CA2747995A1 - Antibiotic compositions for the treatment of gram negative infections - Google Patents

Antibiotic compositions for the treatment of gram negative infections Download PDF

Info

Publication number
CA2747995A1
CA2747995A1 CA2747995A CA2747995A CA2747995A1 CA 2747995 A1 CA2747995 A1 CA 2747995A1 CA 2747995 A CA2747995 A CA 2747995A CA 2747995 A CA2747995 A CA 2747995A CA 2747995 A1 CA2747995 A1 CA 2747995A1
Authority
CA
Canada
Prior art keywords
compound
formula
polymyxin
compounds
antibacterial
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA2747995A
Other languages
French (fr)
Inventor
Richard A. Leese
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Biosource Pharm Inc
Original Assignee
Biosource Pharm Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Biosource Pharm Inc filed Critical Biosource Pharm Inc
Publication of CA2747995A1 publication Critical patent/CA2747995A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07KPEPTIDES
    • C07K7/00Peptides having 5 to 20 amino acids in a fully defined sequence; Derivatives thereof
    • C07K7/50Cyclic peptides containing at least one abnormal peptide link
    • C07K7/54Cyclic peptides containing at least one abnormal peptide link with at least one abnormal peptide link in the ring
    • C07K7/60Cyclic peptides containing at least one abnormal peptide link with at least one abnormal peptide link in the ring the cyclisation occurring through the 4-amino group of 2,4-diamino-butanoic acid
    • C07K7/62Polymyxins; Related peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • A61K38/04Peptides having up to 20 amino acids in a fully defined sequence; Derivatives thereof
    • A61K38/12Cyclic peptides, e.g. bacitracins; Polymyxins; Gramicidins S, C; Tyrocidins A, B or C
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K38/00Medicinal preparations containing peptides
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • YGENERAL TAGGING OF NEW TECHNOLOGICAL DEVELOPMENTS; GENERAL TAGGING OF CROSS-SECTIONAL TECHNOLOGIES SPANNING OVER SEVERAL SECTIONS OF THE IPC; TECHNICAL SUBJECTS COVERED BY FORMER USPC CROSS-REFERENCE ART COLLECTIONS [XRACs] AND DIGESTS
    • Y02TECHNOLOGIES OR APPLICATIONS FOR MITIGATION OR ADAPTATION AGAINST CLIMATE CHANGE
    • Y02ATECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE
    • Y02A50/00TECHNOLOGIES FOR ADAPTATION TO CLIMATE CHANGE in human health protection, e.g. against extreme weather
    • Y02A50/30Against vector-borne diseases, e.g. mosquito-borne, fly-borne, tick-borne or waterborne diseases whose impact is exacerbated by climate change

Abstract

Provided herein are novel compounds and novel protected compounds that can be derived from polymyxin, including, e.g., polymyxin Bi and [lie7] polymyxin Bi. The novel compounds have antibacterial properties against a diverse range of gram negative bacteria and reduced toxicity compared to polymyxins such as polymyxin B. Also provided are antibacterial pharmaceutical compositions containing the novel compounds and novel protected compounds, as well as methods for preparing the antibacterial compounds and protected compounds.

Description

ANTIBIOTIC COMPOSITIONS FOR THE
TREATMENT OF GRAM NEGATIVE INFECTIONS
RELATED APPLICATIONS
[001] This application claims the benefit of U.S. provisional patent application 61/140,408, filed December 23, 2008, incorporated herein by reference in its entirety.

TECHNICAL FIELD
[002] This application describes antibacterial compounds, and related methods of treatment and methods of manufacture.

BACKGROUND
[003] Gram-negative bacteria that are resistant to aminoglycoside, [3-lactam, and fluoroquinolone antibiotics are increasingly common. These bacteria are often only susceptible to the polymyxins and related peptides having antibacterial properties. As a result, there is renewed interest in the use of polymyxins for the treatment of multidrug-resistant gram-negative bacterial infections in humans.
[004] Peptides such as polymyxin B and the related colistin (polymyxin E) have been administered to humans as antibacterial agents. However, their use has been previously limited because of their toxicity. These peptides comprise a seven amino acid cyclic peptide attached to an exocyclic three amino acid chain, wherein the N-terminal amine of the exocyclic chain is linked to a "side chain" or "tail". The tail is an acyl group.
[005] Renal toxicity has been observed with the recommended dosing of polymyxin B in some patients. Neurotoxicity or neuropathy has also been observed in patients with compromised renal functions, with an overall incidence of 7.3%
reported in one large study with colistin (see, e.g., Evans, et al. (1999) Ann.
Pharmacother. 33:960-967). When the acyl exocyclic chain and the adjacent N-terminal 2,4-diaminobutanoic acid (Dab) residue are enzymatically removed from polymyxin, this yields the corresponding polymyxin nonapeptide. The in vivo toxicity of the nonapeptide of polymyxin B is significantly less than that of polymyxin B itself (see, e.g., Kimura, et al. (1992) J. Antibiot., 45, 742-749). The toxicity of the nonapeptide in cell culture is reduced by about 100-fold relative to polymyxin B.
However, the antibacterial activity of the nonapeptide is also reduced by about 2 - 64 fold relative to polymyxin B (see, e.g., Duwe, et al. (1986) Antimicrob.
Agents Chemother, 30:340-341).
[006] Attempts have been made to chemically modify polymyxin and colistin in order to obtain peptides with improved antibacterial properties and reduced toxicity. For example, the total synthesis of polymyxin B and four analogues was previously accomplished by a combination of solid phase peptide syntheses to obtain linear structures, followed by removal from the resin and condensation in solution at high dilution to obtain the cyclic peptide structure (see, e.g., Tsubery (2001) Peptides. 22: 1675-1681). The derivatives, however, were less active than polymyxin B. A more recent total synthesis of polymyxin B and a few closely related compounds was accomplished only by solid phase peptide synthesis (see, e.g., DeVisser, et al. (2003) J. Peptide Res. 61, 298-306 and Kline, et al. (2001) J. Pept.
Res., 57: 175-187). Although both of these solid phase total synthetic approaches can provide new derivatives of polymyxin, these methods appear limited since the quantities of antibiotic produced are small and require large amounts of amino acid precursors. Any scale up of these methods for clinical studies may prove to be difficult and prohibitively costly. Further, neither of these methods is known to have produced novel derivatives of polymyxin having both improved antibacterial and toxicity properties relative to polymyxin B.
[007] Thus, there is a need for new peptide compounds having equivalent antibacterial properties to polymyxin B, but with significantly lower toxicity, such as renal toxicity, as well as methods of manufacturing such antibacterial compounds.
SUMMARY
[008] Antibacterial compounds of Formula (I) where R7 is an alkyl moiety such as isobutyl or sec-butyl have antibacterial activity against a diverse range of gram negative bacteria with unexpected lower renal toxicity than polymyxin compounds such as polymyxin B.

I
O

CI O O O
H H H

`'~
IfI H H
O
O
NH HN :CO
H

O
HO
[009] Pharmaceutical compositions containing the antibacterial compounds of Formula (I) are disclosed herein, as well as methods for preparing the antibacterial compounds, amine-protected analogs of the antibacterial compounds, and methods of using the antibacterial compounds. The antibacterial compounds of Formula (I) can be derived from a polymyxin such as polymyxin B, and [Ile7] polymyxin B1.
Pharmaceutical compositions comprising one or more compounds of Formula (I), or pharmaceutically acceptable salts and/or derivatives thereof, are useful, for example, in treating bacterial infections arising from a variety of gram negative pathogens.
Methods for treating an infection in a subject can include administering to the subject a therapeutically effective amount of at least one compound of Formula (I), or a pharmaceutically acceptable salt thereof. The antibacterial compounds can also be used in the manufacture of medicaments for treatment of infections.

BRIEF DESCRIPTIONS OF THE FIGURES
[010] Figure 1 is a synthetic scheme useful for preparing compounds of Formula (I) and other compounds.
[011] Figure 2 is a scatter plot of total renal histopathology scores in monkeys.

DETAILED DESCRIPTION
Definitions [012] As used herein and unless otherwise indicated, the following words, phrases and symbols shall have the meanings as set forth below, except to the extent that the context in which they are used indicates otherwise.
[013] A dash ("-") that is not between two letters or symbols is used to indicate a point of attachment for a substituent. For example, -CONH2 is attached through the carbon atom.
[014] "Amino protecting group," as used herein, refers to any substituent that may be used to prevent an amino group on a molecule from undergoing a chemical reaction while chemical change occurs elsewhere in the molecule. An amino protecting group can be removed under the appropriate chemical conditions.
Numerous amino protecting groups are well known to those skilled in the art, and examples of amino protecting groups, methods for their addition, and methods for their removal can be found in, for example, "Protective Groups in Organic Synthesis"
by Theodora W. Greene, John Wiley and Sons, New York, 1991, the disclosure of which is incorporated herein by reference.
[015] "Amino protecting group reagents," as used herein, refer to addition reagents that can react with an amino group such as the N-terminus of a peptide, thereby chemically modifying said amino group by addition of an amino protecting group.
[016] As used herein to refer to an amino protecting group containing at least one "acidic substituent," the term "acidic substituent," refers to a portion of the amino protecting group (e.g., a substituent) containing a donatable hydrogen.
Exemplary acidic substituents include the acid form of sulfo, sulfate, sulfonate, carboxy, carboxylate, phosphonate, and phosphate. In one embodiment, the protecting group comprises an aryl or heteroaryl substituted with the acidic substituent.
[017] As used herein, the term "water soluble" refers to the compound with sufficient water solubility for an intended purpose. Water soluble compounds with an amino protecting group with an acidic substituent can have a first water solubility adequate to permit deacylation of such compounds in an aqueous medium (e.g., 0.1-g/L, preferably about 1 g/L or higher). Similarly, water soluble antibiotic compounds can have a second water solubility adequate to permit dissolution of therapeutically effective amounts of the antibiotic compounds in an aqueous pharmaceutical composition (e.g., about 100-500 g/L, including at least about g/L).
[018] "Aryl," as used herein, refers to a mono-, bi-, or other multi-carbocyclic, aromatic ring system, including aromatic ring systems fused to one or more rings selected from aryl, cycloalkyl, and heterocyclyl, as well as aryl moieties having from 5-14 ring members. Nonlimiting examples of aryl groups include phenyl, naphthyl, biphenyl, and anthracenyl.
[019] "Carboxy," as used herein, refers to a COOH radical.
[020] "Fmoc" is a 9-fluorenylmethoxycarbonyl group.
[021] "Halo," as used herein, refers to bromo, chloro, fluoro, and iodo, and the term "halogen" includes fluorine, chlorine, bromine, and iodine.
[022] "Heteroaryl," as used herein, refers to mono-, bi-, or other multi-carbocyclic, aromatic ring systems having from one to four non-carbon atoms or hetero groups containing one or more non-carbon atoms or groups selected from 0, N, NH, S, or SO forming a portion of at least one position in the aromatic ring system. Heteroaryl ring systems can have, for example, five to fifteen ring members.
Nonlimiting examples of heteroaryl groups include indolyl, pyridinyl, thiazolyl, thiadiazoyl, isoquinolinyl, pyrazolyl, oxazolyl, oxadiazolyl, triazolyl, and pyrrolyl groups.
[023] "[Ile7]-polymyxin BI," as used herein, refers to a compound of the Formula:

o H

O O
H H

H H
O O
NH HN O
H

O
HO and commercially available salts thereof, such as a sulfate salt.
[024] "Polymyxin B1," as used herein, refers to a compound of the Formula:
'o H 4q ` ,. ,J~ H
IY `
H
H O NH O
O
O O
H H
N N HN NHz H H
O O
H HN
H
NH2 NH=
O NH, HO
and commercially available salts thereof, such as a sulfate salt.
[025] "Sulfo," as used herein, refers to the -SO3H moiety.
[026] The term "prodrug," as used herein, represents compounds that are rapidly transformed in vivo to the parent compound of the antibacterial compounds described herein, for example, by hydrolysis in blood. A discussion is provided in T.
Higuchi and V. Stella, Pro-drugs as Novel Delivery Systems, Vol. 14 of the ACS
Symposium Series, and in Edward B. Roche, ed., Bioreversible Carriers in Drug Design, American Pharmaceutical Association and Pergamon Press, 1987, both of which are incorporated herein by reference.
[027] The term "pharmaceutically acceptable prodrugs" as used herein, represents those prodrugs of the compounds of the present invention that are, within the scope of sound medical judgment, suitable for use in an intended therapeutic application (e.g., suitable for contact with the tissues of humans and lower animals) without undue toxicity, irritation, allergic response, commensurate with a reasonable benefit/risk ratio, and effective for their intended use, as well as the ionic (e.g.
zwitterionic) forms and pharmaceutically acceptable derivatives (e.g., esters) of antibacterial compounds.
[028] As used herein, the term "pharmaceutically-acceptable salt" refers to pharmaceutical salts that are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, and allergic response, and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically-acceptable salts are well known in the art, such as a sulfate salt. For example, S. M. Berge, et al. describes pharmaceutically acceptable salts in J. Pharm. Sci., 1977, 66:1-19.
[029] As used herein, the phrase "pharmaceutically-acceptable carrier" refers generally to solvents, dispersion media, excipients, coatings, matrices, stabilizers, buffers, absorption enhancers, adjuvents, controlled release media, and the like, that are compatible with an intended use, such as pharmaceutical administration.
The use of such carriers for pharmaceutically active substances is well known in the art.
Nonlimiting examples of carriers include corn starch or gelatin, lactose, sucrose, microcrystalline cellulose, kaolin, mannitol, dicalcium phosphate, sodium chloride and alginic acid. The compositions may contain croscarmellose sodium, microcrystalline cellulose, corn starch, sodium starch glycolate and alginic acid.
[030] The term "therapeutically effective amount," as used herein refers to an amount of an antibacterial compound that is effective to perform the function being sought by the researcher or clinician without unduly harming the tissues of the subject to which the agent is administered.
[031] The term "subject," as used herein, refers to a mammal, a plant, a lower animal, or a cell culture. In one embodiment, a subject is a human or other animal patient in need of antibacterial treatment.
[032] Unless otherwise indicated, the following abbreviations as used herein have the following meanings:
AUC = area under the curve ADME = absorption, distribution, metabolism, excretion BID = bis in die, meaning to give medication twice a day BUN = blood urea nitrogen cfu = colony forming units CH3CN = acetonitrile Cmax = the maximum plasma concentration CV = cardiovascular CYP = Cytochrome P450 EC50 = half maximal effective concentration ED = effective dose EDTA = ethylenediaminetetraacetic acid EtOH = ethanol HOAc = acetic acid HPLC = high pressure liquid chromatography MeOH = methanol MIC = minimum inhibitory concentration to inhibit growth of the test organism MIC90 = MIC required to inhibit growth of 90% of the strains of an organism tested MTD = maximum tolerated dose NaH2PO4 = sodium phosphate NOAEL = no observable adverse effect level PK = pharmacokinetics PMB = polymyxin B
PME = polymyxin E (colistin) QD = quaque die (every day) TID = ter in die, meaning to give medication three times a day OD600 = optical density measured at 600 nm Antibacterial Compounds [033] Antibacterial compositions can include compounds of Formula (I) where R7 is an alkyl moiety selected from isobutyl and sec-butyl, as well as pharmaceutically acceptable derivatives or pharmaceutically acceptable salts thereof:

N NH

CI O O O`-H H H

N ),N N N N ::x02 H

O Y~~~ NH2 O
HO
[034] For example, pharmaceutical compositions with antibacterial activity can include one or more compounds of Formula (I), such as a compound of Formula (Ia), a compound of Formula (lb), specific enantiomers of Formula (la) or Formula (lb), or any combination thereof, or pharmaceutically acceptable salts (e.g., a sulfate salt) or derivatives (e.g., esters) thereof:

O

N NH

H

H H
O
O NH HN O
H

O yl---~ NH2 O
(I} H O
O

N NH
H

O
H H H
N`N N HN N H 2 +I H H
O
O
NH HN
H

O yl---~ NH2 O
(lb) HO
[035] One particular example of an antibacterial compound of Formula (la) is described by Formula (Ic) and Formula (Ic') below.

L-Leu7 ' L-Dabs D-Phe6 L-Dabs t 1 L-Dab5 L-Thr10 L-Dab4 t L-Dab3 t L-Thr2 t L-Dab1 t NH
O)-N
H CI
(Ic) O
N

O

N N N v `O \NH HN0 ~ = H O = N
O

(Ic") [036] The antibacterial compounds of Formula (I) have antibacterial activity against a range of gram negative bacteria, including antibacterial activity and spectrum similar to certain polymyxin compounds such as colistin (polymyxin E) and/or polymyxin B. For example, as discussed in the Examples, compounds of Formula (I) are active against colistin susceptible P. aeruginosa and Acinetobacter spp., including multi-drug resistant strains, while also having antibacterial activity against E. coli, K. pneumonia, Citrobacterspp. and some activity against Enterobacterspp. isolates. As detailed in the Examples, pharmacological studies confirmed in vivo antibacterial activity of a compound of Formula (I) comparable to polymyxin B in both a murine lung infection model using P. aeruginosa #44 and K.
pneumoniae bacteria#21, as well as a murine thigh infection model using A.
baumannii #1570. In addition, the compounds of Formula (I) are as active or more acitve than polymyxin B against mult-drug resistant bacteria. Furthermore, the compound of Formula (I) designated as compound 5 in the examples below, is as active as polymyxin B against a bacterial infection that may be caused or exacerbated by P. aeruginosa. One preferred antibacterial compound of Formula (I) is (S)-4-amino-N-((2S,3R)-1 -((S)-4-amino-1 -oxo-1 -((3S,6S,9S, 12S,15R,18S,21 S)-6,9,18-tris(2-aminoethyl)-15-benzyl-3-((R)-1-hydroxyethyl)-12-isobutyl-2,5,8,11,14,17,20-heptaoxo-1,4,7,10,13,16,19-heptaazacyclotricosan-21-ylamino)butan-2-ylamino)-3-hydroxy-1-oxobutan-2-yl)-2-(3-(2-chlorophenyl)ureido)butanamide.
[037] Surprisingly, the antibacterial compounds of Formula (I) have reduced toxicity compared to other compounds of Formula (II) and polymyxins such as polymyxin B. For example, certain compounds of Formula (I) (i.e., compounds 5 and 6 in Table 1) were substantially less toxic in the rat kidney proximal tubule cell culture cytotoxicity assay of Example 4 than either polymyxin B or the structurally similar 3-chlorophenyl analog designated compound 4. Compounds 5 and 6 of Formula (I) have an EC50 value of > 1,000 g/mL compared to EC50 values of 318 g/mL measured for polymyxin B and 691 .1g/mL measured for compound 4. The Formula (I) compound designated compound 5 in Table 1 also showed reduced renal toxicity compared to polymyxin B in the seven-day repeat dose safety study in cynomologus monkeys detailed in Example 6.

Synthesis of Compounds [038] The compounds of Formula (II), including the compounds described in Formula (I), can be prepared from a polymyxin starting material according to the synthetic scheme shown in Figure 1:

O
H

.

O ~ D
H H H
R I N X-C 'N I R3 Nd N ::x0 O

H
NH NH HN

H
O
(II} HO '"
wherein:
R1 is chosen from H, and -C(O)NHRA, wherein RA is chosen from benzyl and phenyl, and wherein both said benzyl and phenyl may be optionally substituted with one or more halo and/or nitro;
R2 is chosen from -CH2CH(CH3)2 and -CH(CH3)CH2CH3; and R3 is H.
[039] As shown in the Examples, certain compounds of Formula (II) are as active or more active against bacteria than polymyxin B. Compounds of this embodiment include those designated as compounds 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 13, 14, and 15 in the Examples below.
[040] The compounds of Formula (II) can possess one or more chiral asymmetric carbon atoms and are thus capable of exhibiting optical activity.
Unless otherwise indicated, Formula (II) describes racemic and individual enantiomeric and/or diastereomeric forms of compounds at each chiral position, including racemic or enantiomerically resolved non-racemic compositions thereof. Unless otherwise indicated, the compounds disclosed herein can be utilized as a single isomer or as a mixture of stereochemical isomeric forms. For example, compounds of Formula (la) can include compounds of Formula (Ic) and Formula (Ic') and/or other diastereomers described by Formula (Ia).
[041] Compounds of Formula (II) can be obtained from any suitable lipopeptide starting material of Formula (III) comprising a cyclic portion with a plurality of 2,4-diaminobutanoic acid residues and an acyl exocyclic tail portion (T):

T(Y1),(Y2)y(Y3)Z-NH H 0 R6 N
N O
H
0 (CH2)2NH2 HN NH2-(CH2)2 0 HN

N N )~~R7 H N
__1Y
Rip 0 O
(CH2)2NH2 (III) wherein Y1 and Y3 are 2,4-diaminobutanoic acid residues; Y2 is a threonine residue;
x, y and z are integers independently equal to 1; R6 is benzyl; R7 is chosen from isobutyl and sec-butyl; and R10 is 1-hydroxy-1-ethyl; T is R'-(C=O)-; and R' is an alkyl or (e.g., hydroxyl-) substituted alkyl, such as 6-methyloctanoyl, 6-methylheptanoyl, octanoyl, heptanoyl, nonanoyl, or 3-hydroxy-6-methyloctanoyl.
Examples of suitable starting materials include a polymyxin B, or a [Ile7]-polymyxin B.
The starting material can be a naturally occurring polymyxin compound isolated from the fermentation of Bacillus polymyxa according to procedures described in, e.g., Hausmann, et at. (1954) J. Am. Chem. Soc. 76, 4892-4896.
[042] Referring to Figure 1, the polymyxin 131 starting material (Ila) is reacted with an amino protecting group reagent (step (a)) to form a protected starting material (Ilb) comprising an amino protecting group (P) attached to the primary amino moieties in the starting material (Ila). Preferably, the amino protecting group (P) comprises an acidic moiety to provide a protected starting material (Ilb) that is sufficiently water soluble to react with a deacylating agent in an aqueous medium (step (b)) to form a deacylated material (Ile). The deacylated material (Ile) can be reacted with an addition reagent (step (c)) to form a protected antibacterial compound (lid). The amino protecting group (P) can be removed from the protected antibacterial compound (lid) to form the compound of Formula (II), including compounds of Formula (I).
[043] The water solubility of the compounds of Formulae (Ila), (Ilb), (Ilc), (lid) and Formula (I) may differ, while each independently being suitable for different intended purposes. For example, a water soluble protected starting material (e.g., compound of Formula (Ilb) in Figure 1) can include amino protecting groups with an acidic substituent (e.g., "P" in Formula (Ilb) in Figure 1) providing sufficient water solubility to perform enzymatic deacylation (e.g., step (b) in Figure 1) of the protected starting material at an acceptable yield in an aqueous medium. A product material can be an antibiotic compound of Formula (I) with sufficient water solubility to form an aqueous pharmaceutical composition comprising a therapeutically effective concentration of the compound of Formula (I).
[044] The protecting group (P) can be selected to provide a desired level of water solubility in the protected starting material (Ilb). For example, the protecting group (P) can include an aryl or heteroaryl moiety and at least one acidic substituent selected from carboxy, sulfo, sulfate, and salts thereof. Exemplary protecting groups include 9-fluorenylmethoxycarbonyl (Fmoc) substituted with acidic substituents or salts thereof, such as 2-sulfo-9-fluorenylmethoxycarbonyl (HSO3-Fmoc), and its sodium salt (NaSO3-Fmoc), 2-carboxymethyl-9-fluorenylmethoxycarbonyl (2-carboxymethyl-Fmoc), 2-carboxy-9-fluorenylmethoxycarbonyl (2-carboxy-Fmoc), and 4-carboxy-9-fluorenylmethoxycarbonyl (4-carboxy-Fmoc). Preferably, the protecting group is a sulfonic acid of 9-fluorenylmethoxycarbonyl, such as 2-sulfo-9-fluorenylmethoxycarbonyl. Additional amino protecting groups include, but are not limited to, the protecting groups disclosed in "Protective Groups in Organic Synthesis" by Theodora W. Greene, John Wiley and Sons, New York, 1991 at pp. 315-348, the disclosure of which is incorporated herein by reference.
[045] The protected starting material (llb) can be reacted with an enzymatic deacylating agent in an aqueous medium to form the deacylated material (11c).
One example of a deacylating enzyme useful for deacylation of the protected starting material (Ilb) is produced by certain microorganisms of the genus family Actinoplanaceae. Some of the known species and varieties of this family include Actinoplanes philippinensis, Actinoplanes armeniacus, Actinoplanes utahensis, Actinoplanes missouriensis, Spirillospora albida, Streptosporangium roseum, Streptosporangium vulgare, Streptosporangium roseum var hollandensi, Streptosporangium album, Streptosporangium viridialbum, Amorphosporangium auranticolor, Ampullariella regularis, Ampullariella campanulata, Ampullariella lobata, Ampullariella digitata, Pilimelia terevasa, Pimelia anulata, Planomonospora parontospora, Planomonospora venezuelensis, Planobispora Iongispora, Planobispora rosea, Dactylosporangium aurantiacum, and Dactylosporangium thailandende. All natural and artificial variants and mutants which are obtained from the Actinoplanacea family and which produce the enzyme may be used in this invention. The deacylating agent is preferably a polymyxin deacylase enzyme, which can be obtained from Actinoplanes utahensis.
[046] The deacylase enzyme can be obtained as a water-soluble freeze-dried solid. In one embodiment, the deacylase is obtained by fermenting Actinoplanes utahensis, separating the cells from the fermentation medium, washing the cells with water, extracting the cells with basic buffer at pH 8-11 for about 20 minutes, adjusting the extract to pH 7-8 and freeze-drying. The powdered form of the enzyme resulting from this process can be relatively stable and can be readily re-dissolved in water for use. Further purification can be obtained by gel filtration, membrane filtration or other types of chromatography. This enzyme can deacylate, for example, a sodium salt N-[2-sulfo-9-fluorenylmethoxycarbonyl]5 polymyxin B
to obtain the deacylated protected peptide having the Formula (IIc) in Figure 1.
In other embodiments, the enzyme from Actinoplanes utahensis can be used as the whole broth from the fermentation or as the washed cells.
[047] The enzyme from Actinoplanes utahensis can also be used as a water-solubilized preparation. The water-solubilized enzyme preparation can be obtained by a relatively strong basic extraction of the washed cells, followed by adjustment of the pH of the clear extract to pH 7-8. This water-solubilized enzyme preparation can be freeze-dried to a solid form.
[048] Referring to step (c) of Figure 1, the deacylated material (IIc) is reacted with an addition reagent selected to react with a primary amino group at the N-terminus of the exocyclic portion of the deacylated material (IIc), thereby chemically modifying the amino group by addition of all, or a component, of the addition reagent to the amino group. For example, an addition reagent may be an acylamino reagent such as R'-(C=O)-LG or a sulfonating reagent such as R'-S02-LG, where LG is a leaving group, that can react with an amino group. An addition reagent may also be, for example, an isocyanate, isothiocyanate, activated ester, acid chloride, sulfonyl chloride, activated sulfonamide, activated heterocycle, activated heteroaryl, chloroformate, cyanoformate, thioacylester, phosphoryl chloride, phosphoramidate, imidate, or lactone. An addition reagent may also be an aldehyde or ketone that reacts with an amine under reductive conditions to form an alkylated amine. An addition reagent may also be an activated amino acid, or an amino acid and a peptide coupling reagent, such as, e.g., PyBOP (behzotriazole-1-yl-oxy-tris-pyrrolidino-phosphonium hexafluorophosphate), HBtU (2-(1 H-benzotriazole-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate), HBtU/HOBt (2-(1 H-benzotriazole-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate / N-hydroxybenzotriazole), or DCC (dicyclohexylcarbodiimide). In the addition reaction shown in step (c) of Figure 1, various addition reagents can be selected to reactively couple the functional substituent "R1-" to the primary amine in the deacylated material (Ilc) to form compounds of Formula (II), where R, is defined in Table 1 of the Examples.
[049] The protected antibacterial compound (lid) in Figure 1 can be converted to a compound of Formula (II) by removing the protecting groups (P) of the protected antibacterial compound (lid) (e.g., by deprotection using standard methods such as those described in "Protective Groups in Organic Synthesis" by Theodora W. Greene, John Wiley and Sons, New York, 1991, the disclosure of which is incorporated herein by reference). As those skilled in the art will recognize, the choice of amino protecting group employed in the first step of the process (step a) will dictate the reagents and procedures used in removing said amino protecting group. When the reagent for the chemical modification contains one or more protecting group(s), those protecting group(s) can also be removed. The choice of the protecting group(s) utilized on the chemical modifying agent substituent(s) will dictate the reagents and procedures used in removing said protecting group(s).
When the protecting group(s) utilized on the modifying agent substituent(s) and the protecting group utilized for the protected compounds are compatible, the protecting groups may be removed in a single step. However, when the protecting group(s) are incompatible multiple steps may be required to remove all of the protecting groups.
[050] After removal of the amino protecting groups (P), the compounds of Formula (I) can be purified by gel filtration, chromatography, or reverse-phase HPLC.
Diastereomers can be separated by conventional means such as chromatography or crystallization. Enantiomers can be obtained by resolution of the racemic mixtures according to conventional processes, for example by formation of diastereomeric salts by treatment with an optically active acid or base. Nonlimiting examples of appropriate acids include tartaric, diacetyltartaric, dibenzoyltartaric, ditoluoyltartaric and camphorsulfonic acid. The mixture of diastereomers can be separated by crystallization followed by liberation of the optically active bases from the optically active salts. Alternative process for separation of enantiomers include the use of a chiral chromatography column optimally chosen to maximize the separation of the enantiomers, or the use of supercritical fluid chromatography (SFC). For example, the compound of Formula (Ic) can be obtained from a starting material with corresponding stereochemical structures (e.g., a polymyxin B1 starting material).
Another method involves synthesis of covalent diastereomeric molecules by treating compounds of the invention with an activated form of an enantiomerically enriched acid or with an enantiomerically enriched isocyanate. The synthesized diastereomers can be separated by conventional means such as chromatography, distillation, crystallization or sublimation, and then hydrolyzed to obtain the enantiomerically enriched compound. Optically active compounds can likewise be obtained by utilizing optically active starting materials. These isomers may be in the form of a free acid, a free base, an ester or a salt.
[051] One particular process for preparing an antibacterial compound can include the following steps: (a) treating a polymyxin such as polymyxin B with an amino protecting group comprising at least one acidic substituent to form a protected compound; (b) treating the protected compound with a deacylating agent, to form at least one deacylated protected compound having the Formula:

P O

HN N NH

O O O

H H
O
NH HN O
H
HNC ~NH N
P P O
NH
~1 Yk-----O P
HO , wherein R2 is isobutyl and P is an amino protecting group;

(c) treating the at least one deacylated protected compound with an isocyanate to form at least one acylated protected compound having the Formula:

H
P "IN N NH
5:~' 1 H
OH O NH O I- H
O O O
N N N
H H HN ~P
ac"
O O
I NH HN O
O
H
,NH
P HNC P O N N,P
H
O
HO ; and (d) removing the amino protecting groups (e.g., by treating the at least one acylated protected compound with an organic base) to form the antibacterial compound having the Formula:

O

H
O

O O O
H H H
aCN

O O
I NH HN O
H

HO
such as a compound of Formula (Ic) and Formula (Ic') above.
[052] The Examples describe one suitable method for obtaining the antibacterial compound of Formula (Ic) and (Ic'). Purification of the antibacterial compounds by the methods of Examples using HPLC can provide a compound of Formula (Ic) and (Ic') with a purity of about 98% (e.g., with up to about 1 %
of Formula (lb)). Alternatively, similar methods using low pressure liquid chromatography can be used to produce an antibacterial compound of Formula (Ic) and Formula (Ic') with a purity of about 92% (e.g., with up to about 3% of a compound of Formula (lb)).

Pharmaceutical Antibacterial Compositions [053] Pharmaceutical compositions can be formed by combining a compound of Formula (I) or a pharmaceutically acceptable derivative or salt thereof with a pharmaceutically acceptable carrier suitable for delivery to a recipient subject (e.g., a human) in accordance with known methods of drug delivery.
Antibacterial pharmaceutical compositions suitable for administration of one or more compounds of Formula (I) can be Formulated. The compound of Formula (I), and/or pharmaceutically acceptable salts or derivatives of Formula (I) can be included in a pharmaceutical antibacterial composition along with one or more carriers.
[054] A compound of Formula (I) can be Formulated as a variety of salts or derivatives to improve stability or toxicological properties of the compound, increase or decrease solubility, improve pharmacokinetic performance of the compound (e.g., Cmax or AUC measurements) or improve storage properties (e.g., to reduce hygroscopicity) of a pharmaceutical composition. Pharmaceutically-acceptable salts of compounds of Formula (I) may be prepared by conventional means from the corresponding compound of the invention by treating, for example, the compound with the appropriate acid or base. Examples of publications describing the selection and formation of pharmaceutically acceptable salts of medicinal compounds include Haynes, Delia A., et al., "Occurrence of Pharmaceutically Acceptable Anions and Cations in the Cambridge Structural Database," Journal of Pharmaceutical Sciences, v. 94, no. 10, 2111-2120 (October 2005), and Stahl, PH, et al., Eds., "Handbook of Pharmaceutical Salts: Properties, Selection and Use," Weinheim/Zurich, Wiley-VCHNHCA, incorporated herein by reference.
[055] A compound of Formula (I) may also be Formulated as a prodrug in a pharmaceutical composition. Pro-drugs can include an antibacterial compound disclosed herein having one or more amino groups protected with HSO3-Fmoc and/or other amino protecting groups (comprising at least one acidic group are antibacterial pro-drugs (e.g., as shown as "P" in compounds of Formula (lid) in Figure 1). The HSO3-Fmoc group can be cleaved after introduction into an animal, such as, e.g., a mammal, including a human, liberating the biologically active compound. Administration of a biologically active compound as the protected pro-drug may result in a slow release mechanism for the antibacterial compound.
Other suitable prodrugs can include esters of a compound of Formula (I). A
discussion of pro-drugs is provided in, e.g., Gershonov, et al. (2000), J. Med. Chem. 43:
(13), 2530-2537, and Schechter, et al. (2002), J. Med. Chem. 45: (19), 4264-4270, which are all incorporated by reference.
[056] The pharmaceutical compositions can be formulated for parenteral delivery, including intravenous, intramuscular, intraperetoneal, subcutaneous, intraocular, intrathecal, intraarticular, intrasynovial, cisternal, intrahepatic, intralesional and intracranial injection, infusion, and/or inhaled routes of administration for the therapeutic treatment of medical conditions, such as bacterial infections.
[057] Pharmaceutical preparations can be prepared in accordance with standard procedures and are administered at dosages that are selected to treat infection (see, e.g., Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, PA and Goodman and Gilman's "The Pharmaceutical Basis of Therapeutics," Pergamon Press, New York, NY, the contents of which are incorporated herein by reference, for a general description of the methods for administering various antimicrobial agents for human therapy).
[058] The pharmaceutical composition can include one or more carriers, as defined above, for an intended medical use. Pharmaceutical compositions for parenteral injection comprise pharmaceutically-acceptable aqueous or nonaqueous solutions of antibacterial compounds of Formula (I) in addition to one or more of the following: pH buffered solutions, adjuvants (e.g. preservatives, wetting agents, emulsifying agents, and dispersing agents), liposomal Formulations, nanoparticles, dispersions, suspensions or emulsions as well as sterile powders for reconstitution into sterile injectable solutions or dispersions just prior to use. In one particular example, a pharmaceutical composition includes an antibacterial active component with about 92-98% w/w of the active component being a compound of Formula (Ic) and Formula (Ic'), and with about 0-3% (including about 0-1%, 0-3%, 1-3%, up to 1 %, and/or up to 3%) w/w of the active component being a compound of Formula (lb), where this active component is added to the other components of the pharmaceutical composition. One particular intravenous Formulation is formed by combining an antibacterial active component with the remaining components of an intravenous pharmaceutical composition, where the active component has about 98% w/w of a compound of Formula (Ic) and Formula (Ic') and about 0-1 % w/w of a compound of Formula (lb). For intravenous (IV) use, the pharmaceutical composition can include any of the commonly used intravenous fluids that can be administered by infusion, such as physiological saline or Ringer's solution.
Injectable depot forms to release the antibacterial agent in situ can be made by forming microencapsulating matrices of the drug in biodegradable polymers such as polylactide-polyglycolide. Depending upon the ratio of drug to polymer and the nature of the particular polymer employed, the rate of drug release can be controlled.
For intramuscular preparations, a sterile Formulation of a compound of the present invention, or a suitable soluble salt form of the compound, for example the hydrochloride salt, can be dissolved and administered in a pharmaceutical diluent such as Water-for-Injection (WFI), physiological saline or 5% glucose.
[059] In some cases, in order to prolong the effect of the drug, it may be desirable to slow the absorption of the drug following subcutaneous or intramuscular injection. This may be accomplished by the use of a liquid suspension of crystalline or amorphous material with poor water solubility. Amorphous material may be used alone or together with stabilizers as necessary. The rate of absorption of the drug then depends upon its rate of dissolution, which in turn, may depend upon crystal size and crystalline form.
[060] The injectable Formulations can be sterilized, for example, by filtration through a bacterial-retaining filter, or by incorporating sterilizing agents in the form of sterile solid compositions, which can be dissolved or dispersed in sterile water or other sterile injectable medium just prior to use.
[061] The pharmaceutical compositions can be in powder form for reconstitution in the appropriate pharmaceutically acceptable carrier at the time of delivery. In another embodiment, the unit dosage form of the compound can be a solution of the antibacterial compound of Formula (I), or a salt thereof, in a suitable diluent in sterile, hermetically sealed ampoules or sterile syringes.

Methods of Using Antibacterial Compositions [062] The antibacterial compounds described herein are useful in the manufacture of antibacterial pharmaceutical compositions, and treatment of bacteria with these compositions. In particular, the antibacterial compounds are useful in treating and eliminating gram negative bacterial infections. Methods of treating bacterial infections in subjects (e.g., humans and animals) can include the administration of a therapeutically effective amount of an antibiotic compound of the Formula (I) or a pharmaceutically acceptable salt or prodrug thereof.
[063] The antibacterial compounds of Formula (I) can be used in vivo, for example, to treat bacterial infections in a subject, as well as in vitro, for example to treat cells (e.g., bacteria) in culture to eliminate or reduce the level of bacterial contamination of a cell culture. In one embodiment, at least one compound of Formula (I), or pharmaceutical compositions thereof, are administered to a cell culture, such as by administering in a nutrient medium.
[064] Methods of treatment of such infections include administering to a subject in need thereof a therapeutically effective amount of an antibacterial compound of Formula (I). The compound can be parenterally administered to a subject having or suspected to have a bacterial infection, such as a gram negative infection.
[065] The antibacterial compounds of Formula (I) are preferably used in vivo to treat an infection in a subject by administering a therapeutically effective amount of a compound of Formula (I) in a pharmaceutical omposition. The method can comprise parenterally administering to a subject in need thereof a pharmaceutical composition comprising at least one compound of Formula (I). Pharmaceutical compositions include compositions comprising compound(s) of Formula (1) in an amount sufficient to achieve the intended purpose, i.e., the treatment or prevent of infectious diseases. The amount and concentration of antibacterial compound of Formula (1) in the pharmaceutical composition, as well as the quantity of the pharmaceutical composition administered to a subject, can be selected based on clinically relevant factors, such as medically relevant characteristics of the subject (e.g., age, weight, gender, other medical conditions, and the like), the solubility of the antibacterial compound in the pharamceutical composition, the potency and activity of the antibacterial compound, and the manner of administration of the pharmaceutical composition. A pharmaceutical composition comprising a therapeutically effective amount of an antibacterial compound of Formula (1) can be administered intravenously to a patient for treatment of gram negative infections in a clinically safe and effective manner, including one or more separate administrations of the composition. For example, about 0.05 mg/kg to about 5.0 mg/kg can be administered to a subject per day in one or more doses (e.g., doses of about 0.05 mg/kg QD, 0.10 mg/kg QD, 0.50 mg/kg QD, 1.0 mg/kg QD, 1.5 mg/kg QD, 2.0 mg/kg QD, 2.5 mg/kg QD, 3.0 mg/kg QD, 0.75 mg/kg BID, 1.5 mg/kg BID or 2.0 mg/kg BID). For certain antibiotic indications, the total daily dose of a compound of Formula (I) can be about 0.05 mg/kg to about 3.0 mg/kg of one or more compounds of Formula (I) administered intravenously to a subject one to three times a day, including administration of total daily doses of about 0.05-3.0, 0.1-3.0, 0.5-3.0, 1.0-3.0, 1.5-3.0, 2.0-3.0, 2.5-3.0, and 0.5-3.0 mg/kg/day of a compound of Formula (la), Formula (Ic) or Formula (Ic') using 60-minute QD, BID or TID intravenous infusion dosing. In one particular example, antibiotic pharmaceutical compositions can be administered to a subject with, e.g., total daily doses of 1.5 mg/kg, 3.0 mg/kg, 4.0 mg/kg of a composition with up to about 92-98% of Formula (la), Formula (Ic) and/or Formula (Ic') are intravenously administered QD or BID. The amount per administered dose or the total amount administered will depend on such factors as the nature and severity of the infection, the age and general health of the patient, the tolerance of the patient to the compound and the microorganism or microorganisms involved in the infection.
[066] In particular, the pharmaceutical compositions comprising antibacterial compounds of Formula (I) can be used to treat a subject having a bacterial infection in which the infection is caused or exacerbated by a gram-negative bacteria.
These gram-negative bacteria include, but are not limited to, Acinetobacterspp.
(including Acinetobacter baumannii), Citrobacter spp., Enterobacter spp., Escherichia spp.
(including Escherichia coli), Haemophilus influenzae, Morganella morganii, Pseudomonas aeruginosa, Klebsiella spp. (including Klebsiella pneumoniae), Salmonella spp., Shigella spp., Yersinia pseudotuberculosis, and all species of Enterobacter, Pasteurella, Brucella, Bordetella, Proteus, Serratia, Providencia, and Edwardsiella. The bacteria infection can also be caused or exacerbated by a gram-negative bacteria chosen from Pseudomonas aeruginosa, Acinetobacter spp, Stenotrophomonas maltophilia, Escherichia coli, Klebsiella pneumoniae, Citrobacter spp, and Enterobacter. In one embodiment, at least one compound of Formula (I) may be used to treat multiple drug resistant bacteria, such as Multiple Drug Resistant (MDR) P. aeruginosa, Extended Spectrum Beta Lactamase (ESBL) K.
pneumonia, ESBL E. coli, and A. baumannii [067] The pharmaceutical compositions can be used to treat a bacterial infection of any organ or tissue in the body caused by gram-negative bacteria.
These organs or tissue include, without limitation, skeletal muscle, skin, bloodstream, kidneys, heart, lung and bone. For example, a pharmaceutical composition comprising at least one compound of Formula (I) can be administered to a subject to treat, without limitation, skin and soft tissue infections (e.g., complex skin infections), bacteremia, interabdominal infections and urinary tract infections (e.g., cUTI). In addition, a compound of Formula (I) may be used to treat community acquired respiratory infections, including, without limitation, otitis media, sinusitis, chronic bronchitis and pneumonia (including community-acquired pneumonia, hospital-acquired pneumonia and ventilator associated pneumonia), including pneumonia caused by drug-resistant H. influenzae. At least one compound of Formula (I) can be administered to a subject to treat mixed infections that comprise different types of gram-negative bacteria, or which comprise both gram-positive and gram-negative bacteria. These types of infections include intra-abdominal infections and obstetrical/gynecological infections. At least one compound of Formula (I) may also be administered to a subject to treat an infection including, without limitation, endocarditis, nephritis, septic arthritis, intra-abdominal sepsis, bone and joint infections and osteomyelitis. At least one compound of Formula (I), or pharmaceutical compositions thereof, may also be directly injected or administered into an abscess, ventricle or joint. Pharmaceutical compositions administered as an aerosol for the treatment of pneumonia or other lung-based infections. In one embodiment, the aerosol delivery vehicle is an anhydrous, liquid or dry powder inhaler.

Examples [068] A series of compounds according to Formula (II) were prepared, as described herein:

O
H
R H NH

OH O NH O
O O
H H H
N N HN N
Rim H H R3 O
NH HN :co-, H
R /NH HN N ~R3 H
3 R3 0-~' N
O
HO
(II) wherein the substituents are selected as indicated in Table 1. These compounds include compounds of Formula (I) (designated Compound 5 and Compound 6) and Formula (II).
Table 1: Compounds of Formula (II), including compounds of Formula (I) Compound No. R, R2 R3 1 -C(O)NH-phenyl -CH2CH(CH3)2 H
2 -C(O)NH-phenyl -CH(CH3)CH2CH3 H
3 -C(O)NH-4- -CH2CH(CH3)2 H
chlorophenyl 4 -C(O)NH-3- -CH2CH(CH3)2 H
chlorophenyl -C(O)NH-2- -CH2CH(CH3)2 H
chlorophenyl 6 -C(O)NH-2- -CH(CH3)CH2CH3 H
chlorophenyl 7 -C(O)NH-2,6- -CH2CH(CH3)2 H
dichlorophenyl 8 -C(O)NH-2,6- -CH(CH3)CH2CH3 H
dichlorophenyl 9 -C(O)NH-4- -CH2CH(CH3)2 H
bromophenyl -C(O)NH-4- -CH2CH(CH3)2 H
fluorophenyl Compound No. R, R2 R3 11 -C(O)NH- -CH2CH(CH3)2 H
pentafluorophenyl 12 -C(O)NH-2- -CH2CH(CH3)2 H
chlorobenzyl 13 -C(O)NH-2- -CH(CH3)CH2CH3 H
chlorobenzyl 14 -C(O)NH-2,4- -CH2CH(CH3)2 H
dichlorobenzyl 15 -C(O)NH-2,4- -CH(CH3)CH2CH3 H
dichlorobenzyl 16 -C(O)NH-2-chloro-4- -CH2CH(CH3)2 H
nitrophenyl 17 -C(O)NH-2-chloro-4- -CH(CH3)CH2CH3 H
nitrophenyl 18 -C(O)NH-phenyl -CH2CH(CH3)2 HSO3-Fmoc 19 -C(O)NH-phenyl -CH(CH3)CH2CH3 HSO3-Fmoc 20 -C(O)NH-4- -CH2CH(CH3)2 HSO3-Fmoc chlorophenyl 21 -C(O)NH-3- -CH2CH(CH3)2 HSO3-Fmoc chlorophenyl 22 -C(O)NH-2- -CH2CH(CH3)2 HSO3-Fmoc chlorophenyl 23 -C(O)NH-2- -CH(CH3)CH2CH3 HSO3-Fmoc chlorophenyl 24 -C(O)NH-2,6- -CH2CH(CH3)2 HSO3-Fmoc dichlorophenyl 25 -C(O)NH-2,6- -CH(CH3)CH2CH3 HSO3-Fmoc dichlorophenyl 26 -C(O)NH-4- -CH2CH(CH3)2 HSO3-Fmoc bromophenyl 27 -C(O)NH-4- -CH2CH(CH3)2 HSO3-Fmoc fluorophenyl 28 -C(O)NH- -CH2CH(CH3)2 HSO3-Fmoc pentafluorophenyl 29 -C(O)NH-2- -CH2CH(CH3)2 HSO3-Fmoc chlorobenzyl 30 -C(O)NH-2- -CH(CH3)CH2CH3 HSO3-Fmoc chlorobenz I
31 -C(O)NH-2,4- -CH2CH(CH3)2 HSO3-Fmoc dichlorobenzyl 32 -C(O)NH-2,4- -CH(CH3)CH2CH3 HSO3-Fmoc dichlorobenzyl 33 -C(O)NH-2-chloro-4- -CH2CH(CH3)2 HSO3-Fmoc nitrophenyl 34 -C(O)NH-2-chloro-4- -CH(CH3)CH2CH3 HSO3-Fmoc nitrophenyl 35 H -CH2CH(CH3)2 HSO3-Fmoc 36 H -CH(CH3)CH2CH3 HSO3-Fmoc [069] Unless otherwise indicated, the antibacterial activities of compounds are indicated in the Examples as their minimum inhibitory concentrations (MICs) against Pseudomonas aeruginosa, Acinetobacter spp, Stenotrophomonas maltophilia, Escherichia coli, Klebsiella pneumoniae, Citrobacter spp, and Enterobacter. MICs can be determined by the conditions disclosed in the Examples, as well as in Jarolmen, H. et al., "Activity of Minocycline Against R-Factor Carrying Enterobacteriaceae," Infectious Immunity, Vol. 1, No. 4, pp. 321-326, 1970, the disclosure of which is incorporated herein by reference.
[070] Accordingly, MIC's refer to the concentration of each antimicrobial agent required to inhibit the growth of a bacterial isolate, as well as the corresponding concentrations required to inhibit 50% and 90% of each group of microorganisms tested (MIC50 and MIC90, respectively, consistent with definitions in the "Instructions to Authors" for Antimicrobial Agents and Chemotherapy (Antimicrobial Agents and Chemotherapy, January 2010, p. 1-23, Vol. 54, No. 1, incorporated herein by reference). When only up to nine isolates of a species are tested, MIC's are reported as the MIC range of each antimicrobial agent tested. MIC
values provided as numerical ranges for testing of a single bacteria strain indicate the range of MIC values obtained in repeated tests of the indicated bacteria strain.
Example 1: Preparation of the Deacylase [071] The deacylase is produced by culturing Actinoplanes utahensis NRRL
12052 under submerged aerobic fermentation conditions. The fermentation protocol employed is known (Boeck, L.D. et al., Journal of Antibiotics 41:(8), 1085-(1998), incorporated herein by reference). A stock culture of the NRRL 12052 variant, preserved in 20% glycerol at -70 C, was introduced into a 25 x 150 mm test tube with a glass rod and Morton closure containing 10 mL of a medium composed of sucrose 2.0%, pre-cooked oatmeal 2.0%, distiller's grains and solubles 0.5%, yeast extract 0.25%, K2HPO4 0.1 %, KCI 0.05%, MgSO4.7H2O 0.05% and FeSO4.7H2O 0.0002% in deionized water. After incubation at 30 C for 72 hrs on a rotary shaker orbiting at 250 rpm the resulting mycelial suspension was transferred into 50 mL of PM3 medium in a 250 mL Erlenmeyer flask. This medium contained sucrose 2.0%, peanut meal 1.0%, K2HP04 0.12%, KH2PO4 0.05% and MgSO4.7H2O

0.025% in tap water. The flask was incubated at a temperature of 30. C for a period of 60 to 90 hrs. The harvest time was determined by an assay which involved HPLC
analyses of the deacylation of N-[2-sulfo-9-fluorenlymethoxycarbonyl]5 polymyxin B
by the whole broth at different times during the fermentation.
[072] Because single-colony isolates from a lyophile of the culture were heterogeneous for both morphology and enzyme production capability, selections were made to recover a stable, high-producing variant. Initially, multiple fermentations were carried out using inocula prepared from strain 12052.
Vegetative growth from the flask yielding the best deacylating activity was plated on a differential agar (CM). CM agar contained corn steep liquor 0.5%, Bacto peptone 0.5%, soluble starch 1.0%, NaCl 0.05%, CaC12-2H20 0.05% and Bacto agar 2.0%.
Colonies were then selected for further evaluation. Isolate No. 18 was selected as a small colony type and shown to be the best deacylase producer of all colonies selected. Comparison was based on conversion of protected polymyxin B to deacylated protected polymyxin B as determined by HPLC. This isolate was routinely used for the production of the deacylase enzyme.

Example 2: Preparation of Compounds of Formula I

Step (a): Preparation of Penta-9-(2-sulfo) fluorenylmethoxycarbonyl-Polymyxin B.
[073] Fluorenylmethoxycarbonyl N-hydroxysuccinimide (1.1982g, 3.55 mmol) dissolved in 15 mL of methylene chloride was stirred in an ice bath, using a drierite tube to maintain a dry atmosphere. A solution of chlorosulfonic acid (0.21 mL, 3.15 mmol) in 6 mL of methylene chloride was added dropwise to give a yellow solution.
The mixture was allowed to come to room temperature and stirred for several hours.
The resulting white precipitate was filtered and washed with cyclohexane-methylene chloride (1:1) then dried in vacuo over phosphorus pentoxide to afford 9-(2-sulfo)fluorenylmethoxycarbonyl N-hydroxysuccinimide (HSO3Fmoc-O-Su). Yield 1.148 g, (hygroscopic white solid). See, e.g., Y. Shechter et al. J. Med.
Chem., 43, 2530 (2000).
[074] Polymyxin B, containing a mixture of polymyxin B, and [Ile7] polymyxin B1 (1.0 g, 0.841 mmol), as purchased from Sigma-Aldrich, was dissolved in a solution of 25 mL saturated sodium bicarbonate, 25 mL of water and 25 mL of tetrahydrofuran. A solution of 2-sulfo-9-fluorenylmethoxy-N-hydroxysuccinimide (2.0 g, 4.8 mmol) in 25 mL of tetrahydrofuran was added in several portions over 45 min.
The reaction mixture was stirred at room temperature overnight and diluted with 50 mL of water. The reaction mixture was then acidified to pH 0.5 - 1 with approximately 30 mL of 6N hydrochloric acid to give an oily precipitate. The mixture was chilled overnight and the aqueous layer was decanted. The oily residue was dissolved in 100 ml of ethanol and the ethanol was evaporated in vacuo (35 C) with the aid of ethyl acetate. The resulting solid was triturated with ethyl acetate, filtered and dried to afford 1.74 g of a mixture containing the protected polymyxin B1 and [Ile7] polymyxin B1 products.
[075] The above procedure can be used with the corresponding polymyxin B
sulfate salt (containing a mixture of polymyxin B1 sulfate and [Ile7]
polymyxin B, sulfate) and 2-sulfo-9-fluorenylmethoxycarbonyl chloride with similar results.

Step (b): Deacylation of Penta-2-sulfo-9-fluorenylmethoxycarbonyl-Polymyxin B.
[076] Penta-2-sulfo-9-fluorenylmethoxycarbonyl-Polymyxin B (1 g) was dissolved in 800 mL of 0.02 M ammonium phosphate buffer (pH 7.2), combined with 200 mL EtOH and 7.44 g EDTA, and then adjusted to pH 8 with 1 N NaOH. The EtOH and EDTA were added to prevent some conversion to the protected nonapeptide and to act as a preservative. To this resulting solution was added mg of deacylase enzyme and the reaction was run at pH 8 and stirred at 30 C
for 8 -16 hours. The reaction was then adjusted to pH 6 with 1 N HCI and the protected polymyxin B decapeptide was added to the resin as described in the next section.
The resulting filtrate contains the enzyme.
[077] The completed 1 L deacylation reaction solution was adjusted to pH 6.
Envi-Chrom P resin was added, the mixture was stirred for 1 hour, and the resin was removed by filtration. The resin was placed in a column, washed with 80 mL of 20%
CH3CN in 0.02 M NaH2PO4 adjusted to pH 6.8 with approximately 40 mL of 22.5%
CH3CN in the buffer and 80 mL of 25% CH3CN in the buffer. The deacylated protected Compounds 69 and 70 were then eluted from the resin with 40% CH3CN
in the buffer as 8 mL fractions were collected. Fractions 4, 5, and 6 were combined, evaporated to remove CH3CN, and the residual aqueous solution was freeze dried to obtain 704 mg of crude product mixture. The crude product mixture was mixed with 20 mL of MeOH, stirred for 0.5 hours, and separated by centrifugation. The decant was evaporated to dryness, water was added to the residue, and the solution was freeze dried to obtain 452 mg of deacylated protected Compounds 69 and 70 as a very light tan powder. This product mixture contained about 85% of the deacylated protected Compound 35 and 15% of the deacylated protected Compound 36.

Step (c): Acylation of deacylated protected Compounds 69 and 70.
[078] The deacylated protected mixture of Compounds 69 and 70, 412 mg (approximately 0.16 mmol) was dissolved at room temperature in 4 mL of dimethylformamide (DMF) and 0.4 mL collidine/2M HCI (5:1 volume ratio) to yield a clear solution which was sampled for HPLC as 0-time reaction reference. To the stirred solution was slowly added 80 pL (0.66 mmol) of 2-chiorophenylisocyanate.
After stirring for 30 minutes at room temperature, the reaction was sampled by analytical HPLC and was found to contain approximately 100% conversion to desired acylated protected Compounds 22 and 23 as a mixture.

Step (d): Deprotection of the acylated protected Compounds 22 and 23 [079] To the reaction mixture in Step (c) was added 1.6 mL MeOH (to reduce mixture viscosity) and 0.4 mL piperidine to remove the protecting groups. The mixture was stirred for 60 minutes at room temperature. The reaction mixture was diluted with 80 mL of Solvent A (see below) plus 0.28 mL HOAc (to ensure neutralization of excess piperidine), to yield a clear solution which contained the deprotected products of Formula (I), which were designated as Compound 5 (i.e., R7 is isobutyl) and Compound 6 (i.e., R7 is sec-butyl).

Example 3: Isolation and Purification of Compounds 5 and 6 Preparation of CM-Sepharose Cartridge [080] Fast Flow a carboxymethyl agarose (e.g. sold under the tradename, CM-SEPHAROSE) slurry (100 mL sample) was diluted with approximately 100 mL of 20% EtOH. Approximately 30 mL of the dilute slurry was poured into a 60 mL
polypropylene solid phase extraction tube and the slurry was allowed to settle. The top frit was carefully put in place to exclude air bubbles to obtain final packing bed dimensions of 22 x 26.5 mm. The excess 20% EtOH was decanted and the packing was rinsed by gravity flow with 60 mL of Solvent A.

Product Isolation [081] The diluted reaction mixture was applied to the cartridge and allowed to flow in by gravity. The product-loaded cartridge was rinsed with 60 mL Solvent A
and then rinsed with 24 mL of 0.05 M ammonium acetate at pH 5Ø The cartridge was stripped with 32 mL of 0.27 M sodium sulfate pH 2.3 buffer (1:2 dilution of Stock Buffer A, see below). 4 mL fractions (#4 through #11) were collected and analyzed by HPLC to evaluate product content profile. Fractions 7 through 10 (containing approximately 99% of a mixture of compound 5 and compound 6) were pooled.
Purification by Preparative HPLC
[082] The column was rinsed at room temperature at 10 mL/min with 100 mL
each, in order, Eluent A, Eluent B, Eluent D and Eluent C. The pooled CM-Sepharose fractions were injected onto the column with two sequential injections, approximately 8 mL each, each being rinsed onto the column with 50 mL of Eluent C
at 5.0 mL/min. The gradient was then initiated and all elutions were done at room temperature. The column eluate was monitored at 281 nm (approximately 1 cm flow cell path length). Fraction collection was initiated as Compound 6 started to elute.
7.5 mL fractions were collected until the apex of the major peak followed by 3 mL
fraction collection. Fractions were evaluated by analytical HPLC and the appropriate fractions were pooled. Fractions 8-10 contained Compound 6 and fractions 14-29 contained essentially pure Compound 5.

Product Desalting and Freeze Drying [083] The desalting cartridge was prepared as follows: 2.0 g of EnviChrom-P
styrene/divinylbenzene resin was slurried in 20 mL of 50% acetonitrile then poured into a 20 mL polypropylene cartridge. The top frit was put in place and the excess 50% acetonitrile was drained off. Using gravity flow for all subsequent steps, the packing was rinsed with 24 mL of 67% acetonitrile and then 24 mL of distilled water.
Preparative HPLC fractions 14 through 29 were pooled and the acetonitrile was removed under vacuum at below 35 C. The desolventized fraction pool was applied to the cartridge and the loaded resin was rinsed with multiple 2 mL then 4 mL
increments of distilled water (16 mL total). The desalted product was stripped from the resin using 48 mL of 67% acetonitrile. The acetonitrile was removed from the strip fraction under vacuum at below 35 C and the desolventized product solution was freeze dried to obtain 119 mg of Compound 5, which appeared to be pure by analytical HPLC. The preparative HPLC fractions containing Compound 6 were treated in a similar manner to obtain 15 mg of Compound 6, which also appeared pure by analytical HPLC.
[084] Solvent A: 65% MeOH 0.04M ammonium acetate/0.02M acetic acid pH 5.0, 650 mL HPLC grade MeOH, 40 mL 1.OOM ammonium acetate/0.50M acetic acid, pH 5.0 (1:10 dilution), and distilled water to 1.OOL.
[085] Stock Buffer A: approximately 0.54 M in total sulfate (as sodium sulfate/bisulfate) pH 2.3 (1:10 dilution), 55.3g (30 mL) concentrated sulfuric acid, 76.4g (50 mL) 50% NaOH (approximately 20M), and distilled water to 1.OOL.
Preparative HPLC Conditions [086] Equipment: Waters Prep 4000 pump with Radial-Pak compression unit, Waters Delta-Pak C18 radial-pak cartridges (100A pore, 2.5 x 21 cm), ABI

UV Detector with heat exchanger removed, 10 mL Loop injector, and Pharmacia fraction collector.
[087] Eluent A: 100% isopropanol, Eluent B: 20% isopropanol [088] Eluent C: 15% acetonitrile 0.04M in sodium sulfate (pH 2.3) 150 mL acetonitrile, 80 mL Stock Buffer A, and distilled water to 1.OOL.
[089] Eluent D: 30% acetonitrile 0.04M in sodium sulfate (pH 2.3) 300 mL
acetonitrile, 80 mL Stock Buffer A, and distilled water to 1.OOL.

Elution Gradient: Time(min) %Eluent C %Eluent D Flow(mL/min) 0 100 0 10.0 45 67 33 10.0 45.1 67 33 5.0*
*Note: flow rate is reduced to accommodate limitation of fraction collector [090] Additional compounds were produced using procedures similar to those described above in Examples 1-3 from the corresponding commercially available isocyanates.

Example 4: Elucidation of Formula (Ic) and (Ic") Chemical Structure [091] The chemical structure of Formula (Ic) and (Ic") (designated Compound 5) was determined to include a 2-chlorophenyl carbamyl linked to the N-terminal 2, 4-Diaminobutyric acid of a 10-amino acid peptide. The C-terminal residue of Compound 5, threonine, is linked to the molecule via an amide bond on the a-amino side chain of 2,4-diaminobutyric acid, and the compound contains 10 amino acids which are 6 residues of 2,4-diaminobutyric acids, 2 residues of threonine, and one each of leucine and phenylalanine. The compound has an average molecular weight of 1216.82 with an empirical Formula of C54H86N17O13C1.
[092] The structure shown in Formula (Ic') was deduced based on the method of synthesis from known starting materials and confirmed by HPLC and spectroscopic measurements.
[093] UV-visible spectroscopy was used to determine the electronic absorption of Compound 5. In water, one maximum absorption wavelength was detected. The maximum absorption wavelength is 236 nm with an extinction coefficient of 10565 M-1 cm-1. The UV-VIS spectrum of Compound 5 was acquired on Agilent 8543 UV-Visible system. The spectrum was collected in the absorbance mode. The light-path length used was 1 cm. Compound 5 was dissolved in water at the final concentration of 0.0735 mg/mL prior to the measurement. From the UV-Visible spectrum, one maximum wavelength was observed at 236nm. The calculated extinction coefficient of Compound 5 at 236 nm was 10565 M"1cm-1 [094] Infrared spectroscopy was used to characterize the functional groups present in the Compound 5 molecule. The Compound 5 FTIR sample was prepared and measured by a KBr method. The Compound 5 infrared spectrum was acquired on a Perkin-Elmer 1600 Series FTIR. The spectrum was recorded between 400 cm-1 and 4000 cm-1. The major bands observed in the IR spectrum are 3271.7 cm-1, associated with the amide, amine, and hydroxy (OH) stretches, the 3066.8 and 2929.6 cm-1 vibrations due to the asymmetric and symmetric CH stretching (aromatic and aliphatic respectively), the 1652.1 and 1539.0 cm-1 stretches, associated to the amide I and II band, and finally the 1108.0 cm-1 band, associated to the alcoholic CO
stretches.
[095] The exact mass of Compound 5 was found to be 1216.6350 Da ([M+H]+, Theoretical, 1216.6352 Da). The exact mass of Compound 5 was measured by positive ion electrospray high-resolution mass spectrometry on Micromass Q-Tof API US hybrid quadrupole/time of flight mass spectrometer. The MS/MS data of Compound 5 was recorded in the positive ionization mode by nanospray ionization on a Sciex Q-Star/Pulsar hybrid quadrupole/time-of-flight mass spectrometer. For MS/MS experiments, the m/z 608.86 ion, assigned as [M+2H]2+
was selected. The y-type ion observed at m/z 762.5 is consistent with the structure of the cyclic peptide portion of the Compound 5. The y-type ions at m/z 963.6 and 989.6 are consistent with the presence of the two amino-acid residues, which is attached to the cyclic peptide. Signals at m/z 662.5, 561.4, 461.3, and 361.3 are consistent with the sequence of amino acids in the cyclic peptide of Compound 5.
An intense signal at m/z 254.1 is consistent with the presence of the CIC6H4NHCONHCH(CH2CH2NH2)CO acyl peptide at the N-terminus of the Compound 5. Other signals e.g., m/z 202.1, 302.2 and 101.1 support the sequential assignment of Compound 5. This amino acid sequence information confirmed that Compound 5 is composed of 10 amino acid residues.
[096] Amino acid analysis of Compound 5 was determined by reversed phase HPLC combined to acid hydrolysis and derivatization with AccQ
fluorescence reagent (6-am inoquinolyl-N-hydroxysuccinim idyl carbamate). Derivatized amino acids were excited at 250 nm and monitored at 395 nm. Compound 5 contains 10 amino acids. The amino acid compositions (ratio) are summarized as follows:
Threonine (2.0); 2,4-diaminobutyric acid (5.7); Leucine (1.1); and Phenylalanine (1.1). The amount of 2,4-diaminobutyric acid residues in Compound 5 were found to be 5.7 residues, which is slightly less than the expected amount (6 residues).
This is due to partial hydrolysis of a urea group in Compound 5. This result was consistent with the data from MS/MS analysis.
[097] The amino acid sequence of Compound 5, confirmed with tandem mass spectroscopy and NMR (2D NOESY), falls into the following order: 2-chlorophenyl carbamyl-1 Dab-2Thr-3Dab-4Dab-5Dab-6Phe-7Leu-8Dab-9Dab-1 OThr, where 4Dab was linked to 1 OThr forming a cyclic peptide. The amino acid composition (ratio) of Compound 5 was determined by amino acid analysis.
Stereochemistry of amino acid constituents in Compound 5 was determined by LCMS using Marfey's reagent. The peptide was transformed into its amino acid constituents by hydrolysis under acidic conditions. Each amino acid was derivatized with Marfey's reagent and analyzed by LC/MS. The amino acid stereochemistry of Compound 5 hydrolysate was observed and compared to the individual derivatized amino acid reference standards. Determination of amino acid stereochemistry was achieved by LC retention time and mass identification of each derivatized amino acid using TM-146. The complete amino acid assignment of Compound 5 is the following: N-2-chlorophenylcarbamyl-N2-L-(2,4)-Dab-L-Thr-N2-L-(2,4)-Dab-N2-(4-&)-L-(2,4)-Dab-N2-L-(2,4)-Dab-D-Phe-L-Leu-N2-L-(2,4)-Dab-N2-L-(2,4)-Dab-L-Thr(&).
The 1 H, COSY, TOCSY and NOESY data were primarily used to assign 1 H peaks, while 13C,13 C DEPT, HSQC and HMBC were used to assign 13C data, and to verify 1H assignment. The results from NMR, consistent with data obtained from MS and AAA, confirm the structure of Compound 5.
[098] Nuclear magnetic resonance spectra were acquired at 5 C on a 600 MHz instrument at Custom NMR Services (Ayer, MA). One-dimensional spectra and two-dimensional COSY, TOCSY (mixing time 60 ms), NOESY (mixing time 150 ms), HSQC, HMBC were obtained for Compound 5 reference standard lot CG-01-003.
The sample was dissolved in 150 mM sodium phosphate buffer, pH 6.5 with 90%
H2O and 10% D20 to a concentration of 58 mg/mL.
[099] The 1H, COSY, TOCSY and NOESY data were primarily used to assign 1H peaks, while 13C, 13C DEPT, HSQC and HMBC were used to assign 13C
data, and to verify 1H assignment. The sequential identification of residues in Compound 5 was executed through the analysis of two-dimensional NOESY
spectra. Compound 5 exhibits NOE peaks from backbone NH of one residue to the side chain of the preceding residue which enables the sequential identification of residues. Table 2A gives the proton and carbon chemical shift assignments of the backbone and side chains of Compound 5.

Table 2A. 1H and 13C peak Assignments for Compound 5 at 5 C
Residue NH C=O aH (aC) PH Others Tail* 8.06 157.03 6.98 (127.53), 6.87 (126.72), 7.15 (128.95), 6.90 (128.28), C-NH 133.31, C-Cl not observable Dabl 7.11 173.67 4.11 1.78, 1.93 yH 2.83 (35.82) (51.24) (28.38) Thr2 8.15 171.86 4.01 3.93 (66.30) yH 0.90 (18.20) (58.63) Dab3 8.43 172.29 4.15 1.86, 1.75 yH 2.72 (35.80) (50.51) (27.60) Dab4 8.31 172.50 3.90 1.55 (29.61) yH 3.05, 2.80 (35.60), 6NH
(50.80) 7.44 DabS 8.28 171.43 4.19 1.78, 1.68 yH 2.63, 2.56 (35.43) (49.76) (29.03) Phe6 8.62 173.17 4.12 2.62, 2.84 7.19 (125.65), 7.15 (128.95), (55.98) (35.60) 7.05 (128.28), 6.87 (126.14), C 134.42 Leu7 8.42 174.52 3.81 1.10, 0.95 yH 0.03 (22.22), 6H1 0.32 (50.80) (38.15) (21.80), 6H2 0.23 (19.40) Dab8 7.90 170.96 3.99 1.95 (27.40) yH 2.87, 2.78 (35.43) (51.37) Dab9 8.31 173.01 3.89 1.61, 1.88 yH 2.74 (35.81) (52.43) (27.94) ThrlO 7.89 171.23 3.92 3.80 (59.18) yH 0.87 (18.51) (65.43) * Tail: 2-chlorophenylcarbamyl [0100] Compound 5 has the physical properties listed in Table 2B.

Table 2B

Appearance: White to off white amorphous solid Odor: Odorless pH: 5.9, at 5 mg/mL in water Melting/boiling range: 260 C
Dissociation constant 8.17, 8.62, 9.01, 9.41 and 9.71 (pKa):
Specific rotation: -107 Solubility Profile: > 300 g/L in water 0.2 g/L in ethanol Lipophilicity (Log P) < -1.0 Hygroscopicity: Very hygroscopic Crystallinity: Non-crystalline powder Salt form: Sulfate salts Example 5: Microbiology Studies [0101] To evaluate the potency and spectrum of compounds of Formula (I), (i.e., Compounds designated 5 and 6), a study was conducted that put together a panel of 455 Gram negative strains, which included multi-drug resistant clinical isolates selected from various surveillance programs. Table 3A lists the strains that were included in the study and the activity of Compound 5, Compound 6, and colistin (polymyxin E, abbreviated PME). Strains with acquired resistance to colistin and/or carbapenems and/or broad spectrum cephalosporins were pre-selected and included in all organism groups evaluated.
[0102] MIC testing was performed according to Clinical and Laboratory Standards Institute (CLSI) M7-A7 (2006) broth microdilution methods. Unless otherwise indicated, MIC values are provided in units of micrograms per milliliter.
[0103] One day prior to testing, individual colonies were isolated by streaking onto rich, non-selective Tryptic Soy agar with 5% lysed sheeps blood (TSAB) with incubation at 35-37 C for 18-24 hours.
[0104] Cultures were prepared by touching 3-5 colonies into 3 mL of Cation adjusted Mueller Hinton Broth (caMHB) in a 14 mL tube (CaMHB was prepared and sterilized according to manufacturer's specifications). Cultures were grown at 200 rpm for approximately four hours prior to density adjustment for addition to the MIC assay.
[0105] OD600 of growing cultures was measured and adjusted to approximately 105 colony forming units per mL (CFU/mL) in caMHB for MIC
inoculation (approximately OD600 0.001).
[0106] Diluted cultures were used to inoculate 50 L per well in broth microdilution assays (final volume 100 pl per well; compounds were prepared in two-fold dilutions).
[0107] Plates were incubated 16-20 hrs at 37 C, with shaking at 200 rpm.
[0108] OD600 was determined for all wells. Growth was defined as OD600 >0.1.
MICs were defined as the lowest concentration producing no visible turbidity (0D6oo<0.1).

Table 3A: MIC90 for Compounds of Formula (I) (Compounds 5 and 6) (micrograms/mL) Species # strains Compound Compound PMB PME

P. aeruginosa 100 8 2 2 2 cinetobacter spp 81 2 4 2 4 S. maltophilia 25 16 >16 8 16 . coli 80 4 2 2 2*
pneumonia 81 4 4 2 4*
Citrobacter spp. 20 1 0.5 1 0.5*
P'terobacter 20 >16 >16 >64 >64*
*some strains tested were multi-drug resistant [0109] As shown by Table 3A, Compound 5 showed in vitro activity and spectrum similar to that of colistin (PME). It was active against colistin susceptible P.
aeruginosa and Acinetobacterspp., including multi-drug resistant strains.
Compound 5 was also active against E. coli, K. pneumonia, Citrobacter spp. and had some activity against Enterobacter spp. isolates. Compound 5 showed limited activity against organisms intrinsically resistant to colistin, such as indole-positive Proteae, P. mirabilis, and S. marcescans.
[0110] As shown by Table 3A (showing MIC90 measurements for the indicated strains), Compound 6 also showed in vitro activity and spectrum similar to those of colistin (PME) and Compound 5. For example, Compound 6 was active against colistin susceptible Acinetobacter spp., including multi-drug resistant strains, and was also active against E. coli, K. pneumonia, Citrobacter spp. Compound 6 also had similar activity to compound 5 against Enterobacter spp. isolates.
However, Compound 6 showed an approximately 4-fold decrease in activity against colistin susceptible P. aeruginosa, whereas Compound 5 exhibited similar activity to that of colistin. Thus, for at least this reason, Compound 5 shows surprising and unexpected properties, despite the similar structural resemblance to Compound 6.
Table 3B: MIC for Compounds of Formula (I) (Compounds 3 and 5) (micrograms/mL) pecies Compound 3 Compound 5 cinetobacter baumannii 1570 8 4 . coli 1699 4 2 P. aeruginosa 44 4 1 pneumonia 21 4 2 [0111 ] As shown by Table 3B, Compound 5 showed superior in vitro antibacterial activity and spectrum compared to Compound 3. For example, Compound 5 was more active than Compound 3 against strains of Acinetobacter baumannii, E. coli, P. aeruginosa, and K. pneumonia. Compound 5 was selected for cell culture cytotoxicity based on superior antibacterial properties compared to Compound 3. The 4-chlorophenyl isomer (Compound 3) was less efficacious in comparison to both Compound 5 and polymyxin B.

Cell culture cytotoxicity assay [0112] Cell culture cytoxicity assays were performed on compounds of Formula (II) having in vitro antibacterial properties comparable or better than that of polymyxin B1 (PMB). Surprisingly, as shown in Table 4, the 3-chlorophenyl analog (Compound 4) exhibited higher toxicity (an EC50 of 691) than the structurally similar 2-chlorophenyl analogue (Compound 5, an EC50 of >1000), while both compounds exhibited antibacterial activity comparable to each other and to Polymyxin B
(e.g., MIC values of 2 micrograms/mL in Table 4) against P. aeruginosa.
[0113] Day 1: 1x105 to 2x105 cells were inoculated (Cell line: LLC-PK1) per flask, containing 4mL/T25 flask of medium DMEM-F12 (50/50) media supplemented with 10%FBS (Fetal Bovine Serum). The cells were then incubated at 37 C, 5%
C02, 80% RH.

[0114] Day 4: The cell cultures were examined to determine 80% confluency.
To trypsinize of the 80% confluent flasks, 4 mL of DMEM-F12-10%FBS media was prepared per flask. The media was aspirated from the flask and 5 mL of Versene was added to rinse the flask. The Versene was aspirated and 2 mL of triple Express solution (trypsin solution) was added. The cells were incubated for 5 min at room temperature and examined via microscopy for the rounding of the cells and their detachment. To speed up the reaction, the flask can be incubated at 37 C for minutes. 4 mL of media (DMEM-F12-10%FBS) was added to stop the reaction.
Using a 5 mL pipette, the completed reaction was pipetted up and down to ensure that the cells were properly detached from the flask and from each others. The cell number was counted using a hemocytometer slide. 100 pL of cell suspension was transferred to a tube and 100 pL of Trypan Blue was added. Inoculum for 96 well is 10,000 cells per well. For one 96 well plate, 26.3 pL x 110 well = 2893 pL
total. 2.9 mL of cell suspension was mixed with 8.1 mL of media DMEM-F12-10%FBS and 100 pL was distributed to each well using a multichannel pipetter. The plate was then Incubated for 24h at 37 C.
[0115] Day 5: DMEM-F12 (no FBS) medium was dispensed into tubes with the test article to achieve final concentrations of 1000 mg/mL, 500 mg/mL, 250 mg/mL and 125 mg/mL. The 96 well plate was taken off the incubator and media was removed from each well using multichannel pipetter. 80 pL of serially diluted compounds of Formula (I) was added to each well. Every set of concentration was done in triplicate. Media and positive controls were added to each plate and the plates were incubated for 24 hours at 37 C.
[0116] Day 6: The plates were taken from the incubator, the media was removed from each well, and 100 pL of fresh media DMEM-F1 2 (this is equivalent to a rinse and the media can be replaced with PBS) was added. The media is then removed and 80 pL of cell lysis solution is added. After 5 minutes, 80 pL of sterile filtered high quality water was added to each well. Using a white or black 96 well reaction plate, 80 pL of 25x diluted ATP mix reagent (Sigma Bioluminescent Somatic Cell Assay Kit [cat # FL-ASC]) was added into one column. 80 pL of diluted ATP
standards (dilutions used: 250, 500, 1000, 2000 and 4000) was added and the plate was read immediately in the topcount with a 2 second read time. In another white or black 96 well reaction plate, 80 pL of 25x diluted ATP mix reagent (plate A) was added and 80 pL of lysis-water solution was transferred to each column of plate A.

The plate was then read immediately in the topcount with a 2 second read time.
This process was repeated for next consecutive plate and the EC50 for certain compounds of Formula I were calculated and provided in Table 4.
Table 4: MIC and EC50 data for certain compounds of Formula (II) Compound No. Ri R2 R3 MIC EC5o Rat kidney P. aeruginosa proximal tubule cells micrograms/ml (micrograms/ml 1 -C(O)NH-phenyl -CH2CH(CH3)2 H 1 >1000 2 -C(O)NH-phenyl -CH(CH3)CH2CH3 H 2 >1000 3 -C(O)NH-4- -CH2CH(CH3)2 H 4 chlorophenyl 4 -C(O)NH-3- -CH2CH(CH3)2 H 2 691 chlorophenyl -C(O)NH-2- -CH2CH(CH3)2 H 2 >1000 chlorophenyl 6 -C(O)NH-2- -CH(CH3)CH2CH3 H 1 >1000 chlorophenyl 7 -C(O)NH-2,6- -CH2CH(CH3)2 H 2 >1000 dichlorophenyl 8 -C(O)NH-2,6- -CH(CH3)CH2CH3 H 2 dichlorophenyl 9 -C(O)NH-4- -CH2CH(CH3)2 H 2 502 bromophenyl -C(O)NH-4- -CH2CH(CH3)2 H 2 >1000 fluorophenyl 11 -C(O)NH- -CH2CH(CH3)2 H 2 738 pentafluorophenyl 12 -C(O)NH-2- -CH2CH(CH3)2 H
chlorobenzyl 13 -C(O)NH-2- -CH(CH3)CH2CH3 H 2 chlorobenzyl 14 -C(O)NH-2,4- -CH2CH(CH3)2 H 1 269 dichlorobenzyl -C(O)NH-2,4- -CH(CH3)CH2CH3 H 1 267 Compound No. Ri R2 R3 MIC EC5o Rat kidney P. aeruginosa proximal tubule cells micrograms/ml (micrograms/ml) dichlorobenzyl 16 -C(O)NH-2-chloro- -CH2CH(CH3)2 H 4 >1000 4-nitrophenyl 17 -C(O)NH-2-chloro- -CH(CH3)CH2CH3 H
4-nitrophenyl PMB NA -CH2CH(CH3)2 H 2 318 [0117] As shown by Table 4, Compound 5 showed comparable efficacy to polymyxin B against P. aeruginosa. In addition, Compound 5 is substantially less toxic (EC50 of greater than 1000 in rat kidney proximal tubule cells) when compared to polymyxin B, which has an EC50 of 318. Compound 5 is believed to be unique despite its structrual resemblance to other halogenated or nitro phenyl derviatives.
Example 6: Pharmacological studies [0118] Compound 5 was tested for efficacy in 8 models (both mouse models and rat models) with 5 different Gram negative pathogens. Efficacy was demonstrated in 7 out of 8 of these models. In the one model where Compound 5 did not show efficacy, none of the polymyxin compounds produced efficacy either. In the remaining 7 models, Compound 5 showed comparable efficacy to both polymyxin B and colistin. Data from two representative animal models is shown below.

In Vivo Efficacy of Compound 5 Against Lung Infection in Mice [0119] Infection was induced in normal immunocompetent or neutropenic mice by intranasal inoculation of P. aeruginosa #44. Compound 5, polymyxin B, colistin, imipenem or ciprofloxacin were administered subcutaneously at 1 and 6 hours post-infection. At 24 hours post-infection, mice were humanely euthanized and the lungs of the mice were removed, homogenized, serially diluted, and plated on agar, to quantitate the bacterial burden. Compound 5, polymyxin B, colistin, ciprofloxacin and imipenem were evaluated for their ability to decrease the bacterial burden from the lungs compared with water-treated control mice.

[0120] P. aeruginosa #44 or K. pneumoniae #21 were grown in cation adjusted Mueller Hinton Broth (caMHB) (catalog# B12322; Fisher Scientific, Pittsburgh, PA) at 37 C. MIC testing was performed according to Clinical and Laboratory Standards Institute [CLSI, formerly National Committee for Clinical Laboratory Standards (NCCLS), Wayne, PA] guidelines for broth microdilution.
[0121 ] All of the test compounds were dissolved in sterile water for administration by subcutaneous (sc) injection. To dissolve ciprofloxacin, 2N
HCI was added to the solution in small amounts until all the powder dissolved completely. A
series of dilutions of test compounds were prepared in water.
[0122] In the lung infection model using P. aeruginosa #44 as the infecting organism, CD-1 mice were made neutropenic by intraperitoneal (ip) administration with 150 mg/kg cyclophosphamide on day -4, followed by a second ip dose of 100 mg/kg cyclophosphamide on day -1. On day 0 a diluted, exponential-phase growing culture of P. aeruginosa #44 corresponding to 1x105 or 2.5x108 CFU
respectively, in 0.1 mL of sterile saline, was inoculated by the intranasal route in mice. Just prior to inoculation, mice were anaesthetized with 60 mg/kg pentobarbital, ip. The actual concentration of bacteria in the inoculum was confirmed by determination of viable counts by dilution plating on tryptic soy agar plates. The number of CFU of bacteria was determined after an incubation of 16 h at 37 C.
[0123] Groups of five mice were used for each dose of the test compounds and the vehicle (water) control group. Each group received sc injections of test compounds or vehicle at 1 hour post-infection, and then at 6 hours post-infection for a total of 2 doses. In each study, there were three to five dose groups per test compound.
[0124] Eighteen hours after the last injection, the mice were euthanized by asphyxiation with CO2. The lungs were removed aseptically, homogenized in 4 mL
of sterile, distilled water and dilution plated on tryptic soy agar plates to quantify bacterial CFU. The number of CFU of bacteria in lungs was determined after an incubation of 16 hours at 37 C (the CFU per milliliter of homogenate).
[0125] The results were expressed as the geometric mean loglo CFU/mL the standard deviation. The limit of detection was 10 CFU per mL of lung homogenate.
Lungs were considered sterile when no CFU were detected on the agar. The efficacy of the test compounds was assessed by comparing the number of loglo CFU/mL
measured in the infected and treated mice with control animals treated with water.

The efficacy of Compound 5 was also compared with the positive comparator antibiotics polymyxin B, colistin, ciprofloxacin and imipenem-cilastatin.
[0126] Dose response curves of test compounds were generated for each isolate. Regression lines were generated in Microsoft Excel and used to calculate the dose expected to produce a 3 loglo reduction in bacterial count as compared to water-treated group [ED_31oglo (mg/kg, sc, BID)].
[0127] MIC testing was performed according to CLSI (formerly NCCLS) guidelines for broth microdilution. MICs of the compounds against P.
aeruginosa #44 are shown in Table 5.

Table 5: MIC of Certain Compounds Against P. aeruginosa #44 Compound MIC (micrograms/ml) against P. aeruginosa #44 Colistin 2-4 Polymyxin B 1-2 Imipenem-Cilastatin 1-2 Ciprofloxacin 0.25 [0128] Compound 5 resulted in reduction of bacterial counts of P. aeruginosa #44 in the lungs of infected mice as shown in Table 6.

Table 6: In vivo Efficacy of Compound 5 Against Lung Infections in Mice Mice/ Total daily dosage of Loglo reduction in Bacterial isolate Compound 5 CFU/mL of lung (mg/kg/day) homogenate Neutropenic mice/ 8 1.0 P. aeruginosa #44 16 1.7 24 2.4 32 4.0 40 4.0 [0129] In these studies, the efficacy of Compound 5 compared favorably to that of polymyxin B, colistin, ciprofloxacin, or imipenem-cilastatin against P.
aeruginosa #44 lung infections in mice (see Table 7).

Table 7: Efficacy of Compound 5 and Comparator Antibiotics versus P.
aeruginosa #44 Lung Infection in Neutropenic Mice Drug Dosage Loglo CFU/mL lung ED_3Ioglo (mg/kg, BID)a homogenate SD (mg/kg, BID) Compound 5 4.0 5.49 1.27 8.0 4.86 1.18 12.0 4.13 1.46 9.05b 16.0 2.56 0.34 20.0 2.51 0.70 Polymyxin B 4.0 6.48 1.59 8.0 5.73 1.92 12.0 3.47 1.16 12.97 16.0 2.88 1.07 20.0 2.71 1.09 Imipenem- 2.5 3.33 0.21 2.34 Cilastatin Water water 6.53 0.35 ---Doses were administered 1 and 6 hours post-infection. Lungs were harvested at hours.
b The dose has been corrected for potency of this lot (#CG-01-002) = 711 g/mg.
The actual concentration of bacteria in the infection inoculum was 1x105 CFU
in 0.1 mL
of sterile saline that was inoculated into each mouse.

In vivo efficacy of Compound 5 against thigh infection in mice [0130] Infections were induced in neutropenic mice by intramuscular inoculation of into the left thigh of each mouse. Compound 5, or a comparator drug, either polymyxin B, colistin or imipenem, were administered subcutaneously at hour and 6 hours post-infection. Eighteen hours after the last treatment, mice were euthanized humanely; the infected thighs were removed, homogenized, serially diluted, and plated on agar. The bacterial burden was then quantitated by colony count. Compound 5, polymyxin B, colistin, and imipenem were evaluated for their ability to decrease the bacterial burden in the infected thighs compared with water-treated control mice.
[0131 ] A. baumannii #1570 were grown in cation adjusted Mueller Hinton Broth (caMHB) (catalog# B12322; Fisher Scientific, Pittsburgh, PA) at 37 C.
MIC
testing was performed according to Clinical and Laboratory Standards Institute [CLSI, formerly National Committee for Clinical Laboratory Standards (NCCLS), Wayne, PA] guidelines for broth microdilution.

[0132] All the test compounds were dissolved in sterile water for administration by subcutaneous (sc) injection. A series of dilutions of test compounds were prepared in sterile water.
[0133] Mice were made neutropenic by ip administration with 150 mg/kg cyclophosphamide on day -4, followed by a second ip dose of 100 mg/kg cyclophosphamide on day -1. A diluted exponential-phase growing culture of A.
baumannii #1570 corresponding to 1x107 or 5x104 CFU respectively, in 0.2 mL of sterile saline, was injected into the left thigh of conscious mice on day 0.
The actual concentration of bacteria in the inoculum was confirmed by determination of viable counts by dilution plating on tryptic soy agar plates. The number of CFU of bacteria was determined after an incubation of 16 hours at 37 C.
[0134] Groups of five mice were used for each dose of the test compounds and the vehicle. Each group received sc injections of test compounds or vehicle at 1 hour post-infection, and then at 6 hours post-infection for a total of 2 doses. In each study, there were three to five dose groups per test compound. Eighteen hours after the last injection, the mice were euthanized by asphyxiation with CO2. The left thighs were removed aseptically, homogenized in 4 mL of distilled water and dilution plated on tryptic soy agar plates to quantify bacterial CFU. The number of CFU of bacteria in thighs was determined after an incubation of 16 hours at 37 C (the CFU per milliliter of homogenate).
[0135] The results were expressed as the geometric mean loglo CFU/mL the standard deviation. The limit of detection was 10 CFU per mL of thigh homogenate.
Thighs were considered sterile when no CFU were detected on the agar. The efficacy of the test compounds was assessed by comparing the number of log10 CFU/mL measured in the treated mice with control animals treated with vehicle (water). The efficacy of Compound 5 was also compared with the positive comparator antibiotics polymyxin B, colistin and imipenem-cilastatin.
[0136] Dose response curves of test compounds were generated for each isolate. Regression lines were generated using Microsoft Excel to calculate the dose administered twice daily (BID) predicted to produce a 3 loglo reduction in bacterial count as compared to the water-treated control group [ED_31og1o (mg/kg, sc, BID)].
[0137] MIC testing was performed according to CLSI (formerly NCCLS) guidelines for broth microdilution. MIC of the compounds against A. baumannii #1570 is shown in Table 8.

Table 8: MIC of the compounds against A. baumannii#1570 Compound MIC (microgramslmi) against A. baumannii #1570 Colistin 2 Polymyxin B 1-2 Imipenem-Cilastatin 1-2 [0138] Compound 5 produced a dose-dependent reduction of bacterial counts of the infected thigh muscles induced by A. baumannii #1570 as shown in Table 9.
Table 9: In vivo Efficacy of Compound 5 Against A. baumannii #1570 Thigh Infections in Neutropenic Mice Bacterial isolate Total Daily Dosage of LoglO reduction in (date of study) Compound 5 CFU/mL of thigh (mg/kg/day) homogenate A. baumannii #1570 4.0 0.8 (10/30/07) 8.0 1.1 16.0 2.7 24.0 3.8 [0139] In these studies, Compound 5 compared favorably to polymyxin B, colistin or imipenem-cilastatin against A. baumannii #1570 thigh infections in mice.

Table 10: Efficacy of Compound 5 and Comparator Antibiotics versus A.
baumannii #1570 Thigh Infection in Neutropenic Mice: Study 2 Drug Dosage Logo CFU/mL thigh ED-31oglO
(mg/kg, BID a homogenate SD (mg/kg, BID) Compound 5 2.0 6.93 0.56 4.0 6.64 0.55 64b 8.0 5.06 0.80 12.0 3.97 0.34 Polymyxin B 2.0 6.04 1.10 4.0 4.94 1.03 48 8.0 4.07 0.41 12.0 4.19 0.59 Imipenem- 2.0 7.41 0.15 Cilastatin 4.0 6.83 0.52 9.6 8.0 4.99 1.06 12.0 4.22 0.40 Water water 7.75 0.35 ---a Doses were administered 1 and 6 hours post-infection. Thighs were harvested at 24 hours.
b The dose has been corrected for potency of this lot (#CG-01-002) = 711 g/mg.
The actual concentration of bacteria in the infection inoculum was 5.3x106 CFU
in 0.2 mL of sterile saline that was injected into each mouse in this study performed on 10/30/07.
Example 7: Reduced renal toxicity of Compound 5 compared to polymyxin B
[0140] A comparative 7 day repeat dose safety study in cynomologous monkeys between polymyxin B and Compound 5 was performed.
[0141] Twenty five female, naive cynomolgus monkeys were assigned to one of six dose groups as shown in the table below. Animals were 2.52 kg to 3.77 kg and were 4.2 to 5.7 years old. During acclimation, a chronic femoral intravenous catheter was implanted into all animals to permit continuous infusion. The femoral vein catheter was routed subcutaneously to exit through a prepared incision in the interscapular region. Animals were fitted with jackets connected to flexible stainless steel tethers that protected the catheters.
[0142] Monkeys were dosed twice or three times daily for seven days with control article (0.9% Sodium Chloride for Injection, USP), 25000 IU/kg Compound 5, 37500 IU/kg Compound 5, or 37500 IU/kg PMB (Alpharma, lot number Al 411079).
Each administration was given via a targeted 30-minute intravenous infusion.
The study design for this experiment is shown in Table 11.

Table 11: Study Design for Safety Evaluation in Cynomologous Monkeys Between Polymyxin B and Compound 5 Group Test Dose Total Dose Level/ Dose Level/ Number of Article Frequency Daily Administration Administration Animals Dose (mg/kg) (IU/kg) (Female) Level (IU/kg) 1 Control TID 0 0 0 5 Article 2 Compound BID 75000 3.2 37500 5 3 Compound TID 75000 2.2 25000 4 4 Compound TID 112500 3.2 37500 3 5 Polymyxin BID 75000 3.8 37500 5 B
6 Polymyxin TID 112500 3.8 37500 3 B

[0143] The following parameters were evaluated: daily clinical observations and qualitative food consumption, body weight (twice prior to the first dosing and at necropsy), urinalysis, hematology, coagulation, serum chemistry, toxicokinetics, gross pathology, organ weights, and histopathology of the kidneys and injection sites, and of the liver and sciatic nerves of control and high dose groups.
Animals were evaluated through Study Day 7 and then terminated.
[0144] In this comparative 7-day monkey study with the approved drug polymyxin B, notably less severe nephrotoxicity was observed with Compound 5 when the two drugs were compared at equivalent antibacterial activity doses (75,000 to 112,500 IU/kg), although higher Cmax and AUC values were apparent with polymyxin B.
[0145] The differences in nephrotoxicity between Compound 5 and polymyxin B are summarized in Figure 2. Figure 2 is a scatter plot showing nephrotoxicity data measured as a cumulative score based upon tubular degeneration, tubular regeneration, tubular dilatation, and tubular casts. Each category was scored on a basis from 0 to 4 based on the severity of each finding.
[0146] Primary test article-related changes occurred in the kidneys, and the severity of findings was lower for animals treated with Compound 5 than with an identical dose of polymyxin B. With Compound 5 findings were limited to very minimal histologic changes (minimal renal tubular degeneration, regeneration, casts or acute renal epithelial cell necrosis) in animals receiving 75,000 IU/kg/day. At the 112,500 IU/kg/day Compound 5 dose, histologic kidney changes consisted of increases in numbers and severity of cortical and medullary tubules containing necrotic or degenerative cells as compared to effects noted at the 75,000 IU/kg dose. In contrast, administration of 75,000 IU/kg/day of polymyxin B resulted in renal tubular histology similar to findings from animals receiving 112,500 IU/kg/day of Compound 5 (with an increase in the number of regenerative tubules observed).
These findings were accompanied by mild increases in serum creatinine and BUN
concentrations. In animals receiving 112,500 IU/kg/day of polymyxin B, the extent of histologic findings progressed to involvement of a majority of cortical and some medullary tubules containing necrotic or degenerate cells. These findings correlated with elevations in serum creatinine and blood urea nitrogen concentrations in animals given 112,500 IU/kg/day of polymyxin B.
[0147] For comparison of Compound 5 and polymyxin B, the nephrotoxicity was measured as a cumulative score based upon tubular degeneration, tubular regeneration, tubular dilatation, and tubular casts. Each category was scored on a basis from 0 to 4 based on the severity of each finding and individual monkeys are represented in the Figure 2.
[0148] Compound 5 exhibited consistent and dose-dependent pharmacokinetic profiles in repeat-dose studies in cynomolgus monkeys, which was observed at clinically-relevant dose levels (i.e., <_ 75,000 iu/kg in cynomolgus monkeys). At higher dose levels, a slightly greater than dose-proportional response was evident in systemic exposure (AUC) and peak plasma concentrations (Cmax).
The decrease in clearance at these higher doses may explain the non-linear systemic exposure to Compound 5. The clearance (CL) is less than hepatic blood flow, which suggests Compound 5 has minimal potential for hepatic extraction.
The volume of distribution in all species was greater than blood volume, which suggests extra-vascular distribution of Compound 5.
[0149] The pharmacokinetic characteristics of Compound 5 as compared with those of polymyxin B, exhibited differences, most notably decreased serum protein binding, and increased plasma clearance and volume of distribution, which may lead to decreased potential for toxicity and potentially enhanced antibiotic efficacy through greater tissue distribution. Treatment with polymyxin B resulted in a systemic exposure (AUC) 2.5-fold greater than that observed with Compound 5 treatment at the same activity-normalized dose. Maximal plasma concentrations (Cmax) of polymyxin B were approximately 2-fold greater than Compound 5 concentrations on Day 1, at the comparable activity dose of 37,500 IU/kg for both compounds. The higher systemic exposures evident at the same activity doses of polymyxin B
could lead to greater potential for adverse effects (nephrotoxicity, and possible anaphylactoid reactions) as compared with Compound 5, at equally efficacious dose levels. Systemic clearance of polymyxin B on Day 1 was 2 to 3-fold less than that observed for Compound 5. Further, the volume of distribution observed following polymyxin B administration was approximately 2-fold lower than that observed following Compound 5 administration. The difference in volumes of distribution suggests that Compound 5 has an increased tissue distribution as compared with polymyxin B, which may lead to higher tissue levels (such as in the lung) for effective treatment of bacterial infection. The differences in clearance and volume of distribution between Compound 5 and polymyxin B may be related, partially, to the lower protein binding of Compound 5 (- 30%) as compared to polymyxin B (-56%).
[0150] The kidney findings in female cynomolgus monkeys following seven days of repeated IV administration of Compound 5 were evident at dose levels of 6 mg/kg BID, 6 mg/kg QD, and 9 mg/kg QD, and to a lesser degree of severity at 3 and 4.5 mg/kg BID. Histopathological changes were characterized by minimal to mild degeneration/necrosis and regeneration of tubular epithelium, dilation of renal tubules, subacute interstitial inflammation, and cellular/granular and proteinaceous casts. There was an associated increase in blood urea nitrogen (BUN) and serum creatinine at dose levels of 6 mg/kg BID, 6 mg/kg QD, with 9 mg/kg QD
exhibiting the most extensive kidney effects. The kidney findings in monkeys following seven days of repeated administration of Compound 5 by intravenous infusion were notably less than the marketed comparator polymyxin B when administered at equivalent antimicrobial doses.
[0151 ] Referring again to Figure 2, Compound 5 was markedly less nephrotoxic as assessed micrscopically than polymyxin B (PMB) on a dose normalized basis. Primary test article-related changes occurred in the kidneys, and the severity of findings was lower for animals treated with Compound 5 than with an identical dosage of PMB. The profile of responses to administration of either Compound 5 or PMB were dose-dependent; minimal responses occurred in animals receiving 6.6 mg/kg/day, whereas responses were more extensive in animals receiving 9.9 mg/kg/day. Responses to Compound 5 were limited to very mild histologic changes (minimal renal tubular degeneration, regeneration, casts or acute renal epithelial cell necrosis) in animals receiving 6.6 mg/kg/day (about 3.3 mg/kg BID or about 2.2 mg/kg TID). Because the extent of the renal changes were limited and similar to background changes evident in the vehicle control animals, these effects were not considered to be adverse. At the 9.9 mg/kg/day (3.3 mg/kg TID) dose, histological kidney changes were increased in numbers and severity of cortical and medullary tubules containing necrotic or degenerative epithelial cells, regenerative tubules, casts, and mild interstitial lymphohistiocytic inflammation as compared to effects noted at the 6.6 mg/kg dose. In contrast, administration of 6.6 mg/kg/day of PMB resulted in renal tubular histology similar to findings from animals receiving 9.9 mg/kg/day of Compound 5 (with an increase in the number of regenerative tubules observed). These findings were accompanied by mild increases in serum creatinine and blood urea nitrogen concentrations. In animals receiving 9.9 mg/kg/day of PMB, the extent of histologic findings progressed to involvement of a majority of cortical and some medullary tubules containing necrotic or degenerate epithelial cells, cast, and mild to moderate interstitial lymphohistiocytic and neutrophilic inflammation. These findings were correlated with the macroscopic observation of pale kidney in one animal as well as elevations in serum creatinine and blood urea nitrogen concentrations in animals given 9.9 mg/kg/day of PMB.
[0152] While some embodiments have been shown and described, various modifications and substitutions may be made thereto without departing from the spirit and scope of the invention. For example, it is not intended that the claims set forth hereinafter be construed narrower that the literal language thereof, nor is it intended that exemplary embodiments from the specification be read into the claims.
Accordingly, it is to be understood that the present invention has been described herein by way of illustration only, and that such descriptions do not constitute limitations on the scope of the claims.

Claims (20)

1. A pharmaceutical composition comprising an antibacterial compound of the Formula:

or a pharmaceutically acceptable salt thereof.
2. The composition of claim 1, further comprising a pharmaceutically acceptable carrier, wherein the antibacterial compound is (S)-4-amino-N-((2S,3R)-1-((S)-4-amino-1-oxo-1-((3S,6S,9S, 12S,15R,18S,21 S)-6,9,18-tris(2-aminoethyl)-15-benzyl-3-((R)-1-hydroxyethyl)-12-isobutyl-2,5,8,11,14,17,20-heptaoxo-1,4,7,10,13,16,19-heptaazacyclotricosan-21-ylamino)butan-2-ylamino)-3-hydroxy-1-oxobutan-2-yl)-2-(3-(2-chlorophenyl)ureido)butanamide.
3. The composition of claim 2, wherein the composition is adapted for intravenous administration.
4. The composition of claim 1, wherein the antibacterial compound has the Formula:

5. A method of treating a bacterial infection comprising administering to a subject in need thereof a therapeutically effective amount of a pharmaceutical composition comprising a compound of the Formula:

6. The method according to claim 5, wherein the subject is a human, an animal, a cell culture, or a plant.
7. The method according to claim 5, further comprising the step of determining the type of bacteria in the bacterial infection.
8. The method of claim 7, wherein the type of bacteria in the bacterial infection includes a gram-negative bacteria.
9. The method according to claim 7, wherein the type of bacteria in the bacterial infection includes a susceptible or multi-drug resistant bacteria.
10. The method of claim 7, wherein the type of bacteria in the bacterial infection includes Pseudomonas aeruginosa, Acinetobacter spp, Stenotrophomonas maltophilia, Escherichia coli, Klebsiella pneumoniae, Citrobacter spp, Enterobacter, or a mixture comprising at least one of said bacteria.
11. The method of claim 7, wherein the type of bacteria in the bacterial infection includes Pseudomonas aeruginosa, Acinetobacter spp, Stenotrophomonas maltophilia, Escherichia coli, Klebsiella pneumoniae, Citrobacter spp, Enterobacter, or a mixture comprising at least one of said bacteria.
12.The method of claim 5, wherein the antibacterial compound has the Formula:
13.A process for preparing an antibacterial compound, the process comprising:
(a) treating a polymyxin B with an amino protecting group comprising at least one acidic substituent to form a protected compound ;
(b) treating the protected compound with a deacylating agent, to form at least one deacylated protected compound;
(c) treating the at least one deacylated protected compound with an isocyanate to form at least one acylated protected compound; and (d) treating the at least one acylated protected compound with an organic base to form the antibacterial compound having the Formula:

14. The process of claim 13, wherein a. the deacylating agent is produced by Actinoplanes utahensis; and b. the protecting group is 2-sulfo-9-fluorenylmethoxycarbonyl.
15. The use of a compound of the Formula:
or a pharmaceutically acceptable salt thereof, in the preparation of an antibacterial pharmaceutical composition.
16. The use of claim 15, wherein the pharmaceutical composition is adapted for intravenous administration.
17. The use of claim 16, wherein the pharmaceutically acceptable salt is a sulfate salt.
18. The use of claim 17, wherein the compound has the Formula:
19.A compound of the Formula:

or a salt thereof.
20.The compound of claim 19, wherein the compound is (S)-4-amino-N-((2S,3R)-1-((S)-4-amino-1-oxo-1-((3S,6S,9S, 12S,15R,18S,21 S)-6,9,18-tris(2-aminoethyl)-15-benzyl-3-((R)-1-hydroxyethyl)-12-isobutyl-2,5,8,11,14,17,20-heptaoxo-1,4,7,10,13,16,19-heptaazacyclotricosan-21-ylamino)butan-2-ylamino)-3-hydroxy-1-oxobutan-2-yl)-2-(3-(2-chlorophenyl)ureido)butanamide.
CA2747995A 2008-12-23 2009-12-22 Antibiotic compositions for the treatment of gram negative infections Abandoned CA2747995A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US14040808P 2008-12-23 2008-12-23
US61/140,408 2008-12-23
PCT/US2009/069247 WO2010075416A1 (en) 2008-12-23 2009-12-22 Antibiotic compositions for the treatment of gram negative infections

Publications (1)

Publication Number Publication Date
CA2747995A1 true CA2747995A1 (en) 2010-07-01

Family

ID=42110218

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2747995A Abandoned CA2747995A1 (en) 2008-12-23 2009-12-22 Antibiotic compositions for the treatment of gram negative infections

Country Status (7)

Country Link
US (2) US8343912B2 (en)
AR (1) AR074874A1 (en)
CA (1) CA2747995A1 (en)
NZ (1) NZ593892A (en)
TW (1) TW201035111A (en)
WO (1) WO2010075416A1 (en)
ZA (1) ZA201105290B (en)

Families Citing this family (20)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060004185A1 (en) 2004-07-01 2006-01-05 Leese Richard A Peptide antibiotics and peptide intermediates for their prepartion
WO2010075416A1 (en) 2008-12-23 2010-07-01 Biosource Pharm, Inc. Antibiotic compositions for the treatment of gram negative infections
US8415307B1 (en) 2010-06-23 2013-04-09 Biosource Pharm, Inc. Antibiotic compositions for the treatment of gram negative infections
ES2708997T3 (en) * 2011-10-26 2019-04-12 Allergan Inc Amide derivatives of N-Urea substituted amino acids as modulators of the receptor analogue to formyl peptide receptor 1 (FPRL-1)
WO2013072695A1 (en) * 2011-11-18 2013-05-23 Novacta Biosystems Limited Polymyxin derivatives
EP2679236A1 (en) 2012-06-29 2014-01-01 Zentrum für biomedizinische Technologie der Donau- Universität Krems Metering guide for lipopeptides that bind endotoxin
CA2939061C (en) 2013-05-22 2023-10-24 New Pharma Licence Holdings Limited Polymyxin derivatives and their use in combination therapy together with different antibiotics
AU2015228898B2 (en) * 2014-03-11 2018-11-15 Spero Therapeutics, Inc. Polymyxin derivatives and their use in combination therapy together with different antibiotics
CN106414480B (en) * 2014-04-01 2020-08-04 莫纳什大学 Polymyxin derivatives as antimicrobial compounds
WO2015172047A1 (en) * 2014-05-08 2015-11-12 Merck Sharp & Dohme Corp. Cyclic peptide compounds and related methods, salts and compositions
WO2016100578A2 (en) 2014-12-16 2016-06-23 Micurx Pharmaceuticals, Inc. Antimicrobial polymyxins for treatment of bacterial infections
AU2016331658B2 (en) * 2015-09-29 2021-04-01 Monash University Antimicrobial polymyxin derivative compounds
EP3344255B1 (en) 2016-06-01 2019-08-28 Gazel, Deniz Antimicrobial effect of methylene blue on colistin resistant acinetobacter baumannii bacteria
WO2017218922A2 (en) * 2016-06-17 2017-12-21 Cidara Therapeutics, Inc. Compositions and methods for the treatment of bacterial infections
US20190315806A1 (en) * 2016-12-16 2019-10-17 Institute Of Medicinal Biotechnology, Chinese Academy Of Medical Sciences Polymyxin derivative, preparation method and application thereof
US20200188476A1 (en) * 2017-04-18 2020-06-18 The Regents Of The University Of California Anti-microbial agent-polymer conjugates and methods of use thereof
MX2020013811A (en) 2018-06-25 2021-03-09 Spero Therapeutics Inc Compounds.
CN109206486B (en) * 2018-09-03 2020-11-10 杭州中美华东制药有限公司 Polymyxin B sulfate impurity and preparation method thereof
EP3636659A1 (en) * 2018-10-08 2020-04-15 Universitat de Barcelona Polymyxin-based compounds useful as antibacterial agents
US20230141981A1 (en) 2020-01-21 2023-05-11 Shanghai Micurx Pharmaceuticals Co., Ltd Novel compounds and composition for targeted therapy of kidney-associated cancers

Family Cites Families (119)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3854480A (en) 1969-04-01 1974-12-17 Alza Corp Drug-delivery system
US3753970A (en) 1970-08-04 1973-08-21 Rhone Poulenc Sa Cyclopeptides derived from polymyxins and their preparation
FR2124060B1 (en) 1971-02-02 1974-04-12 Rhone Poulenc Sa
US4091092A (en) 1976-09-20 1978-05-23 E. R. Squibb & Sons, Inc. Polymyxin F and process of producing polymyxin F
IE52535B1 (en) 1981-02-16 1987-12-09 Ici Plc Continuous release pharmaceutical compositions
US5366734A (en) 1981-02-16 1994-11-22 Zeneca Limited Continuous release pharmaceutical compositions
ES8403890A1 (en) 1982-02-27 1984-04-01 Beecham Group Plc Antibacterial 1-normon-2-yl-heterocyclic compounds.
ATE37983T1 (en) 1982-04-22 1988-11-15 Ici Plc DELAYED RELEASE AGENT.
USRE32310E (en) 1982-05-21 1986-12-16 Eli Lilly And Company Derivatives of A-21978C cyclic peptides
USRE32311E (en) 1982-05-21 1986-12-16 Eli Lilly And Company Derivatives of A-21978C cyclic peptides
US4524135A (en) 1982-05-21 1985-06-18 Eli Lilly And Company A-21978C cyclic peptides
US4482487A (en) 1982-05-21 1984-11-13 Eli Lilly And Company A-21978C cyclic peptides
US4537717A (en) 1982-05-21 1985-08-27 Eli Lilly And Company Derivatives of A-21978C cyclic peptides
US4399067A (en) 1982-05-21 1983-08-16 Eli Lilly And Company Derivatives of A-21978C cyclic peptides
GB2128617A (en) 1982-10-06 1984-05-02 Martti Vaara Polypeptides for use in antibacterial therapy
US4452775A (en) 1982-12-03 1984-06-05 Syntex (U.S.A.) Inc. Cholesterol matrix delivery system for sustained release of macromolecules
US4601893A (en) 1984-02-08 1986-07-22 Pfizer Inc. Laminate device for controlled and prolonged release of substances to an ambient environment and method of use
US4895566A (en) 1986-07-25 1990-01-23 C. R. Bard, Inc. Coating medical devices with cationic antibiotics
EP0258672A3 (en) 1986-08-07 1989-07-26 MEDICEChem.-Pharm. Fabrik Pütter GmbH & Co. KG Cationic surfactants and pharmaceutical compositions containing them
US4830860A (en) 1986-10-30 1989-05-16 Pfizer Inc. Stressed polymeric device for controlled release of a substance to an ambient environment
US5755788A (en) 1987-02-19 1998-05-26 Rutgers, The State University Prosthesis and implants having liposomes bound thereto and methods of preparation
US20010021697A1 (en) * 1987-09-14 2001-09-13 Henning Lowenstein Methods and compositions for the treatment of mammalian infections employing medicaments comprising hymenoptera venom, proteinageous or polypeptide components thereof, or analogues of such proteinaceous or polypeptide components
US5039789A (en) 1988-04-11 1991-08-13 Eli Lilly And Company A54145 cyclic peptides
US5028590A (en) 1988-04-11 1991-07-02 Eli Lilly And Company Derivatives of A54145 cyclic peptides
US5254535A (en) 1989-04-17 1993-10-19 The Children's Hospital Of Pennsylvania Composition and treatment with biologically active peptides and antibiotic
US5112614A (en) 1989-09-14 1992-05-12 Alza Corporation Implantable delivery dispenser
US5459237A (en) 1990-02-08 1995-10-17 Magainin Pharmaceuticals Inc. Peptide compositions and uses therefor
US5082653A (en) 1990-10-31 1992-01-21 Warner-Lambert Company Anti-plaque compositions comprising a combination of morpholinoamino alcohol and antibiotic
JPH04167172A (en) 1990-10-31 1992-06-15 Nec Corp Vector processor
US5686065A (en) 1991-03-27 1997-11-11 Special Advanced Biomaterials, Inc. Topical siloxane sunscreen compositions having enhanced performance and safety
AU654714B2 (en) 1991-04-08 1994-11-17 Magainin Pharmaceuticals, Inc. Novel peptide compositions and uses therefor
EP0667871A1 (en) 1991-09-13 1995-08-23 Magainin Pharmaceuticals Inc. Biologically active amphiphilic peptide compositions and uses therefor
TW274552B (en) 1992-05-26 1996-04-21 Hoechst Ag
US6348445B1 (en) 1992-06-01 2002-02-19 Magainin Pharmaceuticals, Inc. Biologically active peptides with reduced toxicity in animals and a method for preparing same
US5283005A (en) 1992-10-08 1994-02-01 Olin Corporation Synergistic biocide combination for industrial fluids
US5424290A (en) 1992-10-26 1995-06-13 Magainin Pharmaceuticals Inc. Biologically active peptides and uses therefor
US5360788A (en) 1992-11-10 1994-11-01 Olin Corporation Personal care composition containing pyrithione and a basic lipopeptide
US5370876A (en) 1993-01-08 1994-12-06 Microbarriers Antimicrobial protective skin composition and method for protecting skin from body fluids
US5654451B1 (en) 1993-01-14 2000-02-22 Magainin Pharma Amino acids and peptides having modified c-terminals and modified n-terminals
CA2112776C (en) 1993-01-21 2002-11-12 Masakazu Tsuchiya Process for inhibiting activity of endotoxin
US5652332A (en) 1993-03-12 1997-07-29 Xoma Biologically active peptides from functional domains of bactericidal/permeability-increasing protein and uses thereof
US20040023884A1 (en) 1993-03-12 2004-02-05 Xoma Corporation Biologically active peptides from functional domains of bactericidal/permeability-increasing protein and uses thereof
US5733872A (en) 1993-03-12 1998-03-31 Xoma Corporation Biologically active peptides from functional domains of bactericidal/permeability-increasing protein and uses thereof
US6180134B1 (en) 1993-03-23 2001-01-30 Sequus Pharmaceuticals, Inc. Enhanced ciruclation effector composition and method
US5635216A (en) 1993-12-16 1997-06-03 Eli Lilly And Company Microparticle compositions containing peptides, and methods for the preparation thereof
JPH09504512A (en) 1993-08-13 1997-05-06 スミスクライン・ビーチャム・パブリック・リミテッド・カンパニー Derivatives of monic acids A and C having antibacterial, antimycoplasmal, antifungal and herbicidal activities
US5587358A (en) 1994-05-09 1996-12-24 Asahi Kasei Kogyo Kabushiki Kaisha Potentiators of antimicrobial activity
FR2735983B1 (en) 1995-06-29 1997-12-05 Centre Nat Rech Scient PEPTIDE FOR MODIFYING THE ACTIVITY OF THE HUMAN OR ANIMAL IMMUNE SYSTEM
US5849761A (en) 1995-09-12 1998-12-15 Regents Of The University Of California Peripherally active anti-hyperalgesic opiates
US20020150964A1 (en) 1995-12-19 2002-10-17 Centre National De La Recherche Scientifique Peptides for the activation of the immune system in humans and animals
US6011008A (en) 1997-01-08 2000-01-04 Yissum Research Developement Company Of The Hebrew University Of Jerusalem Conjugates of biologically active substances
US6004573A (en) 1997-10-03 1999-12-21 Macromed, Inc. Biodegradable low molecular weight triblock poly(lactide-co-glycolide) polyethylene glycol copolymers having reverse thermal gelation properties
US6846478B1 (en) 1998-02-27 2005-01-25 The Procter & Gamble Company Promoting whole body health
US6350438B1 (en) 1998-02-27 2002-02-26 The Procter & Gamble Company Oral care compositions comprising chlorite and methods
US6350738B1 (en) 1998-03-06 2002-02-26 Brigham Young University Steroid derived antibiotics
US6767904B2 (en) 1998-03-06 2004-07-27 Bringham Young University Steroid derived antibiotics
NZ510690A (en) 1998-09-25 2002-10-25 Cubist Pharm Inc Methods for administration of antibiotics
IT1308180B1 (en) 1999-02-12 2001-12-07 Chiesi Farma Spa SYNTHETIC PEPTIDES HAVING THE ABILITY TO DECREASE SURFACE TENSION AND THEIR USE IN THE PREPARATION OF A SURFACTANT
US6515104B1 (en) 1999-06-25 2003-02-04 Xoma Technology Ltd. Therapeutic peptide-based constructs derived from domain II of bactericidal/permeability-increasing protein
US6964948B2 (en) 1999-06-25 2005-11-15 Xoma Technology Ltd. Therapeutic peptide-based constructs derived from Domain II of bactericidal/permeability-increasing protein
WO2001005815A1 (en) 1999-07-15 2001-01-25 Eli Lilly And Company Process for deacylation of lipodepsipeptides
US6380356B1 (en) 1999-12-07 2002-04-30 Advanced Medicine, Inc. Multivalent polymyxin antibiotics
WO2001044272A2 (en) 1999-12-15 2001-06-21 Cubist Pharmaceuticals, Inc. Daptomycin analogs as antibacterial agents
BR0017028A (en) 1999-12-15 2003-01-07 Cubist Pharm Inc Lipopeptides as antibacterial agents
IL150225A0 (en) 1999-12-15 2002-12-01 Cubist Pharm Inc Lipopeptides, pharmaceutical compositions containing the same and methods for the production thereof
US7074392B1 (en) 2000-03-27 2006-07-11 Taro Pharmaceutical Industries Limited Controllled delivery system of antifungal and keratolytic agents for local treatment of fungal infections
EP1284992A2 (en) 2000-05-30 2003-02-26 Viridis Biotech Inc. Polyubiquitin based hydrogel and uses thereof
CA2415068C (en) 2000-06-30 2009-03-10 The Procter & Gamble Company Oral compositions comprising antimicrobial agents
AU7893301A (en) 2000-07-17 2002-01-30 Intrabiotics Pharmaceuticals Antimicrobial sulfonamide derivatives of lipopeptide antibiotics
US6511962B1 (en) 2000-07-17 2003-01-28 Micrologix Biotech Inc. Derivatives of laspartomycin and preparation and use thereof
DE60239316D1 (en) 2001-01-16 2011-04-14 Univ Ramot HYBRID PEPTIDES FOR THE TREATMENT OF BACTERAEMY AND SEPSIS
CN1489588A (en) 2001-01-31 2004-04-14 �Ʒ� Thiazolyl-oxazolyl-pyrrolyl-and imidazolyl-acid amide derivatives useful as inbibitors of PDE 4 isozymes
WO2002074316A1 (en) 2001-03-15 2002-09-26 Enteron Pharmaceuticals, Inc. Method of treating inflammatory disorders of the gastrointestinal tract using topical active corticosteroids
WO2003014147A1 (en) 2001-08-06 2003-02-20 Cubist Pharmaceuticals, Inc. Novel depsipeptides and process for preparing same
US6681765B2 (en) 2001-12-18 2004-01-27 Sheree H. Wen Antiviral and antibacterial respirator mask
US7345018B2 (en) 2002-04-25 2008-03-18 Reception Aps Method of treating side effects induced by therapeutic agents
EP1360961A1 (en) 2002-05-07 2003-11-12 AM-Pharma B.V. Use of antimicrobial peptides for potentiating the activity of antimicrobial agents
US6767718B2 (en) 2002-05-10 2004-07-27 Biosource Pharm, Inc. Lipodepsipeptide antibiotics and methods of preparation
NZ538628A (en) 2002-08-12 2008-06-30 Dynavax Tech Corp Immunomodulatory compositions, methods of making, and methods of use thereof
US20040037895A1 (en) 2002-08-23 2004-02-26 Alex Zhu Methods of treating involuntary facial spasms and facial wrinkles
US20060104968A1 (en) 2003-03-05 2006-05-18 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminogly ycanases
US20090123367A1 (en) 2003-03-05 2009-05-14 Delfmems Soluble Glycosaminoglycanases and Methods of Preparing and Using Soluble Glycosaminoglycanases
US7871607B2 (en) 2003-03-05 2011-01-18 Halozyme, Inc. Soluble glycosaminoglycanases and methods of preparing and using soluble glycosaminoglycanases
WO2004101017A2 (en) 2003-05-16 2004-11-25 Blue Membranes Gmbh Medical implants comprising biocompatible coatings
CN1886128A (en) * 2003-09-30 2006-12-27 柯西有限公司 Compositions and methods for treating burns
WO2005046746A2 (en) 2003-11-10 2005-05-26 Angiotech International Ag Medical implants and fibrosis-inducing agents
US7364747B1 (en) 2004-02-02 2008-04-29 S.S. Steiner, Inc. Process and product for inhibiting or preventing bacterial infections
US20060004185A1 (en) 2004-07-01 2006-01-05 Leese Richard A Peptide antibiotics and peptide intermediates for their prepartion
WO2006024741A2 (en) 2004-07-30 2006-03-09 Palumed S.A. Hybrid qa molecules wherein q is an aminoquinoline and a is an antibiotic residue, their synthesis and their uses as antibacterial agent
US7541046B1 (en) 2005-01-04 2009-06-02 Gp Medical, Inc. Nanoparticles for protein drug delivery
WO2006089236A1 (en) 2005-02-18 2006-08-24 The Cleveland Clinic Foundation Apparatus and methods for replacing a cardiac valve
US20060259135A1 (en) 2005-04-20 2006-11-16 The Cleveland Clinic Foundation Apparatus and method for replacing a cardiac valve
ATE442108T1 (en) 2005-07-15 2009-09-15 Cleveland Clinic Foundation DEVICE FOR REMODELING A HEART VALVE RING
WO2007041677A2 (en) 2005-10-03 2007-04-12 Combinatorx, Incorporated Soft tissue implants and drug combination compositions, and use thereof
US20070299043A1 (en) 2005-10-03 2007-12-27 Hunter William L Anti-scarring drug combinations and use thereof
US20070197957A1 (en) 2005-10-03 2007-08-23 Hunter William L Implantable sensors, implantable pumps and anti-scarring drug combinations
US20070208134A1 (en) 2005-10-03 2007-09-06 Hunter William L Anti-scarring drug combinations and use thereof
KR100750658B1 (en) 2005-12-09 2007-08-20 한국생명공학연구원 Polymyxin synthetase and gene cluster thereof
WO2007089864A2 (en) 2006-01-30 2007-08-09 Angiotech Pharmaceuticals, Inc. Sutures and fibrosing agents
US9155792B2 (en) 2006-02-13 2015-10-13 Trustees Of Boston University RecA inhibitors with antibiotic activity, compositions and methods of use
WO2007095631A2 (en) 2006-02-15 2007-08-23 The Regents Of The University Of California New drug delivery system for crossing the blood brain barrier
US20070197658A1 (en) 2006-02-22 2007-08-23 David Sunil A Polyamines and their use as antibacterial and sensitizing agents
US8585753B2 (en) 2006-03-04 2013-11-19 John James Scanlon Fibrillated biodegradable prosthesis
US20070243275A1 (en) 2006-04-13 2007-10-18 Gilbard Jeffrey P Methods and compositions for the treatment of infection or infectious colonization of the eyelid, ocular surface, skin or ear
US8652201B2 (en) 2006-04-26 2014-02-18 The Cleveland Clinic Foundation Apparatus and method for treating cardiovascular diseases
US8771712B2 (en) 2006-05-09 2014-07-08 Emisphere Technologies, Inc. Topical administration of acyclovir
CA2659391A1 (en) 2006-08-03 2008-02-07 Prosensa Technologies B.V. Antibiotic composition
US7807637B2 (en) 2006-08-11 2010-10-05 Northern Antibiotics Oy Polymyxin derivatives and uses thereof
SI2057185T1 (en) 2006-08-11 2016-10-28 Northern Antibiotics Oy Polymyxin derivatives and uses thereof
US20100028334A1 (en) 2006-12-15 2010-02-04 Trustees Of Boston University Compositions and methods to potentiate colistin activity
CA2682745C (en) 2007-04-02 2014-12-23 Sideromics, Llc Methods for preventing or treating infectious diseases caused by extracellular microorganisms, including antimicrobial-resistant strains thereof, using gallium compounds
US8066689B2 (en) 2007-07-11 2011-11-29 Apollo Endosurgery, Inc. Methods and systems for submucosal implantation of a device for diagnosis and treatment with a therapeutic agent
US20090048155A1 (en) 2007-08-15 2009-02-19 Endacea, Inc. Methods for preventing and treating tissue injury and sepsis associated with Yersinia pestis infection
US20090214601A1 (en) 2007-09-28 2009-08-27 Chappa Ralph A Porous Drug Delivery Devices and Related Methods
US20090227018A1 (en) 2007-10-25 2009-09-10 Revalesio Corporation Compositions and methods for modulating cellular membrane-mediated intracellular signal transduction
US8329645B2 (en) 2008-02-08 2012-12-11 Northern Antibiotics Ltd. Polymyxin derivatives and uses thereof
US8193148B2 (en) 2008-02-08 2012-06-05 Northern Antibiotics Ltd. Short fatty acid tail polymyxin derivatives and uses thereof
WO2010075416A1 (en) 2008-12-23 2010-07-01 Biosource Pharm, Inc. Antibiotic compositions for the treatment of gram negative infections
US8415307B1 (en) 2010-06-23 2013-04-09 Biosource Pharm, Inc. Antibiotic compositions for the treatment of gram negative infections

Also Published As

Publication number Publication date
AR074874A1 (en) 2011-02-16
NZ593892A (en) 2013-11-29
US20130345121A1 (en) 2013-12-26
US8937040B2 (en) 2015-01-20
TW201035111A (en) 2010-10-01
US8343912B2 (en) 2013-01-01
US20100160215A1 (en) 2010-06-24
WO2010075416A1 (en) 2010-07-01
ZA201105290B (en) 2012-03-28

Similar Documents

Publication Publication Date Title
US8937040B2 (en) Antibiotic compositions for the treatment of gram negative infections
US8415307B1 (en) Antibiotic compositions for the treatment of gram negative infections
US9234006B2 (en) Compounds
JP7117021B2 (en) Polymyxin derivatives as antibacterial compounds
US8889826B2 (en) Peptide antibiotics and methods for making same
CA2865791A1 (en) Antifungal agents and uses thereof
US20180282374A1 (en) Antimicrobial polymyxin derivative compounds
JPH04217698A (en) Lipopeptide derivative
JPH09500103A (en) Cyclohexapeptidyl aminoalkyl ether
JPH04217699A (en) Lipopeptide derivative
CA2949328C (en) Low substituted polymyxins and compositions thereof
JP2003505396A (en) Pseudomycin prodrug
EP0539088B1 (en) Echinocandin b derivative
EP3197908A1 (en) Novel peptide derivatives and uses thereof

Legal Events

Date Code Title Description
FZDE Dead

Effective date: 20141223