Bone-targeting antimicrobial oxazolidinone-related compounds, formulations thereof, and uses thereof

文档序号:883391 发布日期:2021-03-19 浏览:10次 中文

阅读说明:本技术 骨靶向抗微生物噁唑烷酮相关化合物、其制剂及其用途 (Bone-targeting antimicrobial oxazolidinone-related compounds, formulations thereof, and uses thereof ) 是由 弗兰克·哈洛克·埃比蒂诺 舒廷·孙 查尔斯·E·麦克纳 凯万·萨德拉菲 菲利普·谢里安 于 2019-06-25 设计创作,主要内容包括:本文描述了双膦酸噁唑烷酮化合物及其缀合物和药物制剂,其可以包括双膦酸和噁唑烷酮(或噁唑烷酮抗微生物剂或抗生素剂、其取代基或衍生物),其中所述噁唑烷酮可以可释放地偶联至所述双膦酸。本文还提供了制造和使用双膦酸噁唑烷酮化合物、其缀合物及药物制剂的方法。本文还提供了使用所述化合物、缀合物和制剂用于治疗骨疾病的方法或用于制备用于治疗骨疾病的制剂的方法。(Bisphosphonate oxazolidinone compounds and conjugates and pharmaceutical formulations thereof are described herein, which may include a bisphosphonate and an oxazolidinone (or an oxazolidinone antimicrobial or antibiotic agent, substituent or derivative thereof), wherein the oxazolidinone may be releasably coupled to the bisphosphonate. Also provided herein are methods of making and using the bisphosphonate oxazolidinone compounds, conjugates thereof, and pharmaceutical formulations. Also provided herein are methods of using the compounds, conjugates, and formulations for treating or for preparing formulations for treating bone diseases.)

1. A method of making a bisphosphonate-antimicrobial conjugate thereof, comprising linking a bisphosphonate with an oxazolidinone antimicrobial agent.

2. The method of claim 1, wherein the antimicrobial agent comprises an oxazolidinone substituent or derivative or 2-oxazolidinone.

3. The method of claim 1 or 2, wherein said bisphosphonic acid is of the formulaWherein R is1Or R2Comprising an alkyl (hydroxyphenyl) group, an alkylhydroxy group, an alkylphenylamino group, an alkylamino group, an alkyl (hydroxyheterocyclic) group or an alkylaminocyclic group.

4. A process according to any one of claims 1 to 3 wherein the bisphosphonate is a methylenebisphosphonic acid and the alpha position of the methylenebisphosphonic acid moiety is substituted with hydroxy, fluoro, chloro, bromo or iodo.

5. A process according to any one or more of claims 1 to 3, wherein the bisphosphonic acid is an ethylidene bisphosphonic acid and the alpha position of the ethylidene bisphosphonic acid is substituted by hydroxy, fluoro, chloro, bromo or iodo.

6. The method of any of claims 1-5, wherein the antimicrobial agent comprises a primary or secondary amine group or a hydroxyl group.

7. The method of any of claims 1-6, wherein the linkage between the bisphosphonate and the antimicrobial agent is reversible in vivo to release the oxazolidinone antimicrobial agent at a therapeutically effective rate and dose at a targeted site.

8. The method according to any one of claims 1-7, wherein a urethane linker, preferably an aryl carbamate linker, is used for the connection between the bisphosphonic acid and the oxazolidinone antimicrobial agent.

9. The method of any one of claims 1-7, wherein a carbonate linker is used for the connection between the bisphosphonate and the oxazolidinone antimicrobial agent.

10. The method of any one of claims 1-7, wherein an ester linker is used for the linkage between the bisphosphonate and the oxazolidinone antimicrobial agent.

11. The method of any one of claims 1-7, wherein a urea (carbamide) linker is used for the linkage between the bisphosphonic acid and the oxazolidinone antimicrobial agent.

12. The method of any one of claims 1-7, wherein an S-thiocarbamate or O-thiocarbamate linker is used for the linkage between the bisphosphonate and the oxazolidinone antimicrobial agent.

13. The method of any one of claims 1-7, wherein a dithiocarbamate linker is used for the linkage between the bisphosphonic acid and the oxazolidinone antimicrobial agent.

14. The method of any one of claims 1-7, wherein a thiourea (thiocarboxamide) linker is used for the linkage between the bisphosphonic acid and the oxazolidinone antimicrobial agent.

15. A method according to any one of claims 1 to 14 wherein the oxazolidinone antimicrobial agent comprises a hydroxymethyl substituent on an oxazolidinone heterocyclic group.

16. The method of any one of claims 1-14, wherein the oxazolidinone antimicrobial agent is tedizolid.

17. The method of any one of claims 1-14, wherein the oxazolidinone antimicrobial agent is linezolid or a deacetylated analog thereof.

18. The method of any one of claims 1-14, wherein the oxazolidinone antimicrobial agent is oxyphenxadone, sutezolid, radizomide, rabezolid, a deacetylated analog thereof, or pasireotide.

19. The method of any one of claims 1-18, wherein said bisphosphonate is 4-aminophenylethylidene bisphosphonate.

20. The method of any one of claims 1-18, wherein said bisphosphonic acid is 4-hydroxyphenylethylidene bisphosphonic acid.

21. A bisphosphonate-antimicrobial agent prepared by the method of any one of the preceding claims.

22. A bisphosphonate-antimicrobial agent consisting of a general formula (BP-L-TD) in which 4-aminophenylethylidene bisphosphonate is linked to Tedizolid (TD) through a carbamate, wherein BP is 4-aminophenylethylidene bisphosphonate, L is-nc (O) O-, and TD is tedizolid.

23. A bisphosphonate-antimicrobial agent consisting of a compound of formula (BP-L-TD) wherein 4-hydroxyphenylethylidene bisphosphonate is linked to Tedizolid (TD) by a carbonate, wherein BP is 4-hydroxyphenylethylidene bisphosphonate, L is-oc (O) O-, and TD is tedizolid.

24. A bisphosphonate-antimicrobial agent consisting of a general formula (BP-L-TD) in which 4-aminophenylethylidene bisphosphonate is linked to Tedizolid (TD) through a carbamate, wherein BP is 4-aminophenylethylidene bisphosphonate, L is-nc(s) O-, and TD is tedizolid.

25. A bisphosphonate-antimicrobial agent consisting of a general formula (BP-L-dLD) in which 4-hydroxyphenylethylidene bisphosphonate is linked to deacetyllinezolid (dLD) via a carbamate, wherein BP is 4-hydroxyphenylethylidene bisphosphonate, L is-oc (o) N-, and dLD is deacetyllinezolid.

26. A bisphosphonate-antimicrobial agent consisting of a general formula (BP-L-dLD) in which 4-hydroxyphenylethylidene bisphosphonate is linked to deacetyllinezolid (dLD) via a carbamate, wherein BP is 4-hydroxyphenylethylidene bisphosphonate, L is-oc(s) N-or-sc (o) N-or-sc(s) N-, and dLD is deacetyllinezolid.

27. A bisphosphonate-antimicrobial agent consisting of a general formula (BP-L-dLD) in which 4-aminophenylethylidene bisphosphonate is linked to deacetyllinezolid (dLD) by urea, wherein BP is 4-aminophenylethylidene bisphosphonate, L is-nc (o) N-or-nc(s) N-, and dLD is deacetyllinezolid.

28. A bisphosphonate-antimicrobial agent consisting of a general formula (BP-L-ED) in which 4-hydroxyphenylethylidene bisphosphonate is linked to hydroxypipexone (ED) through a carbamate, wherein BP is 4-hydroxyphenylethylidene bisphosphonate, L is-nc (O) O-, and ED is hydroxypipexone.

29. A bisphosphonate-antimicrobial agent consisting of a general formula (BP-L-ED) in which 4-hydroxyphenylethylidene bisphosphonate is linked to hydroxypipexone (ED) through a carbamate, wherein BP is 4-hydroxyphenylethylidene bisphosphonate, L is-nc(s) O-, and ED is hydroxypipexone.

30. A bisphosphonate-antimicrobial agent consisting of a general formula (BP-L-ED) in which 4-hydroxyphenylethylidene bisphosphonate is linked to hydroxypipexone (ED) by a carbonate, wherein BP is 4-hydroxyphenylethylidene bisphosphonate, L is-oc (O) O-, and ED is hydroxypipexone.

31. A bisphosphonate-antimicrobial agent consisting of a general formula (BP-L-dED) in which 4-hydroxyphenylethylidene bisphosphonate is linked to deacetylhydroxypiperone (dED) via a carbamate, wherein BP is 4-hydroxyphenylethylidene bisphosphonate, L is-oc (o) N-, and dED is deacetylhydroxypiperone.

32. A bisphosphonate-antimicrobial agent consisting of a bisphosphonate having the general formula (BP-L-dED) 4-hydroxyphenylethylidene bisphosphonate linked to deacetylhydroxypiperone (dED), wherein BP is 4-hydroxyphenylethylidene bisphosphonate, L is-oc(s) N-or-sc (o) N-or-sc(s) N-, and dED is deacetylhydroxypiperone.

33. A bisphosphonate-antimicrobial agent consisting of a general formula (BP-L-dED) in which 4-aminophenylethylidene bisphosphonate is linked to deacetylhydroxypiperone (dED) by urea (dED), wherein BP is 4-aminophenylethylidene bisphosphonate, L is-nc (o) N-or-nc(s) N-, and dED is deacetylhydroxypiperone.

34. A bisphosphonate-antimicrobial agent consisting of a compound of formula (BP-L-dSD) in which 4-hydroxyphenylethylidene bisphosphonate is linked to deacetylated sutezolid (dSD) by a carbamate, wherein BP is 4-hydroxyphenylethylidene bisphosphonate, L is-oc (o) N-, and dSD is deacetylated sutezolid.

35. A bisphosphonate-antimicrobial agent consisting of a bisphosphonate having the general formula (BP-L-dSD) in which 4-hydroxyphenylethylidenebisphosphonate is linked to deacetylated sutezolid (dSD), wherein BP is 4-hydroxyphenylethylidenebisphosphonate, L is-OC (S) N-or-SC (O) N-or-SC (S) N-, and dSD is deacetylated sutezolid.

36. A bisphosphonate-antimicrobial agent consisting of a bisphosphonate of formula (BP-L-dSD) in which 4-aminophenylethylidene bisphosphonate is linked to deacetylated sutazolid (dSD) by urea, wherein BP is 4-aminophenylethylidene bisphosphonate, L is-nc (o) N-or-nc(s) N-, and dSD is deacetylated sutazolid.

37. A bisphosphonate-antimicrobial agent consisting of a general formula (BP-L-RD) in which 4-aminophenylethylidene bisphosphonate is linked to ladzolamide (dRD) via a carbamate, wherein BP is 4-aminophenylethylidene bisphosphonate, L is-oc (o) N-, and RD is ladzolamide.

38. A bisphosphonate-antimicrobial agent consisting of a general formula (BP-L-RD) in which 4-aminophenylethylidene bisphosphonate is linked to ladzolamide (dRD) via a carbamate, wherein BP is 4-aminophenylethylidene bisphosphonate, L is-oc(s) N-or-sc (o) N-or-sc(s) N-, and RD is ladzolamide.

39. A bisphosphonate-antimicrobial agent consisting of a general formula (BP-L-RD) in which 4-aminophenylethylidene bisphosphonate is linked to deacetyl ladzolamide (dRD) by urea, wherein BP is 4-aminophenylethylidene bisphosphonate, L is-nc (o) N-or-nc(s) N-, and RD is ladzolamide.

40. A bisphosphonate-antimicrobial agent consisting of a general formula (BP-L-dRD) in which 4-aminophenylethylidene bisphosphonate is linked to deacetyl radizole (dRD) via a carbamate, wherein BP is 4-aminophenylethylidene bisphosphonate, L is-oc (o) N-, and dRD is deacetyl radizole.

41. A bisphosphonate-antimicrobial agent consisting of a bisphosphonate with 4-aminophenylethylidene bisphosphonate linked to deacetyl radizole (dRD) (BP-L-dRD), where BP is 4-aminophenylethylidene bisphosphonate, L is-oc(s) N-or-sc (o) N-or-sc(s) N-, and dRD is deacetyl radizole.

42. A bisphosphonate-antimicrobial agent consisting of a general formula (BP-L-dRD) in which 4-aminophenylethylidene bisphosphonate is linked to deacetyl ladzolamide (dRD) by urea (BP-L-dRD), wherein BP is 4-aminophenylethylidene bisphosphonate, L is-nc (o) N-or-nc(s) N-, and dRD is deacetylladzolamide.

43. A bisphosphonate-antimicrobial agent consisting of a compound of formula (BP-L-dRbD) wherein BP is 4-hydroxyphenylethylidene bisphosphonate, L is-oc (o) N-, and dRbD is deacetylated rankzolide, linked by carbamate to deacetylated rankzolide (dRbD).

44. A bisphosphonate-antimicrobial agent consisting of a bisphosphonate having 4-hydroxyphenylethylidene bisphosphonate attached to a deacetylated rankzolid (dRbD) of the general formula (BP-L-dRbD), wherein BP is 4-hydroxyphenylethylidene bisphosphonate, L is-OC (S) N-or-SC (O) N-or-SC (S) N-, and dRbD is deacetylated rankzolid.

45. A bisphosphonate-antimicrobial agent consisting of a bisphosphonate of the general formula (BP-L-dRbD) in which 4-aminophenylethylidene bisphosphonate is linked by urea to deacetylated rankbendazole (dRbD), wherein BP is 4-aminophenylethylidene bisphosphonate, L is-nc (o) N-or-nc(s) N-, and dRbD is deacetylated rabenzolid.

46. A bisphosphonate-antimicrobial agent consisting of a general formula (BP-L-PD) in which 4-aminophenylethylidene bisphosphonate is linked to Pasiclazole (PD) through a carbamate, wherein BP is 4-aminophenylethylidene bisphosphonate, L is-nc (O) O-, and PD is pasiclazole.

47. A bisphosphonate-antimicrobial agent consisting of a general formula (BP-L-PD) in which 4-aminophenylethylidene bisphosphonate is linked to Pasiclazole (PD) through a carbamate, wherein BP is 4-aminophenylethylidene bisphosphonate, L is-nc(s) O-, and PD is pasiclazole.

48. A bisphosphonate-antimicrobial agent consisting of a general formula (BP-L-PD) in which 4-hydroxyphenylethylidene bisphosphonate is linked to Pasiclazole (PD) by a carbonate, wherein BP is 4-hydroxyphenylethylidene bisphosphonate, L is-oc (O) O-, and PD is pasiclazole.

49. A bisphosphonate-antimicrobial agent as substantially described by any one of the conjugates in figure 7 herein.

50. The bisphosphonate-antimicrobial agent according to any one of the preceding claims for use in the treatment of bone diseases.

51. The bisphosphonate-antimicrobial agent of claim 50, wherein the bone infectious disease is selected from the group consisting of: osteomyelitis, osteolytic bone infection, osteonecrosis, diabetic chronic osteomyelitis, diabetic foot, periodontitis and other jaw bone infections.

52. The bisphosphonate-antimicrobial agent according to any of the preceding claims for use in the treatment of surroundings associated and not associated with bone-related infections (non-bone infections).

53. A compound comprising a bisphosphonate-antimicrobial agent according to any one or more of claims 21-50 or prepared by a process according to any one of claims 1-20, configured for delivery and release of a target molecule at a bone site.

54. A formulation comprising a bisphosphonate-antimicrobial agent according to any one of claims 21-50 or prepared by a process according to any one of claims 1-20, provided in the form of: oral irrigation solutions, buffer solutions for intravenous or parenteral use, or microspheres in the form of powders/tablets for enteral administration or placed directly at the site of bone infection during surgery.

55. A formulation comprising a bisphosphonate-antimicrobial agent according to any one of claims 21-50 or prepared by a process according to any one of claims 1-20, wherein the formulation comprises an effective amount of a bisphosphonate-antimicrobial agent for the treatment of a bone infectious disease.

56. A method of treating a bone infection comprising delivering an effective amount of a bisphosphonate-antimicrobial agent according to any one of claims 21-50 or prepared by the method according to any one of claims 1-20 for the treatment of a bone infectious disease.

57. Use of a bisphosphonate-antimicrobial agent according to any one of claims 21-50 or prepared by a process according to any one of claims 1-20 for the preparation of a formulation for the treatment of bone diseases.

58. The bisphosphonate-antimicrobial agent for use according to claim 57, wherein the bone infectious disease is selected from the group consisting of: osteomyelitis, osteolytic bone infection, osteonecrosis, diabetic chronic osteomyelitis, diabetic foot, periodontitis and other jaw bone infections.

59. The bisphosphonate-antimicrobial agent for use according to claim 57, wherein the formulation is used to treat a surrounding (non-bone infection) related and unrelated to bone-related infections.

60. Use of a bisphosphonate-antimicrobial agent according to any one or more of claims 21-50 or prepared by a process according to one of claims 1-20 for the preparation of a formulation configured for delivery and release of a target antimicrobial molecule at a bone site.

61. A bone graft composition comprising: a bone graft material and a bisphosphonate-antimicrobial agent according to any one or more of claims 21-50 or prepared by a method according to any one of claims 1-20, wherein the bisphosphonate-antimicrobial agent is attached, bound, chemisorbed or mixed with the bone graft material.

62. The bone graft composition of claim 61, wherein the bone graft material is an autograft bone material, an allograft bone material, a xenograft bone material, a synthetic bone graft material, or any combination thereof.

63. A method, comprising: implanting a bone graft composition according to any one of claims 61-62 into a subject in need thereof.

64. A method of preventing biofilm infection at a bone or an implant surgical site or at a surgical site where a bone graft is performed, wherein the method comprises: administering to a subject in need thereof a bisphosphonate-antimicrobial agent according to one or more of claims 21-50 or prepared by a method according to one of claims 1-20.

65. A method of preventing biofilm infection at a bone or an implant surgical site or at a surgical site where a bone graft is performed, wherein the method comprises:

implanting a bone graft composition according to any one of claims 61-62 into a subject in need thereof.

66. A compound according to formula (5),

wherein R is1Can be an oxazolidinone antibiotic substituent or a known component of an oxazolidinone derivative antimicrobial or antibiotic compound, and can include alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, phenyl, substituted phenyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, halogen, hydroxy, alkoxy, substituted alkoxy, phenoxy, substituted phenoxy, aryloxy, substituted aryloxy, alkylthio, substituted alkylthio, phenylthio, substituted phenylthio, arylthio, substituted arylthio, cyano, isocyano, substituted isocyano, carbonyl, substituted carbonyl, carboxyl, substituted carboxyl, amino, substituted amino, amido, substituted amido, sulfonyl, substituted sulfonyl, sulfonic acid, phosphoryl, substituted phosphoryl, phosphonyl, substituted phosphonyl, or substituted phosphonyl, Polyaryl, substituted polyaryl, C3-C20Cyclic, substituted C3-C20Rings, heterocycles, substituted heterocycles, amino acids, peptides and polypeptide groups; and is

Wherein R is2May be a substituent comprising a releasable linker as described herein involved in the linkage to a bisphosphonate as described herein, and may include alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, phenyl, substituted phenyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, halogen, hydroxy, alkoxy, substituted alkoxy, phenoxy, substituted phenoxy, aryloxy, substituted aryloxy, alkylthio, substituted alkylthio, phenylthio, substituted phenylthio, arylthio, substituted arylthio, cyano, isocyano, substituted isocyano, carbonyl, substituted carbonyl, carboxyl, substituted carboxyl, amino, substituted aryl, substituted heteroaryl, halogen, hydroxy, alkoxy, substituted alkoxy, phenoxy, substituted phenoxy, aryloxy, substituted alkylthio, phenylthio, substituted phenylthio, arylthio, cyano, substituted isocyano, carbonyl, substituted carbonyl, carboxyl, substituted carboxyl, amino, substituted cyanoAmino, amido, substituted amido, sulfonyl, substituted sulfonyl, sulfonic acid, phosphoryl, substituted phosphoryl, phosphonyl, substituted phosphonyl, polyaryl, substituted polyaryl, C3-C20Cyclic, substituted C3-C20Rings, heterocycles, substituted heterocycles, amino acids, peptides and polypeptide groups.

67. The compound of claim 66, wherein the linker is a carbamate linker or a carbonate linker.

68. The compound of claim 66 or 67, wherein the linker is selected from the group consisting of: an aryl carbamate linker, a thiocarbamate-O-aryl ester linker, a thiocarbamate-S-aryl ester linker, a phenyl carbamate linker, a thiocarbamate linker, an O-thiocarbamate linker, an S-thiocarbamate linker, an ester linker, or a dithiocarbamate.

69. A compound according to any of claims 66-68, wherein said bisphosphonic acid is selected from the group consisting of: hydroxyphenylalkyl or aryl bisphosphonic acids, hydroxyphenyl (or aryl) alkylhydroxy bisphosphonic acids, aminophenyl (or aryl) alkyl bisphosphonic acids, aminophenyl (or aryl) alkylhydroxy bisphosphonic acids, hydroxyalkyl hydroxy bisphosphonic acids, hydroxyalkylphenyl (or aryl) alkyl bisphosphonic acids, hydroxyphenyl (or aryl) alkylhydroxy bisphosphonic acids, aminophenyl (or aryl) alkyl hydroxy bisphosphonic acids, hydroxyalkyl hydroxy bisphosphonic acids, hydroxypyridyl alkyl bisphosphonic acids, pyridylalkyl bisphosphonic acid, hydroxyimidazolylalkyl bisphosphonic acid, imidazolylalkyl bisphosphonic acid, etidronic acid, pamidronic acid, neridronic acid, olpadronic acid, alendronic acid, ibandronic acid, risedronic acid, zoledronic acid, minodronic acid, and combinations thereof, wherein all compounds may be optionally further substituted or unsubstituted.

70. The compound of any of claims 66-68, wherein the BP is selected from the group consisting of: etidronic acid, methylenehydroxy bisphosphonic acid (MHDP), pamidronic acid, alendronic acid, risedronic acid, zoledronic acid, minodronic acid, and combinations thereof.

71. The compound of any of claims 66-70, wherein said oxazolidinone is an oxazolidinone antimicrobial or antibiotic compound or agent.

72. A compound according to any of claims 66-71, wherein said oxazolidinone compound is 2-oxazolidinone.

73. A compound according to any of claims 66-71, wherein said oxazolidinone compound is selected from the group consisting of: linezolid, tedizolid, hydroxypipexone, pasirezolid, radizolid, sutezolid, antimicrobial or antibiotic substituents or derivatives thereof and combinations thereof.

74. A compound according to any of claims 66-70, wherein said oxazolidinone compound is tedizolid or a substituent or derivative thereof.

75. A compound according to any of claims 66-74, wherein said bisphosphonate is an inert bisphosphonate.

76. The compound of claim 75, wherein said linker is an aryl carbamate linker.

77. A compound according to any of claims 66-74, wherein said bisphosphonate is an active bisphosphonate.

78. A compound according to claim 77, wherein the bisphosphonic acid has a geminal hydroxyl group and the linker is attached to the hydroxyl group of the bisphosphonic acid.

79. A compound of formula (4)

Background

Bone and joint infections affect millions of adults and children worldwide. The overall incidence in the united states is 3-6 million people, with a particular population having different risks. For diabetic patients, the annual incidence of foot ulcers is about 1 in 30, up to two-thirds of cases with underlying osteomyelitis. In children, the annual incidence has recently been reported in the range of 1/4000 to 1/15000. However, in the Pediatric Health Information System (PHIS) database of the management data of the american pediatric hospital, we found that during the 5 years 2009-2013, 10,245 (0.5%) of 2,247,889 was diagnosed as osteomyelitis at the time of discharge and the gross annual incidence was about 1/1100 hospitalization.

Many gram-positive and gram-negative bacteria as well as fungi and mycobacteria can cause bone and joint infections. To date, the most common organism implicated in bone and joint infections is Staphylococcus aureus (staphyloccus aureus), both methicillin-sensitive (MSSA) and methicillin-resistant (MRSA).

The standard of care for bone and joint infections generally requires systemic administration of antibiotics. For acute infections, the prescription is usually: the antibiotic is injected intravenously for 2-6 weeks. For chronic infections or infections associated with retained implanted hardware, a prolonged course of oral antibiotics may be followed. For both acute and chronic infections, these extended courses of treatment may lead to drug-related adverse events in a significant percentage of patients — 15% in one estimate of the cohort for treatment of infected MSSA. Furthermore, it is known that nephrotoxicity of vancomycin, the most common treatment of MRSA infections, occurs in up to 43% of patients and increases with the duration of treatment.

Persistent bone infections such as osteomyelitis of the jaw bone, osteomyelitis and osteonecrosis in other skeletal sites eventually lead to significant bone resorption and destruction of bone and Hydroxyapatite (HA) minerals. Bone and HA resorption is thought to be induced and mediated not only by osteocytes (i.e., osteoclasts), but also by microbial biofilm pathogens in combination with host inflammatory responses and osteoclastogenesis activity. The treatment of infectious bone diseases is mainly based on clinical pathological factors with or without surgical intervention for antimicrobial therapy. Surgery may include conservative removal of the affected bone or a more aggressive approach, such as resection.

To overcome many of the challenges associated with treating bone infections, it has become routine for clinicians to use local delivery systems to achieve higher therapeutic antibacterial concentrations in bone. For example, polymethylmethacrylate beads represent the most non-biodegradable carrier systems for delivering antibiotics to orthopedic infections, but they require surgical removal after drug release is complete. They also tend to release the antibiotic in an initial burst mode that quickly consumes the majority of the drug from the carrier beads, followed by a slow release at a lower concentration that may not be sufficient to control the infection and may promote the development of drug resistance. These concerns limit the usefulness of this approach in most bone and joint infections.

Dentistry has used local delivery of antimicrobial agents to treat infected jaw bone associated with conditions such as periodontal bone loss, jaw osteomyelitis and osteonecrosis to achieve high local concentrations of the drug, but without surgical intervention, these approaches are generally ineffective and the bone bioavailability of antibiotics is poor. Antibiotic impregnated cementum, which is used primarily when debriding infected implants for the first time to improve control of infection, is not generally used to treat bone and joint infections of natural bone without implanted hardware. Concerns regarding prolonged sub-therapeutic antibiotic concentrations and selection of drug-resistant bacteria also apply to cementum.

Local delivery of antimicrobial agents to bone is an important advance in the treatment of infectious bone diseases, but still has osmotic limitations and potential eukaryotic cellular cytotoxicity; therefore, there is a high need to research and develop more efficient delivery systems with physiological targeting. The ideal antibiotic delivery system is one that targets bone tissue without the need for surgical implantation or removal. This targeting also minimizes systemic dose and exposure of tissues other than bone to antibiotics, thus reducing the risk of selective stress that adversely affects or promotes the emergence of resistant bacteria. Another potential major benefit is that by achieving prolonged antibiotic concentrations at the site of infection, it is possible to reduce the frequency of administration.

Disclosure of Invention

Provided herein in various aspects are BP oxazolidinone (oxazolidinone) (or oxazolidinone) antibiotic compounds, conjugates, and formulations to address the aforementioned needs. BP oxazolidinone antibiotic compounds, conjugates, and formulations can comprise bisphosphonic acids (BPs) that can be releasably conjugated to oxazolidinone antibiotics, such as tedizolid. In any one or more embodiments, the BP oxazolidinone conjugate can be administered systemically to a subject to selectively deliver the oxazolidinone to the bone, and in particular to an infected bone site, or locally to the subject when combined with a bone graft or bone graft substitute (i.e., can target bone, bone infection, or other high bone metabolic site). In any one or more embodiments, the BP oxazolidinone compound or conjugate can release an oxazolidinone, particularly an oxazolidinone antibiotic compound, a substituent, or a derivative thereof. Also provided herein are methods of synthesizing BP oxazolidinone compounds, conjugates, and methods of treating or preventing osteomyelitis or other bone infections with one or more BP oxazolidinone compounds, conjugates, and/or formulations provided herein.

In any one or more aspects, the compound or conjugate can be a compound according to general formula (1) below.

In any one or more aspects, the compound or conjugate can have the formula:

in a particular embodiment, the compound or conjugate may be a tedizolid analog linked to a bisphosphonate, as in formula (3) or (4) below.

Also provided herein are pharmaceutical compositions comprising a compound or conjugate according to formula (1), or, in particular, formula (2), formula (3), or formula (4), and a pharmaceutically acceptable carrier.

Also provided herein are methods of treating a bone infection in a subject in need thereof, which may include the steps of: administering to a subject in need thereof an amount of a compound or conjugate according to formula (1), or in particular formula (2), formula (3) or formula (4), or a pharmaceutical formulation comprising a compound according to formula (1), or in particular formula (2), formula (3) or formula (4).

Also provided herein are compounds, conjugates, and antimicrobial and antibiotic agents comprising a Bisphosphonate (BP) and an oxazolidinone compound, wherein the oxazolidinone compound is releasably or reversibly coupled to the bisphosphonate through a linker, as described herein. Preferred releasable linkers are more or less stable in the blood stream shortly after administration and more or less slowly lyse in the bone/skeletal zones of the body to slowly release locally the oxazolidinone antibiotic compound, substituent or derivative.

In any one or more aspects herein, the BP can be selected from the group consisting of: hydroxyphenylalkyl or aryl bisphosphonic acids, hydroxyphenyl (or aryl) alkylhydroxy bisphosphonic acids, aminophenyl (or aryl) alkylphosphonic acids, aminophenyl (or aryl) alkylhydroxy bisphosphonic acids, hydroxyalkyl bisphosphonic acids, hydroxyalkylhydroxy bisphosphonic acids, hydroxyalkylphenyl (or aryl) alkylphosphonic acids, hydroxyphenyl (or aryl) alkylhydroxy bisphosphonic acids, aminophenyl (or aryl) alkylphosphonic acids, aminophenyl (or aryl) alkylhydroxy bisphosphonic acids, hydroxyalkyl bisphosphonic acids, hydroxyalkylhydroxy bisphosphonic acids, hydroxypyridinylalkyl bisphosphonic acids, pyridylalkyl bisphosphonic acids, hydroxyimidazolylalkyl bisphosphonic acids, imidazolylalkyl bisphosphonic acids, etidronic acid (etidronate), pamidronate, neridronate, olpadronate, alendronate, ibandronate, risedronate, Zoledronate (zoledronate), minodronate (minodronate), and combinations thereof, wherein all compounds may be optionally further substituted or unsubstituted. In particular, the BP may be selected from the group consisting of: etidronic acid, methylenehydroxy bisphosphonic acid (MHDP), pamidronic acid, alendronic acid, risedronic acid, zoledronic acid, minodronic acid, and combinations thereof.

In any one or more aspects herein, the oxazolidinone (or oxazolidinone) is an oxazolidinone antimicrobial or antibiotic compound or agent. The oxazolidinone compound may be a compound containing 2-oxazolidinone (also referred to as 2-oxazolidinone). The oxazolidinone compound may be selected from the group consisting of: linezolid, tedizolid, hydroxypipexone, pasiclolide, redazolide, rabezolid, sutezolid and combinations thereof. The oxazolidinone may be a substituent, such as a suitable substituent, or a derivative of any of the foregoing, particularly an antimicrobial or antibiotic substituent.

In some aspects, the BP is etidronic acid. In some aspects, the oxazolidinone is tedizolid. In other aspects, the oxazolidinone is tedizolid, and the BP can be other BPs described herein, such as pamidronic acid, neridronic acid, olpadronic acid, alendronic acid, ibandronic acid, minodronic acid, risedronic acid, zoledronic acid, hydroxymethylbisphosphonic acid, and combinations thereof.

Analogs of the oxazolidinone compounds of the present disclosure can have a structure according to formula (5),

wherein R is1May be oxazolidinone antibiotic substituents or known components of oxazolidinone derivative antimicrobial or antibiotic compounds and may include alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkylAlkynyl, phenyl, substituted phenyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, halogen, hydroxy, alkoxy, substituted alkoxy, phenoxy, substituted phenoxy, aryloxy, substituted aryloxy, alkylthio, substituted alkylthio, phenylthio, substituted phenylthio, arylthio, substituted arylthio, cyano, isocyano, substituted isocyano, carbonyl, substituted carbonyl, carboxyl, substituted carboxyl, amino, substituted amino, amido, substituted amido, sulfonyl, substituted sulfonyl, sulfonic acid, phosphoryl, substituted phosphoryl, phosphonyl, substituted phosphonyl, polyaryl, substituted polyaryl, C3-C20Cyclic, substituted C3-C20Rings, heterocycles, substituted heterocycles, amino acids, peptides and polypeptide groups; and is

Wherein R is2May be a substituent comprising a releasable linker as described herein involved in the linkage to a bisphosphonate as described herein, and may include alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, phenyl, substituted phenyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, halogen, hydroxy, alkoxy, substituted alkoxy, phenoxy, substituted phenoxy, aryloxy, substituted aryloxy, alkylthio, substituted alkylthio, phenylthio, substituted phenylthio, arylthio, substituted arylthio, cyano, isocyano, substituted isocyano, carbonyl, substituted carbonyl, carboxyl, substituted carboxyl, amino, substituted amino, amido, substituted amido, sulfonyl, substituted sulfonyl, sulfonic acid, phosphoryl, substituted phosphoryl, phosphonyl, substituted phosphonyl, or, Polyaryl, substituted polyaryl, C3-C20Cyclic, substituted C3-C20Rings, heterocycles, substituted heterocycles, amino acids, peptides and polypeptide groups.

The linker may be a cleavable compound, meaning that it reversibly couples an oxazolidinone antimicrobial or antibiotic compound, in particular an oxazolidinone antimicrobial or antibiotic substituent or a derivative thereof, to the BP. As used herein, the term "cleavable" may mean a group that is chemically or biochemically labile under physiological conditions. In any one or more aspects, the linker can be a carbamate having the structure or formula (6)

For the preparation of oxazolidinones, R1With BP, R2Is coupled, as described herein, and R3Can be substituted and unsubstituted alkyl, acetyl, benzoyl or other amides, phenyl and substituted phenyl, preferably H.

In any one or more aspects, the linker can be a carbonate having the structure or formula (7)

For the preparation of oxazolidinones, R1With BP, R2Coupling, as described herein.

In any one or more aspects, the linker can be an aryl carbamate linker. The linker may be a thiocarbamic-O-aryl ester linker. The linker may be a thiourethane-S-aryl ester linker. The linker may be a phenyl carbamate linker. The linker may be a thiocarbamate linker. The linker may be an O-thiocarbamate linker. The linker may be an S-thiocarbamate linker. The linker may be an ester linker. The linker may be a dithiocarbamate. The linker may be a urea linker. The joint may be attached to R of formula (5)2Group and further coupled to BP, as described herein. In any one or more aspects, the linker can be exemplified by any one of formulae (8) -formula (12) below, wherein: r1Can be oxazolidinone or oxazolidinone substituents or derivatives and R2May be BP, both as described herein; and R is3May be substituted or unsubstituted alkyl, acetyl, benzoyl or other amides,Phenyl and substituted phenyl, preferably H.

In any one or more aspects, the alpha position of the methylene bisphosphonic acid (methylene bisphosphonates) moiety may be substituted with hydroxy, fluoro, chloro, bromo, or iodo. In some aspects, the bisphosphonic acid may include a p-hydroxyphenyl ethylidene (ethylidene) group or a derivative thereof. In some aspects, the ethylidene bisphosphonic acid does not contain an alpha-hydroxy group at the alpha position.

In any one or more aspects, the compound or conjugate has a formula according to formula (1) above, or more specifically formula (2), formula (3), formula (4), or formula (5).

Also provided herein are pharmaceutical formulations that can comprise a bisphosphonate and an oxazolidinone antibiotic compound and a pharmaceutically acceptable carrier, wherein the oxazolidinone antibiotic compound is releasably coupled to the bisphosphonate through a linker. Preferred releasable linkers are more or less stable in the blood stream shortly after administration and more or less slowly lyse in the bone/skeletal zones of the body to slowly release locally the oxazolidinone antibiotic compound, substituent or derivative.

In any one or more aspects, the bisphosphonate may be selected from the group of: hydroxyphenylalkyl or aryl bisphosphonic acids, hydroxyphenyl (or aryl) alkylhydroxy bisphosphonic acids, aminophenyl (or aryl) alkyl bisphosphonic acids, aminophenyl (or aryl) alkylhydroxy bisphosphonic acids, hydroxyalkyl hydroxy bisphosphonic acids, hydroxyalkylphenyl (or aryl) alkyl bisphosphonic acids, hydroxyphenyl (or aryl) alkylhydroxy bisphosphonic acids, aminophenyl (or aryl) alkyl hydroxy bisphosphonic acids, hydroxyalkyl hydroxy bisphosphonic acids, hydroxypyridyl alkyl bisphosphonic acids, pyridylalkyl bisphosphonic acid, hydroxyimidazolylalkyl bisphosphonic acid, imidazolylalkyl bisphosphonic acid, etidronic acid, pamidronic acid, neridronic acid, olpadronic acid, alendronic acid, ibandronic acid, risedronic acid, zoledronic acid, minodronic acid, and combinations thereof, wherein all compounds may be optionally further substituted or unsubstituted. In particular, the BP may be selected from the group consisting of: etidronic acid, methylenehydroxy bisphosphonic acid (MHDP), pamidronic acid, alendronic acid, risedronic acid, zoledronic acid, minodronic acid, and combinations thereof.

In any one or more aspects herein, the oxazolidinone (or oxazolidinone) is an oxazolidinone antimicrobial or antibiotic compound or agent. The oxazolidinone compound may be a compound containing 2-oxazolidinone. The oxazolidinone compound may be selected from the group consisting of: linezolid, tedizolid, hydroxypipexone, pasiclolide, redazolide, rabezolid, sutezolid and combinations thereof. The oxazolidinone may be a substituent, such as a suitable substituent, or a derivative of any of the foregoing, particularly an antimicrobial or antibiotic substituent.

In some aspects, the BP is etidronic acid. In some aspects, the oxazolidinone is tedizolid. In other aspects, the oxazolidinone is tedizolid, and the BP can be other BPs described herein, such as pamidronic acid, neridronic acid, olpadronic acid, alendronic acid, ibandronic acid, minodronic acid, risedronic acid, zoledronic acid, hydroxymethylbisphosphonic acid, and combinations thereof.

The analogue of the oxazolidinone antibiotic compound may have a structure according to formula (5)

Wherein R is1Can be an oxazolidinone antibiotic substituent or a known component of oxazolidinone derivative antimicrobial or antibiotic compounds, and can include alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, phenyl, substituted phenyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, halogen, hydroxy, alkoxy, substituted alkoxy, phenoxy, substituted phenoxy, aryloxy, substituted aryloxy, alkylthio, substituted alkoxy, substituted aryl, substitutedAlkylthio, phenylthio, substituted phenylthio, arylthio, substituted arylthio, cyano, isocyano, substituted isocyano, carbonyl, substituted carbonyl, carboxyl, substituted carboxyl, amino, substituted amino, amido, substituted amido, sulfonyl, substituted sulfonyl, sulfonic acid, phosphoryl, substituted phosphoryl, phosphonyl, substituted phosphonyl, polyaryl, substituted polyaryl, C3-C20Cyclic, substituted C3-C20Rings, heterocycles, substituted heterocycles, amino acids, peptides and polypeptide groups; and is

Wherein R is2May be a substituent comprising a releasable linker as described herein involved in the linkage to a bisphosphonate as described herein, and may include alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, phenyl, substituted phenyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, halogen, hydroxy, alkoxy, substituted alkoxy, phenoxy, substituted phenoxy, aryloxy, substituted aryloxy, alkylthio, substituted alkylthio, phenylthio, substituted phenylthio, arylthio, substituted arylthio, cyano, isocyano, substituted isocyano, carbonyl, substituted carbonyl, carboxyl, substituted carboxyl, amino, substituted amino, amido, substituted amido, sulfonyl, substituted sulfonyl, sulfonic acid, phosphoryl, substituted phosphoryl, phosphonyl, substituted phosphonyl, or, Polyaryl, substituted polyaryl, C3-C20Cyclic, substituted C3-C20Rings, heterocycles, substituted heterocycles, amino acids, peptides and polypeptide groups.

The linker may be a cleavable compound, meaning that it reversibly couples the oxazolidinone antimicrobial compound to the BP. In any one or more aspects, the linker can be a carbamate having the structure or formula (6) above that is used to couple the oxazolidinone, R1Coupling to BP, R2As described herein, and R3Can be substituted and unsubstituted alkyl, acetyl, benzoyl or other amides, phenyl and substituted phenyl, preferably H.

In any one or more aspects, the linker can be a carbonate having the structure or formula (7) above, which is used to couple the oxazolidinone, R1Coupling to BP, R2As described herein.

In any one or more aspects, the linker can be a carbamate linker. The linker may be an aryl carbamate linker. The linker may be a thiocarbamic-O-aryl ester linker. The linker may be a thiourethane-S-aryl ester linker. The linker may be a phenyl carbamate linker. The linker may be a thiocarbamate linker. The linker may be an O-thiocarbamate linker. The linker may be an S-thiocarbamate linker. The linker may be an ester linker. The linker may be a dithiocarbamate. The linker may be a urea linker. The joint may be attached to R of formula (5)2Group and further coupled to BP, as described herein. In any one or more aspects, the linker can be exemplified by any one of formulae (8) -formula (12) above, wherein: r1Can be oxazolidinone or oxazolidinone substituents or derivatives and R2May be BP, both as described herein; and R is3Can be substituted and unsubstituted alkyl, acetyl, benzoyl or other amides, phenyl and substituted phenyl, preferably H.

In any one or more aspects, the alpha position of the methylene bisphosphonic acid moiety can be substituted with hydroxy, fluoro, chloro, bromo, or iodo. In some aspects, the bisphosphonic acid may include a p-hydroxyphenyl ethylidene group or a p-hydroxyphenyl alkylidene group or derivative thereof. In some aspects, the hydroxyethylidene bisphosphonate/hydroxyphenyl alkylidene does not include an alpha-hydroxy group in the alpha position.

In some aspects, the formulation may include a compound or conjugate having a formula according to formula (1) above, or more particularly formula (2), formula (3), formula (4), or formula (5).

The amount of the compound or conjugate in the pharmaceutical formulation may be an amount effective to kill or inhibit bacteria. The amount of the compound or conjugate in the pharmaceutical formulation may be an amount effective to treat, inhibit or prevent bone disease. The amount of the compound or conjugate in the pharmaceutical formulation may be an amount effective to treat, inhibit or prevent osteomyelitis, osteonecrosis, peri-implantitis and/or periodontitis. The amount of the compound or conjugate in the pharmaceutical formulation may be an effective amount for prophylactic treatment of any of the foregoing.

Also provided herein are methods of treating bone diseases, such as hematogenous or local osteomyelitis including juvenile osteomyelitis and infections associated with prosthetic joint replacement or osteonecrosis, in a subject in need thereof, which may comprise the steps of: administering to the subject in need thereof an amount of a compound as provided herein or a pharmaceutical formulation thereof.

Also provided herein are methods of treating peri-implantitis or periodontitis in a subject in need thereof, comprising administering to the subject in need thereof an amount of a compound as provided herein or a pharmaceutical formulation thereof.

Also provided herein are methods of treating bone infections (including diabetic foot disease) in a diabetic patient in a subject in need thereof, comprising administering to the subject in need thereof an amount of a compound as provided herein or a pharmaceutical formulation thereof. The associated reduction in amputation, debridement of limbs and affected bone sites will result from these more powerful local antibiotic treatment modalities.

Also provided herein are bone graft compositions that can include a bone graft material and a compound or pharmaceutical formulation thereof as described herein, wherein the compound or pharmaceutical formulation thereof is attached, bound, chemisorbed, or mixed with the bone graft material. The bone graft material may be an autograft bone material, an allograft bone material, a xenograft bone material, a synthetic bone graft material, or any combination thereof.

Also provided herein are methods that can include the step of implanting a bone graft composition as described herein into a subject in need thereof.

Also provided herein are methods of prophylactic or preventative treatment of a biofilm infection at a bone or an implant surgical site or at a surgical site where a bone graft is performed, wherein the methods may comprise the step of administering to a subject in need thereof a compound as described herein.

Also provided herein are methods of prophylactic or preventative treatment of biofilm infection at a bone or implant surgical site or at a surgical site where a bone graft is performed, wherein the methods may comprise the step of implanting a bone graft composition as described herein into a subject in need thereof.

Further, in an embodiment, there is provided a method of preparing a bisphosphonate-antimicrobial conjugate thereof, comprising linking a bisphosphonate with an oxazolidinone antimicrobial agent. In any one or more aspects thereof, the antimicrobial agent can include an oxazolidinone substituent or derivative or a 2-oxazolidinone. The bisphosphonic acid may have the formulaWherein R is1Or R2Comprising an alkyl (hydroxyphenyl) group, an alkylhydroxy group, an alkylphenylamino group, an alkylamino group, an alkyl (hydroxyheterocyclic) group or an alkylaminocyclic group. The bisphosphonate may be a methylenebisphosphonic acid, and the alpha position of the methylenebisphosphonic acid moiety is substituted with hydroxy, fluoro, chloro, bromo, or iodo. The bisphosphonate may be an ethylidene bisphosphonate, and the alpha position of the ethylidene bisphosphonate is substituted with hydroxy, fluoro, chloro, bromo, or iodo. The antimicrobial agent may comprise a primary or secondary amine group or a hydroxyl group. The linkage between the bisphosphonate and the antimicrobial agent is reversible in vivo to release the oxazolidinone antimicrobial agent at a therapeutically effective rate and dosage at the targeted site. A carbamate linker, preferably an aryl carbamate linker, is used for the linkage between the bisphosphonate and the oxazolidinone antimicrobial agent. A carbonate linker is used for the linkage between the bisphosphonic acid and the oxazolidinone antimicrobial agent. An ester linker is used for the linkage between the bisphosphonate and the oxazolidinone antimicrobial agent. Urea (carbamide) linkers are used for the linkage between the bisphosphonic acid and the oxazolidinone antimicrobial agent. S-thiocarbamates or O-thioamino groupsA formate linker is used for the linkage between the bisphosphonic acid and the oxazolidinone antimicrobial agent. A dithiocarbamate linker is used for the linkage between the bisphosphonic acid and the oxazolidinone antimicrobial agent. A thiourea (thiocarbamide) linker is used for the linkage between the bisphosphonic acid and the oxazolidinone antimicrobial agent. The oxazolidinone antimicrobial agents may include a hydroxymethyl substituent on the oxazolidinone heterocyclic group. The oxazolidinone antimicrobial agent can be tedizolid. The oxazolidinone antimicrobial agent can be linezolid or a deacetylated analog thereof. The oxazolidinone antimicrobial agent may be hydroxypipexone, sutezolid, radizolid, a deacetylated analogue thereof, or pasireotide. The bisphosphonate may be 4-aminophenylethylidene bisphosphonate.

In any one or more embodiments herein, there is provided a bisphosphonate-antimicrobial agent prepared by any one or more aspects of the foregoing method.

In any one or more embodiments herein, a bisphosphonate-antimicrobial agent is provided. The bisphosphonate-antimicrobial agent may be a general formula (BP-L-TD) in which 4-aminophenylethylidene bisphosphonate is linked to Tedizolid (TD) through a carbamate, wherein BP is 4-aminophenylethylidene bisphosphonate, L is-nc (O) O-, and TD is tedizolid. The bisphosphonate-antimicrobial agent may consist of a general formula (BP-L-TD) in which 4-hydroxyphenylethylidene bisphosphonate is linked to Tedizolid (TD) by a carbonate, wherein BP is 4-hydroxyphenylethylidene bisphosphonate, L is-oc (O) O-, and TD is tedizolid. The bisphosphonate-antimicrobial agent may consist of a general formula (BP-L-TD) in which 4-aminophenylethylidene bisphosphonate is linked to Tedizolid (TD) through a carbamate, wherein BP is 4-aminophenylethylidene bisphosphonate, L is-nc(s) O-, and TD is tedizolid. The bisphosphonate-antimicrobial agent may consist of general formula (BP-L-dLD) wherein BP is 4-hydroxyphenylethylidene bisphosphonate, L is-oc (o) N-, and dLD is deacetyllinezolid, linking 4-hydroxyphenylethylidene bisphosphonate to deacetyllinezolid (dLD) via a carbamate. The bisphosphonate-antimicrobial agent may consist of general formula (BP-L-dLD) wherein BP is 4-hydroxyphenylethylidene bisphosphonate, L is-oc(s) -N-or-sc (o) N-or-sc(s) N-, and dLD is deacetyllinezolid (dLD) by linking 4-hydroxyphenylethylidene bisphosphonate to deacetyllinezolid (dLD) via a carbamate. The bisphosphonate-antimicrobial agent may consist of general formula (BP-L-dLD) in which 4-aminophenylethylidene bisphosphonate is linked to deacetyllinezolid (dLD) by urea, where BP is 4-aminophenylethylidene bisphosphonate, L is-nc (o) N-or-nc(s) N-, and dLD is deacetyllinezolid. The bisphosphonate-antimicrobial agent may consist of a general formula (BP-L-ED) in which 4-hydroxyphenylethylidene bisphosphonate is linked to hydroxypipexone (ED) by a carbamate, wherein BP is 4-hydroxyphenylethylidene bisphosphonate, L is-nc (O) O-, and ED is hydroxypipexone. The bisphosphonate-antimicrobial agent may consist of a general formula (BP-L-ED) in which 4-hydroxyphenylethylidene bisphosphonate is linked to hydroxypipexone (ED) by a carbamate, wherein BP is 4-hydroxyphenylethylidene bisphosphonate, L is-nc(s) O-, and ED is hydroxypipexone. The bisphosphonate-antimicrobial agent may consist of a general formula (BP-L-ED) in which 4-hydroxyphenylethylidene bisphosphonate is linked to hydroxypipexone (ED) by a carbonate, wherein BP is 4-hydroxyphenylethylidene bisphosphonate, L is-oc (O) O-, and ED is hydroxypipexone. The bisphosphonate-antimicrobial agent may consist of general formula (BP-L-dED) where 4-hydroxyphenylethylidene bisphosphonate is linked to deacetylhydroxypipexone (dED) by a carbamate, where BP is 4-hydroxyphenylethylidene bisphosphonate, L is-oc (o) N-, and dED is deacetylhydroxypipexone. The bisphosphonate-antimicrobial agent may consist of general formula (BP-L-dED) wherein BP is 4-hydroxyphenylethylidene bisphosphonate, L is-oc(s) -N-or-sc (o) N-or-sc(s) N-, and dED is deacetylhydroxypipexone, linking 4-hydroxyphenylethylidene bisphosphonate to deacetylhydroxypipexone (dED) via a carbamate. The bisphosphonate-antimicrobial agent may consist of a general formula (BP-L-dED) in which 4-aminophenylethylidene bisphosphonate is linked to deacetylhydroxypipexone (dED) by urea, where BP is 4-aminophenylethylidene bisphosphonate, L is-nc (o) N-or-nc(s) N-, and dED is deacetylhydroxypipexone. The bisphosphonate-antimicrobial agent may consist of a general formula (BP-L-dSD) in which 4-hydroxyphenylethylidene bisphosphonate is linked to deacetylated sutazolid (dSD) by carbamate, where BP is 4-hydroxyphenylethylidene bisphosphonate, L is-oc (o) N-, and dSD is deacetylated sutazolid. The bisphosphonate-antimicrobial agent may consist of a general formula (BP-L-dSD) in which 4-hydroxyphenylethylidene bisphosphonate is linked to deacetylated sutazolid (dSD) by carbamate, wherein BP is 4-hydroxyphenylethylidene bisphosphonate, L is-OC (S) N-or-SC (O) N-or-SC (S) N-, and dSD is deacetylated sutazolid. The bisphosphonate-antimicrobial agent may consist of a general formula (BP-L-dSD) in which 4-aminophenylethylidene bisphosphonate is linked to deacetylated sutazolid (dSD) by urea, wherein BP is 4-aminophenylethylidene bisphosphonate, L is-nc (o) N-or-nc(s) N-, and dSD is deacetylated sutazolid. The bisphosphonate-antimicrobial agent may consist of a general formula (BP-L-RD) in which 4-aminophenylethylidene bisphosphonate is linked to ladzolamide (dRD) by carbamate, wherein BP is 4-aminophenylethylidene bisphosphonate, L is-OC (O) N-, and RD is ladzolamide. The bisphosphonate-antimicrobial agent may consist of a general formula (BP-L-RD) in which 4-aminophenylethylidene bisphosphonate is linked to ladzolamide (dRD) by carbamate, wherein BP is 4-aminophenylethylidene bisphosphonate, L is-OC (S) -or-SC (O) N-or-SC (S) N-, and RD is ladzolamide. The bisphosphonate-antimicrobial agent may consist of a general formula (BP-L-RD) in which 4-aminophenylethylidene bisphosphonate is linked to deacetyl radizole (dRD) by urea, wherein BP is 4-aminophenylethylidene bisphosphonate, L is-NC (O) N-or-NC (S) N-, and RD is radizole. The bisphosphonate-antimicrobial agent may consist of general formula (BP-L-dRD) wherein BP is 4-aminophenylethylidene bisphosphonate, L is-oc (o) N-, and dRD is deacetyl-ridazole, linking the 4-aminophenylethylidene bisphosphonate to deacetyl-ridazole (dRD) via a carbamate. The bisphosphonate-antimicrobial agent may consist of general formula (BP-L-dRD) wherein BP is 4-aminophenylethylidene bisphosphonate, L is-oc(s) -N-or-sc (o) N-or-sc(s) N-, and dRD is deacetyl-ridazole, linking the 4-aminophenylethylidene bisphosphonate to deacetyl-ridazole (dRD) via a carbamate. The bisphosphonate-antimicrobial agent may consist of general formula (BP-L-dRD) in which 4-aminophenylethylidene bisphosphonate is linked to deacetyl radizole (dRD) by urea, where BP is 4-aminophenylethylidene bisphosphonate, L is-nc (o) N-or-nc(s) N-, and dRD is deacetyl radizole. The bisphosphonate-antimicrobial agent may consist of a general formula (BP-L-dRbD) in which 4-hydroxyphenylethylidene bisphosphonate is linked to deacetylated rankzolid (dRbD) by carbamate, where BP is 4-hydroxyphenylethylidene bisphosphonate, L is-oc (o) N-, and dRbD is deacetylated rankzolid. The bisphosphonate-antimicrobial agent may consist of a compound of the general formula (BP-L-dRbD) wherein BP is 4-hydroxyphenylethylidene bisphosphonate, L is-OC (S) N-or-SC (O) N-or-SC (S) N-, and dRbD is deacetylated rankolinylid by carbamate linking 4-hydroxyphenylethylidene bisphosphonate to deacetylated rankylid (dRbD). The bisphosphonate-antimicrobial agent may consist of a compound of the general formula (BP-L-dRbD) in which 4-aminophenylethylidene bisphosphonate is linked to deacetylated rankzotide (dRbD) by urea, wherein BP is 4-aminophenylethylidene bisphosphonate, L is-NC (O) N-or-NC (S) N-, and dRbD is deacetylated rankzotide. The bisphosphonate-antimicrobial agent may consist of a general formula (BP-L-PD) in which 4-aminophenylethylidene bisphosphonate is linked to Pasiclazole (PD) through a carbamate, wherein BP is 4-aminophenylethylidene bisphosphonate, L is-nc (O) O-, and PD is pasiclazole. The bisphosphonate-antimicrobial agent may consist of a general formula (BP-L-PD) in which 4-aminophenylethylidene bisphosphonate is linked to Pasiclazole (PD) through a carbamate, wherein BP is 4-aminophenylethylidene bisphosphonate, L is-nc(s) O-, and PD is pasiclazole. The bisphosphonate-antimicrobial agent may consist of a general formula (BP-L-PD) in which 4-hydroxyphenylethylidene bisphosphonate is linked to Pasiclazole (PD) by a carbonate, wherein BP is 4-hydroxyphenylethylidene bisphosphonate, L is-oc (O) O-, and PD is pasiclazole. The bisphosphonate-antimicrobial agent may be as substantially described by any one of the conjugates in figure 7 herein.

In any one or more embodiments herein, the bisphosphonate-antimicrobial agent of any one or more aspects herein may be useful or may be used in the preparation of a formulation for the treatment of bone diseases. The bone infectious disease may be selected from the group consisting of: osteomyelitis, osteolytic bone infection, osteonecrosis, diabetic chronic osteomyelitis, diabetic foot, periodontitis and other jaw bone infections. The bisphosphonate-antimicrobial agent may be used to treat surroundings associated and not associated with bone-related infections (non-bone infections).

In any one or more embodiments herein, the aforementioned bisphosphonate-antimicrobial agent of any one or more aspects herein may be configured to deliver and release a target molecule at a bone site. Formulations comprising a bisphosphonate-antimicrobial agent as described in any one or more aspects herein may be provided in the form of: oral irrigation solutions, buffer solutions for intravenous or parenteral use, or microspheres in the form of powders/tablets for enteral administration or placed directly at the site of bone infection during surgery. Formulations including the bisphosphonate-antimicrobial agent may include an effective amount of the bisphosphonate-antimicrobial agent for treating bone infectious diseases.

In any one or more embodiments herein, there is provided a bone graft composition comprising: a bone graft material and the bisphosphonate-antimicrobial agent, wherein the bisphosphonate-antimicrobial agent is attached, bound, chemisorbed or mixed with the bone graft material. The bone graft material may be an autograft bone material, an allograft bone material, a xenograft bone material, a synthetic bone graft material, or any combination thereof.

In any one or more embodiments herein, there is provided a method comprising: a method of implanting the foregoing bone graft composition into a subject in need thereof.

In any one or more embodiments herein, there is provided a method of preventing biofilm infection at a bone or surgical site of implantation or bone grafting, wherein the method comprises: administering a bisphosphonate-antimicrobial agent as described in any one or more aspects herein to a subject in need thereof, or wherein the method comprises: implanting the bone graft composition into a subject in need thereof.

Other compounds, compositions, conjugates, formulations, methods, features and advantages of the disclosure will be or become apparent to one with skill in the art upon examination of the following figures and detailed description. It is intended that all such additional systems, methods, features and advantages be included within this description, be within the scope of the present disclosure, and be protected by the accompanying claims.

Drawings

Further aspects of the present disclosure will be more readily understood after a review of the following detailed description of various embodiments of the disclosure taken in conjunction with the accompanying drawings.

Figure 1 depicts exemplary BP-TD conjugates and tedizolid release mechanisms of the present disclosure.

Figure 2 depicts an example of pharmacologically inert BP for conjugation: medium (A/E), high (B/F) and low (C/G) affinity BP and longer phenyl alkyl chain BP (D/H).

Figure 3 depicts an example of a pharmacologically active BP that can be used in the conjugates of the invention.

Figure 4A depicts α -hydroxy modified risedronic acid and zoledronic acid; and fig. 4B depicts 1) bp modified by substitution or removal of the a-hydroxy group (ppyrebp); 2) BP (p-RIS) modified by substitution at the para position of the pyridine ring. The circled H is attached to the alpha carbon of the bisphosphonate-substituted carbon chain.

Figure 5 depicts an example of an oxazolidinone antibacterial agent that can be used to conjugate with bisphosphonic acids.

Figure 6 depicts an example of a bisphosphonate-oxazolidinone conjugate of the present disclosure.

FIG. 7 depicts an exemplary synthesis of a BP-L-TD (L: -OC (O) N-) conjugate.

FIG. 8 depicts an exemplary synthesis of a BP-L-dLD (L: -OC (O) N-) conjugate.

FIG. 9 depicts an exemplary synthesis of a BP-L-TD (L: -OC (O) O-) conjugate.

FIG. 10 depicts an exemplary synthesis of a BP-L-dLD (L: -NC (O) N-) conjugate.

FIG. 11 depicts an exemplary synthesis of a BP-L-TD (L: -OC (S) N-) conjugate.

FIG. 12 depicts an exemplary synthesis of a BP-L-dLD (L: -OC (S) N-) conjugate.

FIG. 13 depicts an exemplary synthesis of a BP-L-dLD (L: -SC (O) N-) conjugate.

FIG. 14 depicts an exemplary synthesis of a BP-L-dLD (L: -SC (S) N-) conjugate.

FIG. 15 depicts an exemplary synthesis of a BP-L-dLD (L: -NC (S) N-) conjugate.

Figure 16 depicts the results of a prophylactic Hydroxyapatite (HA) experiment with test compounds against staphylococcus aureus.

Figure 17 depicts the results of HA eradication experiments with test compounds against staphylococcus aureus biofilms.

Figure 18A is a picture of rat femurs saturated with antibiotics or conjugates after introduction into a solution of staphylococcus aureus in our ex vivo prophylactic experimental setup. The more red the bone, the more staphylococcal biofilm is attached to the surface of these areas. Femoral 1-negative control sample (no antimicrobial, no staphylococcus); 2-positive control sample (no added antimicrobial, staphylococcus solution); 3-bone saturated with tedizolid and introducing the bone into a staphylococcal solution; 4-bone saturated with ciprofloxacin and introduction of bone into staphylococcal solution; 5-bone saturated with BET conjugate (BP-L-TD, L: -C (O) O-, as exemplified in FIG. 7 continuation (19)) and introduced into the staphylococcal solution; and figure 18B depicts the results of the staphylococcus aureus biofilm prevention and eradication settings, showing that the conjugate BET significantly prevented or eradicated biofilm growth in the isolated femur, as did tedizolid alone, with the maximum efficacy observed with the BET conjugate in both settings (K-W test, × p < 0.01; comparative control).

Detailed Description

Before the present disclosure is described in more detail, it is to be understood that this disclosure is not limited to particular embodiments described, and as such may, of course, vary. It is also to be understood that the terminology used herein is for the purpose of describing particular embodiments only and is not intended to be limiting.

Where a range of values is provided, it is understood that each intervening value, to the tenth of the unit of the lower limit unless the context clearly dictates otherwise, between the upper and lower limit of that range and any other stated or intervening value in that stated range is encompassed within the disclosure. The upper and lower limits of these smaller ranges may independently be included in the smaller ranges, and are also encompassed within the disclosure, subject to any specifically excluded limit in the stated range. Where the range includes one or both of the limits, ranges excluding either or both of those included limits are also included in the disclosure.

Unless defined otherwise, all technical and scientific terms used herein have the same meaning as commonly understood by one of ordinary skill in the art to which this disclosure belongs. Although any methods and materials similar or equivalent to those described herein can also be used in the practice or testing of the present disclosure, the preferred methods and materials are now described.

All publications and patents cited in this specification are herein incorporated by reference as if each individual publication or patent were specifically and individually indicated to be incorporated by reference and were incorporated by reference to disclose and describe the methods and/or materials in connection with which the publications were cited. The citation of any publication is a publication prior to the filing date thereof and should not be construed as an admission that the present disclosure is not entitled to antedate such publication by virtue of prior publication. Further, the dates of publication provided may be different from the actual publication dates which may need to be independently confirmed.

It will be apparent to those of skill in the art upon reading this disclosure that each of the individual embodiments described and illustrated herein has discrete components and features which may be readily separated from or combined with the features of any of the other several embodiments without departing from the scope or spirit of the present disclosure. Any listed methods may be performed in the order of the events listed or in any other order that is logically possible.

Unless otherwise indicated, embodiments of the present disclosure will employ techniques in the art of molecular biology, microbiology, nanotechnology, pharmacology, organic chemistry, biochemistry, botany, and the like. These techniques are fully described in the literature.

Definition of

Unless otherwise stated herein, the following definitions are provided.

As used herein, "about", "approximately", and the like, when used in connection with a numerical variable, generally refer to the value of the variable as well as all values of the variable that are within experimental error (e.g., within 95% confidence interval of the mean) or within ± 10% of the specified value (whichever is larger).

As used interchangeably herein, "subject," "individual," or "patient" refers to a vertebrate, preferably a mammal, more preferably a human. Mammals include, but are not limited to, murines, simians, humans, domestic animals, sports animals, and pets. The term "pet" includes dogs, cats, guinea pigs, mice, rats, rabbits, ferrets, and the like. The term "livestock" includes horses, sheep, goats, chickens, pigs, cattle, donkeys, llamas, alpacas, turkeys, and the like.

As used herein, "control" may refer to an alternative subject or sample used in an experiment for comparison purposes and is included to minimize or distinguish the effects of variables other than independent variables.

As used herein, an "analog" or "analog" such as an analog of a bisphosphonate, as described herein, may refer to a member that is structurally similar to the parent molecule or an additional parent molecule, such as a bisphosphonate.

As used herein, "conjugated" may refer to two or more compounds directly attached to each other by one or more covalent or non-covalent bonds. The term "conjugated" as used herein may also refer to two or more compounds indirectly attached to each other through an intermediate compound (such as a linker).

As used herein, "pharmaceutical formulation" refers to the combination of an active agent, compound or ingredient with a pharmaceutically acceptable carrier or excipient, making the composition suitable for diagnostic, therapeutic or prophylactic use in vitro, in vivo or ex vivo.

As used herein, "pharmaceutically acceptable carrier or excipient" refers to a carrier or excipient that is generally safe, non-toxic, and not biologically or otherwise undesirable for use in the preparation of pharmaceutical formulations, and includes carriers or excipients that are acceptable for veterinary as well as human pharmaceutical use. As used in the specification and claims, "pharmaceutically acceptable carrier or excipient" includes both one and more than one such carrier or excipient.

As used herein, "pharmaceutically acceptable salt" refers to any acid or base addition salt, the counter ion of which is non-toxic to a subject administered a pharmaceutical dose of the salt.

As used herein, "active agent" or "active ingredient" refers to one or more components of a composition to which all or part of the effect of the composition is attributed.

As used herein, "dose," "unit dose," or "amount" refers to discrete units suitable for use on the body of a subject, each unit comprising a predetermined amount of a BP conjugate, such as a BP quinolone conjugate, composition, or formulation described herein, calculated to produce a desired response or a response associated with administration thereof.

As used herein, "derivative" refers to any compound having the same or similar core structure as the compound, but with at least one structural difference, including substitution, deletion, and/or addition of one or more atoms or functional groups. The term "derivative" does not imply that the derivative is synthesized from the parent compound as a starting material or intermediate, although this may be the case. The term "derivative" can include prodrugs or metabolites of the parent compound. The derivatives include compounds in which the free amino groups in the parent compound have been derivatized to form amine salts, p-toluenesulfonamides, benzyloxycarboxamides, t-butoxycarboxamides, ethyl thiocarbamate derivatives, trifluoroacetamides, chloroacetamides or carboxamides. Derivatives include compounds in which the carboxyl group of the parent compound has been derivatized to form methyl and ethyl esters or other types of esters, amides, hydroxamic acids or hydrazides. Derivatives include those in which the hydroxy group in the parent compound has been derivatized to form an O-acyl, O-carbamoyl or O-alkyl derivative. Derivatives include compounds in which a hydrogen bond donor group in the parent compound is replaced by another hydrogen bond donor group (such as OH, NH or SH). Derivatives include the replacement of a hydrogen bond accepting group in the parent compound with another hydrogen bond accepting group such as esters, ethers, ketones, carbonates, tertiary amines, imines, thiones, sulfones, tertiary amides, and sulfides. "derivatives" also include extensions such as the replacement of the cyclopentane ring with saturated or unsaturated cyclohexane or other more complex, e.g., nitrogen-containing, rings, as well as extensions of these rings with various groups.

As used herein, "administering" means orally, topically, intravenously, subcutaneously, transdermally, intramuscularly, intra-articularly, parenterally, intra-arteriolar, intradermally, intraventricularly, intracranially, intraperitoneally, intralesionally, intranasally, rectally, vaginally, by inhalation, or by implantable drug depot. The term "parenteral" includes subcutaneous, intravenous, intramuscular, intraarticular, intrasynovial, intrasternal, intrathecal, intrahepatic, intralesional and intracranial injection or infusion techniques.

The term "substituted" as used herein refers to all permissible substituents of the compounds described herein. In a broad sense, the permissible substituents include acyclic and cyclic, branched and unbranched, carbocyclic and heterocyclic, aromatic and nonaromatic substituents of organic compounds. Exemplary substituents include, but are not limited to, halogen, hydroxyl group, or any other organic group containing any number of carbon atoms (e.g., 1-14 carbon atoms), and also optionally include one or more heteroatoms, such as oxygen, sulfur, or nitrogen, grouped in linear, branched, or cyclic structural formats. Representative substituents include alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, phenyl, substituted phenyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, halogen, hydroxy, alkoxy, substituted alkoxy, phenoxy, substituted phenoxy, aryloxy, substituted aryloxy, alkylthio, substituted alkylthio, phenylthio, substituted phenylthio, arylthio, substituted arylthio, phenyl, substituted phenyl, phenyl,Cyano, isocyano, substituted isocyano, carbonyl, substituted carbonyl, carboxyl, substituted carboxyl, amino, substituted amino, amido, substituted amido, sulfonyl, substituted sulfonyl, sulfonic acid, phosphoryl, substituted phosphoryl, phosphonyl, substituted phosphonyl, polyaryl, substituted polyaryl, C3-C20Cyclic, substituted C3-C20Rings, heterocycles, substituted heterocycles, amino acids, peptides and polypeptide groups.

As used herein, "substituent" or "suitable substituent" means a chemically and pharmaceutically acceptable group, i.e., a moiety that does not significantly interfere with the preparation of, or negate the efficacy of, the compounds of the present invention. Such suitable substituents may be routinely selected by those skilled in the art. Suitable substituents include, but are not limited to, the following: halogen, C1-C6Alkyl radical, C2-C6Alkenyl radical, C1-C6Haloalkyl, C1-C6Alkoxy radical, C1-C6Haloalkoxy, C2-C6Alkynyl, C3-C8Cycloalkenyl group, (C)3-C8Cycloalkyl) C1-C6Alkyl, (C)3-C8Cycloalkyl) C2-C6Alkenyl, (C)3-C8Cycloalkyl) C1-C6Alkoxy radical, C3-C7Heterocycloalkyl group, (C)3-C7Heterocycloalkyl) C1-C6Alkyl, (C)3-C7Heterocycloalkyl) C2-C6Alkenyl, (C)3-C7Heterocycloalkyl) C1-C6Alkoxy, hydroxy, carboxy, oxo, sulfonyl, C1-C6Alkylsulfonyl, aryl, heteroaryl, aryloxy, heteroaryloxy, aralkyl, heteroaralkyl, aralkoxy, heteroaralkoxy, nitro, cyano, amino, C1-C6Alkylamino, di- (C)1-C6Alkyl) amino, carbamoyl, (C)1-C6Alkyl) carbonyl (C)1-C6Alkoxy) carbonyl (C)1-C6Alkyl) aminocarbonyl, di- (C)1-C6Alkyl) aminocarbonyl, arylcarbonyl, aryloxycarbonyl, (C)1-C6Alkyl) sulfonyl and arylsulfonyl. The groups listed above as suitable substituents are as defined below, except that suitable substituents may not be further optionally substituted.

The term "alkyl" refers to the radical of a saturated aliphatic group (i.e., an alkane from which one hydrogen atom has been removed), including straight-chain alkyl groups, branched-chain alkyl groups, cycloalkyl (alicyclic) groups, alkyl-substituted cycloalkyl groups, and cycloalkyl-substituted alkyl groups.

In some embodiments, the linear or branched alkyl group may have 30 or fewer carbon atoms in its backbone (e.g., C for linear chain)1-C30And for the side chain is C3-C30). In other embodiments, the linear or branched alkyl group may contain 20 or fewer, 15 or fewer, or 10 or fewer carbon atoms in its backbone. Likewise, in some embodiments, cycloalkyl groups may have 3 to 10 carbon atoms in their ring structure. In some of these embodiments, the cycloalkyl group can have 5, 6, or 7 carbons in the ring structure.

The term "alkyl" (or "lower alkyl") as used herein is intended to include both "unsubstituted alkyls" and "substituted alkyls," the latter of which refers to alkyl moieties having one or more substituents replacing a hydrogen on one or more carbons of the hydrocarbon backbone. Such substituents include, but are not limited to, halogen, hydroxyl, carbonyl (such as carboxyl, alkoxycarbonyl, formyl, or acyl), thiocarbonyl (such as thioester, thioacetate, or thioformate), alkoxy, phosphoryl, phosphate, phosphonate, phosphinate, amino, amide, amidine, imine, cyano, nitro, azido, sulfhydryl, alkylthio, sulfate, sulfonate, sulfamoyl, sulfonamide, sulfonyl, heterocyclyl, aralkyl, or an aromatic or heteroaromatic moiety.

As used herein, "lower alkyl" means an alkyl group as defined above, but having one to ten carbons in its backbone structure, unless the number of carbons is otherwise specified. Likewise, "lower alkenyl" and "lower alkynyl" have similar chain lengths.

It will be appreciated by those skilled in the art that the moiety substituted on the hydrocarbon chain may itself be substituted, if appropriate. For example, substituents of substituted alkyl groups may include halogen, hydroxy, nitro, thiol, amino, azido, imino, amido, phosphoryl (including phosphonato and phosphonato), sulfonyl (including sulfate, sulfonamido, sulfamoyl and sulfonate), and silyl groups, as well as ether, alkylthio, carbonyl (including ketones, aldehydes, carboxylic acid groups and esters), -CF3CN, -CN, etc. Cycloalkyl groups may be substituted in the same manner.

The term "heteroalkyl," as used herein, refers to a straight or branched chain or cyclic carbon-containing group, or a combination thereof, that contains at least one heteroatom. Suitable heteroatoms include, but are not limited to, O, N, Si, P, Se, B, and S, wherein the phosphorus and sulfur atoms are optionally oxidized, and the nitrogen heteroatom is optionally quaternized. The heteroalkyl group may be substituted as defined above for the alkyl group.

The term "alkylthio" refers to an alkyl group as defined above having a thio group attached thereto. In a preferred embodiment, an "alkylthio" moiety is represented by one of-S-alkyl, -S-alkenyl, and-S-alkynyl. Representative alkylthio groups include methylthio, ethylthio, and the like. The term "alkylthio" also includes cycloalkyl groups, alkene and cycloalkene groups, and alkyne groups. "arylthio" refers to an aryl or heteroaryl group. The arylthio group may be substituted as defined above for the alkyl group.

The terms "alkenyl" and "alkynyl" refer to unsaturated aliphatic groups that are similar in length and possible substitution as alkyl groups described above, but each contain at least one double or triple bond.

The term "alkoxy" or "alkoxy" as used herein refers to an alkyl group as defined above having an oxy group attached thereto. Representative alkoxy groups include methoxy, ethoxy, propoxy, tert-butoxy and the like. An "ether" is two hydrocarbons covalently linked by oxygen. Thus, substituents that render an alkyl group as an ether or an alkyl group similar to an alkoxy group, such as may be represented by one of-O-alkyl, -O-alkenyl, and-O-alkynyl. The terms "aryloxy" and "aryloxy", as used interchangeably herein, may be represented by-O-aryl or O-heteroaryl, wherein aryl and heteroaryl are defined as follows. Alkoxy and aryloxy groups may be substituted as described above for alkyl groups.

The terms "amine" and "amino" (and protonated forms thereof) are well known in the art and refer to both unsubstituted and substituted amines, e.g., moieties that can be represented by the general formula:

wherein R, R 'and R' each independently represent hydrogen, alkyl, alkenyl, (CH2)m-RCOr R and R' together with the N atom to which they are attached form a heterocyclic ring having from 4 to 8 atoms in the ring structure; rCRepresents aryl, cycloalkyl, cycloalkenyl, heterocycle or polycycle; and m is zero or an integer in the range of 1 to 8. In some embodiments, only one of R or R 'may be a carbonyl group, e.g., R, R' and the nitrogen together do not form an imide. In other embodiments, the term "amine" does not include amides, e.g., where one of R and R' represents a carbonyl group. In a further embodiment, R and R' (and optionally R ") each independently represent hydrogen, alkyl or cycloalkyl, alkenyl or cycloalkenyl or alkynyl. Thus, the term "alkylamine" as used herein means an amine group as defined above having a substituted (as described above for alkyl) or unsubstituted alkyl group attached thereto, i.e. at least one of R and R' is an alkyl group.

The term "amido" is known in the art as an amino-substituted carbonyl and includes moieties that can be represented by the general formula:

wherein R and R' are as defined above.

As used herein, "aryl" refers to C5-C10-aromatic, heterocyclic, fused aromatic, fused heterocyclic, biaromatic or diheterocyclic systems. As broadly defined, "aryl" as used herein includes 5-, 6-, 7-, 8-, 9-, and 10-membered monocyclic aromatic groups that may include from zero to four heteroatoms, such as benzene, pyrrole, furan, thiophene, imidazole, oxazole, thiazole, triazole, pyrazole, pyridine, pyrazine, pyridazine, pyrimidine, and the like. Those aryl groups having heteroatoms in the ring structure may also be referred to as "aryl heterocycles" or "heteroaromatics". The aromatic ring may be substituted at one or more ring positions with one or more substituents including, but not limited to, halogen, azide, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxy, alkoxy, amino (or quaternized amino), nitro, mercapto, imino, amido, phosphonato, carbonyl, carboxy, silyl, ether, alkylthio, sulfonyl, sulfonamido, ketone, aldehyde, ester, heterocyclyl, aromatic or heteroaromatic moieties, -CF3CN, -CN, and combinations thereof. The term "aryl" includes phenyl.

The term "aryl" also includes polycyclic ring systems having two or more cyclic rings in which two or more carbons are common to two adjoining rings (i.e., "fused rings"), wherein at least one of the rings is aromatic, e.g., the other cyclic ring or rings can be cycloalkyl, cycloalkenyl, cycloalkynyl, aryl, and/or heterocyclic. Examples of heterocycles include, but are not limited to, benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, benzoxazolinyl, benzothiazolyl, benzotriazolyl, benzotetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolinyl, carbazolyl, 4aH carbazolyl, carbolinyl, chromanyl, benzopyranyl, cinnolinyl, decahydroquinolinyl, 2H,6H-1,5, 2-dithiazinyl, dihydrofuro [2,3b ] tetrahydrofuran, furanyl, furazanyl, imidazolidinyl, imidazolinyl, imidazolyl, 1H-indazolyl, pseudoindolyl (indolynyl), indolinyl, indolizinyl, indolyl, 3H-indolyl, isatinoyl (isatinoyl), isobenzofuranyl, isobenzodihydropyranyl, isoindolinyl, isoindolyl, isoquinolyl, isothiazolyl, isoxazolyl, methylenedioxyphenyl, morpholinyl, naphthyridinyl, octahydroisoquinolyl, oxadiazolyl, 1,2, 3-oxadiazolyl, 1,2, 4-oxadiazolyl, 1,2, 5-oxadiazolyl, 1,3, 4-oxadiazolyl, oxazolidinyl, oxazolyl, oxindolyl, pyrimidinyl, phenanthridinyl, phenanthrolinyl, phenazinyl, phenothiazinyl, phenoxathiin, phenoxazinyl, phthalazinyl, piperazinyl, piperidinyl, piperidonyl, 4-piperidonyl, piperonyl, pteridinyl, purinyl, pyranyl, pyrazinyl, pyrazolidinyl, pyrazolinyl, pyrazolyl, pyridazinyl, pyridooxazole, pyridoimidazole, pyridothiazole, pyridyl (pyridinyl), pyridyl (pyridylal), pyrimidinyl, pyrrolidinyl, pyrrolinyl, pyrazolinyl, pyrazolidinyl, pyridylal, and the like, 2H-pyrrolyl, quinazolinyl, quinolinyl, 4H-quinolizinyl, quinoxalinyl, quinuclidinyl, tetrahydrofuryl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, tetrazolyl, 6H-1,2, 5-thiadiazolyl, 1,2, 3-thiadiazolyl, 1,2, 4-thiadiazolyl, 1,2, 5-thiadiazolyl, 1,3, 4-thiadiazolyl, thianthrenyl, thiazolyl, thienyl, thienothiazolyl, thienooxazolyl, thienoimidazolyl, thienyl, and xanthenyl. One or more of the rings may be substituted as defined above for "aryl".

The term "aralkyl" as used herein refers to an alkyl group substituted with an aryl group (e.g., an aromatic or heteroaromatic group).

The term "aralkoxy" may be represented by-O-aralkyl, wherein aralkyl is as defined above.

The term "carbocycle" as used herein refers to one or more aromatic or non-aromatic rings in which each atom of the ring or rings is carbon.

As used herein, "heterocycle" or "heterocyclic" refers to a monocyclic or bicyclic structure comprising 3 to 10 ring atoms (and in some embodiments 5 to 6 ring atoms) wherein the ring atoms are carbon and one to four heteroatoms each selected from the following group: non-peroxidized oxygen, sulfur and N (Y), wherein Y is absent or is H, O, (C)1-C10) Alkyl, phenyl or benzylOptionally containing 1-3 double bonds, and optionally substituted with one or more substituents. Examples of heterocycles include, but are not limited to, benzimidazolyl, benzofuranyl, benzothiofuranyl, benzothiophenyl, benzoxazolyl, benzoxazolinyl, benzothiazolyl, benzotriazolyl, benzotetrazolyl, benzisoxazolyl, benzisothiazolyl, benzimidazolinyl, carbazolyl, 4aH carbazolyl, carbolinyl, chromanyl, benzopyranyl, cinnolinyl, decahydroquinolinyl, 2H,6H-1,5, 2-dithiazinyl, dihydrofuro [2,3b ] group]Tetrahydrofuran, furyl, furazanyl, imidazolidinyl, imidazolinyl, imidazolyl, 1H-indazolyl, isoindolyl, indolinyl, indolizinyl, indolyl, 3H-indolyl, isatoiyl, isobenzofuryl, isobenzodihydropyranyl, isoindolyl, isoindolinyl, isoindolyl, isothiazolyl, isoxazolyl, methylenedioxyphenyl, morpholinyl, naphthyridinyl, octahydroisoquinolyl, oxadiazolyl, 1,2, 3-oxadiazolyl, 1,2, 4-oxadiazolyl, 1,2, 5-oxadiazolyl, 1,3, 4-oxadiazolyl, oxazolidinyl, oxazolyl, oxepanyl, oxetanyl, oxindolyl, pyrimidinyl, phenanthridinyl, phenanthrolinyl, phenazinyl, phenothiazinyl, phenoxazinyl, phenazinyl, phthalazinyl, isoquinolyl, isothiazolyl, isoxazolyl, methylindolyl, morpholinyl, isoquinolyl, isothiazolyl, isoxazolyl, 1, 4-oxadiazolyl, morpholinyl, naphthyridinyl, octahydroisoquinolyl, oxadiazolyl, and phenanthridinyl, Piperazinyl, piperidinyl, piperidonyl, 4-piperidonyl, piperonyl, pteridinyl, purinyl, pyranyl, pyrazinyl, pyrazolidinyl, pyrazolinyl, pyrazolyl, pyridazinyl, pyridooxazole, pyridoimidazole, pyridothiazole, pyridyl (pyridinyl), pyridyl (pyridil), pyrimidinyl, pyrrolidinyl, pyrrolinyl, 2H-pyrrolyl, quinazolinyl, quinolinyl, 4H-quinolizinyl, quinoxalinyl, quinuclidinyl, tetrahydrofuranyl, tetrahydroisoquinolinyl, tetrahydroquinolinyl, tetrazolyl, 6H-1,2, 5-thiadiazinyl, 1,2, 3-thiadiazolyl, 1,2, 4-thiadiazolyl, 1,2, 5-thiadiazolyl, 1,3, 4-thiadiazolyl, thianthrenyl, thiazolyl, thienyl, pyranyl, pyrazinyl, pyrazolidinyl, quinazolinyl, quinolizyl, quinoliz, Thienothiazolyl, thienooxazolyl, thienoimidazolyl, thienyl and xanthyl groups. The heterocyclic group may be optionally substituted at one or more positions with one or more substituents as defined above for alkyl and aryl groupsFor example, halogen, alkyl, aralkyl, alkenyl, alkynyl, cycloalkyl, hydroxy, amino, nitro, mercapto, imino, amido, phosphate, phosphonate, phosphinate, carbonyl, carboxyl, silyl, ether, alkylthio, sulfonyl, ketone, aldehyde, ester, heterocyclyl, aromatic or heteroaromatic moieties, -CF3CN, -CN, etc. The term "heterocycle" or "heterocyclic" may be used to describe a compound that may include a heterocycle or heterocyclic ring.

The term "carbonyl" is art-recognized and includes such moieties as may be represented by the general formula:

wherein X is a bond or represents oxygen or sulphur and R' are as defined above. When X is oxygen and R or R' is not hydrogen, the formula represents an "ester". When X is oxygen and R is as defined above, the moiety is referred to herein as a carboxyl group, and in particular when R is hydrogen, the formula represents a "carboxylic acid". When X is oxygen and R' is hydrogen, the formula represents "formate". Typically, when the oxygen atom in the above formula is replaced by sulfur, the formula represents a "thiocarbonyl" group. When X is sulfur and R or R' is not hydrogen, the formula represents a "thioester". When X is sulfur and R is hydrogen, the formula represents a "thiocarboxylic acid". When X is sulfur and R' is hydrogen, the formula represents "thiocarboxylate". On the other hand, when X is a bond and R is not hydrogen, the above formula represents a "ketone" group. When X is a bond and R is hydrogen, the above formula represents an "aldehyde" group.

The term "heteroatom" as used herein means an atom of any element other than carbon or hydrogen. Exemplary heteroatoms include, but are not limited to, boron, nitrogen, oxygen, phosphorus, sulfur, silicon, arsenic, and selenium. Heteroatoms, such as nitrogen, may have hydrogen substituents and/or any permissible substituents of organic compounds described herein that satisfy the valencies of the heteroatoms. It is understood that the implication of "substituted" or "substituted" is that such substitution is in accordance with the allowed valences of the atoms and substituents being replaced, and that the substitution results in a stable compound, i.e. a compound that does not spontaneously undergo a transformation such as rearrangement, cyclization, elimination, etc.

The term "hydroxy" refers to the "-OH" group.

As used herein, the term "nitro" refers to-NO2(ii) a The term "halogen" means-F, -Cl, -Br, or-I; the term "mercapto" refers to-SH; the term "hydroxy" refers to-OH; and the term "sulfonyl" refers to-SO2-。

As used herein, "carbamate" may be used to refer to a compound derived from carbamic acid (NH)2COOH) and may include carbamates. "carbamates" may have the following general structure:

wherein R is1、R2And R3May be any permissible substituents.

As used herein, "carbonate" may be used to refer to a compound derived from carbonic acid (H)2CO3) Derivatized compounds, and may include carbonates. "carbonates" can have the following general structure:

as used herein, an "effective amount" may refer to an amount of a composition described herein or a pharmaceutical formulation described herein that will elicit the desired biological or medical response of a tissue, system, animal, plant, protozoan, bacteria, yeast or human that is being sought by a researcher, veterinarian, medical doctor or other clinician. The desired biological response may be modulation of bone formation and/or remodeling, including but not limited to modulation of bone resorption and/or uptake of a BP conjugate (such as a BP quinolone conjugate described herein). The effective amount will vary depending on the exact chemical structure of the composition or pharmaceutical preparation, the pathogen being treated or prevented and/or the severity of its infection, disease, disorder, syndrome or symptom, the route of administration, time of administration, rate of excretion, drug combination, the judgment of the treating physician, the dosage form, and the age, weight, general health, sex, and/or diet of the subject to be treated. An "effective amount" can refer to an amount of a composition described herein that is effective to inhibit the growth or reproduction of a microorganism (including, but not limited to, a bacterium or population thereof). An "effective amount" can refer to an amount of a composition described herein that kills a microorganism (including, but not limited to, a bacterium or population thereof). An "effective amount" can refer to an amount of a composition described herein that is effective to treat and/or prevent osteomyelitis in a subject in need thereof.

The terms "oxazolidinone antimicrobial molecule," "oxazolidinone antimicrobial agent," or "substituent" or "derivative" thereof, and related terms have the same meaning and refer to an antimicrobial agent that is part of the well known class of "oxazolidinones," as well as "oxazolidinones," as described in more detail herein.

As used herein, "therapeutic" may generally refer to treating, curing and/or ameliorating a disease, condition, disorder or side effect, or reducing the rate of progression of a disease, condition, disorder or side effect. The term also includes within its scope enhancement of normal physiological function, palliative treatment, and partial remediation of its disease, disorder, condition, side effect, or symptom.

The term "antibacterial" includes those compounds that inhibit, prevent or reverse bacterial growth, those compounds that inhibit, prevent or reverse the activity of bacterial enzymes or biochemical pathways, those compounds that kill or damage bacteria, and those compounds that block or slow the development of bacterial infections.

As used herein, the terms "treating" and "treatment" may generally refer to obtaining a desired pharmacological and/or physiological effect. The effect may be prophylactic in terms of preventing or partially preventing a disease, symptom or condition thereof. And as used herein is intended to mean alleviation of a disease condition associated with at least a bacterial infection in a subject (including a mammal, such as a human being), which disease condition is alleviated by reducing the growth, replication and/or reproduction of any bacteria, such as gram-positive organisms, and which alleviation includes completely or partially curing, inhibiting, alleviating, ameliorating and/or alleviating the disease condition.

The term "preventing" is intended to mean at least reducing 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 halting the progression of a disease condition associated with a bacterial infection in a mammal, preferably a human. In particular, these terms are relevant to the treatment of mammals to reduce the likelihood ("prevention") or prevent the occurrence of bacterial infections, such as may occur during or after surgery involving bone repair or replacement. These terms also include reducing the likelihood of a bacterial infection ("prophylaxis") or preventing a bacterial infection when a mammal is found to be predisposed to a disease condition but has not yet been diagnosed as having a disease condition. For example, by administering a compound of formula (1) and/or formula (2), or a pharmaceutically acceptable prodrug, salt, active metabolite, or solvate thereof, before such infection occurs, the likelihood of a bacterial infection in a mammal can be reduced or prevented.

As used herein, "synergistic effect," "synergistic effect," or "synergy" refers to an effect produced between two or more molecules, compounds, substances, factors, or compositions that is greater than or different from the sum of their respective effects.

As used herein, "additive effect" refers to an effect produced between two or more molecules, compounds, substances, factors or compositions that is equal or equivalent to the sum of their respective effects.

As used herein, the term "biocompatible" means that the material, along with any metabolites or degradation products thereof, is generally non-toxic to a recipient and does not cause any significant adverse effects to the recipient. Generally, a biocompatible material is one that does not cause a significant inflammatory or immune response when administered to a patient.

As used herein, the term "osteomyelitis" may refer to acute or chronic osteomyelitis, and/or diabetic foot osteomyelitis, diabetic chronic osteomyelitis, prosthetic joint infection, periodontitis, peri-implantitis, osteonecrosis, and/or hematogenous osteomyelitis and/or other bone infections.

Discussion of the related Art

Methylenebisphosphonic acid or substituted methylenebisphosphonic acid moieties, commonly referred to as "bisphosphonic acids" (BPs) which are therapeutic agents, are used to treat a number of bone conditions. The bisphosphonate P-C-P group mimics the P-O-P bond of the naturally occurring mediator inorganic pyrophosphate of bone metabolism. The following shows the structural relationship between acid-form pyrophosphoric acid and methylenebisphosphonic acid. Each BP may be formed by covalently attached substituents R1And R2And (4) limiting.

The bridging carbon of the bisphosphonic acids may be modified (R)1、R2) Substitution to confer specific biological properties to the derivative. BP exhibits strong binding affinity for HA (the major inorganic material found in bone, especially at sites of high bone turnover), and they are exceptionally stable against both chemical and biological degradation. It is generally not known that BP also traverses the soft and hard tissues of the body (e.g., endothelium, periosteum, HA) to target bone and small tubular networks and vascular pathways in bone. These highly specific bone targeting properties of BP make it an ideal vehicle for drug or macromolecule delivery to the bone surface.

Oxazolidinone antibiotics are ideal small molecule antibiotics effective against the major pathogens associated with bone infections. This series is headed by tedizolid, a newly approved active drug against staphylococcus aureus (MSSA and MRSA). Tedizolid is a second generation oxazolidinone following the first generation linezolid. As a treatment for acute bacterial skin and skin structure infections (abssi), tedizolid obtained FDA approval for the us market in 6 months 2014. In both phase 3 studies, tedizolid was taken once daily for a period of six days not less than 10 days for the twice daily administration of linezolid. The drug appears to have fewer gastrointestinal side effects and hematological inhibition than linezolid, although a course of more than 6 days has not been published, and therefore the true incidence of toxicity remains unknown if the drug is administered for a prolonged typical course of bone and joint infection. Nevertheless, the overall efficacy and apparent safety of this drug against staphylococcus aureus, coupled with the desirable chemical properties, makes it an attractive candidate for specific delivery to bone for the treatment of infectious bone diseases.

From the foregoing, there is an important but unmet medical need for improved antimicrobial agents for the treatment of bone infections. Embodiments of the present disclosure provided herein may provide bone-targeted antimicrobial agents to treat bone infections, and in some embodiments, overcome the deficiencies of traditional therapeutic compositions and methods. Generally, the compositions described herein may include a bisphosphonate-antimicrobial (BP-Ab) conjugate comprising an antimicrobial agent, e.g., an oxazolidinone, particularly an oxazolidinone antimicrobial or an antibiotic substituent or derivative, such as Tedizolid (TD), onto a Bisphosphonate (BP).

The compositions provided herein may use a "targeting and release linker" strategy, wherein releasable and bone-specific targeted bisphosphonate-antimicrobial (BP-Ab) conjugates can be made by attaching an antimicrobial agent, e.g., an oxazolidinone, to a pharmacologically inert BP or pharmacologically active BP using a cleavable or reversible linker, such as a carbamate, hydrazone, or the like, or a carbonate ester, such that the antimicrobial agent can be released upon binding to the bone surface, typically found at the active site of bone resorption or infection, through a reduced pH and/or enzymatic environment. An exemplary BP-oxazolidinone release mechanism is depicted in fig. 1, using Tedizolid (TD) as an exemplary oxazolidinone. This BP-Ab conjugate may have the ability to specifically deliver and release antimicrobial agents to targeted infectious osteolytic sites, thereby providing unique treatment options by providing higher concentrations of antimicrobial agents and relatively lower systemic levels at the disease site. Other oxazolidinone-BP compounds and conjugates as described herein can have the same or similar activity.

Also provided herein are formulations that can include an amount of a compound, conjugate, or composition described herein and an additional compound, such as, but not limited to, a carrier, diluent, or other active agent or ingredient. The formulation may be a pharmaceutical formulation which may comprise a pharmaceutically acceptable carrier. The compositions and/or formulations can be administered to a subject. The subject may have a bone infection. The compositions and formulations provided herein can be used to treat and/or prevent bone infections. In some embodiments, the compositions and formulations provided herein can provide bone-specific delivery of antimicrobial agents.

The general concept of using BP to target active drug substances to bone regions has been discussed in a number of reports. However, no drug has been developed, as early attempts resulted in systemically unstable prodrugs or non-cleavable conjugates, which were found to be largely inactive by interfering with pharmacodynamic requirements. This suggests that the target and release strategies may be chemical class dependent (taking into account the compatibility of the functional groups of each component) as well as biochemical target dependent, and that the design of any particular chemical class must be tailored for its use. Accordingly, provided herein are embodiments of novel methods of developing bone-targeted antimicrobial agents with bonds that are metabolically stable in the bloodstream and metabolically unstable on bone to facilitate proper release.

In particular, in any one or more aspects herein, the bonds used herein are designed to allow maximum local antibacterial efficacy at the site of infection where there is a higher bone turnover, while also limiting exposure to lower turnover bone sites, non-bone sites, and distal compartments throughout the body, from any adverse effects due to antibiotics or bisphosphonate components or conjugates. Thus, for example, the aryl carbamate linkers herein are specifically selected to have maximum stability in blood while still being sensitive to chemical cleavage and release of oxazolidinone antibiotics at the bacterial infected skeletal sites due to their sensitivity to the enzymatic processes and pH characteristics of that environment. In addition, selected, but not all embodiments in this disclosure include the use of bisphosphonic acids that have no significant pharmacological activity with respect to the targeting component of these drug conjugates. These "non-or weakly-resorptive bisphosphonates have the feature of targeting antibiotics only to the described bone compartment and do not have the properties that otherwise directly affect bone metabolism. Examples include aryl carbamates and aryl thiocarbamates derived from substituted and unsubstituted 2- [ 4-aminophenyl ] ethane 1,1 bisphosphonic acids and 2- [ 4-hydroxyphenyl ] ethane 1,1 bisphosphonic acids. Also included are carbamates derived from substituted and unsubstituted 2- [ 3-aminophenyl ] ethane 1,1 bisphosphonic acids and 2- [ 3-hydroxyphenyl ] ethane 1,1 bisphosphonic acids, substituted and unsubstituted 2- [ 2-aminophenyl ] ethane 1,1 bisphosphonic acids and 2- [ 2-hydroxyphenyl ] ethane 1,1 bisphosphonic acids. In addition, aryl dithiocarbamates derived from substituted and unsubstituted 2- [ 4-thiophenyl ] ethane 1,1 bisphosphonic acids, 2- [ 3-thiophenyl ] ethane 1,1 bisphosphonic acids, and 2- [ 2-thiophenyl ] ethane 1,1 bisphosphonic acids.

In any one or more aspects, the BP of the conjugate can be a pharmacologically inactive BP. An example of a pharmacologically inert BP that can be conjugated with an oxazolidinone as described herein is shown in figure 2.

As an example, the inert BP set used for conjugation may be 4-hydroxyphenylethylidene BP (FIG. 2A) or 4-aminophenylethylidene BP (FIG. 2E) with moderate mineral affinity. Additional analogs, such as hydroxyl BP (fig. 2B and 2F) (higher mineral affinity) and methyl BP (fig. 2C and 2G) (lower mineral affinity) can be used to modulate the concentration of BP-Ab conjugate at bone. Phenylalkyl BPs with different chain lengths, such as in fig. 2D and 2H (propyl or butyl versus ethylphenyl), can also be used to optimize conjugation chemistry yield and conjugate stability.

Bisphosphosphosphosphosphosphonoate (BP) oxazolidinone conjugates and formulations thereof

BP oxazolidinone conjugates

Provided herein are BP oxazolidinone compounds, conjugates, and formulations thereof. BP may be conjugated to oxazolidinone through a linker. In an embodiment, the linker is a releasable linker. The oxazolidinone may be releasably attached to the BP through a linker. Thus, in any one or more embodiments, the BP oxazolidinone compound or conjugate can selectively deliver and release oxazolidinone, particularly oxazolidinone antimicrobial or antibiotic substituents or derivatives, at or near a bone, bone graft, or bone graft substitute (fig. 1). In other words, the BP conjugates can provide targeted delivery of oxazolidinones, such as Tedizolid (TD), to bone and/or regions proximal to bone.

The BP of the BP oxazolidinone conjugates provided herein can be any BP, including but not limited to, hydroxyphenylalkyl or aryl bisphosphonic acids, hydroxyphenyl (or aryl) alkylhydroxy bisphosphonic acids, aminophenyl (or aryl) alkylbisphosphonic acids, aminophenyl (or aryl) alkylhydroxy bisphosphonic acids, hydroxyalkyl hydroxybisphosphonic acids, hydroxyalkylphenyl (or aryl) alkylphosphonic acids, hydroxyphenyl (or aryl) alkylhydroxy bisphosphonic acids, aminophenyl (or aryl) alkylphosphonic acids, aminophenyl (or aryl) alkylhydroxy bisphosphonic acids, hydroxyalkyl hydroxybisphosphonic acids (all of the former being further substituted or unsubstituted), etidronic acid, pamidronic acid, neridronic acid, olpadronic acid, alendronic acid, ibandronic acid, minophosphonic acid, selidronic acid, zoledronic acid, hydroxymethylbisphosphonic acid, and combinations thereof. Bisphosphonic acids may also be substituted with phosphonophosphinic acids or phosphonocarboxylic acids. In embodiments, BP can be pamidronic acid, alendronic acid, risedronic acid, zoledronic acid, minodronic acid, neridronic acid, etidronic acid, which can be unmodified or modified as described herein.

BP can be modified to contain an alpha-hydroxy group (e.g., alpha-hydroxy modified risedronic acid and zoledronic acid, fig. 4A). Other BPs can be modified in the same manner. In some embodiments, BP can be modified by substitution or removal of the α -hydroxy group. (FIG. 4B, for example, p-PyrEBP). Removal or substitution of the α -hydroxy group may reduce or eliminate the anti-absorption effect of BP as compared to the unmodified equivalent BP. Thus, in any one or more embodiments, the BP conjugates provided herein can comprise a BP lacking an a-hydroxy group or having a substituted a-hydroxy group. Suitable substitutions of the alpha-hydroxy group may include, but are not limited to, H, alkyl, aryl, alkylaryl. In addition additional molecules conjugated to BP can also affect the anti-absorption effect. For example, when an oxazolidinone and/or linker is coupled to a BP with a side change in para-substitution, the anti-absorption effect can be significantly reduced or eliminated. In any one or more embodiments, BP can be modified to include both α -hydroxy deleted or substituted and para-substituted side chains.

In BP comprising an aryl or phenyl group, the aryl or phenyl group may be substituted at any position on the ring with a suitable substituent. In any one or more embodiments, the aryl or phenyl ring of the BP is substituted with one or more electron donating species (e.g., F, N and Cl).

Non-pharmacologically active BP variants may also be used for the purpose of delivery without BP effect.

The oxazolidinones can have a general structure according to formula (5), wherein R1Can be a known component of oxazolidinone antibiotic or antibiotic resistant substituent or derivative compounds and can include alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, phenyl, substituted phenyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, halogen, hydroxy, alkoxy, substituted alkoxy, phenoxy, substituted phenoxy, aryloxy, substituted aryloxy, alkylthio, substituted alkylthio, phenylthio, substituted phenylthio, arylthio, substituted arylthio, cyano, isocyano, substituted isocyano, carbonyl, substituted carbonyl, carboxyl, substituted carboxyl, amino, substituted amino, amido, substituted amido, sulfonyl, substituted sulfonyl, sulfonic acid, phosphoryl, substituted phosphoryl, phosphonyl, substituted phosphonyl, polyaryl, substituted aryl, halogen, hydroxy, alkoxy, substituted alkoxy, phenoxy, substituted phenoxy, aryloxy, substituted alkylthio, substituted alkylthio, phenylthio, substituted phenylthio, arylthio, cyano, isocyano, substituted isocyano, carbonyl, substituted carbonyl, carboxyl, substituted carboxyl, amino, substituted amido, sulfonyl, sulfonic acid, phosphoryl, C3-C20Cyclic, substituted C3-C20Rings, heterocycles, substituted heterocycles, amino acids, peptides and polypeptide groups; and is

Wherein R is2May be a substituent comprising a releasable linker as described herein involved in the linkage to a bisphosphonate as described herein, and may include alkyl, substituted alkyl, alkenyl, substituted alkenyl, alkynyl, substituted alkynyl, phenyl, substituted phenyl, aryl, substituted aryl, heteroaryl, substituted heteroaryl, halo, hydroxy, alkoxy, substituted alkoxy, phenoxy, substituted phenoxy, aryloxy, substituted alkoxy, substituted phenoxy, aryloxy, or pharmaceutically acceptable salts thereofAryloxy, alkylthio, substituted alkylthio, phenylthio, substituted phenylthio, arylthio, substituted arylthio, cyano, isocyano, substituted isocyano, carbonyl, substituted carbonyl, carboxyl, substituted carboxyl, amino, substituted amino, amido, substituted amido, sulfonyl, substituted sulfonyl, sulfonic acid, phosphoryl, substituted phosphoryl, phosphonyl, substituted phosphonyl, polyaryl, substituted polyaryl, C3-C20Cyclic, substituted C3-C20Rings, heterocycles, substituted heterocycles, amino acids, peptides and polypeptide groups.

The BP may be conjugated to an oxazolidinone, particularly an oxazolidinone antimicrobial or antibiotic substituent or derivative, via a releasable linker, as described herein. In any one or more aspects, the linker can be a carbamate or carbonate of the structure of formula (6) or formula (7), respectively. In some embodiments, the releasable linker may be a phenyl carbamate linker. The releasable linker may be an aryl carbamate linker. In some embodiments, the linker may be an aryl thiocarbamate linker. In some embodiments, the linker may be a phenylthiocarbamate linker. In some embodiments, the thiocarbamate linker may be an O-thiocarbamate linker. In some embodiments, the thiocarbamate linker may be an S-thiocarbamate linker. In some embodiments, the linker may be a urea linker. In some embodiments, the linker may be an aryl dithiocarbamate linker. In some embodiments, the linker may be an ester linker.

In some aspects, the α -hydroxy containing BP can be conjugated to an oxazolidinone (or oxazolidinone), such as Tedizolid (TD), at the geminal OH group on the BP, as shown below. This can be achieved, for example, using hydroxy-derivatized carbamates. This may be particularly advantageous for conjugation to pharmacologically active BP variants as shown in figure 3. In some aspects, an oxazolidinone, such as TD, can be directly conjugated (e.g., without a linker) to a geminal OH group of a BP. In some aspects, the oxazolidinone may be indirectly conjugated through a linker at the geminal OH group of the BP.

In some aspects, an aminophenyl-containing BP, such as any of the pharmacologically inert BP variants shown in fig. 2, may be conjugated to an oxazolidinone, such as Tedizolid (TD), preferably through an aryl carbamate linker, as shown below. This may have the advantage of controlling the release of oxazolidinone antimicrobial compounds compared to oxazolidinone compounds conjugated to, for example, the active BP variant of fig. 3. The inactive BP variants may have fewer side effects than the active BP variants, such as lower potential toxicity than the active BP variants, in which case higher concentrations or amounts of compounds and conjugates incorporating the inactive BP variants may be used as compared to compounds and conjugates incorporating the active BP variants.

In some aspects, the compound may have a formula according to formula (2), formula (3), formula (4), or formula (5).

BP oxazolidinone conjugate drug formulations

Also described herein are formulations, including pharmaceutical formulations, which may comprise an amount of a BP oxazolidinone conjugate or compound as described elsewhere herein. The amount may be an effective amount. The amount is effective to inhibit the growth and/or reproduction of bacteria. This amount is effective to kill the bacteria. The amount is effective for treating or inhibiting bone diseases, particularly infectious bone diseases, or as a prophylactic agent against bone diseases, particularly infectious bone diseases. The amount is effective for treating or inhibiting osteomyelitis, osteolytic bone infection, osteonecrosis, diabetic chronic osteomyelitis, diabetic foot, periodontitis, and other jaw bone infections.

Formulations, including pharmaceutical formulations, can be formulated for delivery by a variety of routes and can include a pharmaceutically acceptable carrier. Techniques and formulations are generally available in Remmington's Pharmaceutical Sciences, Meade Publishing Co., Easton, Pa. (20)thEd.,2000), the entire disclosure of which is incorporated herein by reference. For systemic administration, injections are available, including intramuscular, intravenous, intraperitoneal, and subcutaneous injections. For injection, the therapeutic compositions of the present invention may be formulated in liquid solutions, for example, in physiologically compatible buffers, such as Hank's solution or Ringer's solution. In addition, the BP oxazolidinone conjugates and/or compounds herein can be formulated in solid form and redissolved or suspended immediately prior to use. Lyophilized forms are also included. Formulations of BP oxazolidinone compounds or conjugates, including pharmaceutical formulations, can be characterized as being at least sterile and pyrogen-free. These formulations include both for human use and veterinary formulations.

Suitable pharmaceutically acceptable carriers include, but are not limited to, water, salt solutions, alcohols, gum arabic, vegetable oils, benzyl alcohols, polyethylene glycols, gelatin, carbohydrates such as lactose, amylose or starch, magnesium stearate, talc, silicic acid, viscous paraffin, perfume oils, fatty acid esters, hydroxymethylcellulose, and polyvinylpyrrolidone, which do not deleteriously react with the BP oxazolidinone compound or conjugate.

The pharmaceutical formulation may be sterilized and, if desired, may be mixed with adjuvants such as lubricants, preservatives, stabilizers, wetting agents, emulsifiers, salts for influencing osmotic pressure, buffers, coloring agents, flavoring agents and/or aromatic substances and the like, which do not deleteriously react with the BP oxazolidinone compound or conjugate.

Another formulation includes the addition of one or more BP oxazolidinone compounds and/or conjugates to bone graft materials or interstitial fillers for the prevention or treatment of osteomyelitis, peri-implantitis or peri-prosthetic infection, and for alveolar protection after tooth extraction.

The pharmaceutical formulation may be formulated to be compatible with its intended route of administration. Examples of routes of administration include parenteral, e.g., intravenous, intradermal, subcutaneous, oral (e.g., inhalation), transdermal (topical), transmucosal, and rectal administration. Solutions or suspensions for parenteral, intradermal, or subcutaneous administration may include the following components: sterile diluents such as water for injection, saline solution, fixed oils, polyethylene glycols, glycerin, propylene glycol or other synthetic solvents; antibacterial agents such as benzyl alcohol or methyl paraben; antioxidants such as ascorbic acid or sodium bisulfite; chelating agents, such as ethylenediaminetetraacetic acid; buffers such as acetate, citrate or phosphate, and agents for adjusting tonicity such as sodium chloride or glucose. The adjustment of the pH can be with an acid or base, such as hydrochloric acid or sodium hydroxide. The parenteral preparation can be enclosed in ampoules, disposable syringes or multiple dose vials made of glass or plastic.

Formulations suitable for injectable use, including pharmaceutical formulations, may include sterile aqueous solutions (where water soluble) or dispersions and sterile powders for the extemporaneous preparation of sterile injectable solutions or dispersions. For intravenous administration, suitable carriers may include physiological saline, bacteriostatic water, Cremophor EMTM(BASF, Parsippany, n.j.) or Phosphate Buffered Saline (PBS). To the extent that easy injection is achieved, the injectable pharmaceutical formulation may be sterile and may be fluid. Injectable pharmaceutical formulations can be stable under the conditions of manufacture and storage and must be preserved against the contaminating action of microorganisms such as bacteria and fungi. The carrier can be a solvent or dispersion medium containing, for example, water, ethanol, and pharmaceutically acceptable polyols such as glycerol, propylene glycol, liquid polyethylene glycol, and suitable mixtures thereof. Proper fluidity can be maintained, for example, by the use of a coating such as lecithin, by the maintenance of the required particle size in the case of dispersion and by the use of surfactants. Prevention of the action of microorganisms can be achieved by various antibacterial and antifungal agents, for example, parabens, chlorobutanol, phenol, ascorbic acid, thimerosalAnd so on. In some embodiments, it may be useful to include isotonic agents (e.g., sugars, polyalcohols such as mannitol, sorbitol, and sodium chloride) in the composition.

Sterile injectable solutions can be prepared by incorporating an amount of any one or more of the BP oxazolidinone compounds or conjugates described herein into a suitable solvent, if desired with a combination of one or more of the ingredients enumerated herein, followed by filter sterilization. In general, the preparation of the dispersion can be carried out by incorporating one or more BP oxazolidinone compounds or conjugates into a sterile vehicle which comprises a base dispersion medium and the required other ingredients from those enumerated herein. In the case of sterile powders for the preparation of sterile injectable solutions, examples of useful methods of preparation are vacuum drying and freeze-drying which yields a powder of the active ingredient plus any additional desired ingredient in a previously sterile-filtered solution thereof.

Systemic administration can also be by transmucosal or transdermal means. For transmucosal or transdermal administration, penetrants appropriate to the barrier to be permeated are used in the formulation. Such penetrants are generally known in the art, and include, for example, for transmucosal administration, surfactants (detergens), bile salts, and fusidic acid derivatives. Transmucosal administration can be accomplished through the use of nasal sprays or suppositories. For transdermal administration, the BP oxazolidinone conjugate may be formulated as an ointment, gel or cream as is generally known in the art. In some embodiments, one or more BP oxazolidinone compounds and/or conjugates can be administered through a transdermal delivery system, which can slowly release the BP oxazolidinone conjugate for transdermal absorption. The penetration enhancer may be used to enhance transdermal penetration of the active factor in the conditioned medium. Transdermal patches are described, for example, in U.S. Pat. Nos. 5,407,713; U.S. patent nos. 5,352,456; U.S. patent nos. 5,332,213; U.S. Pat. Nos. 5,336,168; U.S. patent nos. 5,290,561; U.S. patent nos. 5,254,346; U.S. patent nos. 5,164,189; U.S. patent nos. 5,163,899; U.S. patent nos. 5,088,977; U.S. patent nos. 5,087,240; U.S. patent nos. 5,008,110; and U.S. patent No.4,921,475.

For oral administration, the formulations as described herein may be presented as capsules, tablets, powders, granules, or as a suspension or solution. The formulations may contain conventional additives such as lactose, mannitol, corn or potato starch, binders, crystalline cellulose, cellulose derivatives, acacia, corn starch, gelatin, disintegrants, potato starch, sodium carboxymethylcellulose, dibasic calcium phosphate, anhydrous or sodium starch glycolate, lubricants and/or magnesium stearate.

For parenteral administration (i.e., administration by a route other than through the digestive tract), the formulations described herein may be combined with a sterile aqueous solution that is isotonic with the blood of the subject. Such formulations may be prepared by dissolving the active ingredient (e.g., BP oxazolidinone compound or conjugate) in water containing a physiologically compatible substance such as sodium chloride, glycine, or the like and having a buffered pH compatible with physiological conditions to produce an aqueous solution, and then rendering the solution sterile. The formulations may be presented in unit-or multi-dose containers, such as sealed ampoules or vials. The formulation may be delivered by injection, infusion or other means known in the art.

For transdermal administration, the formulations described herein may be combined with skin penetration enhancers, such as propylene glycol, polyethylene glycol, isopropyl alcohol, ethanol, oleic acid, N-methyl pyrrolidone, and the like, which increase the permeability of the skin to the nucleic acid vectors (vectors) of the present invention and allow the nucleic acid vectors to permeate into the bloodstream through the skin. The formulations and/or compositions described herein may be further combined with a polymeric substance, such as ethyl cellulose, hydroxypropyl cellulose, ethylene/vinyl acetate, polyvinylpyrrolidone, and the like, to provide a composition in the form of a gel, which may be dissolved in a solvent such as methylene chloride, evaporated to a desired viscosity and then applied to a base material to provide a patch.

The formulations described herein may be combined with any xenograft (bovine), autograft (autologous) or allograft (cadaveric) material or synthetic bone substitute, especially a hydroxyapatite containing substance, material or surface, in order to be included in bone graft substitutes or bone gap fillers to prevent local post-operative infection or post-operative graft failure and to provide sustained local release of antibiotics at the site of the graft. For example, the treating surgeon or clinician may premix the powder formulation with any commercially available bone graft substitute or with autograft at the bedside/couch side. This formulation may be further combined with any of the previously described formulations, and may be combined with products comprising hydroxyapatite, tricalciumphosphate, collagen, aliphatic polyesters, polylactic acids (PLA), polyglycolic acids (PGA), and Polycaprolactone (PCL), Polyhydroxybutyrate (PHB), methacrylates, polymethylmethacrylate, resins, monomers, polymers, cancellous bone allograft, human fibrin, platelet rich plasma, platelet rich fibrin, plaster of paris, apatite, synthetic hydroxyapatite, coral hydroxyapatite, wollastonite (calcium silicate), calcium sulfate, bioactive glass, ceramics, titanium, devitalized bone matrix, non-collagenous protein, collagen, and autolytic de-antigenic allogeneic bone. In this embodiment, the bone graft material combined with the BP-oxazolidinone conjugate can be in the formulation of a paste, powder, cement, gel, hydrogel, matrix, granule, lyophilized powder, lyophilized bone, demineralized lyophilized bone, fresh or fresh frozen bone, pith mixture, pellet, strip, embolic agent (plugs), membrane, lyophilized powder reconstituted to form a wet paste, pellet, sponge, block, briquette (morsels), rod, wedge, cement (cement) or amorphous granule; of these, many can also be in an injectable formulation or as a combination of two or more of the above (e.g., an injectable paste of a sponge).

In another embodiment, one or more BP-oxazolidinone compounds or conjugates can be combined with a factor-based bone graft comprising a natural or recombinant growth factor, such as transforming growth factor-beta (TGF- β), Platelet Derived Growth Factor (PDGF), Fibroblast Growth Factor (FGF), and/or Bone Morphogenic Protein (BMP). In another embodiment, one or more BP oxazolidinone compounds or conjugates as described herein can be combined with cell-based bone grafts for regenerative medicine and dentistry including embryonic and/or adult stem cells, tissue specific stem cells, hematopoietic stem cells, epidermal stem cells, epithelial stem cells, gingival stem cells, periodontal ligament stem cells, adipose stem cells, bone marrow stem cells and blood stem cells. Thus, bone grafts having properties of osteoconduction, osteoinduction, osteopromotion, osteogenesis or any combination thereof may be used in combination with one or more BP oxazolidinone compounds or conjugates for clinical or therapeutic use.

Dosage forms

The BP oxazolidinone compounds, conjugates, and formulations described herein can be provided in unit dosage forms, such as tablets, capsules, single dose injection or infusion vials, or as a pre-determined dose in a formulation as described above for mixing with bone graft material. Where appropriate, the dosage forms described herein may be microencapsulated. The dosage form may also be formulated to prolong or sustain the release of any of the ingredients. In some embodiments, the complex active agent may be a delayed release component. In other embodiments, the release of the secondary ingredient is delayed. Suitable methods for delaying release of the ingredients include, but are not limited to, coating or embedding the ingredients in a polymer, wax, gel, or like material. Delayed release dosage formulations may be prepared as described in standard references, such as "Pharmaceutical dosage forms" or "eds. Liberman et al. (New York, Marcel Dekker, Inc.,1989)," Remington-The science and practice of medicine ", 20th ed., Lippincott Williams & Wilkins, Baltimore, MD,2000, and" Pharmaceutical dosage forms and drug delivery systems ", 6th Edition, Ansel et al. (Media, PA: Williams and Wilkins, 1995). These references provide excipients, materials, equipment and processes for preparing tablets and capsules, and delayed release dosage forms of tablets and pellets, capsules and granules. The delayed release may be any time from about one hour to about 3 months or more.

Coatings may be formed from water-soluble polymers, water-insoluble polymers and/or pH-dependent polymers, with or without water-insoluble/water-soluble non-polymeric excipients, in varying ratios to produce the desired release profile. Coatings may be applied to the dosage form (matrix or simply) including, but not limited to, tablets (compressed with or without coated beads), capsules (with or without coated beads), beads, granular compositions, the ingredients themselves formulated as, but not limited to, a suspension or sprinkled dosage form.

Examples of suitable coating materials include, but are not limited to, cellulosic polymers such as cellulose acetate phthalate, hydroxypropyl cellulose, hydroxypropyl methylcellulose phthalate, and hydroxypropyl methylcellulose acetate succinate; polyvinyl acetate phthalate, acrylic polymers and copolymers, and commercially available under the trade name(Roth Pharma, Westerstadt, Germany), zein, shellac, and polysaccharides.

An effective amount

The formulations may comprise an effective amount of a BP oxazolidinone conjugate as described herein (effective to inhibit and/or kill bacteria). In some embodiments, an effective amount of a BP oxazolidinone compound or conjugate described herein ranges from about 0.001pg to about 1,000g or more. In some embodiments, an effective amount of a BP oxazolidinone compound or conjugate described herein can range from about 0.001mg/kg body weight to about 1,000mg/kg body weight. In yet other embodiments, the effective amount of the BP oxazolidinone compound or conjugate can range from about 1% w/w to about 99% or more w/w, w/v, or v/v of the total formulation. In some embodiments, the effective amount of the BP oxazolidinone compound or conjugate is effective to kill bacteria that are causative agents of osteomyelitis and all subtypes thereof (e.g., diabetic foot osteomyelitis), jaw necrosis, and periodontitis, including, but not limited to, Staphylococcus (Staphylococcus), Pseudomonas (Pseudomonas), agglomerans (agregabacter), Actinomyces (Actinomyces), Streptococcus (Streptococcus), Haemophilus (Haemophilus), Salmonella (Salmonella), Serratia (serrrata), Enterobacter (Enterobacter), clostridium (Fusobacterium), Bacteroides (Bacteroides), Porphyromonas (Porphyromonas), Prevotella (Prevotella), weinpercoccus (veillia), Campylobacter (Campylobacter), peptostroccus (peptostroccus), echocysticerus (eellus), Treponema (Treponema), Treponema (Yersinia), Yersinia (Yersinia), etc, Any strain or species of the genera tanneria (tannorella) and Escherichia (Escherichia).

Methods of using BP oxazolidinone compounds and conjugates

An amount (including an effective amount) of the BP oxazolidinone compounds, conjugates, and formulations thereof described herein can be administered to a subject in need thereof. In some embodiments, a subject in need thereof can have a bone infection, disease, disorder, or symptom thereof. In some embodiments, a subject in need thereof may be suspected of having, or otherwise susceptible to having, a bone infection, disease, disorder, or symptom thereof. In some embodiments, a subject in need thereof may be at risk of developing osteomyelitis, osteonecrosis, periprosthetic infection, and/or peri-implantitis. In embodiments, the disease or condition may be osteomyelitis and all subtypes thereof, osteonecrosis, peri-implantitis, or periodontitis. In some embodiments, a subject in need thereof has bone infected with a microorganism (such as a bacterium). In some embodiments, the bacterium can be any strain or species of staphylococcus, pseudomonas, coacervate bacillus, actinomyces, streptococcus, haemophilus, salmonella, serratia, enterobacter, clostridium, bacteroides, porphyromonas, prevotella, veyonococcus, campylobacter, peptostreptococcus, akerma, treponema, alisteribacter, micromonospora, yersinia, tannophila, and escherichia. In some embodiments, the bacteria may form a biofilm. In some embodiments, osteomyelitis in a subject in need thereof can be treated by administering to a subject in need thereof an amount (such as an effective amount) of a BP oxazolidinone compound, conjugate, or formulation described herein. In some embodiments, the compositions, compounds, and formulations provided herein may be used for osteonecrosis treatment and/or prevention, stretch osteogenesis, fracture repair, repair of critical upper socket defects, jaw bone reconstruction, and any other reconstruction or repair of bone and/or joint.

Administration of the BP oxazolidinone compound and conjugate is not limited to a single route, but may include administration by a variety of routes. For example, exemplary administration by various routes includes a combination of intradermal and intramuscular administration, or a combination of intradermal and subcutaneous administration, among others. The multiple administrations may be sequential or simultaneous. Other modes of administration by various routes will be apparent to those skilled in the art.

The pharmaceutical formulation may be administered to a subject by any suitable method that allows the agent to exert its effect on the subject in vivo. For example, the formulations and compounds and compositions described herein can be administered to a subject by known procedures, including, but not limited to, by oral administration, sublingual or buccal administration, parenteral administration, transdermal administration, by inhalation, by nasal delivery, vaginal, rectal, and intramuscular. The formulations and compounds and compositions described herein may be administered parenterally, by extrafascial, intracapsular, intradermal (intracutaneous), subcutaneous, intradermal, intrathecal, intramuscular, intraperitoneal, intrasternal, intravascular, intravenous, parenchymal, and/or sublingual delivery. Delivery may be by injection, infusion, catheter delivery, or some other means, such as by tablet or spray. In the case of infection-resistant bone graft materials at the surgical site, delivery may also be by a carrier such as hydroxyapatite or bone. Delivery may be by attachment or other bonding with bone graft material.

Examples

Having now described embodiments of the present disclosure, the following examples, in general, describe some additional embodiments of the present disclosure. While embodiments of the present disclosure have been described in connection with the following examples and the corresponding text and drawings, there is no intent to limit embodiments of the present disclosure to such illustrations. On the contrary, the intent is to cover all alternatives, modifications and equivalents as included within the spirit and scope of the embodiments of the disclosure.

In any one or more aspects, the BP of the conjugate can be a pharmacologically inactive BP. An example of a pharmacologically inert BP that can be conjugated with an oxazolidinone as described herein is shown in figure 2. As an example, the inert BP set used for conjugation (FIG. 2) may be 4-hydroxyphenylethylidene BP (FIG. 2A) or 4-aminophenylethylidene BP (FIG. 2E). Additional analogs, such as hydroxyl BP (fig. 2B and 2F) (higher mineral affinity) and methyl BP (fig. 2C and 2G) (lower mineral affinity) can be used to modulate the concentration of BP-Ab conjugate at bone. Phenylalkyl BPs with different chain lengths, such as shown in fig. 2D and 2H (propyl or butyl versus ethyl or phenyl) can also be used to optimize conjugation chemistry yield and conjugate stability.

By way of example, BPs with high or moderate pharmacological activity, such as etidronic acid, methylenehydroxy bisphosphonic acid (MHDP), pamidronic acid, alendronic acid, risedronic acid, zoledronic acid, minodronic acid, etc., depicted in fig. 3, can also be used in conjugation to provide a bisphosphonate component or dual active agent that is studied to protect adequately from bone loss and bone infection, as bone resorption is known to occur at the site of infection.

As an example, the antimicrobial agent of the conjugates and compounds of the invention can be an oxazolidinone analog of formula (5) herein or any oxazolidinone agent shown in figure 5. The oxazolidinone antimicrobial or antibiotic agent can be a substituent or derivative of an oxazolidinone. For example, in the case of linezolid, the terminal amide or amino substituted carbonyl group may be substituted with a linker to provide for the release of deacetylated (or deacetylated) linezolid as the active agent. The same applies to oxyphenone, radizomide, rabezolid and sutezolid. Oxazolidinone antimicrobials remain the mainstay for the treatment of skin infections in adults, tedizolid (TD, fig. 5) or linezolid (LD, fig. 5). Introduction of an active antimicrobial or antibiotic compound such as TD or LD into the BP molecule can enhance its ability to bind to, focus on, and retain infected bone (high bone turnover sites that are often difficult to treat). Some bisphosphonate-oxazolidinone conjugates of the present disclosure are exemplified in figure 6.

The synthesis of the BP-linker-tedizolid (BP-L-TD, L: -OC (O) N-) conjugate is illustrated in the synthetic scheme depicted in FIG. 7. The chemistry is also applicable to other oxazolidinones such as oxyphenxadone, pasireotide, and the like.

The synthesis of the BP-linker-deacetyllinezolid (BP-L-dLD, L: -OC (O) N-) conjugate is illustrated in the synthetic scheme depicted in FIG. 8. This chemistry is also applicable to other deacetylated oxazolidinones such as oxyphenone, radizole, rabezolid, sutezolid, and the like.

The synthesis of the BP-linker-tedizolid (BP-L-TD, L: -OC (O)) conjugate is illustrated in the synthetic scheme depicted in FIG. 9. This chemistry is also applicable to other oxazolidinones such as oxyphenxadone, pasireotide, and the like.

The synthesis of the BP-linker-deacetyllinezolid (BP-L-dLD, L: -NC (O) N-) conjugate is illustrated in the synthetic scheme depicted in FIG. 10. This chemistry is also applicable to other deacetylated oxazolidinones such as oxyphenone, radizole, rabezolid, sutezolid, and the like.

As an example, the releasable linker may be a urethane linkage, an aryl carbamate, a benzoic carbamate linkage, a thiocarbamate, a dithiocarbamate, or a hydrazone linkage. These BP-Ab conjugates are stable in the bloodstream following in vivo injection, but are unstable at the bone surface to release the antimicrobial agent at effective concentrations, providing detectable antibacterial activity. The release rate of the antimicrobial agent and the amount of surface-localized drug relative to the amount administered can be adjusted by changing the chain length of the phenylalkyl BP or using a different bone affinity BP.

A detailed synthesis of BP-linker-tedizolid (BP-L-TD, L: -OC (O) N-) and (BP-L-TD, L: -C (O) O-) conjugates can be as follows:

4- (2, 2-bis (diisopropyloxyphosphoryl) ethyl) benzoic acid methyl ester (1)

Under nitrogen atmosphereIn a 250mL round bottom flask, THF (35mL) was added to a 60% dispersion of NaH in mineral oil (0.976g, 24.40 mmol). The suspension was cooled to 0 ℃ while stirring, and tetraisopropyl methylenebisphosphate (6.480mL,20.33mmol) was gradually added. The reaction was allowed to reach ambient temperature and once the hydrogen bubbles from the reaction mixture stopped, the solution was cooled again to 0 ℃. Methyl 4- (bromomethyl) benzoate (4.660g, 20.33mmol) was dissolved in THF (35mL) and added dropwise to the reaction. The resulting solution was allowed to stir overnight while slowly reaching ambient temperature. The reaction mixture was then cooled to 0 ℃ and quenched with MeOH (2 mL). Reacting NH4Cl(aqueous solution)(1M,80mL) was added to the crude and the product was extracted with DCM (3X 100mL), the combined organics were washed with brine (100mL) over MgSO4Drying, filtering, concentrating under reduced pressure, and purifying by using ACN CHCl3Gradient (0-100%) silica gel column chromatography gave 1 as a clear oil (58% yield).

4- (2, 2-bis (diisopropyloxyphosphoryl) ethyl) benzoic acid (2)

To a solution of 1(1.968g,3.996mmol) in MeOH (20mL) in a 100mL round bottom flask was added LiOH H2O (0.838g,19.98mmol), and the resulting solution was stirred at room temperature overnight. The reaction mixture was evaporated to dryness, the residue was dissolved in water (70mL), and HCl was added slowly(aqueous solution)(1M) to reach pH 3. With CHCl3The resulting mixture was extracted (3X 60 mL). The combined organics were washed with water over MgSO4Dry above and concentrate under reduced pressure to give a thick pale yellow oil which turns to an off-white solid in the refrigerator (100% yield).

(2- (4- (azidocarbonyl) phenyl) ethan-1, 1-diyl) bis (phosphonic acid) tetraisopropyl ester (3)

Compound 2(1.913g, 3.998mmol) was dissolved in anhydrous dioxane (19 mL). Diphenylphosphoryl azide (1.100g,3.998mmol) and trimethylamine (0.445g,4.400mmol) were added, and the reaction was stirred at room temperature for 2 hours. Ultrapure deionized water (20mL) was added to the reaction mixture and the product was extracted using hexane (4 x 40 mL). The hexane extracts were combined and MgSO4Drying, filtration and concentration in vacuo afforded 4, which was used without further purification (92% yield).

In use of N2In a rinsed 8Dr glass vial, compound 3(84.00mg,0.167mmol) and tedizolid (61.80mg,0.167mmol) were dissolved in anhydrous DMF (1.5mL) and a catalytic amount of DMAP (8.200mg,0.067mmol) was added to the solution. The reaction vial was covered with foil and stirred at 80 ℃ for 3 hours. By TLC (1:1 ACN/CHCl)3) The progress of the reaction was monitored. The solvent was removed under reduced pressure and purified by using ACN CHCl3Gradient (25-50%) silica gel column chromatography gave 4 as a white foam (81% yield).

(R) - (2- (4- ((((3- (3-fluoro-4- (6- (2-methyl-2H-tetrazol-5-yl) pyridin-3-yl) phenyl) -2-oxooxazolidin-5-yl) methoxy) carbonyl) amino) phenyl) ethan-1, 1-diyl) bis (phosphonic acid) (5)

In an 8Dr glass vial, Compound 4(0.104g,0.123mmol) was dissolved in DCM (1.5mL) and BTMS (324.5 μ L,2.459mmol) was added. The vial was capped and heated at 35 ℃ overnight while covered with foil and stirred. The next day, the solvent was removed in vacuo and the crude quenched with MeOH (3 mL). The resulting solution was stirred at room temperature for 30 minutes. The solvent was removed under reduced pressure and the product was precipitated in MeOH and the resulting suspension was filtered using a medium pore glass funnel. The resulting solid was further washed with MeOH to give a light yellow powder (81% yield).

4- (2, 2-bis (diisopropyloxyphosphoryl) ethyl) benzoic acid (R) - (3- (3-fluoro-4- (6- (2-methyl-2H-tetrazol-5-yl) pyridin-3-yl) phenyl) -2-oxooxazolidin-5-yl) methyl ester (6)

In use of N2In a rinsed 1.5Dr glass vial, compound 2(100mg,0.209), tedizolid (77.4mg,0.209mmol) and EDC (64.9mg,0.418mmol) were dissolved in anhydrous DMF (2mL) and a catalytic amount of 4-DMAP (28.1mg,0.230mmol) was added to the solution. The reaction vial was covered with foil and stirred at room temperature overnight. By TLC (1:1 ACN/CHCl)3) The progress of the reaction was monitored. The solvent was removed under reduced pressure and purified by using 1:1CAN/CHCl3The mixture was purified by silica gel column chromatography as eluent to give 6 as a pale yellow viscous oil (yield 95%).

(R) - (2- (4- (((3- (3-fluoro-4- (6- (2-methyl-2H-tetrazol-5-yl) pyridin-3-yl) phenyl) -2-oxooxazolidin-5-yl) methoxy) carbonyl) phenyl) ethan-1, 1-diyl) bis (phosphonic acid) (7)

In a 1Dr glass vial, compound 6(0.096g,0.116mmol) was dissolved in chloroform (1mL) and BTMS (610 μ L,4.62mmol) was added. The vial was capped and heated at 55 ℃ overnight while covered with foil and stirred. The next day, the solvent was removed in vacuo and the crude quenched with MeOH (2 mL). The resulting solution was stirred at room temperature for 30 minutes. The solvent was removed under reduced pressure and the product was precipitated in MeOH and the resulting suspension was filtered using a medium pore glass funnel. The resulting solid was further washed with MeOH to give a yellow powder (70% yield).

The synthesis of the BP-linker-tedizolid (BP-L-TD, L: -OC (S) N-) conjugate is illustrated in the synthetic scheme depicted in FIG. 11. This chemistry is also applicable to other oxazolidinones such as oxyphenxadone, pasireotide, and the like.

The synthesis of the BP-linker-linezolid (BP-L-dLD, L: -OC (S) N-) conjugate is exemplified in the synthetic scheme depicted in FIG. 12. This chemistry is also applicable to other deacetylated oxazolidinones such as oxyphenone, radizomide, rabezolid, and sutezolid, among others.

The synthesis of the BP-linker-linezolid (BP-L-dLD, L: -SC (O) N-) conjugate is exemplified in the synthetic scheme depicted in FIG. 13. This chemistry is also applicable to other deacetylated oxazolidinones such as oxyphenone, radizomide, rabezolid, and sutezolid, among others.

The synthesis of the BP-linker-linezolid (BP-L-dLD, L: -SC (S) N-) conjugate is exemplified in the synthetic scheme depicted in FIG. 14. This chemistry is also applicable to other deacetylated oxazolidinones such as oxyphenone, radizomide, rabezolid, and sutezolid, among others.

The synthesis of the BP-linker-linezolid (BP-L-dLD, L: -NC (S) N-) conjugate is exemplified in the synthetic scheme depicted in FIG. 15. This chemistry is also applicable to other deacetylated oxazolidinones such as oxyphenone, radizomide, rabezolid, and sutezolid, among others.

Table 1 below depicts antimicrobial sensitivity results against experimental pathogens in polystyrene wells using a standard assay, wherein M-minocycline; c-ciprofloxacin; x-moxifloxacin; t-tedizolid; v-vancomycin; BCT-bisphosphonic acid-carbamate-tedizolid conjugate (BP-L-TD, L: -NC (O) O-, as exemplified in FIG. 6); a BET-bisphosphonate-tedizolid conjugate ((BP-L-TD, L: -C (O) O-, as exemplified in FIG. 6, continuation (19));*the spectrum of antimicrobial activity does not encompass this particular pathogen and does not support use against particular bacteria.

TABLE 1

Figure 16 depicts the results of a prophylactic Hydroxyapatite (HA) experiment with test compounds against staphylococcus aureus. The BET conjugate (BP-L-TD, L: -C (O) O-, as exemplified in FIG. 6 (19)) showed statistically significant efficacy in preventing staphylococcal cell growth. (K-W test,. p < 0.01; comparative ═ control). Results were obtained using standard assays.

Figure 17 depicts the results of HA eradication experiments with test compounds against staphylococcus aureus biofilms. With the exception of vancomycin and minocycline, all compounds tested were significantly effective in eradicating staphylococcal cells. (K-W test,. p < 0.01; comparative ═ control). The BET conjugate is BP-L-TD, L: -C (O) O-, as exemplified in FIG. 7 continuation (19). Results were also obtained by using standard assays.

Figure 18A depicts a picture of rat femurs saturated with antibiotics or conjugates after introduction of the rat femurs into a solution of staphylococcus aureus in our ex vivo prophylactic experimental setup. The more red the bone, the more staphylococcal biofilm is attached to the surface of these areas. Femoral 1-negative control sample (no antimicrobial, no staphylococcus); 2-positive control sample (no added antimicrobial, staphylococcus solution); 3-bone saturated with tedizolid and introducing the bone into a staphylococcal solution; 4-bone saturated with ciprofloxacin and introduction of bone into staphylococcal solution; 5-bone saturated with BET conjugate (BP-L-TD, L: -C (O) O-, as exemplified in FIG. 7 continuation (19)) and introduced into the staphylococcal solution; figure 18B depicts the staphylococcus aureus biofilm prevention and eradication settings, the conjugate BET significantly prevented or eradicated biofilm growth of the ex vivo femurs as did tedizolid alone, with the BET conjugate observed for maximum efficacy in both settings (K-W test, × p < 0.01; comparator ═ control).

Ratios, concentrations, amounts, and other numerical data herein may be presented in a range format. It is to be understood that such a range format is used for convenience and brevity, and should be interpreted in a flexible manner to include not only the numerical values explicitly recited as the limits of the range, but also to include all the individual numerical values or sub-ranges encompassed within that range as if each numerical value and sub-range is explicitly recited. For purposes of this specification, a concentration range of "about 0.1% to about 5%" should be interpreted to include not only the explicitly recited concentration of about 0.1% to about 5%, but also include individual concentrations (e.g., 1%, 2%, 3%, and 4%) and the sub-ranges within the indicated range (e.g., 0.5%, 1.1%, 2.2%, 3.3%, and 4.4%). In embodiments, the term "about" may include rounding according to the convention for the significant digit of the numerical value. In addition, the phrase "about 'x' to 'y'" includes "about 'x' to about 'y'".

It should be emphasized that the above-described embodiments are merely examples of possible implementations. Many variations and modifications may be made to the above-described embodiments without departing from the principles of the present disclosure. All such modifications and variations are intended to be included herein within the scope of this disclosure and protected by the following claims.

70页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:作为肿瘤免疫应答的生物标志物的重复RNA

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!