Skip to main content
Advances in Wound Care logoLink to Advances in Wound Care
. 2020 Jun 9;9(7):378–395. doi: 10.1089/wound.2019.1113

Are Antimicrobial Peptide Dendrimers an Escape from ESKAPE?

Yayoi Kawano 1, Olivier Jordan 2, Takehisa Hanawa 1, Gerrit Borchard 2, Viorica Patrulea 2,*
PMCID: PMC7307686  PMID: 32320368

Abstract

Significance: The crisis of antimicrobial resistance (AMR) increases dramatically despite all efforts to use available antibiotics or last resort antimicrobial agents. The spread of the AMR, declared as one of the most important health-related issues, warrants the development of new antimicrobial strategies.

Recent Advances: Antimicrobial peptides (AMPs) and AMP dendrimers (AMPDs), as well as polymer dendrimers are relatively new and promising strategies with the potential to overcome drug resistance issues arising in ESKAPE pathogens (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species) colonizing chronic wounds.

Critical Issues: AMPs–AMPDs suffer from limited efficacy, short-lasting bioactivity, and concerns of toxicity. To circumvent these drawbacks, their covalent coupling to biopolymers and/or encapsulation into different drug carrier systems is investigated, with a special focus on topical applications.

Future Directions: Scientists and the pharmaceutical industry should focus on this challenging subject to either improve the activity of existing antimicrobial agents or find new drug candidates. The focus should be put on the discovery of new drugs or the combination of existing drugs for a better synergy, taking into account all kinds of wounds and existing pathogens, and more specifically on the development of next-generation antimicrobial peptides, encompassing the delivery carrier toward improved pharmacokinetics and efficacy.

Keywords: chronic wounds, ESKAPE microbial infection, topical antimicrobials, chitosan derivatives, antimicrobial peptide dendrimers, nanoparticles


graphic file with name wound.2019.1113_figure5.jpg

Viorica Patrulea, PhD

Scope and Significance

Microbial infection has become a major global threat due to the emergence of antimicrobial resistance (AMR). This spurred the need for innovative strategies to fight multidrug-resistant (MDR) bacteria.

This review summarizes the most relevant available antimicrobial agents related to topical therapy. We discuss antiseptics and antibiotics commonly used in wound care and summarize the shortcomings of their application in certain patients. Furthermore, we review innovative strategies relying on linear and dendrimeric antimicrobial peptides (AMPs), including the drug delivery approaches that may improve their efficacy against AMR.

Translational Relevance

Treatment of infections resulting from bacteria belonging to the ESKAPE (Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species) collection is a clinical challenge. This article describes commercially available topical antimicrobials and antiseptics, emphasizing their bacterial activity and limitations. Key AMPs tested in clinical trials are presented. To cope with the urgent need for innovative solutions, novel dendrimer peptides, chemical conjugates, and nanocarriers are discussed, which may allow for an improved activity against MDR bacterial infections.

Clinical Relevance

The care and management of infected wounds is a burden not only for patients, who suffer from severe pain, but for clinicians as well. AMR remains a critical issue and novel AMPs and AMP dendrimers (AMPDs) offer solutions for eradicating MDR bacteria, allowing healing to occur.

Background

Skin has essential roles in maintaining homeostasis, preventing microbial invasion, and providing a barrier between body tissues and the external environment. However, burns, and traumatic or chronic wounds, such as venous, diabetic, and pressure ulcers, compromise the protective barrier that skin offers and subsequently facilitate the risk of bacterial infections.1 Microbial infection due to AMR is one of the major global threats that continues to worsen despite the efforts in finding solutions. Around 0.7 million people die each year due to the emergence and persistence of MDR bacteria or so-called superbugs.2 It is estimated that by the end of 2050, AMR could lead to more than 10 million deaths annually, compared with more than 8.2 million deaths attributable to cancer.2

MDR hinders the healing process in wounds, as most of the wounds would develop infections at some point.1 One of the most challenging MDR bacteria is P. aeruginosa, which is part of the problematic bacterial collection called ESKAPE. Among other virulence factors such as adhesion, quorum sensing, or toxin production, the success of ESKAPE pathogens in escaping from the antibiotic treatment lies in the different posttranslational modifications found in their proteome.3 With the emergence of multidrug-resistant bacteria and reemerging infectious diseases, development of new antibacterial agents has become an urgent task.

The World Health Organization (WHO) called on member states to act against AMR since 1998. WHO calls for improvement of surveillance and testing systems, appropriate use of antibacterial agents, and strengthening countermeasures against infectious diseases. WHO has recently listed nosocomial ESKAPE among the 12 families of MDR bacteria. Moreover, AMR negatively affects the global economy and especially poor countries where the health care system is underdeveloped.4 Recent calculations estimate that by the end of 2050, global MDR costs will go above $100 trillion.2

Clinical and Laboratory Standards Institute (CLSI) guidelines established a list of available antibiotics specifically active against ESKAPE bacteria (Table 1). Surprisingly, many antibiotics and combination of antibiotics have been removed from the list due to their side effects or lack of efficacy. Even for relatively new antibiotics, incidences of resistance are reported and for some bacteria, such as A. baumannii, K. pneumoniae, and P. aeruginosa, resistance has not yet been overcome. These latter were generally kept in the CLSI guideline. Some antimicrobial agents were added since 2010 due to evidence of absence of resistance against specific strains.

Table 1.

Antimicrobial agents against ESKAPE added, removed, or kept from the CLSI guidelines over the period 2010–2019

Pathogen Available Antibiotics EfficacyResistance Reported Ref.
Enterococcus faecium Dalbavancin, Oritavancin, Tedizolid, Telavancin No resistance reported 5
Vancomycin Resistance found
Staphylococcus aureus Dalbavancin, Oritavancin, Tedizolid, Telavancin No resistance reported 6,7
Ceftaroline Resistance found
Amikacin, Amoxicillin–clavulanate, Ampicillin–sulbactam, Cefaclor, Cefamandole, Cefazolin, Cefdinir, Cefepime, Cefmetazole, Cefonicid, Cefoperazone, Cefotaxime, Cefotetan, Cefpodoxime, Cefprozil, Ceftazidime, Ceftizoxime, Ceftriaxone, Cefuroxime, Cephalothin, Doripenem, Ertapenem, Kanamycin, Imipenem, Loracarbef, Meropenem, Moxalactam, Netilmicin, Oxacillin, Piperacillin–tazobactam, Ticarcillin–clavulanate, Tobramycin Removed from CLSI guidelines
Klebsiella pneumoniae Aztreonam, Cefazolin, Cefepime, Ceftazidime, Cefotaxime, Ceftaroline, Ceftazidime–avibactam, Ceftizoxime, Ceftolozane–tazobactam, Ceftriaxone, Doripenem, Ertapenem, Imipenem, Meropenem Resistance found 8
Cephalothin, Nalidixic acid, Ticarcillin Removed from CLSI guidelines
Acinetobacter baumannii Doripenem, Ertapenem, Imipenem, Meropenem Resistance reported 9–11
Mezlocillin, Ticarcillin Removed from CLSI guidelines
Pseudomonas aeruginosa Colistin, Doripenem, Imipenem, Meropenem, Piperacillin, Piperacillin–tazobactam, Ticarcillin–clavulanate Resistance reported 12,13
Cefoperazone, Cefotaxime, Ceftizoxime, Ceftriaxone, Moxalactam, Ticarcillin Removed from CLSI guidelines
Enterobacter spp. Aztreonam No resistance reported 14,15
Cefazolin, Cefepime, Cefotaxime, Ceftaroline, Ceftazidime, Ceftazidime–avibactam, Ceftizoxime, Ceftolozane–tazobactam, Ceftriaxone, Doripenem, Ertapenem, Imipenem, Meropenem Resistance reported
Cephalothin, Nalidixic acid, Ticarcillin Removed from CLSI guidelines

CLSI, Clinical and Laboratory Standards Institute.

Lastly, AMPs have attracted high interest, since they are less likely to induce MDR. AMPs are short peptides consisting of 10–50 residues and produced by all living forms, including protozoa, bacteria, animals, and humans. They are important mediators of innate immune defense. Their amino acid sequence and secondary structure are diverse, but most of the antibacterial peptides are amphipathic with both basic and hydrophobic clusters, and bacterial cell membranes rich in acidic lipids are used for ATP production. Most AMPs possess cationic properties. They have shown a broad activity against a wide range of pathogens, including MDR bacteria by lysing cell membrane through electrostatic interactions.

However, AMPs are rapidly degraded once in contact with human serum (short plasma half-life), losing their activity and most of them are toxic, besides having high production costs.16 Another promising approach is the use of AMPDs, which showed better activity than conventional AMPs, but still show quite high toxicity and poor stability in human serum of only a few hours.17 There is therefore an urgent need to develop new strategies of application of these AMPs and AMPDs avoiding their degradation, while reducing their toxicity at therapeutic concentrations. Chemical conjugation of these potent molecules to different polymers may offer a solution to overcome these drawbacks. Still, the exact antimicrobial mechanism of the polymer–peptide conjugate needs to be further evaluated, as there is a lack of clinical studies describing the healing of infected wounds upon application of AMPs or AMPDs.

Discussion

Infection and biofilm formation

In general, microorganisms colonize all open wounds, although not all wounds will show clinical signs of infection.18,19 Upon infection, microbes create a cytotoxic environment, which often leads to chronic wounds and eventually to gangrene with successive amputation of the infected limb or even to the death of the patient due to sepsis.20 However, the likelihood that a wound will be infected is not only related to the presence of the microorganisms, but to the depth, size, and location of the wound, as well. For example, purulent secretions or local expressions of inflammation are clear indications that an infection has occured.18 Nevertheless, the inflammation may be caused by conditions unrelated to a wound, such as diabetic neuropathy, venous insufficiency, or ischemia.20

As a rule, wounds can be classified as acute or chronic. Acute wounds are a result of an injury, surgery, or the use of intravascular devices. Acute wounds heal within a very short period of time, following successive phases of inflammation, proliferation, migration of keratinocytes and fibroblasts, and final tissue maturation.21 Wounds that fail healing within 3 months through the normal healing process are categorized as chronic.22 This type of wounds show a persistent inflammation phase, which is characterized by a continuous influx of polymorphonuclear neutrophils leading to impaired wound healing.23

Moreover, as wounds grow deeper and become more complex, they can infect the underlying tissue and bone causing osteomyelitis.24 For instance, surgical-site infections, such as superficial incisional, deep incisional, organ, or interorgan space infections, will show postoperative signs of infection typically within the first 10 days, occasionally only after month(s).25

Diabetic wounds, such as foot ulcer or venous ulcer, result from uncontrolled glycemia, leading to microvascular complications (retinopathy, nephropathy, and neuropathy) and very high incidence of infection.20,26 The potential of the wounds to be infected or to heal depends on the surrounding skin and mucous membranes. Actually, the longer the wounds are exposed to the bacteria, the easier it is for the bacteria to proliferate and colonize.

Wounds are ideal hosts for bacterial colonization, providing a warm environment and nutrients. This may lead to local or dangerous systemic infections.27 The propensity for a wound to become infected is directly proportional to the pathogenicity or virulence of the microorganism, and inversely related to both local and systemic resistance of the host.28 Local factors refer to wound size and depth, degree of chronicity, contamination, type of wound, presence of necrotic tissue, anatomic location, and compromised sterilization of the materials; while systemic factors relate to diabetes, obesity, smoking, age, alcoholism, malnutrition, radiation, medication (with steroids, chemotherapy), or inherited neutrophil defects. Usually, infected wounds are accompanied by foul odor, necrotic tissue, wound pain, and impaired healing. It is generally considered that wound colonization occurs at bacterial loads <105 bacteria per gram tissue and infection when >105 bacteria per gram tissue are found.23

Interestingly, critically ill patients have higher rates of MDR microorganisms compared with other patients.29 Most common MDR bacteria are methicillin-resistant S. aureus (MRSA), vancomycin-resistant enterococci, and MDR Gram-negative bacteria (i.e., extended-spectrum β-lactamases; AmpC-type β-lactamases, and metallo-β-lactamase).30,31 Importantly, a rational administration of topical antimicrobial agents should be considered to prevent any resistance development. For this, the use of systemic antibiotics is indicated only when infection is evident or in case of ascending limb infection, sepsis, or incision wounds spreading cellulitis.32 Bite wounds, depending on their severity, should be treated with oral antibiotics.33 Care should be taken to limit the duration of antibiotic administration than to the recommended one, to avoid the development of AMR.34

Bacteria within biofilms are 100–1,000 times more tolerant to antibiotics, disinfectants, or mechanical stresses; thus impeding conventional antibiotic therapy and delaying wound healing in chronic infections.35 Pathogenic bacteria amplify the AMR issue by creating a 3D bacterial biofilm network, which can strongly enhance the chronicity of the wounds. Biofilms are formed of communities with a high bacterial cell density that are enclosed in a self-produced matrix of extracellular polymeric substance. This matrix composed of exopolysaccharides, proteins, and DNA confers additional resistance to bacteria.

The formation of biofilm (Fig. 1) begins when planktonic (free swimming) cells find their way to a surface to which they attach, followed by their rearrangement to form and maturate the biofilm. Biofilm dispersion, also referred as cell detachment, is followed by release of planktonic bacteria that will restart formation of a new biofilm at distant sites.

Figure 1.

Figure 1.

Biofilm formation, including the four stages: (1) Cell attachment, (2) colony formation, (3) mature biofilm, and (4) dispersion of the cells. Color images are available online.

Examples of biofilm-related infections include colonization of almost any surface, including abiotic surfaces (hospital walls, medical devices, implants, catheters, etc.), as well as of biotic surfaces (surgical sites, wounds, lungs, urinary tract, cardiac tissues, bones, etc.).36

Several mechanisms have been proposed to understand the tolerance of bacteria in biofilms to antibiotic treatment37:

  • 1.

    Limited antibiotic penetration into the biofilm

  • 2.

    Gene mutation

  • 3.

    Reduced metabolic rate, growth rate, and division rate

  • 4.

    Presence of slowly growing persister cells that could reactivate after the antibiotic treatment

  • 5.

    Overexpression of bacterial efflux pump in biofilm, which leads to increased resistance to antibiotics

  • 6.

    Protection by the self-secreted matrix of extracellular polymer substance.

There are few antimicrobial agents in clinical use to specifically target biofilms, probably due to the poor understanding of biofilm formation. Resistance ability can be explained if combining the before-mentioned mechanisms. For instance, P. aeruginosa, which frequently leads to biofilm-associated infections, adapts easily to the hostile habitat by producing adapted phenotypes and mutations. On the other hand, the geometry of P. aeruginosa's colonies in a shape of tall ridges or wrinkles (referred as colony rugosity), facilitates their oxygen supply and allow them to grow taller.35

Current biofilm-related infection diagnosis are based on (i) clinical wound characteristics: edema, erythema, warmth, and purulence; (ii) laboratory-based analyses: microbiological tests of wound swabs; and (iii) technical methods: scanning electron microscopy, gas chromatography–mass spectrometry, epifluorescence microscopy, colorimetric methods, and metabolic and biomass assays.38 In the context of AMR, adequate diagnosis and design of efficient treatments against biofilm-associated infection is critically needed.

Topical antimicrobial and aseptic agents

Despite recent advances in wound management, very few topical therapies (Fig. 2) proved their efficacy in promoting wound healing. They led to a better understanding of factors influencing the process of wound healing and protection against bacterial infection. However, these methods have met with challenges, as well.39,40

Figure 2.

Figure 2.

Current treatment methods for skin regeneration. Color images are available online.

Many wound dressings have been developed in an attempt to combine anti-infective properties and promotion of wound healing. Hydrophilic hydrogels and foams absorb wound exudates and keep the wound hydrated. However, they can act as a perfect breeding ground for bacterial growth. Bruises and scrapes result from frequent bandage change, which may lead to new skin injuries.41 Cotton gauze dressing has been considered for years as the standard of wound care, along with many other commercially available products, such as alginates, collagen dressings, regenerated cellulose, and honey, among others. However, the gold standard for treatment of chronic wounds is yet to be identified.42 Integration of antibiotics into conventional wound dressings has been of high interest.

Topical antimicrobial agents, including disinfectants, antiseptics, and antibiotics, have the ability to kill microorganisms, inhibit their growth, or reduce their number. Disinfectants are very potent against most microorganisms; however, their high toxicity toward all healthy tissues limits their application to inanimate objects and materials, such as surgical instruments and surfaces. Antiseptics have a broad antimicrobial activity and may be used even for some open wounds, but toxicity was reported as well.43

In contrast, antibiotics have high bactericidal activity through a specific cell-targeting mechanism—still limited by the AMR, which is very common nowadays. Moreover, administration of some antibiotics is accompanied by several side effects, such as pain, rash, cytotoxicity, toxic effects to kidneys, liver, and other organs.44 Several studies reported toxic encephalopathy-induced nonconvulsive status epilepticus,45 seizures,46 chronic myeloid leukemia, and renal failure,47 in the case of cephalosporin use.

Chronic wounds are treated using different topical antibacterial or antiseptic formulations with or without antibiotics as listed in Table 2, depending on the severity of the wound. Besides topical agents, treatment with dermal substitutes has shown effective to heal specific wounds. However, these substitutes generally lack the antimicrobial properties required for a durable outcome.

Table 2.

Commercially available topical antimicrobials and antiseptics, their bacterial activity and limitations

TAA Formulation(s) Activity Against Microorganism(s) Limitations Ref.
Acetic acid Solution 0.5% Gram (+), (−) bacteria Limited activity against biofilms; in vitro toxicity 48
Amoxicillin Gel, solution Gram (+), (−) bacteria Resistance found 49
Bacitracin Ointment Gram (+), less active in Gram (−); resistance found in Enterobacter spp. and Pseudomonas spp. Allergic reactions; may lead to overgrowth of drug-resistant organisms; cross-sensitization with neomycin; anaphylaxis 50
Cadexomer iodine Gels, ointment, dressings Gram (+), (−) bacteria; viruses; high wound exudates absorption Cytotoxic; postapplication pain; renal failure; thyroid dysfunction; MRSA 51,52
Cephalosporins Cream, gel, ointment Gram (+), (−) bacteria Renal dysfunction; gastrointestinal disorders; hematologic reactions; neurotoxicity; seizures; encephalopathy 45–47
Cerium nitrate Cream, dressings Gram (+), (−) bacteria Methemoglobinemia; hematology alterations; weight loss 53
Chlorhexidine Solution, sponge, brush, foam Gram (+), (−) bacteria; less effective against P. aeruginosa Cytotoxicity; allergic reactions, including anaphylaxis; resistance reported; injury to eyes and middle ear 28
Clotrimazole Cream, ointment Yeast; fungi Recurring infection 54
Fusidic acid Cream Gram (+) Rapid resistance; multiple applications per day 55
Gauze Vaseline gauze, silicone gauze, sterile gauze Nonbacterial Wound drying; creates new injuries on changing; significant pain 56
Gentamicin Cream and ointment Gram (+), (−) bacteria May lead to resistance; multiple application per day; ototoxicity 57,58
Honey Dressings Inhibits >50 bacterial species, including some MRSA Nonmedical honey to be avoided (as it may contain spores) 28,59,60
H2O2 Solution, cream Gram (+), (−) bacteria; fungi; viruses Cytotoxicity; oxygen gas embolism 61
Mafenide acetate (Sulfamylon) Cream, solution, ointment Gram (+), (−) bacteria Prone to cause pain upon application; allergic reactions 62
Mupirocin Ointment MRSA infection Potential for developing resistance 48
Neomycin Ointment, cream, powder Gram (−) and some Gram (+) bacteria Allergic contact dermatitis; may cause systemic toxicity; ototoxicity; nephrotoxicity 63,64
Neosporin Ointment Gram (+), (−) bacteria Allergen 65,66
Nystatin Cream Fungi Resistance reported 65,67
PHMB Gel, solution, and dressing Gram (+), (−) bacteria and fungi Cytotoxicity; anaphylaxis 68
Polymyxin B (Colistin) Ointment Gram (−) Last-resort; hypersensitivity reactions; neurotoxicity; renal acute tubular necrosis 48,69
Polysporin Ointment Gram (+), (−) bacteria and fungi Potential for allergy if neomycin crosssensitization 65,66
Povidone/iodine Solution, ointment, surgical scrub, cream, hydrogel Gram (+), (−) bacteria; viruses; fungi; and yeast Contact dermatitis; metabolic acidosis; delayed wound healing 70–72
Silver dressings Foams, nanoparticle gel Gram (+), (−) bacteria and fungi, including MRSA and VRE Possible silver staining of tissues; delayed epithelialization (debated) 28,42
AgNO3 Cream, solution, sticks Gram (+), (−) bacteria and fungi Frequent reapplication due to short acting; methemoglobinemia; allergies; bacterial resistance 48,73
Silver sulfadiazine Cream Gram (+), (−) bacteria Mild skin sensitiveness 74,75
Sodium hypochlorite Dakin's solution More active on Gram (+) than Gram (−) bacteria, fungi, and viruses Cytotoxicity; postgraft bleeding; dissolve clots 76,77
Retapamulin Ointment Some Gram (+) and very few Gram (−) bacteria May cause local reactions; several applications 78
Xeroform petrolatum Dressing Gram (+), (−) bacteria; yeast Disputed antibactericidal activity 79,80
Zinc oxide-Scarlet Red Fine mesh gauze, cream, ointment Some Gram (+), (−) bacteria; fungi Potential irritation 48

+, positive; −, negative; AgNO3, silver nitrate; H2O2, hydrogen peroxide; MRSA, methicillin-resistant S. aureus; PHMB, polyhexa-methylene biguanide; TAA, topical antimicrobials and antiseptics; VRE, vancomycin-resistant enterococci.

Alternative agents to antibiotics

Nowadays, we face an urgent need to identify new antibacterial drugs to overcome AMR of different microorganisms.

Antimicrobial peptides

The AMPs, also referred as host defense peptides, were identified as good candidates to limit resistance-induced microorganisms. They are abundant in prokaryotes (produced by Gram-positive and Gram-negative bacteria) and in all eukaryotic organisms (fungi, algae, plants, insects, and mammals) and well distributed in cells and tissues as the front fighting line against pathogens.81 The first AMP, a tyrothricin compound was extracted from Bacillus strain by Dubos and Gause in 1939 independently from each other in their respective laboratories. It proved to be effective against pneumococci infection in mice.82 Soon after, it was found that the first AMP contained two different molecules: 80% tyrocidine and 20% gramicidin. Tyrocidine was very effective against both Gram-positive and Gram-negative bacteria, despite being highly toxic against mammalian cells.83 Gramicidin was applied for the treatment of infected wounds and especially ulcers during the Second World War.

Inspired by natural AMPs, many synthetic or semisynthetic analogs were recently developed. The main focus is on synthetic AMPs with higher antimicrobial activity and lower risks of toxicity toward host cells than their natural analogs. An online antibacterial peptide database (APD3) lists more than 3,130 AMPs originating from all species,84,85 out of which 134 are identified as human host defense peptides with more than 100 of those exhibiting antibacterial activity.86 Their classification depends on the charge, length, sequence of amino acids, and their secondary structure as shown in Table 3. They may have either amphiphilic or cationic domains, for example, human AMPs have a net charge range from anionic (rare) to cationic (most often), which ranges from −3 to +20.86

Table 3.

The four antimicrobial peptide families with their type of conformation and examples

Family Type Type of Secondary Structure Examples Ref.
α-Helix α-Helical conformation Cryptdin-4, human intestinal α-defensin HD5 and HD6, LL-37, Magainin 1 and 2, Moricin 87,88
β-Sheet At least two β-sheets and two to four disulfide bridges hBD-1, hBD-2, hBD-3, hBD-4, Pg1, Tachyplesin I 88
Loop Single bond (either disulfide, amide, or isopeptide) Thanatin 88
Extended family Neither α-helical nor β-sheets Indolicidin, Indolicidin analog (CP10A), Tritrpticin 89

hBD, human β-defensin; Pg1, protegrin-1.

Considering the critical situation of ESKAPE pathogens, AMPs are used to date as an effective therapy. They have the advantage of fast acting, bactericidal, multifunctional (stimulate the immune system and inhibit bacterial growth), and anti-inflammatory and/or wound healing promotor. AMPs, such as human β-defensins (hBD-1, hBD-2, and hBD-3)90 and cathelicidin LL-37,91 originating from epithelial tissues are factors of the innate immune system. They protect skin from infections caused by several microorganisms, such as K. pneumoniae, MRSA, P. aeruginosa, Escherichia coli, and Neisseria gonorrhoeae.92

Moreover, LL-37 is reported as a safe agent for clinical use as it successfully showed promotion of wound healing in hard-to-heal venous leg ulcers during short-term treatment.93 Histatin 5, human salivary peptide, has a strong antibacterial activity (≥70%) against five out of six ESKAPE pathogens, except K. pneumoniae. It also showed a strong in vitro antibiofilm formation in P. aeruginosa (60% killing) but less in A. baumannii and S. aureus.94

Cellular distress is usually noticed when exposing bacteria to antibiotics, although no bacterial adaptation nor resistance development was shown when treating E. coli for several hours with AMPs, such as cecropin A, melittin, magainin II, pexiganan, and LL-37 at 50% minimal inhibitory concentration (MIC). Also, treatment with these AMPs did not show any changes in mutation rate nor differential expression of genes related to stress-induced mutagenesis, while ampicillin, ciprofloxacin, and kanamycin antibiotics increased the mutation rate by threefold to fourfold.95

Bacterial resistance to AMPs and virulence was noticed in case of AMP proteolytic degradation. For instance, S. aureus together with an aureolysin metalloprotease could degrade LL-37 AMP by cleaving C-terminus bonds of the peptide and in turn contributing to resistance.95 ZapA metalloprotease could inhibit bacterial activity of LL-37 and hBD-1 against Proteus mirabilis, responsible for urinary tract infections, by at least 7- and 30-fold, respectively. Surprisingly, the same protease did not inhibit the activity of hBD-2, which has differences in amino acid sequence than hBD-1.96 P. aeruginosa, Enterococcus faecalis, and Streptococcus pyogenes use a common mechanism to escape α-defensins by secreting extracellular proteoglycans, which releases dermatan sulfate. Released compound further binds to α-defensins by completely inhibiting its activity.95

The process of biofilm formation is another mechanism of AMR to escape AMPs. It was reported that the DNA found in P. aeruginosa biofilm induces resistances to both polymyxin B and colistin by inducing lipopolysaccharide (LPS) modification.95 On the contrary, no resistance was found when treating protease-resistant P. aeruginosa biofilms with LL-37.97 In spite of these few reported resistance to pathogens, AMPs remain a promising tool to fight resistance, benefiting from their broad activity spectrum and the variety of their mechanisms of action.

Antimicrobial peptides mechanism of action

Bacteria fall into two main categories depending on their cell wall structure: Gram-negative and Gram-positive bacteria. Gram-positive cells have an outer bacterial cell wall; whereas Gram-negative possess an additional outer membrane adapted with several porins and LPSs. These differences in the cell membrane will confer different susceptibility to various antimicrobial agents.98 For instance, some of the AMPs were reported to be more effective against Gram-negative than Gram-positive species.

Even though the mechanisms of action are currently debated among scientists, most agree on the key role of electrostatic forces between positively charged AMPs and negatively charged bacterial membrane, which in turns leads to bacterial leakage and death.99 Other proposed mechanisms are: (i) disruptive, such as “barrel-stave” and “toroidal pore” models of pore formation in the bacterial membrane; “carpet–detergent,” by which peptides can form micelles with the membrane components, (ii) nondisruptive, for example, bacterial membrane thinning, depolarization, or aggregation, and (iii) mediated by the “stringent response,” which is the stress response by the bacteria, involving secondary messenger metabolites.98 A detailed description of the modes of action is given by Bahar and Ren100 In addition, the AMP mechanisms of action depend on their concentration, pH, or temperature.99,101

Antimicrobial peptides toxicity and efficacy: preclinical and clinical data

Despite their high and broad antimicrobial activity, AMPs may suffer from their toxicity toward mammalian cells. Toxicity against red blood cells (RBCs), or the ability of AMPs to lyse RBCs, also referred as hemolysis, is another major concern. Therefore, the selectivity toward bacterial cells is generally defined by the ratio of HC50/MIC, where HC50 is the concentration necessary to lyse 50% of RBCs and MIC is the minimal concentration to inhibit the growth of a given microorganism, for example, to obtain a bacteriostatic effect.102 Another important parameter is the minimal bactericidal concentration (MBC) of AMPs, which indicates the ability to eliminate (kill) bacteria. Antibacterial agents are usually regarded as bactericidal if the MBC is no more than four times the MIC.103

For the clinical use of AMPs, one should consider their mechanisms of action, stability under physiological conditions, and the balance between their efficacy and toxicity. Currently, only some AMPs with the ability to combat MDR bacteria have been approved by the FDA and already routinely used, such as Gramicidin (date of approval: 2005),104 Micafungin (2005),105 Anidulafungin (2006),106 Telavancin (2009),106 Ceftaroline (2010),5 Dalbavancin (2014),106 Oritavancin (2014),106 Caspofungin (2017),106 Ozenoxacin (2017),5 Tedizolid Phosphate (2015),5 and Omadacycline (2018).5 Most of them are against bacterial infection and administered either I.V. or topically.107,108 Some AMPs, not yet FDA approved, are being tested in clinical trials and listed in Table 4.

Table 4.

Novel antimicrobial peptides for topical application tested in preclinical and clinical trials to eradicate multidrug-resistant bacteria

Peptide Producer Description Application Adminis-tration Phase Comments Ref.
Brilacidin® Innovation Pharmaceuticals, Inc. Defensin mimetic Acute skin and soft tissue infections in oral mucositis I.V. >II Reduces oral mucositis in HNC patients 109,110
Dusquetide (SGX942) Soligenix First-in-class innate defense regulators Oral mucositis I.V. III Significantly reduces oral mucositis in HNC patients 111
hLF1–11 AM-Pharma Lactoferricin-based peptide Bacterial and fungal infections and for prophylaxis in hematopoietic stem cell transplantation I.V. I/II Low antimicrobial efficacy and stem cell transplantation-associated infections in immunocompromised patients was reported; Company suspended trials 112,113
Human LL-37 (OP145) ProMore Pharma Human cathelicidin Leg ulcer Topical gel >II Significantly better than placebo 93,114
Lytixar (LTX-109) Lytix Biopharma AM Synthetic peptidomimetic Skin infection; nasal colonization with S. aureus and impetigo Topical hydrogel I/II Studies in progress 115
Novarifyn® (NP432) NovaBiotics Synthetic peptide MRSA, P. aeruginosa, Clostridium difficile, A. baumannii, Escherichia coli Topical PC Ongoing studies 116
Novexatin NovaBiotics Cyclic cationic peptide Fungal nail infection Topical brush II Ongoing studies. No side effects reported yet 114
PAC-113 Pacgen; Demegen Synthetic histatin 3 Oral mouth rinse for Candidiasis in HIV patients Topical solution II High efficacy for oral candidiasis 117
Pexiganan (Locilex®, MSI-78) Magainin Pharmaceuticals Magainin 2 analog Diabetic foot ulcer Topical cream III One of the debated peptide for its poor antibacterial efficacy compared with other antibiotics 16,26,118,119
Polymyxin Athenex Cyclic cationic lipopeptides Urinary tract infection, mucositis, ocular and wound infection treatment I.M., I.T., I.V., ophthalmic II/III Used as a “last resort” due to its high toxicity (neurotoxicity and nephrotoxicity); excluded from CLSI list 120,121

hLF, human lactoferrin fragment; HNC, head and neck cancer; I.M., intramuscular; I.T., intrathecal; I.V., intravenous injection; PC, preclinical.

Most of the AMPs cannot reach the clinical phase due to their systemic toxicity, fast degradation, short half-life, and/or reduced activity in the presence of salts or divalent cations.122 Several AMPs in clinical trials failed to show a better activity than conventional antibiotics or exhibited adverse side effects, although in general one should consider the trade-off between toxicity and efficacy. Some examples are Iseganan (withdrawn after phase III) intended for the treatment of oral mucositis117,123 and Omiganan (withdrawn after phase III), a topical gel for prevention of catheter infections, acne and rosacea.122,124 These failures have spurred the development of encapsulation strategies of AMPs into different delivery systems, such as nanoparticles and liposomes, to enhance their stability and half-life.

Besides clinically approved antimicrobial and antiseptic agents, synthetic dendritic polymers and peptide dendrimers have recently shown promising developments, detailed in the dedicated paragraph below. In addition, other approaches such as encapsulation into nanocarriers or chemical coupling to other molecules are used to reduce AMPs toxicity and increase their efficacy.

Polymer dendrimers for topical application

Poly(amidoamine)

Synthetic poly(amidoamine) (PAMAM) dendrimers are available up to the 10th generation and mostly studied for their possible antimicrobial efficacy. The most common polymeric dendrimers are PAMAM, polypropyleneimine, poly-l-lysine, carbosilane, polyglycerol, poly(bencyl ether), and phosphorus dendrimers.125 Polymer dendrimers have a three-dimensional structure with different density of functional groups and have been found as effective antibacterials. Starting from the inner core of the dendrimer toward the external side, each step of ramification is identified as a generation (Gx), as illustrated in Fig. 3.

Figure 3.

Figure 3.

Schematic representation of branched dendrimers and their delivery systems. Color images are available online.

The modification of one unit will affect the properties of the whole dendrimer construct. Calabretta et al. have reported for the first time the effectiveness of fifth-generation (G5) amino-terminated PAMAM dendrimers against both P. aeruginosa and S. aureus at very low concentrations (MIC of 1.5 and 20.8 μg/mL, respectively).126 However, G5 PAMAM exerted higher toxicity (25% survival at 10 μg/mL) toward human corneal epithelial cells compared with LL-37 (significant toxicity at 25 μg/mL), potentially due to the highly branched cationic dendrimers.

Interestingly, a smaller PAMAM generation (G3) was found to have an enhanced activity against P. aeruginosa and S. aureus when compared with G5 PAMAM (G3: 6.3 μg/mL vs. G5: 12.5 μg/mL), or LL-37 (1.3–12.5 μg/mL).127 This suggests that the number of amino groups displayed by the dendrimers—higher for G5 than for G3—is not the sole factor for the antimicrobial activity observed.

PEGylation of the functional groups in PAMAM dendrimers was reported to reduce both the toxicity and bactericidal activity, while complete polyethylene glycol replacement of functional groups inhibited the activity of the dendrimer against P. aeruginosa, most probably due to decreased number of amino groups.126,127 Another approach to reduce the toxicity of PAMAM dendrimers is to modify the functional groups into amino-, hydroxyl-, and carboxyl-terminated G4-PAMAM.128

The antibacterial activity against E. coli in vitro was found to decrease from G4-PAMAM-NH2 to G4-PAMAM-OH to G4-PAMAM-COOH (IC50 of 3.8 μg/mL, 5.4 mg/mL, and 22 mg/mL, respectively). Topical vaginal and cervical application of G4-PAMAM-OH in a pregnant guinea pig model of chorioamnionitis (intrauterine infection by E. coli) lead to major changes to the outer membrane of E. coli, while G4-PAMAM-NH2 induced changes to both inner and outer bacterial membranes.128 However, G4-PAMAM-NH2 was dropped for further potential application due to its very high toxicity, while G4-PAMAM-OH was barely transported across placental membrane model, suggesting safety for pregnant women.129

Lower generation, G1 PAMAM-disaccharide galabiose modified exhibited a 3,000-fold increased potency against Streptococcus suis with an MIC of 0.3 nM. G1 dendrimer was able to inhibit the adhesion of S. suis.130 Actually, increasing the number of generations in amino-PAMAM from G3 to G7 significantly decreased in vitro viability and inhibited differentiation of human neural progenitor cells and damaged DNA at a concentration of 5 μg/mL. In contrast, G0, G1, and G2 at the same concentration did not show any cytotoxicity.131 Therefore, lower generation PAMAM hold promise to improve the efficacy–toxicity ratio of the dendrimers, paving the way to clinical applications.

AMPDs for topical applications

Compared with linear AMPs, AMPDs show a three-dimensional, regularly branched structure built by covalent bonds, a very low polydispersity and a higher density of surface groups.17 Their structure is very similar to the polymeric dendrimers, except that they have only one-side branches, which makes them more flexible for chemical coupling or incorporation into different delivery systems (Fig. 4). The synthetic flexibility and high density of the functional groups found in AMPDs make them very attractive for use as delivery systems for drugs and bioactive principles. Designing peptide-based agents is strongly supported by the high potency of the AMPDs not only to kill bacteria, but also to reduce the toxicity against mammalian cells.132

Figure 4.

Figure 4.

Amino acid sequence in AMPD (e.g., G3KL), which is based on a divalent lysine core whose α and ɛ amines along with leucine double geometrically with each ramification building up a new generation. AMPD, antimicrobial peptide dendrimer. Color images are available online.

The displayed functional groups will govern the mechanism of bacterial killing. AMPDs, which bear charged ends, are believed to act by penetrating the cell membrane inducing leakage of intracellular materials resulting in bacterial death. Therefore, the AMPD mechanism of action against bacteria is related to the number of functional surface groups and their ability to cross the cell membrane.132

AMPDs show increased activity, which is usually attributed to the higher local concentration of bioactive units in such assemblies, and to their greater stability against peptidases and proteases. For instance, dendrimeric peptides were shown to be selective for microbial surfaces with a broad antimicrobial and low hemolytic activity. A family of AMPDs based on R4 tetrapeptide (RLYR) and R8 octapeptide (RLYR-KVYG), were tested against 10 different microbial strains. Both R4 and R8-based dendrimers of fifth and eighth generation exhibited high activity with MICs <1 μM against Gram-positive and Gram-negative bacteria as well as fungi.133

Besides, these AMPDs were resistant to proteolytic degradation or to protease inhibition, which has been attributed to their dendrimeric structure. A lipodimeric peptide, SB056, was investigated for its antimicrobial activity against a range of bacteria, including Gram-positive and Gram-negative strains. The in vitro assays showed high antimicrobial activity with MIC in the range of 2–32 μg/mL against A. baumannii, Enterobacter cloacae, E. coli, K. pneumoniae, and P. aeruginosa, which is comparable with the activity of polymyxin B.

The SB056 AMPD showed strong activity against E. coli and S. aureus strains as well as strong Staphylococcus epidermidis biofilm inhibition.134 Further improvements on the amphipathic part of the SB056 resulted in more ordered β-strands with a stronger antimicrobial activity against both Gram-positive and Gram-negative bacteria.135 A study on series of tryptophan-ending dendrimers showed that amphiphilic AMPDs can be an effective therapy of E. coli infections. Most of the tryptophan-anchored AMPDs were able to inhibit the growth of antibiotic-resistant E. coli strains, sometimes better than polymyxin B or even indolicidin, besides showing stability in plasma along with low hemolysis and genotoxicity.136

Recently, a novel G3KL (containing repetitive units of lysine and leucine) AMPD showed high potency at low MIC against 35 strains of P. aeruginosa (8–32 μg/mL), 32 strains of A. baumannii (16 μg/mL), E. coli (8 μg/mL), and K. pneumoniae (16–64 μg/mL).17 G3KL is a peptide dendrimer of third generation, which acts as a membrane-disrupting agent against bacteria. G3RL (with repetitive units of arginine and leucine) showed lower efficiency than G3KL against P. aeruginosa (8–32 μg/mL) and Bacillus subtilis (11 μg/mL). Once in contact with the serum, the biological activity of G3RL is inhibited.132

Moreover, both G3KL and G3RL within biological bandages have shown high efficacy against P. aeruginosa, absence of toxicity, and no gene alteration in progenitor fibroblast cells at a concentration of 100 μg/mL. Especially G3KL showed enhanced angiogenesis in human umbilical vein endothelial cells and chorioallantoic membrane assays, as a proof of further potency to enhance wound healing.137 A second-generation AMPD, such as TNS18, has the same activity as G3KL against Gram-negative bacteria, except K. pneumoniae and against Gram-positive MRSA (MIC = 8–16 μg/mL).17

Moreover, D-enantiomeric dendrimers dG3KL and dTNS18 have shown high killing effect against different P. aeruginosa biofilm strains (90.2–100%) in vivo on larvae.138 Therefore, the topology and the sequence of the dendrimers can not only affect their antimicrobial potential, but can also alter their proangiogenic effect, as well. Moreover, the same group of Reymond have developed two different glycopeptide dendrimers: a fucosylated peptide dendrimer (FD2) and two galactosylated dendrimer (GalAG2 and GalBG2), which proved to be potent against P. aeruginosa biofilm formation in vitro.139

The AMPDs show strong potency against multiple bacterial strains and biofilms. Further research is warranted to optimize their delivery to the wounded site, for a potential clinical translation.

Delivery strategies for AMPs

The limitations of AMPs in terms of efficacy, fast degradation, or toxicity require adequate delivery strategies to tackle these challenges. As AMPDs are a relatively new class of antimicrobials, most of the research has been performed on AMPs, which were covalently coupled to biopolymers or encapsulated into nanoparticles or liposomes. The design of AMP nanocarriers could serve as an example of how to render AMPDs even more effective while preserving their bioactivity.

Covalent coupling of AMPs to chitosan–chitosan derivatives

AMPs can be favorable drug candidates to be coupled to biopolymers, such as chitosan or chitosan derivatives to reduce hemolytic effects and/or enhance antibacterial activity, also benefiting from chitosan's bacteriostatic properties. These conjugates have the advantage of increased stability against proteases and peptidases, low immunogenicity, high efficiency and selectivity, and relatively small size that allows AMPs to disrupt bacterial wall.140

  • Anoplin–chitosan: Anoplin (derived from wasp venom) covalently coupled to chitosan showed enhanced in vitro bioactivity and absence of hemolysis. The activity of anoplin–chitosan conjugates against S. aureus and E. coli increased proportionally with their degree of substitution (MIC of anoplin peptide of 1.9 μg/mL against E. coli).141

  • HHC10–chitosan: Cysteine-HHC10 AMP coupled to chitosan showed enhanced bioactivity against S. aureus and S. epidermidis; almost no hemolysis and lower toxicity than HHC10 alone.142

  • hLF1–11–chitosan: Human hLF1–11 covalently coupled to a thiol-derivatized chitosan film lead to a significant increase in S. aureus adhesion against implant-related infections.143

  • Dhvar-5–chitosan: This peptide was immobilized to chitosan films for S. aureus elimination.144

These studies suggest a potential for chitosan–peptide conjugates to improve activity and decrease toxicity compared with the parent peptide. Still, we need further investigations to validate this experimental approach and reveal the mechanisms behind this improvement.

Nanocarrier systems

Encapsulation of peptides into nano- or microcarriers systems can be an efficient approach to lower cytotoxicity, preserve activity by reducing their degradation and enhance their selectivity.145 Among these, liposomes, micelles, polymer nanoparticles, and microparticles have met success for drug delivery. This approach has been applied to some AMPs, potentially improving their pharmacokinetic profile.

Liposomes

Liposomes are self-assembled colloidal systems composed of one or more phospholipid bilayers. They have been studied in the last decades as suitable vehicles for drug delivery due to their encapsulation ability and biocompatibility.145 Liposomes have the advantage of encapsulating both hydrophobic and hydrophilic compounds. Moreover, these drug release systems may protect AMPs against degradation, decrease cytotoxicity, and enhance their stability and bioactivity. For example, Yang et al. incorporated a WLBU2 peptide (24-amino acids) using a modified liposome delivery system with high efficiency against both Gram-positive and Gram-negative bacteria (P. aeruginosa and S. aureus, respectively) and against Chlamydia trachomatis.

Furthermore, WLBU2-modified liposomes were safe to human skin fibroblasts and the activity of the peptide was preserved even in the presence of human serum and blood.146 This AMP-modified liposome system could be potentially used for local infections. An I.V. injection of tuftsin-loaded liposomes in infected animals resulted in site-specific delivery of AMP and stimulation of liver and spleen macrophage functions against antibacterial–antiparasitic infections, such as tuberculosis and leishmaniasis.147

Vancomycin and chitosan-loaded liposomes were shown to not only improve the pharmacokinetic profile of the peptide, but to also reduce nephrotoxicity in mice. Injectable vancomycin liposomes showed high antibacterial efficacy against Gram-positive bacterial infections, a sustained release profile, and prolonged systemic circulation.145 This prevented a vancomycin burst release, which could lead to different side effects.

Micelles

A DP7 (12-amino acids) cationic and hydrophilic AMP, incorporated into a micellar system, showed potent therapeutic benefits in different in vivo disease models and proved to be safe through I.V. injection in mice. The AMP–micellar construct showed reduced hemolysis and high antibacterial activity against S. aureus both in vitro (MIC of DP7-micelles >1,024 μg/mL against P. aeruginosa, S. aureus, and E. coli) and in vivo.

In P. aeruginosa-infected zebrafish embryos and S. aureus-infected mice, DP7-micelles showed high efficacy and therapeutic safety comparable to vancomycin. After an I.V. (80 mg/kg body weight) administration of DP7-micelles, all mice survived and no liver bleeding or pulmonary hemorrhage was observed.148 These AMP-micelle formulations may potentially be used for bacterial infections (in both Gram-positive and Gram-negative species) as they were shown to significantly stimulate defensive immune reactions in vivo as well.

Micro- and nanoparticles

Vancomycin has been loaded into polycaprolactone polymer microparticles to minimize the side effects of vancomycin. These microparticles were coadministered with calcium phosphate bone substitutes for preventing postsurgery infection. The encapsulation of vancomycin into microparticles resulted in prolonged peptide release in vitro over several weeks.149 Vancomycin was successfully encapsulated into poly(lactide-co-glycolide) (PLGA) polymers to prevent external–internal ocular bacterial infections.150 Thus, vancomycin encapsulated into nanodelivery systems may successfully be used as an alternative treatment of infections caused by MDR bacteria.

Piras et al. could formulate an efficient nanoparticles system against S. epidermidis by ionic gelation method. Peptide LLPIVGNLLKSLL-amide (called TB) was added to chitosan to form NPs. The encapsulation of TB peptide exhibited high bactericidal properties against S. epidermidis strains and significantly reduced the toxicity against mammalian cells.151 Another RBRBR peptide was encapsulated into chitosan NPs by a similar method resulting in an enhanced activity against S. aureus and significantly reduced hemolysis and cytotoxicity.152 Thus, chitosan may act as an antimicrobial activity enhancer and/or significantly limit the toxicity of the AMPs.

d'Angelo et al. engineered a colistin-loaded PLGA nanoparticles system for sustained delivery of the peptide against P. aeruginosa in lung infection. Colistin encapsulated into PLGA NPs could easily penetrate an artificial mucus layer during the first 6 h and successfully eliminated P. aeruginosa biofilm in vitro within 72 h at 7.5 and 15 μg/mL.153

These few studies of AMPs covalently coupled or noncovalently associated to different biopolymers suggest that AMPs are potent candidates to eradicate MDR bacterial infections at an enhanced antimicrobial activity and lower toxicity. Additionally, achieving optimum drug–AMP loading, using the right and safe polymer, storing the new formulation while preserving the bioactivity and stability of the AMP are definitely to be further evaluated.

Summary

The occurrence of AMR has changed the landscape of the drugs used in clinics, more specifically to treat ESKAPE-related infections. In addition, the complexity of the factors affecting wound healing renders the choice of adequate antimicrobial agents difficult. In this context, alternative strategies to overcome AMR are proposed.

Besides clinically approved antimicrobial and antiseptic agents, synthetic dendritic polymers and novel peptide dendrimers (AMPDs) have recently shown promising results in preclinical models of infection. Further strategies are also available to improve their activity and decrease toxicity compared with the parent peptides: the conjugation with (bio)polymers, or the incorporation into carriers such as liposomes, nano- or microparticles. These strategies may allow for a sustained pharmacokinetic profile and improve the activity against MDR bacterial infections, paving the way toward their use in a clinical setup.

Take-Home Messages

  • The occurrence of AMR has changed the landscape of the drugs used in clinics, more specifically to treat ESKAPE-related infections.

  • Antimicrobial peptidic agents are highly potent with a broad activity against Gram (+) and (−) bacteria and microorganisms.

  • AMPs are good candidates to limit resistance-induced microorganisms, benefiting from their broad activity spectrum and the variety of their mechanisms of action.

  • The performance of AMPs can be further enhanced by several strategies such as: chemical conjugation to biopolymers or organization in a dendritic structure.

  • Nanocarrier technology may further improve pharmacokinetic profile to increase antimicrobial effect and reduce toxicity.

  • AMPDs show strong potency against multiple bacterial strains and biofilms. Further research is warranted to optimize their delivery to the wounded site, for a potential clinical translation.

Acknowledgment and Funding Sources

No external funding sources were used in the preparation of this article.

Abbreviations and Acronyms

AgNO3

silver nitrate

AMP

antimicrobial peptide

AMPD

antimicrobial peptide dendrimer

AMR

antimicrobial resistance

CLSI

Clinical and Laboratory Standards Institute

ESKAPE

Enterococcus faecium, Staphylococcus aureus, Klebsiella pneumoniae, Acinetobacter baumannii, Pseudomonas aeruginosa, and Enterobacter species

FDA

U.S. Food and Drug Administration

Gram (+)

Gram-positive

Gram (−)

Gram-negative

H2O2

hydrogen peroxide

hBD

human β-defensin

HC

hemolytic concentration

hLF

human lactoferrin fragment

HNC

head and neck cancer

I.M.

intramuscular

I.T.

intrathecal

I.V.

intravenous injection

LPS

lipopolysaccharide

MBC

minimal bactericidal concentration

MDR

multidrug resistant

MIC

minimal inhibitory concentration

MRSA

methicillin-resistant S. aureus

PAMAM

poly(amidoamine)

PC

preclinical

Pg1

protegrin-1

PHMB

polyhexa-methylene biguanide

PLGA

poly(lactide-co-glycolide)

RBC

red blood cell

TAA

topical antimicrobials and antiseptics

VRE

vancomycin-resistant enterococci

WHO

World Health Organization

Author Disclosure and Ghostwriting

All authors confirm no conflict of interest and no ghost writers were used to write this article.

About the Authors

Yayoi Kawano, PhD, is Associate Professor at Faculty of Pharmaceutical Sciences, Tokyo University of Science, Japan. Her research interests are to incorporate poorly water-soluble drugs into different drug delivery systems, and to evaluate formulations for wound healing. Olivier Jordan, PhD, is Senior Lecturer at the University of Geneva. His research interests lie in the field of innovative carriers for drug, peptide, or protein delivery based on biopolymers, including hyaluronic acid or chitosan. Takehisa Hanawa, PhD, is Professor at the Faculty of Pharmaceutical Sciences of Tokyo University of Science. His research interests are to prepare and apply nanoparticle, hydrogel, and film formulations for various external formulations. Gerrit Borchard, PharmD, PhD, is a Full Professor in Biopharmaceutics at the University of Geneva. His group s research focuses on the interaction of drug and vaccine formulations with biological systems, including immunotherapy of cancer and infectious diseases, development and characterization of nanomedicines applied in these indications. Viorica Patrulea, PhD, is a postdoctoral fellow at the University of Geneva. She has developed a new technological platform based on her expertise in polymer chemistry and complemented by the knowledge of biological and pharmaceutical aspects of the development of formulations for wound healing and protection against microbial infection.

References

  • 1. Ambekar RS, Kandasubramanian B. Advancements in nanofibers for wound dressing: a review. Eur Polym J 2019;117:304–336 [Google Scholar]
  • 2. Jasovský D, Littmann J, Zorzet A, Cars O. Antimicrobial resistance—a threat to the world's sustainable development. Ups J Med Sci 2016;121:159–164 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 3. Tiwari V. Post-translational modification of ESKAPE pathogens as a potential target in drug discovery. Drug Discov Today 2019;24:814–822 [DOI] [PubMed] [Google Scholar]
  • 4. Laxminarayan R, Duse A, Wattal C, et al. Antibiotic resistance—the need for global solutions. Lancet Inf Dis 2013;13:1057–1098 [DOI] [PubMed] [Google Scholar]
  • 5. Koulenti D, Xu E, Mok IYS, et al. Novel antibiotics for multidrug-resistant gram-positive microorganisms. Microorganisms 2019;7:270–294 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 6. Nigo M, Diaz L, Carvajal LP, et al. Ceftaroline-resistant, daptomycin-tolerant, and heterogeneous vancomycin-intermediate methicillin-resistant Staphylococcus aureus causing infective endocarditis. Antimicrob Agents Chemother 2017;61:e01235-16 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7. Long SW, Olsen RJ, Mehta SC, et al. PBP2a mutations causing high-level Ceftaroline resistance in clinical methicillin-resistant Staphylococcus aureus isolates. Antimicrob Agents Chemother 2014;58:6668–6674 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 8. Santajit S, Indrawattana N. Mechanisms of antimicrobial resistance in ESKAPE pathogens. Biomed Res Int 2016;2016:2475067. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 9. Caio C, Maugeri G, Zingali T, et al. Extensively drug-resistant ArmA-producing Acinetobacter baumannii in an Italian intensive care unit. New Microbiol 2018;41:159–161 [PubMed] [Google Scholar]
  • 10. Chuang L, Ratnayake L. Overcoming challenges of treating extensively drug-resistant Acinetobacter baumannii bacteraemic urinary tract infection. Int J Antimicrob Agents 2018;52:521–522 [DOI] [PubMed] [Google Scholar]
  • 11. Nowak J, Zander E, Stefanik D, et al. High incidence of pandrug-resistant Acinetobacter baumannii isolates collected from patients with ventilator-associated pneumonia in Greece, Italy and Spain as part of the MagicBullet clinical trial. J Antimicrob Chemother 2017;72:3277–3282 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12. Mohapatra DP, Debata NK, Singh SK. Extensively drug-resistant and pandrug-resistant Gram-negative bacteria in a tertiary-care hospital in Eastern India: a 4-year retrospective study. J Glob Antimicrob Resist 2018;15:246–249 [DOI] [PubMed] [Google Scholar]
  • 13. Gill JS, Arora S, Khanna SP, Kumar KH. Prevalence of multidrug-resistant, extensively drug-resistant, and Pandrug-resistant Pseudomonas aeruginosa from a tertiary level intensive care unit. J Glob Infect Dis 2016;8:155–159 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 14. Kulengowski B, Rutter WC, Campion JJ, et al. Effect of increasing meropenem MIC on the killing activity of meropenem in combination with amikacin or polymyxin B against MBL- and KPC-producing Enterobacter cloacae. Diagn Microbiol Infect Dis 2018;92:262–266 [DOI] [PubMed] [Google Scholar]
  • 15. Alves PH, Boff RT, Barth AL, Martins AF. Synergy of polymyxin B, tigecycline and meropenem against carbapenem-resistant Enterobacter cloacae complex isolates. Diagn Microbiol Infect Dis 2019;94:81–85 [DOI] [PubMed] [Google Scholar]
  • 16. Giuliani A, Rinaldi AC. Beyond natural antimicrobial peptides: multimeric peptides and other peptidomimetic approaches. Cell Mol Life Sci 2011;68:2255–2266 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 17. Siriwardena TN, Lüscher A, Köhler T, et al. Antimicrobial peptide dendrimer chimera. Helv Chim Acta 2019;102:e1900034 [Google Scholar]
  • 18. Han G, Ceilley R. Chronic wound healing: a review of current management and treatments. Adv Ther 2017;34:599–610 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 19. Lipsky BA, Dryden M, Gottrup F, et al. Antimicrobial stewardship in wound care: a Position Paper from the British Society for Antimicrobial Chemotherapy and European Wound Management Association. J Antimicrob Chemother 2016;71:3026–3035 [DOI] [PubMed] [Google Scholar]
  • 20. Frykberg RG, Banks J. Challenges in the treatment of chronic wounds. Adv Wound Care 2015;4:560–582 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 21. Patrulea V, Ostafe V, Borchard G, Jordan O. Chitosan as a starting material for wound healing applications. Eur J Pharm Biopharm 2015;97:417–426 [DOI] [PubMed] [Google Scholar]
  • 22. Halim AS, Khoo TL, Saad AZM. Wound bed preparation from a clinical perspective. Indian J Plast Surg 2012;45:193–202 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23. Bjarnsholt T, Kirketerp-Møller K, Jensen PØ, et al. Why chronic wounds will not heal: a novel hypothesis. Wound Repair Regen 2008;16:2–10 [DOI] [PubMed] [Google Scholar]
  • 24. Fritz JM, McDonald JR. Osteomyelitis: approach to diagnosis and treatment. Physician Sportsmed 2008;36:nihpa116823. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 25. Hitchman LH, Smith GE, Chetter IC. Prosthetic infections and high-risk surgical populations. Surgery (Oxford) 2019;37:38–44 [Google Scholar]
  • 26. Uçkay I, Aragón-Sánchez J, Lew D, Lipsky BA. Diabetic foot infections: what have we learned in the last 30 years? Int J Infect Dis 2015;40:81–91 [DOI] [PubMed] [Google Scholar]
  • 27. Strohal R, Mittlbock M, Hammerle G. The management of critically colonized and locally infected leg ulcers with an acid-oxidizing solution: a pilot study. Adv Skin Wound Care 2018;31:163–171 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28. Lipsky BA, Hoey C. Topical antimicrobial therapy for treating chronic wounds. Clin Infect Dis 2009;49:1541–1549 [DOI] [PubMed] [Google Scholar]
  • 29. Edwardson S, Cairns C. Nosocomial infections in the ICU. Anaesth Intens Care Med 2019;20:14–18 [Google Scholar]
  • 30. Wu Y-L, Yang X-Y, Ding X-X, et al. Exposure to infected/colonized roommates and prior room occupants increases the risks of healthcare-associated infections with the same organism. J Hosp Inf 2019;101:231–239 [DOI] [PubMed] [Google Scholar]
  • 31. Beceiro A, Tomás M, Bou G. Antimicrobial resistance and virulence: a successful or deleterious association in the bacterial world? Clin Microb Rev 2013;26:185–230 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32. Filius PM, Gyssens IC. Impact of increasing antimicrobial resistance on wound management. Am J Clin Dermatol 2002;3:1–7 [DOI] [PubMed] [Google Scholar]
  • 33. Contreras-Marín M, Sandoval-Rodríguez JI, García-Ramírez R, Morales-Yépez HA. Mammal bite management [in Spanish]. Cir Cir 2016;84:525–530 [DOI] [PubMed] [Google Scholar]
  • 34. Pouwels KB, Hopkins S, Llewelyn MJ, et al. Duration of antibiotic treatment for common infections in English primary care: cross sectional analysis and comparison with guidelines. BMJ 2019;364:l440. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 35. Omar A, Wright JB, Schultz G, Burrell R, Nadworny P. Microbial biofilms and chronic wounds. Microorganisms 2017;5:E9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36. Konai MM, Bhattacharjee B, Ghosh S, Haldar J. Recent progress in polymer research to tackle infections and antimicrobial resistance. Biomacromolecules 2018;19:1888–1917 [DOI] [PubMed] [Google Scholar]
  • 37. Rabin N, Zheng Y, Opoku-Temeng C, et al. Biofilm formation mechanisms and targets for developing antibiofilm agents. Future Med Chem 2015;7:493–512 [DOI] [PubMed] [Google Scholar]
  • 38. Ashrafi M, Novak-Frazer L, Bates M, et al. Validation of biofilm formation on human skin wound models and demonstration of clinically translatable bacteria-specific volatile signatures. Sci Rep 2018;8:9431. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39. Kanji S, Das H. Advances of stem cell therapeutics in cutaneous wound healing and regeneration. Mediators Inflamm 2017;2017:5217967. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40. Abdel-Sayed P, Hirt-Burri N, de Buys Roessingh A, Raffoul W, Applegate LA. Evolution of biological bandages as first cover for burn patients. Adv Wound Care 2019;8:555–564 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41. Qu J, Zhao X, Liang Y, et al. Antibacterial adhesive injectable hydrogels with rapid self-healing, extensibility and compressibility as wound dressing for joints skin wound healing. Biomaterials 2018;183:185–199 [DOI] [PubMed] [Google Scholar]
  • 42. Erring M, Gaba S, Mohsina S, Tripathy S, Sharma RK. Comparison of efficacy of silver-nanoparticle gel, nano-silver-foam and collagen dressings in treatment of partial thickness burn wounds. Burns 2019;45:1888–1894 [DOI] [PubMed] [Google Scholar]
  • 43. Punjataewakupt A, Napavichayanun S, Aramwit P. The downside of antimicrobial agents for wound healing. Eur J Clin Microbiol Inf Dis 2019;38:39–54 [DOI] [PubMed] [Google Scholar]
  • 44. Dumville JC, Lipsky BA, Hoey C, et al. Topical antimicrobial agents for treating foot ulcers in people with diabetes. Cochrane Database Syst Rev 2017;6:CD011038. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 45. Bora I, Demir AB, Uzun P. Nonconvulsive status epilepticus cases arising in connection with cephalosporins. Epilepsy Behav Case Rep 2016;6:23–27 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46. Suemaru K, Yoshikawa M, Aso H, Watanabe M. 5-Fluorouracil exacerbates cefepime-induced convulsions in pentylenetetrazol-kindled mice. Epilepsy Res 2019;157:106195. [DOI] [PubMed] [Google Scholar]
  • 47. Triplett JD, Lawn ND, Chan J, Dunne JW. Cephalosporin-related neurotoxicity: metabolic encephalopathy or non-convulsive status epilepticus? J Cli Neurosci 2019;67:163–166 [DOI] [PubMed] [Google Scholar]
  • 48. Greenhalgh DG. Topical antimicrobial agents for burn wounds. Clin Plast Surg 2009;36:597–606 [DOI] [PubMed] [Google Scholar]
  • 49. Khan MA, Bahadar S, Ullah N, et al. Distribution and antimicrobial resistance patterns of Clostridium perfringens isolated from vaccinated and unvaccinated goats. Small Ruminant Res 2019;173:70–73 [Google Scholar]
  • 50. Oh E, Bae J, Kumar A, Choi H-J, Jeon B. Antioxidant-based synergistic eradication of methicillin-resistant Staphylococcus aureus (MRSA) biofilms with bacitracin. Int J Antimicrob Agents 2018;52:96–99 [DOI] [PubMed] [Google Scholar]
  • 51. Jaffe L, Wu SC. Dressings, topical therapy, and negative pressure wound therapy. Clin Podiatr Med Sur 2019;36:397–411 [DOI] [PubMed] [Google Scholar]
  • 52. Beers EH. Palliative wound care: less is more. Surg Clin N Am 2019;99:899–919 [DOI] [PubMed] [Google Scholar]
  • 53. Rachid A, Christophe M, Marc B-M, et al. Methemoglobinemia by cerium nitrate poisoning. Burns 2006;32:1060–1061 [DOI] [PubMed] [Google Scholar]
  • 54. Banaeian S, Sereshti M, Rafieian M, Farahbod F, Kheiri S. Comparison of vaginal ointment of honey and clotrimazole for treatment of vulvovaginal candidiasis: a random clinical trial. J Mycol Med 2017;27:494–500 [DOI] [PubMed] [Google Scholar]
  • 55. Davey RX, Tong SYC. The epidemiology of Staphylococcus aureus skin and soft tissue infection in the southern Barkly region of Australia's Northern Territory in 2017. Pathology 2019;51:308–312 [DOI] [PubMed] [Google Scholar]
  • 56. Genuino GAS, Baluyut-Angeles KV, Espiritu APT, Lapitan MCM, Buckley BS. Topical petrolatum gel alone versus topical silver sulfadiazine with standard gauze dressings for the treatment of superficial partial thickness burns in adults: a randomized controlled trial. Burns 2014;40:1267–1273 [DOI] [PubMed] [Google Scholar]
  • 57. Pasmooij AMG. Topical gentamicin for the treatment of genetic skin diseases. J Investig Dermatol 2018;138:731–734 [DOI] [PubMed] [Google Scholar]
  • 58. Tsai C-C, Yang P-S, Liu C-L, et al. Comparison of topical mupirocin and gentamicin in the prevention of peritoneal dialysis-related infections: a systematic review and meta-analysis. Am J Sur 2018;215:179–185 [DOI] [PubMed] [Google Scholar]
  • 59. Zeleníková R, Vyhlídalová D. Applying honey dressings to non-healing wounds in elderly persons receiving home care. J Tissue Viability 2019;28:139–143 [DOI] [PubMed] [Google Scholar]
  • 60. Wang C, Guo M, Zhang N, Wang G. Effectiveness of honey dressing in the treatment of diabetic foot ulcers: a systematic review and meta-analysis. Complement Ther Clin Pract 2019;34:123–131 [DOI] [PubMed] [Google Scholar]
  • 61. Kiran S, Marwah S, Bansal T, Gupta N. Venous air/oxygen embolism due to hydrogen peroxide in anal fistulectomy. J Anaesthesiol Clin Pharmacol 2018;34:555–557 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 62. Ibrahim A, Fagan S, Keaney T, et al. A simple cost-saving measure: 2.5% mafenide acetate solution. J Burn Care Res 2014;35:349–353 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 63. Story S, Skriba MJ, Maiti K, et al. Synthesis, antimicrobial activity, attenuation of aminoglycoside resistance in MRSA, and ribosomal A-site binding of pyrene-neomycin conjugates. Eur J Med Chem 2019;163:381–393 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64. Sun P, Yu F, Lu J, et al. In vivo effects of neomycin sulfate on non-specific immunity, oxidative damage and replication of cyprinid herpesvirus 2 in crucian carp (Carassius auratus gibelio). Aquacult Fish 2019;4:67–73 [Google Scholar]
  • 65. Leon-Villapalos J, Jeschke MG, Herndon DN. Topical management of facial burns. Burns 2008;34:903–911 [DOI] [PubMed] [Google Scholar]
  • 66. Thornton Spann C, Taylor SC, Weinberg JM. Topical antimicrobial agents in dermatology. Dis Mon 2004;50:407–421 [DOI] [PubMed] [Google Scholar]
  • 67. AbouSamra MM, Basha M, Awad GEA, Mansy SS. A promising nystatin nanocapsular hydrogel as an antifungal polymeric carrier for the treatment of topical candidiasis. J Drug Deliv Sci Technol 2019;49:365–374 [Google Scholar]
  • 68. Kautz O, Schumann H, Degerbeck F, Venemalm L, Jakob T. Severe anaphylaxis to the antiseptic polyhexanide. Allergy 2010;65:1068–1070 [DOI] [PubMed] [Google Scholar]
  • 69. Thomas VM, Brown RM, Ashcraft DS, Pankey GA. Synergistic effect between nisin and polymyxin B against pandrug-resistant and extensively drug-resistant Acinetobacter baumannii. Int J Antimicrob Agents 2019;53:663–668 [DOI] [PubMed] [Google Scholar]
  • 70. Leung AM, Braverman LE. Consequences of excess iodine. Nat Rev Endocrinol 2013;10:136. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 71. Sato S, Miyake M, Hazama A, Omori K. Povidone-iodine-induced cell death in cultured human epithelial HeLa cells and rat oral mucosal tissue. Drug Chem Toxicol 2014;37:268–275 [DOI] [PubMed] [Google Scholar]
  • 72. Vogt PM, Reimer K, Hauser J, et al. PVP-iodine in hydrosomes and hydrogel—a novel concept in wound therapy leads to enhanced epithelialization and reduced loss of skin grafts. Burns 2006;32:698–705 [DOI] [PubMed] [Google Scholar]
  • 73. Sterling JP. Silver-resistance, allergy, and blue skin: truth or urban legend? Burns 2014;40:S19–S23 [DOI] [PubMed] [Google Scholar]
  • 74. Moyano AJ, Mas CR, Colque CA, Smania AM. Dealing with biofilms of Pseudomonas aeruginosa and Staphylococcus aureus: in vitro evaluation of a novel aerosol formulation of silver sulfadiazine. Burns 2020;46:128–135 [DOI] [PubMed] [Google Scholar]
  • 75. Mohseni M, Shamloo A, Aghababaie Z, et al. A comparative study of wound dressings loaded with silver sulfadiazine and silver nanoparticles: in vitro and in vivo evaluation. Int J Pharm 2019;564:350–358 [DOI] [PubMed] [Google Scholar]
  • 76. Salazar-Mercado SA, Torres-León CA, Rojas-Suárez JP. Cytotoxic evaluation of sodium hypochlorite, using Pisum sativum L as effective bioindicator. Ecotoxicol Environ Saf 2019;173:71–76 [DOI] [PubMed] [Google Scholar]
  • 77. Luddin N, Ahmed HMA. The antibacterial activity of sodium hypochlorite and chlorhexidine against Enterococcus faecalis: a review on agar diffusion and direct contact methods. J Conserv Dent 2013;16:9–16 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 78. Bohaty BR, Choi S, Cai C, Hebert AA. Clinical and bacteriological efficacy of twice daily topical retapamulin ointment 1% in the management of impetigo and other uncomplicated superficial skin infections. Int J Womens Dermatol 2015;1:13–20 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 79. Iliescu Nelea M, Paek L, Dao L, et al. In-situ characterization of the bacterial biofilm associated with Xeroform™ and Kaltostat™ dressings and evaluation of their effectiveness on thin skin engraftment donor sites in burn patients. Burns 2019;45:1122–1130 [DOI] [PubMed] [Google Scholar]
  • 80. Barillo DJ, Barillo AR, Korn S, Lam K, Attar PS. The antimicrobial spectrum of Xeroform®. Burns 2017;43:1189–1194 [DOI] [PubMed] [Google Scholar]
  • 81. Roncevic T, Puizina J, Tossi A. Antimicrobial peptides as anti-infective agents in pre-post-antibiotic era? Int J Mol Sci 2019;20:5713. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 82. Dubos RJ. Studies on a bactericidal agent extracted from a soil bacillus: I. Preparation of the agent. Its activity in vitro. J Exp Med 1939;70:1–10 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 83. Dubos RJ, Hotchkiss RD. The production of bactericidal substances by aerobic sporulating bacili. J Exp Med 1941;73:629–640 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84. Wang G, Li X, Wang Z. APD3: the antimicrobial peptide database as a tool for research and education. Nucleic Acids Res 2016;44:D1087–D1093 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 85. The Antimicrobial Peptide Database (APD). http://aps.unmc.edu/AP (last accessed October1, 2019)
  • 86. Andersson DI, Hughes D, Kubicek-Sutherland JZ. Mechanisms and consequences of bacterial resistance to antimicrobial peptides. Drug Resist Updat 2016;26:43–57 [DOI] [PubMed] [Google Scholar]
  • 87. Kuroda K, Okumura K, Isogai H, Isogai E. The human cathelicidin antimicrobial peptide LL-37 and mimics are potential anticancer drugs. Front Oncol 2015;5:144. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 88. Powers J-PS, Hancock REW. The relationship between peptide structure and antibacterial activity. Peptides 2003;24:1681–1691 [DOI] [PubMed] [Google Scholar]
  • 89. Khandelia H, Kaznessis YN. Cation-pi interactions stabilize the structure of the antimicrobial peptide indolicidin near membranes: molecular dynamics simulations. J Phys Chem B 2007;111:242–250 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 90. Li X, Duan D, Yang J, et al. The expression of human β-defensins (hBD-1, hBD-2, hBD-3, hBD-4) in gingival epithelia. Arch Oral Biol 2016;66:15–21 [DOI] [PubMed] [Google Scholar]
  • 91. Vandamme D, Landuyt B, Luyten W, Schoofs L. A comprehensive summary of LL-37, the factotum human cathelicidin peptide. Cell Immunol 2012;280:22–35 [DOI] [PubMed] [Google Scholar]
  • 92. Bergman P, Johansson L, Asp V, et al. Neisseria gonorrhoeae downregulates expression of the human antimicrobial peptide LL-37. Cell Microbiol 2005;7:1009–1017 [DOI] [PubMed] [Google Scholar]
  • 93. Grönberg A, Mahlapuu M, Ståhle M, Whately-Smith C, Rollman O. Treatment with LL-37 is safe and effective in enhancing healing of hard-to-heal venous leg ulcers: a randomized, placebo-controlled clinical trial. Wound Repair Regen 2014;22:613–621 [DOI] [PubMed] [Google Scholar]
  • 94. Du H, Puri S, McCall A, et al. Human salivary protein Histatin 5 has potent bactericidal activity against ESKAPE pathogens. Front Cell Infect Microbiol 2017;7:41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 95. Maria-Neto S, de Almeida KC, Macedo MLR, Franco OL. Understanding bacterial resistance to antimicrobial peptides: from the surface to deep inside. Biochim Biophys Acta 2015;1848(Pt B):3078–3088 [DOI] [PubMed] [Google Scholar]
  • 96. Belas R, Manos J, Suvanasuthi R. Proteus mirabilis ZapA metalloprotease degrades a broad spectrum of substrates, including antimicrobial peptides. Infect Immun 2004;72:5159–5167 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 97. Overhage J, Campisano A, Bains M, et al. Human host defense peptide LL-37 prevents bacterial biofilm formation. Infect Immun 2008;76:4176–4182 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98. Buch PJ, Chai Y, Goluch ED. Treating polymicrobial infections in chronic diabetic wounds. Clin Microbiol Rev 2019;32:e00091-00018 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99. Lee EY, Lee MW, Fulan BM, Ferguson AL, Wong GCL. What can machine learning do for antimicrobial peptides, and what can antimicrobial peptides do for machine learning? Interface Focus 2017;7:20160153. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 100. Bahar AA, Ren D. Antimicrobial peptides. Pharmaceuticals (Basel) 2013;6:1543–1575 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 101. Sani M-A, Separovic F. How membrane-active peptides get into lipid membranes. Acc Chem Res 2016;49:1130–1138 [DOI] [PubMed] [Google Scholar]
  • 102. Sahariah P, Benediktssdóttir BE, Hjálmarsdóttir MÁ, et al. Impact of chain length on antibacterial activity and hemocompatibility of quaternary N-alkyl and N,N-dialkyl chitosan derivatives. Biomacromolecules 2015;16:1449–1460 [DOI] [PubMed] [Google Scholar]
  • 103. Steigbigel RT, Steigbigel NH. Static vs cidal antibiotics. Clin Infect Dis 2019;68:351–352 [DOI] [PubMed] [Google Scholar]
  • 104. Wenzel M, Rautenbach M, Vosloo JA, et al. The multifaceted antibacterial mechanisms of the pioneering peptide antibiotics tyrocidine and gramicidin S. mBio 2018;9:e00802-18 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 105. Kane LE, Muzevich KM. Micafungin in the treatment of candiduria: a case series. Med Mycol Case Rep 2016;11:5–8 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 106. Food and Drug Administration (FDA) Center for drug evaluation and research. Approved drug products with Therapeutic Equivalence Evaluations. https://www.fda.gov/media/71474/download (last accessed September29, 2019)
  • 107. Lei J, Sun L, Huang S, et al. The antimicrobial peptides and their potential clinical applications. Am J Transl Res 2019;11:3919–3931 [PMC free article] [PubMed] [Google Scholar]
  • 108. Koo HB, Seo J. Antimicrobial peptides under clinical investigation. Pept Sci 2019;111:e24122 [Google Scholar]
  • 109. Mensa B, Howell GL, Scott R, DeGrado WF. Comparative mechanistic studies of brilacidin, daptomycin, and the antimicrobial peptide LL16. Antimicrob Agents Chemother 2014;58:5136–5145 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110. Kuppusamy R, Willcox M, Black DS, Kumar N. Short cationic peptidomimetic antimicrobials. Antibiotics (Basel) 2019;8:44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111. Kudrimoti M, Curtis A, Azawi S, et al. Dusquetide: a novel innate defense regulator demonstrating a significant and consistent reduction in the duration of oral mucositis in preclinical data and a randomized, placebo-controlled phase 2a clinical study. J Biotechnol 2016;239:115–125 [DOI] [PubMed] [Google Scholar]
  • 112. Hamill P, Brown K, Jenssen H, Hancock RE. Novel anti-infectives: is host defence the answer? Curr Opin Biotechnol 2008;19:628–636 [DOI] [PubMed] [Google Scholar]
  • 113. van der Velden WJFM, van Iersel TMP, Blijlevens NMA, Donnelly JP. Safety and tolerability of the antimicrobial peptide human lactoferrin 1–11 (hLF1–11). BMC Med 2009;7:44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 114. Ciumac D, Gong H, Hu X, Lu JR. Membrane targeting cationic antimicrobial peptides. J Colloid Interface Sci 2019;537:163–185 [DOI] [PubMed] [Google Scholar]
  • 115. U.S. National Library of Medicine. Clinical trials. https://clinicaltrials.gov/ct2/show/NCT01803035 (last accessed September29, 2019)
  • 116. Felicio MR, Silva ON, Goncalves S, Santos NC, Franco OL. Peptides with dual antimicrobial and anticancer activities. Front Chem 2017;5:5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117. Boto A, Pérez de la Lastra JM, González CC. The road from host-defense peptides to a new generation of antimicrobial drugs. Molecules 2018;23:311. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 118. U.S. National Library of Medicine. Clinical trials. https://clinicaltrials.gov/ct2/show/results/NCT01594762 (last accessed September29, 2019)
  • 119. Lipsky BA, Holroyd KJ, 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 [DOI] [PubMed] [Google Scholar]
  • 120. Rabanal F, Cajal Y. Recent advances and perspectives in the design and development of polymyxins. Nat Prod Rep 2017;34:886–908 [DOI] [PubMed] [Google Scholar]
  • 121. Biswaro LS, da Costa Sousa MG, Rezende TMB, Dias SC, Franco OL. Antimicrobial peptides and nanotechnology, recent advances and challenges. Front Microbiol 2018;9:855. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 122. Mahlapuu M, Håkansson J, Ringstad L, Björn C. Antimicrobial peptides: an emerging category of therapeutic agents. Front Cell Infect Microbiol 2016;6:194. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 123. Molchanova N, Hansen PR, Franzyk H. Advances in development of antimicrobial peptidomimetics as potential drugs. Molecules 2017;22:E1430. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 124. U.S. National Library of Medicine. Clinical trials. https://clinicaltrials.gov/ct2/show/NCT02571998 (last accessed September29, 2019)
  • 125. Martin-Serrano Á, Gómez R, Ortega P, de la Mata FJ. Nanosystems as vehicles for the delivery of antimicrobial peptides (AMPs). Pharmaceutics 2019;11:448. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 126. Calabretta MK, Kumar A, McDermott AM, Cai C. Antibacterial activities of poly(amidoamine) dendrimers terminated with amino and poly(ethylene glycol) groups. Biomacromolecules 2007;8:1807–1811 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 127. Lopez AI, Reins RY, McDermott AM, Trautner BW, Cai C. Antibacterial activity and cytotoxicity of PEGylated poly(amidoamine) dendrimers. Mol Biosyst 2009;5:1148–1156 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 128. Wang B, Navath RS, Menjoge AR, et al. Inhibition of bacterial growth and intramniotic infection in a guinea pig model of chorioamnionitis using PAMAM dendrimers. Int J Pharm 2010;395:298–308 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 129. Menjoge AR, Navath RS, Asad A, et al. Transport and biodistribution of dendrimers across human fetal membranes: implications for intravaginal administration of dendrimer-drug conjugates. Biomaterials 2010;31:5007–5021 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130. Pieters RJ. Intervention with bacterial adhesion by multivalent carbohydrates. Med Res Rev 2007;27:796–816 [DOI] [PubMed] [Google Scholar]
  • 131. Zeng Y, Kurokawa Y, Win-Shwe TT, et al. Effects of PAMAM dendrimers with various surface functional groups and multiple generations on cytotoxicity and neuronal differentiation using human neural progenitor cells. J Toxicol Sci 2016;41:351–370 [DOI] [PubMed] [Google Scholar]
  • 132. Stach M, Siriwardena TN, Köhler T, et al. Combining topology and sequence design for the discovery of potent antimicrobial peptide dendrimers against multidrug-resistant Pseudomonas aeruginosa. Angew Chem 2014;53:12827–12831 [DOI] [PubMed] [Google Scholar]
  • 133. Tam JP, Lu Y-A, Yang J-L. Antimicrobial dendrimeric peptides. Eur J Biochem 2002;269:923–932 [DOI] [PubMed] [Google Scholar]
  • 134. Scorciapino MA, Serra I, Manzo G, Rinaldi AC. Antimicrobial dendrimeric peptides: structure, activity and new therapeutic applications. Int J Mol Sci 2017;18:542. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 135. Scorciapino Mariano A, Pirri G, Vargiu Attilio V, et al. A novel dendrimeric peptide with antimicrobial properties: structure-function analysis of SB056. Biophys J 2012;102:1039–1048 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 136. Sowińska M, Laskowska A, Guśpiel A, et al. Bioinspired amphiphilic peptide dendrimers as specific and effective compounds against drug resistant clinical isolates of E. coli. Bioconjugate Chem 2018;29:3571–3585 [DOI] [PubMed] [Google Scholar]
  • 137. Abdel-Sayed P, Kaeppeli A, Siriwardena T, et al. Anti-microbial dendrimers against multidrug-resistant P. aeruginosa enhance the angiogenic effect of biological burn-wound bandages. Sci Rep 2016;6:22020. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 138. Pompilio A, Geminiani C, Mantini P, et al. Peptide dendrimers as “lead compounds” for the treatment of chronic lung infections by Pseudomonas aeruginosa in cystic fibrosis patients: in vitro and in vivo studies. Infect Drug Resist 2018;11:1767–1782 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 139. Reymond J-L, Bergmann M, Darbre T. Glycopeptide dendrimers as Pseudomonas aeruginosa biofilm inhibitors. Chem Soc Rev 2013;42:4814–4822 [DOI] [PubMed] [Google Scholar]
  • 140. Vlieghe P, Lisowski V, Martinez J, Khrestchatisky M. Synthetic therapeutic peptides: science and market. Drug Discov Today 2010;15:40–56 [DOI] [PubMed] [Google Scholar]
  • 141. Sahariah P, Sørensen KK, Hjálmarsdóttir MÁ, et al. Antimicrobial peptide shows enhanced activity and reduced toxicity upon grafting to chitosan polymers. Chem Commun (Camb) 2015;51:11611–11614 [DOI] [PubMed] [Google Scholar]
  • 142. Pranantyo D, Xu LQ, Kang E-T, Chan-Park MB. Chitosan-based peptidopolysaccharides as cationic antimicrobial agents and antibacterial coatings. Biomacromolecules 2018;19:2156–2165 [DOI] [PubMed] [Google Scholar]
  • 143. Costa F, Maia S, Gomes J, Gomes P, Martins MC. Characterization of hLF1–11 immobilization onto chitosan ultrathin films, and its effects on antimicrobial activity. Acta Biomater 2014;10:3513–3521 [DOI] [PubMed] [Google Scholar]
  • 144. Costa FM, Maia SR, Gomes PA, Martins MC. Dhvar5 antimicrobial peptide (AMP) chemoselective covalent immobilization results on higher antiadherence effect than simple physical adsorption. Biomaterials 2015;52:531–538 [DOI] [PubMed] [Google Scholar]
  • 145. Yang Z, Liu J, Gao J, Chen S, Huang G. Chitosan coated vancomycin hydrochloride liposomes: characterizations and evaluation. Int J Pharm 2015;495:508–515 [DOI] [PubMed] [Google Scholar]
  • 146. Yang K, Gitter B, Rüger R, et al. Antimicrobial peptide-modified liposomes for bacteria targeted delivery of temoporfin in photodynamic antimicrobial chemotherapy. Photochem Photobiol Sci 2011;10:1593–1601 [DOI] [PubMed] [Google Scholar]
  • 147. Gupta CM, Haq W. Tuftsin-Bearing Liposomes as Antibiotic Carriers in Treatment of Macrophage Infections. Methods Enzymol 2005;391:291–304 [DOI] [PubMed] [Google Scholar]
  • 148. Zhang R, Wu F, Wu L, et al. Novel self-assembled micelles based on cholesterol-modified antimicrobial peptide (DP7) for safe and effective systemic administration in animal models of bacterial infection. Antimicrob Agents Chemother 2018;62:e00368-18 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 149. Iooss P, Le Ray AM, Grimandi G, Daculsi G, Merle C. A new injectable bone substitute combining poly(ɛ-caprolactone) microparticles with biphasic calcium phosphate granules. Biomaterials 2001;22:2785–2794 [DOI] [PubMed] [Google Scholar]
  • 150. Gavini E, Chetoni P, Cossu M, et al. PLGA microspheres for the ocular delivery of a peptide drug, vancomycin using emulsification/spray-drying as the preparation method: in vitro/in vivo studies. Eur J Pharm Biopharm 2004;57:207–212 [DOI] [PubMed] [Google Scholar]
  • 151. Piras AM, Maisetta G, Sandreschi S, et al. Chitosan nanoparticles loaded with the antimicrobial peptide temporin B exert a long-term antibacterial activity in vitro against clinical isolates of Staphylococcus epidermidis. Front Microbiol 2015;6:372. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 152. Almaaytah A, Mohammed GK, Abualhaijaa A, Al-Balas Q. Development of novel ultrashort antimicrobial peptide nanoparticles with potent antimicrobial and antibiofilm activities against multidrug-resistant bacteria. Drug Des Devel Ther 2017;11:3159–3170 [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153. d'Angelo I, Casciaro B, Miro A, et al. Overcoming barriers in Pseudomonas aeruginosa lung infections: engineered nanoparticles for local delivery of a cationic antimicrobial peptide. Colloids Surf B Biointerfaces 2015;135:717–725 [DOI] [PubMed] [Google Scholar]

Articles from Advances in Wound Care are provided here courtesy of Mary Ann Liebert, Inc.

RESOURCES