Next Article in Journal
Glycated Albumin versus Glycated Hemoglobin as a Glycemic Indicator in Diabetic Patients on Peritoneal Dialysis
Previous Article in Journal
TNFSF15 Inhibits Blood Retinal Barrier Breakdown Induced by Diabetes
 
 
Font Type:
Arial Georgia Verdana
Font Size:
Aa Aa Aa
Line Spacing:
Column Width:
Background:
Review

New Is Old, and Old Is New: Recent Advances in Antibiotic-Based, Antibiotic-Free and Ethnomedical Treatments against Methicillin-Resistant Staphylococcus aureus Wound Infections

1
Institute of Pathogenic Biology, School of Basic Medical Sciences, Lanzhou University, Lanzhou 730000, China
2
Spinal Surgery Department, Affiliated Hospital of Gansu University of Chinese Medicines, Lanzhou 730020, China
*
Author to whom correspondence should be addressed.
Int. J. Mol. Sci. 2016, 17(5), 617; https://doi.org/10.3390/ijms17050617
Submission received: 31 December 2015 / Revised: 18 April 2016 / Accepted: 19 April 2016 / Published: 25 April 2016
(This article belongs to the Section Biochemistry)

Abstract

:
Staphylococcus aureus is the most common pathogen of wound infections. Thus far, methicillin-resistant S. aureus (MRSA) has become the major causative agent in wound infections, especially for nosocomial infections. MRSA infections are seldom eradicated by routine antimicrobial therapies. More concerning, some strains have become resistant to the newest antibiotics of last resort. Furthermore, horizontal transfer of a polymyxin resistance gene, mcr-1, has been identified in Enterobacteriaceae, by which resistance to the last group of antibiotics will likely spread rapidly. The worst-case scenario, “a return to the pre-antibiotic era”, is likely in sight. A perpetual goal for antibiotic research is the discovery of an antibiotic that lacks resistance potential, such as the recent discovery of teixobactin. However, when considering the issue from an ecological and evolutionary standpoint, it is evident that it is insufficient to solve the antibiotic dilemma through the use of antibiotics themselves. In this review, we summarized recent advances in antibiotic-based, antibiotic-free and ethnomedical treatments against MRSA wound infections to identify new clues to solve the antibiotic dilemma. One potential solution is to use ethnomedical drugs topically. Some ethnomedical drugs have been demonstrated to be effective antimicrobials against MRSA. A decline in antibiotic resistance can therefore be expected, as has been demonstrated when antibiotic-free treatments were used to limit the use of antibiotics. It is also anticipated that these drugs will have low resistance potential, although there is only minimal evidence to support this claim to date. More clinical trials and animal tests should be conducted on this topic.

1. Introduction

Staphylococcus aureus is the most common pathogen involved in skin and soft tissue infections and is the principal cause of surgical site infections (SSI) [1,2]. Wound infections often have negative impacts on patient outcomes, most commonly including a delay or deterioration of wound healing and potentially leading to sepsis. Furthermore, colonized staphylococcal cells are potential sources for cross-contamination, whereby S. aureus becomes a major nosocomial pathogen. Nosocomial infection is a major cause of surgical morbidity and mortality [3,4], and SSIs have a reported incidence rate of 2%–20% [5,6]. Evidence supports the use of S. aureus decolonization in surgical patients to prevent S. aureus infection, and this intervention has been associated with low rates of postoperative S. aureus infection. The staphylococcal carriage is most commonly eradicated by intranasal application of mupirocin either alone or in combination with antiseptic soaps or systemic antimicrobial agents [7]. However, the major cause of nosocomial infection is methicillin-resistant S. aureus (MRSA) [8,9,10], which is hard to eradicate [7] despite reports of some cases treated by warming therapy [4]. Furthermore, the efficacy of eradication in patients with community-associated MRSA has not been established, and the necessity of routine decolonization is not supported by data [7]. MRSA outbreaks have created a significant challenge for surgery and clinical practice in recent decades; the failure of traditional antimicrobial treatments has gradually become a worldwide problem [11,12,13], especially in the developing world [14]. Thus, effective therapeutic options to combat S. aureus infection, with an emphasis on MRSA, are urgently needed.

2. Antibiotics Developed to Combat MRSA

There are several therapeutic strategies, including clinical strategies and some still under development, to combat drug-resistant S. aureus: antibiotic-based treatments, alternative treatments, antibiotic-free treatments, immunotherapy, therapeutic vaccines and possible combinations of these options. [15,16,17]. Among these strategies, the use of antibiotics is effective and the most historically important. However, an unsettling trend has been witnessed in recent decades: increased antibiotic resistance and decreased antibiotic research and development [18,19,20]. From 1968 to 2003, only two novel classes of systemic antibiotics were developed: linezolid (2000) and daptomycin (2003) [19]. There are two primary contributors to this situation. Commercial interests with low profit motivations have failed to garner enthusiasm for antibiotic development because of their short-term use [13]. Second, the high level of difficulty in conducting clinical trials against drug-resistant strains dampens the hope of many candidates for final approval [18]. However, a new trend has emerged after the U.S. FDA (Food and Drug Administration)’s reboot of antibiotic development [20] involving an acceleration of drug approvals and documented antibiotic development. Some newly approved antibiotics (Table 1) are effective in preventing and controlling MRSA wound infections.
Antibiotic development is new, hard and time-consuming. However, a cost-effective and evidence-based strategy should be implemented to identify new therapeutic indications of clinically used drugs, i.e., drug repurposing, rescue and repositioning [65]. This approach, although old, is both easy and time-saving. Ebselen is an organoselenium compound [66], and celecoxib is a marketed inhibitor of cyclooxygenase-2 [67]. Both exhibit demonstrated antimicrobial activity, especially for MRSA and vancomycin-resistant S. aureus (VRSA). Furthermore, ebselen has been shown to be remarkably active, to significantly reduce established staphylococcal biofilms and to act synergistically with traditional antimicrobials [66]. Simvastatin has been predicted to exhibit activity as a topical antimicrobial against skin bacterial infection due to its broad-spectrum (including MRSA) antibacterial, anti-staphylococcal biofilm, anti-inflammatory and wound healing activities [68]; furthermore, simvastatin can also synergistically act with traditional antimicrobials. Recently, tamoxifen, a selective estrogen receptor modulator widely used for the treatment of breast cancer, has been repositioned to enhance MRSA clearance by boosting neutrophil bactericidal capacity [69].
However, there is an intrinsic obstacle to antibiotic development: antibiotics are the only drugs for which widespread use decreases their utility, i.e., “fighting back by microbes” [70]. There is almost no exception among the novel antibiotics (Table 1). A dilemma of antibiotics thus arises: antibiotics themselves are the cause of many healthcare-acquired infections (HCAI), further leading to “a return to the pre-antibiotic era”, as the World Health Organization has warned [71]. If this situation were to come to pass, once-curable infections would become fatal. Frighteningly, [72] horizontal transfer of a polymyxin resistance gene, mcr-1, has been identified in Enterobacteriaceae, and it is believed that this marks the beginning of the spread of resistance to the last group of antibiotics, leading to increased numbers of pan-drug-resistant strains. However, there is some hope. Teixobactin [73], discovered in a screen of uncultured bacteria, is a novel antibiotic that may not trigger the development of resistance. The biosynthetic gene cluster has been identified for future synthetic approaches. No resistant mutants of S. aureus were obtained by either serial passage at sub-MIC (minimum inhibitory concentration) levels of teixobactin or after culture on media with four-fold MIC of teixobactin. However, these results merely imply the rare occurrence of chromosomal mutation; as noted [72], a plasmid-mediated transfer mechanism contributes significantly to multi- and even pan-drug resistance. Furthermore, before it is introduced to clinical practice, drug resistance should be considered from an ecological and evolutionary view [74]. For example, a biofilm is a complex community with spatial and physiological heterogeneity in which the evolution of an exploitative interaction occurs [75,76]. Such evolution provokes marked changes in the symbiotic nature of heterogeneous strains/species and affects community function [75], e.g., antibiotic resistance. A previous study [77] revealed that clinically relevant resistance to the last class of antibiotics can be derived from competitive interactions between bacterial cells in a staphylococcal biofilm. Taken together, these data indicate that the concern of “a return to the pre-antibiotic era” has not been lifted. Alternative antibiotic-free treatments are therefore urgently required.

3. Topical Antibiotic-Free Treatments against MRSA

Although their single use rarely eradicates MRSA infections, topical antibiotic-free treatments can alter microbial burden, inhibit biofilm formation and disrupt formed biofilms. Because biofilm formation confers antibiotic resistance to bacteria [78], these treatments also play crucial roles in combating drug-resistant microbes through the inhibition and disruption of biofilms. Furthermore, these treatments improve wound healing through the removal of necrotic tissue, the alteration of the microbial burden, the reduction of local edema, and tissue repair and regeneration through wound contraction and remodeling, increasing angiogenesis and blood flow [79,80,81,82,83]. Among these effects, debridement is essential for successful wound care. Debridement has been shown [82] to effectively reduce the microbial burden in MRSA wound infections in several modalities, including traditional sharp debridement, hydrosurgery and plasma-mediated bipolar radiofrequency ablation (PBRA). Combinations of these antibiotic-free treatments are believed to be a promising approach to cure drug-resistant S. aureus wound infections. For example, debridement can improve the topical efficacy of species-specific bacteriophages against S. aureus biofilm-infected wounds by disrupting the matrix mass to aid the penetration of these bacteriophages [83].
The most common topical treatment in wound care is the use of a dressing. The wound dressing improves wound healing and tissue restoration; furthermore, modern wound dressings often exhibit excellent antimicrobial activity conferred by either non-antibiotics or antibiotics or a combination of the two [84]. Recently, an electrochemical scaffold (“e-scaffold”) made of conductive carbon fabric was proposed as an alternative antibiotic-free wound dressing to eliminate difficult-to-treat biofilms [85]. Low and constant concentrations of H2O2 are generated by the dressing to destroy biofilms through the electrochemical conversion of oxygen by applying an electric potential to the “e-scaffold”. Although the test strain was multidrug-resistant Acinetobacter baumannii, the dressing is expected to be used as a treatment against MRSA and MRSA-formed biofilms. Another antibiotic-free dressing is used to deliver human β-defensin 3 (hBD-3) by a bioengineered skin tissue; the dressing contains NIKShBD-3 stably expressing hBD-3 after a NIKS human keratinocyte cell line was transfected ex vivo with a construct containing an epidermis-specific promoter driving hBD-3. Use of the dressing has been shown to result in a therapeutically relevant reduction in growth of an MRSA mutant (also a resistant to daptomycin and vancomycin) in an animal model of infected third-degree burn wounds [86].
Negative pressure wound therapy (NPWT) and NPWTi (NPWT with instillation) are complicated dressings to manage large complex injuries to soft tissue and have been shown to effectively reduce the infection rate of both chronic non-healing and acute contaminated open wounds [79,80,81]. Despite the lack of definitive conclusions concerning their use, partially due to the lack of large randomized controlled trials (RCTs), evidence from a small number of retrospective comparative cohorts demonstrates an association between the use of NPWT and a reduction in SSI [87]. Furthermore, clinical practices indicate their relevance in the prevention and control of MRSA infections [79,88,89]. Combined with antibiotics, NPWT successfully treated a pump pocket MRSA infection after a left ventricular assist device implantation [88] and a peri-prosthetic MRSA infection after incisional herniorrhaphy [89]. However, the results of a previous study demonstrate that NPWT reduces the effectiveness of local antibiotic depots, which is an adjunctive therapy for open wounds [79]. Large prospective RCTs of NPWT are therefore needed to support the current evidence that it is effective in treating MRSA with the adjunction of antibiotic therapy.
A previous case report indicated that acupuncture and moxibustion improved staphylococcal wound healing and resulted in full recovery with a partial withdrawal of antibiotics in a patient with a poor response to antibiotics, suggesting their complementary role in staphylococcal wound care [90]. Interestingly, larval therapy has been used as an alternative treatment for hard-to-heal chronic and infected wounds [91]. Some anti-MRSA factors have been identified in larval excretions/secretions [92]. Furthermore, larval therapy can eliminate S. aureus biofilms with the adjunction of antibiotic treatments [93]. These treatments provide clues to combat MRSA infection using natural means, on which ethnomedicine depends, for the long term.

4. Ethnomedical Treatments against MRSA

Antibiotics are gradually losing the war against microbes worldwide [11,12,13,70,71,72], and greater suffering can be observed in the developing world [14]. The desperate situation urges people to revisit the “old-fashioned” but still in use ethnomedicine, which roots from practical, empirical and “almost forgotten” knowledge about naturally derived materials. The resources are primarily based on botanical materials (herbs) but may include animal and mineral materials. Many active compounds have been identified from these materials; furthermore, some of them can be engineered and synthesized. In fact, natural products contribute significantly to modern healthcare. However, most ethnomedical treatments and remedies are yet to be verified, and more evidence is required to support their modern use. For example, acupuncture and moxibustion represent evidence-based complementary therapies for some diseases [94,95,96]. Although they have been reported to successfully treat MRSA infections [90], no evidence-based recommendations have been reported, nor are there ongoing clinical trials in PubMed, Cochrane Library and CenterWatch.
The unsuitability of natural product-centered paradigms in ethnomedical study is perhaps a responsible factor for the slow translation of ethnomedicine to evidence-based medicine. Ethnomedicinal materials may contain complex active compounds that result in a broad but vague spectrum of their ethnomedical indications due to pleiotropic effects. Additionally, the prescribed formulas may have different recipes to improve their therapeutic efficacy through altering the proportions of ingredients (in Chinese Traditional Medicine, the so-called “pattern differentiation and treatment determination, Bianzheng lunzhi”) [97]. “Treat as a whole” is the superior approach in the pharmacological study of those materials.
In accordance with the historical context and philosophy of their ethnomedical treatment, people in the developing world continue to use some naturally derived materials as topical wound healing agents, following the “treat it as a whole” approach. To facilitate audience reading, we have summarized in Table 2 some agents (either single or formula) that exhibit anti-staphylococcal activities. These agents exhibit complex activities on either planktonic microbes or biofilms or both. Their bactericidal effects are mediated by the inhibition of essential survival factors [98] or the microbial enzymatic activities [99,100] of the pathogens, by the antiseptics (e.g., H2O2 and methylglyoxal) that they contain [101], produce [102,103,104] and stabilize [105], and by harnessing physiological factors through active compounds, such as antimicrobial peptides [106] and alkaloids [107]. At the biofilm level, these agents target all formation stages, including the planktonic stage, initial adhesion to surfaces and sessile micro-colony formation. The agents inhibit the expression of key genes involved in biofilm formation [108], act as quorum sensing (QS) inhibitors [109,110], inhibit microbial adhesion by either impairing adherence ability of those biofilm-forming microbes [111] or by covering microbes and/or surfaces [78] and decreasing the produced biomass [112], thereby inhibiting formation and improving the eradication of biofilms.
Ethnomedical drugs may combat drug-resistant strains through their anti-biofilm activities because biofilm formation confers antibiotic resistance to bacteria [78]. For example, the medicinal plant Duabanga grandiflora has been demonstrated to inhibit MRSA biofilm formation through the reduction of cell-surface attachment and the attenuation of the level of penicillin-binding protein 2a (PBP2a) [143]. PBP2a, encoded by mec A, is a protein that confers β-lactam antibiotic resistance to S. aureus, thereby promoting the emergence of MRSA [144]. Furthermore, naturally derived materials may contain some inhibitors of multidrug efflux pumps [145], which are used to detoxify antibiotics by multi- and pan-resistant S. aureus [146]. Together, naturally derived materials can be used to reverse microbial antibiotic resistance and therefore aid in limiting the overuse of antibiotics. However, whether they themselves have a low resistance potential remains unclear. Indeed, “MRSA” is a medical term but not an ethnomedical one. The use of ethnomedicine remains insufficiently supported by evidence: ethnomedical drugs effectively treat MRSA infections with a low resistance potential.

5. A Feasible and Cost-Effective but Challenging Way: To Use Ethnomedical Drugs Topically

The topical use of ethnomedical drugs is not only an ethnomedical practice but a paradigm for ethnomedicine study. The complexity of their effects may decrease to a large extent while in topical use. A reverse proof is deduced from the selection between systemic and topical antibiotics for wound infection. Systemic antibiotics are often the putative preferred choice, perhaps because the antibiotic bioavailability is more stable than that of topical treatments when loci ischemia occurs [147]. Using ethnomedical drugs topically in wound dressings is therefore feasible and, given the ease of obtaining these drugs, cost-effective.
Based on naturally derived materials or natural products, several new wound dressings have been under development through either combination with other antimicrobial agents or when used alone [112]. For example, aloe vera inner gel has been demonstrated to exhibit antimicrobial activities against some Gram-negative (G) bacteria and fungi [112]. Silver [148,149] and honey [101], both well-known, naturally derived antimicrobials, have also been used in the development of new wound dressings. Polymer films containing silver nanoparticles can confer an anti-MSSA (methicillin-sensitive strains of Staphylococcus aureus) effect on biological dressings [150]. Manuka honey synergistically enhanced the effectiveness of several antibiotics against MRSA and Pseudomonas aeruginosa [103] and can also exert positive effects on wound dressings when combined with silver, thereby lowering antimicrobial resistance and limiting the overuse of antibiotics [151]. Moreover, several herbs have been demonstrated to enhance silver nanoparticles (AgNPs) [152,153], and some of these herbs can also act as stabilizers of AgNPs [152]. Indeed, silver is generally accepted as an evidence-based topical wound antimicrobial that is used as a preparation of either a solution or nanoparticles in wound dressings [148,149]; however, silver is suggested to treat G bacteria more effectively, whereas mupirocin is the recommendation for MRSA wound infection [154].
A prerequisite of using ethnomedical drugs to combat MRSA infections more robustly than antibiotics is to make fewer mistakes in their use than were made in antibiotic use. A mistake that is still made is to make poor choices between systemic and topical administrations for wound infection. Systemic antibiotics are ideally never applied in topical use due to the risks of promoting both resistance and allergy [155], whereas topical antibiotics are not recommended for systemic use due to serious adverse effects [156]. Few topical antibiotics have been proven to be effective in clinical trials [156,157], although topical treatment with retapamulin and mupirocin is significantly more effective than systemic treatment with linezolid and vancomycin in eradicating MRSA in a murine superficial skin wound infection model [158]. Moreover, the topical use of antibiotics may result in systemic toxicity, allergy, wound healing delay and normal flora dysfunction [157,159]. Hence, it is crucial in wound care to rationally use systemic and topical antibiotics. Otherwise, overuse and misuse of antibiotics will drastically promote antibiotic resistance. One evidence-based suggestion is that topical antibiotic therapy might be an effective alternative to oral antibiotic therapy in treating diabetic patients with a mildly infected foot ulcer and might reduce the risk of selecting antimicrobial-resistant bacteria [159]. The overuse and misuse of topical antibiotics is perhaps due to an optimism bias that topical use rarely enhances the risk of antibiotic resistance. However, irrespective of whether the antibiotics are systemic or topical, widespread use is associated with the emergence of resistant bacterial strains [147]. In fact, topical antibiotics have long been known to promote the antibiotic resistance of S. aureus [155]. Clearly, the appropriate use of these agents remains a great challenge for antibiotic therapies for MRSA wound infections but a greater one for ethnomedical treatments. Nonetheless, there is still no clear definition of “systemic/topical” and “sensitive/resistant” in ethnomedicine.
To prevent the reoccurrence of a similar dilemma, the development and application of ethomedical drugs should avoid mistakes once made with antibiotics. Ethnomedical drugs should be divided into systemic and topical categories through evaluating their potential systemic adverse effects, allergies and toxicity, as well as local hypersensitivity and bio-absorbance, thereby avoiding misuse. To a large extent, the avoidance of antimicrobial misuse will result in a low resistance potential. However, the emergence of resistant mutants is the essential factor responsible for drug resistance. Thus, drug resistance should be monitored from the beginning of and throughout the development and application of ethnomedical drugs. Additionally, well-designed clinical trials and high-quality animal tests should be conducted because most of the ethnomedical studies on MRSA infection are conducted at the levels of case studies and in vitro assays (Table 2). The lack of evidence dampens the translation of ethnomedicine into evidence-based therapies.

6. Conclusions

Some newly developed antibiotics exhibit high effectiveness in combating MRSA infection, as do candidates under development. With a combination of debridement and modern wound dressings, these agents can successfully treat MRSA wound infections on the basis of limiting their usage. However, antibiotic resistance rapidly spreads, resulting in increasing numbers of multidrug- and even pan-drug-resistant strains. In addition to the development of novel antimicrobials and antibiotic-free treatments, the verification and validation of ethnomedical drugs is a feasible and cost-effective approach to address this issue. The topical use of ethnomedical drugs represents both the development of new wound dressings as well as a platform to study ethnomedical drugs in a “treat it as a whole” approach, which improves the translation of ethnomedicine into evidence-based practice. Naturally derived materials can therefore act as evidence-based drugs for modern medicine. The medical use of honey and silver is a successful paradigm. Despite the limited efficacy of their single use, their combined use in modern wound dressings contributes significantly to combating MRSA infections. Significantly, some naturally derived materials used in ethnomedicine may reverse the antibiotic resistance of MRSA through the attenuation of their PBP2a levels and the inhibition of their multidrug efflux pumps. The medicinal plant Duabanga grandiflora is therefore worthy of significant study. However, most ethnomedical materials are not well-studied, although they exhibit some relevance in combating MRSA infections based on in vitro assays. Furthermore, the tested strains were MSSA in some studies. Well-designed clinical trials and high-quality animal tests are therefore required in the future. Additionally, to avoid creating a similar crisis point in the future, the development and the application of ethnomedical drugs should learn the lessons of previous antibiotic treatment use.

Acknowledgments

This work was jointly supported by a grant from National Natural Science Foundation of China (81160444) and a grant from Natural Science Foundation of Gansu Province, China (1208RJZA182).

Author Contributions

All authors contributed equally.

Conflicts of Interest

The authors declare no conflict of interest.

References

  1. Chahoud, J.; Kanafani, Z.; Kanj, S.S. Surgical site infections following spine surgery: Eliminating the controversies in the diagnosis. Front. Med. 2014, 1. [Google Scholar] [CrossRef]
  2. Munoz, P.; Hortal, J.; Giannella, M.; Barrio, J.M.; Rodriguez-Creixems, M.; Perez, M.J.; Rincon, C.; Bouza, E. Nasal carriage of S. aureus increases the risk of surgical site infection after major heart surgery. J. Hosp. Infect. 2008, 68, 25–31. [Google Scholar] [CrossRef] [PubMed]
  3. Lee, C.Y.; Chen, P.Y.; Huang, F.L.; Lin, C.F. Microbiologic spectrum and susceptibility pattern of clinical isolates from the pediatric intensive care unit in a single medical center—6 years’ experience. J. Microbiol. Immunol. Infect. 2009, 42, 160–165. [Google Scholar] [PubMed]
  4. Ellis, S.L.; Finn, P.; Noone, M.; Leaper, D.J. Eradication of methicillin-resistant Staphylococcus aureus from pressure sores using warming therapy. Surg. Infect. 2003, 4, 53–55. [Google Scholar] [CrossRef] [PubMed]
  5. Negi, V.; Pal, S.; Juyal, D.; Sharma, M.K.; Sharma, N. Bacteriological profile of surgical site infections and their antibiogram: A study from resource constrained rural setting of Uttarakhand State, India. J. Clin. Diagn. Res. JCDR 2015, 9, DC17–DC20. [Google Scholar] [CrossRef] [PubMed]
  6. Hohmann, C.; Eickhoff, C.; Radziwill, R.; Schulz, M. Adherence to guidelines for antibiotic prophylaxis in surgery patients in German hospitals: A multicentre evaluation involving pharmacy interns. Infection 2012, 40, 131–137. [Google Scholar] [CrossRef] [PubMed]
  7. Simor, A.E. Staphylococcal decolonisation: An effective strategy for prevention of infection? Lancet Infect. Dis. 2011, 11, 952–962. [Google Scholar] [CrossRef]
  8. Tong, S.Y.; Holden, M.T.; Nickerson, E.K.; Cooper, B.S.; Koser, C.U.; Cori, A.; Jombart, T.; Cauchemez, S.; Fraser, C.; Wuthiekanun, V.; et al. Genome sequencing defines phylogeny and spread of methicillin-resistant Staphylococcus aureus in a high transmission setting. Genome Res. 2015, 25, 111–118. [Google Scholar] [CrossRef] [PubMed]
  9. Falagas, M.E.; Karageorgopoulos, D.E.; Leptidis, J.; Korbila, I.P. MRSA in Africa: Filling the global map of antimicrobial resistance. PLoS ONE 2013, 8, e68024. [Google Scholar] [CrossRef] [PubMed]
  10. Nickerson, E.K.; West, T.E.; Day, N.P.; Peacock, S.J. Staphylococcus aureus disease and drug resistance in resource-limited countries in South and East Asia. Lancet Infect. Dis. 2009, 9, 130–135. [Google Scholar] [CrossRef]
  11. Fry, D.E. The continued challenge of Staphylococcus aureus in the surgical patient. Am. Surg. 2013, 79, 1–10. [Google Scholar] [PubMed]
  12. Bahemia, I.A.; Muganza, A.; Moore, R.; Sahid, F.; Menezes, C.N. Microbiology and antibiotic resistance in severe burns patients: A 5 year review in an adult burns unit. Burns J. Int. Soc. Burn Inj. 2015, 41, 1536–1542. [Google Scholar] [CrossRef] [PubMed]
  13. Arias, C.A.; Murray, B.E. Antibiotic-resistant bugs in the 21st century—A clinical super-challenge. N. Engl. J. Med. 2009, 360, 439–443. [Google Scholar] [CrossRef] [PubMed]
  14. Reardon, S. Antibiotic resistance sweeping developing world. Nature 2014, 509, 141–142. [Google Scholar] [CrossRef] [PubMed]
  15. McKenna, M. Vaccine development: Man vs. MRSA. Nature 2012, 482, 23–25. [Google Scholar] [CrossRef] [PubMed]
  16. Zhao, Z.; Sun, H.Q.; Wei, S.S.; Li, B.; Feng, Q.; Zhu, J.; Zeng, H.; Zou, Q.M.; Wu, C. Multiple B-cell epitope vaccine induces a Staphylococcus enterotoxin B-specific IgG1 protective response against MRSA infection. Sci. Rep. 2015, 5, 12371. [Google Scholar] [CrossRef] [PubMed]
  17. Bal, A.M.; Gould, I.M. Antibiotic resistance in Staphylococcus aureus and its relevance in therapy. Expert Opin. Pharmacother. 2005, 6, 2257–2269. [Google Scholar] [CrossRef] [PubMed]
  18. Boucher, H.W.; Talbot, G.H.; Bradley, J.S.; Edwards, J.E.; Gilbert, D.; Rice, L.B.; Scheld, M.; Spellberg, B.; Bartlett, J. Bad bugs, no drugs: No ESKAPE! An update from the Infectious Diseases Society of America. Clin. Infect. Dis. 2009, 48, 1–12. [Google Scholar] [CrossRef] [PubMed]
  19. Spellberg, B.; Powers, J.H.; Brass, E.P.; Miller, L.G.; Edwards, J.E., Jr. Trends in antimicrobial drug development: Implications for the future. Clin. Infect. Dis. 2004, 38, 1279–1286. [Google Scholar] [CrossRef] [PubMed]
  20. Shlaes, D.M.; Sahm, D.; Opiela, C.; Spellberg, B. The FDA reboot of antibiotic development. Antimicrob. Agents Chemother. 2013, 57, 4605–4607. [Google Scholar] [CrossRef] [PubMed]
  21. U.S. Food and Drug Administration. FDA Approved Drugs for Infections and Infectious Diseases. Available online: http://www.accessdata.fda.gov/scripts/cder/drugsatfda/index.cfm?fuseaction=Search.DrugDetails (accessed on 20 December 2015).
  22. Corey, G.R.; Kabler, H.; Mehra, P.; Gupta, S.; Overcash, J.S.; Porwal, A.; Giordano, P.; Lucasti, C.; Perez, A.; Good, S.; et al. Single-dose oritavancin in the treatment of acute bacterial skin infections. N. Engl. J. Med. 2014, 370, 2180–2190. [Google Scholar] [CrossRef] [PubMed]
  23. Dunbar, L.M.; Milata, J.; McClure, T.; Wasilewski, M.M.; Team, S.S. Comparison of the efficacy and safety of oritavancin front-loaded dosing regimens to daily dosing: An analysis of the SIMPLIFI trial. Antimicrob. Agents Chemother. 2011, 55, 3476–3484. [Google Scholar] [CrossRef] [PubMed]
  24. Mlynarczyk, A.; Mlynarczyk, B.; Kmera-Muszynska, M.; Majewski, S.; Mlynarczyk, G. Mechanisms of the resistance and tolerance to β-lactam and glycopeptide antibiotics in pathogenic Gram-positive cocci. Mini Rev. Med. Chem. 2009, 9, 1527–1537. [Google Scholar] [CrossRef] [PubMed]
  25. Zhanel, G.G.; Love, R.; Adam, H.; Golden, A.; Zelenitsky, S.; Schweizer, F.; Gorityala, B.; Lagace-Wiens, P.R.; Rubinstein, E.; Walkty, A.; et al. Tedizolid: A novel oxazolidinone with potent activity against multidrug-resistant Gram-positive pathogens. Drugs 2015, 75, 253–270. [Google Scholar] [CrossRef] [PubMed]
  26. Thomson, K.S.; Goering, R.V. Activity of tedizolid (TR-700) against well-characterized methicillin-resistant Staphylococcus aureus strains of diverse epidemiological origins. Antimicrob. Agents Chemother. 2013, 57, 2892–2895. [Google Scholar] [CrossRef] [PubMed]
  27. Prokocimer, P.; de Anda, C.; Fang, E.; Mehra, P.; Das, A. Tedizolid phosphate vs. linezolid for treatment of acute bacterial skin and skin structure infections: The ESTABLISH-1 randomized trial. JAMA 2013, 309, 559–569. [Google Scholar] [CrossRef] [PubMed]
  28. Prokocimer, P.; Bien, P.; Surber, J.; Mehra, P.; DeAnda, C.; Bulitta, J.B.; Corey, G.R. Phase 2, randomized, double-blind, dose-ranging study evaluating the safety, tolerability, population pharmacokinetics, and efficacy of oral torezolid phosphate in patients with complicated skin and skin structure infections. Antimicrob. Agents Chemother. 2011, 55, 583–592. [Google Scholar] [CrossRef] [PubMed]
  29. Moran, G.J.; Fang, E.; Corey, G.R.; Das, A.F.; de Anda, C.; Prokocimer, P. Tedizolid for 6 days versus linezolid for 10 days for acute bacterial skin and skin-structure infections (ESTABLISH-2): A randomised, double-blind, phase 3, non-inferiority trial. Lancet Infect. Dis. 2014, 14, 696–705. [Google Scholar] [CrossRef]
  30. Sahm, D.F.; Deane, J.; Bien, P.A.; Locke, J.B.; Zuill, D.E.; Shaw, K.J.; Bartizal, K.F. Results of the surveillance of Tedizolid activity and resistance program: In vitro susceptibility of Gram-positive pathogens collected in 2011 and 2012 from the United States and Europe. Diagn. Microbiol. Infect. Dis. 2015, 81, 112–118. [Google Scholar] [CrossRef] [PubMed]
  31. Barnea, Y.; Lerner, A.; Aizic, A.; Navon-Venezia, S.; Rachi, E.; Dunne, M.W.; Puttagunta, S.; Carmeli, Y. Efficacy of dalbavancin in the treatment of MRSA rat sternal osteomyelitis with mediastinitis. J. Antimicrob. Chemother. 2015, 71, 460–463. [Google Scholar] [CrossRef] [PubMed]
  32. Esposito, S.; Noviello, S.; Leone, S. Dalbavancin for the treatment of acute bacterial skin and skin structure infections. Expert Opin. Pharmacother. 2015, 23, 313–317. [Google Scholar]
  33. Zhanel, G.G.; Calic, D.; Schweizer, F.; Zelenitsky, S.; Adam, H.; Lagace-Wiens, P.R.; Rubinstein, E.; Gin, A.S.; Hoban, D.J.; Karlowsky, J.A. New lipoglycopeptides: A comparative review of dalbavancin, oritavancin and telavancin. Drugs 2010, 70, 859–886. [Google Scholar] [CrossRef] [PubMed]
  34. Goldstein, E.J.; Citron, D.M.; Merriam, C.V.; Tyrrell, K.L. Ceftaroline versus isolates from animal bite wounds: Comparative in vitro activities against 243 isolates, including 156 Pasteurella species isolates. Antimicrob. Agents Chemother. 2012, 56, 6319–6323. [Google Scholar] [CrossRef] [PubMed]
  35. Friedland, H.D.; O’Neal, T.; Biek, D.; Eckburg, P.B.; Rank, D.R.; Llorens, L.; Smith, A.; Witherell, G.W.; Laudano, J.B.; Thye, D. CANVAS 1 and 2: Analysis of clinical response at day 3 in two phase 3 trials of ceftaroline fosamil versus vancomycin plus aztreonam in treatment of acute bacterial skin and skin structure infections. Antimicrob. Agents Chemother. 2012, 56, 2231–2236. [Google Scholar] [CrossRef] [PubMed]
  36. Biedenbach, D.J.; Alm, R.A.; Lahiri, S.D.; Reiszner, E.; Hoban, D.J.; Sahm, D.F.; Bouchillon, S.K.; Ambler, J.E. In vitro activity of ceftaroline against Staphylococcus aureus isolated in 2012 from Asia-Pacific countries: AWARE surveillance program. Antimicrob. Agents Chemother. 2015, 60, 343–347. [Google Scholar] [CrossRef] [PubMed]
  37. Yanik, K.; Guluzade, E.; Bilgin, K.; Karadag, A.; Eroglu, C.; Birinci, A.; Gunaydin, M. Ceftaroline activity on certain respiratory tract and wound infection agents at the minimum inhibitory concentration level. J. Infect. Dev. Ctries. 2015, 9, 1086–1090. [Google Scholar] [CrossRef] [PubMed]
  38. Lahiri, S.D.; Alm, R.A. Potential of Staphylococcus aureus isolates carrying different PBP2a alleles to develop resistance to ceftaroline. J. Antimicrob. Chemother. 2016, 71, 34–40. [Google Scholar] [CrossRef] [PubMed]
  39. Zhanel, G.G.; Adam, H.J.; Baxter, M.R.; Fuller, J.; Nichol, K.A.; Denisuik, A.J.; Lagace-Wiens, P.R.; Walkty, A.; Karlowsky, J.A.; Schweizer, F.; et al. Antimicrobial susceptibility of 22746 pathogens from Canadian hospitals: Results of the CANWARD 2007-11 study. J. Antimicrob. Chemother. 2013, 68 (Suppl. 1), i7–i22. [Google Scholar] [CrossRef] [PubMed]
  40. Stryjewski, M.E.; Barriere, S.L.; O’Riordan, W.; Dunbar, L.M.; Hopkins, A.; Genter, F.C.; Corey, G.R. Efficacy of telavancin in patients with specific types of complicated skin and skin structure infections. J. Antimicrob. Chemother. 2012, 67, 1496–1502. [Google Scholar] [CrossRef] [PubMed]
  41. Pfaller, M.A.; Mendes, R.E.; Sader, H.S.; Jones, R.N. Telavancin activity against Gram-positive bacteria isolated from respiratory tract specimens of patients with nosocomial pneumonia. J. Antimicrob. Chemother. 2010, 65, 2396–2404. [Google Scholar] [CrossRef] [PubMed]
  42. Brinkman, M.B.; Fan, K.; Shiveley, R.L.; van Anglen, L.J. Successful treatment of polymicrobial calcaneal osteomyelitis with telavancin, rifampin, and meropenem. Ann. Pharmacother. 2012, 46, e15. [Google Scholar] [CrossRef] [PubMed]
  43. Souli, M.; Karaiskos, I.; Galani, L.; Maraki, S.; Perivolioti, E.; Argyropoulou, A.; Charissiadou, A.; Zachariadou, L.; Tsiplakou, S.; Papaioannou, V.; et al. Nationwide surveillance of resistance rates of Staphylococcus aureus clinical isolates from Greek hospitals, 2012–2013. Infect. Dis. 2016, 48, 287–292. [Google Scholar] [CrossRef] [PubMed]
  44. Eckmann, C.; Heizmann, W.; Bodmann, K.F.; von Eiff, C.; Petrik, C.; Loeschmann, P.A. Tigecycline in the treatment of patients with necrotizing skin and soft tissue infections due to multiresistant bacteria. Surg. Infect. 2015, 16, 618–625. [Google Scholar] [CrossRef] [PubMed]
  45. Wong, S.Y.; Manikam, R.; Muniandy, S. Prevalence and antibiotic susceptibility of bacteria from acute and chronic wounds in Malaysian subjects. J. Infect. Dev. Ctries. 2015, 9, 936–944. [Google Scholar] [CrossRef] [PubMed]
  46. Romanowski, E.G.; Kowalski, T.A.; O’Connor, K.E.; Yates, K.A.; Mah, F.S.; Shanks, R.M.; Kowalski, R.P. The in vitro evaluation of tigecycline and the in vivo evaluation of RPX-978 (0.5% Tigecycline) as an ocular antibiotic. J. Ocul. Pharmacol. Ther. 2016, 32, 119–126. [Google Scholar] [CrossRef] [PubMed]
  47. Ramirez, M.S.; Traglia, G.M.; Perez, J.F.; Muller, G.L.; Martinez, M.F.; Golic, A.E.; Mussi, M.A. White and blue light induce reduction in susceptibility to minocycline and tigecycline in Acinetobacter spp. and other bacteria of clinical importance. J. Med. Microbiol. 2015, 64 Pt 5, 525–537. [Google Scholar] [CrossRef] [PubMed]
  48. Heidari, H.; Emaneini, M.; Dabiri, H.; Jabalameli, F. Virulence factors, antimicrobial resistance pattern and molecular analysis of Enterococcal strains isolated from burn patients. Microb. Pathog. 2016, 90, 93–97. [Google Scholar] [CrossRef] [PubMed]
  49. O'Riordan, W.; Mehra, P.; Manos, P.; Kingsley, J.; Lawrence, L.; Cammarata, S. A randomized phase 2 study comparing two doses of delafloxacin with tigecycline in adults with complicated skin and skin-structure infections. Int. J. Infect. Dis. IJID 2015, 30, 67–73. [Google Scholar] [CrossRef] [PubMed]
  50. Pinheiro, L.; Brito, C.I.; Pereira, V.C.; Oliveira, A.; Bartolomeu, A.R.; Camargo, C.H.; Cunha, M.L. Susceptibility profile of Staphylococcus epidermidis and Staphylococcus haemolyticus isolated from blood cultures to vancomycin and novel antimicrobial drugs over a period of 12 years. Microb. Drug Resist. 2015. [Google Scholar] [CrossRef] [PubMed]
  51. Niveditha, N.; Sujatha, S. Worrisome trends in rising minimum inhibitory concentration values of antibiotics against methicillin resistant Staphylococcus aureus—Insights from a tertiary care center, South India. Braz. J. Infect. Dis. 2015, 19, 585–589. [Google Scholar] [CrossRef] [PubMed]
  52. Lupien, A.; Gingras, H.; Leprohon, P.; Ouellette, M. Induced tigecycline resistance in Streptococcus pneumoniae mutants reveals mutations in ribosomal proteins and rRNA. J. Antimicrob. Chemother. 2015, 70, 2973–2980. [Google Scholar] [CrossRef] [PubMed]
  53. Cogo, A.; Gonzalez-Ruiz, A.; Pathan, R.; Hamed, K. Real-world treatment of complicated skin and soft tissue infections with daptomycin: Results from a large European Registry (EU-CORE). Infect. Dis. Ther. 2015, 4, 273–282. [Google Scholar] [CrossRef] [PubMed]
  54. Dhand, A.; Bayer, A.S.; Pogliano, J.; Yang, S.J.; Bolaris, M.; Nizet, V.; Wang, G.; Sakoulas, G. Use of antistaphylococcal β-lactams to increase daptomycin activity in eradicating persistent bacteremia due to methicillin-resistant Staphylococcus aureus: Role of enhanced daptomycin binding. Clin. Infect. Dis. 2011, 53, 158–163. [Google Scholar] [CrossRef] [PubMed]
  55. Stone, P.A.; AbuRahma, A.F.; Campbell, J.R.; Hass, S.M.; Mousa, A.Y.; Nanjundappa, A.; Srivastiva, M.; Modak, A.; Emmett, M. Prospective randomized double-blinded trial comparing 2 anti-MRSA agents with supplemental coverage of cefazolin before lower extremity revascularization. Ann. Surg. 2015, 262, 495–501. [Google Scholar] [CrossRef] [PubMed]
  56. Fernandez-Natal, I.; Saez-Nieto, J.A.; Medina-Pascual, M.J.; Albersmeier, A.; Valdezate, S.; Guerra-Laso, J.M.; Rodriguez, H.; Marrodan, T.; Parras, T.; Tauch, A.; et al. Dermabacter hominis: A usually daptomycin-resistant Gram-positive organism infrequently isolated from human clinical samples. New Microbes New Infect. 2013, 1, 35–40. [Google Scholar] [CrossRef] [PubMed]
  57. Saravolatz, L.D.; Pawlak, J.; Johnson, L.B. In vitro activity of oritavancin against community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA), vancomycin-intermediate S. aureus (VISA), vancomycin-resistant S. aureus (VRSA) and daptomycin-non-susceptible S. aureus (DNSSA). Int. J. Antimicrob. Agents 2010, 36, 69–72. [Google Scholar] [CrossRef] [PubMed]
  58. Wunderink, R.G.; Niederman, M.S.; Kollef, M.H.; Shorr, A.F.; Kunkel, M.J.; Baruch, A.; McGee, W.T.; Reisman, A.; Chastre, J. Linezolid in methicillin-resistant Staphylococcus aureus nosocomial pneumonia: A randomized, controlled study. Clin. Infect. Dis. 2012, 54, 621–629. [Google Scholar] [CrossRef] [PubMed]
  59. Yayan, J.; Ghebremedhin, B.; Rasche, K. No outbreak of Vancomycin and linezolid resistance in Staphylococcal pneumonia over a 10-year period. PLoS ONE 2015, 10, e0138895. [Google Scholar] [CrossRef] [PubMed]
  60. Akhi, M.T.; Ghotaslou, R.; Beheshtirouy, S.; Asgharzadeh, M.; Pirzadeh, T.; Asghari, B.; Alizadeh, N.; Ostadgavahi, A.T.; Somesaraei, V.S.; Memar, M.Y. Antibiotic susceptibility pattern of aerobic and anaerobic bacteria isolated from surgical site infection of hospitalized patients. Jundishapur J. Microbiol. 2015, 8, e20309. [Google Scholar] [CrossRef] [PubMed]
  61. Herrero, I.A.; Issa, N.C.; Patel, R. Nosocomial spread of linezolid-resistant, vancomycin-resistant Enterococcus faecium. N. Engl. J. Med. 2002, 346, 867–869. [Google Scholar] [CrossRef] [PubMed]
  62. Cidral, T.A.; Carvalho, M.C.; Figueiredo, A.M.; de Melo, M.C. Emergence of methicillin-resistant coagulase-negative staphylococci resistant to linezolid with rRNA gene C2190T and G2603T mutations. APMIS 2015, 123, 867–871. [Google Scholar] [CrossRef] [PubMed]
  63. Caballero, J.D.; Pastor, M.D.; Vindel, A.; Maiz, L.; Yague, G.; Salvador, C.; Cobo, M.; Morosini, M.I.; del Campo, R.; Canton, R.; et al. Emergence of cfr-mediated linezolid resistance in a methicillin-resistant Staphylococcus aureus epidemic clone isolated from patients with cystic fibrosis. Antimicrob. Agents Chemother. 2015. [Google Scholar] [CrossRef]
  64. Cafini, F.; Nguyen, L.T.; Higashide, M.; Roman, F.; Prieto, J.; Morikawa, K. Horizontal gene transmission of the cfr gene to MRSA and Enterococcus: Role of Staphylococcus epidermidis as a reservoir and alternative pathway for the spread of linezolid resistance. J. Antimicrob. Chemother. 2015. [Google Scholar] [CrossRef]
  65. Savoia, D. New antimicrobial approaches: Reuse of old drugs. Curr. Drug Targets 2016, 17, 731–738. [Google Scholar] [CrossRef] [PubMed]
  66. Thangamani, S.; Younis, W.; Seleem, M.N. Repurposing ebselen for treatment of multidrug-resistant staphylococcal infections. Sci. Rep. 2015, 5, 11596. [Google Scholar] [CrossRef] [PubMed]
  67. Thangamani, S.; Younis, W.; Seleem, M.N. Repurposing celecoxib as a topical antimicrobial agent. Front. Microbiol. 2015, 6. [Google Scholar] [CrossRef] [PubMed]
  68. Thangamani, S.; Mohammad, H.; Abushahba, M.F.; Hamed, M.I.; Sobreira, T.J.; Hedrick, V.E.; Paul, L.N.; Seleem, M.N. Exploring simvastatin, an antihyperlipidemic drug, as a potential topical antibacterial agent. Sci. Rep. 2015, 5, 16407. [Google Scholar] [CrossRef] [PubMed]
  69. Corriden, R.; Hollands, A.; Olson, J.; Derieux, J.; Lopez, J.; Chang, J.T.; Gonzalez, D.J.; Nizet, V. Tamoxifen augments the innate immune function of neutrophils through modulation of intracellular ceramide. Nat. Commun. 2015, 6, 8369. [Google Scholar] [CrossRef] [PubMed]
  70. Taubes, G. The bacteria fight back. Science 2008, 321, 356–361. [Google Scholar] [CrossRef] [PubMed]
  71. Buru, A.S.; Pichika, M.R.; Neela, V.; Mohandas, K. In vitro antibacterial effects of Cinnamomum extracts on common bacteria found in wound infections with emphasis on methicillin-resistant Staphylococcus aureus. J. Ethnopharmacol. 2014, 153, 587–595. [Google Scholar] [CrossRef] [PubMed]
  72. Liu, Y.Y.; Wang, Y.; Walsh, T.R.; Yi, L.X.; Zhang, R.; Spencer, J.; Doi, Y.; Tian, G.; Dong, B.; Huang, X.; et al. Emergence of plasmid-mediated colistin resistance mechanism MCR-1 in animals and human beings in China: A microbiological and molecular biological study. Lancet Infect. Dis. 2015, 16, 161–168. [Google Scholar] [CrossRef]
  73. Ling, L.L.; Schneider, T.; Peoples, A.J.; Spoering, A.L.; Engels, I.; Conlon, B.P.; Mueller, A.; Schaberle, T.F.; Hughes, D.E.; Epstein, S.; et al. A new antibiotic kills pathogens without detectable resistance. Nature 2015, 517, 455–459. [Google Scholar] [CrossRef] [PubMed]
  74. Stewart, P.S.; Costerton, J.W. Antibiotic resistance of bacteria in biofilms. Lancet 2001, 358, 135–138. [Google Scholar] [CrossRef]
  75. Hansen, S.K.; Rainey, P.B.; Haagensen, J.A.; Molin, S. Evolution of species interactions in a biofilm community. Nature 2007, 445, 533–536. [Google Scholar] [CrossRef] [PubMed]
  76. Wimpenny, J.; Manz, W.; Szewzyk, U. Heterogeneity in biofilms. FEMS Microbiol. Rev. 2000, 24, 661–671. [Google Scholar] [CrossRef] [PubMed]
  77. Koch, G.; Yepes, A.; Forstner, K.U.; Wermser, C.; Stengel, S.T.; Modamio, J.; Ohlsen, K.; Foster, K.R.; Lopez, D. Evolution of resistance to a last-resort antibiotic in Staphylococcus aureus via bacterial competition. Cell 2014, 158, 1060–1071. [Google Scholar] [CrossRef] [PubMed]
  78. Segev-Zarko, L.; Saar-Dover, R.; Brumfeld, V.; Mangoni, M.L.; Shai, Y. Mechanisms of biofilm inhibition and degradation by antimicrobial peptides. Biochem. J. 2015, 468, 259–270. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  79. Stinner, D.J.; Hsu, J.R.; Wenke, J.C. Negative pressure wound therapy reduces the effectiveness of traditional local antibiotic depot in a large complex musculoskeletal wound animal model. J. Orthop. Trauma 2012, 26, 512–518. [Google Scholar] [CrossRef] [PubMed]
  80. Hasan, M.Y.; Teo, R.; Nather, A. Negative-pressure wound therapy for management of diabetic foot wounds: A review of the mechanism of action, clinical applications, and recent developments. Diabet. Foot Ankle 2015, 6, 27618. [Google Scholar] [CrossRef] [PubMed]
  81. Kim, P.J.; Attinger, C.E.; Oliver, N.; Garwood, C.; Evans, K.K.; Steinberg, J.S.; Lavery, L.A. Comparison of outcomes for normal saline and an antiseptic solution for negative-pressure wound therapy with instillation. Plast. Reconstr. Surg. 2015, 136, 657e–664e. [Google Scholar] [CrossRef] [PubMed]
  82. Nusbaum, A.G.; Gil, J.; Rippy, M.K.; Warne, B.; Valdes, J.; Claro, A.; Davis, S.C. Effective method to remove wound bacteria: Comparison of various debridement modalities in an in vivo porcine model. J. Surg. Res. 2012, 176, 701–707. [Google Scholar] [CrossRef] [PubMed]
  83. Seth, A.K.; Geringer, M.R.; Nguyen, K.T.; Agnew, S.P.; Dumanian, Z.; Galiano, R.D.; Leung, K.P.; Mustoe, T.A.; Hong, S.J. Bacteriophage therapy for Staphylococcus aureus biofilm-infected wounds: A new approach to chronic wound care. Plast. Reconstr. Surg. 2013, 131, 225–234. [Google Scholar] [CrossRef] [PubMed]
  84. Jin, S.G.; Yousaf, A.M.; Jang, S.W.; Son, M.W.; Kim, K.S.; Kim, D.W.; Li, D.X.; Kim, J.O.; Yong, C.S.; Choi, H.G. In vivo wound-healing effects of novel benzalkonium chloride-loaded hydrocolloid wound dressing. Drug Dev. Res. 2015, 76, 157–165. [Google Scholar] [CrossRef] [PubMed]
  85. Sultana, S.T.; Atci, E.; Babauta, J.T.; Mohamed Falghoush, A.; Snekvik, K.R.; Call, D.R.; Beyenal, H. Electrochemical scaffold generates localized, low concentration of hydrogen peroxide that inhibits bacterial pathogens and biofilms. Sci. Rep. 2015, 5, 14908. [Google Scholar] [CrossRef] [PubMed]
  86. Gibson, A.L.; Thomas-Virnig, C.L.; Centanni, J.M.; Schlosser, S.J.; Johnston, C.E.; van Winkle, K.F.; Szilagyi, A.; He, L.K.; Shankar, R.; Allen-Hoffmann, B.L. Nonviral human β defensin-3 expression in a bioengineered human skin tissue: A therapeutic alternative for infected wounds. Wound Repair Regen. 2012, 20, 414–424. [Google Scholar] [CrossRef] [PubMed]
  87. Sandy-Hodgetts, K.; Watts, R. Effectiveness of negative pressure wound therapy/closed incision management in the prevention of post-surgical wound complications: A systematic review and meta-analysis. JBI Database Syst. Rev. Implement. Rep. 2015, 13, 253–303. [Google Scholar]
  88. Kimura, M.; Nishimura, T.; Kinoshita, O.; Okada, S.; Inafuku, H.; Kyo, S.; Ono, M. Successful treatment of pump pocket infection after left ventricular assist device implantation by negative pressure wound therapy and omental transposition. Ann. Thorac. Cardiovasc. Surg. 2014, 20, 842–845. [Google Scholar] [CrossRef] [PubMed]
  89. Steenvoorde, P.; de Roo, R.A.; Oskam, J.; Neijenhuis, P. Negative pressure wound therapy to treat peri-prosthetic methicillin-resistant Staphylococcus aureus infection after incisional herniorrhaphy. A case study and literature review. Ostomy Wound Manag. 2006, 52, 52–54. [Google Scholar]
  90. Diogenes, M.S.; Carvalho, A.C.; Tabosa, A.M. Acupuncture and moxibustion as fundamental therapeutic complements for full recovery of staphylococcal skin infection after a poor 50-day treatment response to antibiotics. J. Altern. Complement. Med. 2008, 14, 757–761. [Google Scholar] [CrossRef] [PubMed]
  91. Diaz-Roa, A.; Gaona, M.A.; Segura, N.A.; Ramirez-Hernandez, A.; Cortes-Vecino, J.A.; Patarroyo, M.A.; Bello, F. Evaluating Sarconesiopsis magellanica blowfly-derived larval therapy and comparing it to Lucilia sericata-derived therapy in an animal model. Acta Trop. 2015, 154, 34–41. [Google Scholar] [CrossRef] [PubMed]
  92. Bexfield, A.; Nigam, Y.; Thomas, S.; Ratcliffe, N.A. Detection and partial characterisation of two antibacterial factors from the excretions/secretions of the medicinal maggot Lucilia sericata and their activity against methicillin-resistant Staphylococcus aureus (MRSA). Microbes Infect. Inst. Pasteur 2004, 6, 1297–1304. [Google Scholar] [CrossRef] [PubMed]
  93. Van der Plas, M.J.; Dambrot, C.; Dogterom-Ballering, H.C.; Kruithof, S.; van Dissel, J.T.; Nibbering, P.H. Combinations of maggot excretions/secretions and antibiotics are effective against Staphylococcus aureus biofilms and the bacteria derived therefrom. J. Antimicrob. Chemother. 2010, 65, 917–923. [Google Scholar] [CrossRef] [PubMed]
  94. Deare, J.C.; Zheng, Z.; Xue, C.C.; Liu, J.P.; Shang, J.; Scott, S.W.; Littlejohn, G. Acupuncture for treating fibromyalgia. In Cochrane Database of Systematic Reviews; John Wiley & Sons, Ltd.: Hoboken, NJ, USA, 2013. [Google Scholar]
  95. Manheimer, E.; Cheng, K.; Wieland, L.S.; Min, L.S.; Shen, X.; Berman, B.M.; Lao, L. Acupuncture for treatment of irritable bowel syndrome. In Cochrane Database of Systematic Reviews; John Wiley & Sons, Ltd.: Hoboken, NJ, USA, 2012. [Google Scholar]
  96. Linde, K.; Allais, G.; Brinkhaus, B.; Manheimer, E.; Vickers, A.; White, A.R. Acupuncture for tension-type headache. In Cochrane Database of Systematic Reviews; John Wiley & Sons, Ltd.: Hoboken, NJ, USA, 2009. [Google Scholar]
  97. Temrangsee, P.; Kondo, S.; Itharat, A. Antibacterial activity of extracts from five medicinal plants and their formula against bacteria that cause chronic wound infection. J. Med. Assoc. Thail. 2011, 94 (Suppl. 7), S166–S171. [Google Scholar]
  98. Zhang, S.; Wang, J.; Xu, W.; Liu, Y.; Wang, W.; Wu, K.; Wang, Z.; Zhang, X. Antibacterial effects of Traditional Chinese Medicine monomers against Streptococcus pneumoniae via inhibiting pneumococcal histidine kinase (VicK). Front. Microbiol. 2015, 6. [Google Scholar] [CrossRef] [PubMed]
  99. Emran, T.B.; Rahman, M.A.; Uddin, M.M.; Dash, R.; Hossen, M.F.; Mohiuddin, M.; Alam, M.R. Molecular docking and inhibition studies on the interactions of Bacopa monnieri’s potent phytochemicals against pathogenic Staphylococcus aureus. Daru 2015, 23. [Google Scholar] [CrossRef] [PubMed]
  100. Duan, F.; Li, X.; Cai, S.; Xin, G.; Wang, Y.; Du, D.; He, S.; Huang, B.; Guo, X.; Zhao, H.; et al. Haloemodin as novel antibacterial agent inhibiting DNA gyrase and bacterial topoisomerase I. J. Med. Chem. 2014, 57, 3707–3714. [Google Scholar] [CrossRef] [PubMed]
  101. Molan, P.; Rhodes, T. Honey: A Biologic Wound Dressing. Wounds 2015, 27, 141–151. [Google Scholar] [PubMed]
  102. Asadollahi, F.; Mehrzad, J.; Chaichi, M.J.; Taghavi Razavizadeh, A. Photoimmunological properties of borage in bovine neutrophil in vitro model. J. Photochem. Photobiol. B Biol. 2015, 151, 270–275. [Google Scholar] [CrossRef] [PubMed]
  103. Jenkins, R.; Cooper, R. Improving antibiotic activity against wound pathogens with manuka honey in vitro. PLoS ONE 2012, 7, e45600. [Google Scholar] [CrossRef] [PubMed]
  104. Natarajan, S.; Williamson, D.; Grey, J.; Harding, K.G.; Cooper, R.A. Healing of an MRSA-colonized, hydroxyurea-induced leg ulcer with honey. J. Dermatol. Treat. 2001, 12, 33–36. [Google Scholar] [CrossRef] [PubMed]
  105. Li, W.Z.; Tan, L.L.; Li, Q.J.; Zhou, B.J.; Gao, Y.X.; Ding, W.J. Stabilizing the bactericidal activity of hydrogen peroxide: A brand new function of certain Chinese herbs. Chin. J. Integr. Med. 2014, 20, 468–473. [Google Scholar] [CrossRef] [PubMed]
  106. Lee, J.; Han, S.Y.; Ji, A.R.; Park, J.K.; Hong, I.H.; Ki, M.R.; Lee, E.M.; Kim, A.Y.; Lee, E.J.; Hwang, J.S.; et al. Antimicrobial effects of coprisin on wounds infected with Staphylococcus aureus in rats. Wound Repair Regen. 2013, 21, 876–882. [Google Scholar] [CrossRef] [PubMed]
  107. Pan, X.; Bligh, S.W.; Smith, E. Quinolone alkaloids from Fructus Euodiae show activity against methicillin-resistant Staphylococcus aureus. Phytother. Res. PTR 2014, 28, 305–307. [Google Scholar] [CrossRef] [PubMed]
  108. Chen, X.; Shang, F.; Meng, Y.; Li, L.; Cui, Y.; Zhang, M.; Qi, K.; Xue, T. Ethanol extract of Sanguisorba officinalis L. inhibits biofilm formation of methicillin-resistant Staphylococcus aureus in an ica-dependent manner. J. Dairy Sci. 2015, 98, 8486–8491. [Google Scholar] [CrossRef] [PubMed]
  109. Brango-Vanegas, J.; Costa, G.M.; Ortmann, C.F.; Schenkel, E.P.; Reginatto, F.H.; Ramos, F.A.; Arevalo-Ferro, C.; Castellanos, L. Glycosylflavonoids from Cecropia pachystachya Trecul are quorum sensing inhibitors. Phytomedicine 2014, 21, 670–675. [Google Scholar] [CrossRef] [PubMed]
  110. Zhao, Y.L.; Zhou, Y.H.; Chen, J.Q.; Huang, Q.Y.; Han, Q.; Liu, B.; Cheng, G.D.; Li, Y.H. Quantitative proteomic analysis of sub-MIC erythromycin inhibiting biofilm formation of S. suis in vitro. J. Proteom. 2015, 116, 1–14. [Google Scholar] [CrossRef] [PubMed]
  111. Limsuwan, S.; Homlaead, S.; Watcharakul, S.; Chusri, S.; Moosigapong, K.; Saising, J.; Voravuthikunchai, S.P. Inhibition of microbial adhesion to plastic surface and human buccal epithelial cells by Rhodomyrtus tomentosa leaf extract. Arch. Oral Biol. 2014, 59, 1256–1265. [Google Scholar] [CrossRef] [PubMed]
  112. Cataldi, V.; di Bartolomeo, S.; di Campli, E.; Nostro, A.; Cellini, L.; di Giulio, M. In vitro activity of Aloe vera inner gel against microorganisms grown in planktonic and sessile phases. Int. J. Immunopathol. Pharmacol. 2015, 28, 595–602. [Google Scholar] [CrossRef] [PubMed]
  113. Budri, P.E.; Silva, N.C.; Bonsaglia, E.C.; Fernandes, A.J.; Araujo, J.P.J.; Doyama, J.T.; Goncalves, J.L.; Santos, M.V.; Fitzgerald-Hughes, D.; Rall, V.L. Effect of essential oils of Syzygium aromaticum and Cinnamomum zeylanicum and their major components on biofilm production in Staphylococcus aureus strains isolated from milk of cows with mastitis. J. Dairy Sci. 2015, 98, 5899–5904. [Google Scholar] [CrossRef] [PubMed]
  114. Chew, Y.L.; Chan, E.W.; Tan, P.L.; Lim, Y.Y.; Stanslas, J.; Goh, J.K. Assessment of phytochemical content, polyphenolic composition, antioxidant and antibacterial activities of Leguminosae medicinal plants in Peninsular Malaysia. BMC Complement. Altern. Med. 2011, 11. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  115. Husain, F.M.; Ahmad, I.; Khan, M.S.; Al-Shabib, N.A. Trigonella foenum-graceum (seed) extract interferes with quorum sensing regulated traits and biofilm formation in the strains of Pseudomonas aeruginosa and Aeromonas hydrophila. Evid. Based Complement. Altern. Med. 2015, 2015, 879540. [Google Scholar] [CrossRef] [PubMed]
  116. Teixeira, E.H.; Napimoga, M.H.; Carneiro, V.A.; de Oliveira, T.M.; Cunha, R.M.; Havt, A.; Martins, J.L.; Pinto, V.P.; Goncalves, R.B.; Cavada, B.S. In vitro inhibition of Streptococci binding to enamel acquired pellicle by plant lectins. J. Appl. Microbiol. 2006, 101, 111–116. [Google Scholar] [CrossRef] [PubMed]
  117. Wu, J.G.; Peng, W.; Yi, J.; Wu, Y.B.; Chen, T.Q.; Wong, K.H.; Wu, J.Z. Chemical composition, antimicrobial activity against Staphylococcus aureus and a pro-apoptotic effect in SGC-7901 of the essential oil from Toona sinensis (A. Juss.) Roem. leaves. J. Ethnopharmacol. 2014, 154, 198–205. [Google Scholar] [CrossRef] [PubMed]
  118. Sianglum, W.; Srimanote, P.; Taylor, P.W.; Rosado, H.; Voravuthikunchai, S.P. Transcriptome analysis of responses to rhodomyrtone in methicillin-resistant Staphylococcus aureus. PLoS ONE 2012, 7, e45744. [Google Scholar] [CrossRef] [PubMed]
  119. Srisuwan, S.; Tongtawe, P.; Srimanote, P.; Voravuthikunchai, S.P. Rhodomyrtone modulates innate immune responses of THP-1 monocytes to assist in clearing methicillin-resistant Staphylococcus aureus. PLoS ONE 2014, 9, e110321. [Google Scholar] [CrossRef] [PubMed]
  120. Visutthi, M.; Srimanote, P.; Voravuthikunchai, S.P. Responses in the expression of extracellular proteins in methicillin-resistant Staphylococcus aureus treated with rhodomyrtone. J. Microbiol. 2011, 49, 956–964. [Google Scholar] [CrossRef] [PubMed]
  121. Sianglum, W.; Srimanote, P.; Wonglumsom, W.; Kittiniyom, K.; Voravuthikunchai, S.P. Proteome analyses of cellular proteins in methicillin-resistant Staphylococcus aureus treated with rhodomyrtone, a novel antibiotic candidate. PLoS ONE 2011, 6, e16628. [Google Scholar] [CrossRef] [PubMed]
  122. Limsuwan, S.; Trip, E.N.; Kouwen, T.R.; Piersma, S.; Hiranrat, A.; Mahabusarakam, W.; Voravuthikunchai, S.P.; van Dijl, J.M.; Kayser, O. Rhodomyrtone: A new candidate as natural antibacterial drug from Rhodomyrtus tomentosa. Phytomedicine 2009, 16, 645–651. [Google Scholar] [CrossRef] [PubMed]
  123. Chusri, S.; Settharaksa, S.; Chokpaisarn, J.; Limsuwan, S.; Voravuthikunchai, S.P. Thai herbal formulas used for wound treatment: A study of their antibacterial potency, anti-inflammatory, antioxidant, and cytotoxicity effects. J. Altern. Complement. Med. 2013, 19, 671–676. [Google Scholar] [CrossRef] [PubMed]
  124. Chusri, S.; Sompetch, K.; Mukdee, S.; Jansrisewangwong, S.; Srichai, T.; Maneenoon, K.; Limsuwan, S.; Voravuthikunchai, S.P. Inhibition of Staphylococcus epidermidis biofilm formation by traditional Thai herbal recipes used for wound treatment. Evid. Based Complement. Altern. Med. 2012, 2012, 159797. [Google Scholar] [CrossRef] [PubMed]
  125. Sasitorn Chusri, N.C. Wanvalit Thongza-ard, Surasak Limsuwan and Supayang Piyawan Voravuthikunchai, in vitro antibacterial activity of ethanol extracts of nine herbal formulas and its plant components used for skin infections in Southern Thailand. J. Med. Plant Res. 2015, 6, 5616–5623. [Google Scholar]
  126. Chusri, S.; Tongrod, S.; Chokpaisarn, J.; Limsuwan, S.; Voravuthikunchai, S.P. Antagonistic interactions of “Ya-Sa-Marn-Phlae” ethanol extract in combination with topical antiseptics against clinical isolates of Staphylococcus aureus. BioMed Res. Int. 2014, 2014, 867603. [Google Scholar] [CrossRef] [PubMed]
  127. Quave, C.L.; Plano, L.R.; Pantuso, T.; Bennett, B.C. Effects of extracts from Italian medicinal plants on planktonic growth, biofilm formation and adherence of methicillin-resistant Staphylococcus aureus. J. Ethnopharmacol. 2008, 118, 418–428. [Google Scholar] [CrossRef] [PubMed]
  128. Fahimi, S.; Abdollahi, M.; Mortazavi, S.A.; Hajimehdipoor, H.; Abdolghaffari, A.H.; Rezvanfar, M.A. Wound healing activity of a traditionally used poly herbal product in a burn wound model in rats. Iran. Red Crescent Med. J. 2015, 17, e19960. [Google Scholar] [CrossRef] [PubMed]
  129. Awolola, G.V.; Koorbanally, N.A.; Chenia, H.; Shode, F.O.; Baijnath, H. Antibacterial and anti-biofilm activity of flavonoids and triterpenes isolated from the extracts of Ficus sansibarica Warb. subsp. sansibarica (Moraceae) extracts. Afr. J. Tradit. Complement. Altern. Med. 2014, 11, 124–131. [Google Scholar] [CrossRef] [PubMed]
  130. Teka, A.; Rondevaldova, J.; Asfaw, Z.; Demissew, S.; van Damme, P.; Kokoska, L.; Vanhove, W. In vitro antimicrobial activity of plants used in traditional medicine in Gurage and Silti Zones, south central Ethiopia. BMC Complement. Altern. Med. 2015, 15, 286. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  131. Anani, K.; Adjrah, Y.; Ameyapoh, Y.; Karou, S.D.; Agbonon, A.; de Souza, C.; Gbeassor, M. Effects of hydroethanolic extracts of Balanites aegyptiaca (L.) Delile (Balanitaceae) on some resistant pathogens bacteria isolated from wounds. J. Ethnopharmacol. 2015, 164, 16–21. [Google Scholar] [CrossRef] [PubMed]
  132. Munyendo, W.L.L.; Orwa, J.A.; Rukunga, G.M.; Bii, C.C. Bacteriostatic and bactericidal activities of Aspilia mossambicensis, Ocimum gratissimum and Toddalia asiatica extracts on selected pathogenic bacteria. Res. J. Med. Plant 2011, 5, 717–727. [Google Scholar] [CrossRef]
  133. Okoli, C.O.; Akah, P.A.; Okoli, A.S. Potentials of leaves of Aspilia africana (Compositae) in wound care: An experimental evaluation. BMC Complement. Altern. Med. 2007, 7, 24. [Google Scholar] [CrossRef] [PubMed]
  134. Palombo, E.A.; Semple, S.J. Antibacterial activity of Australian plant extracts against methicillin-resistant Staphylococcus aureus (MRSA) and vancomycin-resistant enterococci (VRE). J. Basic Microbiol. 2002, 42, 444–448. [Google Scholar] [CrossRef]
  135. Sadgrove, N.J.; Jones, G.L. A possible role of partially pyrolysed essential oils in Australian Aboriginal traditional ceremonial and medicinal smoking applications of Eremophila longifolia (R. Br.) F. Muell (Scrophulariaceae). J. Ethnopharmacol. 2013, 147, 638–644. [Google Scholar] [CrossRef] [PubMed]
  136. Hobby, G.H.; Quave, C.L.; Nelson, K.; Compadre, C.M.; Beenken, K.E.; Smeltzer, M.S. Quercus cerris extracts limit Staphylococcus aureus biofilm formation. J. Ethnopharmacol. 2012, 144, 812–815. [Google Scholar] [CrossRef] [PubMed]
  137. Holler, J.G.; Sondergaard, K.; Slotved, H.C.; Guzman, A.; Molgaard, P. Evaluation of the antibacterial activity of Chilean plants traditionally used for wound healing therapy against multidrug-resistant Staphylococcus aureus. Planta Med. 2012, 78, 200–205. [Google Scholar] [CrossRef] [PubMed]
  138. Edmondson, M.; Newall, N.; Carville, K.; Smith, J.; Riley, T.V.; Carson, C.F. Uncontrolled, open-label, pilot study of tea tree (Melaleuca alternifolia) oil solution in the decolonisation of methicillin-resistant Staphylococcus aureus positive wounds and its influence on wound healing. Int. Wound J. 2011, 8, 375–384. [Google Scholar] [CrossRef] [PubMed]
  139. Sherry, E.; Boeck, H.; Warnke, P.H. Percutaneous treatment of chronic MRSA osteomyelitis with a novel plant-derived antiseptic. BMC Surg. 2001, 1, 1. [Google Scholar] [CrossRef] [PubMed] [Green Version]
  140. Shrestha, G.; Raphael, J.; Leavitt, S.D.; St Clair, L.L. In vitro evaluation of the antibacterial activity of extracts from 34 species of North American lichens. Pharm. Biol. 2014, 52, 1262–1266. [Google Scholar] [CrossRef] [PubMed]
  141. Wu, X.; Santos, R.R.; Fink-Gremmels, J. Analyzing the antibacterial effects of food ingredients: Model experiments with allicin and garlic extracts on biofilm formation and viability of Staphylococcus epidermidis. Food Sci. Nutr. 2015, 3, 158–168. [Google Scholar] [CrossRef] [PubMed]
  142. Viswanathan, V.; Phadatare, A.G.; Mukne, A. Antimycobacterial and antibacterial activity of Allium Sativum bulbs. Indian J. Pharm. Sci. 2014, 76, 256–261. [Google Scholar] [PubMed]
  143. Santiago, C.; Lim, K.H.; Loh, H.S.; Ting, K.N. Inhibitory effect of Duabanga grandiflora on MRSA biofilm formation via prevention of cell-surface attachment and PBP2a production. Molecules 2015, 20, 4473–4482. [Google Scholar] [CrossRef] [PubMed]
  144. Kreiswirth, B.; Kornblum, J.; Arbeit, R.D.; Eisner, W.; Maslow, J.N.; McGeer, A.; Low, D.E.; Novick, R.P. Evidence for a clonal origin of methicillin resistance in Staphylococcus aureus. Science 1993, 259, 227–230. [Google Scholar] [CrossRef] [PubMed]
  145. Bharate, J.B.; Singh, S.; Wani, A.; Sharma, S.; Joshi, P.; Khan, I.A.; Kumar, A.; Vishwakarma, R.A.; Bharate, S.B. Discovery of 4-acetyl-3-(4-fluorophenyl)-1-(p-tolyl)-5-methylpyrrole as a dual inhibitor of human P-glycoprotein and Staphylococcus aureus Nor A efflux pump. Org. Biomol. Chem. 2015, 13, 5424–5431. [Google Scholar] [CrossRef] [PubMed]
  146. Costa, S.S.; Viveiros, M.; Amaral, L.; Couto, I. Multidrug Efflux Pumps in Staphylococcus aureus: An Update. Open Microbiol. J. 2013, 7, 59–71. [Google Scholar] [CrossRef] [PubMed]
  147. White, R.J.; Cutting, K.; Kingsley, A. Topical antimicrobials in the control of wound bioburden. Ostomy Wound Manag. 2006, 52, 26–58. [Google Scholar] [CrossRef]
  148. Dai, T.; Huang, Y.Y.; Sharma, S.K.; Hashmi, J.T.; Kurup, D.B.; Hamblin, M.R. Topical antimicrobials for burn wound infections. Recent Pat. Anti-Infect. Drug Discov. 2010, 5, 124–151. [Google Scholar] [CrossRef]
  149. Barajas-Nava, L.A.; Lopez-Alcalde, J.; Roque i Figuls, M.; Sola, I.; Bonfill Cosp, X. Antibiotic prophylaxis for preventing burn wound infection. Cochrane Database Syst. Rev. 2013, 6, CD008738. [Google Scholar] [PubMed]
  150. Guthrie, K.M.; Agarwal, A.; Tackes, D.S.; Johnson, K.W.; Abbott, N.L.; Murphy, C.J.; Czuprynski, C.J.; Kierski, P.R.; Schurr, M.J.; McAnulty, J.F. Antibacterial efficacy of silver-impregnated polyelectrolyte multilayers immobilized on a biological dressing in a murine wound infection model. Ann. Surg. 2012, 256, 371–377. [Google Scholar] [CrossRef] [PubMed]
  151. Wang, Y.L.; Yu, Q.H.; Chen, S.K.; Wang, Y.H. In-vitro activity of honey and topical silver in wound care management. Drug Res. 2015, 65, 592–596. [Google Scholar] [CrossRef] [PubMed]
  152. Sun, W.; Qu, D.; Ma, Y.; Chen, Y.; Liu, C.; Zhou, J. Enhanced stability and antibacterial efficacy of a traditional Chinese medicine-mediated silver nanoparticle delivery system. Int. J. Nanomed. 2014, 9, 5491–5502. [Google Scholar]
  153. Murugan, K.; Senthilkumar, B.; Senbagam, D.; Al-Sohaibani, S. Biosynthesis of silver nanoparticles using Acacia leucophloea extract and their antibacterial activity. Int. J. Nanomed. 2014, 9, 2431–2438. [Google Scholar]
  154. Glasser, J.S.; Guymon, C.H.; Mende, K.; Wolf, S.E.; Hospenthal, D.R.; Murray, C.K. Activity of topical antimicrobial agents against multidrug-resistant bacteria recovered from burn patients. Burns 2010, 36, 1172–1184. [Google Scholar] [CrossRef] [PubMed]
  155. Boot, J.R. Antibiotic resistance and topical treatment. Br. Med. J. 1978, 2, 649–650. [Google Scholar]
  156. Heal, C.F.; van Driel, M.L.; Lepper, P.D.; Banks, J.L. Topical antibiotics for preventing surgical site infection in wounds healing by primary intention. In Cochrane Database of Systematic Reviews; John Wiley & Sons, Ltd.: Hoboken, NJ, USA, 2014. [Google Scholar] [Green Version]
  157. Lipsky, B.A.; Hoey, C. Topical antimicrobial therapy for treating chronic wounds. Clin. Infect. Dis. 2009, 49, 1541–1549. [Google Scholar] [CrossRef] [PubMed]
  158. Lundberg, C.V.; Frimodt-Moller, N. Efficacy of topical and systemic antibiotic treatment of meticillin-resistant Staphylococcus aureus in a murine superficial skin wound infection model. Int. J. Antimicrob. Agents 2013, 42, 272–275. [Google Scholar] [CrossRef] [PubMed]
  159. Lipsky, B.A.; Holroyd, K.J.; Zasloff, M. Topical versus systemic antimicrobial therapy for treating mildly infected diabetic foot ulcers: A randomized, controlled, double-blinded, multicenter trial of pexiganan cream. Clin. Infect. Dis. 2008, 47, 1537–1545. [Google Scholar] [CrossRef] [PubMed]
Table 1. Some U.S. Food and Drug Administration (FDA) approved antibiotics to combat methicillin-resistant Staphylococcus aureus (MRSA) infection [21].
Table 1. Some U.S. Food and Drug Administration (FDA) approved antibiotics to combat methicillin-resistant Staphylococcus aureus (MRSA) infection [21].
AntibioticsIndicationsTherapeutic RelevanceResistant Strains ReportedMonth/Year Approved
For MRSA InfectionsFor Wound Infections
OritavancinFor the treatment of acute bacterial skin and skin structure infections[22,23][22,23]Low potential [24]August 2014
SivextroFor the treatment of acute bacterial skin and skin structure infections[25,26][27,28,29]Low potential [25,30]June 2014
DalvanceFor the treatment of acute bacterial skin and skin structure infections[31,32][31,32]VanA vancomycin-resistant enterococci [33]May 2014
TeflaroFor the treatment of bacterial skin infections and bacterial pneumonia[34,35,36][34,35,37]A S. aureus [38]October 2010
TelavancinFor the treatment of complicated skin and skin structure infections[39,40,41][40,42]VanA vancomycin-resistant enterococci [33]September 2009; June 2013
TigecyclineFor the treatment of complicated skin and skin structure and intra-abdominal infections and bacterial pneumonia[39,43,44,45,46][45,47,48,49]Staphylococcus spp. [50], an MRSA [51], an S. pneumoniae [52]June 2005
DaptomycinFor the treatment of complicated skin and skin structure infections[39,43,45,53,54,55][45,53]Dermabacter hominis [56], Staphylococcus spp. [45], a daptomycin non-susceptible S. aureus [57]September 2003
LinezolidFor the treatment of infections, including pneumonia, infections of the skin and infections caused by a resistant bacterium (Enterococcus faecium)[39,43,45,58,59][45,60]Vancomycin-resistant Enterococcus faecium. [61]; MRSA [62,63,64]April 2000
Table 2. Some ethnomedicinal materials with ethnomedical purposes for wound healing.
Table 2. Some ethnomedicinal materials with ethnomedical purposes for wound healing.
RegionsResources of MaterialsEffectsNoteReferences
Anti-BiofilmAnti-MRSA
BangladeshBacopa monnieri Linn. (Plantaginaceae)Unknown+In vitro assays[99]
MalaysiaCinnamomum spp.+[113]+[71]1. Better effects against MRSA than MSSA [71][71,113]
2. In vitro assays
Leguminosae familyUnknown+[114]1. Anti-non-S.A. biofilm [115,116][114,115,116]
2. In vitro assays
ChinaFructus EuodiaeUnknown+1. Active compounds: quinolone alkaloids[107]
2. In vitro assays
Sanguisorba officinalis L.++1. In vitro assays[108]
2. Inhibiting MRSA biofilm formation in an ica-dependent manner
Toona sinensis (A. Juss.) Roem. (TSL)Unknown+1. Active compounds: sesquiterpenes[117]
2. In vitro assays
ThailandRhodomyrtus tomentosa (Aiton) Hassk.+[111]+[118,119,120,121,122]1. Active compound: rhodomyrtone[111,118,119,120,121,122]
2. Inhibiting microbial adherence ability to sustain surfaces
Herbal formulas+[123]+[124,125]1. In vitro assays[123,124,125,126]
2. Ethnomedical purposes can be clues for their medical applications
3. Antagonistic interactions in combination with topical antiseptics
ItalySome medicinal plants++Ethnomedical purposes can be clues for their medical applications[127]
IranMalva sylvestris L., Solanum nigrum L. and Rosa damascene Mill.+[127]+[127]In vitro assays[127,128]
AfricanFicus sansibarica Warb. Subsp. Sansibarica (Moraceae)++In vitro assays[129]
EthiopiaGuizotia schimperi Sch. Bip. ex Walp.Unknown+In vitro assays[130]
TogoBalanites aegyptiaca (L.) Delile (Balanitaceae)Unknown+In vitro assays[131]
AfricanAspilia africana C. D Adams (Compositae)Unknown+[132]Including animal tests[133]
AustraliaEremophila longifolia (R. Br.) F. MuellUnknown+[134]1. The first known Western scientific justification for the smoking ceremonies involving leaves of Eremophila longifolia[135]
2. In vitro assays
MediterraneanQuercus cerris L., Fagaceae++In vitro assays[136]
ChileanSome medicinal plantsUnknown+In vitro assays[137]
ExtensiveTea treeUnknown+[138]1. Clinical trial [138][138,139]
2. Case study [139]
North AmericanLichensUnknown+In vitro assays[140]
WorldwideGarlic+[141]+In vitro assays[141,142]
MSSA: Methicillin-sensitive Staphylococcus aureus.

Share and Cite

MDPI and ACS Style

Dou, J.-L.; Jiang, Y.-W.; Xie, J.-Q.; Zhang, X.-G. New Is Old, and Old Is New: Recent Advances in Antibiotic-Based, Antibiotic-Free and Ethnomedical Treatments against Methicillin-Resistant Staphylococcus aureus Wound Infections. Int. J. Mol. Sci. 2016, 17, 617. https://doi.org/10.3390/ijms17050617

AMA Style

Dou J-L, Jiang Y-W, Xie J-Q, Zhang X-G. New Is Old, and Old Is New: Recent Advances in Antibiotic-Based, Antibiotic-Free and Ethnomedical Treatments against Methicillin-Resistant Staphylococcus aureus Wound Infections. International Journal of Molecular Sciences. 2016; 17(5):617. https://doi.org/10.3390/ijms17050617

Chicago/Turabian Style

Dou, Jian-Lin, Yi-Wei Jiang, Jun-Qiu Xie, and Xiao-Gang Zhang. 2016. "New Is Old, and Old Is New: Recent Advances in Antibiotic-Based, Antibiotic-Free and Ethnomedical Treatments against Methicillin-Resistant Staphylococcus aureus Wound Infections" International Journal of Molecular Sciences 17, no. 5: 617. https://doi.org/10.3390/ijms17050617

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