract (the part of plant used, extraction method, etc.) also have an impact on EOs’ antimicrobial activity [53,54].
The aim of this review is to investigate the current level of knowledge regarding the six WHO HPPs: vancomycin-resistant E. faecium (VREF); clarithromycin-resistant H. pylori (CRHP); fluoroquinolone-resistant Campylobacter spp. (FRC); cephalosporin-resistant, fluoroquinolone-resistant N. gonorrhoeae (CRNG, FRNG); fluoroquinolone-resistant Salmonellae spp. (FRS); and methicillin-resistant, vancomycin-intermediate and -resistant S. aureus (MRSA, VISA, VRSA). To our knowledge, there have not been any recent publications that systematically present the antimicrobial activity of EOs against WHO priority 2 pathogens. Thus, our two main objectives are to present (1) HPPs’ key resistance mechanisms and (2) the latest EOs that exert antimicrobial activity on the aforementioned pathogens, with an emphasis on the AB-resistant strains.

2. Methods

2.1. Review’s Objective and Design

The aim of this review was to investigate the current level of knowledge regarding the six WHO HPPs (priority 2 list). This study was designed as a follow-up of a previous paper that addressed WHO critical priority pathogens (priority 1 list) [42]. Each of the six bacteria were discussed in terms of: pathogenesis, mechanism of AMR (with an emphasis on ABs indicated by WHO as increasingly inefficacious) and EOs as potential treatment candidates (with an emphasis on EOs that are effective against resistant strains). A working team was set up with members having experience in the area of interest: microbiology, clinical laboratory, clinical pharmacy, botany and pharmacognosy.

2.2. Literature Search Strategy

A flow diagram of screened records is provided in Figure 1. We conducted an independent search for each bacterium on three electronic databases (PubMED, Web of Science, Elsevier’s Scopus) and one web search engine (Google Scholar) from April 2022 to September 2022. The primary keyword combination used to perform the search was the following: WHO critical pathogen strain AND AB resistance AND essential oils (e.g., Enterococcus faecium AND vancomycin-resistant AND essential oils). Searches were conducted separately in each database and, after removing the duplicates, the records were exported to the citation software. Articles included in the study were those that: (1) focused on WHO priority 2 strains (primarily aiming for MDR strains); (2) investigated the activity of EOs against pathogens; (3) were published in the last 5 years; and (4) were written in English. The articles that failed to meet the inclusion criteria were excluded from the study, as well as the ones that fell under one of the following categories: book chapter, abstract, short communication, technical note, letter. Publications that appeared to be methodologically flawed and provided insufficient details or confusing outcomes were also dismissed.

2.3. Data Extraction

Pertinent articles were closely evaluated by reviewers and the following data were extracted: (1) study team and year of publication; (2) bacterial strain under investigation (including its resistance, if mentioned); (3) EOs/pure phytocompound proposed as having antimicrobial activity; method(s) of assessing the antimicrobial activity; and (4) main results. Any disagreements or queries were settled by a researcher with expertise in microbiology (D.M.).

3. Results and Discussion

3.1. Enterococcus Faecium, Vancomycin-Resistant

Enterococcus spp. are Gram-positive bacteria that are part of human faecal microbiota, as commensals of the intestinal tract [52]. However, some strains, particularly Enterococcus faecalis and Enterococcus faecium, may cause nosocomial outbreaks and opportunistic infections in hospitalized patients [53]. While most enterococci infections are caused by E. faecalis (nearly 90%), E. faecium has a higher rate of AB resistance, especially in case of bloodstream infections [54]. Plasmid transfer and homologous recombination, mediated by insertion sequence elements, are two important mechanisms through which E. faecium manages to escape effective therapy [55]. A series of genes (ace, acm, scm and ecb) code for a subset of adhesion factors that mediate the pathogen’s initial attachment. Aggregation substances, such as Enterococcal surface protein, favour E. faecium’s ability to form biofilms. Moreover, exoenzymes, including gelatinase, hyaluronidase and cytolysin, are secreted externally and can damage host cells by triggering inflammation [56].
Until recently, vancomycin was considered the gold standard in the therapy of beta-lactam resistant Gram-positive cocci. However, AMR is rising: in the United States, 30% of enterococci are vancomycin-resistant [57], while in some European countries the proportion of VREF is even higher (Table 1). VREF carried by patients is commonly shared within the hospital environment, as it may persist despite standard cleaning; thus, patients can acquire VREF if they are admitted to a room previously occupied by a VREF-positive patient [58]. VREF can be detected by identifying at least one of nine resistance genes—vanA, B, C, D, E, G, L, M and N—present in the mobile genetic elements (vanA, vanB) or located on the chromosome (vanC) [59]. The most predominant genotypes are vanA and vanB. vanA E. faecium is highly resistant to vancomycin and teicoplanin, while vanB E. faecium shows high resistance to vancomycin and susceptibility to teicoplanin [60]. Although linezolid has been considered a good alternative in treating VREF infections, resistance to this drug is gradually increasing, with G2576 T mutation within the 23S rDNA being one of the major resistance mechanisms [57]. Teicoplanin or last-line ABs such as daptomycin, tigecycline or quinupristin/dalfopristin remain an option for some VREF infections, but resistance has been reported [61,62].
There are several EOs that have shown activity against VREF (Appendix B, Table A2). Saki et al. investigated the antibacterial effects of cinnamon bark (Cinnamomum zeylanicum) EO on XDR isolates and determined that VREF was sensitive to this EO [63]. Five other EOs, namely bitter orange (Citrus aurantium v. amara), lemon (Citrus × limon), blue gum eucalyptus (Eucalyptus globulus), tea tree (Melaleuca alternifolia) and Mediterranean cypress (Cupressus sempervirens) were evaluated by Iseppi and collaborators. They observed that M. alternifolia EO was the most effective EO and C. aurantium the least effective EO, while the other three EOs displayed antibacterial activity against all strains, to a certain extent. What is more, EO–EO and EO–AB associations showed a synergistic antimicrobial activity in most tests and were even effective against biofilm formation [64]. Synergistic interactions were also investigated by Owen et al. in their study on oregano (Origanum compactum), rosewood (Aniba roseadora) and cumin (Cuminum cymimum) EOs. They found out that the combination of carvacrol and cuminaldehyde could re-establish susceptibility to vancomycin in VREF, resulting in bactericidal activity [65]. Their previous studies also indicated that these particular EOs exhibited zones of inhibition against VSEf and VREF in the Kirby–Bauer disc diffusion method, with minimal inhibitory concentrations (MICs) ranging from 0.29 to 37.20 mg/mL [66].
When looking into the effect of five EOs on VREF isolated from wastewater treatment plants, clinical samples and reference strains, Sakkas et al. discovered that origanum (Thymus capitatus), thyme (Thymus vulgaris) and tea tree (Melaleuca alternifolia) EOs were effective against the pathogenic strains [67]. Di Vito et al. compared the antimicrobial activity of EOs and hydrolates extracted from lavender (Lavandula angustifolia and Lavandula intermedia), origanum (Origanum hirtum), winter savory (Satureja montana), scarlet beebalm/bergamot (Monarda didyma) and beebalm/wild bergamot (Monarda fistulosa). They determined that the antimicrobial activity of hydrolates is milder than that of the corresponding EOs, with higher MICs. In contrast to hydrolates, which must be used at concentrations of 25–50% v/v to achieve the same antimicrobial activity, EOs are active at lower concentrations: 50% of the strains are susceptible at concentrations of 0.125–2% v/v [68].
Further data from these studies are presented in Appendix B, Table A2.

3.2. Helicobacter Pylori, Clarithromycin-Resistant

Helicobacter pylori are Gram-negative bacteria that colonize the stomach and duodenum, and might be a key contributor in diseases such as chronic gastritis, peptic ulcer, gastric adenocarcinoma, mucosa-associated lymphoid tissue lymphoma and iron deficiency anaemia [69]. While the most common sources of H. pylori contamination are water, environment and animals [70], it appears that a plant-based diet is associated with a lower prevalence in adults; in contrast, higher consumption of fried food and well water are considered risk factors for infection [71]. Overall, prevalence of H. pylori infection seems to be declining globally, with Oceania having the lowest prevalence rates (24.4%); however, in some parts of the world, the prevalence remains quite stable and reaches a rate of 79.1% in Africa [72]. H. pylori can cause lifelong infection without eradication, with recurrence appearing either by recrudescence or reinfection [73]. It has been established that H. pylori gastritis should be considered an infectious disease regardless of the absence of symptoms, complications or resultant illnesses [74]. Several specific virulence factors can lead to a more severe outcome: cytotoxin-associated gene A (CagA, especially EPIYA-D and EPIYA-C) and type IV secretion system (CagL polymorphism), the genotypes of vacuolating cytotoxin A (vacA, s1/i1/m1 types), and blood group antigen binding adhesin (BabA, low-producer or chimeric with BabB) [75].
The classical triple-therapy regimen against H. pylori infection includes a proton-pump inhibitor (PPI), amoxicillin and clarithromycin, taken together for 7–14 days [76]. The addition of nitroimidazole (metronidazole or tinidazole) adds benefits to the regimen, requiring fewer doses of ABs and being more effective [77]. Due to its ability to inhibit bacterial protein synthesis, clarithromycin has been the mainstay of therapy in H. pylori infections; the AB has a good mucosal diffusion, low MIC and a minimal impact on gastric acidity [78]. However, vertically transmitted point mutations in the peptidyl transferase loop of the V domain of 23S rRNA gene are leading to the emergence of CRHP strains [79]. In developed countries, 90% of CRHP appear as a result of mutations in two specific adjacent nucleotide positions—A2142G and A2143—which are the most prevalent and well-documented point mutations in this pathogen [80]. Efflux pumps and outer membrane proteins are also involved in clarithromycin-resistance. Notably, PPIs are structurally similar to efflux pump inhibitors, such as Phe-Arg-β-naphthylamide—a molecule able to decrease the MIC of ABs. As CRHP continues to escape efficient therapy, new drugs have been developed, such as vonoprazan—a novel potassium-competitive acid blocker that strongly inhibits H+/K+ ATPase-mediated gastric acid secretion [81]. In May 2022, The US Food and Drug Administration has approved two vonoprazan-based treatments, both superior to PPI-based triple or dual therapy [82].
Elkousy et al. demonstrated that both marjoram (Origanum majorana) and mandarin (Citrus reticulata) EOs exhibited antimicrobial activity against H. pylori. The authors stipulated that, thanks to its high content of oxygenated compounds (trans-sabinene hydrate, terpinen-4-ol, linalyl acetate, caryophyllene oxide and a-terpineol), marjoram EO displayed a lower MIC than the other EO. Additionally, the combination of the two EOs had a synergistic effect against H. pylori, with a lower MIC, equal to clarithromycin’s MIC [83] (Appendix B, Table A3). A study that investigated the effectiveness of four Piper spp. EOs against H. pylori determined that long pepper (Piper longum) EO recorded the same MIC as clarithromycin, followed by white pepper EO, tailed pepper and then black pepper. This study shows that, although the EOs came from plants in the same genus, individual components in each volatile oil led to variation in MIC values [84]. The same is true for different Pinus species, as Gad et al. determined that, among four pine EOs, P. pinea EO exhibited the highest anti-H. pylori activity, with a MIC comparable to that of clarithromycin [85]. In a study by Mariem et al., 54.54% of gastric biopsy H. pylori isolates showed resistance to at least one of the five tested ABs (erythromycin, clarithromycin, ciprofloxacin, levofloxacin or metronidazole), while mastic tree (Pistacia lentiscus) EO showed anti-H. pylori activity against all tested strains [86]. Several other EOs (wild thyme [87], cinnamon [88], cedarwood, oregano [89]) exhibited strong antimicrobial activity against H. pylori strains, while other EOs (common sage, lemon balm, English lavender [87], clove, thyme, rosemary [88], guabiraba [90]) showed good to mild antimicrobial effects. Various active compounds/EOs, such as geraniol [91], α-pinene (from P. atlantica) [92] or β-caryophyllene [93] are increasingly being tested on animal models, uncovering their anti-H. pylori activity (Appendix B, Table A3). However, data regarding EOs’ activity against CRHP are scarce.

3.3. Campylobacter spp., Fluoroquinolone-Resistant

Campylobacter spp. includes several commensal Gram-negative species, but also pathogenic strains of Campylobacter jejuni and Campylobacter coli [94]. C. jejuni alone is the leading cause of bacterial gastroenteritis in humans, surpassing Escherichia coli, Shigella spp. and Salmonella [95]. The main sources of contamination are raw or undercooked meat, unpasteurized milk or dairy products, contaminated vegetables, natural mineral water, shellfish and flies [96]. In developing countries, Campylobacter infection is generally limited to children and the clinical symptoms are usually indistinguishable from those caused by other enteric pathogens [97]. In some instances—usually in immunocompromised, pregnant and elderly patients—Campylobacter can trigger extraintestinal manifestations, including abscesses, meningitis, endocarditis and bacteraemia [98]. Moreover, a number of patients develop chronic sequelae, such as irritable bowel syndrome, reactive arthritis, Reiter’s Syndrome, Guillain–Barré Syndrome, Crohn’s disease and ulcerative colitis [99]. The main virulence factors of Campylobacter spp. are their ability to adhere and colonize (cadF, racR, virB11, pldA and dnaJ genes), invade intestinal epithelial cells (ciaB and ceuE genes) and produce toxins (cdtA, cdtB, cdtC genes) [100].
While self-limiting intestinal manifestations are treated using replacement of fluids and electrolytes, severe extraintestinal infections need treatment with the ABs of choice (macrolides and fluoroquinolones [101]) or with alternative solutions (tetracyclines and aminoglycosides), for which all have reported AMR [102]. Fluoroquinolones act by inhibiting DNA gyrase (GyrA and GyrB subunits) and topoisomerase IV (ParC and ParE subunits)—two enzymes crucial for bacterial survival, involved in DNA replication and repair, transcription and recombination [103]. Point mutations (T86I, T86K, A70T and D90N) in the quinolone-resistance-determining region (QRDR) of DNA gyrase’s GyrA subunit led to the emergence of FRC [104]. Moreover, drug efflux pump CmeABC works synergistically with the gyrA mutation, leading to the appearance of FRC; however, in the absence of the gyrA gene mutation, CmeABC over-expression does not correlate with ciprofloxacin resistance [105]. Nevertheless, fluoroquinolone resistance cannot be attributed to mutations in parC and parE genes (since Campylobacter lacks these particular genes); similarly, it appears that mutations in the gyrB gene are not related to the emergence of FRC [106].
Gahamanyi et al. evaluated the susceptibility of C. jejuni and C. coli to various natural products (plant extracts, EOs and active compounds) and frontline ABs. They determined that, among the tested products, cinnamon (Cinnamomum cassia) EO and its main compound (E)-Cinnamaldehyde, clove (Syzygium aromaticum) EO and its main compound eugenol, and baicalein had the lowest MIC and MBC values (25–100 µg/mL) [107] (Appendix B, Table A4). Duarte et al. tested the activity of coriander (Coriandrum sativum) EO and its major compound linalool against the two above-mentioned strains; once again both products showed comparable activities: inhibition of biofilm formation, reduction in quorum-sensing and inhibition of the microbial growth [108]. The anti-C. jejuni activity of clove EO and eugenol was supported by another study showing that the products perturb the expression of virulence factors, alter the morphology and induce oxidative stress in C. jejuni [109]. Ahmed et al. confirmed that clove and cinnamon EOs were effective against C. jejuni and S. aureus, but garlic EO did not share the same efficacy [110]. El Baaboua et al. determined that MDR Campylobacter spp. were sensitive to oregano (Origanum compactum), mint (Mentha pulegium) and lavender (Lavandula stoechas) EOs, which interfered with the microbial ability to form biofilms. Lavender and oregano acted synergistically with tetracycline or ampicillin, reducing the effective doses of EOs, tetracycline and ampicillin [111]. One interesting study investigated the potential of thyme (Thymus vulgaris) EO/gelatin nanofibers to inhibit C. jejuni growth in chicken. Food packaging containing thyme EO β-cyclodextrin ε-polylysine nanoparticles as antibacterial agents readily damaged C. jejuni cell membranes and reduced the microbial population [112]. Another noteworthy approach was that of Lin et al. who prepared chrysanthemum EO–chitosan–pectin triple-layer liposomes and determined that the product exhibited high anti-C. jejuni activity [113]. Lastly, even though some active compounds present promising in vitro results, further in vivo studies fail to reach the same outcomes. For instance, although (-)-α-pinene was able to reduce ciprofloxacin’s MICs (when used in combination), it did not manage to impede fluoroquinolone resistance development when added to enrofloxacin in broiler chickens [114] (Appendix B, Table A4).

3.4. Neisseria gonorrhoeae, Cephalosporin-Resistant, Fluoroquinolone-Resistant

Neisseria gonorrhoeae is a Gram-negative diplococcus and represents the etiological agent of gonorrhoea, the second most common sexually transmitted infection that affects both men and women [115]. According to WHO, N. gonorrhoeae causes significant morbidity and economic costs around the world, with 82.4 million new cases of gonorrhoea among adults and adolescents each year [116]. While the majority of gonorrhoea cases are asymptomatic, untreated infections may lead to serious complications such as endometritis, salpingitis, sterility, chronic pelvic pain, ectopic pregnancy, neonatal infections and increased risk of acquiring HIV [117]. Moreover, there are cases when the pathogen disseminates, causing skin, joint or tendon infection and, rarely, endocarditis or meningitis [118]. The primary step in N. gonorrhoeae pathogenesis is the bacterial adherence to the epithelium of the mucosa, mediated through surface structures: type IV pili, opacity (Opa) proteins, LOS and PorB—the major outer membrane protein porin [119]. After adhering, the pathogen replicates, forming microcolonies and biofilms, and sometimes even invades epithelial cells by transcytosis [120].
For many years, gonorrhoea was considered relatively easy to treat in monotherapy; however, due to AB overuse, AMR has emerged for most classes (sulphonamides, penicillins, tetracyclines, macrolides, fluoroquinolones and early-generation cephalosporins) [121]. Today, the most recommended gonorrhoea treatment is dual therapy: a single dose of a third-generation cephalosporin (250–500 mg intramuscular ceftriaxone or 400 mg peroral cefixime) in combination with azithromycin (1–2 g peroral) [122]. Concerningly, in 2018, ceftriaxone-resistant and azithromycin-resistant N. gonorrhoeae strains have been isolated [123]. Cephalosporin resistance is caused by mutations in various chromosomal regions that encode for important microbial proteins such as the transpeptidase domain of the PBP2 protein (penA gene), porin B subunit (porB1b gene) and PBP1 protein (ponA gene) [124], as well as overexpression of MtrCDE membrane pump proteins [125]. Fluoroquinolone resistance is acquired through mutations in QRDR: a single amino acid change in the GyrA subunit (positions 91 or 95) leads to intermediary resistance level, while three or more changes in the GyrA subunit (positions 91, 95 and 102), ParC (position 87 and 91) and/or Par E (position 439) proteins may lead to even higher MICs [126]. There is a pressing need to develop new diagnostic strategies and novel antimicrobials in order to preserve ceftriaxone, as it is the last empirical first-line monotherapy for gonorrhoea [127]. In 2021, WHO issued a general protocol called the Enhanced Gonococcal Antimicrobial Surveillance Programme, which aims to strengthen the quality, comparability and timeliness of gonococcal AMR data across multiple countries [128]. As the global burden rises, new collaborators join this programme to combat AMR in gonorrhoea [129].
Propolis extract has been shown to exhibit antimicrobial activity against ciprofloxacin-sensitive and ciprofloxacin-resistant N. gonorrhoeae strains, in a study by Vică et al.; depending on the harvesting region, the extracts presented with various inhibition zone diameters and MICs [130] (Appendix B, Table A5). Umaru et al. studied the effect of Molineria capitulate fruit EO on various pathogens and showed that both M. capitulate EO and its major component, myrcene, displayed antimicrobial activity against N. gonorrhoeae strains [131]. Soliman et al. investigated the antimicrobial properties of two guava (Psidium spp.) EOs and determined that both EOs showed good antibacterial effects, but Psidium cattleianum displayed preferential activity against N. gonorrhoeae [132]. Other publications showed that Eclipta alba EO is highly effective against N. gonorrhoeae strains [133], while Ferula tingitana EO displays a more modest antibacterial activity [134] (Appendix B, Table A5).

3.5. Salmonellae, Fluoroquinolone-Resistant

Salmonella spp. is a genus of facultative anaerobe Gram-negative bacilli having peritrichous flagella [135]. The genus Salmonella belongs to the family Enterobacteriaceae and includes two main species: Salmonella enterica and Salmonella bongori [136]. Around 99% of the Salmonella strains that cause infection in humans or other mammals belong to the S. enterica strains [137]. S. enterica is further subdivided into six-subspecies, serotypes, serogroups and serovars, according to the expression of somatic lipopolysaccharide O and flagellar H antigens [138]. The three major diseases caused by Salmonella in humans are: non-invasive non-typhoidal salmonellosis (niNTS), invasive non-typhoidal salmonellosis (iNTS) and typhoid fever [139]. Additionally, Salmonella serotypes can asymptomatically colonize humans’ gallbladders, thus making them chronic carriers and potential disseminators [136].
Non-typhoidal salmonellosis (NTS) is among the most prevalent global cause of foodborne illnesses [140] and includes infections caused by all Salmonella spp., with the exception of the distinct typhoidal serotypes: Typhi and Paratyphi A-C [139]. While niNTS may have a variety of clinical manifestations, the most common is gastroenteritis and is usually self-limiting [140]. Given that both humans and animals are potential hosts for niNTS [141], the infection is mainly transmitted via the consumption of animal products, but also unpasteurized dairy products, seafood and fruits [140]. The incidence of gastroenteritis due to niNTS peaks in the developing world, but it is also of considerable importance in developed countries; for instance, in the European Union (EU), salmonellosis is the second-most reported gastrointestinal infection in humans after campylobacteriosis [140]. When non-typhoidal Salmonella spp. (S. typhimurium and S. enteritidis) go beyond the gastrointestinal tract and invade normally sterile sites causing bacteraemia, iNTS occurs [142]. Contrary to niNTS, iNTS typically manifests as a febrile systemic illness (where diarrhoea is often absent) and lower respiratory tract disorders, due to co-infections with Mycobacterium tuberculosis and Streptococcus pneumoniae [139]. The global burden of iNTS is estimated by Ao et al., who evaluated 3.4 million cases of iNTS and approximated the annual death toll to around 700,000 [143]. In a similar study that excluded patients with HIV-associated iNTS, it was established that more than 60,000 deaths occurred in 600,000 cases of iNTS [142]. The most severe Salmonella spp. infections, typhoid and paratyphoid enteric fevers, are caused by Salmonella enterica subspecies enterica serovars Typhi and Paratyphi A, B and C [137,141]. Contrary to NTS broad host specificity, S. typhi is found only in humans [139]. For reasons not fully understood, it is estimated that around 5% of infected individuals will fail to clear the infection within a year, and will instead progress to a chronic carrier state where the bacteria will primarily reside in the hepatobiliary tract and gallbladder [139]. These systemic diseases cause more than 200,000 deaths globally, sub-Saharan Africa and Asia accounting for around 46% and 32% of typhoid fever cases, respectively [137]. Worryingly from 1990 to 2010 annual mortality from typhoid fever has increased by 39% [139].
Antibiotic therapy is not needed for Salmonella-induced gastroenteritis while, for invasive Salmonella infections, ampicillin, chloramphenicol and trimethoprim-sulfamethoxazole are used as first-line treatment [136]. However [139], a large proportion of AB-resistant Salmonella are acquired through the consumption of contaminated food of animal origin [144]. Poultry are the main source of human salmonellosis. In order to move forward through the food chain, Salmonella must be resistant to different environmental stress conditions such as heat, desiccation, nutrient starvation or biocides, and Salmonella spp. use quorum sensing to control processes such as luminescence, sporulation, virulence or biofilm formation [135].
AMR can be achieved by mutations in different chromosomal loci that are part of a core set of genes, such as genomic islands and through exogenous resistance genes carried by mobile genetic elements that can diffuse horizontally [145]. Salmonella spp. mediate resistance to ABs by three major mechanisms: drug inactivation, protection of the AB target sites, and removal of ABs using efflux pumps or multidrug pumps. Firstly, the main mechanism, drug inactivation, is characterized by destruction of antimicrobial agents (quinolones, macrolides) through chemical modification using enzymes that catalyse reactions such as acetylation, phosphorylation and adenylation [144]. Secondly, Salmonella spp. can protect the target sites of ABs, which are typically either enzymes or other specific cell structures [144,146]. For instance, the plasmid-encoded quinolone resistance protein (Qnr) confers resistance to quinolones by acting as a DNA homolog that competes for the binding of DNA gyrase and topoisomerase IV [144,146]. Thirdly, Salmonella spp. can use relatively nonspecific efflux pumps (such as AcrAB-TolC) encoded by genes within mobile elements to reject fluoroquinolones, β-lactams and carbapenems [144,146].
Emerging MDR Salmonella spp. have changed the treatment regimen towards using second-line ABs such as fluoroquinolones (ciprofloxacin) and third-generation cephalosporins [136]. However, as fluoroquinolones are extensively exploited for animal production in several countries [147], WHO notifies that the prevalence of FRS is growing quickly [136]. Under normal conditions, quinolones enter bacteria through porins and then exert their bacteriostatic activity by binding to the gyrase/topoisomerase IV–DNA complex [144]. Resistance to nalidixic acid, driven by a single mutation within gyrA, is a precursor to resistance to all quinolones [148]. Additionally, the fluoroquinolone resistance in Salmonella is caused either by chromosomal mutations in the QRDRs of the gyr and par genes [149], or by the acquisition of several plasmid-mediated quinolone resistance (PMQR) genes [143]: (1) qnr (quinolone resistance proteins); (2) aac(6′)-lb-cr (aminoglycoside-modifying acetyltransferase); and (3) oqxAB and qepA [141,144]. Increased expression of the AcrAB-TolC multidrug efflux system was also shown to play an important role in the development of high-level fluoroquinolone resistance [149]. Generally, resistance to ciprofloxacin in enteric bacteria is acquired through gyrA mutations, while PMQR genes also induce low-level resistance. In contrast, in Salmonella spp., mutations in transferable PMQR gene qnr were observed in all ciprofloxacin-resistant isolates, whereas gyrA mutations were often found in isolates with reduced ciprofloxacin susceptibility but not in all ciprofloxacin-resistant isolates [150].
To our knowledge, there are no articles describing the antimicrobial action of EOs on FRS.

3.6. Staphylococcus aureus, Methicillin-Resistant, Vancomycin-Intermediate and -Resistant

Staphylococcus aureus is a ubiquitous Gram-positive, coagulase-positive, facultative anaerobe, nonmotile and spherical bacterium (grape-like cluster), possessing not a flagella but a capsule [151]. It can be present either as a commensal member of the microbiota (usually localized in the upper respiratory tract, gut or on the skin) or as an opportunistic pathogen [152]. S. aureus may cause mild to severe infections, depending on the localization of the infection (skin, soft tissues or blood) and patient’s characteristics (age—more aggressive in infants and older people; comorbidities—immunosuppression, diabetes, heart or renal diseases; and others factors—implantable medical devices, low social economic status or intravenous drug use) [153,154]. The disorders caused by S. aureus range from skin infections (pimples, impetigo, folliculitis, boils, cellulitis, carbuncles, scalded skin syndrome, abscesses) to more severe and life-threatening diseases, including pneumonia, osteomyelitis, endocarditis, meningitis, toxic shock syndrome, bacteremia or sepsis [151,155].
While most people who are colonized with S. aureus will not develop an invasive infection, S. aureus infections are overall extremely frequent and particularly problematic due to AB resistance and the ability to form biofilms [153]. In 1942, shortly after the introduction of penicillin into clinical practice, S. aureus started hydrolysing the beta-lactam ring and establishing penicillin resistance. Then, in the late 1950s, the appearance of the semi-synthetic beta-lactamase-resistant AB, methicillin, led to the emergence of the first MRSA strain only 2 years after its introduction [156] due to mutations in the gene encoding for penicillin-binding protein 2a or 2′ (PBP2a; PBP2′) (mecA) [152]. Nowadays, S. aureus remains one of the most common resistant pathogens worldwide [154,157]. According to the American Centers for Disease Control and Prevention, S. aureus has evolved into a major health-threatening pathogen, as invasive infections caused by MRSA have a high mortality rate and its remarkable ability of acquiring AB resistance against multiple drug classes significantly complicates the treatment [158]. Although the rates of AB resistance are widely variable depending on the country (relatively low AMR in Scandinavian countries and high AMR in Southern Europe, USA and China, due to differences in hygiene and surveillance measures), they are still on the rise in poorly developed countries (South America and some countries in Africa) [151,159].
The beginning of the 1960s was marked by the discovery of the SCCmec complex, a key factor that enabled S. aureus to acquire resistance to most of the beta-lactam ABs [151]. Currently 12 SCCmec complexes are known, and they are divided by (1) the group of cassette chromosome recombinase (ccr) complex and (2) the category of mec complex. With the exception of type XI SCCmec which contains homologue mecC, all SCCmec types include mecA, a component that encodes for PBP2a [160]. PBP2a is responsible for the transpeptidase action in the biosynthesis of peptidoglycan, in the presence of beta-lactam AB, inhibiting the function of PBP 1, 2, 3 and 4. While mecC is a variant of mecA which encodes for PBP2aLGA251 (named after MRSA strain LGA 251), mecB is a plasmid-developed methicillin resistant form with unclear mechanism of resistance [161,162]. mecA’s expression depends on regulators encoded by mecI, mecR1 and mecR2, and on regulators of gene expression, such as blaZ, blaI and blaRI [162]. The auxiliary fem genes seem to also have an important influence on the resistance phenotypes [163].
Over time S. aureus has also gained resistance to vancomycin, the AB used as a first-line treatment for the past six decades [164,165]. The feasible alternative options to vancomycin treatment include high doses of daptomycin co-administered with either gentamicin, rifampicin, linezolid, trimethoprim + sulfamethoxazole or beta-lactam. If response to daptomycin is inadequate, secondary options include monotherapy or co-administration with a number of possible ABs: quinupristin + dalfopristin, linezolid, telavancin or trimethoprim + sulfamethoxazole [166]. In 2002, VRSA strains were identified in the USA; it is thought that resistance was mediated by vanA gene acquired from E. faecalis on the plasmid-borne transposon Tn1546 [167]. Given that VRSA has a preference for diabetic wounds where vancomycin-resistant enterococci reside, there is a clear opportunity for horizontal gene transfer of Tn1546 accommodating vanA [168].
The study performed by Oo T et al. evaluated the antimicrobial efficacy of crude extract and EO obtained from nutmeg (Myristica fragrans Houtt.) on S. aureus efflux pump systems (chromosomal norA and mepA) involved in the resistance mechanism of MRSA. They found that elemicin, myristicin, methoxyeugenol and asarone can work as efflux pump inhibitors, thus potentiating the antimicrobial activity of classical ABs. They observed a synergistic activity of ciprofloxacin and the two nutmeg formulations, concluding that both the extract and the EO act as efflux pump inhibitors, while ciprofloxacin acts as an efflux system [169] (Appendix B, Table A6). Alharbi NS et al. highlighted the effect of two different concentrations of tailed pepper (Piper cubeba L.) EO against MRSA ATCC 43300. While the higher concentration induced serious microscopic deteriorations of the bacterial cell, the lower concentration had no observable microscopic effects; however, significant modifications within the cell wall were observed at a nanoscopic level. The authors concluded that the EO induced an antibacterial action on both methicillin- and oxacillin-resistant S. aureus strains through its action upon the cell wall and the cytoplasmic membrane [170] (Appendix B, Table A6).
Piasecki B et al. tested 19 EOs extracted from Cymbopogon spp. and determined that C. flexuosus (lemongrass) EO exhibited the highest antibacterial activity, while citronellol stood out as the most powerful active compound (from citronellol, geraniol and citral). Moreover, all tested EOs manifested antibiofilm properties, with a MBIC ranging from 1 to 4 mg/mL. Nonetheless, after 48 h of treatment at a maximum concentration, cardiotoxicity and shortened tail were observed in zebrafish, with C. martini var. motia showing the most toxic potential (about 20 times more toxic than C. winterianus) [171]. Merghni A et al. also showed that both blue gum (Eucalyptus globulus Labill.) EO and its main active compound, 1,8-cineole, present excellent antibiofilm properties and bacteriostatic effects, with the EO inducing a more potent effect on quorum sensing [172] (Appendix B, Table A6).
It is well known that Gram-negative bacteria are more resistant to ABs and toxins than Gram-positive bacteria, due to their perfected cell wall. Specifically, they have a multi-layered, complex cell wall, covered by a hydrophilic membrane abundant in lipopolysaccharides. Moreover, the periplasmic space of Gram-negative bacteria contains enzymes capable of degrading exogenous molecules, thus preventing the access of inhibitors. Several studies have also emphasized that there is a difference in the antibacterial activity of EOs in Gram-positive versus Gram-negative bacteria [173] (Appendix B, Table A6).
Predoi D et al. investigated the activity of EOs in hydroxyapatite, a calcium phosphate compound best known for its similarities with human hard tissues and its applicability in medicine (dental applications, bone regeneration). Their work was based on the premise that, by incorporating antibacterial agents in hydroxyapatite, the risks of postoperative infections would be reduced. Thus, they highlight the physio-chemical properties and antimicrobial activity of two nanocomposites of hydroxyapatite embedded with basil and lavender EOs, with the latter exhibiting the best antibacterial properties [174]. The team continued to look into the effectiveness of hydroxyapatite associations with EOs in further studies [175,176] (Appendix B, Table A6).
Interestingly, Mouwakeh A and collaborators postulated the fact that black caraway (Nigella sativa L.) EO and its active components (carvacrol and p-cymene) could be used as MRSA modifiers of resistance. In their study they hypothesized that the hydroxyl group in carvacrol and thymoquinone might play a key role in their antimicrobial activity [177].
De Moura et al. focused on evaluating the antioxidant, antibacterial and antibiofilm activity of nerolidol, an acyclic sesquiterpene present in many species (usually the trans isomer) such as wood oil, red oil, cabreuva oil and balm of Peru [178]. It is of valuable importance in the cosmetic industry (as a preservative agent to fixate perfumes) and pharmaceutical industry (as a stimulating agent to increase the concentration of active ingredients in transdermal formulations) [179]. Nerolidol has a broad spectrum of pharmacological/biological actions, including antineoplastic, anti-inflammatory, antinociceptive, larvicidal and leishmanicidal activity [179], and, recently, its implication for neurodegenerative disorders has been described [180]. De Moura and colleagues found that nerolidol is effective for both MSSA and MRSA at the same MIC of 2 mg/mL [178].
Two EOs of lemon verbena (Aloysia citriodora Palau) collected from different Palestinian regions were tested for their antimicrobial, antioxidant, cytotoxic and cyclooxygenase (COX) inhibitory effects by Jaradat N et al. Both EOs showed antimicrobial activity against MRSA, P. vulgaris and C. albicans, but the Baqa al-Gharbiyye EO manifested stronger antioxidant, cytotoxic and anti-cyclooxygenase activities, compared with the Umm al-Fahm EO [181] (Appendix B, Table A6).
The work of Adrian Man and collaborators investigated the antimicrobial effect of some well-known EOs in micellar and aqueous extracts. They assessed the antibacterial activity of oregano, lemon, thyme, myrtle and frankincense EOs against S. aureus, E. faecalis, E. coli, K. pneumoniae and P. aeruginosa, and determined that gram-positive bacteria (including MRSA) were more susceptible compared with P. aeruginosa which was found to be the most resistant. The authors highlighted that micellar suspensions of EOs (especially those containing high concentrations of terpenes and terpenoids—i.e., oregano, thyme, lemon EOs) can be introduced in new topical formulations to enhance the penetrability of EOs and, thus, their action [41] (Appendix B, Table A6).
The study of Kwiatkowski P. addressed the efficacy of several active compounds in EOs against mupirocin-susceptible and low-level mupirocin-resistant MRSA. Notably, mupirocin is an AB synthesized by Pseudomonas fluorescens and has medical applications. It is used on nasal mucosa as an ointment in order to decolonize S. aureus. It is applied for 5–14 days (especially before a surgical intervention) to reduce the risk of postoperative wound infection and prevent the spread of bacteria to medical staff’s hands [182]. Screening of S. aureus is mandatory in patients undergoing cardiac or orthopaedic surgery, especially for MRSA. Moreover, all patients prior to surgical procedures must have a full-body shower (with/without disinfectants such as chlorhexidine) to reduce the number of surgical site infections and complications caused by this highly pathogenic and resistant microorganism [183]. The mechanism by which mupirocin exerts its antibacterial activity consists of the inhibition of isoleucyl-tRNA (ended by ileS gene on the chromosome) and, thus, the blocking of protein synthesis within the bacteria. Two types of mupirocin resistance have been described: low-level (MupRL, MIC: 8–256 mg/L) and high-level (MupHR, MIC: 512 mg/L). Usually, the MIC of mupirocin on sensible strains is around 4 mg/L. If MupRL is caused by ileS gene mutation, MupRH is induced by a plasmid encoded in the ileS2 gene (responsible for encoding a different isoleucyl-tRNA synthase) which will trigger a low affinity for mupirocin [182]. In the study performed, carvacrol highlighted the best inhibitory action on the tested MRSA strains and 1,8-cineole induced a synergistic action against MupRL MRSA with penicillin G. The authors suggested that high precision technology that trigger crucial checkpoints for staphylococcal resistance is needed [184] (Appendix B, Table A6).
The group of Manzuoerh et al. investigated the in vivo effect of topical dill (Anethum graveolens) EO versus mupirocin on a MRSA-induced infection in a BALB/c mouse model. They used two different concentrations of dill EO and determined that the main active components were α-phellandrene (47.3%), p-cymene (18.5%) and carvone (14.1%). The topical administration of dill EO decreased the inflammation and stimulated re-epithelialization, angiogenesis and collagen and fibroblast sedimentation. The topical effects were the result of an increased expression of p53 and caspases-3, in the case of the anti-inflammatory activity, and Blc-2, VEGF and FGF-2 expression in the case of the proliferative activity. Together with the stimulation of collagen synthesis through increases of ERα expression level, all these effects led to improvements in wound healing and reduction of the infection [185]. Mahboubi M et al. performed a similar study on EOs obtained from the aerial parts of Oliveria decumbens and Pelargonium graveolens. The main active substances in Oliveria decumbens EO were thymol (50.1%), γ -terpinene (20.7%) and p-cymene (17.6%), while beta-citronellol (39.3%) and geraniol (23.6%) were present in Pelargonium graveolens EO. Both the herbal cream containing the two EOs and the mupirocin formulation diminished log CFU (colony-forming units). Although further clinical and toxicological studies are required, topical formulations of EOs could be used in the treatment of wound infections, given their antimicrobial activity and healing effects [186] (Appendix B, Table A6).
In 2020, Chen J and collaborators presented the metabolomics analysis of antibacterial activity of camphor leaves EO (Cinnamomum camphora) [187]. It is worth mentioning that metabolomics analysis contributes to a multifactorial description of a drug mechanism of action and identifies eventual dynamic changes in metabolites as a response to drug treatment [188]. Moreover, key marker identification through pathway analysis can be used in order to identify metabolites’ changes in the course of antimicrobial activity. The camphor EO was rich in linalool (26.6%) and eucalyptol (16.8%), as well as α-terpineol (8.7%), isoborneol (8.1%), β-phellandrene (5.1%) and camphor (5.0%). The metabolomics analysis revealed 74 different metabolites (29 upregulated and 45 downregulated). The EO stimulated the activity of isocitrate dehydrogenase (47.35%) and decreased the activity of malate dehydrogenase (72.63%), succinate dehydrogenase (31.52%) and α-ketoglutarate dehydrogenase (63.29%). The authors concluded that the antimicrobial activity was the result of an imbalance in the amino acid metabolism, a rise in the apoptosis rate and a disruption in cell wall and membrane with the efflux of DNA, RNA and proteins from the MRSA cell [187] (Appendix B, Table A6).
There is a scarcity of studies describing the antimicrobial properties of EOs against VRSA and VISA strains. Vasconcelos SECB et al. investigated the antibacterial activity of Mexican mint (Plectranthus amboinicus) EO on VRSA and oxacillin-resistant S. aureus and determined that the strains were more sensitive when both carvacrol and EO were used [189] (Appendix B, Table A6).
Further data from these and other studies are provided in Appendix B, Table A6.

4. Conclusions

This systematic review presents the most recent studies of EOs’ activity against pathogens on WHO’s priority 2 list. While the emergence of AMR is a natural evolutionary process in bacteria, the widespread use and misuse of ABs has had an amplifying effect on this process.
Due to their selectivity on pathogenic bacteria and relatively low toxicity, EOs have been proposed as good alternatives and valuable adjuvants in a variety of infections. However, testing and evaluating the antimicrobial activity of EOs is difficult because of their volatility, water insolubility and complexity. Moreover, long incubation times during the testing period may result in the evaporation or decomposition of some of the active components. EOs’ phytocompound composition can vary and is influenced by plant subspecies, geographical location, growing conditions, growth phase, extraction method, and exposure to light, temperature and humidity.
There is a need for a consensus on the methodology used to assess the antimicrobial activity of EOs. In our paper, we refrained from a strict assessment of the results in the reviewed studies, given the heterogeneity of research design and techniques. Further research incorporating standardization methods to form a universal consensus are needed to allow a critical examination of EOs’ antimicrobial action. As a general consideration, some authors interpret the findings as follows: MIC: <100 µg/mL—highly active; MIC = 100–500 µg/mL—active; MIC = 500–1000 µg/mL—moderately active; MIC = 1–2 mg/mL—low activity; and MIC: >2 mg/mL—inactive [141,149].
Interestingly, suggestions that might enable the use of EOs in therapy have been mentioned as limitations in some of studies cited above. Thus, in order to provide cohesive results and to facilitate the use of EOs in clinical practice, the following should be considered: (1) assurance that the techniques used to analyse EOs’ composition are compliant with pharmacopeial requirements; (2) investigation of at least 10 different strains of the same microbial species in the same study, as they vary greatly from one another; (3) inclusion of anti-biofilm activity; (4) implementation of cytotoxicity assays for each study as there are limited data regarding EO dosage and safety, especially in humans; (5) increased standardization in methodology; and (6) intensive monitoring over time. Until further research elicits sufficient data for the large-scale use of EO phytocompounds in the treatment of infection, the general population might incorporate EOs into their daily diet for prophylaxis. For instance, not all cultures take the best benefits from using spices, even though they have good, non-specific and multidirectional antimicrobial activity.
In conclusion, while there is growing evidence for the introduction of EOs into clinical practice, especially those which observed no toxic effects, they are currently underused in practice. Based on their efficacy from the combined action of antimicrobial compounds and their synergistic activity with conventional ABs or food preservatives, EOs have the potential to lessen the burden of AMR. The development of standardized techniques for analysing the in vivo antimicrobial activity of EOs is a critical first step.

Author Contributions

Conceptualization, V.O.B., M.R., D.M. and C.D.; methodology, V.O.B., D.M., C.O., B.B., A.T. and O.D.; validation, D.M., M.R., C.D., O.C., R.F. and M.A.; data curation, M.R., V.O.B., A.T., B.B., D.M., A.L. and G.D.C.; writing—original draft preparation, M.R., V.O.B., A.L., A.T., C.O. and B.B; writing—review and editing, O.D., M.A., S.L.B., D.M. and C.D. All authors have read and agreed to the published version of the manuscript.

Funding

This research received no external funding. The APC was funded by “Victor Babes” University of Medicine and Pharmacy, Timisoara, Romania.

Institutional Review Board Statement

Not applicable.

Informed Consent Statement

Not applicable.

Data Availability Statement

All the extracted data is presented within the manuscript.

Conflicts of Interest

The authors declare no conflict of interest.

Abbreviations

ABsAntibiotics
AMRAntimicrobial resistance
ARGAntibiotic resistant genes
ATPAdenosine triphosphate
CRHPClarithromycin-resistant H. pylori
CRNGCephalosporin-resistant N. gonorrhoeae
EOsEssential oils
ESBLExtended spectrum beta-lactamase
ESKAPEE. faecium, S. aureus, K. pneumoniae, A. baumanni, P. aeruginosa and E. cloacae
EUEuropean Union
FICFractional inhibitory concentration
FRNGFluoroquinolone-resistant N. gonorrhoeae
FRSFluoroquinolone-resistant Salmonellae spp.
GLASSGlobal Antimicrobial Resistance Surveillance System
HPPHigh priority pathogen
IC50Half-maximal inhibitory concentration
iNTSInvasive nontyphoidal salmonellosis
KPCK. Pneumoniae carbapenemase
MBCMinimum bactericidal concentration
MBEC50%Minimum biofilm inhibitory concentration
MDRMultidrug resistant
MICMinimum inhibitory concentration
MRSAMethicillin-resistant Staphylococcus aureus
MSSAMethicillin-sensitive Staphylococcus aureus
niNTSNon-invasive non-typhoidal salmonellosis
NTSNontyphoidal salmonellosis
PDRPandrug-resistant
PMQRPlasmid-mediated quinolone resistance
PPIProton-pump inhibitor
RNARibonucleic acid
QRDRQuinolone resistance-determining region
VISAVancomycin-intermediate Staphylococcus aureus
VREFVancomycin-resistant Enterococcus faecium
VSEFVancomycin-sensitive Enterococcus faecium
VRSAVancomycin-resistant S. aureus
WHOWorld Health Organization
XDRExtensively drug-resistant
ZOIZone of inhibition

Appendix A

Table A1. Top 5 European countries in terms of AMR to HPPs, according to the Surveillance Atlas of Infectious Diseases [32].
Table A1. Top 5 European countries in terms of AMR to HPPs, according to the Surveillance Atlas of Infectious Diseases [32].
BacteriaAB Resistance (Year)Resistant Strains Proportion (%)Country
E. faeciumVancomycin (2020)56.6Lithuania
44.2Cyprus
41.8Greece
40.0Slovakia
39.3Romania
S. aureusMethicillin (2020)49.1Cyprus
47.3Romania
40.2Greece
33.5Italy
29.7Portugal
N. gonorrhoeaeCiprofloxacin (2019)100.0Cyprus
100.0Croatia
80.0Italy
78.5Hungary
75.0Estonia
Cefixime (2019)50.0Cyprus
11.1Croatia
3.0Italy
2.5Belgium
1.8Portugal
Ceftriaxone (2019)0.9Portugal
0.6Belgium
0.2Norway
0.0Austria
0.0Germany

Appendix B

Table A2. Studies assessing the antimicrobial activity of essential oils against vancomycin-resistant Enterococcus faecium.
Table A2. Studies assessing the antimicrobial activity of essential oils against vancomycin-resistant Enterococcus faecium.
Study Team and YearBacterial StrainEssential Oil(s)Method(s)ResultsReference
Saki M et al., 2020VREF, MRSACinnamomum zeylanicumAgar disc diffusion
Broth microdilution
C. zeylanicum EO (bark; containing: eugenol, trans-cinnamaldehyde, coumarin, benzaldehyde, diacetone alcohol, benzylcarboxaldehyde, styrene and phenol) showed potent antibacterial effects on the bacterial isolates. The most sensitive isolate was MRSA, followed by VREF (MIC = 0.15–1.25 μL/mL for S. aureus; MIC = 0.15–2.5 μL/mL for E. faecium).[63]
Iseppi R et al., 2021Vancomycin-resistant Enterococcus spp.,
MRSA
Citrus aurantium subsp. amara Engler, Citrus × limon L. Osbeck, Eucalyptus globulus Labill., Melaleuca alternifolia (Maiden and Betche) and Cupressus sempervirens L. (Mill.)Agar disc diffusion
Broth microdilution
All EOs displayed antibacterial effect against all strains to different extents, but M. alternifolia EO was the most effective and C. aurantium showed the lowest activity. EO–EO and EO–AB associations showed a synergistic outcome in most tests and were effective against biofilm formation.[64]
Sakkas H et al., 2018Vancomycin-resistant Enterococcus spp.,
MRSA
Ocimum basilicum L. (estragole), Matricaria chamomilla L. (bisabolol and trans-b-farnesene), Thymus capitatus L. (carvacrol and thymol), Melaleuca alternifolia (terpinen-4-oland p-cymene) and Thymus vulgaris L. (thymol, linalool and p-cymene)Broth microdilutionT. capitatus EO yielded the best antimicrobial results followed by T. vulgaris, M. alternifolia and O. basilicum, while M. chamomilla EO exhibited weak antibacterial properties (MIC for S. aureus = 0.06–0.5% (v/v) for T. capitatus; 0.06–1% (v/v) for T. vulgaris; 0.12–1% (v/v) for M. alternifolia; 0.25–4% (v/v) for O. basilicum; and 2->4% (v/v) for M. chamomilla; MIC for Enterococcus spp. = 0.25–1% (v/v) for T. capitatus; 0.5–2% (v/v) for T. vulgaris; 1–4% (v/v) for M. alternifolia; >4% (v/v) for O. basilicum; and >4% (v/v) for M. chamomilla).[67]
Owen L et al., 2019Vancomycin-sensitive E. faecium (VSEF), VREF, MSSA, MRSAOriganum compactum Benth (carvacrol), Aniba roseadora Ducke (linalool) and Cuminum cymimum L. (cuminaldehyde)Kirby–Bauer disc diffusion
Thin layer chromatography-direct bioautography
O. compactum EO exhibited strong antimicrobial activity against S. aureus and E. faecium (MIC = 0.29–1.15 mg/mL). C. cymimum EO was strongly antimicrobial against MSSA and MRSA (MIC = 0.58–2.33 mg/mL), but had weaker activity on E. faecium (MIC = 18.60–37.20 mg/mL). A. roseadora EO was relatively inactive against S. aureus and E. faecium (MIC = 8.80–35.20 mg/mL).[66]
Owen L et al., 2020VSSA, VREFOriganum compactum Benth (carvacrol), Aniba roseadora Ducke (linalool) and Cuminum cymimum L. (cuminaldehyde)Checkerboard method
Time kill assay
Transcriptomic analysis
Gradient plate method
Sodium chloride tolerance
Galleria mellonella treatment assays
β-galactosidase leakage
The EO–vancomycin combination is able to kill clinical isolates of VRE (2.33–5.25 log10 CFU/mL reduction). However, in vivo G. mellonella larvae assay showed no antimicrobial activity of the EO–vancomycin combination.[65]
Di Vito M et al., 2021Methicillin-sensitive E. faecium, MRSALavandula angustifolia, Lavandula intermedia, Origanum hirtum, Satureja montana, Monarda didyma, Monarda fistulosaBroth microdilutionS. montana and O. hirtum EOs exhibit the highest activity (IR90 = 0.25–1% v/v). O. hirtum and M. didyma hydrolates were more active than the other three (IR90 = 50% v/v). Hydrolates need to be 25–200 times more concentrated than EOs to reach the same antimicrobial activity.[68]
Table A3. Studies assessing the antimicrobial activity of essential oils against clarithromycin-resistant Helicobacter pylori.
Table A3. Studies assessing the antimicrobial activity of essential oils against clarithromycin-resistant Helicobacter pylori.
Study Team and YearBacterial StrainEssential Oil(s)Method(s)ResultsReference
Elkousy R et al., 2022H. pylori ATCC 43504Origanum majorana L. (marjoram)
Citrus reticulata L. (mandarin)
Micro-well dilutionO. majorana is more effective against H. pylori (MIC = 11.4 mg/mL), while C. reticulata possess a slightly lower antibacterial activity (MIC = 31.25 mg/mL). The combination of the two EOs exhibited a synergistic inhibitory effect against H. pylori (MIC = 1.95 mg/mL).[83]
Al-Sayed E et al., 2021H. pylori RCMB 031124, ATCC 43504Piper nigrum L. (black and white pepper)
Piper longum L. (long pepper)
Piper cubeba L.F. (tailed pepper)
Micro-well dilutionP. longum EO had the highest anti-H. pylori activity (MIC = 1.95 μg/mL). White pepper EO was next in terms of antimicrobial effect (MIC = 3.90 μg/mL), followed by P. cubeba and P. nigrum EO (MIC = 7.81 μg/mL).[84]
Meriem M et al., 2016H. pylori isolated from gastric biopsyPistacia lentiscus var. Chia (mastic tree)Disc diffusionP. lentiscus showed a strong antimicrobial activity, confirmed by the MIC (1/5000, v/v) and the inhibition diameters (32 mm, 26.66 mm, 19.67 and 12.33 for the pure and diluted oil to 1/2, 1/4 and 1/8). The effect on H. pylori can be attributed to its content in monoterpenes (α-pinene, limonene and β-myrcene).[86]
Knezevic P et al., 2018Metronidazole-resistant H. pylori,
H. pylori ATCC 26695 (ATCC 700392)
Juniperus communis L.,
Hyssopus officinalis L.,
Salvia officinalis L.,
Melissa officinalis L.,
Lavandula angustifolia Mill.,
Ocimum basilicum L.
Thymus serpyllum L.
Broth microdilutionJ. communis, H. officinalis and O. basilicum EOs did not show any antibacterial activity, with the highest applied concentrations. S. officinalis EO inhibits growth of H. pylori ATCC 26695 (MIC = 4 µL/mL), while L. angustifolia and M. officinalis EOs have anti-H. pylori activity depending on oil composition and strain properties. The most active EO was T. serpyllum (MIC = 2.0–4.0 µL/mL).[87]
Ali S et al., 2022H. pylori isolated from gastric biopsySyzygium aromaticum L. (clove)
Thymus vulgaris L. (thyme)
Rosmarinus officinalis L. (rosemary)
Matricaria recutita L. (chamomile)
Cinnamomum zeylanicum L. (cinnamon)
Disc diffusion
Agar well diffusion
Cytotoxicity test
C. zeylanicum EO has the strongest anti-H. pylori activity, with a mean inhibition zone of 23.4 mm, higher than the levofloxacin positive control. Mean inhibition zones for S. aromaticum, T. vulgaris and R. officinalis EOs were 19.8 mm, 15.5 mm and 9.8 mm, respectively.[88]
Korona-Glowniak I et al., 2020H. pylori ATCC 43504
H. pylori isolated from clinical settings (both sensitive and resistant)
Pinus sylvestris L. (pine needle),
Citrus lemon L. (lemon),
Abies alba Mill (silver fir),
Thymus vulgaris L. (thyme),
Cymbopogon schoenanthus (L.) Spreng (lemongrass),
Juniperus virginiana L. (cedarwood)
Melissa officinalis L. (lemon balm),
Melaleuca alternifolia Maiden et Betche (tea tree),
Origanum vulgare L. (oregano)
Urease inhibition activity
Antioxidant activity analysis
Broth microdilution
The most active EOs were T. vulgaris, C. schoenanthus, J. virginiana and M. officinalis (MIC = 15.6 mg/L), followed by O. vulgare (MIC = 31.3 mg/L), M. alternifolia (MIC = 62.5 mg/L) and P. sylvestris, A. alba, and C. lemon (MIC = 125 mg/L) EOs. Regarding urease inhibition activity, the most efficient EO was J. virginiana EO (IC50 = 5.3 mg/L), inhibiting urease at sub-MIC concentrations. [89]
Gad H. et al., 2021H. pylori RCMB 031124, ATCC 43504Pinus canariensis C. Sm. ex D.C.,
Pinus halepensis Miller,
Pinus pinea L.
Pinus roxburghii Sarg.
Well dilutionP. pinea EO exhibited the highest antimicrobial activity (MIC = 3.9 μg/mL), comparable to that of clarithromycin. P. halepensis, P. roxburghii and P. canariensis EOs showed a milder anti-H. pylori activity (MIC = 15.6 μg/mL). [85]
Neves NCV C et al., 2022Clarithromycin-sensitive H. pylori, CRHP—type cultures and clinical isolate strainsCampomanesia lineatifolia R. and P.Broth microdilutionC. lineatifolia EO inhibits the growth of all H. pylori strains at the lowest concentration tested (MIC = 6 µL/mL). [90]
Bhattamisra S et al., 2019H. pylori ATCC 43504Geraniol (active component found in the lemongrass, rose, palmarosa, ginger, orange, lavender, citronella and nutmeg EOs)Measurement of gastric juice volume, pH and total acidity
Determination of myeloperoxidase
Determination of total glutathione
Rapid urease test
Histopathological evaluation
Geraniol presents antiulcer effect and anti-H. pylori action, associated with decreased myeloperoxidase activity, gastric secretion and bacterial load, as well as increased glutathione levels and gastric pH. [91]
Memariani Z et al., 2017Clinical H. pylori strainsPistacia atlantica Desf.Disc diffusion assay
Micro-well dilution assay
Microscopic evaluation of gastric ulcer
Measurement of ulcer index and calculation of protection rate
Acute toxicity
P. atlantica EO (rich in α-pinene) was safe up to 2000 mg/kg and no clinical sign of toxicity was observed. All H. pylori strains were sensitive to this EO (inhibition zone = 26–35 mm, MIC = 275–1100 µg/mL). The highest dose of EO (100 mg/kg) protected significantly better against peptic ulcer than that of the ranitidine treated group.[92]
Jung D et al., 2020H. pylori KCTC12083β-caryophyllene (active compound found in cloves, basil, cinnamon, and copaiba)Histopathological assessment
In vitro quantification of colony forming units
β-caryophyllene dose-dependently diminished H. pylori levels and inflammation in gastric mucosa. In vitro test determined that a concentration of 1 M β-caryophyllene was able to eradicate 99.9% of H. pylori strains but other doses did not have the same antimicrobial efficacy.[93]
Table A4. Studies assessing the antimicrobial activity of essential oils against fluoroquinolone-resistant Campylobacter spp.
Table A4. Studies assessing the antimicrobial activity of essential oils against fluoroquinolone-resistant Campylobacter spp.
Study Team and YearBacterial StrainEssential Oil(s)Method(s)ResultsReference
Gahamanyi N et al., 2020C. jejuni strains ATCC 33560TM and MT947450
C. coli strains ATCC 33559TM and MT947451
Cinnamomum cassia (L.) J. Presl (cinnamon) extract and EO,
Salvia plebeia R. Br (common sage) extract,
Mentha canadensis L. (wild mint) extract,
Scutellaria baicalensis Georgi (skullcap) extract,
Meehania urticifolia (Miq.) Makino (nettle-leaf mint) extract,
Syzygium aromaticum L. (clove) EO,
(E)-Cinnamaldehyde, eugenol, baicalein, kuraridin and emodin
Broth microdilutionThe highest anti-Campylobacter activity was recorded for C. cassia EO and phytochemical (E)-cinnamaldehyde (MIC = 25–50 µg/mL). Clove oil and its major compound eugenol showed as well activity against Campylobacter spp. (MIC = 50–100 µg/mL). Other active compounds displayed good antimicrobial effects: baicalein (MIC = 32–64 µg/mL) and kuraridin (MIC = 48 µg/mL), while emodin had the lowest activity (MIC = 50 µg/mL for C. jejuni and 200 µg/mL for C. coli).[107]
Imunović K et al., 2019C. jejuni(-)-α-pineneBroth microdilution
Quorum-sensing inhibition in vitro
Broiler chicken colonization with C. jejuni broiler
By itself, (-)-α-pinene showed low antimicrobial activity (MIC50 = 2000 mg/L); however, when in combinations, (-)-α-pinene was able to reduce ciprofloxacin’s and erythromycin’s MICs. When treating cultures with three subinhibitory concentrations of (-)-α-pinene, a reduction in quorum-sensing signalling molecules is observed. However, (-)-α-pinene did not manage to impede fluoroquinolone resistance development when added to enrofloxacin in broiler chickens.[114]
Lin L et al., 2018C. jejuniThymus vulgaris L. (thyme)Plate count
Measurement of inhibition zone
Sodium dodecyl sulphate-polyacrylamide gel electrophoresis (SDS-PAGE)
Transmission electron microscopy (TEM)
The population of C. jejuni was lower in the group treated with thyme EO β-cyclodextrin ε-polylysine nanoparticles (TCPNs) than in control group, both in chicken soup (difference = 2.74 log CFU/mL, inhibition zone = 25.10 mm) and commercially available chicken meat (difference = 1.38 log CFU/g). Due to the encapsulation and presence of ε-polylysine, the prepared TCPNs exhibited better and prolonged anti-C. jejuni activity than thyme EO/β-cyclodextin inclusion complex and free thyme EO. Damage in cell membrane and protein leakage in C. jejuni treated with TCPNs were emphasized by TEM and SDS-PAGE analysis.[112]
Duarte A et al., 2016C. jejuni ATCC 33560 and 225421,
C. coli ATCC 33559 and 873
Coriandrum sativum L. (coriander), linaloolBroth microdilution
Anti-biofilm activity
Disc diffusion assay
Vapour diffusion assay
Broth assay
Evaluation of the antioxidant activity
Anti-quorum-sensing activity
Both EO and linalool showed anti-Campylobacter activity (diameter of inhibition > 85 mm, MIC = 0.5–1 µL/mL), inhibited biofilm formation and produced quorum-sensing inhibition.[108]
Kovács J et al., 2016C. jejuni NCTC 11168Syzygium aromaticum L. (clove), eugenol, beta-caryophyllene and alpha-humuleneBroth microdilution
Time kill assay
S. aromaticum EO (MIC = 200 µg/mL and MBC = 800 µg/mL) and eugenol showed antimicrobial properties and altered C. jejuni morphology, whereas beta-caryophyllene and alpha-humulene had no such effects.[109]
Ahmed J et al., 2016C. jejuni ATCC 33291
S. aureus ATCC 6538
Cinnamomum cassia Presl (cinnamon),
Allium sativum L. (garlic) and
Syzygium aromaticum L. (clove) EOs
Agar disc diffusionZone of inhibition showed that both S. aureus and C. jejuni were sensitive to clove and cinnamon EOs, while garlic EO was found to be less effective. Same outcome resulted from survivor counts (CFU/mL).[110]
El Baaboua A et al., 2022MDR Campylobacter spp.Origanum compactum Benth (oregano),
Rosmarinus officinalis L. (rosemary),
Mentha pulegium L. (mint) and
Lavandula stoechas L. (lavender)
Agar well diffusion
Broth microdilution
Biofilm detection
Diameters of inhibition showed that O. compactum and L. stoechas EOs had high anti-campylobacterial effect (diameter = 15 -> 80 mm and 24 -> 80 mm, respectively). High sensitivity of Campylobacter spp. toward O. compactum, M. pulegium and L. stoechas was also sustained by MIC, with the lowest value recorded of 0.063% (v/v).[111]
Lin L et al., 2019C. jejuni CICC 22,936ChrysanthemumTransmission electron microscopy (TEM)The C. jejuni population of treated samples decreased from 3.2 log CFU/mL to 0 log CFU/mL at 12 °C for 12 days. However, at 25 °C and 37 °C the population of C. jejuni was around 1.2 log CFU/mL to 2.3 log CFU/mL.[113]
Table A5. Studies assessing the antimicrobial activity of essential oils against cephalosporin-resistant, fluoroquinolone-resistant Neisseria gonorrhoeae.
Table A5. Studies assessing the antimicrobial activity of essential oils against cephalosporin-resistant, fluoroquinolone-resistant Neisseria gonorrhoeae.
Study Team and YearBacterial StrainEssential Oil(s)Method(s)Results Reference
Vică M et al., 2021Ciprofloxacin-sensitive N. gonorrhoeae,
Ciprofloxacin-intermediate-resistant N. gonorrhoeae,
Ciprofloxacin-resistant N. gonorrhoeae,
Aqueous extracts of propolis samplesDisc diffusion
Broth microdilution
The propolis extracts possessed anti-N. gonorrhoeae activity. The mean diameter of the inhibition zones for propolis extracts was 39.75 mm (27–42 mm), in some cases exceeding ciprofloxacin’s zone of inhibition. For most extracts, MIC was 6.25 µg/mL, while some propolis samples exhibited MIC of 12.5 µg/mL or 25.0 µg/mL.[130]
Umaru I et al., 2020MDR S. aureus
MDR N. gonorrhoea
Molineria capitulata Lour., myrceneDisc diffusionAt 500 µg/mL, M. capitulata EO exhibited ZOI = 23.28 ± 0.13 mm on S. aureus and ZOI = 22.3 ± 0.37 mm on N. gonorrhoeae. Myrcene showed similar results, with ZOI = 23.53 ± 0.13 mm on S. aureus and ZOI = 20.50 ± 0.17 mm on N. gonorrhoeae.[131]
Soliman F et al., 2016Neisseria gonorrhoeae 19424,
Staphylococcus aureus ATCC 12600
Psidium guajava L. (guava leaf), Psidium cattleianum Sabine (strawberry guava)Agar disc diffusion
Agar dilution
S. aureus: ZOI = 16 ± 0.15 mm (P. guajava) and 10 ± 0.28 mm (P. cattleianum); MIC = 6.75 µg/mL (P. guajva).
N. gonorrhoeae: ZOI = 12 ± 0.22 mm (P. guajava) and 13 ± 0.29 mm (P. cattleianum); MIC = 13.01 µg/mL (P. cattleianum).
[132]
Zaman G et al., 2021N. gonorrhoeae,
S. aureus
Eclipta alba L. (kenharaj),
Atriplex hortensis L. (pahari palang),
Hedyotis scandens (Roxb.) (bhedeli-lota),
Leucas linifolia Spreng (doron bon),
Murraya koenigii L.(narasingha),
Phlogacanthus thyrsiflorus Nees (tita-phul)
Disc diffusion
Broth microdilution
The antimicrobial activity varied as follows: E. alba (for N. gonorrhoeae: ZOI = 22.60 ± 2.50 mm, MIC = 1.50 ± 0.20 μg/mL, MBC = 3.06 ± 0.40 μg/mL; for S. aureus: ZOI = 17.20 ± 1.47 mm, MIC = 0.05 ± 0.02 μg/mL, MBC = 0.11 ± 0.03 μg/mL); A. hortensis (for N. gonorrhoeae: MIC = 6.0 ± 0.26 μg/mL, MBC = 12.07 ± 0.65 μg/mL; for S. aureus: ZOI = 12.90 ± 2.55 mm, MIC = 3.53 ± 0.35 μg/mL, MBC = 8.73 ± 2.62); H. scandens (for N. gonorrhoeae: ZOI = 12.60 ± 1.43 mm, MIC = 7.50 ± 0.36 μg/mL, MBC = 15.30 ± 0.43 μg/mL; for S. aureus: ZOI = 20.50 ± 3.10 mm, MIC = 6.66 ± 0.40 μg/mL, MBC = 13.0 ± 0.65); L. linifolia (for N. gonorrhoeae: ZOI = 18.20 ± 1.22 mm, MIC = 3.50 ± 0.20 μg/mL, MBC = 7.06 ± 0.40 μg/mL; for S. aureus: ZOI = 8.0 ± 0.81 mm, MIC = 4.33 ± 0.25 μg/mL, MBC = 8.73 ± 0.61); M. koenigii (for N. gonorrhoeae: ZOI = 15.50 ± 1.08 mm, MIC = 5.46 ± 0.20 μg/mL, MBC = 11.07 ± 0.37 μg/mL; for S. aureus: ZOI = 7.40 ± 1.17 mm, MIC = 6.10 ± 0.43 μg/mL, MBC = 12.50 ± 0.79); and P. thyrsiflorus (for N. gonorrhoeae: ZOI = 18.40 ± 0.96 mm, MIC = 5.10 ± 0.17 μg/mL, MBC = 10.20 ± 0.34 μg/mL; for S. aureus: ZOI = 18.30 ± 0.82 mm, MIC = 4.70 ± 0.20 μg/mL, MBC = 9.40 ± 0.40)[133]
Elghwaji W et al., 2017N. gonorrhoeae ATCC 19424,
S. aureus ATCC 12600
Ferula tingitana L.Agar disc diffusion
Evaluation of cytotoxic activity
N. gonorrhoeae: ZOI = 12 mm (F. tingitana flower-derived EO), ZOI = 13 mm (F. tingitana leaves-derived EO); S. aureus: ZOI = 9 mm (F. tingitana flower-derived EO), ZOI = 11 mm (F. tingitana leaves-derived EO). IC50 for F. tingitana flower-derived EO = 6.9 µg/mL, 8.6 µg/mL and 4.4 µg/mL; for F. tingitana leaves-derived EO = 4.8 µg/mL, 4.2 µg/mL and 10.9 µg/mL, on breast, liver and cervical carcinoma cell lines. The results were comparable to the standard cytotoxic drug doxorubicin.[134]
Table A6. Studies assessing the antimicrobial activity of essential oils against methicillin resistant, vancomycin-intermediate and -resistant S. aureus (a non-exhaustive list).
Table A6. Studies assessing the antimicrobial activity of essential oils against methicillin resistant, vancomycin-intermediate and -resistant S. aureus (a non-exhaustive list).
Study Team and YearBacterial StrainEssential OilsMethod(s)Results Reference
Ben Abdallah F et al., 2020MRSAOriganum majorana L. (origanum), Rosmarinus officinalis L. (rosemary) and Thymys zygis L. (common thyme)Disc diffusion
MIC and MBC methods
Crystal violet
Potent antimicrobial effect shown by all tested EOs, T. zygis having the most powerful effect (MIC = 0.39 mg/mL–0.78 mg/mL; MBC = 3.125 mg/mL), followed by O. majorana (MIC = 0.78 mg/mL–1.56 mg/mL; MBC = 3.125 mg/mL–12.5 mg/mL) and R. officinalis (MIC = 0.78 mg/mL–1.56 mg/mL; MBC = 12.5 mg/mL). Regarding the biofilm inhibition and eradication effect, O. majorana EO presented the most potent activity (inhibition effect: from 10.29 to 95.91% and eradication effect: from 18.31 to 98.01%).[190]
Gomez-Sequeda N et al., 2020MRSA
E coli O157:H7
Lippia origanoides KunthBroth microdilution
Cytotoxicity assay
Scanning electron microscopy
Thymol and carvacrol chemotypes from Lippia origanoides exhibited the best antimicrobial action for MRSA (MIC = 1.2 and 0.6 mg/mL) and for E. coli O157:H7 (MIC = 0.9 and 0.3 mg/mL), as well as the best antibiofilm inhibition (>70.3%), but the analysis performed on the selectivity index (≤3) highlighted the fact that further studies are required in order to reduce its in vitro toxicity.[191]
Ekhtelat M. et al., 2020MRSA
Yersinia enterocolitica
Cuminum cyminum L. (cumin)
Mentha spicata L. (spearmint)
Mentha longifolia L. (horse mint)
Single or in combination with sodium benzoate
Agar disc diffusion
Micro-well dilution assay
M. longifolia L. (main component: pulegone) and C. cyminum L. (main component: aldehyde) presented the best antibacterial effect against the tested strains. The association with sodium benzoate exhibited a more potent antibacterial effect, compared with the use of sodium benzoate alone as antimicrobial agent, and therefore can reduce the high doses required if used alone for preservation of food or drug products, and thus its toxicity.[192]
Tang C. et al., 2020MRSAAmomum villosum LourLabel-free quantitative proteomics
Sodium dodecyl sulphate-polyacrylamide gel electrophoresis analysis
Scanning electron microscopy
The antibacterial effect expressed by the tested EO demonstrated its mechanism of action: cell membrane lesion which affects its integrity, intracellular leakage of substances, protein inhibition and biofilm synthesis inhibition.[193]
Rubini D. et al., 2018MRSAPogostemon heyneanus Benth.
Cinnamomum tamala Nees and Eberm (Indian bay leaf)
Confocal laser scanning microscopy
Scanning electron microscopy
Both EOs affected the MRSA preformed biofilms and were successful in reducing virulence factors such as staphyloxanthin and hemolysin, with (E)-nerolidol having a higher affinity for dehydroxysqualene synthase (responsible for the synthesis of staphyloxanthin).[194]
Utegenova GA et al., 2019MRSAFerula ovina (Boiss.) Boiss,
Ferula iliensis Krasn. ex. Korovin,
Ferula akitschkensis B. Fedtsch. Ex Koso-Pol.
Pulse-field gel electrophoresis
Colony count technique
Broth microdilution
F. ovina EOs (from roots and stems at fruiting stages) expressed the most powerful antibacterial activity dependent of the concentration, with IC50 = 19.1, 20.9 and 22.9 mcg/mL. The main components with antibacterial activity being eremophilene and trans-verbenol (single or associated), although they were not the major constituents of the EOs.[195]
Oo T et al., 2021MRSAMyristica fragrans Houtt. (nutmeg) crude extract and EODisc diffusion
PCR
Checkerboard titration assay
The association of nutmeg preparations with ciprofloxacin led to synergistic action on efflux pump system (chromosomal norA and mepA) in MRSA, mepA being incriminated for the efflux pump inhibition of EO.[169]
Sreepian A et al., 2022MRSA
MSSA
Citrus reticulata Blanco (mandarin orange)
Citrus × aurantiifolia (Christm.) Swingle (key lime)
alone and in combination with gentamicin
Agar disc diffusion
Resazurin-based microdilution
Checkerboard titration assay
Both EOs manifested inhibitory effects against the tested strains. The most potent antimicrobial activity was observed for C. reticulata EO and limonene (the major compound of both EOs). Synergistic effect with gentamicin was observed for C. reticulata EO (FIC index = 0.012–0.258) and limonene (FIC index = 0.012–0.375) on both tested strains.[196]
Dalli M et al., 2021MRSA
E. coli
P. aeruginosa
A. baumannii
Nigella sativa L. (black caraway/cumin)Agar diffusion method
Microdilution method
The tested EOs, although from 4 different countries (India, Saudi Arabia, Morocco, Syria), had similar compositions and were slightly more potent against MRSA compared with the Gram-negative bacteria tested (MIC/MBC = 3–10 µg/mL).[197]
Alharbi NS et al., 2016MRSA and oxacillin-resistant S. aureusPiper cubeba L. f. (tailed pepper)Atomic force microscopy
Transmission electron microscopy
Microdilution assay
Severe damage of S. aureus ATCC 43300 cells was observed at microscopic levels with 50 mcg/mL EO when compared to 25 mcg/mL, although the latter produced important modification within the cell wall, at nanoscopic levels. Therefore, both EOs exhibited antibacterial effects on the cell wall, as well as on the plasma membrane.[170]
Tang C et al., 2021MRSAAmomum villosum LourMetabolomics analysisThe tested EO induced an antibacterial effect by blocking the amino acid metabolism and tricarboxylic acid cycle (the activity of key enzymes was inhibited) of MRSA. Moreover, it inhibited the synthesis of reactive oxygen species and adenosine triphosphate, leading to bacterial cell death.[198]
Piasecki B et al., 2021MRSACymbopogon spp.Microdilution
Direct bioautography
Danio rerio “Zebrafish” model assay
Broth microdilution assay
C. flexuosus (lemongrass EO) expressed the highest antibacterial activity from all the 19 tested EOs of Cymbopogon spp. (MIC/MBC = 0.5 mg/mL).
Citronellol showed powerful antibacterial activity (MIC/MBC = 0.25 mg/mL).
Antibiofilm effects were also observed for all tested EOs (MBIC = 1 mg/mL–4 mg/mL). C. martini var. motia expressed the most toxic potential (about 20 times more toxic than C. winterianus) on zebrafish model assay.
[171]
Iseppi R et al., 2021MRSA
vancomycin-resistant enterococci
ESBL-producing E. coli
Citrus × aurantium L. (bitter orange)
Citrus x limon L. (lemon)
Eucalyptus globulus Labill. (blue gum)
Melaleuca alternifolia (Maiden and Betche) Cheel (tea tree)
Cupressus sempervirens L. (Italian cypress)
Agar disc diffusion
MIC assay
Checkerboard method
Tea tree oil (M. alternifolia) was the most effective EO, although all tested EOs presented antibacterial activity. Synergistic action was observed when EOs were associated with other EOs or with classical ABs. Good antibiofilm activity was observed when the EOs were used in monotherapy or in combination[64]
Merghni A et al., 2018MRSAEucalyptus globulus Labill. (blue gum)
and 1,8-cineole
Test tube method
Crystal violet staining assay
Disc diffusion test
MIC/MBC methods
E. globulus, as well as its main component: 1,8 cineole, presented high antibiofilm activity, with E. globulus EO having a better anti-quorum-sensing potential (even at low concentrations) (MBC < 50 mg/mL) than 1,8-cineole alone (MBC > 50 mg/mL).[172]
Al-Maharik N et al., 2021MRSA
S. aureus ATCC 25923
E. faecium ATCC 700221
K. pneumoniae ATCC 13883
Proteus vulgaris ATCC 700221
E. coli ATCC 25922
P. aeruginosa ATCC 27853
C. albicans ATCC 90028
Satureja nabateorum (Danin and Hedge) BräuchlerBroth microdilution assay
Cell culture cytotoxicity assay
Both the fresh and the air-dried EOs of S. nabateorum presented good and similar antimicrobial and fungicidal activity. EO obtained from the air-dried sample manifested a higher antimicrobial activity against MRSA (MIC = 6.25 µg/mL) than ciprofloxacin (MIC = 12.5 µg/mL). Both EOs showed cytotoxic activity against HeLa and HepG2 cancer cells and were proposed as potential alternatives to bactericides and fungicides of chemical origin, as well as natural preservatives and conservation substances.[199]
Khamis AS et al., 2021MRSA ATCC 33591
E. coli NCTC 10418
P. aeruginosa NCTC 10662
Bacillus subtilis ATCC 6059
Micrococcus luteus ATCC 9341
S. aureus NCTC 6571
Juniperus chinensis L. (Chinese juniper)
Juniperus seravschanica Kom. (Pashtun juniper) versus their methanolic crude extracts
Disc diffusionAlthough all EOs tested showed antimicrobial activity, increased activity was observed against M. luteus and B. subtilis. Only the crude methanolic extracts manifested an increased antibacterial activity against MRSA and S. aureus. A higher antimicrobial activity was observed for methanolic crude extract of J. seravschanica (from Oman) against MRSA.[173]
Predoi D et al., 2018MRSA
S. aureus ATCC 0364
E. coli ATCC 25922
Hydroxyapatite coated with basil (Hap-B)
Hydroxyapatite coated with lavender (HAp-L) EOs
Fourier transform infrared spectroscopy
Broth microdilution
HAp-L was found to exhibit the highest inhibitory growth activity against the tested strains, with MIC = 0.039 mg/mL for MRSA and E. coli ATCC 25922, and 0.02 mg/mL for S. aureus.[174]
Mouwakeh A et al., 2019MRSA
MSSA
Nigella sativa L. (black caraway)
Thymoquinone
Carvacrol
p-cymene
Broth microdilution
Ethidium bromide accumulation assay
Real-time reverse transcriptase quantitative PCR
Crystal violet assay
The tested strains were susceptible to N. sativa EO, carvacrol and thymoquinone (but not p-cymene), and each of the tested compound affected the membrane integrity of MRSA (including p-cymene). P-cymene was found to down-regulate the expression of EP gene mepA in MRSA, therefore decreasing its virulence.[177]
Donadu MG et al., 2020MRSA
MSSA
S. epidermidis
E. faecalis
Candida tropicalis
Candida albicans
Candida glabrata
Candida epidermidis
C. parapsilosis
E. coli
P. aeruginosa
K. pneumoniae
T. vaginalis strain G3
Enterovirus—A71 strain
Hornstedtia bella SkornickBroth microdilution
Cell and cytotoxic assay
MIC/MLC = 1–4% v/v for MRSA, MSSA, S. epidermidis (β-Pinene, E-β-caryophyllene and α-humulene); MIC/MLC = 2–16% v/v for C. tropicalis and C. parapsilosis; MIC/MLC = 4–16% v/v for E. faecalis; and MIC/MLC = 8–16% v/v for the remaining tested strains. Low cytotoxicity against Vero 76 and MRC-5 for leaf oil, without any toxic effect for rhizomes and whole-plant oils on the cells, as well as no action against enterovirus (EV-A71).[200]
Bay M et al., 2019MRSA
E. coli
P. aeruginosa
S. mutans
S. pyogenes
Trypanosoma cruzi
Bocageopsis multiflora R.E.Fr.,
Duguetia quitarensis Benth.,
Fusaea longifolia (Aubl.) Saff.,
Guatteria punctata (Aubl.) RAHoward
Broth microdilution
Trypanocidal activity assays
Cytotoxicity assays
Potent antimicrobial activity manifested by the tested EOs against S. mutans (MIC = 4.68–37.5 µg/mL. G. punctata EO was the most effective against T. cruzi, therefore having a good trypanocidal activity, 34 times more active than the reference drug benznidazole (IC50 = 0.029 µg/mL, SI = 32)[201]
Aelenei P et al., 2019MRSA
MSSA
S. epidermidis
P. aeruginosa
E. coli
Coriandrum sativum L. (coriander) and linalool, both associated with ABsBroth microdilution
Checkerboard assay
Synergistic interactions were observed between coriander EO and its major component, linalool, with ABs such as amoxicillin, oxacillin, gentamicin, tetracycline and ciprofloxacin, their MICs being drastically reduced.
FICI ≤ 0.5 was obtained for coriander EO + gentamicin or amoxicillin against MRSA.
[202]
Leal ALAB et al., 2021S. aureus SA1199B (overexpressing norA gene)
S. aureus K2068 (overexpressing mepA gene)
S. aureus K4100 (overexpressing qacC gene)
C. albicans ATCC 10231
E. coli ATCC 25922
S. aureus ATCC 25923
Piper caldense C.DC. alone or in combination with norfloxacinMicrodilutionAntimicrobial effect against S. aureus strains was observed only in association with norfloxacin, effect observed at subinhibitory concentrations. In contrast, the EO was active against C. albicans, inhibiting an important mechanism of virulence of the fungi, the hypae formation, therefore having a good antifungal activity. The EO was able to act as efflux pump inhibitor on norA, mepA and qacC.[203]
Predoi D. et al., 2018MRSA 1144
S. aureus 1426
ESBL E. coli 4493
E. coli ATCC 25922
Ocimum basilicum L. (basil)
Lavandula augustifolia Mill (lavender)

(linalool being the major compound in both EOs)
Broth microdilution
Flow cytometric assay
Lavender EO expressed a good antibacterial action (MIC < 0.1% mg/mL for E. coli strains and up to 0.78% mg/mL for S. aureus strains; MBC < 0.1% mg/mL up to 1.56% mg/mL). The hydroxyapatite solution with lavender EO expressed an increased antibacterial activity (MIC = 0.31 mg/mL; MBC = 0.62 mg/mL for MRSA 1144), making hydroxyapatite a possible vehicle for lavender EO solutions in low concentrations.[175]
de Jesus GS et al., 2020S. aureus NEWP0023
E. coli NEWP 0022
MRSA mecA mediated
S. warneri β-lactamase producer
S. intermedius mecA mediated methicillin resistance
Pectis substriata Rusby
alone or in combinations with ABs
Broth microdilution
Checkerboard microtiter test
The EO exhibited antibacterial activity depending on the strains with potent antimicrobial activity against S. warne-ri and moderate activity against S. aureus standard strain and S. intermedius. Synergistic actions were observed when associated with ampicillin and kanamycin.[204]
Cui ZH et al., 2021MRSA 43300
E. coli ATCC 25922
S. typhimurium ATCC 14028
K. pneumoniae ATCC 700603
29 plant EOsModified well diffusion
Checkerboard assay
Standard time-killing assay
Orange oil + amikacin and petitgrain oil + tetracycline exhibited synergistic actions against S. aureus, S. typhimurium and K. pneumoniae. The same action was observed for petitgrain EO + tetracycline against E. coli tested strain.[205]
de Moura DF et al., 2021MRSA—clinical isolate
MSSA—clinical isolate
P. aeruginosa—clinical isolate
E. faecalis ATCC 14 506
E. coli ATCC 25 922
K. pneumoniae ATCC 29 665
P. aeruginosa ATCC 9029
S. aureus ATCC 6538
S. epidermidis ATCC 12 228
S. mutans ATCC 10 499
NerolidolMicrodilution
Crystal violet method
Nerolidol was effective against MRSA (MIC = 2 mg/mL), P. aeruginosa and K. pneumoniae carbapenemase (MIC = 0.5 mg/mL). It showed dose-dependent antioxidant, antibacterial and antibiofilm activity (the percentage of inhibition being 51–98% at concentrations varying from 0.5 to 4 mg/mL).[178]
Jaradat N et al., 2021MRSA
S. aureus ATCC 25923
C. albicans ATCC 90028
P. aeruginosa ATCC 9027
E. coli ATCC 25922
K. pneumonia ATCC 13883
P. vulgaris ATCC 8427
Aloysia citriodora Palau (lemon verbena)Broth microdilution assayTwo EOs were obtained from lemon verbena (Umm al-Fahm and Bawa al-Gharbiyye). α-Citral (geranial) was determined to be their main component. Potent antimicrobial activity was observed against MRSA and P. vulgaris (MIC = 2.5 µg/mL), using ciprofloxacin and ampicillin as reference. Good antifungal activity was noticed against Candida albicans (MIC = 0.312–0.625 µg/mL), where fluconazole was used as reference. The cytotoxicity was tested on HeLa tumour cells versus doxorubicin: at a concentration of 500 µg/mL, the viability was 98.13 and 96.09% for the two EOs. The EO from Baqa al-Gharbiyye highlighted a more potent cytotoxic activity (IC50 = 84.5 ± 0.24 µg/mL) vs. doxorubicin (IC50 = 22.01 ± 1.4 µg/mL), as well as a stronger inhibition activity against COX-1 and COX-2 enzymes (IC50 = 52.93 ± 0.13; 89.31 ± 0.21 µg/mL).[181]
Fahed L. et al., 2016MRSA ATCC 33591
S. aureus ATCC 29213
C. albicans ATCC 10231
P. aeruginosa CIP 82118
T. rubrum SNB-TR1
Tricophyton mentagrophytes SNB-TM1
Tricophyton violaceum SNB-TV1
Tricophyton tonsurans SNB-TT
Salvia multicaulis VahlBroth microdilution assayZahlé EO MIC = 128 µg/mL for both S. aureus strains. Good antimicrobial activity was observed for nerolidol, the major active compound (MIC = 128 µg/mL for S. aureus strains and 64 µg/mL for Tricophyton rubrum).[206]
Mahdavi B et al., 2017MRSA
S. aureus ATCC 25923
B. thuringiensis TACC 10792
B. subtilis ATCC 11774
S. epidermidis ATCC 1228
E. faecalis ATCC 14506
A. hydrophilia ATCC 7966
E. coli ATCC 10536
E. aerogenes ATCC 13048
P. mirabilis ATCC 12453
P. vulgaris ATCC 33420
S. typhimurium ATCC 51812
S. sonnei ATCC 29930
S. marcescens ATCC 13880
V. parahaemolyticus ATCC 17802
P. aeruginosa ATCC 10145
C. albicans ATCC 90028
C. parapsilosis ATCC 22019
Etlingera sayapensis A.D. Poulsen and IbrahimDisc diffusion assay
MIC method
The EO obtained from the rhizome of E. sayapensis presented potent antimicrobial activity against 13 of 18 tested strains, including MRSA, S. aureus, E. coli, P. mirabilis, B. subtilis, C. albicans.[207]
Gadisa E et al., 2019MRSA
E. coli—MDR strain
K. pneumoniae—MDR strain
S. aureus ATCC 25922
E. coli ATCC 25922
K. pneumoniae ATCC 700603
Blepharis cuspidata Qoree waraantii
Boswellia ogadensis Vollesen
Thymus schimper
used in combination
Broth microdilution assay
FIC index
The combination of B. cuspidata and T. schimperi EOs highlighted a MIC/MBC = 0.39 µg/mL against MRSA. Moreover, association of B. cuspidata and B. ogadensis had a MIC of 1.56 µg/mL for MRSA. A FICI of 0.38 (a synergistic effect) was obtained for MRSA using B. cuspidata + B. orgadensis, and of 0.25 for K. pneumoniae MDR, for the same association.[208]
Bano S et al., 2020MRSA
E. coli
P. aeruginosa—total drug resistant
K. pneumoniae
Terminalia arjuna (Roxb.) Wight and Arn.Not mentionedMIC = 0.32 mg/mL for leaf oil and 0.16 mg/mL for fruit oil (against MRSA).[209]
Ding L. et al., 2020MRSA 134/94 R9
S. aureus SG 511
P. aeruginosa K799/61
BG137 B7
B. subtilis ATCC 6633
Mycobacterium vaccae IMET 10670
Vancomycin-resistant E. faecalis 1528 R10
E. coli SG458
Sporobolomyces salmonicolor SBUG 549
C albicans
Penicillium notatum JP36
Geosmin compounds
Compound 6: 4β,10α-eudesmane-5β,11-diol EO
Agar diffusion assayThe 4β,10α-eudesmane-5β,11-diol (compound 6) was reported as the active substance in EO from the aromatic grass Cymbopogon distans (an oil used in folk medicine to treat microbial infections, inflammations and colds or to protect books from insects and fungi, in ancient China). Compound 6 exhibited large antimicrobial activity against all tested strains (inhibition zone for MRSA and P. aeruginosa = 11 mm).[210]
Jaradat N et al., 2017MRSA
S. aureus ATCC 25923
E. coli ATCC 25922
P. aeruginosa ATCC 27853
C. albicans
Ruta chalepensis L. (fringed rue)Broth microdilution assay
MIC assay
Linalyl acetate and β-linalol were determine to be the major active compounds having good antibacterial and antioxidant activities. The best antimicrobial activity was obtained for E. coli, P. aeruginosa, S. aureus and MRSA (MIC for R. chalepensis from Jerusalem = 4 mg/mL vs. gentamycin 1.5 mg/mL).[211]
Man A et al., 2019MSSA ATCC 29213,
MRSA ATCC 43300
E. faecalis ATCC 29212
E. coli ATCC 25922
K. pneumoniae ATCC 13883
P. aeruginosa ATCC 27853
Micellar and aqueous extracts of EOs:
Boswellia sacra Flueck. resin (Frankincense)
Myrtus communis L. (common myrtle)
Thymus vulgaris L. (common thyme)
Citrus limon L.
Origanum vulgare Linnaeus (oregano)
Lavandula augustifolia Mill. (English lavender)
MIC and MBC determination methodsGood antibacterial activity was observed for oregano, thyme, lemon and lavender EOs, with Gram-positive cocci including MRSA being the most susceptible bacteria and P. aeruginosa being the most resistant. The best antibacterial activity was observed for oregano EO (decreased MIC/MBC ratio up to 64 times). It was observed that colloid or micelle suspensions of EOs (oregano, thyme and lemon whole EOs) may be more efficient than antimicrobial agents (in the case of MRSA).[41]
Kot B et al., 2017MRSAThyme oil
trans-cinnamaldehyde
Ferulic acid
p-coumaric acid
caffeic acid
lavender oil
geranium oil
tea tree oil
Not mentionedAfter 48 h of treatment, thyme oil decreased the biofilm mass by up to 85% and the metabolic activity of biofilms by up to 88.7%.[212]
Kwiatkowski P et al., 2020MRSA ATCC 433001,8-cineole
eugenol
carvacrol
linalool
linalyl acetate
trans-anethole
thymol
menthone
menthol
β-caryophyllene
Antibiotic susceptibility testing: Kirby–Bauer method and D-test
mecA gene detection
Broth microdilution
Checkerboard method
FTIR spectroscopic analyses
Good anti-MRSA activity for 8 of the 10 tested EOs was observed. Thymol expressed the most powerful inhibitory activity against the tested strain.
Linalyl acetate alone or especially in combination with methicillin was effective against low- and high-level beta-lactam resistant MRSA strains, effect attributed to the 2 methoxy moieties present in its molecule. Further in vivo studies are required to assess cytotoxicity and establish the safe concentration of linalyl acetate-methicillin.
[213]
Kwiatkowski P et al., 2019MRSA
(S. aureus ATCC 43300 mutated strains)
1,8-cineole
eugenol
carvacrol
linalool
(-)-menthone
linalyl acetate
trans-anethole versus mupirocin-susceptible (MupS) and low-level mupiocin-resistant (MupRL) MRSA
Broth microdilution
Checkerboard method
Both strains of MRSA showed sensibility to the tested EOs, with carvacrol expressing the highest inhibitory activity (MIC = 0.48–0.95 mg/mL). 1,8-cineole expressed synergistic activity with penicillin G against MupRL MRSA, therefore increasing its activity against both tested strains (MupS and MupRL MRSA).[184]
Cui H et al., 2016MRSA ATCC 43300Cinnamon oil encapsulated in liposomesBroth microdilution
Time-kill assay
MBIC and MBEC assays
MTT staining method
Biofilm CFU counting
Plate colony-counting method
Potent antibacterial and antibiofilm action was observed for the tested EO against MRSA, after at least 4h treatment (MIC/MBC = 0.25 mg/mL; MBIC/MBEC = 1.00 mg/mL). The bactericidal effect was highlighted by the irreversible destruction of MRSA cell membrane (rough, wrinkled and irregular). A potent antibiofilm activity was observed at 1.0 mg/mL cinnamon oil. Anti-biofilm properties of liposomes containing cinnamon oil highlighted a gradual and consecutive decrease of viable MRSA cells, depending on the conditions (stainless steel, gauze, nylon membrane, exposure time).[214]
Perez AP et al., 2019MRSA isolates (Cordobes clone, DOS61, DOS90 and DOS59)Thymus vulgaris L. (common thyme) nanovesicular formulationMTT assayThe antibiofilm effect was observed for nanoarchaeosomes (MIC = 4 mg/mL), this nanovesicular formulation demonstrated good stability during storage. Moreover, the macrophages (J774A.1) viability was diminished after 24 h of incubation for both formulations (including nanoliposomes) at 0.4 mg/mL.[215]
Farias KS et al., 2019MRSA ATCC 43300
P. aeruginosa ATCC 27853
T. vaginalis ATCC 30236
S. epidermidis ATCC 35984
K. pneumoniae clinical isolate
Nectandra amazonum
Nectrandra cuspidate
Nectandra gardineri
Nectranda hihua
Nectandra megapotamica
Crystal violet assay
Anti-Trichomonas vaginalis assay
(+)-α-bisabolol (found in a concentration of 93.7% in N. megapotamica leaves) presented a potent antibiofilm effect against MRSA (10 µg/mL) and P. aeruginosa (100 µg/mL). Good anti-trichomoniasis activity was found (IC50 = 98.7 µg/mL), together with cytotoxic and haemolytic actions in Vero cells and human erythrocytes.
[216]
Eid AM et al., 2021MRSA
P. aeruginosa ATCC 9027
K. pneumoniae ATCC 13883
E. coli ATCC 25922
S. aureus ATCC 25923
(P. mirabilis?)
C. albicans ATCC 90028
Coriandrum sativum L. (coriander) nanoemulgelAgar diffusionGood antibacterial and anticancer effects observed for the nanoemulgel, compared with the crude coriander oil. (MRSA MIC = 6.5 µg/mL; IC50 = 28.84 µg/mL for MCF-7 (human breast cancer cells), 24.54 µg/mL for HeLa (human cervical epithelioid carcinoma cells); and 28.18 µg/mL for Hep3B (hepatocellular carcinoma cells))[217]
Bako C et al., 2021MRSA 4262
P. aeruginosa ATCC 27853
Salvia sclarea L. (clary sage)Brain–heart infusion broth
Thin layer chromatography without separation
Post-chromatographic detection
Clary sage exhibited a 7.57 mm inhibition zone for MRSA and 7.51 mm for P. aeruginosa.[218]
Bushra Jamil et al., 2016MRSA
β-lactamase producing E. coli
Elettaria cardamomum (L.) Maton (green cardamom) oil chitosan nano-particlesKirby–Bauer disc diffusion
Agar well diffusion
Broth dilution
Non-cytotoxic (on human corneal epithelial cells) and non-haemolytic effects were observed for the cardamom oil loaded in chitosan nano-particles, as well as potent antibacterial properties against the tested strains.[219]
Khoury M et al., 2019MRSA ATCC 33591
E. coli ATCC 25922
S. aureus ATCC 29213
C. albicans ATCC 10231
C. parapsilosis ATCC 22019
Cryptococcus neoformans ATCC SNB-CN1
Trichophyton rubrum SNB-TR1
T. violaceum
T. soudanense SNB-TS1
T. tonsurans SNB-TT1
T. mentagrophytes SNB-AF1
Aspergillus fumigatus ATCC SNB-AF1
Hirtellina lobelia DC.Broth microdilution method
Checkerboard assay
Good bactericidal effects against S. aureus strains, including MRSA (MIC/MBC = 128 µg/mL). Synergism was observed in association with vancomycin (against S. aureus). The main compound, α-bisabolol, was found to have potent antimicrobial potential (including anti-fungal).[220]
Viktorova J et al., 2020MRSA DBM 12
S. aureus ATCC 25923
Salmonella enterica CCM 4420
Proteus vulgaris DBM 3022
Mycobacterium smegmatis ATCC 70084
P. aeruginosa CCM 3955
C. albicans DBM 2186
C. famata DBM 23
Cryptococcus albidus DBM 4
Cymbopogon citratus (DC.) Stapf (lemon grass)Broth microdilution
Autoinducer bioassay
Pgp-Glo assay
Static antibiofilm assay
Resazurin assay
Mature biofilm assay
Citral (the main component of lemongrass EO—63%) was almost 100 times more active than lemongrass EO. Both disrupted bacterial communication and adhesion during biofilm formation (in S. aureus and P. aeruginosa), with citral having the highest effect (citral IC50 was 7–70 times lower that lemongrass IC50). Citral had 5 times more potent antibiofilm activity on P. aeruginosa than on MRSA. Lemongrass EO (and not citral alone) induced a sensitizing action on MRSA and on ovarian carcinoma cells resistant to doxorubicin, probably through the inhibition of P-glycoprotein efflux pump.
[221]
Manzuoerh R et al., 2019MRSA clinical isolate
Methicillin-sensitive S. aureus ATCC 25923
Methicillin-resistant S. aureus ATCC 33591
Anethum graveolens L. (dill)Broth nutrition
Wound model
Dill EO application on wounds inhibited bacterial growth and diminished wound area compared to the control group. It reduced the inflammatory process (by decreasing p53 and caspase-3 expression) and enhanced re-epithelization, angiogenesis, collagen and fibroblast deposition after topical administration. The expression of Bcl-2, p53 caspase-3, VEGF and FGF-2 was increased in the group treated with the EO.[185]
Taha AM et al., 2017MRSA RCMB 2658
E. coli RCMB 010052
Geotrichum candidum RCMB 05097
P. aeruginosa RCMB 010043
B. subtilis RCMB 010067
H. pylori RCMB 088452
A. fumigatus RCMB 02568
M. tuberculosis
Cinnamomum glanduliferum (Wall) Nees (false camphor tree)Agar-well diffusion
Resazurin microtiter assay
Broth microdilution
The antibacterial activity of bark EO was good against MRSA (MIC = 7.81 µg/mL) and strong against E. coli (activity index = 1 and MIC = 0.49 µg/mL). Toxicity against colon (HCT-116), liver (HepG2) and breast (MCF-7) carcinoma cell lines was observed (IC50 = 9.1; 42.4; and 57.3 µg/mL). It was suggested that the antimicrobial and cytotoxic effects were due to eucalyptol (65.87%), as well as terpinene-4-ol (7.57%), α-terpineol (7.39%) and others.[222]
Jaradat N et al., 2016MRSA
S. aureus ATCC 25923
E. coli ATCC 25922
P. aeruginosa ATCC 27853
C. albicans—clinical isolate
Pheretima posthuma
Thymus bovei Benth.MIC assay
Broth microdilution
Antihelmintic assay
Trans-geraniol (35.38%), α-citral (20.37%), β-citral (14.76%) and cis-geraniol (7.38%) were the major identified phytocompounds (MRSA and E. coli MIC = 0.5 mg/mL). The most potent antimicrobial activity was observed in the case of P. aeruginosa, S. aureus and C. albicans (MIC = 0.25 mg/mL).
Strong antihelmintic activity was observed for the tested EO (compared with the piperazine citrate reference standard).
[223]
Lahmar A et al., 2016MRSA 138; 760; 753
ESBL E. coli
Ceftazidime-resistant A. baumannii
Pituranthos chloranthus
Teucruim ramosissimum
Pistacia lentiscus
alone and in combination with ABs
Broth microdilution
Broth microdilution checkerboard
Time-kill assay
MRSA MIC values varied as follows: 0.25–0.5 mg/mL for P. chloranthus EO; 0.25–1 mg/mL for T. ramosissimum; and 0.125–1 mg/mL for P. lentiscus. MIC values for E. coli and A. baumannii were found to be higher. Higher antibacterial effect against E. coli (synergic action) was observed when EOs were associated with ofloxacin and novobiocin (MIC was reduces up to 64 times). EOs enhanced the effect of all ABs used for MRSA strains (especially for MRSA 760).[224]
Shehadeh M et al., 2019S. aureus ATCC 25923
MRSA
E. faecium ATCC 700221
E. coli ATCC 25922
P. aeruginosa ATCC 27853
Origanum syriacum L. (bible hyssop)Broth microdilutionThe most effective EOs against MRSA and other S. aureus strains were those rich in thymol (MIC = 390 µg/mL). The chemotypes rich in α-terpinene were effective against P. aeruginosa (MIC = 1560 µg/mL) and EOs rich in gamma-terpinene expressed the highest antibacterial properties against E. faecium (MIC = 97 µg/mL), as well as good antioxidant effect.[225]
Demirci F et al., 2018MRSA ATCC 700699
S. aureus ATCC BAA-1026
S. epidermidis ATCC 14990
S. pyogenes ATCC 19615
S. pneumoniae ATCC 10015
P. aeruginosa ATCC 10145
H. influenzae ATCC 49247
M. catarrhalis ATCC 23245
Thymus sipyleus Boiss.Agar diffusion
Broth microdilution
Vapour diffusion
The best antibacterial effect for thyme scented lemon was noticed against S. aureus, S. pyogenes and M. catarrhalis (MIC = 12–13 µg/mL). MRSA MIC = 310 µg/mL and P. aeruginosa MIC = 1250 µg/mL (lowest inhibitory effect). The anti-inflammatory effect was 12.1 ± 1.8% in 100 µg/mL, both effects being required for efficient inhalations in the treatment of rhinosinusitis.[226]
Mahboubi M et al., 2016MRSAOliveria decumbens vent
Pelargonium graveolens L’Hér. vs. mupirocin
Skin wound infection in miceSimilar potent antibacterial effects against MRSA were obtained for both mupirocin and the herbal cream containing den EO (from Oliveria decumbens aerial part) and geranium EO (from Pelargonium graveolens leaves), with log CFU = 2.46 ± 0.32 and 2.5 ± 0.26, respectively, compared to 5.9 ± 0.26 and 5.65 ± 0.23 for placebo and control groups.[186]
Salameh N et al., 2020MRSA
P. mirabilis
S. aureus clinical isolates
E. coli ATCC 25922
E. faecium ATCC 700221
K. pneumoniae ATCC 13883
P. aeruginosa ATCC 27853
Shigella sonnei ATCC 25931
S. aureus ATCC 25923
C. albicans ATCC 90028
Epidermophyton floccosum ATCC 52066
Micromeria fruticosa serpyllifoliaBroth microdilution
Agar dilution
Low antimicrobial activity against MRSA (MIC = 3.125–6.250 mg/mL). The EO samples from different parts of Palestine exhibited different antimicrobial and antioxidant effects.[227]
Jaradat N et al., 2019MRSA
S. aureus ATCC 25923
E. faecium ATCC 700221
E. coli ATCC 25922
P. aeruginosa ATCC 27853
Shigella sonnie ATCC 25931
C. albicans ATCC 90028
Epidermophyton floccosum ATCC 10231
Stachys viticina Boiss.Microdilution assayThe strongest antibacterial activity of EO from leaves was observed against MRSA (MIC = 0.039 mg/mL). Cytotoxicity was observed for the 2 cancer cell lines: HeLa (cervical adenocarcinoma, with a 95% inhibition at 1.25 mg/mL) and Colo-205 (colon, with a 90% inhibition at 0.5 mg/mL). High COX1,2 inhibitory activity (similar to that of NSAID etodolac), as well as antioxidant activity (IC50 = 19.95 µg/mL), were determined.[228]
Kwiatkowski P et al., 2019MRSA
S. aureus ATCC 43300 (control)
Lavandula augustifolia Mill. associated with octenidine dihydrochlorideBroth microdilution
Checkerboard assay
Time-kill curve assay
Lavender EO (flowering herb) showed activity against MRSA (MIC = 13.72 µg/mL). Synergistic activity observed with octenidine dihydrochloride (FICI = 0.11–0.26). Lavender EO appears to modify the bacterial cell wall structure and might be used for enhancing the activity of conventional antiseptics.[229]
Chen J et al., 2020MRSA ATCC 43300
S. aureus ATCC 25923
E. faecalis ATCC 29212
B. subtilis ATCC 21332
P. aeruginosa ATCC 27853
S. gallinarum CVCC 79207
E. coli ATCC 25922
Cinnamomum camphora (Linn.) Presl (camphor)Broth microdilution
Field emission scanning electron microscopy
Transmission electron microscopy
EO showed a certain activity against MRSA (MIC = 0.8 mg/mL, MBC = 1.6 mg/mL) and good antibacterial effects on the other strains, dependent on the concentration.[187]
Noumi E et al., 2018MRSA—28 strains
S. aureus ATCC 6538
S. aureus ATCC 4330
P. aeruginosa
C. violaceum ATCC 12 472
Melaleuca alternifolia (Maiden and Betche) Cheel (tea tree)
Terpinen-4-ol
Disc diffusion assay
Microdilution assay
Semi-quantitative adherence assay
Crystal violet assay
Violacein inhibition assay
Swarming inhibition assay
Sa 442 gene was identified in all confirmed 28 MRSA strains (mecA gene positive, MIC = 0.048–3.125 mg/mL for tea tree EO and 0.048–1.52 mg/mL for the terpinen-4-ol; MBC = 25–50 mg/mL for tea tree EO and 6.25–50 mg/mL for terpinen-4-ol).
Both tea tree EO and terpinene-4-ol exhibited the adhesivity of MRSA on polystyrene (MIC/16 = 0.003 mg/mL).
Terpinen-4-ol showed anti-biofilm activity of 73.70%, while low concentrations of tea tree EO inhibited the formation of biofilm and cell communication.
[230]
Gradinaru AC et al., 2018P. aeruginosa ATCC 27853
S. aureus ATCC 25923
S. pneumoniae ATCC 49619
Penicillin-resistant S. pneumoniae (ARPA 2351)
S. aureus (MRSA 37, 4185)
S. pneumoniae (PRSP 4423, 4546, 4566)
Trachyspermum ammi (L.) Sprague ex Turrill
(ajwain, ajowan)
Broth microdilution
Checkerboard method
A synergistic action of ajowan EO/thymol + amoxicillin was observed on MRSA (FICI = 0.37–0.50). The same effect was observed for the association of EO + ciprofloxacin in the case of P. aeruginosa, S. aureus and penicillin-resistant S. pneumoniae (FICI = 0.37–0.5).[231]
Marino A et al., 2020S. aureus ATCC 6538
S. aureus ATCC 43300
S. epidermidis ATCC 35984
L. monocytogenes ATCC 13932
B. subtilis ATCC 6633
S. aureus 7786
MRSA (S. aureus 815)
S. aureus 74CCH-MRSA
P. aeruginosa ATCC 9027
Candida sp.
Coridothymus capitatus (L.) Reichenb. fil. Hydrolate alone or in association with tetracycline/itraconazoleCheckerboard method
Broth microdilution
Propidium iodide and
MitoTracker staining
Spanish oregano (also known as Thymus capitatus (L.) Hoffmanns. and Link) EO obtained from flowers was used. Antimicrobial activity of the prepared hydrolates (alone or in combination with tetracyline and itraconazole) was assessed. The hydrolate exhibited good antimicrobial activity, as well as a synergistic action (alteration of mitochondrial function) with itraconazole against C. krusei and an additive effect (alteration of membrane permeability) with tetracycline against MRSA strains.[232]
Tadic V et al., 2017MRSA clinical strain
MSSA ATCC 29213
E coli ATCC 25922
K pneumoniae cabapenem susceptible
K pneumoniae cabapenem resistant
C albicans ATCC 14053
Sideritis romana L. subsp. purpurea (Tal. ex Benth.) Heywood (purple ironwort)Mueller Hinton brothForty-three potentially active compounds were identified, the most abundant being bicyclogermacrene (23.8%), germacrene F (8%), (E)-caryophyllene (7.9%) and spathulenol (5.5%). High activity was noticed against both MSSA (MIC = 0.307 mg/mL, MBC = 0.615 mg/mL) and MRSA (MIC = 0.307 mg/mL and MBC = 0.153 mg/mL), at high and low inoculum. The same was observed for both extracts in 1.2-dichloroethane and methanol.[233]
Ramirez-Rueda RY et al., 2019MRSA ATCC 43300
E faecalis vancomycin-resistant ATCC 51299
Chrysopogon zizaniodes (L.) Roberty known as Vetiveria zizanioides (L.) Nash (vetiver grass)Mueller Hinton brothThe root extract EO showed activity against MRSA (MIC = 62.5 µg/mL) and VREF (MIC = 125 µg/mL). Cedr-8-en-13-ol was proposed as the most important compound exhibiting antimicrobial activity.[234]
Brun P et al., 2019MRSA
P aeruginosa
C glabrata
H simplex virus type 1 strain 16
Melaleuca alternifolia (Maiden and Betche) Cheel (tea tree)Broth microdilutionTen commercially available tea tree EO products were tested against strains grown in planktonic mode or biofilms. Regarding MRSA, MIC varied from 0.027–2.5% v/v for the tested substances. The authors concluded that the antimicrobial activity could not be attributed to terpinene-4-ol alone and stipulated that it is a consequence of synergism among different constituents of the EOs.[235]
Jaradat NA et al., 2016S. aureus ATCC 25923
E coli ATCC 25922
P aeruginosa ATCC 27853
MRSA
C. albicans clinical isolates
Trichodesma africanum (L.) Sm.Broth microdilutionMRSA MIC = 3 mg/mL. The microwave-ultrasonic extraction technique yielded the best results.[236]
Saidi M et al., 2016MRSA
E coli ESBL producing
P aeruginosa MBL producing
Thymus daenensis Celak.Disc diffusionMRSA MIC: 25 mg/mL. Good cyto-tolerability, as well as antioxidant properties were found.[237]
Wang B et al., 2017MRSA ATCC 43300
MRSA ATCC 33591
E. coli ATCC 25922
E. coli ATCC 44102
S. aureus ATCC 25923
S. aureus ATCC 26003
Pogostone—from Pogostemon cablin (Blanco) Beneth. (patchouli)
Broth microdilutionMolecular docking studies of pogostone (obtained from patchouli EO) with pentaerythritol tetranitrate reductase were performed and the structure–activity relationship was analysed. Compound 3h exhibited the highest antimicrobial activity against MRSA (MIC = 8 µg/mL), similar to that of levofloxacin and vancomycin that were used as positive controls.[238]
S. aureus—vancomycin resistant
Vasconcelos SECB et al., 2017VRSA
(S aureus strains isolated from food) and oxacillin resistant S aureus
S aureus ATCC 6538
Plectranthus amboinicus Lour. (Mexican mint)Disc diffusion
Microdilution
Microtiter-plate technique
Crystal violet assay
Counting viable cells
EO obtained from the leaves and stem was used to evaluate the antimicrobial and antibiofilm activity. Carvacrol was determined as the major component in the EO. All tested strains were sensitive to carvacrol and EO, and the best activity (no viable cells on the biofilm) was noticed for the combination of both products (MIC = 0.5 mg/mL).[189]

References

  1. Hutchings, M.I.; Truman, A.W.; Wilkinson, B. Antibiotics: Past, present and future. Curr. Opin. Microbiol. 2019, 51, 72–80. [Google Scholar] [CrossRef] [PubMed]
  2. Zhuang, M.; Achmon, Y.; Cao, Y.; Liang, X.; Chen, L.; Wang, H.; Siame, B.A.; Leung, K.Y. Distribution of antibiotic resistance genes in the environment. Environ. Pollut. 2021, 285, 117402. [Google Scholar] [CrossRef] [PubMed]
  3. Mohr, K.I. History of Antibiotics Research. In How to Overcome the Antibiotic Crisis; Springer: Cham, Switzerland, 2016; pp. 237–272. [Google Scholar] [CrossRef]
  4. Aslam, B.; Wang, W.; Arshad, M.I.; Khurshid, M.; Muzammil, S.; Rasool, M.H.; Nisar, M.A.; Alvi, R.F.; Aslam, M.A.; Qamar, M.U.; et al. Antibiotic resistance: A rundown of a global crisis. Infect. Drug Resist. 2018, 11, 1645–1658. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  5. Van Duijkeren, E.; Schink, A.K.; Roberts, M.C.; Wang, Y.; Schwarz, S. Mechanisms of Bacterial Resistance to Antimicrobial Agents. Microbiol. Spectr. 2018, 6. [Google Scholar] [CrossRef]
  6. Blair, J.M.A.; Webber, M.A.; Baylay, A.J.; Ogbolu, D.O.; Piddock, L.J.V. Molecular mechanisms of antibiotic resistance. Nat. Rev. Microbiol. 2015, 13, 42–51. [Google Scholar] [CrossRef]
  7. Wang, J.; Chen, X. Removal of antibiotic resistance genes (ARGs) in various wastewater treatment processes: An overview. Crit. Rev. Environ. Sci. Technol. 2020, 52, 571–630. [Google Scholar] [CrossRef]
  8. Chokshi, A.; Sifri, Z.; Cennimo, D.; Horng, H. Global contributors to antibiotic resistance. J. Glob. Infect. Dis. 2019, 11, 36–42. [Google Scholar] [CrossRef]
  9. Frieri, M.; Kumar, K.; Boutin, A. Antibiotic resistance. J. Infect. Public Health 2017, 10, 369–378. [Google Scholar] [CrossRef] [Green Version]
  10. Mancuso, G.; Midiri, A.; Gerace, E.; Biondo, C. Bacterial Antibiotic Resistance: The Most Critical Pathogens. Pathogens 2021, 10, 1310. [Google Scholar] [CrossRef] [PubMed]
  11. Malik, B.; Bhattacharyya, S. Antibiotic drug-resistance as a complex system driven by socio-economic growth and antibiotic misuse. Sci. Rep. 2019, 9, 9788. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  12. King, L.M.; Fleming-Dutra, K.E.; Hicks, L.A. Advances in optimizing the prescription of antibiotics in outpatient settings. BMJ 2018, 363, k3047. [Google Scholar] [CrossRef]
  13. López-Vázquez, P.M.; Vazquez-Lago, J.; Figueiras, A. Misprescription of antibiotics in primary care: A critical systematic review of its determinants. J. Eval. Clin. Pract. 2011, 18, 473–484. [Google Scholar] [CrossRef]
  14. Grigoryan, L.; Germanos, G.; Zoorob, R.; Juneja, S.; Raphael, J.L.; Paasche-Orlow, M.K.; Trautner, B.W. Use of Antibiotics Without a Prescription in the U.S. Population: A Scoping Review. Ann. Intern. Med. 2019, 171, 257–263. [Google Scholar] [CrossRef]
  15. Gerber, J.S.; Jackson, M.A.; Tamma, P.D.; Zaoutis, T.E.; Maldonado, Y.A.; O’leary, S.T.; Banerjee, R.; Barnett, E.D.; Campbell, J.D.; Caserta, M.T.; et al. Antibiotic Stewardship in Pediatrics. Pediatrics 2021, 147, e2020040295. [Google Scholar] [CrossRef]
  16. Allel, K.; Day, L.; Hamilton, A.; Lin, L.; Furuya-Kanamori, L.; Moore, C.E.; Van Boeckel, T.; Laxminarayan, R.; Yakob, L. Global antimicrobial-resistance drivers: An ecological country-level study at the human–animal interface. Lancet Planet. Health 2023, 7, e291–e303. [Google Scholar] [CrossRef] [PubMed]
  17. Zalewska, M.; Błażejewska, A.; Czapko, A.; Popowska, M. Antibiotics and Antibiotic Resistance Genes in Animal Manure—Consequences of Its Application in Agriculture. Front. Microbiol. 2021, 12, 610656. [Google Scholar] [CrossRef] [PubMed]
  18. Martin, M.J.; Thottathil, S.E.; Newman, T.B. Antibiotics Overuse in Animal Agriculture: A Call to Action for Health Care Providers. Am. J. Public Health 2015, 105, 2409–2410. [Google Scholar] [CrossRef]
  19. Landers, T.F.; Cohen, B.; Wittum, T.E.; Larson, E.L. A Review of Antibiotic Use in Food Animals: Perspective, Policy, and Potential. Public Health Rep. 2012, 127, 4–22. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  20. Kirchhelle, C. Pharming animals: A global history of antibiotics in food production (1935–2017). Palgrave Commun. 2018, 4, 96. [Google Scholar] [CrossRef] [Green Version]
  21. Larsson, D.G.J.; Flach, C.-F. Antibiotic resistance in the environment. Nat. Rev. Microbiol. 2022, 20, 257–269. [Google Scholar] [CrossRef]
  22. Wang, J.; Wang, L.; Zhu, L.; Wang, J.; Xing, B. Antibiotic resistance in agricultural soils: Source, fate, mechanism and attenuation strategy. Crit. Rev. Environ. Sci. Technol. 2022, 52, 847–889. [Google Scholar] [CrossRef]
  23. Silver, L.L. Challenges of Antibacterial Discovery. Clin. Microbiol. Rev. 2011, 24, 71–109. [Google Scholar] [CrossRef] [Green Version]
  24. Terreni, M.; Taccani, M.; Pregnolato, M. New Antibiotics for Multidrug-Resistant Bacterial Strains: Latest Research Developments and Future Perspectives. Molecules 2021, 26, 2671. [Google Scholar] [CrossRef]
  25. Miethke, M.; Pieroni, M.; Weber, T.; Brönstrup, M.; Hammann, P.; Halby, L.; Arimondo, P.B.; Glaser, P.; Aigle, B.; Bode, H.B.; et al. Towards the sustainable discovery and development of new antibiotics. Nat. Rev. Chem. 2021, 5, 726–749. [Google Scholar] [CrossRef]
  26. Mendelson, M.; Matsoso, M.P. The World Health Organization Global Action Plan for antimicrobial resistance. S. Afr. Med. J. 2015, 105, 325. [Google Scholar] [CrossRef] [Green Version]
  27. World Health Organization Global Priority List of Antibiotic-Resistant Bacteria to Guide Research, Discovery, and Development of New Antibiotics. Cad. Pesqui. 2017, 43, 348–365.
  28. 2021 Antibacterial Agents in Clinical and Preclinical Development: An Overview and Analysis. Available online: https://www.who.int/publications/i/item/9789240047655 (accessed on 12 September 2022).
  29. Political Leadership and Action on Antimicrobial Resistance: The Road to the 2024 UNGA High-Level Meeting on AMR. Available online: https://www.unep.org/events/conference/political-leadership-and-action-antimicrobial-resistance-road-2024-unga-high (accessed on 28 May 2023).
  30. One Health Trust—Resistance Map. Available online: https://resistancemap.onehealthtrust.org/AntibioticResistance.php (accessed on 24 May 2023).
  31. Surveillance Atlas of Infectious Diseases. Available online: https://atlas.ecdc.europa.eu/public/index.aspx?Dataset=27&HealthTopic=4 (accessed on 9 May 2022).
  32. Antimicrobial Resistance Surveillance in Europe 2022–2020 Data. Available online: https://www.ecdc.europa.eu/en/publications-data/antimicrobial-resistance-surveillance-europe-2022-2020-data (accessed on 8 July 2022).
  33. Yu, Z.; Tang, J.; Khare, T.; Kumar, V. The alarming antimicrobial resistance in ESKAPEE pathogens: Can essential oils come to the rescue? Fitoterapia 2020, 140, 104433. [Google Scholar] [CrossRef] [PubMed]
  34. Chouhan, S.; Sharma, K.; Guleria, S. Antimicrobial Activity of Some Essential Oils—Present Status and Future Perspectives. Medicines 2017, 4, 58. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  35. Guimarães, A.C.; Meireles, L.M.; Lemos, M.F.; Guimarães, M.C.C.; Endringer, D.C.; Fronza, M.; Scherer, R. Antibacterial Activity of Terpenes and Terpenoids Present in Essential Oils. Molecules 2019, 24, 2471. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  36. Chandra, H.; Bishnoi, P.; Yadav, A.; Patni, B.; Mishra, A.P.; Nautiyal, A.R. Antimicrobial Resistance and the Alternative Resources with Special Emphasis on Plant-Based Antimicrobials—A Review. Plants 2017, 6, 16. [Google Scholar] [CrossRef] [Green Version]
  37. Freires, I.A.; Denny, C.; Benso, B.; De Alencar, S.M.; Rosalen, P.L. Antibacterial Activity of Essential Oils and Their Isolated Constituents against Cariogenic Bacteria: A Systematic Review. Molecules 2015, 20, 7329–7358. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  38. Mahizan, N.A.; Yang, S.-K.; Moo, C.L.; Song, A.A.-L.; Chong, C.-M.; Chong, C.-W.; Abushelaibi, A.; Erin Lim, S.-H.; Lai, K.-S. Terpene Derivatives as a Potential Agent against Antimicrobial Resistance (AMR) Pathogens. Molecules 2019, 24, 2631. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  39. Bassanetti, I.; Carcelli, M.; Buschini, A.; Montalbano, S.; Leonardi, G.; Pelagatti, P.; Tosi, G.; Massi, P.; Fiorentini, L.; Rogolino, D. Investigation of antibacterial activity of new classes of essential oils derivatives. Food Control 2017, 73, 606–612. [Google Scholar] [CrossRef]
  40. Dhifi, W.; Bellili, S.; Jazi, S.; Bahloul, N.; Mnif, W. Essential Oils’ Chemical Characterization and Investigation of Some Biological Activities: A Critical Review. Medicines 2016, 3, 25. [Google Scholar] [CrossRef] [Green Version]
  41. Man, A.; Santacroce, L.; Iacob, R.; Mare, A.; Man, L. Antimicrobial Activity of Six Essential Oils Against a Group of Human Pathogens: A Comparative Study. Pathogens 2019, 8, 15. [Google Scholar] [CrossRef] [Green Version]
  42. Badescu, B.; Buda, V.; Romanescu, M.; Lombrea, A.; Danciu, C.; Dalleur, O.; Dohou, A.M.; Dumitrascu, V.; Cretu, O.; Licker, M.; et al. Current State of Knowledge Regarding WHO Critical Priority Pathogens: Mechanisms of Resistance and Proposed Solutions through Candidates Such as Essential Oils. Plants 2022, 11, 1789. [Google Scholar] [CrossRef]
  43. Ambrosio, C.M.S.; de Alencar, S.M.; de Sousa, R.L.M.; Moreno, A.M.; Da Gloria, E.M. Antimicrobial activity of several essential oils on pathogenic and beneficial bacteria. Ind. Crop. Prod. 2017, 97, 128–136. [Google Scholar] [CrossRef]
  44. Nair, A.; Mallya, R.; Suvarna, V.; Khan, T.A.; Momin, M.; Omri, A. Nanoparticles—Attractive Carriers of Antimicrobial Essential Oils. Antibiotics 2022, 11, 108. [Google Scholar] [CrossRef]
  45. Li, Y.-X.; Erhunmwunsee, F.; Liu, M.; Yang, K.; Zheng, W.; Tian, J. Antimicrobial mechanisms of spice essential oils and application in food industry. Food Chem. 2022, 382, 132312. [Google Scholar] [CrossRef]
  46. El-Tarabily, K.A.; El-Saadony, M.T.; Alagawany, M.; Arif, M.; Batiha, G.E.; Khafaga, A.F.; Elwan, H.A.M.; Elnesr, S.S.; El-Hack, M.E.A. Using essential oils to overcome bacterial biofilm formation and their antimicrobial resistance. Saudi J. Biol. Sci. 2021, 28, 5145–5156. [Google Scholar] [CrossRef]
  47. Ortega-Ramirez, L.A.; Gutiérrez-Pacheco, M.M.; Vargas-Arispuro, I.; González-Aguilar, G.A.; Martínez-Téllez, M.A.; Ayala-Zavala, J.F. Inhibition of Glucosyltransferase Activity and Glucan Production as an Antibiofilm Mechanism of Lemongrass Essential Oil against Escherichia coli O157:H7. Antibiotics 2020, 9, 102. [Google Scholar] [CrossRef] [Green Version]
  48. Evangelista, A.G.; Corrêa, J.A.F.; Pinto, A.C.S.M.; Luciano, F.B. The impact of essential oils on antibiotic use in animal production regarding antimicrobial resistance—A review. Crit. Rev. Food Sci. Nutr. 2021, 62, 5267–5283. [Google Scholar] [CrossRef]
  49. Swamy, M.K.; Akhtar, M.S.; Sinniah, U.R. Antimicrobial properties of plant essential oils against human pathogens and their mode of action: An updated review. Evid. Based Complement. Altern. Med. 2016, 2016, 3012462. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  50. Quave, C.L.; Horswill, A.R. Flipping the switch: Tools for detecting small molecule inhibitors of staphylococcal virulence. Front. Microbiol. 2014, 5, 706. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  51. Reyes-Jurado, F.; Franco-Vega, A.; Ramírez-Corona, N.; Palou, E.; López-Malo, A. Essential Oils: Antimicrobial Activities, Extraction Methods, and Their Modeling. Food Eng. Rev. 2015, 7, 275–297. [Google Scholar] [CrossRef]
  52. Ayobami, O.; Willrich, N.; Reuss, A.; Eckmanns, T.; Markwart, R. The ongoing challenge of vancomycin-resistant Enterococcus faecium and Enterococcus faecalis in Europe: An epidemiological analysis of bloodstream infections. Emerg. Microbes Infect. 2020, 9, 1180–1193. [Google Scholar] [CrossRef] [PubMed]
  53. Lee, B.J.; Vu, B.N.; Seddon, A.N.; Hodgson, H.A.; Wang, S.K. Treatment Considerations for CNS Infections Caused by Vancomycin-Resistant Enterococcus faecium: A Focused Review of Linezolid and Daptomycin. Ann. Pharmacother. 2020, 54, 1243–1251. [Google Scholar] [CrossRef]
  54. Shiadeh, S.M.J.; Pormohammad, A.; Hashemi, A.; Lak, P. Global prevalence of antibiotic resistance in blood-isolated Enterococcus faecalis and Enterococcus faecium: A systematic review and meta-analysis. Infect. Drug Resist. 2019, 12, 2713–2725. [Google Scholar] [CrossRef] [Green Version]
  55. Zhou, X.; Willems, R.J.L.; Friedrich, A.W.; Rossen, J.W.A.; Bathoorn, E. Enterococcus faecium: From microbiological insights to practical recommendations for infection control and diagnostics. Antimicrob. Resist. Infect. Control. 2020, 9, 130. [Google Scholar] [CrossRef]
  56. Lee, T.; Pang, S.; Abraham, S.; Coombs, G.W. Antimicrobial-resistant CC17 Enterococcus faecium: The past, the present and the future. J. Glob. Antimicrob. Resist. 2018, 16, 36–47. [Google Scholar] [CrossRef] [Green Version]
  57. Olearo, F.; Both, A.; Campos, C.B.; Hilgarth, H.; Klupp, E.-M.; Hansen, J.L.; Maurer, F.P.; Christner, M.; Aepfelbacher, M.; Rohde, H. Emergence of linezolid-resistance in vancomycin-resistant Enterococcus faecium ST117 associated with increased linezolid-consumption. Int. J. Med. Microbiol. 2021, 311, 151477. [Google Scholar] [CrossRef] [PubMed]
  58. Gouliouris, T.; Coll, F.; Ludden, C.; Blane, B.; Raven, K.E.; Naydenova, P.; Crawley, C.; Török, M.E.; Enoch, D.A.; Brown, N.M.; et al. Quantifying acquisition and transmission of Enterococcus faecium using genomic surveillance. Nat. Microbiol. 2020, 6, 103–111. [Google Scholar] [CrossRef] [PubMed]
  59. Vivas, R.; Barbosa, A.A.T.; Dolabella, S.S.; Jain, S. Multidrug-Resistant Bacteria and Alternative Methods to Control Them: An Overview. Microb. Drug Resist. 2019, 25, 890–908. [Google Scholar] [CrossRef]
  60. Moosavian, M.; Ghadri, H.; Samli, Z. Molecular detection of vanA and vanB genes among vancomycin-resistant enterococci in ICU-hospitalized patients in Ahvaz in southwest of Iran. Infect. Drug Resist. 2018, 11, 2269–2275. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  61. Satlin, M.J.; Nicolau, D.P.; Humphries, R.M.; Kuti, J.L.; Campeau, S.A.; Ii, J.S.L.; Weinstein, M.P.; Jorgensen, J.H.; Clinical and Laboratory Standards Institute Subcommittee on Antimicrobial Susceptibility Testing and Ad Hoc Working Group on Revision of Daptomycin Enterococcal Breakpoints. Development of Daptomycin Susceptibility Breakpoints for Enterococcus faecium and Revision of the Breakpoints for Other Enterococcal Species by the Clinical and Laboratory Standards Institute. Clin. Infect. Dis. 2019, 70, 1240–1246. [Google Scholar] [CrossRef]
  62. Gorrie, C.; Higgs, C.; Carter, G.; Stinear, T.P.; Howden, B. Genomics of vancomycin-resistant Enterococcus faecium. Microb. Genom. 2019, 5, e000283. [Google Scholar] [CrossRef]
  63. Saki, M.; Seyed-Mohammadi, S.; Montazeri, E.A.; Siahpoosh, A.; Moosavian, M.; Latifi, S.M. In vitro antibacterial properties of Cinnamomum zeylanicum essential oil against clinical extensively drug-resistant bacteria. Eur. J. Integr. Med. 2020, 37, 101146. [Google Scholar] [CrossRef]
  64. Iseppi, R.; Mariani, M.; Condò, C.; Sabia, C.; Messi, P. Essential Oils: A Natural Weapon against Antibiotic-Resistant Bacteria Responsible for Nosocomial Infections. Antibiotics 2021, 10, 417. [Google Scholar] [CrossRef]
  65. Owen, L.; Webb, J.P.; Green, J.; Smith, L.J.; Laird, K. From formulation to in vivo model: A comprehensive study of a synergistic relationship between vancomycin, carvacrol, and cuminaldehyde against Enterococcus faecium. Phytotherapy Res. 2020, 34, 1638–1649. [Google Scholar] [CrossRef]
  66. Owen, L.; White, A.W.; Laird, K. Characterisation and screening of antimicrobial essential oil components against clinically important antibiotic-resistant bacteria using thin layer chromatography-direct bioautography hyphenated with GC-MS, LC-MS and NMR. Phytochem. Anal. 2018, 30, 121–131. [Google Scholar] [CrossRef]
  67. Sakkas, H.; Economou, V.; Gousia, P.; Bozidis, P.; Sakkas, V.A.; Petsios, S.; Mpekoulis, G.; Ilia, A.; Papadopoulou, C. Antibacterial Efficacy of Commercially Available Essential Oils Tested Against Drug-Resistant Gram-Positive Pathogens. Appl. Sci. 2018, 8, 2201. [Google Scholar] [CrossRef] [Green Version]
  68. Di Vito, M.; Smolka, A.; Proto, M.R.; Barbanti, L.; Gelmini, F.; Napoli, E.; Bellardi, M.G.; Mattarelli, P.; Beretta, G.; Sanguinetti, M.; et al. Is the Antimicrobial Activity of Hydrolates Lower than that of Essential Oils? Antibiotics 2021, 10, 88. [Google Scholar] [CrossRef]
  69. Sun, Y.; Zhang, J. Helicobacter pylori recrudescence and its influencing factors. J. Cell. Mol. Med. 2019, 23, 7919–7925. [Google Scholar] [CrossRef] [Green Version]
  70. Saeidi, E.; Sheikhshahrokh, A. vacA Genotype Status of Helicobacter pylori Isolated from Foods with Animal Origin. BioMed Res. Int. 2016, 2016, 8701067. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  71. Amaral, O.; Fernandes, I.; Veiga, N.; Pereira, C.; Chaves, C.; Nelas, P.; Silva, D. Living Conditions and Helicobacter pylori in Adults. BioMed Res. Int. 2017, 2017, 9082716. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  72. Sjomina, O.; Pavlova, J.; Niv, Y.; Leja, M. Epidemiology of Helicobacter pylori infection. Helicobacter 2018, 23, e12514. [Google Scholar] [CrossRef] [Green Version]
  73. Hu, Y.; Wan, J.-H.; Li, X.-Y.; Zhu, Y.; Graham, D.Y.; Lu, N.-H. Systematic review with meta-analysis: The global recurrence rate of Helicobacter pylori. Aliment. Pharmacol. Ther. 2017, 46, 773–779. [Google Scholar] [CrossRef] [Green Version]
  74. Sugano, K.; Tack, J.; Kuipers, E.J.; Graham, D.Y.; El-Omar, E.M.; Miura, S.; Haruma, K.; Asaka, M.; Uemura, N.; Malfertheiner, P. Kyoto global consensus report on Helicobacter pylori gastritis. Gut 2015, 64, 1353–1367. [Google Scholar] [CrossRef] [Green Version]
  75. Chang, W.-L.; Yeh, Y.-C.; Sheu, B.-S. The impacts of H. pylori virulence factors on the development of gastroduodenal diseases. J. Biomed. Sci. 2018, 25, 68. [Google Scholar] [CrossRef] [Green Version]
  76. Matta, A.J.; Zambrano, D.C.; Pazos, A.J. Punctual mutations in 23S rRNA gene of clarithromycin-resistant Helicobacter pylori in Colombian populations. World J. Gastroenterol. 2018, 24, 1531–1539. [Google Scholar] [CrossRef]
  77. Greenberg, E.R.; Anderson, G.L.; Morgan, D.R.; Torres, J.; Chey, W.D.; Bravo, L.E.; Dominguez, R.L.; Ferreccio, C.; Herrero, R.; Lazcano-Ponce, E.C.; et al. 14-day triple, 5-day concomitant, and 10-day sequential therapies for Helicobacter pylori infection in seven Latin American sites: A randomised trial. Lancet 2011, 378, 507–514. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  78. Mégraud, F. Current Recommendations for Helicobacter pylori Therapies in a World of Evolving Resistance. Gut Microbes 2013, 4, 37–41. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Xuan, S.H.; Wu, L.P.; Zhou, Y.G.; Xiao, M.B. Detection of Clarithromycin-Resistant Helicobacter pylori in Clinical Specimens by Molecular Methods: A Review. J. Glob. Antimicrob. Resist. 2016, 4, 35–41. [Google Scholar] [CrossRef] [PubMed]
  80. Marques, A.T.; Vítor, J.M.B.; Santos, A.; Oleastro, M.; Vale, F.F. Trends in Helicobacter pylori Resistance to Clarithromycin: From Phenotypic to Genomic Approaches. Microb. Genomics 2020, 6, 1–11. [Google Scholar] [CrossRef]
  81. Li, M.; Oshima, T.; Horikawa, T.; Tozawa, K.; Tomita, T.; Fukui, H.; Watari, J.; Miwa, H. Systematic Review with Meta-Analysis: Vonoprazan, a Potent Acid Blocker, Is Superior to Proton-Pump Inhibitors for Eradication of Clarithromycin-Resistant Strains of Helicobacter pylori. Helicobacter 2018, 23, e12495. [Google Scholar] [CrossRef]
  82. FDA Approves Two Vonoprazan Therapies for H pylori Eradication. Available online: https://www.medscape.com/viewarticle/973403?icd=ssl_login_success_220916 (accessed on 28 June 2022).
  83. Elkousy, R.H.; Mostafa, N.M.; Abd-Alkhalek, A.M.; Hassab, M.A.E.; Al-Rashood, S.T.; Eldehna, W.M.; Eldahshan, O.A.; Abd-Alkhalek C, A.M. GC/MS Analysis and Potential Synergistic Effect of Mandarin and Marjoram Oils on Helicobacter pylori. J. Enzyme Inhib. Med. Chem. 2022, 37, 1610–1619. [Google Scholar] [CrossRef]
  84. Al-Sayed, E.; Gad, H.A.; El-Kersh, D.M. Characterization of Four Piper Essential Oils (GC/MS and ATR-IR) Coupled to Chemometrics and Their Anti-Helicobacter pylori Activity. ACS Omega 2021, 6, 25652–25663. [Google Scholar] [CrossRef]
  85. Gad, H.; Al-Sayed, E.; Ayoub, I. Phytochemical Discrimination of Pinus Species Based on GC–MS and ATR-IR Analyses and Their Impact on Helicobacter pylori. Phytochem. Anal. 2021, 32, 820–835. [Google Scholar] [CrossRef]
  86. Meriem, M.; Rachida, A.; Housseyn, M.; Farida, T.; Abdelaziz, M.; Fouzia, M. Antimicrobial Activity of the Essential Oil Isolated from Pistacia lentiscus Leaves Against Helicobacter pylori Algerian Clinical Isolates. J. Essent. Oil-Bear. Plants 2016, 19, 466–474. [Google Scholar] [CrossRef]
  87. Knezevic, P.; Aleksic Sabo, V.; Simin, N.; Lesjak, M.; Mimica-Dukic, N. A Colorimetric Broth Microdilution Method for Assessment of Helicobacter pylori Sensitivity to Antimicrobial Agents. J. Pharm. Biomed. Anal. 2018, 152, 271–278. [Google Scholar] [CrossRef] [PubMed]
  88. Ali, S.S.; Abd Elnabi, M.K.; Alkherkhisy, M.M.; Hasan, A.; Li, F.; Khalil, M.; Sun, J.; El-Zawawy, N. Exploring the Potential of Cinnamomum zeylanicum Oil against Drug Resistant Helicobacter pylori-Producing Cytotoxic Genes. J. Appl. Biomed. 2022, 20, 22–36. [Google Scholar] [CrossRef] [PubMed]
  89. Korona-Glowniak, I.; Glowniak-Lipa, A.; Ludwiczuk, A.; Baj, T.; Malm, A. The in Vitro Activity of Essential Oils against Helicobacter pylori Growth and Urease Activity. Molecules 2020, 25, 586. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  90. Neves, N.C.V.; de Mello, M.P.; Smith, S.M.; Boylan, F.; Caliari, M.V.; Castilho, R.O. Chemical Composition and In Vitro Anti-Helicobacter pylori Activity of Campomanesia lineatifolia Ruiz & Pavón (Myrtaceae) Essential Oil. Plants 2022, 11, 1945. [Google Scholar] [CrossRef]
  91. Bhattamisra, S.K.; Yean Yan, V.L.; Koh Lee, C.; Hui Kuean, C.; Candasamy, M.; Liew, Y.K.; Sahu, P.S. Protective Activity of Geraniol against Acetic Acid and Helicobacter pylori- Induced Gastric Ulcers in Rats. J. Tradit. Complement. Med. 2019, 9, 206–214. [Google Scholar] [CrossRef]
  92. Memariani, Z.; Sharifzadeh, M.; Bozorgi, M.; Hajimahmoodi, M.; Farzaei, M.H.; Gholami, M.; Siavoshi, F.; Saniee, P. Protective Effect of Essential Oil of Pistacia Atlantica Desf. on Peptic Ulcer: Role of α-Pinene. J. Tradit. Chin. Med. 2017, 37, 57–63. [Google Scholar] [CrossRef]
  93. Jung, D.H.; Park, M.H.; Kim, C.J.; Lee, J.Y.; Keum, C.Y.; Kim, I.S.; Yun, C.H.; Kim, S.K.; Kim, W.H.; Lee, Y.C. Effect of β-Caryophyllene from Cloves Extract on Helicobacter pylori Eradication in Mouse Model. Nutrients 2020, 12, 1000. [Google Scholar] [CrossRef] [Green Version]
  94. Man, S.M. The Clinical Importance of Emerging Campylobacter Species. Nat. Rev. Gastroenterol. Hepatol. 2011, 8, 669–685. [Google Scholar] [CrossRef]
  95. Costa, D.; Iraola, G. Pathogenomics of Emerging Campylobacter Species. Clin. Microbiol. Rev. 2019, 32, 1–24. [Google Scholar] [CrossRef] [PubMed]
  96. Koutsoumanis, K.; Allende, A.; Álvarez-Ordóñez, A.; Bolton, D.; Bover-Cid, S.; Chemaly, M.; Davies, R.; De Cesare, A.; Herman, L.; Hilbert, F.; et al. Role Played by the Environment in the Emergence and Spread of Antimicrobial Resistance (AMR) through the Food Chain. EFSA J. 2021, 19, e06651. [Google Scholar] [CrossRef]
  97. Hansson, I.; Sandberg, M.; Habib, I.; Lowman, R.; Engvall, E.O. Knowledge Gaps in Control of Campylobacter for Prevention of Campylobacteriosis. Transbound. Emerg. Dis. 2018, 65, 30–48. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  98. Dai, L.; Sahin, O.; Grover, M.; Zhang, Q. New and Alternative Strategies for the Prevention, Control, and Treatment of Antibiotic-Resistant Campylobacter. Transl. Res. 2020, 223, 76–88. [Google Scholar] [CrossRef]
  99. Esan, O.B.; Pearce, M.; van Hecke, O.; Roberts, N.; Collins, D.R.J.; Violato, M.; McCarthy, N.; Perera, R.; Fanshawe, T.R. Factors Associated with Sequelae of Campylobacter and Non-Typhoidal Salmonella Infections: A Systematic Review. EBioMedicine 2017, 15, 100–111. [Google Scholar] [CrossRef] [Green Version]
  100. Gharbi, M.; Béjaoui, A.; Ben Hamda, C.; Ghedira, K.; Ghram, A.; Maaroufi, A. Distribution of Virulence and Antibiotic Resistance Genes in Campylobacter jejuni and Campylobacter coli Isolated from Broiler Chickens in Tunisia. J. Microbiol. Immunol. Infect. 2022, 55, 1273–1282. [Google Scholar] [CrossRef]
  101. Ammar, A.M.; Abd El-Hamid, M.I.; El-Malt, R.M.S.; Azab, D.S.; Albogami, S.; Al-Sanea, M.M.; Soliman, W.E.; Ghoneim, M.M.; Bendary, M.M. Molecular Detection of Fluoroquinolone Resistance among Multidrug-, Extensively Drug-, and Pan-Drug-Resistant Campylobacter Species in Egypt. Antibiotics 2021, 10, 1342. [Google Scholar] [CrossRef] [PubMed]
  102. Khademi, F.; Sahebkar, A. Prevalence of Fluoroquinolone-Resistant Campylobacter Species in Iran: A Systematic Review and Meta-Analysis. Int. J. Microbiol. 2020, 2020, 8868197. [Google Scholar] [CrossRef] [PubMed]
  103. Wieczorek, K.; Osek, J. Antimicrobial Resistance Mechanisms among Campylobacter. BioMed. Res. Int. 2013, 2013, 340605. [Google Scholar] [CrossRef] [Green Version]
  104. Osaili, T.M.; Alaboudi, A.R. Antimicrobial Resistance of Campylobacter sp. In Foodborne Pathogens and Antibiotic Resistance; Wiley: Hoboken, NJ, USA, 2016; pp. 417–429. [Google Scholar] [CrossRef]
  105. Sproston, E.L.; Wimalarathna, H.M.L.; Sheppard, S.K. Trends in fluoroquinolone resistance in Campylobacter. Microb. Genom. 2018, 4, e000198. [Google Scholar] [CrossRef] [PubMed]
  106. Tang, Y.; Fang, L.; Xu, C.; Zhang, Q. Antibiotic Resistance Trends and Mechanisms in the Foodborne Pathogen, Campylobacter. Anim. Health Res. Rev. 2017, 18, 87–98. [Google Scholar] [CrossRef] [Green Version]
  107. Gahamanyi, N.; Song, D.-G.; Cha, K.H.; Yoon, K.-Y.; Mboera, L.E.; Matee, M.I.; Mutangana, D.; Amachawadi, R.G.; Komba, E.V.; Pan, C.-H. Susceptibility of Campylobacter Strains to Selected Natural Products and Frontline Antibiotics. Antibiotics 2020, 9, 790. [Google Scholar] [CrossRef]
  108. Duarte, A.; Luís, Â.; Oleastro, M.; Domingues, F.C. Antioxidant properties of coriander essential oil and linalool and their potential to control Campylobacter spp. Food Control 2016, 61, 115–122. [Google Scholar] [CrossRef]
  109. Kovács, J.K.; Felső, P.; Makszin, L.; Pápai, Z.; Horváth, G.; Ábrahám, H.; Palkovics, T.; Böszörményi, A.; Emődy, L.; Schneider, G. Antimicrobial and Virulence-Modulating Effects of Clove Essential Oil on the Foodborne Pathogen Campylobacter jejuni. Appl. Environ. Microbiol. 2016, 82, 6158–6166. [Google Scholar] [CrossRef] [Green Version]
  110. Ahmed, J.; Hiremath, N.; Jacob, H. Antimicrobial, Rheological, and Thermal Properties of Plasticized Polylactide Films Incorporated with Essential Oils to Inhibit Staphylococcus aureus and Campylobacter jejuni. J. Food Sci. 2016, 81, E419–E429. [Google Scholar] [CrossRef]
  111. El Baaboua, A.; El Maadoudi, M.; Bouyahya, A.; Belmehdi, O.; Kounnoun, A.; Cheyadmi, S.; Ouzakar, S.; Senhaji, N.S.; Abrini, J. Evaluation of the combined effect of antibiotics and essential oils against Campylobacter multidrug resistant strains and their biofilm formation. S. Afr. J. Bot. 2022, 150, 451–465. [Google Scholar] [CrossRef]
  112. Lin, L.; Zhu, Y.; Cui, H. Electrospun thyme essential oil/gelatin nanofibers for active packaging against Campylobacter jejuni in chicken. LWT 2018, 97, 711–718. [Google Scholar] [CrossRef]
  113. Lin, L.; Gu, Y.; Sun, Y.; Cui, H. Characterization of chrysanthemum essential oil triple-layer liposomes and its application against Campylobacter jejuni on chicken. LWT 2019, 107, 16–24. [Google Scholar] [CrossRef]
  114. Šimunović, K.; Sahin, O.; Kovač, J.; Shen, Z.; Klančnik, A.; Zhang, Q.; Smole Možina, S. (-)-α-Pinene reduces quorum sensing and Campylobacter jejuni colonization in broiler chickens. PLoS ONE 2020, 15, e0230423. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Horst, A.L.; Rosenbohm, J.M.; Kolluri, N.; Hardick, J.; Gaydos, C.A.; Cabodi, M.; Klapperich, C.M.; Linnes, J.C. A paperfluidic platform to detect Neisseria gonorrhoeae in clinical samples. Biomed. Microdevices 2018, 20, 35. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  116. WHO Guidelines for the Treatment of Neisseria Gonorrhoeae. Available online: https://www.who.int/publications/i/item/9789241549691 (accessed on 2 July 2022).
  117. Chan, P.A.; Robinette, A.; Montgomery, M.; Almonte, A.; Cu-Uvin, S.; Lonks, J.R.; Chapin, K.C.; Kojic, E.M.; Hardy, E.J. Extragenital Infections Caused by Chlamydia trachomatis and Neisseria gonorrhoeae: A Review of the Literature. Infect. Dis. Obstet. Gynecol. 2016, 2016, 5758387. [Google Scholar] [CrossRef] [Green Version]
  118. Rice, P.A.; Shafer, W.M.; Ram, S.; Jerse, A.E. Neisseria gonorrhoeae: Drug Resistance, Mouse Models, and Vaccine Development. Annu. Rev. Microbiol. 2017, 71, 665–686. [Google Scholar] [CrossRef] [Green Version]
  119. Quillin, S.J.; Seifert, H.S. Neisseria gonorrhoeae host adaptation and pathogenesis. Nat. Rev. Genet. 2018, 16, 226–240. [Google Scholar] [CrossRef]
  120. Lim, K.Y.L.; Mullally, C.A.; Haese, E.C.; Kibble, E.A.; McCluskey, N.R.; Mikucki, E.C.; Thai, V.C.; Stubbs, K.A.; Sarkar-Tyson, M.; Kahler, C.M. Anti-Virulence Therapeutic Approaches for Neisseria gonorrhoeae. Antibiotics 2021, 10, 103. [Google Scholar] [CrossRef] [PubMed]
  121. Wi, T.; Lahra, M.M.; Ndowa, F.; Bala, M.; Dillon, J.-A.R.; Ramon-Pardo, P.; Eremin, S.R.; Bolan, G.; Unemo, M. Antimicrobial resistance in Neisseria gonorrhoeae: Global surveillance and a call for international collaborative action. PLoS Med. 2017, 14, e1002344. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  122. Suay-García, B.; Pérez-Gracia, M.T. Future Prospects for Neisseria gonorrhoeae Treatment. Antibiotics 2018, 7, 49. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  123. Whiley, D.M.; Jennison, A.; Pearson, J.; Lahra, M.M. Genetic characterisation of Neisseria gonorrhoeae resistant to both ceftriaxone and azithromycin. Lancet Infect. Dis. 2018, 18, 717–718. [Google Scholar] [CrossRef] [PubMed]
  124. 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. 2019, 209, 95–108. [Google Scholar] [CrossRef]
  125. Ayala, J.C.; Schmerer, M.W.; Kersh, E.N.; Unemo, M.; Shafer, W.M. Gonococcal Clinical Strains Bearing a Common gdhR Single Nucleotide Polymorphism That Results in Enhanced Expression of the Virulence Gene lctP Frequently Possess a mtrR Promoter Mutation That Decreases Antibiotic Susceptibility. mBio 2022, 13, e00276-22. [Google Scholar] [CrossRef] [PubMed]
  126. Da Costa-Lourenço, A.P.R.; dos Santos, K.T.B.; Moreira, B.M.; Fracalanzza, S.E.L.; Bonelli, R.R. Antimicrobial resistance in Neisseria gonorrhoeae: History, molecular mechanisms and epidemiological aspects of an emerging global threat. Braz. J. Microbiol. 2017, 48, 617–628. [Google Scholar] [CrossRef]
  127. Low, N.; Unemo, M. Molecular tests for the detection of antimicrobial resistant Neisseria gonorrhoeae: When, where, and how to use? Curr. Opin. Infect. Dis. 2016, 29, 45–51. [Google Scholar] [CrossRef] [Green Version]
  128. Enhanced Gonococcal Antimicrobial Surveillance Programme (EGASP): GENERAL PROTOCOL. Available online: https://www.who.int/publications/i/item/9789240021341 (accessed on 5 May 2022).
  129. WHO New Partnership to Combat Antimicrobial Resistance in Gonorrhoea. Available online: https://www.who.int/news/item/08-12-2021-new-partnership-to-combat-antimicrobial-resistance-in-gonorrhoea (accessed on 19 July 2022).
  130. Vică, M.L.; Glevitzky, I.; Glevitzky, M.; Siserman, C.V.; Matei, H.V.; Teodoru, C.A. Antibacterial Activity of Propolis Extracts from the Central Region of Romania against Neisseria gonorrhoeae. Antibiotics 2021, 10, 689. [Google Scholar] [CrossRef]
  131. Umaru, I.J.; Umaru, H.A.; Ahuchaogu, C.E.; Usman, M.A. Phytochemical, Characterization and Antimicrobial Studies of Molineria Capitulata Fruits Essential Oil Against Multidrug Resistance Pathogens. Solid State Technol. 2020, 63, 149–152. [Google Scholar]
  132. Soliman, F.M.; Fathy, M.M.; Salama, M.M.; Saber, F.R. Comparative study of the volatile oil content and antimicrobial activity of Psidium guajava L. and Psidium cattleianum Sabine leaves. Bull. Fac. Pharm. Cairo Univ. 2016, 54, 219–225. [Google Scholar] [CrossRef] [Green Version]
  133. Zaman, G.S. Antibacterial potency of extracted essential oils of some plant species against common gram-positive and gram-negative bacteria. King Khalid Univ. J. Health Sci. 2021, 6, 18. [Google Scholar] [CrossRef]
  134. Elghwaji, W.; El-Sayed, A.M.; El-Deeb, K.S.; ElSayed, A.M. Chemical Composition, Antimicrobial and Antitumor Potentiality of Essential Oil of Ferula Tingitana L. Apiaceae Grow in Libya. Pharmacogn. Mag. 2017, 13 (Suppl. S3), S446–S451. [Google Scholar] [CrossRef] [PubMed]
  135. Sholpan, A.; Lamas, A.; Cepeda, A.; Franco, C.M. Salmonella spp. quorum sensing: An overview from environmental persistence to host cell invasion. AIMS Microbiol. 2021, 7, 238–256. [Google Scholar] [CrossRef] [PubMed]
  136. Khademi, F.; Vaez, H.; Ghanbari, F.; Arzanlou, M.; Mohammadshahi, J.; Sahebkar, A. Prevalence of fluoroquinolone-resistant Salmonella serotypes in Iran: A meta-analysis. Pathog. Global Health 2020, 114, 16–29. [Google Scholar] [CrossRef] [PubMed]
  137. Fuche, F.J.; Sow, O.; Simon, R.; Tennant, S.M. Salmonella Serogroup C: Current Status of Vaccines and Why They Are Needed. Clin. Vaccine Immunol. 2016, 23, 737–745. [Google Scholar] [CrossRef] [Green Version]
  138. Cui, M.; Zhang, P.; Li, J.; Sun, C.; Song, L.; Zhang, C.; Zhao, Q.; Wu, C. Prevalence and Characterization of Fluoroquinolone Resistant Salmonella Isolated from an Integrated Broiler Chicken Supply Chain. Front. Microbiol. 2019, 10, 1865. [Google Scholar] [CrossRef] [Green Version]
  139. Kurtz, J.R.; Goggins, J.A.; McLachlan, J.B. Salmonella infection: Interplay between the bacteria and host immune system. Immunol. Lett. 2017, 190, 42–50. [Google Scholar] [CrossRef]
  140. Mellou, K.; Gkova, M.; Panagiotidou, E.; Tzani, M.; Sideroglou, T.; Mandilara, G. Diversity and Resistance Profiles of Human Non-typhoidal Salmonella spp. in Greece, 2003–2020. Antibiotics 2021, 10, 983. [Google Scholar] [CrossRef]
  141. Cuypers, W.L.; Jacobs, J.; Wong, V.; Klemm, E.J.; Deborggraeve, S.; Van Puyvelde, S. Fluoroquinolone resistance in Salmonella: Insights by whole-genome sequencing. Microb. Genom. 2018, 4, 1–9. [Google Scholar] [CrossRef]
  142. Parisi, A.; Crump, J.A.; Stafford, R.; Glass, K.; Howden, B.; Kirk, M. Increasing incidence of invasive nontyphoidal Salmonella infections in Queensland, Australia, 2007–2016. PLoS Neglected Trop. Dis. 2019, 13, e0007187. [Google Scholar] [CrossRef] [PubMed]
  143. Stanaway, J.D.; Parisi, A.; Sarkar, K.; Blacker, B.F.; Reiner, R.C.; Hay, S.I.; Nixon, M.R.; Dolecek, C.; James, S.L.; Mokdad, A.H.; et al. The global burden of non-typhoidal Salmonella invasive disease: A systematic analysis for the Global Burden of Disease Study 2017. Lancet Infect. Dis. 2019, 19, 1312–1324. [Google Scholar] [CrossRef] [Green Version]
  144. Castro-Vargas, R.E.; Herrera-Sánchez, M.P.; Rodríguez-Hernández, R.; Rondón-Barragán, I.S. Antibiotic resistance in Salmonella spp. isolated from poultry: A global overview. Vet. World 2020, 13, 2070–2084. [Google Scholar] [CrossRef]
  145. Jajere, S.M. A review of Salmonella enterica with particular focus on the pathogenicity and virulence factors, host specificity and antimicrobial resistance including multidrug resistance. Vet. World 2019, 12, 504–521. [Google Scholar] [CrossRef] [Green Version]
  146. Mthembu, T.P.; Zishiri, O.T.; El Zowalaty, M.E. Molecular Detection of Multidrug-Resistant Salmonella Isolated from Livestock Production Systems in South Africa. Infect. Drug Resist. 2019, 12, 3537–3548. [Google Scholar] [CrossRef] [Green Version]
  147. Pribul, B.R.; Festivo, M.L.; Rodrigues, M.S.; Costa, R.G.; Rodrigues, E.C.d.P.; de Souza, M.M.S.; Rodrigues, D.d.P. Characteristics of Quinolone Resistance in Salmonella spp. Isolates from the Food Chain in Brazil. Front. Microbiol. 2017, 8, 299. [Google Scholar] [CrossRef] [PubMed]
  148. Yin, X.; Dudley, E.G.; Pinto, C.N.; M’Ikanatha, N.M. Fluoroquinolone sales in food animals and quinolone resistance in non-typhoidal Salmonella from retail meats: United States, 2009–2018. J. Glob. Antimicrob. Resist. 2022, 29, 163–167. [Google Scholar] [CrossRef]
  149. Chang, M.-X.; Zhang, J.-F.; Sun, Y.-H.; Li, R.-S.; Lin, X.-L.; Yang, L.; Webber, M.A.; Jiang, H.-X. Contribution of Different Mechanisms to Ciprofloxacin Resistance in Salmonella spp. Front. Microbiol. 2021, 12, 663731. [Google Scholar] [CrossRef]
  150. Song, Q.; Xu, Z.; Gao, H.; Zhang, D. Overview of the development of quinolone resistance in Salmonella species in China, 2005–2016. Infect. Drug Resist. 2018, 11, 267–274. [Google Scholar] [CrossRef] [Green Version]
  151. Lee, A.S.; de Lencastre, H.; Garau, J.; Kluytmans, J.; Malhotra-Kumar, S.; Peschel, A.; Harbarth, S. Methicillin-resistant Staphylococcus aureus. Nat. Rev. Dis. Prim. 2018, 4, 18033. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  152. Guo, Y.; Song, G.; Sun, M.; Wang, J.; Wang, Y. Prevalence and Therapies of Antibiotic-Resistance in Staphylococcus aureus. Front. Cell. Infect. Microbiol. 2020, 10, 107. [Google Scholar] [CrossRef] [Green Version]
  153. Kwiecinski, J.M.; Horswill, A.R. Staphylococcus aureus bloodstream infections: Pathogenesis and regulatory mechanisms. Curr. Opin. Microbiol. 2020, 53, 51–60. [Google Scholar] [CrossRef] [PubMed]
  154. Lakhundi, S.; Zhang, K. Methicillin-Resistant Staphylococcus aureus: Molecular Characterization, Evolution, and Epidemiology. Clin. Microbiol. Rev. 2018, 31, e00020-18. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  155. Cheung, G.Y.C.; Bae, J.S.; Otto, M. Pathogenicity and virulence of Staphylococcus aureus. Virulence 2021, 12, 547–569. [Google Scholar] [CrossRef]
  156. Harkins, C.P.; Pichon, B.; Doumith, M.; Parkhill, J.; Westh, H.; Tomasz, A.; de Lencastre, H.; Bentley, S.D.; Kearns, A.M.; Holden, M.T.G. Methicillin-resistant Staphylococcus aureus emerged long before the introduction of methicillin into clinical practice. Genome Biol. 2017, 18, 130. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  157. Mediavilla, J.R.; Chen, L.; Mathema, B.; Kreiswirth, B.N. Global epidemiology of community-associated methicillin resistant Staphylococcus aureus (CA-MRSA). Curr. Opin. Microbiol. 2012, 15, 588–595. [Google Scholar] [CrossRef]
  158. Deadly Staph Infections Still Threaten the U.S. CDC Online Newsroom. CDC. Available online: https://www.cdc.gov/media/releases/2019/p0305-deadly-staph-infections.html (accessed on 18 May 2022).
  159. European Centre for Disease Prevention and Control—Partners and Networks. Available online: https://www.ecdc.europa.eu/en/about-us/ecdcs-partnerships-and-networks (accessed on 20 November 2022).
  160. Ito, T.; Kuwahara-Arai, K.; Katayama, Y.; Uehara, Y.; Han, X.; Kondo, Y.; Hiramatsu, K. Staphylococcal Cassette Chromosome mec (SCCmec) Analysis of MRSA. Methods Mol. Biol. 2014, 1085, 131–148. [Google Scholar] [CrossRef]
  161. Mlynarczyk-Bonikowska, B.; Kowalewski, C.; Krolak-Ulinska, A.; Marusza, W. Molecular Mechanisms of Drug Resistance in Staphylococcus aureus. Int. J. Mol. Sci. 2022, 23, 8088. [Google Scholar] [CrossRef]
  162. Youssef, C.R.B.; Kadry, A.A.; El-Ganiny, A.M. Investigating the relation between resistance pattern and type of Staphylococcal cassette chromosome mec (SCCmec) in methicillin-resistant Staphylococcus aureus. Iran. J. Microbiol. 2022, 14, 56–66. [Google Scholar] [CrossRef]
  163. McCarthy, H.; Rudkin, J.; Black, N.; Egallagher, L.; O’Neill, E.; O’Gara, J.P. Methicillin resistance and the biofilm phenotype in Staphylococcus aureus. Front. Cell. Infect. Microbiol. 2015, 5, 1. [Google Scholar] [CrossRef] [Green Version]
  164. Lin, C.-Y.; Wang, J.-H.; Lin, K.-H.; Ho, Y.-L.; Ho, C.-M. Methicillin-resistant Staphylococcus aureus with reduced vancomycin susceptibility in Taiwan. Ci Ji Yi Xue Za Zhi = Tzu Chi Med. J. 2018, 30, 135–140. [Google Scholar] [CrossRef]
  165. Sakoulas, G.; Moellering, J.R.C.; Eliopoulos, G.M. Adaptation of Methicillin-Resistant Staphylococcus aureus in the Face of Vancomycin Therapy. Clin. Infect. Dis. 2006, 42, S40–S50. [Google Scholar] [CrossRef]
  166. Liu, C.; Bayer, A.; Cosgrove, S.E.; Daum, R.S.; Fridkin, S.K.; Gorwitz, R.J.; Kaplan, S.L.; Karchmer, A.W.; Levine, D.P.; Murray, B.E.; et al. Clinical practice guidelines by the Infectious Diseases Society of America for the treatment of methicillin-resistant Staphylococcus aureus infections in adults and children. Clin. Infect. Dis. 2011, 52, e18–e55. [Google Scholar] [CrossRef] [Green Version]
  167. Panthee, S.; Hamamoto, H.; Paudel, A.; Sekimizu, K. Genomic analysis of vancomycin-resistant Staphylococcus aureus VRS3b and its comparison with other VRSA isolates. Drug Discov. Ther. 2017, 11, 78–83. [Google Scholar] [CrossRef] [Green Version]
  168. McGuinness, W.A.; Malachowa, N.; DeLeo, F.R. Vancomycin Resistance in Staphylococcus aureus. Yale J. Biol. Med. 2017, 90, 133–152. [Google Scholar]
  169. Oo, T.; Saiboonjan, B.; Srijampa, S.; Srisrattakarn, A.; Sutthanut, K.; Tavichakorntrakool, R.; Chanawong, A.; Lulitanond, A.; Tippayawat, P. Inhibition of Bacterial Efflux Pumps by Crude Extracts and Essential Oil from Myristica fragrans Houtt. (Nutmeg) Seeds against Methicillin-Resistant Staphylococcus aureus. Molecules 2021, 26, 4662. [Google Scholar] [CrossRef] [PubMed]
  170. Alharbi, N.S.; Khaled, J.M.; Alzaharni, K.E.; Mothana, R.A.; Alsaid, M.S.; Alhoshan, M.; Dass, L.A.; Kadaikunnan, S.; Alobaidi, A.S. Effects of Piper cubeba L. essential oil on methicillin-resistant Staphylococcus aureus: An AFM and TEM study. J. Mol. Recognit. 2016, 30, e2564. [Google Scholar] [CrossRef] [PubMed]
  171. Piasecki, B.; Biernasiuk, A.; Skiba, A.; Skalicka-Woźniak, K.; Ludwiczuk, A. Composition, Anti-MRSA Activity and Toxicity of Essential Oils from Cymbopogon Species. Molecules 2021, 26, 7542. [Google Scholar] [CrossRef]
  172. Merghni, A.; Noumi, E.; Hadded, O.; Dridi, N.; Panwar, H.; Ceylan, O.; Mastouri, M.; Snoussi, M. Assessment of the antibiofilm and antiquorum sensing activities of Eucalyptus globulus essential oil and its main component 1,8-cineole against methicillin-resistant Staphylococcus aureus strains. Microb. Pathog. 2018, 118, 74–80. [Google Scholar] [CrossRef]
  173. Khamis, A.-D.S.; Chai, L.C. Chemical and Antimicrobial Analyses of Juniperus chinensis and Juniperus seravschanica Essential Oils and Comparison with Their Methanolic Crude Extracts. Int. J. Anal. Chem. 2021, 2021, 9937522. [Google Scholar] [CrossRef]
  174. Predoi, D.; Groza, A.; Iconaru, S.L.; Predoi, G.; Barbuceanu, F.; Guegan, R.; Motelica-Heino, M.S.; Cimpeanu, C. Properties of Basil and Lavender Essential Oils Adsorbed on the Surface of Hydroxyapatite. Materials 2018, 11, 652. [Google Scholar] [CrossRef] [Green Version]
  175. Predoi, D.; Iconaru, S.L.; Buton, N.; Badea, M.L.; Marutescu, L. Antimicrobial Activity of New Materials Based on Lavender and Basil Essential Oils and Hydroxyapatite. Nanomaterials 2018, 8, 291. [Google Scholar] [CrossRef] [Green Version]
  176. Badea, M.L.; Iconaru, S.L.; Groza, A.; Chifiriuc, M.C.; Beuran, M.; Predoi, D. Peppermint Essential Oil-Doped Hydroxyapatite Nanoparticles with Antimicrobial Properties. Molecules 2019, 24, 2169. [Google Scholar] [CrossRef] [Green Version]
  177. Mouwakeh, A.; Kincses, A.; Nové, M.; Mosolygó, T.; Mohácsi-Farkas, C.; Kiskó, G.; Spengler, G. Nigella sativa essential oil and its bioactive compounds as resistance modifiers against Staphylococcus aureus. Phytother. Res. 2019, 33, 1010–1018. [Google Scholar] [CrossRef] [PubMed]
  178. De Moura, D.F.; Rocha, T.A.; Barros, D.D.M.; da Silva, M.M.; Santana, M.D.S.; Neta, B.M.; Cavalcanti, I.M.F.; Martins, R.D.; da Silva, M.V. Evaluation of the antioxidant, antibacterial, and antibiofilm activity of the sesquiterpene nerolidol. Arch. Microbiol. 2021, 203, 4303–4311. [Google Scholar] [CrossRef]
  179. Chan, W.-K.; Tan, L.T.-H.; Chan, K.-G.; Lee, L.-H.; Goh, B.-H. Nerolidol: A Sesquiterpene Alcohol with Multi-Faceted Pharmacological and Biological Activities. Molecules 2016, 21, 529. [Google Scholar] [CrossRef] [Green Version]
  180. De Carvalho, R.B.F.; De Almeida, A.A.C.; Campelo, N.B.; Lellis, D.R.O.D.; Nunes, L.C.C. Nerolidol and its Pharmacological Application in Treating Neurodegenerative Diseases: A Review. Recent Patents Biotechnol. 2018, 12, 158–168. [Google Scholar] [CrossRef]
  181. Jaradat, N.; Hawash, M.; Abualhasan, M.N.; Qadi, M.; Ghanim, M.; Massarwy, E.; Abu Ammar, S.; Zmero, N.; Arar, M.; Hussein, F.; et al. Spectral characterization, antioxidant, antimicrobial, cytotoxic, and cyclooxygenase inhibitory activities of Aloysia citriodora essential oils collected from two Palestinian regions. BMC Complement. Med. Ther. 2021, 21, 143. [Google Scholar] [CrossRef] [PubMed]
  182. Dadashi, M.; Hajikhani, B.; Darban-Sarokhalil, D.; van Belkum, A.; Goudarzi, M. Mupirocin resistance in Staphylococcus aureus: A systematic review and meta-analysis. J. Glob. Antimicrob. Resist. 2019, 20, 238–247. [Google Scholar] [CrossRef] [PubMed]
  183. Sharara, S.L.; Maragakis, L.L.; Cosgrove, S.E. Decolonization of Staphylococcus aureus. Infect. Dis. Clin. N. Am. 2020, 35, 107–133. [Google Scholar] [CrossRef] [PubMed]
  184. Kwiatkowski, P.; Pruss, A.; Wojciuk, B.; Dołęgowska, B.; Wajs-Bonikowska, A.; Sienkiewicz, M.; Mężyńska, M.; Łopusiewicz, Ł. The Influence of Essential Oil Compounds on Antibacterial Activity of Mupirocin-Susceptible and Induced Low-Level Mupirocin-Resistant MRSA Strains. Molecules 2019, 24, 3105. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  185. Manzuoerh, R.; Farahpour, M.R.; Oryan, A.; Sonboli, A. Effectiveness of topical administration of Anethum graveolens essential oil on MRSA-infected wounds. Biomed. Pharmacother. 2018, 109, 1650–1658. [Google Scholar] [CrossRef]
  186. Mahboubi, M.; Feizabadi, M.M.; Khamechian, T.; Kazempour, N.; Zadeh, M.R.; Sasani, F.; Bekhradi, M. The Effect of Oliveria decumbens and Pelargonium graveolens on Healing of Infected Skin Wounds in Mice. World J. Plast. Surg. 2016, 5, 259–264. [Google Scholar]
  187. Chen, J.; Tang, C.; Zhang, R.; Ye, S.; Zhao, Z.; Huang, Y.; Xu, X.; Lan, W.; Yang, D. Metabolomics analysis to evaluate the antibacterial activity of the essential oil from the leaves of Cinnamomum camphora (Linn.) Presl. J. Ethnopharmacol. 2020, 253, 112652. [Google Scholar] [CrossRef] [PubMed]
  188. Rinschen, M.M.; Ivanisevic, J.; Giera, M.; Siuzdak, G. Identification of bioactive metabolites using activity metabolomics. Nat. Rev. Mol. Cell Biol. 2019, 20, 353–367. [Google Scholar] [CrossRef] [PubMed]
  189. Vasconcelos, S.E.C.B.; Melo, H.M.; Cavalcante, T.T.A.; Júnior, F.E.A.C.; De Carvalho, M.G.; Menezes, F.G.R.; De Sousa, O.V.; Costa, R.A. Plectranthus amboinicus essential oil and carvacrol bioactive against planktonic and biofilm of oxacillin- and vancomycin-resistant Staphylococcus aureus. BMC Complement. Altern. Med. 2017, 17, 462. [Google Scholar] [CrossRef] [Green Version]
  190. Ben Abdallah, F.; Lagha, R.; Gaber, A. Biofilm Inhibition and Eradication Properties of Medicinal Plant Essential Oils against Methicillin-Resistant Staphylococcus aureus Clinical Isolates. Pharmaceuticals 2020, 13, 369. [Google Scholar] [CrossRef]
  191. Gómez-Sequeda, N.; Cáceres, M.; Stashenko, E.E.; Hidalgo, W.; Ortiz, C. Antimicrobial and Antibiofilm Activities of Essential Oils against Escherichia coli O157:H7 and Methicillin-Resistant Staphylococcus aureus (MRSA). Antibiotics 2020, 9, 730. [Google Scholar] [CrossRef]
  192. Ekhtelat, M.; Borujeni, F.K.; Siahpoosh, A.; Ameri, A. Chemical composition and antibacterial effects of some essential oils individually and in combination with sodium benzoate against methicillin-resistant Staphylococcus aureus and Yersinia enterocolitica. Vet. Res. Forum. 2020, 11, 333–338. [Google Scholar] [CrossRef]
  193. Tang, C.; Chen, J.; Zhang, L.; Zhang, R.; Zhang, S.; Ye, S.; Zhao, Z.; Yang, D. Exploring the antibacterial mechanism of essential oils by membrane permeability, apoptosis and biofilm formation combination with proteomics analysis against methicillin-resistant Staphylococcus aureus. Int. J. Med. Microbiol. 2020, 310, 151435. [Google Scholar] [CrossRef]
  194. Rubini, D.; Banu, S.F.; Nisha, P.; Murugan, R.; Thamotharan, S.; Percino, M.J.; Subramani, P.; Nithyanand, P. Essential oils from unexplored aromatic plants quench biofilm formation and virulence of Methicillin resistant Staphylococcus aureus. Microb. Pathog. 2018, 122, 162–173. [Google Scholar] [CrossRef] [PubMed]
  195. Utegenova, G.A.; Pallister, K.B.; Kushnarenko, S.V.; Özek, G.; Özek, T.; Abidkulova, K.T.; Kirpotina, L.N.; Schepetkin, I.A.; Quinn, M.T.; Voyich, J.M. Chemical Composition and Antibacterial Activity of Essential Oils from Ferula L. Species against Methicillin-Resistant Staphylococcus aureus. Molecules 2018, 23, 1679. [Google Scholar] [CrossRef] [Green Version]
  196. Sreepian, A.; Popruk, S.; Nutalai, D.; Phutthanu, C.; Sreepian, P.M. Antibacterial Activities and Synergistic Interaction of Citrus Essential Oils and Limonene with Gentamicin against Clinically Isolated Methicillin-Resistant Staphylococcus aureus. Sci. World J. 2022, 2022, 8418287. [Google Scholar] [CrossRef] [PubMed]
  197. Dalli, M.; Azizi, S.-E.; Benouda, H.; Azghar, A.; Tahri, M.; Bouammali, B.; Maleb, A.; Gseyra, N. Molecular Composition and Antibacterial Effect of Five Essential Oils Extracted from Nigella sativa L. Seeds against Multidrug-Resistant Bacteria: A Comparative Study. Evid.-Based Complement. Altern. Med. 2021, 2021, 6643765. [Google Scholar] [CrossRef] [PubMed]
  198. Tang, C.; Chen, J.; Zhou, Y.; Ding, P.; He, G.; Zhang, L.; Zhao, Z.; Yang, D. Exploring antimicrobial mechanism of essential oil of Amomum villosum Lour through metabolomics based on gas chromatography-mass spectrometry in methicillin-resistant Staphylococcus aureus. Microbiol. Res. 2020, 242, 126608. [Google Scholar] [CrossRef]
  199. Al-Maharik, N.; Jaradat, N. Phytochemical Profile, Antimicrobial, Cytotoxic, and Antioxidant Activities of Fresh and Air-Dried Satureja nabateorum Essential Oils. Molecules 2021, 27, 125. [Google Scholar] [CrossRef]
  200. Donadu, M.G.; Le, N.T.; Ho, D.V.; Doan, T.Q.; Le, A.T.; Raal, A.; Usai, M.; Marchetti, M.; Sanna, G.; Madeddu, S.; et al. Phytochemical Compositions and Biological Activities of Essential Oils from the Leaves, Rhizomes and Whole Plant of Hornstedtia bella Škorničk. Antibiotics 2020, 9, 334. [Google Scholar] [CrossRef]
  201. Bay, M.; Souza de Oliveira, J.V.; Ademar Sales Junior, P.; Fonseca Murta, S.M.; dos Santos, A.R.; dos Ssantos Bastos, I.; Puccinelli Orlandi, P.; Teixeira de Sousa Junior, P. In Vitro Trypanocidal and Antibacterial Activities of Essential Oils from Four Species of the Family Annonaceae. Chem. Biodivers. 2019, 16, e1900359. [Google Scholar] [CrossRef]
  202. Aelenei, P.; Rimbu, C.M.; Guguianu, E.; Dimitriu, G.; Aprotosoaie, A.C.; Brebu, M.; Horhogea, C.E.; Miron, A. Coriander essential oil and linalool—Interactions with antibiotics against Gram-positive and Gram-negative bacteria. Lett. Appl. Microbiol. 2018, 68, 156–164. [Google Scholar] [CrossRef]
  203. Leal, A.L.A.B.; Bezerra, C.F.; Confortin, C.; da Silva, L.E.; Marinho, E.M.; Marinho, M.M.; Vasconcelos, M.A.; da Silva, T.G.; Marinho, E.S.; Teixeira, A.M.R.; et al. Chemical composition and potentiating action of Norfloxacin mediated by the essential oil of Piper caldense C.D.C. against Staphylococcus aureus strains overexpressing efflux pump genes. Arch. Microbiol. 2021, 203, 4727–4736. [Google Scholar] [CrossRef]
  204. De Jesus, G.S.; Micheletti, A.C.; Takahashi, K.M.; Matayoshi, T.; Pott, A.; Yoshida, N.C. Antimicrobial potential of Pectis substriata essential oil (Asteraceae) against drug-resistant Staphylococcus strains. An. Acad. Bras. Cienc. 2020, 92, e20200456. [Google Scholar] [CrossRef]
  205. Cui, Z.-H.; He, H.-L.; Wu, S.-B.; Dong, C.-L.; Lu, S.-Y.; Shan, T.-J.; Fang, L.-X.; Liao, X.-P.; Liu, Y.-H.; Sun, J. Rapid Screening of Essential Oils as Substances Which Enhance Antibiotic Activity Using a Modified Well Diffusion Method. Antibiotics 2021, 10, 463. [Google Scholar] [CrossRef] [PubMed]
  206. Fahed, L.; Stien, D.; Ouaini, N.; Eparvier, V.; El Beyrouthy, M. Chemical Diversity and Antimicrobial Activity of Salvia multicaulis VahlEssential Oils. Chem. Biodivers. 2016, 13, 591–595. [Google Scholar] [CrossRef] [Green Version]
  207. Mahdavi, B.; Yaacob, W.A.; Din, L.B. Chemical composition, antioxidant, and antibacterial activity of essential oils from Etlingera sayapensis A.D. Poulsen & Ibrahim. Asian Pac. J. Trop. Med. 2017, 10, 819–826. [Google Scholar] [CrossRef]
  208. Gadisa, E.; Weldearegay, G.; Desta, K.; Tsegaye, G.; Hailu, S.; Jote, K.; Takele, A. Combined antibacterial effect of essential oils from three most commonly used Ethiopian traditional medicinal plants on multidrug resistant bacteria. BMC Complement. Altern. Med. 2019, 19, 24. [Google Scholar] [CrossRef]
  209. Bano, S.; Intisar, A.; Rauf, M.; Ghaffar, A.; Yasmeen, F.; Zaman, W.-U.; Intisar, U.; Kausar, G.; Muhammad, N.; Aamir, A. Comparative analysis of oil composition and antibacterial activity of aerial parts of Terminalia arjuna (Roxb.). Nat. Prod. Res. 2019, 34, 1311–1314. [Google Scholar] [CrossRef] [PubMed]
  210. Ding, L.; Hertweck, C. Oxygenated Geosmins and Plant-like Eudesmanes from a Bacterial Mangrove Endophyte. J. Nat. Prod. 2020, 83, 2207–2211. [Google Scholar] [CrossRef] [PubMed]
  211. Jaradat, N.; Adwan, L.; K’aibni, S.; Zaid, A.N.; Shtaya, M.J.Y.; Shraim, N.; Assali, M. Variability of Chemical Compositions and Antimicrobial and Antioxidant Activities of Ruta chalepensis Leaf Essential Oils from Three Palestinian Regions. BioMed Res. Int. 2017, 2017, 2672689. [Google Scholar] [CrossRef] [Green Version]
  212. Kot, B.; Wierzchowska, K.; Grużewska, A.; Lohinau, D. The effects of selected phytochemicals on biofilm formed by five methicillin-resistant Staphylococcus aureus. Nat. Prod. Res. 2017, 32, 1299–1302. [Google Scholar] [CrossRef]
  213. Kwiatkowski, P.; Łopusiewicz, Ł.; Pruss, A.; Kostek, M.; Sienkiewicz, M.; Bonikowski, R.; Wojciechowska-Koszko, I.; Dołęgowska, B. Antibacterial Activity of Selected Essential Oil Compounds Alone and in Combination with β-Lactam Antibiotics Against MRSA Strains. Int. J. Mol. Sci. 2020, 21, 7106. [Google Scholar] [CrossRef] [PubMed]
  214. Cui, H.; Li, W.; Li, C.; Vittayapadung, S.; Lin, L. Liposome containing cinnamon oil with antibacterial activity against methicillin-resistant Staphylococcus aureus biofilm. Biofouling 2016, 32, 215–225. [Google Scholar] [CrossRef]
  215. Perez, A.P.; Perez, N.; Lozano, C.M.S.; Altube, M.J.; de Farias, M.A.; Portugal, R.V.; Buzzola, F.; Morilla, M.J.; Romero, E.L. The anti MRSA biofilm activity of Thymus vulgaris essential oil in nanovesicles. Phytomedicine 2019, 57, 339–351. [Google Scholar] [CrossRef]
  216. Farias, K.S.; Kato, N.N.; Boaretto, A.G.; Weber, J.I.; Brust, F.R.; Alves, F.M.; Tasca, T.; Macedo, A.J.; Silva, D.B.; Carollo, C.A. Nectandra as a renewable source for (+)-α-bisabolol, an antibiofilm and anti-Trichomonas vaginalis compound. Fitoterapia 2019, 136, 104179. [Google Scholar] [CrossRef] [PubMed]
  217. Eid, A.M.; Issa, L.; Al-Kharouf, O.; Jaber, R.; Hreash, F. Development of Coriandrum sativum Oil Nanoemulgel and Evaluation of Its Antimicrobial and Anticancer Activity. BioMed Res. Int. 2021, 2021, 5247816. [Google Scholar] [CrossRef]
  218. Bakó, C.; Balázs, V.L.; Takács, G.; Pallos, J.P.; Pál, S.; Kocsis, B.; Pethő, D.R.; Horváth, G. Combination of Analytical and Statistical Methods in Order to Optimize Antibacterial Activity of Clary Sage Supercritical Fluid Extracts. Molecules 2021, 26, 6449. [Google Scholar] [CrossRef] [PubMed]
  219. Jamil, B.; Abbasi, R.; Abbasi, S.; Imran, M.; Khan, S.U.; Ihsan, A.; Javed, S.; Bokhari, H. Encapsulation of Cardamom Essential Oil in Chitosan Nano-composites: In-vitro Efficacy on Antibiotic-Resistant Bacterial Pathogens and Cytotoxicity Studies. Front. Microbiol. 2016, 7, 1580. [Google Scholar] [CrossRef]
  220. Khoury, M.; El Beyrouthy, M.; Ouaini, N.; Eparvier, V.; Stien, D. Hirtellina lobelii DC. essential oil, its constituents, its combination with antimicrobial drugs and its mode of action. Fitoterapia 2019, 133, 130–136. [Google Scholar] [CrossRef] [PubMed]
  221. Viktorová, J.; Stupák, M.; Řehořová, K.; Dobiasová, S.; Hoang, L.; HajšLová, J.; Van Thanh, T.; Van Tri, L.; Van Tuan, N.; Ruml, T. Lemon Grass Essential Oil does not Modulate Cancer Cells Multidrug Resistance by Citral—Its Dominant and Strongly Antimicrobial Compound. Foods 2020, 9, 585. [Google Scholar] [CrossRef]
  222. Taha, A.M.; Eldahshan, O.A. Chemical Characteristics, Antimicrobial and Cytotoxic Activities of the Essential Oil of Egyptian Cinnamomum glanduliferum Bark. Chem. Biodivers. 2017, 14, e1600443. [Google Scholar] [CrossRef]
  223. Jaradat, N.; Adwan, L.; K’Aibni, S.; Shraim, N.; Zaid, A.N. Chemical composition, anthelmintic, antibacterial and antioxidant effects of Thymus bovei essential oil. BMC Complement. Altern. Med. 2016, 16, 418. [Google Scholar] [CrossRef] [Green Version]
  224. Lahmar, A.; Bedoui, A.; Mokdad-Bzeouich, I.; Dhaouifi, Z.; Kalboussi, Z.; Cheraif, I.; Ghedira, K.; Chekir-Ghedira, L. Reversal of resistance in bacteria underlies synergistic effect of essential oils with conventional antibiotics. Microb. Pathog. 2017, 106, 50–59. [Google Scholar] [CrossRef]
  225. Shehadeh, M.; Jaradat, N.; Al-Masri, M.; Zaid, A.N.; Hussein, F.; Khasati, A.; Suaifan, G.; Darwish, R. Rapid, cost-effective and organic solvent-free production of biologically active essential oil from Mediterranean wild Origanum syriacum. Saudi Pharm. J. 2019, 27, 612–618. [Google Scholar] [CrossRef]
  226. Demirci, F.; Karaca, N.; Tekin, M.; Demirci, B. Anti-inflammatory and antibacterial evaluation of Thymus sipyleus Boiss. subsp. sipyleus var. sipyleus essential oil against rhinosinusitis pathogens. Microb. Pathog. 2018, 122, 117–121. [Google Scholar] [CrossRef] [PubMed]
  227. Salameh, N.; Shraim, N.; Jaradat, N.; El Masri, M.; Adwan, L.; K’aibni, S.; Alkowni, R.; Radwan, A.; AbuAlhasan, M. Screening of Antioxidant and Antimicrobial Activity of Micromeria fruticosa serpyllifolia Volatile Oils: A Comparative Study of Plants Collected from Different Regions of West Bank, Palestine. BioMed Res. Int. 2020, 2020, 4851879. [Google Scholar] [CrossRef] [PubMed]
  228. Jaradat, N.; Al-Maharik, N. Fingerprinting, Antimicrobial, Antioxidant, Anticancer, Cyclooxygenase and Metabolic Enzymes Inhibitory Characteristic Evaluations of Stachys viticina Boiss. Essential Oil. Molecules 2019, 24, 3880. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  229. Kwiatkowski, P.; Łopusiewicz, Ł.; Kostek, M.; Drozłowska, E.; Pruss, A.; Wojciuk, B.; Sienkiewicz, M.; Zielińska-Bliźniewska, H.; Dołęgowska, B. The Antibacterial Activity of Lavender Essential Oil Alone and In Combination with Octenidine Dihydrochloride against MRSA Strains. Molecules 2020, 25, 95. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  230. Noumi, E.; Merghni, A.; Alreshidi, M.M.; Haddad, O.; Akmadar, G.; De Martino, L.; Mastouri, M.; Ceylan, O.; Snoussi, M.; Al-Sieni, A.; et al. Chromobacterium violaceum and Pseudomonas aeruginosa PAO1: Models for Evaluating Anti-Quorum Sensing Activity of Melaleuca alternifolia Essential Oil and Its Main Component Terpinen-4-ol. Molecules 2018, 23, 2672. [Google Scholar] [CrossRef] [Green Version]
  231. Grădinaru, A.C.; Trifan, A.; Şpac, A.; Brebu, M.; Miron, A.; Aprotosoaie, A.C. Antibacterial activity of traditional spices against lower respiratory tract pathogens: Combinatorial effects of Trachyspermum ammi essential oil with conventional antibiotics. Lett. Appl. Microbiol. 2018, 67, 449–457. [Google Scholar] [CrossRef]
  232. Marino, A.; Nostro, A.; Mandras, N.; Roana, J.; Ginestra, G.; Miceli, N.; Taviano, M.F.; Gelmini, F.; Beretta, G.; Tullio, V. Evaluation of antimicrobial activity of the hydrolate of Coridothymus capitatus (L.) Reichenb. fil. (Lamiaceae) alone and in combination with antimicrobial agents. BMC Complement. Med. Ther. 2020, 20, 89. [Google Scholar] [CrossRef]
  233. Tadić, V.; Oliva, A.; Božović, M.; Cipolla, A.; De Angelis, M.; Vullo, V.; Garzoli, S.; Ragno, R. Chemical and Antimicrobial Analyses of Sideritis romana L. subsp. purpurea (Tal. ex Benth.) Heywood, an Endemic of the Western Balkan. Molecules 2017, 22, 1395. [Google Scholar] [CrossRef] [Green Version]
  234. Ramírez-Rueda, R.Y.; Marinho, J.; Salvador, M.J. Bioguided identification of antimicrobial compounds from Chrysopogon zizaniodes (L.) Roberty root essential oil. Futur. Microbiol. 2019, 14, 1179–1189. [Google Scholar] [CrossRef]
  235. Brun, P.; Bernabè, G.; Filippini, R.; Piovan, A. In Vitro Antimicrobial Activities of Commercially Available Tea Tree (Melaleuca alternifolia) Essential Oils. Curr. Microbiol. 2018, 76, 108–116. [Google Scholar] [CrossRef] [PubMed]
  236. Jaradat, N.; Zaid, A.; Abuzant, A.; Shawahna, R. Investigation the efficiency of various methods of volatile oil extraction from Trichodesma africanum and their impact on the anti-oxidant and anti-microbial activities. J. Intercult. Ethnopharmacol. 2016, 5, 250–256. [Google Scholar] [CrossRef] [PubMed]
  237. Saidi, M.; Sadeghifard, N.; Kazemian, H.; Sekawi, Z.; Badakhsh, B.; Friadian, S.; Ghafourian, S. Ex Vivo Evaluation of Thymus daenensis as an Antioxidant and Antibacterial Medicinal Herb. Drug Res. 2016, 66, 657–659. [Google Scholar] [CrossRef] [PubMed]
  238. Wang, B.; Huang, W.; Zhou, J.; Tang, X.; Chen, Y.; Peng, C.; Han, B. Drug design based on pentaerythritol tetranitrate reductase: Synthesis and antibacterial activity of Pogostone derivatives. Org. Biomol. Chem. 2017, 15, 6548–6556. [Google Scholar] [CrossRef] [Green Version]
Figure 1. PRISMA 2020 flow diagram for new systematic reviews which included searches of databases and registers only: following database identification, duplicates were removed and the remaining records were screened for meeting the inclusion criteria. Retrieval of the full text was not possible for all publications. Before extracting the data from the studies, a final assessment was carried out to eliminate publications whose findings were ambiguous, according to the exclusion criteria.
Figure 1. PRISMA 2020 flow diagram for new systematic reviews which included searches of databases and registers only: following database identification, duplicates were removed and the remaining records were screened for meeting the inclusion criteria. Retrieval of the full text was not possible for all publications. Before extracting the data from the studies, a final assessment was carried out to eliminate publications whose findings were ambiguous, according to the exclusion criteria.
Ijms 24 09727 g001
Table 1. Top 5 countries in terms of AMR to HPPs, according to One Health Trust’s Resistance Map [31].
Table 1. Top 5 countries in terms of AMR to HPPs, according to One Health Trust’s Resistance Map [31].
BacteriaAB ResistanceYearResistant Strains Proportion (%)Country
E. faeciumVancomycin201769 (62–75)Argentina
201668 (65–71)USA
201666 (60–71)Taiwan
201960 (52–67)Serbia
201251 (42–60)Venezuela
S. aureusMethicillin201988 (77–95)Egypt
201773 (69–77)Vietnam
201968 (66–69)India
201866 (60–72)Nigeria
201965 (59–70)Pakistan
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

Romanescu, M.; Oprean, C.; Lombrea, A.; Badescu, B.; Teodor, A.; Constantin, G.D.; Andor, M.; Folescu, R.; Muntean, D.; Danciu, C.; et al. Current State of Knowledge Regarding WHO High Priority Pathogens—Resistance Mechanisms and Proposed Solutions through Candidates Such as Essential Oils: A Systematic Review. Int. J. Mol. Sci. 2023, 24, 9727. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24119727

AMA Style

Romanescu M, Oprean C, Lombrea A, Badescu B, Teodor A, Constantin GD, Andor M, Folescu R, Muntean D, Danciu C, et al. Current State of Knowledge Regarding WHO High Priority Pathogens—Resistance Mechanisms and Proposed Solutions through Candidates Such as Essential Oils: A Systematic Review. International Journal of Molecular Sciences. 2023; 24(11):9727. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24119727

Chicago/Turabian Style

Romanescu, Mirabela, Camelia Oprean, Adelina Lombrea, Bianca Badescu, Ana Teodor, George D. Constantin, Minodora Andor, Roxana Folescu, Delia Muntean, Corina Danciu, and et al. 2023. "Current State of Knowledge Regarding WHO High Priority Pathogens—Resistance Mechanisms and Proposed Solutions through Candidates Such as Essential Oils: A Systematic Review" International Journal of Molecular Sciences 24, no. 11: 9727. https://0-doi-org.brum.beds.ac.uk/10.3390/ijms24119727

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