Mini Review

Antimicrobial Peptides: A Potential Therapeutic Option for Surgical Site Infections

Berthony Deslouches1,2 and Y Peter Di1*
1Department of Environmental and Occupational Health, University of Pittsburgh Graduate School of Public Health, Pittsburgh, PA, USA
2Department of Microbiology and Molecular Genetics, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA


*Corresponding author: Y Peter Di, Department of Environmental and Occupational Health, 100 Technology Drive, Bridgeside Point I, Room 331, Pittsburgh, PA 15206, USA


Published: 16 Nov, 2017
Cite this article as: Deslouches B, Peter Di Y. Antimicrobial Peptides: A Potential Therapeutic Option for Surgical Site Infections. Clin Surg. 2017; 2: 1740.

Abstract

Surgical Site Infections (SSI) represents one of the most common hospital-associated infections worldwide, and many cases of SSI are due to multidrug-resistant bacteria with the propensity to attach to tissues and form biofilm on post-surgical sites. While systemic antibiotic treatment (prophylactically and therapeutically) is usually effective, SSI can be difficult to treat when associated with drug resistance. Antimicrobial Peptides (AMPs) are an untapped resource that could serve as an effective therapeutic option, as they display broad-spectrum antimicrobial activity regardless of antibiotic resistance. In the last decade, it has become increasingly clear that AMPs also display antibiofilm properties. We reviewed herein the potential of AMPs as promising therapeutics for SSI and the need for structural optimization to develop AMPs for clinical applications.

Introduction

Natural and Synthetic Antimicrobial Peptides (AMPs) exhibit great potential as new therapeutic agents for surgical site infections. AMPs rapidly kill their bacterial targets by membrane-disruption, or less commonly by interference with vital intracellular functions (e.g., DNA, RNA, protein synthesis) [1-3]. AMPs also display anti-biofilm activities [4,5]. However, after more than three decades of AMP discoveries, they are yet to be clinically established as practical antibiotics against common Multi-Drug Resistant (MDR) bacteria. The alarming emergence of MDR pathogenic microorganisms [6-8], coupled with a decrease in the pharmaceutical industry research pipeline for novel antimicrobial agents, has created an urgent need to develop new strategies to address the pressing problems associated with current infectious diseases [9]. Thus, infections associated with MDR pathogens constitute an imminent health crisis (World Health Organization, 2014) [6,10]. These infections are of particular importance to surgical patients, as hospital-associated transmission of MDR bacteria in these patients can frequently lead to life-threatening complications [11-13]. Surgical Site Infections (SSI) refer to infections of the skin and subcutaneous or deep tissues that occur at a manipulated site within 30 days after an operation (WHO, 2016) [14,15]. The mean incidence of SSI is 7 to 15 cases per 100 hospitalized patients, with higher incidence in lower-income nations. However, even in higher-income countries, SSIs remain the most or second most common nosocomial infections [15]. The most frequent etiologic agents are Staphylococcus aureus (30%) and coagulase-negative staphylococci (12%), which are associated with the skin flora. Other causative agents include the Enterobacteriaceae Escherichia coli, Enterobacter spp, and Klebsiella pneumoniae as well as Enterococcus faecalis and Pseudomonas aeruginosa, which all belong to the group of MDR bacteria referred to as ESKAPE pathogens [14-17]. In addition to the propensity to develop drug resistance, when attached to tissues, these organisms tend to adopt a biofilm mode of growth [18], which usually exacerbates the progression of the wound to a chronic, non-healing state eventually resulting in the sequelae of sepsis [19]. Importantly, biofilm tends to be inherently resistant to current antibiotics [5,20]. We and other investigators have postulated that AMPs possess unique properties that warrant their development as potential countermeasures to combat MDR pathogens.

Properties of Natural AMPs

Most AMPs are short cationic peptides (10-50 amino acids long) with an amphipathic (cationic and hydrophobic domains) structure that are ribosomally synthesized in most life forms, including humans [21-26]. Hence, ubiquitous in nature, AMPs represent the first line of defense against a variety of microbial pathogens (e.g., bacteria, fungi, parasites, viruses) [5,27-33]. AMPs are structurally diverse (α-helix, β-sheets, loop structures; reviewed in detail elsewhere) [32-36], with the amphipathic structure as a consensus motif required for antimicrobial activity [37]. AMPs generally recognize their bacterial targets via electrostatic interactions with negatively charged bacterial membrane lipids [38-43]. For the α-helical AMPs in particular (e.g., the human helical AMP LL37) [4,22,44-46], these membrane interactions are required for induction of the secondary structure, which is random coil in aqueous solutions. Although bacterial killing commonly occurs via membrane perturbation mechanisms (e.g., LL37, magainin), other antimicrobial mechanisms of AMPs have been demonstrated including bacterial cell penetration (e.g., proline-rich AMPs) and interference with vital intracellular processes [47-50]. AMPs may elicit an anti-infective host immune response and possess the ability to neutralize endotoxins, suggesting potential efficacy in septic shock [42,51-54]. Importantly, their anti-biofilm properties may confer efficacy against infections associated with wounds, medical implants, and chronic respiratory illnesses. In contrast to standard antibiotics, AMPs have a number of properties that confer the ability to overcome common resistance mechanisms of MDR pathogens. (1) AMPs generally do not require metabolic processes for antimicrobial activity [55,56]; therefore, they are effective against both quiescent and actively growing bacteria. (2) They display rapid (seconds or minutes) killing kinetics, which would allow limited time for extensive growth and for mutations to occur [57]. (3) In sharp contrast to conventional antibiotics, AMPs demonstrate a low propensity to invoke selection of bacterial resistance [44,57-60]. Despite these unique properties, the clinical developments of AMPs have been largely unsuccessfully.

Limitations of Natural AMPs

Several limitations of natural AMPs have delayed their successful development for clinical use. Natural AMPs display (1) contextual activity with potential inhibition in the presence of acidic pH, saline, divalent cations, and serum or plasma [57,60]; (2) insufficient evidence for systemic efficacy in animal models, which is particularly important in the context of sepsis as a complication of MDR-associated wound infections; (3) potential for susceptibility to protease digestion, which may limit their applications; (4) unclear Pharmacokinetic (PK) properties; (5) potential immunogenicity; (6) potential host toxicity or safety concerns; (7) potential resistance to AMPs, although far less common than resistance to current antibiotics (e.g., by lipid modification toward a reduced density of negative charges) [61,62]. These limitations can be overcome by structural design optimization, as shown by studies of engineered peptides [20,37,57,60].

Structural Optimization of AMPs

Inherently, AMPs are more preventive than therapeutic and are not dedicated antibiotics, whereas antibiotics typically cure diseases. AMPs have evolved to perform multiple functions, which may explain the diversity in amino acid compositions. However, as we (and others) have shown, a diverse amino acid composition is not essential for antimicrobial activity and may even interfere with structural optimization for antibiotic function. Therefore, it is important to approach AMPs differently from the way we approach discoveries of standard antibiotics to optimize their structures based on how they work. In that regard, several studies have already demonstrated that AMPs can be best optimized by reducing the diversity in amino acid composition and focusing mainly on the incorporation of cationic and hydrophobic amino acids, which are more specifically responsible for the amphipathic structure and antimicrobial properties. Such an approach has resulted in optimized AMPs that retain activities in environments of high ionic strength, low pH, and may even work systemically. This is the case of the engineered AMP WLBU2, which demonstrates efficacy in a P. aeruginosa septicemia model when systemically administered. WLBU2 is also very effective against P. aeruginosa when directly delivered to the airway in a murine pneumonia model [63]. WLBU2 is composed of only three amino acids (Arg, Val, and Trp) and designed to fold into an idealized amphipathic helix. In addition, AMPs can be modified to enhance PK properties by D-amino acid substitution [64,65], AMP mimic (e.g., peptoids) [66], cyclization [67], and end-terminus modification [46,68,69].
Anti-Biofilm Properties of AMPS
For SSI, anti-biofilm properties are crucial. When bacteria grow in rich nutrient broth in the laboratory, they grow rapidly (exponentially) in a planktonic mode until the nutrients in the growth medium are largely consumed. However, some bacteria do not grow under such conditions or may adopt an alternate mode of growth called biofilm depending on the growth condition. Bacteria have the capacity to attach to surfaces and secrete an extracellular matrix as a shield against stringent environmental conditions. Similarly, bacteria tend to colonize surfaces of human tissues and adopt a biofilm mode of growth, which is often much less susceptible to antibiotics than bacteria growing in planktonic form. The property to form biofilm enhances the ability of bacteria to “escape” standard treatment, and trauma or post-surgical patients are at high risk of developing these types of infections. Several AMPs display antibiofilm properties, and anti-biofilm prevention and disruption properties can be enhanced in engineered AMPs such as WLBU2 and others [5,20,70]. Appropriate SSI in vivo testing models remain to be developed for further advancement of AMPs for the treatment of SSI and other biofilmrelated infections.

Concluding Remarks

The field of AMPs has been developed for more than three decades, but its success at a clinical level is still negligible. While numerous studies have addressed the limitations of AMPs, they are either scattered or incremental. Based on characteristics of AMPs (especially designed or modified AMPs), they hold a great promise to overcome MDR bacteria-associated SSI. To establish AMPs as a reliable source of effective therapeutics, systematic studies are needed to serve as a reference for multiple groups of investigators dedicated to the continuous development of AMPs.

Conclusion

This research is supported by NIH awards R01 HL-091938, HL- 125128, AI-133351, and a grant (CIA-123062) from Flight Attendant Medical Research Institute.

References

  1. Epand RF, Maloy WL, Ramamoorthy A, Epand RM. Probing the "charge cluster mechanism" in amphipathic helical cationic antimicrobial peptides. Biochemistry. 2010;49(19):4076-84.
  2. Deslouches B, Steckbeck JD, Craigo JK, Doi Y, Mietzner TA, Montelaro RC. Rational design of engineered cationic antimicrobial peptides consisting exclusively of arginine and tryptophan, and their activity against multidrug-resistant pathogens. Antimicrob Agents Chemother. 2013;57(6):2511-21.
  3. Rokitskaya TI, Kolodkin NI, Kotova EA, Antonenko YN. Indolicidin action on membrane permeability: carrier mechanism versus pore formation. Biochim Biophys Acta. 2011;1808(1):91-7.
  4. Hell E, Giske CG, Nelson A, Romling U, Marchini G. Human cathelicidin peptide LL37 inhibits both attachment capability and biofilm formation of Staphylococcus epidermidis. Lett Appl Microbiol. 2010;50(2):211-5.
  5. Melvin JA, Lashua LP, Kiedrowski MR, Yang G, Deslouches B, Montelaro RC, et al. Simultaneous Antibiofilm and Antiviral Activities of an Engineered Antimicrobial Peptide during Virus-Bacterium Coinfection. mSphere. 2016;1(3):e00083-16.
  6. Collignon PC, Conly JM, Andremont A, McEwen SA, Aidara-Kane A; World Health Organization Advisory Group BMoISoAR. World Health Organization Ranking of Antimicrobials According to Their Importance in Human Medicine: A Critical Step for Developing Risk Management Strategies to Control Antimicrobial Resistance from Food Animal Production. Clin Infect Dis. 2016;63(8):1087-93.
  7. Thabet L, Zoghlami A, Boukadida J, Ghanem A, Messadi AA. Comparative study of antibiotic resistance in bacteria isolated from the burned patients during two periods (2005-2008, 2008-2011) and in two hospitals (Hospital Aziza Othmana, trauma and burn center). Tunis Med. 2013;91(2):138-42.
  8. Soto SM. Role of efflux pumps in the antibiotic resistance of bacteria embedded in a biofilm. Virulence. 2013;4(3):223-9.
  9. Munoz-Davila MJ. Role of Old Antibiotics in the Era of Antibiotic Resistance. Highlighted Nitrofurantoin for the Treatment of Lower Urinary Tract Infections. Antibiotics (Basel). 2014;3(1):39-48.
  10. Tanwar J, Das S, Fatima Z, Hameed S. Multidrug resistance: an emerging crisis. Interdiscip Perspect Infect Dis. 2014;2014:541340.
  11. Chandrasekera RM, Lesho EP, Chukwuma U, Cummings JF, Waterman PE. The state of antimicrobial resistance surveillance in the military health system: a review of improvements made in the last 10 years and remaining surveillance gaps. Mil Med. 2015;180(2):145-50.
  12. Lesho EP, Waterman PE, Chukwuma U, McAuliffe K, Neumann C, Julius MD, et al. The antimicrobial resistance monitoring and research (ARMoR) program: the US Department of Defense response to escalating antimicrobial resistance. Clin Infect Dis. 2014;59(3):390-7.
  13. Cappiello F, Di Grazia A, Li-Av SZ, Scali S, Ferrera L, Galietta L, et al. Esculentin-1a-derived peptides promote clearance of P. aeruginosa internalized in cystic fibrosis bronchial cells as well as lung cells migration: Biochemical properties and a plausible mode of action. Antimicrob Agents Chemother. 2016;60(12):7252-62.
  14. Leaper DJ, Edmiston CE. World Health Organization: global guidelines for the prevention of surgical site infection. J Hosp Infect. 2017;95(2):135-6.
  15. World Health Organization. Global Guidelines for the Prevention of Surgical Site Infection. Geneva. 2016.
  16. Sisirak M, Zvizdic A, Hukic M. Methicillin-resistant Staphylococcus aureus (MRSA) as a cause of nosocomial wound infections. Bosn J Basic Med Sci. 2010;10(1):32-7.
  17. Tekin R, Dal T, Bozkurt F, Deveci O, Palanc Y, Arslan E, et al. Risk factors for nosocomial burn wound infection caused by multidrug resistant Acinetobacter baumannii. J Burn Care Res. 2014;35(1):e73-80.
  18. Halstead FD, Thwaite JE, Burt R, Laws TR, Raguse M, Ralf M, et al. Antibacterial Activity of Blue Light against Nosocomial Wound Pathogens Growing Planktonically and as Mature Biofilms. Appl Environ Microbiol. 2016;82(13):4006-16.
  19. Argenta A, Satish L, Gallo P, Liu F, Kathju S. Local Application of Probiotic Bacteria Prophylaxes against Sepsis and Death Resulting from Burn Wound Infection. PloS One. 2016;11(10):e0165294.
  20. Lashua LP, Melvin JA, Deslouches B, Pilewski JM, Montelaro RC, Bomberger JM. Engineered cationic antimicrobial peptide (eCAP) prevents Pseudomonas aeruginosa biofilm growth on airway epithelial cells. J Antimicrob Chemother. 2016;71(8):2200-7.
  21. Wuerth K, Hancock RE. New insights into cathelicidin modulation of adaptive immunity. Eur J Immunol. 2011;41(10):2817-9.
  22. Steinstraesser L, Ring A, Bals R, Steinau HU, Langer S. The human host defense peptide LL37/hCAP accelerates angiogenesis in PEGT/PBT biopolymers. Ann Plast Surg. 2006;56(1):93-8.
  23. McPhee JB, Lewenza S, Hancock RE. Cationic antimicrobial peptides activate a two-component regulatory system, PmrA-PmrB, that regulates resistance to polymyxin B and cationic antimicrobial peptides in Pseudomonas aeruginosa. Mol Microbiol. 2003;50(1):205-17.
  24. Hancock RE. Cationic peptides: effectors in innate immunity and novel antimicrobials. T Lancet Infect Dis. 2001;1(3):156-64.
  25. Scott MG, Yan H, Hancock RE. Biological properties of structurally related alpha-helical cationic antimicrobial peptides. Infect Immun. 1999;67(4):2005-9.
  26. Pirtskhalava M, Gabrielian A, Cruz P, Alekseev V, Rosenthal A, Tartakovsky M, et al. DBAASP v.2: an enhanced database of structure and antimicrobial/cytotoxic activity of natural and synthetic peptides. Nucleic Acids Res. 2016;44(D1):D1104-12.
  27. Boulanger N, Munks RJ, Hamilton JV, Brun R, Lehane MJ, Bulet P, et al. Epithelial innate immunity. A novel antimicrobial peptide with antiparasitic activity in the blood-sucking insect Stomoxys calcitrans. J Biol Chem. 2002;277(51):49921-6.
  28. Hancock RE. Cationic antimicrobial peptides: towards clinical applications. Expert Opin Investig Drugs. 2000;9(8):1723-9.
  29. Hancock RE, Diamond G. The role of cationic antimicrobial peptides in innate host defences. Trends Microbiol. 2000;8(9):402-10.
  30. Kamysz E, Sikorska E, Karafova A, Dawgul M. Synthesis, biological activity and conformational analysis of head-to-tail cyclic analogues of LL37 and histatin 5. J Pept Sci. 2012;18(9):560-6.
  31. Ryu S, Choi SY, Acharya S, Armstrong CA, Song PI, Kim BJ, et al. Antimicrobial and anti-inflammatory effects of Cecropin A(1-8)-Magainin2(1-12) hybrid peptide analog p5 against Malassezia furfur infection in human keratinocytes. The J Invest Dermatol. 2011;131(8):1677-83.
  32. Lourenzoni MR, Namba AM, Caseli L, Degreve L, Zaniquelli ME. Study of the interaction of human defensins with cell membrane models: relationships between structure and biological activity. J Phys Chem B. 2007;111(38):11318-29.
  33. Bauer F, Schweimer K, Kluver E, Rösch P, Adermann K, Sticht H, et al. Structure determination of human and murine beta-defensins reveals structural conservation in the absence of significant sequence similarity. Protein Sci. 2001;10(12):2470-9.
  34. Braff MH, Hawkins MA, Di Nardo A, Dorschner R, Leung DY, Gallo RL, et al. Structure-function relationships among human cathelicidin peptides: dissociation of antimicrobial properties from host immunostimulatory activities. J Immunol. 2005;174(7):4271-78.
  35. Lehrer RI, Ganz T. Antimicrobial peptides in mammalian and insect host defence. Curr Opin Immunol. 1999;11(1):23-7.
  36. van Hoek ML. Antimicrobial peptides in reptiles. Pharmaceuticals (Basel). 2014;7(6):723-53.
  37. Deslouches B, Gonzalez IA, DeAlmeida D, Steele C, Montelaro RC, Mietzner TA, et al. De novo-derived cationic antimicrobial peptide activity in a murine model of Pseudomonas aeruginosa bacteraemia. J Antimicrob Chemother. 2007;60(3):669-72.
  38. Thennarasu S, Tan A, Penumatchu R, Shelburne CE, Heyl DL, Ramamoorthy A. Antimicrobial and membrane disrupting activities of a peptide derived from the human cathelicidin antimicrobial peptide LL37. Biophys J. 2010;98(2):248-57.
  39. Zhang L, Rozek A, Hancock RE. Interaction of cationic antimicrobial peptides with model membranes. J Biol Chem. 2001;276(38):35714-22.
  40. Dean RE, O'Brien LM, Thwaite JE, Fox MA, Atkins H, Ulaeto DO. A carpet-based mechanism for direct antimicrobial peptide activity against vaccinia virus membranes. Peptides. 2010;31(11):1966-72.
  41. Sato H, Feix JB. Peptide-membrane interactions and mechanisms of membrane destruction by amphipathic alpha-helical antimicrobial peptides. Biochim Biophys Acta. 2006;1758(9):1245-56.
  42. Li P, Wohland T, Ho B, Ding JL. Perturbation of Lipopolysaccharide (LPS) Micelles by Sushi 3 (S3) antimicrobial peptide. The importance of an intermolecular disulfide bond in S3 dimer for binding, disruption, and neutralization of LPS. J Biol Chem. 2004;279(48):50150-6.
  43. Hancock RE, Rozek A. Role of membranes in the activities of antimicrobial cationic peptides. FEMS Microbiol Lett. 2002;206(2):143-9.
  44. Deslouches B, Steckbeck JD, Craigo JK, Doi Y, Burns JL, Montelaro RC. Engineered cationic antimicrobial peptides to overcome multidrug resistance by ESKAPE pathogens. Antimicrob Agents Chemother. 2015;59(2):1329-33.
  45. Gustafsson A, Sigel S, Ljunggren L. The antimicrobial peptide LL37 and its truncated derivatives potentiates proinflammatory cytokine induction by lipoteichoic acid in whole blood. Scand J Clin Lab Invest. 2010;70(7):512-8.
  46. Deslouches B, Di YP. Antimicrobial peptides with selective antitumor mechanisms: prospect for anticancer applications. Oncotarget. 2017;8(28):46635-51.
  47. Hale JD, Hancock RE. Alternative mechanisms of action of cationic antimicrobial peptides on bacteria. Expert Rev Anti Infect Ther. 2007;5(6):951-9.
  48. Ostorhazi E, Holub MC, Rozgonyi F, Cassone M, Wade JD, Otvos L Jr. Broad-spectrum antimicrobial efficacy of peptide A3-APO in mouse models of multidrug-resistant wound and lung infections cannot be explained by in vitro activity against the pathogens involved. Int J Antimicrob Agents. 2011;37(5):480-4.
  49. Yang ST, Lee JY, Kim HJ, Shin SY, Hahm KS, Kim JI, et al. Contribution of a central proline in model amphipathic alpha-helical peptides to self-association, interaction with phospholipids, and antimicrobial mode of action. FEBS J. 2006;273(17):4040-54.
  50. Friedrich CL, Rozek A, Patrzykat A, Hancock RE. Structure and mechanism of action of an indolicidin peptide derivative with improved activity against gram-positive bacteria. J Biol Chem. 2001;276(26):24015-22.
  51. Brandenburg K, Garidel P, Fukuoka S, Howe J, Koch MH, Gutsmann T, et al. Molecular basis for endotoxin neutralization by amphipathic peptides derived from the alpha-helical cationic core-region of NK-lysin. Biophys Chem. 2010;150(1-3):80-7.
  52. Lin QP, Zhou LF, Li NN, Li BC, Cai YF, Zhang SQ, et al. Lipopolysaccharide neutralization by the antibacterial peptide CM4. European journal of pharmacology. 2008;596(1-3):160-5.
  53. Ryder MP, Wu X, McKelvey GR, McGuire J, Schilke KF. Binding interactions of bacterial lipopolysaccharide and the cationic amphiphilic peptides polymyxin B and WLBU2. Colloids Surf B Biointerfaces. 2014;120:81-7.
  54. Paranjape SM, Lauer TW, Montelaro RC, Mietzner TA, Vij N. Modulation of proinflammatory activity by the engineered cationic antimicrobial peptide WLBU-2. F1000Res. 2013;2:36.
  55. Deslouches B, Hasek ML, Craigo JK, Steckbeck JD, Montelaro RC. Comparative functional properties of engineered cationic antimicrobial peptides consisting exclusively of tryptophan and either lysine or arginine. J Med Microbiol. 2016;65(6):554-65.
  56. Hicks RP, Abercrombie JJ, Wong RK, Leung KP. Antimicrobial peptides containing unnatural amino acid exhibit potent bactericidal activity against ESKAPE pathogens. Bioorg Med Chem. 2013;21(1):205-14.
  57. Deslouches B, Islam K, Craigo JK, Paranjape SM, Montelaro RC, Mietzner TA. Activity of the de novo engineered antimicrobial peptide WLBU2 against Pseudomonas aeruginosa in human serum and whole blood: implications for systemic applications. Antimicrob Agents Chemother. 2005;49(8):3208-16.
  58. Steckbeck JD, Deslouches B, Montelaro RC. Antimicrobial peptides: new drugs for bad bugs? Expert Opin Biol Ther. 2014;14(1):11-4.
  59. Deslouches B, Steckbeck JD, Craigo JK, Doi Y, Mietzner TA, Montelaro RC. Rational Design of Engineered Cationic Antimicrobial Peptides Consisting Exclusively of Arginine and Tryptophan: WR eCAP Activity against Multidrug-Resistant Pathogens. Antimicrob Agents Chemother. 2013;57(6):2511-21.
  60. Deslouches B, Phadke SM, Lazarevic V, Islam K, Montelaro RC, Mietzner TA, et al. De novo generation of cationic antimicrobial peptides: influence of length and tryptophan substitution on antimicrobial activity. Antimicrob Agents Chemother. 2005;49(1):316-22.
  61. Flamm RK, Rhomberg PR, Simpson KM, Farrell DJ, Sader HS, Jones RN. In vitro spectrum of pexiganan activity when tested against pathogens from diabetic foot infections and with selected resistance mechanisms. Antimicrob Agents Chemother. 2015;59(3):1751-4.
  62. Saar-Dover R, Bitler A, Nezer R. D-alanylation of lipoteichoic acids confers resistance to cationic peptides in group B streptococcus by increasing the cell wall density. PLoS pathogens. 2012;8(9):e1002891.
  63. Chen C, Deslouches B, Montelaro R, Di Y. Enhanced efficacy of the engineered antimicrobial peptide WLBU2 via direct airway delivery in a murine model of P. aeruginosa pneumonia. Clin Microbiol Infect. 2017.
  64. Mangoni ML, Papo N, Saugar JM, Shai Y, Simmaco M, Rivas L, et al. Effect of natural L- to D-amino acid conversion on the organization, membrane binding, and biological function of the antimicrobial peptides bombinins H. Biochemistry. 2006;45(13):4266-76.
  65. Hamamoto K, Kida Y, Zhang Y, Shimizu T, Kuwano K. Antimicrobial activity and stability to proteolysis of small linear cationic peptides with D-amino acid substitutions. Microbiol Immunol. 2002;46(11):741-9.
  66. Mojsoska B, Zuckermann RN, Jenssen H. Structure-activity relationship study of novel peptoids that mimic the structure of antimicrobial peptides. Antimicrob Agents Chemother. 2015;59(7):4112-20.
  67. Junkes C, Harvey RD, Bruce KD, Dolling R, Bagheri M, Dathe M. Cyclic antimicrobial R-, W-rich peptides: the role of peptide structure and E. coli outer and inner membranes in activity and the mode of action. Eur Biophys J. 2011;40(4):515-28.
  68. Mura M, Wang J, Zhou Y, Pinna M, Zvelindovsky AV, Dennison SR, et al. The effect of amidation on the behaviour of antimicrobial peptides. Eur Biophys J. 2016;45(3):195-207.
  69. Dennison SR, Harris F, Bhatt T, Singh J, Phoenix DA. The effect of C-terminal amidation on the efficacy and selectivity of antimicrobial and anticancer peptides. Mol Cell Biochem. 2009;332(1-2):43-50.
  70. De Zoysa GH, Cameron AJ, Hegde VV, Raghothama S, Sarojini V. Antimicrobial peptides with potential for biofilm eradication: synthesis and structure activity relationship studies of battacin peptides. J Med Chem. 2015;58(2):625-39.