Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
COMPOUNDS AND METHODS OF TREATING INFECTIONS
Document Type and Number:
WIPO Patent Application WO/2014/176636
Kind Code:
A1
Abstract:
The invention provides compounds of Formula (I), and methods of treating or preventing a bacterial infection in a subject using a compound of Formula (I). The invention also provides the use of a compound of Formula (I) in the manufacture of a medicament for the treatment of a bacterial infection in a subject. The invention further provides a medical device when used in a method of treating or preventing a bacterial infection in a subject and to a medical device comprising the composition of the invention.

Inventors:
PAGE STEPHEN (AU)
GARG SANJAY (AU)
KEENAN MARTINE (AU)
MCCLUSKEY ADAM (AU)
STEVENS ANDREW (AU)
Application Number:
PCT/AU2014/000483
Publication Date:
November 06, 2014
Filing Date:
May 01, 2014
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
NEOCULI PTY LTD (AU)
PAGE STEPHEN (AU)
GARG SANJAY (AU)
KEENAN MARTINE (AU)
MCCLUSKEY ADAM (AU)
STEVENS ANDREW (AU)
International Classes:
C07C279/02; A61K31/12; A61K31/155; A61K31/341; A61K31/381; A61K31/4045; A61K31/4192; A61K31/44; A61K31/498; A61P31/04; C07C241/04; C07C249/14; C07C275/68; C07C281/08; C07C281/18; C07C337/08; C07D401/12; C07D403/12; C07D407/12; C07D409/12
Domestic Patent References:
WO2008014266A22008-01-31
WO2008014266A22008-01-31
Foreign References:
US20090285772A12009-11-19
Other References:
DATABASE REGISTRY [online] 3 June 2008 (2008-06-03), "CARBONIMIDIC DIHYDRAZIDE, 2,2'-BIS(2-NAPHTHALENYLMETHYLENE)- (CA INDEX NAME)", XP055294316, retrieved from STN Database accession no. 1024798-17-1
DATABASE REGISTRY [online] 17 August 2006 (2006-08-17), "CARBONIMIDIC DIHYDRAZIDE, 2,2'-BIS[(4-METHYLPHENYL)METHYLENE]- (CA INDEX NAME)", XP055294318, retrieved from STN Database accession no. 902275-25-6
DATABASE REGISTRY [online] 28 January 2002 (2002-01-28), "CARBONIMIDIC DIHYDRAZIDE, BIS[(4-HYDROXY-3-METHOXYPHENYL)METHYLENE]- (9CI)(CA INDEX NAME)", XP055294320, retrieved from STN Database accession no. 387363-92-0
DATABASE REGISTRY [online] 24 June 2009 (2009-06-24), "INDEX NAME NOT YET ASSIGNED", XP055294321, retrieved from STN Database accession no. 1159783-84-2
DATABASE REGISTRY [online] 24 June 2009 (2009-06-24), "INDEX NAME NOT YET ASSIGNED OTHER NAMES:", XP055294324, retrieved from STN Database accession no. 1159808-94-2
DATABASE REGISTRY [online] 26 April 1991 (1991-04-26), "1H-1,2,4-TRIAZOL-5-AMINE, N-(4-CHLOROPHENYL)-3-[4-(TRIFLUOROMETHYL)PHENYL]-(CA INDEX NAME)", XP055294326, retrieved from STN Database accession no. 133476-56-9
MAKKI M.S.T. ET AL: "Synthesis of New Bioactive Sulfur Compounds Bearing Heterocyclic Moiety and Their Analytical Applications", INTERNATIONAL JOURNAL OF CHEMISTRY, vol. 3, no. 1, 2011, pages 181 - 192, XP055294334
TEHRANI K.H.M.E. ET AL: "Synthesis and Antimycobacterial Activity of Symmetric Thiocarbohydrazone Derivatives against Mycobacterium bovis BCG", IRANIAN JOURNAL OF PHARMACEUTICAL RESEARCH, vol. 12, no. 2, 2013, pages 331 - 346, XP055294471
PELTTARI, E. ET AL.: "Carbohydrazones of Substituted Salicylaldehydes as Potential Lead Compounds for the Development of Narrow-Spectrum Antimicrobials", ZEITSCHRIFT FÜR NATURFORSCHUNG C, vol. 62, 2007, pages 483 - 486, XP055294474
PANDEY, A. ET AL.: "Synthesis of Schiff Bases of 2-amino-5-aryl-1,3,4-thiadiazole and Its Analgesic, Anti-Inflammatory and Anti-Bacterial Activity", JOURNAL OF CHEMISTRY, vol. 9, no. 4, 2012, pages 2524 - 2531, XP055294477
HOSNY, M. A. ET AL.: "A New Type of Synthesis of 1,2,3- Thiadiazole and 1,2,3- Diazaphosphole Derivatives Via-Hurd-Mori Cyclization", JOURNAL OF CHEMISTRY, vol. 9, no. 3, pages 1276 - 1287, XP055294478
KUMAR, A. ET AL.: "Syntheses, Antibacterial and Antifungal Screening of Some 1,3,4- Oxadiazole Analogues.", INTERNATIONAL JOURNAL OF PHARMACY AND PHARMACEUTICAL SCIENCES, vol. 4, no. SUPPLE, 2012, pages 440 - 443, XP055328273
SHI, Z. ET AL.: "Synthesis of new deoxycholic acid bis thiocarbazones under solvent-free conditions using microwave irradiation", JOURNAL OF CHEMICAL RESEARCH, vol. 35, no. 4, 2011, pages 198 - 201, XP008182579
KUMAR, S. ET AL.: "Synthesis and biological activity of 5-substituted-2-amino-1,3,4- oxadiazole derivatives", TURKISH JOURNAL OF CHEMISTRY, vol. 35, 2011, pages 99 - 108, XP055294512
ONIGA, O. ET AL.: "Synthesis of Some 2-(Acetophenon-hydrazin)-thiazoles and 2-(4- thiazolyl-methynhydrazin)-thiazoles as potential antibacterial and antifungal agents", FARMACIA, vol. 58, no. 6, 2010, pages 825 - 833, XP055294515
PAVAN, F. R. ET AL.: "Thiosemicarbazones, semicarbazones, dithiocarbazates and hydrazide/hydrazones: Anti - Mycobacterium tuberculosis activity and cytotoxicity", EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY, vol. 45, 2010, pages 1898 - 1905, XP026976509
CASTILLO-GARIT, J. A. ET AL.: "Ligand-based discovery of novel trypanosomicidal drug-like compounds: In silico identification and experimental support", EUROPEAN JOURNAL OF MEDICINAL CHEMISTRY, vol. 46, 2011, pages 3324 - 3330, XP028377954
INFECTIOUS DISEASES SOCIETY OF AMERICA, CLINICAL INFECTIOUS DISEASES,, vol. 50, no. 8, 2010, pages 1081 - 1083
HANSEN ET AL., CHEMOSPHERE, vol. 86, 2012, pages 212 - 215
HANSEN ET AL., ENVIRONMENTAL POLLUTION, vol. 157, 2009, pages 474 - 480
SIEVERT DMRICKS PEDWARDS JR ET AL.: "Antimicrobial-resistant pathogens associated with healthcare-associated infections: summary of data reported to the National Healthcare Safety Network at the Centers for Disease Control and Prevention, 2009-2010", INFECT CONTROL HOSP EPIDEMIOL, vol. 34, 2013, pages 1 - 14
"Remington's Pharmaceutical Sciences", 1985, MACK PUBLISHING COMPANY, pages: 1418
ADVANCED DRUG DELIVERY REVIEWS, vol. 62, pages 491 - 502
AKIYAMA, H.H. KANZAKIY. ABEJ. TADAJ. ARATA: "Staphylococcus aureus infection on experimental croton oil-inflamed skin in mice", JOURNAL OF DERMATOLOGICAL SCIENCE, vol. 8, no. 1, 1994, pages 1 - 10
"CLSI M7-A7", 2006, article "Methods for Dilution Antimicrobial Susceptibility Tests for Bacteria That Grow Aerobically"
"CLSI M31-A2", 2002, article "Performance Standards for Antimicrobial Disk and Dilution Susceptibility Tests for Bacteria Isolated from Animals"
"Performance Standards for Antimicrobial Susceptibility Testing", CLSI M2-A9, 2006
"CLSI document M07-A8", 2009, CLINICAL AND LABORATORY STANDARDS INSTITUTE, article "Methods for Dilution Antimicrobial Susceptibility Tests for Bacteria That Grow Aerobically"
"CLSI document M100-S20", 2010, CLINICAL AND LABORATORY STANDARDS INSTITUTE, article "Performance Standards for Antimicrobial Susceptibility Testing; Nineteenth Informational Supplement"
FRANCIS ET AL., INFECT IMMUN, vol. 69, 2001, pages 3350 - 2358
BRILES ET AL., J. INFEC. DISEASES., vol. 188, 2003, pages 339 - 348
BRILES ET AL., INFECT IMMUN, vol. 68, 2000, pages 796 - 800
TETTLELIN ET AL., SCIENCE, vol. 293, 2001, pages 498 - 506
BRILES ET AL., J. EXP MED., vol. 153, 1981, pages 694 - 705
AVERY ET AL., NATURE REVIEWS MICROBIOLOGY, vol. 8, 2010, pages 260 - 271
HAMMERSCHMIDT, S. ET AL., INFECT IMMUN, vol. 73, 2005, pages 4653 - 4667
BEMIS, D. A.R. D. JONES ET AL.: "Evaluation of cefoxitin disk diffusion breakpoint for detection of methicillin resistance in Staphylococcus pseudintermedius isolates from dogs", JOURNAL OF VETERINARY DIAGNOSTIC INVESTIGATION, vol. 24, no. 5, 2012, pages 964 - 967
GUNICS ET AL., INT. J. ANTIMICROB. AGENTS., vol. 14, 2000, pages 239 - 42
MCKAY ET AL., J. ANTIMICROB. CHEMOTHER., vol. 63, no. 6, 2009, pages 1191 - 1199
STIERITZ, D. D.A. BONDID. MCDERMOTTE. B. MICHAELS: "A burned mouse model to evaluate anti-pseudomonas activity of topical agents", JOURNAL OF ANTIMICROBIAL CHEMOTHERAPY, vol. 9, no. 2, 1982, pages 133 - 140
MCRIPLEY, R. J.R. R. WHITNEY: "Characterization and Quantitation of Experimental Surgical-Wound Infections Used to Evaluate Topical Antibacterial Agents", ANTIMICROBIAL AGENTS AND CHEMOTHERAPY, vol. 10, no. 1, 1976, pages 38 - 44
KUGELBERG, E.T. NORSTROMT. K. PETERSENT. DUVOLDD. I. ANDERSSOND. HUGHES: "Establishment of a Superficial Skin Infection Model in Mice by Using Staphylococcus aureus and Streptococcus pyogenes", ANTIMICROBIAL AGENTS AND CHEMOTHERAPY, vol. 49, no. 8, 2005, pages 3435 - 3441
GUO, Y.R. I. RAMOSJ. S. CHON. P. DONEGANA. L. CHEUNGL. S. MILLER: "In Vivo Bioluminescence Imaging To Evaluate Systemic and Topical Antibiotics against Community-Acquired Methicillin-Resistant Staphylococcus aureus-Infected Skin Wounds in Mice", ANTIMICROBIAL AGENTS AND CHEMOTHERAPY, vol. 57, no. 2, 2013, pages 855 - 863
Attorney, Agent or Firm:
WRAYS (West Perth, Western Australia 6005, AU)
Download PDF:
Claims:
165

CLAIMS

A compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof:

Formula I wherein F , is H, cycloalkyl, Formula II, or Formula III;

Formula II Formula III

wherein R3 is H, NH2, NHNH2, 0-CH2-CH3, NH-CH(0)-phenyl, NH-chlorophenyl, NH-CH2-chlorophenyl, NH-N-CH-cycloalkyl, Formula IV, Formula V or Formula VI;

Formula IV Formula V Formula VI wherein A0 is N, C, CH, or A0 is C and A0 is bonded to R4 to form a triazole ring; wherein Ai is N, C, NH, -(CH)2-N-, -(C6H5)C-CH-N-, or Formula VII;

Substitute Sheet

(Rule 26) RO/AU 166

Formula VII

A2 is N, C, NH, N-C(0)-phenyl or Formula VII; wherein A3, A4, A5, Αβ, A7, As, An , A12, A13, A14, A15, A16 , Ai7, A 8, A19, Α20, A2i A23, A24, A25, A26 and A27 are independently C, O, N, NH, S; wherein A9 is C, O, N, NH, N-C(0)-0-CH2-CH3, N-C(0)-0-CH(CH3)2, N-C(0)-NH- CH2-CH3, N-C(0)-NH-CH2-phenyl, N-C(0)-CH2-CH2-CH2-CH2-CH2-CH3, N-C(O)- CH2-furan-2-yl; wherein A10 is C, NH, -N-(CH)2-, -N-CH-C(C6H5)-; wherein A22 is -CH(CH3)-, -N-CH-, -N-C(CH3)-, N-C(CH2OH)-;

R2 is H, COOH, CH2NH2, CH2OH, CH2NHNH2, methyl, ethyl, propyl, butyl, cyclopentyl, or Formula VII and R2 are R4 are bonded together to form a pyrimidine, pyrazine or triazine ring, or R2 and Rg are bonded together to form a pyrrolidinyl oxindole ring; wherein R4 is N, NH, O, S, or R4 and A0 are bonded together to form a triazole ring, or R4 is N and R4 and R2 are bonded together to form a pyrimidine ring; wherein R7 is H, CI, Br, F, OH, CH3, OCH3, SCH3, CN, CCH, CF3, OCF3, SCF3, N02, butyl, i-butyl, dimethylamino, phenyl, n-propyl, /'-propyl, -NH-C(0)-CH3, - (CH)2-COOH, piperazin-1-yl, or R7 and R8 are bonded together to form a substituted or unsubstituted, saturated or unsaturated aliphatic ring, heterocyclic ring or benzene ring; wherein R6, R8, Ri4, R16, R25 and R27 are independently H, OH, CI, F, Br, CH3,

CN, OCH3, COOH, N02, CF3, R8 and R7 bond together to form a substituted or unsubstituted, saturated or unsaturated aliphatic ring, heterocyclic ring, or benzene ring, R14 and R15 are bonded together to form a substituted or unsubstituted, saturated or unsaturated aliphatic ring, heterocyclic ring or benzene ring, R8 and R9 are bonded together to form a substituted or

Substitute Sheet

(Rule 26) RO/AU 167 unsubstituted, saturated or unsaturated aliphatic ring, heterocyclic ring or benzene ring, or R14 and R13 are bonded together to form a substituted or unsubstituted saturated or unsaturated aliphatic ring, heterocyclic ring or benzene ring; wherein R5, R9, R17, R24 and R28 are independently H, O, OH, CI, F, Br, NH2, CH3, CF3, OCH3, CN, N02, phenyl, -NH-CH(OH)-CH3, -NH-C(0)-CH3, or R9 and R8 are bonded together to form a substituted or unsubstituted, saturated or unsaturated aliphatic ring, heterocyclic ring or benzene ring, or R13 and R14 are bonded together to form a substituted or unsubstituted saturated or unsaturated aliphatic ring, heterocyclic ring or benzene ring; wherein R10, Rn, Ri9, R20, R22 and R23 are independently H, CI, or Br, or R10 and R11 are bonded together to form a substituted or unsubstituted, saturated or unsaturated aliphatic ring, heterocyclic ring or benzene ring, or R 9 and R20 are bonded together to form a substituted or unsubstituted, saturated or unsaturated aliphatic ring, heterocyclic ring or benzene ring, or R22 and R23 are bonded together to form a substituted or unsubstituted, saturated or unsaturated aliphatic ring, heterocyclic ring or benzene ring; wherein R12, R18 and R21 are independently H, COOH, CH2NH2, CH2OH, methyl, ethyl, propyl, butyl, cyclopentyl, or R12 and R13 are bonded together to form a pyrrolidinyl oxindole ring; wherein R15 and R26 are independently H, CI, Br, F, OH, CH3, OCH3, SCH3, CN, CF3, OCF3, SCF3, N02, CCH, n-butyl, f-butyl, dimethylamino, phenyl, n-propyl, /- propyl, -NH-C(0)-CH3, -(CH)2-COOH, piperazin-1-yl, or R15 and R14 are bonded together to form a substituted or unsubstituted, saturated or unsaturated aliphatic ring, heterocyclic ring or benzene ring; and wherein "— " is a double bond or a single bond.

2. A compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, selected from the compounds presented in Figure 1.

3. A compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein A0 is C; wherein 1 is N; or Formula VII;

Substitute Sheet

(Rule 26) RO/AU 168 wherein A2 is N; or NH; wherein A3j A4, Ae, A7, An , A12, A14, A15, are N; or C; wherein A5, A13, A23, A24, A25, A26 and A27 are C; wherein A8 and A21 are S; wherein A9 is NH; wherein A10 is N; wherein A22 is -N-CH-; -N-C(CH3)-; or -N-C(CH2OH)-; wherein is H; Formula II; Formula III; cycloalkyl; wherein R2 is H; methyl; ethyl; CH2NHNH2; CH2OH; butyl; cyclopentyl; or Formula VII and R2 is bonded to R4, to form a pyrimidine ring; wherein R3 is NH2; Formula IV; Formula V; Formula VI; NH2, NH-N-CH- cycloalkyl; or 0-CH2-CH3; wherein R4 is NH; O; S; or R4 is N and R4 and R2 are bonded together to form a pyrimidine ring; wherein R7 is H; F; CI; CF3; methyl; R7 and R8 are bonded together to form an unsubstituted, benzene ring; OH; i-butyl; phenyl; dimethylamino; /-propyl; n- propyl; CN; CCH; n-butyl; SCH3; R7 and R8 are bonded together to form an unsubstituted, unsaturated heterocyclic ring; OCH3; Br; OCF3; piperazin-1-yl; or SCF3; wherein R6, R8, R14, and R16 are independently H; OH; F; OCH3; CF3; methyl; CI; CN; Br; R8 and R7 are bonded together to form an unsubstituted, benzene ring; R8 and R are bonded together to form an unsubstituted, unsaturated heterocyclic ring; R14 and R15 are bonded together to form an unsubstituted, benzene ring; or R14 and R15 are bonded together to form an unsubstituted, unsaturated heterocyclic ring; wherein R5, R9, R13, and R17 are independently H; OH; NH2; CI; F; OCH3; OH; - NH-CH(OH)-CH3; wherein R12 is H; methyl; ethyl; CH2OH; or cyclopentyl;

Substitute Sheet

(Rule 26) RO/AU 169

wherein R 5 is H; F; CI; CF3; methyl; R7 and R8 are bonded together to form an unsubstituted, benzene ring; OH; f-butyl; phenyl; dimethylamino; /-propyl; n- propyl; CN; CCH; n-butyl; SCH3; R15 and R14 are bonded together to form an unsubstituted, unsaturated heterocyclic ring; OCH3; Br; OCF3; piperazin-1-yl; or SCF3; wherein R24 and R28 are independently H; OH; or CI; wherein R25 and R2 are independently H; or OH; wherein R26 is H; CH3; Br; CI; OH; dimethylamino; -0-P(0)(OEt)2; CF3; or F; and wherein "— " is independently a single or a double bond.

4. A compound according to any one of the preceding claims, wherein the compound is

and NCL223.

5. The compound according to any one of the preceding claims, wherein the compound is selected from the group comprising: NCL040; NCL078; NCL079; NCL080; NCL081 ; NCL084; NCL088; NCL089; NCL097; NCL099; NCL123; NCL146; NCL157; NCL158; NCL177; NCL179; NCL188; NCL193; NCL195; NCL196; NCL197; NCL199; NCL202; NCL204; NCL205; NCL215; NCL216; NCL217; NCL219; and NCL221.

6. The compound according to any one of the preceding claims, wherein the compound is selected from the group comprising: NCL078; NCL079; NCL080; NCL081 ; NCL084;

Substitute Sheet

(Rule 26) RO/AU 170

NCL089; NCL097; NCL157; NCL158; NCL179; NCL188; NCL193; NCL195; NCL196; NCL199; NCL204; NCL216; NCL217; NCL219; and NCL221.

7. The compound according to any one of the preceding claims, wherein the compound is selected from the group comprising: NCL089; NCL097; NCL157; NCL179; NCL188; NCL193; NCL195; NCL196; NCL216; NCL219; and NCL221.

8. The compound according to any one of the preceding claims, wherein the compound is selected from the group comprising: NCL097; NCL157; NCL179; NCL188; NCL195; and NCL196.

9. The compound according to any one of the preceding claims, wherein the compound is a chloride salt.

10. The compound according to any one of the preceding claims, wherein the compound is not a compound selected from the group comprising: NCL812, NCL001 , NCL002, NCL003, NCL004, NCL005, NCL006, NCL007, NCL010, NCL011 , NCL012, NCL013, NCL014, NCL015, NCL016, NCL017, NCL018, NCL019, NCL020, NCL021 , NCL022, NCL024, NCL027, NCL028, NCL030, NCL031 , NCL032, NCL033, NCL034, NCL035, NCL038, NCL041 , NCL042, NCL043, NCL044, NCL045, NCL046, NCL047, NCL048, NCL049, NCL051 , NCL052, NCL053, NCL054, NCL055, NCL056, NCL057, NCL058, NCL059, NCL060, NCL062, NCL063, NCL066, NCL067, NCL069, NCL070, NCL071 , NCL072, NCL073, NCL074, NCL077, NCL082, NCL083, NCL087, NCL091 , NCL093, NCL096, NCL100, NCL102, NCL103, NCL107, NCL109, NCL110, NCL 13, NCL117, NCL120, NCL127, NCL128, NCL129, NCL134, NCL142, NCL143, NCL149, NCL151 , NCL155, NCL160, NCL163, NCL175, NCL182, NCL191 , NCL192, and NCL214.

11. A method of treating or preventing a bacterial colonisation or infection in a subject, the method including the step of administering a therapeutically effective amount of a compound of any one of claims 1 to 10, or a therapeutically acceptable salt thereof, to the subject, wherein the bacterial infection is caused by a bacterial agent.

12. The method according to claim 11 , wherein the subject is an animal most preferably selected from the group comprising: human, canine, feline, bovine, ovine, caprine, porcine, avian, piscine and equine species.

13. The method according to either claim 11 or claim 12, wherein the compound is administered to the subject in a dose in the range of 0.1 mg/kg to 250 mg/kg bodyweight.

14. The method according to any one of claims 11 to 13, wherein the bacterial agent is gram positive.

Substitute Sheet

(Rule 26) RO/AU 171

15. The method according to claim 14, wherein the bacterial agent is selected from the group comprising: Abiotrophia defectiva, Acholeplasma spp., Actinobaculum suis, Actinomyces bovis, Actinomyces europaeus, Actinomyces georgiae, Actinomyces gerencseriae, Actinomyces graevenitzii, Actinomyces hordeovulneris, Actinomyces israelii serotype II, Actinomyces israelii, Actinomyces meyeri, A ctinomyces naeslundii, Actinomyces neuii, Actinomyces odontolyticus, Actinomyces radingae, Actinomyces spp, Actinomyces turicensis, Actinomyces viscosus, Alloscardovia omnicolens, Anaerococcus hydrogenalis, Anaerococcus lactolyticus, Anaerococcus murdochii, Anaerococcus octavius, Anaerococcus prevotii, Anaerococcus tetradius, Anaerococcus vaginalis, Arcanobacterium (Actinomyces) bernardiae, Arcanobacterium (Actinomyces) pyogenes, Arcanobacterium bernardiae, Arcanobacterium cardiffensis, Arcanobacterium funkei, Arcanobacterium haemolyticum, Arcanobacterium houstonensis, Arcanobacterium lingnae, Arcanobacterium pyogenes (Actinomyces pyogenes), Arthrobacter, Atopobium minutum, Atopobium parvulum, Atopobium rimae, Atopobium spp, Atopobium vaginae, Bacillus anthracis, Bacillus cereus, Bacillus circulans, Bacillus licheniformis, Bacillus megaterium, Bacillus melaninogenicus, Bacillus pumilus, Bacillus sphaericus, Bacillus subtilis, beta haemolytic Steptococcus spp, Bifidobacteria adolescentis, Bifidobacteria dentium, Bifidobacteria scardovii, Bifidobacteria, Brevibacillus brevis, Brevibacillus laterosporus, Brevibacterium, Bulleidia extructa, Catabacter hongkongensis, CDC coryneform groups F-1 and G, Clostridiium tetani, Clostridium baratii, Clostridium bifermentans, Clostridium botulinum (types A, B, C, D, E, F, G), Clostridium botulinum, Clostridium butyricum, Clostridium chauvoei, Clostridium colinum, Clostridium difficile, Clostridium haemolyticum, Clostridium histolyticum, Clostridium novyi type A, Clostridium novyi type B, Clostridium novyi, Clostridium perfringens type A, Clostridium perfringens types A-E, Clostridium perfringens, Clostridium piliforme, Clostridium ramosum, Clostridium septicum, Clostridium sordelli, Clostridium sphenoides, Clostridium spiroforme, Clostridium spp, Clostridium tertium, Clostridium tetani, Collinsella aerofaciens, Corynebacterium accolens, Corynebacterium afermentans afermentans, Corynebacterium afermentans lipophilum, Corynebacterium amycolatum, Corynebacterium argentoratense, Corynebacterium aurimucosum, Corynebacterium auris, Corynebacterium bovis, Corynebacterium confusum, Corynebacterium cystidis, Corynebacterium diphtheria, Corynebacterium freneyi, Corynebacterium glucuronolyticum, Corynebacterium jeikeium, Corynebacterium kroppenstedtii, Corynebacterium kutscheri, Corynebacterium lipophiloflavum, Corynebacterium macginleyi, Corynebacterium matruchotii, orynebacterium minutissimum, Corynebacterium pilosum, Corynebacterium propinquum, Corynebacterium pseudodiphtheriticum, Corynebacterium pseudotuberculosis, Corynebacterium renale,

Corynebacterium riegelii, Corynebacterium simu!ans, Corynebacterium striatum,

Substitute Sheet

(Rule 26) RO/AU 172

Corynebacterium sundvallense, Corynebacterium thomssensii, Corynebacterium tuberculostearum, Corynebacterium ulcerans, Corynebacterium urealyticum, Corynebacterium xerosis, Crossiella equi, Dermabacter, Dermatophilus congolense, Dermatophilus congolensis, Eggerthella brachy, Eggerthella hongkongensis, Eggerthella infirmum, Eggerthella lenta, Eggerthella minutum, Eggerthella nodatum, Eggerthella saphenum, Eggerthella sinensis, Eggerthella sulci, Eggerthella tenue, Eggerthella, Enterococcus avium, Enterococcus bovis, Enterococcus casseliflavus/flavescens, Enterococcus cecorum, Enterococcus dispar, Enterococcus durans, Enterococcus faecalis, Enterococcus faecium, Enterococcus gallinarum, Enterococcus gilvus, Enterococcus hirae, Enterococcus italicus, Enterococcus malodoratus, Enterococcus mundtii, Enterococcus pallens, Enterococcus pseudoavium, Enterococcus raffinosus, Enterococcus sanguinicola, Enterococcus spp, Erysipelothrix rhusiopathiae, Eubacterium, Filifactor alocis, Finegoldia magna, Gallicola barnesae, Gemella asaccharolytica, Gemella bergeri, Gemella cuniculi, Gemella haemolysans, Gemella morbillorum, Gemella palaticanis, Gemella sanguinis, Gordonia spp., Granulicatella adiacens, Granulicatella elegans, Granulicatella para-adiacens, Kytococcus schroeteri, Lactobacillus acidophilus, Lactobacillus casei, Lactobacillus fermentum, Lactobacillus gasseri, Lactobacillus iners, Lactobacillus plantarum, Lactobacillus rhamnosus, Lactobacillus species, Lactobacillus ultunensis, Leifsonia aquatic, Leuconostoc citreum, Leuconostoc lactis, Leuconostoc mesenteroides, Leuconostoc paramesenteroides, Leuconostoc pseudomesenteroides, Listeria grayi, Listeria innocua, Listeria ivanovii, Listeria monocytogenes, Listeria seeligeri, Listeria welshimeri, Microbacterium,M obiluncus curtisii, Mobiluncus mulieris, Mobiluncus spp, Mogibacterium timidum, Mogibacterium vescum, Moryella indoligenes, Mycobacterium senegalense, Mycobacterium abscessus, Mycobacterium africanum, Mycobacterium arupense, Mycobacterium asiaticum, Mycobacterium aubagnense, Mycobacterium avium complex, Mycobacterium avium subsp paratuberculosis, Mycobacterium avium, Mycobacterium bolletii, Mycobacterium bovis, Mycobacterium branderi, Mycobacterium canettii, Mycobacterium caprae, Mycobacterium celatum, Mycobacterium chelonae, Mycobacterium chimaera, Mycobacterium colombiense, Mycobacterium conceptionense, Mycobacterium conspicuum, Mycobacterium elephantis, Mycobacterium farcinogenes, Mycobacterium florentinum, Mycobacterium fortuitum group, Mycobacterium genavense, Mycobacterium goodii, Mycobacterium haemophilum, Mycobacterium heckeshornense, Mycobacterium heidelbergense, Mycobacterium houstonense, Mycobacterium immunogenum, Mycobacterium interjectum, Mycobacterium intracellulare, Mycobacterium kansasii, Mycobacterium lacus, Mycobacterium lentiflavum, Mycobacterium leprae, Mycobacterium lepraemurium, Mycobacterium mageritense,

Mycobacterium malmoense, Mycobacterium marinum, Mycobacterium massiliense,

Substitute Sheet

(Rule 26) RO/AU 173

Mycobacterium microti, Mycobacterium montefiorense (eels), Mycobacterium moracense, Mycobacterium mucogenicum, Mycobacterium nebraskense,

Mycobacterium neoaurum, Mycobacterium novocastrense, Mycobacterium palustre,

Mycobacterium paratuberculosis (Johne's Disease), Mycobacterium parmense,

Mycobacterium phlei, Mycobacterium phocaicum, Mycobacterium pinnipedii,

Mycobacterium porcinum, Mycobacterium pseudoshottsii (fish), Mycobacterium pseudotuberculosis, Mycobacterium saskatchewanense, Mycobacterium scrofulaceum,

Mycobacterium senuense, Mycobacterium septicum, Mycobacterium simiae,

Mycobacterium smegmatis, Mycobacterium spp, Mycobacterium szulgai, Mycobacterium terrae/chromogenicum complex, Mycobacterium triplex, Mycobacterium tuberculosis,

Mycobacterium tusciae, Mycobacterium ulcerans, Mycobacterium wolinskyi,

Mycobacterium xenopi, Mycobacterium, Nocardia asteroides, Nocardia brasiHensis,

Nocardia farcinica, Nocardia nova, Nocardia otitidiscaviarum, Nocardia spp, Nocardia transvalensis, Oerskovia, Olsenella oral spp, Olsenella profuse, Olsenella uli,

Oribacterium sinus, Paenibacillus alvei, Parvimonas micra, Pediococcus, Peptococcus indolicus, Peptococcus niger, Peptoniphilus asaccharolyticus, Peptoniphilus gorbachii,

Peptoniphilus harei, Peptoniphilus indolicus, Peptoniphilus ivorii, Peptoniphilus lacrimalis, Peptoniphilus olsenii, Peptostreptococcus anaerobius, Peptostreptococcus stomatis, Propionibacterium acnes, Propionibacterium granulosum, Propionibacterium propionicum, Propionibacterium, Pseudoramibacter alactolyticus, Rhodococcus equi,

Rhodococcus erythropolis, Rhodococcus fasciens, Rhodococcus rhodochrous, Rothia,

Ruminococcus productus, Slackia exigua, Slackia heliotrinireducens, Solobacterium moorei, Staphylococcus arlettae, Staphylococcus aureus subsp. anaerobius,

Staphylococcus aureus, Staphylococcus auricularis, Staphylococcus capitis subsp. capitis, Staphylococcus capitis subsp. urealyticus, Staphylococcus capitis,

Staphylococcus caprae, Staphylococcus carnosus, Staphylococcus caseolyticus,

Staphylococcus chromogenes, Staphylococcus cohnii subsp. cohnii, Staphylococcus cohnii subsp. urealyticus, Staphylococcus cohnii, Staphylococcus condimenti,

Staphylococcus delphini, Staphylococcus epidermidis, Staphylococcus equorum,

Staphylococcus felis, Staphylococcus fleurettii, Staphylococcus gallinarum,

Staphylococcus haemolyticus, Staphylococcus hominis, Staphylococcus hyicus,

Staphylococcus intermedius, Staphylococcus kloosii, Staphylococcus lentus,

Staphylococcus lugdunensis, Staphylococcus lutrae, Staphylococcus muscae,

Staphylococcus nepalensis, Staphylococcus pasteuri, Staphylococcus pettenkoferi,

Staphylococcus piscifermentans, Staphylococcus pseudintermedius, Staphylococcus pulvereri, Staphylococcus saccharolyticus, Staphylococcus saprophytics,

Staphylococcus schleiferi subsp. coagulans, Staphylococcus schleiferi, Staphylococcus sciuri, Staphylococcus simiae, Staphylococcus simulans, Staphylococcus spp,

Substitute Sheet

(Rule 26) RO/AU 174

Staphylococcus succinus, Staphylococcus vitulinus, Staphylococcus warneri, Staphylococcus xylosus, Staphylococcus vitulinus, Stomatococcus mucilaginosus (reclassified as Rothia mucilaginosa), Streptococcus agalactiae, Streptococcus anginosus species group (Streptococcus intermedius, Streptococcus constellatus, and Streptococcus anginosus), Streptococcus bovis species group (S. gallolyticus subsp. gallolyticus (formerly S. bovis biotype I), Streptococcus bovis, Streptococcus canis, Streptococcus dysgalactiae subsp. dysgalactiae, S. equi subsp. equi, S. equi subsp. zooepidemicus, S. porcinus, S. canis, S. suis, S. iniae), Streptococcus dysgalactiae subsp. equisimilis, Streptococcus dysgalactiae, Streptococcus equi (Streptococcus equi subsp equi), Streptococcus equi subsp. zooepidemicus, Streptococcus equi, Streptococcus equinus, Streptococcus equisimilis (Streptococcus dysgalactiae subsp equisimilis), Streptococcus gallolyticus subsp. pasteurianus (formerly S. bovis biotype 11/2), Streptococcus infantarius subsp Infantarius, Streptococcus lutetiensis (formerly S. bovis biotype 11/1), Streptococcus mitis species group (S. cristatus , S. infantis, S. mitis, S. oralis, S. peroris, S. orisratti), and Streptococcus mutans species group (S. cricetus, S. downei, S. ferus, S. hyovaginalis, S. macaccae, S. mutans, S. ratti, S. sobrinus, Sanguinis Group, S. gordonii, S. parasanguinis, S. sanguinis), Streptococcus pneumoniae, Streptococcus porcinus, Streptococcus pyogenes, Streptococcus salivarius species group (S. alactolyticus, S. hyointestinalis, S. infantarius, S. salivarius, S. thermophilus, S. vestibularis), Streptococcus spp, Streptococcus suis, Streptococcus uberis, Streptococcus zooepidemicus (Streptococcus equi subsp zooepidemicus), Streptococcus zooepidemicus, Trueperella abortisuis, Trueperella bernardiae, Trueperella bialowiezensis, Trueperella bonasi, Trueperella pyogenes (Arcanobacterium pyogenes), Tsukamurella spp., Turicella, and Turicibacter sanguine.

16. The method according to claim 15, wherein the bacterial agent is selected from the group comprising: Staphylococcus aureus, Staphylococcus pseudintermedius, Streptococcus pneumoniae, Streptococcus pyogenes, Streptococcus agalactiae, Enterococcus faecium, Enterococcus faecalis, and Clostridium difficile.

17. The method according to any one of claims 1 1 to 13, wherein the bacterial agent is gram negative.

18. The method according to claim 17, wherein the bacterial agent is selected from the group comprising: Acetobacteraceae:- Roseomonas cervicalis; Roseomonas fauriae; Roseomonas gilardii. - Aeromonadaceae:- Aeromonas allosaccharophila; Aeromonas aquariorum; Aeromonas caviae; Aeromonas hydrophila (and subspecies); Aeromonas salmonicida; Aeromonas shubertii; Aeromonas veronii biovar sobria (Aeromonas sobria).

— Alcaligenaceae:- Achromobacter xylosoxidans; Alcaligenes faecalis; Bordetella

Substitute Sheet

(Rule 26) RO/AU 175 ansorpii; Bordetella avium; Bordetella bronchiseptica; Bordetella hinzii; Bordetella holmesii; Bordetella parapertussis; Bordetella pertussis; Bordetella petrii; Bordetella trematum; Oligella ureolytica; Oligella urethralis. - Anaplasmataceae:- Anaplasma phagocytophilum; Anaplasma platys; Anaplasma bovis; Anaplasma centrale; Anaplasma marginale; Anaplasma odocoilei; Anaplasma ovis; Ehrlichia canis; Ehrlichia chaffeensis; Ehrlichia ewingii; Ehrlichia muris; Ehrlichia ovina; Ehrlichia ruminantium; Neoehrlichia lotoris; Neoehrlichia mikurensis; Neorickettsia helminthoeca; Neorickettsia risticii; Neorickettsia sennetsu; Wolbachia pipientis. - Armatimonadaceae:- Armatimonas rosea. - Bacteroidaceae:- Bacteroides forsythus; Bacteroides fragilis; Bacteroides melaninogenicus; Bacteroides ruber; Bacteroides urealtyicus. - Bartonellaceae:- Bartonella alsatica; Bartonella australis; Bartonella bacilliformis; Bartonella birtlesii; Bartonella bovis; Bartonella capreoli; Bartonella chomelii; Bartonella clarridgeiae; Bartonella doshiae; Bartonella elizabethae; Bartonella grahamii; Bartonella henselae; Bartonella koehlerae; Bartonella peromysci; Bartonella phoceensis; Bartonella quintana; Bartonella rattimassiliensis; Bartonella rochalimae; Bartonella schoenbuchensis; Bartonella talpae; Bartonella tamiae; Bartonella taylorii; Bartonella tribocorum; Bartonella vinsonii subsp . berkhoffii; Bartonella vinsonii subsp. arupensis; Bartonella vinsonii subsp. vinsonii. - Bdellovibrionaceae:- Bdellovibrio spp. ~ Brachyspiraceae:- Brachyspira spp including Brachyspira hampsonii, Brachyspira hyodysenteriae, Brachyspira murdochii, Brachyspira pilosicoli. - Brucellaceae:- Brucella abortus; Brucella canis; Brucella ceti; Brucella melitensis; Brucella ovis; Brucella pinnipedialis; Brucella suis; Ochrobactrum anthropi; Ochrobactrum intermedium. - Burkholderiaceae:- Burkholderia aboris; Burkholderia ambifaria (genomovar VII); Burkholderia anthina (genomovar VIII); Burkholderia cenocepacia (genomovar III); Burkholderia cepacia (genomovar I); Burkholderia diffusa; Burkholderia dolosa (genomovar VI); Burkholderia latens; Burkholderia mallei; Burkholderia metallica; Burkholderia multivorans (genomovar II); Burkholderia pseudomallei; Burkholderia pyrrocinia (genomovar IX); Burkholderia seminalis; Burkholderia stabilis (genomovar IV); Burkholderia ubonensis (genomovar X); Burkholderia vietnamiensis (genomovar V); Cupriavidus pauculus; Cupriavidus gilardii; Ralstonia pickettii; Ralstonia mannitolilytica; Sphaerotilus hippei; Sphaerotilus montanus; Sphaerotilus natans. - Campylobacteraceae:- Arcobacter spp includng Arcobacter skirrowii; Campylobacter coli; Campylobacter concisus; Campylobacter curvus; Campylobacter fetus; Campylobacter gracilis; Campylobacter helveticus; Campylobacter hominis; Campylobacter hyointestinalis; Campylobacter insulaenigrae; Campylobacter jejuni; Campylobacter lanienae; Campylobacter lari; Campylobacter laridis; Campylobacter mucosalis; Campylobacter rectus; Campylobacter showae; Campylobacter sputorum; Campylobacter upsaliensis. - Candidatus:- Piscichlamydia salmonis. - Cardiobacteriaceae;- Cardiobacterium hominis; Cardiobacterium valvarum;

Substitute Sheet

(Rule 26) RO/AU 176

Dichelobacter nodosus. - Chlamydiaceae:- Chlamydia spp including Chlamydia avium, Chlamydia gallinacea, Chlamydia muridarum, Chlamydia suis, Chlamydia trachomatis; Chlamydophila spp including Chlamydophila pneumoniae, Chlamydophila pecorum, Chlamydophila psittaci, Chlamydophila abortus, Chlamydophila caviae, and Chlamydophila felis. - Chthonomonadaceae:- Chthonomonas calidirosea. - Comamonadaceae:- Comamonas testosteroni; Verminephrobacter spp. - Coxiellaceae:- Coxiella burnetii. - Cytophagaceae:- Cytophaga columnaris; Cytophaga hutchinsonii; Flexibacter echinicida; Flexibacter elegans; Flexibacter flexilis; Flexibacter litoralis; Flexibacter polymorphus; Flexibacter roseolus; Flexibacter ruber. Desulfovibrionaceae:- Bilophila wadsworthia; Lawsonia intracellularis. Enterobacteriaceae:- Cedecea davisae; Cedecea lapagei; Cedecea neteri; amalonaticus; Citrobacter diversus; Citrobacter freundii; Citrobacter koseri; Cronobacter condimenti; Cronobacter dublinensis; Cronobacter helveticus; Cronobacter malonaticus; Cronobacter muytjensii; Cronobacter pulveris; Cronobacter sakazakii; Cronobacter turicensis; Cronobacter universalis; Cronobacter zurichensis; Edwardsiella ictaluri; Edwardsiella tarda; Enterobacter aerogenes; Enterobacter agglomerans; Enterobacter cloacae; Enterobacter cowanii; Escherichia albertii; Escherichia coli, including A I EC = adherent invasive E. coli, EaggEC = enteroaggregative E. coli; EHEC = enterohemorrhagic E. coli; EIEC = enteroinvasive E. coli; EPEC = enteropathogenic E. coli; ETEC = enterotoxigenic E. coli; ExPEC = extraintestinal pathogenic E. coli, NMEC = neonatal meningitis E. coli, NTEC = necrotoxigenic E. coli, OPEC = uropathogenic E. coli.; Escherichia fergusonii; Ewingella americana; Hafnia alvei; Hafnia paralvei; Klebsiella granulomatis; Klebsiella oxytoca; Klebsiella pneumoniae; Kluyvera ascorbata; Kluyvera cryocrescens; Morganella morganii; Pantoea (formerly Enterobacter) agglomerans; Photorhabdus asymbiotica; Plesiomonas shigelloides; Proteus mirabilis; Proteus penneri; Proteus vulgaris; Providencia alcalifaciens; Providencia rettgeri; Providencia stuartii; Raouitella electrica; Raouitella ornithinolytica; Raouitella planticola; Raouitella terrigena; Salmonella bongori, Salmonella enterica subspecies enterica (many serotypes); Serratia liquifaciens; Serratia marcesans; Shigella boydii; Shigella dysenteriae; Shigella flexneri; Shigella sonnei; Yersinia enterocolitica; Yersinia pestis; Yersinia pseudotuberculosis; Yersinia ruckeri. - Fimbriimonadaceae:- Fimbriimonas ginsengisoli. - Flavobacteriaceae:- Bergeyella zoohelcum; Capnocytophaga canimorsus; Capnocytophaga cynodegmi; Capnocytophaga gingivalis; Capnocytophaga granulosa; Capnocytophaga haemolytica; Capnocytophaga leadbetteri; Capnocytophaga ochracea; Capnocytophaga sputigena; Chryseobacterium indologenes; Chryseobacterium piscicola; Elizabethkingia meningoseptica; Flavobacterium branchiophilum; Flavobacterium columnare; Flavobacterium oncorhynchi;

Flavobacterium piscicida; Flavobacterium psychrophilum; Myroides odoratus; Myroides

Substitute Sheet

(Rule 26) RO/AU 177 odoratimimus; Ornithobacterium rhinotracheale; Riemerella anatipestifer; Riemerella columbina; Riemerella columbipharyngis; Tenacibaculum dicentrarchi; Tenacibaculum discolour; Tenacibaculum gallaicum; Tenacibaculum maritimum; Tenacibaculum soleae; Weeksella virosa. — Francisellaceae:- Francisella tularensis subsp. tularensis; Francisella tularensis subsp. holarctica; Francisella tularensis subsp. novicida; Francisella philomiragia; Francisella noatunensis; Francisella noatunensis subsp. orientalis (also termed Francisella asiatica). - Fusobacteriaceae:- Fusobacterium spp. including Fusobacterium necrophorum, Fusobacterium nucleatum, Fuso-bacterium polymorphum. - Helicobacteraceae:- Helicobacter cinaedi; Helicobacter fennelliae; Helicobacter pylori. - Legionellaceae:- Legionella pneumophila and other species including; Legionella anisa; Legionella birminghamensis; Legionella bozemannii; Legionella cincinnatiensis; Legionella dumoffii; Legionella feeleii; Legionella gormanii; Legionella hackeliae; Legionella jordanis; Legionella lansingensis; Legionella longbeachae; Legionella maceachernii; Legionella micdadei; Legionella oakridgensis; Legionella parisiensis; Legionella sainthelens; Legionella tusconensis; Legionella wadsworthii; Legionella waltersii. - Leptospiraceae:- Leptospira alexanderi (including Leptospira alexanderi serovar Hebdomadis, Leptospira alexanderi serovar Manhao 3); Leptospira alstoni (including Leptospira alstoni serovar Pingchang, Leptospira alstoni serovar Sichuan); Leptospira biflexa (including Leptospira biflexa serovar Ancona, Leptospira biflexa serovar Canela); Leptospira borgpetersenii (including Leptospira borgpetersenii serovar Hardjo, Leptospira borgpetersenii serovar Hardjo-bovis, Leptospira borgpetersenii serovar Pomona, Leptospira borgpetersenii serovar Tarassovi); Leptospira broomii (including Leptospira broomii serovar Hurstbridge); Leptospira fainei (including Leptospira fainei serovar Hurstbridge); Leptospira idonii; Leptospira inadai (including Leptospira inadai serovar Lyme, Leptospira inadai serovar Malaya); Leptospira interrogans (including Leptospira interrogans serovar Australis, Leptospira interrogans serovar Autumnalis, Leptospira interrogans serovar Bratislava, Leptospira interrogans serovar Canicola, Leptospira interrogans serovar Grippotyphosa, Leptospira interrogans serovar Hardjo, Leptospira interrogans serovar Hardjo-bovis, Leptospira interrogans serovar lcterohaemorrhagiae, Leptospira interrogans serovar Pomona, Leptospira interrogans serovar Pyrogenes, Leptospira interrogans serovar Tarassovi); Leptospira kirschneri (including Leptospira kirschneri serovar Bulgarica, Leptospira kirschneri serovar Cynopteri, Leptospira kirschneri serovar Grippotyphosa); Leptospira kmetyi; Leptospira licerasiae; Leptospira meyeri (including Leptospira meyeri serovar Sofia); Leptospira noguchii (including Leptospira noguchii serovar Panama, Leptospira noguchii serovar Pomona); Leptospira santarosai; Leptospira terpstrae; Leptospira vanthielii; Leptospira weilii (including Leptospira weilii serovar Celledoni,

Leptospira weilii serovar Sarmin); Leptospira wolbachii; Leptospira wolffii; Leptospira

Substitute Sheet

(Rule 26) RO/AU 178

yanagawae. - Leptotrichiaceae:- Leptotrichia buccalis; Streptobacillus moniliformis. - Methylobacteriaceae:- Methylobacterium extorquens group; Methylobacterium fujisawaense; Methylobacterium mesophilicum; Methylobacterium zatmanii. Moraxellaceae:- Acinetobacter baumannii (genomic species 2); Acinetobacter baylyi; Acinetobacter bouvetii; Acinetobacter calcoaceticus (genomic species 1); Acinetobacter gerneri; Acinetobacter grimontii; Acinetobacter haemolyticus (genomic species 4); Acinetobacter johnsonii (genomic species 7); Acinetobacter junii (genomic species 5); Acinetobacter Iwoffi (genomic species 8/9); Acinetobacter parvus; Acinetobacter radioresistens (genomic species 12); Acinetobacter schindleri; Acinetobacter tandoii; Acinetobacter tjernbergiae; Acinetobacter towneri; Acinetobacter ursingii; Acinetobacter venetianus; Moraxella atlantae; Moraxella boevrei; Moraxella bovis; Moraxella bovoculi; Moraxella canis; Moraxella caprae; Moraxella catarrhalis; Moraxella caviae; Moraxella cuniculi; Moraxella equi; Moraxella lacunata; Moraxella lincolnii; Moraxella macacae; Moraxella nonliquefaciens; Moraxella oblonga; Moraxella osloensis; Moraxella ovis; Moraxella phenylpyruvica; Moraxella pluranimalium; Moraxella porci. - Moritellaceae:- Moritella abyssi; Moritella dasanensis; Moritella japonica; Moritella marina; Moritella profunda; Moritella viscosa; Moritella yayanosii. - Neisseriaceae:- Chromobacterium violaceum; Eikenella corrodens; Kingella denitrificans, Kingella kingae, Kingella oralis, Kingella potus; Neisseria cinerea; Neisseria elongata; Neisseria flavescens; Neisseria gonorrhoeae; Neisseria lactamica; Neisseria meningitidis; Neisseria mucosa; Neisseria polysaccharea; Neisseria sicca; Neisseria subflava; Neisseria weaver; Vitreoscilla spp. - Nitrosomonadaceae:- Nitrosomonas eutropha; Nitrosomonas halophila; Nitrosomonas oligotropha. - Pasteurellaceae:- Actinobacillus actinomycetemcomitans; Actinobacillus equuli; Actinobacillus lignieresii; Actinobacillus pleuropneumoniae; Actinobacillus seminis; Actinobacillus succinogenes; Actinobacillus ureae; Aggregatibacter actinomycetemcomitans, Aggregatibacter segnis, Aggregatibacter aphrophilus; Avibacterium avium; Avibacterium endocarditidis; Avibacterium gailinarum; Avibacterium paragallinarum; Avibacterium volantium; Bibersteinia trehalose; Gallibacterium anatis; Gallibacterium genomospecies 1; Gallibacterium genomospecies 2; Gallibacterium genomospecies 3; Gallibacterium group V; Gallibacterium melopsittaci; Gallibacterium salpingitidis; Gallibacterium trehalosifermentans; Haemophilus aegyptius; Haemophilus avium; Haemophilus ducreyi; Haemophilus haemolyticus; Haemophilus influenzae; Haemophilus parahaemolyticus; Haemophilus parainfluenzae; Haemophilus parasuis; Histophilus somni; Mannheimia caviae; Mannheimia glucosida; Mannheimia granulomatis; Mannheimia haemolytica; Mannheimia ruminalis; Mannheimia varigena; Nicoletella semolina; Pasteurella aerogenes; Pasteurella bettyae; Pasteurella caballi; Pasteurella canis; Pasteurella dagmatis; Pasteurella multocida (subspecies multocida, septicum, gallicida); Pasteurella nneumotropica; Pasteurella stomatis; Pasteurella

Substitute Sheet

(Rule 26) RO/AU 179 trehalosi. - Piscirickettsiaceae:- Piscirickettsia salmonis. - Plesiomonadaceae:- Plesiomonas shigelloides. — Polyangiaceae:- Sorangium cellulosum. — Porphyromonadaceae:- Dysgonomonas capnocytophagoides; Dysgonomonas gadei; Dysgonomonas hofstadii; Dysgonomonas mossii; Dysgonomonas oryzarvi; Dysgonomonas wimpennyi; Porphyromonas gingivalis. — Prevotellaceae:- Prevotella spp.including Prevotella intermedia, Prevotella melaninogenica. - Pseudomonadaceae:- Chryseomonas luteola; Pseudomonas aeruginosa; Pseudomonas luteola; Pseudomonas fluorescens; Pseudomonas putida; Pseudomonas stutzeri; Pseudomonas oryzihabitans. - Rhizobiaceae:- Agrobacterium tumefaciens; Rhizobium radiobacter. - Rickettsiaceae:- Orientia chuto; Orientia tsutsugamushi; Rickettsia aeschlimannii; Rickettsia africae; Rickettsia akari; Rickettsia argasii; Rickettsia asiatica; Rickettsia australis; Rickettsia bellii; Rickettsia canadensis; Rickettsia conorii; Rickettsia cooleyi; Rickettsia felis; Rickettsia heilongjiangensis; Rickettsia helvetica; Rickettsia honei; Rickettsia hoogstraalii; Rickettsia hulinensis; Rickettsia hulinii; Rickettsia japonica; Rickettsia marmionii; Rickettsia martinet; Rickettsia massiliae; Rickettsia monacensis; Rickettsia montanensis; Rickettsia monteiroi; Rickettsia moreli; Rickettsia parkeri; Rickettsia peacockii; Rickettsia philipii; Rickettsia prowazekii; Rickettsia raoultii; Rickettsia rhipicephali; Rickettsia rickettsii; Rickettsia sibirica subgroup; Rickettsia slovaca; Rickettsia tamurae; Rickettsia typhi. - Shewanellaceae:- Shewanella putrefaciens. - Sphingomonadaceae:- Sphingobacterium multivorum; Sphingobacterium spiritivorum; Sphingomonas paucimobilis. - Spirillaceae:- Spirillum minus; Spirillum volutans; Spirillum winogradskyi. — Spirochaetaceae:- Borrelia afzelii; Borrelia anserina; Borrelia bissettii; Borrelia burgdorferi; Borrelia coriaceae; Borrelia duttonii; Borrelia garinii; Borrelia hermsii; Borrelia hispanica; Borrelia japonica; Borrelia lonestari; Borrelia lusitaniae; Borrelia miyamotoi; Borrelia parkeri; Borrelia persica; Borrelia recurrentis; Borrelia spielmanii; Borrelia turicatae; Borrelia turicatae; Borrelia valaisiana; Treponema carateum; Treponema pallidum ssp. endemicum; Treponema pallidum ssp. pallidum; Treponema pallidum ssp. pertenue. — Succinivibrionaceae:- Anaerobiospirillum spp. — Sutterellaceae:- Sutterella spp including Sutterella wadsworthia. — Thermaceae:- Meiothermus spp. — Thermotogaceae:- Thermotoga neapolitana. — Veillonellaceae:- Dialister spp; Megamonas spp; Megasphaera spp; Pectinatus spp; Pelosinus spp; Propionispora spp; Sporomusa spp; Veillonella spp.; Zymophilus spp.— Vibrionaceae:- Photobacterium damselae; Vibrio adaptatus; Vibrio alginolyticus; Vibrio azasii; Vibrio campbellii; Vibrio cholera; Vibrio damsel; Vibrio fluvialis; Vibrio furnisii; Vibrio hollisae; Vibrio metchnikovii; Vibrio mimicus; Vibrio parahaemolyticus; Vibrio vulnificus. — Wolbachieae:- Wolbachia spp. - Xanthomonadaceae:- Luteimonas aestuarii; Luteimonas aquatica; Luteimonas composti; Luteimonas lutimaris; Luteimonas marina;

Luteimonas mephitis; Luteimonas vadosa; Pseudoxanthomonas broegbernensis;

Substitute Sheet

(Rule 26) RO/AU 180

Pseudoxanthomonas japonensis; Stenotrophomonas maltophilia; and Stenotrophomonas nitritireducens.

19. The method of claim 18, wherein the bacterial agent is selected from the group comprising: Acinetobacter species, Aeromonas hydrophila, Citrobacter species, Enterobacter species, Escherichia coli, Klebsiella pneumoniae, Morganella morganii, Pseudomonas aeruginosa, and Stenotrophomonas maltophilia.

20. The method according to any one of the preceding claims, wherein the compound of Formula I, is administered together with a compound that reduces the integrity of the cell wall of the bacterial agent.

21. The method according to any one of claims 11 to 13, wherein the bacterial agent has no cell wall.

22. The method according to claim 21 , wherein the bacterial agent is selected from the group comprising: Mycoplasma spp, Mycoplasma agalactiae, Mycoplasma alkalescens, Mycoplasma amphoriforme, Mycoplasma arginini, Mycoplasma bovigenitalum, Mycoplasma bovirhinis, Mycoplasma bovis, Mycoplasma bovoculi, Mycoplasma buccale, Mycoplasma californicum, Mycoplasma canadense, Mycoplasma capricolum subsp. capricolum, Mycoplasma capricolum subsp. capripneumoniae, Mycoplasma conjunctivae, Mycoplasma cynos, Mycoplasma dispar, Mycoplasma equigenitalium, Mycoplasma faucium, Mycoplasma felis, Mycoplasma fermentans (incognitus str), Mycoplasma gallisepticum (MG), Mycoplasma gateae, Mycoplasma genitalium, Mycoplasma haemocanis, Mycoplasma haemofelis, Mycoplasma haemosuis (formerly Eperythrozoon suis), Mycoplasma hominis, Mycoplasma hyopneumoniae, Mycoplasma hyorhinis, Mycoplasma hyosynoviae, Mycoplasma iowae meleagridis (MM), Mycoplasma iowae, Mycoplasma leachii, Mycoplasma lipophilum, Mycoplasma meleagridis, Mycoplasma mycoides subsp capri, Mycoplasma mycoides subsp mycoides, Mycoplasma mycoides subsp. mycoides (such as Contagious bovine pleuropneumonia CBPP), Mycoplasma orale, Mycoplasma ovipneumoniae, Mycoplasma ovis, Mycoplasma penetrans, Mycoplasma pirum, Mycoplasma pneumoniae, Mycoplasma primatum, Mycoplasma putrefaciens, Mycoplasma salivarium, Mycoplasma spermatophilum, Mycoplasma suis, Mycoplasma synoviae (MS), Mycoplasma wenyonii, Mycoplasma, Ureaplasma spp, Ureaplasma parvum, Ureaplasma urealyticum, Ureaplasma, and Ureoplasma diversum.

23. The method according to any one of claims 11 to 13, wherein the bacterial agent is gram positive, gram negative or does not have a cell wall and is selected from the group

Substitute Sheet

(Rule 26) RO/AU 181 comprising, but not limited to, the following livestock pathogens: Actinobaculum suis, Actinomyces bovis, Arcanobacterium pyogenes, Bacillus anthracis, cereus, licheniformis, pumilus, melaninogenicus, subtilis, Clostridium botulinum, chauvoei, haemolyticum, novyi, perfringens, septicum, sordellii, tetani, colinum, Corynebacterium pseudotuberculosis, renale, Dermatophilus congolensis, Enterococcus spp (such as E. faecalis, E. faecium, E. durans, E. avium, E. hirae), Erysipelothrix rhusiopathiae, Listeria ivanovii, grayi, innocua, seeligeri, welshimeri, monocytogenes, Mycobacterium avium, bovis, paratuberculosis (Johne's Disease), Mycoplasma Csuch as capricolum subsp. capripneumoniae, subsp. capricolum, M. mycoides subsp mycoides, M. agalactiae, M. ovipneumoniae, M. conjunctivae, M. arginini, M. bovis, and M. putrefaciens) Mycoplasma bovis, dispar, mycoides subsp. mycoides (such as Contagious bovine pleuropneumonia CBPP) Mycoplasma gallisepticum (MG), iowae meleagridis (MM), synoviae (MS) Mycoplasma haemosuis (formerly Eperythrozoon suis), alkalescens, bovigenitalum, bovirhinis, bovoculi, californicum, canadense, cynos, equigenitalium, gateae, haemocanis, haemofelis, hyopneumoniae, hyorhinis, hyosynoviae, iowae, leachii, meleagridis, mycoides subsp capri, wenyonii, suis, Rhodococcus equi, Staphylococcus epidermidis, Staphylococcus simulans, Staphylococcus felis, Staphylococcus xylosus, Staphylococcus chromogenes, Staphylococcus warneri, Staphylococcus haemolyticus, Staphylococcus sciuri, Staphylococcus saprophytics, Staphylococcus hominis, Staphylococcus caprae, Staphylococcus cohnii subsp. cohnii, Staphylococcus cohnii subsp. urealyticus, Staphylococcus capitis subsp. capitis, Staphylococcus capitis subsp. urealyticus, Staphylococcus hyicus, Staphylococcus aureus, Staphylococcus pseudintermedius, Staphylococcus delphini, Staphylococcus schleiferi subsp. coagulans, Staphylococcus aureus subsp. anaerobius, Streptococcus uberis, Streptococcus canis, Streptococcus agalactiae, Streptococcus dysgalactiae, Streptococcus pyogenes, Streptococcus bovis, Streptococcus equi subsp. Zooepidemicus, Streptococcus equinus, Streptococcus equi (Streptococcus equi subsp equi), Streptococcus equisimilis (Streptococcus dysgalactiae subsp equisimilis), porcinus, suis, zooepidemicus, Streptococcus zooepidemicus (Streptococcus equi subsp zooepidemicus), Streptococcus dysgalactiae subsp. equisimilis, Propionibacterium acnes, Propionibacterium granulosum, Eubacterium, Peptococcus indolicus, and Peptostreptococcus anaerobius; Actinobacillus, Aeromonas, Anaplasma, Arcobacter, Avibacterium, Bacteroides, Bartonella, Bordetella, Borrelia, Brachyspira, Brucella, Campylobacter, Capnocytophaga, Chlamydia, Chlamydophila, Chryseobacterium, Coxiella, Cytophaga, Dichelobacter, Edwardsiella, Ehrlichia, Escherichia, Flavobacterium, Francisella, Fusobacterium, Gallibacterium, Haemophilus, Histophilus, Klebsiella, Lawsonia, Leptospira, Mannheimia, Megasphaera, Moraxella, Neorickettsia,

Ornithobacterium, Pasteurella, Photobacterium, Piscichlamydia, Piscirickettsia,

Substitute Sheet

(Rule 26) RO/AU 182

Porphyromonas, Prevotella, Proteus, Pseudomonas, Rickettsia, Riemerella, Salmonella, Streptobacillus, Tenacibaculum, Vibrio, and Yersinia.

24. The method according to any one of claims 11 to 13, wherein the infection or colonisation is caused by a mixture of at least two bacterial agents selected from the group comprising: gram postive bacteria, gram negative bacteria and bacteria with no cell wall.

25. The method according to any one of claims 11 to 24, wherein the bacterial agent is resistant to one or more compounds selected from the group comprising: penicillins, cephalosporins, carbapenems, monobactams and other beta lactam antibiotics, fusidanes, aminoglycosides, fluoroquinolones, streptogramins, tetracyclines, glycylcyclines, chloramphenicol and other phenicols, macrolides and ketolides, lincosamides, oxazolidinones, aminocyclitols, polymyxins, glycopeptides, lipopeptides, bacitracin, mupiricin, pleuromutilins, rifamycins, sulphonamides and trimethoprim.

26. The method according to claim 25, wherein the bacterial agent is resistant to one or more compounds selected from the group comprising: beta lactams, glycopeptides, lipopeptides, macrolides, oxazolidinones and tetracyclines.

27. The method according to claims 25 or 26, wherein the bacterial agent is resistant to the compound when the compound is at a concentration range selected from the following: 0.001 pg/mL - 10,000 pg/mL; 0.01 pg/mL - 1000 pg/mL; 0.10 pg/mL - 100 pg/mL; and 1 pg/mL - 50 pg/mL

28. The method according to any one of claims 11 to 27, wherein the bacterial infection or colonisation in the subject substantially causes an indication selected from the group comprising: nosocomial pneumonia caused by Staphylococcus aureus or Streptococcus pneumoniae; complicated skin and skin structure infections caused by Staphylococcus aureus, Streptococcus pyogenes, or Streptococcus agalactiae; invasive pneumococcal diseases including pneumonia, bronchitis, acute sinusitis, otitis media, conjunctivitis, meningitis, bacteremia, sepsis, osteomyelitis, septic arthritis, endocarditis, peritonitis, pericarditis, cellulitis, and brain abscess caused by Streptococcus pneumonia; uncomplicated skin and skin structure infections caused by Staphylococcus aureus or Streptococcus pyogenes; community-acquired pneumonia caused by Streptococcus pneumoniae, Staphylococcus aureus or Staphylococcus aureus; bloodstream infections (bacteraemia) caused by methicillin-susceptible and methicillin-resistant isolates; vancomycin-resistant Enterococcus infections; Clostridium difficile associated diarrhea (CDAD); and gram negative, gram positive or mixed bacterial bone and joint infections;

Substitute Sheet

(Rule 26) RO/AU 183

central nervous system infections; eye infections; gastrointestinal tract infections; genital infections; intra-abdominal infections; respiratory tract infections; otitis externa; otitis media; sepsis; systemic infections; intra-abdominal infections (lAls); urinary tract infections (UTIs); and bacteraemia.

29. The method according to any one of claims 11 to 28, wherein the therapeutically effective amount of compound of Formula I, or a therapeutically acceptable salt thereof, is administered to the subject by oral administration.

30. The method according to any one of claims 11 to 28, wherein the therapeutically effective amount of compound of Formula I, or a therapeutically acceptable salt thereof, is administered to the subject by parenteral administration.

31. The method according to any one of claims 11 to 28, wherein the therapeutically effective amount of compound of Formula I, or a therapeutically acceptable salt thereof, is administered to the subject by topical administration.

32. An antibacterial pharmaceutical composition comprising a therapeutically effective amount of a compound of Formula I, or a therapeutically acceptable salt thereof, and optionally a pharmaceutically acceptable excipient or carrier.

33. An antibacterial veterinary composition comprising a therapeutically effective amount of a compound of Formula I, or a therapeutically acceptable salt thereof, and optionally a veterinary acceptable excipient or carrier.

34. The composition according to either claim 32 or claim 33, wherein the composition comprises a further antimicrobial agent selected from the group comprising: antibacterial and antifungal agents.

35. The composition according to either claim 32 or claim 33, wherein the composition is adapted for oral administration.

36. The composition according to either claim 32 or claim 33, wherein the composition is adapted for parenteral administration.

37. The composition according to either claim 32 or claim 33, wherein the composition is adapted for topical administration.

38. Use of a compound of Formula I, or a therapeutically acceptable salt thereof, in the manufacture of a medicament for the treatment of a bacterial colonisation or infection in a subject.

Substitute Sheet

(Rule 26) RO/AU 184

39. The use according to claim 38, wherein the use comprises administering a therapeutically effective amount of compound of Formula I, or a therapeutically acceptable salt thereof, to the subject.

40. The use according to claim 39, wherein the compound of Formula I is administered to the subject in a dose in the range of 0.1 mg/kg to 250 mg/kg body weight.

41. The use according to any one of claims 38 to 40, wherein the medicament is administered to the subject by oral administration.

42. The use according to any one of claims 38 to 40, wherein the medicament is administered to the subject by parenteral administration.

43. The use according to any one of claims 38 to 40, wherein the medicament is administered to the subject by topical administration.

44. A medical device when used in a method of treating or preventing a bacterial colonisation or infection in the subject, wherein the medical device comprises the composition according to any one of claims 34 to 37.

45. The medical device according to claim 44, wherein the medical device is in a form selected from the group comprising: a plaster, a bandage, a dressing or implant applied to a bacterial or colonisation infection in a subject.

46. A method of killing bacteria, the method including the step of contacting the bacteria with a compound of Formula I, or a therapeutically acceptable salt thereof.

47. Use of a compound of Formula I, or a therapeutically acceptable salt thereof, to kill bacteria, said use comprising the step of contacting the bacteria with a compound of Formula I, or a therapeutically acceptable salt thereof.

48. A method, a composition, device or a use, substantially as described herein with reference to the accompanying Examples and Figures.

Substitute Sheet

(Rule 26) RO/AU

Description:
COMPOUNDS AND METHODS OF TREATING INFECTIONS

TECHNICAL FIELD

[0001] This invention relates to compounds of Formula I, methods of treating or preventing a bacterial infection in a subject using a compound of Formula I, the use of a compound of Formula I in the manufacture of a medicament for the treatment of a bacterial infection in a subject, and medical devices when used in a method of treating or preventing a bacterial infection in a subject.

BACKGROUND ART

[0002] A marked increase in prevalence of multi-drug resistance in disease-causing Gram- positive (G+ve) {Staphylococcus aureus, Enterococcus spp. and Streptococcus pneumoniae) and Gram negative (G-ve) pathogens {Escherichia co///, Enterobacter spp. , Salmonella spp., Acinetobacter baumannii, Klebsiella pneumoniae and Pseudomonas aeruginosa) has coincided with an unprecedented global decline in investment in new anti-infective drugs. There are few currently registered alternatives for multidrug resistant (MDR) bacterial infections, forcing clinicians to consider older generation drugs such as colistin with narrow spectrum and considerable potential for toxic side-effects. In addition, there are fewer novel classes of antiinfective therapeutics moving through the drug development pipeline.

[0003] Since the year 2000, a period of almost 15 years, only 5 novel mode of action (MOA) antibacterial agents have been approved by the US FDA - linezolid (an oxazolidinone) in 2000, daptomycin (a lipopeptide) in 2003, retapamulin (a pleuromutilin) in 2007, fidaxomicin (a macrolide tiacumicin) in 2011 , and bedaquiline (a diarylquinoline) in 2012. Notably, none of these agents has signficiant activity against gram negative bacteria. No novel MOA antibacterial agents were approved in 2013 and to date in 2014 only tedizolid and dalbavancin, both analogs of existing classes, have been recommended for approval in the US. While there are more than 300 anti-infective medicines in various stages of development, the large majority of these medicines are previously approved antibacterial compounds or their derivatives that are undergoing studies for new indications.

[0004] Furthermore, the prevalence of multidrug-resistance in animal-specific pathogens together with greater regulation of the registration and usage of antimicrobials in animals, has caused veterinarians to become increasingly reliant on the traditional classes of antimicrobial agents. The risk of transfer of MDR zoonotic organisms from animals to humans has also led to calls for further restrictions on the usage of some recently registered antibacterial drugs such as the fluoroquinolones and the third and fourth generation cephalosporins. Epidemiology of antibacterial resistance development in pathogens of humans and animals

[0005] Much of the evolution in resistance development is driven by changes in the epidemiology of key MDR organisms. Once only restricted to human hospitals and aged care facilities, methicillin resistant Staphylococcus aureus (MRSA) strains are now being isolated from the community in alarming proportions. Furthermore, community-acquired MRSA strains are more likely to carry the Panton-Valentine leukocidin (PVL) toxin, a virulence factor linked to skin and soft tissue lesions as well as a rapid, fulminating, necrotizing pneumonia with significant associated mortality. Recently MRSA strains have become host-adapted in several key animal species including livestock, horses and companion animals and regular cases of human-to-animal and animal-to-human transfer are being documented. This has important consequences for strain transmission and public health. A recent survey of 751 Australian veterinarians for MRSA nasal carriage found that a remarkable 21.4% of equine veterinarians were MRSA-positive compared to 4.9% of small animal veterinarians and 0.9% of veterinarians with little animal contact. These ecological shifts of MRSA together with the emergence of resistance to new drugs developed specifically for MRSA such as linezolid, confirm that new MRSA anti-infectives are urgently needed. Furthermore, hospitals that use vancomycin for treating MRSA then have to contend with outbreaks of vancomycin-resistant enterococci (VRE) infections in their patients, once again with limited alternative antimicrobial choices.

[0006] The global emergence and spread within the community of highly virulent MDR Gram- negative (G-ve) bacteria such as E. coli 025b:ST131 confirms that bacterial pathogens can simultaneously evolve both virulence and resistance determinants. Echoing recent MRSA epidemiology, E. coli 025b:ST131 , a major cause of urinary tract and bloodstream infections in humans, has now been isolated from extraintestinal infections in companion animals, and poultry. The increasing significance of E. coli 025b:ST131 and other MDR Enterobacteriaceae with combined resistance to fluoroquinolones and extended spectrum β-lactams and carbapenems is another worrying trend, especially considering there have been few recent breakthroughs in the development of G-ve spectrum anti-infectives apart from incremental advances in the carbapenem family.

[0007] The World Health Organisation has identified antibiotic resistance as one of the three major future threats to global health. A recent report from the US Centers for Disease Control and Prevention (CDC) estimated that "in the United States, more than two million people are sickened every year with antibiotic-resistant infections, with at least 23,000 dying as a result." The extra medical costs, in the USA alone, associated with treating and managing a single case of antibiotic-resistant infection are estimated to be between US$18,588 and US$29,069 per year resulting in an overall direct cost to the US health system of over US$20 billion annually. In addition, the cost to US households in terms of lost productivity is estimated at over US$35 billion per annum. Twenty five thousand patients in the European Union (EU) still die annually from infection with MDR bacteria despite many EU countries having world's best practice hospital surveillance and infection control strategies. The EU costs from health care expenses and lost productivity associated with MDR infections are estimated to be at least€1.5 billion per year.

[0008] There is an unmet clinical need for antibacterial agents with novel mechanisms of action to supplement and replace currently available antibacterial agents, the efficacy of which is increasingly undermined by antibacterial resistance mechanisms. There additionally remains a need for alternative antibacterials in the treatment of infection by multi-resistant bacteria. However, as reported by the Infectious Diseases Society of America and the European Centre for Disease Control and Prevention, few new drugs are being developed that offer promising results over existing treatments (Infectious Diseases Society of America 2010, Clinical Infectious Diseases, 50(8): 1081 -1083).

[0009] It is an object of the present invention to overcome at least one of the failings of the prior art.

[0010] The discussion of the background art set out above is intended to facilitate an understanding of the present invention only. The discussion is not an acknowledgement or admission that any of the material referred to is or was part of the common general knowledge as at the priority date of the application.

SUMMARY OF INVENTION

[001 1] According to one aspect of the invention, there is provided a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof:

Formula I

[0012] In one preferred embodiment, Ri is H, cycloalkyl, Formula II, or Formula III;

wherein R 3 is H, NH 2 , NHNH 2> 0~CH 2 -CH 3 , NH-CH{0)-phenyl, NH-chlorophenyl, NH-CH 2 -chlorophenyl, NH-N-CH-cycloalkyl, Formula IV, Formula V or Formula VI;

Formula IV Formula V Formula VI wherein A 0 is N, C, CH, or Ao is C and A 0 is bonded to f¾ to form a triazole ring; wherein is N, C, NH, -<CH) 2 -N-, -<C 6 H 5 )C-CH-N-, or Formula VII;

Formula VII

A 2 is N, C, NH, N-C(0)-phenyl, or Formula VII; wherein A 3 , A4, A 5l A 6 , A 7 , A 8 , An, A 12 , A 13 , A 14 , A 15 , A 16 , A 17 , A 18 , A 19 , A 20 , A 21 A 23 , A 24 , A 25 , A 26 and A 27 are independently C, O, N, NH, S; wherein A 9 is C, O, N, NH, N-C(0)-0-CH 2 -CH 3 , N-C(0)-0-CH(CH 3 ) 2 , N-C(0)-NH- CH2-CH 3 , N-C(0)-NH-CH 2 -phenyl, N-C(0)-CH 2 -CH 2 -CH 2 -CH 2 -CH 2 -CH 3 , N-C(O)- CH 2 -furan-2-yl; wherein A 10 is C, NH, -N-(CH) 2 -, -N~CH-C(C 6 H 5 )~; wherein A 22 is -CH(CH 3 )-, -N-CH-, -N-C(CH 3 )-, N-C(CH 2 OH)-;

R 2 is H, COOH, CH 2 NH 2 , CH 2 OH, CH 2 NHNH 2 , methyl, ethyl, propyl, butyl, cyclopentyl, or Formula VII and R 2 are R 4 are bonded together to form a pyrimidine, pyrazine or triazine ring, or R 2 and R 9 are bonded together to form a pyrrolidinyl oxindole ring; wherein R 4 is N, NH, O, S, or R 4 and A 0 are bonded together to form a triazole ring, or R 4 is N and R 4 and R 2 are bonded together to form a pyrimidine ring; wherein R 7 is H, CI, Br, F, OH, CH 3 , OCH 3 , SCH 3 , CN, CCH, CF 3 , OCF 3 , SCF 3 , N0 2 , butyl, f-butyl, dimethylamino, phenyl, n-propyl, /-propyl, -NH-C(0)-CH 3 , - (CH) 2 -COOH, piperazin-1-yl, or R 7 and R 8 are bonded together to form a substituted or unsubstituted, saturated or unsaturated aliphatic ring, heterocyclic ring or benzene ring; wherein R 6 , Rs, Ri 4 , Rie, R25 and R 27 are independently H, OH, CI, F, Br, CH 3 , CN, OCH 3 , COOH, N0 2 , CF 3 , R 8 and R 7 bond together to form a substituted or unsubstituted, saturated or unsaturated aliphatic ring, heterocyclic ring, or benzene ring, R 14 and R 15 are bonded together to form a substituted or unsubstituted, saturated or unsaturated aliphatic ring, heterocyclic ring or benzene ring, R 8 and R 9 are bonded together to form a substituted or unsubstituted, saturated or unsaturated aliphatic ring, heterocyclic ring or benzene ring, or R 14 and Ri 3 are bonded together to form a substituted or unsubstituted saturated or unsaturated aliphatic ring, heterocyclic ring or benzene ring; wherein R 5 , R 9 , Ri 7 , R 24 and R 28 are independently H, O, OH, CI, F, Br, NH 2 , CH 3 , CF 3 , OCH 3 , CN, N0 2 , phenyl, ~NH-CH(OH)-CH 3 , -NH-C(0)-CH 3 , or R 9 and R 8 are bonded together to form a substituted or unsubstituted, saturated or unsaturated aliphatic ring, heterocyclic ring or benzene ring, or R 3 and Ri are bonded together to form a substituted or unsubstituted saturated or unsaturated aliphatic ring, heterocyclic ring or benzene ring; wherein R 0 , Rn , R-ig, R20, R22 and R23 are independently H, CI, or Br, or R 0 and R-J 1 are bonded together to form a substituted or unsubstituted, saturated or unsaturated aliphatic ring, heterocyclic ring or benzene ring, or R 19 and R20 are bonded together to form a substituted or unsubstituted, saturated or unsaturated aliphatic ring, heterocyclic ring or benzene ring, or R 22 and R23 are bonded together to form a substituted or unsubstituted, saturated or unsaturated aliphatic ring, heterocyclic ring or benzene ring; wherein R 12, R« and R 2 i are independently H, COOH, CH 2 NH 2 , CH 2 OH, methyl, ethyl, propyl, butyl, cyclopentyl, or R 12 and R 3 are bonded together to form a pyrrolidinyl oxindole ring; wherein R 15 and R 26 are independently H, CI, Br, F, OH, CH 3 , OCH 3 , SCH 3 , CN, CF 3 , OCF 3 , SCF 3 , N0 2 , CCH, n-butyl, i-butyl, dimethylamino, phenyl, n-propyl, /- propyl, -NH-C(0)-CH 3 , -(CH) 2 -COOH, piperazin-1-yl, or R 16 and R 14 are bonded together to form a substituted or unsubstituted, saturated or unsaturated aliphatic ring, heterocyclic ring or benzene ring; and wherein "— " is a double bond or a single bond.

[0013] The compound of Formula I is preferably a chloride salt.

[0014] In another aspect of the invention, there is provided a compound, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, selected from the list of compounds presented in Figure 1. Where a salt is presented in Figure 1 , the invention covers both the salt as presented and the freebase of that salt, and stereoisomers, tautomers, other pharmaceutically acceptable salts, and also other prodrugs of the freebase.

[0015] Preferably, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein A 0 is C; wherein is N; or Formula VII; wherein A 2 is N; or NH; wherein A 3 , A4, A 6 , A 7 , An , A i 2 , A 1 , A 15 , are N; or C; wherein A 5l A 13 , A 23 , A 24 , A 25 , A 26 and A 27 are C; wherein A 8 and A 2 i are S; wherein A 9 is NH; wherein A-i 0 is N; wherein A 22 is -N-CH-; -N-C(CH 3 )-; or ~N-C<CH 2 OH)-; wherein Ri is H; Formula II; Formula III; cycloalkyl; wherein R 2 is H; methyl; ethyl; CH 2 NHNH 2 ; CH 2 OH; butyl; cyclopentyl; or Formula VII and R 2 is bonded to R 4 , to form a pyrimidine ring; wherein R 3 is NH 2 ; Formula IV; Formula V; Formula VI; NH 2 , NH-N-CH- cycloalkyl; or 0-CH 2 -CH 3 ; wherein R is NH; O; S; or R 4 is N and R 4 and R 2 are bonded together to form a pyrimidine ring;; wherein R 7 is H; F; CI; CF 3 ; methyl; R 7 and R 8 are bonded together to form an unsubstituted, benzene ring; OH; f-butyl; phenyl; dimethylamino; /-propyl; n- propyl; CN; CCH; /7-butyl; SCH 3 ; R 7 and R 8 are bonded together to form an unsubstituted, unsaturated heterocyclic ring; OCH 3 ; Br; OCF 3 ; piperazin-1 -yl; or SCF 3 ; wherein R 6 , R 8 , Ri 4 , and R 16 are independently H ; OH; F; OCH 3 ; CF 3 ; methyl; CI; CN; Br; R 8 and R 7 are bonded together to form an unsubstituted, benzene ring; R 8 and R 7 are bonded together to form an unsubstituted, unsaturated heterocyclic ring; Ri 4 and Ri 5 are bonded together to form an unsubstituted, benzene ring; or R 14 and R 15 are bonded together to form an unsubstituted, unsaturated heterocyclic ring; wherein R 5 , R 9 , Ri 3 , and R 17 are independently H; OH; NH 2 ; CI; F; OCH 3 ; OH; - NH-CH(OH)-CH 3 ; wherein Ri 2 is H; methyl; ethyl; CH 2 OH; or cyclopentyl; wherein R15 is H; F; CI; CF 3 ; methyl ; R and R 8 are bonded together to form an unsubstituted, benzene ring; OH; f-butyl; phenyl; dimethylamino; /-propyl; n- propyl; CN; CCH; n-butyl; SCH 3 ; R 5 and Ri 4 are bonded together to form an unsubstituted, unsaturated heterocyclic ring; OCH 3 ; Br; OCF 3 ; piperazin-1-yl; or SCF 3 ; wherein R 24 and R 28 are independently H; OH; or CI; wherein R 25 and R 27 are independently H; or OH; wherein R 26 is H; CH 3 ; Br; CI; OH; dimethylamino; -0-P{0){OEt) 2 ; CF 3 ; or F; and wherein is independently a single or a double bond.

[0016] More preferably, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, selected from the group comprising: NCL008; NCL009; NCL023; NCL025; NCL026; NCL029; NCL036; NCL037;

and NCL230.

[0017] Even more preferably, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, selected from the group comprising: NCL040; NCL078; NCL079; NCL080; NCL081 ; NCL084; NCL088; NCL089; NCL097; NCL099; NCL123; NCL146; NCL157; NCL158; NCL177; NCL179; NCL188; NCL193; NCL195; NCL196; NCL197; NCL199; NCL202; NCL204; NCL205; NCL215; NCL216; NCL217; NCL219; and NCL221.

[0018] Even more preferably, the compound is a compound of Formula I , or a stereoisomer, tautomer, pharmaceutically acceptable sait, or prodrug thereof, selected from the group comprising: NCL078; NCL079; NCL080; NCL081 ; NCL084; NCL089; NCL097; NCL157; NCL158; NCL179; NCL188; NCL193; NCL195; NCL196; NCL199; NCL204; NCL216; NCL217; NCL219; and NCL221 .

[0019] Even more preferably, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, selected from the group comprising: NCL089; NCL097; NCL157; NCL179; NCL188; NCL193; NCL195; NCL196; NCL216; NCL219; and NCL221.

[0020] Most preferably, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, selected from the group comprising: NCL097; NCL157; NCL179; NCL188; NCL195; and NCL196.

[0021 ] In one preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein the compound is not a compound selected from the group consisting of: NCL812, NCL001 , NCL002, NCL003, NCL004, NCL005, NCL006, NCL007, NCL010, NCL011 , NCL012, NCL013, NCL014, NCL015, NCL016, NCL017, NCL018, NCL019, NCL020, NCL021 , NCL022, NCL024, NCL027, NCL028, NCL030, NCL031 , NCL032, NCL033, NCL034, NCL035, NCL038, NCL041 , NCL042, NCL043, NCL044, NCL045, NCL046, NCL047, NCL048, NCL049, NCL051 , NCL052, NCL053, NCL054, NCL055, NCL056, NCL057, NCL058, NCL059, NCL060, NCL062, NCL063, NCL066, NCL067, NCL069, NCL070, NCL071 , NCL072, NCL073, NCL074, NCL077, NCL082, NCL083, NCL087, NCL091 , NCL093, NCL096, NCL100, NCL102, NCL103, NCL107, NCL109, NCL1 10, NCL1 13, NCL1 17, NCL120, NCL127, NCL128, NCL129, NCL134, NCL142, NCL143, NCL149, NCL151 , NCL155, NCL160, NCL163, NCL175, NCL182, NCL191 , NCL192, and NCL214.

[0022] In a preferred aspect of the invention, the compound of Formula I is not robenidine {also referenced in this specification as NCL812 and also known as 1 ,3-bis[{£)-(4- chlorophenyl)methyleneamino]guanidine), which has a structure as follows:

[0023] In one preferred embodiment of the invention, the compound is a compound of Formula I , or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein Ri is Formula II ; R 3 is Formula IV; A-i and A 0 are N; A 2 and A 9 are NH; A 0 , A 3 , A4, A 5 , A 6 , A 7 , An , A 2 , Ai 3 , An and A 15l are C; R 2 , R 5 , R 6 , R7, Rg, R12, R13, Ri5, Rie, R17 are H; R 4 is O; R 8 and Ri 4 are CF 3 ; and "— " in Formula I between A 0 and A^ all Formula I I and all Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0024] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein is Formula II ; R 3 is NH 2 ; is N and A 2 is NH; A 0 , A 3 , A4, A 5 , As, and A 7 are C; R 2 , R 5 , R 6 , R 7 , and R 8 are H; R 4 is NH; R 9 is CI; and "— " in Formula I between A 0 and A and all Formula I I "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0025] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein Ri is Formula I I; R3 is Formula iV; A^ and A 0 are N; A 2 and A 9 are NH; A 0 , A 3 , A*, A 5 , AQ, A 7 , An, A 12 , A 13 , A 14 and A 16 , are C; R 2 , R 5 , R 6 , R 8 , R 9 , R 12 , R 13 , Ri 4 , Ri 6 , Ri 7 are H; R 4 is NH; R 7 and R15 are F; and in Formula I between A 0 and A-, , all Formula I I and all Formula IV "— -" are double bonds. An example of a compound of this embodiment of the invention includes-.

[0026] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein R-, is Formula II; R 3 is Formula IV; Ai and A 10 are N; A 2 and A 9 are NH; A 0 , A 3 , A4, A 5 , As, A 7 , A„, A 12 , A 13 , A 14 and A 15 , are C; R 2 , R 5 , R 6 , R 7 , R 9 , R 12 , R 13 , Ri 5 , R 16 , R 17 are H; R 4 is NH; Ra and Ri 4 are F; and "— " in Formula I between A 0 and A 1t all Formula II and all Formula IV "— -" are double bonds. An example of a compound of this embodiment of the invention includes:

[0027] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein Ri is Formula II ; R 3 is Formula IV Ai and A 10 are N; A 2 and A g are NH; A 0l A 3 , *, A 5 , Ae, A 7 , An, A 12 , A 13 , A 14 and A 15 , are C; R 2 , R 5 , R 6 , R 7 , R 8 , Ri 2 , R14, R15, Rie, R17 are H; R 4 is NH; R 9 and Ri 3 are OCH 3 ; and "— " in Formula I between A 0 and A^ all Formula I I and all Formula IV "- — " are double bonds. An example of a compound of this embodiment of the invention includes:

[0028] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein is Formula II; R 3 is Formula IV; Ai and A 10 are N; A 2 and A 9 are NH; Ao, A 3 , A4, A 5 , Ae, A 7 , An , A 12 , A 13 , A 14 and A 15 , are C; R 2l R 5 , R 6 , R?, Rg, R12, R13, R15, R16, R17 are H; R 4 is NH; R 8 and R 14 are OCH 3 ; and "— " in Formula I between A 0 and Ai , all Formula II and all Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0029] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein R-i is Formula I I; R 3 is NH 2 ; A 1 is N; A 2 is NH; A 0 , A 3 , A4, A 5 , A 6l and A 7 are C; R 2 , R 5 , R 6 , R 8 and R 9 are H; R 4 is NH; R 7 is CI; and "— " in Formula I between A 0 and A^ and all Formula II "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0030] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein Ri is Formula II ; R 3 is Formula IV; Ai and Ai 0 are N; A 2 and A 9 are NH; A 0 , A 3) A4, A 5 , Ae, A 7 , A , A 12 , A 13 , A 14 and A 15 , are C; R 2 , R 5 , R 6 , R 8 , R 9 , R12, Ri 3 , Ri 4 , R16, R17 are H; R 4 is NH; R 7 and R 5 are CF 3 ; and "— " in Formula I between A 0 and Ai , all Formula II and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0031 ] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein is Formula II ; R 3 is Formula IV; and A 10 are N; A 2 and A 9 are NH; A 0 , A 3 , A4, A 5 , Ae. A7. An, A12, Ai3, A14 and A 15 , are C; R 2 , R 5 , R 6 , R 8 , R 9 , R12. R13. Ri . Rie. R17 are H; R4 is NH; R 7 and R 15 are methyl; and "— -" in Formula l between A 0 and Ai, all Formula U and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0032] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein R., is Formula I I; R 3 is Formula IV; A-, and A 10 are N; A 2 and A 9 are NH; A 0 , A 3 , A4, A 5 , As, A 7 , An , A 12 , A 13 , A 14 and A 16 , are C; R 2 , R 5 , R 6 , R 7 , R 8 , R 12 , R i4 , R 15 , Rie, R17 are H; R4 is NH; R 9 and R 13 are methyl; and "— " in Formula I between A 0 and Ai , all Formula II and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0033] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein R^ is Formula II; R 3 is Formula IV; and A 10 are N; A 2 and A g are NH; A 0 , A 3 , A4, A 5l As, A 7> An, A 12 , A 13 , A 14 and A 15 , are C; R 2) R 5 , R 6 , R 7 , R 9 , i 2 , i 3l R15, Ri 6 and R 17 are H; is NH; R 8 and R 14 are methyl; and "— " in Formula I between A 0 and At, all Formula U and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0034] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein R is Formula II; R 3 is NH 2 ; Ai is N; A 2 is NH; A 0 , A 3 , A 4 , A 5 , A 6 , and A 7 are C; R 2 , R 5 , R 6 , R 8 , and R 9 are H; R 4 is N H; R 7 is CF 3 ; and "— " in Formula I between A 0 and A-,, and all Formula II "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0035] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein is Formula II; R 3 is NH 2 ; A-i is N; A 2 is NH; A 0 , A 3 , A , A 5 , A 6 , and A 7 are C; R 2 , R5, Re, R 7l and R 9 are H; R 4 is NH; R 8 is CF 3 ; and "— " in Formula I between A 0 and A 1 t and all Formula II "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0036] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein Ri is Formula II; R3 is NH 2 ; Ai is N; A 2 is NH; A 0 , A 3 , A 4 , A 5 , A 6 , and A 7 are C; R 2 , R5, Re, R 8 , and R 9 are H; R 4 is NH; R 7 is methyl; and "— " in Formula I between A 0 and A 1 ( and all Formula I I "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0037] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein R-, is Formula II; R 3 is NH 2 ; Ai is N; A 2 is NH; A 0 , A 3 , A4, Α δ , Ae, and A 7 are C; R 2 , R5, Re, R 7 , and R 9 are H; R4 is NH; R 8 is CI; and "— " in Formula I between A 0 and A 1 ( and all Formula II "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0038] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein Ri is Formula II; R 3 is Formula IV; Ai and A 10 are N; A 2 and A 9 are NH; Ao, A 3 , A4, A 5 , Αβ, A 7 , An , A 12 , A 13 , A 14 and A 15) are C; R 2 , R 5 , R 6 , R 7 , Rg, Ri2, Ri 3 , Ri5, i6, Ri 7 are H; R4 is NH; Ra and R-K are CI; and "— " in Formula I between A 0 and A t all Formula II and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0039] In another preferred embodiment of the invention, the compound is a compound of Formula I , or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein R'i is Formula I I ; R 3 is Formula IV; ! and A 10 are N; A 2 and A 9 are NH; A 0 , A 3 , A4, A 5 , A 6 , A 7 , An , A 12 , A 13 , A 14 and A 15 , are C; R 2 and R 12 are methyl; R 5 , R e , R a , R 9 , R13, i , Rie, R17 are H; R 4 is NH; R 7 and R i S are CF 3 ; and "— " in Formula I between A 0 and A : all Formula I I and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0040] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein is Formula I I ; R 3 is Formula IV; and Ai 0 are N; A 2 and A 9 are NH; A 0 , A 3l A l A 5 , A 6 , A 7 , An , A 12 , A 13 , A 14 and A 15 , are C; R 2 and R i2 are methyl; R 6 , R 6 , R 8l R 9 , R 13 , R 14 , R 16 and R 17 are H; R 4 is NH; R 7 and Ri 6 are CI; and "-— " in Formula I between A 0 and An, all Formula I I and Formula IV "-— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0041 ] In another preferred embodiment of the invention, the compound is a compound of Formula I , or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein Ri is Formula I I ; R 3 is NHNH 2 ; A is N; A 2 is NH; A 0 , A 3 , A4, A 5 , A 6 , and A 7 are C; R 2 is methyl, R 5 , Rs, Rs and R 8 are H; R4 is NH; R 7 is CI; and "— " in Formula I between Ao and A, , and all Formula I I "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0042] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein R-, is Formula II ; R3 is Formula IV; At and A 0 are N; A 2 and A 9 are NH; A 0 , A3, A4, A 5 , A 6 , A 7 , An , A12, Ai3, A 14 and A 16 , are C; R 2 , R 5 , R 6 , R 7 , R 9 , R12, Ria, R15, R16, and R 17 are H; R 4 is S; R 8 and R 14 are CI; and "— " in Formula I between A 0 and Ai, all Formula II and Formula IV "-- -" are double bonds. An example of a compound of this embodiment of the invention includes:

[0043] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein R 1 is Formula II; R 3 is NH 2 ; A t is N;A 2 is NH; A 0 , A 3 , At, A 5 , A 6 , and A 7 are C; R 2 , R 5 , R 6 , Ra, and R 9 are H; R 4 is NH; R 7 is CI; and "— " in Formula I between A 0 and Ai, and all Formula II "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0044] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein R is Formula II; R 3 is NHNH 2 ; Ai is N; A 2 is NH; A 0 , A 3 , A4, A 5 , Ae, and A 7 are C; R 2 is methyl; R 5 , R 6 , R 8l and R 9 are H; R 4 is NH; R 7 is CF 3 ; and "-— " in Formula I between Ao and A^ and all Formula II "-— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0045] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein Rt is Formula II; R3 is Formula IV; Ai and A 10 are N; A 2 and A 9 are NH; A 0 , A 3 , t, A 5 , A 6 , A 7 , An , Ai2, A 13 , A 14 and Ai 5 , are C; R 2 , R 5 , R 6 , R 8 , R 9 , R 12 , Ri 3 , Ri 4 , and R 6 are H; R 4 is NH; R 7 , R 15 and R-t? are CI; and "— " in Formula I between A 0 and A^ all Formula II and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0046] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein Ri is Formula II ; R 3 is Formula IV; and A 10 are N ; A 2 and A 9 are NH; A 0 , A 3 , A4, A 5 , A 6 , A 7 , An, A 12 , A 13 , A 14 and A 5 , are C; R 2 , R 6 , R 6 , R 8 , R_, R12, R13, R14, and R i 6 are H; R 4 is NH; R 7 is CI; R 15 is CF 3 ; Ri 7 is F; and "— " in Formula I between A 0 and A-,, all Formula II and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes (NCL078):

[0047] In another preferred embodiment of the invention, the compound is a compound of Formula I , or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein Ri is Formula II; R 3 is Formula IV; At and A 1D are N; A 2 and A s are NH; A D , A 3 , At, A 5 , As, A 7 , An , A 12 , A 13 , A 14 and A 15 , are C; R 2 , R 5 , R e , R 3 , R 9 , R12, R13, Ri 4 , R i6 and Ri 7 are H; R 4 is NH; R 7 is CI; R 15 is F; and "— " in Formula I between A 0 and Ai , all Formula II and Formula IV "-- -" are double bonds. An example of a compound of this embodiment of the invention includes (NCL079):

[0048] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein Ri is Formula II; R 3 is Formula IV; Ai and A 0 are N; A 2 and A 9 are NH; A 0 , A 3 , A4, A 5 , Ae, A 7 , A , A 12 , A 13 , A 14 and A 15 , are C; R 2 , R 5 , R 8 , R a , R 9 , R 13 , R 1 , R 6 and R 17 are H; R 4 is NH; R 7 is CI; Ri2 is methyl; R i 5 is CF 3 ; and "— " in Formula I between A 0 and A n , all Formula II and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes (NCL080):

[0049] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein R-, is Formula II ; R 3 is Formula IV; and A 10 are N; A 2 and A 9 are NH; Ao, A 3 , A4, A 5 , As, A 7 , An , A 12 , An, A 14 and A 15 , are C; R 2 , R s , R 6 , R 8 , Rg, R13, Ri 4 , R16 and Ri 7 are H; R 4 is NH; R 7 and R-| 5 are CI; R 12 is methyl; and "— " in Formula I A 0 and Ai , all Formula II and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0050] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein R is Formula II; R 3 is Formula IV; Ai and A 10 are N; A 2 and A 9 are NH; A 0 , A 3 , At, A 5 , Ae, A 7 , An , A 12 , A 13 , A 4 and A 5 , are C; R 2 , R s , R 6 , R 8 , R 9 , R 12 , R 13 , R 1 , R 15 , and R 6 are H; R 4 is NH; R 7 and R 7 are CI; and "-— " in Formula I between A 0 and A 1 : all Formula I I and Formula IV "- — " are double bonds. An example of a compound of this embodiment of the invention includes:

[0051 ] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein R-, is Formula II ; R 3 is Formula IV; and A10 are N; A 2 and A 9 are NH; A 0 , A 3 , A 4 , As, A 6 , A 7 , An , A 12 , A 13 , A 14 and A 15 , are C; R 2 , R 5 , R e , R 8 , R 9 , R 12 , R 13 , R 14 , and Ri 6 are H; R 4 is NH; R 7 and R15 are CI; R 7 is F; and "— " in Formula I between Ao and A^ all Formula II and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes (NCL084):

10052] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein Ri is Formula II; R 3 is Formula IV; A-, and A 10 are N; A 2 and A 9 are NH; A 0 , A 3 , A¾, A 5 , Ae, A 7 , An , A 12 , A 13 , A 14 and A 15 , are C; R 2 , R 5 , Rs, R 8 , R9, R12, R13, R15, R16, and R i7 are H; R 4 is NH; R 7 is CI; R 4 is CN; and "— " in Formula I between A 0 and Ai , all Formula II and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes:

I0053] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein is Formula II; R3 is Formula IV; Ai and A10 are N; A 2 and A 9 are NH; A 0 , A3, A , A 5 , A 6 , A 7 , An , A 12 , Ai 3, A 14 and A 15 , are C; R 2 , R 5 , R 6 , R 8) R 9 , R 12 , R 13 , Ri 4 , R15, and Ri 6 are H; R 4 is NH; R 7 is CI; R 17 is F; and "— " in Formula I between A 0 and Ai , all Formula 11 and Formula IV "-- -" are double bonds. An example of a compound of this embodiment of the invention includes:

[0054] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein is Formula I I; R 3 is Formula IV; Ai and A 10 are N; A 2 and A 9 are NH; Ao, A 3 , Ai, A 5 , A 6 , A 7 , A , A 2 , A 13 , A 14 and A 15 , are C; R 2 and Ri 2 are methyl; R 5 , R 6 , R 8 , R 9 , R13, Ri 4 , Ris, and R 17 are H; R 4 is NH; R 7 is CI; R 15 is CF 3 ; and "— " in Formula I between A 0 and Ai , all Formula II and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes (NCL089):

[0055] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein is Formula II; R3 is Formula IV; Ai and A 10 are N; A 2 and A 9 are NH; A 0 , A 3 , A4, A 5 , A 6 , A 7 , An, A 12 , A 13 , A 14 and A 15 , are C; R 2 , R 5 , R 6 , Rg, R12, R13, Rie, and Ri 7 are H; R 4 is NH; R 7 and R 8 are bonded together to form an unsubstituted, benzene ring; R 14 and R15 are bonded together to form an unsubstituted, benzene ring; and "— " in Formula I between A 0 and Α·,, all Formula I I and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0056] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein A 0 is C; A-, is N; A 2 is NH; Ri is cyclohexyl; R 3 is NH-N-CH-cyclohexyl; R 4 is NH; R 2 is H; and in Formula I between A 0 and A^ is a double bond. An example of a compound of this embodiment of the invention includes:

[0057] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein R is Formula II; R 3 is Formula IV; A, and A 10 are N; A 2 and A 9 are NH; A 0 , A 3 , A 4 , A 5 , A 6 , A 7 , , A 2 , A 13 , A 14 and A 15 , are C; R 2 , R 5 , Rg, R 12 , R13 and R 17 are H; R 4 is NH; R 6 , R 7 , R 8 , R14, R 15 , and R i6 are OH; and "— " in Formula I between A 0 and Α·,, all Formula II and Formula IV "-- -" are double bonds. An example of a compound of this embodiment of the invention includes (NCL097):

[0058] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein is Formula II; R 3 is Formula IV; A^ and A 0 are N; A 2 and A 9 are NH; A 0 , A 3 , A , A 5 , Ae, A 7 , An, A 12 , A 13 , A 14 and A S , are C; R 2 , R 5 , R 6 , R 8l R 9 , R 12 , R 13 , R 14 , R 16 and R i7 are H; R 4 is NH; R 7 and R 15 are f-butyl; and "— " in Formula I between A 0 and A-, , all Formula II and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0059] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein R-, is Formula li; R 3 is Formula IV; Ai and A 10 are N; A 2 and A 9 are NH; A 0 , A 3 , A , A 5 , As, A 7 , An , A 12 , A 13 , A 14 and A 15 , are C; R 2 , R 7 , R 8> R 9 , R12, R13, R14, and R 15 are H; R4 is NH; R 5 , R 8 , R16, and R 17 are OH; and "— " in Formula I between A 0 and A^ all Formula II and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0060] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein Ri is Formula ll; R 3 is Formula IV; A : and A10 are N; A 2 and A 9 are NH; A 0 , A 3 , A 4 , A 5 , As, A 7 , An , A 12 , A 13 , A 14 and A 15 , are C; R 2 , R 5 , R 8 , R i2 , R14, and R 17 are H; ¾ is NH; R 6 , R , R 9 , R 3 , R 15 , and R 16 are OH; and "— " in Formula I between A 0 and At , all Formula I I and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0061 ] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein is Formula II; R 3 is Formula IV; and A 10 are N; A 2 and A 9 are NH; A 0 , A 3 , A , A 5 , A 6 , A 7 , A 11 t A 12 , A 13 , A 14 and A 15 , are C; R 2 , R 5 , R 6 , Ri 2 , Ri 6 , and R 17 are H; R 4 is NH; R 7 , R 8 , R 9 , Ri3, R , and R 15 are OH; and "— " in Formula I between Ao and Ai , all Formula II and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes (NCL097):

[0062] In another preferred embodiment of the invention, the compound is a compound of Formula I , or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein Ri is Formula II; R3 is Formula IV; A-i and A 10 are N; A 2 and A 9 are NH; A 0 , A 3 , A4, A 5 , Ag, A 7 , An , A 12 , A 13 , A 14 and A 15 , are C; R 2 , R 6 , R 7 , R 8 , R 9 , R12, R13, Ri 4 , Ris, and R 16 are H; R 4 is NH; R 5 and R17 are OH; and "— " in Formula I between A 0 and At , all Formula II and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0063] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein Ri is Formula II; R 3 is Formula IV; Ai and A 10 are N; A 2 and A s are NH; A 0 , A 3 , A , A 5 , As, A 7 , An , Ai2, A 13 , A 14 and A 15 , are C; R 2 , R 5 , R 7 , R 8 , R 9 , Ri 2 , R13, Ri 4 , R15, and R i7 are H; R 4 is NH; R 6 and Rt 6 are OH; and "— " in Formula I between A 0 and A ( all Formula II and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0064] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein Ri is Formula I I; R 3 is Formula IV; and A 10 are N; A 2 and A 9 are NH ; Ao, A 3 , A 4 , As, As, A 7j An , A12, A 13 , A 14 and A 15 , are C; R 2 , R 6 , R 8 , R 9 , R12, Ris, Ri 4 , and Ri 7 are H; R 4 is NH; R 6 , R 7 , R 15 , and R 6 are OH; and "— " in Formula I between A 0 and A 1 f all Formula II and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0065] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein F is Formula II; R3 is Formula IV; At and A 10 are N ; A 2 and A 9 are N H; A 0 , A 3 , A4, A 5 , Ae, A 7 , An , A 12 , A,3, Aw and A 15 , are C; R 2 , R 5 , R 6 , R 8 , e , R12, R13, Ru, R16, and R 17 are H; R 4 is NH; R 7 and R 5 are phenyl; and "— " in Formula I between A 0 and Ai , all Formula II and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0066] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein Ri is Formula II; R3 is Formula IV; and A 0 are N; A 2 and A 9 are NH; A 0 , A 3 , A4, A 5 , Ae, A 7 , An , A 2 , A 3 , A 14 and A 5 , are C; R 2 , R 5 , Re, e, R9, R12, R13, Ri 4 , R-ie, and R 17 are H; R 4 is NH; R 7 and R 15 are dimethylamino; and "— " in Formula I between A 0 and A-, , all Formula II and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0067] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein R is Formula II; R3 is Formula IV; Ai and A 10 are N; A 2 and A 9 are NH; A 0 , A 3 , A,, A 5 , Ae, A 7 , An , A 12 , Aw, A 4 and A 15 , are C; R 2 , R 5 , Re, R9, R12, R13, RM > and R 17 are H; R 4 is NH; R 6 and R16 are OCH 3 ; R 7 and R15 are OH; and "— " in Formula I between A 0 and A-,, all Formula II and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0068] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein Ri is Formula II; R 3 is Formula IV; and A 10 are N ; A 2 and A 9 are N H; Ao, A 3 , A4, A 5 , A 6l A 7 , A , A12, A 13 , A 14 and A 15) are C; R 2 , R 5 , Re, Ra, R9, R12, R13, 14, R-m, and R 17 are H; R 4 is NH; R 7 and R15 are /-propyl; and "— " in Formula I between A 0 and A 1 : all Formula II and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0069] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein is Formula II; R 3 is Formula IV; Ai and A 10 are N; A 2 and A 9 are NH; Ao, A 3 , A,,, A 5 , A B , A 7 , An , A 12 , A 13 , A 14 and A 15 , are C; R 2 , R 5 , R 6 , Ra, Rg, R12, Ris, i 4 , R16, and Ri 7 are H; R 4 is NH; R 7 and Ri 5 are n-propyl; and "— " in Formula I between A 0 and A 1 : all Formula II and Formula IV "— -" are double bonds. An example of a compound of this embodiment of the invention includes:

[0070] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein Ri is Formula II ; R 3 is Formula IV; Ai and A 10 are N ; A 2 and A 9 are NH; Ao, A 3 , A 4 , A 5 , As, A 7 , An , A 12 , A 13 , A 14 and A 15 , are C; R 2 , R 5 , Ra, R., R12, Ri 3 , i4, and R 7 are H; R 4 is NH; R 6 , R7, R13, and R16 are F; and "— " in Formula I between A 0 and Ai , all Formula II and Formula IV are double bonds. An example of a compound of this embodiment of the invention includes: F F

F HCI F

10071] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein Ri is Formula II ; R 3 is Formula IV; A 1 and A 10 are N; A 2 and A 9 are NH; Ao, A 3 , A4, A 5 , Ae, A 7 , An , A 2l A 13 , A 14 and A 15> are C; R 2 , R 5 , R 6 , Ra, R9, R12, Ria, RH, Rie, and Ri 7 are H; R 4 is NH; R 7 and R15 are CCH; and "— " in Formula I between A 0 and Ai , all Formuia II and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes:

I0072] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein Ri is Formula II ; R3 is Formula IV; Ai and A 10 are N; A 2 and A 9 are N H; A 0 , A 3 , A 4 , A 5 , Ae, A 7 , An , A 12 , A 13 , A 14 and A 16 , are C; R 2 , R 5 , R 7l R 8 , R 9 , Ri 2 , R13, Ri 4 , R15, and R i7 are H; R 4 is NH; R 6 and R e are Br; and "— " in Formula I between A 0 and Ai , all Formula II and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0073] In another preferred embodiment of the invention, the compound is a compound of Formula I , or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein Ri is Formula II ; R 3 is Formula IV; and A 10 are N; A 2 and A 9 are NH; A 0 , A 3l A 4 , A 5 , As, A 7 , An , Ai 2 , A-I3, A14 and A 15 , are C; R 2 , R 5 , R 6 , R 8 , Rg, R12, R13, RH, Rie, and Ri 7 are H; R 4 is NH; R 7 and R15 are butyl; and "— " in Formula I between A 0 and A 1 ( all Formula II and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0074] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein , is Formula II ; R 3 is Formula IV; A is -C(C 6 H 5 )~CH-N- and A 10 is -N-CH-C(C a H 5 )-; A 2 and A 9 are NH; A 0 , A 3 , A4, A 5 , As, A 7 , A 11 s Ai 2 , A 13 , A 14 and A 15 , are C; R 2 , R 5 , R 6 , R7, Rs, R9, R12, Ri 3, Ri4, R15, R16, and R 17 are H; R 4 is NH; and "— " in Formula I between A 0 and A 1 t all Formula II and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0075] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein is Formula II; R 3 is Formula IV; Ai and A-| 0 are N; A 2 and A 9 are NH; A 0 , A 3 , A 4 , A 5 , Ag, A 7 , An , A 12 , A 13 , A 14 and Ai 5 , are C; R 2 , R 5 , R e , R 8 , R 9 , R12, i 3 , i 4 , Rie, and R 17 are H; R 4 is NH; R 7 and R 15 are CH 3 S; and "— " in Formula I between A 0 and Ai , all Formula II and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0076] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein is Formula II I; R 3 is Formula VI; A 0 is C; R 2 and R 2 i are H; Ai and A 20 are N; A 2 and A 19 are NH; A 8 and A 2 i are S; R 4 is NH; R 10 and Rn are bonded together to form a substituted benzene ring; R 22 and R 23 are bonded together to form a substituted benzene ring; and "— " in Formula ! between A 0 and A 1 : and all Formula II I and Formula VI "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0077] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein is Formula II; R 3 is Formula IV; and A 10 are N; A 2 and A9 are NH; Ao, A3, A4, A5, A3, A 7) An, A 12) A,3, A and A 15 , are C; R 2 and R 12 are methyl; R 5 , R 6 , R 7 , Re, Rg, Rl3. Rl , Rl5, R16, and Ri 7 are H; R 4 is NH; and "— " in Formula I between A 0 and A^ all Formula II and Formula IV "-— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0078] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein is Formula II; R3 is Formula IV; and A^ are N; A 2 and A 9 are NH; A 0 , A 3 , A4, A 5 , A 6 , A 7 , A , A 12 , A 13l A 14 and A 15 , are C; R 2l R 5 , Rg, R 9 , R 12 , R 13 , R 16 , and R i7 are H; R is NH; R 7 and R 8 are bonded together to form an unsubstituted, heterocyclic ring; R 14 and R15 are bonded together to form an unsubstituted, unsaturated heterocyclic ring; and "— " in Formula I between A 0 and Ai, all Formula II and Formula IV "-— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0079] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein Ri is Formula II; R 3 is Formula IV; Ai is -{CH) 2 -N- and A 10 is -N-(CH) 2 -; A 2 and A 9 are NH; Ao, A 3 , A4, A 5 , A 6 , A , An, Ai 2 , A 13 , A 14 and A-| 5 , are C; R 2 , R 5 , R6, Rs, R9, R12, R13. Ri , R16, and Ri 7 are H; R 4 is NH; R 7 and R 15 are OCH 3 ; and "— " in Formula I between A 0 and Ai , all Formula I I and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes:

10080] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein Ri is Formula I I; R 3 is Formula IV; A-i and A 10 are N; A 2 and A 9 are NH; A 0 , A 3 , A4, A 5| A 6 , A 7 , An, A 12 , A 13 , A 14 and A 15 , are C; R 2 , R 5 , R 6 , Rs, R., R12, R13, Ri 4 , R16, and R 17 are H; R4 is NH; R 7 and R 15 are OH; and "— " in Formula I between A 0 and Ai , all Formula I! and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes:

10081 ] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein Rt is Formula I I; R 3 is Formula IV; A^ and A 10 are N; A 2 and A 9 are NH; A 0 , A 3 , A 4 , A 5 , As, A 7 , An , A 2 , A 13 , A 14 and A 15 , are C; R 2 and R 12 are ethyl; R 5 , Rs, Rs, Rg, Ri 3 , i 4 , 16, and Ri 7 are H; R is NH; R 7 and R 15 are CI; and "— " in Formula I between A 0 and A^ all Formula II and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0082] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein Ri is Formula I I; R 3 is Formula IV; Ai and A 10 are N; A 2 and A 9 are NH; A 0) A 3 , A4, A 5 , A 6 , A 7 , An, Ai 2 , A 3 , A 14 and A15, are C; R 2 and Ri 2 are methyl; R 5 , R 6 , R 8 , R9, Ri 3 , i 4 , Rie, and R 17 are H; R 4 is NH; R 7 and R 15 are Br; and "— " in Formula I between A 0 and A-i , all Formula II and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0083] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein is Formula I I; R3 is Formula IV; A-i and A 10 are N; A 2 and A 9 are NH; A 0 , A 3 , A4, As, Ae, A7. An, A 12 , Ai3, A 14 and A 15 , are C; R 2 , R 5 , R 6 , R 8 , R12, i 4 , Rie, and R 7 are H; R 4 is NH; R 7 and Ri 5 are CI; R 9 and R 13 are NH 2 ; and "— " in Formula I between A 0 and A 1 ( all Formula II and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes (NCL157):

[0084] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein R-i is Formula II; R 3 is Formula IV; and A 10 are N; A 2 and A 9 are NH; A 0 , A 3 , A 4 , A 5 , A 6 , A 7 , An, A 12 , A 13 , A 4 and A 15 , are C; R 2 and R 12 are ethyl; R 5 and Ri 7 are OH; R 6 , R 8 , R 9 , R 13 , Ri4, and R 16 are H; R is NH; R 7 and R15 are CI; and "— " in Formula I between A 0 and A-,, all Formula II and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes (NCL158):

[0085] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein is Formula II; R3 is Formula IV; Ai and A 0 are N; A 2 and A 9 are NH; Ao, A 3 , A^ A 5 , A3, A 7 , An, A 12 , A 13 , A 4 and A 15 , are C; R 2 and Ri 2 are cyclopentyl; R 5 and Ri 7 are OH; R 6 , R 8] Rg, Ri 3 , i4, and R 16 are H; R 4 is NH; R 7 and Ri 5 are CI; and "— " in Formula I between A 0 and A 1 ( all Formula II and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0086] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein R-ι is Formula II; R3 is Formula IV; Ai and A 10 are N; A 2 and A 9 are NH; A 0 , A 3l A 4 , A 5 , g, A 7 , An , A 12 , A 13l A 14 and A 16 , are C; R 2 , R 6 , R 6 , Rs, Rs>, R12, R13, u. R« and R 17 are H; R 4 is NH; R 7 and R 15 are OCF 3 ; and "— " in Formula I between A 0 and Ai , all Formula II and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0087] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein is Formula II; R3 is Formula IV; A-, and A 10 are N ; A 2 and A 9 are N H; A 0 , A 3 , A4, A 5l As, A 7 , An , A-| 2 , Af 3 , A-j and A 15 , are C; R 2 and R 12 are methyl; R 5 , R 6 , Re, Rg, Ri3 > Ru, R« and Ri 7 are H; R 4 is N H; R 7 and Ris are piperazin-1 -yl; and "— " in Formula I between A 0 and At , all Formula II and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0088] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein R., is Formula II; R 3 is 0-C H 2 -CH 3 ; A 1 is N; A 2 is N H; A 0 , A 3 , At, As, A 6 and A 7 are C; R 2 is methyl; R 5 , R 6 , Rs, and R 9 are H ; R is N H; R 7 is CI; and "— " in Formula I between A 0 and At , and all Formula II "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0089] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein is Formula I I; R 3 is Formula IV; A-i and A 10 are N ; A 2 and A 9 are N H; A 0 , A 3 , A 4 , A 5 , Ae, A 7) An , A 12 , A 13 , A 14 and A 15 , are C; R 2 , R 5 , R 6 , R 8l R 9 , 12, R13. R14. R ie and R 17 are H; R4 is N H; R 7 and R15 are SCF 3 ; and "— " in Formula I between A 0 and A : all Formula II and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes:

[0090] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein R-i is Formula II; R 3 is Formula IV; and A 10 are N; A 2 and A 9 are NH; A 0 , A 3l A , A 5l A 6 , A 7 , An , A 12 , A 13 , A 14 and A 16 , are C; R 2 , R 6 , Rs, R a , R 12 , R13, Ru, and Ri 6 are H; R is NH; R 7 and R 15 are CI; R 5 and R i 7 are -NH-CH(OH)-CH 3 ; and "— -" in Formula I between A 0 and At, all Formula I I and Formula IV are double bonds. An example of a compound of this embodiment of the invention includes:

[0091] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein A 0 is C; Ri is H; A 2 and R 4 are N; R 3 is NH 2 ; Ai is Formula VII; R 2 is Formula VII and R 2 is bonded to R4, forming a pyrimidine ring; "— -" in Formula I between R 2 and A 0 , and between Ai and A 2 are double bonds; A 22 is -N-CH-; R 24 , R 25 , R 27 and R 28 are H; A 23 , A 2 , A 25 , A 26 and A 27 are C; and R 26 is CI. An example of a compound of this embodiment of the invention includes (NCL179):

[0092] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein is Formula I I; R 3 is NH 2 ; Ai is N; A 2 and R 4 are NH; Ao, A 3 , A4, A 5 , Ae, and A 7 are C; R5, Re, Rs, and R 9 are H; R 2 is butyl; R 7 is CI; and "— -" in Formula I between A 0 and Ai, and all Formula II "— " are double bonds. An example of a compound of this embodiment of the invention includes (NCL1

[0093] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein A 0 is C; R-i is H; A 2 and R 4 are N; R 3 is NH 2 ; Ai is Formula VII; R 2 is Formula VII and R 2 is bonded to R 4 , forming a pyrimidine ring; "— " in Formula I between R 2 and A 0 , and between A : and A 2 are double bonds; A∞ is -N-CH-; R 24 , R 25 , R 27 and R 28 are H; A 23 , A 24] A 25 , A 26 and A 27 are C; and R 2a is CH 3 . An example of a compound of this embodiment of the invention includes (NCL195):

[0094] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein A 0 is C; is H; A 2 and R 4 are N; R 3 is NH 2 ; Ai is Formula VII; R 2 is Formula VII and R 2 is bonded to R 4 , forming a pyrimidine ring; "— " in Formula I between R 2 and A 0 , and between Ai and A 2 are double bonds; A 22 is -N-CH-; R 24 , R 25 , R 27 and R 28 are H; A 23 , A 2 , A 25 , A 26 and A 27 are C; and R 26 is OH. An example of a compound of this embodiment of the invention includes

[0095] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein A 0 is C; Ri is H; A 2 and R 4 are N; R 3 is NH 2 ; A-ι is Formula VII; R 2 is Formula VI I and R 2 is bonded to R 4 , forming a pyrimidine ring; "— " in Formula I between R 2 and A 0 , and between Ai and A 2 are double bonds; A 22 is -N-CH-; R 24 , R 25 , R 27 and R 29 are H; A 23 , A 24 , A 25 , A 26 and A 27 are C; and R 2e is Br. An example of a compound of this embodiment of the invention includes (NCL193):

[0096] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein A 0 is C; Ri is H; A 2 and R 4 are N; R 3 is NH 2 ; A^ is Formula VII; R 2 is Formula VII and R 2 is bonded to R 4 , forming a pyrimidine ring; "— " in Formula I between R 2 and A 0 , and between A and A 2 are double bonds; A 22 is -N-CH-; R 24 , R 25 , R 26 , R 27 and R 28 are H; and A 23 , A 24 , A 25 , A 26 and A 27 are C. An example of a compound of this embodiment of the invention includes (NCL199):

[0097] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein A 0 is C; Ri is H; A 2 and R 4 are N; R 3 is NH 2 ; A^ is Formula VII; R 2 is Formula VII and R 2 is bonded to R , forming a pyrimidine ring; "— " in Formula I between R 2 and A 0 , and between and A 2 are double bonds; A is -N-C(CH 3 )-; R 24 , R 25 , R 27 and R 28 are H; A 23 , A 24 , A 25 , A 26 and A 27 are C; and R 26 is CI. An example of a compound of this embodiment of the invention includes (NCL204):

[0098] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein Ri is Formula II; R 3 is Formula IV; A^ and A 10 are N; A 2 and A 9 are NH; A 0 , A 3 , A , A 5 , Ae, A 7 , An, A 12 , A 13 , A 14 and A i5 , are C; R 2 , R 6 , R 8 , R 9 , R i2 , Ri 3 , Ri 4 , and R 16 are H; R is NH; R 7 and R15 are CI; R 5 and R 17 are F; and "— " in Formula I between A 0 and A^ all Formula II and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes (NCL216):

[0099] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein R-, is Formula II; R 3 is Formula IV; A^ and A 0 are N; A 2 and A 9 are NH; A 0 , A 3 , A 4 , A 5 , A 6 , A 7l An, A 12 , A 13 , A 14 and A 5 , are C; R 2 and R 12 are methyl; R 5 , R 6 , R s , Rg, R13, i4, Rie and R 17 are H; R 4 is NH; R 7 and Ri 5 are CH 3 ; and "— " in Formula I between A 0 and A-, , all Formula II and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes (NCL217):

[00100] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein R is Formula II; R 3 is Formula IV; Ai and A 10 are N; A 2 and A 9 are NH; Ao, A 3 , A4, A5, A 6 , A 7 , An , Ai 2 , Ai 3 , A 14 and Ai 6 , are C; R 2 and Ri 2 are methyl; R 5 , R 6 , R 6 , R9, R13, i , Rie and Ri7 are H; R 4 is NH; R 7 and Ri 6 are i-butyl; and "— " in Formula I between A 0 and Ai , all Formula II and Formula IV "— " are double bonds. An example of a compound of this embodiment of the invention includes (NCL219):

[00101 ] In another preferred embodiment of the invention, the compound is a compound of Formula I, or a stereoisomer, tautomer, pharmaceutically acceptable salt, or prodrug thereof, wherein A 0 is C; R-i is H; A 2 and R4 are N; R 3 is NH 2 ; Ai is Formula VII; R 2 is Formula VII and R 2 is bonded to R 4> forming a pyrimidine ring; "— " in Formula I between R 2 and A 0 , and between A-, and A 2 are double bonds; A 22 is -N-CH-; R 24 , R 25 , R 27 and R 28 are H; A 23 , A 24 , A 26 , A 26 and A 27 are C; and R 26 is CF 3 . An example of a compound of this embodiment of the invention includes (NCL221):

[00102] According to another aspect of the invention, there is provided a method of treating or preventing a bacterial colonisation or infection in a subject, the method comprising the step of administering a therapeutically effective amount of a compound of Formula I, or a therapeutically acceptable salt thereof, to the subject. In this aspect, the bacterial infection is caused by a bacterial agent. The method of treating or preventing a bacterial infection or colonisation in a subject, may also comprise the administration of the pharmaceutical or veterinary compositions of the invention.

[00103] According to a further aspect of the invention, there is provided the use of a compound of Formula I, or a therapeutically acceptable salt thereof, in the manufacture of medicament the treatment of a bacterial colonisation or infection in a subject. In this aspect, the bacterial infection is caused by a bacterial agent.

[00104] The subject may be any subject capable of colonisation and infection by bacteria. The subject may be mammalian, or may be piscine or avian. Preferably, the subject is selected from the group comprising, but not limited to, human, canine, feline, bovine, ovine, caprine, other ruminant species, porcine, equine, avian, or piscine.

[00105] The compound of Formula I may be administered to the subject in a dose selected from the group comprising 0.1 mg/kg to 250 mg/kg body weight, preferably 1 mg/kg to 100 mg/kg body weight, and more preferably 5 mg/kg to 50 mg/kg body weight. The compound of Formula I may be administered to the subject using a dosing schedule selected from the group consisting of: hourly, 3 times daily; twice daily; daily; every second day; twice weekly; once weekly; once fortnightly; once monthly; once every two months or by constant rate or variable rate infusion. Preferably, the compound of Formula I is administered until colonisation or the signs and symptoms of infection or colonisation have at least been partially treated or alleviated.

[00106] In one embodiment, the concentration of compound of Formula I (or a metabolite) in the subject's blood after treatment is within a range selected from the group comprising, but not limited to: between 0.1 and 10 ug/mL at 2 hours, 1 and 200 ug/mL after 12 hours; between 0.1 and 5 ug/mL after 24 h; between 0.01 and 2 ug/mL after 48 hours; between 0.0001 and 1 ug/mL after 72 h. Preferably, the concentration is selected from the group comprising, but not limited to: less than 200 ug/mL after 12 hours; less than 5 ug/mL after 24 hours; less than 1 ug/L after 48 hours and less than 0.5 ug/mL after 72 hours.

[00107] The agent causing the bacterial infection is a bacterial agent. I n one preferred embodiment, the agent is not a protozoan species. In one preferred embodiment, the agent is not a coccidian protozoan. More preferably, the agent is not Clostridium perfringens nor a heterotrophic bacterial species present in soil samples collected by Hansen et al from Jyndevad Denmark as discussed in the following papers: Hansen et al. 2012, Chemosphere, 86:212-215; and Hansen et al. 2009, Environmental Pollution 157:474-480.

[00108] In another embodiment, the bacterial agent is gram negative. In another embodiment, the bacterial agent is gram positive. In another embodiment, the bacterial agent has no celi wall. In another embodiment, the bacterial infection is caused by a mixture of at least two agents selected from the group consisting of: gram negative, gram positive and bacterial agents with no cell wall.

[00109] The bacterial agent causing the bacterial infection may be a gram positive bacterial agent selected from the group comprising, but not limited to, Staphylococcus spp, Streptococci, Enterococcus spp, Leuconostoc spp, Corynebacterium spp, Arcanobacteria spp, Trueperella spp, Rhodococcus spp, Bacillus spp, Anaerobic Cocci, Anaerobic Gram-Positive Nonsporulating Bacilli, Actinomyces spp, Clostridium spp, Nocardia spp, Erysipelothrix spp, Listeria spp, Kytococcus spp, Mycoplasma spp, Ureaplasma spp, and Mycobacterium spp.

[00110] In one embodiment, the bacterial agent is gram positive and is selected from the group comprising, but not limited to, Staphylococcus spp. Examples of Staphylococcus spp include Staphylococcus epidermidis, Staphylococcus haemolyticus, Staphylococcus lugdunensis, Staphylococcus saprophytics, Staphylococcus auricularis, Staphylococcus capitis, Staphylococcus caprae, Staphylococcus carnosus, Staphylococcus cohnii, Staphylococcus hominis, Staphylococcus pasteuri, Staphylococcus pettenkoferi, Staphylococcus pulvereri, Staphylococcus saccharolyticus, Staphylococcus simulans, Staphylococcus schieiferi, Staphylococcus warneri, Staphylococcus xyiosus, Staphylococcus arlettae, Staphylococcus caseolyticus, Staphylococcus chromogenes, Staphylococcus condimenti, Staphylococcus delphini, Staphylococcus equorum, Staphylococcus felis, Staphylococcus fleurettii, Staphylococcus gallinarum, Staphylococcus hyicus, Staphylococcus intermedius, Staphylococcus kloosii, Staphylococcus lentus, Staphylococcus lutrae, Staphylococcus muscae, Staphylococcus nepalensis, Staphylococcus piscifermentans, Staphylococcus pseudintermedius, Staphylococcus sciuri, Staphylococcus simiae, Staphylococcus succinus, and Staphylococcus vitulinus.

[001 11] In another embodiment, the bacterial agent is gram positive and is selected from the group comprising, but not limited to, Streptococcus spp. Examples of Streptococcus spp include Streptococcus agalactiae, Streptococcus alactolyticus, Streptococcus anginosus, Streptococcus canis, Streptococcus constellatus, Streptococcus cricetus, Streptococcus cristatus , Streptococcus downei, Streptococcus dysgalactiae subs p. dysgalactiae, Streptococcus dysgalactiae subsp. equisimilis, Streptococcus equi subsp. equi, Streptococcus equi subsp. zooepidemicus, Streptococcus ferus, Streptococcus gallolyticus subsp. gallolyticus (formerly Streptococcus bovis biotype i), Streptococcus gallolyticus subsp. pasteurianus (formerly Streptococcus bovis biotype ii/2), Streptococcus gordonii, Streptococcus hyointestinalis, Streptococcus hyovaginalis, Streptococcus infantarius, Streptococcus infantarius subsp infantarius, Streptococcus infantis, Streptococcus iniae, Strepto∞ccus intermedius, Streptococcus lutetiensis (formerly Streptococcus bovis biotype ii.1), Streptococcus macaccae, Streptococcus mitis, Streptococcus mutans, Streptococcus oralis, Streptococcus orisratti, Streptococcus parasanguinis, Streptococcus peroris, Streptococcus pneumoniae, Streptococcus porcinus, Streptococcus pseudintermedius, Streptococcus pyogenes, Streptococcus ratti, Streptococcus salivarius, Streptococcus sanguinis, Streptococcus sobrinus, Streptococcus suis, Streptococcus thermophilus, Streptococcus vestibularis, and Nutritionally Variant (Deficient) Streptococci (Abiotrophia defectiva, Granulicatella adiacens, Granulicatella elegans, and Granulicatella para-adiacens) and related species such as Rothia mucilaginosa (formerly Stomatococcus mucilaginosus ) and Pediococcus.

1001 12] In another embodiment, the bacterial agent is gram positive and selected from the group comprising, but not limited to, Enterococcus spp. Examples of Enterococcus spp include Enterococcus faecalis, Enterococcus faecium, Enterococcus gallinarum, Enterococcus durans, Enterococcus avium, Enterococcus raffinosus, Enterococcus pallens, Enterococcus gilvus, Enterococcus cecorum, Enterococcus malodoratus, Enterococcus italicus, Enterococcus sanguinicola, Enterococcus mundtii, Enterococcus casseliflavus/flavescens, Enterococcus dispar, Enterococcus hirae, Enterococcus pseudoavium, and Enterococcus bovis.

[001 13] In another embodiment, the bacterial agent is gram positive and selected from the group comprising, but not limited to, Leuconostoc spp. Examples of Leuconostoc spp include Leuconostoc mesenteroides, Leuconostoc pseudomesenteroides, Leuconostoc paramesenteroides, Leuconostoc citreum, and Leuconostoc lactis.

1001 14] In another embodiment, the bacterial agent is gram positive and selected from the group comprising, but not limited to, Corynebactenum spp. Examples of Corynebactenum spp include nonlipophilic, fermentative Corynebactenum spp such as Corynebactenum ulcerans, Corynebactenum pseudotuberculosis, Corynebactenum xerosis, Corynebactenum striatum, Corynebactenum minutissimum, Corynebactenum amycolatum, Corynebactenum glucuronolyticum, Corynebactenum argentoratense, Corynebactenum matruchotii, Corynebactenum riegelii, Corynebactenum confusum, Corynebactenum cystidis, Corynebactenum diphtheria, Corynebactenum simulans, Corynebactenum sundvallense, Corynebactenum thomssensii, Corynebactenum freneyi, and Corynebactenum aurimucosum, nonlipophilic, nonfermentative Corynebactenum spp such as Corynebactenum afermentans afermentans, Corynebactenum auris, Corynebactenum pseudodiphtheriticum, and Corynebactenum propinquum and lipophilic Corynebactenum spp such as Corynebactenum jeikeium, Corynebactenum urealyticum, Corynebactenum afermentans lipophilum, Corynebactenum accolens, Corynebactenum macginleyi, Corynebactenum tuberculostearum, Corynebactenum kroppenstedtii, Corynebactenum kutscheri, Corynebactenum pilosum, Corynebactenum bovis, CDC coryneform groups F-1 and G, and Corynebactenum lipophiloflavum, and other Corynebacterium spp such as Turicella, Arthrobacter, Brevibacterium, Dermabacter, Rothia, Oerskovia, Microbacterium, and Leifsonia aquatica.

[001 15] In another embodiment, the bacterial agent is gram positive and selected from the group comprising, but not limited to, Arcanobacteria spp. Examples of Arcanobacteria spp include A. haemolyticum, A. pyogenes ('now known as Trueperella pyogenes, originally known as Actinomyces pyogenes), and A. bernardiae.

[00116] In another embodiment, the bacterial agent is gram positive and selected from the group comprising, but not limited to, Rhodococcus spp. Examples of Rhodococcus spp include Rhodococcus equi, Rhodococcus erythropolis, Rhodococcus fasciens, and Rhodococcus rhodochrous.

[001 17] In another embodiment, the bacterial agent is gram positive and selected from the group comprising, but not limited to, Gordonia spp.

[001 18] in another embodiment, the bacterial agent is gram positive and selected from the group comprising, but not limited to, Tsukamurella spp.

[001 19] In another embodiment, the bacterial agent is gram positive and selected from the group comprising, but not limited to, Acholeplasma spp.

[00120] In another embodiment, the bacterial agent is gram positive and selected from the group comprising, but not limited to, Actinobacteria such as Crossiella equi.

[00121] In another embodiment, the bacterial agent is gram positive and selected from the group comprising, but not limited to, Bacillus spp. Examples of Bacillus spp include Bacillus anthracis, Bacillus cereus, Bacillus circulans, Bacillus licheniformis, Bacillus megaterium, Bacillus pumilus, Bacillus sphaericus, Bacillus subtilis, Brevibacillus brevis, Brevibacillus laterosporus, and Paenibacillus alvei.

[00122] In another embodiment, the bacterial agent is gram positive and selected from the group comprising, but not limited to, Anaerobic Cocci. Examples of Anaerobic Cocci include Anaerococcus murdochii, Anaerococcus prevotii, Anaerococcus tetradius, Anaerococcus octavius, Anaerococcus hydrogenalis, Anaerococcus lactolyticus, Anaerococcus vaginalis, Atopobium parvulum, Finegoldia magna, Gallicola barnesae, Gemella asaccharolytica, Gemella bergeri, Gemella cuniculi, Gemella haemolysans, Gemella morbillorum, Gemella palaticanis, Gemella sanguinis, Parvimonas micra, Peptococcus niger, Peptoniphilus asaccharolyticus, Peptoniphilus gorbachii, Peptoniphilus indolicus, Peptoniphilus harei, Peptoniphilus ivorii, Peptoniphilus lacrimalis, Peptoniphilus olsenii, Peptostreptococcus stomatis, Peptostreptococcus anaerobius, Ruminococcus productus, Slackia heliotrinireducens, and Staphylococcus saccharolyticus.

[00123] In another embodiment, the bacterial agent is gram positive and selected from the group comprising, but not limited to, Anaerobic Gram-Positive Nonsporulating Bacilli. Examples of Anaerobic Gram-Positive Nonsporulating Bacilli include Alloscardovia omnicolens, Atopobium species (such as Atopobium minutum, Atopobium rimae, Atopobium parvulum, and Atopobium vaginae), Bifidobacteria (such as Bifidobacteria adolescentis, Bifidobacteria dentium, Bifidobacteria scardovii), Catabacter hongkongensis, Collinsella aerofaciens, Eggerthella (such as Eggerthella lenta, Eggerthella hongkongensis and Eggerthella sinensis), Eubacterium and related species (such as Eubacterium nodatum, Eubacterium tenue, Eubacterium brachy, Eubacterium infirmum, Eubacterium minutum, Eubacterium nodatum, Eubacterium saphenum, Eubacterium sulci, Filifactor alocis, Mogibacterium timidum, Mogibacterium vescum, Pseudoramibacter alactolyticus, Bulleidia extructa, and Solobacterium moorei), Lactobacillus species (such as Lactobacillus rhamnosus, Lactobacillus casei, Lactobacillus fermentum, Lactobacillus gasseri, Lactobacillus plantarum, Lactobacillus acidophilus, Lactobacillus iners and Lactobacillus ultunensis), Mobiluncus species (such as Mobiluncus curtisii, Mobiluncus mulieris), Moryella indoligenes, Olsenella oral species (such as Olsenella uli and Olsenella profuse), Oribacterium sinus, Propionibacterium (such as Propionibacterium acnes and Propionibacterium propionicum), Slackia exigua, and Turicibacter sanguine.

[00124] In another embodiment, the bacterial agent is gram positive and selected from the group comprising, but not limited to, Actinomyces spp. Examples of Actinomyces spp include Actinomyces israelii, Actinomyces naeslundii, Actinomyces viscosus, Actinomyces odontolyticus, Actinomyces meyeri, and /Aci/nomyces gerencseriae (formerly Actinomyces israelii serotype I I), Actinomyces europaeus, Actinomyces neuii, Actinomyces radingae, Actinomyces graevenitzii, Actinomyces hordeovulneris, Actinomyces turicensis, Actinomyces georgiae, Arcanobacterium {Actinomyces) pyogenes, Arcanobacterium (Actinomyces) bernardiae, Actinomyces funkei, Actinomyces lingnae, Actinomyces houstonensis, and Actinomyces cardiffensis.

[00125] In another embodiment, the bacterial agent is gram positive and selected from the group comprising, but not limited to, Clostridium spp. Examples of Clostridium spp include Clostridium baratii, Clostridium bifermentans, Clostridium botulinum, Clostridium botulinum (types A, B, C, D, E, F, G), Clostridium butyricum, Clostridium difficile, Clostridium histolyticum, Clostridium novyi (type A), Clostridium novyi (type B), Clostridium perfringens, Clostridium perfringens (types A-E), Clostridium ramosum, Clostridium septicum, Clostridium sordelli, Clostridium sphenoides, Clostridium tertium, and Clostridium tetani. [00126] In another embodiment, the bacterial agent is gram positive and selected from the group comprising, but not limited to, Nocardia spp. Examples of Nocardia spp include Nocardia asteroides, Nocardia brasiliensis, Nocardia farcinica, Nocardia nova, Nocardia otitidiscaviarum, and Nocardia transvalensis.

[00127] In another embodiment, the bacterial agent is gram positive and selected from the group comprising, but not limited to, Erysipelothrix spp, such as Erysipelothrix rhusiopathiae.

[00128] In another embodiment, the bacterial agent is gram positive and selected from the group comprising, but not limited to, Listeria spp, such as Listeria monocytogenes.

[00129] In another embodiment, the bacterial agent is gram positive and selected from the group comprising, but not limited to, Kytococcus spp, such as K fococcus schroeteri.

[00130] In another embodiment, the bacterial agent is gram positive and selected from the group comprising, but not limited to, Mycobacterium spp. Examples of Mycobacterium spp include Mycobacterium abscessus, Mycobacterium arupense, Mycobacterium asiaticum, Mycobacterium aubagnense, Mycobacterium avium complex, Mycobacterium bolletii, Mycobacterium bolletii, Mycobacterium branderi, Mycobacterium canettii, Mycobacterium caprae, Mycobacterium celatum, Mycobacterium chelonae, Mycobacterium chimaera, Mycobacterium colombiense, Mycobacterium conceptionense, Mycobacterium conspicuum, Mycobacterium elephantis, Mycobacterium farcinogenes, Mycobacterium florentinum, Mycobacterium fortuitum group, Mycobacterium genavense, Mycobacterium goodii, Mycobacterium haemophilum, Mycobacterium heckeshornense, Mycobacterium heidelbergense, Mycobacterium houstonense, Mycobacterium immunogenum, Mycobacterium interjectum, Mycobacterium intracellulare, Mycobacterium senegalense, Mycobacterium africanum, Mycobacterium avium subsp paratuberculosis, Mycobacterium kansasii, Mycobacterium lacus, Mycobacterium lentiflavum, Mycobacterium leprae, Mycobacterium lepraemurium, Mycobacterium mageritense, Mycobacterium malmoense, Mycobacterium marinum, Mycobacterium massiliense, Mycobacterium microti, Mycobacterium montefiorense (eels), Mycobacterium moracense, Mycobacterium mucogenicum, Mycobacterium nebraskense, Mycobacterium neoaurum, Mycobacterium novocastrense, Mycobacterium palustre, Mycobacterium parmense, Mycobacterium phlei, Mycobacterium phocaicum, Mycobacterium pinnipedii, Mycobacterium porcinum, Mycobacterium pseudoshottsii (fish), Mycobacterium pseudotuberculosis, Mycobacterium saskatchewanense, Mycobacterium scrofulaceum, Mycobacterium senuense, Mycobacterium septicum, Mycobacterium simiae, Mycobacterium smegmatis, Mycobacterium szulgai, Mycobacterium terrae/chromogenicum complex, Mycobacterium triplex, Mycobacterium tuberculosis, Mycobacterium tusciae, Mycobacterium ulcerans, Mycobacterium wolinskyi, and Mycobacterium xenopi. [00131] In another embodiment, the bacterial agent is gram positive and selected from the group comprising, but not limited to, Trueperella spp. Examples of Trueperella spp include Trueperella abortisuis, Trueperella bernardiae, Trueperella bialowiezensis, Trueperella bonasi, Trueperella pyogenes (Arcanobacterium pyogenes).

[00132] In another embodiment, the bacterial agent is gram positive, gram negative or does not have a cell wall and selected from the group comprising, but not limited to, livestock pathogens. Examples of livestock pathogens include Actinobaculum suis, Actinomyces bovis, Arcanobacterium pyogenes, Bacillus anthracis, cereus, licheniformis, pumilus, melaninogenicus, subtilis, Clostridium botulinum, chauvoei, haemolyticum, novyi, perfringens, septicum, sordellii, tetani, colinum, Corynebacterium pseudotuberculosis, renale, Dermatophilus congolensis, Enterococcus spp (such as E. faecalis, E. faecium, E. durans, E. avium, E. hirae), Erysipelothrix rhusiopathiae, Listeria ivanovii, grayi, innocua, seeligeri, welshimeri, monocytogenes, Mycobacterium avium, bovis, paratuberculosis {Johne's Disease), Mycoplasma (such as capricolum subsp. capripneumoniae, subsp. capricolum, M. mycoides subsp mycoides, M. agalactiae, M. ovipneumoniae, M. conjunctivae, M. arginini, M. bovis, and M. putrefaciens) Mycoplasma bovis, dispar, mycoides subsp. mycoides (such as Contagious bovine pleuropneumonia CBPP) Mycoplasma gallisepticum (MG), iowae meleagridis (MM), synoviae (MS) Mycoplasma haemosuis (formerly Eperythrozoon suis), alkalescens, bovigenitalum, bovirhinis, bovoculi, californicum, canadense, cynos, equigenitalium, gateae, haemocanis, haemofelis, hyopneumoniae, hyorhinis, hyosynoviae, iowae, leachii, meleagridis, mycoides subsp capri, wenyonii, suis, Rhodococcus equi, Staphylococcus epidermidis, Staphylococcus simulans, Staphylococcus felis, Staphylococcus xylosus, Staphylococcus chromogenes, Staphylococcus warneri, Staphylococcus haemolyticus, Staphylococcus sciuri, Staphylococcus saprophytics, Staphylococcus hominis, Staphylococcus caprae, Staphylococcus cohnii subsp. cohnii, Staphylococcus cohnii subsp. urealyticus, Staphylococcus capitis subsp. capitis, Staphylococcus capitis subsp. urealyticus, Staphylococcus hyicus, Staphylococcus aureus, Staphylococcus pseudintermedius, Staphylococcus delphini, Staphylococcus schleiferi subsp. coagulans, Staphylococcus aureus subsp. anaerobius, Streptococcus uberis, Streptococcus canis, Streptococcus agalactiae, Streptococcus dysgalactiae, Streptococcus pyogenes, Streptococcus bovis, Streptococcus equi subsp. Zooepidemicus, Streptococcus equinus, Streptococcus equi (Streptococcus equi subsp equi), Streptococcus equisimills (Streptococcus dysgalactiae subsp equisimilis), porcinus, suis, zooepidemicus, Streptococcus zooepidemicus (Streptococcus equi subsp zooepidemicus), Streptococcus dysgalactiae subsp. equisimills, Propionibacterium acnes, Propionibacterium granulosum, Eubacterium, Peptococcus indolicus, and Peptostreptococcus anaerobius; and various species of the following Gram negative genera: Actinobacillus, Aeromonas, Anaplasma, Arcobacter, Avibacterium, Bacteroides, Bartonella, Bordetella, Borrelia, Brachyspira, Brucella, Campylobacter, Capnocytophaga, Chlamydia, Chlamydophila, Chryseobacterium, Coxiella, Cytophaga, Dichelobacter, Edwardsiella, Ehrlichia, Escherichia, Flavobacterium, Francisella, Fusobacterium, Gallibacterium, Haemophilus, Histophilus, Klebsiella, Lawsonia, Leptospira, Mannheimia, Megasphaera, Moraxella, Neorickettsia, Nicoletella, Ornithobacterium, Pasteurella, Photobacterium, Piscichlamydia, Piscirickettsia, Porphyromonas, Prevotella, Proteus, Pseudomonas, Rickettsia, Riemerella, Salmonella, Streptobacillus, Tenacibaculum, Vibrio, and Yersinia.

[00133] In another embodiment, the bacterial agent is selected from the group comprising, but not limited to, pathogens of companion animal species such as cats, dogs and horses. Examples of such pathogens include equine pathogens such as Streptococcus equi, Streptococcus zooepidemicus, Rhodococcus equi, Clostridium difficile, Clostridium perfringens, Corynebacterium pseudotuberculosis, Clostridium piliforme, Actinomyces bovis, Staphylococcus aureus, β-haemolytic Steptococcus spp, Dermatophilus congolense, Clostridiium tetani, and Clostridium botulinum. Further examples include pathogens of dogs and cats such as Staphylococcus spp, Streptococcus spp, Clostridium spp, Actinomyces spp, Enterococcus spp, Nocardia spp, Mycoplasma spp, and Mycobacterium spp.

[00134] In another embodiment, the bacterial agent is gram negative and selected from the group consisting of the following representee families and species: /4cefo0acteraceae Roseomonas cervicalis; Roseomonas fauriae; Roseomonas gilardii. — Aeromonadaceae:- Aeromonas allosaccharophila; Aeromonas aquariorum; Aeromonas caviae; Aeromonas hydrophila (and subspecies); Aeromonas salmonicida; Aeromonas shubertii; Aeromonas veronii biovar sobria (Aeromonas sobria). -- Alcaligenaceae:- Achromobacter xylosoxidans; Alcaligenes faecalis; Bordetella ansorpii; Bordetella avium; Bordetella bronchiseptica; Bordetella hinzii; Bordetella holmesii; Bordetella parapertussis; Bordetella pertussis; Bordetella petrii; Bordetella trematum; Oligella ureolytica; Oligella urethralis. - Anaplasmataceae:- Anaplasma phagocytophilum; Anaplasma platys; Anaplasma bovis; Anaplasma centrale; Anaplasma marginale; Anaplasma odocoilei; Anaplasma ovis; Ehrlichia canis; Ehrlichia chaffeensis; Ehrlichia ewingii; Ehrlichia muris; Ehrlichia ovina; Ehrlichia ruminantium; Neoehrlichia lotoris; Neoehrlichia mikurensis; Neorickettsia helminthoeca; Neorickettsia risticii; Neorickettsia sennetsu; Wolbachia pipientis.— Armatimonadaceae:- Armatimonas rosea. -- Bacteroidaceae:- Bacteroides forsythus; Bacteroides fragilis; Bacteroides melaninogenicus; Bacteroides ruber; Bacteroides urealtyicus. - Bartonellaceae:- Bartonella alsatica; Bartonella australis; Bartonella bacilliformis; Bartonella birtlesii; Bartonella bovis; Bartonella capreoli; Bartonella chomelii; Bartonella clarridgeiae; Bartonella doshiae; Bartonella elizabethae; Bartonella grahamii; Bartonella henselae; Bartonella koehlerae; Bartonella peromysci; Bartonella phoceensis; Bartonella quintana; Bartonella rattimassiliensis; Bartonella rochalimae; Bartonella schoenbuchensis; Bartonella talpae; Bartonella tamiae; Bartonella taylorii; Bartonella tribocorum; Bartonella vinsonii subsp . berkhoffii; Bartonella vinsonii subsp. arupensis; Bartonella vinsonii subsp. vinsonii. - Bdellovibrionaceae:- Bdellovibrio spp.— Brachyspiraceae - Brachyspira spp including Brachyspira hampsonii, Brachyspira hyodysenteriae, Brachyspira murdochii, Brachyspira pilosicoli. - Brucellaceae:- Brucella abortus; Brucella canis; Brucella ceti; Brucella melitensis; Brucella ovis; Brucella pinnipedialis; Brucella suis; Ochrobactrum anthropi; Ochrobactrum intermedium. - Burkholderiaceae:- Burkholderia aboris; Burkholderia ambifaria (genomovar VII); Burkholderia anthina (genomovar VIII); Burkholderia cenocepacia (genomovar III); Burkholderia cepacia (genomovar I); Burkholderia diffusa; Burkholderia dolosa (genomovar VI); Burkholderia latens; Burkholderia mallei; Burkholderia metallica; Burkholderia multivorans (genomovar II); Burkholderia pseudomallei; Burkholderia pyrrocinia (genomovar IX); Burkholderia seminalis; Burkholderia stabilis (genomovar IV); Burkholderia ubonensis (genomovar X); Burkholderia vietnamiensis (genomovar V); Cupriavidus pauculus; Cupriavidus gilardii; Ralstonia pickettii; Ralstonia mannitolilytica; Sphaerotilus hippei; Sphaerotilus montanus; Sphaerotilus natans. - Campylobacteraceae:- Arcobacter spp includng Arcobacter skirrowii; Campylobacter coli; Campylobacter concisus; Campylobacter curvus; Campylobacter fetus; Campylobacter gracilis; Campylobacter helveticus; Campylobacter hominis; Campylobacter hyointestinalis; Campylobacter insulaenigrae; Campylobacter jejuni; Campylobacter lanienae; Campylobacter lari; Campylobacter laridis; Campylobacter mucosalis; Campylobacter rectus; Campylobacter showae; Campylobacter sputorum; Campylobacter upsaliensis. - Candidatus:- Piscichlamydia salmonis. - Cardiobacteriaceae - Cardiobacterium hominis; Cardiobacterium valvarum; Dichelobacter nodosus. - Chlamydiaceae:- Chlamydia spp including Chlamydia avium, Chlamydia gallinacea, Chlamydia muridarum, Chlamydia suis, Chlamydia trachomatis; Chlamydophila spp including Chlamydophila pneumoniae, Chlamydophila pecorum, Chlamydophila psittaci, Chlamydophila abortus, Chlamydophila caviae, and Chlamydophila felis. - Chthonomonadaceae:- Chthonomonas calidirosea. - Comamonadaceae:- Comamonas testosteroni; Verminephrobacter spp. - Coxiellaceae:- Coxiella burnetii. - Cytophagaceae:- Cytophaga columnaris; Cytophaga hutchinsonii; Flexibacter echinicida; Flexibacter elegans; Flexibacter flexilis; Flexibacter litoralis; Flexibacter polymorphus; Flexibacter roseolus; Flexibacter ruber. - Desulfovibrionaceae:- Bilophila wadsworthia; Lawsonia intracellularis. - Enterobacteriaceae:- Cedecea davisae; Cedecea lapagei; Cedecea neteri; amalonaticus; Citrobacter diversus; Citrobacter freundii; Citrobacter koseri; Cronobacter condimenti; Cronobacter dublinensis; Cronobacter helveticus; Cronobacter malonaticus; Cronobacter muytjensii; Cronobacter pulveris; Cronobacter sakazakii; Cronobacter turicensis; Cronobacter universalis; Cronobacter zurichensis; Edwardsiella ictaluri; Edwardsiella tarda; Enterobacter aerogenes; Enterobacter agglomerans; Enterobacter cloacae; Enterobacter cowanii; Escherichia albertii; Escherichia coli, including AIEC = adherent invasive E. coli, EaggEC = enteroaggregative E. coli; EHEC = enterohemorrhagic E. coli; EIEC = enteroinvasive E. coli; EPEC = enteropathogenic E. coli; ETEC = enterotoxigenic E. coli; ExPEC = extraintestinal pathogenic E. coli, NMEC = neonatal meningitis E. coli, NTEC = necrotoxigenic E. co/ , UPEC = uropathogenic E. coll.; Escherichia fergusonii; Ewingella americana; Hafnia alvei; Hafnia paralvei; Klebsiella granulomatis; Klebsiella oxytoca; Klebsiella pneumoniae; Kluyvera ascorbata; Kluyvera cryocrescens; Morganella morganii; Pantoea (formerly Enterobacter) agglomerans; Photorhabdus asymbiotica; Plesiomonas shigelloides; Proteus mirabilis; Proteus penneri; Proteus vulgaris; Providencia alcalifaciens; Providencia rettgeri; Providencia stuartii; Raoultella electrica; Raoultella ornithinolytica; Raoultella planticola; Raoultella terrigena; Salmonella bongori, Salmonella enterica subspecies enterica (many serotypes); Serratia liquifaciens; Serratia marcesans; Shigella boydii; Shigella dysenteriae; Shigella flexneri; Shigella sonnei; Yersinia enterocolitica; Yersinia pestis; Yersinia pseudotuberculosis; Yersinia ruckeri. - Fimbriimonadaceae:- Fimbriimonas ginsengisoli. - Flavobacteriaceae:- Bergeyella zoohelcum; Capnocytophaga canimorsus; Capnocytophaga cynodegmi; Capnocytophaga gingivalis; Capnocytophaga granulosa; Capnocytophaga haemolytica; Capnocytophaga leadbetteri; Capnocytophaga ochracea; Capnocytophaga sputigena; Chryseobacterium indologenes; Chryseobacterium piscicola; Elizabethkingia meningoseptica; Flavobacterium branchiophilum; Flavobacterium coiumnare; Flavobacterium oncorhynchi; Flavobacterium piscicida; Flavobacterium psychrophilum; Myroides odoratus; Myroides odoratimimus; Ornithobacterium rhinotracheale; Riemerella anatipestifer; Riemerella columbina; Riemerella columbipharyngis; Tenacibaculum dicentrarchi; Tenacibaculum discolour; Tenacibaculum gallaicum; Tenacibaculum maritimum; Tenacibaculum soleae; Weeksella virosa. - Francisellaceae - Francisella tularensis subsp. tularensis; Francisella tularensis subsp. holarctica; Francisella tularensis subsp. novicida; Francisella philomiragia; Francisella noatunensis; Francisella noatunensis subsp. orientalis (also termed Francisella asiatica). - Fusobacteriaceae:- Fusobacterium spp. including Fusobacterium necrophorum, Fusobacterium nucleatum, Fuso-bacterium polymorphum. - Helicobacteraceae:- Helicobacter cinaedi; Helicobacter fennelliae; Helicobacter pylori. -- Legionellaceae:- Legionella pneumophila and other species including; Legionella anisa; Legionella birminghamensis; Legionella bozemannii; Legionella cincinnatiensis; Legionella dumoffii; Legionella feeleii; Legionella gormanii; Legionella hackeliae; Legionella jordanis; Legionella lansingensis; Legionella longbeachae; Legionella maceachernii; Legionella micdadei; Legionella oakridgensis; Legionella parisiensis; Legionella sainthelens; Legionella tusconensis; Legionella wadsworthii; Legionella waltersii. - Leptospiraceae:- Leptospira alexanderi (including Leptospira alexanderi serovar Hebdomadis, Leptospira alexanderi serovar Manhao 3); Leptospira alstoni (including Leptospira alstoni serovar Pingchang, Leptospira alstoni serovar Sichuan); Leptospira biflexa (including Leptospira biflexa serovar Ancona, Leptospira biflexa serovar Canela); Leptospira borgpetersenii (including Leptospira borgpetersenii serovar Hardjo, Leptospira borgpetersenii serovar Hardjo-bovis, Leptospira borgpetersenii serovar Pomona, Leptospira borgpetersenii serovar Tarassovi); Leptospira broomii (including Leptospira broomii serovar Hurstbridge); Leptospira fainei (including Leptospira fainei serovar Hurstbridge); Leptospira idonii; Leptospira inadai (including Leptospira inadai serovar Lyme, Leptospira inadai serovar Malaya); Leptospira interrogans (including Leptospira interrogans serovar Australis, Leptospira interrogans serovar Autumnalis, Leptospira interrogans serovar Bratislava, Leptospira interrogans serovar Canicola, Leptospira interrogans serovar Grippotyphosa, Leptospira interrogans serovar Hardjo, Leptospira interrogans serovar Hardjo-bovis, Leptospira interrogans serovar lcterohaemorrhagiae, Leptospira interrogans serovar Pomona, Leptospira interrogans serovar Pyrogenes, Leptospira interrogans serovar Tarassovi); Leptospira kirschneri (including Leptospira kirschneri serovar Bulgarica, Leptospira kirschneri serovar Cynopteri, Leptospira kirschneri serovar Grippotyphosa); Leptospira kmetyi; Leptospira licerasiae; Leptospira meyeri (including Leptospira meyeri serovar Sofia); Leptospira noguchii (including Leptospira noguchii serovar Panama, Leptospira noguchii serovar Pomona); Leptospira santarosai; Leptospira terpstrae; Leptospira vanthielii; Leptospira weilii (including Leptospira weilii serovar Celledoni, Leptospira weilii serovar Sarmin); Leptospira wolbachii; Leptospira wolffii; Leptospira yanagawae. - Leptotrichiaceae:- Leptotrichia buccalis; Streptobacillus moniliformis. - Methylobacteriaceae - Methylobacterium extorquens group; Methylobacterium fujisawaense; Methylobacterium mesophilicum; Methylobacterium zatmanii. - Moraxellaceae:- Acinetobacter baumannii (genomic species 2); Acinetobacter baylyi; Acinetobacter bouvetii; Acinetobacter ca/coacef/ci/s (genomic species 1); Acinetobacter gerneri; Acinetobacter grimontii; Acinetobacter haemolyticus (genomic species 4); Acinetobacter johnsonii (genomic species 7); Acinetobacter junii (genomic species 5); Acinetobacter Iwoffi (genomic species 8/9); Acinetobacter parvus; Acinetobacter radioresistens (genomic species 12); Acinetobacter schindleri; Acinetobacter tandoii; Acinetobacter tjernbergiae; Acinetobacter towneri; Acinetobacter ursingii; Acinetobacter venetianus; Moraxella atlantae; Moraxella boevrei; Moraxella bovis; Moraxella bovoculi; Moraxella canis; Moraxella caprae; Moraxella catarrhalis; Moraxella caviae; Moraxella cuniculi; Moraxella equi; Moraxella lacunata; Moraxella lincolnii; Moraxella macacae; Moraxella nonliquefaciens; Moraxella oblonga; Moraxella osloensis; Moraxella ovis; Moraxella phenylpyruvica; Moraxella pluranimalium; Moraxella pore/. ~ Moritellaceae:- Moritella abyssi; Moritella dasanensis; Moritella japonica; Moritella marina; Moritella pro-funda; Moritella viscosa; Moritella yayanosii. - Neisseriaceae:- Chromobacterium violaceum; Eikenella corrodens; Kingella denitrificans, Kingella kingae, Kingella oralis, Kingella potus; Neisseria cinerea; Neisseria elongata; Neisseria flavescens; Neisseria gonorrhoeae; Neisseria /acfam/ca; Neisseria meningitidis; Neisseria mucosa; Neisseria polysaccharea; Neisseria sicca; Neisseria subflava; Neisseria weaver; Vitreoscilla spp.— Nitrosomonadaceae:- Nitrosomonas eutropha; Nitrosomonas halophila; Nitrosomonas oligotropha. Paste urellaceae:- Actinobacillus actinomycetemcomitans; Actinobacillus equuli; Actinobacillus lignieresii; Actinobacillus pleuropneumoniae; Actinobacillus seminis; Actinobacillus succinogenes; Actinobacillus ureae; Aggregatibacter actinomycetemcomitans, Aggregatibacter segnis, Aggregatibacter aphrophilus; Avibacterium avium; Avibacterium endocarditidis; Avibacterium gallinarum; Avibacterium paragallinarum; Avibacterium volant/um; Bibersteinia trehalose; Gallibacterium anatis; Gallibacterium genomospecies 1; Gallibacterium genomospecies 2; Gallibacterium genomospecies 3; Gallibacterium group V; Gallibacterium melopsittaci; Gallibacterium salpingitidis; Gallibacterium trehalosifermentans; Haemophilus aegyptius; Haemophilus avium; Haemophilus ducreyi; Haemophilus haemolyticus; Haemophilus influenzae; Haemophilus parahaemolyticus; Haemophilus parainfluenzae; Haemophilus parasuis; Histophilus somni; Mannheimia cav/ ' ae; Mannheimia glucosida; Mannheimia granulomatis; Mannheimia haemolytica; Mannheimia ruminalis; Mannheimia varigena; Nicoletella semolina; Pasteurella aerogenes; Pasteurella bettyae; Pasteurella caballi; Pasteurella canis; Pasteurella dagmatis; Pasteurella multocida (subspecies multocida, septicum, gallicida); Pasteurella pneumotropica; Pasteurella stomatis; Pasteurella trehalosi. - Piscirickettsiaceae:- Piscirickettsia salmonis. - Plesiomonadaceae:- Plesiomonas shigelloides. - - Polyangiaceae:- Sorangium cellulosum. -- Porphyromonadaceae:- Dysgonomonas capnocytophagoides; Dysgonomonas gadei; Dysgonomonas hofstadii; Dysgonomonas mossii; Dysgonomonas oryzarvi; Dysgonomonas wimpennyi; Porphyromonas gingivalls. Prevotellaceae:- Prevotella spp. including Prevotella intermedia, Prevotella melaninogenica. ~ Pseudomonadaceae - Chryseomonas luteola; Pseudomonas aeruginosa; Pseudomonas luteoia; Pseudomonas fluorescens; Pseudomonas putida; Pseudomonas stutzeri; Pseudomonas oryzihabitans. - Rhizobiaceae:- Agrobacterium tumefaciens; Rhizobium radiobacter. -- Rickettsiaceae:- Orientia chuto; Orientia tsutsugamushi; Rickettsia aeschlimannii; Rickettsia africae; Rickettsia akari; Rickettsia argasii; Rickettsia asiatica; Rickettsia australis; Rickettsia bellii; Rickettsia canadensis; Rickettsia conorii; Rickettsia cooleyi; Rickettsia felis; Rickettsia heilongjiangensis; Rickettsia helvetica; Rickettsia honei; Rickettsia hoogstraalii; Rickettsia hulinensis; Rickettsia hulinii; Rickettsia japonica; Rickettsia marmionii; Rickettsia martinet; Rickettsia massiliae; Rickettsia monacensis; Rickettsia montanensis; Rickettsia monteiroi; Rickettsia moreli; Rickettsia parkeri; Rickettsia peacockii; Rickettsia philipii; Rickettsia prowazekii; Rickettsia raoultii; Rickettsia rhipicephali; Rickettsia rickettsii; Rickettsia sibirica subgroup; Rickettsia slovaca; Rickettsia tamurae; Rickettsia typhi. - Shewanellaceae:- Shewanella putrefaciens. ~ Sphingomonadaceae - Sphingobacterium multivorum; Sphingobacterium spiritivorum; Sphingomonas paucimobilis. - Spirillaceae:- Spirillum minus; Spirillum volutans; Spirillum winogradskyi. - Spirochaetaceae:- Borrelia afzelii; Borrelia anserina; Borrelia bissettii; Borrelia burgdorferi; Borrelia coriaceae; Borrelia duttonii; Borrelia garinii; Borrelia hermsii; Borrelia hispanica; Borrelia japonica; Borrelia lonestari; Borrelia lusitaniae; Borrelia miyamotoi; Borrelia parkeri; Borrelia persica; Borrelia recurrentis; Borrelia spielmanii; Borrelia turicatae; Borrelia turicatae; Borrelia valaisiana; Treponema carateum; Treponema pallidum ssp. endemicum; Treponema pallidum ssp. pallidum; Treponema pallidum ssp. pertenue. - Succinivibrionaceae:- Anaerobiospirillum spp. - Sutterellaceae:- Sutterella spp including Sutterella wadsworthia. — Thermaceae:- Meiothermus spp. - Thermotogaceae - Thermotoga neapolitana. - Veillonellaceae:- Dialister spp; Megamonas spp; Megasphaera spp; Pectinatus spp; Pelosinus spp; Propionispora spp; Sporomusa spp; Veillonella spp.; Zymophilus spp. -- Vibrionaceae:- Photobacterium damselae; Vibrio adaptatus; Vibrio alginolyticus; Vibrio azasii; Vibrio campbellii; Vibrio cholera; Vibrio damsel; Vibrio fluvialis; Vibrio furnisii; Vibrio hollisae; Vibrio metchnikovii; Vibrio mimicus; Vibrio parahaemolyticus; Vibrio vulnificus. — Wolbachieae - Wolbachia spp. - Xanthomonadaceae:- Luteimonas aestuarii; Luteimonas aquatica; Luteimonas composti; Luteimonas lutimaris; Luteimonas marina; Luteimonas mephitis; Luteimonas vadosa; Pseudoxanthomonas broegbernensis; Pseudoxanthomonas japonensis; Stenotrophomonas maltophilia; Stenotrophomonas nitritireducens.

[00135] Most preferably, the bacterial agent causing the bacterial infection is gram negative and is selected from the group comprising: Acinetobacter species, Aeromonas hydrophila, Citrobacter species, Enterobacter species, Escherichia co/ , Klebsiella pneumoniae, Morganella morganii, Pseudomonas aeruginosa, and Stenotrophomonas maltophilia.

[00136] In another preferred embodiment, the bacteria agent causing the bacterial colonisation or infection is resistant to a conventional antibiotic used to treat the colonisation or infection. In one preferred embodiment, the bacterial agent is resistant to a compound selected from the group comprising: one or more of aminoglycosides (for example gentamicin, tobramycin, amikacin, or netilmicin); anti-MRSA cephalosporins (for example ceftaroline); antipseudomonal penicillins + β-lactamase inhibitors (for example ticarcillin-clavulanic acid or piperacillin-tazobactam); carbapenems (for example ertapenem, imipenem, meropenem or doripenem); non-extended spectrum cephalosporins; 1st and 2nd generation cephalosporins (for example cefazolin or cefuroxime); extended-spectrum cephaiosporins; 3rd and 4th generation cephalosporins (for example cefotaxime or ceftriaxone); cephamycins (for example cefoxitin or cefotetan); fluoroquinolones (for example ciprofloxacin); folate pathway inhibitors (for example trimethoprim-sulphamethoxazole); glycylcyclines (for example tigecycline); monobactams (for example aztreonam); penicillins (for example ampicillin); penicillins + β- lactamase inhibitors (for example amoxicillin-clavulanic acid or ampicillin-sulbactam); phenicols (for example chloramphenicol); phosphonic acids (for example fosfomycin); polymyxins (for example colistin); and tetracyclines (for example tetracycline, doxycycline or minocycline. Preferably, the bacterial agent resistant to these compounds is gram negative.

[00137] Preferably, the bacterial agent is resistant to a compound selected from the group comprising: penicillins, cephalosporins, carbapenems, monobactams and other β-lactam antibiotics, fusidanes, aminoglycosides, fluoroquinolones, streptogramins, tetracyclines, glycylcyclines, chloramphenicol and other phenicols, macrolides and ketolides, lincosamides, oxazolidinones, aminocyclitols, polymyxins, glycopeptides, lipopeptides, bacitracin, mupiricin, pleuromutilins, rifamycins, sulphonamides and trimethoprim. Preferably, the compound is selected from the group comprising: beta lactams, glycopeptides, lipopeptides, macrolides, oxazolidinones and tetracyclines. Preferably, the bacterial agent is resistant to the compound when the compound is at a concentration range selected from the following: 0.001 μg/mL - 10,000 μg mL; 0.01 μg/mL - 1000 μg/mL; 0.10 μg/mL - 100 μg mL; and 1 μg/mL - 50 μg/mL.

100138] In another preferred embodiment, the bacterial agent causing the bacterial infection is selected from the group comprising, but not limited to, gram positive bacteria. The microbe is preferably a gram positive bacterial agent selected from the group comprising Staphylococcus aureus, Staphylococcus pseudintermedius, Streptococcus pneumoniae, Streptococcus pyogenes, Streptococcus agalactiae, Streptococcus uberis, Enterococcus faecium, Enterococcus faecalis, and Clostridium difficile.

[00139] In one preferred embodiment, the bacterial agent has no cell wall. Preferably, the bacterial agent is selected from the group comprising: Mycoplasma spp, Mycoplasma agalactiae, Mycoplasma alkalescens, Mycoplasma amphoriforme, Mycoplasma arginini, Mycoplasma bovigenitalum, Mycoplasma bovirhinis, Mycoplasma bovis, Mycoplasma bovoculi, Mycoplasma buccale, Mycoplasma californicum, Mycoplasma canadense, Mycoplasma capricoium subsp. capricoium, Mycoplasma capricoium subsp. capripneumoniae, Mycoplasma conjunctivae, Mycoplasma cynos, Mycoplasma dispar, Mycoplasma equigenitalium, Mycoplasma faucium, Mycoplasma felis, Mycoplasma fermentans (incognitus str.), Mycoplasma gallisepticum (MG), Mycoplasma gateae, Mycoplasma genitalium, Mycoplasma haemocanis, Mycoplasma haemofelis, Mycoplasma haemosuis (formerly Eperythrozoon suis), Mycoplasma hominis, Mycoplasma hyopneumoniae, Mycoplasma hyorhinis, Mycoplasma hyosynoviae, Mycoplasma iowae meleagridis (MM), Mycoplasma iowae, Mycoplasma leachii, Mycoplasma lipophilum, Mycoplasma meleagridis, Mycoplasma mycoides subsp capri, Mycoplasma mycoides subsp mycoides, Mycoplasma mycoides subsp. mycoides (such as Contagious bovine pleuropneumonia CBPP), Mycoplasma orale, Mycoplasma ovipneumoniae, Mycoplasma ovis, Mycoplasma penetrans, Mycoplasma pirum, Mycoplasma pneumoniae, Mycoplasma primatum, Mycoplasma putrefaciens, Mycoplasma salivarium, Mycoplasma sperm atophil urn, Mycoplasma suis, Mycoplasma synoviae (MS), Mycoplasma wenyonii, Mycoplasma, Ureaplasma spp, Ureaplasma parvum, Ureaplasma urealyticum, Ureaplasma, and Ureoplasma diversum.

[00140] In another most preferred embodiment, the bacterial agent is Staphylococcus aureus.

[00141] In another preferred embodiment, the bacterial agent is resistant to a compound selected from the group comprising: one or more of aminoglycosides (for example gentamicin); ansamycins {for example rifampicin); anti-MRSA cephalosporins (for example ceftaroline); anti- staphylococcal β-lactams (or cephamycins) (for example oxacillin or cefoxitin); carbapenems (for example ertapenem, imipenem, meropenem or doripenem); non-extended spectrum cephalosporins; 1 st and 2nd generation cephalosporins (for example cefazolin or cefuroxime); extended-spectrum cephalosporins; 3rd and 4th generation cephalosporins (for example cefotaxime or ceftriaxone); cephamycins (for example cefoxitin or cefotetan); fluoroquinolones (for example ciprofloxacin or moxifloxacin); folate pathway inhibitors (for example trimethoprim- sulphamethoxazole); fucidanes (for example fusidic acid); glycopeptides (for example vancomycin, teicoplanin or telavancin); glycylcyclines (for example tigecycline); lincosamides (for example clindamycin); lipopeptides (for example daptomycin); macrolides (for example erythromycin); oxazolidinones (for example linezolid or tedizolid); phenicols (for example chloramphenicol); phosphonic acids (for example fosfomycin); streptogramins (for example quinupristin-dalfopristin); and tetracyclines (for example tetracycline, doxycycline or minocycline). Preferably, the bacterial agent resistant to these compounds is gram positive.

[00142] In another most preferred embodiment, the bacterial agent is Streptococcus pneumoniae. The Streptococcus pneumoniae may be a strain that is resistant to one or more of β-lactams and macrolides.

[00143] In another most preferred embodiment, the bacterial agent is Streptococcus pyogenes.

[00144] I n another most preferred embodiment, the bacterial agent is Streptococcus agalactiae.

[00145] In another most preferred embodiment, the bacterial agent is either Enterococcus faecium or Enterococcus faecalis. The Enterococcus faecium or Enterococcus faecalis may be a strain that is resistant to aminoglycosides (for example gentamicin (high level) or streptomycin (for example streptomycin (high level)); carbapenems (for example imipenem, meropenem or doripenem); fluoroquinolones (for example ciprofloxacin, levofloxacin or moxifloxacin); glycopeptides (for example vancomycin or teicoplanin); glycylcyclines (for example tigecycline); lipopeptides (for example daptomycin); oxazolidinones (for example linezolid); penicillins (for example ampiciilin); streptogramins (for example quinupristin-dalfopristin); tetracycline (for example doxycycline or minocycline).

[00146] In another most preferred embodiment, the bacterial agent is Clostridium difficile.

[00147] The bacterial infection in the subject may cause a disease selected from the group comprising, but not limited to, nosocomial pneumonia caused by Staphylococcus aureus (MDR, XDR, PDR or methicillin-susceptible or -resistant strains), or invasive pneumococcal diseases such as pneumonia, bronchitis, acute sinusitis, otitis media, conjunctivitis, meningitis, bacteremia, sepsis, osteomyelitis, septic arthritis, endocarditis, peritonitis, pericarditis, cellulitis, and brain abscess caused by Streptococcus pneumoniae (including multi-drug resistant strains [MDRSP] such as those resistant to β-lactams and macrolides), complicated skin and skin structure infections, including diabetic foot infections, with or without concomitant osteomyelitis, caused by Staphylococcus aureus (methicillin-susceptible and -resistant strains), Streptococcus pyogenes, or Streptococcus agalactiae, uncomplicated skin and skin structure infections caused by Staphylococcus aureus (methicillin-susceptible and -resistant strains) or Streptococcus pyogenes, community-acquired pneumonia caused by Streptococcus pneumoniae (including multi-drug resistant strains [MDRSP], including cases with concurrent bacteraemia, or Staphylococcus aureus (methicillin-susceptible and -resistant strains) and Staphylococcus aureus bloodstream Infections (bacteraemia), including those with right-sided infective endocarditis, caused by methicillin-susceptible and methicillin-resistant isolates, vancomycin- resistant enterococcus infections, including cases with concurrent bacteraemia, and treatment of Clostridium d/ f/c//e-associated diarrhea (CDAD).

[00148] Gram negative organisms are important causes of many infectious diseases in humans and other animal species. Bone and joint infections (Gram-negative organisms or mixed bacteria, are an important cause of vertebral osteomyelitis and septic arthritis), cardiovascular system infections (including endocarditis caused by the HACEK group - Haemophilus parainfluenzae, Haemophilus aphrophilus, Aggregatibacter actinomycetemcomitans, Cardiobacterium hominis, Eikenella corrodens, Kingella kingae), central nervous system infections (the commonest causes of bacterial meningitis are Neisseria meningitidis, Streptococcus pneumoniae and, in nonvaccinated young children, Haemophilus influenzae type b (Hib), in neonates and infants less than 3 months of age, Streptococcus agalactiae (group B streptococcus), Escherichia coli and other aerobic Gram-negative rods are important pathogens, brain abscess or subdural empyema, the infecting organism(s) vary with the underlying predisposing cause but where the likely site of origin is the ear, enteric Gram- negative bacilli are commonly involved), eye infections (common pathogens include Haemophilus influenza, Neisseria gonorrhoeae or Chlamydia trachomatis), gastrointestinal tract infections (a wide range of pathogens are implicated including enterotoxigenic Escherichia coli (ETEC), Salmonella, Campylobacter, Shigella, Vibrio cholera and Yersinia enterocolitica), genital infections (bacterial vaginosis is a polymicrobial clinical syndrome with high concentrations of anaerobic (eg Mobiluncus species) and other fastidious bacteria (including Gardnerella vaginalis and Atopobium vaginae), and Mycoplasma hominis; non-sexually acquired pelvic inflammatory disease (PID) is usually caused by mixed vaginal flora, including anaerobes, facultative Gram-negative bacteria and Mycoplasma hominis, while sexually acquired PID is usually initiated by C. trachomatis or N. gonorrhoeae with growing evidence that M. genitalium infection is involved in a significant minority of cases), intra-abdominal infections (peritonitis due to perforated viscus is usually a polymicrobial infection with aerobic and anaerobic bowel flora while spontaneous bacterial peritonitis (SBP) is usually caused by enteric Gram-negative bacilli, such as Escherichia co// and Klebsiella species, Klebsiella pneumoniae is an increasingly identified cause of liver abscess), community-acquired pneumonia (Mycoplasma pneumoniae, Chlamydophila (Chlamydia) pneumoniae, Chlamydophila (Chlamydia) psittaci, Haemophilus influenza, aerobic Gram-negative bacilli including Klebsiella pneumonia, Pseudomonas aeruginosa, Acinetobacter baumannii, Burkholderia pseudomallei), otitis externa (including acute diffuse) (bacterial cultures commonly yield Pseudomonas aeruginosa, Staphylococcus aureus, and Proteus and Klebsiella species), otitis media (including acute) (common bacterial pathogens include Streptococcus pneumoniae, Haemophilus influenzae and Moraxella catarrhalis), sepsis (including severe) (including Acinetobacter baumannii, disseminated gonococcal sepsis, Gram-negative enteric bacteria, Neisseria meningitidis (meningococcal sepsis) and Pseudomonas aeruginosa), Systemic infections (Spotted fevers (Rickettsia) and scrub typhus (Orientia), Brucellosis, Cat-scratch disease and other Bartonella infections, Leptospirosis, Lyme disease, Melioidosis, Q fever, Typhoid and paratyphoid fevers (enteric fevers), urinary tract infections (acute cystitis, acute pyelonephritis, recurrent urinary tract infections and atheter-associated bacteriuria and urinary tract infections).

[00149] In humans gram negative bacteria are common causes of intra-abdominal infections (lAls), urinary tract infections (UTIs), hospital acquired pneumonia, and bacteraemia. Escherichia coli (E. coli), Klebsiella pneumoniae (K. pneumoniae), and Pseudomonas aeruginosa (P. aeruginosa) are important pathogens in the hospital setting, accounting for 27% of all pathogens and 70% of all Gram-negative pathogens causing healthcare-associated infections [Sieved DM, Ricks P, Edwards JR, et al. Antimicrobial-resistant pathogens associated with healthcare-associated infections: summary of data reported to the National Healthcare Safety Network at the Centers for Disease Control and Prevention, 2009-2010. Infect Control Hosp Epidemiol. 2013;34: 1-14.].

[00150] Gram negative bacteria are showing rising rates of resistance to current therapies. The production of extended-spectrum β-lactamase (ESBL) enzymes is a common mechanism of resistance. Rates of ESBL-producing E. coli and K. pneumoniae have risen substantially, with the result that these bacteria are increasingly resistant to widely used antimicrobials.

[00151 ] P. aeruginosa is the most common Gram-negative cause of nosocomial pneumonia and the second most common cause of catheter- related UTIs in the U.S.

[00152] E. coli is the most common cause of UTIs. Cases of UTI caused by ESBL- producing E. coli and K. pneumonia as well as P. aeruginosa, including MDR strains, are increasing. ESBL-producing E. coli and K. pneumoniae are also frequently isolated in patients with complicated IAI (clAI). [00153] P. aeruginosa is a clinically challenging and virulent pathogen that can be a cause of common infections in humans such as nosocomial pneumonia, UTI, IAI, and bloodstream infections. P. aeruginosa is the most common Gram-negative organism causing ventilator associated pneumonia and the second most common cause of catheter-associated UTIs.

[00154] The increase in the number of infections caused by Gram-negative bacteria is being accompanied by rising rates of resistance. Treatment options to meet this challenge are increasingly limited. There is a critical need for new antibiotics to meet the needs of patients now and in the future.

[00155] In one preferred aspect, more than one compound of the invention is administered to the subject.

[00156] In another preferred embodiment, a compound of the invention, or a therapeutically acceptable salt thereof, is administered together with a compound or agent that removes or substantially removes or reduces the integrity of the cell wall of the bacterial agent. As an example, the compound is selected from the group consisting of: β lactams, fosfomycin, lysozyme, polymyxins and chelating agents such as ethylenediaminetetraacetic acid (EDTA). As an example, the agent is an immunological agent {such as an antibody or vaccine) that reduces the integrity of the cell wall. In one preferred embodiment, the compound, or a therapeutically acceptable salt thereof, is administered together with a compound that removes or substantially removes or weakens the integrity of the outer cell wall of a gram negative bacterial agent.

[00157] According to another aspect of the invention, there is provided an antibacterial pharmaceutical composition comprising a therapeutically effective amount of a compound of Formula I, or a therapeutically acceptable salt thereof. Preferably, the composition is an antibacterial pharmaceutical composition.

[00158] According to another aspect of the invention, there is provided an antibacterial veterinary composition comprising a therapeutically effective amount of a compound of Formula I, or a therapeutically acceptable salt thereof. Preferably, the composition is an anti-bacterial veterinary composition.

[00159] The pharmaceutical composition may optionally include a pharmaceutically acceptable excipient or carrier. The veterinary composition may optionally include a veterinary acceptable excipient or carrier.

[00160] The pharmaceutical or veterinary composition of the invention preferably contains a compound of Formula I, or a pharmaceutically acceptable salt, at a concentration of selected from the groupd consisting of: 1 mg/g to 500 mg/g; 5 mg to 400 mg/g; 10 mg/g to 200 mg/g; 20 mg/g to 100 mg/g; 30 mg/g to 70 mg/g; and 40 mg/g to 60 mg/g.

[00161 ] In another embodiment, the pharmaceutical or veterinary composition comprises impurities, wherein the quantity of impurities as a percentage of the total weight of the composition is selected from the group consisting of: less than 20% impurities (by total weight of the composition); less than 15% impurities; less than 10% impurities; less than 8% impurities; less than 5% impurities; less than 4% impurities; less than 3% impurities; less than 2% impurities; less than 1 % impurities; less than 0.5% impurities; less than 0.1 % impurities. In one embodiment, the pharmaceutical or veterinary composition comprises microbial impurities or secondary metabolites, wherein the quantity of microbial impurities as a percentage of the total weight of the composition is selected from the group consisting of: less than 5%; less than 4%; less than 3%; less than 2%; less than 1 %; less than 0.5%; less than 0.1 %; less than 0.01 %; less than 0.001 %. In one embodiment, the pharmaceutical or veterinary composition is sterile and stored in a sealed and sterile container. In one embodiment, the pharmaceutical or veterinary composition contains no detectable level of microbial contamination.

[00162] The pharmaceutical or veterinary composition of the invention may comprise a further antimicrobial agent. The further antimicrobial agent may be an antifungal agent or antibacterial agent. The method of treating or preventing a bacterial infection or colonisation in a subject, may also comprise the administration of a compound of the invention with a further antimicrobial agent.

[00163] The pharmaceutical or veterinary composition of the invention may comprise more than one compound of the invention. For example, a combination of compounds. The method of treating or preventing a bacterial infection or colonisation in a subject, may also comprise the administration of more than one compound of the invention.

[00164] In one embodiment, the antifungal agent is selected from the group comprising, but not limited to naturally occurring agents including Echinocandins (Anidulafungin, Caspofungin, Micafungin), Polyenes (Amphotericin B, Candicidin, Filipin, Fungichromin (Pentamycin), Hachimycin, Hamycin, Lucensomycin, Mepartricin, Natamycin, Nystatin, Pecilocin, Perimycin), and other naturally occurring antifungal agents including Griseofulvin, Oligomycins, Pyrrolnitrin, Siccanin, and Viridin. The antifungal agent may be a synthetic compound selected from the group comprising, but not limited to Allylamines (Butenafine, Naftifine, Terbinafine) Imidazoles (Bifonazole, Butoconazole, Chlormidazole, Climbazole, Croconazole (Cloconazole), Clotrimazole, Eberconazole, Econazole, Enilconazole, Fenticonazole, Flutrimazole, Fosfluconazole, Isoconazole, Ketoconazole, Lanoconazole, Luliconazole, Miconazole, Neticonazole, Omoconazole, Oxiconazole Nitrate, Parconazole, Sertaconazole, Sulconazole, Tioconazole), Thiocarba mates (Liranaftate, Tolciclate, Tolindate, Tolnaftate), Triazoles (Fluconazole, Isavuconazole, Itraconazole, Posaconazole, Ravuconazole, Saperconazole, Terconazole, Voriconazole), and other synthetic agents such as Acrisorcin, Amorolfine, Bromosalicylchloranilide (Bromochlorosalicylanilide), Buclosamide, Calcium Propionate, Chlorphenesin, Ciclopirox, Cloxyquin (Cloxiquine), Coparaffinate, Exalamide, Flucytosine, Haloprogin, Hexetidine, Loflucarban, Nifuratel, Nifuroxime, Piroctone, Potassium Iodide, Propionic Acid, Pyrithione, Salicylanilide, Sodium Parachlorobenzoate, Sodium Propionate, Sulbentine, Tenonitrozole, Triacetin, Trimetrexate, Undecylenic Acid (Undecenoic Acid), and Zinc Propionate.

[00165] The composition of the invention may comprise an antibiotic adjunct selected from the group comprising, but not limited to, β-Lactamase Inhibitors (Avibactam, Clavulanic Acid, Sulbactam, Sultamicillin, Tazobactam), Renal Dipeptidase Inhibitors (Cilastatin), and Renal Protectant (Betamipron).

[00166] In one embodiment, the composition of the invention comprises a further antibiotic selected from the group comprising, but not limited to, 2,4-DIAMINOPYRIMIDI NES, including Baquiloprim, Brodimoprim, lclaprim, Ormetoprim, Pyrimethamine, Tetroxoprim, Trimethoprim; AMINOCOUMARI NS, including Novobiocin; AMI NOCYCLITOLS, including Spectinomycin,; AMINOGLYCOSIDES, including Amikacin, Apramycin, Arbekacin, Bekanamycin, Butirosin, Dibekacin, Dihydrostreptomycin, Etimicin, Fortimicins (Astromicin), Framycetin, Gentamicin, Hygromycin B, Isepamicin, Kanamycin, Micronomicin, Neomycin, Netilmicin, Paromomycin, Plazomicin, Ribostamycin, Sisomicin, Streptomycin, Tobramycin, Verdamicin; AMI NOMETHYLCYCLI NES, including Omadacycline; AMPHENICOLS, including Azidamfenicol, Chloramphenicol, Florfenicol, Thiamphenicol; ANSAMYCINS, including Rifabutin, Rifamide, Rifampin (Rifampicin), Rifamycin, Rifapentine, Rifaximin; ANTISEPTIC AGENTS, including Acridine derivatives (including acriflavine, aminoacridine, ethacridine, proflavine), Bispyridines (including octenidine dihydrochloride), Brominated salicylanilides (including bromsalans), Chlorhexidine, Phenol derivatives (including thymol and triclosan), Quarternary ammonium compounds (including Alkyldimethylethylbenzyi Ammonium Chloride, benzalkonium chloride, cetylpyridinium chloride, benzethonium chloride, cetrimonium); ANTITUBERCULAR AGENTS, including Cycloserine, Delamanid, Ethambutol, Ethionamide, Isoniazid (Ftivazide), Morinamide, p-Aminosalicylic Acid (PAS), Protionamide, Pyrazinamide, Terizidone, Thioacetazone, Tiocarlide; ARSENICALS, including Arsanilic Acid, Roxarsone; BACTERIOCINS, including Nisin, Brilacidin (PMX-30063); β-LACTAM CARBACEPHEMS, including Loracarbef; β-LACTAM CARBAPENEMS, including Biapenem, Doripenem, Ertapenem, Faropenem, Imipenem, Meropenem, Panipenem, Razupenem, Ritipenem, Sulopenem, Tebipenem, Tomopenem; β-LACTAM CEPHALOSPORINS, including Cefacetrile, Cefaclor, Cefadroxil, Cefalexin, Cefaloglycin, Cefalonium, Cefaloridine, Cefalothin, Cefamandole, Cefapirin, Cefatrizine, Cefazaflur, Cefazedone, Cefazolin, Cefcapene, Cefdinir, Cefditoren, Cefepime, Cefetamet, Cefixime, Cefmenoxime, Cefodizime, Cefonicid, Cefoperazone, Ceforanide, Cefose!is, Cefotaxime, Cefotiam, Cefovecin, Cefozopran, Cefpimizole, Cefpiramide, Cefpirome, Cefpodoxime, Cefprozil, Cefquinome, Cefradine, Cefroxadine, Cefsulodin, Ceftaroline, Ceftazidime, Cefteram, Ceftezole, Ceftibuten, Ceftiofur, Ceftizoxime, Ceftobiprole, Ceftolozane, Ceftradine, Ceftrezole, Ceftriaxone, Ceftroxadine, Cefuroxime, Cefuzonam, Pivcefalexin; β-LACTAM CEPHAMYCINS, including Cefbuperazone, Cefmetazole, Cefminox, Cefotetan, Cefoxitin; β-LACTAM MONOBACTAMS, including Aztreonam, Carumonam, Tigemonam; β- LACTAM OXACEPHEMS, including Flomoxef, Latamoxef, Moxalactam; β-LACTAM PENICILLINS, including Amdinocillin (Mecillinam), Amoxicillin, Ampicillin, Apalcillin, Aspoxicillin, Azidocillin, Azlociliin, Bacampicillin, Carbenicillin, Carindacillin, Ciclacillin, Clemizole Penicillin, Clometocillin, Cloxacillin, Cyclacillin, Dicloxacillin, Epicillin, Fenbenicillin, Floxacillin (Flucloxacillin), Hetacillin, Lenampicillin, Mecillinam, Metampicillin, Methicillin Sodium, Mezlocillin, Nafcillin, Oxacillin, Penamecillin, Penethamate Hydriodide, Penicillin G, Penicillin G Benzathine, Penicillin G Procaine, Penicillin N, Penicillin O, Penicillin V, Phenethicillin Potassium, Piperacillin, Pivampicillin, Pivmecillinam, Propicillin, Quinacillin, Sulbenicillin, Sultamicillin, Talampicillin, Temocillin, Ticarcillin; BICYCLOMYCINS, including Bicozamycin; BORON CONTAINING ANTIBACTERIAL AGENTS, including AN3365 (aminomethylbenzoxaboroles), GSK2251052 (leucyl-tRNA synthetase inhibitors); CYCLIC ESTERS, including Fosfomycin; FATTY ACID SYNTHESIS INHIBITORS (Fabl), AFN-1252, MUT056399, FAB-001 FLUOROQUINOLONES, including Avarofloxacin, Balofloxacin, Besifloxacin, Chinfloxacin, Cinoxacin, Ciprofloxacin, Clinafloxacin, Danofloxacin, Delafloxacin, Difloxacin, Enoxacin, Enrofloxacin, Finafloxacin, Fleroxacin, Flumequine, Garenoxacin, Gatifloxacin, Gemifloxacin, Grepafloxacin, Ibafloxacin, Levofloxacin, Lomefioxacin, Marbofloxacin, Miloxacin, Moxifloxacin, Nadifloxacin, Norfloxacin, Ofloxacin, Orbifloxacin, Pazufloxacin, Pefloxacin, Pradofloxacin, Prulifloxacin, Rosoxacin, Rufloxacin, Sarafloxacin, Sitafloxacin, Sparfloxacin, Temafloxacin, Tosufloxacin, Trovafloxacin, Zabofloxacin; FUSIDANES, including Fusidic Acid; GLYCOLIPODEPSIPEPTIDE, including Ramoplanin; GLYCOPEPTIDES, including Avoparcin, Dalbavancin, Norvancomycin, Oritavancin, Teicoplanin, Telavancin, Vancomycin,; GLYCOPHOSPHOLIPIDS, including Bambermycins (bambermycin, moenomycins, flavophospholipol); GLYCYLCYCLINES, including Tigecycline; HYBRIDS, Cadazolid (Oxazolidinone-quinolone), TD-1792 (glycopeptide-cephalosporin); LINCOSAMIDES, including Clindamycin, Lincomycin, Pirlimycin; LIPOPEPTIDES, including Daptomycin, Surotomycin; MACROLIDES, including Azithromycin, Carbomycin, Cethromycin, Clarithromycin, Dirithromycin, Erythromycin, Fidaxomicin, Flurithromycin, Gamithromycin, Josamycin, Kitasamycin, Leucomycin, Meleumycin, Midecamycins, Miokamycin, Mirosamycin, Oleandomycin, Primycin, Rokitamycin, Rosaramicin, Roxithromycin, Sedecamycin, Solithromycin, Spiramycin, Telithromycin, Terdecamycin, Tildipirosin, Tilmicosin, Troleandomycin, Tulathromycin, Tylosin, Tylvalosin; NITROFURANS, including Furaltadone, Furazidin, Furazolidone, Furazolium Chloride, Nifuratel, Nifurfoline, Nifuroxazide, Nifurpirinol, Nifurtoinol, Nifurzide, Nitrofural, Nitrofurantoin, Nitrofurazone; NITRO I IMIDAZOLES, including Dimetridazole, Metronidazole, Ornidazole, Ronidazole, Secnidazole, Tinidazole; OLIGOSACCHARIDES, including Avilamycin, Everninomicin; OTHER ANTIBACTERIAL AGENTS, including Auriclosene, Chloroxine, Chlorquinaldol, Clioquinol, Clofoctol, Halquinol, Lotilibcin, Mandelic Acid, Methenamine (hexamine), Nitazole, Nitroxoline, Perchlozone, Taurolidine, Thenoic Acid, Xibornol; OXAZOLID1NONES, including Eperezolid, Linezolid, Posizolid, Radezolid, Sutezolid, Tedizolid (Torezolid); PEPTIDE DEFORMYLASE INHIBITORS, including GSK1322322; PEPTIDES, including Omiganan, Pexiganan; PLEUROMUTILINS, including Retapamulin, Tiamulin, Valnemulin; POLYETHER IONOPHORES, including Laidlomycin, Lasalocid, Maduramicin, Monensin, Narasin, Salinomycin, Semduramicin; POLYMYXI NS, including Colistin, Polymyxin B; POLYPEPTIDES, including Amphomycin, Bacitracin, Capreomycin, Enduracidin, Enramycin, Enviomycin, Fusafungine, Gramicidin(s), Iseganan, Magainins, Nosiheptide, Ristocetin, Thiostrepton, Tuberactinomycin, Tyrocidine, Tyrothricin, Viomycin; PSEUDOMONIC ACIDS, including Mupirocin; QUINOLONES, including Nalidixic Acid, Nemonoxacin, Oxolinic Acid, Ozenoxacin, Pipemidic Acid, Piromidic Acid; QUI NOXALI NES, including Carbadox, Olaquindox; RIMINOFENAZINES, including Clofazimine; STATI NS, including Atorvastatin, Fluvastatin, Lovastatin, Mevastatin, Pitavastatin, Pravastatin, Rosuvastatin, Simvastatin; STREPTOGRAMINS, including Dalfopristin, Flopristin, Linopnstin, Pristinamycin, Quinupristin, Virginiamycin; STREPTOTHRICINS, including Nourseothricin; SULFONAMIDES, including Acetyl Sulfamethoxypyrazine, Chloramine-B, Chloramine-T, Dichloramine T, Formosulfathiazole, Mafenide, N4-Sulfanilylsulfanilamide, Noprylsulfamide, N- Sulfanilyl-3,4-xylamide, Ormaosulfathiazole, Phthalylsulfacetamide, Phthalylsulfathiazole, Salazosulfadimidine, Succinylsulfathiazole, Sulfabenzamide, Sulfacarbamide, Sulfacetamide, Sulfachlorpyridazine, Sulfachrysoidine, Sulfaclozine, Sulfacytine, Sulfadiazine, Sulfadicramide, Sulfadimethoxine, Sulfadimidine, Sulfadoxine, Sulfaethidole, Sulfaguanidine, Sulfaguanole, Sulfalene, Sulfaloxic Acid, Sulfamerazine, Sulfameter, Sulfamethazine, Sulfamethizole, Sulfamethomidine, Sulfamethoxazole, Sulfamethoxypyridazine, Sulfamethylthiazole, Sulfametopyrazine, Sulfametrole, Sulfamidochrysoidine, Sulfamonomethoxine, Sulfamoxole, Sulfanilamide, Sulfanilylurea, Sulfaperine, Sulfaphenazole, Sulfaproxyline, Sulfapyrazine, Sulfapyridine, Sulfaquinoxaline, Sulfathiazole, Sulfathiourea, Sulfatroxazole, Sulfisomidine, Sulfisoxazole (Sulfafurazole); SULFONES, including Acediasulfone, Dapsone, Glucosulfone Sodium, p-Sulfanilylbenzylamine, Succisulfone, Sulfanilic Acid, Sulfoxone Sodium, Thiazolsulfone; TETRACYCLI NES, including Chlortetracycline, Clomocycline, Demeclocycline, Doxycycline, Eravacycline, Guamecycline, Lymecycline, Meclocycline, Methacycline, Minocycline, Oxytetracycline, Penimepicycline, Pipacycline, Rolitetracycline, Sarecycline, and Tetracycline. [00167] The composition of the invention may further comprise an excipient selected from the group comprising, but not limited to, binders and compression aids, coatings and films, colouring agents diluents and vehicles disintegrants, emulsifying and solubilising agents, flavours and sweeteners, repellents, glidants and lubricants, plasticisers, preservatives, propellants, solvents, stabilisers, suspending agents and viscosity enhancers.

[00168] According to a further aspect of the invention, there is provided a medical device when used in a method of treating or preventing a bacterial infection in the subject.

[00169] According to further aspect of the invention, there is provided a medical device comprising the composition of the invention. The composition of the invention may be any slow release form, and/or in the form of a coating of the medical device.

[00170] The medical device may be in a form selected from the group comprising: an implant, a plaster, a bandage, and other dressing applied to a bacterial infection in a subject.

[00171] According to further aspect of the invention, there is provided a method of killing bacteria, the method including the step of contacting the bacteria with a compound of the invention, or a therapeutically acceptable salt thereof.

[00172] According to further aspect of the invention, there is provided the use of a compound of the invention, or a therapeutically acceptable salt thereof, to kill bacteria, said use comprising the step of contacting the bacteria with a compound of the invention, or a therapeutically acceptable salt thereof.

[00173] Terms used herein will have their customary meanings in the art unless specified. As used herein, the term robenidine, NCL812 (also known as 1 ,3-bis[(E)-{4- chlorophenyl)methyleneamino]guanidine) refers to a compound having the following chemical structure:

BRIEF DESCRIPTION OF THE DRAWINGS

[00174] Further features of the present invention are more fully described in the following description of several non-limiting embodiments thereof. This description is included solely for the purposes of exemplifying the present invention. It should not be understood as a restriction on the broad summary, disclosure or description of the invention as set out above. The description will be made with reference to the accompanying drawings in which: Figure 1 presents the chemical name and chemical structure of the compounds NCL001 to NCL230;

Figure 2 shows a graph of the effect of NCL812 on DNA macromolecular synthesis in

Staphylococcus aureus according to example 2;

Figure 3 shows a graph of the effect of NCL812 on RNA macromolecular synthesis in

Staphylococcus aureus according to example 2;

Figure 4 shows a graph of the effect of NCL812 on protein macromolecular synthesis in

Staphylococcus aureus (ATCC29213) according to example 2;

Figure 5 shows a graph of the effect of NCL812 on cell wall macromolecular synthesis in

Staphylococcus aureus (ATCC29213) according to example 2;

Figure 6 shows a graph of the effect of NCL812 on lipid macromolecular synthesis in

Staphylococcus aureus (ATCC29213) according to example 2;

Figure 7 shows a graph summarising the effect of NCL812 on macromolecular synthesis in Staphylococcus aureus (ATCC29213) according to example 2;

Figure 8 shows a g raph of the effect of NCL812 on ATP release from Staphylococcus aureus (ATCC29213) according to example 3;

Figure 9 shows a graph showing the average melting point peaks for the negative derivative dF/dT after real-time polymerase chain reaction of the mecA gene in methicillin-resistant S. aureus isolates grouped by mec gene complexes, A (n=4), B (n=10), C2 (n=4) and unclassified (n=2). Groups indicated with different superscripts are significantly different (P<0.05), according to example 4;

Figure 10 shows a graph of the optical densities of the unsupplemented growth control, ampicillin and different concentrations of antibacterial agent NCL812 against methicillin-susceptible S. aureus ATCC 49775 using broth microdilution methodology according to example 4. The concentrations of NCL812 tested were at the MIC and four times the MIC determined under test conditions, up to 24 h incubation. Ampicillin was tested at the MIC. Bactericidal activity was tested at 0, 1 , 2, 4, 8, 12, and 24 h for antibacterials;

Figure 1 1 shows a graph of kill kinetic curves for methicillin-susceptible S. aureus ATCC

49775 demonstrating bactericidal activity of NCL812 using the Clinical and Laboratory Standards Institute macrodilution methodology in a 10 ml vial according to example 4. The concentrations of antibacterials tested were at 1 χ and x the MIC determined under test conditions. Bactericidal activity was determined at 0, 1 , 2, 4, 8, 12 and 24 h after antibacterial addition. Bactericidal activity was defined as a 3log i0 (99.9%) decrease in the number viable bacteria from the initial inoculum size;

Figure 12 shows a graph indicating the change of pH during macro-broth dilution assay for S. pneumoniae strain D39 exposed to 4 Mg/mL in NCL812 and 0.0023 μg mL ampicillin according to example 5;

Figure 13 shows a graph illustrating the 48-hour time-kill of S. pneumoniae strain D39 treated with NCL812 according to example 5;

Figure 14 shows a graph illustrating the 48-hour time-kill of S. pneumoniae strain D39 treated with NCL062 according to example 5;

Figure 15 shows a graph illustrating in the 14-hour time-kill of S. pneumoniae strain D39 treated with NCL812 according to example 5;

Figure 16 shows a graph illustrating in the 14-hour time-kill of S. pneumoniae strain D39 treated with NCL062 according to example 5;

Figure 17 shows a graph illustrating the 14-hour time-kiil of S. pneumoniae strain D39 treated with ampicillin according to example 5;

Figure 18 shows a graph illustrating the 12-hour time-kill of S. pneumoniae strain D39 treated with NCL812, adopted from the Figure 43, according to example 5;

Figure 19 shows a graph illustrating the 12-hour time-kill of S. pneumoniae strain D39 treated with NCL062, adopted from the Figure 44, according to example 5;

Figure 20 shows a graph illustrating the 48-hour time-kill of S. pneumoniae strain D39 treated with ampicillin according to example 5;

Figure 21 shows a graph illustrating the 48-hour time-kill of S. pneumoniae strain D39 treated with erythromycin according to example 5;

Figure 22 shows a graph illustrating the 48-hour time-kill of S. pneumoniae strain D39 treated with NCL812 and 5% choline chloride;

Figure 23 shows a graph illustrating the 12-hour time-kill of S. pneumoniae strain D39 treated with NCL812 and 5% choline chloride according to example 5; Figure 24 shows a graph illustrating the 48-hour time-kill of S. pneumoniae strain D39 treated with NCL062 and 5% choline chloride according to example 5;

Figure 25 shows a graph illustrating the 12-hour time-kill of S. pneumoniae strain D39 treated with NCL062 and 5% choline chloride according to example 5;

Figure 26 shows a graph illustrating the 48-hour time-kill of S. pneumoniae strain D39 treated with ampicillin and choline chloride according to example 5;

Figure 27 shows a graph of the relative MBC of D39 treated with NCL812 or NCL062 for

48 hours according to example 5;

Figure 28 shows a graph illustrating the relative minimal bactericidal concentration

(MBC) of S. pneumoniae strain D39 treated with ampicillin over a 48 h time period according to example 5;

Figure 29 shows a graph illustrating the relative MBC for S. pneumoniae strain D39 treated with erythromycin over a 48 h time period according to example 5;

Figure 30 shows a graph illustrating the viable count (log 10 CFU/ml) of S. pneumoniae strain D39 treated with NCL812 from a macro-broth dilution of time-kill over 24 hours according to example 5;

Figure 31 shows a graph illustrating the viable count (log 10 CFU/ml) of S. pneumoniae strain D39 treated with ampicillin from a macro-broth dilution of time-kill over 24 hours according to example 5;

Figure 32 is a bar graph illustrating the mean cell membrane thickness of treated and untreated D39 according to example 5;

Figure 33 is a bar graph illustrating the mean width of periplasmic space of treated (16 μg/mL NCL812) and untreated D39 samples according to example 5;

Figure 34 shows the kill kinetics of MRSA 580 isolate obtained at different concentrations of NCL812 over a period of 8 hours according to example 7;

Figure 35 shows the kill kinetics of MRSA 580 in different concentrations of NCL812 over a period of 24 h according to example 7;

Figure 36 shows the kill kinetics of MRSA 698 in different concentrations of NCL812 over a period of 24 h according to example 7; Figure 37 shows the kill kinetics of VRE 26c(dc) at different concentrations of NCL812 over a period of 24 h according to example 7;

Figure 38 shows the kill kinetics of VRE 16c(dc) at different concentrations of NCL812 over a period of 24 h according to example 8;

Figure 39 shows the kill kinetics assay of Staphylococcus aureus KC01 at different concentrations of NCL812, up to 24 h incubation according to example 8;

Figure 40 shows the kill kinetics assay of Enterococcus faecalis USA01 at different concentrations of NCL812, up to 24 h incubation according to example 8; and

Figure 41 is a graph illustrating the cumulative release of NCL812 and NCL099 from

Formulation B according to example 10.

DESCRIPTION OF EMBODIMENTS

General

[00175] Before describing the present invention in detail, it is to be understood that the invention is not limited to particular exemplified methods or compositions disclosed herein. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments of the invention only, and is not intended to be limiting.

[00176] All publications referred to herein, including patents or patent applications, are incorporated by reference in their entirety. However, applications that are mentioned herein are referred to simply for the purpose of describing and disclosing the procedures, protocols, and reagents referred to in the publication which may have been used in connection with the invention. The citation of any publications referred to herein is not to be construed as an admission that the invention is not entitled to antedate such disclosure by virtue of prior invention.

[00177] In addition, the carrying out of the present invention makes use of, unless otherwise indicated, conventional microbiological techniques within the skill of the art. Such conventional techniques are known to the skilled worker.

[00178] As used herein, and in the appended claims, the singular forms "a", "an", and "the" include the plural unless the context clearly indicates otherwise.

[00179] Unless otherwise indicated, all technical and scientific terms used herein have the same meanings as commonly understood by one of ordinary skill in the art to which this invention belongs. Although any materials and methods similar to, or equivalent to, those described herein may be used to carry out the present invention, the preferred materials and methods are herein described.

[00180] The invention described herein may include one or more ranges of values {e.g. size, concentration, dose etc). A range of values will be understood to include all values within the range, including the values defining the range, and values adjacent to the range that lead to the same or substantially the same outcome as the values immediately adjacent to that value which define the boundary of the range.

[00181] The pharmaceutical or veterinary compositions of the invention may be administered in a variety of unit dosages depending on the method of administration, target site, physiological state of the patient, and other medicaments administered. For example, unit dosage form suitable for oral administration include solid dosage forms such as powder, tablets, pills, and capsules, and liquid dosage forms, such as elixirs, syrups, solutions and suspensions. The active ingredients may also be administered parenterally in sterile liquid dosage forms. Gelatin capsules may contain the active ingredient and inactive ingredients such as powder carriers, glucose, lactose, sucrose, mannitol, starch, cellulose or cellulose derivatives, magnesium stearate, stearic acid, sodium saccharin, talcum, magnesium carbonate, and the like.

[00182] The phrase "therapeutically effective amount" as used herein refers to an amount sufficient to inhibit bacterial growth associated with a bacterial infection or colonisation. That is, reference to the administration of the therapeutically effective amount of a compound of Formula I according to the methods or compositions of the invention refers to a therapeutic effect in which substantial bacteriocidal or bacteriostatic activity causes a substantial inhibition of bacterial infection. The term "therapeutically effective amount" as used herein, refers to a sufficient amount of the composition to provide the desired biological, therapeutic, and/or prophylactic result. The desired results include elimination of bacterial infection or colonisation or reduction and/or alleviation of the signs, symptoms, or causes of a disease, or any other desired alteration of a biological system. An effective amount in any individual case may be determined by one of ordinary skill in the art using routine experimentation. In relation to a pharmaceutical or veterinary composition, effective amounts can be dosages that are recommended in the modulation of a diseased state or signs or symptoms thereof. Effective amounts differ depending on the composition used and the route of administration employed. Effective amounts are routinely optimized taking into consideration pharmacokinetic and pharmacodynamic characteristics as well as various factors of a particular patient, such as age, weight, gender, etc and the area affected by disease or disease causing microbes.

[00183] As referred to herein, the terms "treatment" or "treating" refers to the full or partial removal of the symptoms and signs of the condition. For example, in the treatment of a bacterial infection or colonisation, the treatment completely or partially removes the signs of the infection. Preferably in the treatment of infection, the treatment reduces or eliminates the infecting bacterial pathogen leading to microbiai cure.

[00184] As referred to herein, the term "bacteria" refers to members of a large domain of prokaryotic microorganisms. Typically a few micrometres in length, bacteria have a number of shapes, ranging from spheres to rods and spirals and can be present as individual cells or present in linear chains or clusters of variable numbers and shape. Preferably the terms "bacteria" and its adjective "bacterial" refer to bacteria such as the Gram positive Staphylococcus spp, Streptocccus spp, Bacillus spp, Enferococcus spp, Listeria spp, and anaerobic bacteria; Gram negative Escherichia coli, Enterobacter spp, Klebsiella spp and Pseudomonas spp; and the cell wall free bacteria such as Mycoplasma spp and Ureaplasma spp. The terms may refer to an antibiotic-sensitive strain or an antibiotic-resistant strain. In a preferred embodiment, the terms refer to MRSA or MRSP. In another preferred embodiment, the terms refer to MDR Staphylococcus spp, Streptococcus spp, Enterococcus spp, Clostridium difficile, Escherichia coli, Enterobacter spp, Klebsiella spp and Pseudomonas spp.

[00185] Referred to herein, the term "methicillin-resistant bacteria" (such as methicillin- resistant Staphylococcus) refers a bacteria isolate that demonstrates resistance at any dose to all β-lactams including penicillins, carbapenems and first to fourth generation cephalosporins, but not to the fifth generation anti-MRSA cephalosporins (for example ceftaroline). Multidrug- resistant (MDR) is defined as acquired non-susceptibility to at least one agent in three or more antimicrobial categories, extensively drug-resistant (XDR) is defined as non-susceptibility to at least one agent in all but two or fewer antimicrobial categories (i.e. bacterial isolates remain susceptible to only one or two categories) and pandrug-resistant (PDR) is defined as non- susceptibility to all agents in all antimicrobial categories currently available.

[00186] An example of susceptible, MDR, XDR and PDR bacteria includes the following. Wiid type, antibacterial unexposed isolates of Staphylococcus aureus that are likely to be susceptible to all of the following antibacterial categories (and agents); aminoglycosides (for example gentamicin); ansamycins (for example rifampicin); anti-MRSA cephalosporins (for example ceftaroline); anti-staphylococcal β-lactams (or cephamycins) (for example oxacillin or cefoxitin); carbapenems (for example ertapenem, imipenem, meropenem or doripenem); non- extended spectrum cephalosporins; 1 st and 2nd generation cephalosporins (for example cefazolin or cefuroxime); extended-spectrum cephalosporins; 3rd and 4th generation cephalosporins (for example cefotaxime or ceftriaxone); cephamycins (for example cefoxitin or cefotetan); fluoroquinolones (for example ciprofloxacin or moxifloxacin); folate pathway inhibitors (for example trimethoprim-sulphamethoxazole); fucidanes (for example fusidic acid); glycopeptides (for example vancomycin, teicoplanin or telavancin); glycylcyclines (for example tigecycline); lincosamides (for example clindamycin); lipopeptides (for example daptomycin); macrolides (for example erythromycin); oxazolidinones (for example linezolid or tedizolid); phenicols (for example chloramphenicol); phosphonic acids (for example fosfomycin); streptogramins (for example quinupristin-dalfopristin; and tetracyclines (for example tetracycline, doxycycline or minocycline). Isolates that are non-susceptible to more than one agent in more than three antimicrobial categories are classified as MDR (all MRSA, for example, meet the definition of MDR). Isolates that are non-susceptible to more than one agent in all but one or two antimicrobial categories are classified as XDR. Isolates that are non-susceptible to all listed antibacterial agents are PDR.

100187] Pharmaceutically and veterinary acceptable salts include salts which retain the biological effectiveness and properties of the compounds of the present disclosure and which are not biologically or otherwise undesirable. In many cases, the compounds disclosed herein are capable of forming acid and/or base salts by virtue of the presence of amino and/or carboxyl groups or groups similar thereto. Acceptable base addition salts can be prepared from inorganic and organic bases. Salts derived from inorganic bases, include by way of example only, sodium, potassium, lithium, ammonium, calcium and magnesium salts. Salts derived from organic bases include, but are not limited to, salts of primary, secondary and tertiary amines, such as by way of example only, alkyl amines, dialkyl amines, trialkyl amines, substituted alkyl amines, di(subsrituted alkyl) amines, tri(substituted alkyl) amines, alkenyl amines, dialkenyl amines, trialkenyl amines, substituted alkenyl amines, di(substituted alkenyl) amines, tri (substituted alkenyl) amines, cycloalkyl amines, di(cycloalkyl) amines, tri(cycloalkyl) amines, substituted cycloalkyl amines, disubstituted cycloalkyl amines, trisubstituted cycloalkyl amines, cycloalkenyl amines, di(cycloalkenyl) amines, tri(cycloalkenyl) amines, substituted cycloalkenyl amines, disubstituted cycloalkenyl amines, trisubstituted cycloalkenyl amines, aryl amines, diary' amines, triaryl amines, heteroaryl amines, diheteroaryl amines, triheteroaryl amines, heterocyclic amines, diheterocyclic amines, triheterocyclic amines, mixed di- and tri-amines where at least two of the substituents on the amine are different and are selected from the group consisting of alkyl, substituted alkyl, alkenyl, substituted alkenyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic, and the like. Also included are amines where the two or three substituents, together with the amino nitrogen, form a heterocyclic or heteroaryl group.

[00188] Pharmaceutically and veterinary acceptable acid addition salts may be prepared from inorganic and organic acids. The inorganic acids that can be used include, by way of example only, hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like. The organic acids that can be used include, by way of example only, acetic acid, propionic acid, glycolic acid, pyruvic acid, oxalic acid, malic acid, malonic acid, succinic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, p-toluenesulfonic acid, salicylic acid, and the like.

[00189] The pharmaceutically or veterinary acceptable salts of the compounds useful in the present disclosure can be synthesized from the parent compound, which contains a basic or acidic moiety, by conventional chemical methods. Generally, such salts can be prepared by reacting the free acid or base forms of these compounds with a stoichiometric amount of the appropriate base or acid in water or in an organic solvent, or in a mixture of the two; generally, nonaqueous media like ether, ethyl acetate, ethanol, isopropanol, or acetonitrile are preferred. Lists of suitable salts are found in Remington's Pharmaceutical Sciences. 17th ed., Mack Publishing Company, Easton, Pa. (1985), p. 1418, the disclosure of which is hereby incorporated by reference. Examples of such acceptable salts are the iodide, acetate, phenyl acetate, trifluoroacetate, acryl ate, ascorbate, benzoate, chlorobenzoate, dinitrobenzoate, hydroxybenzoate, methoxybenzoate, methyl benzoate, o-acetoxybenzoate, naphthalene-2- benzoate, bromide, isobutyrate, phenylbutyrate, γ-hydroxybutyrate, β-hydroxybutyrate, butyne- l,4-dioate, hexyne-l,4-dioate, hexyne- 1 ,6-dioate, caproate, caprylate, chloride, cinnamate, citrate, decanoate, formate, fumarate, glycollate, heptanoate, hippurate, lactate, malate, maleate, hydroxymaleate, malonate, mandelate, mesylate, nicotinate, isonicotinate, nitrate, oxalate, phthalate, terephthalate, phosphate, monohydrogenphosphate, dihydrogenphosphate, metaphosphate, pyrophosphate, propiolate, propionate, phenylpropionate, salicylate, sebacate, succinate, suberate, sulfate, bisulfate, pyrosulfate, sulfite, bisulfite, sulfonate, benzenesulfonate, p-bromophenylsulfonate, chlorobenzenesulfonate, propanesu!fonate, ethanesulfonate, 2- hydroxyethanesulfonate, merhanesulfonate, naphthalene-l-sulfonate, naphthalene-2-sulfonate, p-toluenesulfonate, xylenesulfonate, tartarate, and the like.

[00190] The pharmaceutical or veterinary compositions of the invention may be formulated in conventional manner, together with other pharmaceutically acceptable excipients if desired, into forms suitable for oral, parenteral, or topical administration. The modes of administration may include parenteral, for example, intramuscular, subcutaneous and intravenous administration, oral administration, topical administration and direct administration to sites of infection such as intraocular, intraaural, intrauterine, intranasal, intramammary, intraperitoneal, intralesional, etc.

[00191] The pharmaceutical or veterinary compositions of the invention may be formulated for oral administration. Traditional inactive ingredients may be added to provide desirable colour, taste, stability, buffering capacity, dispersion, or other known desirable features. Examples include red iron oxide, silica gel, sodium laurel sulphate, titanium dioxide, edible white ink, and the like. Conventional diluents may be used to make compressed tablets. Both tablets and capsules may be manufactured as sustained-release compositions for the continual release of medication over a period of time. Compressed tablets may be in the form of sugar coated or film coated tablets, or enteric-coated tablets for selective disintegration in the gastrointestinal tract. Liquid dosage forms for oral administration may contain colouring and/or flavouring to increase patient compliance. As an example, the oral formulation comprising compounds of the invention may be a tablet comprising any one, or a combination of, the following excipients: calcium hydrogen phosphate dehydrate, microcrystalline cellulose, lactose, hydroxypropyl methyl cellulose, and talc.

[00192] The compositions described herein may be in the form of a liquid formulation. Examples of preferred liquid compositions include solutions, emulsions, injection solutions, solutions contained in capsules. The liquid formulation may comprise a solution that includes a therapeutic agent dissolved in a solvent. Generally, any solvent that has the desired effect may be used in which the therapeutic agent dissolves and which can be administered to a subject. Generally, any concentration of therapeutic agent that has the desired effect can be used. The formulation in some variations is a solution which is unsaturated, a saturated or a supersaturated solution . The solvent may be a pure solvent or may be a mixture of liquid solvent components. In some variations the solution formed is an in situ gelling formulation. Solvents and types of solutions that may be used are well known to those versed in such drug delivery technologies. *

[00193] The composition described herein may be in the form of a liquid suspension. The liquid suspensions may be prepared according to standard procedures known in the art. Examples of liquid suspensions include micro-emulsions, the formation of complexing compounds, and stabilising suspensions. The liquid suspension may be in undiluted or concentrated form. Liquid suspensions for oral use may contain suitable preservatives, antioxidants, and other excipients known in the art functioning as one or more of dispersion agents, suspending agents, thickening agents, emulsifying agents, wetting agents, solubilising agents, stabilising agents, flavouring and sweetening agents, colouring agents, and the like. The liquid suspension may contain glycerol and water.

[00194] The composition described herein may be in the form of an oral paste. The oral paste may be prepared according to standard procedures known in the art.

[00195] The composition is described herein may be in the form of a liquid formulation for injection, such as intra-muscular injection, and prepared using methods known in the art. For example, the liquid formulation may contain polyvinylpyrrolidone K30 and water.

[00196] The composition is described herein may be in the form of topical preparations. The topical preparation may be in the form of a lotion or a cream, prepared using methods known in the art. For example, a lotion may be formulated with an aqueous or oily base and may include one or more excipients known in the art, functioning as viscosity enhancers, emulsifying agents, fragrances or perfumes, preservative agents, chelating agents, pH modifiers, antioxidants, and the like. For example, the topical formulation comprising one or more compounds of the invention may be a gel comprising anyone, or a combination of, the following excipients: PEG 8000, PEG 4000, PEG 200, glycerol, propylene glycol. The NCL812 compound may further be formulated into a solid dispersion using SoluPlus {BASF, www.soluplus.com) and formulated with anyone, or a combination of, the following excipients: PEG 8000, PEG 4000, PEG 200, glycerol, and propylene glycol.

[00197] For aerosol administration, the composition of the invention is provided in a finely divided form together with a non-toxic surfactant and a propellant. The surfactant is preferably soluble in the propellant. Such surfactants may include esters or partial esters of fatty acids.

[00198] The compositions of the invention may alternatively be formulated for delivery by injection. As an example, the compound is delivered by injection by any one of the following routes: intravenous, intramuscular, intradermal, intraperitoneal, and subcutaneous.

[00199] The compositions of the invention may alternatively be formulated using nanotechnology drug delivery techniques such as those known in the art. Nanotechnology- based drug delivery systems have the advantage of improving bioavailability, patient compliance and reducing side effects.

[00200] The formulation of the composition of the invention includes the preparation of nanoparticles in the form of nanosuspensions or nanoemulsions, based on compound solubility. Nanosuspensions are dispersions of nanosized drug particles prepared by bottom-up or top- down technology and stabilised with suitable excipients. This approach may be applied to the compounds of the invention which can have poor aqueous and lipid solubility, in order to enhance saturation solubility and improve dissolution characteristics. An example of this technique is set out in Sharma and Garg (2010) (Pure drug and polymer-based nanotechnologies for the improved solubility, stability, bioavailability, and targeting of anti-HIV drugs. Advanced Drug Delivery Reviews, 62: p. 491-502). Saturation solubility will be understood to be a compound-specific constant that depends on temperature, properties of the dissolution medium, and particle size (<1 -2 μιτι).

[00201] The composition of the invention may be provided in the form of a nansuspension. For nanosuspensions, the increase in the surface area may lead to an increase in saturation solubility. Nanosuspensions are colloidal drug delivery systems, consisting of particles below 1 μιη. Compositions of the invention may be in the form of nanosuspensions including nanocrystalline suspensions, solid lipid nanoparticles (SLNs), polymeric nanoparticles, nanocapsules, polymeric micelles and dendrimers. Nanosuspensions may be prepared using a top-down approach where larger particles may be reduced to nanometre dimensions by a variety of techniques known in the art including wet-milling and high-pressure homogenisation. Alternatively, nanosuspensions may be prepared using a bottom-up technique where controlled precipitation of particles may be carried out from solution.

100202] The composition of the invention may be provided in the form of a nanoemulsion. Nanoemulsions are typically clear oil-in-water or water-in-oil biphasic systems, with a droplet size in the range of 100-500 nm, and with compounds of interest present in the hydrophobic phase. The preparation of nanoemulsions may improve the solubility of the compounds of the invention described herein, leading to better bioavailability. Nanosized suspensions may include agents for electrostatic or steric stabilisation such as polymers and surfactants. Compositions in the form of SLNs may comprise biodegradable lipids such as triglycerides, steroids, waxes and emulsifiers such as soybean lecithin, egg lecithin, and poloxamers. The preparation of a SLN preparation may involve dissolving/dispersing drug in melted lipid followed by hot or cold homogenisation. If hot homogenisation is used, the melted lipidic phase may be dispersed in an aqueous phase and an emulsion prepared. This may be solidified by cooling to achieve SLNs. If cold homogenisation is used, the lipidic phase may be solidified in liquid nitrogen and ground to micron size. The resulting powder may be subjected to high-pressure homogenisation in an aqueous surfactant solution.

I00203] The Compounds of Formula I as described herein may be dissolved in oils/liquid lipids and stabilised into an emulsion formulation. Nanoemulsions may be prepared using highland low-energy droplet reduction techniques. High-energy methods may include high-pressure homogenisation, ultrasonication and microfluidisation. If the low-energy method is used, solvent diffusion and phase inversion will generate a spontaneous nanoemulsion. Lipids used in nanoemulsions may be selected from the group comprising triglycerides, soybean oil, safflower oil, and sesame oil. Other components such as emulsifiers, antioxidants, pH modifiers and preservatives may also be added.

[00204] The composition may be in the form of a controlled-release formulation and may include a degradable or non-degradable polymer, hydrogel, organogel, or other physical construct that modifies the release of the compound. It is understood that such formulations may include additional inactive ingredients that are added to provide desirable colour, stability, buffering capacity, dispersion, or other known desirable features. Such formulations may further include liposomes, such as emulsions, foams, micelles, insoluble monolayers, liquid crystals, phospholipid dispersions, lamellar layers and the like. Liposomes for use in the invention may be formed from standard vesicle-forming lipids, generally including neutral and negatively charged phospholipids and a sterol, such as cholesterol. 100205] The formulations of the invention may have the advantage of increased solubility and/or stability of the compounds, particularly for those formulations prepared using nanotechnology techniques. Such increased stability and/or stability of the compounds of Formula I may improve bioavailability and enhance drug exposure for oral and/or parenteral dosage forms.

Throughout this specification, unless the context requires otherwise, the word "comprise" or variations such as "comprises" or "comprising", will be understood to imply the inclusion of a stated integer or group of integers but not the exclusion of any other integer or group of integers.

EXAMPLES

EXAMPLE 1 : The minimum inhibitory concentrations (MIC) for NCL812 in methicillin- resistant Staphylococcus aureus (MRSA), vancomycin-resistant Enferococcus spp. (VRE) and Sfrepfococcus pneumoniae.

Specific

[00206] As is apparent from the preceding summary of the invention, the invention relates to compounds of Formula I, methods of treatment of a bacterial infection, uses and medical devices.

[00207] This study was undertaken to determine minimum inhibitory concentrations (MIC) for antibacterial agent NCL812. The antibacterial agent represents a potentially new class of drug with a perceived narrow spectrum of activity against bacteria and a novel mechanism of action. This study focused on recent isolates of three major opportunistic pathogens of humans where the development of antibacterial resistance to existing antibacterial classes is problematic: methicillin-resistant Staphylococcus aureus (MRSA), vancomycin-resistant Enterococcus spp. (VRE) and Sfrepfococcus pneumoniae.

[00208] In this example, NCL812 minimum inhibitory concentrations (MICs) were determined for 61 Australian clinical isolates (comprised of 21 MRSA, 20 putative VRE and 20 S. pneumoniae isolates). The MIC profiles for NCL812 were found to be remarkably consistent, with MIC 50 and MIC 90 values of 4 μg/mL recorded for each of the species tested.

Materials and Methods

Bacterial Isolate Collection and Identification

[00209] Sixty one test isolates were sourced from clinical diagnostic microbiology laboratories. The MRSA isolates were originally cultured on selective Brilliance MRSA Chromogenic Agar (Oxoid). Suspect colonies were selected on the basis of their colony appearance on this agar and identification as Staphylococcus aureus was determined using colony characteristics on non-selective Sheep Blood Agar (SBA) and phenotypic characteristics such as Gram stain, positive catalase test, positive coagulase test (tube coagulase test using rabbit plasma) and clumping factor (agglutination with the Oxoid Staphytect latex test), positive Voges Proskauer test, and the ability to produce acid from trehalose. A positive cefoxitin resistance screen confirmed the isolates as MRSA. All Enterococcus isolates underwent a standard biochemical identification. Biochemical profiling provisionally identified four of the VRE isolates as Enterococcus faecalis and the remainder as Enterococcus faecium. All S. pneumoniae isolates were identified on the basis of standard biochemical profiling.

Preparation of Antibacterials

[00210] Analytical grade NCL812 (batch 20081214) with a defined potency of 1000 mg/g

(ie 100%) was obtained. The powder was stored at a temperature of -20 °C. Aliquots (1 mL) of stock solution (25.6 mg/mL) were prepared in DMSO and stored at -80 °C and defrosted immediately before use.

Preparation of 96 well microtitre plates for broth microdilution MIC test using NCL812

[0021 1] Cation adjusted Mueller Hinton Broth (CAMHB) was prepared using 100 mL sterile Mueller Hinton broth (pH adjusted). To each 100 mL volume, 125 μί of calcium stock solution (10 mg Ca 2+ per ml) and 43 μί magnesium stock solution (10 mg Mg 2+ per mL) was aseptically added. Sufficient broth is made up for daily use, with unused portions being stored at 4 °C overnight.

[00212] Microdilution trays with 4% lysed horse blood in CAMHB was prepared by lysing horse blood (Oxoid) by repeated freezing and thawing (3-4 times) and aseptically mixing the lysed horse blood (LHB) 50:50 with sterile distilled water. A cell free suspension was obtained by centrifuging 50% LHB at 16,000*g (7000 rpm) for 20 min. The supernatant was decanted, recentrifuged and stored frozen. 50% LHB was diluted with CAMHB to obtain a final concentration of 4% (7 mL LHB into 93 mL CAMHB). 4% LHB-CAMHB was used instead of CAMHB in all steps in the preparation of the microdilution trays and preparation of antimicrobial solutions for Streptococcus species.

[00213] A stock antibiotic working solution of NCL812 was prepared to a concentration of 25.60 mg/mL. Potency was described as 1000 mg/g or 100%. The powder was dissolved in 10 mL DMSO and 1 mL volumes were aliquoted into eppendorf tubes and stored at -80 °C. When added to CAMHB, a fine cloudy precipitate formed, and was shaken well before and during aliquoting. [00214] A stock solution of ampicillin was prepared to the concentration of 25.60 mg/mL.

Ampicillin was used for internal quality control. The powdered ampicillin was dissolved in 4 mL of phosphate buffer pH 8.0, 0.1 mol/L, then diluted in 6 mL phosphate buffer pH 6.0, 0.1 mol/L. 1 mL volumes were aliquoted into eppendorf tubes and stored at -80 °C.

[00215] For Staphylococcus aureus, a working solution of 256 μg/mL was prepared by diluting stock solutions as described above 1 :100 in CAMHB {100 μΐ into 9.9 mL). When 90 μί was added to each well 12, there was a 1 :2 dilution so that well 12 had 128 μg mL of antibiotic. The range of antimicrobial was calculated as 0.25 μg/mL (well 3) to 128 μg/mL (well 12).

[00216] For Enterococcus species, a working solution of 64 μg/mL was prepared by diluting stock solutions as described above 1 :400 in CAMHB (100 ί into 9.9 mL, then further dilute this 1 :4). When 90 μL was added to well 12 there was a 1 :2 dilution so that well 12 had 32 μg/mL of antibiotic.

[00217] For Streptococcus pneumoniae, a working solution of 64 μg mL was prepared by diluting stock solutions as described above 1 :400 in 4% LHB-CAMHB (100 μί into 9.9 mL, then further diluting this 1 :4). When 90 il Mars added to well 12 there was a 1 :2 dilution so that well 12 had 32 μg/mL of antibiotic.

[00218] Serial dilutions were prepared in 96 well plates were set up in a safety cabinet according to methods standard in the art. Brifely: 90 [iL of the working antibiotic solution was added to each well in Column 12 of the plate, and mixed well, before 90 [iL was transferred to column 1 1. The solutions were mixed again, and then transferred to the next column as before, continuing the dilutions through to column 3. Mixing the well requires the pick up and expulsion of 90 [iL in each well 3-4 times before picking up and transferring the 90 μί to the next well. Column 2 (bacterial positive control) and column 1 (negative control) did not form part of the serial dilution. The trays were set up as follows: 2 strains were tested in duplicate in one tray, such that strain 1 was located in rows A to D, strain 2 was located in rows E to H, etc. The MIC ^g/mL) Interpretive Standard for Ampicillin using Control strains is shown in Table 1 below. Staphylococcus aureus ATCC 29213 Acceptable MIC range for Ampicillin = 0.5 to 2 μg mL ; Enterococcus faecalis ATCC 29212 Acceptable MIC range for Ampicillin = 0.5 to 2 g/mL, Strepto∞ccus pneumoniae ATCC 49619 Acceptable MIC range for Ampicillin = 0.06 to 0.25 μg/mL.

Table 1 : MIC ^g/mL) Interpretive Standard for Ampicillin using Control strains according to Example 1 . Sensitive Intermediate Resistant

Staphylococcus aureus < 0.25 > 0.5

Enterococcus species < 8 > 16

Streptococcus pneumoniae < 2 4 > 8

Preparation of bacterial suspension for broth microdilution MIC method

[00219] Fresh cultures of bacteria were prepared for testing on sheep blood agar (SBA), and overnight incubation at 37 °C as follows; 2-3 colonies of each strain in 7 mL sterile saline, and the OD SO o measured as an indication of the density (approx. 0.5x 108 CFU/mL or 0.5 McFarland Standard). The bacterial suspension was adjusted to a final absorbance of 0.08 to 0.100, using saline to achieve correct density, and as the blank. Within 15 minutes of preparation, the adjusted bacterial suspension with sterile saline 1 :20 {1 mL into 19 mL sterile saline) to achieve a final bacterial concentration of 4 to 5*10 6 CFU/mL. The bacterial solution was placed into a sterile trough and 10 μί of bacterial solution added into wells 2 through to 12 on each required row {dilution of 1 : 10, with final concentration of bacteria in wells = 5x105 CFU/mL). The tray was sealed and incubated at 37 °C for 18-24 h. The purity of bacterial suspension was confirmed by streaking out 50 μί of the 1 :20 dilution onto a SBA plate, which was incubated for 37 °C for 18 h and examined. Viable counts were carried out to ensure that the correct concentration of bacteria and been added to the wells. The diluted bacterial solution {4 to 5 x10 6 CFU/mL) was diluted 1 : 10 downwards by adding 100 μί to 900 μί of sterile saline in sterile tubes, and the serial dilutions continued 1 : 10 for 5 tubes. 100 μί (4-5 drops) of the 4 th and 5 th dilutions {tube 4 = 105 and tube 5 = 106 CFU/ml) was plated around in duplicate on pre- dried PCA agar plates and incubated at 37°C overnight. The following day the number of colonies on the plates was counted and the average count in 100 μί obtained. The study was multiplied by 10 to obtain a viable bacterial count per mL.

Description and identification of isolates

[00220] The MRSA isolates were originally cultured on selective Brilliance MRSA Chromogenic Agar (Oxoid). Suspect colonies were selected on the basis of their colony appearance on this agar and identification as Staphylococcus aureus was determined using colony characteristics on non-selective SBA and phenotypic characteristics such as Gram stain, positive catalase test, positive coagulase test (tube coagulase test using rabbit plasma) and clumping factor (agglutination with the Oxoid Staphytect latex test), positive Voges Proskauer test, and the ability to produce acid from trehalose. A positive cefoxitin resistance screen confirmed the isolates as MRSA.

[00221 ] MRSA clonal complexes were determined by rapid molecular typing. Two of the strains could not be typed using the rapid method, as shown in Table 2 below. Table 2: a table showing the MRSA clonal complexes according to Example 1.

NA: Not Applicable; TBD: Isolates could not be typed using the rapid method and are currently being identified using traditional methodology.

[00222] All Enterococcus isolates underwent a simplified biochemical identification based on Quinn et al. (1994, Clinical Veterinary Microbiology, Mosby Ltd, New York). Biochemical profiling provisionally identified four of the VRE isolates as Enterococcus faecalis and the remainder as possibly Enterococcus faecium. All S. pneumoniae isolates were identified on the basis of standard biochemical profiling.

Test product and storage

[00223] Analytical grade NCL812 (batch 20081214) with a defined potency of 1000 mg/g (ie 100%) was obtained and the powder was stored at a temperature of -20 °C. Aliquots (1 mL) of stock solution (25.6 mg/mL) were prepared in DMSO and stored at -80 °C and defrosted immediately before use.

Minimum inhibitory concentration determination [00224] Minimum inhibitory concentrations ( g mL) were determined using the broth microdilution method recommended by the Clinical and Laboratory Standards Institute (CLSI) (Methods for Dilution Antimicrobial Susceptibility Tests for Bacteria That Grow Aerobically; Approved Standard - Seventh Edition. CLSI M7-A7, 2006; Performance Standards for Antimicrobial Disk and Dilution Susceptibility Tests for Bacteria Isolated from Animals; Approved Standard - Second Edition. CLSI M31-A2, 2002; Performance Standards for Antimicrobial Susceptibility Testing; CLSI M2-A9, 2006).

[00225] The MIC was regarded as the the lowest concentration of an antimicrobial agent that completely inhibited growth of the organism in the microdilution wells as detected by the unaided eye. MIC breakpoints were determined by visual assessment and then confirmed using an ELISA plate reader, measuring absorbance levels at 450 nm. Bacterial growth (turbidity) in the wells with antimicrobial was compared with the amount of growth (turbidity) in the growth- control well (containing no antimicrobial). All isolates were tested in duplicate, if there was a difference of greater than one two-fold dilution in the results, the test was repeated a third time. The purity of the isolates was closely monitored during testing by subculturing the prepared bacterial inoculum onto SBA. Control organisms (Enterococcus faecalis strain ATCC 29212, S. aureus strain ATCC 29213 and S. pneumoniae strain ATCC 49619) were used throughout the testing to monitor quality control. The MICs of the control strains for the antimicrobial ampicillin (range 1.0, 2.0 and 0.06 μg/mL, respectively) were determined for each testing run as an internal quality control. The MIC 50 , MIC go and MIC range (minimum and maximum) were calculated for each of the bacterial groups.

Results

[00226] Ampicillin MIC values obtained for the ATCC control strains were within the normal range expected on the basis of CLSI recommendations. The NCL812 and ampicillin MIC values for each isolate are indicated in Table 3 (MRSA isolates), Table 4 (VRE isolates) and Table 5 (S. pneumoniae isolates) below. MIC 50 , MIC 90 , MIC mode and MIC range for each of the species of bacteria tested are shown in Table 6 below. The MIC 50 is considered to be the lowest concentration which inhibits visible growth for 50% of the isolates. The MlC go is considered to be the lowest concentration which inhibits visible growth for 90% of the isolates. The MIC mode is the most commonly occurring MIC value and MIC range the minimum and maximum MIC values obtained.

Table 3: The Minimum Inhibitory Concentrations for the individual Staphylococcus aureus isolates according to Example 1. MIC Testing - AMP/NCL812 Test 1 Test 2 Test Test 2

AMP Ampieillin

MIC90 I >128 Mg/ml | >128 Mg/ml I 4 Mg ml I 4 Mg/m

Table 4: The Minimum Inhibitory Concentrations for the individual Enterococcus isolates according to Example 1.

AMP Ampieillin

JC90 I 2 ug/ml [ 2 ug/ml [ 4 ug mi | 4 ug ml

Table 5: The Minimum Inhibitory Concentrations for the individual Streptococcus pneumoniae isolates according to Example 1.

Table 6: The NCL812 MIC 50 , MIC S0 , MIC mode and MIC range for Australian isolates of MRSA, VRE and S. pneumoniae.

Made— the most commonly occurring MTC value.

§ Comparative ampicillin MICs are slicmii in parenthesis.

[00227] NCL812 MIC values were consistent within and between each of the three species. MIC 50 and MIC 90 values were both equal (4 pg/ml) for MRSA, VRE and S. pneumoniae isolates, with less than 10% of isolates showing MIC values either 1 -2 dilutions below or only one dilution above this figure.

[00228] On the basis of these results, NCL812 represents a new antibacterial.

EXAMPLE 2: Effect of NCL812 on Staphylococcus aureus Macromolecular Synthesis Materials and Methods

Test compounds

[00229] Test compound NCL812 was transported to the experimental facility under conditions of ambient temperature and then stored at 2-8 °C until assayed. Stock solutions were made by dissolving NCL812 dry powder in 100% DMSO to a concentration of 6,400 μg mL. Vancomycin {Cat. # 1 134335), Rifampicin {Cat. # R-7382) and Cerulenin {Cat. # C-2389) were all obtained from Sigma, Ciprofloxacin was obtained from USP {Cat. # 1134335) and Linezolid was obtained from ChemPacific {Cat. # 35710).

Minimal Inhibitory Concentration Testing

[00230] The MIC assay method followed the procedure described by the Clinical and Laboratory Standards Institute, or CLSI (Clinical and Laboratory Standards Institute. Methods for Dilution Antimicrobial Susceptibility Tests for Bacteria That Grow Aerobically; Approved Standard— Eighth Edition. CLSI document M07-A8 [ISBN 1-56238-689-1]. Clinical and Laboratory Standards Institute, 940 West Valley Road, Suite 1400, Wayne, Pennsylvania 19087-19898 USA, 2009), and employed automated liquid handlers to conduct serial dilutions and liquid transfers. The medium employed for the MIC assay was Mueller Hinton II Broth (MHB II- Becton Dickinson, Sparks, MD; Cat No 212322; Lot 904441 1). S. aureus ATCC 29213 served as the quality control strain, and linezolid was utilized as the quality control antibiotic to validate the assay. NCL812 and linezolid were both dissolved in 100% DMSO before addition to the growth medium.

Macromolecular Synthesis Assays

Bacteria and growth conditions

[00231 ] The effect of NCL812 on whole cell DNA, RNA, cell wall, protein and lipid synthesis was investigated using S. aureus ATCC 29213. Cells were grown at 35 °C overnight on Trypticase Soy agar. A colony from the plate was used to inoculate 10 ml of Mueller Hinton broth II {MHBI I), and the culture was grown to early exponential growth phase (OD 6 oo = 0.2 to 0.3) while incubating in a shaker at 35 °C and 200 rpm.

DNA, RNA, and protein synthesis

[00232] When cells reached early exponential phase, 100 μί of culture was added to triplicate wells containing various concentrations of test compound or control antibiotics (5 μί) at 20* the final concentration in 100% DMSO. A 5% DMSO treated culture served as the "no drug" control for all experiments. Cells were added in MHBII at 105% to account for the volume of drug added to each reaction or in M9 minimal medium for protein synthesis reactions. Following 15 min incubation at room temperature, either [ 3 H] thymidine (DNA synthesis), [ 3 H] uridine (RNA synthesis) or [ 3 H] leucine (protein synthesis) was added at 0.5-1.0 μθί per reaction, depending on the experiment. Reactions were allowed to proceed at room temperature for 15-30 min and then stopped by adding 12 μΙ_ of cold 5% trichloroacetic acid (TCA) or 5% TCA/2% casamino acids (protein synthesis only). Reactions were incubated on ice for 30 min and the TCA precipitated material was collected on a 25 mm GF/A filter. After washing three times with 5 ml of cold 5% TCA, the filters were rinsed two times with 5 mL 100% ethanol, allowed to dry, and then counted using a Beckman LS3801 liquid scintillation counter.

Cell wall synthesis

[00233] Bacterial cells in early exponential growth phase were transferred to M9 minimal medium and added to 1.5 mL eppendorf tubes (100 μ-Jtube) containing various concentrations of test compound or control antibiotics (5 μί) at 20* the final concentration in 100% DMSO as described above. Following a 5 min incubation at 37 °C, [ 4 C]N-acetylglucosamine (0.4 μΰί/ΓβθθΙίοη) was added to each tube and incubated for 45 min in a 37 °C heating block. Reactions were stopped through the addition of 100 μί of 8% SDS to each tube. Reactions were then heated at 95 °C for 30 min in a heating block, cooled, briefly centrifuged, and spotted onto pre-wet HA filters (0.45 μΜ). After washing three times with 5 mL of 0.1% SDS, the filters were rinsed two times with 5 ml of deionized water, allowed to dry, and then counted using a Beckman LS3801 liquid scintillation counter.

Lipid synthesis

[00234] Bacterial cells were grown to early exponential growth phase in MHBI I broth and added to 1 .5 mL eppendorf tubes (in triplicate) containing various concentrations of test compound or control antibiotics as described above. Following a 5 min incubation at room temperature, [ 3 H] glycerol was added at 0.5 μθΐ per reaction.

[00235] Reactions were allowed to proceed at room temperature for 15 min and then stopped through the addition of 375 μί chloroform/methanol (1 :2) followed by vortexing for 20 seconds after each addition. Chloroform (125 μί) was then added to each reaction, vortexed, followed by the addition of 125 μί dH 2 0 and vortexing. Reactions were centrifuged at 13,000 rpm for 10 min, and then 150 μί of the organic phase was transferred to a scintillation vial and allowed to dry in a fume hood for at least 1 hr. Samples were then counted via liquid scintillation counting.

Results

[00236] Susceptibility testing was conducted with NCL812 and S. aureus ATCC 29213 to determine the concentrations of drug needed in the macromolecular synthesis assays. 100237] Table 7 shows that the MIC for NCL812 was 4 Mg/mL, while the quality control agent linezolid was within the CLSI-established quality control range {Clinical and Laboratory Standards Institute. Performance Standards for Antimicrobial Susceptibility Testing; Nineteenth Informational Supplement. CLSI document M100-S20 [ISBN 1-56238-716-2]. Clinical and Laboratory Standards Institute, 940 West Valley Road, Suite 1400, Wayne, Pennsylvania 19087-1898 USA, 2010). Precipitation of NCL812 was observed at >8 μg/mL in plates that were prepared in an identical fashion, but did not receive an inoculum of S. aureus. Macromolecuiar synthesis inhibition studies were performed using concentrations of NCL812 that were equivalent to 0, 0.25, 0.5, 1 , 2, 4 or 8-fold the MIC value (4 Mg/ml) for S. aureus ATCC 29213 (Figures 11 -16).

Table 7: Minimum Inhibitory Concentrations values for NCL812 (robenidine) and linezolid against Staphylococcus aureus ATCC29213 according to Example 2.

[00238] Figure 2 shows the effect of NCL812 on DNA synthesis. NCL812 demonstrated no inhibition at 0.25 fold the MIC, 40% inhibition at 0.5 fold, and approximately 95% inhibition at the MIC. This is compared to the control ciprofloxacin which showed approximately 51 % at 8 fold the MIC {0.5 g/mL). The results for NCL812 inhibition of RNA synthesis were very similar to the DNA synthesis study, with rifampicin serving as the positive control {Figure 3. It should be noted that precipitation was observed at 4 to 8 fold the MIC in the Mueller Hinton broth II utilized in the DNA and RNA synthesis assays.

[00239] Protein synthesis was inhibited in a dose-dependent manner at 0.25, 0.5, and 1 fold the MIC value of NCL812 showing up to 97% inhibition at the MIC (Figure 4. Linezolid demonstrated approximately 61 % inhibition of protein synthesis at 8 fold the MIC (2 μg/mL). Precipitation of NCL812 occurred at 4 and 8 fold the MIC in the protein synthesis assay.

[00240] In Figure 5 NCL812 also showed a somewhat dose-dependent inhibition of cell wall synthesis, though there was a large increase in inhibition from 1 to 2 fold the MIC. However, inhibition dropped to approximately 68% and 52% at 4 fold and 8 fold the MIC, respectively. Precipitation of NCL812 occurred at 2, 4, and 8 fold the MIC in the M9 minimal medium used for the cell wall synthesis assay, and that is the likely cause of the decline in inhibition. In comparison, the positive control vancomycin showed 96% inhibition at 8 fold the MIC (2 ig/mL). NCL812 demonstrated a similar inhibition profile against lipid synthesis as that shown for DNA and RNA synthesis, reaching approximately 90% inhibition at the MIC {Figure 6). The positive control inhibitor cerulenin demonstrated 72% inhibition at 8 fold the MIC (32 Mg/mL).

[00241 ] Figure 7 represents a composite of all five macromolecular synthesis reactions. It can be observed that the inhibition curves were similar for each pathway, suggesting a global inhibition of several pathways simultaneously by NCL812. It is possible that NCL812 targets the cell membrane, causing leakage of essential ions and/or metabolites, thereby leading to a global shutdown of the cell synthesis pathways.

[00242] In summary, NCL812 inhibited DNA, RNA, protein, cell wall, and lipid pathways in a growing culture of S. aureus. Though some instances of dose-dependent inhibition of pathways was observed, all five macromolecular synthesis reactions were similarly sensitive to NCL812.

EXAMPLE 3: Effect of NCL812 on ATP Release from Staphylococcus aureus

Materials and Methods

Test compounds

[00243] The test compound NCL812 was shipped under conditions of ambient temperature and then stored at 2-8 °C until assayed. Stock solutions were made by dissolving NCL812 dry powder in 100% DMSO to a concentration of 1 ,600 μg/mL. Polymyxin B was obtained from Sigma (Cat. # P-4932).

Test Organism

[00244] S. aureus ATCC 29213 was originally acquired from the American Type Culture Collection (Manassas, VA).

A TP Release Assay

[00245] The CellTiter-Glo Luminescent Cell Viability Assay (Promega) was utilized to measure the leakage of ATP from bacteria. Cultures were grown to early exponential phase (0.2 - 0.3 OD eoo ) in Mueller-Hinton Broth II and then treated with seven different concentrations of either NCL812 or polymyxin B (positive contra!) utilizing the MIC for each compound as a guide (0, 0.25, 0.5, 1 , 2, 3, 4, or 8 fold the MIC). The negative control received 2% DMSO, which represented the final DMSO concentration in each assay. After a 30 min exposure to drug, cells were sedimented by centrifugation and the supernatant was analyzed for the presence of ATP. Results were expressed as ATP concentration released to the medium (μΜ).

Results [00246] The MIC for NCL812 has been previously determined to be 4 μ9/ιηΙ_. The ATP release assay is conducted by growing S. aureus to exponential phase and then adding drug at multiples of the MIC in an effort to detect a dose-dependent response.

[00247] As shown in Figure 8 the positive control polymyxin B released ATP from S. aureus cells in a dose-dependent fashion with maximal release of approximately 0.34 μΜ ATP at 8 fold the MIC (256 μg/mL). ATP release in the presence of NCL812 was dose-dependent at 0.5-1 fold the MIC, resulting in maximal release (0.33 μΜ) observed at the MIC (4 μg ml). ATP release actually decreased thereafter at 2 to 8 fold the MIC. It should be noted that in previous studies precipitation of NCL812 was observed at 4 to 8 fold the MIC in Mueller Hinton broth II.

[00248] In summary, NCL812 demonstrated dose-dependent release of ATP from actively growing S. aureus cells. ATP release from the cells into the growth medium reached maximum levels at the MIC value, and this was followed by a decrease in ATP release at higher doses. The data indicated that NCL812 may interact with the cell membrane of S. aureus, causing leakage of vital metabolites such as ATP.

EXAMPLE 4: In vitro antibacterial activity of NCL812 against methicillin-resistant and methicillin-susceptible Staphylococcus aureus

Materials and Methods

Antimicrobial agents

[00249] Aliquots of stock solution of NCL812 (25.6 mg/ml) was prepared in DMSO, stored at -80 °C and defrosted immediately before use. Ampicillin stock was obtained from Sigma- Aidrich (Australia). Antimicrobial discs were obtained from Thermo Fisher Scientific (Australia).

Microorganisms

[00250] Clinical isolates of MRSA that represented the most common sequence types of both hospital-acquired (HA) and community-associated (CA) MRSA in Australia were obtained and are described in Table 8 below. The S. aureus control organism ATCC 49775 was used. Isolate identification was confirmed by conventional phenotypic methodologies, including the slide coagulase test, Vogues-Proskauer test, polymyxin B sensitivity (300 units), and Staphytect Plus Protein A latex slide agglutination (Thermo Fisher Scientific Australia). Bacteria were stored at -80 °C in 40% glycerol broth and routinely grown from stock on sheep blood agar (SBA) incubated at 37 °C. In subsequent experiments, only fresh cultures <24 h were used. Table 8: Staphylococcus aureus clone/isolate name ! type, source, antibiogram, clindamycin resistance status, multi-locus sequence type fMLST), staphylococcal cassette chromosome (SCCmec) type, clonal complex, Panton-Valentine leukocidin staius (PVL), and spa type for isolates used according to example 4

>

MSSA; methicillin-susceptible S. aureus. HA-MRSA: hospital-acquired methicillin-resistant S, aur&us. CA-MRSA; community-associated met iciliin- resistant S. aureus, Em; Erythromycin. Ci; Ciprofloxacin, Gn; Gentamicin. Tm; Trimethoprim. Te; Tetracycline. FA; Fusidic Acid. Rf: Rifampidn. Mp; Mu irocin

Isolate resistotyping

100251 ] Antibiotic-susceptibility profiling of the isolate collection was undertaken using Kirby-Bauer disc diffusion, as recommended by the Clinical and Laboratory Standards Institute (CLSI) on Mueller-Hinton agar. Isolates were grown overnight on SBA at 37 °C. Colonies were suspended in physiological saline. Turbidity was adjusted to a 0.5 McFarland standard and suspensions were spread over the medium. Antibiotic discs according to Table 9 below were transferred onto the inoculated medium and analysed after 24 h incubation at 37 °C. Isolates labelled as MRSA that were not β-lactam-resistant on the basis of the Kirby-Bauer test were grown from stock on plate count agar supplemented with 5 pg/ml ampicillin and subject to repeat testing, as PBP2a expression can be induced by exposure to β-lactam antimicrobials.

Table 9: Antibacterial agent zone diameter interpretive sizes for Kirby-Bauer disc diffusion, as used in Example 4.

Disk Zone diameter interpretive sizes (mm)

Antimicrobial Class Agent

content Resistant Intermediate Susceptible

Aminoglycoside Gentamicin 10 pg ≤12 13-14 ≥15

Ansamycin Rifampicin 5 pg ≤16 17-19 ≥20

Cephalosporin Cephalexin 30 pg ≤14 15-17 ≥18

Cephamycin Cefotetan 30 pg ≤12 13-15 >16

Folate pathway Trimethoprim- 1.25/23.75

≤10 11-15 >16 inhibitor sulfamethoxazole

Glycopeptide Vancomycin 30 pg - - ≥15

Lincosamide Clindamycin 2 pg ≤14 15-20 ≥21

Macrolide Erythromycin 15 pg ≤13 14-22 ≥23

Fluoroquinolone Ciprofloxacin 5 pg ≤15 16-20 ≥21

Tetracycline Tetracycline 30 pg ≤14 15-18 ≥19 β-lactam/penicillin Penicillin-G 10 units ≤28 - ≥29 β-lactam/penicillin- Oxacillin 1 ≤10 11-12 >13 stable penicillin p-lactam/p-lactamase Amoxicillin-

20/10 ig ≤19 - >20 inhibitor combination clavulanic acid

Molecular detection of the protein A and mecA genes to confirm MRSA status

[00252] Isolate identities were confirmed genotypically using a duplex polymerase chain reaction (PCR) test targeting the spa (protein A) and mecA (methicillin resistance) genes. In addition, the isolates were tested in a mecA and spa Sybr green real-time PCR. Approximately ten colonies of each overnight bacterial subculture was suspended in 1 * phosphate buffered saline (pH 7.4) and vortexed. Isolates were subject to DNA extraction using the QIAamp® DNA Mini Kit (Qiagen, Australia) following the manufacturers protocols. Template DNA was eluted in 50 μΙ_ of elution buffer and either used directly in PCR, or stored at -20 °C prior to DNA amplification using the spa forward (5'-TGATACAGTAAATGACATTG~3') and reverse (5 - TTCTTATCAACAACAAGTTC-3') primers and mecA forward (5 - TTCGTGTCTTTTAATAAGTGAGG-3') and reverse (5'-ATGAAGTGGTAAATGGTAATATCG-3') primers (Invitrogen, Australia). Conventional PCR amplification was performed in a 20 μΙ_ volume containing 10 μΙ_ HotStarTaq Plus Master Mix (Qiagen, Australia), 0.5 μΜ of each spa primer, 0.2 μΜ of each mecA primer, and 3 μΙ_ of extracted DNA. An automated thermal cycler (T100 Thermal Cycler, Bio-Rad) was used for PCR amplification of the spa and mecA genes according to the following conditions: PCR Stage (Enzyme activation at 95 °C for 300 s, followed by 38 amplification rounds of 94 °C for 30 s (denaturation), 50 °C for 30 s (annealing) and 72 °C for 38 s (extension) and then a cooling stage of 20 °C until required); Real-time PCR Stage (Enzyme activation at 95 °C for 300 s, followed by 40 amplification rounds of 95 °C for 15 s (denaturation), 50 °C for 20 s (annealing) and 70 °C for 40 s (extension), a single round at 95 °C for 5 s, a sinlge round at 55 °C for 20 s, continuous melting curve from 95 °C to 0 °C and a cooling period of 40 °C for 30 s. The mecA and spa amplified products of 325 and 120 bp, respectively, were detected by GelRed staining followed by electrophoresis in 2% agarose gels.

Minimum inhibitory concentration testing

[00253] The in vitro activities of NCL812 and ampicillin as a positive control were determined by broth microdilution as recommended by the CLSI in cation-adjusted Mueller- Hinton I I broth. Microtiter plates containing two-fold dilutions of each antimicrobial agent were inoculated with -10 5 CFU/ml of each isolate in a 100 μΙ_ final volume. Plates were incubated for 24 h at 37 °C. Turbidity (absorbance at OD 60 o) was measured using a Bio-Rad Benchmark Plus microplate spectrophotometer in Microplate Manager® version 5.2.1 (Bio-Rad). Minimum inhibitory concentration (MIC) endpoints were defined as the lowest antimicrobial concentration assessed by the spectrophotometer that inhibited bacterial growth. ATCC 49775 was included in the isolate collection as a control organism using breakpoints defined by the CLSI. The MIC 50 , MICgo (concentrations that inhibited growth of the lower 50% and 90% of total organisms, respectively), and MIC range (minimum and maximum) were calculated to profile the antimicrobial susceptibility of the isolate collection.

Bactericidal activity

[00254] The bactericidal activity of NCL812 was established by determination of the minimum bactericidal concentration (MBC) and time-kill analyses using CLSI guidelines. The MBC was defined as the lowest drug concentration at which 99.95% of the original inoculum was eliminated.

[00255] Time-kill assays for ATCC 49775 were performed in cation-adjusted Mueller- Hinton I I broth in Microtiter plates and again in 10 ml volumes for macrodilution assays at antimicrobial concentrations equivalent to 1 * and 4 χ the MIC. Bactericidal activity in macrodilution assays was identified as a 3log 0 decrease from the initial inoculum size. Bacteria were cultured overnight at 37 °C on SBA. Colonies were suspended in broth and the turbidity was adjusted to a 0.5 McFarland standard to obtain a bacterial suspension of ~10 5 CFU/ml. Bacterial suspensions were incubated at 37 °C with shaking. Aliquots were removed at 0, 1 , 2, 4, 8, 12, and 24 h after antimicrobial addition, diluted, plated onto SBA and incubated for 48 h at 37 °C for viable count determination. Turbidimetric growth curves for S. aureus were obtained for Microtiter plate assays by monitoring optical density changes using a Bio-Rad Benchmark Plus microplate spectrophotometer at 600 nm. Optical densities were measured at 0, 1 , 2, 4, 8, 12, and 24 h after antimicrobial addition.

Statistical methodology

[00256] Microbiological data was interpreted using CLSI guidelines. Data was examined using the student's t-test, Fisher's exact test, analysis of variance, and a generalized linear model for tests of between-subjects effects where appropriate. Differences were considered significant at the 0.05 level in IBM SPSS® version 19.0.

Results

Confirmation of Staphylococcus aureus identity and mecA status

[00257] Latex agglutination tests confirmed that all 30 isolates were protein A positive. The isolates tested positive for coagulase activity using slide agglutination. Voges-Proskauer and polymyxin B resistance tests confirmed that all isolates were S. aureus except for a single methicillin-susceptible isolate; MSSA DE-25, as shown in Table 10 below. Based on spa gene PCR amplification, this isolate was not identified as a S. aureus isolate despite testing positive in the protein A latex agglutination and slide coagulase tests. This canine-origin Staphylococcus spp. was identified as Staphylococcus pseudintermedius based on biochemical characteristics. mecA conventional and real-time PCR results confirmed that 66.66% of the isolates were classified as methicillin-resistant on the basis of possession of the mecA gene. There were no significant differences between the ability of conventional and real-time PCR to detect the mecA gene (P>0.05).

Table 10: Percentage of presumptively identified S. aureus isolates reporting positive to selected phenotyptc and genotypic tests according to Example 4.

HA-MRSA; hospital-acquired S. aureus . GA-MRSA; community-associated S. aureus . S. aureus isolates were identified as testing positive to protein A latex agglutination (Protein A), slide coaguiase, Voges-Proskauer and polymyxin B resistance tests, as weii as testing positive for polymerase chain reaction (PCR) and real-time PGR amplification of the spa gene. Methicillin-resistant S, aureus isolates were identified as isolates testing positive to the criteria described above, as well as positive for PGR and real-time PGR of the mecA gene.

Table 11 : Resistance of S, aureus Isolates to antibacterial agents using the Kirby-Bauer disc diffusion method according to example 4

HA-MRSA; hospital-acquired methicillin-res'istant S, aureus. CA-MRSA; community-associated methicillin-resistant S. aureus

Staphylococcus aureus antimicrobial susceptibility profiles

[00258] Antimicrobial susceptibility assays revealed that HA-MRSA isolates had the highest mean prevalence of resistance to multiple antimicrobial classes (P<0.000). CA-MRSA isolates were next most resistant (PO.007), followed by methicillin-susceptible staphylococci (PO.037), as shown in Table 11 above. Oxacillin resistance was expressed in only 80.00% and 10.00% of HA-MRSA and CA-MRSA isolates, respectively. Cefotetan resistance was expressed in 80.00% and 20.00% of HA-MRSA and CA-MRSA isolates, respectively. Although oxacillin and cefotetan did not significantly differ in their ability to detect MRSA (P>0.05), detection was significantly improved when using the mecA PCR when compared to disc diffusion (PO.013). The majority of HA-MRSA isolates expressed resistance to amoxicillin-clavulanic acid, cefotetan, cephalexin, clindamycin, erythromycin, oxacillin, and penicillin-G, whereas the majority of CA-MRSA isolates were resistant to only clindamycin, erythromycin, and penicillin-G. None of the isolates tested were vancomycin resistant. Overall, the most prevalent resistance phenotypes were penicillin-G (83.33%), erythromycin (73.33%), and clindamycin (43.33%), whilst only single isolates (3.33%) were resistant to trimethoprim-sulfamethoxazole and rifampicin. mec gene complex interactions

[00259] All MRSA isolates belonging to mec gene complex A expressed resistance to both oxacillin and cefotetan, as shown in Table 12 below. However, only 20% of mec gene complex B MRSA isolates were phenotypically resistant to these antimicrobials. Of the MRSA isolates belonging to mec gene complex C2, only a single isolate expressed methicillin resistance to oxacillin and only two isolates expressed resistance to cefotetan. Unclassified MRSA isolates expressed full resistance to oxacillin and cefotetan.

Table 12: Number and percentage of identified mec gene complexes in 20 S. aureus strains classified as methicillin-resistant according to example 4 mec gene complex SCCmec Oxacillin resistant Cefotetan resistant

(number of isolates) types isolates isolates

II 3 (100%) 3 (100%)

A

III 1 (100%) 1 (100%)

(n=4)

Overall 4 (100%) 4 (100%)

I 1 (50%) 1 (50%)

B

IV 1 (12.5%) 1 (12.5%)

(n=10)

Overall 2 (20%) 2 (20%) C2 V 1 (25%) 2 (50%)

<n=4) Overall 1 (25%) 2 (50%)

Unclassified

Overall 2 (100%) 2 (100%) <n=2)

Respective staphylococcal cassette chromosome (SCCmec) complexes and types expressing phenotypic resistance to oxacillin and cefotetan are indicated as well as real-time mecA status, and the average negative dF/dT peak obtained from melting point analysis from real-time PCR of the mecA gene

[00260] Melting point peaks for the mecA real-time PCR negative derivative plot -dF/dT differed between mec gene complex (PO.003) (Figure 9. On average, mec gene complex B and unclassified isolates demonstrated higher melting point peaks than other SCCmec types (P<0.012).

Physical properties of test antimicrobials and comparison of minimum inhibitory concentration results from initial analogue testing

[00261 ] Test antimicrobials were selected on the basis of solubility and antimicrobial activity from preliminary studies. Cloudy precipitates were observed when both NCL812 were dissolved in cation-adjusted Mueller-Hinton II broth, as shown in Table 13 below. Following initial structure-activity testing on each synthesized analogue, NCL812 was found to have consistent MIC values in this present study.

Table 13: Characteristics of antibacterial NCL812 and the β-Iactam antibacterial ampicillin according to Example 4.

Mueller-Hinton II broth (CAMHB), and average minimum inhibitory concentrations (MIC) ^g/ml at 24 h) against methicillin-resistant S. aureus (MRSA) determined from preliminary studies and those determined during this present study. ATCC 49775; methicillin-susceptible S. aureus isolate and ATCC control strain. MRSA580; methicillin-resistant S. aureus isolate #580. MRSA698; methicillin-resistant S. aureus isolate #698

In vitro antibacterial activities: minimum inhibitory concentrations [00262] MI Cso and MIC g0 values for lead compound NCL812 (4- and 4-8 ig/ml) are shown in Table 14 below. MIC values differed by S. aureus classification (susceptible, HA- or CA-MRSA) (P<0.005). In many cases, NCL812 had significantly increased activity against CA- MRSA and methicillin-susceptible staphylococci by one dilution when compared to HA-MRSA (PO.002 and P<0.020, respectively), however there were no significant differences between MIC values for methicillin-susceptible staphylococci and CA-MRSA (P>0.05). Ampicillin MIC values obtained for the ATCC control strain were within the normal range expected on the basis of CLSI guidelines

Table 14: In vitro activities of the novel antibacterial NCL812 and the β-lactam antibacterial ampicillin against S. aureus clinical isolates according to Example 4.

HA-MRSA; hospital-acquired methicillin-resistant S. aureus. CA-MRSA; community-associated methicillin-resistant S. aureus. MIC; minimum inhibitory concentration (Mg/ml). MBC; minimum bactericidal concentration (Mg/ml). MIC/MBCrange; minimum and maximum MIC/MBC for all isolates. MIC/MBC 50 ; MIC/MBC at which 50% of isolates are inhibited. MIC/MBC 90 ; MIC/MBC at which 90% of isolates are inhibited

In vitro antibacterial activities: minimum bactericidal concentrations

[00263] The MBCs determined from NCL812 were equivalent to the MIC for 93.33% and 83.33% of S. aureus isolates, respectively (Table 14). In all remaining cases, MBCs were one dilution higher. For NCL812, MBCs ranged from 2-8 μg/mL· and 4-16 pg/mL, respectively. Time-kill studies

100264] In comparison to the turbidimetric growth curve of ATCC 49775, no visible bacterial growth was observed when ATCC 49775 was inoculated into cation-adjusted Mueller Hinton I I broth supplemented with NCL812 at 1 * and 4* the MIC in microdilution assays (PO.033 and P<0.038, respectively) (Figure 10).

[00265] When analysed in 10 mL macrodilution assays, broth supplemented with antimicrobials at 1 * and 4x the MIC and inoculated with ATCC 49775 displayed significantly reduced viable counts for both NCL812 concentrations when compared to the growth control (0.000<P<0.008) (Figure 11 ). Additionally, the time-kill profiles of each concentration of NCL812 did not significantly differ (P>0.05). Both concentrations remained bactericidal until approximately 8-12 h after antimicrobial addition, where bacterial regrowth was observed. Considerable variation in the killing activity of NCL812 was observed from 8-24 h. Although NCL812 was no longer bactericidal by 24 h, viable counts observed at 1 * the MIC remained significantly lower than those obtained from unsupplemented broth (PO.046).

[00266] In summary, the example set out above demonstrates bactericidal activity against both methicillin-susceptible staphylococci and MRSA. MIC and MBC values were consistently low across the selection of isolates (MIC ran ga 2-8 μg/mL). NCL812 retained good in vitro antimicrobial activity against common, multidrug-resistant MRSA isolates, including the epidemic UK EMRSA-15, EMRSA-16, and EMRSA-17, Irish EMRSA-1 , AUS EMRSA-3, NY/JAPAN HA-MRSA, and predominant CA-MRSA clones. NCL812 was also active against one S. pseudintermedius isolate that was originally identified as a S. aureus strain.

[00267] Preliminary testing suggests that NCL812 targets the S. aureus cell membrane, causing dose-dependent release of vital metabolites such as ATP. Disruption of the bacterial membrane bilayer or proteins that are integral to membrane function in bacteria is a target for numerous large antimicrobials which are ubiquitous in nature; including glycolipids, lipopeptides, lipoproteins, fatty acids, neutral lipids, phospholipids, and biosurfactants. Although NCL812 is a low molecular mass (≤500 Da) synthetic compound, it does appear to exert bactericidal activity in a similar manner to other antimicrobials which target the Gram-positive cell membrane, including the high molecular weight cyclic lipodepsipeptide antimicrobial agent daptomycin, or the low molecular mass quinolone-derived HT61 , whose chemical structure is not currently available. Many of these lipophilic antibacterial agents are also not effective against Gram- negative microorganisms due to the presence of the outer lipid bilayer membrane, which contains narrow porin channels reducing the net penetration of some compounds into the cell.

[00268] The insolubility of NCL812 at even low concentrations in microbiological media may reflect the amphipathic and oligomeric nature of this antimicrobial and suggests that the real MIC may be much lower than observed, as it is likely that it is only NCL812 in solution that is biologically active. In time-kill studies, NCL812 exerted rapid in vitro bactericidal activity against ATCC 49775. Again, these findings are consistent with a time-kill profile of cell membrane function inhibitors such as daptomycin and HT61.

[00269] Importantly, the apparent short in vitro half-life of this antimicrobial resulted in bacterial regrowth observed at 12 h after antimicrobial addition. This suggests that if a viable bacterial population survives the initial exposure to NCL812 prior to antimicrobial inactivation, bacterial regrowth will occur. The development of resistance to NCL812 in these studies was ruled out as test bacteria remained susceptible to NCL812 following harvesting, washing and MIC testing. Whilst the apparent short in vitro half-life of NCL812 may be a desirable characteristic for future in vivo application, it does suggest that NCL812 should be administered every 8 h in future in vivo safety and efficacy experiments to maintain adequate systemic concentrations, though it would appear from the time-kill profile that the NCL compound series are concentration-dependent rather than time-dependent antimicrobials.

100270] To overcome the methicillin-susceptible phenotype, extending disc diffusion incubation time from 24 to 48 h compensates for the slow derepression of the mecR gene. Although the effects of longer incubation were not examined, and the smali sample size of MRSA isolates prevented further investigation into mec complex interactions; genetic techniques were of significantly improved sensitivity when compared to phenotypic methods for confirmation of the mecA status of the isolates in this study. Although genetic techniques are not always employed as a routine method for detecting MRSA, real-time PCR identification of the presence of the mecA gene in a Staphylococcus spp. isolate remains the diagnostic gold standard.

EXAMPLE 5: In vitro pharmacodynamics of a new antimicrobial agent for Streptococcus pneumoniae.

Materials and Methods

Pneumococcal antimicrobial susceptibility Pneumococcal strains and growth conditions

[00271 ] Twenty pneumococcal isolates that comprised six characterised laboratory strains and 14 clinical isolates were the subject of this study (P9/6A, P21/3, WCH16/6A, WCH43/4, WCH46/4, WCH57/8, WCH77/5, WCH86/4, WCH89/7, WCH92/4, WCH 137/6A, WCH158/19F, WCH 184/19F and WCH211/1 1 ; strain/serotype, respectively). Other isolates used in this example were: A66.1/3 (Francis et a/., 2001 . Infect Immun. 69: 3350-2358); EF3030/19F (Briles et a/. , 2003 J. Infec. Diseases. 188:339-348); L82016/6B (Briles et a/., 2000 Infect Immun. 68:796-800); TIGR4/4 (Tettlelin ef a/. , 2001 Science 293:498-506); and WU2/3 (Bri!es ef a/., 1981 J. Exp Med. 153:694-705). See Table 15 below for the phenotypic characteristics of the isolates used in this study. The National Collection of Type Cultures (NCTC) control strain D39 (Avery ei a/., 2010 Nature Reviews Microbiology 8:260-271 ) was used as a growth control for all MIC and MBC assays. D39 was later designated for kill kinetics, point of resistance assays and transmission electron microscopy (TEM) studies as it is a well documented laboratory strain with a defined in vivo pathogenesis (Table 15) that displayed consistent NCL812 MICs and MBCs.

Table 15: Pneumococcal isolates and their phenotypic description according to Example 5.

ND = Not determined

[00272] For all in vitro assays, fresh pneumococcal isolates were grown overnight (O/N) on horse blood agar (HBA) plates (39 g/L Columbia blood agar base lOxoid] 5% Iv/v] defribinated horse blood lOxoid] at 37 °C with 5% supplemented C0 2 ). Mueller-Hinton blood agar with 5% defibrinated sheep blood (MHSBA Roseworthy Media and Blood Service) was used for disk diffusion analysis as directed by Clinical Laboratory Standards Institute (CLSI) standards. Pneumococci were routinely grown in broth consisting of 4% lysed horse blood (LHB) with Cation Adjusted Mueller Hinton Broth (CAHMB, [Difco]) at 37 °C with 5% supplemented C0 2 . Horse serum broth (HSB, 10% (v/v) donor horse serum in nutrient broth 110 g/L peptone, 10 g/L Lab Lemco (Oxiod) and 5 g/L NaCI]) was also used in some MIC assays. Isolates were stored in HSB at -80 °C.

Antibiotic stocks and reagents

[00273] NCL812 was provided in dry powder form. A total of 256 mg was dispensed into 10 mL of 100% D SO to make a stock of 25.6 mg/mL, which was then diluted 1 : 100 in CAHMB to make a final working stock of 256 μg mL. Ampicillin dry powder was from Sigma A0166. The original 25.6 mg/mL stock was diluted in saline 1 :100, 1 :4, 1 :20 and finally 1 :16 in CAMHB to make a final working stock of 0.18 μg/mL. Erythromycin was purchased from Sigma Aldrich and choline chloride was from Roche Diagnostics. Twenty micro litres of 0.05 μg/mL erythromycin was diluted 1 :25 in 4.980 mL of CAMHB to give a final working stock of 0.2 μg/mL. Choline chloride (0.5%) was added to 4% LHB:CAMHB for specific kill kinetic assays.

Defining antimicrobial susceptibility of pneumococcal isolates

[00274] Isolate susceptibility to 12 different antimicrobials (Table 16) was determined by CLSI and European Committee on Antimicrobial Susceptibility Testing (EUCAST) methods. Antimicrobials were selected based upon the CLSI and EUCAST guidelines. Standardised bacterial suspensions were spread onto MHSBA using a sterile cotton swab. Bacterial suspensions from of Streptococcus pneumoniae were standardised to an OD 600 between 0.08 and 0.1 using a spectrophotometer and then diluted 1 :20. Bacterial colonies were taken from an O/N horse blood agar plate. To ensure the purity of the 1 :20 bacterial suspension, 50 μί was spread plated onto horse blood agar and incubated O/N at 37 °C with 5% C0 2 . The CFU was calculated and compared to the initial plate counts. Antibiotic disks (Purchased from Sigma Aldrich) were placed using a disk dispenser (Purchased from Oxoid) according to CLSI standards. MHSBA plates were incubated for 16 h - 24 h at 37 °C in 5% C0 2 . Zones of complete inhibition were measured in triplicate to the nearest millimetre using a ruler on natural light-reflected growth, and the mode was represented as the diameter for each isolate. Pneumococcal isolates were categorised as sensitive, intermediate (I) or resistant (R) by CLSI standards and quality control (QC) ranges (Table 16).

Table 16: Antibacterials used for disc diffusion analysis with interpretive standards of zone diameters (mm) according to Example 5.

were determined by CLSI standards .

*Zone diameters for Ciprofioxacin antimicrobial susceptibility to S. pneumoniae were determined by EUCAST .

Determination of NCL812 MIC S0 , MIC 90 , MIC range and MBC 60 , MBC 90 , MBC range

[00276] MICs for NCL812 for all isolates listed in Table 15 were determined by measuring OD 60 o (Spectramax spectrophotometer, Molecular Devices Corporation) as an indicator of bacterial growth using 96 well microtitre trays after incubation for 24 h at 37 °C in 5% C0 2 . [Micro-broth dilutions and 96 well trays are prepared by the following method: 90 pL of 4% LHB:CAMHB is aliquotted into all wells using a multichannel pipette. 90 pL of working antimicrobial stocks were no serial diluted down the tray by a 1 :2 dilution. Negative broth controls and dilution control were taken into account when planning the set up of a 96 well tray.] 10 pL of bacterial suspension was then added to the appropriate wells in the 96 well tray. Appropriate positive (no antimicrobial), negative (no antimicrobial or bacteria) and negative dilution (a serial dilution control of antimicrobial and broth) controls were included in each assay. MBC and plate counts for kill kinetic assays were determined by aliquotting 20 pL from each well of the 96 well microtitre tray onto HBA, and incubating at 37 °C with 5% C0 2 . The MBC was determined by a 99.95% inhibition of S. pneumoniae, taking into account the dilution factor. MICs and MBCs were determined in quadruplicate and the mode was taken as the representative value. The MIC 50 , MIC 90 and MIC range and MBC 50 , MBC 90 and MBC range were determined according to CLSI standards. The MIC 50 and MIC 90 , or MBC 50 and MBC 90 , are defined by the lowest concentrations which, when all the MICs and MBCs of the isolates are arranged from lowest to highest, inhibited the 50th and 90th percentile of the total amount of isolates, respectively.

Micro-broth dilution time kill studies with NCL812 using strain D39

[00277] Bacterial suspensions were added in triplicate to a 96 well microtitre tray containing NCL812 with a starting concentration of 128 pg/mL and serially diluted 1 :2 sequentially to a concentration of 0.25 pg/mL. Negative dilution controls were subtracted from the median growth value to obtain a suitable indicator of overall bacterial production. The 96 well tray was incubated at 37 °C in 5% C0 2 and OD 60 o read every 2 h for the first 12 h followed by final reads at 24 and 48 h. To further supplement this data, a separate experiment in which a 96 well tray was read automatically at half hourly intervals using a spectrophotometer (Spectramax spectrophotometer, Molecular Devices Corporation) for 14 h was performed to confirm the trends in growth curves observed from original micro-broth dilution studies.

MBC time kill studies with NCL812 using strain D39

[00278] MBC kill kinetics assays involved the preparation of three 96 well microtitre trays. At specific time points, aliquots obtained from these trays provided viable counts following incubation at 37 °C in 5% C0 2 on HBA, and the MBC was determined after 24 h of growth.

Macro-broth dilution time kill studies of D39 with NCL812

[00279] Bacterial suspensions and working antibiotic stocks were prepared as described above. [For preparing macro-broth dilutions, 20 mL tubes were filled each with 9 mL of 4% LHB:CAMHB. 9 mL of a working antimicrobial stock was diluted 1 :2 when added to to one of the tubes, and then serial diluted down from a high to low concentration of antimicrobial. 1 mL of S. pneumoniae bacterial suspension was added to the appropriate tubes, including the positive control. Tubes were incubated at 37 °C with 5% C0 2 with gentle manual tilting of the tubes treated with NCL812 every 10 min for the first 12 h. At every 2-3 h during the first 12 h of growth and then at 24 h and 48 h, 50 μΙ_ of each bacterial suspension was spread plated onto HBA and incubated at 37 °C with 5% C0 2 for 16-24 h.]

[00280] Table 17 below indicates the concentrations used for each antimicrobial. Cultures were incubated at 37 °C in 5% C0 2 with gentle manual tilting every 10 min for the first 12 h. Viable counts from 50 μΙ_ aliquots of each concentration were read following incubation at 37 °C in 5% C0 2 for 24 h. The pH of each sample was measured at specific time points using pH indicator strips. Confluent growth was defined when more than 1000 colonies were counted per plate. A bactericidal effect was defined as a 1000 fold reduction (99.9%) of the original cell suspension determined at 24 h for each concentration.

Table 17: Antibacterial agent concentrations used in macro-broth dilution assays according to Example 5.

Point of resistance assay for NCL812

[00281] Macro-broth dilutions were prepared as above. Broth cultures of strain D39 {10 mL) were incubated in the presence of 2 μ9/ιηΙ_ and 4 μg/mL of NCL812, and 0.022 μ9/ιηΙ_ of Ampicillin for 6 h at 37 °C in 5% C0 2 . Samples were centrifuged at a relative centrifugal force (RCF) of 101.45xg for 10 min and washed in 50 mL of phosphate buffered saline (PBS) twice to remove any residual antimicrobial, and/or bacterial end products and media. Washed bacteria were resuspended and MICs were performed.

Effect of NCL812 on D39 cell membrane ultrastructure

Transmission Electron Microscopy

[00282] Morphological appearance and morphometric analysis of the cell membrane was determined using transmission electron microscopy (TEM). Bacterial suspensions and 10 mL cultures of D39 were prepared as before. Samples were incubated at 37 °C in 5% C0 2 with gentle manual tilting of the cultures every 10 min. Cultures were exposed to either 1 g/mL, 4 μg/mL or 16 μg/mL of NCL812 and harvested at 6 or 12 h by centrifugation at RCF of 101.45*g for 20 min and washed twice in 50 mL of PBS. Critical time points for TEM work were determined by analysing trends in the growth curves produced from the kill kinetics studies. Samples were resuspended in PBS containing 20% glycerol and stored at -80 °C until required. Before fixation, 20% glycerol was removed by centrifugation and washing on ice three times in 50 mL of PBS.

[00283] Samples were fixed using modified protocols defined by a previous study examining cell wall ultrastructure of S. pneumoniae (Hammerschmidt, S. et al. 2005. Infect Immun 73:4653-4667). A lysine acetate-based formaldehyde-glutaraldehyde ruthenium red- osmium fixation procedure involved fixing the bacterial pellets with a cacodylate buffer solution containing 2% formaldehyde, 2.5% glutaraldehyde, 0.075% ruthenium red and 0.075 M of lysine acetate for 1 h. After washing with cacodylate buffer containing 0.075% ruthenium red three times, a second fixation in cacodylate buffer solution containing 2% formaldehyde, 2.5% glutaraldehyde and 0.075% ruthenium red was undertaken for 1.5 h. Cells were subsequently washed three times with cacodylate buffer containing 0.075% ruthenium red and underwent a final fixation in 1 % osmium tetroxide in cacodylate containing 0.075% ruthenium red for 1 h. The samples were then washed three times in cacodylate buffer containing 0.075% ruthenium red only. 100284] Samples were washed and dehydrated using a graded series of ethanol (70, 90, 95 and 100%) for 10-20 min, two times for each step. Samples were infiltrated using 50:50 LR White resin in 100% ethanol for 1 h, and subsequently washed with 100% LR White resin for 1 h and left O/N in a third change of 100% LR white to ensure adequate infiltration of resin. The samples were then embedded in fresh LR White resin and incubated at 50 °C for 48 h. Sections were cut to 1 μιτι using a glass knife, stained with Toluidene Blue and viewed under a light microcrope at 400* to identify the presence of stained pneumococci. At least four ultra-thin sections were then cut to 90 nm using a diamond knife and placed on matrix grids, one section per grid. Ultra-thin sections were then stained with uranyl acetate and lead citrate alternatively at 5 min intervals, followed by three washes with distilled water in-between each exposure. Stained sections were then placed on grids and viewed between 25000* and 130000* on a Philips CM100 Transmission Electron Microscope. Images were obtained at 130000* magnification and analysed using analysis [Olympus Soft Imaging Systems].

Statistical analysis

[00285] Statistical analyses were conducted using statistics program GraphPad Prism (5th ed, GraphPad Software Inc.) for Windows. For growth curves, data presented were the mean and standard error of mean (SEM) (represented as error bars) for each data point except for macro-broth dilution studies where multiple replicates could not be obtained due to the high costs involved in this assay. Two tailed, unpaired f-tests were performed.

Results

Pharmacodynamics of NCL812 in S. pneumoniae

Quality control disk diffusion analysis for 20 S. pneumoniae isolates

[00286} Although nine out of the twelve antimicrobials used for disk diffusion analysis had established QC ranges by EUCAST, QC ranges were not defined for amoxicillin-clavulanate, clarithromycin and clindamycin (Table 18 and Table 19). WCH 16 and WCH184 were both resistant to at least two antimicrobials whereas EF3030 and WCH 137 were intermediate and resistant to trimethoprim-sulphamethoxazole respectively (Table 19). The other remaining sixteen isolates were sensitive to all twelve antimicrobials. Sensitivity to ampicillin was confirmed for each isolate, enabling the use of ampicillin as a positive control in later micro- broth dilution assays (Table 18), Table 18: Artti bacteria! susceptibility of 20 S. pneumoniae isolates for six different antibacterials according to Example 5.

Blue ceils represent senstitive isolates; orange cells represent intermediate (i) isolates; green cells represent resistant isolates.

Table 19: Antibacterial susceptibiiity of 20 S. pneumoniae isolates for six different antibacterials according to exampte 5

Blu (R) isolates

Solubility and activity of NCL812 and NCL062 in different media

[00287] NCL812 visually appeared to have higher solubility in 100% DMSO compared to NCL062 and only developed turbidity when it was further diluted into CAMHB or PBS {Table 20). Although a CAMHB diluent for NCL062 appeared to be transparent by visual inspection (Table 20), further studies on NCL062 with a CAMHB diluent resulted in complete confluence in microbroth dilution assays for six S. pneumoniae isolates in comparison to growth with the DMSO diluents (Table 21 and Table 22).

Table 20: Visual analysis of NCL812 and NCL062 and ampicillin solubility according to Example 5.

Table 21 : Individual MICs of NCL062 for each pneumococcal isolate according to Example 5

Table 22: Difference in activity of NCL812 and NCL062 in different media using micro-broth dilution to obtain an MIC as a predictor according to Example 5.

NCL062 Ampicillin Isolate Resistance status to Ampicillin MIC MBC MIC MBC

(Mg/ml) (Mg/ml) ( g/ml) (Mg/ml)

*

D39 Sensitive Confluent * 0.0225

* *

A66.1 Sensitive Confluent 0.045

WU-2 Sensitive Confluent * 0.0225 *

P21 *

Sensitive Confluent 0.0225 0.045

WCH158 Sensitive Confluent 64 0.0225 0.022

WCH57 Sensitive Confluent *

0.09 0.09

[00288] Growth of S. pneumoniae strain D39 in an MIC assay for NCL812 and NCL062 using 10% HSB {220 mL of horse serum is filtered to 10% in 180 mL of Lemco nutrient broth) resulted in a threefold increase in the MIC for D39 treated with NCL812 and NCL062 {Table 23) with a twofold increase for the positive ampicillin control. There was no notable change in MIC for D39 with differing storage conditions of pre-prepared 96 well microtitre trays (Table 24). During macro-broth dilutions, the pH of the media did not change compared to appropriate controls (Figure 12.

Table 23: Growth of S. pneumoniae strain D39 in an MIC assay for NCL812 and NCL062 using horse serum supplemented broth

[00289] Table 24: Storage of prepared micro-titer trays for micro-broth dilution does not change MIC of D39 according to Example 5.

Determination of S. pneumoniae in vitro susceptibility to NCL812 and NCL062 Determination of NCL812 and NCL062 MIC 50 , MIC 90 , MIC range

[00290] NCL812 exhibited a MIC 50 and MIC 90 MIC 90 of 8 Mg/mL and MIC range of 4-8 Mg/mL whereas for NCL062 these values were higher and more variable (Table 25 and Table 26). The MIC for ampicillin was comparable to recent published findings using micro-broth dilution as an endpoint for antimicrobial resistance in pneumococcal isolates, thus confirming the accuracy of MICs obtained for NCL812 and NCL062 (Tables 25 to 26 and Figure 13).

Table 25: MIC 50 , MIC 90 , MBC 50 , MBC 90 and MIC range for all isolates treated with NCL812, NCL062, and ampicillin according to Example 5.

Table 26: MICs of NCL812 of each pneumococcal isolate according to Example 5.

Determination of NCL812 and NCL062 MBC 50 , MBC 90 , MBC range

[00291] Minimum bactericidal concentrations (MBC 50 , MBC 90 and MBC range respectively) were determined for NCL812 and ampicillin for all twenty isolates (Tables 25 to Table 26). The MBC 50 , MBC 90 , and MBC range was lower and more consistent for NCL812 compared with NCL062 (Table 25). Micro-broth dilution time kill studies of D39 treated with NCL812 and NCL062

I00292] D39 exposed to sub-inhibitory concentrations (< 2 pg/ml) of NCL812 or NCL062 grew similarly to unexposed controls over a 48 h period (Figure 13 and 14. Higher concentrations of NCL812 and NCL062 (> 16 pg/mL) resulted in no bacterial growth for 48 h (Figure 14 and 15. These growth characteristics were validated by a micro-broth kill kinetic study using a Spectramax spectrophotometer, which measured growth (represented as OD 60 o) at half-hourly intervals for 14 h for NCL812, NCL062 and ampicillin (Figures 15 to 17. There was an approximate six hour difference between the commencement of exponential growth for D39 treated with NCL812 and D39 treated with NCL062 (Figures 13, 14, 18 and 19).

[00293] The growth of D39 treated with NCL812 or NCL062 was compared to D39 treated with ampicillin or erythromycin over 48 h (Figures 20 and 21). D39 treated with ampicillin exhibited similar growth to D39 exposed to NCL812 or NCL062 over 48 h (Figure 20). Erythromycin-treated D39 produced very different growth curves from NCL812 and NCL062 where a larger difference in growth between concentrations was observed (Figure 21). The addition of 5% choline chloride to the media over a 48 h period resulted in no significant difference in growth for NCL812 and NCL062 compared to positive and growth controls (Figures 22 to 26).

Point of resistance testing

[00294] D39 treated with≤4 pg/mL NCL812 entered a log phase of growth at 6 h (Figure 13 and 18), as shown in four independent experiments. The possibility of antimicrobial resistance to NCL812 between 5 and 6 h was investigated by determining further MlCs on D39 exposed to 2 pg/mL NCL8 2, 4 pg/mL NCL8 2 and 0.0225 pg/mL ampicillin for 6 h. Results showed no significant increase in MIC for all samples of D39 exposed to NCL812 compared to growth controls, and ampicillin (Table 27).

Table 27: MICs of D39 exposed to 2 pg/mL or 4 pg/mL of NCL812 for 6 h according to Example 5.

D39 growth 2 ** | 8 Mg.mL "1 j 8 Mg.mL " 8 Mg.mL 8 g.mL "1

*D39 growth control: S. pneumoniae strain D39 grown for 6 hrs in 4 LHB:CAMHB.

** D39 growth2 control: S. pneumoniae strain D39 on HBA O/N, resuspended in saline

(0.1 OD 600 ) and diluted 1/20 in sterile saline.

Micro-broth dilutions by measuring relative MBC at specific time points

[00295] Relative MBCs were determined at specific time intervals from using broth dilution assays incubated for 48 h for NCL812 and NCL062 (Figure 27) and control antimicrobials ampicillin and erythromycin (Figures 20 and 21). MICs of ampicillin and erythromycin for D39 were determined (Tables 26 and 28). The comparative features of the growth of ampicillin, and erythromycin are described (Figures 28 and 29). Ampicillin and erythromycin demonstrated a time-dependent reduction in bacteria. NCL062 exhibited rapid bactericidal action, with an immediate (within the first 10 min of administration) MBC of 8 μg/mL (Figure 27). Although there were inconsistencies in the MBCs for NCL062 between 5 and 12 h, NCL062 maintained a constant bactericidal concentration (4 μg/mL) between 24 and 48 h. NCL812 exhibited fast bactericidal action, evidenced by an approximate 3 fold decrease in MBC within 5 h (Figure 27). A consistent bactericidal concentration (8 μ9/ηιΙ_) was maintained for the full 48 h for NCL812.

I00296] Table 28: MIC and MBC for erythromycin with D39 according to Example 5.

Macro-broth dilution time kill studies of D39 with NCL812 and NCL062

[00297] Viable counts for each time point were represented as a iog 0 CFU/mL reduction for NCL812 (Figure 30) and ampicillin (Figure 31 ). Consistent confluent growth (determined by a limit of 2x 104 CFU) was observed for unexposed controls and 2 \ig/mL NCL812. Complete bactericidal activity (defined by a 3log 10 reduction in CFU) for 128 μ9/ιτιΐ_ of NCL812 was observed by a 4log 10 reduction of CFU in 3 h and concentrations between 16 μg/mL and 64 μg/mL NCL812 were effective at eliminating bacterial growth within 8 h (Figure 30). NCL812 at 4 μg/mL and 8 μ9/ηηΙ_ appeared to be inactivated at 1 1 h post-exposure, as increased growth of strain D39 after this time point was observed (Figure 20 and 31 ). The viable counts of strain D39 treated with ampicillin demonstrated consistency for this particular assay by showing a constant diminished time-dependant killing over 48 h (Figure 31 ).

Transmission electron microscopy [00298] Morphometric analysis revealed significant changes to the cell membrane in strain D39 exposed to 16 pg/mL NCL812 for 6 h compared to growth controls. Samples treated with 4 g/mL as well as 12 h cultures were not considered for morphometric analysis due to the lack of bacterial cells available in each section. Treated samples possessed significantly thicker cell membranes (6.43 ± 0.29 nm) compared to untreated samples (4.35 ± 0.24 nm) (pO.0001 ) (Figures 32 and 29). The periplasmic space (intracellular space between the cell membrane and the cell wall) of D39 treated with 16 μg/mL NCL812 was significantly wider (4.54 ±0.096 nm) compared to untreated samples (3.91 ± 0.14 nm) (p<0.001 ) (Figures 29 and 33).

[00299] Table 29: Morphometric studies on the ultra structures of D39 treated with NCL812 for 6 hours according to Example 5.

[00300] In summary, NCL812 produced highly consistent MICs and equivalent MBCs for the S. pneumoniae strain collection, confirming that it is bactericidal against this organism. In kill kinetics experiments, which measured the relative MBC over a 48 h period, a consistent bactericidal effect was elicited in D39 after 6 h from initial exposure to NCL812.

[00301] This demonstration of bactericidal activity is the first to be observed in S. pneumoniae. This demonstrates that NCL812 is effective against pneumococcal in vitro.

[00302] Competitive binding between components in blood, serum or broth decreased the antimicrobial activity of NCL. This was reflected in the increase of MIC observed between different broth types and diluents. Following the completion of these studies, recent independent research confirmed precipitation of NCL812 in PBS and reported complete solubility in water containing 4% DMSO, following initial dilution in 100% DMSO. A water-soluble NCL812 will greatly improve in vivo bioavailability and negative interaction between blood or serum proteins.

[00303] Based on the findings of this study, NCL812 exhibits a mechanism of action against S. pneumoniae that is different from β-lactam or macrolide classes, as it appears to exhibit concentration-dependent bactericidal activity as opposed to time-dependant qualities. Identifying the maximum pharmacokinetic serum concentration of NCL812 in vivo will assist confirmation of its concentration-dependant pharmacodynamic activity. Furthermore, the addition of choline chloride to the media confirmed that the mechanism of action for NCL is not associated with the affinity to cell wall choline binding proteins, and therefore may not be cell wall associated.

[00304] Morphometric analysis of the cell membrane and periplasmic space of D39 treated with 16 9/ιτιΙ_ NCL812 for 6 h showed that the cell membrane and periplasmic space was larger in treated samples, compared to control samples. The apparent increase in membrane size could be due to an accumulation of electron dense intracellular material beneath the cell membrane. The increase in the size of the periplasmic space may be have been due to disruption of the cell membrane, potentially by depolarisation or ATP inhibition. The mechanism of action of NCL812 may not be calcium-dependant as it appears that no competitive binding between NCL812 and ruthenium red, a calcium channel inhibitor of lipid bilayers, was observed in electron micrographs.

[00305] In conclusion, this in vitro study has demonstrated that NCL812 has many desirable characteristics as a fast-acting concentration-dependent bactericidal antimicrobial that appears to target the cell membrane of S. pneumoniae. These characteristics are desirable to treat acute pneumococcal infections. As NCL812 may possess a mechanism of action that targets the cell membrane, it will act much more quickly than time-dependent antimicrobials such as β-lactams and macrolides and potentially more effective than other bactericidal concentration-dependent antimicrobials such as fluoroquinolones which have intracellular targets.

EXAMPLE 6: Characterization of methicillin-susceptible and methicillin-resistant isolates of

Staphylococcus pseudintermedius from Australia and preliminary in vitro efficacy of a new anti-staphylococcal compound

Materials and Methods

Sample collection and identification of methicillin susceptible Staphylococcus pseudintermedius (MSSP) and methicillin resistant Staphylococcus pseudintermedius (MRSP)

[00306] A total of 23 Staphylococcus pseudintermedius isolates were obtained from dogs {Table 30). Table 30: Staphylococcus pseudintermedius isolates tested according to Example 6. cr

0

n i 5

[00307] Ten methicillin susceptible and 13 methicillin resistant Staphylococcus pseudintermedius were collected for the study. Isolates were phenotypically classified as methicillin resistant on the basis of in vitro resistance to oxacillin and genetically for the presence of mecA gene according to standard procedures.

I00308] Oxacillin and cefoxitin susceptibility testing using disk diffusion technique and Epsilometer testing were performed. Identification of mecA gene was performed using polymerase chain reaction (PCR)

[00309] CLSI disk diffusion susceptibility testing was performed on the 23 Sp. isolates for the following antimicrobials: penicillin, amoxicillin, erythromycin, gentamicin, clindamycin, ciprofloxacin, cephalexin, chloramphenicol, tetracycline, oxytetracycline, vancomycin, cefotetan, moxifloxacin and rifampin.

[00310] Minimum inhibitory concentration (MIC) and minimum bactericidal concentration (MBC) testing was undertaken using CLSI methodology for NCL812 and included ampicillin as a control. Anti-staphylococcal compounds were then tested against all 23 isolates and minimum inhibitory concentrations (MIC) were determined according to standard protocols. After the MICs were determined, the minimum bactericidal concentrations were performed to determine if these compounds are bacteriostatic or bacteriocidal.

Results

[0031 1 ] The mecA gene was present in 13 isolates of MRSP and negative in 10 MSSP {Tables 30 and 31). All MRSP isolates were resistant to oxacillin based on disc diffusion (<17 mm) and E-test MIC (>0.5 mg/L).

Table 31 : Staphylococcus pseudlntermedius isolates tested: according to Example 6.

[00312] When cefoxitin resistance breakpoint was set at≤24 mm, 3/13 (23%) and 5/13 (38%) of MRSP tested respectively were susceptible to cefoxitin. When cefoxitin resistance break point was set at <30 mm, only 1/13 (7.7%) of MRSP tested was susceptible (Tables 30 and 31).

[00313] The MRSP isolates were resistant to multiple antibiotic classes. Of the 13 MRSP isolates, all 13 were susceptible to rifampin. 3/13 (23%) were susceptible to chloramphenicol; 10/13 (77%) were susceptible to vancomycin (Tables 30 and 31).

[00314] Interestingly, 3/13 (23%) of the MRSP isolates were susceptible to amoxicillin; 8/13 (62%) were susceptible to cephalothin; 12/13 (92%) susceptible to cefotetan and 12/13 (92%) susceptibe to moxifloxacin (Tables 30 and 31).

[00315] All 23 isolates were susceptible to NCL812 based on MICs. In addition, NCL812 has been shown to be bactericidal based on minimal bactericidal concentrations (MBC).

[00316] The MIC range of NCL812 against the Staphylococcus pseudintermedius isolates was found to be between 1 Mg/mL and 4 pg/mL (Table 32). The MIC 50 and MIC 90 of NCL812 against the Staphylococcus pseudintermedius isolates was found to be 2 μg mL and 4 μg/mL respectively (Table 33). The MIC mode and MIC range of NCL812 against the Staphylococcus pseudintermedius isolates was found to be 2 μg/mL and 1 -4 μg/mL respectively (Table 33).

Table 32: MICs of NCL812 and ampicillin against Staphylococcus pseudintermedius isolates according to Example 6.

Table 33: MIC50, MIC90, the MIC mode, an MIC range of NCL812 against Staphylococcus pseudintermedius isolates according to Example 6.

[00317] Methicillin resistant Staphylococcus pseudintermedius (MRSP) is an emerging problem in dogs, cats and horses. Two major clonal MRSP lineages have been reported from dogs in Europe (ST 71 ) and North America (ST 68). There were also reports of MRSP affecting dogs in Japan and a single case of MRSP in veterinary personnel in Hong Kong.

[00318] In this study, MRSP isolates were determined using a combination of presence of mecA gene and in vitro resistance to oxacillin. Cefoxitin susceptibility has been used as a substitute for oxacillin for methicillin resistant Staphylococcus aureus. However, cefoxitin disk diffusion tests using interpretive guidelines recommended for human isolates of methicillin resistant Staphylococcus aureus and coagulase negative staphylococci are unreliable in identifying MRSP. A cefoxitin breakpoint resistance of ≤30 mm=resistant and≥31 =susceptible has been proposed by Bemis et al, 2012[Bemis, D. A. , R. D. Jones, et al. (2012). "Evaluation of cefoxitin disk diffusion breakpoint for detection of methicillin resistance in Staphylococcus pseudintermedius isolates from dogs." Journal of Veterinary Diagnostic Investigation 24(5): 964- 967]. This study is in agreement that this breakpoint may be more reliable in predicting methicillin resistant Staphylococcus pseudintermedius. [00319] MRSP isolates are generally resistant to multiple antibiotic classes. Bacterial culture and antibiotic susceptibilities are therefore recommended for all suspect MRSP infections to allow appropriate selection of antibiotics. A limitation noted in this study is the apparent in vitro susceptibility of MRSP isolates to amoxicillin and cephalosporins (cephalothin and cefotetan).

[00320] NCL812 was effective against all 23 isolates of both MSSP and MRSP. A larger scale study is warranted to confirm the effectiveness of NCL812 against Staphylococcus pseudintermedius as it may provide a safe alternative antibiotic option for emerging MRSP infections in domestic animals.

EXAMPLE 7: Preparation and testing of NCL812 analogues (also known as compounds of the invention).

Materials and Methods

NCL812

Analytical grade NCL812 with a defined potency of 960 mg/g (i.e. 96%) was obtained. The powder was stored in a sealed sample container out of direct sunlight and at room temperature at the study site. Aliquots (1 ml.) of stock solution (containing 25.6 mg/mL of NCL812 in DMSO) were prepared and stored at -80 °C and defrosted immediately before use.

Synthesising and Testing of NCL812 Analogues

[00321] Analogues NCL001 to NCL230, as identified in Figure 1 , were synthesised using standard methods in the art. As an example, the methods used to manufacture compounds NCL097; NCL157; NCL179; NCL188; NCL195; and NCL196 are as follows:

NCL 097 (2,2'-bis[(3,4,5-trihydroxyphenyl)methylene]carbonimidic dihydrazide hydrochloride)

[00322] A suspension of 3,4,5-trihydroxybenzaldehyde (412.0 mg, 2.673 mmol, 2.21 eq.) and /V,/V'-diaminoguanidine hydrochloride (152.0 mg, 1.21 1 mmol) in EtOH (5 ml_) was subjected to microwave irradiation (150 W) at 100 °C for 10 min. The reaction was then allowed to cool to ambient temperature. The resulting precipitate was collected and washed with chilled EtOH (5 ml_) and Et 2 0 (5 ml_) to afford the carbonimidicdihydrazide (369.0 mg, 77%) as a pale brown solid.M.P. 292°C (Decomp.). 1 H NMR (300 MHz, DMSO-d6) δ 9.06 (br s, 6H), 8.25 - 8.01 (m, 4H), 6.83 (s, 4H). 13 C NMR (75 MHz, DMSO-d6) δ 152.2, 149.7, 146.2, 136.5, 123.7, 107.4. LRMS(ESr): 361.95 [M + 1 ] + .

NCL157 (2,2'-bis[(2-amino-4-chlorophenyl)methylene]carbonimidic dihydrazide hydrochloride) 100323] Synthesis of 2-amino-4-chloro-N-methoxy-N-methylbenzamide. To a solution of 2-amino-4-chlorobenzoic acid (5.6691 g, 33.041 mmol), Λ/, Ο-dimethylhydroxylamine hydrochloride (5.7504 g, 58.954 mmol, 1.78 eq.), /S/-(3-dimethylaminopropyl)-/V'- ethylcarbodiimide hydrochloride (7.7925 g, 40.649 mmol, 1 .23 eq.) and /V-hydroxybenzotriazole hydrate (5.2371 g, 38.793 mmol (anhydrous basis), 1.17 eq.) in DMF (100 mL) was added diisopropylethyiamine (18.0 mL, 13.4 g, 104 mmol, 3.15 eq.) and the brown solution stirred at ambient temperature for 7 h. The reaction was then concentrated in vacuo before dilution with 1 M NaOH (100 mL) and extracting with CH 2 CI 2 (3 χ 100 mL) The combined organic extracts were washed with 1 M HCI (100 mL) before drying over MgS0 4 and concentrating in vacuo to afford a brown syrup. This oil was then further dried at 60 °C under high vacuum to afford the crude Weinreb amide (7.021 g, 99%) as a brown syrup that crystallised on standing. The crude material was used without further purification. 1 H NMR (400 MHz, CDCI 3 ) δ 7.24 (d, J = 8.4 Hz, 1 H), 6.62 (d, J = 18 Hz, 1 H), 6.54 (dd, J = 8.4, 1.9 Hz, 1 H), 4.75 (s, 2H), 3.48 (s, 3H), 3.24 (s, 3H). 13 C NMR (101 MHz, CDCI 3 ) δ 169.2, 148.4, 137.1 , 130.6, 1 16.6, 1 16.1 , 1 15.0, 61 .1 , 34.0.

[00324] Synthesis of 2-amino-4-chlorobenzaldehyde. Crude 2-amino-4-chloro-/V- methoxy-/ -methylbenzamide (751.1 mg, 3.532 mmol) was broken up into ca. 120 mg batches and each dissolved in THF (10 mL) and cooled to 0 °C before LiAIH 4 (2M in THF, 0.5 mL) was added to each and the solutions stirred for 16 h, allowing the reactions to achieve room temperature. The reactions were quenched with saturated NH 4 CI (1 mL) before being combined, diluted with saturated NaHC0 3 (160 mL) and extracted with CHCI 3 (2 x 150 mL, 1 χ 75 mL). The combined organics were dried over MgS0 4 and concentrated in vacuo to afford the crude benzaldehyde (463.3 mg, 85%) as yellow/orange crystals. The material was used without further purification. 1 H (400 MHz, CD 3 OD) 9.77 (d, J = 0.7 Hz, 1 H), 7.46 (d, J = 8.3 Hz, 1 H), 6.83 - 6.71 (m, 1 H), 6.63 (dd, J = 8.4, 1 .9 Hz, 1 H). 13 C NMR (101 MHz, CD 3 OD) δ 194.6, 153.0, 142.5, 138.4, 1 18.3, 1 16.8, 1 16.1 .

[00325] Synthesis of 2,2'-bis[(2-amino-4-chlorophenyl)methylene]carbonimidic dihydrazide hydrochloride. A suspension of 2- amino- 4- chlorobenzaldehyde (128.0 mg, 0.823 mmol, 1.78 eq.) and Λ/,Λ/'-diaminoguanidine hydrochloride (58.0 mg, 0.462 mmol) in EtOH (2 mL) was subjected to microwave irradiation (1 00 W) at 60°C for 5 minutes. Most solvent was then removed in vacuo, EtOH (1 mL) was added and the flask was transferred to the freezer to effect crystallisation. The resulting precipitate was collected and washed with EtOH (1 mL) to afford the carbonimidicdihydrazide (21 .0 mg, 13%) as a pale yellow solid. 1 H NMR (400 MHz, DMSO-ds) δ 1 1 .71 (br s, 2H), 8.40 (s, 2H), 8.37 (s, 2H), 7.29 (d, J = 8.4 Hz, 2H), 6.87 (d, J = 2.0 Hz, 2H), 6.73 (br s, 4H), 6.59 (dd, J = 8.3, 2.0 Hz, 2H). 13 C NMR (101 MHz, DMSO-d a ) δ 152.1 , 151.5, 148.9, 136.0, 134.7, 1 15.1 , 1 14.5, 1 12.8.

NCL179 (4 > 6-bis(2-((£)-4-chlorobenzylidene)hydrazinyl)pyrimidin -2-amine) [00326] A suspension of 2-amino-4,6-dihydrazinylpyrimidine (67.3 mg, 0.434 mmol) and 4-chlorobenzaldehyde (198.8 mg, 1 .414 mmol, 3.26 eq.) in EtOH (25 mL) was heated at reflux for 16 h. After this time, the condenser was removed and the solution concentrated to approx. 1 mL and the resulting precipitate filtered hot and washed with Et 2 0 (10 mL) to afford the aminopyrimidine (42.8 mg, 25%) as an off-white amorphous powder. M.P. 275 °C (Decomp.). H NMR (400 MHz, DMSO) δ 10.70 (s, 2H), 8.02 (s, 2H), 7.67 (d, J = 8.4 Hz, 4H), 7.52 (d, J = 8.4 Hz, 4H), 6.28 (s, 1 H), 5.85 (s, 2H). 13 C NMR (101 MHz, DMSO) δ 162.8, 162.6, 138.8, 134.1 , 133.1 , 128.9, 127.6, 73.5.

NCL188 ((E)-2-(1-(4-chlorophenyt)pentylidene)hydrazine-1 -carboximidamide hydrochloride)

[00327] A suspension of 1 -(4-chlorophenyl)pentanone (1.8319 g, 9.3146 mmol, 1.95 eq.) and aminoguanidine hydrochloride (527.6 mg, 4.773 mmol) in EtOH (15 mL) was heated at 65 °C for 16 h. The crude was cooled to ambient temperature before being diluted with Et 2 0 (60 mL) and cooled to 0 °C to precipitate unreacted aminoguanidine hydrochloride (174.5 mg). The mother liquors were then concentrated in vacuo and the residue dissolved in Et 2 0 (20 mL). The solution was then boiled and hexanes (10 mL) added to afford the caboximidamide as a cream solid. H NMR (400 MHz, DMSO) δ 1 1 .54 (s, 1 H), 7.99 (d, J = 8.7 Hz, 2H), 7.90 (s, 3H), 7.47 (d, J = 8.6 Hz, 2H), 2.91 - 2.82 (m, 2H), 1.48 - 1.32 (m, 4H), 0.89 - 0.84 (m, 3H). 3 C NMR (101 MHz, DMSO) δ 156.2, 153.8, 134.8, 134.4, 128.7, 128.4, 28.1 , 26.6, 22.0, 13.8

NCL195 (4,6-bis(2-((E)-4-methyibenzylidene)hydrazinyl)pyrimidin-2-a mine)

A suspension of 2-amino-4,6-dihydrazinopyrimidine (58.9 mg, 0.380 mmol) and 4- methylbenzaldehyde (0.10 mL, 100 mg, 0.832 mmol, 2.19 eq.) in EtOH (4 mL) was heated at reflux for 16 h. The reaction mixture was cooled to ambient temperature before collecting the pellet-like precipitate, washing with Et 2 0 (20 mL). The 'pellets' were then crushed and the solid further washed with Et 2 0 (10 mL) to afford the pyrimidine (85.8 mg, 63%) as a white 'fluffy' powder. M.P. 274-276 °C. 1 H NMR (400 MHz, DMSO) δ 10.51 (s, 2H), 8.00 (s, 2H), 7.54 (d, J = 8.0 Hz, 4H), 7.26 (d, J = 7.9 Hz, 4H), 6.26 (s, 1 H), 5.77 (s, 2H), 2.34 (s, 6H). 3 C NMR (101 MHz, DMSO) δ 162.8, 162.6, 140.1 , 138.4, 132.5, 129.4, 126.0, 73.3, 21 .0.

NCL196 (4,4'-((i£, rE)-((2-aminopyrimidine-4,6-diy!)bis(hydrazin-2-yl-1

ylidene))bis(methanylylidene))diphenol)

[00328] A suspension of 2-amino-4,6-dihydrazinopyrimidine (70.4 mg, 0.454 mmol) and 4-hydroxybenzaldehyde (140.3 mg, 1 .149 mmol, 2.53 eq.) in EtOH (3 mL) was heated at reflux for 16 h. The reaction mixture was cooled to ambient temperature before collecting the precipitate, washing with Et 2 0 (25 mL), to afford the pyrimidine (91 .4 mg, 55%) as an off-white powder. M.P. 298 °C (Decomp.). 1 H NMR (400 MHz, DMSO) δ 10.31 (s, 2H), 9.74 (s, 2H), 7.94 (s, 2H), 7.48 (d, J = 8.6 Hz, 4H), 6.83 (d, J = 8.6 Hz, 4H), 6.20 (s, 1 H), 5.70 (s, 2H). 13 C (101 MHz, DMSO) 6 162.7, 162.5, 158.3, 140.5, 127.7, 126.3, 1 15.7, 73.0.

MIC tests

[00329] Minimum inhibitory concentrations ( g/ml) were determined using the broth microdilution method recommended by the Clinical and Laboratory Standards Institute (CLSI). MIC breakpoints were determined by visual assessment and then confirmed using an ELISA plate reader, measuring absorbance levels at 600 nm. Bacterial growth (turbidity) in the wells with antimicrobial was compared with the amount of growth (turbidity) in the growth-control well (containing no antimicrobiai). All isolates were tested in duplicate, if there was a difference of greater than one two-fold dilution in the results, the test was repeated a third time. The purity of the isolates was closely monitored during testing by subculturing the prepared bacterial inoculum onto SBA (sheep blood agar). The MICs of the control strains for the antimicrobial ampicillin were determined for each testing run as an internal quality control. The MIC 50 , MIC 90 and MIC range (minimum and maximum) were calculated for each of the bacterial groups.

Activity of NCL812 and MIC against Gram-negative bacteria

[00330] The activity of NCL812 against Gram-negative bacteria was assessed using the broth microdilution method recommended by the Clinical and Laboratory Standards Institute (CLSI), and the MICs (pg/mL) for NCL812 and ampicillin were determined.

Determination of Minimum Bactericidal Concentration (MBC)

CLSI methodology

[00331 ] Briefly, 10 of the contents of each well starting at the MIC was inoculated on to a Columbia SBA plate and incubated at 37 °C for 48 h. Plates were examined at 24 and 48 h and the MBC was recorded as the lowest concentration of NCL812 at which no colonies of bacteria were observed on the plate (or significant inhibition of growth was observed compared to the control) (CLSI 2005).

Kill kinetics assays (MRSA & VRE)

[00332] MRSA VRE were grown overnight on Columbia SBA at 37 °C. A few colonies of bacteria were then suspended in CAMHB and adjusted to an optical density of 0.08 to 0.10. The bacterial suspension was diluted 1 : 10. One millilitre of the bacteria was added to 9 mL of CAMHB containing various concentrations (up to 4 x MIC) of NCL812, to achieve a final bacterial concentration of 1 to 3x10 e CFU/ml. The tubes were incubated at 37 °C. In order to determine the number of viable bacteria present at various time points, a 100 aliquot was removed from each tube and diluted in normal saline. Then, 100 L of each dilution was spread onto colony count agar, in duplicate, and incubated for 48 h at 37 °C. After 24 h the numbers of colonies present on each plate were counted and therefore the number of viable bacteria present in the original suspension enumerated. Plates were re-checked after 48 hours.

Synergy studies with other classes of antimicrobial agent.

[00333] The checkerboard method (Gunics et al., 2000 Int. J. Antimicrob. Agents. 14:239- 42) was used to find interactions (synergy, antagonism, no effect) of NCL812 in combination with tetracycline, chloramphenicol, erythromycin (macrolide), ampicillin (β-lactam broad- spectrum), gentamicin (aminoglycoside), ciprofloxacin (fluoroquinolone), sulfamethoxazole (sulphonamide), or penicillin G (β-lactam narrow-spectrum). For initial experiments, a laboratory strain of Staphylococcus aureus T3-129 was used, however this strain gave inconsistent results for some of the antimicrobials and a new strain of Staphylococcus spp. designated MK1 (definitive species identification currently in progress) that was sensitive to all tested antimicrobials was used in subsequent tests.

100334] Firstly, the MIC of each antibiotic alone was determined in accordance to CLSI standard guidelines. Secondly, the combination of NCL812 with each of above antibiotics was tested in duplicate. To evaluate the effect of the combination the fractional inhibitory concentration (FIC) was calculated for each antibiotic as follows:

FIC of tested antibiotic = MIC of tested antibiotic in combination / MIC of antibiotic alone.

FIC of NCL812 = MIC of NCL812 in combination / MIC of NCL812 alone.

FIC, = FIC index = FIC of NCL812 + FIC of each tested antibiotic.

100335] According to the checkerboard guidelines, Synergy (S) was defined as an FIC|<0.5. No effect (NE) was defined as 0.5<FIC|<4. Antagonism (A) was defined as a 4<FIC[.

Testing of NCL812 Analogues

[00336] NCL812 analogues were stored at 4 °C until assayed. MICs were determined against two MRSA strains, two VRE strains and one strain each of E. co// ' and Pseudomonas aeruginosa.

Results

Determination of Minimum inhibitory Concentration (MIC)

[00337] The comparative NCL812 and ampicillin MIC values (Mg/mL) for 21 MRSA isolates were obtained. The results for the original experiments (phase I), and repeat testing (phase II) are shown in Table 34. Each MIC test was performed in duplicate. Table 34: NCL8 2 and ampicil!ln I.C values (pg/ml) for 21 MRSA isolates obtained according to Example 7,

Table 35: Comparative NCL812 arid ampiciilin MIC values (pg/ml) for 13 VRE isolates obtained according to Example 7.

100338] The comparative NCL812 and ampicillin MIC values (Mg/ml) for 13 VRE isolates were obtained. The results for the original experiments (phase I), and repeat testing (phase I I) are shown in Table 35. Each MIC test was performed in duplicate.

[00339] NCL812 MIC 50 , MIC 90 , MIC mode and MIC range were obtained for Australian isolates of MRSA and VRE, as shown in Table 6 Comparative MIC values for ampicillin are shown in parentheses.

Activity of NCL812 and MIC against Gram-negative bacteria

[00340] Comparative NCL812 and ampicillin MIC values ( g/mL) for Escherichia coli, Pseudomonas aeruginosa and Salmonella arizonae were obtained, as shown in Table 36. Each MIC test was performed in duplicate.

Table 36: Comparative NCL812 and ampicillin MIC values (Mg/ml) for Escherichia coli, Pseudomonas aeruginosa and Salmonella arizonae obtained according to Example 7.

[00341 ] The antimicrobial activity of NCL812 against the selected Gram-negative bacteria was >128 Mg/ml.

Determination of Minimum Bactericidal Concentration (MBC)

[00342] MBC results for MRSA isolates are shown in Table 37 which shows NCL812 MBC values (Mg/mL) for 20 MRSA isolates. Each MBC test was performed in duplicate starting from the NCL812 MIC concentration to 16 times the MIC. For all isolates, the MBC was equal to the MIC. However, inconsistent growth on agar plates was recorded for some concentrations.

Table 37: NCL812 MBC values (Mg/ml) for 20 MRSA isolates according to Example 7.

GB = Bacterial Growth on Sheep Blood Agar

N** = Not cultured on Sheep Blood Agar

[00343] The results for 10 VRE isolates are shown in Table 38, Each MBC test was performed in duplicate starting from NCL812 MIC concentration to 32 times the MIC. As with the MRSA isolates tested, the MBC appears to be equal to the MIC. However, with the VRE isolates, an anomaly was observed at higher NCL812 concentrations. There is significant inhibition of growth at concentrations near to the M!C, but as the NCL812 concentration increases, bacteria appear to be less subject to inhibition. High numbers of bacteria were observed on the plates at NCL812 concentrations≥16 μg/mL.

Table 38: NCL812 MBC values (Mg/ml) for 10 VRE isolates according to Example 7

*Number of bacteria growing after 24 hours per ml of sample (CFU/ml); M = many bacteria growing on the plate (too many to count)

I00344] NCL812 was found to be bactericidal against Gram-positive bacteria at concentrations equivalent to the MIC.

Kill kinetics assays (MRSA & VRE)

[00345] In preliminary experiments, colony counts were performed at t = 15, 30, 45 and 60 min. No significant changes in the bacterial concentration were observed at these time points, suggesting that NCL812 is not rapidly bactericidal {by comparison, the lipoglycopeptide oritavancin (McKay et al. (2009) J. Antimicrob. Chemother. 63 (6): 1 191 -1 199) caused a 3log 10 reduction in viable count within an hour of exposure to a concentration equivalent to the C mgx ). Therefore, for future experiments, sampling time points were extended out to one and then two hour intervals.

100346] In the initial experiments, for MRSA, at 4 h a reduction of at least 2.5log 10 CFU/mL was observed in comparison to the growth control. At 8 h there was at least a 3.5log 10 CFU/mL difference between the control and the bacteria exposed to NCL812. After 24 h the numbers of bacteria present in all NCL812 concentrations was not significantly different to the control. There was a consistent reduction in bacterial numbers at NCL812 concentrations from 4-16 μg mL, up to 8 h, but the same was not observed for concentrations greater than 16 μg mL. By comparison, most bactericidal antimicrobial agents, used or being developed for the treatment of MRSA and VRE (oritavancin, daptomycin, vancomycin) are rapidly bactericidal achieving similar log reductions within 1 h of exposure in a concentration dependent manner (McKay et al., 2009). In kill kinetics experiments bacteriostatic antimicrobials recommended for the treatment of MRSA and VRE infections (teicoplanin and linezolid only marginally decrease the viable count and growth).

[00347] For VRE the observed decrease in the CFU/mL of bacteria exposed to NCL812 was less than for MRSA. At 4 h there was approximately a 2log 0 reduction in the viable count compared to the control, and at 8 h there was approximately a 2.5log 10 reduction. However, at 24 h the growth of bacteria was no longer as significantly reduced in comparison to the control. Bacterial numbers increased after 8 h incubation and this affect appeared to be more pronounced with increasing concentrations of NCL812.

[00348] The kill kinetics of MRSA 580 were obtained at different concentrations of NCL812 over a period of 8 h, as shown in Figure 34 The kill kinetics of MRSA 580 in different concentrations of NCL812 over a period of 24 h are shown in Figure 35 After 4 h of incubation the media was changed to fresh media containing the same concentration of NCL812.

[00349] The kill kinetics of MRSA 698 in different concentrations of NCL812 over a period of 24 h is shown in Figure 36 After 4 h of incubation the media was changed to fresh media containing the same concentration of NCL812.

[00350] The kili kinetics of VRE 26c(dc) at different concentrations of NCL812 over a period of 24 h are shown in Figure 37.

[00351] The kill kinetics of VRE 16c(dc) at different concentrations of NCL812 over a period of 24 h are shown in Figure 38.

Test for bacterial resistance to NCL812

[00352] Preliminary tests were carried out to determine whether bacterial resistance may account for the observations of bacterial growth at higher concentrations of NCL812 and the increase in bacterial numbers in kill kinetics experiments at 24 h incubation. Bacteria (MRSA) growing at high concentrations of NCL812, in the 96 well micro titre tray, were subcultured on to SBA and incubated for 24 h, then MIC testing was performed. There was no change in the MIC of these bacteria. Bacteria growing in broth used for kill kinetics experiments was also tested for any change in the MIC. No change was observed.

[00353] Additionally, bacteria exposed to NCL812 at high concentrations were then subcultured on to plate count agar containing NCL812 (64 μg/mL and 128 μg/mL) and incubated for 24 h at 37 °C. Bacteria growing on the plate were then used for running an MIC test. There was still no change in the MIC of the bacteria.

[00354] In summary, NCL812 has bactericidal activity against MRSA which is less pronounced against VRE strains. The bactericidal effect is not rapid in comparison to bactericidal antimicrobials developed for MRSA and VRE infections (daptomycin, oratovancin, vancomycin). Aberrant bactericidal results at higher concentrations of NCL812 are not indicative of resistance development, but may be suggestive of loss of activity. Stability testing of the compound in broth media should therefore be undertaken before exploring these interesting but currently unexplained results. This will include a detailed examination of the literature to determine if this phenomenon is observed in other classes of antimicrobial agent. In this case, closer examination of the kill kinetics between 8 and 24 h will be required. NCL812 kill curves for MRSA and VRE suggest more bactericidal activity in comparison to bacteriostatic antimicrobials (linezolid, teicoplanin). Kill curves should now be generated for Streptococcus pneumoniae once stability issues of NCL812 are investigated, as for example with linezolid, as some antibacterial agents can be bacteriostatic against some bacteria and bactericidal against others.

Synergy studies with other classes of antimicrobial agent.

[00355] MICs, FICs, FI and the interaction between NCL812 and eight antibiotics is shown in Table 39. None of the eight tested compounds, representing distinct classes of antimicrobial agent showed either positive (synergism) or negative (antagonism) interaction with NCL812 consistent with an additive effect when antibacterial agents are added to NCL812.

Table 39: MICs, FICs, FIC, and the interaction between NCL812 and eight antibiotics according to Example 7.

0.062 0.125 0.5 2 4 0.5 1 NE

1 st 0.062 0.125 0.5 2 4 0.5 1 NE 2 2 nd 0.031 0.25 0.125 2 4 0.5 0.625 NE

1 S. aureus strain T3-29

2 Staphylococcus spp. Strain MK1

FIC ! = MIC of anitbiotic in combination with NCL812/MIC of antibiotic alone

FIC 2 = MIC of NCL812 in combination with antibiotic/MIC of NCL812 alone

FIC, = FIC index

Testing of NCL812 Analogues

[00356] The chemical structures of analogues NCL001 to NCL230 are shown in Figure 1.

[00357] M lCs for NCL812 and analogues NCL001-070 are shown in Table 40. [00358] MlCs for analogues NCL071 to 171 are shown in Table 41.

[00359] M lCs for analogues NCL171 to 230 are shown in Table 42.

Table 40: MiCs for NCL812 and analogues NCL001-070 according to Example 7.

5*3 C 5

o cr

P t 5

re

Substitute Sheet (Rule 26) RO/AU

Substitute Sheet (Rule 26) RO/AU

Subsiitirte Sheet (Rule 26) RO/AU Table 41 : MICs for analogues NCL071-170 according to Example 7.

MIC ( g/ml) at 24 hours

NCL Compound Code

RSA 1 RSA 2 VRE 1 VRE 2 E. coli P. aeruginosa

>128 > 128 >128 >128 >128 >128

NCL071

>128 > 128 >128 >128 >128 >128

32 64 64 64 >128 >128

NCL072

32 64 64 64 >128 >128

128 128 16 64 >128 >128

NCL073

128 128 16 64 >128 >128

16 16 2 8 >128 >128

NCL074

16 16 1 4 >128 >128

> 128 >128 > 28 >128 >128 >128

NCL075

> 128 >128 >128 >128 >128 >128

> 128 >128 >128 >128 >128 >128

NCL076

> 128 >128 >128 >128 >128 >128

> 128 >128 >128 >128 >128 >128

NCL077

>128 >128 >128 >128 >128 >128

4 16 4 4 >128 >128

NCL078

4 8 2 2 >128 >128

4 4 2 4 >128 >128

NCL079

4 8 2 4 >128 >128

4 4 2 4 >128 >128

NCL080

4 8 2 4 >128 >128

4 8 1 4 >128 >128

NCL081

4 4 1 4 >128 >128

8 8 4 2 > 128 >128

NCL082

4 8 4 4 >128 >128

> 128 >128 >128 > 128 >128 >128

NCL083

> 128 >128 >128 >128 >128 >128

2 8 2 2 >128 >128

NCL084

2 4 2 2 >128 >128

>128 >128 >128 >128 >128 >128

NCL085

>128 >128 >128 >128 >128 >128

>128 >128 >128 128 >128 >128

NCL086

> 128 >128 >128 128 >128 >128

> 128 >128 >128 >128 >128 >128

NCL087

> 128 >128 >128 >128 >128 >128

16 8 4 4 >128 >128

NCL088

8 8 4 4 >128 >128

4 4 0.5 1 >128 >128

NCL089

8 4 0.5 1 >128 >128

>128 >128 >128 >128 >128 >128

NCL090

>128 >128 >128 >128 >128 >128

> 128 >128 >128 >128 >128 >128

NCL091

> 128 >128 >128 > 128 >128 >128

>128 >128 >128 >128 >128 >128

NCL092

>128 > 128 >128 >128 >128 >128

64 128 64 >128 >128 >128

NCL093

64 128 64 >128 >128 >128

128 128 64 128 >128 >128

NCL094

128 128 64 128 >128 >128

>128 >128 >128 >128 >128 >128

NCL095

>128 >128 >128 >128 >128 >128

>128 >128 >128 > 128 >128 >128

NCL096

>128 >128 >128 >128 >128 >128

128 32 32 32 >128 128

NCL097

128 32 32 32 >128 >128

> 128 >128 >128 >128 >128 >128

NCL098

> 128 >128 >128 > 128 >128 >128

8 4 8 8 > 128 >128

NCL099

8 4 8 8 >128 > 128

NCL100 I >128 >128 >128 >128 >128 > 128 >128 >128 >128 >128 >128 >128

32 32 64 128 >128 >128

N L1U1

32 32 64 128 >128 >128

>128 >128 >128 >128 >128 >128

NCL102

>128 >128 >128 >128 >128 >128

>128 >128 >128 >128 >128 >128

NCL103

>128 >128 >128 >128 >128 >128

64 64 128 >128 >128 >128

NCL104

64 64 128 >128 >128 >128

64 >128 64 >128 >128 >128

NCL105

64 >128 64 >128 >128 >128

>128 >128 >128 >128 >128 >128

NCL106

>128 >128 >128 >128 >128 >128

8 16 32 128 >128 >128

NCL107

8 16 32 128 >128 >128

64 64 64 128 >128 >128

NCL108

64 64 64 128 >128 >128

>128 >128 >128 >128 >128 >128

NCL109

>128 >128 >128 >128 >128 >128

>128 >128 >128 >128 >128 >128

NCL110

>128 >128 >128 >128 >128 >128

64 64 64 >128 >128 >128

NCL111

64 64 64 >128 >128 >128

>128 128 64 32 >128 >128

NCL112

>128 128 64 32 >128 >128

16 8 8 16 >128 >128

NCL113

16 8 8 16 >128 >128

>128 >128 >128 >128 >128 >128

NCL114

>128 >128 >128 >128 >128 >128

>128 >128 >128 >128 >128 >128

NCL 115

>128 >128 >128 >128 >128 >128

>128 >128 >128 >128 >128 >128

NCL 116

>128 >128 >128 >128 >128 >128

64 128 64 64 >128 >128

NCL 117

64 128 64 64 >128 >128

>128 >128 >128 >128 >128 >128

NCL 118

>128 >128 >128 >128 >128 >128

>128 >128 >128 >128 >128 >128

NCL 119

>128 >128 >128 >128 >128 >128

8 4 2 8 >128 >128

NCL 120

8 8 2 8 >128 >128

32 32 8 32 >128 >128

NCL 121

32 32 8 64 >128 >128

>128 >128 >128 >128 >128 >128

NCL 122

>128 >128 >128 >128 >128 >128

16 4 4 4 >128 >128

NCL123

32 8 4 4 >128 >128

>128 >128 >128 >128 >128 >128

NCL 124

>128 >128 >128 >128 >128 >128

>128 >128 >128 >128 >128 >128

NCL 125

>128 >128 >128 >128 >128 >128

>128 >128 4 >128 >128 >128

NCL 126

>128 >128 8 >128 >128 >128

>128 >128 >128 >128 >128 >128

NCL 127

>128 >128 >128 >128 >128 >128

>128 >128 >128 >128 >128 >128

NCL 128

>128 >128 >128 >128 >128 >128

>128 >128 >128 >128 >128 >128

NCL 129

>128 >128 >128 >128 >128 >128

>128 >128 >128 >128 >128 >128

NCL 130

>128 >128 >128 >128 >128 >128

>128 >128 4 >128 >128 >128

NCL 131

>128 >128 4 >128 >128 >128

NCL 132 >128 >128 >128 >128 >128 >128 >128 >128 > 128 >128 > 128 >128 All >128 >128 > 128 >128 >128 >128

>128 >128 >128 > 128 >128 >128 Λ ΊΛ >128 >128 >128 >128 >128 >128

> 128 >128 >128 >128 >128 >128

>128 >128 >128 >128 >128 > 128

NCL 135

>128 >128 >128 >128 >128 > 128

64 128 32 128 >128 >128

NCL 136

64 128 32 128 >128 >128

>128 >128 > 128 > 128 >128 >128

NCL 137

>128 >128 > 128 >128 >128 >128

>128 32 >128 >128 >128 >128

NCL 138

>128 32 >128 >128 >128 >128

>128 >128 >128 >128 >128 >128

NCL139

>128 >128 >128 >128 >128 >128

>128 16 2 8 >128 >128

NCL 140

>128 32 2 8 >128 > 128

>128 >128 16 >128 > 128 > 128

NCL 141

>128 >128 16 >128 >128 >128

>128 >128 >128 >128 >128 >128

NCL 142

>128 >128 >128 >128 >128 >128

16 16 8 8 >128 >128

NCL 143

16 16 8 8 >128 >128

>128 >128 >128 >128 >128 >128

NCL 144

>128 >128 >128 >128 >128 >128

>128 >128 >128 >128 > 128 > 128

NCL 145

>128 >128 >128 >128 >128 >128

8 16 16 16 >128 >128

NCL 146

8 16 16 16 >128 >128

>128 >128 >128 >128 >128 >128

NCL 147

>128 >128 >128 >128 >128 >128

>128 >128 >128 >128 >128 >128

NCL 148

>128 >128 >128 >128 >128 >128

>128 >128 > 128 >128 >128 >128

NCL 149

>128 >128 >128 >128 >128 > 128

128 128 >128 >128 >128 >128

NCL 150

128 128 >128 >128 >128 >128

64 64 128 64 >128 >128

NCL 151

64 64 128 64 >128 >128

>128 >128 >128 >128 > 128 >128

NCL 152

> 128 >128 >128 >128 >128 > 128

32 16 8 8 >128 >128

NCL 153

32 16 8 8 >128 >128

>128 >128 >128 >128 >128 >128

NCL 154

>128 >128 >128 >128 >128 >128

8 8 16 8 >128 >128

NCL 155

8 8 16 8 >128 >128

>128 >128 >128 > 128 >128 > 128

NCL 156

>128 >128 >128 >128 >128 >128

2 2 2 2 >128 >128

NCL 157

2 2 2 4 >128 >128

8 16 16 16 > 128 >128

NCL 140

8 16 16 16 >128 >128

8 8 8 8 >128 >128

NCL 038

8 8 8 8 >128 >128

4 4 4 4 >128 >128

NCL 158

4 4 4 4 >128 >128

64 128 16 128 >128 >128

NCL 159

64 128 16 128 > 128 >128

32 128 16 64 >128 >128

NCL 160

64 128 16 64 >128 >128

64 128 128 128 >128 >128

NCL 161

128 128 128 128 > 128 >128

NCL 162 >128 128 >128 > 128 >128 >128 >128 128 >128 >128 >128 >128

>128 >128 >128 > 128 >128 >128

NCL 163

>128 >128 >128 > 128 >128 >128

>128 >128 >128 >128 >128 >128

NCL 164

>128 >128 >128 >128 >128 >128

>128 >128 >128 >128 >128 >128

NCL 165

>128 >128 >128 >128 >128 >128

32 64 8 128 >128 >128

NCL 166

32 64 8 128 >128 >128

>128 >128 >128 > 128 >128 >128

NCL 167

>128 >128 >128 > 128 >128 >128

32 32 64 32 > 128 >128

NCL 168

32 32 64 32 > 128 >128

32 64 32 32 >128 >128

NCL 169

32 64 32 32 >128 >128

>128 >128 >128 > 128 >128 >128

NCL 170

>128 >128 >128 > 128 >128 >128

Table 42: M!Cs for analogues NCL171 -230 according to Example 7.

NCL202 8 8 8 8 >128 >128

8 8 8 >128 >128

NCL203 32 64 32 32 >128 >128

32 64 64 32 >128 >128

NCL204 4 4 64 >128 >128 >128

8 8 64 >128 >128 >128

NCL205 4 4 64 >128 >128 >128

8 8 64 >128 >128 >128

NCL206 >128 >128 >128 >128 >128 >128

>128 >128 >128 >128 >128 >128

NCL207 16 32 >128 >128 >128 >128

32 32 >128 >128 >128 >128

NCL208 >128 >128 >128 >128 >128 >128

>128 >128 >128 >128 >128

NCL209 >128 >128 >128 >128 >128 >128

>128 >128 >128 >128 >128 >128

NCL210 >128 >128 128 32 >128 >128

>128 >128 128 64 >128 >128

NCL211 >128 >128 >128 >128 >128 >128

>128 >128 >128 >128 >128 >128

NCL212 >128 >128 32 16 >128 >128

>128 128 32 16 >128 >128

NCL213 >128 >128 >128 >128 >128 >128

>128 >128 > 28 >128 >128 >128

NCL214 >128 >128 >128 >128 >128 >128

>128 >128 >128 >128 >128 >128

NCL215 8 8 2 4 >128 >128

16 8 4 8 >128 >128

NCL216 2 2 2 4 >128 >128

4 2 4 >128 >128

NCL217 4 4 2 4 >128 >128

4 4 2 4 >128 >128

NCL218 >128 >128 16 >128 >128 >128

>128 >128 16 >128 >128 >128

NCL219 2 2 16 16 >128 >128

2 2 16 16 >128 >128

NCL220 16 16 32 32 >128 >128

16 16 32 32 >128 >128

NCL221 4 2 64 64 >128 >128

4 2 64 64 >128 >128

NCL222 >128 >128 >128 >128 >128 >128

>128 >128 >128 >128 >128 >128

NCL223 >128 >128 >128 >128 >128 >128

>128 >128 >128 >128 >128 >128

NCL224 >128 >128 >128 >128 >128 >128 NCL225 >128 >128 >128 >128 >128 >128

NCL226 >128 >128 >128 >128 >128 >128

NCL227 >128 >128 >128 >128 >128 >128

NCL228 >128 >128 >128 >128 >128 >128

NCL229 >128 >128 >128 >128 >128 >128

NCL230 >128 >128 >128 >128 >128 >128

[00360] The NCL analogues showing the highest level of Gram-negative antibacterial activity included NCL030 (especially Pseudomonas), NCL041 , NCL043, NCL044 (especially Pseudomonas), NCL052 (especially Pseudomonas), and NCL053 (especially Pseudomonas), NCL097 (especially Pseudomonas), NCL196 and NCL188 (especially E. co//).

[00362] The NCL analogues showing greatest activity against VRE included: NCL01 1

NCL021 NCL023 NCL029 NCL030; NCL035; NCL038 NCL041 ; NCL043

NCL044 NCL052 NCL054 NCL061 ; NCL062; NCL069 NCL073; NCL074

NCL078 NCL079 NCL080 NCL081 ; NCL082; NCL084 NCL094; NCL097

NCL099 NCL101 NCL105 NCL107; NCL108; NCL1 1 1 NCL1 17; NCL120

NCL121 NCL123 NCL126 NCL131 ; NCL136; NCL140 NCL146; NCL151

NCL153 NCL155 NCL157 NCL158; NCL159; NCL160 NCL168; NCL169

NCL171 NCL173 NCL174 NCL175; NCL177; NCL17 NCL181 ; NCL 182

NCL183 NCL184 NCL185 NCL186; NCL187; NCL18 NCL191 ; NCL192

NCL193 NCL195 NCL196 NCL197; NCL202; NCL20 NCL210; NCL212

NCL215 NCL216 NCL217 NCL218; NCL219; NCL220; and NCL221 .

[00363] The bioassay ranking of the analogues tested is shown in Table 43.

Table 43: The bioassay ranking of the analogues tested according to Example 7.

G- M+R M+R MRSA MRSA VRE VRE

NCL ID Rank NCL ID mcg/ml rank NCL ID mcg/ml rank NCL ID mcg/ml rank

NCL097 1 NCL062 2.25 1 NCL157 2 1 NCL089 0.75 1

NCL196 1 NCL157 2.25 2 NCL195 2 2 NCL062 1.5 2

NCL030 1 NCL812 2.375 3 NCL179 2 3 NCL812 1.75 3

NCL041 1 NCL195 2.75 4 NCL219 2 4 NCL084 2 4

NCL043 1 NCL216 2.75 5 NCL216 2.5 5 NCL157 2.5 5

NCL044 1 NCL089 2.875 6 NCL193 2.5 6 NCL081 2.5 6

NCL052 1 NCL084 3 7 NCL062 3 7 NCL216 3 7

NCL053 1 NCL179 3 8 NCL812 3 8 NCL217 3 8 NCL054 3.5 9 NCL054 3 9 NCL079 3 9

NCL217 3.5 10 NCL221 3 10 NCL080 3 10

NCL081 3.75 1 1 NCL199 3.5 11 NCL078 3 11

NCL193 3.75 12 NCL084 4 12 NCL195 3.5 12

NCL079 4 13 NCL217 4 13 NCL082 3.5 13

NCL080 4 14 NCL158 4 14 NCL074 3.75 14

NCL158 4 15 NCL089 5 15 NCL179 4 15

NCL082 5.25 16 NCL081 5 16 NCL054 4 16

NCL078 5.5 17 NCL079 5 17 NCL158 4 17

NCL120 6 18 NCL080 5 18 NCL088 4 18

NCL088 7 19 NCL099 6 19 NCL123 4 19

NCL099 7 20 NCL204 6 20 NCL215 4.5 20

NCL177 7 21 NCL205 6 21 NCL193 5 21

NCL215 7.25 22 NCL082 7 22 NCL120 5 22

NCL197 7.5 23 NCL120 7 23 NCL035 5 23

NCL040 8 24 NCL078 8 24 NCL140 5 24

NCL038 9 25 NCL177 8 25 NCL177 6 25

NCL202 9 26 NCL197 8 26 NCL197 7 26

NCL219 9 27 NCL040 8 27 NCL099 8 27

NCL123 9.5 28 NCL038 8 28 NCL040 8 28

NCL074 9.875 29 NCL155 8 29 NCL202 8 29

NCL155 10 30 NCL088 10 30 NCL021 8 30

NCL035 10.5 31 NCL215 10 31 NCL143 8 31

NCL021 12 32 NCL202 10 32 NCL153 8 32

NCL113 12 33 NCL113 12 33 NCL038 10 33

NCL143 12 34 NCL146 12 34 NCL155 12 34

NCL146 14 35 NCL107 12 35 NCL113 12 35

NCL153 16 36 NCL123 15 36 NCL219 16 36

NCL188 16 37 NCL074 16 37 NCL146 16 37

NCL173 17 38 NCL035 16 38 NCL188 16 38

NCL023 20 39 NCL021 16 39 NCL173 18 39

NCL220 24 40 NCL143 16 40 NCL174 18 40

NCL174 25 41 NCL188 16 41 NCL039 20 41

NCL190 28 42 NCL173 16 42 NCL023 24 42

NCL039 30 43 NCL023 16 43 NCL212 24 43

NCL121 30 44 NCL220 16 44 NCL121 28 44

NCL029 32 45 NCL153 24 45 NCL220 32 45

NCL221 33.5 46 NCL190 24 46 NCL190 32 46

NCL168 40 47 NCL207 28 47 NCL029 32 47

NCL169 40 48 NCL174 32 48 NCL169 32 48

NCL203 44 49 NCL121 32 49 NCL180 32 49

NCL107 46 50 NCL029 32 50 NCL184 32 50

EiXAMPLE 8: The effects of NCL812 on antimicrobial sensitive isolates of Staphylococcus aureus and Enterococcus faecalis

Materials and Methods

Strain information

[00364] Two Staphylococcus aureus isolates were used in the following experiments; S. aureus MK01 a human skin strain, and S. aureus KC01 an equine skin strain. These isolates were identified by Gram stain and biochemical methods, including the Remel Staphaurex commercial kit. One Enterococcus faecalis isolate (USA01 ), was not identified as a VRE strain. As this isolate has previously been speciated, it was not subjected to further testing, except for observation of pure, characteristic growth on blood agar.

Investigation of Minimum Bactericidal Concentration (MBC)

CLSI methodology

[00365] As in previous experiments, 10 μΙ_ of the contents of each well starting at the MIC was inoculated on to a Columbia SBA plate and incubated at 37 °C for 48 h. Plates were examined at 24 and 48 h and the MBC was recorded as the lowest concentration of NCL812 at which no colonies of bacteria were observed on the plate (or significant inhibition of growth was observed compared to the control) (CLSI 2005).

Kill kinetics assays for S. aureus KC01 & E. faecalis USA01 Method

[00366] S. aureus KC01 and E. faecalis USA01 , not determined to be MRSA or VRE, respectively, were grown overnight on Columbia SBA at 37 °C. A few colonies of bacteria were then suspended in CAMHB (cation-adjusted Mueller Hinton broth) and adjusted to OD 60 o of 0.08 to 0.10. The bacterial suspension was diluted 1 : 10. One millilitre of the bacteria were added to 9 mL of CAMHB containing various concentrations (up to 4*M!C) of NCL, to achieve a final bacterial concentration of 1 to 3x 106 CFU/mL. The tubes were incubated at 37 °C, with constant shaking. In order to determine the number of viable bacteria present at various time points, a 100 μΙ_ aliquot was removed from each tube and diluted. Then, 100 μΙ_ of each dilution were spread onto colony count agar, in duplicate, and incubated for 48 h at 37 °C. After 24 h the numbers of colonies present on each plate were counted and therefore the number of viable bacteria present in the original suspension enumerated. Plates were re-checked after 48 hours.

Results

Minimum Inhibitory Concentration (MIC)

[00367] The NCL812 MIC for isolates S. aureus MK01 and KC01 , and E. faecalis USA01 was investigated. The results were: S. aureus MK01 = 4-8 pg/mL, S. aureus KC01 = 2 pg/mL, E. faecalis USA 01 = 4 pg/mL

[00368] S. aureus isolates MK01 and KC01 were investigated and no growth, or growth only at low concentrations of NCL812 (2 pg/ml), was observed, indicating that NCL812 is bactericidal against S. aureus. For the E. faecalis isolate tested (USA01) however, growth of bacteria was observed at all concentrations of NCL812 tested. There was an obvious reduction in the number of bacteria with increasing concentration, but growth was present compared with no growth for S. aureus. A summary of these results can be seen in Table 45. Table 45 shows the results for NCL812 MBC tests on two non-MRSA S. aureus isolates and one non-VRE E. faecalis isolate. Each MBC test was performed in duplicate. No change in the results was observed at 48 h. Table 37 shows NCL812 MBC values (pg/mL) for 20 MRSA isolates. Each MBC test was performed in duplicate starting from NCL812 MIC concentration to 16 times of MIC. Table 38 shows NCL812 MBC values (pg/ml) for 10 VRE isolates. Each MBC test was performed in duplicate starting from NCL812 MIC concentration to 32 times the MIC.

Table 45: NCL812 MBC tests on two non-MRSA Staphylococcus aureus isolates and one non- VRE Enterococcus faecalis isolate according to Example 8.

+ = Growth on Sheep Blood Agar; 0 = No Growth on Sheep Blood Agar; N = Not Cultured; Numbers in Parenthesis are the Number of Bacteria Growing after 24 hours per ml of sample (CFU/ml)

Kill kinetics assays for S. aureus KC01 & E. faecalis US A01 Method

[00369] Colony counts were performed at t = 0, 120, 240, and 360 min, then again at 24 h. At the 2 h time point S. aureus KC01 showed a minimum of a 2.5log 10 reduction in bacterial numbers from initial numbers, and greater than a 3log 10 reduction in comparison to the control at the same time point. A minimum of a 2log 0 reduction was still evident at 6 h incubation, however after 24 h the numbers of bacteria present had increased and this was not significantly different to the control.

[00370] Similar results were obtained with E. faecalis USA01 , however the reduction in bacteria! numbers observed was less than for S. aureus KC01 . A 2log 10 reduction in CFU/mL was observed at 2 h, compared to the growth control. However, the reduction in CFU/mL compared to the original bacterial numbers was only just greater than 1 log 10 . At concentrations of 4-16 pg/mL of NCL812 this reduction in bacterial numbers remained consistent until the 6 h time point. At concentrations of 32 and 64 pg/mL however, there was approximately a 1 log 10 rise in bacterial numbers over the same time period. At 24 h bacterial numbers at all concentrations had increased to almost the same level as the growth control.

[00371 ] The results observed with these strains of S. aureus and E. faecalis are consistent with the results observed for the kill kinetics assay for all MRSA and VRE isolates tested. The kill kinetics assay of Staphylococcus aureus KC01 at different concentrations of NCL812, up to 24 h incubation are shown in Figure 39. The kill kinetics assay of Enterococcus faecalis USA01 at different concentrations of NCL812, up to 24 h incubation are shown in Figure 40.

[EXAMPLE 9: Formulations of Compounds

[00372] The following formulations were prepared using standard methods in the art.

Formulation A - Topical Formulation - PEG-based Gel with compounds of the invention

4.0g PEG 4000;

3.5g PEG 200;

0.6g propylene glycol;

1.9g water; and

0.204g of Compound (for example, NCL099)

[00373] PEG 4000, PEG 200 and propylene glycol were mixed and heated to 150 °C and until all solid crystals were dissolved. Compound was added to water and sonicated for 30 minutes until fully suspended. The Compound solution and gel solutions were mixed and allowed to cool and solidify. Formulation A will likely demonstrate acceptable viscosity, ease of skin application, uniform suspension and consistent and fine texture.

Formulation B - Topical Formulation - PEG-based Gel with compounds of the invention

3.0g PEG 4000;

1.0g PEG 8000;

3.0g PEG 200;

1.0g propylene glycol;

1.9g water; and

0.202g of Compound (for example, NCL099)

[00374] PEG 4000, PEG 8000, PEG 200 and propylene glycol were mixed and heated to 150 °C and until all solid crystals were dissolved. Compound (for example, NCL099) was added to water and sonicated for 30 minutes until fully suspended. The Compound solution and gel solutions were mixed and allowed to cool and solidify. Formulation B demonstrated acceptable viscosity, ease of skin application, uniform suspension and consistent and fine texture.

Formulation C - Topical Formulation ~ PEG-based Gel with CompoundSoluplus

2.5g PEG 4000;

4.0g PEG 200;

2.5g propylene glycol;

1.0g water; and

1.8g solid dispersion of Compound-SoluPlus. [00375] Solup!us was purchased from BASF (www.soluplus.com). Compound-SoluPlus was prepared using standard methods in the art. PEG 4000, PEG 200, Compound-SoluPlus and propylene glycol were mixed and heated to 150 °C and until all solid crystals were dissolve. Water was added and then the solution was sonicated. The solution was allowed to cool and solidify. Formulation C demonstrated acceptable viscosity, ease of skin application, uniform suspension and consistent and fine texture.

Formulation D - Tablet Formulation

30mg Calcium hydrogen phosphate dehydrate;

80mg Microcrystalline cellulose;

50mg Lactose;

8mg Hydroxypropyl methyl cellulose

1 .5mg Talc

10mg of compound (for example NCL099)

[00376] The excipients were weighed and mixed for 5 minutes. The mixture was fed into a feed hopper of a tablet press machine and the machine was operated according to standard procedures in the art. Formulation D demonstrated acceptable tablet hardness, disintegration and frability.

Formulation E - Oral Suspension

2.0 ml Glycerol;

1.5ml Absolute ethanol;

600mg NCL812; and

To 60ml Vehicle (Ora Sweet and Ora Plus, 1 : 1 ).

[00377] NCL 812 powder was sieved through a 75 μηη sieve. 600 mg of sieved NCL 812 was mixed with 2.0 ml glycerol and 1 .5 ml absolute ethanol. The mixture was placed in a mortar and manually milled until all NCL 812 was suspended uniformly. The suspension was sonicated for 30 minutes. Vehicle (55 ml of Ora Sweet and Ora Plus mixture) was then added to the suspension and milled for another 10 minutes. Volume was made up with the Ora plus and Ora sweet mixture to 60 ml by transferring to a measuring cylinder

[00378] Formulation E demonstrated acceptable suspension and demonstrated acceptable short term stability.

Formulation F - Intramuscular Injection

20mg/ml Polyvinylpyrrolidone K30 (PVPK30);

0.09mg/ml NCL812; and

50ml water. [00379] Two percent of w/v PVP K30 solution was prepared by the addition of 1 .0 g of PVP K30 to 50 ml of MilliQ water. The solution was then placed in a sonicator for 30 minutes to equilibrate and 4.5 mg of NCL 812 was added to the PVP solution and placed on an incubator shaker at a maximum speed of 10 rpm over a period of 24 hours, with controlled temperature of 25± C. Solution was transferred to 5 ml vials and checked for clarity, appearance, pH and short-term stability. The pH of solution was 7.25.

[00380] Formulation F demonstrated acceptable transparency and short term stability. EXAMPLE 10: Release of NCL812 and NCL099 from Formulation B.

[00381 ] The objective of this study was to measure the release of NCL812 and NCL099 from Formulation B prepared in Example 9.

[00382] Franz diffusion cells were utilized to quantify the release rate of NCL 812 and NCL099 from its topical formulations. Five millilitres of absolute ethanol, which was chosen as the desired release medium, was loaded into the receptor chamber. Temperature of the receptor fluid was kept constant, at 32±1 °C using a water jacket. Acetyl cellulose membranes, with pore size of 0.45 μιτι (Pall Corporation) was selected and placed between donor and receptor chamber. Followed by that, a number of test samples (Formulation B) was loaded into the donor chamber. One millilitre of receptor fluid was collected at regular time intervals of 0.25, 0.50, 0.75, 1 , 2, 3, 4, 5, 6, 7, 8 and 24 hours through the sampling port. One millilitre of fresh absolute ethanol was immediately returned to the receptor chamber. UV-HPLC was utilized to analyse the content of the receptor fluids attained.

[00383] Figure 41 presents the cumulative release of NCL812 and NCL099 over time. This study demonstrates that Formulation B provides an acceptable release profile for NCL812 and NCL099.

EXAMPLE 11 : NMR Specroscopy Lists of Compounds NCL81 2, NCL001-NCL230

[00384] NMR Spectroscopy was performed on compounds NCL812, NCL001-NCL230 using standard methods in the art. The lists of the NMR spectroscopy are presented in Table 46. Table 46: NMR Specroscopy Lists of Compounds NCL812, NCL001-NCL230

NCL8 112 1H NMR (400 MHz, DMSO) δ 12.04 (br. s, 2H), 8.48 (br. s, 1H), 8.37 (br. s, 2H), 7.97 (d, J = 8.6 Hz, 4H), 7.57 (d, J = 8.6 H

NCL001 1 H NMR (400 MHz, DMSO) δ 10.84 (br. s, 2H), 8.17 (br. s, 2H), 7.77 (d, J = 8.2 Hz, 4H), 7.50 (d, J = 8.6 Hz, 4H)

NCL002 1 H NMR (400 MHz, DMSO) δ 11.06 (s, 2H), 8.58 (br. s, 2H), 8.17 (br. s, 2H), 7.50 - 7.52 (m, 2H), 7.41 - 7.45 (m, 4H)

NCL003 1 H NMR (400 MHz, DMSO) δ 10.71 (s, 2H), 8.17 (br. S, 2H), 7.73 - 7.88 (m, 4H), 7.28 (t, J = 8.8 Hz, 4H)

NCL004 1 H NMR (400 MHz, DMSO) δ 10.89 (br. s, 2H), 8.19 (br. s, 2H), 7.65 (br. s, 2H), 7.43 - 7.56 (m, 4H), 7.19 - 7.27 (m, 2H)

NCL005 1 H NMR (400 MHz, DMSO) δ 10.94 (br. S, 2H), 8.43 (br. s, 2H), 8.10 (br. S, 2H), 7.39 - 7.52 (m, 2H), 7.21 - 7.35 (m, 4H)

NCL006 1 H NMR (400 MHz, DMSO) δ 10.50 (s, 2H), 8.11 (br. s, 2H), 7.68 (d, J = 8.6 Hz, 4H), 6.99 (d, J = 8.6 Hz, 4H), 3.80 (s, 6H)

NCL007 1 H NMR (400 MHz, DMSO) δ 11.10 (br. S, 2H), 8.24 (br. s, 2H), 7.81 - 8.03 (m, 8H)

NCL008 1 H NMR (400 MHz, DMSO) δ 11.24 (br. S, 2H), 8.51 (br. s, 2H), 8.18 - 8.29 (m, 2H), 7.90 (d, J = 7.4 Hz, 2H), 7.80 (t, J = 7.

NCL009 1 H NMR (400 MHz, DMSO) δ 11.02 (s, 2H), 8.26 (br. S, 4H), 8.07 (d, J = 7.8 Hz, 2H), 7.85 (d, J = 7.8 Hz, 2H), 7.65 (t, J =

NCL010 1 H NMR (400 MHz, DMSO) δ 10.74 (br. S, 2H), 8.15 (br. s, 2H), 7.25 - 7.39 (m, 6H), 6.94-7.01 (m, 2H), 3.82 (s, 6H)

NCL011 1 H NMR (400 MHz, DMSO) δ 11.02 (s, 2H), 8.28 (br. s, 2H), 8.13 (s, 2H), 8.04 (d, J = 7.4 Hz, 2H), 7.75 (d, J = 8.0 Hz, 2H),

NCL012 1H NMR (400 MHz, DMSO) δ 11.04 (br. s, 2H), 8.27 (br. s, 2H), 7.97 (d, J = 7.8 Hz, 4H), 7.80 (d, J = 8.2 Hz, 4H)

NCL013 1 H NMR (400 MHz, DMSO) δ 11.22 (br. S, 2H), 8.55 (br. s, 2H), 8.35 (d, J = 7.0 Hz, 2H), 7.73 - 7.82 (m, 4H), 7.57 - 7.65 (

NCL014 1 H NMR (400 MHz, DMSO) δ 10.30 (s, 1 H), 7.81 (s, 1 H), 7.76 (d, J = 8.6 Hz, 2H), 7.43 (d, J = 8.6 Hz, 2H), 6.53 (br. s, 2H)

NCL015 1 H NMR (400 MHz, DMSO) δ 12.02 (br. s, 1 H), 8.55 (s, 1 H), 8.27-8.33 (m, 1H), 7.79 (br. s, 3H), 7.51 - 7.56 (m, 1 H), 7.39 -

NCL016 1 H NMR (400 MHz, DMSO) δ 11.98 (br. s, 1 H), 8.39 (s, 1 H), 8.19-8.26 (m, 1 H), 7.80 (br. s, 3H), 7.46 - 7.58 (m, 1 H), 7.20 -

NCL017 1 H NMR (400 MHz, DMSO) δ 11.79 (br. s, 1 H), 8.17 (s, 1 H), 7.87 (d, J = 9.8 Hz, 1 H), 7.71 (br. s, 3H), 7.62 (d, J = 7.4 Hz, 1

NCL018 1H NMR (400 MHz, DMSO) δ 10.66 (s, 2H), 8.47 (br. S, 2H), 7.91-8.00 (m, 2H), 7.19 - 7.32 (m, 6H), 2.42 (s, 6H)

NCL019 1 H NMR (400 MHz, DMSO) δ 10.68 (br. s, 2H), 8.15 (br. s, 2H), 7.57 (s, 2H), 7.52 (d, J = 7.4 Hz, 2H), 7.32 (t, J = 7.6 Hz, 2

NCL020 1 H NMR (400 MHz, DMSO) δ 12.37 (br. S, 2H), 8.83 (br. s, 2H), 8.63 (br. s, 2H), 8.39 - 8.44 (m, 2H), 7.55 - 7.60 (m, 2H), 7.

NCL021 1 H NMR (400 MHz, DMSO) δ 12.11 (br. s, 1 H), 8.52 (br. s, 2H), 8.40 (br. s, 2H), 8.02 (t, J = 8.6, 4H), 7.35 (t, J = 8.8 Hz, 4H

NCL022 1 H NMR (400 MHz, DMSO) δ 12.19 (br. s, 2H), 8.65 (br. s, 2H), 8.58 (br. s, 1 H), 8.34 (t, J = 7.6 Hz, 2H), 7.51 - 7.60 (m, 2H

NCL023 1 H NMR (400 MHz, DMSO) δ 12.08 (br. S, 2H), 8.38 (br. s, 2H), 7.92-8.00 (m, 2H), 7.65-7.71 (m, 2H), 7.50 - 7.58 (m, 2H),

NCL024 1H NMR (400 MHz, DMSO) δ 12.32 (br. s, 2H), 8.67 (br. s, 2H), 8.44 (br. s, 2H), 8.15 (d, J = 8.6 Hz, 4H), 7.98 (d, J = 8.6 Hz, 4H)

NCL025 1H NMR (400 MHz, DMSO) δ 8.75 (br. s, 2H), 8.50 (d, J = 8.2 Hz, 2H), 7.97 (d, J = 7.4 Hz, 2H), 7.85 (t, J = 7.6 Hz, 2H), 7.68 (t, J = 7.6 Hz, 2H)

NCL026 1H NMR (400 MHz, DMSO) δ 12.26 (br. s, 1H), 8.66 (br. s, 1H), 8.55 (s, 2H), 8.43 (br. s, 2H), 8.21 (d, J = 7.8 Hz, 2H), 7.94 (d, J = 7.8 Hz, 2H), 7.71 (t, J = 7.8 Hz, 2H) NCL027 1H NMR (400 MHz, DMSO) δ 11.78 (br. s, 2H), 8.31 (br. s, 3H), 7.87 (d, J = 8.6 Hz, 4H), 7.04 (d, J = 8.6 Hz, 4H), 3.83 (s, 6H)

1H NMR (400 MHz, DMSO) δ 12.00 (br. s, 2H), 8.75 (br. s, 2H), 8.39 (br. s, 2H), 8.22 (d, J = 6.7 Hz, 2H), 7.44 - 7.52 (m, 2H), 7.14 (d, J = 8.2 Hz, 2H), 7.05 (t, J = 7.

NCL028

Hz, 2H), 3.89 (s, 6H)

NCL029 1H NMR (400 MHz, DMSO) δ 11.98 (br. s, 2H), 8.48 (br. s, 2H), 8.36 (br. s, 2H), 7.56 (s, 2H), 7.35 - 7.49 (m, 4H), 7.04-7.10 (m, 2H), 3.84 (s, 6H)

NCL030 1H NMR (400 MHz, DMSO) δ 11.83 (br. s, 1H), 8.16 (s, 1H), 7,91 (d, J = 8.2 Hz, 2H), 7.75 (br. s, 1H), 7.53 (d, J = 8.2 Hz, 2H)

NCL031 1H NMR (400 MHz, DMSO) δ 11.91 (br. s, 1H), 8.22 (s, 1H), 8.09 (d, J = 8.2 Hz, 2H), 7.93 (d, J = 8.2 Hz, 2H)

1H NMR (400 MHz, DMSO) δ 12.12 (s, 1H), 8.48 (s, 1H), 8.38 (d, J = 7.8 Hz, 1H), 7.94 (d, J = 7.8 Hz, 1H), 7.86 (br. s, 2H), 7.80 (t, J = 7.8 Hz, 2H), 7.64 (t, J = 7.6 H

NCL032

1H)

NCL033 1H NMR (400 MHz, DMSO) δ 11.93 (s, 1H), 8.50 (s, 1H), 8.20 (s, 1H), 8.14 (d, J = 7.8 Hz, 1H), 7.90 (d, J = 7.8 Hz, 1H), 7.66 (t, J = 7.8 Hz, 1H)

1H NMR (400 MHz, DMSO) δ 11.87 (br. s, 1H), 8.48 (s, 1H), 8.09 (d, J = 7.8 Hz, 1H), 7.70 (br. s, 2H), 7.38 - 7.49 (m, 1H), 7.11 (d, J = 8.6 Hz, 1H), 7.01 (t, J = 7.4 H

NCL034

1H), 3.86 (s, 3H)

NCL035 1H NMR (400 MHz, DMSO) δ 12.32 (br. s, 2H), 8.69 (br. s, 2H), 8.49 (br. s, 2H), 8.18 (d, J = 7.8 Hz, 4H), 7.86 (d, J = 8.2 Hz, 4H)

NCL036 1H NMR (400 MHz, DMSO) δ 12.51 (br. s, 1 H), 8.80 (br. s, 2H), 8.72 (br. s, 1 H), 8.59 (d, J = 7.8 Hz, 2H), 7.78 - 7.91 (m, 4H), 7.71 (t, J = 8.0 Hz, 2H)

NCL037 1H NMR (400 MHz, DMSO) δ 12.28 (br. s, 2H), 8.70 (br. s, 2H), 8.50 (br. s, 2H), 8.38 (s, 2H), 8.22 (d, J = 7.8 Hz, 2H), 7.85 (d, J = 7.8 Hz, 2H), 7.74 (t, J = 7.8 Hz, 2H) NCL038 1H NMR (400 MHz, DMSO) δ 11.92 (br. s, 2H), 8.41 (br. s, 2H), 8.36 (br. s, 2H), 7.83 (d, J = 8.2 Hz, 4H), 7.31 (d, J = 7.8 Hz, 4H), 2.37 (s, 6H)

NCL039 1H NMR (400 MHz, DMSO) δ 11.99 (br. s, 2H), 8.73 (br. s, 2H), 8.41 (br. S, 2H), 8.19 (d, J = 7.8 Hz, 2H), 7.37 (t, J = 8.0 Hz, 2H), 7.30 (t, J = 7.8 Hz, 4H), 2.46 (s, 6H)

1H NMR (400 MHz, DMSO) δ 11.97 (br. s, 2H), 8.44 (br. s, 2H), 8.37 (br. s, 2H), 7.76 (s, 2H), 7.71 (d, J = 7.8 Hz, 2H), 7.38 (t, J = 7.8 Hz, 2H), 7.31 (d, J = 7.8 Hz, 2H)

NCL040

2.38 (s, 6H)

NCL041 1H NMR (400 MHz, DMSO) δ 11.94 (s, 1H), 8.25 (s, 1H), 8.11 (d, J = 7.8 Hz, 2H), 7.71 - 7.91 (m, 4H)

NCL042 1H NMR (400 MHz, DMSO) δ 12.04 (s, 1H), 8.46-8.56 (m,2H), 7.70-7.93 (m, 5H), 7.66 (t, J = 7.8 Hz, 1H)

NCL043 1H NMR (400 MHz, DMSO) δ 11.88 (s, 1H), 8.33 (s, 1H), 8.26 (s, 1H), 8.14 (d, J - 7.8 Hz, 1H), 7.80 (d, J =7.8 Hz, 1H), 7.69 (t, J = 7.8 Hz, 1H)

NCL044 1H NMR (400 MHz, DMSO) δ 11.71 (br. s, 1H), 8.13 (s, 1H), 7.76 (d, J = 8.2 Hz, 2H), 7.27 (d, J = 7.8 Hz, 2H), 2.35 (s, 3H)

NCL045 1H NMR (400 MHz, DMSO) δ 11.69 (br. s, 1H), 8.45 (s, 1H), 8.06 (d, J = 7.4 Hz, H), 7.67 (br. s, 2H), 7.30 - 7.39 (m, 1H), 7.20 - 7.29 (m, 2H), 2.42 (s, 3H)

NCL046 1H NMR (400 MHz, DMSO) δ 11.64 (br. s, 1H), 8.12 (s, 1H), 7.53 - 7.77 (m, 4H), 7.34 (t, J = 7.8 Hz, 1H), 7.27 (d, J = 7.8 Hz, 1H), 2.35 (s, 3H)

NCL047 1H NMR (400 MHz, DMSO) δ 10.47 (s, 1H), 8.23 (s, 1H), 8.16 - 8.21 (m, 1H), 7.42 - 7.50 (m, 1H), 7.30 - 7.40 (m, 2H), 6.57 (br. s, 2H)

NCL048 1H NMR (400 MHz, DMSO) δ 10.40 (s, 1H), 8.08-8.15 (m, 1H), 8.05 (s, 1H), 7.34-7.44 (m, 1H), 7.17-7.28 (m, 2H), 6.54 (br. s, 2H)

NCL049 1H NMR (400 MHz, DMSO) δ 10.51 (s, 1H), 7.93 (d, J = 8.2 Hz, 1H), 7.86 (s, 1H), 7.83 (d, J = 8.2 Hz, 1H), 6.63 (br. s, 2H)

1H NMR (400 MHz, DMSO) δ 10.66 (s, 1H), 8.13 (d, J = 8.0 Hz, 1H), 8.11 (s, 1H), 7.85 (d, J = 7.4 Hz, 1H), 7.71 (t, J = 7.8 Hz, 1H), 7.52 (t, J = 7.0 Hz, 1H),6.60(br.

NCL050

2H)

NCL051 1H NMR (400 MHz, DMSO) δ 10.43 (s, 1H), 8.37 (s, 1H), 7.98 (d, J = 7.8 Hz, 1H), 7.84 (s, 1H), 7.77 (d, J = 7.8 Hz, 1H), 7.57 (t, J = 7.8 Hz, 1H)

NCL052 1H NMR (400 MHz, DMSO) δ 11.95 (s, 1H), 8.16 (s, 1H), 8.10 (s, 1H), 7.80 (br. s, 2H), 7.76 (d, J = 7.0 Hz, 1 H), 7.40-7.55 (m, 2H)

NCL053 1H NMR (400 MHz, DMSO) δ 11.88 (s, 1H), 8.17 (s, 1H), 7.95 (dd, J = 8.8, 5.7 Hz, 2H), 7.76 (br. s, 1H), 7.30 (t, J = 9.0 Hz, 2H)

NCL054 1H NMR (400 MHz, DMSO) δ 12.17 (br. s, 2H), 8.61 (br. s, 2H), 8.39 (br. s, 2H), 8.16 (s, 2H), 7.83 (d, J = 7.0 Hz, 2H), 7.45 - 7.61 (m, 4H)

NCL055 1H NMR (400 MHz, DMSO) δ 10.34 (s, 1H), 7.93 (s, H), 7.80 (s, H), 7.54 - 7.67 (m, 1H), 7.32 - 7.46 (m, 2H), 6.58 (br. s, 1H)

NCL056 1H NMR (400 MHz, DMSO) δ 10.23 (s, 1H), 7.82 (s, H), 7.78 (dd, J = 8.8, 5.7 Hz, 2H), 7.21 (t, J = 8.8 Hz, 1H), 6.49 (br. s, 2H)

NCL057 1H NMR (400 MHz, DMSO) δ 10.33 (s, 1H),7.82 (s, 1H), 7.71 (d, J =9.8 Hz, 1H), 7.35 - 7.53 (m, 2H), 7.10-7:23 (m, 1H),6.57(br. s, 1H)

NCL058 1H NMR (400 MHz, DMSO) δ 10.08 (s, 1H), 7.78 (s, 1H), 7.65 (d, J =9.0 Hz, 2H), 6.94 (d, J = 8.6 Hz, 2H), 6.40 (br. S, 2H), 3.78 (s, 3H)

1H NMR (400 MHz, DMSO) δ 10.23 (s, 1H), 7.80 (s, 1H), 7.34 (s, 1H), 7.29 (t, J = 7.8 Hz, 1H), 7.21 (d, J = 7.8 Hz, 1H), 6.91 (dd, J = 7.8, 2.0 Hz, 1H), 6.51 (br. s, 2H

NCL059

3.79 (s, 3H)

1H NMR (400 MHz, DMSO) δ 10.56 (s, 1H), 8.41 (d, J = 7.8 Hz, 1H), 8.19 (br. s, 1H), 7.74 (d, J = 7.8 Hz, 1H), 7.67 (t, J = 7.6 Hz, 1H), 7.54 (t, J = 7.8 Hz, 1H), 6.62 (b

NCL060

S,2H)

NCL061 1H NMR (400 MHz, DMSO) δ 11.71 (br. s, 2H), 8.91 (br. s, 1H), 8.28 (d, J = 7.8 Hz, 4H), 7.82 (d, J = 8.2 Hz, 4H), 2.49 (br. s, 6H)

NCL062 1H NMR (400 MHz, DMSO) δ 11.68 (br. S, 2H), 8.78 (br. s, 1H), 8.10 (d, J = 8.6 Hz, 4H), 7.52 (d, J = 8.6 Hz, 4H), 2.43 (s, 6H)

NCL063 1H NMR (400 MHz, DMSO) δ 7.99 (br. s, 4H), 7.83 (d, J = 7.8 Hz, 6H)

NCL064 1H NMR (400 MHz, DMSO) δ 7.94 (d, J = 7.8 Hz, 4H), 7.84 (t, J = 7.6 Hz, 4H), 7.64 (t, J = 7.6 Hz, 4H)

NCL065 1H NMR (400 MHz, DMSO) δ 12.20 (br. s, 1H), 11.86 (br. s, H), 8.65 (br. s, 1H), 8.50 (br. s, 1H), 8.03-8.27 (m, 3H), 7.90 (d, J = 7.8 Hz, 2H), 7.68 (t, J = 7.6 Hz, 2H) NCL066 1H NMR (400 MHz, DMSO) δ 7.43 - 7.67 (m, 7H), 7.23 - 7.34 (m, 2H)

NCL067 1H NMR (400 MHz, DMSO) δ 10.40 (s, 1H), 8.16 (s, 1H), 7.98 (d, J = 7.4 Hz, 1H),7.90(s, 1H), 7.67 (d, J = 7.3 Hz, 1H), 7.60 (t, J = 7.4 Hz, 1H)

NCL068 1H NMR (400 MHz, DMSO) δ 8.47 (br. s, 1H), 8.18 (d, J = 7.8 Hz, 3H), 8.07 (d, J = 8.6 Hz, 2H), 7.85 (d, J = 7.8 Hz, 2H), 7.53 (d, J = 8.6 Hz, 2H), 2.40 (s, 3H) NCL069 1H NMR (400 MHz, DMSO) δ 9.44 (br. s, 1H), 8.03 (d, J = 8.2 Hz, 2H), 7.94 (br. s, 2H), 7.48 (d, J = 8.6 Hz, 2H), 2.31 (s, 3H)

NCL070 1H NMR (400 MHz, DMSO) δ 8.12 (br. s, 1 H), 7.71 (br. s, 3H), 7.49 (br. s, 6H)

1H NMR (400 MHz, DMSO) δ 11.43 (s, 1H), 8.40 (s, 1H), 8.16 (br. s, 1H), 8.10 (d, J = 7.4 Hz, 1H), 7.95 (br. s, 1H), 7.37 (t, J = 7.8 Hz, 1H), 7.06 (d, J = 8.2 Hz, 1H)

NCL071

6.95 (t, J = 7.4 Hz, 1H), 3.82 (s, 3H)

NCL072 1H NMR (400 MHz, DMSO) δ 7.91 (br. d, J = 8.2 Hz, 3H), 7.52 (d, J = 8.6 Hz, 2H)

NCL073 1H NMR (400 MHz, DMSO) δ 9.51 (br. s, 1H), 8.22 (d, J = 8.3 Hz, 2H), 8.01 (br. s, 2H), 7.77 (d, J = 8.3 Hz, 2H), 2.36 (s, 3H)

1H NMR (400 MHz, DMSO) δ 12.46 (br. s, 1H), 8.79 (br. s, 1H), 8.66 (br. s, 2H), 8.46 (d, J = 8.2 Hz, 2H), 7.99 (d, J = 7.0 Hz, 2H), 7.77 (br. s, 1H), 7.58 (d, J = 6.7 Hz NCL074

3H)

NCL075 1H NMR (400 MHz, DMSO) δ 8.48 (br. s, 1H), 8.26 (d, J = 7.4 Hz, 2H), 8.19 (d, J = 7.4 Hz, 2H), 7.75-7.93 (m, 4H), 2.46 (s, 3H)

NCL076 1H NMR (400 MHz, DMSO) δ 8.53 (br. s, 2H), 8.29 - 8.46 (m, 2H), 7.99 (d, J = 7.8 Hz, 2H), 7.83 (d, J = 7.8 Hz, 2H), 7.57 (d, J = 7.8 Hz, 2H), 7.31 (d, J = 7.8 Hz, 2H)

2.37 (s, 3H)

1 H NMR (400 MHz, DMSO) δ 12.28 (br. S, 1 H), 8.67 (br. S, 1 H), 8.49 (br. S, 1 H), 8.42 (br. s, 1 H), 8.18 (d, J = 7.8 Hz, 2H), 8.00 (d, J = 8.2 Hz, 2H), 7.86 (d, J = 8.2 H

NCL077

2H), 7.58 (d, J = 7.8 Hz, 2H)

1 H NMR (400 MHz, DMSO) δ 12.33 (br. s, 1 H), 8.74 (br. s, 2H), 8.58 (t, J = 7.6 Hz, 1 H), 8.42 (br. s, 1 H), 8.00 (d, J = 8.2 Hz, 2H), 7.87 (d, J = 10.2 Hz, 1H), 7.74 (d, J

NCL078

8.2 Hz, 1 H), 7.58 (d, J = 8.2 Hz, 2H)

NCL079 1 H NMR (400 MHz, DMSO) δ 12.08 (br. s, 1 H), 8.54 (br. s, 1 H), 8.39 (br. s, 2H), 7.93 - 8.09 (m, 4H), 7.57 (d, J = 8.2 Hz, 2H), 7.35 (t, J = 8.6 Hz, 2H)

1 H NMR (400 MHz, DMSO) δ 12.04 (br. s, 1 H), 11.36 (br. s, 1 H), 8.40 (br. s, 1H), 8.25 (d, J = 8.2 Hz, 2H), 8.00 (d, J = 7.0 Hz, 2H), 7.82 (d, J = 7.4 Hz, 2H), 7.57 (d, J

NCL080

6.7 Hz, 2H), 2.44 (s, 3H)

NCL081 1 H NMR (400 MHz, DMSO) δ 8.39 (br. s, 1H), 8.07 (d, J = 8.6 Hz, 2H), 7.99 (d, J = 8.6 Hz, 2H), 7.57 (d, J = 8.2 Hz, 2H), 7.53 (d, J = 8.6 Hz, 2H), 2.39 (s, 3H)

1 H NMR (400 MHz, DMSO) δ 12.40 (br. S, 1 H), 8.84 (br. s, 1 H), 8.63 (br. s, 2H), 8.42 (d, J = 7.8 Hz, 2H), 7.99 (d, J = 8.6 Hz, 2H), 7.58 (d, J = 8.2 Hz, 3H), 7.43 - 7.5 NCL082

(m, 2H)

NCL083 1 H NMR (400 MHz, DMSO) δ 12.30 (br. s, 1 H), 8.64 (br. s, 2H), 8.41 (br. s, 2H), 8.17 (s, 1H), 7.99 (d, J = 8.6 Hz, 2H), 7.82 (d, J = 6.7 Hz, 1 H), 7.45 - 7.64 (m, 4H)

1 H NMR (400 MHz, DMSO) δ 12.38 (br. s, 1 H), 8.64 (br. s, 2H), 8.30 - 8.50 (m, 2H), 7.99 (d, J = 8.6 Hz, 2H), 7.62 (d, J = 10.4, 1.4 Hz, 1 H), 7.57 (d, J = 8.6 Hz, 2H

NCL084

7.46 (d, J = 8.6 Hz, 1H)

1 H NMR (400 MHz, DMSO) δ 8.77 (br. s, 1 H), 8.68 (br. s, 2H), 8.52 (d, J = 8.2 Hz, 1 H), 8.46 (br. s, 1 H), 7.92 - 8.06 (m, 3H), 7.84 (t, J = 7.6 Hz, 1 H), 7.68 (t, J = 7.6 H NCL085

1 H), 7.58 (d, J = 8.2 Hz, 2H)

1 H NMR (400 MHz, DMSO) δ 12.39 (br. s, 1 H), 8.68 (br. s, 2H), 8.57 (s, 1 H), 8.43 (br. s, 2H), 8.21 (d, J = 7.4 Hz, 1 H), 8.00 (d, J = 8.2 Hz, 2H), 7.94 (d, J = 7.0 Hz, 1 H NCL086

7.70 (t, J = 7.2 Hz, 1 H), 7.58 (d, J = 7.8 Hz, 2H)

NCL087 1 H NMR (400 MHz, DMSO) δ 8.71 (br. s, 2H), 8.48 (br. s, 1H), 8.43 (br. s, 1 H), 8.16 (d, J = 8.2 Hz, 2H), 7.99 (t, J = 8.0 Hz, 4H), 7.58 (d, J = 8.6 Hz, 2H)

1 H NMR (400 MHz, DMSO) δ 12.33 (br. s, 1 H), 8.68 (br. s, 1H), 8.61 (br. s, 2H), 8.42 (br. s, 1 H), 8.35 (t, J = 7.4 Hz, 1 H), 7.99 (d, J

NCL088

7.27 - 7.41 (m, 2H)

NCL089 1 H NMR (400 MHz, DMSO) δ 11.45 (br. S, 1 H), 8.76 - 8.94 (m, 1 H), 8.11 (d, J = 8.0 Hz, 4H), 7.53 (d, J = 8.0 Hz, 4H), 2.42 (br. s, 6H)

NCL090 1 H NMR (400 MHz, DMSO) δ 10.60 (br. s, 1 H), 8.68 (s, 1H), 8.09 (br. s, 1 H), 7.95 (br. s, 4H), 7.32 - 7.71 (m, 10H)

NCL091 1 H NMR (400 MHz, DMSO) δ 9.61 (br. s, 1H), 8.19 (d, J = 6.3 Hz, 2H), 7.90 (br. s, 2H), 7.56 - 7.73 (m, 2H), 7.26 - 7.40 (m, 2H)

NCL092 1 H NMR (400 MHz, DMSO) δ 12.38 (br. S, 1 H), 7.82 - 7.94 (m, 2H), 7.18 - 7.63 (m, 7H), 4.39 (br. s, 2H)

NCL 093 1 H NMR (300 MHz, DMSO) δ 8.64 (s, 4H), 8.35 - 8.24 (m, 4H), 8.06 - 7.93 (m, 6H), 7.64 - 7.54 (m, 4H).

NCL 094 1 H NMR (300 MHz, CDCI3) 6 7.16 (d, J = 4.7 Hz, 2H), 6.10 (br S, 3H), 2.27 - 2.14 (m, 2H), 1.84 - 1.61 (m, 10H), 1.37 - 1.13 (m, 10H)

NCL 095 1H NMR (300 MHz, DMSO) δ 8.39 (s, 2H), 8.36 - 8.11 (m, 4H), 7.78 (s, 2H), 7.12 (d, J = 1.4 Hz, 2H).

NCL 096 1 H NMR (300 MHz, MeOD) δ 8.08 (d, J = 8.2 Hz, 2H), 7.61 - 7.53 (m, 4H), 7.45 - 7.30 (m, 6H), 7.17 - 6.97 (m, 4H).

1 H NMR (300 MHz, DMSO) δ 9.06 (br s, 6H), 8.25 - 8.01 (m, 4H), 6.83 (s, 4H).

NCL 097

13C NMR (75 MHz, DMSO) δ 152.2, 149.7, 146.2, 136.5, 123.7, 107.4.

NCL 098 1 H NMR (300 MHz, DMSO) δ 8.65 (s, 2H), 8.53 (s, 2H), 8.40 (s, 2H), 8.24 (d, J = 7.3 Hz, 2H), 8.09 - 7.98 (m, 2H), 7.62 (t, J = 7.7 Hz, 2H).

1 H NMR (300 MHz, DMSO) δ 8.56 - 8.32 (m, 4H), 7.85 (d, J = 8.3 Hz, 4H), 7.49 (d, J = 8.3 Hz, 4H), 1.31 (s, 18H).

NCL 099

13C NMR (75 MHz, DMSO) δ 153.7, 152.7, 148.8, 130.7, 127.8, 125.6, 34.7, 31.0.

NCL 100 1 H NMR (300 MHz, DMSO) δ 12.39 (br s, 2H), 8.55 (s, 2H), 8.46 (s, 2H), 8.01 - 7.88 (m, 4H), 7.55 - 7.41 (m, 6H).

1 H NMR (300 MHz, DMSO) δ 12.06 (br s, 2H), 9.71 (br s, 2H), 9.21 (br s, 2H), 8.70 (s, 2H), 8.30 (s, 2H), 7.50 (d, J = 7.9 Hz, 2H), 6.90 (d, J = 7.7 Hz, 2H), 6.70 (t, J = 7. NCL 101

Hz, 2H).

NCL 102 1 H NMR (300 MHz, DMSO) δ 12.86 (br s, 2H), 8.89 (s, 2H), 8.77 (s, 2H), 8.52 (d, J = 7.9 Hz, 2H), 8.11 (d, J = 8.1 Hz, 2H), 7.91 - 7.78 (m, 2H), 7.77 - 7.65 (m, 2H). NCL 103 1 H NMR (300 MHz, DMSO) δ 11.81 (br s, 2H), 10.32 - 9.85 (m, 4H), 8.52 (s, 2H), 8.12 (s, 2H), 7.85 (d, J = 8.4 Hz, 2H), 6.43 (s, 2H), 8.33 (d, J = 8.5 Hz, 2H).

NCL 104 1 H NMR (300 MHz, DMSO) δ 11.72 (br s, 2H), 9.78 (br s, 2H), 9.45 (s, 2H), 8.48 (s, 2H), 8.34 (br s, 2H), 8.04 (s, 2H), 7.33 (s, 2H), 6.44 (s, 2H).

NCL 105 1 H NMR (300 MHz, DMSO) δ 11.75 (br s, 2H), 9.71 (br s, 2H), 9.15 (br s, 2H), 8.86 - 8.40 (m, 4H), 8.13 (s, 2H), 7.33 (d, J = 8.6 Hz, 2H), 6.42 (d, J = 8.6 Hz, 2H). NCL 106 1 H NMR (300 MHz, DMSO) δ 9.12 (br s, 4H), 8.29 (s, 2H), 8.20 (s, 2H), 7.10 (s, 2H), 6.93 (s, 2H), 3.84 (s, 6H).

1 H NMR (300 MHz, DMSO) δ 12.19 (s, 2H), 10.25 (s, 2H), 8.70 (s, 2H), 8.34 (s, 2H), 8.06 (d, J = 7.8 Hz, 2H), 7.35 - 7.23 (m, 2H), 7.00 (d, J = 8.2 Hz, 2H), 6.87 (t, J

NCL 107

7.5 Hz, 2H).

NCL 108 1 H NMR (300 MHz, DMSO) δ 8.00 (s, 2H), 7.26 - 7.08 (m, 6H), 6.98 - 6.43 (m, 4H).

NCL 109 1 H NMR (300 MHz, DMSO) δ 8.60 (d, J = 1.2 Hz, 2H), 8.33 - 8.19 (m, 4H), 8.15 (d, J = 8.0 Hz, 2H), 7.74 - 7.61 (m, 2H), 7.12 (s, 2H).

NCL 110 1 H NMR (300 MHz, DMSO) δ 8.85 (s, 2H), 8.58 (s, 2H), 8.31 (d, J = 8.7 Hz, 4H), 8.23 (d, J = 8.9 Hz, 4H).

1 H NMR (300 MHz, DMSO) δ 11.80 (br s, 2H), 10.30 - 9.80 (m, 4H), 8.52 (s, 2H), 8.12 (s, 2H), 7.84 (d, J = 8.6 Hz, 2H), 6.42 (d, J = 1.8 Hz, 2H), 6.33 (d, J = 8.6 Hz

NCL 111

2H).

NCL 112 1 H NMR (300 MHz, DMSO) δ 12.48 (br s, 2H), 8.62 (s, 2H), 8.51 (s, 2H), 8.04 (d, J = 7.5 Hz, 4H), 7.85 - 7.69 (m, 8H), 7.54 - 7.36 (m, 6H).

NCL 113 1 H NMR (400 MHz, DMSO) δ 11.92 (br s, 2H), 8.24 (s, 2H), 8.16 (s, 2H), 7.71 (d, J = 8.9 Hz, 4H), 6.74 (d, J = 8.9 Hz, 4H), 2.98 (s, 12H).

NCL 114 1 H NMR (400 MHz, DMSO) δ 12.67 (br s, 2H), 8.81 (s, 2H), 8.40 (s, 2H), 8.06 (d, J = 1.8 Hz, 4H), 7.68 (t, J = 1.8 Hz, 2H).

1 H NMR (400 MHz, DMSO) δ 12.09 (br s, 2H), 8.44 (s, 2H), 8.34 (s, 2H), 7.63 (d, J = 1.4 Hz, 2H), 7.33 (dd, J = 8.3, 1.6 Hz, 2H), 7.03 (d, J = 8.4 Hz, 2H), 3.86 (s, 6H)

NCL 115

3.81 (s, 6H).

NCL 116 1 H NMR (400 MHz, DMSO) δ 12.27 (br s, 2H), 8.49 (s, 2H), 8.44 - 8.27 (m, 4H), 7.57 - 7.44 (m, 10H), 7.41 - 7.33 (m, 6H).

NCL 117 1 H NMR (400 MHz, DMSO) δ 9.77 (br S, 2H), 8.36 (s, 2H), 8.29 (s, 2H), 7.58 (d, J = 1.5 Hz, 2H), 7.23 (dd, J = 8.1 , 1.2 Hz, 2H), 6.87 (d, J = 8.1 Hz, 2H), 3.86 (s, 6H). NCL 118 1 H NMR (400 MHz, DMSO) δ 12.83 (br s, 2H), 8.76 (s, 2H), 8.69 (s, 2H), 8.31 - 8.21 (m, 2H), 7.45 - 7.31 (m, 4H).

NCL 119 1 H NMR (400 MHz, DMSO) δ 12.13 (br s, 2H), 10.35 (s, 2H), 8.39 (s, 2H), 8.35 (s, 2H), 7.85 (d, J = 8.7 Hz, 4H), 7.72 (d, J = 8.6 Hz, 4H), 2.08 (s, 6H).

NCL 120 1 H NMR (400 MHz, DMSO) δ 8.62 - 8.29 (m, 4H), 7.85 (d, J = 8.2 Hz, 4H), 7.33 (d, J = 8.2 Hz, 4H), 2.98 - 2.87 (m, 2H), 1.21 (d, J = 6.9 Hz, 12H).

NCL 121 1 H NMR (400 MHz, DMSO) δ 8.60 - 8.30 (m, 4H), 7.84 (d, J = 8.1 Hz, 4H), 7.28 (d, J = 8.1 Hz, 4H), 2.59 (t, J = 7.5 Hz, 4H), 1.64 - 1.54 (m, 4H), 0.88 (t, J = 7.3 Hz, 6H). NCL 122 1 H NMR (400 MHz, DMSO) δ 8.56 (s, 2H), 8.42 (d, J = 2.0 Hz, 2H), 8.38 (s, 2H), 8.09 (dd, J = 8.7, 2.0 Hz, 2H), 7.29 (d, J = 8.7 Hz, 2H).

1 H NMR (400 MHz, DMSO) δ 8.68 (s, 2H), 8.43 (s, 2H), 8.28 - 8.16 (m, 2H), 7.77 - 7.64 (m, 2H), 7.58 - 7.46 (m, 2H).

NCL 123 13C NMR (101 MHz, DMSO) δ 153.0, 150.8 (dd, J = 250.6, 13.0 Hz), 149.9 (dd, J = 245.9, 13.2 Hz), 146.6, 131.2 (dd, J = 6.4, 3.4 Hz), 126.0 (dd, J = 6.4, 2.8 Hz), 117.

(d, J = 17.7 Hz), 115.7 (d, J = 18.5 Hz).

1 H NMR (400 MHz, DMSO) δ 10.84 (br s, 2H), 9.29 (s, 2H), 8.80 (d, J = 8.6 Hz, 2H), 8.43 (s, 2H), 7.94 (d, J = 9.0 Hz, 2H), 7.87 (d, J = 7.9 Hz, 2H), 7.64 - 7.56 (m, 2H)

NCL 124

7.43 - 7.36 (m, 2H), 7.34 (d, J = 8.9 Hz, 2H).

NCL 125 H NMR (400 MHz, DMSO) δ 9.21 (br s, 2H), 8.42 - 8.17 (m, 4H), 7.43 (d, J = 1.9 Hz, 2H), 7.25 (dd, J = 8.4, 1.9 Hz, 2H), 6.99 (d, J = 8.4 Hz, 2H), 3.83 (s, 6H).

NCL 126 1 H NMR (400 MHz, DMSO) δ 12.27 (br s, 2H), 8.58 (s, 2H), 8.42 (s, 2H), 7.96 (d, J = 8.3 Hz, 4H), 7.58 (d, J = 8.2 Hz, 4H), 4.39 (s, 2H).

NCL 127 H NMR (400 MHz, DMSO) δ 12.84 (br s, 2H), 8.84 (s, 2H), 8.74 (s, 2H), 8.46 (d, J = 8.6 Hz, 2H), 7.71 (d, J = 1.6 Hz, 2H), 7.54 (dd, J = 8.6, 1.2 Hz, 2H).

NCL 128 1 H NMR (400 MHz, DMSO) δ 8.64 (s, 2H), 8.36 (s, 2H).

NCL 129 H NMR (400 MHz, DMSO) δ 8.84 (s, 2H), 8.69 (s, 2H), 8.41 (dd, J = 7.8, 1.7 Hz, 2H), 7.70 (dd, J = 8.0, 1.0 Hz, 2H), 7.48 (t, J = 7.3 Hz, 2H)*, 7.44 - 7.37 (m, 2H)*.

NCL 130 H NMR (400 MHz, DMSO) δ 8.66 (s, 2H), 8.37 (s, 2H), 7.79 (d, J = 1.6 Hz, 2H), 7.68 - 7.62 (m, 2H), 3.92 (s, 6H), 3.78 (s, 6H).

NCL 131 H NMR (400 MHz, DMSO) δ 12.45 (br s, 2H), 8.68 (s, 2H), 8.41 (s, 2H), 8.29 (s, 2H), 7.87 (d, J = 7.8 Hz, 2H), 7.70 - 7.63 (m, 2H), 7.44 (t, J = 7.9 Hz, 2H).

NCL 132 1 H NMR (400 MHz, DMSO) δ 12.69 (br s, 2H), 8.61 (s, 4H), 7.34 (dd, J = 9.0, 2.8 Hz, 2H), 7.22 (td, J = 8.6, 2.9 Hz, 2H), 7.03 (dd, J = 8.8, 4.6 Hz, 2H), 5.29 (s, 4H). NCL 133 1 H NMR (400 MHz, DMSO) δ 12.51 (br s, 2H), 8.49 - 8.28 (m, 4H), 8.16 (s, 2H), 7.35 (s, 2H).

NCL 134 1 H NMR (400 MHz, DMSO) δ 12.42 (br s, 2H), 8.60 (s, 2H), 8.42 (s, 2H), 7.91 (d, J = 8.5 Hz, 4H), 7.69 (d, J = 8.5 Hz, 4H).

NCL 135 1 H NMR (400 MHz, DMSO) δ 12.24 (br s, 2H), 8.66 (s, 2H), 8.49 (br s, 2H), 7.79 (s, 2H), 7.24 (s, 2H), 3.88 (s, 6H), 3.85 (s, 6H).

1 H NMR (400 MHz, DMSO) δ 12.21 (br s, 2H), 8,44 (s, 2H), 8.39 (s, 2H), 7.83 (d, J = 8.2 Hz, 4H), 7.30 (d, J = 8.2 Hz, 4H), 2.63 (t, J = 7.7 Hz, 4H), 1.61 - 1.52 (m, 4H)

NCL 136

1.36 - 1.26 (m, 4H), 0.90 (t, J = 7.3 Hz, 6H).

NCL 137 1 H NMR (400 MHz, DMSO) δ 12.94 (br s, 2H), 8.68 (s, 2H), 8.33 (s, 2H), 7.60 (d, J = 7.9 Hz, 4H)*, 7.49 (dd, J = 8.7, 7.4 Hz, 2H)*.

NCL 138 1 H NMR (400 MHz, DMSO) δ 12.12 (br s, 2H), 8.21 (s, 2H), 7.97 (s, 2H), 7.54 - 7.47 (m, 6H), 7.42 - 7.36 (m, 6H), 7.31 - 7.21 (m, 8 H), 6.84 (d, J = 9.8 Hz, 2H).

NCL 139 1 H NMR (400 MHz, DMSO) δ 12.65 (br s, 2H), 9.63 (d, J = 1.8 Hz, 2H), 8.88 - 8.58 (m, 6H), 8.13 - 8.01 (m, 4H), 7.88 - 7.79 (m, 2H), 7.68 (t, J = 7.4 Hz, 2H).

NCL 140 1 H NMR (400 MHz, DMSO) δ 12.17 (br s, 2H), 8.46 (s, 2H), 8.37 (s, 2H), 7.86 (d, J = 8.2 Hz, 4H), 7.34 (d, J = 8.2 Hz, 4H), 2.53 (s, 6H).

NCL 141 1 H NMR (400 MHz, DMSO) δ 8.66 (s, 2H), 8.60 (d, J = 1.9 Hz, 2H), 8.52 (br s, 2H), 8.49 (s, 2H), 8.09 (d, J = 8.6 Hz, 2H), 7.50 (dd, J = 8.6, 2.0 Hz, 2H).

NCL 142 H NMR (400 MHz, DMSO) δ 8.80 (s, 2H), 7.41 - 7.20 (m, 12H), 5.56 (s, 2H), 3.79 (d, J = 3.4 Hz, 4H).

NCL 143 1 H NMR (400 MHz, DMSO) δ 1 .91 (br s, 2H), 8.66 (br s, 2H), 8.10 - 8.00 (m, 4H), 7.51 - 7.41 (m, 6H), 2.45 (s, 6H).

NCL 144 H NMR (400 MHz, DMSO) δ 12.36 (br s, 2H), 8.38 - 8.23 (m, 4H), 7.16 (d, J = 3.5 Hz, 2H), 6.82 (d, J = 3.5 Hz, 2H).

NCL 145 H NMR (400 MHz, DMSO) δ 12.41 (br s, 2H), 8.41 - 8.21 (m, 4H), 7.20 (d, J = 3.5 Hz, 2H), 6.73 (d, J = 3.5 Hz, 2H).

H NMR (400 MHz, DMSO) δ 11.97 (br s, 2H), .45 (s, 2H), 8.47 (s, 2H), 8.30 (s, 2H), 8.02 (s, 2H), 7.80 (dd, J = 8.6, 0.9 Hz, 2H), 7.48 (d, J = 8.5 Hz, 2H), 7.45 - 7.4 NCL 146 (m, 2H), 6.53 (s, 2H).

13C NMR (101 MHz, DMSO) δ 152.4, 150.6, 137.4, 127.6, 126.7, 124.5, 121.8, 120.3, 111.9, 102.0.

NCL 147 H NMR (400 MHz, DMSO) δ 13.09 (br s, 2H), 9.97 (s, 2H), 9.01 (s, 2H), 8.73 (s, 2H), 8.19 - 8.09 (m, 4H), 7.95 - 7.84 (m, 4H).

κ,^ι 1Η NMR (400 MHz, DMSO) δ 12.44 (s, 4H), 8.60 (s, 2H), 8.44 (s, 2H), 7.99 (d, J = 8.3 Hz, 4H), 7.81 (d, J = 8.3 Hz, 4H), 7.63 (d, J = 16.0 Hz, 2H), 6.66 (d, J = 16.0 H N L 148

NCL 149 1H NMR (400 MHz, DMSO) δ 8.75 (d, J = 6.1 Hz, 4H), 8.68 (s, 2H), 8.47 (s, 2H), 8.07 (d, J = 6.1 Hz, 4H).

1H NMR (400 MHz, DMSO) δ 12.10 (brs,2H), 8.23-8.07 (m, 4H), 7.55 (d, J =8.7 Hz, 4H), 7.13 (d, J = 16.0 Hz, 2H), 6.99 (d, J = 8.7 Hz, 4H), 6.81 (dd, J = 16.0, 9. NUL10U HZi 2H), 3.79 (S, 6H).

NCL 151 1H NMR (400 MHz, DMSO) δ 11.92 (brs, 2H), 10.13 (brs, 2H), 8.28 (s, 4H), 7.75 (d, J = 8.5 Hz, 4H), 6.86 (d, J = 8.5 Hz, 4H).

1H NMR (400 MHz, DMSO) δ 12.08 (brs, 2H), 9.35 (s, 2H), 8.66 (s, 2H), 8.48 (s, 2H), 7.67 (d, J = 7.2 Hz, 2H), 7.25 (d, J = 7.2 Hz, 2H), 6.88 (t, J = 7.6 Hz, 2H), 2.23 (

NOL ID.:

NCL 153 1H NMR (400 MHz, DMSO) δ 11.68 (s,2H),8.78 (s, 2H), 8.09 (d, J = 8.4 Hz, 4H), 7.52 (d, J = 8.6 Hz, 4H), 2.92 (q, J = 7.5 Hz, 4H), 1.12 (t, J = 7.4 Hz, 6H).

1H NMR (400 MHz, DMSO) δ 12.09 (s, 2H), 8.69 (s, 2H), 8.07 (d, J = 8.6 Hz, 4H), 7.50 (d, J = 8.6 Hz, 4H), 3.01 -2.88 (m, 4H), 1.49-1.39 (m, 8H), 0.88 (t, J = 6.6 H NOL 1i>4 6H).

NCL 155 1 H NMR (400 MHz, DMSO) δ 11.63 (s, 2H), 8.76 (s, 2H), 8.01 (d, J = 8.6 Hz, 4H), 7.65 (d, J = 8.6 Hz, 4H), 2.41 (s, 6H).

NCL 156 1H NMR (40 ° MHz ' DMS °) 512 - 18 (s, 2H),8.71 (s, 2H), 8.08 (d, J =8.0 Hz, 4H), 7.50 (d, J = 8.3 Hz, 4H), 3.01 -2.87 (m, 4H), 1.58- 1.46 (m, 4H), 1.01 (t, J = 7.1 H

6H).

1H NMR (400 MHz, DMSO) δ 11.71 (brs, 2H), 8.40 (s, 2H), 8.37 (s, 2H), 7.29 (d, J = 8.4 Hz, 2H), 6.87 (d, J = 2.0 Hz, 2H), 6.73 (br s, 4H), 6.59 (dd, J = 8.3, 2.0 Hz, 2H) 13C NMR (101 MHz, DMSO) δ 152.1, 151.5, 148.9, 136.0, 134.7, 115.1, 114.5, 112.8.

1H NMR (400 MHz, DMSO) δ 13.03 (brs, 1H), 10.49 (brs, 1H), 9.35 (brs, 1H), 7.24 (d, J = 7.6 Hz, 2H), 7.11 -6.61 (m, 6H), 2.5 (Contains CH2 groups determined b COSY, however eclipsed by solvent signal), 0.93 (s, 6H).

NCL 158 13CNMR(101 MHz, DMSO) δ 157.8, 155.4, 152.9, 134.4, 130.1, 118.7, 115.6,29.9, 10.8.

*COSY was used to determine that the signal due to the methylene protons appears under the DMSO signal. Line broadening is apparent in the 13C-NMR (due t tautomerisation effects) making carbon allocation difficult.

NCL 159 1H NMR (400 MHz, DMSO) δ 12.69 (brs, 1H), 10.44 (brs, 1H), 9.15 (brs, 1H), 7.21 (s, 2H), 7.09-6.60 (m, 6H), 2.98 (p, J = 7.7 Hz, 2H), 1.84-1.01 (m, 16H).

NCL 160 1H NMR (400 MHz, DMSO) δ 12.49 (brs, 2H), 8.65 (s, 2H), 8.49 (s, 2H), 8.10 (d, J = 8.7 Hz, 4H) * , 7.47 (d, J = 8.3 Hz, 4H) * .

NCL 161 1 H NMR (400 MHz, DMSO) δ 7.73 (d, J = 8.8 Hz, 2H), 6.92 (d, J = 8.9 Hz, 2H), 3.22 - 3.13 (m, 4H), 2.99 - 2.86 (m, 4H), 2.24 (s, 3H).

NCL162 1H NMR (400 MHz, CDCI3) δ 7.76-7.70 (m, 2H), 7.35 - 7.30 (m, 2H), 4.99 (s, 2H), 4.34 (q, J = 7.1 Hz, 2H), 2.39 (s, 3H), 1.35 (t, J = 7.1 Hz, 3H).

1H NMR ( 40 ° MHz > DMSO) δ 13.88 (s, 1H), 13.21 (s, 1H), 11.50 (s, 2H), 9.70 (s, 2H), 7.70 (s, 2H), 7.43 (t, J = 7.7 Hz, 2H), 7.14 (t, J = 7.5 Hz, 2H), 7.00 (d, J = 7.8 H

AH).

ΝΓΙ 1R4 1H NMR ( 40 ° MHz ' DMS °) 5 0 - 85 (s, 2H), 9.38 (s, 2H), 8.64 (s, 3H), 7.90 (s, 3H), 7.34 (d, J = 8.5 Hz, 2H), 7.19 (s, 3H), 6.83 (d, J = 2.1 Hz, 2H), 6.57 (dd, J = 8.5, 2.

N _1b4 H2j 2H)

NCL165 1H NMR (400 MHz, DMSO) δ 10.87 (brs, 1H), 9.53 (brs, 1H), 8.77 (brs, 3H), 7.61-7.44 (m, 8H), 7.40 (brs, 3H), 7.27 (brs, 3H), 4.07 (brs, 2H).

NCL 66 1H NMR (400 MHz, DMSO) δ 12.70 (br s, 2H), 8.75 (br s, 2H), 8.55 (br s, 2H), 8.13 (d, J = 8.2 Hz, 4H), 7.81 (d, J =8.1 Hz, 4H).

NCL167 1H NMR (400 MHz, DMSO) δ 14.09 (s, 1H), 8.12 (d, J = 6.9 Hz, 2H), 7.97 (dd, J = 6.4, 3.0 Hz, 2H), 7.56 (ddd, J = 8.2, 6.7, 2.9 Hz, 7H).

1H NMR (400 MHz, DMSO) δ 11.86 (brs, 2H), 8.42 (brs, 4H), 7.35 (d, J = 6.8 Hz, 4H), 6.75 (s, 2H), 6.67 (d, J = 7.9 Hz, 2H), 3.44 (d, J = 6.9 Hz, 2H)*, 1.24 (t, J = 5. NOUbb 6H) * g jgna| part|y edipsed by in DMSO

NCL170 1 H NMR (400 MHz, DMSO) δ 12.29 (s, 2H), 10.29 (s, 2H), 8.55 (s, 2H), 8.45 (s, 2H), 8.13 (d, J = 8.5 Hz, 2H), 7.71 (s, 2H), 7.35 (dd, J = 8.5, 1.8 Hz, 2H), 2.12 (s, 6H).

1 H NMR (400 MHz, DMSO) δ 11.63 (s, 2H), 9.84 (s, 2H), 8.46 (s, 2H), 8.02 (s, 2H), 7.74 (d, J = 8.8 Hz, 2H), 6.30 (d, J = 7.4 Hz * , 2H), 6.17 (s, 2H), 2.94 (s, 12H)

NCL171

*Poorly resolved doublet gives reduced coupling constant.

1 H NMR (400 MHz, DMSO) δ 12.00 (s, 2H), 8.92 (s, 2H), 8.64 (dd, J = 4.7, 0.6 Hz, 2H), 8.57 (d, J = 8.1 Hz, 2H), 7.89 (td, J = 8.0, 1.6 Hz, 2H), 7.50 - 7.41 (m, 2H), 2.5

NCL172

(s, 6H).

H NMR (400 MHz, DMSO) δ 11.49 (br s, 2H), 10,62 (br s, 2H), 8.55 (br s, 2H), 7.59 (d, J = 8.3 Hz, 2H), 6.98 (d, J = 1.9 Hz, 2H), 6.92 (dd, J = 8.4, 2.0 Hz, 2H), 2.37 (s NCL 173 6H).

NCL174 1 H NMR (400 MHz, DMSO) δ 12.02 (s, 2H), 10.81 (s, 2H), 8.63 (s, 2H), 8.38 (s, 2H), 8.12 (d, J = 8.3 Hz, 2H), 7.13 - 6.84 (m, 4H).

NCL 175 1H NMR (400 MHz, DMSO) δ 12.63 (br s, 2H), 8.90 (d, J = 2.1 Hz, 2H), 8.74 (s, 2H), 8.56 - 8.42 (m, 4H), 7.66 (d, J = 8.4 Hz, 2H).

1 H NMR (400 MHz, DMSO) δ 11.94 (br s, 2H), 8.44 (s, 2H), 8.36 (s, 2H), 8.07 (dd, J = 4.8, 1.7 Hz, 2H), 7.72 (dd, J = 7.6, 1.4 Hz, 2H), 7.19 (s, 4H), 6.67 (dd, J = 7.5, 4. NCL 176

Hz, 2H).

NCL 177 1 H NMR (400 MHz, DMSO) δ 8.67 (s, 2H), 7.97 (s, 4H), 7.50 (d, J = 8.6 Hz, 4H), 4.81 (s, 4H).

NCL178 1 H NMR (400 MHz, DMSO) δ 10.17 (s, 2H), 8.24 (s, 1H), 7.83 (d, J = 8.6 Hz, 4H), 7.50 (d, J = 8.6 Hz, 4H), 6.97 (s, 1 H), 2.32 (s, 6H).

1 H NMR (400 MHz, DMSO) δ 10.70 (s, 2H), 8.02 (s, 2H), 7.67 (d, J = 8.4 Hz, 4H), 7.52 (d, J = 8.4 Hz, 4H), 6.28 (s, 1 H), 5.85 (s, 2H).

NCL179

13C NMR (101 MHz, DMSO) δ 162.8, 162.6, 138.8, 134.1, 133.1 , 128.9, 127.6, 73.5.

1 H NMR (400 MHz, DMSO) δ 10.62 (s, 2H), 8.22 (d, J = 0.9 Hz, 1 H), 7.82 - 7.74 (m, 4H), 7.53 - 7.47 (m, 4H), 6.93 (d, J = 0.6 Hz, 1H), 5.85 (t, J = 5.3 Hz, 2H), 4.74 (d

NCL180

J = 5.2 Hz, 4H).

NCL181 1 H NMR (400 MHz, DMSO) δ 11.20 (s, 2H), 8.17 (s, 1H), 8.09 (s, 2H), 7.72 (d, J = 7.4 Hz, 4H), 7.54 (d, J = 7.6 Hz, 4H), 6.83 (s, 1H).

NCL182 1 H NMR (400 MHz, CDCI3) δ 7.44 - 7.19 (m, 6H), 5.67 (s, 1H), 5.42 (s, 1 H), 5.06 (s, 2H), 4.95 - 4.63 (m, 1 H), 1.52 (d, J = 6.8 Hz, 3H).

NCL183 1H NMR (400 MHz, CDC13) δ 8.28 (s, 1H), 7.43 - 7.13 (m, 10H), 6.19 (s, 1H), 5.94 (s, 1H), 4.95 - 4.48 (m, 2H), 1.56 (d, J = 6.8 Hz, 4H), 1.50 (d, J = 6.5 Hz, 2H). NCL184 1 H NMR (400 MHz, MeOD) δ 7.44 - 7.04 (m, 10H), 5.26 - 4.53 (m, 7H), 1.51 - 1.35 (m, 6H).

1 H NMR (400 MHz, DMSO) δ 11.54 (s, 1 H), 7.99 (d, J = 8.7 Hz, 2H), 7.90 (s, 3H), 7.47 (d, J = 8.6 Hz, 2H), 2.91 - 2.82 (m, 2H), .48 - 1.32 (m, 4H), 0.89 - 0.84 (m, 3H)

NCL188

13C NMR (101 MHz, DMSO) δ 156.2, 153.8, 134.8, 134.4, 128.7, 128.4, 28.1 , 26.6, 22.0, 13.8.

NCL190 1 H NMR (400 MHz, DMSO) δ 11.42 (s, 1 H), 8.06 (d, J = 8.7 Hz, 3H), 8.01 - 7.71 (m, 5H), 7.53 (d, J = 8.7 Hz, 3H), 4.90 (s, 2H), 3.69 (br s, 3H).

NCL191 1 H NMR (400 MHz, DMSO) δ 11.51 (s, 1 H), 8.85 (s, 3H), 7.99 (d, J = 8.6 Hz, 7H), 7.46 (d, J = 8.6 Hz, 4H), 2.35 (s, 3H).

NCL192 1 H NMR (400 MHz, DMSO) δ 11.65 (s, 1 H), 8.21 (s, 4H), 7.82 (dd, J = 7.6, 1.9 Hz, 2H), 7.53 - 7.39 (m, 3H).

1 H NMR (400 MHz, DMSO) δ 10.71 (s, 2H), 8.00 (s, 2H), 7.66 (d, J = 8.5 Hz, 4H), 7.60 (d, J = 8.6 Hz, 4H), 6.27 (s, 1 H), 5.86 (s, 2H).

NCL193

13C NMR (101 MHz, DMSO) δ 162.7, 162.6, 138.8, 134.5, 131.8, 127.9, 121.7, 73.5.

NCL194 1 H NMR (400 MHz, DMSO) δ 12.01 (br s, 1 H), 10.84 (s, 2H), 9.98 (br s, 1 H), 9.79 (br s, 1 H), 7.96 (br s, 3H), 7.79 (br s, 2H), 7.67 - 7.37 (m, 6H), 5.37 (br s, 1 H).

H NMR (400 MHz, DMSO) δ 10.51 (s, 2H), 8.00 (s, 2H), 7.54 (d, J = 8.0 Hz, 4H), 7.26 (d, J = 7.9 Hz, 4H), 6.26 (s, 1H), 5.77 (s, 2H), 2.34 (s, 6H).

NCL195

13C NMR (101 MHz, DMSO) δ 162.8, 162.6, 140.1 , 138.4, 132.5, 129.4, 126.0, 73.3, 21.0 .

1H NMR (400 MHz, CDCI3) δ 10.31 (s, 2H), 9.74 (s, 2H), 7.94 (s, 2H), 7.48 (d, J = 8.6 Hz, 4H), 6.83 (d, J = 8.6 Hz, 4H), 6.20 (s, 1H), 5.70 (s, 2H).

NCL196

13C NMR (101 MHz, CDCI3) δ 162.7, 162.5, 158.3, 140.5, 127.7, 126.3, 115.7, 73.0.

1 H NMR (400 MHz, DMSO) δ 10.51 (s, 2H), 9.55 (s, 2H), 7.95 (s, 2H), 7.22 (t, J = 7.9 Hz, 2H), 7.11 - 7.04 (m, 4H), 6.76 (d, J = 8.4 Hz, 2H), 6.23 (s, 1H), 5.80 (s, 2H).

NCL197

13C NMR (101 MHz, DMSO) δ 162.8, 162.6, 157.7, 140.4, 136.4, 129.9, 117.4, 116.1 , 112.4, 73.3.

1 H NMR (400 MHz, DMSO) δ 10.60 (s, 2H), 8.04 (s, 2H), 7.66 (d, J = 7.5 Hz, 4H), 7.45 (t, J = 7.1 Hz, 4H), 7.40 - 7.34 (m, 2H), 6.30 (s, 1 H), 5.82 (s, 2H).

NCL199

13C NMR (101 MHz, DMSO) δ 163.3, 163.1 , 140.5, 135.7, 129.3, 129.2, 126.5, 73.9.

NCL200 1H NMR (400 MHz, DMSO) δ 10.51 (s, 2H), 8.02 (s, 2H), 7.58 (d, J = 8.3 Hz, 4H), 7.47 (d, J = 8.3 Hz, 4H), 6.25 (s, 1H), 5.77 (s, 2H), 1.31 (s, 18H).

NCL201 1H NMR (400 MHz, DMSO) δ 10.19 (s, 2H), 7.91 (s, 2H), 7.47 (d, J = 8.8 Hz, 4H), 6.77 (d, J = 8.9 Hz, 4H), 6.16 (s, 1 H), 5.63 (s, 2H).

1H NMR (600 MHz, DMSO) δ 13.43 (s, 1 H), 11.45 (s, 2H), 10.28 (br. s, 1 H), 9.47 (s, 1 H), 8.38 (s, 2H), 7.71 (dd, J = 7.7, 1.3 Hz, 1 H), 7.67 (br d, J = 7.0 Hz, 2H), 7.53 - 7.48 (m, 1 H), 7.28 - 7.23 (m, 2H), 7.03 - 6.98 (m, 2H), 6.96 - 6.90 (m, 4H), 6.56 (br S, H).

NCL202 *Due to tautomers and rotamers associated with proximity of the phenol to the hydrazone >17 protons are observed.

13C NMR (151 MHz, DMSO) δ 167.7, 163.2, 162.6, 156.7*, 141.4*, 135.3, 133.9, 131.0, 130.5, 120.7*, 119.95, 119.8, 118.9, 117.6, 116.7, 116.6 * .

*Line broadening due to tautomers and rotamers made signals difficult to assign.

NCL203 1H NMR (400 MHz, DMSO) δ 12.60 - 10.94 (m, 3H), 7.79 - 7.39 (m, 3H), 2.25 (s, 2H), 1.82 - 1.59 (m, 10H), 1.35 - 1.15 (m, 10H).

. 1 H NMR (400 MHz, DMSO) δ 9.58 (d, J = 7.6 Hz, 2H), 7.82 (s, 4H), 7.50 (s, 4H), 6.45 (d, J = 9.6 Hz, 1 H), 5.75 (s, 2H), 2.28 (d, J = 9.3 Hz, 6H).

NL,L_iU4 13C NMR ^ 101 MHz DMS0) δ 63 _ 48 162.11 , 143.34, 137.68, 132.88, 128.34, 127.16, 75.15, 13.01.

1 H NMR (600 MHz, DMSO) δ 10.92 (s, 2H), 8.42 (s, 2H), 7.98 (d, J = 7.5 Hz, 2H), 7.50 (d, J = 7.8 Hz, 2H), 7.44 (t, J = 7.4 Hz, 2H), 7.38 (t, J = 7.3 Hz, 2H), 6.35 (s, 1 H)

NCL205 5.95 (s, 2H).

13C NMR (151 MHz, DMSO) δ 162.75, 162.68, 136.0, 132.4, 132.0, 130.1 , 129.9, 127.6, 126.2, 73.7.

NCL206 1 H NMR (400 MHz, DMSO) δ 10.75 (s, 2H), 8.09 (s, 2H), 7.52 (d, J = 8.1 Hz, 4H), 7.24 (d, J = 8.0 Hz, 4H), 6.71 (s, 1 H), 2.33 (s, 6H).

NCL207 1 H NMR (400 MHz, DMSO) δ 11.43 (s, 2H), .05 (s, 2H), 8.31 (s, 2H), 7.42 (s, 2H), 7.29 - 7.18 (m, 2H), 6.89 (t, J = 7.7 Hz, 4H), 6.74 (s, 2H).

NCL208 1 H NMR (400 MHz, DMSO) δ 9.63 (s, 2H), 7.82 (d, J = 8.5 Hz, 4H), 7.46 (d, J = 8.5 Hz, 4H), 6.73 (s, 2H), 2.29 (s, 6H).

NCL209 1 H NMR (400 MHz, DMSO) δ 10.75 (s, 2H), 9.55 (s, 2H), 8.04 (s, 2H), 7.21 (t, J = 7.8 Hz, 2H), 7.06 (s, 2H), 7.02 (d, J = 7.6 Hz, 2H), 6.84 - 6.57 (m, 4H).

NCL210 1 H NMR (400 MHz, DMSO) δ 11.16 (s, 2H), 8.22 (s, 2H), 7.85 (d, J = 8.3 Hz, 4H), 7.80 (d, J = 8.5 Hz, 4H), 6.92 (s, 2H).

NCL211 1H NMR (400 MHz, DMSO) δ 10.56 (s, 2H), 9.78 (s, 2H), 8.02 (s, 2H), 7.45 (d, J = 8.6 Hz, 4H), 6.80 (d, J = 8.6 Hz, 4H), 6.62 (s, 2H).

NCL212 1 H NMR (400 MHz, DMSO) δ 10.94 (s, 2H), 8.10 (s, 2H), 7.67 - 7.60 (m, 4H), 7.61 - 7.54 (m, 4H), 6.80 (s, 2H).

NCL213 1 H NMR (400 MHz, DMSO) δ 10.23 (br S, 2H), 7.27 (s, 2H), 2.27 - 1.98 (m, 2H), 1.78 - 1.42 (m, 10H), 1.33 - 1.00 (m, 10H).

NCL214 1 H NMR (400 MHz, DMSO) δ 10.84 (s, 2H), 8.14 (s, 2H), 7.63 (d, J = 7.4 Hz, 4H), 7.46 - 7.33 (m, 3H), 6.76 (s, 2H).

NCL215 1 H NMR (600 MHz, DMSO) δ 12.06 (s, 2H), 8.72 (s, 2H), 7.97 (t, J = 8.4 Hz, 2H), 7.56 (dd, J = 11.1 , 1.9 Hz, 2H), 7.38 (dd, J = 8.5, 1.9 Hz, 2H), 2.42 (d, J = 2.9 Hz, 6H).

1 H NMR (400 MHz, DMSO) δ 12.43 (br s, 2H), 8.66 (br s, 2H), 8.62 (br s, 2H), 8.38 (t, J = 8.3 Hz, 2H), 7.61 (dd, J = 10.5, 1.9 Hz, 2H), 7.45 (dd, J = 8.6, 1.6 Hz, 2H).

NCL216 13C NMR (101 MHz, DMSO) δ 160.7 (d, J = 254.5 Hz), 152.8*, 140.8 * , 136.3 (d, J = 10.8 Hz), 128.5, 125.3, 120.2 (d, J = 10.0 Hz), 116.7 (d, J = 24.7 Hz).

*Broad signals

1H NMR (400 MHz, DMSO) δ 11.66 (s, 2H), 8.61 (s, 2H), 7.94 (d, J = 8.2 Hz, 4H), 7.25 (d, J = 8.1 Hz, 4H), 2.41 (s, 6H), 2.35 (s, 6H).

NCL217

13C NMR (101 MHz, DMSO) δ 154.0, 153.3, 139.7, 133 9, 129.0, 127.0, 21.0, 14.9.

1 H NMR (400 MHz, DMSO) δ 10.73 (s, 2H), 8.05 (s, 2H), 7.70 (d, J = 8.58 Hz, 4H), 7.31 - 7.27 (m, 4H), 6.24 (s, 1H), 5.92 (s, 2H), 4.17 (dq, J = 7.06, 8.70 Hz, 8H), 1.2 NCL218

(td, J = 1.01 , 7.05 Hz, 12H).

1 H NMR (400 MHz, DMSO) δ 11.74 (s, 2H), 8.60 (s, 2H), 7.95 (d, J = 8.6 Hz, 4H), 7.45 (d, J = 8.6 Hz, 4H), 2.42 (s, 6H), 1.31 (s, 18H).

NCL219

13C MR (101 MHz, DMSO) 6 154.1 , 153.3, 152.6, 134.0, 126.8, 125.0, 34.5, 31.0, 14.9.

NCL220 1 H NMR (400 MHz, DMSO) δ 10.61 (s, 2H), 8.03 (s, 2H), 7.70 (dd, J = 8.7, 5.6 Hz, 4H), 7.29 (t * , J = 8.9 Hz, 4H), 6.27 (s, 1 H), 5.82 (s, 2H).

NCL221 1 H NMR (600 MHz, DMSO) δ 10.89 (s, 2H), 8.11 (s, 2H), 7.86 (d, J = 8.2 Hz, 4H), 7.81 (d, J = 8.4 Hz, 4H), 6.34 (s, 1 H), 5.94 (s, 2H).

NCL222 1 H NMR (400 MHz, DMSO) δ 10.76 (s, 2H), 7.99 (s, 2H), 7.70 (td, J = 1.7, 9.2 Hz, 2H), 7.54 - 7.41 (m, 4H), 6.30 (s, 1 H), 5.87 (s, 2H).

NCL223 1 H NMR (400 MHz, DMSO) δ 10.48 (s, 2H), 10.07 (s, 2H), 7.98 (s, 2H), 7.65 (d, J = 8.7 Hz, 4H), 7.58 (d, J = 8.7 Hz, 4H), 6.24 (s, 1 H), 5.76 (s, 2H), 2.07 (s, 6H).

NCL224 1 H NMR (400 MHz, DMSO) δ 8.36 (s, 1 H), 7.96 - 7.90 (m, 2H), 7.86 (s, 1 H), 7.79 - 7.73 (m, 2H), 7.54 - 7.47 (m, 6H), 4.26 (q, J = 7.1 Hz, 2H), 1.27 (t, J = 7.1 Hz, 3H).

1H NMR (400 MHz, DMSO) δ 8.35 (s, 1H), 7.98 - 7.91 (m, 2H), 7.87 (s, 1H), 7.79 - 7.74 (m, 2H), 7.51 (dd, J = 2.8, 8.5 Hz, 6H), 3.99 (d, J = 6.5 Hz, 2H), 1.95 (hept, J

NCL225 6.7 Hz, 1 H), 0.93 (d, J = 6.7 Hz, 6H).

1 H NMR (400 MHz, DMSO) δ 10.95 (s, 1 H), 9.18 (t, J = 5.6 Hz, 1 H), 8.38 (d, J = 10.1 Hz, 2H), 8.32 (s, 1 H), 7.99 - 7.92 (m, 2H), 7.83 (d, J = 8.5 Hz, 2H), 7.57 - 7.46 (m NCL226

5H), 3.30 - 3.20 (m, 2H), 1.14 (t, J = 7.2 Hz, 3H).

1 H NMR (400 MHz, DMSO) δ 11.00 (s, 1 H), 9.68 (s, 1 H), 8.52 (s, 1 H), 8.41 (s, 1 H), 8.32 (s, 1 H), 8.01 - 7.80 (m, 4H), 7.56 - 7.46 (m, 4H), 7.36 (d, J = 6.00 Hz, 4H) NCL227

7.30 - 7.21 (m, 1 H), 4.56 - 4.33 (m, 2H).

1 H NMR (400 MHz, DMSO) δ 10.95 (s, 1 H), 9.22 (t, J = 5.6 Hz, 1 H), 8.40 (s, H), 8.34 (s, 1 H), 8.32 (s, 1H), 7.98 - 7.90 (m, 2H), 7.87 - 7.80 (m, 2H), 7.55 - 7.48 (m NCL228

4H), 3.22 (q, J = 6.6 Hz, 2H), 1.56 - 1.46 (m, 2H), 1.39 - 1.25 (m, 6H), 0.88 (t, J = 6.6 Hz, 3H).

1 H NMR (400 MHz, DMSO) δ 10.98 (s, 1 H), 9.60 (s, 1H), 8.54 (s, 1H), 8.40 (s, 1H), 8.30 (s, 1 H), 8.01 - 7.89 (m, 2H), 7.85 (d, J = 8.4 Hz, 2H), 7.64 - 7.59 (m, 1 H), 7.5 NCL229

- 7.47 (m, 4H), 6.42 (dd, J = 1.8, 3.3 Hz, 1 H), 6.31 (m, 1 H), 4.44 (d, J = 5.5 Hz, 2H).

NCL230 1 H NMR (400 MHz, DMSO) δ 10.41 (s, 2H), 7.98 (s, 2H), 7.59 (d, J = 8.9 Hz, 4H), 7.02 (d, J = 8.9 Hz, 4H), 6.23 (s, 1 H), 5.73 (s, 2H), 3.80 (s, 6H).

EXAMPLE 12: Treatment of a bacterial infection in vivo by the administration of NCL812 or NCL099.

[00385] The objective of this study was to determine the efficacy of an Investigational Veterinary Product containing NCL812 or NCL099 in the treatment of a skin infection in mice

[00386] Summary of the Model: A useful animal model system should be clinically relevant, experimentally robust, ethically acceptable, convenient to perform and should provide reliable and reproducible results. There are many animal models of topical skin infection that have been described including the croton oil-inflamed skin model (Akiyama, H. , H. Kanzaki, Y. Abe, J. Tada and J. Arata (1994). "Staphylococcus aureus infection on experimental croton oil- inflamed skin in mice." Journal of Dermatological Science 8(1 ): 1-10), the burnt skin model (Stieritz, D. D., A. Bondi, D. McDermott and E. B. Michaels (1982). "A burned mouse model to evaluate anti-pseudomonas activity of topical agents." Journal of Antimicrobial Chemotherapy 9(2): 133-140), the skin suture-wound model (McRipley, R. J. and R. R. Whitney (1976). "Characterization and Quantitation of Experimental Surgical-Wound Infections Used to Evaluate Topical Antibacterial Agents." Antimicrobial Agents and Chemotherapy 10(1 ): 38-44), the skin tape-stripping model (Kugelberg, E., T. Norstrom, T. K. Petersen, T. Duvold, D. I. Andersson and D. Hughes (2005). "Establishment of a Superficial Skin Infection Model in Mice by Using Staphylococcus aureus and Streptococcus pyogenes." Antimicrobial Agents and Chemotherapy 49(8): 3435-3441) and the linear full thickness scalpel cut method (Guo, Y. , R. I. Ramos, J. S. Cho, N. P. Donegan, A. L. Cheung and L. S. Miller (2013). "In Vivo Bioluminescence Imaging To Evaluate Systemic and Topical Antibiotics against Community-Acquired Methicillin-Resistant Staphylococcus aureus-lnfected Skin Wounds in Mice." Antimicrobial Agents and Chemotherapy 57(2): 855-863)

[00387] Preliminary studies prior to the conduct of the current study established a new method of skin infection arising from a detailed study of the models mentioned above. Briefly, study mice are anaesthetised, a patch of dorsal skin is clipped to reveal the skin and a circular area of skin is removed with a hand held punch, leaving a wound on the dorsum with a central cavity. The wound is infected with a known number of the challenge organism. Approximately four to six hours after infection, the wound is either treated topically with a vehicle formulation or an active formulation. The infected skin wound is retreated every 12 hours for a total of 14 treatments. Mice are humanely euthanased, the area of the original infected wound is dissected and removed and its bacterial content quantified by standard microbiologic tests. In this way, the change in bacterial concentration due to treatment with the active formulation can be readily determined by examining the reduction in bacterial burden compared with the vehicle control. Materials and Methods

Preparation of Infection Inoculum

[00388] Fresh cultures of bacteria (Staphylococcus aureus) were grown on Sheep Blood Agar at 37 °C for 16 - 18 hours. A few typical colonies were selected and suspended in 10 ml of Tryptic Soy Broth and incubated overnight in a shaking incubator (240 rpm) at 37 °C. The overnight suspension was vortexed and diluted (1 : 100) in fresh Tryptic soy broth (100 μΙ [0.1 ml] in 9.9 ml broth). The fresh suspension was incubated for 3 hours in a shaking incubator (as above) in order to obtain mid-logarithmic phase bacteria. Bacteria were pelleted through centrifugation at 7,500 rpm for 10 mins. Broth supernatant was removed and bacteria suspended in 10 ml Phosphate Buffered Saline (PBS). These steps were repeated a further two times. The density of the suspension was checked by measuring absorbance at 600 nm, using a spectrophotometer with saline as a blank, to confirm the target density at a reading of approximately 0.100, consistent with a bacterial density of 2.5 x 10 7 CFU/ml. The suspension was placed into a rack placed into a lockable transport box with an ice brick to maintain refrigeration during transport, followed by storage in cool room upon arrival at the mouse skin infection laboratory. Final suspension was mixed thoroughly before inoculating the skin wounds created in mice.

[00389] I n order to ensure the purity and accuracy of the suspension, the following steps were performed prior to placement into lock box.

[00390] Purity of bacterial suspension ensured by spreading 100 μΙ of the final suspension onto a SBA (sheep blood agar) plate which was incubated at 37 °C for 18 hours and examined to confirm uniform growth of one colony type. Viable counts were performed on final suspension by prepping saline in Eppendorf tubes (approximately 900 μΙ per tube), removing 100 μΙ sample and adding to first Eppendorf tube, vortexing the mixture and repeating using 2 nd Eppendorf tube containing saline. This process was continued for 5 - 6 tubes. Finally, 100 μΙ of 5 th and 6 th dilutions were plated out on plate count agar, incubated at 37 °C for 18 hours and colony counts performed to confirm that the CFU/ml was approximately 2.5 x 10 7 . Following inoculation of the wounds, this process was repeated to ensure that no contamination or decrease in viable counts had occurred during the time of the surgery.

Skin Wound Surgical Procedure

[00391] Each mouse was placed into induction chamber and anaesthesia induced using 2% isoflurane. Eyes of each anaesthetised mouse were covered with veterinary eye lubricant in order to prevent corneal dehydration. Each mouse removed from induction chamber and placed onto surgical area, in front of individual aesthetic nose cone. While under anaesthesia each mouse was monitored for assessment of depth of anaesthesia (response to pain, blink reflex, skeletal muscle tone) and respiratory and cardiac function. Back skin hair was shaved from surgical area with mechanical clippers. Shaved area was cleaned using 70% ethanol applied to paper towel followed by 10% w/v povidone-iodine solution. Once the iodine solution was dry, a subcutaneous injection of the nonsteroidal anti-inflammatroy agent meloxicam was administered. Dorsal skin was pinched gently to allow creation of a circular full-thickness wound using ear punch/biopsy punch. Vehicle control and NCL812 and NCL099 mice had wounds inoculated with 10 μΙ of bacterial suspension using a micropipette (2.5 x 10 5 CFU/10 μΙ). Once the bacterial suspension was dry, mice were placed into individual recovery boxes labelled with the mouse number. The time of inoculation was recorded. Initial body weights of each mouse were recorded on the appropriate score sheet. Mice recovered to full consciousness within 5 minutes. Recovered mice were returned to individual housing and monitored hourly for postsurgical or anaesthetic complications.

Post-Surgical Care (4 hours post-surgery)

[00392] Mice were assessed for post-surgical complications and observations were recorded on clinic record sheet. Each mouse was carefully removed from IVC and placed into an assessment container, avoiding excessive handling or touching of the surgical site. Once the mouse was inside assessment container, it was assessed and the observations recorded on the post-surgical clinical record sheet. Whenever the suggested wellness breakpoints were reached, post-operative analgesia was administered and recorded on the clinical record sheet.

Animal Monitoring and Daily Care

[00393] Antibiotic Administration (7am and 6pm). The first administration of vehicle or NCL812 or NCL099 ointment occurred 4 hours post-surgically. Each ointment container was weighted prior to administration and the weight recorded. Each mouse was carefully restrained. Ointment (vehicle or NCL812 or NCL099) was applied to the lesion area and the treated mouse was returned to IVC where each mouse was observed to ensure ointment was not immediately removed by grooming. The weight of the ointment container post-administration was recorded. The vehicle and active NCL products were applied to the skin wound each 12 hours following the first administration for a total of 14 consecutive treatments. Both the NCL812 and NCL099 products (Formulation B, as presented in Example 9) contained their respective active ingredients at a concentration of 20 mg/g. Approximately 0.1 -0.2 g of ointment was applied on each occasion, delivering a total topical dose of NCL812 or NCL099 between 28 and 56 mg to mice weighing between 18 g and 25 g.

[00394] Daily Monitoring. Monitoring of each mouse took place once daily at around 12pm. Each mouse carefully removed from IVC and placed into observation container, avoiding excessive handling or touching surgical site. The coat, posture, eyes, behaviour, vocalisation and activity whilst in the container were carefully assessed and observations recorded on assessment sheet. Mouse faeces (either on floor of cage or in container) were checked for consistency and observations recorded. The weight of each mouse was determined whilst it was in the container and change in body weight calculated and recorded. The observation container was disinfected with ethanol and set aside to dry while a fresh container was used for the next mouse. For every second day, mice were again anaesthetised using 2% isoflurane and photographed using a ruler for size referencing. These photos were used to assess lesion size and infection progression during the trial period.

Tissue analysis and assessment of antibacterial efficacy

[00395] At the end of the 7 day skin wound assessment period, all test mice were euthanased prior to wound collection for post mortem examination. The skin wound was dissected from the dorsum of each mouse. The sample was placed in a sample tube and weighed before 1 ml PBS and sterile tissue homogenisation beads were added. Tissue samples were homogenised for 10 mins using a tissue homogeniser (Next Advance Bullet Blender) and then vortexed for approximately 30 seconds. 100 μΙ of supernatant was removed and placed into an Eppendorf tube containing 900 μΙ of PBS. This procedure was repeated using serial dilutions for a total of 8 dilutions. Finally, 100 μΙ of each dilution was pipetted onto a plate count agar in duplicate and incubated overnight at 37 °C. Ten microlitres of original suspension was placed onto sheep blood agar to assess culture purity and incubated overnight at 37 °C. The following day, viable counts were performed using incubated plate count agar plates and the identity of Staphylococcus aureus (the challenge organisms) as the harvested strain was confirmed.

Results

[00396] The mean colony count per g of tissue observed in vehicle treated group was 5,888,436 (6.77 log 10 ). The mean colony count per g of tissue observed in NCL812 group was 141 ,254 (5.15 log 0 ). The mean colony count per g of tissue observed in NCL099 treated mice was 1 ,318 (3.12log 10 ). The log 10 colony forming units per gram of tissue and % reduction are summarised in the following table. Table 47: Log 10 colony forming units per gram of tissue and percentage reduction following topical administration of vehicle and treatment.

[00397] It is clear from this table that treatment with either NCL812 or NCL099 leads to high level reductions in the number of infecting Staphylococcus aureus. These results demonstrate effective treatment of a bacterial colonisation or infection in vivo by administration of compounds of the invention.