Skip to main content
NIHPA Author Manuscripts logoLink to NIHPA Author Manuscripts
. Author manuscript; available in PMC: 2021 Dec 20.
Published in final edited form as: Curr Pharm Des. 2015;21(16):2073–2088. doi: 10.2174/1381612821666150310102702

Antimicrobial Peptides and Peptidomimetics – Potent Therapeutic Allies for Staphylococcal Infections

Haroon Mohammad 1, Shankar Thangamani 1, Mohamed N Seleem 1,*
PMCID: PMC8686049  NIHMSID: NIHMS1762181  PMID: 25760338

Abstract

The pervasiveness of bacterial resistance to conventional antibiotics, particularly those associated with staphylococcal infections, has become a global epidemic. However, research involving antimicrobial peptides (AMPs) and their synthetic analogues has unearthed a potentially novel class of antibacterials for the treatment of an array of diseases caused by pathogenic bacteria, including staphylococci. AMPs have several unique advantages over traditional antibiotics including the projected slow emergence of bacterial resistance to these agents and their capability to modulate the host immune response to infection. Unfortunately, their susceptibility to proteolytic degradation, loss of antimicrobial activity due to serum binding or physiological concentration of salts, and toxicity to host tissues has limited their use as systemic agents thus far. Additionally, the presence of economic and regulatory obstacles have hindered the translation of AMPs, as antimicrobials, from the bench to the clinic. The present review delves further into the benefits and challenges of utilizing AMPs as antibacterial agents (particularly for staphylococcal infections), methods which have been utilized to overcome their limitations, their successes and failures in clinical trials, and future avenues for researchers to pursue to develop AMPs as novel therapeutic allies in the treatment of bacterial infections.

Keywords: Antimicrobial peptides, peptidomimetics, antibiotics, multidrug-resistance, staphylococci, MRSA, bacterial resistance, antibacterial, immunomodulatory agents

1. INTRODUCTION

Multidrug-resistant pathogens pose a significant global public health challenge and have been identified in every geographic region of the world (1). Annually, in the United States alone, these pathogens negatively impact the lives of over two million people at a cost of $20 billion to the healthcare system and result in over 23,000 deaths (2). Of these fatalities, nearly half are attributed to a single bacterial pathogen, methicillin-resistant Staphylococcus aureus. While once restricted to the healthcare setting (referred to as healthcare-associated MRSA or HA-MRSA), MRSA infections have become a major problem in the community (referred to as community-acquired MRSA or CA-MRSA) affecting a diverse population including healthcare workers, prison inmates, members of the military, athletes, the homeless population, intravenous drug users, newborn babies, and young children (312). Furthermore, CA-MRSA infections are typically associated with more severe morbidity and mortality than their HA-MRSA counterparts (13). While CA-MRSA is a leading cause of skin and soft-tissue infections, MRSA has also been associated with more complicated medical diseases including necrotizing pneumonia, osteomyelitis, and sepsis (1418).

For nearly 70 years, natural product antibiotics and their synthetic analogues have been the gold standard for treatment of infections caused by bacterial pathogens. However, the emergence of bacterial strains exhibiting resistance to numerous antibiotics has resulted in treatment failure. Indeed, clinical isolates of both CA-MRSA and HA-MRSA have been documented which exhibit resistance to nearly all antibiotic classes including the β-lactams, macrolides, quinolones, tetracyclines, and lincosamides (1923). Further exacerbating the problem, are strains have emerged that exhibit resistance to both first-line antibiotics and drugs deemed agents of last resort (such as linezolid and vancomycin) (2426). Though prudent use of effective antimicrobials is a critical step to alleviate complications and costs associated with MRSA infections, alternative strategies to combat this global challenge must be explored. One potential alternative for novel therapeutic agents to treat infections caused by multidrug-resistant pathogens that has shown significant promise in recent years is the isolation of natural and development of synthetic antimicrobial peptides (AMPs).

Since their initial discovery, AMPs have been isolated from bacteria (termed bacteriocins), marine animals, plants, birds, insects, frogs, and the human innate immune response (2730). AMPs traditionally consist of a short chain of 12 to 100 amino acids linked by peptide bonds (31). They typically are cationic (with at least two positive charges) which permits their interaction with negatively-charged components present in the bacterial membrane and cell wall; furthermore, AMPs are amphipathic which is thought to permit their ability to target and partition into bacterial cell membranes (30, 3234). In addition to their membrane-disrupting abilities, antibacterial peptides have also been shown to attack intracellular targets in bacteria including inhibiting macromolecular synthesis, inhibiting nucleic acid or protein synthesis, binding directly to nucleic acids, or interfering with cell wall synthesis (31, 3537).

In addition to possessing potent antibacterial activity, AMPs have several unique advantages over traditional antibiotics which led to the excitement in trying to isolate and develop synthetic analogues of peptides as novel therapeutic agents. Amongst these selective advantages include a low rate of bacterial resistance emerging, ability to neutralize virulence factors released by pathogens which can trigger a pro-inflammatory immune response in the host (leading to sepsis or toxic shock syndrome), and the proven ability of AMPs (or more specifically, host defense peptides) to stimulate or modulate the host immune response which consequently helps infected hosts to achieve more rapid healing and repair of compromised tissues (38). However while they possess several advantages over traditional antibiotics, AMPs also have several critical limitations that have hindered their ability to be translated into clinical use, particularly for treatment of invasive infections. Among the weaknesses identified thus far for AMPs as antimicrobials include toxicity to host tissues, rapid degradation by proteases, sensitivity to changes in salt concentration, and extensive binding to serum proteins (29, 31, 3942). Though much research has been undertaken to overcome these limitations, a critical analysis of these findings is needed to transform these promising novel antibacterials into therapeutic compounds capable of being used in the clinic, particularly for infections attributed to multidrug-resistant staphylococci.

An extensive review of published literature revealed more than twenty AMPs that possess activity against staphylococci (both in vitro and in vivo in an animal study) have been isolated/developed in the past two decades. The source (from where each peptide was derived), amino acid sequence, in vitro biological activity (denoted as the minimum inhibitory concentration (MIC) capable of inhibiting bacterial growth), and toxicity (to host tissues) of the most promising peptides are outlined in Table 1. The present review will delve into the potential use of AMPs as novel anti-staphylococcal agents by focusing on their unique modes of action against bacteria, advantages and limitations of AMPs as therapeutic agents, current AMPs in clinical trials for the treatment of staphylococcal infections, and future avenues, clinically, for the development of AMPs as anti-staphylococcal agents.

Table 1 -.

Antimicrobial peptides and peptidomimetics with biological activity against S. aureus.

Peptides Source Sequence MIC Toxicity References
Indolicidin Bovine neutrophil granules ILPWKWWPWRR 5–16 μg/ml LD50 of indolicidin in MT-2 cells is 33 μg/ml (4345)
Omiganan (MX-226) Synthetic cationic peptide derived from indolicidin ILRWPWWPWRRK 16–32 μg/ml 1% omiganan gel resulted in no adverse local or systemic effects in repeated-dose (14-day and 28-day in rat and rabbit models) (4650)
LL-37 Human neutrophil granules, epithelial cells (skin, lung, gut, mammary gland and epididymis) LLGDFFRKSKEKIGKEFKRIVQRIKDFLRNLVPRTES 1–8 μg/ml Low dose (100 μg /kg of body weight) did not induce observable toxicity but a high dose (3,000 μg/kg) resulted in adverse effects in rats (5153)
Novispirin G10 Variant of ovispirin-1 from sheep PKNLRRIIRKGIHIIKKYG 4–12 μg/ml 2.5% hemolysis of human erythrocytes at 80 μg/ml (54, 55)
Temporin A Frog (Rana ornativentris) skin FLPLASLFSRLL 5 μM Cytotoxic effect not observed in keratinocytes below 10 μM (56, 57)
Iseganan (IB-367) Derivative of protegrin, from porcine neutrophils RGGLCYCRGRFCVCVGR 4 μg/ml Intraperitoneal injection of IB-367 at a dose of 1mg/kg body weight caused no systemic toxicity in rats (5860)
Brilacidin (PMX-30063) Defensin-peptide mimetic N.A 1–2 μg/ml EC50 for RBCs is greater than 500μg/ml (61, 62) PolyMedix Inc.
Protegrin-1 (PG-1) Porcine leukocytes RGGRLCYCRRRFCVCVGR 1–2 μg/ml 42% hemolysis at 100μg/ml (63, 64)
HB1345 Synthetic lipohexapeptide Decanoyl- KFKWPW 0.5–1 μg/ml Not available Helix Biomedix
Pexiganan acetate (MSI-78)/ Magainin peptide Skin of the African clawed frog (Xenopus laevis) GIGKFLKK AKKFGKAF VKILKK 16–64 μg/ml 250 μg/ml are necessary to induce 100% hemolysis (65, 66)
Plectasin Defensin-like peptide from saprophytic ascomycete Pseudoplectania nigrella. GFGC1NGPWDEDDMQC2HNHC3KSIKGYKGGYCAKGGFVC2KC3Y 16–32 μg/ml Hemolytic dose EC50 for human erythrocytes is 400 μg/ml (67)
NZ2114 Derivative of plectasin N.A 2–4 μg/ml Less than 1% cytotoxicity observed in THP-1 cells at 128μg/ml (68, 69)
Fowlicidin-1 (6–26) An α-helical cathelicidin from chicken RVKRVWPLVIRTVIAGYNLYRAIKKK 1–2 μM Hemolytic dose EC50 for human erythrocytes is ~40 μM (70)
hLF1–11 Derivative of Lactoferrin (LF), an iron-binding glycoprotein present in various secretory fluids such as milk, saliva, tears, and mucosal secretions GRRRRSVQWCA 4–16 μM No toxicity observed in doses up to 10 mg/kg body weight.(at least 10,000 times higher than the dose proposed for treatment in humans) (7175)
Lytixar (LTX-109) Synthetic peptidomimetic N.A 2–4 μg/ml Toxicity not observed in topical application of 5% LTX-109 for 14 days in rats and mini-pigs (7678) Lytix Biopharma
RR Synthetic peptide WLRRIKAWLRR 8–32 μM Less than 10% hemolysis noticed at 300 μM and toxicity not observed in HeLa cells up to 64 μM (79)
Nisin Lantibiotic from Lactococcus lactis N.A 2–16 μg/ml 4.93% hemolysis of human erythrocytes was noticed at 2.5 μg/ml (8082)
RRIKA Synthetic peptide WLRRIKAWLRRIKA 2–4 μM Less than 10% hemolysis noticed at 300 μM and toxicity not observed in HeLa cells up to 32 μM (79)
MU1140 22-amino acid bacteriocin from Streptococcus mutans N.A 8–32 μg/ml Not available (83, 84)
Epinecidin-1 Peptide from fish (Epinephelus coioides) CFHIIKGLFHAGKMIHGLVTRRRHGV 6.25 μg/ml Epinecidin-1 at 2 μg/ml caused lysis of RAW264.7 cells (8587)
Peptide 4 Cyclic D,L-α-peptide KQRWLWLW 3 μg/ml Toxicity not observed in mice at the maximum tested dose of 12 mg/kg with intra-peritoneal injection (88)
Pardaxin (GE33) Red Sea flatfish (Pardachirus marmoratus) GFFALIPKIISSPLFKTLLSAVGSALSSSGGQE 6.25 μg/ml IC50 for HeLa cells was found to be 12.36 μg/ml (8991)
DASamP1 Synthetic peptide FFGKVLKLIRKIF 3.1 μM 50% red blood cells lysed at 25 μM (92)
Human β-defensin 3 (hBD3) Human keratinocytes, airway epithelial cells GIINTLQKYYSRVRGGR 4–8 μg/ml 50% of HepG2 and MDCK cells showed toxicity to hBD3 at 25–50 μg/ml (9397)
Lysostaphin Antibacterial enzyme from Staphylococcus simulans Zinc-containing metalloenzyme of 27 kD 0.002–0.032 μg/ml 50% of normal human epidermal keratinocyte cells (NHEK) are toxic at 1.6 g/L (98100)
HHC-10 Synthetic peptide KRWWKWIRW 1.5–3μM Very low hemolysis (less than 20%) of human erythrocytes was noticed at 188 μg/ml (101)
HHC-36 Synthetic peptide KRWWKWWRR 1.5–3μM Very low hemolysis (less than 20%) of human erythrocytes was noticed at 188 μg/ml (101)

2. MECHANISM OF ACTIONS OF AMPs

Attaining a proper understanding of the biological target upon which antibacterials exhibit their activity is critical to propel them into the latter stages of clinical development. With regards to AMPs, the primary mechanism of action involves targeted disruption of the bacterial cytoplasmic membrane (102). The positive charge present in certain residues within the AMP serves as a point of attraction towards anionic components present on the bacterial cell membrane (such as the phospholipid head-groups). This electrostatic interaction ultimately leads to perturbation of the bacterial cell membrane (102). Membrane disruption often occurs rapidly and is very non-specific. Four predominant models (the toroidal-pore model, the barrel-stave model, the aggregate model, and the carpet model) have been proposed to describe the events that occur as a result of interaction of AMPs with the bacterial membrane (103105). These models have been classified based upon the method the AMP employs to disrupt the membrane which can occur via (i) transient channel formation, (ii) disintegration of the integrity of the bacterial membrane, or (iii) transport through the membrane (102). Irrespective of the method utilized, eventually AMPs cause membrane depolarization and bacterial lysis. The presence of negatively-charged phospholipids in the bacterial membrane is an important factor for the selective action of AMPs upon bacteria over eukaryotic cells, where neutrally charged lipids are found to be predominant in mammalian cells (31).

Antimicrobial peptides are also capable of targeting important components that play a role in bacterial cell wall synthesis. Nisin, a bacteriocin isolated from Lactococcus lactis, binds to lipid II and prevents peptidoglycan formation, resulting in inhibition of cell wall synthesis (106); additionally, nisin also causes membrane disruption. Interestingly, vancomycin also targets the same biosynthetic pathway for cell wall synthesis as nisin; however, this peptide is thought to interact with a different moiety within lipid II, as compared to vancomycin. This is further supported by the fact that vancomycin-resistant strains of S. aureus (VRSA) are susceptible to nisin (81). Plectasin, a fungal defensin, also binds to lipid II and inhibits bacterial cell wall biosynthesis (107). Therefore, in addition to causing membrane depolarization, it is likely that AMPs attack more than one biological target in bacteria. This complexity in their mechanism of action makes it very difficult to isolate bacterial mutants exhibiting resistance to peptides, as will be explained further in the latter portions of this review (108).

In addition to membrane disruption and interfering with bacterial cell wall synthesis, well-established studies have identified that peptides are also capable of attacking internal targets present in the bacterial cytoplasm. These particular peptides, at the minimal effective concentration, do not permeabilize the bacterial membrane but instead, they translocate into the cells and moderate cell death by targeting critical processes inside the bacterial cell. For example, dermaseptin, an AMP isolated from frog skin, inhibits RNA and DNA synthesis without causing membrane depolarization (31). This mechanism of macromolecular synthesis disruption appears similar to that of the antibiotic rifampicin, a RNA polymerase inhibitor (109). Similarly, PR-39, an antimicrobial peptide identified from the pig intestine, and pleurocidin, isolated from fish, inhibit DNA replication and protein synthesis (35, 109). Human Neutrophil Peptide-1 (HNP-1) has also been shown to inhibit nucleic acid synthesis and protein synthesis (110). Indolicidin, derived from bovine neutrophils, blocks DNA synthesis in addition to causing cell membrane disruption (111, 112).

3. IMMUNOMODULATORY ACTIVITY OF AMPs

3.1. Cytokine regulation and immune cell chemotaxis

As described above, AMPs possess several unique methods to exhibit their antibacterial action on bacteria ultimately resulting in the resolution of an infection. However, an alternative mechanism by which AMPs play a role in the clearance of a bacterial infection involves a direct modulation of the host immune response against an invading pathogen. Human defensin peptides form an important component for the immune response against infections, which is supported by the fact that mice lacking the AMP cathelicidin are found to be more susceptible to bacterial infection (113). The ability to modulate host immune responses, either by inhibition or stimulation, varies significantly based on numerous factors; however, this has proven to be a very useful strategy for consideration for the prevention and treatment of bacterial infections. For example, innate defense regulator peptide-1 (IDR-1) has no innate antimicrobial activity in vitro, but it has been shown to protect mice from antibiotic-resistant S. aureus infection (114). IDR-1 selectively suppresses pro-inflammatory cytokines, such as TNF-α and IL-6 at the site of infection, and IDR-1 also increases the production of the anti-inflammatory cytokine IL-10 in the blood (114). Additionally, IDR-1 stimulates production of different chemokines such as RANTES and MCP-1 which recruit both monocytes and macrophages to the site of infection (114). RANTES and MCP-1 play an important role in protection against infection (114116). IDR-1 also protects monocytes from the cytotoxic action of S. aureus (114). Given the excellent immunomodulatory properties of IDR-1, this agent has strong potential for use as a therapeutic agent to clear S. aureus infections. In addition to IDR-1, the peptide hLF1–11, a lactoferrin derivative, exhibits immunomodulatory activity in its ability to bind to human monocytes and subsequently inhibit generation of the myeloperoxidase (MPO) enzyme, a negative regulator of IL-10 (117). hLF1–11 also enhances granulocyte-macrophage colony-stimulating factor (GM-CSF)-derived monocyte differentiation to macrophages which exhibit increased antimicrobial activity against S. aureus (118).

Cathelicidins derived from humans (LL-37), cows (indolicidin), and pigs (PR-39) have been shown to facilitate the recruitment of various immune cells such as neutrophils, monocytes, T cells, and mast cells to the site of infection (119121). LL-37 induces various chemokines such as MCP-1, RANTES, and IL-8 to promote chemotaxis of immune cells (122124). Cathelicidins from humans and cows also have been shown to be capable of suppressing pro-inflammatory cytokine production (including cytokines such as IL-1β and TNF-α) while also increasing anti-inflammatory cytokine production (including IL-10) (119, 120, 125). IB-367 from porcine neutrophils increases the migration of CD11b and Gr-1 positive cells to the site of infection and enhances bacterial clearance (59). Thus, the overall net result of the immunomodulatory functions of peptides and AMPs leads to the control of inflammatory responses in the affected host without altering or compromising the immune responses that are essential for the clearance of the bacterial infection. This dual role of AMPs as antibacterial and immunomodulatory agents has led to their exploration as alternative therapeutics for the treatment of multi-drug resistant S. aureus infections.

3.2. Wound healing and angiogenesis

The utilization of antimicrobial peptides as topical agents to treat both acute and chronic staphylococcal infections (including LTX-109 and Brilacidin (PMX-30063)) has been steadily gaining momentum as can be seen by the increasing number of AMPs that are entering into clinical trials. AMPs can play a direct role in wound healing and angiogenesis, two desirable physiological effects for the resolution of staphylococcal skin infections. A classic example of this are the human defensin peptides (hBDP1–4) which have been shown to enhance chemotaxis of immune cells to wounded tissues and increase keratinocyte migration and proliferation, thereby stimulating tissue regeneration (126, 127). This is further supported by the fact that the lack of hBDs leads to delayed tissue regeneration and impaired re-epithelialization in chronic wounds (128, 129).

Chronic wound healing in diabetic foot ulcer patients is a challenging medical issue, particularly wounds infected with bacteria. S. aureus-infected diabetic foot ulcers are common and hard to treat; proper healing of such infected wounds is often complicated by impaired functioning of immune cells (130136). As stated earlier, hBDPs are known to accelerate healing of wounds including those present in patients with diabetic foot ulcers. For example, one study demonstrated that human β-defensin 3 (hBD3) enhances tissue regeneration and closure of an infected wound in a diabetic porcine model (137). In addition to this, considerably higher hBD3 expression was found in human primary keratinocytes treated with heat inactivated S. aureus, which supports the fact that hBD3 may aid in healing of infected skin wounds (93). Hence, considering their anti-inflammatory, wound healing, and angiogeneic properties, AMPs may have potential for further exploration for the treatment of S. aureus infected chronic wounds.

4. ADVANTAGES OF USING PEPTIDES FOR THE TREATMENT OF BACTERIAL INFECTIONS

In addition to their capability to modulate the host immune response, antimicrobial peptides possess several unique advantages over empirical antibiotics used for the treatment of multidrug-resistant staphylococcal infections. These advantages include (1) potent bactericidal activity and (2) low frequency of bacterial resistance emerging. Combined with AMPs’ anti-inflammatory properties, these unique assets permit AMPs to be utilized in multiple unique therapeutic applications as will be discussed in more detail below and in latter portions of this review.

4.1. Potent antibacterial and rapid bactericidal activity against Gram-positive pathogens, including S. aureus

Numerous peptides have shown potent in vitro antimicrobial activity against important Gram-positive pathogens, including MRSA. Indeed, many published reports have indicated that these AMPs have activity that is similar to or better than conventional antibiotics including vancomycin (28). Nisin has been used as a food preservative for more than 50 years with limited development of bacterial resistance to this agent (31). This same AMP has been shown to inhibit the growth of Gram-positive bacteria at concentrations in the nanomolar range (31). In addition to nisin, AMPs derived from the horseshoe crab (including polyphemusin) have been found to exhibit excellent antibacterial activity, inhibiting bacterial growth at concentrations below 2 μg/mL (138). Maganins, isolated from the skin of frogs and other amphibians, have also shown powerful antimicrobial activity against Gram-positive bacteria. They were the template for the development of the first clinical AMPs but were ultimately found to be unsuccessful for use as therapeutic agents (31). Cathelicidins, isolated from the immune system of many mammals (such as BMAP-28, a bovine AMP), have also been shown to possess excellent antibacterial activity against Gram-positive pathogens (139).

When peptides were examined for their rate of bactericidal activity, many were found to be capable of complete elimination of an initial bacterial inoculum within a few hours. Pexiganan (a synthetic derivative of magainin currently in clinical trials), at a concentration of 256 μg/mL, was shown in one study to completely eliminate S. aureus ATCC 29213 (starting inoculum of 106 CFU/mL) within two hours (140). Importantly, this peptide retained its bactericidal activity against isolates of S. aureus exhibiting resistance to ofloxacin, oxacillin, cefazolin, and imipenem, an additional benefit that has been noted with AMPs. Furthermore the porcine-derived peptide, protegrin-1 (PG-1), at concentrations equivalent to 1 × or 2.5 × MIC, was found to be capable of producing a 3 to 5-log10 reduction in MRSA (both in stationary and log phases of growth) growth, within 8 minutes of exposure, via a standard time-kill assay (63).

4.2. Low frequency of bacterial resistance emerging

As noted by Maria Papagianni in her thorough review of ribosomally-synthesized AMPs, peptide-based antibiotics are thought to be a potential solution to the burgeoning issue of bacterial resistance that has emerged to traditional antibiotics (34). Bacterial resistance to AMPs is unlikely to occur rapidly (as compared to conventional antibiotics), in part, due to the mode of action of these peptides. Given that peptides act on critical structural features present on the surface of bacterial cells (namely the cell membrane), in addition to attacking multiple intracellular targets, this complex multi-target attack on bacteria supports the notion that resistance to AMPs will be slow to develop (31). This postulate has been supported by recent publications examining the propensity of microbial resistance to emerge to different AMPs after serial passage of these compounds at subinhibitory concentrations, as discussed below.

Ge et al noted that when an isolate of S. aureus was exposed to a subinhibitory concentration of pexiganan over eight passages, only a slight increase (from 5.8 μg/mL to 8.3 μg/mL) in the MIC was found (140). Interestingly, no increase in the MIC was observed when the same experiment was repeated for coagulase-negative staphylococci, confirming that bacterial resistance to this particular peptide is slow to develop. A second published report by Steinberg et al discovered that when the peptide PG-1 (at ½ × MIC) was repeatedly exposed to the same strain of MRSA (for 18 passages), no resistance was observed (63). In contrast, when the same analysis was completed with the antibiotic norfloxacin, an 85-fold increase in MIC was observed. Moreover, cross-resistance between peptides and antibiotics (or peptides with other peptides) even after exposure of bacteria to multiple passages of AMPs has not been observed in multiple studies (108, 141). This is in contrast to traditional antibiotics where cross-resistance between multiple antibiotics has been observed in a single drug-resistant bacterial strain; a classic example of this is bacterial production of extended spectrum β-lactamases which confers cross-resistance to multiple antibiotic classes including penicillins and cephalosporins (142, 143). It is also worthy to note here that the peptide nisin, as mentioned earlier, has been used commercially for many years as a food preservative with limited reports of bacterial resistance to this agent being published. These results collectively support the proposition that bacterial resistance, in particular by MRSA, to AMPs would be expected to emerge much slower than what has been observed with traditional antibiotics. Additionally, should resistance to a particular AMP be noted, simple modifications in the structural residues can be utilized to overcome this issue, rendering the AMP still effective. Furthermore, should resistance develop to the antibacterial component of AMPs (via structural modifications to the biological cell membrane, activation of efflux pumps, or through bacterial proteolytic degradation of AMPs), it doesn’t affect their potential to be used alternatively as immunomodulatory agents (to stimulate the host immune response to eliminate these pathogens).

4.3. Anti-inflammatory activity of AMPs

In addition to the potent antibacterial activities reported for many natural and synthetic peptides, several peptides have a dual advantage as antibacterial and anti-inflammatory agents. Of these published reports, Zhang et al demonstrated that synthetic peptides (such as HB43, HB55, and HBPM4) screened for activity against pathogens (including S. aureus) which commonly infect cystic fibrosis patients, possessed excellent antibacterial activity against MRSA (MIC of the three best peptides was 4 μg/mL) (108). Inflammation is a major problem in patients afflicted with cystic fibrosis and is thought to contribute significantly to lung tissue destruction. Interestingly, the authors also discovered that one of their peptides (HBCM2) produced a significant reduction in inflammation in vivo in a murine ear edema model. Furthermore, another study found that the peptide HBPM4 was capable of neutralizing endotoxin (108), which can lead to severe clinical diseases including sepsis or septic shock. One drawback of conventional antibiotics is they can actually stimulate the release of endotoxins by bacteria (144, 145); thus peptides possessing both anti-inflammatory activity and the capability to inhibit toxin production possess a unique advantage over traditional antibiotics.

5. CURRENT LIMITATIONS OF USING PEPTIDES TO TREAT INVASIVE MRSA INFECTIONS

Though AMPs possess multiple desirable properties as therapeutic agents, their ability to be used systemically has been hindered due to a number of different physical limitations. This is problematic, as it pertains to multidrug-resistant staphylococci, because invasive MRSA infections still pose a significant challenge to the healthcare system. Currently available antibiotics fail to treat these systemic infections due to emerging resistance by MRSA to these agents, poor pharmacokinetic profile of many antibiotics (including agents of last resort like vancomycin), and inability to cross cellular membranes of host cells to reach intracellular niches where MRSA colonize and prolong infection (such as macrophages). While there was once great hope that AMPs would be able to overcome the challenges faced by traditional antibiotics, these peptides have significant limitations as well. Peptides cannot be administered systemically for the treatment of invasive MRSA infections for several major reasons including (1) toxicity to host tissues, (2) lack of oral bioavailability, (3) loss of antibacterial activity in the presence of physiological concentration of salts and divalent cations, (4) loss of antimicrobial activity due to serum binding, (5) rapid degradation of peptides by proteases, and (6) inability of peptides to kill MRSA present inside host cells. A more detailed explanation of these drawbacks will be presented in the following paragraphs.

5.1. Toxicity to host tissues

A significant challenge facing researchers trying to develop peptides as novel antibacterials is the fact that many AMPs are toxic to mammalian cells (29). Indeed toxicity to the liver, kidneys, and eyes (for ophthalmic agents) has been reported for many AMPs (40). Additionally, a high degree of toxicity has been observed in mammals when peptides are injected directly into the circulatory system. Furthermore, given the mode of action of several AMPs (including melittin from bees) as nonspecific membrane-disrupting agents, they can attack the integrity of host cells including red blood cells, leading ultimately to hemolysis (31, 40).

The toxicity associated with natural AMPs which are part of the innate immune response/host defense system are often controlled by various host processes including sequestration of peptides in granules in immune cells, generation of nontoxic propeptides that require enzymatic cleavage to achieve the biologically active peptide, and programmed deactivating mechanisms to limit damage to host tissues (40, 146). However AMPs as therapeutic agents are often given as is (at concentrations higher than those found in the human body); given that many AMPs have a low therapeutic index, this creates problems with toxicity when higher doses of peptides are administered. Thus one appropriate mechanism to address this issue would be to develop vehicles to transport and release AMPs at the site of infection to minimize toxic side effects to healthy tissues. However, limited success has been observed in this area as it pertains to cationic AMPs (147).

5.2. Limited bioavailability of peptides administered orally or intravenously

Some AMPs (such as mersacidin) are not soluble in water resulting in limited bioavailability of the AMP in host tissues when administered orally or intravenously (28). Though degradation of peptides by proteases or serum binding are one explanation (as discussed in more detail below), another explanation is that AMPs cannot cross membrane barriers present in host tissues (such as the intestinal tract) to reach the bloodstream and ultimately reach the site of infection. This presents challenges in terms of drug delivery and penetration across biological membranes. A publication assessing the effect of the antibacterial peptide PG-1 found the AMP exhibited potent activity against MRSA ATCC 33591 in vitro (MIC of 2 μg/mL). However, when mice were subjected to an intravenous dose (4 mg/kg) of PG-1, the peptide concentration diminished rapidly and was not detectable in the plasma after two hours (63). The authors postulated that this most likely was due to limited tissue penetration of PG-1. This hypothesis was confirmed when pharmacokinetic experiments with PG-1 were performed via an intramuscular injection (8 mg/kg) of the peptide in mice; no PG-1 was able to be detected in the plasma even ten minutes after the injection was administered. Thus the peptide was not able to diffuse through the muscular tissue and enter the bloodstream. The researchers concluded that bacteria that concentrated in host tissues following intravenous dosing were most likely not exposed to an effective concentrations of PG-1 to kill the bacteria. One solution to resolve the issue of aqueous solubility of peptides is to incorporate a salt (such as an alkali metal like potassium) into the peptide structure; however, this may compromise the antimicrobial activity of the peptide. Another potential solution would be to design a vehicle that would permit the peptide to be delivered directly to the site of infection but to date no such success has been reported.

5.3. Loss of antibacterial activity due to serum binding

A third limitation of AMPs for systemic applications is, given these molecules are peptides, they tend to bind strongly to proteins in the serum. Human serum albumin is the predominant transport protein present in blood plasma; a recent study demonstrated that the hydrophobic residues present in AMPs have a high affinity (in μM concentration) for binding to drug site II in human serum albumin (42). The binding affinity of peptides to serum proteins reduces the distribution and concentration of the active drug that can reach the site of infection. Drugs that bind strongly to serum proteins (>95% bound) present several issues including requiring a higher concentrations to achieve a therapeutic effect in vivo, slow distribution to the target site of infection and issues with elimination of the drug from the host (148, 149). Though increasing the size/concentration of the dose of an AMP given or the frequency of administration would appear a simple solution to this conundrum, this can lead to severe toxicity in host tissues. It has been postulated that the cationic charge of peptides contributes to their binding to tissues and proteins and their overall stability; thus studies have been undertaken to explore modified peptides where these positively-charged residues were removed. However, this resulted in complete loss of activity, indicating the necessity of these residues to achieve an antibacterial effect (150).

5.4. Loss of antibacterial activity in the presence of physiological concentrations of salts and divalent ions

Additionally, the presence of high concentrations of salts (100 mM NaCl) or monovalent and divalent cations (1–2 mM) in the body (such as in serum/plasma) can interfere with cationic antimicrobial peptides’ ability to interact and kill pathogenic bacteria (27, 41, 151). For example, the activity of defensins, weakly antibacterial peptides derived from mammals, have been shown to be negatively affected by increasing salt concentrations (31). Similar results have been reported for other peptides including clavanins, histatins such as P-113, gramicidins, and magainins (33, 150, 152). Several published reports have also mentioned that peptides tested in standard nutrient-rich growth medium were capable of inhibiting bacterial growth at very low concentrations; however when the peptides were tested in solutions/buffers containing increasing concentration of salts or divalent cations (such as 50–100 mM NaCl or MgCl2 to mimic conditions present inside the human body), the antibacterial activities of these peptides were significantly reduced or completely abolished (95). This is problematic for assessing the true potential of AMPs against bacteria given that different culture media (depending on the presence or absence of salts) can produce confounding results (95). Given the presence of such concentrations of salts in the human serum and different regions of the human body where infections can persist, it points to a problem with using AMPs systemically; namely that their antimicrobial activity will most likely be lost due to serum binding.

5.5. Degradation by host and bacterial proteases and bacterial adaptation to minimize peptide effect

Another major limitation of AMPs as systemic agents is the structural integrity of these peptides can be compromised as they are often rapidly degraded by host proteases (particularly those present in the GI tract) (31). In addition to the proteases present in the GI tract, AMPs can be degraded by peptidases present in the blood, as has been shown for other protein drugs (153). This has a profound effect on the pharmacokinetic profile of these peptides, particularly minimizing the quantity of drug available to be absorbed into the bloodstream (once again limiting the use of peptides for systemic applications). Antimicrobial peptides contain basic amino acids in their core structure which makes them susceptible to proteases such as the chymotrypsin-like enzymes (41). Several researchers have tried to address the issue of proteolysis of peptides by introducing covalent modifications to the bonds between the amino acids, insertion or substitution of unusual amino acids (such as D-amino acids which protects peptides from proteolytic degradation), altering the secondary structure of the AMPs, utilizing non-peptide backbones (peptidomimetics), and the creation of prodrug molecules (154157). However, this has produced mixed results with regards to an increase, decrease, or no change observed in the antimicrobial activity of these AMPs against MRSA and other strains of staphylococci (154, 155, 157). Additionally, the introduction of these modifications, in particular the D-amino acids, comes at a price as these peptides cost more to develop (158). Furthermore, no information is presented regarding their potential toxicity to host tissues, stability in physiological salt concentrations, serum binding profile, or impact of the modifications on the overall pharmacokinetic profile of these peptides. Thus limited information is available to assess the true potential of these modified AMPs for use systemically.

In addition to rapid degradation by host proteases, the antibacterial effect of AMPs can be suppressed by bacteria that secrete effector molecules that degrade peptides, prevent peptides from interacting with the bacterial cell membrane (blocking their ability to target and enter into the cell), or suppress the antibacterial effect of peptides altogether. An example of this is the staphylokinase protein secreted by S. aureus which is capable of binding directly to, and neutralizing, α-defensins secreted by human neutrophils (159). In addition to staphloykinase, S. aureus can secrete proteases, including aureolysin, which has been shown to directly interact and breakdown the peptide LL-37 (160). Additionally, given that the cationic moieties of peptides (particularly of defensins, cathelicidins, and their synthetic analogues) play an integral role in interacting with the negatively-charged regions present on the surface of bacterial cells, this electrostatic interaction can be reduced or revered by alterations made to surface components on bacterial cells. For example S. aureus can incorporate an l-lysine modification within phosphatidylglycerol or a d-alanine modification to the teichoic acid component of cell walls, reducing the negative charge present in the cell wall (161, 162). These modifications will reduce the affinity and adsorption of AMPs to these bacterial cell membranes. Furthermore, a limited number of S. aureus strains have been identified which contain a plasmid (pSK1) encoding a gene (qacA) for an efflux pump (163). Though this efflux pump appears to have limited effect on the removal of most natural AMPs present in the human body (such as α-defensin), its identification does raise concern that efflux pumps may be a potential challenge that future synthetic AMPs may have to overcome. Most research, to date, on bacterial adaptation to AMPs has focused on natural peptides derived from humans and animals. More work needs to be employed to understand potential mechanisms of bacterial adaptation to synthetic analogues of these AMPs.

Recent reports have presented several solutions to enhance the stability of antimicrobial peptides to the effect of proteases, limit serum binding, and mitigate the loss of antimicrobial activity in the presence of physiological concentration of salts. Given the simplicity and structural versatility of AMPs, it was thought that simple modifications to the basic structure of the peptide could mitigate stability and pharmacokinetic issues identified with many AMPs. Methods employed included the use of hydrophobic oligopeptide end tags and substitution of tryptophan- and histidine-rich peptides with the amino acid β-naphthylalanine (152, 164). While these techniques provided valuable insight into improvements to enhance the stability and activity of antimicrobial peptides, they did not address or effectively resolve all of the limitations described above.

5.6. Inability to penetrate intracellular compartments to kill S. aureus

S. aureus has been shown to be capable of escaping the host immune response to infection by modulating its gene expression in a manner that permits it to reside inside phagocytic cells, including macrophages (165). This action protects S. aureus from the effect of many antibiotics (incapable of gaining entry into these intracellular niches) and can lead to recurring infections in affected patients, including those suffering from osteomyelitis. Though peptides are smaller in size relative to proteins (which should permit their rapid diffusion across biological membranes), their amphipathic nature creates problems in crossing biological membranes, including those present across host cells and in the intestinal tract (153). This observation has been confirmed in several studies including a report published by Brinch et al with the peptide NZ2114. While the peptide possessed excellent activity against MRSA in vitro (MIC was 4 mg/L), similar to daptomycin and vancomycin, the peptide proved unable to effectively clear MRSA present inside infected THP-1 monocytes (69). Though the authors did not record the actual drug concentration present inside the monocytes, the inability of NZ2114 to effectively enter and survive in the infected monocytes is a plausible explanation. A more recent study utilizing short β-sheet folding synthetic peptides tried to address this issue of intracellular penetration. One of the engineered peptides, Iκ8-all D possessed excellent in vitro activity against S. aureus (MIC of 3.9 mg/L), demonstrated limited hemolytic activity (less than 10% even at a concentration of 1500 mg/L), was not degraded in the presence of proteases, and produced a 5–6 log10 reduction in S. aureus CFU/mL in an infected murine macrophage cell line (166). However none of the assays performed were under true physiological conditions or were confirmed in an animal model. Thus these results, while promising, must be translated into animal studies to confirm these synthetic peptides are in fact capable of eliminating S. aureus present in intracellular compartments.

5.7. Additional challenges

Additional challenges pharmaceutical companies and academic research groups will need to overcome with AMPs to translate their in vitro potential to actual clinical applications (particular systemically) include addressing issues pertaining to the sensitivity of AMPs to temperature, the economic feasibility to manufacture AMPs at a large-scale, and selecting appropriate packaging material and storage conditions to enhance the shelf-life and stability of the AMPs. As an example of the sensitivity of AMPs to temperature and storage conditions, one study assessing the efficacy of synthetic AMPs for eradication of pathogens causing an ocular infection noted that the AMPs developed were sensitive both to the storage medium and temperature. When the storage temperature was decreased from 23°C to 4°C, the AMP lost complete antimicrobial activity against S. aureus (146). This presents a significant problem with regards to maintaining appropriate storage conditions to ensure AMPs do not lose antimicrobial activity prior to administration for treatment.

With regards to the economic feasibility of manufacturing AMPs, they are expensive to manufacture in bulk and there are considerable challenges for small companies and university researchers to develop large enough quantities to test for further applications (29). A single gram of peptide (sufficient for an average daily dose orally or via intravenous injection) may cost upwards of $600 to manufacture (41). This significantly hinders researchers in their pursuit of developing peptides as novel anti-infective agents. Furthermore, several commercial antibiotics, such as vancomycin and erythromycin, still retain more potent activity than most AMPs (MIC values of AMPs are 15–25 fold higher than the antibiotics) in vitro and in in vivo models (more animals survive after treatment) (167). This points to the argument that conventional antibiotics are still a better option, currently, for the treatment of invasive infections compared to antimicrobial peptides. Collectively these points raise significant concerns and challenges to utilizing AMPs to treat systemic infections, particularly those caused by multidrug-resistant staphylococci such as MRSA.

6. PEPTIDOMIMETICS – ALTERNATIVES TO AMPs FOR TREATMENT OF S. aureus INFECTIONS

The limitations observed with AMPs spurred the development of synthetic mimics of these peptides, the so-called peptidomimetics. Peptidomimetics resemble a peptide structure with the exception that the backbone is not limited to the presence of α-amino acids (158). This non-traditional backbone has a selective advantage over natural peptides in that the unique structure of peptidomimetics is protected from the effect of proteases. An additional benefit, peptidomimetics are less expensive alternatives to manufacture over peptides (41). The mechanism of action of peptidomimetics mirrors that of peptides in that both often target the cell membranes of bacteria though alternative modes of action for specific compounds have been proposed. Different methods have been employed to develop peptidomimetics with antimicrobial activity; one method which led to the development of one of the first peptidomimetics to reach clinical trials utilized Protein Epitope Mimetic (PEM) technology. This technology led to the development of iseganan (IntraBiotics Phamaceutical, Inc.), a synthetic analogue of the AMP protegrin I. It was indicated for use in treating oral mucositis but the peptide failed in clinical trials. It was also tested in a phase III trial as a prophylactic agent to prevent ventilator-associated pneumonia; however the trial was stopped after a higher rate of ventilator-associated pneumonia and death were observed in patients after treatment with iseganan as compared to the control (158). This finding tempered initial expectations that peptidomimetics could overcome the limitations associated with clinical failure of AMPs.

A second technique to develop peptidomimetic agents that has shown great promise is in silico design of synthetic mimics of antimicrobial peptides (SMAMPS) which utilize economical synthetic oligomers (158). This effort has led to the development of antimicrobial compounds (termed defensin-mimetics) exhibiting improved toxicity, proteolytic stability, and pharmacokinetic properties relative to AMPs. One of these compounds, brilacidin (PMX-30063), developed by PolyMedix, Inc. (acquired by Cellceutix Corp. in 2013) demonstrated efficacy, even better than vancomycin, against MRSA infections in animal models. Based upon the preliminary studies conducted, brilacidin was proposed to be used as an injectable formulation for several infections caused by S. aureus, including the treatment of skin and soft tissue infections. Recently, a phase II clinical trial was completed; this trial demonstrated a high clinical response rate (as compared to daptomycin) in patients treated for acute bacterial skin and skin structure infections caused by MRSA (168). Furthermore, limited toxicity to patients undergoing treatment was reported from this study. These results, combined with previous studies indicating the drug has a low frequency of bacterial resistance, demonstrate the great promise peptidomimetics have as novel, safe alternative antibacterials which can be developed in the near future.

7. ANTIMICROBIAL PEPTIDES AND PEPTIDOMIMETICS IN CLINICAL TRIALS

The development of peptide-based therapies is gaining momentum, evidenced by the increased number of US Food and Drug Administration (FDA)-approved peptide-based therapeutics, which currently exceeds 100 different drugs. Although there are a few antimicrobial peptides (AMPs) and peptidomimetics that have reached various stages of clinical trials for the treatment and prevention of S. aureus infections, none have received regulatory approval to date (Table 2). Among the clinically-tested peptides, pexiganan showed the most promising results for the treatment of mildly infected diabetic foot ulcers (DFU) in two clinical trials (NCT00563433 and NCT00563394). Pexiganan proved as effective as oral ofloxacin for the treatment of mildly infected DFU though with fewer side effects and reduced likelihood of bacterial resistance developing (169). After several years and several successful clinical trials, pexiganan did not receive FDA approval. However, topical pexiganan cream 0.8% is currently being tested again in two Phase III clinical trials for the treatment of mild infections of DFU (NCT01590758 and NCT01594762).

Table 2 -.

Antimicrobial peptides against S. aureus in clinical trials.

Peptide name Sequence/description Indication Phase Clinical trial identifiers Ref
Pexiganan (MSI-78) GIGKFLKKAKKFGKAFVKILKK Infected Diabetic Ulcers III NCT00563433
NCT00563394
NCT01590758
NCT01594762
(169)
Iseganan (IB-367) RGGLCYCRGRFCVCVGR Ventilator-associated Pneumonia III NCT00118781 (172)
Omiganan (MBI 226) ILRWPWWPWRRK Topical Skin Antisepsis and Preventing Local Catheter Site Infections and III NCT00608959
NCT00231153
(173)
Lytixar (LTX-109) peptidomimetic Arg-Tbt-Arg-NH-EtPh (arginine-tertbutyl tryptophan-arginine-phenylethan) Gram-positive Skin Infection and MRSA nasal colonization II NCT01223222
NCT01158235
NCT01803035
(77)
Brilacidin (PMX-30063) peptidomimetic defensin-mimetic S. aureus skin infection II NCT01211470 (174)

7.1. The demand for topical treatments for infected DFU

The damaging role of S. aureus in DFU is well recognized since it is the most frequently isolated microorganism in infected DFU patients. Additionally, infections caused by multidrug-resistant S. aureus strains are associated with the worst patient outcomes and result in more frequent amputation of limbs (130135). The increasing prevalence of infected DFU and the increasing resistance among associated S. aureus isolates highlights the dearth of adequate antimicrobials to treat DFU infections. Therefore, there is an urgent need to develop new topical therapies capable of treating and promoting the healing of infected DFU. The estimated healthcare-associated costs of $15 billion annually for treatment of chronic wounds (170) such as DFU should create an economic incentive for pharmaceutical companies to invest in and develop AMPs capable of filling this unmet medical need for new antimicrobials. Additionally, independent of their antimicrobial effect, the recognized anti-inflammatory, immunomodulatory, and antibiofilm activity of certain AMPs should be a major advantage in the topical treatment of S. aureus skin infections in general and of infected non-healing DFU, in particular.

7.2. Future of clinical applications of AMPs

The major impediments to the translation of AMPs to the clinic thus far appear to center heavily on the stringent clinical trial designs required by regulatory agencies. Additionally the lengthy waiting process which follows the completion of trials followed by failure to receive FDA approval has slowed the progression of AMP development into the healthcare setting. This issue is further compounded by the fact that the recent FDA “reboot” pledge (171) to encourage the development of new antibiotics is unlikely to entice pharmaceutical companies to invest in this long and risky process of development of new antimicrobial drugs, including AMPs.

It is difficult to predict the possibility of AMPs receiving approval for use against S. aureus-related infections in the next few years. However, the low number of AMPs that are currently in clinical trials (Table 2) would suggest that the likelihood of an AMP receiving approval for use against S. aureus infections will not occur in the immediate future. Though AMPs as antibacterial agents may not make it to the clinical setting soon, work must continue to address their limitations and identify new avenues to pursue to propel their development as novel therapeutic agents.

8. POTENTIAL FUTURE APPLICATIONS FOR PEPTIDES IN TREATMENT OF S. aureus INFECTIONS

Though limited success has been achieved thus far in the development of AMPs for clinical use as antibacterials, they still hold great promise for use as therapeutic agents to treat staphylococcal infections. Their unique advantages over natural product antibiotics combined with a better understanding of their limitations has permitted a more focused strategy to be initiated to identify future applications for AMPs currently in development. Among their potential applications include (1) as topical ointments to treat skin infections and enhance wound healing, (2) use in combination with conventional antibiotics or peptides to produce a synergistic effect against S. aureus, (3) as immunomodulatory agents to clear bacterial infections, and (4) as decolonizing agents in both the healthcare and community settings. We will briefly touch upon the use of AMPs to address these strategies against MRSA in the subsequent paragraphs.

8.1. Topical ointments to treat skin infections

S. aureus is the most common bacterial pathogen linked to patients with skin and soft-tissue infections (SSTIs) (175). More than half (58%) of all SSTIs treated in the United States alone were caused by S. aureus per one epidemiological study of one national health care system (176). The Infectious Diseases Society of America recommends treatment of moderate to severe skin infections caused by MRSA to involve the combination of incision and drainage of the affected region with administration of empirical antibiotics (including mupirocin) (177, 178). However, highly-resistant strains of MRSA, including to mupirocin have been documented presenting significant concern that such therapies may be rendered ineffective in the future (22, 179, 180).

Antimicrobial peptides have the potential to be strong allies in the battle against MRSA skin infections given their potent anti-MRSA activity and low frequency of bacterial resistance emerging. Additionally, AMPs have been shown to possess anti-inflammatory properties and to enhance wound healing, both qualities which are critical for treatment of patients affected by a skin infection. Development of AMPs for topical applications has several appealing qualities including (1) direct delivery to the site of infection (thus avoiding the necessity of a vehicle or carrier as with systemic applications), (2) high local tissue concentration of the drug to be achieved through aggressive dosing combined with AMPs rapid bactericidal activity (permitting rapid clearance of an infection), (3) using a subinhibitory concentration of AMPs in combination with traditional antibiotics to achieve a synergistic effect (limiting potential irritation and toxicity to the skin), and (4) avoid issues pertaining to serum binding and proteolysis which can render AMPs ineffective (particularly when used systemically). Furthermore, toxicity to host issues (which is a significant problem for AMPs developed for systemic applications) can be mitigated by altering the dosage, frequency of administration, and formulation of the final product.

The success of AMPs developed for topical applications which have reached human clinical trials supports the proposition of developing AMPs for the treatment of MRSA skin infections. Among this list includes AMPs such as pexiganan, MBI 594AN, and D2A21. Pexiganan is a synthetic analogue of maganin which showed broad-spectrum activity against over 3000 clinical isolates of bacteria in vitro (including MRSA and VRSA) and, as stated earlier, achieved the same cure rate (85–90%) and wound healing as the control antibiotic ofloxacin in the treatment of patients with diabetic foot ulcers in a Phase III clinical trial (40, 181). MBI 594AN, an indolicidin-like peptide was developed as a topical ointment for the treatment of acne (primarily caused by the bacterium Propionibacterium acnes); in a Phase IIb clinical trial, this ointment produced a statistically significant reduction in the size of inflammatory acne lesions on the skin when compared to the control vehicle (40). D2A21 is another peptide under development as a topical antibiotic that has shown potent activity against MRSA in vitro (MICs between 0.25 – 4 μg/mL) (182); when tested in a rat model (against Pseudomonas aeruginosa), D2A21 demonstrated potential for use in the treatment of both skin infections (183) and burn wounds (184). Though none of these AMPs have reached the market yet, their success in in vivo animal models and human clinical trials, lends credence to the potential for AMPs to be used in the treatment of MRSA skin infections.

The rapid and potent antibacterial activity of peptides also has potential to limit the severity of skin infections by preventing bacterial migration into systemic circulation leading to invasive MRSA infections. Invasive MRSA infections are of particular concern as they can lead to pneumonia, sepsis, and eventually death (185). Thus development of AMPs as topical ointments to treat MRSA skin infections is a viable therapeutic strategy that warrants further investigation for researchers.

8.2. Use as decolonizing agents

A second potential application for AMPs against MRSA is using them as decolonizing agents. MRSA has shown the ability to colonize body surfaces including the skin, nares, and mucosal membranes (186, 187). Additionally, MRSA is capable of colonizing medical devices, in particular catheters, which can have serious consequences to many patients including those undergoing surgery, requiring dialysis, and patients suffering from atopic dermatitis (MRSA colonization leads to more severe skin lesions developing) (187). Breaches in the skin, a first-line of defense against bacterial infection, can permit the pathogen to gain access to the bloodstream and result in potentially fatal diseases, including sepsis. One study demonstrated that 19–25% of patients who were colonized with MRSA in the healthcare setting subsequently developed a MRSA infection (186). Another study found 29% of patients colonized with MRSA in the healthcare setting developed a MRSA infection within 18 months after being discharged from the hospital (188). Such infections have been linked to lengthier hospital stays and higher costs due to infection control guidelines (including patient isolation/decolonization) and therapeutic intervention required to treat the disease (189). This is of particular concern to the pediatric and geriatric populations who are more susceptible to infection (with more severe morbidity) due to a less developed/weaker immune system. One way to deal with this challenge is to utilize agents to reduce MRSA colonization of human tissues, reducing the transmission of MRSA between people and decreasing the likelihood of an infection from developing.

Antibiotics, as decolonizing agents, have been shown to have a profound effect on decreasing MRSA colonization of host tissues (187). Mupirocin has been the drug of choice as a decolonization agent for many years (190). However, as highlighted earlier, strains of MRSA exhibiting moderate to high level of resistance to mupirocin have emerged. Of even greater concern are isolates of the predominant strain of MRSA (USA300) in the United States that have been documented to exhibit resistance to mupirocin (191). Mupirocin-resistant strains will naturally result in decolonization failure when this antibiotic is used. Thus alternative decolonization agents to mupirocin are critically needed.

The combination of potent antibacterial activity with low level of bacterial resistance emerging makes AMPs an attractive alternative to antibiotics as decolonizing agents. Successful experiments performed in animal models and clinical trials have given further support to the use of AMPs as decolonizing agents. A phase IIIa clinical trial with the peptide MBI-226 (Omiganin), developed by Migenix, demonstrated a 40% reduction in MRSA catheter colonization (31). This result lends further credibility to the usage of AMPs as decolonizing agents in addition to alternative topical antibiotics for the treatment of skin infections and immunomodulating agents for the treatment of MRSA infections.

8.3. AMPs for use in combination therapy

The development of novel antimicrobials has not been able to keep pace as the rate of bacterial resistance to antibiotics has continued to rise. Identification of new antimicrobials for use as single agents is a challenging and costly venture that has achieved very limited success in the past four decades. New antimicrobials which receive regulatory approval are often modified versions/derivatives of previously discovered antibiotics with more broad-spectrum activity (such as 4th- and 5th-generation cephalosporins). Though identifying novel lead antimicrobial compounds with a unique mechanism of action is one potential treatment strategy for bacterial infections, other methods have been employed. One of these methods which has observed considerable success is the use of combination therapy with two or more antibiotics (192). Using two antimicrobials together has multiple benefits including the usage of lower concentrations of each drug thus limiting potential issues associated with toxicity to host tissues (as higher concentrations of each drug given alone may trigger side effects at the effective therapeutic dose) and protection from bacterial resistance emerging rapidly to both antimicrobials given together. Currently, combination therapy involving the antibiotics vancomycin and rifampin in addition to sulfamethoxazole and trimethoprim have been successfully utilized against MRSA (193). A similar strategy employing an AMP in combination with a conventional antibiotic has received more attention in recent years (79, 95, 194).

There are multiple characteristics to look for in a suitable partner to test with a particular antimicrobial for combination therapy. AMPs possess two important characteristics that have been identified as important factors for using an agent in combination with another antibiotic including a low frequency of bacterial resistance and an antimicrobial that can rapidly kill the bacteria of interest (193). The fact that AMPs target different biological targets from most traditional antibiotics ensures they can complement the efforts of these conventional therapeutic agents to eradicate difficult pathogens, like MRSA. Several studies have highlighted a synergistic relationship being present when an AMP is combined with a natural product antibiotic. A study performed by Midorikawa et al, compared the efficacy of different AMPs produced by keratinocytes (including human β-defensin-1 (hBD1), hBD2, hBD3, and CAP18) both as stand-alone agents and in combination with one another (95). Interestingly, they found that when β-defensins were combined with CAP18, a synergistic relationship was observed against MRSA. Furthermore, when hBD3 or CAP18 were combined with methicillin at subinhibitory concentrations, a synergistic relationship was observed against ten MRSA strains.

Research conducted by our lab has identified salt-resistant synthetic peptides (RRIKA and RR) that exhibit potent activity in vitro and in vivo (in a murine skin infection model) against MRSA (79, 194). These peptides were found to be nontoxic to mammalian cells at concentrations 4- to 8-fold higher than their MICs against MRSA and exhibited limited hemolysis against human red blood cells (up to a concentration of 300 μM). Even more interesting was the fact that when these peptides were combined with the cell wall-disrupting antibiotic lysostaphin, a synergistic relationship was observed (with a fractional inhibitory concentration (FIC) value of 0.26, identified via the checkerboard assay). Furthermore, an additive effect was observed when these two peptides were combined with conventional antibiotics (vancomycin and linezolid) used to treat MRSA infections. This lends further support to the potential use of AMPs as combinatory agents with antibiotics against MRSA, particularly with skin infections.

8.4. Use as immunomodulatory or immunostimulatory agents

Naturally-occurring peptides, such as the defensins in humans, have been shown to play an important role in the immune response generated against invading pathogens. These molecules play an important role in innate immunity as they are often constitutively expressed or produced in response to microbial virulence factors such as lipopolysaccharide (in Gram-negative bacteria). However, studies have also shown that, in the presence of certain cytokines, leukocytes (including human natural killer cells, B cells, and γδT cells) can be induced to produce peptides such as α-defensins indicating they may play a vital role as effector molecules in adaptive immunity (195, 196). While defensins have been shown to possess potent antibacterial activity at low concentrations against S. aureus (196, 197), it points to a potential alternative use for peptides – as modulators or stimulators of the host immune response to clear invading pathogens. Thus peptides possessing limited or no antimicrobial activity but with immunostimulatory properties (to selectively upregulate the innate immune response to an infection without overstimulation of pro-inflammatory mediators) have potential use as therapeutic agents against S. aureus (particular for clearance of intracellular infections). This action can be combined with a subtherapeutic dose of a traditional antibiotic to achieve a two-fold attack on the bacterial infection – direct action against the pathogen (achieved by the antibiotic) combined with stimulation of the immune response (by the peptide) to clear the infection. Inimex Pharmaceuticals Inc. successfully demonstrated this strategy using a peptide (to enhance the immune response) in combination with a traditional antibiotic that worked in a murine S. aureus infection model to significantly reduce the bacterial load in vivo (40).

The immunomodulatory effect of AMPs is promising because it involves a different method of use over antibacterial action. One such peptide, LL-37 which has low to moderate antibacterial activity against S. aureus, was shown to be capable of reducing the bacterial load of S. aureus in a mouse model. This effect was postulated to be linked to LL-37’s ability to modulate the host immune response. LL-37 plays a role in the induction of chemokine production (IL-8, monocyte-chemoattractant proteins 1 and 3) necessary to attract immune cells including monocytes, neutrophils, and dendritic cells to the site of infection; thus it was proposed that these immune cells (induced by LL-37) were responsible for helping to clear the infection (151).

The immunomodulatory function of AMPs (such as IDR-1018) and AMP fragments (including HB-107) lacking antibacterial properties, has also been shown to accelerate wound repair (198), including wounds infected with S. aureus in animal models (199). Additionally, IDR-1018 was shown to possess both immunomodulatory activity (inducing the release of chemokines) while simultaneously suppressing a pro-inflammatory immune response (200). Controlled stimulation of the innate immune response by AMPs is critical; triggering the innate immune response often leads to excessive production of pro-inflammatory cytokines and effector molecules that can cause severe tissue damage or even lead to sepsis, a disease that kills 140,000 people every year in North America (41, 201).

9. CONCLUSION

Multidrug-resistant infections, in particular those caused by S. aureus, are a daunting public health challenge that requires serious attention. The diminishing utility of current antibiotics in the face of rising bacterial resistance further underscores the urgent need for the development of alternative therapeutic options and strategies to conventional antibiotics. Antimicrobial peptides present a strong potential ally in the battle against bacterial infections given their complex mode of action makes bacterial resistance to AMPs unlikely to rapidly emerge. Though these peptides possess several limitations which need to be addressed to use them in systemic applications (including stability in physiological conditions and limited ability to penetrate intracellular compartments where S. aureus tends to escape the host immune response), success in animal studies and human clinical trials supports the usage of AMPs as decolonizing agents and topical agents (either alone or in combination with conventional antibiotics) for the treatment of S. aureus skin and soft-tissue infections. Additionally, given antimicrobial peptides have the ability to modulate the host immune response, their potential use as immunomodulatory agents to stimulate the host immune response to clear an infection caused by multidrug-resistant bacteria presents another exciting avenue for researchers to pursue for the future. Though regulatory approval of AMPs for antibacterial application has yet to be achieved, the number of natural and synthetic peptides currently in clinical trials, and under development, presents hope that these compounds will be important clinical allies in the future for the treatment of bacterial infections.

References

  • 1.Antimicrobial Resistance: Global Report on Surveillance 2014. France: World Health Organization, 2014. [Google Scholar]
  • 2.Prevention CfDCa. Antibiotic Resistance Threats in the United States, 2013. 2013.
  • 3.Aiello AE, D Lowy F, Wright LN, Larson EL. Meticillin-resistant Staphylococcus aureus among US prisoners and military personnel: review and recommendations for future studies. Lancet Infect Dis. 2006;6(6):335–41. [DOI] [PubMed] [Google Scholar]
  • 4.Cohen PR. Cutaneous community-acquired methicillin-resistant Staphylococcus aureus infection in participants of athletic activities. South Med J. 2005;98(6):596–602. [DOI] [PubMed] [Google Scholar]
  • 5.Craven DE, Rixinger AI, Goularte TA, Mccabe WR. Methicillin-Resistant Staphylococcus-Aureus Bacteremia Linked to Intravenous Drug-Abusers Using a Shooting Gallery. Am J Med. 1986;80(5):770–6. [DOI] [PubMed] [Google Scholar]
  • 6.Gilbert M, MacDonald J, Gregson D, Siushansian J, Zhang K, Elsayed S, et al. Outbreak in Alberta of community-acquired (USA300) methicillin-resistant Staphylococcus aureus in people with a history of drug use, homelessness or incarceration. Can Med Assoc J. 2006;175(2):149–54. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 7.Herold BC, Immergluck LC, Maranan MC, Lauderdale DS, Gaskin RE, Boyle-Vavra S, et al. Community-acquired methicillin-resistant Staphylococcus aureus in children with no identified predisposing risk. Jama-J Am Med Assoc. 1998;279(8):593–8. [DOI] [PubMed] [Google Scholar]
  • 8.Karamatsu ML, Thorp AW, Brown L. Changes in Community-Associated Methicillin-Resistant Staphylococcus aureus Skin and Soft Tissue Infections Presenting to the Pediatric Emergency Department Comparing 2003 to 2008. Pediatr Emerg Care. 2012;28(2):131–5. [DOI] [PubMed] [Google Scholar]
  • 9.Kazakova SV, Hageman JC, Matava M, Srinivasan A, Phelan L, Garfinkel B, et al. A clone of methicillin-resistant Staphylococcus aureus among professional football players. New Engl J Med. 2005;352(5):468–75. [DOI] [PubMed] [Google Scholar]
  • 10.Malcolm B The rise of methicillin-resistant staphylococcus aureus in U.S. correctional populations. Journal of correctional health care : the official journal of the National Commission on Correctional Health Care. 2011;17(3):254–65. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 11.Regev-Yochay G, Rubinstein E, Barzilai A, Carmeli Y, Kuint J, Etienne J, et al. Methicillin-resistant Staphylococcus aureus in neonatal intensive care unit. Emerging infectious diseases. 2005;11(3):453–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 12.Kassis C, Hachem R, Raad II, Perego CA, Dvorak T, Hulten KG, et al. Outbreak of community-acquired methicillin-resistant Staphylococcus aureus skin infections among health care workers in a cancer center. Am J Infect Control. 2011;39(2):112–7. [DOI] [PubMed] [Google Scholar]
  • 13.Tristan A, Ferry T, Durand G, Dauwalder O, Bes M, Lina G, et al. Virulence determinants in community and hospital meticillin-resistant Staphylococcus aureus. The Journal of hospital infection. 2007;65 Suppl 2:105–9. [DOI] [PubMed] [Google Scholar]
  • 14.Bocchini CE, Hulten KG, Mason EO, Gonzalez BE, Hammerman WA, Kaplan SL. Panton-Valentine leukocidin genes are associated with enhanced inflammatory response and local disease in acute hematogenous Staphylococcus aureus osteomyelitis in children. Pediatrics. 2006;117(2):433–40. [DOI] [PubMed] [Google Scholar]
  • 15.David MZ, Daum RS. Community-Associated Methicillin-Resistant Staphylococcus aureus: Epidemiology and Clinical Consequences of an Emerging Epidemic. Clinical microbiology reviews. 2010;23(3):616-+. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 16.Gillet Y, Issartel B, Vanhems P, Fournet JC, Lina G, Bes M, et al. Association between Staphylococcus aureus strains carrying gene for Panton-Valentine leukocidin and highly lethal necrotising pneumonia in young immunocompetent patients. Lancet. 2002;359(9308):753–9. [DOI] [PubMed] [Google Scholar]
  • 17.Odell CA. Community-associated methicillin-resistant Staphylococcus aureus (CA-MRSA) skin infections. Current opinion in pediatrics. 2010;22(3):273–7. [DOI] [PubMed] [Google Scholar]
  • 18.Stryjewski ME, Chambers HF. Skin and soft-tissue infections caused by community-acquired methicillin-resistant Staphylococcus aureus. Clin Infect Dis. 2008;46 Suppl 5:S368–77. [DOI] [PubMed] [Google Scholar]
  • 19.Chambers HF. Community-associated MRSA--resistance and virulence converge. The New England journal of medicine. 2005;352(14):1485–7. [DOI] [PubMed] [Google Scholar]
  • 20.Frazee BW, Lynn J, Charlebois ED, Lambert L, Lowery D, Perdreau-Remington F. High prevalence of methicillin-resistant Staphylococcus aureus in emergency department skin and soft tissue infections. Annals of emergency medicine. 2005;45(3):311–20. [DOI] [PubMed] [Google Scholar]
  • 21.Fridkin SK, Hageman JC, Morrison M, Sanza LT, Como-Sabetti K, Jernigan JA, et al. Methicillin-resistant staphylococcus aureus disease in three communities. New Engl J Med. 2005;352(14):1436–44. [DOI] [PubMed] [Google Scholar]
  • 22.Han LL, McDougal LK, Gorwitz RJ, Mayer KH, Patel JB, Sennott JM, et al. High frequencies of clindamycin and tetracycline resistance in methicillin-resistant Staphylococcus aureus pulsed-field type USA300 isolates collected at a Boston ambulatory health center. J Clin Microbiol. 2007;45(4):1350–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 23.Moran GJ, Krishnadasan A, Gorwitz RJ, Fosheim GE, McDougal LK, Carey RB, et al. Methicillin-resistant S-aureus infections among patients in the emergency department. New Engl J Med. 2006;355(7):666–74. [DOI] [PubMed] [Google Scholar]
  • 24.Hiramatsu K Vancomycin-resistant Staphylococcus aureus: a new model of antibiotic resistance. Lancet Infect Dis. 2001;1(3):147–55. [DOI] [PubMed] [Google Scholar]
  • 25.Locke JB, Morales G, Hilgers M, Kedar GC, Rahawi S, Picazo JJ, et al. Elevated Linezolid Resistance in Clinical cfr-Positive Staphylococcus aureus Isolates Is Associated with Co-Occurring Mutations in Ribosomal Protein L3. Antimicrobial agents and chemotherapy. 2010;54(12):5352–5. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 26.Wilson P, Andrews JA, Charlesworth R, Walesby R, Singer M, Farrell DJ, et al. Linezolid resistance in clinical isolates of Staphylococcus aureus. J Antimicrob Chemoth. 2003;51(1):186–7. [DOI] [PubMed] [Google Scholar]
  • 27.Aldridge KE. Invitro Antistaphylococcal Activities of 2 Investigative Fluoroquinolones, Ci-960 and Win-57273, Compared with Those of Ciprofloxacin, Mupirocin (Pseudomonic Acid), and Peptide-Class Antimicrobial Agents. Antimicrobial agents and chemotherapy. 1992;36(4):851–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 28.Chatterjee S, Chatterjee S, Lad SJ, Phansalkar MS, Rupp RH, Ganguli BN, et al. Mersacidin, a new antibiotic from Bacillus. Fermentation, isolation, purification and chemical characterization. The Journal of antibiotics. 1992;45(6):832–8. [DOI] [PubMed] [Google Scholar]
  • 29.Fox JL. Antimicrobial peptides stage a comeback. Nature biotechnology. 2013;31(5):379–82. [DOI] [PubMed] [Google Scholar]
  • 30.Tincu JA, Menzel LP, Azimov R, Sands J, Hong T, Waring AJ, et al. Plicatamide, an antimicrobial octapeptide from Styela plicata hemocytes. The Journal of biological chemistry. 2003;278(15):13546–53. [DOI] [PubMed] [Google Scholar]
  • 31.Jenssen H, Hamill P, Hancock RE. Peptide antimicrobial agents. Clinical microbiology reviews. 2006;19(3):491–511. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 32.Pinheiro da Silva F, Machado MC. Antimicrobial peptides: clinical relevance and therapeutic implications. Peptides. 2012;36(2):308–14. [DOI] [PubMed] [Google Scholar]
  • 33.Lee IH, Cho Y, Lehrer RI. Effects of pH and salinity on the antimicrobial properties of clavanins. Infection and immunity. 1997;65(7):2898–903. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 34.Papagianni M Ribosomally synthesized peptides with antimicrobial properties: biosynthesis, structure, function, and applications. Biotechnology advances. 2003;21(6):465–99. [DOI] [PubMed] [Google Scholar]
  • 35.Boman HG, Agerberth B, Boman A. Mechanisms of action on Escherichia coli of cecropin P1 and PR-39, two antibacterial peptides from pig intestine. Infection and immunity. 1993;61(7):2978–84. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Lehrer RI, Barton A, Daher KA, Harwig SS, Ganz T, Selsted ME. Interaction of human defensins with Escherichia coli. Mechanism of bactericidal activity. The Journal of clinical investigation. 1989;84(2):553–61. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 37.Park CB, Kim HS, Kim SC. Mechanism of action of the antimicrobial peptide buforin II: buforin II kills microorganisms by penetrating the cell membrane and inhibiting cellular functions. Biochemical and biophysical research communications. 1998;244(1):253–7. [DOI] [PubMed] [Google Scholar]
  • 38.Schuerholz T, Brandenburg K, Marx G. Antimicrobial peptides and their potential application in inflammation and sepsis. Crit Care. 2012;16(2). [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Aldridge KE. In vitro antistaphylococcal activities of two investigative fluoroquinolones, CI-960 and WIN 57273, compared with those of ciprofloxacin, mupirocin (pseudomonic acid), and peptide-class antimicrobial agents. Antimicrobial agents and chemotherapy. 1992;36(4):851–3. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 40.Gordon YJ, Romanowski EG, McDermott AM. A review of antimicrobial peptides and their therapeutic potential as anti-infective drugs. Curr Eye Res. 2005;30(7):505–15. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Hancock REW, Sahl HG. Antimicrobial and host-defense peptides as new anti-infective therapeutic strategies. Nature biotechnology. 2006;24(12):1551–7. [DOI] [PubMed] [Google Scholar]
  • 42.Sivertsen A, Isaksson J, Leiros HKS, Svenson J, Svendsen JS, Brandsdal BO. Synthetic cationic antimicrobial peptides bind with their hydrophobic parts to drug site II of human serum albumin. Bmc Struct Biol. 2014;14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 43.Falla TJ, Karunaratne DN, Hancock RE. Mode of action of the antimicrobial peptide indolicidin. The Journal of biological chemistry. 1996;271(32):19298–303. [DOI] [PubMed] [Google Scholar]
  • 44.Ghiselli R, Giacometti A, Cirioni O, Mocchegiani F, Orlando F, Silvestri C, et al. Efficacy of the bovine antimicrobial peptide indolicidin combined with piperacillin/tazobactam in experimental rat models of polymicrobial peritonitis. Critical care medicine. 2008;36(1):240–5. [DOI] [PubMed] [Google Scholar]
  • 45.Robinson WE Jr., McDougall B, Tran D, Selsted ME. Anti-HIV-1 activity of indolicidin, an antimicrobial peptide from neutrophils. Journal of leukocyte biology. 1998;63(1):94–100. [DOI] [PubMed] [Google Scholar]
  • 46.Fritsche TR, Rhomberg PR, Sader HS, Jones RN. In vitro activity of omiganan pentahydrochloride tested against vancomycin-tolerant, -intermediate, and -resistant Staphylococcus aureus. Diagnostic microbiology and infectious disease. 2008;60(4):399–403. [DOI] [PubMed] [Google Scholar]
  • 47.Anderegg TR, Fritsche TR, Jones RN, Quality Control Working G. Quality control guidelines for MIC susceptibility testing of omiganan pentahydrochloride (MBI 226), a novel antimicrobial peptide. Journal of clinical microbiology. 2004;42(3):1386–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 48.Fritsche TR, Rhomberg PR, Sader HS, Jones RN. Antimicrobial activity of omiganan pentahydrochloride against contemporary fungal pathogens responsible for catheter-associated infections. Antimicrobial agents and chemotherapy. 2008;52(3):1187–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 49.Melo MN, Castanho MA. Omiganan interaction with bacterial membranes and cell wall models. Assigning a biological role to saturation. Biochimica et biophysica acta. 2007;1768(5):1277–90. [DOI] [PubMed] [Google Scholar]
  • 50.Rubinchik E, Dugourd D, Algara T, Pasetka C, Friedland HD. Antimicrobial and antifungal activities of a novel cationic antimicrobial peptide, omiganan, in experimental skin colonisation models. International journal of antimicrobial agents. 2009;34(5):457–61. [DOI] [PubMed] [Google Scholar]
  • 51.Bowdish DM, Davidson DJ, Lau YE, Lee K, Scott MG, Hancock RE. Impact of LL-37 on anti-infective immunity. Journal of leukocyte biology. 2005;77(4):451–9. [DOI] [PubMed] [Google Scholar]
  • 52.Noore J, Noore A, Li B. Cationic antimicrobial peptide LL-37 is effective against both extra- and intracellular Staphylococcus aureus. Antimicrobial agents and chemotherapy. 2013;57(3):1283–90. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 53.Fukumoto K, Nagaoka I, Yamataka A, Kobayashi H, Yanai T, Kato Y, et al. Effect of antibacterial cathelicidin peptide CAP18/LL-37 on sepsis in neonatal rats. Pediatric surgery international. 2005;21(1):20–4. [DOI] [PubMed] [Google Scholar]
  • 54.Jacobsen F, Mohammadi-Tabrisi A, Hirsch T, Mittler D, Mygind PH, Sonksen CP, et al. Antimicrobial activity of the recombinant designer host defence peptide P-novispirin G10 in infected full-thickness wounds of porcine skin. The Journal of antimicrobial chemotherapy. 2007;59(3):493–8. [DOI] [PubMed] [Google Scholar]
  • 55.Sawai MV, Waring AJ, Kearney WR, McCray PB Jr., Forsyth WR, Lehrer RI, et al. Impact of single-residue mutations on the structure and function of ovispirin/novispirin antimicrobial peptides. Protein engineering. 2002;15(3):225–32. [DOI] [PubMed] [Google Scholar]
  • 56.Simonetti O, Cirioni O, Goteri G, Ghiselli R, Kamysz W, Kamysz E, et al. Temporin A is effective in MRSA-infected wounds through bactericidal activity and acceleration of wound repair in a murine model. Peptides. 2008;29(4):520–8. [DOI] [PubMed] [Google Scholar]
  • 57.Wade D, Silberring J, Soliymani R, Heikkinen S, Kilpelainen I, Lankinen H, et al. Antibacterial activities of temporin A analogs. FEBS letters. 2000;479(1–2):6–9. [DOI] [PubMed] [Google Scholar]
  • 58.Mosca DA, Hurst MA, So W, Viajar BS, Fujii CA, Falla TJ. IB-367, a protegrin peptide with in vitro and in vivo activities against the microflora associated with oral mucositis. Antimicrobial agents and chemotherapy. 2000;44(7):1803–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Cirioni O, Silvestri C, Pierpaoli E, Barucca A, Kamysz W, Ghiselli R, et al. IB-367 pre-treatment improves the in vivo efficacy of teicoplanin and daptomycin in an animal model of wounds infected with meticillin-resistant Staphylococcus aureus. Journal of medical microbiology. 2013;62(Pt 10):1552–8. [DOI] [PubMed] [Google Scholar]
  • 60.Giacometti A, Cirioni O, Ghiselli R, Mocchegiani F, D’Amato G, Del Prete MS, et al. Administration of protegrin peptide IB-367 to prevent endotoxin induced mortality in bile duct ligated rats. Gut. 2003;52(6):874–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 61.Fitzgerald-Hughes D, Devocelle M, Humphreys H. Beyond conventional antibiotics for the future treatment of methicillin-resistant Staphylococcus aureus infections: two novel alternatives. FEMS immunology and medical microbiology. 2012;65(3):399–412. [DOI] [PubMed] [Google Scholar]
  • 62.Kumar K, Chopra S. New drugs for methicillin-resistant Staphylococcus aureus: an update. The Journal of antimicrobial chemotherapy. 2013;68(7):1465–70. [DOI] [PubMed] [Google Scholar]
  • 63.Steinberg DA, Hurst MA, Fujii CA, Kung AH, Ho JF, Cheng FC, et al. Protegrin-1: a broad-spectrum, rapidly microbicidal peptide with in vivo activity. Antimicrobial agents and chemotherapy. 1997;41(8):1738–42. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 64.Tran D, Tran P, Roberts K, Osapay G, Schaal J, Ouellette A, et al. Microbicidal properties and cytocidal selectivity of rhesus macaque theta defensins. Antimicrobial agents and chemotherapy. 2008;52(3):944–53. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 65.Lamb HM, Wiseman LR. Pexiganan acetate. Drugs. 1998;56(6):1047–52; discussion 53–4. [DOI] [PubMed] [Google Scholar]
  • 66.Gottler LM, Ramamoorthy A. Structure, membrane orientation, mechanism, and function of pexiganan--a highly potent antimicrobial peptide designed from magainin. Biochimica et biophysica acta. 2009;1788(8):1680–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 67.Mygind PH, Fischer RL, Schnorr KM, Hansen MT, Sonksen CP, Ludvigsen S, et al. Plectasin is a peptide antibiotic with therapeutic potential from a saprophytic fungus. Nature. 2005;437(7061):975–80. [DOI] [PubMed] [Google Scholar]
  • 68.Xiong YQ, Hady WA, Deslandes A, Rey A, Fraisse L, Kristensen HH, et al. Efficacy of NZ2114, a novel plectasin-derived cationic antimicrobial peptide antibiotic, in experimental endocarditis due to methicillin-resistant Staphylococcus aureus. Antimicrobial agents and chemotherapy. 2011;55(11):5325–30. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Brinch KS, Tulkens PM, Van Bambeke F, Frimodt-Moller N, Hoiby N, Kristensen HH. Intracellular activity of the peptide antibiotic NZ2114: studies with Staphylococcus aureus and human THP-1 monocytes, and comparison with daptomycin and vancomycin. The Journal of antimicrobial chemotherapy. 2010;65(8):1720–4. [DOI] [PubMed] [Google Scholar]
  • 70.Bommineni YR, Achanta M, Alexander J, Sunkara LT, Ritchey JW, Zhang G. A fowlicidin-1 analog protects mice from lethal infections induced by methicillin-resistant Staphylococcus aureus. Peptides. 2010;31(7):1225–30. [DOI] [PubMed] [Google Scholar]
  • 71.Faber C, Stallmann HP, Lyaruu DM, Joosten U, von Eiff C, van Nieuw Amerongen A, et al. Comparable efficacies of the antimicrobial peptide human lactoferrin 1–11 and gentamicin in a chronic methicillin-resistant Staphylococcus aureus osteomyelitis model. Antimicrobial agents and chemotherapy. 2005;49(6):2438–44. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 72.Stallmann HP, Faber C, Bronckers AL, Nieuw Amerongen AV, Wuisman PI. Osteomyelitis prevention in rabbits using antimicrobial peptide hLF1–11- or gentamicin-containing calcium phosphate cement. The Journal of antimicrobial chemotherapy. 2004;54(2):472–6. [DOI] [PubMed] [Google Scholar]
  • 73.Nibbering PH, Ravensbergen E, Welling MM, van Berkel LA, van Berkel PH, Pauwels EK, et al. Human lactoferrin and peptides derived from its N terminus are highly effective against infections with antibiotic-resistant bacteria. Infection and immunity. 2001;69(3):1469–76. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 74.Brouwer CP, Welling MM. Various routes of administration of (99m)Tc-labeled synthetic lactoferrin antimicrobial peptide hLF 1–11 enables monitoring and effective killing of multidrug-resistant Staphylococcus aureus infections in mice. Peptides. 2008;29(7):1109–17. [DOI] [PubMed] [Google Scholar]
  • 75.Brouwer CP, Rahman M, Welling MM. Discovery and development of a synthetic peptide derived from lactoferrin for clinical use. Peptides. 2011;32(9):1953–63. [DOI] [PubMed] [Google Scholar]
  • 76.Saravolatz LD, Pawlak J, Johnson L, Bonilla H, Saravolatz LD 2nd, Fakih MG, et al. In vitro activities of LTX-109, a synthetic antimicrobial peptide, against methicillin-resistant, vancomycin-intermediate, vancomycin-resistant, daptomycin-nonsusceptible, and linezolid-nonsusceptible Staphylococcus aureus. Antimicrobial agents and chemotherapy. 2012;56(8):4478–82. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 77.Isaksson J, Brandsdal BO, Engqvist M, Flaten GE, Svendsen JS, Stensen W. A synthetic antimicrobial peptidomimetic (LTX 109): stereochemical impact on membrane disruption. Journal of medicinal chemistry. 2011;54(16):5786–95. [DOI] [PubMed] [Google Scholar]
  • 78.Butler MS, Blaskovich MA, Cooper MA. Antibiotics in the clinical pipeline in 2013. The Journal of antibiotics. 2013;66(10):571–91. [DOI] [PubMed] [Google Scholar]
  • 79.Mohamed MF, Hamed MI, Panitch A, Seleem MN. Targeting methicillin-resistant Staphylococcus aureus with short salt-resistant synthetic peptides. Antimicrobial agents and chemotherapy. 2014;58(7):4113–22. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 80.Heunis TD, Smith C, Dicks LM. Evaluation of a nisin-eluting nanofiber scaffold to treat Staphylococcus aureus-induced skin infections in mice. Antimicrobial agents and chemotherapy. 2013;57(8):3928–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 81.Brumfitt W, Salton MR, Hamilton-Miller JM. Nisin, alone and combined with peptidoglycan-modulating antibiotics: activity against methicillin-resistant Staphylococcus aureus and vancomycin-resistant enterococci. The Journal of antimicrobial chemotherapy. 2002;50(5):731–4. [DOI] [PubMed] [Google Scholar]
  • 82.Vaucher RA, da Motta Ade S, Brandelli A. Evaluation of the in vitro cytotoxicity of the antimicrobial peptide P34. Cell biology international. 2010;34(3):317–23. [DOI] [PubMed] [Google Scholar]
  • 83.Ghobrial OG, Derendorf H, Hillman JD. Pharmacodynamic activity of the lantibiotic MU1140. International journal of antimicrobial agents. 2009;33(1):70–4. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 84.Ghobrial O, Derendorf H, Hillman JD. Pharmacokinetic and pharmacodynamic evaluation of the lantibiotic MU1140. Journal of pharmaceutical sciences. 2010;99(5):2521–8. [DOI] [PubMed] [Google Scholar]
  • 85.Pan CY, Chen JY, Cheng YS, Chen CY, Ni IH, Sheen JF, et al. Gene expression and localization of the epinecidin-1 antimicrobial peptide in the grouper (Epinephelus coioides), and its role in protecting fish against pathogenic infection. DNA and cell biology. 2007;26(6):403–13. [DOI] [PubMed] [Google Scholar]
  • 86.Huang HN, Rajanbabu V, Pan CY, Chan YL, Wu CJ, Chen JY. Use of the antimicrobial peptide Epinecidin-1 to protect against MRSA infection in mice with skin injuries. Biomaterials. 2013;34(38):10319–27. [DOI] [PubMed] [Google Scholar]
  • 87.Lin WJ, Chien YL, Pan CY, Lin TL, Chen JY, Chiu SJ, et al. Epinecidin-1, an antimicrobial peptide from fish (Epinephelus coioides) which has an antitumor effect like lytic peptides in human fibrosarcoma cells. Peptides. 2009;30(2):283–90. [DOI] [PubMed] [Google Scholar]
  • 88.Fernandez-Lopez S, Kim HS, Choi EC, Delgado M, Granja JR, Khasanov A, et al. Antibacterial agents based on the cyclic D,L-alpha-peptide architecture. Nature. 2001;412(6845):452–5. [DOI] [PubMed] [Google Scholar]
  • 89.Huang HN, Pan CY, Chan YL, Chen JY, Wu CJ. Use of the antimicrobial peptide pardaxin (GE33) to protect against methicillin-resistant Staphylococcus aureus infection in mice with skin injuries. Antimicrobial agents and chemotherapy. 2014;58(3):1538–45. [DOI] [PMC free article] [PubMed] [Google Scholar] [Retracted]
  • 90.Oren Z, Shai Y. A class of highly potent antibacterial peptides derived from pardaxin, a pore-forming peptide isolated from Moses sole fish Pardachirus marmoratus. European journal of biochemistry / FEBS. 1996;237(1):303–10. [DOI] [PubMed] [Google Scholar]
  • 91.Huang TC, Chen JY. Proteomic analysis reveals that pardaxin triggers apoptotic signaling pathways in human cervical carcinoma HeLa cells: cross talk among the UPR, c-Jun and ROS. Carcinogenesis. 2013;34(8):1833–42. [DOI] [PubMed] [Google Scholar]
  • 92.Menousek J, Mishra B, Hanke ML, Heim CE, Kielian T, Wang G. Database screening and in vivo efficacy of antimicrobial peptides against methicillin-resistant Staphylococcus aureus USA300. International journal of antimicrobial agents. 2012;39(5):402–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 93.Harder J, Bartels J, Christophers E, Schroder JM. Isolation and characterization of human beta -defensin-3, a novel human inducible peptide antibiotic. The Journal of biological chemistry. 2001;276(8):5707–13. [DOI] [PubMed] [Google Scholar]
  • 94.Zhu C, Tan H, Cheng T, Shen H, Shao J, Guo Y, et al. Human beta-defensin 3 inhibits antibiotic-resistant Staphylococcus biofilm formation. The Journal of surgical research. 2013;183(1):204–13. [DOI] [PubMed] [Google Scholar]
  • 95.Midorikawa K, Ouhara K, Komatsuzawa H, Kawai T, Yamada S, Fujiwara T, et al. Staphylococcus aureus susceptibility to innate antimicrobial peptides, beta-defensins and CAP18, expressed by human keratinocytes. Infection and immunity. 2003;71(7):3730–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 96.Li Q, Zhou Y, Dong K, Guo X. Potential therapeutic efficacy of a bactericidal-immunomodulatory fusion peptide against methicillin-resistant Staphylococcus aureus skin infection. Applied microbiology and biotechnology. 2010;86(1):305–9. [DOI] [PubMed] [Google Scholar]
  • 97.Jung S, Mysliwy J, Spudy B, Lorenzen I, Reiss K, Gelhaus C, et al. Human beta-defensin 2 and beta-defensin 3 chimeric peptides reveal the structural basis of the pathogen specificity of their parent molecules. Antimicrobial agents and chemotherapy. 2011;55(3):954–60. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 98.Kokai-Kun JF, Walsh SM, Chanturiya T, Mond JJ. Lysostaphin cream eradicates Staphylococcus aureus nasal colonization in a cotton rat model. Antimicrobial agents and chemotherapy. 2003;47(5):1589–97. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 99.Desbois AP, Gemmell CG, Coote PJ. In vivo efficacy of the antimicrobial peptide ranalexin in combination with the endopeptidase lysostaphin against wound and systemic meticillin-resistant Staphylococcus aureus (MRSA) infections. International journal of antimicrobial agents. 2010;35(6):559–65. [DOI] [PubMed] [Google Scholar]
  • 100.Kumar JK. Lysostaphin: an antistaphylococcal agent. Applied microbiology and biotechnology. 2008;80(4):555–61. [DOI] [PubMed] [Google Scholar]
  • 101.Cherkasov A, Hilpert K, Jenssen H, Fjell CD, Waldbrook M, Mullaly SC, et al. Use of artificial intelligence in the design of small peptide antibiotics effective against a broad spectrum of highly antibiotic-resistant superbugs. ACS chemical biology. 2009;4(1):65–74. [DOI] [PubMed] [Google Scholar]
  • 102.Brogden KA. Antimicrobial peptides: pore formers or metabolic inhibitors in bacteria? Nature reviews Microbiology. 2005;3(3):238–50. [DOI] [PubMed] [Google Scholar]
  • 103.Wu M, Maier E, Benz R, Hancock RE. Mechanism of interaction of different classes of cationic antimicrobial peptides with planar bilayers and with the cytoplasmic membrane of Escherichia coli. Biochemistry. 1999;38(22):7235–42. [DOI] [PubMed] [Google Scholar]
  • 104.Wu M, Hancock RE. Interaction of the cyclic antimicrobial cationic peptide bactenecin with the outer and cytoplasmic membrane. The Journal of biological chemistry. 1999;274(1):29–35. [DOI] [PubMed] [Google Scholar]
  • 105.Matsuzaki K, Murase O, Fujii N, Miyajima K. An antimicrobial peptide, magainin 2, induced rapid flip-flop of phospholipids coupled with pore formation and peptide translocation. Biochemistry. 1996;35(35):11361–8. [DOI] [PubMed] [Google Scholar]
  • 106.Hasper HE, Kramer NE, Smith JL, Hillman JD, Zachariah C, Kuipers OP, et al. An alternative bactericidal mechanism of action for lantibiotic peptides that target lipid II. Science. 2006;313(5793):1636–7. [DOI] [PubMed] [Google Scholar]
  • 107.Schneider T, Kruse T, Wimmer R, Wiedemann I, Sass V, Pag U, et al. Plectasin, a fungal defensin, targets the bacterial cell wall precursor Lipid II. Science. 2010;328(5982):1168–72. [DOI] [PubMed] [Google Scholar]
  • 108.Zhang L, Parente J, Harris SM, Woods DE, Hancock RE, Falla TJ. Antimicrobial peptide therapeutics for cystic fibrosis. Antimicrobial agents and chemotherapy. 2005;49(7):2921–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 109.Patrzykat A, Friedrich CL, Zhang L, Mendoza V, Hancock RE. Sublethal concentrations of pleurocidin-derived antimicrobial peptides inhibit macromolecular synthesis in Escherichia coli. Antimicrobial agents and chemotherapy. 2002;46(3):605–14. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 110.Xiong YQ, Yeaman MR, Bayer AS. In vitro antibacterial activities of platelet microbicidal protein and neutrophil defensin against Staphylococcus aureus are influenced by antibiotics differing in mechanism of action. Antimicrobial agents and chemotherapy. 1999;43(5):1111–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 111.Subbalakshmi C, Sitaram N. Mechanism of antimicrobial action of indolicidin. FEMS microbiology letters. 1998;160(1):91–6. [DOI] [PubMed] [Google Scholar]
  • 112.Nan YH, Park KH, Park Y, Jeon YJ, Kim Y, Park IS, et al. Investigating the effects of positive charge and hydrophobicity on the cell selectivity, mechanism of action and anti-inflammatory activity of a Trp-rich antimicrobial peptide indolicidin. FEMS microbiology letters. 2009;292(1):134–40. [DOI] [PubMed] [Google Scholar]
  • 113.Nizet V, Ohtake T, Lauth X, Trowbridge J, Rudisill J, Dorschner RA, et al. Innate antimicrobial peptide protects the skin from invasive bacterial infection. Nature. 2001;414(6862):454–7. [DOI] [PubMed] [Google Scholar]
  • 114.Scott MG, Dullaghan E, Mookherjee N, Glavas N, Waldbrook M, Thompson A, et al. An anti-infective peptide that selectively modulates the innate immune response. Nature biotechnology. 2007;25(4):465–72. [DOI] [PubMed] [Google Scholar]
  • 115.Cavaillon JM, Adib-Conquy M, Fitting C, Adrie C, Payen D. Cytokine cascade in sepsis. Scandinavian journal of infectious diseases. 2003;35(9):535–44. [DOI] [PubMed] [Google Scholar]
  • 116.Zisman DA, Kunkel SL, Strieter RM, Tsai WC, Bucknell K, Wilkowski J, et al. MCP-1 protects mice in lethal endotoxemia. The Journal of clinical investigation. 1997;99(12):2832–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 117.van der Does AM, Hensbergen PJ, Bogaards SJ, Cansoy M, Deelder AM, van Leeuwen HC, et al. The human lactoferrin-derived peptide hLF1–11 exerts immunomodulatory effects by specific inhibition of myeloperoxidase activity. Journal of immunology. 2012;188(10):5012–9. [DOI] [PubMed] [Google Scholar]
  • 118.van der Does AM, Bogaards SJ, Ravensbergen B, Beekhuizen H, van Dissel JT, Nibbering PH. Antimicrobial peptide hLF1–11 directs granulocyte-macrophage colony-stimulating factor-driven monocyte differentiation toward macrophages with enhanced recognition and clearance of pathogens. Antimicrobial agents and chemotherapy. 2010;54(2):811–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 119.Yang D, Chertov O, Oppenheim JJ. Participation of mammalian defensins and cathelicidins in anti-microbial immunity: receptors and activities of human defensins and cathelicidin (LL-37). J Leukoc Biol. 2001;69(5):691–7. [PubMed] [Google Scholar]
  • 120.Niyonsaba F, Iwabuchi K, Someya A, Hirata M, Matsuda H, Ogawa H, et al. A cathelicidin family of human antibacterial peptide LL-37 induces mast cell chemotaxis. Immunology. 2002;106(1):20–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 121.Tjabringa GS, Ninaber DK, Drijfhout JW, Rabe KF, Hiemstra PS. Human cathelicidin LL-37 is a chemoattractant for eosinophils and neutrophils that acts via formyl-peptide receptors. International archives of allergy and immunology. 2006;140(2):103–12. [DOI] [PubMed] [Google Scholar]
  • 122.Mookherjee N, Brown KL, Bowdish DM, Doria S, Falsafi R, Hokamp K, et al. Modulation of the TLR-mediated inflammatory response by the endogenous human host defense peptide LL-37. J Immunol. 2006;176(4):2455–64. [DOI] [PubMed] [Google Scholar]
  • 123.Montreekachon P, Chotjumlong P, Bolscher JG, Nazmi K, Reutrakul V, Krisanaprakornkit S. Involvement of P2X(7) purinergic receptor and MEK1/2 in interleukin-8 up-regulation by LL-37 in human gingival fibroblasts. Journal of periodontal research. 2011;46(3):327–37. [DOI] [PubMed] [Google Scholar]
  • 124.Mookherjee N, Lippert DN, Hamill P, Falsafi R, Nijnik A, Kindrachuk J, et al. Intracellular receptor for human host defense peptide LL-37 in monocytes. Journal of immunology. 2009;183(4):2688–96. [DOI] [PubMed] [Google Scholar]
  • 125.Tjabringa GS, Ninaber DK, Drijfhout JW, Rabe KF, Hiemstra PS. Human cathelicidin LL-37 is a chemoattractant for eosinophils and neutrophils that acts via formyl-peptide receptors. Int Arch Allergy Imm. 2006;140(2):103–12. [DOI] [PubMed] [Google Scholar]
  • 126.Niyonsaba F, Ushio H, Nakano N, Ng W, Sayama K, Hashimoto K, et al. Antimicrobial peptides human beta-defensins stimulate epidermal keratinocyte migration, proliferation and production of proinflammatory cytokines and chemokines. The Journal of investigative dermatology. 2007;127(3):594–604. [DOI] [PubMed] [Google Scholar]
  • 127.Ramos R, Silva JP, Rodrigues AC, Costa R, Guardao L, Schmitt F, et al. Wound healing activity of the human antimicrobial peptide LL37. Peptides. 2011;32(7):1469–76. [DOI] [PubMed] [Google Scholar]
  • 128.Heilborn JD, Nilsson MF, Kratz G, Weber G, Sorensen O, Borregaard N, et al. The cathelicidin anti-microbial peptide LL-37 is involved in re-epithelialization of human skin wounds and is lacking in chronic ulcer epithelium. The Journal of investigative dermatology. 2003;120(3):379–89. [DOI] [PubMed] [Google Scholar]
  • 129.Koczulla R, von Degenfeld G, Kupatt C, Krotz F, Zahler S, Gloe T, et al. An angiogenic role for the human peptide antibiotic LL-37/hCAP-18. The Journal of clinical investigation. 2003;111(11):1665–72. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 130.Dang CN, Prasad YD, Boulton AJ, Jude EB. Methicillin-resistant Staphylococcus aureus in the diabetic foot clinic: a worsening problem. Diabetic medicine : a journal of the British Diabetic Association. 2003;20(2):159–61. [DOI] [PubMed] [Google Scholar]
  • 131.Lipsky BA, Stoutenburgh U. Daptomycin for treating infected diabetic foot ulcers: evidence from a randomized, controlled trial comparing daptomycin with vancomycin or semi-synthetic penicillins for complicated skin and skin-structure infections. The Journal of antimicrobial chemotherapy. 2005;55(2):240–5. [DOI] [PubMed] [Google Scholar]
  • 132.Tentolouris N, Jude EB, Smirnof I, Knowles EA, Boulton AJ. Methicillin-resistant Staphylococcus aureus: an increasing problem in a diabetic foot clinic. Diabetic medicine : a journal of the British Diabetic Association. 1999;16(9):767–71. [DOI] [PubMed] [Google Scholar]
  • 133.Fejfarova V, Jirkovska A, Skibova J, Petkov V. [Pathogen resistance and other risk factors in the frequency of lower limb amputations in patients with the diabetic foot syndrome]. Vnitrni lekarstvi. 2002;48(4):302–6. [PubMed] [Google Scholar]
  • 134.Lipsky BA, Berendt AR, Cornia PB, Pile JC, Peters EJ, Armstrong DG, et al. 2012 Infectious Diseases Society of America clinical practice guideline for the diagnosis and treatment of diabetic foot infections. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 2012;54(12):e132–73. [DOI] [PubMed] [Google Scholar]
  • 135.Galkowska H, Podbielska A, Olszewski WL, Stelmach E, Luczak M, Rosinski G, et al. Epidemiology and prevalence of methicillin-resistant Staphylococcus aureus and Staphylococcus epidermidis in patients with diabetic foot ulcers: focus on the differences between species isolated from individuals with ischemic vs. neuropathic foot ulcers. Diabetes research and clinical practice. 2009;84(2):187–93. [DOI] [PubMed] [Google Scholar]
  • 136.Rosenberg CS. Wound healing in the patient with diabetes mellitus. The Nursing clinics of North America. 1990;25(1):247–61. [PubMed] [Google Scholar]
  • 137.Hirsch T, Spielmann M, Zuhaili B, Fossum M, Metzig M, Koehler T, et al. Human beta-defensin-3 promotes wound healing in infected diabetic wounds. The journal of gene medicine. 2009;11(3):220–8. [DOI] [PubMed] [Google Scholar]
  • 138.Zhang L, Scott MG, Yan H, Mayer LD, Hancock RE. Interaction of polyphemusin I and structural analogs with bacterial membranes, lipopolysaccharide, and lipid monolayers. Biochemistry. 2000;39(47):14504–14. [DOI] [PubMed] [Google Scholar]
  • 139.Skerlavaj B, Gennaro R, Bagella L, Merluzzi L, Risso A, Zanetti M. Biological characterization of two novel cathelicidin-derived peptides and identification of structural requirements for their antimicrobial and cell lytic activities. Journal of Biological Chemistry. 1996;271(45):28375–81. [DOI] [PubMed] [Google Scholar]
  • 140.Ge Y, MacDonald DL, Holroyd KJ, Thornsberry C, Wexler H, Zasloff M. In vitro antibacterial properties of pexiganan, an analog of magainin. Antimicrobial agents and chemotherapy. 1999;43(4):782–8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 141.Ge Y, MacDonald D, Henry MM, Hait HI, Nelson KA, Lipsky BA, et al. In vitro susceptibility to pexiganan of bacteria isolated from infected diabetic foot ulcers. Diagn Microbiol Infect Dis. 1999;35(1):45–53. [DOI] [PubMed] [Google Scholar]
  • 142.Barker KF. Antibiotic resistance: a current perspective. British journal of clinical pharmacology. 1999;48(2):109–24. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 143.Nordmann P, Guibert M. Extended-spectrum beta-lactamases in Pseudomonas aeruginosa. The Journal of antimicrobial chemotherapy. 1998;42(2):128–31. [DOI] [PubMed] [Google Scholar]
  • 144.Cohen J, McConnell JS. Antibiotic-induced endotoxin release. Lancet. 1985;2(8463):1069–70. [DOI] [PubMed] [Google Scholar]
  • 145.Shenep JL, Barton RP, Mogan KA. Role of antibiotic class in the rate of liberation of endotoxin during therapy for experimental gram-negative bacterial sepsis. J Infect Dis. 1985;151(6):1012–8. [DOI] [PubMed] [Google Scholar]
  • 146.Mannis MJ. The use of antimicrobial peptides in ophthalmology: an experimental study in corneal preservation and the management of bacterial keratitis. Transactions of the American Ophthalmological Society. 2002;100:243–71. [PMC free article] [PubMed] [Google Scholar]
  • 147.Fraser J, West M, Krieger T, Taylor R, Erfle D. Compositions and methods for treating infections using analogues of indolicidin. Google Patents; 2004. [Google Scholar]
  • 148.Ghuman J, Zunszain PA, Petitpas I, Bhattacharya AA, Otagiri M, Curry S. Structural basis of the drug-binding specificity of human serum albumin. Journal of molecular biology. 2005;353(1):38–52. [DOI] [PubMed] [Google Scholar]
  • 149.Herve F, Urien S, Albengres E, Duche JC, Tillement JP. Drug binding in plasma. A summary of recent trends in the study of drug and hormone binding. Clinical pharmacokinetics. 1994;26(1):44–58. [DOI] [PubMed] [Google Scholar]
  • 150.Rothstein DM, Spacciapoli P, Tran LT, Xu T, Roberts FD, Dalla Serra M, et al. Anticandida activity is retained in P-113, a 12-amino-acid fragment of histatin 5. Antimicrobial agents and chemotherapy. 2001;45(5):1367–73. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 151.Bowdish DME, Davidson DJ, Lau YE, Lee K, Scott MG, Hancock REW. Impact of LL-37 on anti-infective immunity. J Leukocyte Biol. 2005;77(4):451–9. [DOI] [PubMed] [Google Scholar]
  • 152.Chu HL, Yu HY, Yip BS, Chih YH, Liang CW, Cheng HT, et al. Boosting Salt Resistance of Short Antimicrobial Peptides. Antimicrobial agents and chemotherapy. 2013;57(8):4050–2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 153.Verhoef JC, Bodde HE, Deboer AG, Bouwstra JA, Junginger HE, Merkus FWHM, et al. Transport of Peptide and Protein Drugs across Biological-Membranes. Eur J Drug Metab Ph. 1990;15(2):83–93. [DOI] [PubMed] [Google Scholar]
  • 154.Houston ME, Kondejewski LH, Karunaratne DN, Gough M, Fidai S, Hodges RS, et al. Influence of preformed alpha-helix and alpha-helix induction on the activity of cationic antimicrobial peptides. J Pept Res. 1998;52(2):81–8. [DOI] [PubMed] [Google Scholar]
  • 155.Osapay K, Tran D, Ladokhin AS, White SH, Henschen AH, Selsted ME. Formation and characterization of a single Trp-Trp cross-link in indolicidin that confers protease stability without altering antimicrobial activity. Journal of Biological Chemistry. 2000;275(16):12017–22. [DOI] [PubMed] [Google Scholar]
  • 156.R TB, Jeffrey A, Siahaan TJ, Gangwar S, Pauletti GM. Improvement of oral peptide bioavailability: Peptidomimetics and prodrug strategies. Advanced drug delivery reviews. 1997;27(2–3):235–56. [DOI] [PubMed] [Google Scholar]
  • 157.Rozek A, Powers JP, Friedrich CL, Hancock RE. Structure-based design of an indolicidin peptide analogue with increased protease stability. Biochemistry. 2003;42(48):14130–8. [DOI] [PubMed] [Google Scholar]
  • 158.Giuliani A, Rinaldi AC. Beyond natural antimicrobial peptides: multimeric peptides and other peptidomimetic approaches. Cell Mol Life Sci. 2011;68(13):2255–66. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 159.Jin T, Bokarewa M, Foster T, Mitchell J, Higgins J, Tarkowski A. Staphylococcus aureus resists human defensins by production of staphylokinase, a novel bacterial evasion mechanism. J Immunol. 2004;172(2):1169–76. [DOI] [PubMed] [Google Scholar]
  • 160.Sieprawska-Lupa M, Mydel P, Krawczyk K, Wojcik K, Puklo M, Lupa B, et al. Degradation of human antimicrobial peptide LL-37 by Staphylococcus aureus-derived proteinases. Antimicrobial agents and chemotherapy. 2004;48(12):4673–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 161.Nizet V Antimicrobial peptide resistance mechanisms of human bacterial pathogens. Current issues in molecular biology. 2006;8(1):11–26. [PubMed] [Google Scholar]
  • 162.Peschel A How do bacteria resist human antimicrobial peptides? Trends Microbiol. 2002;10(4):179–86. [DOI] [PubMed] [Google Scholar]
  • 163.Kupferwasser LI, Skurray RA, Brown MH, Firth N, Yeaman MR, Bayer AS. Plasmid-mediated resistance to thrombin-induced platelet microbicidal protein in staphylococci: role of the qacA locus. Antimicrobial agents and chemotherapy. 1999;43(10):2395–9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 164.Schmidtchen A, Pasupuleti M, Morgelin M, Davoudi M, Alenfall J, Chalupka A, et al. Boosting Antimicrobial Peptides by Hydrophobic Oligopeptide End Tags. Journal of Biological Chemistry. 2009;284(26):17584–94. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 165.Garzoni C, Francois P, Huyghe A, Couzinet S, Tapparel C, Charbonnier Y, et al. A global view of Staphylococcus aureus whole genome expression upon internalization in human epithelial cells. Bmc Genomics. 2007;8. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 166.Ong ZY, Cheng J, Huang Y, Xu K, Ji Z, Fan W, et al. Effect of stereochemistry, chain length and sequence pattern on antimicrobial properties of short synthetic beta-sheet forming peptide amphiphiles. Biomaterials. 2014;35(4):1315–25. [DOI] [PubMed] [Google Scholar]
  • 167.Chatterjee S, Chatterjee DK, Jani RH, Blumbach J, Ganguli BN, Klesel N, et al. Mersacidin, a new antibiotic from Bacillus. In vitro and in vivo antibacterial activity. The Journal of antibiotics. 1992;45(6):839–45. [DOI] [PubMed] [Google Scholar]
  • 168.PolyMedix Announces Positive Results From Phase 2 Clinical Trial With PMX-30063 First-in-Class Defensin-Mimetic Antibiotic [press release]. April 23, 2012.
  • 169.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. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 2008;47(12):1537–45. [DOI] [PubMed] [Google Scholar]
  • 170.Markova A, Mostow EN. US skin disease assessment: ulcer and wound care. Dermatologic clinics. 2012;30(1):107–11, ix. [DOI] [PubMed] [Google Scholar]
  • 171.Shlaes DM, Sahm D, Opiela C, Spellberg B. The FDA reboot of antibiotic development. Antimicrob Agents Chemother. 2013;57(10):4605–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 172.Kollef M, Pittet D, Sanchez Garcia M, Chastre J, Fagon JY, Bonten M, et al. A randomized double-blind trial of iseganan in prevention of ventilator-associated pneumonia. American journal of respiratory and critical care medicine. 2006;173(1):91–7. [DOI] [PubMed] [Google Scholar]
  • 173.Fritsche TR, Rhomberg PR, Sader HS, Jones RN. Antimicrobial activity of omiganan pentahydrochloride tested against contemporary bacterial pathogens commonly responsible for catheter-associated infections. The Journal of antimicrobial chemotherapy. 2008;61(5):1092–8. [DOI] [PubMed] [Google Scholar]
  • 174.Mensa B, Howell GL, Scott R, DeGrado WF. Comparative Mechanistic Studies of Brilacidin, Daptomycin, and the Antimicrobial Peptide LL16. Antimicrobial agents and chemotherapy. 2014;58(9):5136–45. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 175.Moran GJ, Krishnadasan A, Gorwitz RJ, Fosheim GE, McDougal LK, Carey RB, et al. Methicillin-resistant S. aureus infections among patients in the emergency department. The New England journal of medicine. 2006;355(7):666–74. [DOI] [PubMed] [Google Scholar]
  • 176.Landrum ML, Neumann C, Cook C, Chukwuma U, Ellis MW, Hospenthal DR, et al. Epidemiology of Staphylococcus aureus blood and skin and soft tissue infections in the US military health system, 2005–2010. JAMA : the journal of the American Medical Association. 2012;308(1):50–9. [DOI] [PubMed] [Google Scholar]
  • 177.Liu C, Bayer A, Cosgrove SE, Daum RS, Fridkin SK, Gorwitz RJ. Clinical Practice Guidelines by the Infectious Diseases Society of America for the Treatment of Methicillin-Resistant Staphylococcus aureus Infections in Adults and Children (vol 52, pg e18, 2011). Clin Infect Dis. 2011;53(3):319- [DOI] [PubMed] [Google Scholar]
  • 178.Stevens DL, Bisno AL, Chambers HF, Dellinger EP, Goldstein EJ, Gorbach SL, et al. Executive summary: practice guidelines for the diagnosis and management of skin and soft tissue infections: 2014 update by the infectious diseases society of america. Clin Infect Dis. 2014;59(2):147–59. [DOI] [PubMed] [Google Scholar]
  • 179.Mulvey MR, MacDougall L, Cholin B, Horsman G, Fidyk M, Woods S, et al. Community-associated methicillin-resistant Staphylococcus aureus, Canada. Emerging infectious diseases. 2005;11(6):844–50. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 180.Simor AE, Stuart TL, Louie L, Watt C, Ofner-Agostini M, Gravel D, et al. Mupirocin-resistant, methicillin-resistant Staphylococcus aureus strains in Canadian hospitals. Antimicrobial agents and chemotherapy. 2007;51(11):3880–6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 181.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(12):1537–45. [DOI] [PubMed] [Google Scholar]
  • 182.Schwab U, Gilligan P, Jaynes J, Henke D. In vitro activities of designed antimicrobial peptides against multidrug-resistant cystic fibrosis pathogens. Antimicrobial agents and chemotherapy. 1999;43(6):1435–40. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 183.Chalekson CP, Neumeister MW, Jaynes J. Treatment of infected wounds with the antimicrobial peptide D2A21. The Journal of trauma. 2003;54(4):770–4. [DOI] [PubMed] [Google Scholar]
  • 184.Chalekson CP, Neumeister MW, Jaynes J. Improvement in burn wound infection and survival with antimicrobial peptide D2A21 (Demegel). Plastic and reconstructive surgery. 2002;109(4):1338–43. [DOI] [PubMed] [Google Scholar]
  • 185.Crunkhorn S Antibacterial drugs: New antibiotics on the horizon? Nature reviews Drug discovery. 2013;12(2):99. [DOI] [PubMed] [Google Scholar]
  • 186.Davis KA, Stewart JJ, Crouch HK, Florez CE, Hospenthal DR. Methicillin-resistant Staphylococcus aureus (MRSA) nares colonization at hospital admission and its effect on subsequent MRSA infection. Clin Infect Dis. 2004;39(6):776–82. [DOI] [PubMed] [Google Scholar]
  • 187.Gong JQ, Lin L, Lin T, Hao F, Zeng FQ, Bi ZG, et al. Skin colonization by Staphylococcus aureus in patients with eczema and atopic dermatitis and relevant combined topical therapy: a double-blind multicentre randomized controlled trial. Brit J Dermatol. 2006;155(4):680–7. [DOI] [PubMed] [Google Scholar]
  • 188.Huang SS, Platt R. Risk of methicillin-resistant Staphylococcus aureus infection after previous infection or colonization. Clin Infect Dis. 2003;36(3):281–5. [DOI] [PubMed] [Google Scholar]
  • 189.Carbon C Costs of treating infections caused by methicillin-resistant staphylococci and vancomycin-resistant enterococci. The Journal of antimicrobial chemotherapy. 1999;44 Suppl A:31–6. [DOI] [PubMed] [Google Scholar]
  • 190.McConeghy KW, Mikolich DJ, LaPlante KL. Agents for the Decolonization of Methicillin-Resistant Staphylococcus aureus. Pharmacotherapy. 2009;29(3):263–80. [DOI] [PubMed] [Google Scholar]
  • 191.McDougal LK, Fosheim GE, Nicholson A, Bulens SN, Limbago BM, Shearer JES, et al. Emergence of Resistance among USA300 Methicillin-Resistant Staphylococcus aureus Isolates Causing Invasive Disease in the United States. Antimicrobial agents and chemotherapy. 2010;54(9):3804–11. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 192.Tegos G, Mylonakis E Antimicrobial Drug Discovery: Emerging Strategies.2012.
  • 193.Deresinski S Vancomycin in combination with other antibiotics for the treatment of serious methicillin-resistant Staphylococcus aureus infections. Clinical infectious diseases : an official publication of the Infectious Diseases Society of America. 2009;49(7):1072–9. [DOI] [PubMed] [Google Scholar]
  • 194.Mohamed MF, Seleem MN. Efficacy of short novel antimicrobial and anti-inflammatory peptides in a mouse model of methicillin-resistant Staphylococcus aureus (MRSA) skin infection. Drug design, development and therapy. 2014;8:1979–83. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 195.Agerberth B, Charo J, Werr J, Olsson B, Idali F, Lindbom L, et al. The human antimicrobial and chemotactic peptides LL-37 and alpha-defensins are expressed by specific lymphocyte and monocyte populations. Blood. 2000;96(9):3086–93. [PubMed] [Google Scholar]
  • 196.Ganz T, Selsted ME, Szklarek D, Harwig SS, Daher K, Bainton DF, et al. Defensins. Natural peptide antibiotics of human neutrophils. The Journal of clinical investigation. 1985;76(4):1427–35. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 197.Eisenhauer PB, Harwig SS, Szklarek D, Ganz T, Selsted ME, Lehrer RI. Purification and antimicrobial properties of three defensins from rat neutrophils. Infection and immunity. 1989;57(7):2021–7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 198.Lee PH, Rudisill JA, Lin KH, Zhang L, Harris SM, Falla TJ, et al. HB-107, a nonbacteriostatic fragment of the antimicrobial peptide cecropin B, accelerates murine wound repair. Wound repair and regeneration : official publication of the Wound Healing Society [and] the European Tissue Repair Society. 2004;12(3):351–8. [DOI] [PubMed] [Google Scholar]
  • 199.Steinstraesser L, Hirsch T, Schulte M, Kueckelhaus M, Jacobsen F, Mersch EA, et al. Innate defense regulator peptide 1018 in wound healing and wound infection. PloS one. 2012;7(8):e39373. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 200.Wieczorek M, Jenssen H, Kindrachuk J, Scott WR, Elliott M, Hilpert K, et al. Structural studies of a peptide with immune modulating and direct antimicrobial activity. Chemistry & biology. 2010;17(9):970–80. [DOI] [PubMed] [Google Scholar]
  • 201.Bacha JD Oreola; Lee Ken; Scott Monisha. The Innate Defense. Modern Drug Discovery. 2004;7(s2):43–6. [Google Scholar]

RESOURCES