Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
ANTIMICROBIAL PEPTIDES AND METHODS OF TREATING GRAM-NEGATIVE PATHOGEN INFECTIONS: POLAR AND NON-POLAR FACE ANALOGS
Document Type and Number:
WIPO Patent Application WO/2018/217880
Kind Code:
A1
Abstract:
Antimicrobial agents, including antimicrobial peptides (AMPs) and uses thereof. Compositions and methods of using dermaseptin-type and piscidin-type antimicrobial peptide variants that demonstrate activity and improved therapeutic indices against microbial pathogens. The peptide compositions demonstrate the ability to not only maintain or improve antimicrobial activity against bacterial pathogens including Gram-negative microorganisms Acinetobacter baumannii and Pseudomonas aeruginosa, but also significantly decrease hemolytic activity against human red blood cells. Specificity determinants within the AMPs change selectivity from broad spectrum antimicrobial activity to Gram-negative selectivity.

Inventors:
HODGES ROBERT S (US)
JIANG ZIQING (US)
GERA LAJOS (US)
MANT COLIN T (US)
Application Number:
PCT/US2018/034114
Publication Date:
November 29, 2018
Filing Date:
May 23, 2018
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
HODGES ROBERT S (US)
JIANG ZIQING (US)
GERA LAJOS (US)
MANT COLIN T (US)
International Classes:
A61K38/16
Domestic Patent References:
WO2010141760A22010-12-09
Other References:
ZIQING JIANG ET AL: "Rational Design of [alpha]-Helical Antimicrobial Peptides to Target Gram-negative Pathogens, Acinetobacter baumannii and Pseudomonas aeruginosa: Utilization of Charge, 'Specificity Determinants,' Total Hydrophobicity, Hydrophobe Type and Location as Design Para", CHEMICAL BIOLOGY & DRUG DESIGN, vol. 77, no. 4, 1 April 2011 (2011-04-01), pages 225 - 240, XP055076608, ISSN: 1747-0277, DOI: 10.1111/j.1747-0285.2011.01086.x
ZIQING JIANG ET AL: "Effects of Hydrophobicity on the Antifungal Activity of [alpha]-Helical Antimicrobial Peptides", CHEMICAL BIOLOGY & DRUG DESIGN, vol. 72, no. 6, 1 December 2008 (2008-12-01), pages 483 - 495, XP055076738, ISSN: 1747-0277, DOI: 10.1111/j.1747-0285.2008.00728.x
YUXIN CHEN ET AL: "Role of Peptide Hydrophobicity in the Mechanism of Action of -alpha--Helical Antimicrobial Peptides", ANTIMICROBIAL AGENTS AND CHEMOTHERAPY, AMERICAN SOCIETY FOR MICROBIOLOGY, US, vol. 51, no. 4, 1 April 2007 (2007-04-01), pages 1398 - 1406, XP008149445, ISSN: 0066-4804, [retrieved on 20061211], DOI: 10.1128/AAC.00925-06
CHEN ET AL: "Rational Design of alpha-Helical Antimicrobial Peptides with Enhanced Activities and SpecificityfTherapeutic Index", JOURNAL OF BIOLOGICAL CHEMISTRY, AMERICAN SOCIETY FOR BIOCHEMISTRY AND MOLECULAR BIOLOGY, US, vol. 280, no. 13, 1 April 2005 (2005-04-01), pages 12316 - 12329, XP008136869, ISSN: 0021-9258, [retrieved on 20050127], DOI: 10.1074/JBC.M413406200
E. W. MARTIN: "Remington's Pharmaceutical Sciences", 1975, MACK PUBLISHING CO.
MERRIFIELD ET AL., ADV. ENZYMOL, vol. 32, 1969, pages 221 - 96
FIELDS ET AL., INT. J. PEPTIDE PROTEIN RES, vol. 35, 1990, pages 161 - 214
W.C. CHAN; P.D. WHITE, FMOC SOLID PHASE PEPTIDE SYNTHESIS
"A Practical Approach (Practical Approach Series", OXFORD UNIVERSITY PRESS
N. LEO BENOITON: "Chemistry of Peptide Synthesis", 2005, CRC PRESS
TSUDA, Y.; OKADA, Y.: "Peptides and Proteins in Organic Chemistry: Building Blocks, Catalysis and Coupling Chemistry", vol. 3, 2010, WILEY-VCH VERLAG GMBH & CO. KGAA, article "Solution-Phase Peptide Synthesis, in Amino Acids"
E. ATHERTON; R.A. SHEPPARD: "Solid-phase peptide synthesis: A practical approach", 1989, OXFORD UNIVERSITY PRESS
SAMBROOK ET AL.: "Molecular Cloning - a laboratory manual", 1989, COLD SPRING HARBOR PRESS
"Methods for Dilution Antimicrobial Susceptibility Tests for Bacteria That Grow Aerobically", APPROVED STANDARD-NINTH EDITION. CLSI DOCUMENT M07-A9, 2012
COAST, J.; SMITH, R.D.; MILLAR, M.R., HEALTH ECONOMICS, vol. 5, 1996, pages 217 - 226
"Science Daily", 9 December 2008, INFECTIOUS DISEASES SOCIETY OF AMERICA
GARZA-GONZALEZ, E.; LLACA-DIAZ, J.M.; BOSQUES-PADILLA, F.J.; GONZALEZ, G.M., CHEMOTHERAPY, vol. 56, 2010, pages 275 - 279
BISWAS, S.; BRUNEI, J.M.; DUBUS, J.C.; ROLAIN, J.M., EXPERT REV. ANTI. INFECT. THER., vol. 10, 2012, pages 917 - 34
YU, Z.; QIN, W.; LIN, J.; FANG, S.; QIN, J., BIOMED RES. INTERNATIONAL, 2015
JENSSEN, H.; HAMILL, P.; HANCOCK, R.E., CLIN MICROBIOL REV., vol. 19, 2006, pages 491 - 511
WADE, D.; BOMAN, A.; WAHLIN, B.; DRAIN, C.M.; ANDREU, D.; BOMAN, H.G.; MERRIFIELD, R.B., PROC. NATL. ACAD. SCI. USA, vol. 87, 1990, pages 4761 - 4765
CRIBBS, D.H.; PIKE, C.J.; WEINSTEIN, S.L.; VELAZQUEZ, P.; COTMAN, C.W., J. BIOL. CHEM., vol. 272, 1997, pages 7431 - 7436
HONG, S.Y.; OH, J.E.; LEE, K.H., BIOCHEM. PHARMACOL., vol. 58, 1999, pages 1775 - 1780
WAKABAYASHI, H.; MATSUMOTO, H.; HASHIMOTO, K.; TERAGUCHI, S.; TAKASE, M.; HAYASAWA, H., ANTIMICROB. AGENTS CHEMOTHER., vol. 43, 1999, pages 1267 - 1269
DE LUCCA, A.J.; BLAND, J.M.; VIGO, C.B.; JACKS, T.J.; PETER, J.; WALSH, T.J., MED. MYCOL., vol. 38, 2000, pages 301 - 308
BLAND, J.M.; DE LUCCA, A.J.; JACKS, T.J.; VIGO, C.B., MOL. CELL. BIOCHEM., vol. 218, 2001, pages 105 - 111
HAMAMOTO, K.; KIDA, Y.; ZHANG, Y.; SHIMIZU, T.; KUWANO, K., MICROBIOL. IMMUNOL., vol. 46, 2002, pages 741 - 749
ELMQUIST, A.; LANGEL, U., BIOL. CHEM., vol. 384, 2003, pages 387 - 393
CHEN, Y.; VASIL, A.I.; REHAUME, L.; MANT, C.T.; BURNS, J.L.; VASIL, M.L.; HANCOCK, R.E.; HODGES, R.S., CHEM. BIOL. DRUG DES., vol. 67, 2006, pages 162 - 173
CHEN, Y.; MANT, C.T.; FARMER, S.W.; HANCOCK, R.E.; VASIL, M.L.; HODGES, R.S., J. BIOL. CHEM., vol. 280, 2005, pages 12316 - 12329
JIANG, Z.; VASIL, A.I.; GERA, L.; VASIL, M.L.; HODGES, R.S., CHEM. BIOL. DRUG DES., vol. 77, 2011, pages 225 - 240
JIANG, Z.; VASIL, A.I.; VASIL, M.L.; HODGES, R.S., PHARMACEUTICALS, vol. 7, 2014, pages 366 - 391
CHEN, Y.; GUARNIERI, M.T.; VASIL, A.I.; VASIL, M.L.; MANT, C.T.; HODGES, R.S., ANTIMICROB. AGENTS CHEMOTHER., vol. 51, 2007, pages 1398 - 1406
JIANG, Z.; KULLBERG, B.J.; VAN DER LEE, H.; VASIL, A.I.; HALE, J.D.; MANT, C.T.; HANCOCK, R.E.W.; VASIL, M.L.; NETEA, M.G.; HODGES, CHEM. BIOL. DRUG DES., vol. 72, 2008, pages 483 - 495
JIANG, Z.; HIGGINS, M.P.; WHITEHURST, J.; KISICH, K.O.; VOSKUIL, M.I.; HODGES, R.S., PROTEIN PEPT. LETT., vol. 18, 2011, pages 241 - 252
JIANG, Z.; GERA, L.; MANT, C.T.; HODGES, R.S.: "Enabling peptide Research from Basic Research to Drug Discovery", 2015, AMERICAN PEPTIDE SOCIETY AND PROPT SCIENTIFIC PUBLISHING, article "Proceedings of the 24th American Peptide Symposium", pages: 245 - 248
EISENBERG, D.; WEISS, R.M.; TERWILLIGER, T.C., NATURE, vol. 299, 1982, pages 371 - 374
KOVACS, J.M.; MANT, C.T.; HODGES, R.S., BIOPOLYMERS, vol. 84, 2006, pages 283 - 297
MANT, C.T.; KOVACS, J.M.; KIM, H.M.; POLLOCK, D.D.; HODGES, R.S., BIOPOLYMERS, vol. 92, 2009, pages 573 - 595
CHEN, Y.; MANT, C.T.; HODGES, R.S., J. PEPTIDE RESEARCH, vol. 59, 2002, pages 18 - 33
ZHOU, N.E.; MANT, C.T.; HODGES, R.S., PEPTIDE RESEARCH, vol. 3, 1990, pages 8 - 20
JIANG, Z.; VASIL, A.I.; HALE, J.D.; HANCOCK, R.E.; VASIL, M.L.; HODGES, R.S., BIOPOLYMERS, vol. 90, 2008, pages 369 - 383
LEE, D.L.; MANT, C.T.; HODGES, R.S., J. BIOL.CHEM., vol. 278, 2003, pages 22918 - 22927
POUNY, Y.; RAPAPORT, D.; MOR, A.; NICOLAS, P.; SHAI, Y., BIOCHEMISTRY, vol. 31, 1992, pages 12416 - 23
Attorney, Agent or Firm:
TRAVER, Robert D. (US)
Download PDF:
Claims:
What is claimed is:

1. An antimicrobial peptide (AMP) comprising the amino acid sequence:

X^L-X^X^L-L-X^X^L-X^X^A-X^X^X^-X11^^12^13^^14^15^^16^17 (SEQ ID NO: 1)

wherein:

each residue is in the D-enantiomeric form;

each of X1 and X15 are independently, amino acids in the D-enantiomeric form selected from Alanine (A; Ala), and Lysine (K; Lys);

each of X2, X3, X4, X6, X7, X9, X10, X12, X14, X16, and X17 are independently, amino acids in the D-enantiomeric form selected from Serine (S; Ser), and Lysine (K; Lys);

each of X5 and X13 are independently, amino acids in the D-enantiomeric form selected from Lysine (K; Lys), Serine (S; Ser) and Threonine (T; Thr);

each of X8 and X11 are independently, amino acids in the D-enantiomeric form selected from Lysine (K; Lys), Arginine (R; Arg), Ornithine (O; Orn) and Diaminobutyric acid (Dbu), and Diaminopropionic acid (Dpr);

wherein the AMP comprises two positively charged residues on the non-polar face and has 8 or 9 positively charged residues on the polar face, has a total charge of +10 or +11; and,

wherein the AMP does not comprise the amino acid sequence

KLK SLLKTL SK AKKKKLKTLLK AL SK (SEQ ID NO:2).

2. An AMP of claim 1 wherein the AMP comprises the amino acid sequence

X^KKLLKKLSSA -X8-SS-XU LSTLLKKLKK (SEQ ID NO: 12)

wherein X1, X8 and X11 are as defined in Claim 1.

3. An AMP of claim 1 wherein the AMP comprises the amino acid sequence

X^KKLLKTLSSA -X8-SS-XU LSKLLKKLKK (SEQ ID NO:27)

wherein X1, X8 and X11 are as defined in Claim 1.

4. An AMP of claim 1 wherein the AMP comprises the amino acid sequence

X^SSLLSKLKKA-X^KK-X11 LKKLLKALSS (SEQ ID NO:30) wherein X1, X8 and X11 are as defined in Claim 1.

5. An AMP of claim 1 wherein the AMP comprises the amino acid sequence

X^KKLLKKLKSA-X^SS-X11 LSTLLSKLKK (SEQ ID NO:33)

wherein X1, X8 and X11 are as defined in Claim 1.

6. An AMP of claim 1 wherein the AMP comprises the amino acid sequence

X^KKLLKKLSSA-X^SS-X11 LSSLLKKLKK (SEQ ID NO:36)

wherein X1, X8 and X11 are as defined in Claim 1.

7. An AMP of claim 1 wherein the AMP comprises the amino acid sequence

X^KSLLKSLSKA-X^KK-X11 LKSLLKALSK (SEQ ID NO:39)

wherein X1, X8 and X11 are as defined in Claim 1.

8. An AMP of claim 1 wherein the AMP comprises the amino acid sequence

ALKSLLKKLSKA-X8-KK-XU LKKLLKALSS (SEQ ID NO:42)

wherein X8 and X11 are as defined in Claim 1.

9. An AMP of claim 1, wherein the AMP comprises an amino acid sequence selected from the group consisting of:

AL S S L L S KL KKAK KKKLKKL L KAL S S 10

ALKKLLKKLKSAKSSKLSTLLSKLKK 11

KLKKLLKKLSSA (Orn) SS (Orn) LSTLLKKLKK 14

KLKKLLKKLSSA (Dbu) SS (Dbu) LSTLLKKLKK 15

KLKKLLKKLSSA (Dpr) SS (Dpr) LSTLLKKLKK 16

KLKKLLKKLSSA (Arg) SS (Arg) LSTLLKKLKK 17

ALKKLLKKLSSA (Orn) SS (Orn) LSTLLKKLKK 19

ALKKLLKKLSSA (Dbu) SS (Dbu) LSTLLKKLKK 20

ALKKLLKKLSSA (Dpr) SS (Dpr) LSTLLKKLKK 21

ALKKLLKKLSSA (Arg) SS (Arg) LSTLLKKLKK 22

AL KS L L KKL S KAK KKKLKKL L KAL S S 23

KLKKLLKKLSSAKSSKLSSLLKKLKK 24

ALKKLLKKLSSAKSSKLSSLLKKLKK 25

KLKSLLKSLSKAKKKKLKSLLKALSK 26

10. The AMP of Claim 1, wherein the amino acid sequence of the AMP comprises a sequence selected from the group consisting of SEQ ID NOs: 3, 5, 6, 8, 10, 14-16, 19-21 and 23-26.

11. The AMP of any one of claims 1-10, wherein the AMP inhibits propagation of a prokaryote.

12. The AMP of Claim 11, wherein the prokaryote is a Gram-negative bacterium.

13. The AMP of Claim 12, wherein the Gram-negative bacterium is at least one of A. baumannii and P. aeruginosa.

14. The AMPs of any one of claims 1-13, wherein the therapeutic index (calculated by the ratio of hemolytic activity and antimicrobial activity (MIC) is at least 50.

15. The AMP of any one of claims 1-13, wherein the AMP exhibits greater antimicrobial activity against Gram-negative P. aeruginosa or Acinetobacter baumannii drug-resistant mutants compared to other AMPs.

16. The AMP of Claim 1, wherein the AMP exhibits at least a 10-fold increased selectivity for prokaryotic cells over eukaryotic cells.

17. The AMP of Claim 1, wherein the AMP exhibits at least a 10-fold increased selectivity for Gram-negative bacteria over Gram-positive bacteria.

18. The AMP of Claim 17, wherein the Gram-negative bacteria may be Acinetobacter baumannii, and the Gram-positive bacteria may be Staphylococcus aureus.

19. The AMP of any one of claims 1-13, wherein the AMP is equally effective in inhibiting the propagation of an antibiotic-resistant prokaryote and an antibiotic-sensitive prokaryote.

20. A pharmaceutical composition comprising at least one peptide of any one of claims 1- 13, and a pharmaceutically acceptable carrier.

21. The pharmaceutical composition of claim 20, comprising a mono-phasic

pharmaceutical composition suitable for parenteral or oral administration consisting essentially of a therapeutically-effective amount of at least one peptide of claim 1, and a pharmaceutically acceptable carrier.

22. A method of preventing or treating a microbial infection comprising administering to a subject in need thereof a therapeutically effective amount of at least one peptide of any one of claims 1-13, or a pharmaceutical composition of claim 20.

23. The method of claim 22, wherein the microbial infection is a bacterial infection.

24. The method of claim 23, wherein the bacterial infection is a Gram-negative bacterial infection.

25. The method of claim 23, wherein the bacterial infection is an antibiotic resistant bacterial infection.

26. The method of claim 23, wherein an infecting microorganism is at least one of Acinetobacter baumannii and Pseudomonas aeruginosa.

27. The method of claim 23, wherein an infecting microorganism is multi-drug resistant Pseudomonas aeruginosa or Acinetobacter baumannii. 28. The method of claim 22, wherein the administration of the peptide or pharmaceutical composition is by an administration route selected from oral, topical, intravenous, intraperitoneal, intramuscular, intradermal, intrasternal, intraarticular injection, intrathecal, and infusion. 29. The method of claim 22, wherein the peptide or pharmaceutical composition is administered in conjunction with one or more additional antimicrobial agents.

30. A method of preventing a microbial infection in an individual at risk of developing an infection comprising administering an effective amount of at least one peptide of any one of claims 1-13, or a pharmaceutical composition of claim 20 to the individual in need thereof.

31. The method of claim 30, wherein the individual is a surgical patient. 32. The method of claim 30, wherein the individual is a hospitalized patient.

33. A method of combating a bacterial infection in a patient, comprising applying at least one peptide of any one of claims 1-13, or a pharmaceutical composition of claim 20 to a body surface of the patient. 34. The method of claim 33, wherein the body surface is a wound.

35. The method of claim 33, wherein the composition is applied following an operation or surgery. 36. At least one peptide of any one of claims 1-13, or a pharmaceutical composition of claim 20 for use in the treatment of a microbial infection.

37. Use of at least one peptide of any one of claims 1-13, or a pharmaceutical composition of claim 20 in the manufacture of a medicament for the prevention or treatment of a microbial infection.

38. The AMP of claim 1, wherein the AMP has one or more improved biological properties relative to Polymyxin B or Polymyxin E, wherein said one or more improved biological properties are selected from improved antimicrobial activity against a Gram- negative microorganism, decreased hemolysis of human red blood cells, decreased binding to human serum proteins, and improved therapeutic index for a Gram-negative

microorganism.

39. A pharmaceutical composition of claim 20 in combination with one or more further therapeutic agents.

40. A pharmaceutical composition according to claim 39 wherein the one or more further therapeutic agents are each independently selected from antibiotics.

41. A peptide of any one of claims 1-13 for use as a pharmaceutical.

Description:
ANTIMICROBIAL PEPTIDES AND METHODS OF TREATING GRAM- NEGATIVE PATHOGEN INFECTIONS: POLAR AND NON-POLAR FACE

ANALOGS FIELD OF THE INVENTION

This disclosure relates to the field of antimicrobial peptides (AMPs) and treatments for microbial infections.

BACKGROUND

The explosion of bacterial resistance to traditional antibiotics and a rapid increase in the incidence of multi-drug resistant microbes have created an urgency to develop new classes of antimicrobial agents. There are now "Superbugs" resistant to most or all antibiotics 1 . The Infectious Diseases Society of America has reported that two-thirds of all health care associated infections are caused by six multi-drug resistant organisms referred to as "ESKAPE" pathogens consisting of two Gram-positive organisms, Enterococcus faecium and Staphylococcus aureus, and four Gram-negative organisms, Klebsiella pneumonia, Acinetobacter baumannii, Pseudomonas aeruginosa and Enterobacter species 2 . A recent study in Mexico demonstrated dramatic increases in the incidence of antibiotic-resistant species 3 . Of 550 clinical isolates of baumannii and 250 clinical isolates of P. aeruginosa, 74% of A. baumannii, and 34% of P. aeruginosa were multi- drug resistant.

Polymyxin B and Polymyxin E (Colistin) are cationic peptides consisting of a cyclic heptapeptide with a tripeptide side chain acylated by a fatty acid chain at the amino terminus. These antibiotics were heavily used in the 1960s, but in the 1970s their clinical use was limited due to serious issues of nephrotoxicity and neurotoxicity 4 5 . The revival of these two peptides began in the mid-1990s, due to the lack of novel antibiotics effective against the increasingly-prevalent multi-drug resistant Gram-negative bacteria. Thus, these compounds have become antibiotics of last resort, needed for drug resistant bacteria but associated with a high incidence of toxicity. Resistance to these polymyxins could become a major global health challenge because virtually no new antibiotics are currently available for treating serious Gram-negative infections caused by polymyxin-resistant "superbugs." Accordingly, there is a great need for additional therapeutic antimicrobial treatments effective against drug-resistant organisms. SUMMARY

Antimicrobial peptides (AMPs) are produced by bacteria, fungi, plants, insects, amphibians, crustaceans, fish and mammals, including humans, either constitutively or in response to the presence of a microbe 6 . AMPs are rapidly bactericidal and generally have broad-spectrum activity. It is believed that the antimicrobial mechanism of action of cationic AMPs does not involve a stereoselective interaction with a chiral enzyme or lipid or protein since enantiomeric forms of AMPs with all-D-amino acids have shown equal activities compared to their all-L-enantiomers 7"15 . Because their mode of action apparently involves non-specific interactions with the cytoplasmic membrane of bacteria, bacteria rarely develop resistance to them. Additionally, all D-enantiomer peptides are resistant to proteolytic enzyme degradation, which enhances their potential use as therapeutic agents in mammals. Unfortunately, native AMPs lack specificity between prokaryotic and eukaryotic cells, and are therefore too toxic to be used for systemic treatment of bacterial infections. This toxicity, which manifests as drug- and dose-limiting hemolysis of human red blood cells, has limited the development of a new class of antimicrobial agents based on these AMPs.

The present inventors have previously used an antimicrobial peptide in the D- enantiomeric configuration with one lysine substitution ("Dl (K13)") as a starting point to design antimicrobial peptides with enhanced biologic properties for Gram-negative pathogens only, rather than broad-spectrum activity 17 . The number and location of positively charged residues on the polar and non-polar face of this AMP were studied, ultimately resulting in the development of four new antimicrobial peptides with improvements in antimicrobial activity against Gram-negative pathogens and dramatic reductions in hemolytic activity and therefore unprecedented improvements in therapeutic indices.

The inventors have also studied the antimicrobial peptides piscidin 1 and dermaseptin S4 for substitution of one or two amino acid(s) to lysine(s) at different positions in the center of their nonpolar faces to investigate the generality of the

"specificity determinant" design concept to enhance or maintain antimicrobial activity and significantly improve the therapeutic index 18 .

The inventors also prepared variants in two native AMPs Piscidin 1 (isolated from mast cells of hybrid striped bass - Morone saxatilis male x Morone chrysops female) and dermaseptin S4 (isolated from the skin of tree-dwelling, South American frogs of the Phyllomedusa species) 22 . These variant peptides were tested for their antimicrobial activity against two different pathogens: 11 and 20 diverse clinical isolates of baumannii, and Staphylococcus aureus (12 Methicillin-sensitive S. aureus strains and 8

Methicillin/Oxacillin-resistant S. aureus strains), respectively. These studies showed that substitution of "specificity determinant(s)" in broad spectrum AMPs, encode selectivity for Gram-negative pathogens and simultaneously remove both Gram-positive activity and hemolytic activity of these two, diverse amphipathic alpha-helical AMPs which differ dramatically in amino acid composition, net positive charge and amphipathicity.

This disclosure provides highly effective and specific antimicrobial agents comprising peptides and peptide-containing compositions, and methods of inhibiting microorganisms, and treating a subject in need of antimicrobial therapy.

The antimicrobial peptides (AMPs) and compositions of this disclosure

demonstrate activity and improved therapeutic indices against bacterial pathogens. These AMPs demonstrate the ability to not only maintain or improve antimicrobial activity against Gram-negative bacterial pathogens, but also significantly decrease the hemolysis of mammalian red blood cells. Thus, improved therapeutic indices are achieved by the AMPs of this disclosure.

To overcome the significant mammalian toxicity of most of the known AMPs, the inventors developed the design concept of the "specificity determinant," which refers to the substitution of positively charged amino acid residue(s) in the non-polar face of amphipathic alpha-helical or cyclic beta-sheet antimicrobial peptides to create selectivity between eukaryotic and prokaryotic membranes; that is, antimicrobial activity is maintained and hemolytic activity or cell toxicity to mammalian cells is substantially decreased or eliminated.

The inventors selected piscidin 1 and dermaseptin S4 as examples of native AMPs to substitute positively charged amino acid(s) at different positions in their non-polar faces to enhance or maintain antimicrobial activity and significantly improve the therapeutic index.

This disclosure provides antimicrobial peptides and antimicrobial peptide compositions, as well as methods of inhibiting microorganisms and treating microbial infections, particularly infections by drug-resistant microorganisms. In an aspect of the claimed methods, a subject is treated by administering an AMP or a composition comprising an AMP of this disclosure. The antimicrobial peptides (AMPs) of this disclosure demonstrate activity and improved therapeutic indices against bacterial pathogens. These AMPs may demonstrate the ability to not only maintain or improve antimicrobial activity against bacterial pathogens, including Gram-negative

microorganisms such as Acinetobacter baumannii and Pseudomonas aeruginosa, but also significantly decrease hemolytic activity against human red blood cells. Thus, the AMPs of this disclosure display improved therapeutic indices. Isolated antimicrobial peptides (AMPs) of this disclosure comprise the amino acid sequence (referring to the single-letter amino acid code) of:

X 1 -L-X 2 -X 3 -L-L-X 4 -X 5 -L-X 6 -X 7 -A-X 8 -X 9 -X 10 -X 11 -L-X 12 -X 13 -L-L-X 14 -X 15 -L-X 16 -X 17

(SEQ ID NCv l)

Wherein:

each residue is in the D-enantiomeric form;

each of X 1 and X 15 are independently, amino acids in the D-enantiomeric form selected from Alanine (A; Ala), and Lysine (K; Lys);

each of X 2 , X 3 , X 4 , X 6 , X 7 , X 9 , X 10 , X 12 , X 14 , X 16 , and X 17 are independently, amino acids in the D-enantiomeric form selected from Serine (S; Ser), and Lysine (K; Lys);

each of X 5 and X 13 are independently, amino acids in the D-enantiomeric form selected from Lysine (K; Lys), Threonine (T; Thr), and Serine (S, Ser);

each of X 8 and X 11 are independently, amino acids in the D-enantiomeric form selected from Lysine (K; Lys), Arginine (R; Arg), Ornithine (O; Orn) and Diaminobutyric acid (Dbu), and Diaminopropionic acid (Dpr); and,

wherein the AMPs of this disclosure do not comprise the amino acid sequence KLK SLLKTL SK AKKKKLKTLLK AL SK (SEQ ID NO:2).

The total charge on these molecules is +10 or +11. The total charge on these molecules without specificity determinants is +8 or +9. This may comprise two positively charged residues on the non-polar face ("specificity determinants") and 8 or 9 positively- charged residues on the polar face. The peptides of this disclosure may include residues that disrupt the continuous hydrophobic surface that stabilizes the alpha-helical structure of AMPs that lack the "specificity determinants" (such as the naturally occurring peptides Piscidin 1 and/or Dermaseptin S4, and/or the all D -enantiomeric forms of these naturally occurring peptides).

In one embodiment, the peptides of this disclosure have dramatically reduced a- helical structure in aqueous environment but inducible a-helical structure when the peptides are in a hydrophobic environment to mimic the hydrophobicity of membrane.

In another embodiment, the peptides of this disclosure may include residues that reduce the hydrophobicity on the non-polar face and overall hydrophobicity of the peptide molecule (as measured by retention time at 25°C by reversed-phase chromatography (RP- HPLC).

In another embodiment, the peptides of this disclosure may include residues that dramatically reduce peptide self-association in aqueous conditions

In another embodiment, the peptides of this disclosure may have dramatically reduced toxicity to normal cells (as measured by hemolytic activity to human red blood cells at 37°C after 1 hours). In this context, HCso hemolytic activity is defined as the concentration of peptide (μΜ) that results in 50% hemolysis after 1 hour at 37°C. A skilled person will appreciate that it is advantageous that this value is as large as possible. In particular, hemolytic activity may be measured by the procedure described in the

Examples section of this disclosure, particularly Example 3. In a further embodiment, the peptides of this disclosure exhibit hemolytic activity expressed as HCso value of greater than 500 μg/ml, in particularly 1000 μg/ml or greater, more particularly greater than 2000 μg/ml.

In another embodiment, the peptides of this disclosure may have similar or substantially enhanced antimicrobial activity (compared to AMPs lacking specificity determinants, such as the naturally occurring peptides Piscidin 1 and/or Dermaseptin S4, and/or the all D-enantiomeric forms of these naturally occurring peptides), and particularly with respect to bactericidal activity towards Gram-negative microbes. Antimicrobial activity is expressed as the MIC (the minimum concentration of peptide required to inhibit growth of bacteria after 16-20 hours at 37°C). In particular, antimicrobial activity may be measured by the procedure described in the Examples section of this disclosure, particularly Example 2.

In another embodiment of the invention, the peptides of this disclosure may have dramatically improved therapeutic indices (calculated by the ratio of hemolytic activity and antimicrobial activity (MIC)) compared to AMPs lacking specificity determinants, such as the naturally occurring peptides Piscidin 1 and/or Dermaseptin S4, and/or the all D-enantiomeric forms of these naturally occurring peptides. The ratio of hemolytic activity/antimicrobial activity defines the therapeutic index for a given AMP and is a measure of specificity of the AMP for bacterial membranes. A skilled person will appreciate that typically the higher the therapeutic index, the more specific the AMP is for prokaryotic cells. In a further embodiment, the peptides of this disclosure exhibit therapeutic index values (expressed as HCso/MIC) greater than 500, in particular greater than 1000. A skilled person will appreciate that the therapeutic index is a ratio and the units of measurement of HCso and MIC are required to be consistent.

In another embodiment, the peptides of this disclosure may have antimicrobial selectivity for Gram-negative pathogens resulting from significantly decreased Gram- positive activity and hemolytic activity (compared to AMPs lacking specificity

determinants, such as the naturally-occurring peptides Piscidin 1 and/or Dermaseptin S4, and/or the all D-enantiomeric forms of these naturally occurring peptides). The peptides of this disclosure may have antimicrobial activity against baumannii bacterial strains resistant to Polymyxin B and/or Polymyxin E (Colistin) antibiotics. The peptides of this disclosure may discriminate between eukaryotic and prokaryotic cell membranes. The peptides of this disclosure may have antimicrobial activity even in the presence of human serum.

In aspects of this disclosure, the claimed antimicrobial peptides comprise the peptides of Table 1 A (each of which comprises the listed amino acids, set forth in the one- letter amino acid code, all in the D-enantiomeric form):

Table 1A

A series of peptides (shown in Table IB) was designed to show the effects of substitutions to the specificity determinants at positions 13 and 16 of SEQ ID: 3. The lysine residues at these positions were substituted with Ornithine (Orn), Diaminobutryic acid (Dbu), Diaminopropionic acid (Dpr) or Arginine (Arg). In Table IB, the peptide sequences are shown using the one-letter code (or three-letter code for Orn, Dbu, Dpr and Arg). Positions 13 and 16 are in the center of the non-polar face. Table IB.

Similarly, a series of peptides (shown in Table 1C) was designed to show the effects of substitutions to the specificity determinants at positions 13 and 16 of the SEQ ID:8. The lysine residues at these positions were substituted with Ornithine (Orn), Diaminobutryic acid (Dbu), Diaminopropionic acid (Dpr) or Arginine (Arg). In Table 1C, the peptide sequences are shown using the one-letter code (or three-letter code for Orn, Dbu, Dpr and Arg). Positions 13 and 16 are in the center of the non-polar face.

Table 1C.

Length Sequence SEQ (mer) ID

NO

13 16

26 18 ALKKLLKKLSSA -X 8 - SS - X 11 LSTLLKKLKK

26 ALKKLLKKLSSA ( Lys ) SS (Lys ) LSTLLKKLKK 8

26 ALKKLLKKLSSA (Orn) SS (Orn) LSTLLKKLKK 19

26 ALKKLLKKLSSA (Dbu) SS (Dbu) LSTLLKKLKK 20

26 ALKKLLKKLSSA (Dpr) SS (Dpr) LSTLLKKLKK 21

26 ALKKLLKKLSSA (Arg) SS (Arg) LSTLLKKLKK 22 In one embodiment, the peptides of this disclosure comprise the amino acid sequence of:

X^KKLLKKLSSA -X 8 -SS-X U LSTLLKKLKK (SEQ ID NO: 12)

wherein X 1 , X 8 and X 11 are as defined hereinabove with respect to SEQ ID NO: 1

In another embodiment, the peptides of this disclosure comprise the amino acid sequence of:

KLKKLLKKLS S A -X 8 -SS-X U LSTLLKKLKK (SEQ ID NO: 13)

wherein X 8 and X 11 are as defined hereinabove with respect to SEQ ID NO: 1

In another embodiment, the peptides of this disclosure comprise the amino acid sequence of:

ALKKLLKKLSSA -X 8 -SS-X U LSTLLKKLKK (SEQ ID NO: 18)

wherein X 8 and X 11 are as defined hereinabove with respect to SEQ ID NO: 1

In another embodiment, the peptides of this disclosure comprise the amino acid sequence of:

X^KKLLKTLSSA -X 8 -SS-X U LSKLLKKLKK (SEQ ID NO:27)

wherein X 1 , X 8 and X 11 are as defined hereinabove with respect to SEQ ID NO: 1

In another embodiment, the peptides of this disclosure comprise the amino acid sequence of:

KLKKLLKTL S S A -X 8 -SS-X U LSKLLKKLKK (SEQ ID NO:28)

wherein X 8 and X 11 are as defined hereinabove with respect to SEQ ID NO: 1

In another embodiment, the peptides of this disclosure comprise the amino acid sequence of:

ALKKLLKTLSSA -X 8 -SS-X U LSKLLKKLKK (SEQ ID NO:29)

wherein X 8 and X 11 are as defined hereinabove with respect to SEQ ID NO: 1

In another embodiment, the peptides of this disclosure comprise the amino acid sequence of: X^SSLLSKLKKA-X^KK-X 11 LKKLLKALSS (SEQ ID NO:30)

wherein X 1 , X 8 and X 11 are as defined hereinabove with respect to SEQ ID NO: 1

In another embodiment, the peptides of this disclosure comprise the amino acid sequence of:

KL S SLL SKLKK A-X 8 -KK-X 11 LKKLLKALSS (SEQ ID NO:31)

wherein X 8 and X 11 are as defined hereinabove with respect to SEQ ID NO: 1

In another embodiment, the peptides of this disclosure comprise the amino acid sequence of:

AL S SLL SKLKK A-X 8 -KK-X 11 LKKLLKALSS (SEQ ID NO:32)

wherein X 8 and X 11 are as defined hereinabove with respect to SEQ ID NO: 1

In another embodiment, the peptides of this disclosure comprise the amino acid sequence of:

X^KKLLKKLKSA-X^SS-X 11 LSTLL SKLKK (SEQ ID NO:33)

wherein X 1 , X 8 and X 11 are as defined hereinabove with respect to SEQ ID NO: 1

In another embodiment, the peptides of this disclosure comprise the amino acid sequence of:

KLKKLLKKLKSA-X 8 -SS-X U LSTLL SKLKK (SEQ ID NO:34)

wherein X 8 and X 11 are as defined hereinabove with respect to SEQ ID NO: 1

In another embodiment, the peptides of this disclosure comprise the amino acid sequence of:

ALKKLLKKLKSA-X 8 -SS-X U LSTLL SKLKK (SEQ ID NO:35)

wherein X 8 and X 11 are as defined hereinabove with respect to SEQ ID NO: 1

In another embodiment, the peptides of this disclosure comprise the amino acid sequence of:

X^KKLLKKLSSA-X^SS-X 11 LSSLLKKLKK (SEQ ID NO:36)

wherein X 1 , X 8 and X 11 are as defined hereinabove with respect to SEQ ID NO: 1 In another embodiment, the peptides of this disclosure comprise the amino acid sequence of:

KLKKLLKKLSSA-X 8 -SS-X U LSSLLKKLKK (SEQ ID NO:37)

wherein X 8 and X 11 are as defined hereinabove with respect to SEQ ID NO: 1

In another embodiment, the peptides of this disclosure comprise the amino acid sequence of:

ALKKLLKKLSSA-X 8 -SS-X U LSSLLKKLKK (SEQ ID NO:38)

wherein X 8 and X 11 are as defined hereinabove with respect to SEQ ID NO: 1

In another embodiment, the peptides of this disclosure comprise the amino acid sequence of:

X^KSLLKSLSKA-X^KK-X 11 LKSLLKALSK (SEQ ID NO:39)

wherein X 1 , X 8 and X 11 are as defined hereinabove with respect to SEQ ID NO: 1

In another embodiment, the peptides of this disclosure comprise the amino acid sequence of:

KLK SLLK SL SK A-X 8 -KK-X 11 LKSLLKALSK (SEQ ID NO:40)

wherein X 8 and X 11 are as defined hereinabove with respect to SEQ ID NO: 1

In another embodiment, the peptides of this disclosure comprise the amino acid sequence of:

ALK SLLK SL SK A-X 8 -KK-X 11 LKSLLKALSK (SEQ ID NO:41)

wherein X 8 and X 11 are as defined hereinabove with respect to SEQ ID NO: 1

In another embodiment, the peptides of this disclosure comprise the amino acid sequence of:

ALKSLLKKLSKA-X 8 -KK-X U LKKLLKALSS (SEQ ID NO:42)

wherein X 8 and X 11 are as defined hereinabove with respect to SEQ ID NO: 1 In another embodiment, the peptides of this disclosure comprise the amino acid sequence of:

X^KSLLKTLSKA-X^KK-X 11 LKTLLKALSK (SEQ ID NO:43)

wherein X 1 , X 8 and X 11 are as defined hereinabove with respect to SEQ ID NO: 1, provided that when X 1 is Lysine, X 8 and X 11 are not both Lysine.

In another embodiment, the peptides of this disclosure comprise the amino acid sequence of:

KLKSLLKTLSKA-X 8 -KK-X U LKTLLKALSK (SEQ ID NO:44)

wherein X 1 , X 8 and X 11 are as defined hereinabove with respect to SEQ ID NO: 1, provided that X 8 and X 11 are not both Lysine

In another embodiment, the peptides of this disclosure comprise the amino acid sequence of:

ALKSLLKTLSKA-X 8 -KK-X U LKTLLKALSK (SEQ ID NO:45)

wherein X 8 and X 11 are as defined hereinabove with respect to SEQ ID NO: 1.

In a further embodiment, X 8 and X 11 are not both Lysine.

In one embodiment, the amino acid sequence of the AMP comprises a sequence selected from the group consisting of SEQ ID NOs: 3-11, 14-17 and 19-26, in particular SEQ ID NOs:3, 5, 6, 8, 10, 14-16, 19-21 and 23-26.

Another aspect of this disclosure provides pharmaceutical compositions comprising at least one of the antimicrobial peptides of this disclosure, and a

pharmaceutically acceptable carrier. In aspects of the claimed pharmaceutical

compositions, the compositions may include one or more AMPs having the amino acid sequence of SEQ ID NOs: 3-11, 14-17 or 19-26.

Another aspect provides a method of preventing or treating an infection in a subject, including administering a therapeutically effective amount of a composition to the subject, wherein the composition comprises at least one antimicrobial peptide of this disclosure, and a pharmaceutically acceptable carrier. In aspects of the claimed methods the infecting microorganism is a Gram-negative bacteria.

In these methods, the infecting microorganism may be an antibiotic resistant microbe. The antibiotic resistant microbe may be a Gram-negative, antibiotic-resistant Acinetobacter baumannii or Pseudomonas aeruginosa pathogen. Alternatively or additionally, the antibiotic infecting microorganism may be a drug-resistant (such as a Polymyxin B and/or Polymyxin E (Colistin)-resistant) Gram-negative pathogen, or a Polymyxin B and/or Polymyxin E sensitive Gram-negative pathogen.

This disclosure also provides methods of inhibiting a microorganism, comprising contacting the microorganism with a composition comprising at least one AMP of this disclosure. In these methods, the AMP may be one or more of the peptides having the amino acid sequence of SEQ ID NOs:3-l 1, 14-17 or 19-26.

One aspect of this disclosure provides an antimicrobial peptide (AMP) comprising an amino acid sequence having at least 85%, or at least 90% or at least 95% homology with a peptide selected from the group consisting of SEQ ID NOs:3-l 1, 14-17 or 19-26 or functional analogues, derivatives or fragments thereof, or pharmaceutically-acceptable salts thereof.

In a preferred aspect of the claimed methods, the amino acid sequence of the administered AMP comprises the sequence of SEQ ID NOs:3-l 1, 14-17 or 19-26. In these methods, the AMP inhibits propagation of a prokaryote. The prokaryote may be a Gram- negative bacterium, which may include at least one of A. baumannii and P. aeruginosa bacterium.

In one embodiment, the AMPs of this disclosure may exhibit a therapeutic index for baumannii (calculated by the ratio of hemolytic activity to antimicrobial activity (MIC)) of at least 85-fold.

In a further embodiment, the AMPs of this disclosure may exhibit at least a 10-fold increased selectivity for Gram-negative bacteria over Gram-positive bacteria. The AMP may exhibit between a 10-fold and a 90-fold increase in selectivity for Gram-negative bacteria over Gram-positive bacteria. In these selectivity measurements, the Gram- negative bacteria may be A. baumannii and the Gram -positive bacteria may be

Staphylococcus aureus. In another embodiment, the AMPs of this disclosure having the sequence of any one of SEQ ID Nos:3-l 1, 14-17 or 19-26 may exhibit at least a 10-fold decrease in hemolysis of human red blood cells (measured as HCso) - compared to hemolysis exhibited by Polymyxin B or Polymyxin E.

Another aspect of this disclosure provides a pharmaceutical composition comprising at least one AMP of this disclosure and a pharmaceutically acceptable carrier. The pharmaceutical composition may be a mono-phasic pharmaceutical composition suitable for parenteral or oral administration consisting essentially of a therapeutically- effective amount of at least one AMP of this disclosure, and a pharmaceutically acceptable carrier. In these embodiments, the AMP may be one or more of the peptides having the sequence of SEQ ID NOs:3-l 1, 14-17 or 19-26.

Another aspect of this disclosure provides methods of preventing or treating a microbial infection comprising administering to a subject in need thereof a therapeutically effective amount of at least one AMP of this disclosure, or a pharmaceutical composition comprising the same. In these methods, the AMP administered may be one or more of the peptides having the sequence of SEQ ID NOs: 3-1 1, 14-17 or 19-26. In these methods, the microbial infection may be the result of an infecting bacteria, fungi, virus, or protozoa. The microbial infection may be a bacterial infection. The bacterial infection may be a Gram-negative bacterial infection. The bacterial infection may be an antibiotic resistant bacterial infection. The infecting microorganism may be at least one of Pseudomonas aeruginosa, Acinetobacter baumannii. The infecting microorganism may be an antibiotic- or multi drug-resistant Pseudomonas aeruginosa or Acinetobacter baumannii bacteria.

In these methods, the administration of the peptide or pharmaceutical composition may be made by an administration route selected from oral, topical, intravenous, intraperitoneal, intramuscular, intradermal, intrasternal, intraarticular injection, intrathecal. The peptides or pharmaceutical compositions of this disclosure may be administered in conjunction with one or more additional antimicrobial agents.

This disclosure also provides methods of preventing a microbial infection in an individual at risk of developing an infection comprising administering an effective amount of at least one AMP of this disclosure, or a pharmaceutical composition comprising the same, to an individual in need thereof. The individual may be a surgical patient. The individual may be a hospitalized patient. This disclosure also provides methods of combating a bacterial infection in a patient comprising applying at least one AMP of this disclosure, or a pharmaceutical composition comprising the same, to a body surface of the patient. The body surface may be a wound. The composition may be applied following an operation or surgery.

This disclosure also provides at least one AMP of this disclosure, or a

pharmaceutical composition comprising the same, for use in the treatment of a microbial infection. This disclosure also provides the use of at least one peptide of this disclosure, or a pharmaceutical composition comprising the same, in the manufacture of a medicament for the prevention or treatment of a microbial infection.

DETAILED DESCRIPTION

The terms and phrases used herein have their art-recognized meaning, which can be found by reference to standard texts, journal references and contexts known to those skilled in the art.

As used herein, the singular forms "a", "an", and "the" include plural reference unless the context clearly dictates otherwise. Thus, for example, reference to "a cell" includes a plurality of such cells and equivalents thereof known to those skilled in the art, and so forth. As well, the terms "a" (or "an"), "one or more" and "at least one" can be used interchangeably herein. It is also to be noted that the terms "comprising",

"including", and "containing" can be used interchangeably. The expression "of any of claims XX- YY" (wherein XX and YY refer to claim numbers) is intended to provide a multiple dependent claim in the alternative form, and in some embodiments, is

interchangeable with the expression "as in any one of claims XX- YY."

Unless defined otherwise, all technical and scientific terms used herein have the same meanings as commonly understood by one of ordinary skill in the pertinent art.

Whenever a range of values is given in the specification, for example, a

temperature range, a time range, or a composition or concentration range, all intermediate ranges and subranges, as well as all individual values included in the ranges given are intended to be included in the disclosure. As used herein, ranges specifically include the values provided as endpoint values of the range. For example, a range of 1 to 100 specifically includes the end point values of 1 and 100. Any subranges or individual values in a range or subrange that are included in this description can be excluded from the claims herein. As used herein, "comprising" is synonymous with "including," "containing," or "characterized by," and is inclusive or open-ended and does not exclude additional, unrecited elements or method steps. As used herein, "consisting of excludes any element, step, or ingredient not specified in the claim element. As used herein, "consisting essentially of does not exclude materials or steps that do not materially affect the basic and novel characteristics of the claim. In each instance herein, any of the terms

"comprising", "consisting essentially of and "consisting of may be optionally replaced with either of the other two terms, thus describing alternative aspects of the scope of the subject matter. The invention illustratively described herein suitably may be practiced in the absence of any element or elements, limitation or limitations which is not specifically disclosed herein.

The following definitions are provided to clarify use of these terms in the context of this disclosure.

When used herein, the term "amino acid" is intended to refer to any natural or unnatural amino acid, whether made naturally or synthetically, including those in the L- or D-enantiomeric configurations. The term can also encompass amino acid analog compounds used in peptidomimetics or in peptoids. The term can include a modified or unusual amino acid or a synthetic derivative of an amino acid, e.g. diamino butyric acid and diamino propionic acid and the like. The antimicrobial peptides comprise amino acids linked together by peptide bonds. The peptides are in general in alpha helical

conformation under hydrophobic conditions. Sequences are conventionally given from the amino terminus to the carboxyl terminus. When all the amino acids are of L-configuration, the peptide is said to be an L-enantiomer. When all the amino acids are of D- configuration, the peptide is said to be a D-enantiomer.

A skilled person will appreciate that when two or more amino acids combine to form a peptide, the elements of water are removed, and what remains of each amino acid is called an amino-acid residue. The amino acid residue is the part of an amino acid that makes it unique from all the others. As such, reference herein to an 'amino acid' in the context of an amino acid sequence contained within a peptide will be understood to refer to the respective amino acid residue as appropriate.

Peptides of this disclosure may be substituted, preferably at the N- or C- terminus, by a further moiety. Such moieties may be added to aid the function of the peptide, its targeting or its synthesis, capture or identification, e.g. a label (e.g. biotin) or lipid molecules. The peptides of this disclosure may be chemically modified, for example, post- translationally modified. For example, they may be glycosylated, pegylated or comprise modified amino acid residues. They can be in a variety of forms of polypeptide derivatives, including amides and conjugates with polypeptides. In particular, the amine group at the N-terminus may be substituted with a carboxyl group, such as acetyl, to yield an amide and/or the carboxylic acid group at the C-terminus may be converted to an amide.

Chemically modified peptides also include those having one or more residues chemically derivatized by reaction of a functional side group. Such derivatized side groups include those which have been derivatized to form amine hydrochlorides, amine alkoate salts for example acetate, p-toluene sulfonyl groups, carbobenzoxy groups, t- butyloxycarbonyl groups, chloroacetyl groups and formyl groups. Free carboxyl groups may be derivatized to form salts, methyl and ethyl esters or other types of esters or hydrazides. Free hydroxyl groups may be derivatized to form O-acyl or O-alkyl derivatives.

The term "hemolytic concentration- 50" or "HCso" refers to the concentration of peptide concentration that results in 50% hemolysis of erythrocytes after 1 hr at 37°C. In particular, hemolytic activity may be measured by the procedure described in the

Examples section of this disclosure, particularly Example 3.

Hemolytic concentration was determined from a plot of percent lysis versus peptide concentration ^g/mL). Hemolysis can be determined with red blood cells (RBC) from various species including human red blood cells (hRBC). A skilled person will appreciate that it is advantageous that the hemolytic concentration value is as large as possible, The HCso value was used for testing and comparison purposes to achieve a measure of safety, with respect to hemolysis, that is consistent and comparable across peptides of this disclosure and those known in the art.

The term "therapeutic index" (TI) is the ratio of HCso over minimal inhibitory concentration (MIC) of an antimicrobial agent. Larger values generally indicate greater antimicrobial specificity.

The term "stability" can refer to an ability to resist degradation, to persist in a given environment, and/or to maintain a particular structure. For example, a peptide property of stability can indicate resistance to proteolytic degradation and to maintain an alpha-helical structural conformation.

The following abbreviations are useful: A, Ala, Alanine; M, Met, Methionine; C, Cys, Cysteine; D, Asp, Aspartic Acid; E, Glu, Glutamic Acid; F, Phe, Phenylalanine; G, Gly, Glycine; H, His, Histidine; I, He, Isoleucine; K, Lys, Lysine; L, Leu, Leucine; N,

Asn, Asparagine; O, Orn, Ornithine; P, Pro, Proline; Q, Gin, Glutamine; R, Arg, Arginine; S, Ser, Serine; T, Thr, Threonine; V, Val, Valine; W, Tip, Tryptophan; Y, Tyr, Tyrosine; Dbu, 2,4-Diaminobutyric acid; Dpr, 2,3-Diaminopropionic acid; Ac denotes N a -acetyl and amide denotes C a -amide, RP-HPLC, reversed-phase high performance liquid

chromatography; MIC, minimal inhibitory concentration; HCso hemolytic concentration- 50; TFE, 2,2,2-trifluoroethanol; TFA, trifluoroacetic acid; RBC, red blood cells; hRBC, human red blood cells.

The term "antimicrobial activity" refers to the ability of a peptide to modify a function or metabolic process of a target microorganism, for example to at least partially affect replication, vegetative growth, toxin production, survival, viability in a quiescent state, or other attribute. The term relates to inhibition of growth of a microorganism. In aspects of the claimed peptides and methods, antimicrobial activity relates to the ability of a peptide to kill at least one bacterial species. The bacterial species may be a Gram- negative bacteria. The term can be manifested as microbicidal or microbistatic inhibition of microbial growth.

The phrase "improved biological property" is meant to indicate that a test peptide exhibits less hemolytic activity and/or better antimicrobial activity, or better antimicrobial activity and/or less hemolytic activity, compared to a control peptide (e.g., D-piscidin 1 or D-dermaseptin S4), when tested by the protocols described herein or by any other art- known standard protocols. In general, the improved biological property of the peptide is reflected in the therapeutic index (TI) value which is better than that of the control peptide.

The term "microorganism" herein refers broadly to bacteria, fungi, viruses, and protozoa. In particular, the term is applicable for a microorganism having a cellular or structural component of a lipid bilayer membrane. The membrane may be a cytoplasmic membrane. Pathogenic bacteria, fungi, viruses, and protozoa as known in the art are generally encompassed. Bacteria can include Gram-negative and Gram-positive bacteria in addition to organisms classified in orders of the class Mollicutes and the like, such as species of the Mycoplasma and Acholeplasma genera. Specific examples of Gram- negative bacteria include, but are not limited to, Escherichia coli, Pseudomonas aeruginosa, Acinetobacter baumannii, Salmonella spp., Haemophilus influenzae,

Neisseria spp., Vibrio cholerae, Vibrio parahaemolyticus and Helicobacter pylori.

Examples of Gram-positive bacteria include, but are not limited to, Staphylococcus aureus, Staphylococcus epidermidis, Streptococcus agalactiae, Group A Streptococcus, Streptococcus pyogenes, Enterococcus faecalis, Group B Gram-positive Streptococcus, Corynebacterium xerosis, and Listeria monocytogenes. Examples of fungi include yeasts such as Candida albicans. Examples of viruses include measles virus, herpes simplex virus (HSV-1 and -2), herpes family members (HIV), hepatitis C, vesicular stomatitis virus (VSV), visna virus, and cytomegalovirus (CMV). Examples of protozoa include Giardia.

"Therapeutically effective amount" as used herein, refers to an amount of formulation, composition, or reagent in a pharmaceutically acceptable carrier or a physiologically acceptable salt of an active compound that is of sufficient quantity to ameliorate the undesirable state of the patient, animal, material, or object so treated.

"Ameliorate" refers to a lessening of the detrimental effect of the disease state or disorder, or reduction in contamination, in the receiver of the treatment.

"Pharmaceutical agent or drug" as used herein, refers to a chemical compound or composition capable of inducing a desired therapeutic or prophylactic effect when properly administered to a subject.

"Pharmaceutically acceptable carrier" as used herein, refers to conventional pharmaceutical carriers useful in the methods disclosed herein. Remington's

Pharmaceutical Sciences, by E. W. Martin, Mack Publishing Co., Easton, Pa., 15th Edition (1975), describes compositions and formulations suitable for pharmaceutical delivery of TCR peptides and additional pharmaceutical agents. In general, the nature of the carrier will depend on the particular mode of administration being employed. For instance, parenteral formulations usually comprise injectable fluids that include pharmaceutically and physiologically acceptable fluids such as water, physiological saline, balanced salt solutions, aqueous dextrose, glycerol or the like as a vehicle. For solid compositions (e.g., powder, pill, tablet, or capsule forms), conventional non-toxic solid carriers can include, for example, pharmaceutical grades of mannitol, lactose, starch, or magnesium stearate. In addition to biologically-neutral carriers, pharmaceutical compositions to be administered can contain non-toxic auxiliary substances, such as wetting or emulsifying agents, preservatives, salts, amino acids, and pH buffering agents and the like, for example sodium or potassium chloride or phosphate, Tween, sodium acetate or sorbitan

monolaurate.

As used herein, the term "specificity determinant(s)" refers to positively charged amino acid residue(s) (including, for example, lysine, arginine, or ornithine) in the non- polar face of AMPs that could decrease hemolytic activity/toxicity but increase or maintain the same level of antimicrobial activity, thus increasing the therapeutic index of the AMP.

Antimicrobial peptides (AMPs) of this disclosure have antimicrobial activity by themselves or when covalently conjugated or otherwise coupled or associated with another molecule, e.g., alkanoyl group, polyethylene glycol, an antibody, a small-molecule antibiotic, a specific bacterial cell-surface targeting molecule or a carrier protein such as bovine serum albumin, so long as the peptides are positioned such that they can come into contact with a cell or unit of the target microorganism. These peptides may be modified by methods known in the art provided that the antimicrobial activity is not destroyed or substantially compromised.

The peptides of this disclosure may be isolated or purified. These peptides may be synthetic and can be produced by peptide synthesis techniques or by recombinant expression technology as understood in the art. As used herein, the term "purified" can be understood in to refer to a state of enrichment or selective enrichment of a particular component relative to an earlier state of crudeness or constituency of another component. This term can be considered to correspond to a material that is at least partially purified as opposed to a state of absolute purity. For example, a peptide composition may be considered purified even if the composition does not reach a level of one hundred percent purity with respect to other components in the composition.

Peptides as described above for use in accordance with the invention may be prepared by conventional modes of synthesis including genetic or chemical means.

Synthetic techniques, such as a solid-phase Merrifield-type synthesis, may be preferred for reasons of purity, antigenic specificity, freedom from unwanted side products and ease of production. Suitable techniques for solid-phase peptide synthesis are well known to those skilled in the art (see for example, Merrifield et al, 1969, Adv. Enzymol 32, 221-96 and Fields et al, 1990, Int. J. Peptide Protein Res, 35, 161-214). Chemical synthesis may be performed by methods well known in the art involving cyclic sets of reactions of selective deprotection of the functional groups of a terminal amino acid and coupling of selectively protected amino acid residues, followed finally by complete deprotection of all functional groups.

Synthesis may be performed in solution or on a solid support using suitable solid phases known in the art (for example by the methodology described in the Examples section of this disclosure, particularly Example 1) W.C. Chan and P.D. White; Fmoc Solid Phase Peptide Synthesis: 2) A Practical Approach (Practical Approach Series), Oxford University Press, U.S.A.; Chemistry of Peptide Synthesis by N. Leo Benoiton; CRC Press 2005; ISBN 9781574444544; 3) Tsuda, Y. and Okada, Y. (2010) Solution-Phase Peptide Synthesis, in Amino Acids, Peptides and Proteins in Organic Chemistry: Building Blocks, Catalysis and Coupling Chemistry, Volume 3 (ed A. B. Hughes), Wiley-VCH Verlag GmbH & Co. KGaA, Weinheim, Germany, doi: 10.1002/9783527631803.ch6; 4) Solid- phase peptide synthesis: A practical approach, E. Atherton, R.A. Sheppard, Oxford University press 1989).

Since the peptides of the invention are intended for use in pharmaceutical compositions it will readily be understood that they are each preferably provided in substantially pure form, for example at least 60% pure, more suitably at least 75% pure and preferably at least 85%, especially at least 98% pure (% are on a weight for weight basis). Impure preparations of the compounds may be used for preparing the more pure forms used in the pharmaceutical compositions; these less pure preparations of the compounds should contain at least 1 %, more suitably at least 5% and preferably from 10 to 59%) of a compound of the invention. In an alternative embodiment a peptide of the invention may be produced from or delivered in the form of a polynucleotide which encodes, and is capable of expressing, it. Such polynucleotides can be synthesised according to methods well known in the art, as described by way of example in Sambrook et al (1989, Molecular Cloning - a laboratory manual; Cold Spring Harbor Press). Such polynucleotides may be used in vitro or in vivo in the production of a peptide of the invention. Such polynucleotides may therefore be administered or used in the treatment of a microbial infection or another disease or condition as described herein. All references cited throughout this application, for example patent documents including issued or granted patents or equivalents; patent application publications; and non-patent literature documents or other source material; are hereby incorporated by reference herein in their entireties, as though individually incorporated by reference, to the extent each reference is at least partially not inconsistent with the disclosure in this application (for example, a reference that is partially inconsistent is incorporated by reference except for the partially inconsistent portion of the reference).

When a group of substituents is disclosed herein, it is understood that all individual members of that group and all subgroups, including any isomers, enantiomers, and diastereomers of the group members, are disclosed separately.

When a Markush group or other grouping is used herein, all individual members of the group and all combinations and subcombinations possible of the group are intended to be individually included in the disclosure. When a compound is described herein such that a particular isomer, enantiomer or diastereomer of the compound is not specified, for example, in a formula or in a chemical name, that description is intended to include each isomer and enantiomer of the compound described individually, or in any combination. Additionally, unless otherwise specified, all isotopic variants of compounds disclosed herein are intended to be encompassed by the disclosure. As a brief illustration, it will be understood that any one or more hydrogens in a molecule disclosed can be replaced with deuterium or tritium.

Isotopic variants of a molecule are generally useful as standards in assays for the molecule and in chemical and biological research related to the molecule or its use.

Methods for making such isotopic variants are known in the art.

One of ordinary skill in the art will appreciate that starting materials, biological and chemical materials, biological and chemical reagents, synthetic methods, purification methods, analytical methods, assay methods, and biological methods other than those specifically exemplified can be employed in the practice of the invention without resort to undue experimentation. All art-known functional equivalents, of any such materials and methods are intended to be included in this disclosure.

The terms and expressions which have been employed herein are used as terms of description and not of limitation, and there is no intention in the use of such terms and expressions of excluding any equivalents of the features shown and described or portions thereof, but it is recognized that various modifications are possible within the scope of the invention claimed. Thus, although the invention has been disclosed by various aspects that may include preferred embodiments and aspects, modifications and variations of the concepts herein disclosed may be resorted to by those skilled in the art. Such

modifications and variations are considered to be within the scope of this invention as defined by the appended claims.

The peptides described herein can be used to prepare therapeutic pharmaceutical compositions, for example, by combining the compounds with a pharmaceutically acceptable diluent, excipient, or carrier. The peptides may be added to a carrier in the form of a salt or solvate. For example, in cases where peptides are sufficiently basic or acidic to form stable non-toxic acid or base salts, administration of the peptides as salts may be appropriate. Examples of pharmaceutically acceptable salts are organic acid addition salts formed with acids that form a physiological acceptable anion, for example, tosylate, methanesulfonate. acetate, citrate, malonate, tartrate, succinate, benzoate, ascorbate, a- ketoglutarate, lactate, phosphate and 3 -glycerophosphate. Suitable inorganic salts may also be formed, including hydrochloride, halide, sulfate, nitrate, bicarbonate, and carbonate salts.

Pharmaceutically acceptable salts may be obtained using standard procedures well known in the art, for example by reacting a sufficiently basic compound such as an amine with a suitable acid to provide a physiologically acceptable ionic compound. Alkali metal (for example, sodium, potassium or lithium) or alkaline earth metal (for example, calcium) salts of carboxylic acids can also be prepared by analogous methods.

Compositions of this disclosure

When employed as pharmaceuticals, especially as antimicrobial agents

administered to mammals, the AMPs of this disclosure are administered in the form of pharmaceutical compositions. These compounds can be administered by a variety of routes including oral, rectal, transdermal, subcutaneous, intravenous, intramuscular, intrathecal, and intranasal. Such pharmaceutical compositions are prepared in a manner well known in the pharmaceutical art and comprise at least one AMP of this disclosure.

The pharmaceutical compositions of the present invention contain, as the active ingredient, one or more of the AMPs of this disclosure, associated with pharmaceutically acceptable formulations. In making the compositions of this invention, the active ingredient is usually mixed with an excipient, diluted by an excipient or enclosed within a carrier which can be in the form of a capsule, sachet, paper or other container. An excipient is usually an inert substance that forms a vehicle for a drug. When the excipient serves as a diluent, it can be a solid, semi-solid, or liquid material, which acts as a vehicle, carrier or medium for the active ingredient. Thus, the compositions can be in the form of solutions, syrups, aerosols (as a solid or in a liquid medium), ointments containing, for example, up to 30% by weight of the active compound, soft and hard gelatin capsules, suppositories, sterile injectable solutions, and sterile packaged powders.

In preparing a formulation, it may be necessary to mill active compounds of this disclosure to provide the appropriate particle size prior to combining with the other ingredients. If the antimicrobial peptide is substantially insoluble, it ordinarily is milled to a particle size of less than 200 mesh. If the compound(s) is substantially water soluble, the particle size is normally adjusted by milling to provide a substantially uniform distribution in the formulation, e.g. about 40 mesh.

Some examples of suitable excipients include lactose, dextrose, sucrose, sorbitol, mannitol, starches, gum acacia, gum Arabic, calcium phosphate, alginates, tragacanth, gelatin, calcium silicate, microcrystalline cellulose, polyvinylpyrrolidone, cellulose, sterile water, syrup, and methylcellulose. The formulations can additionally include: lubricating agents such as talc, magnesium stearate, and mineral oil; wetting agents; emulsifying and suspending agents; preserving agents such as methyl- and propylhydroxy-benzoates; sweetening agents; and flavoring agents. The compositions of this disclosure can be formulated to provide quick, sustained or delayed release of the active ingredient after administration to the patient by employing procedures known in the art.

For preparing solid compositions such as tablets, the principal active ingredient is mixed with a pharmaceutical excipient to form a solid preformulation composition containing a homogeneous mixture of a peptide of the present invention. When referring to these preformulation compositions as homogeneous, it is meant that the active ingredient is dispersed evenly throughout the composition so that the composition may be readily subdivided into equally effective unit dosage forms such as tablets, pills and capsules. This solid preformulation is then subdivided into unit dosage forms of the type described above containing from, for example, 0.1 to about 500 mg of the active ingredient of the present invention.

Formulations of this disclosure suitable for oral administration may be in the form of capsules, cachets, pills, tablets, powders, granules or as a solution or a suspension in an aqueous or non-aqueous liquid, or an oil-in-water or water-in-oil liquid emulsions, or as an elixir or syrup, or as pastilles (using an inert base, such as gelatin and glycerin, or sucrose and acacia), and the like, each containing a predetermined amount of a compound or compounds of the present invention as an active ingredient. A compound or compounds of the present invention may also be administered as bolus, electuary or paste.

In solid dosage forms of this disclosure for oral administration (capsules, tablets, pills, dragees, powders, granules and the like), the active ingredient is mixed with one or more pharmaceutically acceptable carriers, such as sodium citrate or dicalcium phosphate, and/or any of the following: (1) fillers or extenders, such as starches, lactose, sucrose, glucose, mannitol, and/or silicic acid; (2) binders, such as, for example,

carboxymethylcellulose, alginates, gelatin, polyvinyl pyrrolidone, sucrose and/or acacia; (3) humectants, such as glycerol; (4) disintegrating agents, such as agar-agar, calcium carbonate, potato or tapioca starch, alginic acid, certain silicates, and sodium carbonate; (5) solution retarding agents, such as paraffin; (6) absorption accelerators, such as quaternary ammonium compounds; (7) wetting agents, such as, for example, cetyl alcohol and glycerol monosterate; (8) absorbents, such as kaolin and bentonite clay; (9) lubricants, such as talc, calcium stearate, magnesium stearate, solid polyethylene glycols, sodium lauryl sulfate, and mixtures thereof; and (10) coloring agents. In the case of capsules, tablets and pills, the pharmaceutical compositions may also comprise buffering agents. Solid compositions of a similar type may be employed as fillers in soft and hard-filled gelatin capsules using such excipients as lactose or milk sugars, as well as high molecular weight polyethylene glycols and the like.

A tablet may be made by compression or molding optionally with one or more accessory ingredients. Compressed tablets may be prepared using binder (for example, gelatin or hydroxypropylmethyl cellulose), lubricant, inert diluent, preservative, disintegrant (for example, sodium starch glycolate or cross-linked sodium carboxymethyl cellulose), surface-active or dispersing agent. Molded tablets may be made by molding in a suitable machine a mixture of the powdered compound moistened with an inert liquid diluent. The tablets, and other solid dosage forms of the pharmaceutical compositions of the present invention, such as dragees, capsules, pills and granules, may optionally be scored or prepared with coatings and shells, such as enteric coatings and other coatings well known in the pharmaceutical-formulating art. They may also be formulated to provide slow or controlled release of the active ingredient therein using, for example, hydroxypropylmethyl cellulose in varying proportions to provide the desired release profile, other polymer matrices, liposomes and/or microspheres. They may be sterilized by, for example, filtration through a bacteria-retaining filter. These compositions may also optionally contain opacifying agents and may release the active ingredient only, or preferentially, in a certain portion of the gastrointestinal tract, optionally, in a delayed manner. Examples of embedding compositions which can be used include polymeric substances and waxes. The active ingredient can also be in microencapsulated form.

The tablets or pills of this disclosure may be coated or otherwise compounded to provide a dosage form affording the advantage of prolonged action. For example, the tablet or pill can comprise an inner dosage and an outer dosage component, the latter being in the form of an envelope over the former. The two components can be separated by an enteric layer which serves to resist disintegration in the stomach and permit the inner component to pass intact into the duodenum or to be delayed in release. A variety of materials can be used for such enteric layers or coatings, such materials including a number of polymeric acids and mixtures of polymeric acids with such materials as shellac, cetyl alcohol, and cellulose acetate.

Liquid dosage forms for oral administration of the compounds of this disclosure include pharmaceutically-acceptable emulsions, microemulsions, solutions, suspensions, syrups and elixirs. In addition to the active ingredient, the liquid dosage forms may contain inert diluents commonly used in the art, such as water or other solvents, solubilizing agents and emulsifiers such as ethyl alcohol, isopropyl alcohol, ethyl carbonate, ethyl acetate, benzyl alcohol, benzyl benzoate, propylene glycol, 1,3-butylene glycol, oils (in particular, cottonseed, groundnut, corn, germ, olive, castor and sesame oils), glycerol, tetrahydrofuryl alcohol, polyethylene glycols and fatty acid esters of sorbitan, and mixtures thereof.

Besides inert diluents, the oral compositions can also include adjuvants such as wetting agents, emulsifying and suspending agents, sweetening, flavoring, coloring, perfuming, and preservative agents.

Suspensions, in addition to the active compounds, may contain suspending agents as, for example, ethoxylated isostearyl alcohols, polyoxyethylene sorbitol and sorbitan esters, microcrystalline cellulose, aluminum metahydroxide, bentonite, agar-agar and tragacanth, and mixtures thereof.

Formulations of the pharmaceutical compositions of this disclosure for rectal or vaginal administration may be presented as a suppository, which may be prepared by mixing one or more compounds of this disclosure with one or more suitable nonirritating excipients or carriers comprising, for example, cocoa butter, polyethylene glycol, a suppository wax or salicylate, and which is solid at room temperature, but liquid at body temperature and, therefore, will melt in the rectum or vagina and release the active compound. Formulations of the present invention which are suitable for vaginal administration also include pessaries, tampons, creams, gels, pastes, foams or spray formulations containing such carriers as are known in the art to be appropriate.

Dosage forms for the topical or transdermal administration of compounds of this invention include powders, sprays, ointments, pastes, creams, lotions, gels, solutions, patches, drops and inhalants. The active ingredient may be mixed under sterile conditions with a pharmaceutically-acceptable carrier, and with any buffers, or propellants which may be required.

The ointments, pastes, creams and gels may contain, in addition to an active ingredient, excipients, such as animal and vegetable fats, oils, waxes, paraffins, starch, tragacanth, cellulose derivatives, polyethylene glycols, silicones, bentonites, silicic acid, talc and zinc oxide, or mixtures thereof.

Powders and sprays can contain, in addition to an active ingredient, excipients such as lactose, talc, silicic acid, aluminum hydroxide, calcium silicates and polyamide powder or mixtures of these substances. Sprays can additionally contain customary propellants such as chlorofluorohydrocarbons and volatile unsubstituted hydrocarbons, such as butane and propane.

Transdermal patches have the added advantage of providing controlled delivery of compounds of this disclosure to the body. Such dosage forms can be made by dissolving, dispersing or otherwise incorporating one or more compounds of this disclosure in a proper medium, such as an elastomeric matrix material. Absorption enhancers can also be used to increase the flux of the compound across the skin. The rate of such flux can be controlled by either providing a rate-controlling membrane or dispersing the compound in a polymer matrix or gel.

Pharmaceutical formulations include those suitable for administration by inhalation or insufflation or for nasal or intraocular administration. For administration to the upper (nasal) or lower respiratory tract by inhalation, the compounds of this disclosure are conveniently delivered from an insufflator, nebulizer or a pressurized pack or other convenient means of delivering an aerosol spray. Pressurized packs may comprise a suitable propellant such as dichlorodifluoromethane, trichlorofluoromethane,

dichlorotetrafluoroethane, carbon dioxide, or other suitable gas. In the case of a pressurized aerosol, the dosage unit may be determined by providing a valve to deliver a metered amount.

Alternatively, for administration by inhalation or insufflation, the composition may take the form of a dry powder, for example, a powder mix of one or more compounds of this disclosure and a suitable powder base, such as lactose or starch. The powder composition may be presented in unit dosage form in, for example, capsules or cartridges, or, e.g., gelatin or blister packs from which the powder may be administered with the aid of an inhalator, insufflator or a metered-dose inhaler.

For intranasal administration, compounds of this disclosure may be administered by means of nose drops or a liquid spray, such as by means of a plastic bottle atomizer or metered-dose inhaler. Typical of atomizers are the Mistometer (Wintrop) and Medihaler (Riker).

Drops, such as eye drops or nose drops, may be formulated with an aqueous or nonaqueous base also comprising one or more dispersing agents, solubilizing agents or suspending agents. Liquid sprays are conveniently delivered from pressurized packs.

Drops can be delivered by means of a simple eye dropper-capped bottle or by means of a plastic bottle adapted to deliver liquid contents dropwise by means of a specially shaped closure.

Pharmaceutical compositions of this invention suitable for parenteral

administration comprise one or more compounds of this disclosure in combination with one or more pharmaceutically-acceptable, sterile isotonic aqueous or non-aqueous solutions, dispersions, suspensions or emulsions, or sterile powders which may be reconstituted into sterile injectable solutions or dispersions just prior to use, which may contain antioxidants, buffers, solutes which render the formulation isotonic with the blood of the intended recipient or suspending or thickening agents.

Examples of suitable aqueous and nonaqueous carriers which may be employed in the pharmaceutical compositions of this disclosure include water, ethanol, polyols (such as glycerol, propylene glycol, polyethylene glycol, and the like), and suitable mixtures thereof, vegetable oils, such as olive oil, and injectable organic esters, such as ethyl oleate. Proper fluidity can be maintained, for example, by the use of coating materials, such as lecithin, by the maintenance of the required particle size in the case of dispersions, and by the use of surfactants.

These compositions may also contain adjuvants such as wetting agents, emulsifying agents, and dispersing agents. It may also be desirable to include isotonic agents, such as sugars, sodium chloride, and the like in the compositions. In addition, prolonged absorption of the injectable pharmaceutical form may be brought about by the inclusion of agents which delay absorption such as aluminum monosterate and gelatin.

In some cases, to prolong the effect of a drug, it is desirable to slow the absorption of the drug from subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material having poor water solubility. The rate of absorption of the drug then depends upon its rate of dissolution which, in turn, may depend upon crystal size and crystalline form. Alternatively, delayed absorption of a parenterally-administered drug is accomplished by dissolving or suspending the drug in an oil vehicle.

Injectable depot forms are made by forming microencapsule matrices of the drug in biodegradable polymers such as polylactide-polyglycolide. Depending on the ratio of drug to polymer, and the nature of the particular polymer employed, the rate of drug release can be controlled. Examples of other biodegradable polymers include

poly(orthoesters) and poly (anhydrides). Depot injectable formulations are also prepared by entrapping the drug in liposomes or microemulsions which are compatible with body tissue. The injectable materials can be sterilized for example, by filtration through a bacterial-retaining filter.

The formulations may be presented in unit-dose or multi-dose sealed containers, for example, ampules and vials, and may be stored in a lyophilized condition requiring only the addition of the sterile liquid carrier, for example water for injection, immediately prior to use. Extemporaneous injection solutions and suspensions may be prepared from sterile powders, granules and tablets of the type described above.

Suitable alkalinizing agents include alkali metal salts and alkaline earth metal salts. The alkali metal salts include sodium carbonate, sodium hydroxide, sodium silicate, disodium hydrogen orthophosphate, sodium aluminate, and other suitable alkali metal salts or mixtures thereof. Suitable alkaline metal salts include calcium carbonate, calcium hydroxide, magnesium carbonate, magnesium hydroxide, magnesium silicate, magnesium aluminate, aluminum magnesium hydroxide or mixture thereof. More particularly, calcium carbonate, potassium bicarbonate, calcium hydroxide, and/or sodium carbonate may be used as alkalinizing agents to obtain a formulation pH within the desired pH range of pH 8 to pH 13. The concentration of the alkalinizing agent is selected to obtain the desired pH, varying from about 0.1% to about 30%, by weight, and more preferably from about 12.5% to about 30%), by weight, of the total weight of the dosage formulation.

Suitable antioxidants may be selected from amongst one or more pharmaceutically acceptable antioxidants known in the art. Examples of pharmaceutically acceptable antioxidants include butylated hydroxyanisole (BHA), sodium ascorbate, butylated hydroxytoluene (BHT), sodium sulfite, citric acid, malic acid and ascorbic acid. The antioxidants may be present in the dosage formulations of the present invention at a concentration between about 0.001%> to about 5%, by weight, of the dosage formulation.

Suitable chelating agents may be selected from amongst one or more chelating agents known in the art. Examples of suitable chelating agents include disodium edetate (EDTA), edetic acid, citric acid and combinations thereof. The chelating agents may be present in a concentration between about 0.001%> and about 5%, by weight, of the dosage formulation.

Methods for Preventing and Treating Microbial Infection

Another aspect of this disclosure provides methods for preventing and treating a microbial infection. These methods include administering to a subject in need thereof a therapeutically effective amount of a peptide or composition of this disclosure that kills or inhibits the growth of infectious microbes, thereby inhibiting or treating the microbial infections. The infecting microorganism may include Gram-negative bacteria. The infecting microorganism may be a Gram-negative bacteria, which may include, but is not limited to, Escherichia coli, Pseudomonas aeruginosa, Acinetobacter baumannii, Salmonella spp., Haemophilus influenzae, Neisseria spp., Vibrio cholerae, Vibrio parahaemolyticus and Helicobacter pylori.

The antimicrobial peptides administered, preferably as a component of a pharmaceutical composition, can include a single antimicrobial peptide of this disclosure, or multiple AMPs of this disclosure. The peptides may include peptides having at least 85%, or at least 90%, or at least 95% homology to a peptide sequence of SEQ ID NOs:3- 11, 14-17 or 19-26 and which effectively treat or prevent a microbial infection. The peptides may include fragments of the peptide of SEQ ID NO: 1 that retain the ability to effectively treat or prevent a microbial infection. Exemplary peptides include the amino acid sequences set forth in SEQ ID NOs:3-l 1, 14-17 or 19-26. Appropriate peptides to use in the methods disclosed herein can be determined by those skilled in the art.

Therapeutic AMPs of this disclosure may be administered by a number of routes, including orally, topically, or parenteral administration including intravenous by injection or infusion, intraperitoneal, intramuscular, intradermal, intrathecal, intrasternal, or intraarticular injection. One of skill in the art can readily determine the appropriate route of administration.

The therapeutically effective amounts of the AMPs of this disclosure that inhibit or kill an infecting microorganism will depend upon the subject being treated, the severity and type of the infection, and the manner of administration. For example, a therapeutically effective amount of a peptide of this disclosure can vary from about 1 microgam/injection up to about lOmg/injection. The exact amount of the peptide is readily determined by one of skill in the art based on the age, weight, sex, and physiological condition of the subject. Effective doses can be extrapolated from dose-response curves derived from in vitro or animal model test systems.

One or more peptides of this disclosure that effectively inhibit or kill an infecting microorganism can be administered in conjunction with one or more additional pharmaceutical agents. The additional pharmaceutical agents can be administered at the same time as, or sequentially with, the peptide(s) of this disclosure. The additional pharmaceutical agent may be an additional antimicrobial agent. When administered at the same time, the additional pharmaceutical agent(s) can be formulated in the same composition that includes the peptide(s) of this disclosure.

In one embodiment, the invention provides a product comprising a peptide of the invention and at least one other therapeutic agent as a combined preparation for simultaneous, separate or sequential use in therapy. In one embodiment, the therapy is the treatment of a microbial infection. Products provided as a combined preparation include a composition comprising the peptide of the invention and the other therapeutic agent(s) together in the same pharmaceutical composition, or the agent of the invention and the other therapeutic agent(s) in separate form, e.g. in the form of a kit.

In the combination therapies of the invention, the peptide of the invention and the other therapeutic agent may be manufactured and/or formulated by the same or different manufacturers. Moreover, the peptide of the invention and the other therapeutic may be brought together into a combination therapy: (i) prior to release of the combination product to physicians (e.g. in the case of a kit comprising the peptide of the invention and the other therapeutic agent); (ii) by the physician themselves (or under the guidance of the physician) shortly before administration; (iii) in the patient themselves, e.g. during sequential administration of the peptide of the invention and the other therapeutic agent.

Accordingly, the invention provides the use of peptide of the invention for treating a microbial infection, wherein the medicament is prepared for administration with another therapeutic agent. The invention also provides the use of another therapeutic agent for treating a microbial infection, wherein the medicament is administered with a peptide of the invention.

The invention also provides a peptide of the invention for use in a method of treating a microbial infection, wherein the peptide of the invention is prepared for administration with another therapeutic agent. The invention also provides another therapeutic agent for use in a method of treating a microbial infection, wherein the other therapeutic agent is prepared for administration with a peptide of the invention. The invention also provides a peptide of the invention for use in a method of treating a microbial infection, wherein the peptide of the invention is administered with another therapeutic agent. The invention also provides another therapeutic agent for use in a method of treating a microbial infection, wherein the other therapeutic agent is administered with a peptide of the invention. The invention also provides the use of a peptide of the invention for treating a microbial infection, wherein the subject has previously (e.g. within 24 hours) been treated with another therapeutic agent. The invention also provides the use of another therapeutic agent for treating a microbial infection, wherein the subject has previously (e.g. within 24 hours) been treated with a peptide of the invention.

Compositions may additionally comprise molecules which assist or augment the action of the agents of the invention, e.g. an antibiotic, which inhibits bacterial or fungal growth or kills bacteria or fungi. A peptide of the invention may be combined in variable or fixed ratio combinations with antibiotics from any known antibiotic class. Specifically, these antibiotics may include antibiotics of the lincomycin family (a class of antibiotic agents originally recovered from streptomyces lincolnensis); antibiotics of the tetracycline family (a class of antibiotic agents originally recovered from streptomyces aureofaciens); and sulfur-based antibiotics such as the sulfonamides. Beta-lactams that can be combined include penems, carbapenems (imipenem, meropenem, ertapenem, doripenem, panipenem, biapenem, and the like), monobactams (aztreonam, tigimonam, carumonam, BAL30072, and the like), as well as a variety of other beta-lactam cell envelope antibiotics (see:

wikipedia.org/wiki/Monobactam).

Specific examples of some suitable antibiotics of the lincomycin family include lincomycin, clindamycin, and clindamycin phosphate. Specific examples of macrolide antibiotics include erythromycin, azithromycin, clarithromycin, dirithromycin, roxithromycin, carbomycin A, josamycin, kitasamycin, midecamycin/midecamycin acetate and troleandomycin.

Specific examples of ketolide antibiotics include telithromycin, cethromycin, solithromycin, spiramycin, ansamycin, oleandomycin, carbomycin, and tylosin. Specific examples of antibiotics of the tetracycline family include tetracycline itself, chlortetracycline, oxytetracycline, demeclocycline, rolitetracycline, methacycline and doxycycline.

Specific examples of sulfur-based antibiotics include the sulfonamides,

sulfacetamide, sulfabenzamide, sulfadiazine, sulfadoxine, sulfamerazine, sulfamethazine, sulfamethizole, sulfisoxazole, and sulfamethoxazole. Further examples of combinable antibiotics include the oxazolidinones such as zyvox (linezolid), peptide antibiotics such as the polymixins, quinolones, fluoroquinolones (wikipedia.orgiwiki/Quinolone antibiotic), aminoglycosides

(wikipedia.org/wiki/Aminoglycoside), and rifamycins (wikipedia.org/wiki/Rifamycin).

Combinable antibiotics can also include various antibacterial agents, antifungal agents, antimycotic agents and antiviral agents; penicillins such as ampicillin or amoxicillin, cephalosporins such as cephalothin and ceclor (cephachlor), aminoglycosides such as, kanamycin, macrolides such as erythromycin, nystatin, and amphotericin; and the antibiotics amikacin, bacillomycin, chloramphenicol, doxorubicin, doxycycline, ethambutol, gentamicin, isoniazid, kanamycin, carbacephalosporins such as lorabid (loracarbef), mupirocin, neomycin, pyrrolnitrin, rifampin, streptomycin, and vancomycin.

Those skilled in the art can determine an appropriate time and duration of therapy that includes the administration of a peptide of this disclosure to achieve the desired preventative or ameliorative effects on the subject treated.

The invention now being generally described will be more readily understood by reference to the following examples, which are included merely for the purposes of illustration of certain aspects of the invention. The examples are not intended to limit the invention, as one of skill in the art would recognize from the above teachings and the following examples that other techniques and methods can satisfy the claims and can be employed without departing from the scope of the claimed invention.

EXAMPLES

Example 1

All peptides were prepared in an analogous manner using the following procedures

SEQ ID SEQUENCE NO.

3 Ac - KLKKLLKKLSSAKSSKLSTLLKKLKK- NH 2

5 Ac - KLSSLLSKL KKAKKKKL KKLLKAL S S - NH 2

6 Ac - KLKKLLKKLKSAKSSKLSTLLSKLKK- NH 2

8 Ac -ALKKLLKKLSSAKSSKLSTLLKKLKK- NH 2 10 Ac -ALSSLLSKL KKAKKKKL KKLLKAL S S - NH 2

14 Ac - KLKKLLKKLSSA ( Orn) SS ( Orn ) LSTLLKKLKK-NH 2

15 Ac - KLKKLLKKLSSA ( Dbu) SS ( Dbu ) LSTLLKKLKK-NH 2

16 Ac - KLKKLLKKLSSA ( Dpr ) SS ( Dpr ) LSTLLKKLKK-NH 2

19 Ac -ALKKLLKKLSSA ( Orn) SS ( Orn ) LSTLLKKLKK-NH 2

20 Ac -ALKKLLKKLSSA ( Dbu) SS ( Dbu ) LSTLLKKLKK-NH 2

21 Ac -ALKKLLKKLSSA ( Dpr ) SS ( Dpr ) LSTLLKKLKK-NH 2

23 Ac -ALKSLLKKLSKAKKKKLKKLLKALSS -NH 2

24 Ac - KLKKLLKKLSSAKSSKLSSLLKKLKK-NH 2

25 Ac -ALKKLLKKLSSAKSSKLSSLLKKLKK-NH 2

26 Ac - KLKSLLKSLSKAKKKKLKSLLKALSK-NH 2

Solid-phase Peptide Synthesis: Standard solid-phase peptide synthesis methodology was conducted using 9-fluorenylmethoxycarbonyl (Fmoc) chemistry and 4-Rinkamide MBHA resin (P3 Biosystems, Louisville, KY) or Rink Amide-ChemMatrix® resin (Biotage, Charlotte, NC) using a Focus-XC peptide synthesizer (Aapptec, Louisville, KY). The coupling procedure used (Benzotriazol-l-yloxy)tris(dimethylamino) phosphonium hexafluorophosphate (Bop)/ 1-hydroxybenzotriazole (HOBt) in dimethylformamide (DMF) with Ν,Ν-diisopropylethylamine (DIPEA) in N-methyl-2-pyrrolidinone (NMP) with the first coupling at room temperature for one hour and the second coupling at 50°C for another hour. The deprotection procedure (removal of Fmoc protecting group) was carried out by treatment of the resin with 0.1 M HOBt in DMF with 20% piperidine. After completion of the synthesis of the desired amino-acid sequence the N-terminus protecting group was removed as described above and the free amine was acetylated with acetic anhydride (Ac 2 0) following the procedure described below.

Acetylation of N-Terminus-NH2

The N-terminal free amino peptide-Rink Amide MBHA resin (0.2 mmol) was suspended in DMF (5 mL) and a solution of Ac 2 0 (0.95 mL, 10 mmol) and DIPEA (1.74 mL, 10 mmol) in DMF (5mL) was added. The sample was mixed for 2 h and the acetylation procedure repeated until the Kaiser test (Ninhydrin Test) was negative. The resin was washed (DMF x 5) and dried by lyophilisation overnight.

Global Deprotection and Cleavage from Resin

The peptide was cleaved from the peptide resin from above with a mixture of 90% trifluoroacetic acid (TFA), 5%> water and 5%> triisopropylsilane (TIS) for 1-2 h. The resin was removed by filtration and the peptide precipitated with ice-cooled diethyl ether on ice for 1-2 h. The pellet was concentrated under reduced pressure, re-dissolved in

acetonitrile/water (1 : 1, with 0.2% TFA) and the solution lyophilized to obtain the crude peptide which was purified by FIPLC as described below.

Analytical and preparative purification by reversed-phase chromatography

Analytical RP-HPLC: Column, Luna CI 8 (2), 250 x 4.6 mm I D., 5 μιη particle size, 100 A pore size from Phenomenex. Run conditions: linear AB gradient (1% acetonitrile/min, starting from 2% acetonitrile) at a flow-rate of 1 mL/min, where eluent A is 0.2%) aq. TFA and eluent B is 0.18% TFA in acetonitrile; temperature, 30°C.

Preparative RP-HPLC: Column, Luna C18 (2), 250 x 30 mm I D., 10 urn particle size, 100 A pore size from Phenomenex. Peptides were dissolved in 0.2%> aq. TFA containing 2%> acetonitrile to a final concentration of 10 mg/mL, filtered sequentially through a 0.45 μπι then 0.22 μπι Millipore filters, and loaded onto the column via multiple 20-mL injections into a 20-mL injection loop at a flow-rate of 10 mL/min. Run conditions: 1%) acetonitrile/min gradient up to an acetonitrile concentration 15%> below that required to elute the peptide during analytical RP-HPLC on the same packing material, then shallow gradient elution (0.1%> acetonitrile/min) at a flow-rate of 10 mL/min (same eluents as shown above for analytical RP-HPLC) at room temperature. The preparative HPLC instrumentation used was obtained from Aapptec, Louisville, KY consisting of two P300 pumps for preparing gradients with flow rates up to 50 mL/min and a variable UV/VIS wavelength detector (UV 3000) connected to a LKB fraction collector. Fractions were analyzed by LC/MS using anAgilent 1100 series HPLC with autosampler connected to an Agilent LC/MSD Trap XT (Agilent Technologies, Inc., Santa Clara, CA). The column used was a HALO 2-C18 column 2.1 x 50 mm, 2 μ particle size from Advanced Materials Technology (Wilmington, DC). Solid-core particles with 0.4-micron thick porous shell and 90 A pore size or an Agilent Zorbax 300SB-C8, 4.6 mm I D. x 100 mm, 300 A pore size, 3.5 micron particle size.

Conversion to Final Acetate Salt of Peptides

The HPLC purified peptide trifluoroacetate (TFA) salt from above was dissolved in 95:5 acetic acid/H20 (15mL) and transferred to a polypropylene conical tube (50 mL) and allowed to stand for 20 minutes to enable counter ion exchange to occur. The sample was frozen (liquid N 2 ) and the peptide lyophilized overnight. This process was repeated twice and the resulting peptide acetate salt was suspended in H2O (10 mL) and lyophylised overnight to give the desired compound as an acetate salt.

Example 2

Antibacterial Activity Gram-Negative and Gram-Positive Bacterial Strains used in this study

Bacterial strains were acquired from the American Type Culture Collection (ATCC, Virginia, USA) and the National Collection of Type Cultures (NCTC, Public Health England, Salisbury, UK). Minimum Inhibitory Concentration (MIC) of peptides versus gram-negative strams-Acinetobacter baumannii NCTC 13420, and Pseudomonas aeruginosa ATCC 27853 and gram-positive strain Staphylococcus aureus ATCC 29213, were determined by broth microdilution as per the guidelines of the Clinical and

Laboratory Standards Institute ("Methods for Dilution Antimicrobial Susceptibility Tests for Bacteria That Grow Aerobically"; Approved Standard-Ninth Edition. CLSI document M07-A9, 2012). Peptides were dissolved in sterile distilled water, which was also used as the drug-free vehicle control when determining peptide MICs. Prior to use in the MIC protocol, bacterial strains were grown on Mueller Hinton agar at 37°C in an ambient atmosphere. The MIC protocol involves the generation of a two-fold dilution series of test peptides in 96-well microtitre plates using cation-adjusted Mueller Hinton broth, giving a typical final concentration range of 0.25-128 μg/mL. A fixed inoculum of the test microorganism (5x10 5 cells/mL) is also added to the plate. The MIC is determined as the lowest concentration of compound that inhibits visible bacterial growth following incubation at 37°C for 16-20 hours. A difference in MIC of two-fold between

experimental replicates is considered to be within the normal variability of this protocol. Table 2: Antimicrobial activity, Hemolytic Activity and Therapeutic Indices

* Not Tested

This above assay was also utilised to assess the impact of plasma protein binding on the susceptibility of test bacteria to antimicrobial peptides. Plasma protein binding can have a dramatic impact on the efficacy of a drug, since it is the 'free' unbound drug that elicits the antibacterial effects. In this case, CA-MHB was supplemented with 20% human serum (v/v; BioIVT, West Sussex, UK) and the MIC assay conducted as described. MIC values obtained from the serum-free assay were compared with serum-supplemented assay to identify the 'serum shift' effect and the fold-change in susceptibility. Less than or equal to two-fold change in MIC between serum-free and serum-supplemented assays suggested little plasma protein binding and good stability in human serum.

Example 3

Hemolytic Activity and Therapeutic Indices

Measurement of Hemolytic Activity:

To determine the extent of haemolytic activity (the ability to lyse red blood cells) in test peptides, a haemolysis assay was conducted using human blood (BioIVT, West Sussex, UK). Human erythrocytes were exposed to a single fixed concentration or a range of concentrations as to determine the % haemolysis (fixed concentration) or the concentration at which 50% haemolysis occurred (HCso), respectively. Prior to the assay, whole human blood was repeatedly washed in phosphate buffered saline (PBS) under cooled conditions to remove the buffy coat, plasma and cell debris. Washed erythrocytes were diluted as to achieve lxlO 8 cells/mL in PBS. Prepared human erythrocyte samples were combined with peptide at a single fixed concentration of 128 μg/mL, or a range of concentrations (two-fold dilution series) from 1000-31.25 μg/mL. Positive and negative controls containing 2% (v/v) Triton X-100 and PBS alone, respectively, were also prepared. Samples were incubated at 37°C for 1 hour and then centrifuged to collect intact erythrocytes. Haemoglobin release was determined by recording the optical density of the supernatant at 540nm. Optical density values were transformed to a percentage of the positive control. Results were recorded as a percentage haemolysis at 128 μg/mL, or by plotting the percentage haemolysis versus concentration and extrapolating the HCso using linear regression analysis (Table 2 above)

Calculation of Therapeutic Index The therapeutic index is a widely-accepted parameter to represent the specificity of antimicrobial peptides for prokaryotic versus eukaryotic cells. It is calculated by the ratio of hemolytic activity and antimicrobial activity (MIC) (Table 2 above); thus, larger values of therapeutic index indicate greater specificity for prokaryotic cells. With the peptides used in this study the inventors used the HC50/MIC ratio value to calculate the therapeutic index.

Example 4

Peptide Hydrophobicity

Retention behavior in RP-HPLC is an excellent method to represent overall peptide hydrophobicity. Retention times of peptides are highly sensitive to the conformational status of the peptides upon interaction with the hydrophobic environment of the column matrix 17 ' 26 . The non-polar faces of amphipathic alpha-helical and amphipathic cyclic beta- sheet peptides represent a preferred binding domain for interaction with the hydrophobic matrix of the reversed-phase column 27 . In this study, the observed peptide hydrophobicity was determined by RP-HPLC retention time as described below and are relative hydrophobicities because they are dependent on the TFA concentration and organic solvent in the mobile phase, gradient rate, temperature, flow rate and the column used. Analytical Purification by Reversed-phase Chromatography: Analytical RP-HPLC:

Column: Zorbax SB-300 C8 (150 X 2.1mm ID; 5-μπι particle size; 30θΑ pore size).

Conditions: linear AB gradient (0.25% B/min from 10% B) at a flow-rate of

0.3ml/min, where Eluent A is 0.2% aq. TFA and Eluent B is 0.18% TFA in

acetonitrile; 30°C.

Table 3 : Relative Hydrophilicity /Hydrophobicity Peptides

References

1. Coast, J.; Smith, R.D.; Millar, M.R., Health Economics 1996, 5, 217-226.

2. Infectious Diseases Society of America., Science Daily 9 December 2008.

(sciencedaily.com/releases/2008/12/081201105706.htm).

3. Garza-Gonzalez, E.; Llaca-Diaz, J.M.; Bosques-Padilla, F.J.; Gonzalez, G.M.,

Chemotherapy 2010, 56, 275-279.

4. Biswas, S.; Brunei, J.M.; Dubus, J.C.; Rolain, J.M., Expert Rev. Anti. Infect. Ther.

2012, 10, 917-34.

5. Yu, Z.; Qin, W.; Lin, 1; Fang, S.; Qin, J., BioMed Res. International 2015

(dx.doi. org/10.1155/2015/679109)

6. Jenssen, H.; Hamill, P.; Hancock, R.E., Clin Microbiol Rev. 2006, 19, 491-511.

7. Wade, D.; Boman, A.; Wahlin, B.; Drain, CM.; Andreu, D.; Boman, H.G.; Merrifield, R.B., Proc. Natl. Acad. Sci. USA 1990, 87, 4761-4765.

8. Cribbs, D.H.; Pike, C.J.; Weinstein, S.L.; Velazquez, P.; Cotman, C.W., J. Biol. Chem.

1997, 272, 7431-7436.

9. Hong, S.Y.; Oh, J.E.; Lee, K.H., Biochem. Pharmacol. 1999, 58, 1775-1780.

10. Wakabayashi, H.; Matsumoto, H.; Hashimoto, K.; Teraguchi, S.; Takase, M.;

Hayasawa, H., Antimicrob. Agents Chemother. 1999, 43, 1267-1269.

1 1. De Lucca, A. J.; Bland, J.M.; Vigo, C.B.; Jacks, T.J.; Peter, J.; Walsh, T.J., Med.

Mycol. 2000, 38, 301-308.

12. Bland, J.M., de Lucca, A.J., Jacks, T.J., Vigo, C.B., Mol. Cell. Biochem. 2001,

218: 105-111.

13. Hamamoto, K.; Kida, Y.; Zhang, Y.; Shimizu, T.; Kuwano, K., Microbiol. Immunol.

2002, 46, 741-749.

14. Elmquist, A.; Langel, U., Biol. Chem. 2003, 384, 387-393.

15. Chen, Y.; Vasil, A.I.; Rehaume, L.; Mant, C.T.; Burns, J.L.; Vasil, M.L.; Hancock, R.E.;

Hodges, R.S., Chem. Biol. Drug Des. 2006, 67, 162-173.

16. Chen, Y.; Mant, C.T.; Farmer, S.W.; Hancock, R.E.; Vasil, M.L.; Hodges, R.S., J.

Biol. Chem. 2005, 280, 12316-12329.

17. Jiang, Z.; Vasil, A.I.; Gera, L.; Vasil, M.L.; Hodges, R.S., Chem. Biol. Drug Des.

2011a, 77, 225-240. 18. Jiang, Z.; Vasil, A.I.; Vasil, M.L.; Hodges, R.S., Pharmaceuticals 2014, 7, 366-391.

19. Chen, Y.; Guarnieri, M.T.; Vasil, A.I.; Vasil, M.L.; Mant, C.T.; Hodges, R.S.,

Antimicrob. Agents Chemother. 2007, 51, 1398-1406.

20. Jiang, Z.; Kullberg, B.J.; van der Lee, H.; Vasil, A.I.; Hale, J.D.; Mant, C.T.; Hancock, R.E.W.; Vasil, M.L.; Netea, M.G.; Hodges, R.S., Chem. Biol. Drug Des. 2008a, 72,

483-495.

21. Jiang, Z.; Higgins, M.P.; Whitehurst, J.; Kisich, K.O.; Voskuil, M.I.; Hodges, R.S., Protein Pept. Lett. 2011b, 18, 241-252.

22. Jiang, Z.; Gera, L.; Mant, C.T.; Hodges, R.S., Proceedings of the 24 th American

Peptide Symposium. In Enabling peptide Research from Basic Research to Drug

Discovery, Orlando, FL (V. Srivastava, A. Yudin and M. Lebl, editors) 2015, pp. 245- 248. Published by the American Peptide Society and Propt Scientific Publishing, 2015).

23. Eisenberg, D.; Weiss, R.M.; Terwilliger, T.C., Nature 1982, 299, 371-374.

24. Kovacs, J.M.; Mant, C.T.; Hodges, R.S., Biopolymers 2006, 84, 283-297.

25. Mant, C.T.; Kovacs, J.M.; Kim, H.M.; Pollock, D.D.; Hodges, R.S., Biopolymers 2009, 92, 573-595.

26. Chen, Y.; Mant, C.T.; Hodges, R.S., J. Peptide Research 2002, 59, 18-33

27. Zhou, N.E.; Mant, C.T.; Hodges, R.S., Peptide Research 1990, 3, 8-20.

28. Jiang, Z.; Vasil, A.I.; Hale, J.D.; Hancock, R.E.; Vasil, M.L.; Hodges, R.S.,

Biopolymers 2008b, 90, 369-383.

29. Lee, D.L.; Mant, C.T.; Hodges, R.S., J. Biol.Chem. 2003, 278,22918-22927.

30. Pouny, Y.; Rapaport, D.; Mor, A.; Nicolas, P.; Shai, Y., Biochemistry 1992, 31 : 12416- 23.