Login| Sign Up| Help| Contact|

Patent Searching and Data


Title:
PHOSPHONATED GLYCOPEPTIDE AND LIPOGLYCOPEPTIDE ANTIBIOTICS AND USES THEREOF FOR THE PREVENTION AND TREATMENT OF BONE AND JOINT INFECTIONS
Document Type and Number:
WIPO Patent Application WO/2008/077241
Kind Code:
A1
Abstract:
The present invention is directed to antimicrobial compounds which have an affinity for binding bones. More particularly, the invention is directed to phosphonated derivatives of glycopeptide or lipoglycopeptide antibiotics. These compounds are useful as antibiotics for the prevention or treatment of bone and joint infections, especially for the prevention and treatment of osteomyelitis.

Inventors:
TANAKA KELLY (CA)
RAFAI FAR ADEL (CA)
CIBLAT STEPHANE (CA)
DIETRICH EVELYNE (CA)
Application Number:
PCT/CA2007/002288
Publication Date:
July 03, 2008
Filing Date:
December 21, 2007
Export Citation:
Click for automatic bibliography generation   Help
Assignee:
TARGANTA THERAPEUTICS INC (CA)
TANAKA KELLY (CA)
RAFAI FAR ADEL (CA)
CIBLAT STEPHANE (CA)
DIETRICH EVELYNE (CA)
International Classes:
C07K9/00; A61K31/663; A61K38/08; A61K38/14; A61P19/08; A61P31/04; C07F9/38; C07K7/06
Domestic Patent References:
WO1996040156A11996-12-19
WO1998015560A11998-04-16
Foreign References:
US20010041689A12001-11-15
US5840684A1998-11-24
US5750509A1998-05-12
US6635618B22003-10-21
Other References:
See also references of EP 2097440A4
Attorney, Agent or Firm:
FASKEN MARTINEAU DUMOULIN LLP (Suite 3400 P.O. Box 242,800 Square Victori, Montréal Québec H4z 1E9, CA)
Download PDF:
Claims:
WHAT IS CLAIMED IS:

1 A compound represented by Formula (I)

or a pharmaceutically acceptable salt, ester or prodrug thereof, wherein A is a glycopeptide or lipoglycopeptide antimicrobial molecule, B is a phosphonated group,

L is a bond or a linker for covalently coupling B to A, and α is 1 , 2, 3, 4, 5, 6 or 7

2 The compound of claim 1 , wherein α is 1 ,2 or 3

3 The compound of claim 1 , wherein B is a bisphosphonate

4 The compound of claim 1 , wherein B has high affinity to osseous tissues

5 The compound of claim 3, wherein said bisphosphonate is selected from the group consisting of

wherein each R* is independently selected from the group consisting of H, lower alkyl, cycloalkyl, aryl and heteroaryl, with the proviso that at least two R* are H,

X is H, OH, NH 2 , or a halo group,

X I are both H, or each is independently selected from the group consisting of H, OH, NH 2 , and a halo group, and

L 1 is the point of attachment to L

6 The compound of claim 1 , wherein L is a hydrolysable linker

7 The compound of claim 1 , wherein B— L— is represented by the following formula (BLi)

wherein

A a indicates the point of attachment to the glycopeptide or lipoglycopeptide antimicrobial molecule A,

W is a covalent bond or is selected from the group of

T is oxygen or sulfur, each R is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic, amino, substituted ammo, hydroxyl, alkoxy, substituted alkoxy, aryloxy, substituted aryloxy, and — R a — Y— R b — Y— R b — B, each R a is independently selected from the group consisting of a covalent bond, alkylene, substituted alkylene, alkenylene, substituted alkenylene, alkynylene, substituted alkynylene, arylene, substituted arylene, —(CO)— alkylene— , substituted —(CO)— alkylene— , —(CO)— alkenylene— , substituted — (CO) — alkenylene — , — (CO) — alkynylene — , substituted — (CO) — alkynylene — , — (CO) — arylene — and substituted — (CO) — arylene — , each R b is independently selected from the group consisting of a covalent bond, alkylene, substituted alkylene, alkenylene, substituted alkenylene, alkynylene, substituted alkynylene, arylene and substituted arylene,

each Y is independently selected from the group consisting of a covalent bond, -CH 2 -, — O— , -S-, -S-S-, — NR C — , -S(O)-, -SO 2 -, -NR 0 C(O)- -OSO 2 -, -OC(O)- — N(R°)S0 2 — — C(O)NR C — , -C(O)O-, -SO 2 NR 0 -, -SO 2 O-, — P(O)(OR°)O— , — P(O)(OR C )NR°— , — OP(O)(OR C )O— , — OP(O)(OR°)NR C — , -OC(O)O-, -NR 0 C(O)O-, -NR 0 C(O)NR 0 -, -OC(O)NR 0 -, -C(O)-, and -N(R C )SO 2 NR°— , each R° is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic and — C(0)R d — , each R d is independently selected from the group consisting of alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl and heterocyclic,

B is a phosphonated group, each Q is independently selected from the group consisting of nitro, chloro, bromo, iodo and fluoro, each X is independently selected from the group consisting of — O — , — S — , and -N(R)-

Z is selected from the group consisting of hydrogen, acyl, substituted acyl, aroyl, substituted aroyl, alkoxycarbonyl, substituted alkoxycarbonyl, aryloxycarbonyl, substituted aryloxycarbonyl,

q is 2 or 3, r is 1 , 2, 3, 4 or 5,

Wi and W 2 are each integers ≥ O such that their sum (wi + w 2 ) is 1 , 2 or 3, a, b, c, d are integers ≥ O such that a+b+c+d ≤7 or null, e and f are integers ≥ O such that e+f = 4, α is O or 1

8 The compound of claim 1 , wherein at least one of said B-L — is coupled to a hydroxyl functionality on said glycopeptide or lipoglycopeptide antibiotic A, and wherein each of said B — L — coupled to a hydroxyl functionality is independently selected from the group consisting of

wherein: B represents said phosphonated group; each p is independently 0 or an integer < 10; each R L is independently selected from the group consisting of H, ethyl and

methyl; q is 2 or 3; n is an integer < 10; r is 1 , 2, 3, 4 or 5; and

W-) and W 2 are each integers > 0 such that their sum (wi + w 2 ) is 1 , 2 or 3.

9. The compound of claim 1 , wherein at least one of said B — L — is coupled to a nitrogen atom on said glycopeptide or lipoglycopeptide antibiotic A, and wherein each of said B — L— coupled to a nitrogen atom is independently selected from the group consisting of:

wherein

B represents said phosphonated group, n is an integer < 10, each p is independently 0 or an integer < 10, each R L IS independently selected from the group consisting of H, ethyl and methyl, q is 2 or 3, r is 1 , 2, 3, 4 or 5,

Wi and W 2 are each integers ≥ 0 such that their sum (W 1 + w 2 ) is 1 , 2 or 3,

X iS -CH 2 -, -CONRL-, -CO-O-CH 2 - Or -CO-O-, each Y is independently selected from the group consisting of — O — , — S — and —NR L —, each Z is indepedently selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, acyl, acyloxy, carboxy, carbamoyl, sulfuryl, sulfinyl, sulfenyl, sulfonyl, mercapto, amino, hydroxyl, cyano and nitro, wherein s is 1 ,

2, 3 or 4, and

Ra is C x H y where x is an integer of 0 to 20 and y is an integer of 1 to 2x+1

10 The compound of claim 1 , wherein at least one of said B — L — is coupled to the carbonyl of a carboxylate group on said glycopeptide or lipoglycopeptide antibiotic A,

and wherein each of said B-L — coupled to the carbonyl of a carboxylate group is independently selected from the group consisting of

wherein n is an integer < 10, p is 0 or an integer < 10,

R L IS H, ethyl or methyl,

R x is -S-, -C(RL) 2 -, — NR L — or —0—,

each Y is independently selected from the group consisting of — O — , — S — , and — NR L — , each Z is independently selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, acyl, acyloxy, carboxy, carbamoyl, sulfuryl, sulfinyl, sulfenyl, sulfonyl, mercapto, ammo, hydroxyl, cyano and nitro, wherein s is 1 ,

2, 3 or 4, and

B represents the phosphonated group

11 The compound of claim 1 , wherein A has a structure represented by the following Formula (A 1 )

or a pharmaceutically acceptable salt, ester or prodrug thereof, wherein

R 1 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic and — R a — Y — R b — (Z) x , or R 1 is a saccharide group optionally substituted with

— R a — Y— R b — (Z) x , — R f , —C(O)R f , or — C(O)- R a — Y— R b — (Z) x ,

R 2 is hydrogen or a saccharide group optionally substituted with

— R a — Y— R b — (Z) x , — R f , — C(O)R f , or — C(O)- R a — Y— R b — (Z) x ,

R 3 is — OR 0 , — NR C R C , — 0— R a — Y— R b — (Z) x , — NR C — R a — Y— R b — (Z) x ,

— NR c R e , or — O— R e ,

R 4 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, — R a — Y— R b — (Z) x ,

— C(O)R d and a saccharide group optionally substituted with

_R a _γ_R b _(Z) x , — R f , or — C(O)- R a — Y— R b — (Z) x , or R 4 and R 5 can be joined, together with the atoms to which they are attached, to form a heterocyclic ring optionally substituted with — NR 0 — R a — Y — R b — (Z) x ,

R 5 is selected from the group consisting of hydrogen, halo, — CH(R 0 )- NR C R C , — CH(R 0 )- NR 0 R 6 , — CH(R 0 )- NR 0 - R a — Y— R b — (Z) x , — CH(R C )— R", and — CH(R 0 )- NR 0 - R a — C(O)- R x ,

R 6 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, — R a — Y— R b — (Z) x ,

— C(O)R d and a saccharide group optionally substituted with _R a _γ_R b _( Z ) )<] _ R f _ — C(O)R f , or — C(O)- R a — Y— R b — (Z) x , or R 5 and R 6 can be joined, together with the atoms to which they are attached, to form a heterocyclic ring optionally substituted with — NR°— R a — Y— R b — (Z) x , R 7 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, — R a — Y— R b — (Z) x , and — C(O)R d ,

R 8 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic and — R a — Y— R b — (Z) x ,

R 9 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl and heterocyclic,

R 10 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl and heterocyclic, or R 8 and R 10 are joined to form — Ar 1 — O — Ar 2 — , where Ar 1 and Ar 2 are independently arylene or heteroarylene,

R 11 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl and heterocyclic, or R 10 and R 11 are joined, together with the carbon and nitrogen atoms to which they are attached, to form a heterocyclic ring,

R 12 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic, — C(0)R d , — C(NH)R d , -C(O)NR 0 R 0 , — C(O)OR d , -C(NH)NR 0 R 0 , _R a _γ_R f _( Z)χ] and -C(O)- R b — Y— R b — (Z) x , or R 11 and R 12 are j oined,

together with the nitrogen atom to which they are attached, to form a heterocyclic ring,

R 13 is hydrogen or — OR 14 ,

R 14 is hydrogen, — C(O)R d or a saccharide group, R a is each independently selected from the group consisting of alkylene, substituted alkylene, alkenylene, substituted alkenylene, alkynylene and substituted alkynylene,

R b is each independently selected from the group consisting of a covalent bond, alkylene, substituted alkylene, alkenylene, substituted alkenylene, alkynylene and substituted alkynylene,

R c is each independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic and — C(O)R d , R d is each independently selected from the group consisting of alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl and heterocyclic,

R e is each a saccharide group, R f is each independently selected from the group consisting of alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, and heterocyclic,

R x is an N-linked amino saccharide or an N-linked heterocycle, X is each independently selected from the group consisting of hydrogen, fluoro, chloro, bromo and iodo,

Y is each independently selected from the group consisting of , — CH 2 — , — O —

, -S-, -S-S-, — NR C — , -S(O)-, -SO 2 -, — NR 0 C(O)- , -OSO 2 -,

-OC(O)- — N(R C )SO 2 — , -C(O)NR 0 -, -C(O)O-, -SO 2 NR 0 -, -SO 2 O-, — P(O)(OR°)O— , — P(O)(OR C )NR C — , — OP(O)(OR°)O— , — OP(O)(OR C )NR C — ,

-OC(O)O-, -NR 0 C(O)O-, -NR 0 C(O)NR 0 -, -OC(O)NR 0 -, -C(O)- and -N(R°)SO 2 NR C — ,

Z is each independently selected from the group consisting of hydrogen, aryl, cycloalkyl, cycloalkenyl, heteroaryl, heterocyclic, and a saccharide, n is O, 1 , or 2, x is 1 or 2, and

12 The compound of claim 1 , wherein A is vancomycin or a derivative thereof

13 The compound of claim 1 , wherein A is teicoplanin or a derivative thereof

14 The compound of claim 1 , wherein A is oπtavancin or a derivative thereof

15 The compound of claim 1 , wherein A is dalbavancin or a derivative thereof

16 The compound of claim 1 , wherein A is telavancin or a derivative thereof

17 The compound of claim 1 , wherein A is selected from the group consisting of compound A35512 A, compound A35512 C, compound A35512 E, compound A35512 F, compound A35512 G, compound A35512 H, compound A40926 A, compound A40926 B, compound A40926 PB, parvodicin B2, parvodicin C1 , parvodicin C3, compound A41030, compound A42867, compound A477, compound A47934, compound A51568A, N-demethylvancomycin, compound A80407, compound A83850, compound A84575, compound AB65, compound AM374, actaplanin, compound A4696, actinoidin, ardacin, aricidin, compound AAD216, avoparcin, compound LL-

AV290, azureomycin, balhimycin, balhimycin V, chloroorienticin, compound A82846B, compound LY264826, chloroeremomycin, chloropeptin, chloropolyspoπn, complestatin, decaplanin, dechlorobalhimycin, dechlorobalhimycin V, chlorobalhimycin, chlorobromobalhimycin, fluorobalhimycin, deglucobalhimycin, N-demethylbalhimycin, N-demethylvancomycin, devancosamine-vancomycin, eremomycin, galacardin, helvecardin, izupeptin, kibdelin, kistamicin, mannopeptin, methylbalhimycin, compound MM47761 , compound MM47766, compound MM47767, compound MM49721 , compound MM49727, compound MM55256, compound MM55260, compound MM55266, compound MM55268, compound MM55270, compound MM55272, compound MM56597, compound MM56598, nogabecin F, compound OA7653, orienticin, dechloroeremomycin, compound PA42867, compound PA45052, chloroorienticin, parvodicin, rhamnosyl-balhimycin, ristocetin, πstomycin, spontin, symnonicin, teichomycin, Targocid, ureido-balhimycin and [T[CH 2 NH]Tpg 4 ]Vancomycιn

18. A compound selected from the group consisting of:

nd

19 A compound represented by the following Formula (II)

or a pharmaceutically acceptable salt, ester or prodrug thereof, wherein

R 1 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic, — R a — Y — R b — (Z) x and — L 1 , or R 1 is a saccharide group optionally substituted with — R a — γ_R b — (Z) x , — R f , -C(O)R,, — C(O)- R a — Y— R b — (Z) x , — C(NL 2 )R f , or — C(NL 3 )- R a — Y— R b — (Z) x ,

R 2 is hydrogen, — L 4 or a saccharide group optionally substituted with — R a — Y— R b — (Z) x , — R f , — C(O)R f , — C(O)- R a — Y— R b — (Z) x , — C(NL 5 )R f , or — C(NL 6 )- R a — Y— R b — (Z) x ,

R 3 is selected from the group consisting of — OR C , — NR 0 R 0 , _0— R a — Y— R b — (Z) x , — NR 0 - R a — Y— R b — (Z) x , — NR°R e , — O— R e , -OL 7 , -NL 8 R 0 , and — NL 9 R 6 ,

R 4 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, — L 10 ,

— R a — Y— R b — (Z) x , — C(O)R d , — C(NL 11 )R d and a saccharide group optionally substituted with — R a — Y— R b — (Z) x , — R f , — C(O)- R a — Y— R b — (Z) x , or — C(NL 12 )- R a — Y— R b — (Z) x , or R 4 and R 5 can be joined, together with the atoms to which they are attached, to form a heterocyclic ring optionally substituted with — NR 0 - R a — Y— R b — (Z) x or -NL 13 — R a — Y— R b — (Z) x , R 5 is selected from the group consisting of hydrogen, halo, — CH(R 0 ) — NR 0 R 0 , — CH(R 0 )- NR°R e , — CH(R 0 )- NR C — R a — Y— R b — (Z) x , — CH(R 0 )- R x , — CH(R 0 )- NR 0 - R a — C(O)- R x , — CH(R 0 )- NL 14 R 0 , — CH(R 0 )- NL 15 R e , — CH(R 0 )- NL 16 - R a — Y— R b — (Z) x , — CH(R 0 )- NL 17 - R a — C(O)- R x and — CH(R C )— NR C — R a — C(NL 18 )- R*

R 6 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, — L 19 , _R a _γ_R b — (Z) x , — C(O)R d , — C(NL 20 )R d and a saccharide group optionally substituted with — R a — Y— R b — (Z) x , — R f , — C(O)R f , — C(O)- R a — Y— R b — (Z) x , — C(NL 21 )R f , or — C(NL 22 )- R a — Y— R b — (Z) x , or R 5 and R 6 can be joined, together with the atoms to which they are attached, to form a heterocyclic ring optionally substituted with — NR C — R a — Y— R b — (Z) x or

-N L 23 — R a — Y— R b — (Z) x ,

R 7 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, — L 24 ,

— R a — Y— R b — (Z) x , — C(O)R d , and — C(NL 25 )R d ,

R 8 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic and — R a — Y— R b — (Z) x ,

R 9 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic, and — L 26 , R 10 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl and heterocyclic, or R 8 and R 10 are joined to form — Ar 1 — O — Ar 2 — , where Ar 1 and Ar 2 are independently arylene or heteroarylene which may optionally be substituted with -OL 27 ,

R 11 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic, and — L 28 , or R 10 and R 11 are joined, together with the carbon and nitrogen atoms to which they are attached, to form a heterocyclic ring which may optionally be substituted with -OL 29 , -CO 2 L 30 or -NL 31 R 0 , R 12 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic, — L 32 , — C(O)R d , — C(NH)R d , -C(O)NR 0 R 0 , — C(O)OR d , -C(NH)NR 0 R 0 ,

_ R a _γ_R b _(Z) x , and — C(O)- R b — Y— R b — (Z) x , — C(NL 33 )R d ,

-C(O)NL 34 R 0 , -C(O)OL 35 , -C(NH)NL 36 R 0 , — C(NL 37 )NR°R°, and — C(NL 38 )- R b — Y— R b — (Z) x , or R 11 and R 12 are joined, together with the nitrogen atom to which they are attached, to form a heterocyclic ring which may optionally be substituted with -OL 39 , -CO 2 L 40 or — NL 41 R 0 , R 13 is hydrogen or —OR 14 ,

R 14 is selected from the group consisting of hydrogen, — L 42 , — C(0)R d , — C(NL 43 )R d and a saccharide group optionally substituted with — R a — Y— R b — (Z) x , — R f , -C(O)R', — C(O)- R a — Y— R b — (Z) x , — C(NL 44 )R f , or — C(NL 45 )- R a — Y— R b — (Z) x , R a is each independently selected from the group consisting of alkylene, substituted alkylene, alkenylene, substituted alkenylene, alkynylene and substituted alkynylene,

R b is each independently selected from the group consisting of a covalent bond, alkylene, substituted alkylene, alkenylene, substituted alkenylene, alkynylene and substituted alkynylene,

R° is each independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic and — C(O)R d , R d is each independently selected from the group consisting of alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl and heterocyclic, R β is each a saccharide group optionally substituted with — R a — Y— R b — (Z) x , — R f , -C(O)R', — C(O)- R a — Y— R b — (Z) x , — C(NL 46 )R f , or

— C(NL 47 )- R a — Y— R b — (Z) x ,

R f is each independently selected from the group consisting of alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, and heterocyclic,

R x is an N-linked amino saccharide or an N-linked heterocycle, either of which may be optionally substituted with — R a — Y— R b — (Z) x , — R', -C(O)R 1 , — C(O)- R a — Y— R b — (Z) x , — C(NL )R,, or — C(NL 49 )- R a — Y— R b — (Z) x , X is each independently selected from the group consisting of hydrogen, fluoro, chloro, bromo and iodo,

Y is each independently selected from the group consisting of -CH 2 — , — O — , -S-, — S— S— , — NR C — , -S(O)-, -SO 2 -, -NR 0 C(O)-, -OSO 2 -,

-OC(O)-, — N(R°)SO 2 — , — C(O)NR C — , -C(O)O-, -SO 2 NR 0 -,

-SO 2 O-, — P(0)(0R°)0— , — P(O)(OR C )NR C — , — OP(O)(OR°)O— ,

— OP(O)(OR C )NR°— , -OC(O)O-, -NR 0 C(O)O-, -NR 0 C(O)NR 0 -,

-OC(O)NR 0 , -C(O)- — N(R°)SO 2 NR C — , -NL 50 -, -NL 51 C(O)-,

-OSO 2 -, -OC(O)-, — N(L 52 JSO 2 -, -C(O)NL 53 -, -SO 2 NL 54 -,

— P(O)(OL 55 )O— , — P(O)(OL 56 )NR°— , — P(O)(OR°)NL 57 — ,

— OP(O)(OL 58 )O— , — OP(O)(OL 59 )NR°— , — OP(0)(OR c )NL βo — ,

-NL 61 C(O)O-, — NL 62 C(O)N R 0 -, -NR 0 C(O)NL 63 -, -OC(O)NL 64 -,

-N(L 65 )SO 2 NR°— and — N(R°)SO 2 NL 66 — ;

Z is each independently selected from the group consisting of hydrogen, aryl, cycloalkyl, cycloalkenyl, heteroaryl, heterocyclic, a saccharide, — L 67 , — L 68 and

_ L 69. n is O, 1 , or 2; x is 1 or 2; and

each L 1 , L 4 , L 10 , L 19 , L 24 , L 27 , L 29 , L 39 , L 42 , and L 67 is a linker independently selected from the group consisting of

wherein:

B represents said phosphonated group; each p is independently 0 or an integer < 10; each R L is independently selected from the group consisting of H, ethyl and methyl; q is 2 or 3; n is an integer < 10; r is 1 , 2, 3, 4 or 5; and W 1 and w 2 are each integers ≥ 0 such that their sum (W 1 + w 2 ) is 1 , 2 or 3;

each L 8 , L 9 , L 13 , L 14 L 15 , L , L 17 , L 23 , L 26 , L 28 , L 31 , L 32 , L 34 , L 36 , L 37 , L 41 , L 50 , L 51 , L 52 , L 53 , L 54 , L 57 , L 60 , L 61 , L 62 , L 63 , L 64 , L 65 , L 66 and L 68 is a linker independently selected from the group consisting of

wherein

B represents said phosphonated group, n is an integer ≤ 10, each p is independently 0 or an integer < 10, each R L IS independently selected from the group consisting of H, ethyl and methyl, q is 2 or 3, r is 1 , 2, 3, 4 or 5, W 1 and W 2 are each integers ≥ 0 such that their sum (W 1 + w 2 ) is 1 , 2 or 3,

X IS -CH 2 , -CONR L -, -CO-O-CH 2 -, or — CO— O— , and

Ra is C x H y where x is an integer of 0 to 20 and y is an integer of 1 to 2x+1 ,

each L 7 , L 30 , L 35 , L 40 , L 55 , L 56 , L 58 , L 59 and L 69 is a linker independently selected from the group consisting of

wherein: n is an integer ≤ 10; p is 0 or an integer ≤ 10;

R L is H, ethyl or methyl;

R x is -S-, -C(RL) 2 -, — NRL- or —0—; each Z is independently selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, acyl, acyloxy, carboxy, carbamoyl, sulfuryl, sulfinyl, sulfenyl, sulfonyl, mercapto, amino, hydroxyl, cyano and nitro, wherein s is 1 ,

2, 3 or 4; and

B represents the phosphonated group;

each L 2 , L 3 , L 5 , L 6 , L 11 , L 12 , L 18 , L 20 , L 21 , L 22 , L 25 , L 33 , L 38 , L 43 , L 44 , L 45 , L 46 , L 47 , L 48 and L 49 is a linker independently

wherein p is 0 or an integer < 10, R L is H, ethyl or methyl, each Z is independently selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, acyl, acyloxy, carboxy, carbamoyl, sulfuryl, sulfinyl, sulfenyl, sulfonyl, mercapto, ammo, hydroxyl, cyano and nitro, wherein s is 1 , 2, 3 or 4, and

B represents the phosphonated group, with the proviso that at least one of L 1 L 2 L 3 L 4 L 5 L 6 L 7 L 8 L 9 L 10 L 11 L 12 L 13 L 14 L 15

■ 16 ■ 17 I 18 I 19 I 20 ■ 21 i 22 i 23 i 24 • 25 i 26 i 27 ■ 28 i 29 i 30 i 31 i 32 i 33 ■ 34 i 35 i 36 i 37 i 38

■ 39 I 40 I 41 I 42 i 43 i 44 i 45 i 46 i 47 i 48 i 49 i 50 ■ 51 |_52 i 53 i 54 i 55 |_56 i 57 i 58 |_59 i 60 i 61 L 62 L 63 L 64 L 65 ,_66 ,_67 ,_6 8 gnd ,_69 |g present

20 The compound of claim 19, wherein B is a phosphonated group selected from the group consisting of

wherein each R* is independently selected from the group consisting of H, lower alkyl, cycloalkyl, aryl and heteroaryl, with the proviso that at least two R* are H,

X is H, OH, NH 2 , or a halo group,

X I are both H, or each is independently selected from the group consisting of H, OH, NH 2 , and a halo group, and

L 1 is the point of attachment to L

21 A pharmaceutical composition comprising a compound of any one of claims 1 to 20, and a pharmaceutically acceptable carrier or excipient

22 A method for treating a bacterial infection in a subject, comprising administering to a subject in need of treatment a pharmaceutically effective amount of a compound of any one of claims 1 to 20

23 A method for treating a bacterial infection in a subject, comprising administering to a subject in need of treatment a pharmaceutically effective amount of a pharmaceutical composition of claim 21

24 A method for preventing a bacterial infection in a subject, comprising administering to a subject in need of prevention a pharmaceutically effective amount of a compound of any one of claims 1 to 20

25 A method for preventing a bacterial infection in a subject, comprising administering to a subject in need of prevention a pharmaceutically effective amount of a pharmaceutical composition of claim 21

26 A method of prophylaxis of a bacterial infection in a subject, comprising administering to a subject in need of such prophylaxis a prophylactically effective amount of a compound of any one of claims 1 to 20

27 A method of prophylaxis of a bacterial infection in a subject, comprising administering to a subject in need of such prophylaxis a prophylactically effective amount of a pharmaceutical composition of claim 21

28 The method of 22 wherein said subject is a human

29 The method of 23 wherein said subject is a human

30 The method of 24 wherein said subject is a human

31 The method of 25 wherein said subject is a human

32 The method of 26 wherein said subject is a human

33 The method of 27 wherein said subject is a human

34 The method of claim 22, further comprising administering a second antibiotic concurrent with administration of said compound

35 The method of claim 24, further comprising administering a second antibiotic concurrent with administration of said compound

36 The method of claim 26, further comprising administering a second antibiotic concurrent with administration of said compound

37 The method of claim 23, further comprising administering a second antibiotic concurrent with administration of said pharmaceutical composition

38 The method of claim 25, further comprising administering a second antibiotic concurrent with administration of said pharmaceutical composition

39 The method of claim 27, further comprising administering a second antibiotic concurrent with administration of said pharmaceutical composition

40 The method of claim 34, wherein said antibiotic is selected from the group consisting of tetracycline, a tetracycline derived antibacterial agent, glycylcycline, a glycylcycline derived antibacterial agent, minocycline, a minocycline derived antibacterial agent, an oxazolidmone antibacterial agent, an aminoglycoside antibacterial agent, a quinolone antibacterial agent, vancomycin, a vancomycin derived antibacterial agent, a teicoplanin, a teicoplanin derived antibacterial agent, eremomycin, an eremomycin derived antibacterial agent, chloroeremomycin, a chloroeremomycin derived antibacterial agent, daptomycin, a daptomycin derived antibacterial agent, Rifamycin, a Rifamycin derived antibacterial agent, Rifampin, a Rifampin derived antibacterial agent, Rifalazil, a Rifalazil derived antibacterial agent, Rifabutin, a Rifabutin derived antibacterial agent, Rifapentin, a Rifapentin derived antibacterial agent, Rifaximin and a Rifaximin derived antibacterial agent

41 The method of claim 35, wherein said antibiotic is selected from the group consisting of tetracycline, a tetracycline derived antibacterial agent, glycylcycline, a glycylcycline derived antibacterial agent, minocycline, a minocycline derived antibacterial agent, an oxazolidmone antibacterial agent, an aminoglycoside antibacterial agent, a quinolone antibacterial agent, vancomycin, a vancomycin derived antibacterial agent, a teicoplanin, a teicoplanin derived antibacterial agent,

eremomycin, an eremomycm derived antibacterial agent, chloroeremomycin, a chloroeremomycin derived antibacterial agent, daptomycin, a daptomycin derived antibacterial agent, Rifamycin, a Rifamycin derived antibacterial agent, Rifampin, a Rifampin derived antibacterial agent, Rifalazil, a Rifalazil derived antibacterial agent, Rifabutin, a Rifabutin derived antibacterial agent, Rifapentin, a Rifapentin derived antibacterial agent, Rifaximin and a Rifaximin derived antibacterial agent

42 The method of claim 36, wherein said antibiotic is selected from the group consisting of tetracycline, a tetracycline derived antibacterial agent, glycylcycline, a glycylcycline derived antibacterial agent, minocycline, a minocycline derived antibacterial agent, an oxazohdinone antibacterial agent, an aminoglycoside antibacterial agent, a quinolone antibacterial agent, vancomycin, a vancomycin derived antibacterial agent, a teicoplanin, a teicoplanin derived antibacterial agent, eremomycin, an eremomycin derived antibacterial agent, chloroeremomycin, a chloroeremomycin derived antibacterial agent, daptomycin, a daptomycin derived antibacterial agent, Rifamycin, a Rifamycin derived antibacterial agent, Rifampin, a Rifampin derived antibacterial agent, Rifalazil, a Rifalazil derived antibacterial agent, Rifabutin, a Rifabutin derived antibacterial agent, Rifapentin, a Rifapentin derived antibacterial agent, Rifaximin and a Rifaximin derived antibacterial agent

43 The method of claim 37, wherein said antibiotic is selected from the group consisting of tetracycline, a tetracycline derived antibacterial agent, glycylcycline, a glycylcycline derived antibacterial agent, minocycline, a minocycline derived antibacterial agent, an oxazohdinone antibacterial agent, an aminoglycoside antibacterial agent, a quinolone antibacterial agent, vancomycin, a vancomycin derived antibacterial agent, a teicoplanin, a teicoplanin derived antibacterial agent, eremomycin, an eremomycin derived antibacterial agent, chloroeremomycin, a chloroeremomycin derived antibacterial agent, daptomycin, a daptomycin derived antibacterial agent, Rifamycin, a Rifamycin derived antibacterial agent, Rifampin, a Rifampin derived antibacterial agent, Rifalazil, a Rifalazil derived antibacterial agent, Rifabutin, a Rifabutin derived antibacterial agent, Rifapentin, a Rifapentin derived antibacterial agent, Rifaximin and a Rifaximin derived antibacterial agent

44 The method of claim 38, wherein said antibiotic is selected from the group consisting of tetracycline, a tetracycline derived antibacterial agent, glycylcycline, a glycylcycline derived antibacterial agent, minocycline, a minocycline derived antibacterial agent, an oxazohdinone antibacterial agent, an aminoglycoside

antibacterial agent, a quinolone antibacterial agent, vancomycin, a vancomycin derived antibacterial agent, a teicoplanin, a teicoplanin derived antibacterial agent, eremomycin, an eremomycin derived antibacterial agent, chloroeremomycin, a chloroeremomycin derived antibacterial agent, daptomycin, a daptomycin derived antibacterial agent, Rifamycin, a Rifamycin derived antibacterial agent, Rifampin, a Rifampin derived antibacterial agent, Rifalazil, a Rifalazil derived antibacterial agent, Rifabutin, a Rifabutin derived antibacterial agent, Rifapentin, a Rifapentin derived antibacterial agent, Rifaximin and a Rifaximin derived antibacterial agent

45 The method of claim 39, wherein said antibiotic is selected from the group consisting of tetracycline, a tetracycline derived antibacterial agent, glycylcycline, a glycylcycline derived antibacterial agent, minocycline, a minocycline derived antibacterial agent, an oxazolidinone antibacterial agent, an aminoglycoside antibacterial agent, a quinolone antibacterial agent, vancomycin, a vancomycin derived antibacterial agent, a teicoplanin, a teicoplanin derived antibacterial agent, eremomycin, an eremomycin derived antibacterial agent, chloroeremomycin, a chloroeremomycin derived antibacterial agent, daptomycin, a daptomycin derived antibacterial agent, Rifamycin, a Rifamycin derived antibacterial agent, Rifampin, a Rifampin derived antibacterial agent, Rifalazil, a Rifalazil derived antibacterial agent, Rifabutin, a Rifabutin derived antibacterial agent, Rifapentin, a Rifapentin derived antibacterial agent, Rifaximin and a Rifaximin derived antibacterial agent

46 A method of accumulating of a glycopeptide or lipoglycopeptide antimicrobial molecule in a bone of a mammal, comprising administering a compound of any one of claims 1 to 20 to a mammal, whereby said compound binds osseous tissue and accumulates in a bone of said mammal

47 A method of accumulating of a glycopeptide or lipoglycopeptide antimicrobial molecule in a bone of a mammal, comprising administering a pharmaceutical composition of claim 21 to a mammal, whereby said compound binds osseous tissue and accumulates in a bone of said mammal

48 A method for prolonging the presence of a glycopeptide or lipoglycopeptide antimicrobial molecule in a bone of a mammal, comprising administering a compound of any one of claims 1 to 20 to a mammal, whereby said compound binds osseous tissue and accumulates in a bone of said mammal, and whereby cleavage of said

linker of said compound is gradual within the bone, thereby prolonging the presence of the glycopeptide or lipoglycopeptide antimicrobial molecule in said bone

49 A method for prolonging the presence of a glycopeptide or lipoglycopeptide antimicrobial molecule in a bone of a mammal, comprising administering a pharmaceutical composition of claim 21 to a mammal, whereby the compound of said pharmaceutical composition binds osseous tissue and accumulates in a bone of said mammal, and whereby cleavage of said linker of said compound is gradual within the bone, thereby prolonging the presence of the glycopeptide or lipoglycopeptide antimicrobial molecule in said bone

Description:

PHOSPHONATED GLYCOPEPTIDE AND LIPOGLYCOPEPTIDE ANTIBIOTICS AND USES THEREOF FOR THE PREVENTION AND TREATMENT OF BONE AND JOINT

INFECTIONS

BACKGROUND OF THE INVENTION a) Field of the invention

The invention relates to phosphonated derivatives of glycopeptide and lipoglycopeptide antibiotics These compounds are useful as antibiotics for prevention and/or the treatment of bone and joint infections, especially for the prophylaxis and/or treatment of osteomyelitis

b) Brief description of the prior art

Osteomyelitis is an inflammation of bone caused by a variety of microorganisms, mainly Staphylococcus aureus (Carek et al , American Family Physician (2001 ), Vo1 12, 12 2413-2420) This painful and debilitating disease occurs more commonly in children Within the adult population, diabetics and kidney dialysis patients are also vulnerable The acute form of the disease is treatable with antibiotics, but requires a lengthy period of daily therapy It can, however, revert to a recurrent or chronic form requiring repeated hospital stays and heavy treatment regimens Glycopeptide and lipoglycopeptide antibiotics are a class of biologically produced or semi-synthetic antimicrobial agents which affect the bacterial cell wall and/or membrane integrity (Williams, D H et al, Angewandte Chemie International Edition in English (1999), 1999, 38, 1172-1 193 Nicolaou, K C et al, Angewandte Chemie International Edition in English (1999), 38, 2097-2152 Kahne, D et al Chemical Reviews (2005), 105, 425 - 448, Pace, J L et al, Biochemical Pharmacology (2006), 71 , 968-980) Best known glycopeptide and lipoglycopeptide antibiotics are certainly vancomycin, teicoplanin, oritavancin (US Patent No 5,840,684), dalbavancin (US patent No 5,750,509) and telavancin (US patent No 6,635,618) The two first drugs were proven clinically and microbiologically to have potent activity against gram- positive organisms and the latter three drugs are in clinical trials Oritavancin, dalbavancin and telavancin possess extremely attractive pharmacological profiles with potent activity against gram-positive organisms, including methicillin-resistant Staphylococcus aureus, intermediate and fully vancomycin-resistant Staphylococcus aureus, vancomycin-resistant Enterococcus spp , and Streptococcus spp Although the use of highly active systemic anti-staphylococcal agents for the treatment of bone and joint infections is becoming attractive, results obtained in animal models using

vancomycin or teicoplanin for the treatment of osteomyelitis have not been convincing (Luu, Q N et al, European Journal of Clinical Microbiology and Infectious Diseases (1989), 8, 562-563 Mader, J T et al Antimicrobial Agents and Chemotherapy (1989), 33, 689-692) The problem may be that although the drug are highly active, the actual intra-bone concentration of the drug is but a fraction of the systemic dose and therefore glycopeptide and lipoglycopeptide antibiotics could prove to be more effective if the proportion of the drug reaching the bone could be increased

Bisphosphonates are well-characterized bone-seeking agents These compounds are known to have a high affinity to the bones due to their ability to bind the Ca 2+ ions found in the hydroxy apatite forming the bone tissues (Hirabayashi and Fujisaki, CIm Pharmacokinet (2003) 42(15) 1319-1330) Therefore, many different types of bisphosphonate-conjugated compounds have been made for targeting drugs selectively to the bone, including proteins (Uludag et al , Biotechnol Prog (2000) 16 1115-11 18), vitamins (US 6,214,812 and WO 02/083150), tyrosine kinase inhibitors (WO 01/44258 and WO 01/44259), hormones (US 5,183,815) and bone scanning agents (US 4,810,486) These and other bisphosphonate derivatives have been used as therapeutic agents for bone diseases such as arthritis (US 4,746,654), osteoporosis (US 5,428,181 and US 6,420,384), hypercalcemia (US 4,973,576), and bone cancers (US 6,548,042) Although some have suggested that bisphosphonate-antibiotics could also be made, only few of such compounds have actually being synthesized, including macrolides (US 5,359,060), fluoroquinolones and β-lactams (US 5,854,227, US 5,880, 11 1 , DE 195 32 235, Pieper and Keppler, Phosphorus, Sulfur and Silicon (2001 ) 170 5-14, and Herczegh et al J Med Chem (2002) 45 2338-41 ) Furthermore, prior to the present invention, no one has ever made or suggested to make phosphonated derivatives of glycopeptide or lipoglycopeptide antibiotics, nor suggested the use of such derivatives for the prevention or treatment of osteomyelitis

In view of the above, there is a need for highly active antibiotics for the prevention and treatment of bone and joint infections More particularly, there is a need for glycopeptide or lipoglycopeptide antibiotics with a higher affinity for bone, and a need for treatment methods wherein the intra-bone concentration of glycopeptide or lipoglycopeptide antibiotics is increased, for an extended period of time, above the minimal effective inhibitory concentrations which are required for killing bacteria The present invention fulfills these needs and also other needs as will be apparent to those skilled in the art upon reading the following specification

SUMMARY OF THE INVENTION

The present invention is directed to antimicrobial compounds which have an affinity for binding bones More particularly, the invention is directed to phosphonated derivatives of glycopeptide or lipoglycopeptide antibiotics These compounds are useful as antibiotics for the prevention or treatment of bone and joint infections, especially for the prevention and treatment of osteomyelitis

In one embodiment, the compounds of the invention are represented by the general Formula (I) as illustrated below

B- -L-

(I) as well as pharmaceutically acceptable salts, esters and prodrugs thereof, wherein

B is a phosphonated group, preferably having a high affinity to osseous tissues,

L is a bond or a linker for coupling B to A,

A is a glycopeptide or lipoglycopeptide antimicrobial molecule, and α is 1 , 2, 3, 4, 5, 6 or 7, preferably 1 , 2 or 3

In a prefered embodiment, B is a bisphosphonate More preferably, B is a bisphosphonate selected from the group consisting of

wherein each R * is independently selected from the group consisting of H, lower alkyl, cycloalkyl, aryl and heteroaryl, with the proviso that at least two R* are H,

X is H, OH, NH 2 , or a halo group,

X I are both H, or each is independently selected from the group consisting of H, OH, NH 2 , and a halo group, and

L 1 is the point of attachment to L

In another prefered embodiment, L is a cleavable linker for covaltently and/or reversibly coupling B to A In a further preferred embodiment, L is a hydrolysable linker In another preferred embodiment, the substructure represented by B — L — is represented by the following formula BL 1

wherein

A 3 indicates the point of attachment to the glycopeptide or lipoglycopeptide antimicrobial molecule A,

W is a covalent bond or is selected from the group of

T is oxygen or sulfur, each R is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic, amino, substituted ammo, hydroxyl, alkoxy, substituted alkoxy, aryloxy, substituted aryloxy, and — R a — Y— R b — Y— R b — B, each R a is independently selected from the group consisting of a covalent bond, alkylene, substituted alkylene, alkenylene, substituted alkenylene, alkynylene, substituted alkynylene, arylene, substituted arylene, —(CO)— alkylene— , substituted —(CO)— alkylene— , —(CO)— alkenylene— , substituted — (CO) — alkenylene — , — (CO) — alkynylene — , substituted —(CO)— alkynylene— , —(CO)— arylene— and substituted — (CO) — arylene — , each R b is independently selected from the group consisting of a covalent bond, alkylene, substituted alkylene, alkenylene, substituted alkenylene, alkynylene, substituted alkynylene, arylene and substituted arylene,

each Y is independently selected from the group consisting of a covalent bond, -CH 2 -, — O— , -S-, -S-S-, — NR 0 - , -S(O)-, -SO 2 -, -NR 0 C(O)- -OSO 2 -, — OC(O)-, — N(R C )SO 2 — , -C(O)NR 0 -, -C(O)O-, -SO 2 NR 0 -, -SO 2 O-, — P(O)(OR°)O— , — P(0)(0R°)NR c — , — OP(O)(OR°)O— , — 0P(0)(0R°)NR c — , -OC(O)O-, -NR 0 C(O)O-, -NR 0 C(O)NR 0 -, -OC(O)NR 0 -, -C(O)-, and -N(R C )SO 2 NR°— ; each R° is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic and — C(0)R d — ; each R d is independently selected from the group consisting of alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl and heterocyclic;

B is a phosphonated group; each Q is independently selected from the group consisting of nitro, chloro, bromo, iodo and fluoro; each X is independently selected from the group consisting of — O — , — S — , and -N(R)-;

Z is selected from the group consisting of hydrogen, acyl, substituted acyl, aroyl, substituted aroyl, alkoxycarbonyl, substituted alkoxycarbonyl, aryloxycarbonyl, substituted aryloxycarbonyl,

q is 2 or 3, r is 1 , 2, 3, 4 or 5,

W 1 and W 2 are each integers ≥ O such that their sum (Wi + w 2 ) is 1 , 2 or 3, a, b, c, d are integers ≥ O such that a+b+c+d ≤7 or null, e and f are integers > O such that e+f = 4, α is O or 1

In preferred embodiments of formula (I), L couples B to A through one or more hydroxyl groups on A, through one or more nitrogen atoms on A, through one or more carboxylic carbonyl groups on A, or through more than one of a combination of hydroxyl groups, nitrogen atoms and carboxylic carbonyl groups on A When L couples B to A through a hydroxyl group on A, preferably L is one or more of the following linkers

wherein: n is an integer ≤ 10, preferably 1 , 2, 3 or 4, more preferably 1 or 2; each p is independently O or an integer < 10, preferably O, 1 , 2, 3 or 4, more preferably O or 1 ; q is 2 or 3 r is 1 , 2, 3, 4 or 5 W 1 and W 2 are integers ≥ 0 such that their sum (W 1 + W 2 ) is 1 ,2 or 3 each R L is independently selected from the group consisting of H, ethyl and methyl, preferably H;

B represents the phosphonated group; and

the substructure A o of the linker represents the hydroxyl moiety of A. When L couples B to A through a nitrogen atom on A, preferably L is one or more of the following linkers:

wherein

B represents said phosphonated group, n is an integer < 10, each p is independently 0 or an integer ≤ 10, each R L IS independently selected from the group consisting of H, ethyl and methyl, q is 2 or 3, r is 1 , 2, 3, 4 or 5,

W 1 and W 2 are each integers ≥ 0 such that their sum (W 1 + W 2 ) is 1 , 2 or 3

X iS -CH 2 -, — CONRL-, -CO-O-CH 2 - Or -CO-O-, each Y is independently selected from the group consisting of -O-, -S- and each Z is indepedently selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, acyl, acyloxy, carboxy, carbamoyl, sulfuryl, sulfinyl, sulfenyl, sulfonyl, mercapto, ammo, hydroxyl, cyano and nitro, wherein s is 1 ,

2, 3, or 4, and

R a is C x Hy where x is an integer of 0 to 20 and y is an integer of 1 to 2x+1

B represents said phosphonated group, and

A a represents the nitrogen atom on A

When L couples B to A through the carbonyl of a carboxylate group on A, preferably L is one or more of the following linkers

wherein n is an integer ≤ 10, preferably 1 , 2, 3 or 4, more preferably 1 or 2, p is O or an integer < 10, preferably O, 1 , 2, 3 or 4, more preferably O or 1 ,

R L is H, ethyl or methyl, preferably H,

R x is -S-, — C(R L ) 2 — , — NR L — or — O— , preferably — NR L — , more preferably -NH-, each Y is independently selected from the group consisting of -O-, -S-, and

-NR L -, each Z is independently selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, acyl, acyloxy, carboxy, carbamoyl, sulfuryl, sulfinyl, sulfenyl, sulfonyl, mercapto, amino, hydroxyl, cyano and nitro, wherein s is 1 ,

2, 3 or 4,

B represents the phosphonated group, and

Ac the substructure Y ° ' of the linker represents the carbonyl of a

carboxy late group of A

In further prefered embodiment, at least one of B-L — is coupled to a hydroxyl functionality on the glycopeptide or lipoglycopeptide antimicrobial molecule A Preferably, when B — L— is coupled to a hydroxyl functionality B — L— is one or more of the following

wherein:

B represents said phosphonated group; each p is independently 0 or an integer ≤ 10, preferably 0, 1 , 2, 3 or 4, more preferably 0 or 1 ; each R L is independently selected from the group consisting of H, ethyl and methyl; q is 2 or 3; n is an integer < 10, preferably 1 , 2, 3 or 4, more preferably 1 or 2; r is 1 , 2, 3, 4 or 5; and

W 1 and W 2 are each integers ≥ 0 such that their sum (wi + w 2 ) is 1 , 2 or 3. In further prefered embodiment, at least one of B-L — is coupled to a nitrogen atom on the glycopeptide or lipoglycopeptide antimicrobial molecule A. Preferably, when B — L — is coupled to a nitrogen atom B-L — is one or more of the following:

wherein

B represents said phosphonated group, n is an integer ≤ 10, each p is independently 0 or an integer ≤ 10, each R L is independently selected from the group consisting of H, ethyl and methyl

q is 2 or 3; r is 1 , 2, 3, 4 or 5;

W 1 and W 2 are each integers ≥ 0 such that their sum (W 1 + w 2 ) is 1 , 2 or 3. X is -CH 2 -, — CONR L -, -CO-O-CH 2 -, or — CO— 0— ; and each Y is independently selected from the group consisting of -O-, -S- and each Z is indepedently selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, acyl, acyloxy, carboxy, carbamoyl, sulfuryl, sulfinyl, sulfenyl, sulfonyl, mercapto, amino, hydroxyl, cyano and nitro, wherein s is 1 , 2, 3 or 4; and

R a is C x H y where x is an integer of 0 to 20 and y is an integer of 1 to 2x+1. B represents said phosphonated group;

In further prefered embodiment of formula (I), at least one of B — L — is coupled to the carbonyl of a carboxylate group on the glycopeptide or lipoglycopeptide antimicrobial molecule A. Preferably, when B — L— is coupled to the carbonyl of a carboxylate group on A, B — L — is one or more of the following:

wherein: n is an integer < 10, preferably 1 , 2, 3 or 4, more preferably 1 or 2; p is 0 or an integer < 10, preferably 0, 1 , 2, 3 or 4, more preferably 0 or 1 ;

R L is H, ethyl or methyl, preferably H;

R x is — S — , — NR L — or — O — ; preferably — NR L — , more preferably — NH — ; each Y is independently selected from the group consisting of — O — , — S — , and —NR L —; each Z is independently selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, acyl, acyloxy, carboxy, carbamoyl, sulfuryl, sulfinyl, sulfenyl, sulfonyl, mercapto, amino, hydroxyl, cyano and nitro; wherein s is 1 , 2, 3 or 4; and

B represents the phosphonated group.

In an additional prefered embodiment of formula (I), α is an integer of 2 to 3, B — L — is coupled to a combination of at least two of a hydroxyl functionality on the glycopeptide or lipoglycopeptide antimicrobial molecule A, a nitrogen " atom on the glycopeptide or lipoglycopeptide antimicrobial molecule A or the carbonyl of a carboxylate group on the glycopeptide or lipoglycopeptide antimicrobial molecule A. Preferably, when B— L— is coupled to a hydroxyl functionality B — L— is one or more of the following:

wherein: B represents said phosphonated group; each p is independently 0 or an integer < 10, preferably 0, 1 , 2, 3 or 4, more preferably 0 or 1 ;

each R L is independently selected from the group consisting of H, ethyl and methyl; q is 2 or 3; n is an integer ≤ 10, preferably 1 , 2, 3 or 4, more preferably 1 or 2, r is 1 , 2, 3, 4 or 5; and

W 1 and W 2 are each integers ≥ 0 such that their sum (W 1 + w 2 ) is 1 , 2 or 3

Preferably, when B — L — is coupled to a nitrogen atom B — L — is one or more of the following.

B represents said phosphonated group, n is an integer ≤ 10, each p is independently 0 or an integer < 10, each R L is independently selected from the group consisting of H, ethyl and methyl, q is 2 or 3, r is 1 , 2, 3, 4 or 5,

W 1 and W 2 are each integers ≥ 0 such that their sum (W 1 + W 2 ) is 1 , 2 or 3

X is -CH 2 -, -CONRL-, -CO-O-CH 2 -, or — CO— 0— , and each Y is independently selected from the group consisting of — O — , — S — and — NR L — , each Z is indepedently selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, acyl, acyloxy, carboxy, carbamoyl, sulfuryl, sulfinyl, sulfenyl, sulfonyl, mercapto, amino, hydroxyl, cyano and nitro, wherein s is 1 ,

2, 3 or 4, and

Ra is C x Hy where x is an integer of 0 to 20 and y is an integer of 1 to 2x+1

Preferably, when B-L — is coupled to the carbonyl of a carboxylate group B— L— is one or more of the following

wherein: n is an integer < 10, preferably 1 , 2, 3 or 4, more preferably 1 or 2; p is O or an integer < 10, preferably 0, 1 , 2, 3 or 4, more preferably 0 or 1 ; R L is H, ethyl or methyl, preferably H;

R x is -S-, -C(R L ) 2 -, — NR L — or — O— ; preferably — NR L — , more preferably — NH — ; each Y is independently selected from the group consisting of — O — , — S- and -NRL-;

each Z is independently selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, acyl, acyloxy, carboxy, carbamoyl, sulfuryl, sulfinyl, sulfenyl, sulfonyl, mercapto, amino, hydroxyl, cyano and nitro; wherein s is 1 , 2, 3 or 4; and B represents the phosphonated group.

In a further preferred embodiment, α is 1 , 2 or 3.

Preferably, the glycopeptide or lipoglycopeptide antimicrobial molecule A has a structure represented by the following Formula (A 1 ):

as well as pharmaceutically acceptable salts, stereoisomers, esters and prodrugs thereof, where:

R 1 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic and — R a — Y — R b — (Z) x ; or R 1 is a saccharide group optionally substituted with — R a — Y— R b — (Z) x , — R f , -C(O)R,, or — C(O)- R a — Y— R b — (Z) x ; R 2 is hydrogen or a saccharide group optionally substituted with — R a — Y— R b — (Z) x , — R f , — C(O)R f , or — C(O)- R a — Y— R b — (Z) x ; R 3 is — OR C , — NR C R C , — O— R a — Y— R b — (Z) x , — NR C — R a — Y— R b — (Z) x , — NR c R e , or — O— R e ;

R 4 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, — R a — Y — R b — (Z) x , — C(O)R d and a saccharide group optionally substituted with — R a — Y— R b — (Z) x , — R f , or — C(O)- R a — Y— R b — (Z) x , or R 4 and R 5 can be

joined, together with the atoms to which they are attached, to form a heterocyclic ring optionally substituted with — NR C — R a — Y— R b — (Z) x , R 5 is selected from the group consisting of hydrogen, halo, — CH(R 0 )- NR C R C , — CH(R C )— NR c R e , — CH(R 0 )- NR 0 - R a — Y— R b — (Z) x , — CH(R 0 )- R x , and _CH(R°)— NR 0 - R a — C(O)- R",

R 6 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, — R a — Y— R b — (Z) x , — C(O)R d and a saccharide group optionally substituted with — R a — Y—R b — (Z) x , — R f , — C(O)R f , or — C(O)- R a — Y— R b — (Z) x , or R 5 and R 6 can be joined, together with the atoms to which they are attached, to form a heterocyclic ring optionally substituted with — NR°— R a — Y— R b — (Z) x , R 7 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, — R a — Y— R b — (Z) x , and — C(0)R d , R 8 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic and — R a — Y— R b — (Z) x , R 9 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl and heterocyclic,

R 10 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl and heterocyclic, or R 8 and R 10 are joined to form — Ar 1 — O — Ar 2 — , where Ar 1 and Ar 2 are independently arylene or heteroarylene,

R 11 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl and heterocyclic, or R 10 and R 11 are joined, together with the carbon and nitrogen atoms to which they are attached, to form a heterocyclic ring, R 12 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic,

— C(O)R d , — C(NH)R d , -C(O)NR 0 R 0 , — C(O)OR d , -C(NH)NR 0 R 0 ,

_R a _γ_R b _(Z) x , and — C(O)- R b — Y— R b — (Z) x , or R 11 and R 12 are joined, together with the nitrogen atom to which they are attached, to form a heterocyclic ring, R 13 is hydrogen or — OR 14 , R 14 is hydrogen, — C(O)R d or a saccharide group,

R a is each independently selected from the group consisting of alkylene, substituted alkylene, alkenylene, substituted alkenylene, alkynylene and substituted alkynylene, R b is each independently selected from the group consisting of a covalent bond, alkylene, substituted alkylene, alkenylene, substituted alkenylene, alkynylene and substituted alkynylene,

R c is each independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic and — C(O)R d ,

R d is each independently selected from the group consisting of alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl and heterocyclic, R e is each a saccharide group,

R f is each independently selected from the group consisting of alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, and heterocyclic, R" is an N-linked amino saccharide or an N-linked heterocycle,

X is each independently selected from the group consisting of hydrogen, fluoro, chloro, bromo and iodo,

Y is each independently selected from the group consisting of , — CH 2 — , — O — , -S-, — S— S— , — NR C — , -S(O)-, -SO 2 -, -NR 0 C(O)-, -OSO 2 -, — OC(O)-, — N(R C )SO 2 — , — C(O)NR C — , -C(O)O-, -SO 2 NR 0 -, -SO 2 O-,

— P(O)(OR°)O— , — P(O)(OR°)NR C — , — OP(O)(OR 0 JO-, — OP(O)(OR°)NR C — , -OC(O)O-, -NR 0 C(O)O-, -NR 0 C(O)NR 0 -, -OC(O)NR 0 -, -C(O)- and -N(R C )SO 2 NR C — , Z is each independently selected from the group consisting of hydrogen, aryl, cycloalkyl, cycloalkenyl, heteroaryl, heterocyclic, and a saccharide, n is O, 1 , or 2,

x is 1 or 2, and

More preferably, the glycopeptide or lipoglycopeptide antimicrobial molecule A is vancomycin, teicoplanin, oritavancin, dalbavancin, telavancin, compound A35512 A, compound A35512 C, compound A35512 E, compound A35512 F, compound A35512 G, compound A35512 H, compound A40926 A, compound A40926 B, compound A40926 PB, parvodicin B2, parvodicin C1 , parvodicin C3, compound A41030, compound A42867, compound A477, compound A47934, compound A51568A, N- demethylvancomycin, compound A80407, compound A83850, compound A84575, compound AB65, compound AM374, actaplanin, compound A4696, actinoidin, ardacin, aricidin, compound AAD216, avoparcin, compound LL-AV290, azureomyαn, balhimycin, balhimycin V, chlorooπenticin, compound A82846B, compound LY264826, chloroeremomycin, chloropeptin, chloropolysporin, complestatin, decaplanin, dechlorobalhimycin, dechlorobalhimycin V, chlorobalhimycin, chlorobromobalhimyαn, fluorobalhimycin, deglucobalhimycin, N-demethylbalhimycin, N-demethylvancomycin, devancosamine-vancomycin, eremomycin, galacardin, helvecardin, izupeptin, kibdelin, kistamicin, mannopeptin, methylbalhimycin, compound MM47761 , compound MM47766, compound MM47767, compound MM49721 , compound MM49727, compound MM55256, compound MM55260, compound MM55266, compound MM55268, compound MM55270, compound MM55272, compound MM56597, compound MM56598, nogabecin F, compound OA7653, oπenticin, dechloroeremomycin, compound PA42867, compound PA45052, chlorooπenticin, parvodicin, rhamnosyl-balhimycin, ristocetin, πstomycin, spontin, symnonicin, teichomycin, Targocid, ureido-balhimycin or [ψ[CH 2 NH]Tpg 4 ]Vancomycιn

In another embodiment, the compounds of the invention are represented by Formula (II)

s well as pharmaceutically acceptable salts, esters and prodrugs thereof, where

R 1 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic,

— R a — Y— R b — (Z) x and — L 1 , or R 1 is a saccharide group optionally substituted with — R a — Y— R b — (Z) x , — R f , -C(O)R f , — C(O)- R a — Y— R b — (Z) x , — C(NL 2 )R f , or — C(NL 3 )- R a — Y— R b — (Z) x , R 2 is hydrogen, — L 4 or a saccharide group optionally substituted with — R a — Y— R b — (Z) x , — R f , — C(0)R f , — C(O)- R a — Y— R b — (Z) x , — C(NL 5 )R f , or

— C(NL 6 )- R a — Y— R b — (Z) x ,

R 3 is selected from the group consisting of — OR C , — NR 0 R 0 , — O— R a — Y— R b — (Z) x , — NR 0 - R a — Y— R b — (Z) x , — NR°R e , — O— R e , -OL 7 , -NL 8 R 0 , and — NL 9 R e , R 4 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, — L 10 ,

— R a — Y— R b — (Z) x , — C(0)R d , — C(NL 11 )R d and a saccharide group optionally substituted with — R a — Y— R b — (Z) x , — R f , — C(O)- R a — Y— R b — (Z) x , or — C(NL 12 )- R a — Y— R b — (Z) x , or R 4 and R 5 can be joined, together with the atoms to which they are attached, to form a heterocyclic ring optionally substituted with — NR 0 - R a — Y— R b — (Z) x or -NL 13 — R a — Y— R b — (Z) x , R 5 is selected from the group consisting of hydrogen, halo, — CH(R 0 ) — NR 0 R 0 , — CH(R 0 )- NR°R e , — CH(R 0 )- NR 0 - R a — Y— R b — (Z) x , — CH(R 0 )- R x , — CH(R 0 )- NR°— R a — C(O)- R x , — CH(R 0 )- NL 14 R 0 , — CH(R 0 )- NL 15 R 6 , — CH(R 0 )- NL 16 - R a — Y— R b — (Z) x , — CH(R 0 )- NL 17 - R a — C(O)- R x and

— CH(R 0 )- NR 0 - R a — C(NL 18 )- R x

R 6 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, — L 19 , — R a — Y— R b — (Z) x , — C(O)R d , — C(NL 20 )R d and a saccharide group optionally substituted with — R a — Y— R b — (Z) x , — R f , — C(O)R f , — C(O)- R a — Y— R b — (Z) x , — C(NL 21 )R f , or — C(NL 22 )- R a — Y— R b — (Z) x , or R 5 and R 6 can be joined, together with the atoms to which they are attached, to form a heterocyclic ring optionally substituted with — NR C — R a — Y— R b — (Z) x or

-NL 23 -R a -Y-R b -(Z) X ,

R 7 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, — L 24 ,

— R a — Y— R b — (Z) x , — C(0)R d , and — C(NL 25 )R d ,

R 8 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic and — R a — Y— R b — (Z) x ,

R 9 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic, and — L 26 , R 10 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl and heterocyclic, or R 8 and R 10 are joined to form — Ar 1 — O — Ar 2 — , where Ar 1 and Ar 2 are independently arylene or heteroarylene which may optionally be substituted with -OL 27 ,

R 11 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic, and — L 28 , or R 10 and R 11 are joined, together with the carbon and nitrogen atoms to which they are attached, to form a heterocyclic ring which may optionally be substituted with -OL 29 , -CO 2 L 30 or -NL 31 R 0 , R 12 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic, — L 32 , — C(0)R d , — C(NH)R d , -C(O)NR 0 R 0 , — C(O)OR d , -C(NH)NR 0 R 0 ,

— R a — Y— R b — (Z) x , and — C(O)- R b — Y— R b — (Z) x , — C(NL 33 )R d ,

-C(O)NL 34 R 0 , -C(O)OL 35 , -C(NH)NL 36 R 0 , — C(NL 37 )NR°R°, and — C(NL 38 )- R b — Y— R b — (Z) x , or R 11 and R 12 are joined, together with the nitrogen atom to which they are attached, to form a heterocyclic ring which may optionally be substituted with -OL 39 , -CO 2 L 40 or — NL 41 R 0 , R 13 is hydrogen or —OR 14 ,

R 14 is selected from the group consisting of hydrogen, — L 42 , — C(O)R d , — C(NL 43 )R d and a saccharide group optionally substituted with _R a _γ_R b _ (Z) x , — R f , — C(O)R', — C(O)- R a — Y— R b — (Z) x , — C(NL 44 )R f , or — C(NL 45 )- R a — Y— R b — (Z) x , R a is each independently selected from the group consisting of alkylene, substituted alkylene, alkenylene, substituted alkenylene, alkynylene and substituted alkynylene,

R b is each independently selected from the group consisting of a covalent bond, alkylene, substituted alkylene, alkenylene, substituted alkenylene, alkynylene and substituted alkynylene,

R c is each independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic and — C(0)R d , R d is each independently selected from the group consisting of alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl and heterocyclic, R e is each a saccharide group optionally substituted with — R a — Y — R b — (Z) x , — R f , — C(O)R f , — C(O)- R a — Y— R b — (Z) x , — C(NL )R f , or

— C(NL 47 )- R a — Y— R b — (Z) x ,

R f is each independently selected from the group consisting of alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, and heterocyclic,

R x is an N-linked amino saccharide or an N-linked heterocycle, either of which may be optionally substituted with — R a — Y— R b — (Z) x , — R f , — C(O)R f , — C(O)- R a — Y— R b — (Z) x , — C(NL 48 )R f , or — C(NL 49 )- R a — Y— R b — (Z) x , X is each independently selected from the group consisting of hydrogen, fluoro, chloro, bromo and iodo,

Y is each independently selected from the group consisting Of -CH 2 -, —0—, -S-, — S— S— , — NR C — , -S(O)-, -SO 2 -, — NR C C(O)— , -OSO 2 -,

— OC(O)-, — N(R C )SO 2 — , -C(O)NR 0 -, -C(O)O-, -SO 2 NR 0 -, -SO 2 O-, -P(O)(OR 0 JO-, — P(O)(OR°)NR°— , — OP(O)(OR°)O— , — OP(O)(OR C )NR C — , -OC(O)O-, -NR 0 C(O)O-, -NR 0 C(O)NR 0 -, -OC(O)NR 0 , -C(O)- — N(R°)SO 2 NR°— , -NL 50 -, -NL 51 C(O)-, -OSO 2 -, — OC(O)-, — N(L 52 )SO 2 — , -C(O)NL 53 -, -SO 2 NL 54 -, — P(O)(OL 55 )O— , — P(O)(OL 56 )NR°— , — P(O)(OR°)NL 57 — , — OP(O)(OL 58 )O— , — OP(O)(OL 59 )NR°— , — OP(O)(OR C )NL 60 — , -NL 61 C(O)O-, — NL 62 C(O)N R 0 -, -NR 0 C(O)NL 63 -, -OC(O)NL 64 -, -N(L 65 )SO 2 NR°— and — N(R°)SO 2 NL 66 — ;

Z is each independently selected from the group consisting of hydrogen, aryl, cycloalkyl, cycloalkenyl, heteroaryl, heterocyclic, a saccharide, — L 67 , — L 68 and

_ L 69. n is O, 1 , or 2; x is 1 or 2; and

each L 1 , L 4 , L 10 , L 19 , L 24 , L 27 , L 29 , L 39 , L 42 , and L 67 is a linker independently selected from the group consisting of

wherein:

B represents said phosphonated group; each p is independently O or an integer ≤ 10; each R L is independently selected from the group consisting of H, ethyl and methyl; q is 2 or 3; n is an integer < 10; r is 1 , 2, 3, 4 or 5; and W 1 and w 2 are each integers ≥ O such that their sum (W 1 + w 2 ) is 1 , 2 or 3;

each L 8 , L 9 , L 13 , L 14 L 15 , L 16 , L 17 , L 23 , L 26 , L 28 , L 31 , L 32 , L 34 , L 36 , L 37 , L 41 , L 50 , L 51 , L 52 L 53 L s4 L 57 L 6o ,_6i ,_62 ,_63 L β 4 L β 5 ,_66 and L 6 β js g |jnker independently selected from the group consisting of

wherein:

B represents said phosphonated group; n is an integer < 10; each p is independently 0 or an integer ≤ 10; each R L is independently selected from the group consisting of H, ethyl and methyl; q is 2 or 3; r is 1 , 2, 3, 4 or 5;

Wi and W 2 are each integers ≥ 0 such that their sum (W 1 + w 2 ) is 1 , 2 or 3;

X is -CH 2 -, —C0NR L —, -CO-O-CH 2 -, or — CO— O— ; and

R a is C x H y where x is an integer of 0 to 20 and y is an integer of 1 to 2x+1 ;

each L 7 , L 30 , L 35 , L 40 , L 55 , L 56 , L 58 , L 59 and L 69 is a linker independently selected from the group consisting of

wherein n is an integer ≤ 10, preferably 1 , 2, 3 or 4, more preferably 1 or 2, p is O or an integer < 10, preferably O, 1 , 2, 3 or 4, more preferably O or 1 ,

R L IS H, ethyl or methyl, preferably H,

R x is — S — , — C(R L ) 2 — , — NR L — or — O — , preferably — NR L — , more preferably — NH — , each Z is independently selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, acyl, acyloxy, carboxy, carbamoyl, sulfuryl, sulfinyl, sulfenyl, sulfonyl, mercapto, amino, hydroxyl, cyano and nitro, wherein s is 1 ,

2, 3 or 4, and

B represents the phosphonated group,

each L 2 , L 3 , L 5 , L 6 , L 11 , L 12 , L 18 , L 20 , L 21 , L 22 , L 25 , L 33 , L 38 , L 43 , L 44 , L 45 , L 46 , L 47 , L 48 and L 49 is a linker independently

wherein p is 0 or an integer < 10, preferably 0, 1 , 2, 3 or 4, more preferably 0 or 1 , R L is H, ethyl or methyl, preferably H, each Z is independently selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, acyl, acyloxy, carboxy, carbamoyl, sulfuryl, sulfinyl, sulfenyl, sulfonyl, mercapto, amino, hydroxyl, cyano and nitro, wherein s is 1 ,

2, 3 or 4, and

B represents the phosphonated group, with the proviso that at least one of L 1 L 2 L 3 L 4 L 5 L 6 L 7 L 8 L 9 L 10 L 11 L 12 L 13 L 14 L 15 L 16 L 17 L 18 L 19 L 20 L 21 L 22 L 23 L 24 L 25 L 26 L 27 L 28 L 29 L 30 L 31 L 32 L 33 L 34 L 35 L 36 L 37 L 38

■ 39 I 40 I 41 1 42 ■ 43 ■ 44 i 45 |_46 ■ 47 i 48 |_49 ■ 50 i 51 |_52 j_53 |_54 |_55 |_56 j ^ 57 |_58 ^9 |_60 |_61

L 62 L 63 L b4 L 65 C L b7 L 6B and l_ bS ιs present

In a preferred embodiment of formula (II), B is a phosphonated group selected from the group consisting of

wherein each R* is independently selected from the group consisting of H, lower alkyl, cycloalkyl, aryl and heteroaryl, with the proviso that at least two R* are H,

X is H, OH, NH 2 , or a halo group,

Xi are both H, or each is independently selected from the group consisting of H,

OH, NH 2 , and a halo group, and

Li is the point of attachment to L

In further preferred embodiments, the compounds of the invention have a structure selected among the structures illustrated below, as well as pharmaceutically acceptable salts, esters, stereoisomers, and prodrugs thereof

and

5 In another aspect of the present invention there are disclosed pharmaceutical compositions comprising one or more of the compounds as defined herein and a pharmaceutically acceptable carrier or excipient.

The present invention encompasses methods for treating a bacterial infection in a subject, comprising administering to a subject having a bacterial infection or otherwise in need of such treatment a pharmaceutically effective amount of one or more of the compounds as defined herein, or a pharmaceutical composition as defined herein The subject may be an animal, preferably a mammal, more preferably a human

The present invention also encompasses methods for preventing a bacterial infection in a subject, comprising administering to a subject at risk for developing a bacterial infection or otherwise in need of such prevention a pharmaceutically effective amount of one or more of the compounds as defined herein, or a pharmaceutical composition as defined herein The subject may be an animal, preferably a mammal, more preferably a human

The present invention further encompasses methods of prophylaxis for a bacterial infection in a subject, comprising administering to a subject in need of such prophylaxis a prophylactically effective amount of one or more of the compounds as defined herein, or a pharmaceutical composition as defined herein The prophylactically effective amount of the compounds or pharmaceutical composition may be administered, for example, to a subject prior to, during, or after an invasive medical treatment The subject may be an animal, preferably a mammal, more preferably a human

The present invention encompasses methods for treating a bacterial infection in a subject, comprising administering to a subject having a bacterial infection or otherwise in need of such treatment a pharmaceutically effective amount of one or more of the compounds as defined herein, or a pharmaceutical composition as defined herein, and concurrently administering a second therapeutic agent The subject may be an animal, preferably a mammal, more preferably a human

The present invention also encompasses methods for preventing a bacterial infection in a subject, comprising administering to a subject at risk for developing a bacterial infection or otherwise in need of such prevention a pharmaceutically effective amount of one or more of the compounds as defined herein, or a pharmaceutical composition as defined herein, and concurrently administering a second therapeutic agent The subject may be an animal, preferably a mammal, more preferably a human

The present invention further encompasses methods of prophylaxis for a bacterial infection in a subject, comprising administering to a subject in need of such prophylaxis a prophylactically effective amount of one or more of the compounds as defined herein, or a pharmaceutical composition as defined herein, and concurrently administering a second therapeutic agent The prophylactically effective amount of the

compounds or pharmaceutical composition, and the second therapeutic agent, may be administered, for example, to a subject prior to, during, or after an invasive medical treatment The subject may be an animal, preferably a mammal, more preferably a human Preferably the second therapeutic agent is an antibiotic More preferably the second therapeutic agent is an antibiotic selected from the group consisting of tetracycline, a tetracycline derived antibacterial agent, glycylcycline, a glycylcycline derived antibacterial agent, minocycline, a minocycline derived antibacterial agent, an oxazohdinone antibacterial agent, an aminoglycoside antibacterial agent, a quinolone antibacterial agent, vancomycin, a vancomycin derived antibacterial agent, a teicoplanin, a teicoplanin derived antibacterial agent, eremomycin, an eremomycin derived antibacterial agent, chloroeremomycin, a chloroeremomycin derived antibacterial agent, daptomycin, a daptomycin derived antibacterial agent, Rifamycin, a Rifamycin derived antibacterial agent, Rifampin, a Rifampin derived antibacterial agent, Rifalazil, a Rifalazil derived antibacterial agent, Rifabutin, a Rifabutin derived antibacterial agent, Rifapentin, a Rifapentin derived antibacterial agent, Rifaximin and a Rifaximin derived antibacterial agent

The invention also provides a method of accumulating a glycopeptide or lipoglycopeptide antimicrobial molecule in a bone of a subject, comprising administering to a subject one or more of the compounds as defined herein, or a pharmaceutical composition as defined herein, whereby the compound or pharmaceutical composition binds osseous tissue and accumulates in the bone of the subject The subject may be an animal, preferably a mammal, more preferably a human The invention further provides a method for prolonging the presence of a glycopeptide or lipoglycopeptide antimicrobial molecule in a bone of a subject, comprising administering to a subject one or more of the compounds as defined herein, or a pharmaceutical composition as defined herein, whereby the compound or pharmaceutical composition binds osseous tissue and accumulates in the bone of the subject, and whereby cleavage of the linker of the compounds is gradual within the bone, thereby prolonging the presence of the glycopeptide or lipoglycopeptide antimicrobial molecule in the bone The subject may be an animal, preferably a mammal, more preferably a human

In a further aspect of the present invention there are provided processes for the preparation of phosphonated glycopeptide and lipoglycopeptide antimicrobial molecule, preferably glycopeptide and lipoglycopeptide antimicrobial molecule of Formula (I) and/or Formula (II) as defined herein

An advantage of the invention is that it provides antimicrobial compounds having an increased binding affinity for bone The invention also provides methods for the unmet medical need of prevention and treatment of bone and joint infections

Additional objects, advantages and features of the present invention will become more apparent upon reading of the following non-restrictive description of preferred embodiments with reference to the accompanying drawings which are exemplary and should not be interpreted as limiting the scope of the present invention

DETAILED DESCRIPTION OF THE INVENTION A) General overview of the invention

The present invention discloses phosphonated derivatives of glycopeptide and lipoglycopeptide antibiotics of structural Formula I and Formula Il as defined above These compounds are useful antimicrobial agents effective against a number of human and veterinary pathogens

The essence of the invention lies in the presence of a phosphonated group attached to a glycopeptide and lipoglycopeptide antibiotic Since phosphonic acid derivatives are known to have a high affinity to bone due to their ability to bind the Ca 2+ ions found in the hydroxyapatite forming bone tissues, the present inventors have hypothesized that it would be possible to increase the binding affinity, adsorption and retention of glycopeptide and lipoglycopeptide antibiotics by the bones by tethering a phosphonated group to such an antibiotic Achieving high concentrations of glycopeptide and lipoglycopeptide antibiotics in vascularized bone (in comparison with the concentrations achieved by administration of a non-phosphonated antibiotic), could prove to increase the concentration of the antibiotic in contiguous devascularized bones (sequestrum) to a level sufficient to eradicate microbes present in this locus of treatment resistance

Actually, the present inventors have synthesized such phosphonated derivatives of glycopeptide and lipoglycopeptide antibiotics and demonstrated that these derivatives have an increased affinity for bony materials The present inventors have also shown that these phosphonated derivatives accumulate in bones of mammals in amounts greater than amounts of a non-phosphonated equivalent of glycopeptide and lipoglycopeptide antimicrobials and that it is possible to prolong the presence of glycopeptide and lipoglycopeptide antimicrobials in the bones by administering such phosphonated derivatives Accordingly, the compounds of the

invention are particularly useful for the prevention and/or the treatment of bone-related infections and bone-related diseases such as osteomyelitis

The present invention discloses phosphonated glycopeptide and lipoglycopeptide antimicrobial molecules, in particular, those phosphonated compounds defined in Formula (I) and Formula (II) as defined above and hereinafter These compounds are useful antimicrobial agents effective against a number of human and veterinary pathogens A phosphonated group is reversibly coupled to a glycopeptide and lipoglycopeptide antimicrobial molecule via a cleavable linker

Phosphonated glycopeptide and lipoglycopeptide antimicrobial molecules have been synthesized and demonstrated to have an increased affinity for osseous materials In vivo, these phosphonated compounds accumulate in bones in amounts greater than amounts of non-phosphonated equivalents The presence of glycopeptide and lipoglycopeptide antimicrobial molecules in the bones can be prolonged by administering phosphonated derivatives of glycopeptide and lipoglycopeptide antimicrobial molecules according to the invention Accordingly, the compounds of the invention are particularly useful for the prevention, prophylaxis and/or treatment of bone and joint-related infections and bone-related diseases such as osteomyelitis

B) Definitions In order to provide an even clearer and more consistent understanding of the invention, including the scope given herein to particular terms, the following general definitions are provided

The term "alkyl" refers to saturated aliphatic groups including straight-chain, branched-chain, cyclic groups, and combinations thereof, having the number of carbon atoms specified, or if no number is specified, having 1 to 12 carbon atoms (preferably 1 to 6) Examples of alkyl groups include, but are not limited to groups such as methyl, ethyl, n-propyl, isopropyl, n-butyl, isobutyl, sec-butyl, t-butyl, n-pentyl, neopentyl, cyclopropyl, cyclobutyl, cyclopentyl, cyclohexyl, cyclobutylmethyl, cyclobutylethyl, cyclopentylmethyl, cyclopentylethyl, and adamantyl The term "cycloalkyl" refers to cyclic alkyl groups (e g cycloalkyl or heterocycloalkyl) consisting of one ring, including, but not limited to, groups such as cycloheptyl, or multiple fused rings, including, but not limited to, groups such as adamantyl or norbornyl

The term "alkylaryl" refers to an alkyl group having the number of carbon atoms designated, appended to one, two, or three aryl groups

The term "N-alkylaminocarbonyl" refers to the radical -C(O)NHR where R is an alkyl group

The term "N,N-dialkylaminocarbonyl " refers to the radical -C(O)NR 3 R b where R 3 and R b are each independently an alkyl group

The term "alkylthio" refers to the radical -SR where R is an alkyl group The term "alkoxy" as used herein refers to an alkyl, alkenyl, or alkynyl linked to an oxygen atom and having the number of carbon atoms specified, or if no number is specified, having 1 to 12 carbon atoms (preferably 1 to 6) Examples of alkoxy groups include, but are not limited to, groups such as methoxy, ethoxy, tert-butoxy, and allyloxy The term "alkoxycarbonyl" refers to the radical -C(O)OR where R is an alkyl The term "alkylsulfonyl" refers to the radical -SO 2 R where R is an alkyl group The term "alkylene" means a saturated divalent aliphatic group including straight-chain, branched-chain, cyclic groups, and combinations thereof, having the number of carbon atoms specified, or if no number is specified, having 1 to 12 carbon atoms (preferably 1 to 6) Examples of alkylene groups include, but are not limited to groups such as methylene, 2,2-dιmethyletylene, ethylene, n-propylene, isopropylene, n-butylene, isobutylene, sec-butylene, t-butylene, n-pentylene, neopentylene, cyclopropylene, cyclobutylene, cyclopentylene, cyclohexylene, cyclobutylmethylene, cyclobutylethylene, cyclopentylmethylene, cyclopentylethylene, and adamantylene

The term "cycloalkylene" refers to cyclic alkylene groups (e g cycloalkylene or heterocycloalkylene) consisting of one ring, including, but not limited to, groups such as cycloheptylene, or multiple fused rings, including, but not limited to, groups such as adamantylene or norbornylene

The term "substituted alkyl" means an alkyl group as defined above that is substituted with one or more substituents, preferably one to three substituents selected from the group consisting of halogen, alkyl, aryl, alkoxy, acyloxy, amino, mono or dialkylamino, hydroxyl, mercapto, carboxy, benzyloxy, phenyl, benzyl, cyano, nitro, thioalkoxy, carboxaldehyde, carboalkoxy and carboxamide, or a functionality that can be suitably blocked, if necessary for purposes of the invention, with a protecting group The phenyl group may optionally be substituted with one to three substituents selected from the group consisting of halogen , alkyl, aryl, alkoxy, acyloxy, amino, mono or dialkylamino, hydroxyl, mercapto, carboxy, benzyloxy, benzyl, cyano, nitro, thioalkoxy, carboxaldehyde, carboalkoxy and carboxamide Examples of substituted alkyl groups include, but are not limited to — CF 3 , — CF 2 -CF 3 , hydroxymethyl, 1- or 2-hydroxyethyl, methoxymethyl, 1- or 2-ethoxyethyl, carboxymethyl, 1- or 2-carboxyethyl, methoxycarbonylmethyl, 1- or 2-methoxycarbonyl ethyl, benzyl, pyrdinylmethyl, thiophenylmethyl, imidazolinylmethyl, dimethylaminoethyl and the like

The term "substituted alkylene" means an alkylene group as defined above that is substituted with one or more substituents, preferably one to three substituents, selected from the group consisting of halogen, alkyl, aryl, alkoxy, acyloxy, amino, mono or dialkylamino, hydroxyl, mercapto, carboxy, benzyloxy, phenyl, benzyl, cyano, nitro, thioalkoxy, carboxaldehyde, carboalkoxy and carboxamide, or a functionality that can be suitably blocked, if necessary for purposes of the invention, with a protecting group The phenyl group may optionally be substituted with one to three substituents selected from the group consisting of halogen, alkyl, aryl, alkoxy, acyloxy, amino, mono or dialkylamino, hydroxyl, mercapto, carboxy, benzyloxy, benzyl, cyano, nitro, thioalkoxy, carboxaldehyde, carboalkoxy and carboxamide Examples of substituted alkyl groups include, but are not limited to -CF 2 — , -CF 2 -CF 2 -, hydroxymethylene, 1- or 2- hydroxyethylene, methoxymethylene, 1- or 2-ethoxyethylene, carboxymethylene, 1- or 2-carboxyethylene, and the like

The term "alkenyl" refers to unsaturated aliphatic groups including straight- chain, branched-chain, cyclic groups, and combinations thereof, having the number of carbon atoms specified, or if no number is specified, having 1 to 12 carbon atoms (preferably 1 to 6), which contain at least one double bond ( — C=C-) Examples of alkenyl groups include, but are not limited to allyl vinyl, -CH 2 — CH=CH — CH 3 , — CH 2 - CH 2 -cyclopentenyl and — CH 2 - CH 2 -cyclohexenyl where the ethyl group can be attached to the cyclopentenyl, cyclohexenyl moiety at any available carbon valence The term "alkenylene" refers to unsaturated divalent aliphatic groups including straight-chain, branched-chain, cyclic groups, and combinations thereof, having the number of carbon atoms specified, or if no number is specified, having 1 to 12 carbon atoms (preferably 1 to 6), which contain at least one double bond (-C=C-) Examples of alkenylene groups include, but are not limited to -CH=CH — , -CH 2 -CH=CH-CH 2 -, — CH 2 - CH(cyclopentenyl)— and the like

The term "alkynyl" refers to unsaturated aliphatic groups including straight- chain, branched-chain, cyclic groups, and combinations thereof, having the number of carbon atoms specified, or if no number is specified, having 1 to 12 carbon atoms (preferably 1 to 6), which contain at least one triple bond ( — C ≡C— ) Examples of alkynyl groups include, but are not limited to acetylene, 2-butynyl, and the like

The term "alkynylene" refers to unsaturated divalent aliphatic groups including straight-chain, branched-chain, cyclic groups, and combinations thereof, having the number of carbon atoms specified, or if no number is specified, having 1 to 12 carbon atoms (preferably 1 to 6), which contain at least one triple bond (— C≡C— ) Examples

of alkynylene groups include, but are not limited to — C≡C— , -C=C-CH 2 -, and the like

The term "substituted alkenyl" or "substituted alkynyl" refers to the alkenyl and alkynyl groups as defined above that are substituted with one or more substituents selected from the group consisting of halogen, alkyl, aryl, alkoxy, acyloxy, amino, hydroxyl, mercapto, carboxy, benzyloxy, phenyl, benzyl, cyano, nitro, thioalkoxy, carboxaldehyde, carboalkoxy and carboxamide, or a functionality that can be suitably blocked, if necessary for purposes of the invention, with a protecting group Examples of substituted alkenyl and alkynyl groups include, but are not limited to — CH=CF 2 , methoxyethenyl, methoxypropenyl, bromopropynyl, and the like

The term "substituted alkenylene" or "substituted alkynylene" refers to the alkenylene and alkynylene groups as defined above that are substituted with one or more substituents selected from the group consisting of halogen, alkyl, aryl, alkoxy, acyloxy, amino, hydroxyl, mercapto, carboxy, benzyloxy, phenyl, benzyl, cyano, nitro, thioalkoxy, carboxaldehyde, carboalkoxy and carboxamide, or a functionality that can be suitably blocked, if necessary for purposes of the invention, with a protecting group

The term "aryl" or "Ar" refers to an aromatic carbocyclic group of 6 to 14 carbon atoms having a single ring (including but not limited to groups such as phenyl) or multiple condensed rings (including but not limited to groups such as naphthyl or anthryl), and includes both unsubstituted and substituted aryl groups Substituted aryl is an aryl group that is substituted with one or more substituents, preferably one to three substituents, selected from the group consisting of alkyl, aryl, alkenyl, alkynyl, halogen, alkoxy, acyloxy, amino, mono or dialkylamino, hydroxyl, mercapto, carboxy, benzyloxy, phenyl, aryloxy, benzyl, cyano, nitro, thioalkoxy, carboxaldehyde, carboalkoxy and carboxamide, or a functionality that can be suitably blocked, if necessary for purposes of the invention, with a protecting group Representative examples include, but are not limited to naphthyl, phenyl, chlorophenyl, iodophenyl, methoxyphenyl, carboxyphenyl, and the like The term "aryloxy" refers to an aryl group linked to an oxygen atom at one of the ring carbons Examples of alkoxy groups include, but are not limited to, groups such as phenoxy, 2-, 3-, or 4-methylphenoxy, and the like The term "arylthio group" refers to the radical — SR C where R 0 is an aryl group The term "heteroarylthio group" refers to the radical — SR d where R d is a heteroaryl

The term "arylene" refers to the diradical derived from aryl (including substituted aryl) as defined above and is exemplified by 1 ,2-phenylene, 1 ,3-phenylene, 1 ,4-phenylene, 1 ,2-naphthylene and the like

The term "amino" refers to the group — NH 2

The term "N-alkylamino" and "N,N-dialkylamino" means a radical -NHR and -NRR' respectively where R and R' independently represent an alkyl group as defined herein Representative examples include, but are not limited to N,N-dιmethylamιno, N- ethyl-N-methylamino, N,N-dι(1-methylethyl)amιno, N-cyclohexyl-N-methylamino, N- cyclohexyl-N-ethylamιno, N-cyclohexyl-N-propylamino, N-cyclohexylmethyl-N- methylammo, N-cyclohexylmethyl-N-ethylamino, and the like

The term "thioalkoxy" means a radical -SR where R is an alkyl as defined above e g , methylthio, ethylthio, propylthio, butylthio, and the like The term "acyl group" means a radical -C(O)R, where R is hydrogen, halogen, alkyl, aryl, heteroaryl, alkoxy, aryloxy, N-alkylamino, N,N-dιalkylamιno, N- arylamino, thioalkoxy, thioaryloxy or substituted alkyl wherein alkyl, aryl, heteroaryl, and substituted alkyl are as defined herein

The term "thioacyl group" means a radical -C(S)R, where R is hydrogen, halogen, alkyl, aryl, heteroaryl, alkoxy, aryloxy, N-alkylamino, N,N-dιalkylamιno, N- arylammo, thioalkoxy, thioaryloxy or substituted alkyl wherein alkyl, aryl, heteroaryl, and substituted alkyl are as defined herein

The term "sulfonyl group" means a radical -SO 2 R, where R is hydrogen, halogen, alkyl, aryl, heteroaryl, alkoxy, aryloxy, N-alkylamino, N,N-dιalkylamιno, N- arylamino, thioalkoxy, thioaryloxy or substituted alkyl wherein alkyl, aryl, heteroaryl, and substituted alkyl are as defined herein

The term "acyloxy" means a radical — OC(=O)R, where R is hydrogen, alkyl, aryl, heteroaryl or substituted alkyl wherein alkyl, aryl, heteroaryl, and substituted alkyl are as defined herein Representative examples include, but are not limited to formyloxy, acetyloxy, cylcohexylcarbonyloxy, cyclohexylmethylcarbonyloxy, benzoyloxy, benzylcarbonyloxy, and the like

The term "heteroalkyl," "heteroalkenyl," and "heteroalkynyl" refers to alkyl, alkenyl, and alkynyl groups respectively as defined above, that contain the number of carbon atoms specified (or if no number is specified, having 1 to 12 carbon atoms, preferably 1 to 6) which contain one or more heteroatoms, preferably one to three heteroatoms, as part of the mam, branched, or cyclic chains in the group Heteroatoms are independently selected from the group consisting of -NR-, -NRR, -S-, -S(O) — , -S(O) 2 -, — O— , -SR, -S(O)R, -S(O) 2 R, —OR —PR—, -PRR, -P(O)R- and -P(O)RR, (where each R is hydrogen, alkyl or aryl) preferably -NR where R is hydrogen or alkyl and/or O Heteroalkyl, heteroalkenyl, and heteroalkynyl groups may be attached to the remainder of the molecule either at a heteroatom (if a

valence is available) or at a carbon atom Examples of heteroalkyl groups include, but are not limited to, groups such as — O— CH 3 , -CH 2 -O-CH 3 , -CH 2 -CH 2 -O-CH 3 , -S-CH 2 -CH 2 -CH 3 , —CH 2 —CH(CH 3 )—S—CH 3 , -CH 2 -CH 2 -NH-CH 2 -CH 3 , 1- ethyl-6-propylpιperιdιno, 2-ethylthιophenyl, piperazino, pyrrolidino, piperidino, morpholino, and the like Examples of heteroalkenyl groups include, but are not limited to groups such as — CH=CH-CH 2 - N(CH 3 ) 2 , and the like

The term "heteroaryl" or "HetAr" refers to an aromatic monovalent monocyclic, bicyclic, or tricyclic radical containing 4, 5, 6, 7, 8, 9, 10, 11 , 12, 13, 14, 15, 16, 17, or 18 -member ring atoms, including 1 , 2, 3, 4, or 5 heteroatoms, preferably one to three heteroatoms including, but not limited to heteroatoms such as N, O, P, or S, within the ring Representative examples include, but are not limited to single ring such as imidazolyl, pyrazolyl, pyrazinyl, pyridazinyl, pyπmidinyl, pyrrolyl, pyπdyl, thiophene, and the like, or multiple condensed rings such as indolyl, quinoline, quinazoline, benzimidazolyl, indolizinyl, benzothienyl, and the like The heteroalkyl, heteroalkenyl, heteroalkynyl and heteroaryl groups can be unsubstituted or substituted with one or more substituents, preferably one to three substituents, selected from the group consisting of alkyl, alkenyl, alkynyl, benzyl, halogen, alkoxy, acyloxy, amino, mono or dialkylamino, hydroxyl, mercapto, carboxy, benzyloxy, phenyl, aryloxy, cyano, nitro, thioalkoxy, carboxaldehyde, carboalkoxy and carboxamide, or a functionality that can be suitably blocked, if necessary for purposes of the invention, with a protecting group Examples of such substituted heteroalkyl groups include, but are not limited to, piperazine, pyrrolidine, morpholine, or pipeπdine, substituted at a nitrogen or carbon by a phenyl or benzyl group, and attached to the remainder of the molecule by any available valence on a carbon or nitrogen, — NH- S(=O) 2 — phenyl, — NH- (C=O)O-alkyl, — NH- C(=O)O-alkyl-aryl, and the like The heteroatom(s) as well as the carbon atoms of the group can be substituted The heteroatom(s) can also be in oxidized form

The term "heteroarylene" refers to the diradical group derived from heteroaryl (including substituted heteroaryl), as defined above, and is exemplified by the groups 2,6-pyrιdιnylene, 2,4-pyrιdιnylene, 1 ,2-quιnolιnylene, 1 ,8-quιnolιnylene, 1 ,4- benzofuranylene, 2,5-pyπdιnylene, 2,5-ιndolenylene, and the like

The term "heteroalkylene", "heteroalkenylene", and "heteroalkynylene" refers to the diradical group derived from heteroalkyl, heteroalkenyl, and heteroalkynyl (including substituted heteroalkyl, heteroalkenyl, and heteroalkynyl) as defined above The term "carboxaldehyde" means — CHO

The term "carboalkoxy" means — C(=O)OR where R is alkyl as defined above and include groups such as methoxycarbonyl, ethoxycarbonyl, and the like

The term "carboxamide" means — C(=O)NHR or — C(=O)NRR' where R and R 1 are independently hydrogen, aryl or alkyl as defined above Representative examples include groups such as aminocarbonyl, N-methylaminocarbonyl, N, N- dimethylaminocarbonyl, and the like

The term "carboxy" refers to the radical -C(O)OH

The term "carbamoyl" refers to the radical -C(O)NH 2

The term "halogen" or "halo" as used herein refer to Cl, Br, F or I substituents, preferably fluoro or chloro

The term "hydroxy" refers to a —OH radical

"Isomers" Compounds that have the same molecular formula (or elemental composition) but differ in the nature or sequence of bonding of their atoms or the arrangement of their atoms in space are termed "isomers" Isomers in which the connectivity between atoms is the same but which differ in the arrangement of their atoms in space are termed "stereoisomers" Stereoisomers that are not mirror images of one another are termed "diastereomers" and those that are non-supenmposable mirror images of each other are termed "enantiomers" When a compound has an asymmetric center, for example which is bonded to four different groups, a pair of enantiomers is possible An enantiomer can be characterized by the absolute configuration of its asymmetric center and is described by the R- and S-sequenαng rules of Cahn, lngold and Prelog, or by the manner in which the molecule rotates the plane of polarized light and designated as dextrorotatory or levorotatory (ι e , as (+) or (-)-ιsomers respectively) A chiral compound can exist as either an individual enantiomer or as a mixture thereof A mixture containing equal proportions of the enantiomers is called a "racemic mixture"

The compounds of this invention may possess one or more asymmetric centers Such compounds can therefore be produced as individual (R)- or (S)- stereoisomers or as mixtures thereof Unless indicated otherwise, the description or naming of a particular compound in the specification and claims is intended to include both individual enantiomers and mixtures, racemic or otherwise, thereof The description is also intended to include all possible diastereomers and mixtures thereof The methods for the determination of stereochemistry and the separation of stereoisomers are well-known in the art (see discussion in Chapter 4 of "Advanced Organic Chemistry", 4th edition J March, John Wiley and Sons, New York, 1992)

"Affinity" represents the tendency of different forms of matter (particles, chemical elements, molecules, ions, supramolecular assemblies) to aggregate or bond According to the present invention, phosphonated glycopeptide or lipoglycopeptides demonstrate affinity for osseous tissues In preferred examples, the phosphonated glycopeptide or lipoglycopeptides demonstrate high affinity for osseous tissues For example, compounds of the invention may bind rapidly and efficiently to bone powder, calcium phosphate or other bone component, such that after a duration of 10 mm to 1 hour of their introduction to a suspension of osseous matter in an aqueous medium at a pH of between 5 and 9, greater than 50% of the amount of a compound of the invention is no longer freely found in solution Such binding demonstrates high affinity for osseous tissues Preferably, greater than 70% of the amount of the phosphonated glycopeptide or lipoglycopeptide molecule is bonded to bone one hour after its introduction to the suspension of osseous matter in the aqueous medium Such binding demonstrates very high affinity for osseous tissues "Optically pure" As generally understood by those skilled in the art, an optically pure compound is one that is enantiomerically pure As used herein, the term "optically pure" is intended to mean a compound which comprises at least a sufficient amount of a single enantiomer to yield a compound having the desired pharmacological activity Preferably, "optically pure" is intended to mean a compound that comprises at least 90% of a single isomer (80% enantiomeric excess), preferably at least 95% (90% e e ), more preferably at least 97 5% (95% e e ), and most preferably at least 99% (98% e e ) Preferably, the compounds of the invention are optically pure

"Protecting group" refers to a chemical group that exhibits the following characteristics 1 ) reacts selectively with the desired functionality in good yield to give a protected substrate that is stable to the projected reactions for which protection is desired, 2) is selectively removable from the protected substrate to yield the desired functionality, and 3) is removable in good yield by reagents compatible with the other functional group(s) present or generated in such projected reactions Examples of suitable protecting groups can be found in Greene et al (1991 ) Protective Groups in Organic Synthesis, 2nd Ed (John Wiley & Sons, lnc , New York) Preferred amino protecting groups include, but are not limited to, benzyloxycarbonyl (CBz), t-butyloxycarbonyl (Boc), t-butyldimethylsilyl (TBDMS), 9-fluorenylmethyl-oxycarbonyl (Fmoc), or suitable photolabile protecting groups such as 6-nιtroveratryloxy carbonyl (Nvoc), nitropiperonyl, pyrenylmethoxycarbonyl, nitrobenzyl, dimethyl dimethoxybenzil, 5-bromo-7-nιtroιndolιnyl, and the like Preferred hydroxyl protecting groups include

acetyl (Ac), benzoyl (Bz), benzyl (Bn), Tetrahydropyranyl (THP), TBDMS, photolabile protecting groups (such as nitroveratryl oxymethyl ether (Nvom)), Mom (methoxy methyl ether), and Mem (methoxy ethoxy methyl ether) Particularly preferred protecting groups include NPEOC (4-nιtrophenethyloxycarbonyl) and NPEOM (4- mtrophenethyloxy-methyloxycarbonyl)

"Prodrug" Phosphonated glycopeptide and lipoglycopeptide antimicrobial molecules of the present invention may be formulated as prodrugs According to the present invention, a prodrug is an inactive (or significantly less active) form of any of the phosphonated glycopeptide and lipoglycopeptide antimicrobial molecule compounds of the present invention Upon in vivo processing, prodrugs of the present invention release an active phosphonated glycopeptide and lipoglycopeptide antimicrobial molecule Prodrugs of phosphonated glycopeptide and lipoglycopeptide antimicrobial molecules of the present invention may be prepared by modifying functional groups present on the phosphonated glycopeptide and lipoglycopeptide antimicrobial molecules in such a way that the modifications may be cleaved in vivo to release the phosphonated glycopeptide and lipoglycopeptide antimicrobial molecules

Prodrugs include compounds of Formula (I) and/or Formula (II) wherein a hydroxyl, carboxyl or amino group in the glycopeptide and lipoglycopeptide antimicrobial molecule portion of the compound is bonded to any group that may be cleaved in vivo to regenerate the free hydroxyl, carboxyl or amino group, respectively Such prodrug groups are in addition to the phosphonated linker that may be coupled to a hydroxy, carboxy and/or amino group of an glycopeptide and lipoglycopeptide antimicrobial molecule Examples of prodrug groups include, but are not limited to, esters (e g , acetate, formate, and benzoate derivatives) and carbamates (e g , N 1 N- dimethylaminocarbonyl) on hydroxy functional groups of the glycopeptide and lipoglycopeptide antimicrobial molecule portion of the phosphonated compounds of the present invention The present invention also includes those prodrugs requiring two or more events in prodrug cleavage According to that embodiment, more complex compounds would release, upon cleavage, a prodrug of a phosphonated glycopeptide and lipoglycopeptide antimicrobial molecule, the latter prodrug being activatable to release a desired phosphonated glycopeptide and lipoglycopeptide antimicrobial molecule The skilled artisan will understand that prodrugs of phosphonated glycopeptide and lipoglycopeptide antimicrobial molecules of the present invention may undergo two cleavage events, one of which cleaves the cleavable linker and thus releases the phosphonate group, the other of which results in the release of the prodrug group

A "pharmaceutically acceptable prodrug" is intended to mean prodrug of phosphonated glycopeptide and lipoglycopeptide antimicrobial molecule, such as a prodrug of a compound of Formula (I) and/or Formula (II), in a formulation that may be administered to a subject, such as a mammal, preferably a human For example, the prodrug may be in a formulation comprising a pharmaceutically acceptable carrier or excipient

A "pharmaceutically acceptable active metabolite" is intended to mean a pharmacologically active product produced through metabolism in the body of a compound of Formula (I) or Formulae (II) as defined herein A "pharmaceutically acceptable solvate" is intended to mean a solvate that retains the biological effectiveness and properties of the biologically active components of compounds of Formula I and/or Formula Il Examples of pharmaceutically acceptable solvates include, but are not limited to water, isopropanol, ethanol, methanol, DMSO, ethyl acetate, acetic acid, and ethanolamine A "pharmaceutically acceptable carrier or excipient" means any compound, solution, substance or material that can be used in a formulation of the compounds of the present invention that may be administered to a subject In particular, carriers and excipients of the present invention are those useful in preparing a pharmaceutical composition that is generally safe, non-toxic and neither biologically nor otherwise undesirable, and that may present pharmacologically favorable profiles and that includes carriers and excipient that are acceptable for veterinary use as well as human pharmaceutical use Suitable pharmaceutically acceptable carriers and excipients are well known in art and can be determined by those of skill in the art as the clinical situation warrants The skilled artisan will understand that diluents are included within the scope of the terms carriers and excipients Examples of suitable carriers and excipients include saline, buffered saline, dextrose, water, glycerol, ethanol, more particularly (1 ) Dulbecco's phosphate buffered saline, pH about 7 4, containing about 1 mg/ml to 25 mg/ml human serum albumin, (2) 0 9% saline (0 9% w/v NaCI), (3) 5% (w/v) dextrose, and (4) water A "pharmaceutically acceptable salt" is intended to mean a salt of phosphonated glycopeptide or lipoglycopeptide antimicrobial molecule, such as a salt of a compound of Formula (I) and/or Formula (II), in a formulation that may be administered to a subject, such as a mammal, preferably a human For example, the salt may be in a formulation comprising a pharmaceutically acceptable carrier or excipient

"Saccharide" represents saturated polyhydroxylated compounds The term is sometimes limited to polyhydroxylated carbon chains possessing an aldehyde or a ketone moiety either free or masked as an acetal or a ketal functionality In this case, it is intended to include monosaccharides, oligosaccharides and polysaccharides as well as substances derived from monosaccharides by reduction of the carbonyl group

(alditols), by oxidation of one or more terminal groups to carboxylic acids, by oxidation of one or more secondary hydroxyl groups to ketones, by replacement of one or more hydroxy group(s) by a hydrogen atom, an amino group, an 0-lιnked ester group, a C- linked ester group, an N-linked amide group, a C-linked amide group, an alkyl group, an aryl group, a thiol group or similar heteroatomic groups and/or by replacement of one or more of the hydrogens bonded to carbons by a C-linked ester group, a C-linked amide group, an alkyl group, an aryl group or other heteroatomic groups It also includes oligomers of modified and unmodified monosaccharides as well as derivatives of these compounds Unmodified, oxidized, reduced or substituted saccharide monoradicals are covalently attached to the glycopeptide via any atom of the saccharide moiety, preferably a carbon Representative saccharide include, by way of illustration, hexoses such as D-glucose, D-mannose, D-xylose, D-galactose, vancosamine, 3-desmethyl- vancosamine, 3-epι-vancosamιne, 4-epι-vancosamιne, acosamine, actinosamine, daunosamine, 3-epι-daunosamιne, πstosamine, D-glucamine, N-methyl-D-glucamιne, D-glucuronic acid, N-acetyl-D-glucosamine, N-acetyl-D-galactosamine, sialyic acid, iduronic acid, L-fucose, and the like, pentoses such as D-πbose or D-arabinose, ketoses such as D-πbulose or D-fructose, disaccharides such as 2-O-(α-L- vancosamιnyl)-β-D-glucopyranose, 2-O-( α-L-vancosamιnyl)-β-D-glucopyranose, 2-O- ( α-L-3-epιvancosamιnyl)-β-D-glucopyranose, 2-O-(3-desmethyl- α-L-vancosamιnyl)-β- D-glucopyranose, sucrose, lactose, or maltose, derivatives such as acetals, amines, acylated, sulfated and phosphorylated sugars, oligosaccharides having from 2 to 10 saccharide units These saccharides are can be either in their open or preferably in their pyranose or furanose forms The saccharide may be linked to the aglycone of the glycopeptide or lipoglycopeptide antimicrobial agent indirectly via an additional spacer such as an ethylene, propylene, butylenes or phenylene group

The term "ammo-containing saccharide group" refers to a saccharide group having an amino substituent Representative ammo-containing saccharide include L- vancosamine, 3-desmethyl-vancosamιne, 3-epι-vancosamιne, 4-epι-vancosamιne,

acosamme, actinosamine, daunosamine, 3-epι-daunosamιne, ristosamine, N-methyl-D- glucamiπe and the like

"Salt" Phosphonated glycopeptide and lipoglycopeptide antimicrobial molecules of the present invention may be in the form of a salt Salts of phosphonated glycopeptide and lipoglycopeptide antimicrobial molecules of the present invention means a salt that retains or improves the biological effectiveness and properties of the free acids and bases of the parent compound as defined herein or that takes advantage of an intrinsically charged functionality on the molecule and that is not biologically or otherwise undesirable Such salts include the following (1) acid addition salts, formed with inorganic acids such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like, or formed with organic acids such as acetic acid, propionic acid, hexanoic acid, cyclopentanepropionic acid, glycolic acid, pyruvic acid, lactic acid, malonic acid, succinic acid, malic acid, maleic acid, fumaric acid, tartaric acid, citric acid, benzoic acid, 3-(4- hydroxybenzoyl)benzoιc acid, cinnamic acid, mandelic acid, methanesulfonic acid, ethanesulfonic acid, 1 ,2-ethane-dιsulfonιc acid, 2-hydroxyethanesulfonιc acid, benzenesulfonic acid, 4-chlorobenzenesulfonιc acid, 2-napthalenesulfonιc acid, 4- toluenesulfonic acid, camphorsulfonic acid, 3-phenyl propionic acid, tπmethylacetic acid, tertiary butylacetic acid, lauryl sulfuric acid, gluconic acid, glutamic acid, hydroxynapthoic acid, salicylic acid, stearic acid, muconic acid, and the like,

(2) salts formed when an acidic proton present in the parent compound either is replaced by a metal ion, e g , an alkali metal ion, an alkaline earth ion, or an aluminum ion, or coordinates with an organic base such as ethanolamme, diethanolamine, triethanolamine, tromethamme, N-methylglucamine, and the like, or (3) salts formed when a charged functionality is present on the molecule and a suitable counterion is present, such as a tetraalkyl(aryl)ammonιum functionality and an alkali metal ion, a tetraalkyl(aryl)phosphonιum functionality and an alkali metal ion, an imidazolium functionality and an alkali metal ion, and the like

As used herein, the terms "bone", "bone tissues" or "osseous tissues" refer to the dense, semi rigid, porous, calcified connective tissue forming the major portion of the skeleton of most vertebrates It also encompasses teeth, osteo-articular tissues and calcifications that are frequently seen in the walls of atherosclerotic vessels

The term "glycopeptide antimicrobial molecule" and "lipoglycopeptide antimicrobial molecule", and related terms, have the same meaning and refer to antimicrobial agents which are part of the well known class of "glycopeptides and lipoglycopeptides" as described in more detail herein

The term "phosphonated group" is intended to mean any compound non-toxic to humans having at least one phosphorus atom bonded to at least three oxygen atoms and having a measurable affinity to osseous tissues as described hereinafter

The term "antibacterial" includes those compounds that inhibit, halt or reverse growth of bacteria, those compounds that inhibit, halt, or reverse the activity of bacterial enzymes or biochemical pathways, those compounds that kill or injure bacteria, and those compounds that block or slow the development of a bacterial infection

The terms "treating" and "treatment" are intended to mean at least the mitigation of a disease condition associated with a bacterial infection in a subject, including mammals such as a human, that is alleviated by a reduction of growth, replication, and/or propagation of any bacterium such as Gram-positive organisms, and includes curing, healing, inhibiting, relieving from, improving and/or alleviating, in whole or in part, the disease condition The term "prophylaxis" is intended to mean at least a reduction in the likelihood that a disease condition associated with a bacterial infection will develop in a mammal, preferably a human The terms "prevent" and "prevention" are intended to mean blocking or stopping a disease condition associated with a bacterial infection from developing in a mammal, preferably a human In particular, the terms are related to the treatment of a mammal to reduce the likelihood ("prophylaxis") or prevent the occurrence of a bacterial infection, such as bacterial infection that may occur during or following a surgery involving bone reparation or replacement The terms also include reducing the likelihood ("prophylaxis") of or preventing a bacterial infection when the mammal is found to be predisposed to having a disease condition but not yet diagnosed as having it For example, one can reduce the likelihood or prevent a bacterial infection in a mammal by administering a compound of Formula (I) and/or Formula (II), or a pharmaceutically acceptable prodrug, salt, active metabolite, or solvate thereof, before occurrence of such infection

The term "subject" is intended to mean an animal, such as a mammal, including humans and animals of veterinary importance, such as dogs, cats, horses, sheep, goats, and cattle

C) Compounds of the invention

As will be described hereinafter in the Exemplification section, the inventors have prepared phosphonated derivatives of glycopeptide or lipoglycopeptide antimicrobial molecules having a high binding affinity to osseous tissues

In one embodiment, the compounds of the invention are represented by the general Formula (I)

as well as pharmaceutically acceptable salts, esters and prodrugs thereof, wherein B is a phosphonated group, preferably having a high affinity to osseous tissues, L is a bond or a linker, preferably covalently coupling B to A, α is 1 , 2, 3, 4, 5, 6 or 7, preferably 1 , 2 or 3, and A is a glycopeptide or lipoglycopeptide antimicrobial molecule,

As mentioned previously, the essence of the invention lies in the presence of a phosphonated group attached to a glycopeptide or lipoglycopeptide antibiotic for increasing the affinity, binding, accumulation and/or retention time of the glycopeptide or lipoglycopeptide antibiotic to or within the bones

Phosphonates

All non-toxic phosphonated groups having anaffmity, preferably a high affinity or a very high affinity, to bone due to their ability to bind the Ca 2+ ions found in the hydroxyapatite forming the bone tissues are suitable according to the present invention Suitable examples of phosphonated groups can be found in WO 04/026315 (Ilex Oncology Research), US 6,214,812 (MBC research), US 5,359,060 (Pfizer), US 5,854,227 and US 6,333,424 (Ehzanor), US 6,548,042 (Arstad and Skattelbol) and WO 2004/089925 (Semaphore Pharmaceuticals) Examples of bisphosphonate and trisphosphonate groups suitable for the present invention include but are not limited to those having the formula

wherein each R* is independently selected from the group consisting of H, lower alkyl, cycloalkyl, aryl and heteroaryl, with the proviso that at least two, preferably

three, R* are H,

R 4 is -CH 2 -, — O— , -S-, or — NH-, each R 5 is independently selected from the group consisting of H, R 6 , OR 6 , NR 6 , and SR 6 , wherein R 6 is H, lower alkyl, cycloalkyl, aryl, heteroaryl or NH 2 , X is H, OH, NH 2 , or a halo group,

X 1 are both H, or each is independently selected from the group consisting of H,

OH, NH 2 , and a halo group, and

Li is the point of attachment to L

Although monophosphonates, bisphosphonates, and tris- or tetraphosphonates could potentially be used, bisphosphonates are preferred More preferably, the bisphosphonate group is the bisphosphonate -CH(P(O)(OH) 2 ) 2 As shown in Example 3 hereinafter, glycopeptide and lipoglycopeptide derivatives possessing such a bisphosphonate group have a strong binding affinity for hydroxyapatite powder Of course, other types of phosphonated group could be selected and synthesized by those skilled in the art For instance the phosphonated group may be an esterase- activated bisphosphonate radical (Vepsalamen J , Current Medicinal Chemistry, 9, 1201 -1208, 2002) or be any other suitable prodrug thereof These and other suitable phosphonated groups are encompassed by the present invention

Glvcopeptide and lipoqlvcopeptide antibiotics

Glycopeptide and lipoglycopeptide antibiotics are a well known class of biologically produced or semi-synthetic Gram-positive antimicrobial agents (Williams, D H et al, Angewandte Chemie International Edition in English (1999), 1999, 38, 1172- 1193 Nicolaou, K C et al, Angewandte Chemie International Edition in English (1999), 38, 2097-2152 Kahne, D et al Chemical Reviews (2005), 105, 425 - 448, Pace, J L et al, Biochemical Pharmacology (2006), 71 , 968-980) Vancomycin and teicoplanin are certainly the best known compounds in this class Both drugs were proven clinically and microbiologically to have potent activity against Gram-positive organisms Oritavancin (US Patent No 5,840,684), dalbavancin (US patent No 5,750,509) and telavancin (US patent No 6,635,618) are recent examples of this class of compounds possessing extremely attractive pharmacological profiles with potent activity against gram-positive organisms, including methicillin-resistant Staphylococcus aureus, intermediate and fully vancomycin-resistant Staphylococcus aureus, vancomycin-resistant Enterococcus spp , and Streptococcus spp The present invention is not restricted to a specific glycopeptide or lipoglycopeptide antibiotic, but encompasses all kinds of glycopeptide or lipoglycopeptide molecules having a suitable

antimicrobial activity including, but not limited to, those disclosed in the above-listed US patents and PCT patent applications (incorporated herein by reference) and other glycopeptide or lipoglycopeptide antibiotic derivatives and hybrids such as glycopeptide-cephalosporin (as described in US patent application No 20050239691 for example)

According to a preferred embodiment, the term "glycopeptide and lipoglycopeptide antimicrobial molecule" includes all compounds having the Formula A 1 illustrated below

as well as pharmaceutically acceptable salts, esters and prodrugs thereof, where R 1 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic and — R a — Y — R b — (Z) x , or R 1 is a saccharide group optionally substituted with — R a — Y— R b — (Z) x , — R f , -C(O)R,, or — C(O)- R a — Y— R b — (Z) x , R 2 is hydrogen or a saccharide group optionally substituted with -R a -Y-R b -(Z) x , — R f , — C(O)R', or — C(O)- R a — Y— R b — (Z) x , R 3 is — OR C , — NR C R C , — O— R a — Y— R b — (Z) x , — NR C — R a — Y— R b — (Z) x , — NR c R e , or — O— R e ,

R 4 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, — R a — Y— R b — (Z) x , — C(O)R d and a saccharide group optionally substituted with _R a — Y— R b _ (Z) x , — R f , or — C(O)- R a — Y— R b — (Z) x , or R 4 and R 5 can be joined, together with the atoms to which they are attached, to form a heterocyclic ring optionally substituted with — NR C — R a — Y— R b — (Z) x ,

R 5 is selected from the group consisting of hydrogen, halo, — CH(R 0 ) — NR C R°, — CH(R 0 )- NR c R e , — CH(R 0 )- NR C — R a — Y— R b — (Z) x , — CH(R 0 )- R x , and — CH(R 0 )- NR°— R a — C(O)- R x ,

R 6 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, — R a — Y— R b — (Z) x ,

— C(0)R d and a saccharide group optionally substituted with — R a — Y— R b — (Z) x , — R f , — C(O)R f , or — C(O)- R a — Y— R b — (Z) x , or R 5 and R 6 can be joined, together with the atoms to which they are attached, to form a heterocyclic ring optionally substituted with — NR°— R a — Y— R b — (Z) x , R 7 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, — R a — Y — R b — (Z) x , and — C(O)R d ,

R 8 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic and — R a — Y— R b — (Z) x ,

R 9 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl and heterocyclic,

R 10 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl and heterocyclic, or R 8 and R 10 are joined to form — Ar 1 — O — Ar 2 — , where Ar 1 and Ar 2 are independently arylene or heteroarylene,

R 11 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl and heterocyclic, or R 10 and R 11 are joined, together with the carbon and nitrogen atoms to which they are attached, to form a heterocyclic ring,

R 12 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic, — C(O)R d , — C(NH)R d , -C(O)NR 0 R 0 , — C(O)OR d , -C(NH)NR 0 R 0 , — R a — Y— R b — (Z) x , and — C(O)- R b — Y— R b — (Z) x , or R 11 and R 12 are joined,

together with the nitrogen atom to which they are attached, to form a heterocyclic ring,

R 13 is hydrogen or — OR 14 ,

R 14 is hydrogen, — C(O)R d or a saccharide group, R a is each independently selected from the group consisting of alkylene, substituted alkylene, alkenylene, substituted alkenylene, alkynylene and substituted alkynylene,

R b is each independently selected from the group consisting of a covalent bond, alkylene, substituted alkylene, alkenylene, substituted alkenylene, alkynylene and substituted alkynylene,

R c is each independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic and — C(O)R d , R d is each independently selected from the group consisting of alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl and heterocyclic,

R e is each a saccharide group, R f is each independently selected from the group consisting of alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, and heterocyclic,

R x is an N-linked amino saccharide or an N-linked heterocycle, X is each independently selected from the group consisting of hydrogen, fluoro, chloro, bromo and iodo,

Y is each independently selected from the group consisting of , — CH 2 — , — O —

, -S-, — S— S— , — NR C — , -S(O)-, -SO 2 -, — NR 0 C(O)- , -OSO 2 -,

-OC(O)-, — N(R C )SO 2 — , -C(O)NR 0 -, -C(O)O-, -SO 2 NR 0 -, -SO 2 O-, — P(O)(OR°)O— , — P(O)(OR°)NR C — , — OP(O)(OR°)O— , — OP(O)(OR C )NR°— ,

-OC(O)O-, -NR 0 C(O)O-, -NR 0 C(O)NR 0 — , -OC(O)NR 0 -, -C(O)-, and -N(R°)SO 2 NR C — ,

Z is each independently selected from the group consisting of hydrogen, aryl, cycloalkyl, cycloalkenyl, heteroaryl, heterocyclic, and a saccharide, n is O, 1 , or 2, x is 1 or 2, and

Those skilled in the art will readily identify, isolate and/or prepare the suitable glycopeptide or lipoglycopeptide antimicrobial molecules according to the invention If necessary they could refer to the numerous literature found in the art, including the US patents and PCT patent applications listed hereinbefore, and more particularly to US Patents No 5,840,684, 5,750,509 and 6,635,618

According to one embodiment, the glycopeptide or lipoglycopeptide antimicrobial molecule is a derivative of vancomycin According to another embodiment, the glycopeptide or lipoglycopeptide antimicrobial molecule is a derivative of teicoplanin According to a third embodiment, the glycopeptide or lipoglycopeptide antimicrobial molecule is a derivative of chloroeremomycin According to a fourth embodiment, the glycopeptide or lipoglycopeptide antimicrobial molecule is a derivative of oritavancin According to a fifth embodiment, the glycopeptide or lipoglycopeptide antimicrobial molecule is a derivative of dalbavancin According to a sixth embodiment, the glycopeptide or lipoglycopeptide antimicrobial molecule is a derivative of telavancin The chemical structures of these six molecules are illustrated hereinafter Arrows indicate preferred sites for attachment of the phosphonated group (direct attachment or via an optional linker), but those skilled in the art will recognize that all hydroxyl amino, amido and carboxyl groups may be possible sites for attachment

Chloroeremomycin Oritavancin

Telavancin

Specific examples of vancomycin and oritavancin derivatives according to the invention are shown in the Exemplification section Even though in the examples phosphonated groups have not been attached to all the preferred attachment sites shown by the arrows, the results presented in the Exemplification section confirm that it is possible to synthesize phosphonated biologically active glycopeptide and lipoglycopeptide derivatives having a highly increased affinity for bony materials Similarly, although not tested, the invention encompasses phosphonated glycopeptide and lipoglycopeptide derivatives having more than just one phosphonated group (one at the carboxy and one at one of the amino groups on the oritavancin molecule for instance) As mentioned previously, the above identified sites of attachment are only preferred sites for tethering a phosphonated group and all other potential sites (on any of the hydroxyl groups for instance) are covered by the present invention

Linkers A cleavable linker L covalently and reversibly couples the phosphonated group

B to glycopeptide or lipoglycopeptide antimicrobial molecules A As used herein, the term "cleavable" refers to a group that is chemically or biochemically unstable under physiological conditions The chemical instability preferably results from spontaneous decomposition due to a reversible chemical process, an intramolecular chemical reaction or hydrolysis (ι e splitting of the molecule or group into two or more new molecules or groups due to the net insertion of one or more water molecules) when it depends on an intermolecular chemical reaction

Cleavage of the linker may range from being very rapid to being very slow For instance, the half-life of the cleavable hker may be of about 1 minute, about 15 minutes, about 30 minutes, about 1 hour, about 5 hours, about 10 hours, about 15 hours, about 1 day or about 48 hours The cleavable linker may be an enzyme- sensitive linker that is cleavable only by selected specific enzymes (e g amidase, esterase, metalloproteinase, etc) or may be susceptible to cleavage by other chemical means, such as but not limited to acid/base catalysis or self-cleavage For instance, it is conceivable according to the invention to have an esterase-sensitive linker that is cleavable only by bone-specific esterases (Goding et al Biochim Biophys Acta (2003), 1638(1 ) 1-19) or bone-specific metalloproteinase (MMP) (Kawabe et al , CIm Orthop (1986) 21 1 244-51 , Tuckermann et al , Differentiation (2001 ), 69(1 ) 49-57, Sellers et al , Biochem J (1978) 171(2) 493-6) or by the action of alkaline phosphatases thereby releasing the glycopeptide or lipoglycopeptide antibiotic at its desired site of action

Similarly, it is conceivable to use a cleavable linker which is not too easily cleavable in

the plasma, thereby permitting a sufficient amount of the phosphonated glycopeptide or lipoglycopeptide antimicrobial molecules to reach and accumulate within the osseous tissues before being cleaved to release the glycopeptide or lipoglycopeptide antimicrobial molecules For instance, the linker may be selected such that only 1 %, 5%, 10%, 15%, 20%, 25%, 30%, 35%, 40%, 50%, 60%, or 70% of the bone-bonded antibiotic is released through a time period extending to 1 minute, 15 minutes, 30 minutes, 1 hour, 5 hours, 10 hours, 15 hours, 1 day, 2 days, 3 days, 4 days, 5 days, 6 days 7 days, one week, two weeks, three weeks or more following administration of the compound of the invention Preferably, the linker is selected such that only about 1 % to about 25% of the bone-bound glycopeptide or lipoglycopeptide antimicrobial molecule is released per day The choice of the linker may vary according to factors such as (ι) the site of attachment of the phosphonated group to the glycopeptide or lipoglycopeptide antimicrobial molecule, (ιι) the type of phosphonated group used, (in) the type of glycopeptide or lipoglycopeptide antimicrobial molecule used, and (ιv) the desired ease of cleavage of the linker and associated release of the glycopeptide or lipoglycopeptide antimicrobial molecule

Preferably, the linker L couples the phosphonated group B to a glycopeptide or lipoglycopeptide antimicrobial molecule A through one or more hydroxyl groups on A, through one or more nitrogen atoms on A, through one or more carboxyl groups on A, or a combination of one or more hydroxyl groups, one or more nitrogen atoms, and/or one or more carboxyl groups, on A Between 1 and 7 phosphonated groups may be coupled to A through any combination of linkers L

The linker is facultative because its presence is dependent upon (ι) the site of attachment of the phosphonated group to the glycopeptide or lipoglycopeptide molecule, (n) the type of phosphonated group used, (in) the type of glycopeptide or lipoglycopeptide used, and (ιv) the desired ease of cleavage of the linker and associated release of the glycopeptide or lipoglycopeptide antibiotic For instance, it is possible to avoid the linker and tether a phosphonated group directly to the carboxyl group of oritavancin Preferably, the bisphosphonated-linker substructure is described by the formula

BL 1

wherein

Aa indicates the point of attachment to the glycopeptide or lipoglycopeptide

antimicrobial molecule A;

W is a covalent bond or is selected from the group of

T is oxygen or sulfur; each R is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic, amino, substituted amino, hydroxyl, alkoxy, substituted alkoxy, aryloxy, substituted aryloxy, and — R a — Y— R b — Y— R b — B; each R a is independently selected from the group consisting of a covalent bond, alkylene, substituted alkylene, alkenylene, substituted alkenylene, alkynylene, substituted alkynylene, arylene, substituted arylene, —(CO)— alkylene— , substituted —(CO)— alkylene— , —(CO)— alkenylene— , substituted — (CO) — alkenylene — , — (CO) — alkynylene — , substituted — (CO) — alkynylene — , — (CO) — arylene — and substituted —(CO)— arylene— ; each R b is independently selected from the group consisting of a covalent bond, alkylene, substituted alkylene, alkenylene, substituted alkenylene, alkynylene, substituted alkynylene, arylene and substituted arylene; each Y is independently selected from the group consisting of a covalent bond, -CH 2 -, — O— , -S-, — S— S— , — NR C — , -S(O)-, -SO 2 -, -NR 0 C(O)- -OSO 2 -, -OC(O)- — N(R°)SO 2 — , -C(O)NR 0 -, -C(O)O-, -SO 2 NR 0 -, -SO 2 O-, — P(O)(OR C )O— , — P(O)(OR C )NR°— , — OP(O)(OR C )O— , — OP(O)(OR C )NR°— , -OC(O)O-, -NR 0 C(O)O-,

— NR C C(O)NR C — , — OC(O)NR C — , -C(O)-, and -N(R C )SO 2 NR C — ; each R c is independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic and — C(O)R d — ; each R d is independently selected from the group consisting of alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl and heterocyclic;

B is a phosphonated group; each Q is independently selected from the group consisting of nitro, chloro, bromo, iodo and fluoro; each X is independently selected from the group consisting of — O — , — S — , and -N(R)-;

Z is selected from the group consisting of hydrogen, acyl, substituted acyl, aroyl, substituted aroyl, alkoxycarbonyl, substituted alkoxycarbonyl, aryloxycarbonyl, substituted aryloxycarbonyl,

q is 2 or 3, r is 1 , 2, 3, 4 or 5,

W 1 and W 2 are each integers ≥ 0 such that their sum (Wi + w 2 ) is 1 , 2 or 3, a, b, c, d are integers ≥ O such that a+b+c+d ≤7 or null, e and f are integers ≥ O such that e+f = 4, α is O or 1

When L couples B to A through a hydroxyl group on A, preferably L is one or more of the following linkers

wherein: n is an integer < 10, preferably 1 , 2, 3 or 4, more preferably 1 or 2; each p is independently O or an integer ≤ 10, preferably O, 1 , 2, 3 or 4, more preferably O or 1 ; q is 2 or 3 r is 1 , 2, 3, 4 or 5

Wi and W 2 are integers ≥ 0 such that their sum (W 1 + w 2 ) is 1 ,2 or 3 each R L is independently selected from the group consisting of H, ethyl and methyl, preferably H;

B represents the phosphonated group; and

the substructure ' A o of the linker represents the hydroxyl moiety of A.

When L couples B to A through a nitrogen atom on A, preferably L is one or more of the following linkers:

wherein:

B represents said phosphonated group, n is an integer < 10, each p is independently 0 or an integer < 10, each R L IS independently selected from the group consisting of H, ethyl and methyl, q is 2 or 3, r is 1 , 2, 3, 4 or 5,

Wi and W 2 are each integers ≥ 0 such that their sum (W 1 + W 2 ) is 1 , 2 or 3 X is -CH 2 -, -CONRL-, -CO-O-CH 2 -, or — CO— O— , and each Y is independently selected from the group consisting Of -O-, -S- and -NR L -, each Z is indepedently selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, acyl, acyloxy, carboxy, carbamoyl, sulfuryl, sulfinyl, sulfenyl, sulfonyl, mercapto, ammo, hydroxyl, cyano and nitro, wherein s is 1 , 2, 3 or 4, and

Ra is C x Hy where x is an integer of 0 to 20 and y is an integer of 1 to 2x+1 B represents said phosphonated group, and A 3 represents the nitrogen atom on A

When L couples B to A through the carbonyl of a carboxyl group on A, preferably L is one or more of the following linkers

wherein n is an integer < 10, preferably 1 , 2, 3 or 4, more preferably 1 or 2, p is 0 or an integer ≤ 10, preferably 0, 1 , 2, 3 or 4, more preferably 0 or 1 , R L IS H, ethyl or methyl, preferably H,

R x is -S-, -C(R L ) 2 -, — NRL- or — O — , preferably — NR L — , more preferably — NH — , each Y is independently selected from the group consisting of — O — , — S — , and —NR L —, each Z is independently selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, acyl, acyloxy, carboxy, carbamoyl, sulfuryl, sulfinyl, sulfenyl, sulfonyl, mercapto, ammo, hydroxyl, cyano and nitro, wherein s is 1 , 2, 3 or 4, and

B represents the phosphonated group, and

A c . / OH carboxylate group T O of A.

According to another particular embodiment, the compounds of the invention are represented by Formula (II)

as well as pharmaceutically acceptable salts, esters and prodrugs thereof, where R 1 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic, — R a — Y — R b — (Z) x and — L 1 , or R 1 is a saccharide group optionally substituted with — R a — Y— R b — (Z) x , — R f , —C(O)R f , — C(O)- R a — Y— R b — (Z) x , — C(NL 2 )R f , or — C(NL 3 )- R a — Y— R b — (Z) x ,

R 2 is hydrogen, — L 4 or a saccharide group optionally substituted with — R a — Y— R b — (Z) x , — R f , — C(O)R f , — C(O)- R a — Y— R b — (Z) x , — C(NL 5 )R f , or — C(NL 6 )- R a — Y— R b — (Z) x ,

R 3 is selected from the group consisting of — OR C , — NR C R C , — 0— R a — Y— R b — (Z) x , — NR C — R a — Y— R b — (Z) x , — NR c R e , — O— R e , -OL 7 , -NL 8 R 0 , and — NL 9 R 6 ,

R 4 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, — L 10 ,

— R a — Y— R b — (Z) x , — C(O)R d , — C(NL 11 )R d and a saccharide group optionally substituted with — R a — Y— R b — (Z) x , — R f , — C(O)- R a — Y— R b — (Z) x , or — C(NL 12 )- R a — Y— R b — (Z) x , or R 4 and R 5 can be joined, together with the atoms to which they are attached, to form a heterocyclic ring optionally substituted with — NR C — R a — Y— R b — (Z) x or -NL 13 — R a — Y— R b — (Z) x , R 5 is selected from the group consisting of hydrogen, halo, — CH(R 0 )- NR C R C , — CH(R 0 )- NR°R e , — CH(R 0 )- NR 0 - R a — Y— R b — (Z) x , — CH(R 0 )- R", — CH(R 0 )- NR°— R a — C(O)- R", — CH(R 0 )- NL 14 R 0 , — CH(R C )— NL 15 R e ,

— CH(R 0 )- NL 16 - R a — Y— R b — (Z) x , — CH(R 0 )- NL 17 - R a — C(O)- R x and — CH(R C )— NR C — R a — C(NL 18 )- R x

R 6 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, — L 19 , — R a — Y— R b — (Z) x , — C(O)R d , — C(NL 20 )R d and a saccharide group optionally substituted with — R a — Y— R b — (Z) x , — R f , — C(0)R f , — C(O)- R a — Y— R b — (Z) x , — C(NL 21 )R f , or — C(NL 22 )- R a — Y— R b — (Z) x , or R 5 and R 6 can be joined, together with the atoms to which they are attached, to form a heterocyclic ring optionally substituted with — NR 0 - R a — Y— R b — (Z) x or -N L 23 — R a — Y— R b — (Z) x ,

R 7 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, — L 24 , _R a _γ_ R b — (Z) x , — C(0)R d , and — C(NL 25 )R d , R 8 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic and — R a — Y— R b — (Z) x ,

R 9 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic, and — L 26 ,

R 10 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl and heterocyclic, or R 8 and R 10 are joined to form — Ar 1 — O — Ar 2 — , where Ar 1 and

Ar 2 are independently arylene or heteroarylene which may optionally be substituted with -OL 27 ,

R 11 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic, and — L 28 , or R 10 and R 11 are joined, together with the carbon and nitrogen atoms to which they are attached, to form a heterocyclic ring which may optionally be substituted with -OL 29 , -CO 2 L 30 or -NL 31 R 0 , R 12 is selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic,

— L 32 , — C(O)R d , — C(NH)R d , — C(O)NR C R C , — C(O)OR d , -C(NH)NR 0 R 0 , — R a — Y— R b — (Z) x , and — C(O)- R b — Y— R b — (Z) x , — C(NL 33 )R d , -C(O)NL 34 R 0 , -C(O)OL 35 , -C(NH)NL 36 R 0 , — C(NL 37 )NR C R C , and — C(NL 38 )- R b — Y— R b — (Z) x , or R 11 and R 12 are joined, together with the nitrogen atom to which they are attached, to form a heterocyclic ring which may optionally be substituted with -OL 39 , -CO 2 L 40 or — NL 41 R 0 , R 13 is hydrogen or — OR 14 ,

R 14 is selected from the group consisting of hydrogen, — L 42 , — C(0)R d , — C(NL 43 )R d and a saccharide group optionally substituted with — R a — Y— R b — (Z) x , — R f , -C(O)R', — C(O)- R a — Y— R b — (Z) x , — C(NL 44 )R f , or

— C(NL 45 )- R a — Y— R b — (Z) x ,

R a is each independently selected from the group consisting of alkylene, substituted alkylene, alkenylene, substituted alkenylene, alkynylene and substituted alkynylene, R b is each independently selected from the group consisting of a covalent bond, alkylene, substituted alkylene, alkenylene, substituted alkenylene, alkynylene and substituted alkynylene,

R c is each independently selected from the group consisting of hydrogen, alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, heterocyclic and — C(0)R d ,

R d is each independently selected from the group consisting of alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl and heterocyclic,

R e is each a saccharide group optionally substituted with — R a — Y — R b — (Z) x , — R f , -C(O)R', — C(O)- R a — Y— R b — (Z) x , — C(NL 46 )R f , or — C(NL 47 )- R a — Y— R b — (Z) x , R f is each independently selected from the group consisting of alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, cycloalkyl, substituted cycloalkyl, cycloalkenyl, substituted cycloalkenyl, aryl, heteroaryl, and heterocyclic,

R* is an N-linked ammo saccharide or an N-linked heterocycle, either of which may be optionally substituted with — R a — Y— R b — (Z) x , — R', — C(O)R f , — C(O)- R a — Y— R b — (Z) x , — C(NL 48 )R f , or — C(NL 49 )- R a — Y— R b — (Z) x ,

X is each independently selected from the group consisting of hydrogen, fluoro, chloro, bromo and iodo,

Y is each independently selected from the group consisting Of -CH 2 — , — O — ,

-S-, — S— S— , — NR C — , -S(O)-, -SO 2 -, -NR 0 C(O)-, -OSO 2 -,

— OC(O)-, — N(R C )SO 2 — , -C(O)NR 0 -, -C(O)O-, -SO 2 NR 0 -,

-SO 2 O-, — P(O)(OR°)O— , — P(O)(OR°)NR°— , — OP(O)(OR°)O— ,

— OP(O)(OR°)NR C — , -OC(O)O-, -NR 0 C(O)O-, -NR 0 C(O)NR 0 -,

-OC(O)NR 0 , — C(O)- ,— N(R c )SO 2 NR 0 — , — NL 50 - , -NL 51 C(O)-,

-OSO 2 -, -OC(O)-, -N(L 52 JSO 2 -, -C(O)NL 53 -, -SO 2 NL 54 -,

— P(O)(OL 55 )O— , — P(O)(OL 56 )NR C — , — P(O)(OR°)NL 57 — ,

— OP(O)(OL 58 )O— , — OP(O)(OL 59 )NR°— , — OP(O)(OR°)NL 60 — ,

-NL 61 C(O)O-, -NL 62 C(O)NR 0 -, -NR 0 C(O)NL 63 -, -OC(O)NL 64 -,

-N(L 65 )SO 2 NR°— and — N(R°)SO 2 NL 66 — ,

Z is each independently selected from the group consisting of hydrogen, aryl, cycloalkyl, cycloalkenyl, heteroaryl, heterocyclic, a saccharide, — L 67 , — L 68 and

_ L 69 n is O, 1 , or 2, x is 1 or 2, and

each L 1 , L 4 , L 10 , L 19 , L 24 , L 27 , L 29 , L 39 , L 42 , and L 67 is a linker independently selected from the group consisting of

wherein:

B represents said phosphonated group; each p is independently 0 or an integer < 10; each R L is independently selected from the group consisting of H, ethyl and methyl; q is 2 or 3; n is an integer < 10; r is 1 , 2, 3, 4 or 5; and Wi and W 2 are each integers ≥ 0 such that their sum (W 1 + w 2 ) is 1 , 2 or 3;

each L 8 , L 9 , L 13 , L 14 , L 15 , L 16 , L 17 , L 23 , L 26 , L 28 , L 31 , L 32 , L 34 , L 36 , L 37 , L 41 , L 50 , L 51 , L 52 , L 53 , L 54 , L 57 , L 60 , L 61 , L 62 , L 63 , L 64 , L 65 , L 66 and L 68 is a linker independently selected from the group consisting of

wherein:

B represents said phosphonated group; n is an integer < 10; each p is independently 0 or an integer < 10; each R L is independently selected from the group consisting of H, ethyl and methyl; q is 2 or 3; r is 1 , 2, 3, 4 or 5;

Wi and W 2 are each integers ≥ 0 such that their sum (W 1 + W 2 ) is 1 , 2 or 3;

X is -CH 2 -, -CONR L -, -CO-O-CH 2 -, or — CO— O— ; and

R a is C x H y where x is an integer of 0 to 20 and y is an integer of 1 to 2x+1 ;

each L 7 , L 30 , L 35 , L 40 , L 55 , L 56 , L 58 , L 59 and L 69 is a linker independently selected from the group consisting of

wherein n is an integer < 10, preferably 1 , 2, 3 or 4, more preferably 1 or 2, p is 0 or an integer < 10, preferably 0, 1 , 2, 3 or 4, more preferably 0 or 1 ,

R L IS H, ethyl or methyl, preferably H,

R x is — S — , — C(R|_) 2 — , — NR L — or — O — , preferably — NR L — , more preferably — NH — , each Z is independently selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, acyl, acyloxy, carboxy, carbamoyl, sulfuryl, sulfinyl, sulfenyl, sulfonyl, mercapto, amino, hydroxyl, cyano and nitro, wherein s is 1 ,

2, 3 or 4, and

B represents the phosphonated group,

each L 2 , L 3 , L 5 , L 6 , L 11 , L 12 , L 18 , L 20 , L 21 , L 22 , L 25 , L 33 , L 38 , L 43 , L 44 , L 45 , L 46 , L 47 , L 48 and L 49 is a linker independently

wherein p is 0 or an integer < 10, preferably 0, 1 , 2, 3 or 4, more preferably 0 or 1 , R L IS H, ethyl or methyl, preferably H, each Z is independently selected from the group consisting of hydrogen, halogen, alkyl, alkoxy, acyl, acyloxy, carboxy, carbamoyl, sulfuryl, sulfinyl, sulfenyl, sulfonyl, mercapto, amino, hydroxyl, cyano and nitro, wherein s is 1 , 2, 3 or 4, and B represents the phosphonated group,

8 i 9 i 10 i 11 I 12 i 13 i 14 i 15 with the proviso that at least one of L 1 L 2 L 3 L 4 L 5 L 6 L 7 L 8 L s L 1U L 11 L" L 13 L 14 L 1

■ 16 I 17 I 18 I 19 I 20 i 21 ι 22 ι 23 i 24 i 25 i 26 i 27 i 28 i 29 ■ 30 |_31 i 32 i 33 ■ 34 |JJ5 i 36 i 37 i 38

■ 39 I 40 I 41 I 42 i 43 i 44 ■ 45 |_46 i 47 i 48 i 49 |_50 | ^ 51 | ^ 52 i 53 |_54 ^55 | ^ 56 ■ 57 [_58 | ^ 59 j ^ 60 i 61

L" L" L 64 L" L bb L 6 ' L and L b8 ιs present

In a preferred embodiment of formula (II), B is a phosphonated group selected from the group consisting of

wherein each R* is independently selected from the group consisting of H, lower alkyl, cycloalkyl, aryl and heteroaryl, with the proviso that at least two R* are H,

X is H, OH, NH 2 , or a halo group,

X I are both H, or each is independently selected from the group consisting of H, OH, NH 2 , and a halo group, and

L 1 is the point of attachment to L

It is also conceivable according to the invention to couple a single phosphonated group to two or more antibacterial molecules In such circumstances, the antibacterial molecules may be the same (e g two molecules of oπtavancin) or different (e g one molecule of the fluoroquinolone antibacterial ciprofloxacin (Cipro®, US 4,670,444) and one molecule of oπtavancin) The phosphonated group may also be tethered to similar groups (e g the ammo groups) or to different groups (e g the

carboxyl group of one fluoroquinolone molecule and the amino group of a glycopeptide or lipoglycopeptide antimicrobial molecule) Examples of potentially useful, cleavable, multi-antibacterial linkers according to the invention include, but are not limited to, those having the structures

wherein each R d is independently an alkyl or an aryl group, p is O or an integer ≤ 10, preferably 0, 1 , 2, 3 or 4, more preferably 0 or 1 ,

the substructure ° of the linker represents the hydroxyl moiety of the glycopeptide or lipoglycopeptide antimicrobial molecule A,

A 3 represents an amine group of the glycopeptide or lipoglycopeptide antimicrobial molecule A, O the substructure F0 - ° of the linker represents the carboxylic moiety of a fluoroquinolone antimicrobial

Because of its high affinity osseous tissues, the phosphonated group B will likely remain bond to the bones for an extended period of times (up to several years) Therefore, it is very important that the phosphonated group be endowed with low or no measurable toxicity According to another embodiment, the phosphonated group B and the linker L are selected such that the linker is hydrolyzed or cleaved in vivo (preferably mostly in osseous tissues) thereby releasing (ι) the glycopeptide or lipoglycopeptide antimicrobial molecule A and (ιι) a chosen non-toxic phosphonated molecule having a

proven bone therapeutic activity Such compounds would thus have a double utility that is to 1 ) provide locally to the bones for an extended period of time and/or at increased concentrations, an antibiotic useful in preventing and/or treating a bacterial bone infection, and 2) provide to the bones a drug stimulating bone regeneration or inhibiting bone resorption, thereby facilitating bone recovery from damages caused by an infection or other injury Suitable phosphonated molecules with proven bone therapeutic activity useful according to the invention include but are not limited to pamidronate, alendronate and incadronate as well as others such as πsedronate, olpadronate, etidronate, ibandronate, zolendronate or nendronate, these molecules being well known bisphosphonate bone resorption inhibitors commonly used for the treatment of osteoporosis

The scheme below illustrates the principles of that embodiment

Additional specific examples of bisphosphonate derivatives according to the invention derived from pamidronate and alendronate are shown hereinafter pamidronate

O (HO) 2 P NH,

HO '

From O P(OH) 2

alendronate o

(HO) 2 P

P(OH) 2

From

It is also conceivable according to the present invention to use a pH-sensitive linker that is cleaved only at a predetermined range of pH In one embodiment, the pH- sensitive linker is a base-sensitive linker that is cleaved at a basic pH ranging from about 7 to about 9 According to another embodiment, the linker is an acid-sensitive linker that is cleaved at an acidic pH ranging from about 7 5 to about 4, preferably from about 6 5 and lower It is hypothesized that such an acid-sensitive linker would allow a specific release of the glycopeptide or lipoglycopeptide antibiotic mostly at a site of bacterial infection because it is known that, acidification of tissues commonly occurs during infection (O'Reilley et al , Antimicrobial Agents and Chemotherapy (1992), 36(12) 2693-97)

A covalent bond or a non-cleavable linker may also covalently couple the phosphonated group B to the glycopeptide or lipoglycopeptide A Such bond or linker would be selected such that it would not be cleaved or would be cleaved mainly by the bacteria present at the actual site of infection It is hypothesized that for such compounds the phosphonated group would remain tethered to the glycopeptide or lipoglycopeptide antibiotic and the whole compound would gradually be released from the bone and absorbed by the bacteria, thereby exerting its antibacterial effect Of course, other types of linkers could be selected and synthesized by those skilled in the art For instance the linker may also contain an in vivo hydrolysable phosphonated group having an affinity to bones as disclosed by Ilex Oncology Research in WO 04/026315 The linker may also contain an active group (e g a releasable group stimulating bone formation or decreasing bone resorption) These and other suitable linkers are encompassed by the present invention

In addition to those compounds described hereinbefore and in the

Exemplification section, additional compounds having the formula [ B~ L~ h α A according to the invention include, but are not limited to, those having the following formulae:

10

0

wherein R is C a H b where a is an integer ≤ 20 and b is a non-null integer <2a+1

Further, the present invention covers the compounds of Formula I and of

Formula II, as well as pharmaceutically acceptable salts, esters and prodrugs thereof Examples of pharmaceutically acceptable salts include, but are not limited to, sulfates, pyrosulfates, bisulfates, sulfites, bisulfites, phosphates, monohydrogenphosphates, dihydrogenphosphates, metaphosphates, pyrophosphates, chlorides, bromides, iodides, acetates, propionates, decanoates, caprylates, acrylates, formates, isobutyrates, caproates, heptanoates, propiolates, oxalates, malonates, succinates, suberates, sebacates, fumarates, maleates, butyne-1 ,4-dιoates, hexyne-1 ,6-dιoates, benzoates, chlorobenzoates, methylbenzoates, dinitrobenzoates, hydroxy benzoates, methoxybenzoates, phthalates, sulfonates, xylenesulfonates, phenylacetates, phenylpropionates, phenylbutyrates, citrates, lactates, gamma-hydroxybutyrates, glycolates, tartrates, methanesulfonates, propanesulfonates, naphthalene-1 -sulfonates, naphthalene-2- sulfonates, and mandelates

If the inventive compound is a base, the desired salt may be prepared by any suitable method known to the art, including treatment of the free base with an inorganic acid, such as hydrochloric acid, hydrobromic acid, sulfuric acid, nitric acid, phosphoric acid, and the like, or with an organic acid, such as acetic acid, maleic acid, succinic acid, mandelic acid, fumaric acid, malonic acid, pyruvic acid, oxalic acid, glycolic acid, salicylic acid, pyranosidyl acids such as glucuronic acid and galacturonic acid, alpha- hydroxy acids such as citric acid and tartaric acid, amino acids such as aspartic acid and glutamic acid, aromatic acids such as benzoic acid and cinnamic acid, sulfonic acids such as p-toluenesulfonic acid or ethanesulfonic acid, or the like

If the inventive compound is an acid, the desired salt may be prepared by any suitable method known to the art, including treatment of the free acid with an inorganic

or organic base, such as an amine (primary, secondary, or tertiary), an alkali metal or alkaline earth metal hydroxide, or the like Illustrative examples of suitable salts include organic salts derived from amino acids such as glycine and arginine, ammonia, primary, secondary and tertiary amines, and cyclic amines such as piperidine, morpholine and piperazine, and inorganic salts derived from sodium, calcium, potassium, magnesium, manganese, iron, copper, zinc, aluminum, and lithium

In the case of compounds, salts, or solvates that are solids, it is understood by those skilled in the art that the inventive compounds, salts, and solvates may exist in different crystal forms, all of which are intended to be within the scope of the present invention

The inventive compounds may exist as single stereoisomers, racemates and/or mixtures of enantiomers and/or diastereomers All such single stereoisomers, racemates and mixtures thereof are intended to be within the scope of the present invention Preferably, the inventive compounds are used in optically pure form It is conceivable that the compounds of the Formula I and/or of Formula Il be administered in the form of a prodrug which is broken down in the human or animal body to give a compound of the Formula I or of Formula Il Examples of prodrugs include in vivo hydrolysable esters of a compound of the Formula I and/or of Formula Il An in vivo hydrolysable ester of a compound of the Formula I and/or of Formula

Il containing carboxy or hydroxy group is, for example, a pharmaceutically-acceptable ester which is hydrolyzed in the human or animal body to produce the parent acid or alcohol Suitable pharmaceutically-acceptable esters for carboxy include (1- 6C)alkoxymethyl esters for example methoxymethyl, (1-6C)alkanoyloxy methyl esters for example pivaloyloxymethyl, phthalidyl esters, (3-8C)cycloalkoxycarbonyloxy(1- 6C)alkyl esters for example 1-cyclohexylcarbonyloxyethyl, 1 ,3-dιoxolen-2-onylmethyl esters for example 5-methyl-1 ,3-dιoxolen-2-onylmethyl, and (1- 6C)alkoxycarbonyloxyethyl esters for example 1 -methoxycarbonyloxyethyl and may be formed at any carboxy group in the compounds of this invention An in vivo hydrolysable ester of a compound of the Formula I and/or of Formula

Il containing a hydroxy group includes inorganic esters such as phosphate esters and alpha-acyloxyalkyl ethers and related compounds which as a result of in vivo hydrolysis of the ester break down to give the parent hydroxy group Examples of alpha- acyloxyalkyl ethers include acetoxymethoxy and 2,2-dιmethylpropιonyloxymethoxy A selection of in vivo hydrolysable ester forming groups for hydroxy include alkanoyl, benzoyl, phenylacetyl and substituted benzoyl and phenylacetyl, alkoxycarbonyl (to

give alkyl carbonate esters), dialkylcarbamoyl and λ/-(dιalkylamιnoethyl)-λ/- alkylcarbamoyl (to give carbamates), dialkylaminoacetyl and carboxyacetyl D) Methods of preparation

The inventive compounds, and their salts, solvates, crystal forms, active metabolites, and prodrugs, may be prepared by employing the techniques available in the art using starting materials that are readily available Certain novel and exemplary methods of preparing the inventive compounds are described in the Exemplification section Such methods are within the scope of this invention

E) Antimicrobial compositions and methods of treatment

A related aspect of the invention concerns the use of compounds of the invention as an active ingredient in a therapeutic or anti-bacterial composition for treatment or prevention purposes

Pharmaceutical compositions

The compounds of the present invention may be formulated as pharmaceutically acceptable compositions

The present invention provides for pharmaceutical compositions comprising a compound of the present invention (e g , those compounds of Formula (I) and (II)) in combination with a pharmaceutically acceptable carrier or excipient Preferably, the compound of the present invention is a therapeutically effective amount of the compound Such carriers include, but are not limited to saline, buffered saline, dextrose, water, glycerol, ethanol, and combinations thereof Acceptable methods of preparing suitable pharmaceutical forms of the pharmaceutical compositions according to the invention are known to those skilled in the art For example, pharmaceutical preparations may be prepared following conventional techniques of the pharmaceutical chemist involving steps such as mixing, granulating, and compressing when necessary for tablet forms, or mixing, filling, and dissolving the ingredients as appropriate, to give the desired products for various routes of administration

The compounds and compositions of the invention are conceived to have a broad spectrum of activity, including antibiotic resistant strains, against both Gram- positive (e g Staphylococcus aureus, Staphylococcus epidermis, Streptococcus pyogenes, Enterococcus faecalis) and Gram-negative bacteria (e g E coll, Chlamydia pneumoniae, Enterobacter sp , H influenza, K pneumoniae, Legionella pneumoniae,

P aeruginosa)

Pharmaceutical compositions and a second therapeutic agent

A wide range of second therapeutic agents, such as antibiotics, can be used in combination with the compounds, compositions and methods of the present invention Antibiotics used as second therapeutic agents may act by interfering with cell wall synthesis, plasma membrane integrity, nucleic acid synthesis, ribosomal function, folate synthesis, etc A non-limiting list of useful antibiotics with which the compounds and compositions might be combined includes Rifamycms, sulfonamides, beta- lactams, tetracyclines, chloramphenicol, aminoglycosides, macrohdes, glycopeptides, streptogramins, quinolones, fluoroquinolones, oxazolidmones and lipopeptides In particular, tetracycline, tetracycline derived antibacterial agents, glycylcycline, glycylcycline derived antibacterial agents, minocycline, minocycline derived antibacterial agents, oxazolidinone antibacterial agents, aminoglycoside antibacterial agents, quinolone antibacterial agents, vancomycin, vancomycin derived antibacterial agents, teicoplanin, teicoplanin derived antibacterial agents, eremomycin, eremomycin derived antibacterial agents, chloroeremomycin, chloroeremomycin derived antibacterial agents, daptomycin, daptomycin derived antibacterial agents, πfamycin and πfamycin derived antibacterial agents are preferred

Methods for inhibiting bacterial growth

According to a related aspect, the present invention concerns methods of inhibiting bacterial growth, and more particularly growth of Gram-positive bacteria The method comprises contacting the bacteria for the purpose of such inhibition with an effective amount of a phosphonated glycopeptide or lipoglycopeptide compound or composition according to the invention (or a pharmaceutically acceptable prodrug, salt, active metabolite, or solvate thereof) For example, one can inhibit cell wall biosynthesis in a Gram-positive bacterium by contacting such a bacterium with a compound of the invention The contacting may be carried out in vitro (in biochemical and/or cellular assays), in vivo in a non-human animal, in vivo in mammals, including humans and/or ex v/vo (e g for sterilization purposes)

The activity of the inventive compounds as inhibitors of cell-wall biosynthesis may be measured by any of the methods available to those skilled in the art, including in vivo and in vitro assays Some examples of suitable assays have been described for measurement of binding to cell-wall fragments (Chu et al Journal of Organic Chemistry (1992), 57 3524-3525 Cooper et al, Chemical Communications (1997), 1625-1626),

binding to whole cell walls (Cegelski et al Journal of Molecular Biology (2006), 357, 1253-1262), inhibition of enzymatic processes leading to cell wall components (Branstrom et a/ FEMS Microbiology Letters (2000), 191 187-190 Leimkuhler et al Journal of the American Chemical Society (2005), 127 3250 - 3251) and inhibition of cell wall biosynthesis at the cellular level (Higgms et al , Antimicrobial Agents and Chemotherapy (2005), 49 1127-1134)

A related aspect of the invention concerns the use of a compound of the invention as an active ingredient in a pharmaceutical, therapeutic or anti-bacterial composition for treatment purposes As defined above, "treating" or "treatgment" means at least the mitigation of a disease condition associated with a bacterial infection in a subject, including mammals such as a human, that is alleviated by a reduction of growth, replication, and/or propagation of any bacterium, such as Gram- positive organisms, and includes curing, healing, inhibiting, relieving from, improving and/or alleviating, in whole or in part, the disease condition The pharmaceutical compositions may be administered in any effective, convenient manner including, for instance, administration by topical, parenteral, oral, anal, intravaginal, intravenous, intraperitoneal, intramuscular, intraocular, subcutaneous, intranasal, intrabronchial, or intradermal routes among others

In therapy or as a prophylactic, the compound(s) of the invention and/or pharmaceutically acceptable prodrugs, salts, active metabolites and solvates may be administered to an individual as an injectable composition, for example as a sterile aqueous dispersion, preferably isotonic Alternatively the composition may be formulated for topical application for example in the form of ointments, creams, lotions, eye ointments, eye drops, ear drops, mouthwash, impregnated dressings and sutures and aerosols, and may contain appropriate conventional additives, including, for example, preservatives, solvents to assist drug penetration, and emollients in ointments and creams Such topical formulations may also contain compatible conventional carriers, for example cream or ointment bases, and ethanol or oleyl alcohol for lotions Such carriers may constitute from about 1 % to about 98% by weight of the formulation, more usually they will constitute up to about 80% by weight of the formulation

Alternative means for systemic administration include transmucosal and transdermal administration using penetrants such as bile salts or fusidic acids or other detergents In addition, if a compound of the present invention can be formulated in an enteric or an encapsulated formulation, oral administration may also be possible

Administration of these compounds may also be topical and/or localized, in the form of salves, pastes, gels, and the like

While the treatment can be administered in a systemic manner through the means described above, it may also be administered in a localized manner For example, the treatment may be administered directly to a bone, such as through an injection into a bone The treatment may also be administered in other localized manners, such as application to a wound through a topical composition or directly into a subcutaneous or other form of wound

The active compound(s) and its pharmaceutically acceptable prodrugs, salts, metabolites and solvates may be also administered to an individual as part of a bone substitute or bone-repair compound such as bone cements or fillers (e g Skelite™, Millenium Biologies, Kingston, ON, Canada) and calcium or hydroxyapatite beads

A dose of the pharmaceutical composition contains at least a pharmaceutically- or therapeutically-effective amount of the active compound (i e , a compound of Formula (I), of Formula (II) and/or a pharmaceutically acceptable prodrug, salt, active metabolite, or solvate thereof), and is preferably made up of one or more pharmaceutical dosage units The selected dose may be administered to a mammal, for example, a human patient, in need of treatment A "therapeutically effective amount" is intended to mean that amount of a compound of Formula (I) and/or of Formula (II) (and/or a pharmaceutically acceptable prodrug, salt, active metabolite, or solvate thereof) that confers a therapeutic effect on the subject treated The therapeutic effect may be objective (ι e measurable by some test or marker (e g lower bacterial count)) or subjective (ι e the subject gives an indication of or feels an effect)

The amount that will correspond to a "therapeutically effective amount" will vary depending upon factors such as the particular compound, the route of administration, excipient usage, the disease condition and the severity thereof, the identity of the mammal in need thereof, and the possibility of co-usage with other agents for treating a disease Nevertheless the therapeutically effective amount can be readily determined by one of skill in the art For administration to mammals, and particularly humans, it is expected that the daily dosage level of the active compound will be from 0 1 mg/kg to 200 mg/kg, typically around 1 -5 mg/kg The physician in any event will determine the actual dosage that will be most suitable for an individual and will vary with the age, weight and response of the particular individual The above dosages are exemplary of the average case There can, of course, be individual instances where higher or lower dosage ranges are merited, and such are within the scope of this invention

The invention provides a method of treating a subject in need of treatment wherein a phosphonated glycopeptide or lipoglycopeptide antimicrobial molecule having high affinity to osseous tissues is administered to the subject Preferably, the phosphonated group is coupled to the glycopeptide or lipoglycopeptide antimicrobial

molecule through a cleavable linker Preferably the subject is a mammal, such as a human The method of treatment may also be applied in a veterinary aspect, to animals such as farm animals including horses, cattle, sheep, and goats, and pets such as dogs, cats and birds Although the invention is preferably directed to the prevention and/or treatment of bone-related infections, the invention encompasses therapeutic and prophylactic methods against other diseases caused by or related to bacterial infection, including but not limited to otitis, conjunctivitis, pneumonia, bacteremia, sinusitis, pleural emphysema and endocarditis, low grade infections in the vicinity of calcifications of atherosclerotic vessels, and meningitis In such methods, an effective therapeutic or prophylactic amount of an antibacterial compound and/or composition as defined hereinbefore, is administered to a mammal (preferably a human) in an amount sufficient to provide a therapeutic effect and thereby prevent or treat the infection of the mammal Exact amounts can be routinely determined by one skilled in the art and will vary depending on several factors, such as the particular bacterial strain involved and the particular antibacterial compound used

Prophylaxis and prevention

An additional use that is particularly contemplated for the compounds invention is for prophylaxis and prevention purposes Indeed, many orthopedic surgeons consider that humans with prosthetic joints should be considered for antibiotic prophylaxis before a treatment that could produce a bacteremia Deep infection is a serious complication sometimes leading to loss of the prosthetic joint and is accompanied by significant morbidity and mortality The compounds and compositions of the invention may therefore be used as a replacement for prophylactic antibiotics in this situation For instance, the compounds and/or compositions of the invention may be administered by injection to achieve a systemic and/or local effect against relevant bacteria shortly before an invasive medical treatment, such as surgery or insertion of an in-dwelling device (e g joint replacement (hip, knee, shoulder, etc ), bone grafting, fracture repair, dental operation or implant Treatment may be continued after invasive medical treatment, such as post-operatively or during the ιn-body time of the device

In addition, the compound and/or composition may also be administered before the invasive medical treatment to permit the accumulation of the compound into the bone tissues prior to the treatment In each instance, the compound(s) of the invention could be administered once, twice, thrice or more, from 1 , 2, 3, 4, 5, 6, 7 days or more, to 10, 9, 8, 7, 6, 5, 4, 3, 2, or

1 hour or less before surgery for permitting an advisable systemic or local presence of the compounds, and/or accumulation in the bones, preferably in the areas potentially exposed to bacterial contamination during the surgical procedure Even more preferably, the phosphonated compounds of the invention would be administered such that they can reach a local concentration of about 5, 10, 20, 30, 40, 50, 75, 100, 500 or even 1000 fold higher concentration than the concentration that would normally be achieved during the administration of the unmodified parent glycopeptide or lipoglycopeptide antimicrobial molecule, i e a non-phosphonated equivalent The compound(s) may be administered after the invasive medical treatment for a period of time, such as 1 , 2, 3, 4, 5 or 6 days, 1 , 2, 3 or more weeks, or for the entire time in which the device is present in the body

Therefore, the invention provides a method of inducing accumulation of an glycopeptide or lipoglycopeptide antimicrobial molecule in bones of a mammal wherein a phosphonated glycopeptide or lipoglycopeptide antimicrobial molecule having high affinity to osseous tissues is administered to a mammal The phosphonated glycopeptide or lipoglycopeptide antimicrobial molecule binds osseous tissues and accumulates in bones of the mammal in amounts greater than amounts of a non- phosphonated equivalent of the glycopeptide or lipoglycopeptide antimicrobial molecule Preferably, the phosphonated group is coupled to the glycopeptide or lipoglycopeptide antimicrobial molecule through a cleavable linker

The invention further provides a method for prolonging the presence of an glycopeptide or lipoglycopeptide antimicrobial molecule in bones of a mammal wherein a phosphonated glycopeptide or lipoglycopeptide antimicrobial molecule having a high affinity to osseous tissues is administered to a mammal The phosphonated group is coupled to the glycopeptide or lipoglycopeptide antimicrobial molecule through a cleavable linker The phosphonated glycopeptide or lipoglycopeptide antimicrobial molecule binds osseous tissues and accumulates in bones of the mammal, and the linker is cleaved gradually within the bones thereby releasing the glycopeptide or lipoglycopeptide antimicrobial molecule and prolonging the presence of the glycopeptide or lipoglycopeptide antimicrobial molecule in the bones

F) In-dwelling devices and products coated with the phosphonated glycopeptide or lipoglycopeptide antimicrobial molecules of the invention

The invention further encompasses in-dwelling devices coated with the compounds of the invention As used herein, the term "in-dwelling device" refers to surgical implants, orthopedic devices, prosthetic devices and catheters, i e , devices

that are introduced to the body of an individual and remain in position for an extended time Such devices include, but are not limited to, artificial joints and implants, heart valves, pacemakers, vascular grafts, vascular catheters, cerebrospinal fluid shunts, urinary catheters, continuous ambulatory peritoneal dialysis (CAPD) catheters According to one embodiment, the in-dwelling device is bathed in or sprayed with a concentration of about 1 mg/ml to about 10 mg/ml of the compound and/or the composition of the invention, before its insertion in the body

According to another embodiment, the in-dwelling device is made of, or pre- coated with, an osseous-like type of material (e g calcium phosphate, Ca-ion and hydroxyapatite (Yoshinan et al , Biomateπals (2001 ), 22(7) 709-715)) Such material is likely to advantageously improve binding of the compounds of the invention to the indwelling device, either during the coating of the device with the compounds of the invention and/or after their local or systemic administration The in-dwelling devices may also be coated with an osseous material pre-loaded with or containing bound bone-targeting compound(s) according to the invention For the above-mentioned embodiments, hydroxyapatite would be preferred as the osseous material More details on coating methods, uses and advantages of hydroxy apatite-coated prostheses are found in the review by Dumbleton and Manly (The Journal of Bone & Joint Surgery (2004) 86A 2526-40) which is incorporated herein by reference

G) Methods of preparation

The inventive compounds, and their salts, solvates, crystal forms, active metabolites, and prodrugs, may be prepared by employing the techniques available in the art using starting materials that are readily available Certain novel and exemplary methods of preparing the inventive compounds are described in the Exemplification section below Such methods are within the scope of this invention

EXAMPLES

The Examples set forth herein below provide exemplary syntheses of certain representative compounds of the invention Also provided are exemplary methods for assaying the compounds of the invention for their activity as inhibitors of protein synthesis, assays for determining the minimum inhibitory concentration (MIC) of the compounds of the invention against microorganisms, and methods for testing in vivo activity and cytotoxicity

Example 1 : Synthesis of Vancomycin and Oritavancin bisphosphonate conjugates

A) General Experimental Procedures

A 1) preparation of bisphosphonate building blocks

IV

Following protocols described in Bioorg Med Chem (1999), 7 901-919, benzyl substituted bisphosphonate building blocks of the general structures III and V can be obtained by alkylation of the anion of I with 4-substιtuted benzyl bromide Il or bromoacetate IV Nitro compound IMa can be converted to aniline IUb by reduction of the nitro group under hydrogenation conditions, using a catalyst such as PtO 2 Esters like IMc and Va can be converted to the corresponding acids IMd or Vb via ester cleavage For example, ester IMc where R' = t-Bu can be treated with TFA to afford the corresponding acid IMd Under similar conditions, ester Va where X = Of-Bu can be converted to acid Vb

Aryl substituted methylene bisphosphonates of general formula IX can be obtained from the parent benzylic halides Vl in a sequence of two Arbuzov reactions separated by a benzylic halogenation The hydroxyl substituted parent molecule IXa can be obtained by the nucleophilic addition of the alkali metal salt of a dialkyl phosphite to 4-hydroxybenzaldehyde as described in Org Biomol Chem (2004), 21 3162-3166

XIIa R = t-Bu, Me XMb R = H

Diethyl (ethoxyphosphιnyl)methylphosphonate X can be prepared using the procedure described in Synth Comm (2002), 32 2951 -2957 and patent US 5,952,478 (1999) It can be coupled with a 4-substιtuted bromobenzene (Xl) to access acid XIIb, following cleavage of the ester intermediate XIIa

XIVa n = 1 XVa n = 1 , R = H XIII XIVb n = 2 XVb n = 2, R = H

MIb m=1 XIVa n = 1 XVIa m=1 , n = 1 IX m=0 XIVb n = 2 XVIb m=1 , n = 2 XVIc m=0, n=1 XVId m=0, π=2

Amines of the general formula XIII can be prepared from dibenzylamine, diallylamine, or other N-benzyl and N-allyl secondary amines, diethyl phosphite and triethyl orthoformate following a protocol described in Synth. Comm. (1996), 26: 2037- 2043 Acylation of XIII with succinic anhydride XIVa or glutaric anhydride XIVb can provide acids XVa and XVb respectively (J. Drug Targeting (1997), 5: 129-138). In a similar fashion, treatment of the previously described IMb or IX with XιV(a-b) results in the succinamic and glutaramic acids XVI(a-d).

I XVII

Olefin XVII can be prepared from I following a protocol described in J. Org. Chem. (1986), 51 3488-3490.

XXIa n = 3 XXIb n = 4

As described in Phosphorus, Sulfur and Silicon (1998), 132 219-229, alcohols of general structure XιX(c-d) and iodides of general structure XXI can be prepared by alkylation of the anion of I by protected ω-hydroxy bromides of various chain length XVIII After deprotection, alcohols can be converted to the corresponding iodides via treatment with in situ generated triphenylphosphine iodine complex These alcohols XιX(c-d) may additionally be converted to acids of general structure XX by conventional methods of oxidation, such as treatment with pyπdinium dichromate

DMF

IMb XXIIa m = 1 IX XXIIb m = O

XIII XXIII

Bromoacetamides XXII(a-b) and XXIII from the parent amines IMb, IX and XIII can be prepared according to a modification of the procedure described in J Drug Targeting (1995), 3 273-282

XXIa n = 3 XXIVa n = 3 XXIb n = 4 XXIVb n = 4

Thiols XXIV(a-b) can be prepared by alkylation of the anion of I with a protected 3-ιodopropane-1 -thiol following the protocol described in Bioorg Med Chem (1999), 7 901-919 Or they can be prepared from iodides XXI(a-b) and an appropriately chosen reagent able to supply the sulfhydryl group, including reagents such as thiourea followed by hydrolysis and thioacetic acid followed by hydrolysis or reduction

IHb XXV

2

XIII XXVI

Thioglycolamides XXV and XXVI can be made through the condensation of amine functionalized bisphosphonates such as IHb and XIII with activated forms of thioglycolic acid, or with thioglycolic acid itself as described for other amines in J lnd Chem Soc (1997), 74 679-682

XXVIlIa n = 3 XXVIIIb n = 4

Vinyl ketones such as XXVIII(a-b) can be prepared through the condensation of the parent (hydroxyphenyl) vinyl ketone XXVII with iodides XXI(a-b) in the presence of an appropriately chosen base

XXlX XXX X = halogen XXXI X = H

C XXXII X = halogen

Diethyl (ethoxyphosphιnyl)methylphosphonate XXIX can be prepared using the procedure described in Synth Comm (2002), 32 2951-2957 and patent US 5,952,478 (1999) It can be coupled with a halogenated 1 ,3-dιoxolone XXX in the presence of a transition metal catalyst to furnish bisphosphonate XXXI This can be followed by a radical halogenation reaction to provide bisphosphonate XXXII

XXXVa n = 3 XXXVIa n = 3 X = cι XXXVb n = 4 S- XXXVIb n = 4, X = cι

V XXXVIIa n = 3, X = 0R X = 0R

XXXIIIa n = 3 XXXIIIb n = 4

Acids XX(a-b) can be converted to activated esters XXXIII(a-b), where R * is p- nitrophenyl or N-succinimidyl, directly by treatment with a coupling agent and p- nitrophenol or N-hydroxysuccinimide or by conversion to the acid chlorides first lodoalkyl thiocarbamates XXXIV can be prepared according to Lund et al (Synthesis (1990) 1 159-1166) They can be treated with acids XX(a-b) in the presence of a non nucleophilic base to provide acyloxymethyl thiocarbamates XXXV(a-b) These can be converted to the parent chloroformates XXXVI(a-b) by treatment with a chlorinating agent such as sulfuryl chloride, and subsequently to the activated carbonates

XXXVII(a-b) where R * OH is p-nitrophenol or N-hydroxysuccinimide

XVa n = 1 XXXVIIIa n = 1 XVb n = 2 XXXVIIIb n = 2

XVIa m=1 , n = 1 XXXIXa m=1 , n = 1 XVIb m=1 , n = 2 XXXIXb m=1 , n = 2 XVIc m=0, n=1 XXXIXc m=0, n=1 XVId m=0, n=2 XXXIXd m=0, n=2

Similarly, acids XV(a-b) and XVI(a-d) can be converted to activated esters XXXVIII(a-b) and XXXIX(a-d) respectively, where R * is N-succinimidyl or p-nitrophenyl, by treatment with a coupling and either p-nitrophenol or N-hydroxysuccinimide

XXIX XL X = halogen XLI

Diethyl (ethoxyphosphιnyl)methylphosphonate XXIX can be coupled with a halogenated dihydrochromenenone XL in the presence of a transition metal catalyst to furnish bisphosphonate XLI This compound can be hydrolyzed in the presence of a base to give the bisphosphonated dihydrocinnamic acid XLII The carboxylic acid

functionality in this compound can be protected as the ester (compound XLIII, where R p is a benzyl or an allyl group), the phenolic hydroxyl can be acylated (compound XLIV) or phosphorylated (compound XLVII) by treatment with an acid chloride or a phosphoryl chloride in the presence of a mild base, and the carboxylic acid can be deprotected under standard conditions to yield acids XLV and XLVIII These acids can be further activated in the form of their p-nitrophenol or their N-hydroxysuccinimide esters (compounds XLVI and IL, R * is p-nitrophenyl or N-succinmidyl) by treatment with a coupling and either p-nitrophenol or N-hydroxysuccinimide

The bisphosphonate building blocks described in this section are in the form of their phosphonic esters, R being Me, Et, /-Pr, allyl or Bn, or as the free bisphosphonic acids and/or free bisphosphonate salts

A-2) Synthesis of vancomycin and oritavancin bisphosphonate conjugates

For the purposes of this discussion, the glycopeptides will be schematically represented, with only the relevant functional groups shown Thus vancomycin will be represent as

Whereby the letters correlate the functional groups on vancomycin and its schematic representation Similarly, oritavancin will be represented as

LIa n = 3

XXXVII(a-b) Lib n = 4

Oπtavancin bisphosphonate conjugates involving amide linkages such as L(a- b), Ll(a-b), Lll(a-b) and Llll(a-d) can be prepared by the treatment of Oπtavancin with bisphosphonated acids XX(a-b), XV(a-b) and XιV(a-d) in the presence of a suitably selected coupling agent, or by treatment with N-hydroxysuccinimide esters XXXIII(a-b), XXXVII(a-b), XXXVIII(a-b) and XXXIX(a-d) with or without the use of a non- nucleophilic base

LVa n = 3

XXXVII(a-b) LVb π = 4

Similarly vancomycin bisphosphonate conjugates involving amide linkages, such as LιV(a-b), LV(a-b), LVI(a-b) and LVII(a-d) can be obtained by the treatment of vancomycin with the same bisphosphonated acids XX(a-b), XV(a-b) and XιV(a-d) in the presence of a suitably selected coupling agent, or the same N-hydroxysuccinimide esters XXXIII(a-b), XXXVII(a-b), XXXVIII(a-b) and XXXIX(a-d) with or without the use of a non-nuclophihc base

LVIII X = CI 1 Br, I, OTs Vb n = 1 LlXa n = 1 OMs, OTf XXa n = 2 LIXb n = 2 XXb n = 3 LIXc n = 3

A spacer can also be introduced between the glycopeptide and the bisphosphonated moiety. Thus treatment of an activated ester of 4-substituted butyric acids, such as LVIII, with bisphosphonated acids Vb and XX(a-b) in the presence of a non-nucleophilic base results in activated esters LιX(a-c). These can react with oritavancin or vancomycin to give bisphosphonated glycopeptides LX(a-c) and LXI(a- c) with or without the use of a non-nuclophilic base.

Treatments of oπtavancin and vancomycin with either bisphosphonated hydrocinnamic acids XLV and XLVIII in the presence of a coupling agent or with either of their activated ester XLVI and IL preferably in the presence of a non-nucleophilic base result in bisphosphonated glycopeptides LXII and LXIII respectively for oritavancin and LXIV and LXV respectively for vancomycin

XXVIII(a-b)

Condenstation of oritavancin and vancomycin with bisphosphonated enones XXVIII(a-b) preferably in the presence of a non-nucleophilic base results in glycopeptides LXVI(a-b) and LXVII(a-b) respectively conjugated to the bisphosphonate moiety through a β-amino ketone linker.

Condenstation of oritavancin and vancomycin with bisphosphonated halomethyl dioxolone XXXII in the presence of a non-nucleophilic base results in bisphosphonated glycopeptides LXVIII and LXIX respectively.

Oritavancin can be protected as the carbamate LXX (Rp is t-butyl, a fluorenylmethyl, a benzylic or an allylic group) by treatment with the corresponding chloroformate, pyrocarbonate or N-hydroxysuccinimide carbonate in the presence of a base. This carbamate can be condensated with bromoacetamides XXII(a-b) and XXIII to furnish glycolamides LXXI(a-b) and LXXII respectively. These bisphosphonated glycopeptides can then be deprotected under standard conditions to provide LXXIII(a-b) and LXXIV respectively.

XXIII

Vancomycin can be likewise protected as the biscarbamate LXXV (Rp is t-butyl, a fluorenylmethyl, a benzylic or an allylic group) by treatment with the corresponding

chloroformate, pyrocarbonate or N-hydroxysuccinimide carbonate in the presence of a base This carbamate can be similarly converted the bisphosphonated glycopeptides LXXVIII(a-b) and LXXIX after condensation with bromoacetamides XXII(a-b) and XXIII to give glycolamides LXXVI(a-b) and LXXVII respectively and their subsequent deprotection

For all bisphosphonated glycopeptide conjugates, deprotection of the phosphonate esters to provide the corresponding phosphonic acids is undertaken according to the nature of R If R = Me, Et or /-Pr, the ester is treated with TMSBr in a solvent such as CH 2 CI 2 , with or without an amine or a heteroaromatic nitrogen containing base, and the resulting silylated intermediate is hydrolysed with water When R = AIIyI, the esters are hydrolyzed by treatment with a strong nucleophile in the presence of Pd(II) catalysts When R = Bn, the esters are cleaved by hydrogenolysis using a catalyst such as Pd on carbon in a solvent such as ethanol

The other protecting groups used can be put on and removed using the coventional methods described in the literature, for instance as reviewed in "Protective Groups in Organic Synthesis", Greene, T W and Wuts, P M G , Wiley-lnterscience, New York, 1999

B) Detailed Experimental Procedures

Scheme 1. Preparation of 3-[(tetraethyl bisphosphonomethyl)carbamoyl]propanoic acid (3a) and 4-[(tetraethyl bisphosphonomethyOcarbamoylJbutanoic acid (3b).

O

Ph-^ O OEt Ph- \ ^P(OEt) 2

NH 2 (EtO) 2 P-H + [-OEt - ^- N-<

Ph- OEt Ph- ' P(OEt) 2 ό

1

Tetraethyl λ/,λ/-Dibenzyl-1-aminomethylenebisphosphonate (1). Compound 1 was prepared according to a modified protocol derived from Synth Comm 1996, 26, 2037-2043 Tπethyl orthoformate (8 89 g, 60 mmol), diethyl phosphite (16 57 g, 120

mmol) and dibenzyl amine (11 80 g, 60 mmol) were combined in a 100 mL round bottom flask fitted with a distillation head The reaction was heated to a temperature of 180-195 0 C for 1 h under Ar When EtOH evolution was complete, the reaction mixture was cooled to room temperature, diluted with CHCI 3 (300 mL), washed with aqueous NaOH (2M, 3 x 60 mL) and brine (2 x 75 mL), then dried over MgSO 4 After evaporation, a crude yield of 25 2 g (87%) was obtained A 4 95 g portion of the crude oil was purified by chromatography (ethyl acetate hexane methanol 14 4 1 ) to yield pure 1 (2 36 g, 41 %) 1 H NMR (400 MHz, CDCI 3 ) δ 1 32 (dt, J=I 0, 7 0, 12H), 3 55 (t, J=25 0, 1 H), 3 95-4 25 (m, 12H), 7 20-7 45 (m, 10H)

Tetraethyl 1-aminomethylenebisphosphonate(2). Compound 1 (2 00 g, 4 14 mmol) was dissolved in EtOH (40 mL) To this solution was added palladium on carbon (10%, 1 5 g) and cyclohexene (2 5 mL, 24 7 mmol) The reaction mixture was refluxed under argon for 15 hours, filtered through celite and evaporated to give 2 as a slightly impure pale yellow oil (1 50 g, 119%), which was used directly in the next step without further purification 1 H NMR (400 MHz, CDCI 3 ) δ 1 35 (t, J=I 0, 12H), 3 58 (t, J=20 3, 1 H), 3 65-3 90 (br s, 2H), 4 20-4 28 (m, 8H)

3-[(tetraethyl bisphosphonomethyl)carbamoyl]propanoic acid (3a). Compound 3a was prepared as described in J Drug Targeting, 1997, 5, 129-138 It was obtained as an oil which slowly solidified, in 57% crude yield from 2 The crude product could be purified by chromatography (10% AcOH / EtOAc) to give a white solid 1 H NMR (400 MHz, CDCI 3 ) δ 1 31 (t, J=I 0, 6H), 1 33 (t, J=I 1 , 6H), 2 61-2 73 (m, 4H), 4 05-4 28 (m, 8H), 5 07 (td, J=21 6, J=9 8, 1 H), 7 90 (d, J=9 4, 1 H)

4-[(tetraethyl bisphosphonomethyl)carbamoyl]butanoic acid (3b). Compound 3b was prepared as described in J Drug Targeting, 1997, 5, 129-138 It was obtained as an orange oil, in 85% crude yield from 2 The crude product could be purified by chromatography (10% AcOH / EtOAc) to give a white solid 1 H NMR (400 MHz, CDCI 3 ) δ 1 30 (t, J=I 0, 6H), 1 34 (t, J=I 0, 6H), 1 92-2 02 (m, 2H), 2 38-2 44 (m, 2H), 2 54 (t, J=I 3, 1 H), 4 04-4 28 (m, 8H), 5 16 (td, J=22 1 , J=10 0, 1 H), 8 45 (d, J=10 2, 1 H)

Scheme 2. Preparation of vancomycin bisphosphonate conjugate 6

4 1 ) TMSBr DMF , - 5 R = Et

2 6-lutιdιne (

2) HF pyridine DMF V -* 6 R = H

Vancomycin bisphosphonated conjugate 5. To a solution of 3a (47 mg 1 16x10 4 mol) and PyBroP (70 mg, 1 35x10 4 mol) ιn 1 mL of DMF was added DIPEA (30 μl_, 1 73x10 4 mol) The mixture was stirred for 15 mm before it was added to a solution of Vanomycin hydrochloride (4, 100 mg, 6 73x10 5 mol) and DIPEA (13 μl_, 7 47x10 5 mol) in 1 5 mL of DMF The flask containing 3a was further rinsed with 1 mL of DMF and the rinse was added to the reaction mixture After stirring under Argon at room temperature for an overnight, the mixture was concentrated in vacuo and the residue was subjected to C18 silica gel chromatography on a Biotage™ flash chromatography system using a gradient of 0-30% acetonitrile in 0 005%TFA in H 2 O as the eluent to furnish 5 (45 mg, 2 45x10 5 mol, 36% yield) as a white solid ESI-MS (M-H) " calculated for C 79 H 1 OoCI 2 N 10 O 32 P 2 1833, found 1833 0

Vancomycin bisphosphonated conjugate 6. Bromotπmethylsilane (243 μl_,

1 84 mmol) was added drop-wise to a stirring solution of 5 (45 mg, 2 45x10 5 mol) and

2 6-lutιdιne (430 μL, 3 70 mmol) in dry DMF (2 5 mL) which was cooled in an ice-bath The resulting mixture was left to come to room temperature on its own and stir there for a total of 20 hr The solvent was removed under reduced pressure to complete dryness The residue was taken up in DMF (2 5 mL) and pyridine (200 μL, 2 48 mmoles) and HF pyridine (70% HF, 32 μL 1 23 mmoles) were added The mixture was stirred for 2h at room temperature before being concentrated in vacuo to dryness The crude residue was purified by C18 silica gel chromatography on a Biotage™ flash chromatography system using a gradient of 0-30% acetonitrile in 0 005%TFA in H 2 O as

the eluent to furnish 6 as a white solid (11 3 mg, 6 56x10 "6 mol, 27% yield) ESI-MS (M-H)- calculated for C 7 IH 84 CI 2 N 10 O 32 P 2 1721 , found 1721 0

Scheme 3. Preparation of oritavancin bisphosphonated conjugate 10

TMSBr 2 6 lutldine then (~ 9 R = Et

HF pyridine

N-succinimidyl 4-[(tetraethyl bisphosphonomethyl)carbamoyl]butanoate (7). To a mixture of acid 3b (1 14g, 2 73 mmoles) and λ/-hydroxysuccιnιmιde (346 mg, 3 00 mmoles) in acetonitrile (14 mL) at O 0 C was added DCC (619 mg, 3 00 mmol)

The mixture was stirred for 1 h at 0 0 C and refrigerated overnight The precipitate was removed by filtration and the filtrate was concentrated then purified by flash chromatography using a gradient of 0-5% MeOH in CH 2 CI 2 to provide compound 7 as a white solid (1 06 g, 76% yield) 1 H NMR (400 MHz, CDCI 3 ) δ 1 33 (2χt, J=I 0 Hz, 12H), 2 12 (quint, J=7 0 Hz, 2H), 2 43 (t, J=6 9 Hz, 2H), 2 69 (t, J= 7 0 Hz, 2H), 2 85 (s, 4H), 4 14-4 27 (m, 8H), 5 05 (dt, J=21 8, 10 1 Hz, 1 H), 6 48 (d, J=9 9 Hz, 1 H)

Oritavancin bisphosphonate conjugate 9. To a suspension of oritavancin bisphosphoπc acid salt (8, 572 mg, 0 29 mmol) in dιoxane/H 2 O (1 1 , 1OmL) was added sodium bicarbonate (48 mg, 0 58 mmol) and the mixture was stirred until complete dissolution of oritavancin Succinimidyl ester 7 (296 mg, 0 58 mmol) was added and the mixture was stirred at room temperature for 18h, after which additional sodium bicarbonate (24 mg, 0 29 mmol) and succinimidyl ester 7 (70 mg, 0 15 mmol) were added Stirring was continued for another 2Oh and the reaction mixture was

concentrated and lyophilized Crude product was purified by C18 silica gel chromatography on a Biotage™ flash chromatography system using 15-100% MeOH in Et 3 N/H 3 PO 4 buffer (0 2% Et 3 N/H 2 O + H 3 PO 4 , pH=3) Pure fractions were combined, concentrated and lyophilized and resulting product was desalted on a second Biotage™ C18 column using 15-80% MeCN in H 2 O (both containing 0 1 %TFA) After concentration and lyophilization of the combined fractions, the dι-TFA salt of the bisphosphonate conjugate 9 was obtained as a white solid (185 mg, 26%) ESI-MS (M+H) calculated for C 100 H 124 CI 3 N 11 O 34 P 2 2192, found 2192 2

Oritavancin bisphosphonated conjugate 10. To a solution of the oπtavancin bisphosphonate conjugate 9 (185 mg, 0 076 mmol) and 2,6-lutιdιne (621 μl_, 5 35 mmol) in DMF (4mL) cooled at -78°C was added TMSBr (454 μl_, 3 44 mmol) The reaction mixture was stirred for 1 h at -78°C, then 23 h at room temperature It was then concentrated to dryness under high vacuum, redissolved in DMF (4 mL) then treated with pyridine (495 μl_, 6 11 mmol) and HF-pyπdιne (77 μl_, 3 06 mmol) After stirring for 1h at room temperature the mixture was concentrated to dryness under high vacuum The crude product was purified by C18 silica gel chromatography on a Biotage™ flash chromatography system using 15-80% MeCN in H 2 O (both containing 0 05% NH 4 OH) Pure fractions were combined, concentrated and lyophilized to provide the tri-ammonium salt of oritavancin bisphosphonate conjugate 10 as a white solid (20 mg, 12%) LCMS 99 3% (254 nm), 97 9% (220 nm), 99 2% (290 nm) ESI- MS (M+H) calculated for C 92 H 108 CI 3 N 11 O 34 P 2 2080, found 2080 5

Scheme 4. Preparation of bisphosphonated bromoacetamides 11 and 14

Tetraethyl 1-(N-2-bromoacetylamino)methylenebisphosphonate (11). A solution of bromoacetyl bromide (O 35 mL, 4 0 mmol) in CH 2 CI 2 (1 mL) was added dropwise to a stirred, cooled (ice-bath) solution of 2 (1 1 g, 3 6 mmol) and pyridine (0 59 mL, 7 3 mmol) in CH 2 CI 2 (10 mL) After stirring at the same temperature for 4 hours, the reaction was quenched by the addition of water The product was extracted with CH 2 CI 2 and the combined organics were washed with 10% aqueous HCI, brine, dried over sodium sulfate and concentrated at reduced pressure The crude yellow oil was purified by silica gel column chromatography (0% to 3% MeOH in CH 2 CI 2 ) resulting in 11 as a colourless solid (0 58 g, 37%) 1 H NMR (400 MHz, CDCI 3 ) δ 1 35 (t, J=I 2, 12H), 3 92 (s, 2H), 4 12-4 28 (m, 8H), 4 92 (dt, J=10 2, 21 7, 1 H), 6 91 (bd, J=10 0, 1 H)

Tetramethyl 1-(4-nitrobenzyl)methylenebisphosphonate (12): Sodium hydride (1 02 g, 25 4 mmol) was added in portions to a stirring solution of tetramethyl methylenebisphosphonate in DMF (40 mL) After 30 mm a solution of 4- mtrobenzylbromide (5 00 g, 23 1 mmol) in THF (5 mL) was added and the resulting mixture was stirred at room temperature for 4 5 hr The reaction was quenched by the addition of saturated aqueous NH 4 CI (20 mL) After the addition of water (100 mL) the product was extracted with EtOAc and the combined organics were washed with brine, dried over MgSO 4 , filtered and concentrated at reduced pressure The crude product was purified by silica gel chromatography (0% to 10% MeOH in EtOAc) resulting in 12 as a colorless solid (2 55 g, 30%) 1 H NMR (400 MHz, CDCI 3 ) δ 2 65 (tt, J=6 5, 23 8,

1 H), 3 31 (dt, J=6 5, 16 5, 2H), 3 73 (d, J=I 0, 6H), 3 75 (d, J=I 0, 6H), 7 42 (d, J=8 9, 2H), 8 15 (d, J=8 9, 2H)

Tetramethyl 1-(4-aminobenzyl)methylenebisphosphonate (13). A mixture of 12 (1 01 g, 2 75 mmol) and PtO 2 (0 035 g, 0 15 mmol) in EtOH (40 mL, 95%) was shaken in a PARR apparatus under 55 p s i of H 2 for 14 hr The catalyst was removed by filtration through glass fiber filter paper and the solvent was removed under reduced pressure to give 13 as a pale yellow solid (0 959 g, 103%) that was used without purification 1 H NMR (400 MHz, CDCI 3 ) δ 2 62 (tt, J=6 3, 23 9, 1 H), 3 12 (dt, J=6 3, 16 2, 2H), 3 70 (d, J=1 9, 6H), 3 73 (d, J=1 9, 6H), 6 61 (d, J=8 5, 2H), 7 04 (d, J=8 5, 2H)

Tetramethyl 1-(4-bromoacetamidobenzyl)methylenebisphosphonate (14):

A solution of 13 (0 959 g, 2 87 mmol) and pyridine (349 μL, 4 31 mmol) in CH 2 CI 2 was cooled in an ice-bath while stirring A solution of bromoacetylbromide (250 μL, 2 87 mmol) in CH 2 CI 2 (5 mL) was added drop-wise and the resulting mixture was stirred for 4 h at that temperature The reaction was quenched by the addition of water and the product was extracted with CH 2 CI 2 The combined organic layers were washed with brine, dried over sodium sulfate, filtered, and concentrated under reduced pressure The crude yellow solid was purified by silica gel chromatography resulting in 14 as a colorless solid (0 897 g, 67%) 1 H NMR (400 MHz, CDCI 3 ) δ 2 65 (tt, J=6 2, 24 4, 1 H), 3 22 (dt, J=6 2, 17 4, 2H), 3 72 (d, J=3 7, 6H), 3 75 (d, J=3 7, 6H), 4 01 (s, 2H), 7 26 (d, J=8 6, 2H), 7 47 (d, J=8 6, 2H), 8 15 (bs, 1H) 31 P (162 MHz, CDCI 3 ) δ 26 33 (s, 2P)

Scheme 5. Preparation of vancomycin bisphosphonate conjugate 18

Fmoc protected Vancomycin 15. To a suspension of vancomycin hydrochloride (4, 5 g, 3 36 mmol) in 200 mL of 1 1 Dioxane H 2 O was added NaHCO 3 (850 mg, 10 1 mmol) and the mixture was stirred to dissolution To this solution was added 9-fluorenylmethyl N-succinimidyl carbonate (4 5 g, 13 3 mmoles) and the resulting mixture was stirred at room temperature for 24h Acetone (1 L) was added, and the precipitated product was filtered, rinsed with acetone and dried in vacuo to furnish 15 (6 8 g, 3 58 mmoles, quantitative yield) as a white solid which was used without further purification

Vancomycin bisphosphonate conjugate 16. To a mixture of Fmoc protected vancomycin 15 (150 mg, 7 92x10 5 mol) and sodium bicarbonate (20 mg, 2 38x10 4 mol) in dry DMF (5 mL), cooled in an ice bath, was added bromoacetamide 11 (50 mg, 1 18x10 4 mol) The mixture was left to come to room temperature and was stirred for a total of 5 days The volatiles were removed in vacuo and the residue was subjected to C18 silica gel chromatography on a Biotage™ flash chromatography system using a gradient of 0-70% acetonitπle in 0 005%TFA in H 2 O as the eluent to furnish 16 (100 mg, 4 47x10 5 mol, 56% yield) as a white solid ESI-MS (M-H) " calculated for C 107 H I 19 CI 2 N 10 O 35 P 2 2236, found 2235 0

Vancomycin bisphosphonate conjugate 17. To a solution of protected vancomycin derivative 16 (100 mg, 4 47x10 "5 mol) in dry DMF (2 mL) was added piperidine (170 μL, 1 18x10 4 mol) The mixture was stirred at room temperature for 4 h and it was concentrated in vacuo The residue was subjected to C18 silica gel chromatography on a Biotage™ flash chromatography system using a gradient of 0- 30% methanol in 0 05%TFA in H 2 O as the eluent to furnish 17 (66 mg, 3 68x10 5 mol, 82% yield) as a white solid ESI-MS (M-H) calculated for C 77 H 98 CI 2 N 10 O 3I P 2 1791 , found 1791 1

Vancomycin bisphosphonated conjugate 18. Bromotrimethylsilane (243 μL,

1 84 mmol) was added drop-wise to a stirring solution of 17 (66 mg, 3 68x10 5 mol) and 2,6-lutιdιne (430 μL, 3 70 mmol) in dry DMF (2 5 mL) which was cooled in an ice-bath The resulting mixture was left to come to room temperature on its own and stir there for a total of 20 hr The solvent was removed under reduced pressure to complete dryness The residue was taken up in DMF (2 5 mL) and pyridine (300 μL, 3 72 mmoles) and HF pyridine (70% HF, 48 μL, 1 85 mmoles) were added The mixture was stirred for 2h at room temperature before being concentrated in vacuo to complete dryness The crude residue was purified by C18 silica gel chromatography on a Biotage™ flash chromatography system using a gradient of 0-30% acetonitrile in 0 005%TFA in H 2 O as the eluent to furnish 18 as a white solid (27 mg, 1 61x10 5 mol, 44% yield) ESI-MS (M-H) calculated for C 69 H 82 CI 2 N 10 O 3I P 2 1679, found 1679 1

Scheme 6. Preparation of vancomycin bisphosphonate conjugate 21

R = Me — H π R r\ = - H

2) HF pyridine, DMF

Vancomycin bisphosphonate conjugate 19. To a mixture of Fmoc protected vancomycin 15 (1 5 g, 7 92x10 4 mol) and sodium bicarbonate (200 mg, 2 38 mmol) in dry DMF (50 ml_), cooled in an ice bath, was added bromoacetamide 14 (814 mg, 1 78 mmol) The mixture was left to come to room temperature and was stirred for a total of 5 days The volatiles were removed in vacuo and the residue was subjected twice to C18 silica gel chromatography on a Biotage™ flash chromatography system using a gradient of 0-100% acetonitrile in 0 005%TFA in H 2 O as the eluent in the first one and a gradient of 0-60% acetonitrile in 0 005%TFA in H 2 O as the eluent in the second one to furnish 19 (1 1 g, 4 84x10 "4 mol, 61% yield) as a white solid ESI-MS (M-H) calculated for C 1 IOHI I 6 CI 2 N 1 OO 35 P 2 2270, found 2269 9

Vancomycin bisphosphonate conjugate 20. To a solution of protected vancomycin derivative 19 (1 1 g, 4 84x10 "4 mol) in dry DMF (30 mL) was added piperidine (1 9 mL, 19 3 mmol) The mixture was stirred at room temperature for 4 h and it was concentrated in vacuo The residue was subjected to C18 silica gel chromatography on a Biotage™ flash chromatography system using a gradient of 0- 50% acetoniutrile in 0 005%TFA in H 2 O as the eluent to furnish 20 (840 mg, 4 6x10 4 mol, 95% yield) as a white solid ESI-MS (M-H) calculated for C 80 H 96 CI 2 N 10 O 31 P 2 1825, found 1824 9

Vancomycin bisphosphonated conjugate 21. Bromotπmethylsilane (3 mL, 1 84 mmol) was added drop-wise to a stirring solution of 20 (840 mg, 4 60x10 "4 mol) and 2,6-lutιdιne (5 3 mL, 45 72 mmol) in dry DMF (30 mL) which was cooled in an ice- bath The resulting mixture was left to come to room temperature on its own and stir there for a total of 20 hr The solvent was removed under reduced pressure to complete dryness The residue was taken up in DMF (30 mL) and pyridine (3 7 mL, 45 81 mmoles) and HF pyridine (70% HF, 600 μL, 23 1 mmoles) were added The mixture was stirred for 2h at room temperature before being concentrated in vacuo to complete dryness The crude residue was purified by C18 silica gel chromatography on a Biotage™ flash chromatography system using a gradient of 0-20% acetonitrile in 0 005%TFA in H 2 O as the eluent to furnish 21 as a white solid (340 mg, 1 92x10 4 mol, 42% yield) ESI-MS (M-H) calculated for C 76 H 88 CI 2 N 10 O 31 P 2 1769, found 1769 1

Scheme 7. Preparation of bisphosphonated bromoacetamides 25 and 28.

AlIyI alcohol Pyridine/ Ph

(AI 26 Y = NO 2 28

Zn NH 4 CI MeCN

27 Y = NH 2

Tetraallyl methylenebisphosphonate (23). To a suspension of tetrachloromethylene bisphosphonate (22, 6 33 g, 25 3 mmol) in toluene (25 mL) at O 0 C was added a mixture of allyl alcohol ( 6 91 mL, 101 mmol) and pyridine (8 20 mL, 101 mmol) with a dropping funnel over 25 mm After the addition, the reaction mixture was warmed to room temperature and stirred for 20 h The precipitate was removed by filtration and the solids were washed with toluene The filtrate was concentrated and purified by flash chromatography on silica gel using 50% acetone/hexanes as eluent Tetraallyl methylenebisphosphonate 23 was obtained as a clear yellowish oil (5 87 g, 17 5 mmol, 69%) 1 H NMR (400 MHz, CDCI 3 ) δ 1 33 (t, J=21 1 Hz, 2H), 4 60-4 63 (m, 8H), 5 23-5 27 (m, 4H), 5 35-5 40 (m, 4H), 5 90-6 00 (m, 4H)

Tetraallyl 1-aminomethylenebisphosphonate (24). To a solution of bisphosphonate 23 (2 0 g, 5 95 mmol) in DMF (6 mL) was added NaH (60% dispersion in mineral oil, 254 mg, 6 35 mmol) portionwise The solution was stirred for 45 mm at room temperature and added to a solution of O-(dιphenylphophιnyl)hydroxylamιne (1 35 g, 5 77 mmol) in THF (40 mL), cooled in a dry ice/acetone bath The resulting mixture was stirred for 10 mm at the same temperature then 18 h at room temperature CH 2 CI 2 (40 mL) was added, the solids were removed by filtration and washed with several portions of CH 2 CI 2 The combined filtrates were concentrated in vacuo and purified by flash chromatography on silica gel using a gradient of 0-5% MeOH/EtOAc to provide aminobisphosphonate 24 as a clear yellow oil (1 24 g, 3 53 mmol, 59%) 1 H NMR (400 MHz, CDCI 3 ) δ 1 69 (bs, 2H), 3 52 (t, J=20 8 Hz, 1 H), 4 64-4 67 (m, 8H), 5 24-5 27 (m, 4H), 5 36-5 42 (m, 4H), 5 92-6 02 (m, 4H)

Tetraallyl 1-bromoacetamidomethylenebisphosphonate (25). To a solution of aminomethylenebisphosphonate 24 (1 24 g, 3 53 mmol) in CH 2 CI 2 (44 mL) at 0°C was added pyridine (428 μl_, 5 29 mmol) followed by bromoacetylbromide (307 μl_, 3 53 mmol) The reaction mixture was stirred at 0 0 C for 30 mm and poured into a saturated aqueous NH 4 CI solution The layers were separated and the aqueous layer was extracted with CH 2 CI 2 (2χ) The combined organic layers were dried over MgSO 4 , filtered and concentrated The crude product was purified by flash chromatography on silica gel using EtOAc as eluant to provide bromide 25 as a clear colorless oil (1 24 g, 2 63mmol, 74%) 1 H NMR (400 MHz, CDCI 3 ) δ 3 91 (s, 2H), 4 61 -4 67 (m, 8H), 5 04

(dt, J=21 7, 10 1 Hz, 1 H), 5 24-5 28 (m, 4H), 5 35-5 40 (m, 4H), 5 88-5 99 (m, 4H), 7 07 (d, J=10 5 Hz, 1 H)

Tetraallyl 1-(4-nitrobenzyl)methylenebisphosphonate (26). To a solution of bisphosphonate 23 (8 02 g, 23 9 mmol) in DMF (24 mL) was added NaH (60% dispersion in mineral oil, 954 mg, 23 9 mmol) portionwise and the solution was stirred for 1 5 h at room temperature A solution of p-nitrobenzyl bromide (7 75 g, 35 9 mmol) in THF (32 mL) was added and the mixture was stirred for 3 h at room temperature It was then poured into a mixture of saturated aqueous NH 4 CI solution and water and extracted with CH 2 CI 2 (3χ) The combined organic layers were washed with saturated brine once, dried over MgSO 4 and concentrated in vacuo The crude product was purified by flash chromatography on silica gel using a gradient of 0-50% EtOAc in CH 2 CI 2 as eluant to provide nitrobenzyl bisphosphonate 26 as a clear yellow oil (4 42 g, 9 38 mmol, 39%) 1 H NMR (400 MHz, CDCI 3 ) δ 2 73 (tt, J=23 8, 6 5 Hz, 1 H), 3 37 (dt, J=16 4, 6 5 Hz, 2H), 4 51-4 61 (m, 8H), 5 21-5 25 (m, 4H), 5 29-5 35 (m, 4H), 5 83-5 93 (m, 4H), 7 44 (d, J=8 8 Hz, 2H), 8 14 (d, J=8 8 Hz, 2H)

Tetraallyl 1-(4-aminobenzyl)methylenebisphosphonate (27) To a solution of nitroarene 26 (460 mg, 0 98 mmol) in MeOH (9 mL) was added saturated aqueous NH 4 CI solution (3 mL) and zinc powder (319 mg, 4 88 mmol) 15 drops of aqueous 1 N HCI were added and the reaction was stirred at room temperature for 18 h EtOAc and saturated NaHCO 3 aqueous solution were added and the mixture was filtered through celite The filtrate was transferred into an extraction funnel and the layers were separated The aqueous layer was extracted with EtOAc (2χ) The combined organic layers were washed with saturated NaCI solution once, dried over MgSO 4 , filtered and concentrated The crude product was purified by flash chromatography on silica gel

using a gradient of 0-5% MeOH in CH 2 CI 2 as eluant to provide aminobenzyl bisphosphonate 27 as a clear yellow oil (374mg, 86%) 1 H NMR (400 MHz, CDCI 3 ) δ 2 69 (tt, J=24 0, 6 1 Hz, 1 H), 3 18 (dt, J=16 7, 6 2 Hz, 2H), 4 46-4 60 (m, 8H), 5 19- 5 22 (m, 4H), 5 29-5 35 (m, 4H), 5 84-5 94 (m, 4H), 6 60 (d, J=8 6 Hz, 2H), 7 06 (d, J=8 4 Hz, 2H)

Tetraallyl 1-(4-bromoacetamidobenzyl)methylenebisphosphonate (28). To a solution of aminobenzyl bisphosphonate 27 (180 mg, 0 41 mmol) in CH 2 CI 2 (5 mL) at 0 0 C was added pyridine (49 μL, 0 61 mmol) followed by bromoacetylbromide (35 μl_, 0 41 mmol) The reaction mixture was stirred at O 0 C for 30 mm and at room temperature for another 2 5 h The reaction mixture was diluted with CH 2 CI 2 , washed with aqueous 1 N HCI solution, saturated NaHCO 3 solution, and saturated NaCI solution, then dried over MgSO 4 , filtered and concentrated The crude product was purified by flash chromatography on silica gel using a gradient of 0-10% MeOH in CH 2 CI 2 as eluant to provide bromide 28 as a clear yellow oil (175 mg, 3 11x10 4 mol, 76%) 1 H NMR (400 MHz, CDCI 3 ) δ 2 72 (tt, J=23 9, 6 2 Hz, 1 H), 3 26 (dt, J= 16 3, 6 1 Hz, 2H), 4 02 (s, 2H), 4 48-4 62 (m, 8H), 5 20-5 24 (m, 4H), 5 29-5 35 (m, 4H), 5 83- 5 94 (m, 4H), 7 25-7 28 (m, 2H), 7 42-7 46 (m, 2H), 8 11 (bs, 1 H)

Scheme 8. Preparation of oritavancin bisphosphonate conjugate 31

Di-W-Alloc oritavancin (29). To oπtavancin bisphosphoric acid salt (8, 2 O g, 1 01 mmol) in DMF (80 mL) and H 2 O (30 mL) was added sodium bicarbonate (676 mg, 8 04 mmol) and the mixture was stirred for 30 mm AIIyI N-succinimidyl carbonate (641 mg, 3 22 mmol) was added and the mixture was stirred at room temperature for 48h A portion of /iBuOH (ca 10-15 mL) was added and the mixture was concentrated under vaccuum to one quarter of its initial volume H 2 O was added and the pH was adjusted to 4 5 by adding aqueous 1 N HCI The precipitate was filtered and washed with H 2 O and dried under vacuum to provide dι-λ/-Alloc oritavancin 29 as a white solid (1 85 g, 8 9x10 4 mol, 93%) which was used without further purification ESI-MS (M+H) calculated for C 94 HiO 5 CI 3 N 10 O 3 O 1961 , found 1961 4

Oritavancin bisphosphonate conjugate 30 To a solution of dι-λ/-Alloc oritavancin 29 (527 mg, 0 25 mmol) in DMF (5 mL) was added sodium bicarbonate (43 mg, 0 51 mmol) and the mixture was stirred for 10 mm after which bromide 25 (120 mg, 0 25 mmol) was added After stirring for 72 h at room temperature, the reaction mixture was concentrated to dryness under vacuum LCMS analysis of crude reaction mixture showed a mixture of starting material 29, desired product 30 and monodeallylated product in a 0 9 1 1 2 ratio C18 silica gel chromatography on a Biotage™ flash chromatography system using 60-100% MeOH in Et 3 NZH 3 PO 4 buffer (0 2% Et 3 N/H 2 O + H 3 PO 4 , pH=3) followed by a second column using 30-100% MeCN in H 2 O (both containing 0 1 % TFA) achieved partial separation of the constituents Fractions containing product 30 and monodeallylated product were combined, concentrated and lyophilized to provide a mixture of product 30 and monodeallylated product (245 mg, 1 1 ratio) which was carried through the next step without further purification ESI-MS (M+H) calculated for C 109 H 128 CI 3 N 11 O 37 P 2 2353, found 2353 2, calculated for C 106 H 124 CI 3 N 11 O 37 P 2 2313, found 2313 4

Oritavancin bisphosphonate conjugate 31. To a solution of the mixture of oritavancin bisphosphonate conjugate 30 and its monodeallylated analog (245 mg) in DMF (2 mL) was added morpholine (1 8 mL, 20 8 mmol) and Pd(PPh 3 ) 4 (24 mg, 0 02 mmol) The mixture was stirred for 4 h at room temperature and concentrated to dryness under vacuum The crude material was purified by C18 silica gel chromatography on a Biotage™ flash chromatography system using 15-80% MeCN in H 2 O (both containing 0 05% NH 4 OH) followed by a second column using 15-80% MeCN in H 2 O (both containing 0 1 % TFA) Pure fractions were combined, concentrated and lyophilized to provide the tπ-TFA salt of oritavancin bisphosphonate

con j ugate 31 as a white solid (58 mg, 2 45x10 5 mol, 24%) ESI-MS (M+H) calculated for C 89 H 104 CI 3 N 11 O 33 P 2 2023, found 2023 8

Scheme 9. Preparation of oritavancin bisphosphonate conjugate 33.

32 R 1 = AIIyI R 2 = AIlOC

Pd(PPh 3 J 4 DMF morpholine

33 R 1 = H R 2 = H

Oritavancin bisphosphonate conjugate 32 To a solution of dι-λ/-Alloc oritavancin 29 (1 g, 0 51 mmol) in DMF (10 mL) was added sodium bicarbonate (86 mg, 1 02 mmol) and the mixture was stirred for 10 mm after which bromide 28 (287 mg, 0 51 mmol) was added After stirring for 48 h at room temperature, additional amounts of sodium bicarbonate (43 mg, 0 51 mmol) and bromide 28 (144 mg, 0 25 mmol) were added and stirring was continued for 48 h, after which the reaction mixture was concentrated to dryness under vacuum The crude reaction mixture was purified by C18 silica gel chromatography on a Biotage™ flash chromatography system using 15- 100% MeOH in Et 3 N/H 3 PO 4 buffer (0 2% Et 3 N/H 2 O + H 3 PO 4 , pH=3) Pure fractions were combined, concentrated and lyophilized and desalted by a second column using 15-100% MeCN in H 2 O (both containing 0 1 % TFA), providing oritavancin bisphosphonate conjugate 32 as a white solid (871 mg, 0 34 mmol, 67%) ESI-MS (M+H) calculated for C 116 H 134 CI 3 N 11 O 37 P 2 2443, found 2443 2

Oritavancin bisphosphonate conjugate 33. To a solution oritavancin bisphosphonate conjugate 31 (871 mg, 0 34 mmol) in DMF (6 mL) was added morpholine (5 9 mL, 68 1 mmol) and Pd(PPh 3 ) 4 (79 mg, 0 068 mmol) The mixture was

stirred for 4 h at room temperature and concentrated to dryness under vacuum The crude material was purified by C18 silica gel chromatography on a Biotage™ flash chromatography system using 15-80% MeCN in H 2 O (both containing 0 05% NH 4 OH), providing the triammonium salt of oπtavancin bisphosphonate conjugate 33 as a white solid (472 mg, 0 21 mmol, 64%) ESI-MS (M+H) calculated for C 96 H 110 CI 3 N 11 O 33 P 2 2115, found 21 15 2

Scheme 10. Preparation of N-succinimidyl 4,4-bis(diethylphosphono)butyrate (37).

34 X = OTHP -N M e0H

^) IRA 120(H + )

35 X = OH NaOCI 2

Tetraethyl 4-(2-Tetrahydro-2H-pyranyloxy)butylene-1,1 -bisphosphonate (34). To a suspension of NaH (60% suspension in mineral oil, 900 mg, 22 0 mmol) in dry THF (20 mL) was added dropwise tetraethyl methylenebisphosphonate (6 46 g, 22 4 mmol) The resulting clear solution was stirred 15 mm at room temperature, after which 2-(3-bromopropoxy)tetrahydro-2/-/-pyran (5 05 g, 22 6 mmol) was added dropwise The reaction mixture was heated to reflux for 6 h, diluted with CH 2 CI 2 (75 mL) and washed with brine (2 x 50 mL), dried (MgSO 4 ) and evaporated It was used as such in the following step

Tetraethyl 4-hydroxybutylene-1 ,1 -bisphosphonate (35). To a stirred solution of the crude product 34 (max 22 4 mmol) in MeOH (40 mL) was added Amberlite IR- 120 (0 6 g) The reaction mixture was heated to 50 0 C for 4 h, filtered and concentrated

in vacuo The crude product was purified by flash chromatography on silica gel with gradient elution from 5-10% methanol/ethyl acetate to give pure 35 (2 67 g, 34% from tetraethyl methylenebisphosphonate) 1 H NMR (400 MHz, CDCI 3 ) δ 1 34 (t, J= 7 1 Hz, 12H), 1 81 (quint, J= 6 5 Hz, 2H), 1 99-2 13 (m, 2H), 2 37 (tt, J= 24 4, 5 6 Hz, 1 H), 2 51 (t, J=5 9 Hz, 2H), 3 66 (q, J= 5 9 Hz, 2H), 4 13-4 22 (m, 8H)

Tetraethyl 3-carboxypropylene-1 ,1-bisphosphonate (36). To a solution of alcohol 35 (12 7 g, 36 7 mmol) in MeCN (200 mL) and phosphate buffer solution (200 ml_, made from mixing equal volumes of 0 67M Na 2 HPO 4 solution and 0 67M NaH 2 PO 4 solution) at 35 °C was added a catalytic amount of TEMPO (430 mg, 2 75 mmol) The reaction flask, maintained at 35 0 C, was fitted with two addition funnels One was filled with a solution of NaCIO 2 (8 3 g, 91 7 mmol) in 75 mL H 2 O The other one was filled with a solution of household bleach (5 25%, 25 mL) in 250 mL H 2 O About 1/5 of the NaCIO 2 solution was added, followed by about 1/5 of the bleach solution to initiate the reaction The remainder of both solutions was added dropwise, simultaneously, with a rate adjusted so that both additions finished concurrently The reaction mixture was stirred at 35 0 C for 4 h, then at room temperature for 18 h The reaction mixture was diluted with 300 mL H 2 O and the pH of the solution was adjusted to 8 0 by adding 1 M NaOH The resulting solution was cooled to 0 0 C and a cold solution of Na 2 SO 3 (6 1 % wt, 185 mL) was added slowly The mixture was stirred at 0 0 C during 30 mm, after which a portion of Et 2 O was added After stirring vigourously, the mixture was poured into an extraction funnel and the Et 2 O layer was separated and discarded The aqueous layer was acidified to pH 3 4 with cone HCI and extracted (3χ) with CHCI 3 //- PrOH mixture (4 1 ) The combined organic layers were dried over MgSO 4 , filtered and concentrated to dryness, yielding 36 as a pale yellow oil (12 9 g, 98%), which could be used without further purification 1 H-NMR (400 MHz, CDCI 3 ) δ 1 34 (t, J =I 0 Hz, 12H), 2 18-2 28 (m, 2H), 2 60 (tt, J= 23 9, 6 5 Hz, 1 H), 2 69 (t, J= 7 3 Hz, 2H), 4 14-4 23 (m, 8H)

N-succinimidyl 4,4-bis(diethylphosphono)butyrate (37). To a solution of 36

(315 mg, 0 874 mmol) and λ/-hydroxysuccιnιmιde (110 mg, 0 960 mmol) in acetonitrile (3 mL) cooled in an ice-bath was added DCC (198 mg, 0 960 mmol) The resulting mixture was stirred at the same temperature for 1 5 hr and stored at 4 ° C overnight The precipitate was filtered off and the filtrate was concentrated to give 37 as a yellow liquid (360 mg, 93%) that was used without purification 1 H NMR (400 MHz, CDCI 3 ) δ

1 34 (t, J =7 0 Hz, 12H), 2 26-2 40 (m, 2H), 2 45 (tt, J= 23 4, 6 1 Hz, 1 H), 2 83 (bs, 4H), 3 01 (t, J=7 3, 2H), 4 15-4 24 (m, 8H) 31 P NMR (162 MHz, CDCI 3 ) δ 23 44 (s, 2P)

Scheme 11. Preparation of oritavancin bisphosphonate conjugate 39.

37 NaHCO 3 H 2 O Dioxane

TMSBr 2 6 lutidine then ( HF pyridine 1 C \ *

Oritavancin bisphosphonate conjugate 38. A suspension of oritavancin diphosphate salt (8, 1 38 g, 0 692 mmol) and NaHCO 3 (1 16 mg, 1 39 mmol) in dioxane/water (1 1 , 20 mL) was stirred at room temperature for 15 mm, at which time oritavancin had fully dissolved 21 (412 mg, 0 901 mmol) was added to the flask and the resulting solution was stirred at room temperature for 16 hr Et 2 O/acetone (1 1 , 20 mL) was added and the resulting precipitate was collected by filtration The crude product was purified by C18 silica gel chromatography on a Biotage™ flash chromatography system using a gradient of 20-80% acetonitπle in 0 05% Formic acid in H 2 O as the eluent to furnish the colourless solid 38 (370 mg, 22%) as the dι-formate salt ESI-MS (M+H) calculated for C 98 H 12 ICI 3 N 10 O 33 P 2 2136, found 2136

Oritavancin bisphosphonate conjugate 39. A solution of 38 (370 mg, 0 173 mmol) and 2,6-lutιdιne (1 41 mL, 12 1 mmol) in DMF (5 mL) was cooled to -70 0 C (2- propanol/dry ice) followed by the drop-wise addition of TMSBr (915 μL, 6 93 mmol) The resulting slurry was stirred for 30 mm at the same temperature then for 38 hr at room temperature The solution was concentrated to dryness in vacuo without heating and the solid was resuspended in DMF (5 mL) followed by the addition of pyridine (1 40 mL, 17 3 mmol) and HF/pyndine (217 μL, 8 66 mmol) The resulting solution was

stirred for 1 hr at room temperature then concentrated to dryness The crude material was dissolved in water/CH 3 CN (1 1 , 3 mL), the pH was adjusted to 3 then the product was partially purified by C18 silica gel chromatography on a Biotage™ flash chromatography system using 15-80% acetonitrile in 0 05% TFA in H 2 O as the eluent The fractions containing the product were lyophilized and the semi-purified material was dissolved in water/CH 3 CN (1 1 , 3 mL), the pH was adjusted to 8 and further subjected to C18 silica gel chromatography on a Biotage™ flash chromatography system using 15-80% acetonitrile in 0 05% NH 4 OH in H 2 O resulting in the dι- ammonium salt of 39 (53 mg, 15%) as a colourless solid ESI-MS (M+H) calculated for C 90 H 105 CI 3 N 10 O 33 P 2 2024, found 2024 7

Scheme 12. Preparation of N-succinimidyl 3,3-bis(diethylphosphono)propanoate (42).

f-Butyl 3,3-bis(diethylphosphono)propanoate (40). To a solution of tetraethyl methylenebisphosphonate (3 0O g, 10 4 mmol) in dry DMF (9 mL) was added NaH (60% suspension in mineral oil, 0 46 g, 11 5 mmol) portionwise The resulting slurry was stirred for 30 mm at room temperature, after which f-butyl bromoacetate (1 7 mL, 11 5 mmol) was quickly added neat The reaction mixture was stirred for 1 h and quenched by adding 2 mL of a saturated solution of NH 4 CI The reaction mixture was evaporated and purified by flash chromatography on silica gel eluting with 5% methanol/ethyl acetate to give pure 40 (2 1 g, 50%) as a clear colourless oil 1 H NMR (400 MHz, CDCI 3 ) δ 1 33 (bt, J=I 0, 12H), 1 46 (s, 9H), 2H), 2 76 (dt, J=16 0, 6 1 , 2H) 3 07 (tt, J=24 0, 6 1 , 1 H), 4 10-4 25 (m, 8H)

3,3-bis(diethylphosphono)propanoic acid (41). Ester 40 (2 1 g, 5 2 mmol) was stirred in TFA (12 mL) for 2 5 mm and concentrated under reduced pressure Crude acid 41 was purified by flash chromatography (gradient elution 100% ethyl acetate - 10% methanol/ ethyl acetate) Acid 41 was obtained as a white solid (1 35 g,

75%) 1 H NMR (400 MHz, CDCI 3 ) δ 1 28-1 39 (m, 12H), 2 86 (dt, >16 1 , 6 3, 2H), 3 12 (tt, J=24 0, 6 3, 1 H), 4 13-4 26 (m, 8H)

N-Succinimidyl 3,3-bis(diethylphosphono)propanoate (42). To a solution of 41 (1 0 g, 2 89 mmol) and λ/-hydroxysuccιnιmιde (366 mg, 3 18 mmol) in acetonitπle (14 mL) cooled in an ice-bath was added DCC (655 mg, 3 18 mmol) The resulting mixture was stirred at the same temperature for 1 h and stored at 4 ° C overnight The precipitate was filtered off and the filtrate was concentrated to give 42 as a white solid (1 29 g, quantitative) that was used without purification 1 H NMR (400 MHz, CDCI 3 ) δ 1 32-1 37 (m, 12H), 2 84 (bs, 4H), 3 03 (tt, J= 23 4, 6 0 Hz, 1 H), 3 17 (dt, J=15 2, 6 0 Hz, 2H), 4 15-4 27 (m, 8H)

Scheme 13. Preparation of oritavancin bisphosphonate conjugate 44.

42 NaHCO 3

H 7 O Dioxane

HF pyridine i f V '

Oritavancin bisphosphonate conjugate 43. To a suspension of oritavancin bisphosphoπc acid salt (8, 1 0 g, 0 50 mmol) in dιoxane/H 2 O (1 1 , 3OmL) was added sodium bicarbonate (84 mg, 1 01 mmol) and the mixture was stirred until complete dissolution of oritavancin Succinimidyl ester 42 (446 mg, 1 01 mmol) was added and the mixture was stirred at room temperature for 24 h, after which additional sodium bicarbonate (42 mg, 0 50 mmol) and succinimidyl ester 42 (223 mg, 0 50 mmol) were added and the stirring was continued for 24 h Additional sodium bicarbonate (42 mg, 0 50 mmol) and succinimidyl ester 42 (223 mg, 0 50 mmol) were added once more and the stirring was continued for 3 d The reaction mixture was concentrated and

lyophilized and the resulting crude product was purified by C18 silica gel chromatography on a Biotage™ flash chromatography system using 15-100% MeOH in Et 3 N/H 3 PO 4 buffer (0 2% Et 3 N/H 2 O + H 3 PO 4 , pH=3) Pure fractions were combined, concentrated and lyophilized and desalted by a second column using 15-80% MeCN in H 2 O (both containing 0 1 % TFA), providing the dι-TFA salt of oritavancin bisphosphonate conjugate 43 as a white solid (276 mg, 12%) ESI-MS (M+H) calculated for C 97 H 119 CI 3 N 10 O 33 P 2 2122, found 2122 2

Oritavancin bisphosphonate conjugate 44. To a solution of the oritavancin bisphosphonate conjugate 43 (274 mg, 0 12 mmol) and 2,6-lutιdιne (975 μl_, 8 4 mmol) in DMF (5mL) cooled at -78°C was added TMSBr (713 μl_, 5 4 mmol) The reaction mixture was stirred for 15 mm at -78 0 C, then 24 h at room temperature It was then concentrated to dryness under high vacuum, redissolved in DMF (5 mL) then treated with pyridine (776 μl_, 9 6 mmol) and HF-pyπdine (120 μl_, 4 8 mmol) After stirring for 1 h at room temperature the mixture was concentrated to dryness under high vacuum The crude product was purified by C18 silica gel chromatography on a Biotage™ flash chromatography system using 15-80% MeCN in H 2 O (both containing 0 1 % TFA) followed by a second column using 15-80% MeCN in H 2 O (both containing 0 05% NH 4 OH) Pure fractions were combined, concentrated and lyophilized to provide the tri-ammonium salt of oritavancin bisphosphonate conjugate 44 as a white solid (34 mg, 14%) ESI-MS (M+H) calculated for C 89 H 103 CI 3 N 10 O 33 P 2 2009, found 2009 6

Scheme 14. Preparation of N-succinimidyl 4-(3,3- bis(diethylphosphono)propanoyloxy) butyrate (48).

O "P(OEt) 2 FKT ^^ P(OEt) 2

N-Succinimidyl 4-bromobutyrate (45). A solution of 4-bromobutyryl chloride (1 0 ml_, 8 69 mmol) in acetonitrile (10 mL) was added dropwise to a solution containing λ/-hydroxysuccιnιmιde (1 0 g, 8 69 mmol) and tπethylamine (1 2 mL, 8 69 mmol) in acetonitrile (10 mL) cooled in an ice bath After stirring for 45 mm at this temperature, the solids were removed by filtration and the filtrate was concentrated under vaccuum The crude product was then dissolved in EtOAc, washed with H 2 O, half saturated NaHCO 3 solution, H 2 O and saturated NaCI solution, dried over Na 2 SO 4 , filtered and concentrated, yielding bromide 45 as a white solid (2 26 g, 99%) that was used without purification 1 H NMR (400 MHz, CDCI 3 ) δ 2 30 (quint, J=6 5 Hz, 2H), 2 83 (t, J=6 4 Hz, 2H), 2 85 (bs, 4H), 3 52 (t, J=6 4 Hz, 2H)

N-Succinimidyl 4-iodobutyrate (46). To bromide 45 (2 26 g, 8 56 mmol) in acetone (43 mL) was added sodium iodide (3 85 g, 25 7 mmol) and the mixture was stirred for 3 h at room temperature, then concentrated under vaccuum The crude product was then dissolved in EtOAc, washed with H 2 O, 5% Na 2 S 2 O 3 solution, H 2 O and saturated NaCI solution, dried over Na 2 SO 4 , filtered and concentrated, yielding iodide 46 as a light yellow solid (2 55 g, 96%) that was used without purification 1 H NMR (400 MHz, CDCI 3 ) δ 2 25 (quint, J=I 0 Hz, 2H), 2 77 (t, J=I 1 Hz, 2H), 2 85 (bs, 4H), 3 29 (t, J=6 7 Hz, 2H)

Potassium 3,3-bis(diethylphosphono)propanoate (47). To a solution of acid 41 (516 mg, 1 49 mmol) in acetonitrile (5 mL) was added an aqueous 1 M solution of KOH (1 64 ml, 1 64 mmol) After stirring for 20 mm, the mixture was concentrated under vaccum and co-evaporated with Et 2 O and CH 2 CI 2 to provide potassium salt 47 as a white foam (550 mg, 96%) 1 H NMR (400 MHz, CDCI 3 ) δ 1 31 (t, J=I 1 Hz, 12H), 2 59 (dt, J=17 7, 5 8 Hz, 2H), 3 17 (tt, J=IA 2, 6 1 , 1 H), 4 13 (2χquιnt, J=I 1 Hz, 8H)

N-Succinimidyl 4-(3,3-bis(diethylphosphono)propanoyloxy)butyrate (48).

A mixture of potassium salt 47 (160 mg, 0 42 mmol) and iodide 46 (129 mg, 0 42 mmol) in DMF (2 mL) was stirred for 3 h, concentrated under vaccuum and purified by flash chromatography on silica gel using 5% MeOH/CH 2 CI 2 as eluent, providing 48 as a pale yellow oil (158 mg, 71 %) 1 H NMR (400 MHz, CDCI 3 ) δ 1 33 (2χt, J=I 1 Hz, 12H), 2 1 1 (quint, J=6 8 Hz, 2H), 2 74 (t, J=I 5 Hz, 2H), 2 81-2 90 (m, 6H), 3 08 (tt, J=IZ 8, 6 4 Hz, 1 H), 4 14-4 23 (m, 10H)

Scheme 15. Preparation of oritavancin bisphosphonate conjugate 50.

TMSBr 2 6 IuuItidine thei 49 R = Et HF py Tiidine "C 50 R = H

Oritavancin bisphosphonate conjugate 49. To a suspension of oritavancin bisphosphoric acid salt (8, 722 mg, 0 36 mmol) in dιoxane/H 2 O (1 1 , 24 mL) was added sodium bicarbonate (61 mg, 0 73 mmol) and the mixture was stirred until complete dissolution of oritavancin Succinimidyl ester 48 (411 mg, 0 73 mmol) was added and the mixture was stirred at room temperature for 48 h, after which the reaction mixture was concentrated and lyophihzed and the resulting crude product was purified by C18 silica gel chromatography on a Biotage™ flash chromatography system using 15-100% MeOH in Et 3 N/H 3 PO 4 buffer (0 2% Et 3 N/H 2 O + H 3 PO 4 , pH=3) Pure fractions were combined, concentrated, lyophihzed and desalted by a second column using 30-100% MeCN in H 2 O (both containing 0 1 % TFA), providing the dι-TFA salt of oritavancin bisphosphonate conjugate 49 as a white solid (451 mg, 51 %) ESI-MS (M+H) calculated for C 1 OiH 125 CI 3 N 10 O 35 P 2 2207, found 2207 4

Oritavancin bisphosphonate conjugate 50. To a solution of the oritavancin bisphosphonate conjugate 49 (450 mg, 0 18 mmol) and 2,6-lutιdιne (1 5 mL, 12 9 mmol) in DMF (1 OmL) cooled in a dry ice/acetone bath was added TMSBr (1 1 mL, 8 31 mmol) The reaction mixture was stirred for 15 mm at the same temperature, then 24 h at room temperature It was then concentrated to dryness under high vacuum, redissolved in DMF (10 mL) then treated with pyridine (1 16 mL, 14 4 mmol) and HF- pyndine (180 μL, 7 20 mmol) After stirring for 1 h at room temperature, the mixture was concentrated to dryness under high vacuum The crude product was purified by C18 silica gel chromatography on a Biotage™ flash chromatography system using 15-

80% MeCN in H 2 O (both containing 0 05% NH 4 OH) Pure fractions were combined, concentrated and lyophihzed to provide the tπ-ammonium salt of oritavancin bisphosphonate conjugate 50 as a white solid (203 mg, 53%) ESI-MS (M+H) calculated for C 93 H 109 CI 3 N 10 O 35 P 2 2096, found 2096 2

Scheme 16. preparation of (5,5-bis(diethylphosphono)pentanoyloxymethyl) N- succinimidyl carbonate (32).

Et) 2 reflux

✓0 (EtO) 2 P'

<Et0)2 λ 1 K2C ° 3 Acetone δ (EtO) 2 P-^- < O

O

4-Bromo-1-butanol (51). To 67 5 mL (832 2 mmol) of refluxing tetrahydrofuran was added 31 mL (274 mmol) of 48% hydrobromic acid dropwise and the yellow solution was allowed to reflux for another 2h After cooled to room temperature, the reaction was carefully neutralized with saturated sodium bicarbonate aqueous solution The resultant mixture was extracted with diethyl ether (3x) and dried over anhydrous sodium sulfate Removal of the solvent afforded the product 51 as a yellow oil (10 7 g, 26%) 1 H NMR (400 MHz, CDCI 3 ) δ 1 69-1 76 (m, 2H), 2 01-1 94 (m, 2H), 3 46 (t, J = 6 6 Hz, 2H), 3 70 (t, J = 6 4 Hz, 2H)

2-(4-Bromobutoxy)-tetrahydro-2H-pyran (52). 3,4-Dιhydro-2/-/-pyran (8 5 mL, 90 96 mmol) was added dropwise to the dichloromethane (20 mL) solution of 51 (10 7 g, 69 93 mmol) and p-toluenesulfonic acid monohydrate (26 5 mg, 0 1372 mmol) The mixture was stirred at room temperature over night After removing the solvent, the residue was purified by flash chromatography on silica gel with 5 1 hexanes/ethyl acetate as the eluent to yield product 52 as a colorless oil (15 3 g, 92%) 1 H NMR (400 MHz, CDCI 3 ) δ 1 48-1 62 (m, 4H), 1 68-1 85 (m, 4H), 1 94-2 02 (m, 2H), 3 40-3 53 (m, 4H), 3 74-3 88 (m, 2H), 4 57-4 59 (m, 1 H)

Tetraethyl 5-(2-Tetrahydro-2H-pyranyloxy)pentylene-1,1-bisphosphonate

(53).To the suspension of sodium hydride (60% in oil, 840 5 mg, 21 0 mmol) in 40 mL of THF was carefully added tetraethyl methylenebisphosphonate (6 16 g, 21 0 mmol) and the resultant pale yellow clear solution was stirred at room temperature for 45 mm Then the bromide 52 (4 97 g, 21 0 mmol) was introduced plus 5 mL of THF rinse The reaction was brought to reflux overnight and allowed to cool to room temperature before being quenched with saturated ammonium chloride aqueous solution Another small amount of water was required to dissolve the solid The mixture was extracted with ethyl acetate (3x), dried over anhydrous sodium sulfate and concentrated in vacuo Flash chromatography on silica gel with 20 1 (v/v) dichloromethane/methanol as the eluent afforded 7 3 g of impure product 53 as a slightly yellow oil The material was used directly in the next step without further purification Selected 1 H NMR (400 MHz, CDCI 3 ) δ 2 28 (tt, J=6 1 , 24 3 Hz, 1 H), 3 37-3 51 (m, 2H), 3 71-3 89 (m, 2H), 4 56-4 58 (m, 1 H)

Tetraethyl 5-hydroxypentylene-1,1-bisphosphonate (54). The crude compound 53 was dissolved in 20 mL of methanol and 74 6 mg (0 386 mmol) of p- toluenesulfonic acid monohydrate was added After overnight stirring at room temperature, the mixture was concentrated and subjected to flash chromatography with gradient elution from 15 1 ethyl acetate/methanol to 8 1 then 6 1 to afford 54 as a colorless oil (3 1 g, 41% over two steps) 1 H NMR (400 MHz, CDCI 3 ) δ 1 24-1 36 (m,

12H), 1 55-1 72 (m, 4H), 1 89-2 03 (m, 2H), 2 16 (bs, 1 H), 2 29 (tt, J=6 1 , 24 3 Hz, 1 H), 3 66 (bs, 2H), 4 11-4 22 (m, 8H)

Tetraethyl 5-carboxypentylene-1 ,1-bisphosphonate (55). To a mixture of alcohol 54 (475 mg, 1 32 mmol), TEMPO (15 mg, 0 095 mmol), MeCN (6 mL) and sodium phosphate buffer (6 mL, 0 67 M, pH = 6 7) heated to 35 °C were added

dropwise a sodium chlorite solution (300 mg in 2 mL of water) and dilute bleach (0 75 ml_ of solution of 1 mL of commercial bleach in 19 mL of water) simultaneously from separate syringes The mixture turned from yellow to red After 5 h, reaction was complete by TLC and 1 H NMR and was cooled to room temperature Water (30 mL) was added and the pH was adjusted to about 9 with the addition of 3 mL of 1 N NaOH The reaction was quenched by pouring into a cold Na 2 SO 3 solution (500 mg in 10 mL of water) and maintained below 20 0 C After 30 mm stirring at the same temperature, 30 mL of diethyl ether was used to extract the mixture and the organic phase was discarded The pH of the aqueous phase was readjusted to between 3-4 by adding 5 mL of 1 N HCI and the mixture was extracted with dichloromethane (3x) The combined extracts were dried over sodium sulfate and concentrated to afford the acid 55 quantitatively, which could be used in the following steps without further purification 1 H NMR (400 MHz, CDCI 3 ) δ 1 34 (t, J =7 0, 12H), 1 86-2 06 (m, 4H), 2 33 (tt, J = 24 2, 5 9, 1 H), 2 36 (t, J = 7 3, 2H), 4 14-4 22 (8H)

S-Ethyl O-(5,5-bis(diethylphosphono)pentanoyloxy)methyl carbonothioate (56): Acid 55 (606 mg, 1 619 mmol), tetrabutylammonium hydrogensulfate (552 mg, 1 626 mmol) and sodium bicarbonate (274 2 mg, 3 264 mmol) were added to the mixture of 4 mL of water and 4 mL of dichloromethane After the evolution of gas stopped, 310 8 mg (1 263 mmol) of S-ethyl 0-ιodomethyl carbonothionate (synthesized according to Folkmann, M , Lund, F J Synthesis, 1990, 1159-1 166) in 1 mL of dichloromethane was added and the mixture was stirred for 2 h The organic phase was separated, washed with water (1x) and dried over sodium sulfate After filtration and concentration, the residue was stirred in ether for 10 mm The solid was removed and the filtrate was concentrated and subjected to flash chromatography on silica gel with elution in 20 1 (v/v) dichloromethane/methanol to afford compound 56 (519 2 mg, 83 %) as a colorless oil 1 HNMR (400 MHz, CDCI 3 ) δ 1 33 (t, J = 7 3, 3H), 1 35 (t, J = 7 7, 12H), 1 86-2 04 (m, 4H), 2 28 (tt, J = 23 8, 6 2, 1 H), 2 40 (t, J = 7 3, 2H), 2 90 (q, J = 7 3, 2H), 4 14-4 22 (m, 8H), 5 80 (s, 2H)

(Carbonochloridoyloxy)methyl 5,5-bis(diethylphosphono)pentanoate (57). Compound 56 neat (519 2 mg, 1 054 mmol) was cooled in an ice/water bath and sulfuryl chloride (128 μL, 1 58 mmol) was carefully added The reaction mixture was allowed to warm to room temperature and was stirred overnight After removal of the excess sulfuryl chloride in vacuo, the crude acid chloride 57 was used directly in the next step without further purification 1 HNMR (400 MHz, CDCI 3 ) δ 1 35 (t, J = 7 0,

12H), 1 86-2 06 (m, 4H), 2 28 (tt, J = 23 8, 6 2, 1 H), 2 45 (t, J = 7 0, 2H), 4 14-4 24 (m, 8H), 5 82 (s, 2H) 31 P NMR (162 MHz, CDCI 3 ) δ 24 34 (s, 2P)

(5,5-bis(diethylphosphono)pentanoyloxymethyl) N-succinimidyl carbonate (58). A solution of /V-hydroxysuccinimide (281 mg, 2 44 mmol) and triethylamine (340μl, 2 44 mmol) in acetonitrile (10 mL) was cooled in an ice-bath A solution of 57 (1 14g, 2 44 mmol) in acetonitrile (2 mL) was added drop-wise over 5 mm After stirring a further 15 mm at the same temperature the solution was filtered and the filtrate was concentrated under reduced pressure The residue was resuspended in EtOAc and washed with water, 5% aqueous Na 2 S 2 O 3 , water and brine then dried over Na 2 SO 4 and concentrated to give 58 (925 mg, 70%) as a yellow liquid that was used without purification 1 H NMR (400 MHz, CDCI 3 ) 5 1 34 (t, J = 7 1 , 12H), 1 87-2 00 (m, 4H), 2 29 (tt, J = 24 1 , 5 4, 1 H), 2 45 (t, J = 7 0, 2H), 2 85 (s, 4H), 4 14-4 24 (m, 8H), 5 86 (s, 2H) 31 P NMR (162 MHz, CDCI 3 ) δ 24 42 (s, 2P)

Scheme 17. Preparation of oritavancin bisphosphonate conjugate 60.

58 NaHCO 3 H 7 O Dioxane

HF pyridine " VC 60 R = H

Oritavancin bisphosphonate conjugate 59. A suspension of oritavancin diphosphate (8, 1 00 g, 0 503 mmol) and NaHCO 3 (84 mg, 1 0 mmol) in dιoxane/H 2 O (1/1 , 10 mL) was stirred at room temperature for 15 mm The homogenous solution was then cooled in an ice-bath and a solution of 58 (341 mg, 0 604 mmol) in acetonitrile (1 mL) was added and the resulting solution was stirred at 0°C for 4 hr Acetone/ether/acetonitπle (1/1/2, 20 mL) were added and the precipitate was collected

by filtration The crude product was purified by C18 silica gel chromatography on a Biotage™ flash chromatography system using 20-80% acetonitrile in 0 05% TFA in H 2 O to furnish the colourless solid 59 (360 mg, 32%) as the dι-TFA salt ESI-MS (M+H) calculated for C 1 OiH 12 SCI 3 N 10 O 36 P 2 2223, found 2223 6

Oritavancin bisphosphonate conjugate 60. A solution of 59 (360 mg, 0 162 mmol) and 2,6-lutιdιne (1 32 ml_, 11 3 mmol) in DMF (5 mL) was cooled to -70°C (2- propanol/dry ice) followed by the drop-wise addition of TMSBr (1 07 mL, 8 10 mmol) The resulting slurry was stirred for 30 mm at the same temperature then for 40 hr at room temperature The solution was concentrated to dryness without heating and the solid was resuspended in DMF (5 mL) followed by the addition of pyridine (1 31 mL, 16 2 mmol) and HF/pyndine (203 μL, 8 10 mmol) The resulting solution was stirred for 1 hr at room temperature then concentrated to dryness The crude material was purified by C18 silica gel chromatography on a Biotage™ flash chromatography system (15% to 80% acetonitrile in 0 05% TFA in H 2 O) resulting in the dι-TFA salt of 60 (160 mg, 44%) as a colourless solid ESI MS (M-H) calculated for C 93 H 109 CI 3 N 10 O 36 P 2 2110, found 2110 7 (M-H) 31 P NMR (162 MHz, CDCI 3 ) δ 21 61 (s, 2P)

Scheme 18. preparation of (4,4-bis(diethylphosphono)butanoyloxymethyl) N- succinimidyl carbonate (63).

(EtO) 2 P.

OH (EtO) 2 P-^^^A. /x λ

Xf "O"

(EtO) 2 P^ 0 (n Bu) 4 NHSO 4 NaHCO 3 (EtO) 2 P^ 0 H 2 O CH 2 CI 2

37 61 X = SEt

SO 2 CI; C 62 X = Cl

S-Ethyl O-(4,4-bis(diethylphosphono)butanoyloxy)methyl carbonothioate (61).

A mixture of 37 (2 00 g, 5 55 mmol), tetrabutylammonium hydrogensulfate (1 88 g, 5 55 mmol) and sodium bicarbonate (933 mg, 11 1 mmol) in H 2 0/dιcholoromethane (1/1 , 60 mL) was stirred at room temperature for 1 5 hr A solution of S-ethyl 0-iodomethyl carbonothionate (1 23 g, 5 00 mmol, Synthesis, 1990, 1159-1166) in dichloromethane (10 mL) was added in 1 mL portions over 1 hr and the resulting mixture was stirred for a further 2 hr The mixture was diluted with dichloromethane (20 mL) and the organic phase was separated, washed with water and brine then dried over sodium sulfate, filtered and concentrated The residue was resuspended in ether, stirred for 1 hr, filtered and concentrated to give 61 (2 03 g, 76%) as a yellow liquid that was used without purification 1 H NMR (400 MHz, CDCI 3 ) δ 1 31 (t, J = 7 5, 3H), 1 32 (t, J = 7 3, 12H), 2 15-2 29 (m, 2H), 2 47 (tt, J = 23 9, 6 7, 1 H), 2 73 (t, J = 7 5, 2H), 2 87 (q, J = 7 3, 2H), 4 12-4 21 (m, 8H), 5 78 (s, 2H) 31 P NMR (162 MHz, CDCI 3 ) δ 23 95 (s, 2P)

(Carbonochloridoyloxy)methyl 4,4-bis(diethylphosphono)butanoate (62). Sulfuryl chloride (687 μL, 8 49 mmol) was added drop-wise to 61 (2 03 g, 4 24 mmol) The neat reaction was stirred at room temperature for 2 5 hr The excess sulfuryl chloride was removed under reduced pressure resulting in chloroformate 62 (1 87 g, 97%) that was used without purification 1 H NMR (400 MHz, CDCI 3 ) δ 1 35 (t, J = 7 1 , 12H), 2 17-2 36 (m, 2H), 2 53 (tt, J = 23 8, 6 5, 1 H), 2 80 (t, J = 7 5, 2H), 4 15-4 24 (m, 8H), 5 82 (s, 2H) 31 P NMR (162 MHz, CDCI 3 ) δ 23 72 (s, 2P)

(4,4-bis(diethylphosphono)butanoyloxymethyl) N-succinimidyl carbonate (63).

A solution of N-hydroxysuccinimide (89 mg, 0 77 mmol) and tπethylamine (108 μL, 0 770 mmol) were dissolved in acetonitrile (2 mL) was cooled in an ice-bath followed by the addition of 62 (350 mg, 0 770 mmol) in acetonitrile (1 mL) over 10 mm The resulting solution was stirred for a further 15 mm at the same temperature then the solution was filtered and the filtrate was concentrated under reduced pressure The residue was resuspended in EtOAc and washed with 5% aqueous Na 2 S 2 O 3 , 5% aqueous NaHCO 3 , water and brine then filtered and dried over Na 2 SO 4 and concentrated to give 63 (410 mg, 100%) as a yellow liquid that was used without purification 1 H NMR (400 MHz, CDCI 3 ) δ 1 34 (t, J = 7 2, 12H), 2 18-2 32 (m, 2H), 2 50 (tt, J = 24 2, 7 6, 1 H), 2 80 (t, J = 7 8, 2H), 2 85 (s, 4H), 4 15-4 23 (m, 8H), 5 87 (s, 2H) 31 P NMR (162 MHz, CDCI 3 ) δ 23 76 (s, 2P)

Scheme 19. Preparation of oritavancin bisphosphonate conjugate 65.

63 NaHCO 3

H 2 O Dioxane

Oritavancin bisphosphonate conjugate 64. A suspension of oritavancin diphosphate (8, 537 mg, 0 270 mmol) and NaHCO 3 (68 mg, 0 81 mmol) in dιoxane/H 2 O (1/1 , 8 ml_) was stirred at room temperature for 15 mm The homogenous solution was then cooled in an ice-bath and a solution of 63 (287 mg, 0 540 mmol) in acetonitrile (1 ml_) was added drop-wise and the resulting solution was stirred at 0 0 C for 4 hr

Acetone/ether/acetonitrile (1/1/2, 20 mL) were added and the precipitate was collected by filtration The crude product was purified by C18 silica gel chromatography on a Biotage™ flash chromatography system (15% to 80% acetonitrile in 0 05%TFA in H 2 O) resulting in the colourless solid 64 (287 mg, 44%) as the dι-TFA salt ESI MS (M+H) calculated for C 1OO H I23 CI 3 N 10 O 36 P 2 2209, found 2209 4

Oritavancin bisphosphonate conjugate 65. A solution of 64 (280 mg, 0 115 mmol) and 2,6-lutιdιne (800 μl_, 6 89 mmol) in DMF (5 mL) was cooled to -70 0 C (2- propanol/dry ice) followed by the drop-wise addition of TMSBr (683 μL, 5 17 mmol) The resulting slurry was stirred for 30 mm at the same temperature then for 28 hr at room temperature The solution was concentrated to dryness without heating and the solid was resuspended in DMF (5 mL) followed by the addition of pyridine (1 31 mL, 16 2 mmol) and HF/pyπdine (203 μL, 8 10 mmol) The resulting solution was stirred for 1 hr at room temperature then concentrated to dryness The crude material was purified by C18 silica gel chromatography on a Biotage™ flash chromatography

system (15% to 80% acetonitrile in 0 05% TFA in H 2 O) resulting in the dι-TFA salt of 65 (90 mg, 54%) as a colourless solid ESI MS (M+H) calculated for C 92 H 107 CI 3 N 10 O 36 P 2 2098, found 2098 3

Example 2: Determination of in vitro antibacterial activity

In vitro antibacterial activity

Susceptibility of S aureus strain ATCC13709 to the commercial antibiotics and synthesized compounds was determined by following the guidelines set by the Clinical and Laboratory Standards Institute (formerly the National Committee for Clinical Laboratory Standards) (M26-A) Compounds were diluted two-fold serially in either DMSO (Vancomycin 4, Oritavancin 8, compounds 10, 31 , 33, 39, 44, 50, 60, 65), DMF (compounds 18 and 21 ) or in PBS (compound 6) and transferred to cation-adjusted Mueller Hinton broth (CAMHB, Becton Dickinson) 50 μL of compounds diluted in CAMHB was mixed with 100 μL of bacteria diluted in CAMHB in 96-well microtiter plates The final number of micro-organisms in the assay was 5x10 5 c f u per mL and the final concentration of DMSO or DMF in the assay, if present, was 1 25% Assays were set up in duplicate and incubated at 37 °C for 18 h The concentration of compound that inhibited visible growth was reported as the minimum inhibitory concentration (MIC)

Susceptibility testing experiments were also carried out in the presence of serum These experiments were carried out similar to the susceptibility testing with the following modifications 75 μL of compounds diluted in CAMHB was mixed with 75 μL of bacteria diluted in 100% serum from any given source (commercial pooled mouse serum (MS) and human serum (HS), Equitech-Bio lnc ) The final concentration of animal serum in the assay was 50% and the concentrations of all other components were identical to those described for susceptibility testing The data is summarized in Table 1

Table 1 : Antibacterial susceptibility of bacteria to selected compounds (Minimum inhibitory concentrations in μg/mL)

a: Cation adjusted Mueller-Hmton broth.

It can be broadly deduced that the bisphosphonated prodrugs 6, 10, 18, 21 , 31 , 33, 39, 44, 50, 60 and 65 possess antibacterial activities which are at least 16 to 32 fold weaker than the parent drugs (vancomycin for 6, 18, and 21 and oritavancin for 10, 31 , 33, 39, 44, 50, 60 and 65) This suggests the introduction of a bisphosphonated moiety to be detrimental to the antibacterial nature of the molecules

The presence of serum greatly impacted the MIC values associated with bisphosphonate conjugated drugs 31 , 33, 44 and 65 in the absence of bone mineral If the antibacterial activity seen results from the parent glycopeptide released from the prodrugs during the course of the assay, this suggests a possible participation from serum components, perhaps hydrolytic enzymes, in the cleavage of the prodrugs In essence, the release of the drug does appear to be less in aqueous buffer than in serum for compounds 31 , 33, 44 and 65 The differences between compounds 18 and

21-prodrugs of vancomycin- and 31 and 33 -prodrugs of oritavancin bearing the same linkers- is particularly stricking and suggests that simple changes in glycopeptide structure greatly impact the ability of the prodrug to regenerate the parent drug

These assay suggest that it is favourable for the bisphosphonated glycopeptides to be prodrugs, given that cleavage to the parent compound would result in raised antibacterial activity

Example 3: Binding of compounds to bone powder in vitro and subsequent regeneration of the parent drug. Bone powder binding

The ability of the molecules from Example 1 to bind to bone powder was established using a microbiological assay for detection An individual compound was dissolved in PBS+2% DMSO and added at a concentration of 1 mg/ml in a slurry of bone meal powder (Now Foods, Bloomingdale, Illinois, USA) in PBS at 10 mg/ml The suspension of drug/prodrug in bone meal powder was incubated at 37 0 C for 1 h to allow for binding, and centrifuged at 13 000 rpm for 2 mm, before recovering the supernatant The bone meal powder pellet was then washed three times with 1 ml of PBS + 2% DMSO The supernatant was assessed for drug content by microbioassays as follows For vancomycin, isolated colonies of the indicator strain Bacillus subtilis 1A754 were resuspended in 0 85% saline to 2 and spread on Cation-adjusted Miller Hinton agar (CAMHA) plates Known volumes of the supernatants were applied to discs and dried The discs were then placed on the seeded CAMHA plates The plates were incubated at 37 0 C for 18 h after which the diameters of the zone of inhibition generated by the discs were measured For oritavancin concentrations were determined with Mueller-Hmton agar containing 5% lysed horse blood and a clinical isolate of Streptococcus pneumoniae (ATCC 700902) Oritavancin standards and samples were placed in wells created in the agar and allowed to diffuse into the agar for 24 h The indicator strain (prepared in saline to an OD 600 of 0 2) was applied to the agar and plates were incubated at 37 0 C in an atmosphere of 5% CO 2 for 24 h, after which diameters of the zones of inhibition were measured.

The amount of prodrug was deduced from standard curves of known amounts of the parent drug (vancomycin 4 or oritavancin 8) that were used as reference for each experiment The results are displayed in Table 2

The results confirm that the bisphosphonated prodrugs are very efficiently removed from solution by osseous matter They also undeniably lend credence to the

use of bisphosphonates as mediators for bone delivery, by comparing prodrugs 18, 21 , 39, 60 and 65 (> 95% bound) to parent drug (binding not detected) It is reasonable to believe that a portion of the unbound material detected not to be bisphosphonated prodrug but contaminating or regenerated parent drug Nevertheless, it is also probable that the extent of binding to the osseous matter is reflective of the kinetics of bone absorption/adsorption There was no zone of inhibition seen for compound 6 in the bioassay, and as such the level of binding could not be determined

Regeneration of drug from bone powder-bound prodrug The ability of the prodrug to release the active entity at the site of infection is paramount for use in vivo This can be partially predetermined by measuring the release of the drug from prodrug bound to osseous matter in vitro

Amounts of parent drug "regenerated" from the phosphonated prodrug were measured as follows Washed bone powder-bound prodrugs from the above experiment were resuspended in 400 μl_ PBS+ 2% DMSO or in 400 μL 50% (v/v in PBS+2% DMSO) human or rat serum The suspension was incubated overnight at 37 0 C, centrifuged at 13,000 rpm for 2 mm and the supernatant was recovered The amount of regenerated parent drug in the supernatant was determined by measurements using the microbiological assays that were previously described for the prodrugs themselves The amount of released drug from prodrug was deduced from standard curves of known amounts of parent drug that were used as reference for each experiment The amount of regenerated drug assessed by this bioassay was corroborated by MIC determination The percentage of drug regenerated in PBS or serum after the overnight incubation (Table 2) was deduced from the difference between the amount of bound prodrug and the amount of regenerated drug (not shown)

Table 2: Bone binding and Conversion of bisphosphonated glycopeptide prodrugs to parent drugs after binding to bone (expressed as % prodrug converted after 24 h incubation)

-: not applicable. 7 ?: cannot be measured using current technique, n.d. : not determined. <L0D: below the limit of detection

The data presented in Table 2 provides evidence as to the importance of the selection of an appropriate bisphosphonate linker on the ability of the prodrugs to release the parent active entity Several trends are revealed by this data First, with

compounds 18 and 21 , both containing glycolamide linkers known to be effective in previous reports (Nielsen et al , Journal of Pharmaceutical Sciences (1988), 77(4) 285- 98 ), there is a fair amount of drug released from the bone matter after 24h On the other hand, no zones of inhibition were observed for bisphosphonated Vancomycin 6, which therefore either does not bind bone powder, and unlikely event given the very high binding of the other compounds, or, more probably, fails to regenerate For the Oritavancin prodrugs, the first trend observed is the very large difference between the rates of cleavage in PBS and those in serum It is possible that the linkers may be very sensitive to the presence of select serum components, but it is also possible that the recovery of Oritavancin in PBS be low when released, as in the experiment, and better when spiked, as in the controls This uncertainty requires the analysis to focus only on values in serum Surprisingly, the simple amide linker in prodrug 39 is still fairly labile, more so than prodrug 60 which has a bisphosphonated version of the known acyloxymethyl carbamate linker (Alexander et al, Journal of Medicinal Chemistry (1988), 31 318-322) Yet, the loss of a single carbon atom in the chain of the linker results in a marked acceleration in the rate of cleavage (see 65 versus 60) As in the case of 39, it appears that proximity of the bisphosphonate group to the site of cleavage results in accelerated hydrolysis, suggesting this moiety to be involved in the mechanism of cleavage These bone binding and regeneration experiments demonstrate both the affinity of the prodrugs for osseous matter and the ability of some of these molecules to release their parent molecule over time This augurs well for these molecules to be effective means of delivering glycopeptide and lipoglycopeptides to the site of infection in the treatment of osteomyelitis It is understood that the examples and embodiments described herein are for illustrative purposes only and that various modifications or changes in light thereof will be suggested to persons skilled in the art and are to be included within the spirit and purview of this application and scope of the appended claims

All documents, publications, patents, books, manuals, articles, papers, abstracts, posters and other materials referenced herein are expressly incorporated herein by reference in their entireties