US20050245491A9 - Tetracycline derivatives and methods of use thereof - Google Patents

Tetracycline derivatives and methods of use thereof Download PDF

Info

Publication number
US20050245491A9
US20050245491A9 US10/264,454 US26445402A US2005245491A9 US 20050245491 A9 US20050245491 A9 US 20050245491A9 US 26445402 A US26445402 A US 26445402A US 2005245491 A9 US2005245491 A9 US 2005245491A9
Authority
US
United States
Prior art keywords
product
alkyl
doxycycline
tetracycline
amino
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Granted
Application number
US10/264,454
Other versions
US20040067912A1 (en
US7214669B2 (en
Inventor
Joseph Hlavka
Richard Ablin
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Tetragenex Pharmaceuticals Inc
Original Assignee
Tetragenex Pharmaceuticals Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Priority to US10/264,454 priority Critical patent/US7214669B2/en
Application filed by Tetragenex Pharmaceuticals Inc filed Critical Tetragenex Pharmaceuticals Inc
Assigned to INNAPHARMA, INC. reassignment INNAPHARMA, INC. ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: ABLIN, RICHARD J., HLAVKA, JOSEPH J.
Publication of US20040067912A1 publication Critical patent/US20040067912A1/en
Priority to US11/076,276 priority patent/US20050267079A1/en
Assigned to TETRAGENEX PHARMACEUTICALS, INC. reassignment TETRAGENEX PHARMACEUTICALS, INC. MERGER (SEE DOCUMENT FOR DETAILS). Assignors: INNAPHARMA, INC.
Publication of US20050245491A9 publication Critical patent/US20050245491A9/en
Priority to US11/673,589 priority patent/US20070142338A1/en
Priority to US11/679,744 priority patent/US20070149488A1/en
Publication of US7214669B2 publication Critical patent/US7214669B2/en
Application granted granted Critical
Anticipated expiration legal-status Critical
Expired - Fee Related legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/65Tetracyclines
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P35/00Antineoplastic agents
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C237/00Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups
    • C07C237/24Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atom of at least one of the carboxamide groups bound to a carbon atom of a ring other than a six-membered aromatic ring of the carbon skeleton
    • C07C237/26Carboxylic acid amides, the carbon skeleton of the acid part being further substituted by amino groups having the carbon atom of at least one of the carboxamide groups bound to a carbon atom of a ring other than a six-membered aromatic ring of the carbon skeleton of a ring being part of a condensed ring system formed by at least four rings, e.g. tetracycline
    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07CACYCLIC OR CARBOCYCLIC COMPOUNDS
    • C07C2603/00Systems containing at least three condensed rings
    • C07C2603/02Ortho- or ortho- and peri-condensed systems
    • C07C2603/40Ortho- or ortho- and peri-condensed systems containing four condensed rings
    • C07C2603/42Ortho- or ortho- and peri-condensed systems containing four condensed rings containing only six-membered rings
    • C07C2603/44Naphthacenes; Hydrogenated naphthacenes
    • C07C2603/461,4,4a,5,5a,6,11,12a- Octahydronaphthacenes, e.g. tetracyclines

Definitions

  • the present invention relates to novel tetracycline derivatives, methods for producing the novel derivatives and methods of using these derivatives.
  • the compound tetracycline exhibits the following general structure
  • the numbering system of the ring nucleus is as follows:
  • Tetracycline as well as the 5-OH (Terramycin) and 7-Cl (Aureomycin) derivatives exist in nature, and are well known antibiotics.
  • Natural tetracycline may be modified without losing their antibiotic properties, although certain elements of the structure must be retained.
  • the modifications that may and may not be made to the basic tetracycline structure have been reviewed by Mitscher in The Chemistry of Tetracyclines, Chapter 6, Marcel Dekker, Publishers, New York (1978). According to Mitscher, the substituents at positions 5-9 of the tetracycline ring system may be modified without the complete loss of antibiotic properties.
  • CMT chemically modified non-antimicrobial tetracyclines
  • Some 4-dedimethylaminotetracycline derivatives are disclosed in U.S. Pat. Nos. 3,029,284 and 5,122,519. They include 6-demethyl-6-deoxy-4-dedimethylaminotetraccyline and 5-hydroxy-6-deoxy-4-dedimethylaminotetracycline with hydrogen and other substituents at C7, and the C9 positions on the D ring. These substitutents include amino, nitro, di (lower alkyl) amino, and mono (lower alkyl) amino or halogen. The 6-demethyl-6-deoxy-4-dedimethylaminotetracycline derivatives and 5-hydroxy-6-deoxy-4-dedimethylaminotetracycline derivatives are said to be useful as antimicrobial agents.
  • Alkylamino (NH-alkyl), and alkylhydrazone (N—NH-alkyl) groups have been substituted on the A ring at the C4 position of 4-dedimethylaminotetracycline. These compounds are known for their antimicrobial properties. See U.S. Pat. Nos. 3,345,370, 3,609,188, 3,622,627, 3,502,660, 3,509,184, 3,502,696, 3,515,731, 3,265,732, 5,122,519, 3,849,493, 3,772,363, and 3,829,453.
  • tetracyclines have been described as having a number of other uses.
  • tetracyclines are also known to inhibit the activity of collagen destructive enzymes, such as matrix metalloproteinases (MMP), including collagenases (MMP-1), gelatinase (MMP-2) and stromelysin (MMP-3).
  • MMP matrix metalloproteinases
  • MMP-1 collagenases
  • MMP-2 gelatinase
  • MMP-3 stromelysin
  • Golub et al. J. Periodont. Res. 20:12-23 (1985); Golub et al. Crit. Revs. Oral Biol. Med. 2:297-322 (1991); U.S. Pat. Nos. 4,666,897; 4,704,383; 4,935,411; 4,935,412.
  • tetracyclines have been known to inhibit wasting and protein degradation in mammalian skeletal muscle, U.S. Pat. No. 5,045,538, and to enhance IL-10
  • tetracyclines were reported to enhance bone protein synthesis in U.S. Pat. No. Re. 34,656, and to reduce bone resorption in organ culture in U.S. Pat. No. 4,704,383.
  • U.S. Pat. No. 5,532,227 to Golub et al discloses that tetracyclines can ameliorate the excessive glycosylation of proteins.
  • tetracyclines inhibit the excessive collagen cross linking which results from excessive glycosylation of collagen in diabetes.
  • Tetracyclines are known to inhibit excessive phospholipase A 2 activity involved in inflammatory conditions such as psoriasis as disclosed in U.S. Pat. No. 5,532,227.
  • tetracyclines are also known to inhibit cycloxygenase-2 (COX-2), tumor necrosis factor (TNF), nitric oxide and IL-1 (interleukin-1).
  • COX-2 cycloxygenase-2
  • TNF tumor necrosis factor
  • IL-1 interleukin-1
  • tetracyclines have been useful in treating a number of diseases.
  • tetracyclines including non-antimicrobial tetracyclines, are effective in treating arthritis. See, for example, Greenwald, et al.
  • Tetracyclines have also been suggested for use in treating skin diseases. For example, White et al., Lancet, April 29, p. 966 (1989) report that the tetracycline minocyline is effective in treating dystrophic epidermolysis bullosa, which is a life-threatening skin condition believed to be related to excess collagenase.
  • tetracyclines have been found to be effective in treating numerous diseases and conditions. Therefore, there is a need for new and even more useful derivatives.
  • Compounds disclosed herein are derivatives of tetracycline, which exhibit antimicrobial and/or anti-cancer activity.
  • the present invention relates to a treatment for inhibiting microbial or tumor growth which comprises adding to a subject in need of said treatment an effective amount of a tetracycline composition or salt thereof, comprising a general formula: wherein R is selected from CH 2 N(R a R b ) or
  • the present invention relates to a treatment for inhibiting microbial or tumor growth which comprises adding to a subject in need of said treatment an effective amount of a tetracycline composition or salt thereof, comprising a general formula: wherein R is selected from CH 2 N(R a R b ) or where R a R b are C 1 to C 6 alkyl;
  • the present invention relates to a treatment for inhibiting microbial or tumor growth which comprises adding to a subject in need of said treatment an effective amount of a tetracycline composition or salt thereof, comprising a general formula: wherein R is selected from CH 2 N(R a R b ) or CH 2 N where R a R b are C 1 -C 6 alkyl;
  • the present invention relates to a treatment for inhibiting microbial or tumor growth which comprises adding to a subject in need of said treatment an effective amount of a tetracycline composition or salt thereof, comprising a general formula: wherein R is selected from CH 2 N(R a R b ) or
  • the present invention relates to a treatment for inhibiting microbial or tumor growth which comprises adding to a subject in need of said treatment an effective amount of a tetracycline composition or salt thereof, comprising a general formula: wherein R 1 is H or N(CH 3 ) 2 ; R 2 is H, CH 3 or OCOCH 3 ; R 3 is H or CH 3 ; R 4 is H; R 5 is H, N(CH 3 ) 2 , NH 2 , N(C 4 H 9 ) 2 , N(C 6 H 13 ) 2 , N(3,3-dimethylbutyl) 2 , N 3 , N 2 + , NO 2 , NHCOCH 3 , NH(n-propyl) 2 , NH-isobutyl, NH-isobutylmethyl, NH(cyclobutyl), NH(cyclobutyl methyl); and R 6 is H, NO 2 , NH 2 , (CH 3 ) 2 CHNH
  • the present invention relates to a treatment for inhibiting microbial or tumor growth which comprises adding to a subject in need of said treatment an effective amount of a tetracycline composition or salt thereof, comprising a general formula: wherein R 1 is H or N(CH 3 ) 2 ; R 2 is H, OH or OCOCH 3 ; R 3 is H or CH 3 ; R 4 is H; R 5 is H, N(CH 3 ) 2 ; N(C 4 H 9 ) 2 , N(C 6 H 13 ) 2 , N(3,3-dimethylbutyl) 2 ; N 3 , N 2 + , NO 2 , NHCOCH 3 , NH(n-propyl) 2 , NH-isobutyl, NH-isobutylmethyl, N(CH 3 CO)(isobutyl), NH(cyclobutyl), NH(cyclobutyl methyl), or NH 2 ; and R 6 is H, NO 2 ;
  • Doxycycline free base (315 mg) was dissolved in 2 mL of methanol and 10 mL of THF, then 1 mL of methyl iodide was added. The reaction mixture was kept for 5 days; no crystallization of the product occurred. The reaction mixture was concentrated and poured into 300 mL of cold ether with stirring. The product that separated was filtered out and dried under vacuum, and 269 mg was obtained; LC/MS shows pure product.
  • Minocycline free base (175 mg, 0.38 mmol) was dissolved in 2 mL of THF, then 0.43 mL of methyl iodide was added. No crystallization of the product occurred.
  • the reaction mixture was concentrated and poured into 200 mL of cold ether/PET ether 40:60 with stirring. The product that separated was filtered out and dried under vacuum; the product was resubjected to the reaction conditions for 3 additional days, then precipitated from ether as before, and 123 mg of minocycline methiodide was obtained.
  • the crude product was dissolved in 2 mL of acetic acid, and 2 mL of water and 60 mg of zinc dust were added with stirring. After 15 minutes, the zinc powder was filtered out. The filtrate was diluted with 15 mL of water containing concentrated hydrochloric acid. The combined organic extracts were dried over magnesium sulfate and concentrated. The solid that separated was filtered out and dried under vacuum, and 150 mg of a yellow product was obtained.
  • MIC Minimum Inhibitory Concentrations
  • the study was an MIC evaluation for five test products, performed using a modification of the Macrodilution Broth Method outlined in NCCLS document M7-A5, Methods for Dilution Antimicrobial Susceptibility Tests for Bacteria that Grow Aerobically, Fifth Edition.
  • Each test product was evaluated, in duplicate, against challenge suspensions of ten microorganism strains. Prior to using each product for testing, it was diluted (w/v) in sterile Mueller-Hinton Broth (MHB) to achieve an initial concentration, based on a potency of 1000 ⁇ g/mg, of 160 ⁇ g/mL.
  • an initial challenge suspension containing approximately 1.0 ⁇ 10 9 CFU/mL was prepared for each microorganism by inoculating a test tube of Sodium Chloride Irrigation with microorganisms taken from the plates of solid media, prepared as described above.
  • the final challenge suspensions containing approximately 1.0 ⁇ 10 6 CFU/mL were prepared for each microorganism species, by placing a 0.2 mL aliquot of the 1.0 ⁇ 10 9 CFU/mL suspension into a sterile 250 mL polypropylene bottle, containing 200 mL of Mueller-Hinton Broth.
  • the final challenge suspensions were mixed thoroughly prior to use in testing.
  • the final plated dilutions were 10 ⁇ 3 , 10 ⁇ 4 and 10 ⁇ 5 . These plates were incubated at 35° ⁇ 2° C. until sufficient growth was observed (Table I).
  • the testing procedure was as follows: 30 mL aliquots of Mueller-Hinton Broth were dispensed into sterile bottles. A 30 mL aliquot of product (initial concentration of 160 ⁇ g/mL) was dispensed into the first of a series of 11 sterile bottles, each containing 30 mL of Mueller-Hinton Broth, and mixed thoroughly in order to achieve the 1:2 (v/v) dilution of product.
  • a 30 mL aliquot was removed from the bottle prepared as described above, and used to initiate a 1:2 (v/v) dilution series through the remaining 10 bottles (1:4, 1:8, 1:16, 1:32, 1:64, 1:128, 1:256, 1:512, 1:1,024 and 1:2,048 product dilutions), each containing 30 mL of Mueller-Hinton Broth. Each bottle was mixed thoroughly prior to removing the 30 mL aliquot required for the next dilution in the series. 1.0 mL aliquots of each product dilution prepared and 1.0 mL aliquots of the initial product preparation (160 ⁇ g/mL product concentration) were transferred to separate sterile test tubes.
  • a positive control tube growth control containing a 1.0 mL aliquot of Mueller-Hinton Broth and a 1.0 mL aliquot of the challenge suspension was prepared for each microorganism (Table I).
  • a negative (media) control tube sterility control; no microbial inoculation) of Mueller-Hinton Broth was also prepared.
  • Product turbidity controls were prepared for each product by transferring 1.0 mL aliquots of each product dilution into separate sterile test tubes. A 1.0 mL aliquot of sterile Mueller-Hinton Broth was introduced into each product dilution tube created and mixed thoroughly using a vortex mixer, thereby resulting in a final product dilution series identical to that described above. The challenge suspension/product dilution tubes and the controls were incubated at 35° ⁇ 2° C. for 20 hours, until good growth was apparent in the positive control tubes.
  • the tubes were examined for growth of the microorganisms, determined visually on the basis of turbidity.
  • MIC Minimum Inhibitory Concentration
  • TSB/TSA/MHB Escherichia coli (Tetracycline-Resistant) 51657 20 Hours 35° ⁇ 2° C.
  • TSB/TSA/MHB Escherichia coli (Tetracycline-Resistant) 51658 20 Hours 35° ⁇ 2° C.
  • TSB/TSA/MHB Escherichia coli (Tetracycline-Resistant) 51659 20 Hours 35° ⁇ 2° C.
  • TSB/TSA/MHB Staphylococcus aureus (Tetracycline- 27217 20 Hours 35° ⁇ 2° C.
  • TSB/TSA/MHB Resistant Staphylococcus aureus (Tetracycline- 27659 20 Hours 35° ⁇ 2° C. TSB/TSA/MHB Resistant) Staphylococcus aureus (Tetracycline- 27660 20 Hours 35° ⁇ 2° C. TSB/TSA/MHB Resistant) Staphylococcus aureus 29213 20 Hours 35° ⁇ 2° C. TSB/TSA/MHB *Initial Population plates were inadvertently incubated at room temperature (20°-27° C.) for approximately 48 hours prior to being incubated at 35° ⁇ 2° C. for approximately 72 hours.
  • Table II presents the Minimum Inhibitory Concentrations, expressed as product dilution, for each test product versus each of the 10 microorganisms tested.
  • Table III presents the Minimum Inhibitory Concentrations, expressed as product concentration ⁇ g/mL), for each test product versus each of the 10 microorganisms tested.
  • the initial solution prepared for each product had a concentration, based on a potency of 1000 ⁇ g/mg, of 160 ⁇ g/mL.
  • MMPs matrix degrading metalloproteinases
  • MMP1 activity was determined by the release of soluble 14 C labelled collagen fragments from 14 C acetylated rat skin type I collagen (Methods in Enzymology 80 711, 1981).
  • MMP2 activity was determined by the release of trichloracetic acid, TCA, soluble fragments from 14 C acetylated rat skin type I gelatin (Methods in Enzymology 248, 470, 1995. Biochem. J. 195, 1981)
  • Incubations were set at 37° C. for 16 hours. The reaction was stopped by cooling the incubations and addition of 50 ⁇ l cold 90% (w/v) trichloracetic acid with careful mixing. TCA precipitation was allowed to proceed for 15 minutes at 4° C. prior to cetrifugation at 10,000 g for 10 minutes at 4° C. 200 ⁇ l of the TCA supernatant was taken for the determination of radioactive content by scintillation counting (Packard Opti Phase Supermix scintillation fluid). Only data derived from within the linear part of the assay (10-70% lysis) were utilized. The percentage inhibition of MMP2 by each tetracycline concentration was plotted to derive the IC50 value, using linear regression analysis.
  • MMP3 activity was determined by the release of trichloracetic acid, TCA, soluble fragments from 14 C acetylated 13 casein (Sigma) as described in Methods in Enzymology 248, 451 (1995).
  • MMP7 activity was determined by the release of trichloracetic acid, TCA, soluble fragments from 14 C acetylated ⁇ casein (Sigma) as described in Methods in Enzymology 248, 451 (1995).
  • Incubations were at 37° C. for 16 hours. The incubation was stopped by the addition of 500 ⁇ l cold 18% TCA and standing on ice for 15 minutes. TCA precipitates were removed by centrifugation at 10,000 g for 10 minutes at 4° C., and 200 ⁇ l supernatant was taken for scintillation counting. Only data derived from within the linear part of the assay (10-60% lysis) were utilized. The percentage inhibition of MMP7 by each concentration of the candidate tetracycline were plotted to derive the IC50 value.
  • MMP9 activity was determined by the release of trichloracetic acid, TCA soluble fragments from 14 C acetylated rat skin type I (Methods in Enzymology 248, 470, 1995. Biochem. J. 195, 1981)
  • Incubations were at 37° C. for 16 hours. The incubations were stopped by cooling the incubations and addition of 50 ⁇ l cold 90% (w/v) trichloracetic acid with careful mixing. TCA precipitation was allowed to proceed for 15 minutes at 4° C. prior to centrifugation at 10,000 g for 10 minutes at 4° C. 200 ⁇ l of the TCA supernatant was taken for the determination of radioactive content by scintillation counting (Packard Opti Phase Supermix scintillation fluid). Only data derived from within the linear part of the assay (10-70% lysis) were utilized. The percentage inhibition of MMP9 by each tetracycline concentration were plotted to derive the IC50 value.
  • MMP14 activity was determined by the release of soluble 14 C labelled collagen fragments from 14 C acetylated rat skin type I collagen (Methods in Enzymology 80 711, 1981).
  • Another objective was to analyze the effects of specific, tetracycline derived compounds on the in-vitro chemo-invasive potential of:
  • the Membrane Invasion Culture System (MICS) in vitro chemo-invasion assay was chosen to measure changes in the invasive potential of specific human cancer cells in response to tetracycline derived compounds.
  • Stock solutions of each compound were hydrated in water containing 2% DMSO and pH 10.0. Upon solubilization, HCl was added to the solution to establish a pH of 7.5-8.0. This solution was then wrapped in foil and kept at 4° C. during the 24 hours of assay. Fresh compound was prepared for each assay.
  • the in vitro chemoinvasion analyses for tumor cell invasiveness are performed using the Membrane Invasion Culture System (MICS) as previously described.
  • the MICS system is a thermally treated plastic manifold system consisting of two matched sets of plates containing fourteen wells, 13 mm in diameter. Interposed between these plates was a polycarbonate filter containing 10 ⁇ m pores and coated with a defined matrix composed of human laminin/collagen IV/gelatin that formed a 35 ⁇ m thick barrier between the top and bottom sections of the wells in MICS. Prior to placing this barrier in place, the lower wells were filled with serum-free RPMI culture medium made 50% in conditioned medium obtained from 2-day-old cultures of human fibroblasts which had been cleared of cells and cellular debris by either centrifugation, or filtering through a 0.45 ⁇ m sterile filter.
  • the barrier was then placed on the lower wells and fresh serum-free medium added to the upper wells after assembly of the system. Fifty thousand tumor cells were then added to the wells either in the presence of the compound, or the DMSO vehicle (the controls). Of the 12 assay wells on each manifold, 3 randomized wells acted as controls, 3 wells received 1 ⁇ g/ml of compound, 3 received 10 ⁇ g/ml and 3 received 25 ⁇ g/ml. After 24 hours, cells and media were removed from the lower wells and replaced by phosphate buffered saline plus 2 mM EDTA. This solution was used to remove all cells from the lower well, and this wash was added to the recovered cells plus medium from the same well.
  • MDA-MB-231 C8161 RPMI-8226 PC-3 ML 1 7 B 20 B 5 B 5 20 35 B 20 10 15 2 47 B 60 B 25 B 25 B 32 B 40 B 49 B 56 B 21 B 21 B 29 B 13 B 24 B 24 B 2 20 15 30 B 20 B 25 B 27 B +8 +10 B +11 20 8 15 1 B +4 B 7 B 3 +8 +2 12 30 B 22 B 15 B +7 +23 B 27 B 3 +1 25 B +22 +3 +24 B 4 5 6 2 27 B 24 B 17 +4 +5 +14 10 +8 3 5 6 +4 46 B 15 27 B 35 B 6 4 36 44 B 35 46 B 17 37 B 42 B 6 +5 +17 B 27 B 18 B 33 B 32 B +6 +25 B +5 21 26 41 B 24 44 B 42 B 8 21 B 33 B 12 B 10 5 13 32 B 30 B 7 +18 +41 B +27 7 14 16 +14 B 12 +2 0 0 3 8 +10 +5 +13 13 B 41 B 40 B 11 3 +2 0 +4 0 9

Abstract

A treatment for inhibiting microbial or tumor growth is disclosed, which comprises adding to a subject in need of treatment an effective amount of one or more tetracycline derivatives.

Description

    FIELD OF INVENTION
  • The present invention relates to novel tetracycline derivatives, methods for producing the novel derivatives and methods of using these derivatives.
  • BACKGROUND OF THE INVENTION
  • The compound tetracycline, exhibits the following general structure
    Figure US20050245491A9-20051103-C00001
  • The numbering system of the ring nucleus is as follows:
    Figure US20050245491A9-20051103-C00002
  • Tetracycline as well as the 5-OH (Terramycin) and 7-Cl (Aureomycin) derivatives exist in nature, and are well known antibiotics. Natural tetracycline may be modified without losing their antibiotic properties, although certain elements of the structure must be retained. The modifications that may and may not be made to the basic tetracycline structure have been reviewed by Mitscher in The Chemistry of Tetracyclines, Chapter 6, Marcel Dekker, Publishers, New York (1978). According to Mitscher, the substituents at positions 5-9 of the tetracycline ring system may be modified without the complete loss of antibiotic properties. Changes to the basic ring system or replacement of the substituents at positions 1-4 and 10-12, however, generally lead to synthetic tetracyclines with substantially less or effectively no antimicrobial activity. Some examples of chemically modified non-antimicrobial tetracyclines (hereinafter CMT) are 4-dedimethylaminotetraccyline, 4-dedimethylaminosancycline (6-demethyl- 6 -deoxy-4-dedimethylaminotetracycline), 4-dedimethylaminominocycline (7-dimethylamino-4-dedimethylaminotetracycline), and 4-dedimethylaminodoxycycline (5-hydroxy-6-deoxy-4-dedimethyaminotetracycline).
  • Some 4-dedimethylaminotetracycline derivatives are disclosed in U.S. Pat. Nos. 3,029,284 and 5,122,519. They include 6-demethyl-6-deoxy-4-dedimethylaminotetraccyline and 5-hydroxy-6-deoxy-4-dedimethylaminotetracycline with hydrogen and other substituents at C7, and the C9 positions on the D ring. These substitutents include amino, nitro, di (lower alkyl) amino, and mono (lower alkyl) amino or halogen. The 6-demethyl-6-deoxy-4-dedimethylaminotetracycline derivatives and 5-hydroxy-6-deoxy-4-dedimethylaminotetracycline derivatives are said to be useful as antimicrobial agents.
  • Other 4-dedimethylaminotetracycline derivatives with an oxime group at the C4 position on the A ring are disclosed in U.S. Pat. Nos. 3,622,627 and 3,824,285. These oxime derivatives have hydrogen and halogen as substitutents at the C7 position and include 7-halo-6-demethyl-6-deoxy-4-dedimethylamino-4-oximinotetracycline, and 7-halo-5-hydroxy-6-deoxy-4-dedimethylamino-4-oximinotetracycline.
  • Alkylamino (NH-alkyl), and alkylhydrazone (N—NH-alkyl) groups have been substituted on the A ring at the C4 position of 4-dedimethylaminotetracycline. These compounds are known for their antimicrobial properties. See U.S. Pat. Nos. 3,345,370, 3,609,188, 3,622,627, 3,502,660, 3,509,184, 3,502,696, 3,515,731, 3,265,732, 5,122,519, 3,849,493, 3,772,363, and 3,829,453.
  • In addition to their antimicrobial properties, tetracyclines have been described as having a number of other uses. For example, tetracyclines are also known to inhibit the activity of collagen destructive enzymes, such as matrix metalloproteinases (MMP), including collagenases (MMP-1), gelatinase (MMP-2) and stromelysin (MMP-3). Golub et al., J. Periodont. Res. 20:12-23 (1985); Golub et al. Crit. Revs. Oral Biol. Med. 2:297-322 (1991); U.S. Pat. Nos. 4,666,897; 4,704,383; 4,935,411; 4,935,412. Also, tetracyclines have been known to inhibit wasting and protein degradation in mammalian skeletal muscle, U.S. Pat. No. 5,045,538, and to enhance IL-10 production in mammalian cells.
  • Furthermore, tetracyclines were reported to enhance bone protein synthesis in U.S. Pat. No. Re. 34,656, and to reduce bone resorption in organ culture in U.S. Pat. No. 4,704,383.
  • Similarly, U.S. Pat. No. 5,532,227 to Golub et al, discloses that tetracyclines can ameliorate the excessive glycosylation of proteins. In particular, tetracyclines inhibit the excessive collagen cross linking which results from excessive glycosylation of collagen in diabetes.
  • Tetracyclines are known to inhibit excessive phospholipase A2 activity involved in inflammatory conditions such as psoriasis as disclosed in U.S. Pat. No. 5,532,227. In addition, tetracyclines are also known to inhibit cycloxygenase-2 (COX-2), tumor necrosis factor (TNF), nitric oxide and IL-1 (interleukin-1).
  • These properties cause the tetracyclines to be useful in treating a number of diseases. For example, there have been a number of suggestions that tetracyclines, including non-antimicrobial tetracyclines, are effective in treating arthritis. See, for example, Greenwald, et al. “Tetracyclines Suppress Metalloproteinase Activity in Adjuvant Arthritis and, in Combination with Flurbioprofen, Ameliorate Bone Damage,” Journal of Rheumatology 19:927-938 (1992); Greenwald et al., “Treatment of Destructive Arthritic Disorders with MMP Inhibitors; Potential Role of Tetracyclines in Inhibition of Matrix Metalloproteinases: Therapeutic Potential,” Annals of the New York Academy of Sciences 732:181-198 (1994); Kloppenburg, et al. “Minocycline in Active Rheumatoid Arthritis,” Arthritis Rheum 37:629-636 (1994); Ryan et al., “Potential of Tetracycline to Modify Cartilage Breakdown in Osteoarthritis,” Current Opinion in Rheumatology 8:238-247 (1996); O'Dell et al, “Treatment of Early Rheumatoid Arthritis with Minocycline or Placebo,” Arthritis Rheum 40:842-848 (1997).
  • Tetracyclines have also been suggested for use in treating skin diseases. For example, White et al., Lancet, April 29, p. 966 (1989) report that the tetracycline minocyline is effective in treating dystrophic epidermolysis bullosa, which is a life-threatening skin condition believed to be related to excess collagenase.
  • Furthermore, studies have also suggested that tetracyclines and inhibitors of metalloproteinases inhibit tumor progression, DeClerck et al., Annals N.Y. Acad. Sci., 732:222-232 (1994), bone resorption, Rifkin et al., Annals N.Y. Acad. Sci., 732:165-180 (1994), angiogenesis, Maragoudakis et al., Br. J. Pharmacol., 111:894-902 (1994), and may have anti-inflammatory properties, Ramamurthy et al., Annals N.Y. Acad. Sci., 732, 427-430 (1994).
  • Based on the foregoing, tetracyclines have been found to be effective in treating numerous diseases and conditions. Therefore, there is a need for new and even more useful derivatives.
  • DESCRIPTION OF THE PREFERRED EMBODIMENTS
  • Compounds disclosed herein are derivatives of tetracycline, which exhibit antimicrobial and/or anti-cancer activity.
  • In a preferred embodiment, the present invention relates to a treatment for inhibiting microbial or tumor growth which comprises adding to a subject in need of said treatment an effective amount of a tetracycline composition or salt thereof, comprising a general formula:
    Figure US20050245491A9-20051103-C00003

    wherein R is selected from CH2N(RaRb) or
    Figure US20050245491A9-20051103-C00004
      • RaRb are C1 to C6 alkyl;
      • Rc and Rd are (CH2)n CHRe, n is 0 or 1 and Re is H, C1 to C6 alkyl, or NH2, and X is NH, S, or CH2;
      • R1 is H or OH;
      • R2 is H, OH, ═O, or OCOR8, where R8 is C1 to C6 alkyl, or alternatively, R2 is N(R9)2, where R9 is hydrogen or C1 to C6 lower alkyl, or R9 CO, with the proviso that when R2 is keto or N(R9)2, then R3 and R4 are H or CH3, and R3 and R4 are not both CH3 or H;
      • 5a hydrogen is α or β;
      • R3 and R4 are H, CH3 or F, with the proviso that when R3 is CH3, then R4 is H or F, and when R4 is CH3, then R3 is H or F and R4 and R5 are not both F;
      • R5 and R7 are halogen, H, NO2, N3, N2 +, C2H5OC(S)S—, CN, NR10R11, where R10 and R11 are H, C1 to C10 alkyl, and R12(CH2)nCO—, where n is 0 to 5 and R9 is H, NH2, mono or disubstituted amino selected from straight, branched or cyclized C1 to C10 alkyl groups, with the proviso that R10 and R11 are not both R12(CH2)nCO—;
      • R7, alternatively, is tertiary butyl; and
      • R6 is halogen, acetylene, R13—C6H5, where R13 is NO2, halogen, acetylamino, amino, phenyl, alkyl, or alkoxy.
  • In a further preferred embodiment, the present invention relates to a treatment for inhibiting microbial or tumor growth which comprises adding to a subject in need of said treatment an effective amount of a tetracycline composition or salt thereof, comprising a general formula:
    Figure US20050245491A9-20051103-C00005

    wherein R is selected from CH2N(RaRb) or
    Figure US20050245491A9-20051103-C00006

    where RaRb are C1 to C6 alkyl;
      • Rc and Rd are (CH2)n CHRe, n is 0 or 1 and Re is H, C1 to C6 alkyl, or NH2, and X is NH, S, or CH2;
      • R1 is H or OH;
      • R2 is H or OH;
      • R3is H or CH3;
      • R4 and R6 are halogen, H, NO2, N3, N2 +, C2H5OC(S)S—, CN, NR7R8, where R7 and R8 are H, C1 to C10 alkyl, and R9(CH2)nCO—, where n is 0 to 5 and R9 is H, NH2, mono or disubstituted amino selected from straight, branched or cyclic C1-C10 alkyl, with the proviso that R7 and R8 are not both R9(CH2)nCO—; and
      • R6 is H or halogen, acetylene, R10—C6H5, where R10 is NO2, halogen, acetylamino, amino, phenyl, alkyl, or alkoxy.
  • In a further preferred embodiment, the present invention relates to a treatment for inhibiting microbial or tumor growth which comprises adding to a subject in need of said treatment an effective amount of a tetracycline composition or salt thereof, comprising a general formula:
    Figure US20050245491A9-20051103-C00007

    wherein R is selected from CH2N(RaRb) or CH2N
    Figure US20050245491A9-20051103-C00008

    where RaRb are C1-C6 alkyl;
      • Rc and Rd are (CH2)n CHRe, n is 0 or 1 and Re is H, C1 to C6 alkyl, or NH2, and X is NH, S, or CH2;
      • R1 and R2 are H or N(R9)2, where R9 is H or C1 to C6 alkyl with the proviso that R1 and R2 are not both N(R9)2, R1 and R2 are also N(R10)3 I, where R10 is C1 to C6 alkyl, with the proviso that R1 and R1 are not both N(R10)3 I;
      • R3 is H;
      • R4 and R5 are H, CH3 or F, with the proviso that when R4 is CH3, then R5 is H or F, and when R5 is CH3, then R4 is H or F, and R4 and R5 are not both F;
      • R6 and R8 are halogen, H, NO2, N3, N2 +, C2H5OC(S)S—, CN, NR11R12, where R11 and R12 are H, C1 to C10 alkyl, and R13(CH2)nCO—, where n is 0 to 5 and R13 is H, NH2, mono or disubstituted amino selected from straight, branched or cyclized C1-C10 alkyl groups, with the proviso that R10 and R11 are not both R12 (CH2)nCO—; and
      • R8 can also be tertiary butyl and R7 is H or halogen.
  • In an additional preferred embodiment, the present invention relates to a treatment for inhibiting microbial or tumor growth which comprises adding to a subject in need of said treatment an effective amount of a tetracycline composition or salt thereof, comprising a general formula:
    Figure US20050245491A9-20051103-C00009

    wherein R is selected from CH2N(RaRb) or
    Figure US20050245491A9-20051103-C00010
      • Ra Rb are C1-C6;
      • Rc and Rd are (CH2)n CHRe, n is 0 or 1, Re is H, alkyl (C1 to C6), NH2, and X is NH, S, or CH2;
      • R1 and R2 are H or N(R9)2 where R9 is hydrogen or lower alkyl (C1 to C6), with the proviso that R1 and R2 can not both be N(R9)2.
      • R3 is H, OH, ═O, OCOR10, where R10 is C1 to C6, or alternatively, R3 is N(R9)2 where R9 is hydrogen or C1 to C6, with the proviso that when R23 is ═O or N(R9)2, then R4 and R5 are H or CH3 and R4 and R5 are not both CH3 or H;
      • 5a hydrogen is α or β;
      • R4 and R5 are H, CH3 or F, with the proviso that when R4 is CH3, then R5 is H or F, and when R5 is CH3 then R4 is H or F and R4 and R5 are not both F;
      • R6 and R8 are halogen, H, NO2, N3, N2 +, C2H5OC(S)S—, CN, NR10R11, where R10 and R11 are H, alkyl (C1 to C10), and R12 (CH2)nCO—, where n is 0 to 5 and R12 is H, NH2, mono or disubstituted amino selected from straight, branched or cyclized C1 to C10 groups, with the proviso that R10 and R11 are both R12 (CH2)nCO—, or, alternatively, R8 is tertiary butyl;
      • R7 is H or halogen, acetylene, R12—C6H5, where R12 is NO2, halogen, acetylamino, amino, phenyl, alkyl, or alkoxy.
  • In a further preferred embodiment, the present invention relates to a treatment for inhibiting microbial or tumor growth which comprises adding to a subject in need of said treatment an effective amount of a tetracycline composition or salt thereof, comprising a general formula:
    Figure US20050245491A9-20051103-C00011

    wherein R1 is H or N(CH3)2; R2 is H, CH3 or OCOCH3; R3 is H or CH3; R4 is H; R5 is H, N(CH3)2, NH2, N(C4H9)2, N(C6H13)2, N(3,3-dimethylbutyl)2, N3, N2 +, NO2, NHCOCH3, NH(n-propyl)2, NH-isobutyl, NH-isobutylmethyl, NH(cyclobutyl), NH(cyclobutyl methyl); and R6 is H, NO2, NH2, (CH3)2 CHNH, N3, NHCOCH3, N2 +, N3 or (CH3)3C.
  • In a further preferred embodiment, the present invention relates to a treatment for inhibiting microbial or tumor growth which comprises adding to a subject in need of said treatment an effective amount of a tetracycline composition or salt thereof, comprising a general formula:
    Figure US20050245491A9-20051103-C00012

    wherein R1 is H or N(CH3)2; R2 is H, OH or OCOCH3; R3 is H or CH3; R4 is H; R5 is H, N(CH3)2; N(C4H9)2, N(C6H13)2, N(3,3-dimethylbutyl)2; N3, N2 +, NO2, NHCOCH3, NH(n-propyl)2, NH-isobutyl, NH-isobutylmethyl, N(CH3CO)(isobutyl), NH(cyclobutyl), NH(cyclobutyl methyl), or NH2; and R6 is H, NO2, NH2, (CH3)2 CHNH, N3, NHCOCH3, N2 +, or (CH3)3C.
  • The following examples describe the preparation of various substances in accordance with the present invention.
  • HPLC analyses were performed on Waters reverse phase C18-symmetry columns using water (containing 0.1% TFA) and acetonitrile as eluents
  • Analytical LC/MS runs were performed on a Hewlett Packard Series 1100 liquid chromatrographer equipped with a series 1100 MSD detector, with ES ionization mode and UV detection at 270 nm, using a 4.6×100 mm column with a flow rate of 0.4 mL/min. Preparative HPLC was performed on a Gilson serial HPLC with UV detection at 270 nm, using a 19×150 mm column and a flow rate of 15 mL/min 1H NMR were recorded on a Bruker 300 MHz spectrometer in deuterated methanol.
  • 9-Nitro-doxycycline sulfate
  • To a stirred solution of doxycycline hydrochloride (1 g, 2.08 mmol) in 30 mL of concentrated sulfuric acid at 0° C. was added potassium nitrate (252 mg, 2.49 mmol). The resulting mixture was stirred for 20 min at 0° C., then poured into 1.2 L cold ether with stirring. The solid which precipitated was filtered out, washed with cold ether and dried under vacuum, and 1.22 g of cream colored product was obtained. If needed, the product was purified by dissolving in methanol, filtering and pouring the filtrate into ether. The solid which separated was filtered out and dried.
  • 9-Amino-doxycycline sulfate
  • To a stirred solution of 9-nitro-doxycycline sulfate (1 g, 1.7 mmol) in 35 mL of ethylene glycol monomethyl ether and 5 mL of methanol was added 700 mg of 10% Pd/C. The reaction mixture was hydrogenated at atmospheric pressure for 6 hrs. The catalyst was filtered off through celite washing with methanol. The filtrate was concentrated, then added dropwise to 800 mL cold ether with stirring. The solid that separated was filtered out, washed with cold ether and dried under vacuum, and 843 mg of cream colored product was obtained.
  • 9-Isopropylamino-doxycycline sulfate
  • To a stirred solution of 9-amino-doxycycline sulfate (100 mg, 0.18 mmol) on 10 mL of ethylene glycol monomethyl ether was added 0.05 ml of concentrated sulfuric acid, 0.5 mL acetone and 100 mg of 10% Pd/C. The resulting mixture was hydrogenated at atmospheric pressure for 1 h 15 min, then the catalyst was filtered through celite, washing with methanol. The filtrate was concentrated, then poured into 150 mL of cold ether with stirring. The solid that separated was filtered out, washed with cold ether and dried under vacuum, and 109 mg of cream colored product was obtained. The product was purified by dissolving in methanol, filtering and pouring the filtrate into ether. The solid which formed was filtered out and dried.
  • 9-Acetamido-doxycycline sulfate
  • To a stirred solution of 9-amino-doxycycline sulfate (72 mg, 0.13 mmol) in 1.5 mL of 1,3-dimethyl-2-imidazolidinone were added 70 mg sodium bicarbonate followed by 0.05 mL of acetyl chloride. The mixture was stirred at room temperature for 1 hour and then filtered. The filtrate was concentrated and poured into 100 ml cold ether with stirring. The solid that separated was filtered out and dried under vacuum, and 92 mg was obtained.
  • 9-Diazonium-doxycycline sulfate hydrochloride
  • To a stirred solution of 9-amino-doxycycline sulfate (300 mg, 0.54 mmol) in 9 mL of 0.1N hydrochloric acid in methanol cooled in an ice bath was added 0.3 mL of n-butyl nitrite. The solution was stirred at 0° C. for 30 minutes then poured into 300 mL of cold ether with stirring. The solid that separated was filtered out and dried under vacuum, and 256 mg of an orange-brown product was obtained.
  • 9-Azido-doxycycline sulfate
  • To a stirred solution of 9-diazonium-doxycycline sulfate hydrochloride (80 mg, 0.14 mmol) in 4.5 mL of 0.1N hydrochloric acid in methanol was added 10 mg of sodium azide. The solution was stirred at room temperature for 2 hours, then poured into 100 mL of cold ether with stirring. The solid that separated was filtered out and dried under vacuum, and 43 mg of product was obtained.
  • 9-(4-flurophenyl)-doxycycline
  • To a solution of 9-diazonium-doxycycline sulfate hydrochloride (358 mg, 0.59 mmol) and 4-flurophenyl boronic acid (107 mg, 0.76 mmol) in 4 mL of methanol was added 18 mg of palladium(II)acetate and the resulting mixture was stirred at room temperature for 16 hours. The catalyst was filtered off and the residue was concentrated and purified by preparative HPLC. The collected fractions from HPLC were concentrated and poured into 20 ml of cold ether. The solid that was separated was filtered out and dried under vacuum, and 32 mg of product was obtained.
  • 7-[1,2-bis(carbobenzyloxy)hydrazino]-doxycycline
  • To a stirred solution of doxycycline hydrochloride (300 mg, 0.62 mmol) in 2.5 ml of THF and 3.2 mL of methanesulfonic acid at 0° C. was added 230 mg of dibenzyazodicarboxylate. The resulting mixture was stirred at 0° C. for 2 hours, then poured into 400 mL of cold ether. The solid that separated was filtered out and dried under vacuum, and 479 mg of an off-white product was obtained.
  • 7-Amino-doxycycline
  • To a solution of 7-[1,2-bis(carbobenzyloxy)hydrazino]-doxycycline (478 mg) in 30 mL of ethylene glycol monomethyl ether was added 200 mg of 10% Pd/C and the resulting mixture was hydrogenated for 3 hours at room temperature. The catalyst was filtered through celite washing with methanol and the filtrate was concentrated and poured into 500 mL cold ether. The solid that separated was filtered out and dried under vacuum, and 268 mg of product was obtained.
  • 7-Dimethylamino-doxycycline sulfate
  • To a solution of 7-amino-doxycycline (100 mg) in 8 mL of ethylene glycol monomethyl ether were added 1 mL of a 37% aqueous formaldehyde solution, 0.05 mL of concentrated sulfuric acid and 100 mg of 10% Pd/C. The resulting mixture was hydrogenated at atmospheric pressure for 6 hours, then the catalyst was filtered through celite, washing with methanol. The residue was concentrated and poured into 100 ml cold ether with stirring. The solid that separated was filtered out with difficulty, being very gluey. The solid was immediately redissolved in methanol and poured into 100 ml cold ether with stirring. The solid that separated was filtered out and dried under vacuum, and 89 mg of product was obtained.
  • 7-Dipropyl-doxycycline sulfate
  • To a solution of 7-amino-doxycycline (90 mg) in 8 ML of ethylene glycol monomethyl ether were added 0.5 mL of propionaldehyde, 0.05 mL of concentrated sulfuric acid and 80 mg of 10% Pd/C. The resulting mixture was hydrogenated at atmospheric pressure for 5 hours, then the catalyst was filtered through celite, washing with methanol. The residue was concentrated and poured into 100 ml cold ether with stirring. The solid that separated was filtered out and dried under vacuum, and 415 mg of product was obtained.
  • 7-Dibutylamino-doxycycline sulfate
  • To a solution of 7-amino-doxycycline (100 mg, 0.2 mmol) in 6 mL of ethylene glycol monomethyl ether were added 0.6 mL of butyraldehyde, 0.05 mL of concentrated sulfuric acid and 100 mg of 10% Pd/C. The resulting mixture was hydrogenated at atmospheric pressure for 4 hours, then the catalyst was filtered through celite, washing with methanol. The residue was concentrated and poured into 100 ml cold ether with stirring. The solid that separated was filtered out and dried under vacuum, and 103 mg of product was obtained.
  • 7-Di(3,3-dimethyl)butylamino-doxycycline sulfate
  • To a solution of 7-amino-doxycycline (100 mg, 0.2 mmol) in 6 mL of ethylene glycol monomethyl ether were added 0.5 mL of 3,3-dimethyl butyraldehyde, 0.05 mL of concentrated sulfuric acid and 100 mg of 10% Pd/C. The resulting mixture was hydrogenated at atmospheric pressure for 4 hrs, then the catalyst was filtered through celite, washing with methanol. The residue was concentrated and poured into 100 ml cold ether with stirring. The solid that separated was filtered out and dried under vacuum, and 122 mg of product was obtained.
  • 7-Dihexylamino-doxycycline sulfate
  • To a solution of 7-amino-doxycycline (100 mg, 0.2 mmol) in 6 mL of ethylene glycol monomethyl ether were added 0.6 mL of hexanal, 0.05 mL of concentrated sulfuric acid and 100 mg of 10% Pd/C. The resulting mixture was hydrogenated at atmospheric pressure for 4 hours, then the catalyst was filtered through celite, washing with methanol. The residue was concentrated and poured into 100 ml cold ether with stirring. The solid that separated was filtered out and dried under vacuum, and 129 mg of product was obtained.
  • 7-Isopropylamino-doxycycline sulfate
  • To a solution of 7-amino-doxycycline (90 mg, 0.2 mmol) in 8 mL of ethylene glycol monomethyl ether were added 0.5 mL of isobutyraldehyde, 0.05 mL of concentrated sulfuric acid and 90 mg of 10% Pd/C. The resulting mixture was hydrogenated at atmospheric pressure for 6 hours, then the catalyst was filtered through celite, washing with methanol. The residue was concentrated and poured into 100 ml cold ether with stirring. The solid that separated was filtered out and dried under vacuum, and 142 mg of product was obtained.
  • 7-Methyl-7-isopropylamino-doxycycline sulfate
  • To a solution of 7-isopropylamino-doxycycline (60 mg) in 4 mL of ethylene glycol monomethyl ether were added 0.4 mL of a 37% aqueous solution of formaldehyde, 0.05 mL of concentrated sulfuric acid and 50 mg of 10% Pd/C. The resulting mixture was hydrogenated at atmospheric pressure for 2 hours, then the catalyst was filtered through celite, washing with methanol. The residue was concentrated and poured into 80 ml cold ether with stirring. The solid that separated was filtered out and dried under vacuum, and 52 mg of product was obtained.
  • 7-Cyclobutylamino-doxycycline sulfate
  • To a solution of 7-amino-doxycycline (80 mg) in 7 mL of ethylene glycol monomethyl ether were added 0.3 mL of cyclobutanone, 0.04 mL of concentrated sulfuric acid and 60 mg of 10% Pd/C. The resulting mixture was hydrogenated at atmospheric pressure for 24 hours, then the catalyst was filtered through celite, washing with methanol. The residue was concentrated and poured into 100 ml cold ether with stirring. The solid that separated was filtered out and dried under vacuum, and 85 mg of product was obtained.
  • 7-Methyl-7-cyclobutylamino-doxycycline sulfate
  • To a solution of 7-cyclobutylamino-doxycycline (40 mg) in 3.5 mL of ethylene glycol monomethyl ether were added 0.3 mL of a 37% aqueous solution of formaldehyde, 0.03 mL of concentrated sulfuric acid and 40 mg of 10% Pd/C. The resulting mixture was hydrogenated at atmospheric pressure for 6 hrs, then the catalyst was filtered through celite, washing with methanol. The residue was concentrated and poured into 80 ml cold ether with stirring. The solid that separated was filtered out and dried under vacuum, and 35 mg of product was obtained.
  • 7-Acetamido-doxycycline sulfate
  • To a stirred solution of 7-amino-doxycycline (200 mg) in 3 mL of 1,3-dimethyl-2-imidazolidinone were added 200 mg sodium bicarbonate followed by 0.09 mL of acetyl chloride. The mixture was stirred at room temperature for 1 hr and then filtered. The filtrate was concentrated and poured into 200 ml cold ether with stirring. The product which precipitated was filtered out and dried under vacuum. Obtained 202 mg of product.
  • 7-acetamido-7-isopropyl-doxycycline sulfate
  • To a stirred solution of 7-isobutylamino-doxycycline (60 mg) in 1.5 mL of 1,3-dimethyl-2-imidazolidinone were added 60 mg sodium bicarbonate followed by 0.05 mL of acetyl chloride. The mixture as stirred at room temperature for 1 hr 30 min and then filtered. The filtrate was concentrated and poured into 80 ml cold ether with stirring. The product which precipitated was filtered out and dried under vacuum.
  • 7-Diazonium-doxycycline hydrochloride or tetrafluoroborate
  • To a stirred solution of 7-amino-doxycycline (200 mg, 0.4 mmol) in 4 mL of 0.1N hydrochloric acid in methanol cooled in an ice bath was added 0.2 mL of n-butyl nitrite. The solution was stirred at 0° C. for 30 minutes, then poured into 200 mL of cold ether with stirring. The solid that separated was filtered out and dried under vacuum, and 173 mg of product was obtained. The diazonium tetrafluoroborate salt was prepared by using tetrafluoroboric acid (54% solution in ether) instead of hydrochloric acid.
  • 7-Azido-doxycycline
  • To a stirred solution of 7-diazonium-doxycycline hydrochloride (80 mg, 0.14 mmol) in 4.5 mL of 0.1N hydrochloric acid in methanol cooled on an ice bath was added 12 mg of sodium azide. The solution was stirred at 0° C. for 2 hours, then poured into 100 mL of cold ether with stirring. The solid that separated was filtered out and dried under vacuum, and 43 mg of product was obtained.
  • 7-Amino-9-nitro-doxycycline sulfate
  • To a stirred solution of 7-amino-doxycycline (220 g, 0.44 mmol) in 4 mL of concentrated sulfuric acid at 0° C. was added potassium nitrate (52 mg, 0.51 mmol). The resulting mixture was stirred for 15 minutes at 0° C., then poured into 300 mL cold ether with stirring. The solid that separated was filtered out, washed with cold ether and dried under vacuum. The product was redissolved in methanol, filtered and the filtrate was poured into ether. The solid that separated was filtered out and dried under vacuum, and 222 g of product was obtained.
  • 7-Dimethylamino-9-nitro-doxycycline sulfate
  • To a stirred solution of 7-dimethylamino-doxycycline (40 g, 0.07 mmol) in 1.5 mL of concentrated sulfuric acid at 0° C. was added potassium nitrate (12 mg, 0.12 mmol). The resulting mixture was stirred for 30 min at 0° C., then poured into 60 mL cold ether with stirring. The solid that separated was filtered out, washed with cold ether and dried under vacuum. LC/MS shows mostly one product containing two nitro groups (possibly a nitrate ester of the desired product).
  • 7-Dimethylamino-9-diazonium-doxycycline sulfate hydrochloride
  • To a stirred solution of 7-dimethylamino-9-amino-doxycycline (50 mg) in 1.5 mL of 0.1N hydrochloric acid in methanol cooled in an ice bath was added 0.05 mL of a n-butyl nitrite. The solution was stirred at 0° C. for 30 minutes, then poured into 80 mL of cold ether with stirring. The solid that separated was filtered out and dried under vacuum, and 38 mg of product was obtained.
  • 7-Acetamido-9-nitro-doxycycline sulfate
  • To a stirred solution of 7-acetamino-doxycycline (60 g, 0.095 mmol) in 2 mL of concentrated sulfuric acid at 0° C. was added potassium nitrate (11 mg, 0.11 mmol). The resulting mixture was stirred for 5 minutes at 0° C., then poured into 80 mL cold ether with stirring. The solid that separated was filtered out, washed with cold ether and dried under vacuum.
  • 7-Acetamido-9-amino-doxycycline sulfate
  • To a stirred solution of 7-acetamido-9-nitro-doxycycline sulfate (77 mg, 0.12 mmol) in 7 mL of ethylene glycol monomethyl ether was added 60 mg of 10% Pd/C. The reaction mixture was hydrogenated at atmospheric pressure for 7 hours. The catalyst was filtered off through celite washing with methanol. The filtrate was concentrated, then added dropwise to 100 mL cold ether with stirring. The solid that separated was filtered out, washed with cold ether and dried under vacuum, and 62 mg of product was obtained.
  • 7-Acetamido-9-diazonium-doxycycline sulfate hydrochloride
  • To a stirred solution of 7-acetamido-9-amino-doxycycline (100 mg) in 3 mL of 0.1N hydrochloric acid in methanol cooled in an ice bath was added 0.1 mL of n-butyl nitrite. The solution was stirred at 0° C. for 30 minutes, then poured into 100 mL of cold ether with stirring. The solid that separated was filtered out and dried under vacuum, and 92 mg of product was obtained.
  • 7-Acetamido-9-azido-doxycycline sulfate
  • To a stirred solution of 7-acetamido-9-diazonium-doxycycline (112 mg, 0.19 mmol) in 6 mL of 0.1N hydrochloric acid in methanol cooled in an ice bath was added 14 mg (0.21 mmol) of sodium azide. The solution was stirred at 0° C. for 35 minutes, then poured into 150 mL of cold ether with stirring. The solid that separated was filtered out and dried under vacuum, and 94 mg of product was obtained.
  • Doxycycline Methiodide
  • Doxycycline free base (315 mg) was dissolved in 2 mL of methanol and 10 mL of THF, then 1 mL of methyl iodide was added. The reaction mixture was kept for 5 days; no crystallization of the product occurred. The reaction mixture was concentrated and poured into 300 mL of cold ether with stirring. The product that separated was filtered out and dried under vacuum, and 269 mg was obtained; LC/MS shows pure product.
  • 5-Acetoxy-doxycycline
  • A solution of doxycycline (100 mg) in 3 mL of a 30% HBr in acetic acid solution was stirred at room temperature for 3.5 days; the reaction mixture was then poured into 100 mL of cold ether with stirring and the product that separated was filtered out. LC/MS showed incomplete conversion of starting material, so the product (86 mg) was resubjected to the reaction conditions for 9 hrs, and then isolated in the same way.
  • 9-tert-butyl-doxycycline
  • A solution of doxycycline (100 mg) in 2 mL of tert-butanol and 3 mL of methanesulfonic acid was stirred at room temperature for 18 hours, then poured into 100 mL of cold water and extracted with n-butanol. The organic extracts were basified with a 1N sodium hydroxide solution and the pH was quickly adjusted to 6.5-7 with concentrated hydrochloric acid. A solid precipitated which was filtered out, then dissolved in methanol, concentrated and added dropwise to 30 mL of an ether/PET ether cold solution with stirring. The solid that separated was filter out and dried under vacuum.
  • 9-tert-butyl-7-nitro-doxycycline
  • A solution of doxycycline hydrochloride (2 g) in 10 mL of tert-butanol and 30 mL of methanesulfonic acid was stirred at room temperature for 3 hours, then 500 mg of potassium nitrate was added and the resulting mixture stirred for an additional hour. The reaction mixture was poured in 100 ml cold solution of water containing 23 g of sodium hydroxide; more dilute NaOH solution was added dropwise until the reaction mixture became basic. The pH was quickly adjusted to 6.5-7 with concentrated hydrochloric acid. The gluey dark solid that separated was filtered out. The aqueous filtrate was extracted once with n-butanol to recover more product. The crude product was dissolved in the minimum amount of methanol and purified by HPLC. The combined fractions from HPLC were concentrated to dryness, dissolved in methanol and added dropwise to 50 mL of a 3:1 mixture of ether/PET ether. The solid that separated was filtered out, and 788 mg of pale yellow product was obtained.
  • 9-tert-butyl-7-amino-doxycycline
  • To a solution of 9-tert-butyl-7-doxycycline (500 mg) in 25 mL of ethylene glycol monomethyl ether was added 350 mg of 10% Pd/C and the resulting mixture was hydrogenated at atmospheric pressure for 17 hours. The catalyst was filtered through celite, washing with methanol. The residue was concentrated, dissolved in THF and added dropwise to a cold 1:1 solution of ether/PET ether with stirring. The product that precipitated was filtered out and dried under vacuum, and 452 mg of product was obtained.
  • 9-tert-butyl-7-diazonium-doxycycline
  • To a stirred solution of 9-tert-butyl-7-amino-doxycycline (93 mg, 0.18 mmol) in 2.5 mL of 0.1N hydrochloric acid in methanol cooled in an ice bath was added 0.1 mL of n-butyl nitrite. The solution was stirred at 0° C. for 30 minutes, and then half was poured into 30 mL of a 3:1 cold mixture of ether/PET ether with stirring. The product stuck to the bottom of the beaker as a gum. It was redissolved in methanol and concentrated, the residue was poured into 20 mL of cold ether and the solid that separated was filtered out immediately and dried under vacuum; 42 mg of product was obtained. The other half of the reaction mixture was used directly for the 9-tert-butyl material below.
  • 9-tert-butyl-7-azido-doxycycline
  • To a stirred solution of 9-tert-butyl-7-diazonium-doxycycline reaction mixture (1.5 mL) at 0° C. were added 8 mg (0.12 mmol) of sodium azide. The solution was stirred for 3 hours while allowed to warm to room temperature. After concentration of the reaction mixture, the residue was poured into a cold 3:1 mixture of ether/PET ether with stirring. No solid separated, so the solution was concentrated to dryness to obtain 35 mg of product.
  • 9-tert-butyl-7-dimethylamino-doxycycline
  • To a stirred reaction mixture from 9-tert-butyl-7-amino-doxycycline containing 167 mg of 9-tert-butyl-7-amino-doxycycline in 10 mL of ethylene glycol monomethyl ether and 115 mg of 10% Pd/C was added 1.2 ml of a 37% aqueous solution of formaldehyde. The resulting mixture was hydrogenated for 3.5 hours at atmospheric pressure, then the catalyst was filtered through celite washing with methanol. The filtrate was concentrated and the residue was poured into 10 mL of cold ether with stirring. The product that separated was filtered out and dried under vacuum. LC/MS showed the expected product, but was contaminated by baseline impurity.
  • 9-tert-butyl-7-acetamido-doxycycline
  • To a stirred solution of 9-tert-butyl-7-amino-doxycycline (100 mg) in 2 mL of 1,3-dimethyl-2-imidazolidinone were added 100 mg of sodium bicarbonate followed by 0.07 mL of acetyl chloride. The mixture was stirred at room temperature for 1.5 hours and then filtered. The filtrate was concentrated and poured into 100 mL of cold ether with stirring. The product that separated was filtered out. The crude product was redissolved in methanol, filtered and added dropwise to a cold 3:1 mixture of ether/PET ether with stirring. The product was collected by filtration and dried under vacuum, and 76 mg of product was obtained.
  • 4-dedimethylamino-doxycycline
  • To a stirred solution of tetracycline methiodide (860 mg) in 12 mL of acetic acid and 12 mL of water were added 432 mg of zinc dust. After stirring for 15 min the zinc powder was filtered out. The filtrate was diluted with 86 mL of water containing 0.86 mL of concentrated hydrochloric acid. The product that separated was filtered out and dried under vacuum, and 419 mg of product was obtained.
  • 4-dedimethylamino-9-nitro-doxycycline
  • To a stirred solution of 4-dedimethylamino-doxycycline (402 mg, 1 mmol) in 20 mL of concentrated sulfuric acid at 0° C. was added 111 mg of potassium nitrate. After stirring for 20 minutes, the reaction mixture was poured into 100 mL of cold water. n-Butanol was added to the water containing the product and the organic layer was separated. Extraction with n-butanol was repeated twice. The combined organic extracts were concentrated and added to cold water with stirring. The product that separated was filtered out; more product was recovered by re-extracting the aqueous filtrate with n-butanol, concentrating, pouring into cold water and filtering. After drying under vacuum, 353 mg of product were obtained.
  • 4-dedimethylamino-9-amino-doxycycline
  • To a stirred solution of 4-dedimethylamino-9-nitro-doxycycline (353 mg) in 17 mL of ethylene glycol monomethyl ether were added 0.1 mL of concentrated sulfuric acid and 200 mg of 10% Pd/C, and the resulting mixture was hydrogenated at atmospheric pressure for 10 hours. The catalyst was filtered through celite, washing with methanol. The filtrate was concentrated and poured into 100 mL of ether. The solid that separated was filtered out and dried under vacuum, and 292 mg of product was obtained.
  • 4-dedimethylamino-9-diazonium-doxycycline hydrochloride
  • To a stirred solution of 4-dedimethylamino-9-amino-doxycycline (220 mg) in 5 mL of 0.1N hydrochloric acid in methanol cooled in an ice bath was added 0.2 mL of n-butyl nitrite. The solution was stirred at 0° C. for 30 minutes, then poured into 150 mL of a 2:1 cold mixture of ether/PET ether with stirring. The solid that separated was filtered out and dried under vacuum, and 191 mg of product was obtained.
  • 4-dedimethylamino-9-azido-doxycycline
  • To a stirred solution of 4-dedimethylamino-9-diazonium-doxycycline (150 mg, 0.32 mmol) in 6.5 mL of 0.1N hydrochloric acid in methanol cooled in an ice bath were added 23 mg of sodium azide, and the resulting mixture was stirred for 1 hour at 0° C. The mixture was then poured into 120 mL of cold ether with stirring. Some solid formed, which was filtered out and found not to be the expected product. The filtrate was concentrated to dryness, taken up in methanol and poured into 50 mL of cold water with stirring. The solid that separated was filtered out and dried under vacuum; more product was obtained by extracting the filtrate with n-butanol, concentrating, pouring into water and filtering out the solid, and 84 mg of product was obtained.
  • 4-dedimethylamino-7-nitro-9-tert-butyl-doxycycline
  • A solution of 4-dedimethylamino-doxycycline (500 mg 1.25 mmol) in 3 mL of tert-butanol and 15 mL of methanesulfonic acid was stirred at room temperature for 15 hours, then 140 mg of potassium nitrate was added and the resulting mixture stirred for an additional hour. The reaction mixture was poured in 200 ml of cold water with stirring. The solid that separated was filtered out and air dried. The crude product was dissolved in the minimum amount of methanol and purified by HPLC. The combined fractions from HPLC were concentrated to dryness, dissolved in methanol and added dropwise to 50 mL of cold water. The solid that separated was filtered out and dried under vacuum, and 217 mg of product was obtained.
  • 4-dedimethylamino-7-amino-9-tert-butyl-doxycycline
  • To a stirred solution of 4-dedimethylamino-7-nitro-9-tert-butyl-doxycycline (217 mg, 0.42 mmol) in 12 mL of ethylene glycol monomethyl ether were added 170 mg of 10% Pd/C, and the resulting mixture was hydrogenated at atmospheric pressure for 13 hours. The catalyst was filtered through celite, washing with methanol. The filtrate was concentrated and poured into 20 of a 3:1 mixture of ether/PET ether, and 40 mg of product was obtained. The orange filtrate was concentrated to dryness to obtain additional 138 mg of product.
  • 4-dedimethylamino-7-diazonium-9-tert-butyl-doxycycline
  • To a stirred 4-dedimethylamino-7-amino-9-tert-butyl-doxycycline (165 mg) in 5 mL of 0.1N hydrocholirc acid in methanol cooled in an ice bath was added 0.15 mL of n-butyl nitrite. The solution was stirred at 0° C. for 30 minutes; half of the material was poured into 50 mL of cold ether with stirring. The solid that separated was filtered out and dried under vacuum, and 50 mg of product was obtained.
  • 4-dedimethylamino-7-amino-doxycycline
  • To a stirred solution of 4-dedimethylamino-doxycycline (400 mg) in 4 mL of THF and 4.5 mL of methanesulfonic acid at 0° C. were added 418 mg (1.4 mmol) of dibenzyl azodicarboxylate, and the resulting mixture was stirred for 4 hours while warming to room temperature. Water and n-butanol were added and two layers separated. The aqueous layer was extracted with a mixture of ether/n-butanol. The combined organic extracts were concentrated to dryness, and 10 mL of ethylene glycol monomethyl ether were added. Then, 430 mg of 10% Pd/C were added and the resulting mixture hydrogenated at atmospheric pressure for 12 hours. The catalyst was filtered off through celite, washing with methanol. The filtrate was concentrated and poured into a 3:1 cold mixture of ether/PET ether with stirring; the product that separated was filtered out and dried under vacuum, and 114 mg of product was obtained.
  • 9-nitro-minocycline sulfate
  • To a stirred solution of minocycline hydrochloride (1 g, 2.02 mmol) in 30 mL of concentrated sulfuric acid at 0° C. was added potassiumnitrate (246 mg). The resulting mixture was stirred for 45 minutes at 0° C., then poured into 1.2 L cold ether with stirring. The solid that separated was filtered out, washed with cold ether and dried under vacuum, and 1.33 g of product was obtained.
  • 9-amino-minocycline sulfate
  • To a stirred solution of 9-nitro-minocycline sulfate (500 g, 0.83 mmol) in 10 mL of ethylene glycol monomethyl ether and 7 mL of methanol were added 250 mg of 10% Pd/C. The reaction mixture was hydrogenated at atmospheric pressure for 4 hours. The catalyst was filtered off through celite, washing with methanol. The filtrate was concentrated, then added dropwise to 600 mL cold ether with stirring. The solid that separated was filtered out, washed with cold ether and dried under vacuum, and 465 mg of product was obtained.
  • 9-diazonium-minocycline sulfate hydrochloride
  • To a stirred solution of 9-amino-minocycline sulfate (100 mg) in 3 mL of 0.1N hydrochloric acid in methanol cooled in an ice bath was added 0.1 mL of tert-butyl nitrite (or n-butyl nitrite). The solution was stirred at 0° C. for 30 minutes, then poured into 100 mL of cold ether with stirring. The solid that separated was filtered out and dried under vacuum, and 87 mg of an orange-brown product was obtained.
  • 9-azido-minocycline sulfate
  • To a stirred solution of 9-diazonium-minocycline sulfate hydrochloride (485 mg, 0.83 mmol) in 18 mL of 0.1N hydrochloric acid in methanol were added 59 mg of sodium azide. The solution was stirred at room temperature for 2 hours, then filtered, and the filtrate was poured into 500 mL of cold ether with stirring. The solid that separated was filtered out and dried under vacuum, and 480 mg of beige product was obtained.
  • 9-acetamido-minocycline
  • To a stirred solution of 9-amino-minocycline sulfate (100 mg, 0.175 mmol) in 1.5 mL of 1,3-dimethyl-2-imidazolidinone were added 100 mg of sodium bicarbonate followed by 0.05 mL of acetyl chloride. The mixture was stirred at room temperature for 45 minutes and then poured into 150 mL of cold ether with stirring. The solid that separated was filtered out and dried under vacuum, and 165 mg of product was obtained.
  • 4-dedimethylamino-minocycline
  • Minocycline free base (175 mg, 0.38 mmol) was dissolved in 2 mL of THF, then 0.43 mL of methyl iodide was added. No crystallization of the product occurred. The reaction mixture was concentrated and poured into 200 mL of cold ether/PET ether 40:60 with stirring. The product that separated was filtered out and dried under vacuum; the product was resubjected to the reaction conditions for 3 additional days, then precipitated from ether as before, and 123 mg of minocycline methiodide was obtained. The crude product was dissolved in 2 mL of acetic acid, and 2 mL of water and 60 mg of zinc dust were added with stirring. After 15 minutes, the zinc powder was filtered out. The filtrate was diluted with 15 mL of water containing concentrated hydrochloric acid. The combined organic extracts were dried over magnesium sulfate and concentrated. The solid that separated was filtered out and dried under vacuum, and 150 mg of a yellow product was obtained.
  • 4-dedimethylamino-9-nitro-minocycline
  • 4-Dedimethylamino minocycline (415 mg, 1 mmol) was dissolved in 30 mL of concentrated sulfuric acid at room temperature. The solution was cooled on an ice bath and potassium nitrate (110 mg, 1.09 mmol) was added with stirring. The resulting mixture was stirred for 15 minutes, then poured into 300 mL cold ether with stirring. The solid that separated was filtered out, washed with cold ether and dried under vacuum. LC/MS showed incomplete conversion of starting material so the product was redissolved in 15 mL of concentrated sulfuric acid, 50 mg of potassium nitrate were added at 0° C. and the resulting mixture was stirred for 45 minutes. The product was isolated as before, and 542 mg of product was obtained.
  • 7,9-dibromo sancycline
  • To a stirred solution of sancycline (50 mg, 0.12 mmol) in 1.5 mL of concentrated sulfuric acid at 0° C. were added 42 mg of N-bromosuccinimide. After 30 minutes, the reaction mixture was poured into 60 mL of cold ether with stirring. The solid that separated was filtered out and dried under vacuum, and 70 mg of yellow product was obtained.
  • Mix of 7-nitro and 9-nitro-sancycline
  • To a stirred solution of sancycline (40 mg, 0.09 mmol) in 1.5 mL of concentrated sulfuric acid at 0° C. was added 10 mg of potassium nitrate. After 20 minutes, the reaction mixture was poured into 50 mL of cold ether with stirring. The solid that separated was filtered out and dried under vacuum, and 59 mg of product was obtained. LC/MS shows a 1.6:1 ratio of two mono-nitrated products.
  • In further testing of the present invention, the Minimum Inhibitory Concentrations (MIC) of five test products when challenged with ten different microorganism strains were evaluated. The study was an MIC evaluation for five test products, performed using a modification of the Macrodilution Broth Method outlined in NCCLS document M7-A5, Methods for Dilution Antimicrobial Susceptibility Tests for Bacteria that Grow Aerobically, Fifth Edition. Each test product was evaluated, in duplicate, against challenge suspensions of ten microorganism strains. Prior to using each product for testing, it was diluted (w/v) in sterile Mueller-Hinton Broth (MHB) to achieve an initial concentration, based on a potency of 1000 μg/mg, of 160 μg/mL.
    Product 1 - Tetracycline Derivative 14.8 mg of this product was dissolved
    Lot Number: INN01182 (MW 615) in 92.5 mL of MHB to achieve the
    initial concentration of 160 μg/mL
    Product 2 - Tetracycline Derivative 13.6 mg of this product was dissolved
    Lot Number: INN01183 (MW 627) in 85.0 mL of MHB to achieve the
    initial concentration of 160 μg/mL
    Product 3 - Tetracycline Derivative 11.9 mg of this product was dissolved
    Lot Number: INN01185 (MW 655) in 74.4 mL of MHB to achieve the
    initial concentration of 160 μg/mL
    Product 4 - Tetracycline Derivative 9.6 mg of this product was dissolved
    Lot Number: INN01189 (MW 625) in 60.0 mL of MHB to achieve the
    initial concentration of 160 μg/mL
    Product 5 - Tetracycline Derivative 13.8 mg of this product was dissolved
    Lot Number: INN01195 (MW 643) in 86.25 mL of MHB to achieve the
    initial concentrations of 160 μg/mL
  • Approximately 48 hours prior to testing, separate sterile tubes of Tryptic Soy Broth were inoculated from lyophilized vials containing each of the challenge microorganisms, as found in Table I, below. The broth cultures were incubated at 35°±2° C. for approximately 24 hours. The broth cultures prepared as described above were inoculated onto the surface of Tryptic Soy Agar contained in Petri plates, and incubated at 35°±2° C. for approximately 24 hours. This produced “lawns” of the microorganisms on the surface of the agar plates, which were used to prepare the challenge suspensions.
  • Prior to initiating testing, an initial challenge suspension containing approximately 1.0×109 CFU/mL was prepared for each microorganism by inoculating a test tube of Sodium Chloride Irrigation with microorganisms taken from the plates of solid media, prepared as described above. The final challenge suspensions containing approximately 1.0×106 CFU/mL were prepared for each microorganism species, by placing a 0.2 mL aliquot of the 1.0×109 CFU/mL suspension into a sterile 250 mL polypropylene bottle, containing 200 mL of Mueller-Hinton Broth. The final challenge suspensions were mixed thoroughly prior to use in testing. The final plated dilutions were 10−3, 10−4 and 10−5. These plates were incubated at 35°±2° C. until sufficient growth was observed (Table I).
  • Following incubation, the colonies on the plates were counted manually using a hand-tally counter. Counts in the 30 to 300 CFU range were used in the data calculations. The Initial Population (CFU/mL) was calculated for each challenge suspension as follows:
    CFU/ML=(C i×10−D)
    Where:
      • Ci=Average of the 2 Plates Counted
      • D=Dilution Factor of the Plate Counts Used
  • The population (CFU/mL) per tube of product/broth following inoculation was calculated for each challenge suspension as follows: Population per Tube ( CFU / mL ) = ( C i × 10 - D ) 2
    Where:
      • Ci=Average of the Two (2) Plates Counted
      • D=Dilution Factor of the Plate Counts Used
      • 2=Total Volume (mL) present in each product/broth tube following inoculation
  • The testing procedure was as follows: 30 mL aliquots of Mueller-Hinton Broth were dispensed into sterile bottles. A 30 mL aliquot of product (initial concentration of 160 μg/mL) was dispensed into the first of a series of 11 sterile bottles, each containing 30 mL of Mueller-Hinton Broth, and mixed thoroughly in order to achieve the 1:2 (v/v) dilution of product. A 30 mL aliquot was removed from the bottle prepared as described above, and used to initiate a 1:2 (v/v) dilution series through the remaining 10 bottles (1:4, 1:8, 1:16, 1:32, 1:64, 1:128, 1:256, 1:512, 1:1,024 and 1:2,048 product dilutions), each containing 30 mL of Mueller-Hinton Broth. Each bottle was mixed thoroughly prior to removing the 30 mL aliquot required for the next dilution in the series. 1.0 mL aliquots of each product dilution prepared and 1.0 mL aliquots of the initial product preparation (160 μg/mL product concentration) were transferred to separate sterile test tubes. A series of 12 tubes, each containing 1.0 mL of the appropriate product dilution (1:1, 1:2, 1:4, 1:8, 1:16, 1:32, 1:64, 1:128, 1:256, 1:512, 1:1,024, and 1:2,048) was prepared for each microorganism evaluated against each product (Table I). This resulted in product concentrations ranging from 160 μg/mL to 0.078 μg/mL.
  • A 1.0 mL aliquot of a suspension containing approximately 1.0×106 CFU/mL was introduced into each product dilution tube in the series, thereby resulting in a final product dilution series of 1:2, 1:4, 1:8, 1:16, 1:32, 1:64, 1:128, 1:256, 1:512, 1:1,024, 1:2,048, and 1:4,096, with each dilution tube containing approximately 5.0×105 CFU/mL of the challenge microorganism. This resulted in final product concentrations ranging from 80 μg/mL to 0.039 μg/mL. The test procedure described above was performed, in duplicate, for each microorganism species tested (Table I) against each test product. A positive control tube (growth control) containing a 1.0 mL aliquot of Mueller-Hinton Broth and a 1.0 mL aliquot of the challenge suspension was prepared for each microorganism (Table I). A negative (media) control tube (sterility control; no microbial inoculation) of Mueller-Hinton Broth was also prepared.
  • Product turbidity controls were prepared for each product by transferring 1.0 mL aliquots of each product dilution into separate sterile test tubes. A 1.0 mL aliquot of sterile Mueller-Hinton Broth was introduced into each product dilution tube created and mixed thoroughly using a vortex mixer, thereby resulting in a final product dilution series identical to that described above. The challenge suspension/product dilution tubes and the controls were incubated at 35°±2° C. for 20 hours, until good growth was apparent in the positive control tubes.
  • Following incubation, the tubes were examined for growth of the microorganisms, determined visually on the basis of turbidity.
  • The Minimum Inhibitory Concentration (MIC) for each test product versus each challenge microorganism was recorded as the highest dilution of test product that completely inhibited growth of the microorganism, as detected by the unaided eye. The results of the duplicate runs for each test product versus each microorganism were recorded and then averaged together to provide the final reported values.
    TABLE I
    Incubation Time* Incubation
    Microorganism Species ATCC # (MIC Tubes) Temp.* Media
    Enterococcus faecalis 29212 20 Hours 35° ± 2° C. TSB/TSA/MHB
    Eseherichia coli 25922 20 Hours 35° ± 2° C. TSB/TSA/MHB
    Escherichia coli (Tetracycline-Resistant) 51657 20 Hours 35° ± 2° C. TSB/TSA/MHB
    Escherichia coli (Tetracycline-Resistant) 51658 20 Hours 35° ± 2° C. TSB/TSA/MHB
    Escherichia coli (Tetracycline-Resistant) 51659 20 Hours 35° ± 2° C. TSB/TSA/MHB
    Staphylococcus aureus (Tetracycline- 27217 20 Hours 35° ± 2° C. TSB/TSA/MHB
    Resistant)
    Staphylococcus aureus (Tetracycline- 27659 20 Hours 35° ± 2° C. TSB/TSA/MHB
    Resistant)
    Staphylococcus aureus (Tetracycline- 27660 20 Hours 35° ± 2° C. TSB/TSA/MHB
    Resistant)
    Staphylococcus aureus 29213 20 Hours 35° ± 2° C. TSB/TSA/MHB

    *Initial Population plates were inadvertently incubated at room temperature (20°-27° C.) for approximately 48 hours prior to being incubated at 35° ± 2° C. for approximately 72 hours.
  • Table II presents the Minimum Inhibitory Concentrations, expressed as product dilution, for each test product versus each of the 10 microorganisms tested. Table III presents the Minimum Inhibitory Concentrations, expressed as product concentration μg/mL), for each test product versus each of the 10 microorganisms tested. The initial solution prepared for each product had a concentration, based on a potency of 1000 μg/mg, of 160 μg/mL.
    TABLE II
    Minimum Inhibitory Concentrations
    (Expressed as Product Dilution)
    Microorganism Species ATCC # Product #1 Product #2 Product #3 Product #4 Product #5
    Enterococcus faecalis 29212 <1:2 1:16 1:16 1:2 1:8
    Escherichia coli 25922  1:2 <1:2  <1:2  <1:2  1:2
    Escherichia coli 51657 <1:2 <1:2  <1:2  <1:2  <1:2 
    (Tetracycline-Resistant)
    Escherichia coli 51658 <1:2 <1:2  <1:2  <1:2  <1:2 
    (Tetracycline-Resistant)
    Escherichia coli 51659 <1:2 <1:2  <1:2  <1:2  <1:2 
    (Tetracycline-Resistant)
    Pseudomonas aeruginosa 27853 <1:2 <1:2  <1:2  <1:2  <1:2 
    Staphylococcus aureus 27217 <1:2 1:32 1:32 1:8  1:16
    (Tetracycline-Resistant)
    Staphylococcus aureus 27659 <1:2 1:32 1:24 1:8  1:16
    (Tetracycline-Resistant)
    Staphylococcus aureus 27660 <1:2 1:32 1:16 1:8  1:32
    (Tetracycline-Resistant)
    Staphylococcus aureus 29213  1:8 1:32 1:32 1:8  1:32
  • TABLE III
    Minimum Inhibitory Concentrations
    (Expressed as Product Dilution)
    Microorganism Species ATCC # Product #1 Product #2 Product #3 Product #4 Product #5
    Enterococcus faecalis 29212 >80 10 10 80 20
    Escherichia coli 25922 80 >80 >80 >80 80
    Escherichia coli 51657 >80 >80 >80 >80 >80
    (Tetracycline-Resistant)
    Escherichia coli 51658 >80 >80 >80 >80 >80
    (Tetracycline-Resistant)
    Escherichia coli 51659 >80 >80 >80 >80 >80
    (Tetracycline-Resistant)
    Pseudomonas aeruginosa 27853 >80 >80 >80 >80 >80
    Staphylococcus aureus 27217 >80 5 5 20 10
    (Tetracycline-Resistant)
    Staphylococcus aureus 27659 >80 5 6.67 20 10
    (Tetracycline-Resistant)
    Staphylococcus aureus 27660 >80 5 10 20 5
    (Tetracycline-Resistant)
    Staphylococcus aureus 29213 20 5 5 20 5
  • In additional studies, the activity of the tetracycline derivatives of the present invention as inhibitors of matrix degrading metalloproteinases (MMPs) that are involved in malignant tumor growth and metastasis were investigated. The studies used the Dunning MAT LyLu tumor model which was developed in Copenhagen rats from a transplantable tumor, transferred from an original prostate tumor from the dorsal prostate of an aged Copenhagen male rat. The MAT LyLu tumor is spontaneously metastatic to lymph nodes and lungs when injected, and produces bone metastases in approximately 80-100% of the recipient animals. Fresh dosing solutions containing 10 mg/ml of each test article in 2% carboxymethylcellulose were prepared daily and used in the testing.
  • The studies initially evaluated toxicity in two groups of five non-tumor bearing animals, each receiving the two tetracycline analogs (9-amino-doxycycline and 9-nitro-doxycycline) at a dose of 40 mg/kg daily for seven days.
  • Three groups of animals (10/group) were dosed by gavage with either vehicle, or one of the two analogs daily for three weeks, and then three times per week until death or sacrifice. Dosing was begun seven days prior to implantation of the MAT LyLu tumor cells by tail vein injection. Approximately 0.1 ml of tumor cell suspension was injected into one of the lateral tail veins. The dose to be chosen was based on two factors: (1) efficacy and (2) morbidity associated with the analog, if any. The following table (Table IV below), demonstrates survival time and weekly body weight of animals in control and treated groups. As shown in the table, 9-amino-doxycycline and 9-nitro-doxycycline demonstrated a marked increase in surviving number after tumor implantation, while also resulting in indistinguishable body weight changes as compared to the control group. This latter finding is particularly significant, in that conventional anti-cancer treatments often result in significant weight loss.
    TABLE IV
    Number of
    Days Post Tumor Surviving Number
    Implantation 9-amino-doxycycline 9-nitro-doxycycline Control
    14 10 8 9
    17 10 8 8
    24 10 8 7
    27 10 8 6
    28 9 8 6
    29 9 6 5
    30 7 4 5
    34 7 4 3
    42 7 4 3
  • In further studies, the inhibitory activity of MMPs, as well as percent inhibition of various cancers by various tetracycline derivative were evaluated. Results appear in Table V, below, and demonstrate the efficacy of the treatments of the present invention. One objective of the studies was to assess a series of tetracycline derivatives as inhibitors of purified human matrix metalloproteinases including MMP1, 2, 3, 7, 9 and 14. The tetracycline derivatives were effective inhibitors in the mM-μM range.
  • EXPERIMENTAL PROTOCOLS 1. Protocol for Measuring MMP1 (Collagenase-1) Inhibition by Tetracycline Derivatives
  • MMP1 activity was determined by the release of soluble 14C labelled collagen fragments from 14C acetylated rat skin type I collagen (Methods in Enzymology 80 711, 1981).
  • Assay constituents:
      • 100 μg 14C labelled rat skin type I collagen (5-6000 dpm)
      • 50 mM Tris HCl pH 7.9
      • 15 mM CaCl2 0.02% azide
      • Human recombinant MMP1 3-4 nM
      • ±Tetracycline (up to 1 mM) in a total volume of 300 μl.
  • Incubations were at 35° C. for 5 hours. Uncleaved collagen fibrils (which form within the first 10 minutes) were removed by centrifugation at 10,000 g, 4° C., 10 minutes. 200 μl supernatant were counted in Packard Opti Phase Supermix scintillation fluid using a scintillation counter. Only data derived from within the linear part of the assay (10-70% lysis of the collagen) were utilized. Based on percentage inhibition of the activity of MMP1 alone by a range of concentrations of each sample, the IC50 value for each tetracycline was calculated by linear regression analysis.
  • 2. Protocol for Measuring MMP2 (Gelatinase A) Inhibition
  • MMP2 activity was determined by the release of trichloracetic acid, TCA, soluble fragments from 14C acetylated rat skin type I gelatin (Methods in Enzymology 248, 470, 1995. Biochem. J. 195, 1981)
  • Assay constituents:
      • 100 μg 14C labelled gelatin (type I collagen denatured 60° C. 20 min)
      • 50 mM Tris HCl pH 7.9
      • 15 mM CaCl2, 0.02% azide
      • Human recombinant MMP1, 0.1-0.5 nM
      • ±Tetracycline (up to 1 MM) in a total volume of 250 μl.
  • Incubations were set at 37° C. for 16 hours. The reaction was stopped by cooling the incubations and addition of 50 μl cold 90% (w/v) trichloracetic acid with careful mixing. TCA precipitation was allowed to proceed for 15 minutes at 4° C. prior to cetrifugation at 10,000 g for 10 minutes at 4° C. 200 μl of the TCA supernatant was taken for the determination of radioactive content by scintillation counting (Packard Opti Phase Supermix scintillation fluid). Only data derived from within the linear part of the assay (10-70% lysis) were utilized. The percentage inhibition of MMP2 by each tetracycline concentration was plotted to derive the IC50 value, using linear regression analysis.
  • 3. Protocol for Measuring MMP3 (Stromelysin 1) Inhibition
  • MMP3 activity was determined by the release of trichloracetic acid, TCA, soluble fragments from 14C acetylated 13 casein (Sigma) as described in Methods in Enzymology 248, 451 (1995).
  • Assay constituents:
      • 100 μg 14C casein (7000 dpm)
      • 50 mM Tris HCl, pH 7.9
      • 15 mM CaCl2, 0.02% azide
      • Human recombinant MMP3, 0.25-1.0 nM
      • ±tetracycline (up to 1 mM) in a total volume of 250 μl.
  • Incubations were at 37° C. for 16 hours. The reaction was stopped by the addition of 500 μl cold 18% TCA and standing on ice for 15 minutes. TCA precipitates were removed by centrifugation at 10,000 g for 10 minutes at 4° C. and 200 μl supernatant was taken for scintillation counting. Only data derived from within the linear part of the assay (10-60% lysis) were utilized. The percentage inhibition of MMP3 by each concentration of the candidate tetracycline was plotted to derive the IC50 value, using linear regression analysis.
  • 4. Protocol for Measuring MMP7 (Matrilysin) Inhibition
  • MMP7 activity was determined by the release of trichloracetic acid, TCA, soluble fragments from 14C acetylated β casein (Sigma) as described in Methods in Enzymology 248, 451 (1995).
  • Assay constituents:
      • 100 μg 14C casein (7000 dpm)
      • 50 mM Tris HCl, pH 7.9
      • 15 mM CaCl2, 0.02% azide
      • Human recombinant MMP7, 1.5 nM
      • ±tetracycline (up to 1 mM) in a total volume of 250 μl.
  • Incubations were at 37° C. for 16 hours. The incubation was stopped by the addition of 500 μl cold 18% TCA and standing on ice for 15 minutes. TCA precipitates were removed by centrifugation at 10,000 g for 10 minutes at 4° C., and 200 μl supernatant was taken for scintillation counting. Only data derived from within the linear part of the assay (10-60% lysis) were utilized. The percentage inhibition of MMP7 by each concentration of the candidate tetracycline were plotted to derive the IC50 value.
  • 5. Protocol for Measuring MMP9 (Gelatinase B) Inhibition
  • MMP9 activity was determined by the release of trichloracetic acid, TCA soluble fragments from 14C acetylated rat skin type I (Methods in Enzymology 248, 470, 1995. Biochem. J. 195, 1981)
  • Assay constituents:
      • 100 μg 14C labelled gelatin (collagen denatured 60° C. 20 min)
      • 50 mM Tris HCl pH 7.9
      • 15 mM CaCl2, 0.02% azide
      • Human recombinant MMP9, 1.2-2 nM
      • ±Tetracycline (up to 1 mM) in a total volume of 250 μl.
  • Incubations were at 37° C. for 16 hours. The incubations were stopped by cooling the incubations and addition of 50 μl cold 90% (w/v) trichloracetic acid with careful mixing. TCA precipitation was allowed to proceed for 15 minutes at 4° C. prior to centrifugation at 10,000 g for 10 minutes at 4° C. 200 μl of the TCA supernatant was taken for the determination of radioactive content by scintillation counting (Packard Opti Phase Supermix scintillation fluid). Only data derived from within the linear part of the assay (10-70% lysis) were utilized. The percentage inhibition of MMP9 by each tetracycline concentration were plotted to derive the IC50 value.
  • 6. Protocol for Measuring MMP14 (MTI-MMP) Inhibition by Tetracycline Derivatives
  • MMP14 activity was determined by the release of soluble 14C labelled collagen fragments from 14C acetylated rat skin type I collagen (Methods in Enzymology 80 711, 1981).
  • Assay constituents:
      • 100 μg 14C labelled collagen (5-6000 dpm)
      • 50 mM Tris HCL pH 7.9
      • 15 mM CaCl2 0.02% azide
      • Human recombinant MMP14 50-100 nM
      • ±Tetracycline (up to 1 mM) in a total volume of 300 μl.
  • Incubations were at 35° C. for 15 hours. Uncleaved collagen fibrils (which form within the first 10 minutes) were removed by centrifugation at 10,000 g 4° C. 10 minutes 200 μl supernatant were counted in Packard Opti Phase Supermix scintillation fluid using a scintillation counter. Only data derived from within the linear part of the assay (10-70% lysis of the collagen) were utilized. Based on percentage inhibition of the activity of MMP1 alone by a range of concentrations of each sample, the IC50 value for each tetracycline was calculated.
  • Another objective was to analyze the effects of specific, tetracycline derived compounds on the in-vitro chemo-invasive potential of:
      • C8161 human melanoma cells
      • MDA-MB-231 human breast cancer cells
      • PC3 human prostate cancer cells
      • RPM18226 human myeloma cancer cells
      • Ark human myeloma cancer cells
      • Arp-1 human myeloma cancer cells
  • The Membrane Invasion Culture System (MICS) in vitro chemo-invasion assay was chosen to measure changes in the invasive potential of specific human cancer cells in response to tetracycline derived compounds. Stock solutions of each compound were hydrated in water containing 2% DMSO and pH 10.0. Upon solubilization, HCl was added to the solution to establish a pH of 7.5-8.0. This solution was then wrapped in foil and kept at 4° C. during the 24 hours of assay. Fresh compound was prepared for each assay. The in vitro chemoinvasion analyses for tumor cell invasiveness are performed using the Membrane Invasion Culture System (MICS) as previously described.
  • The MICS system is a thermally treated plastic manifold system consisting of two matched sets of plates containing fourteen wells, 13 mm in diameter. Interposed between these plates was a polycarbonate filter containing 10 μm pores and coated with a defined matrix composed of human laminin/collagen IV/gelatin that formed a 35 μm thick barrier between the top and bottom sections of the wells in MICS. Prior to placing this barrier in place, the lower wells were filled with serum-free RPMI culture medium made 50% in conditioned medium obtained from 2-day-old cultures of human fibroblasts which had been cleared of cells and cellular debris by either centrifugation, or filtering through a 0.45 μm sterile filter. The barrier was then placed on the lower wells and fresh serum-free medium added to the upper wells after assembly of the system. Fifty thousand tumor cells were then added to the wells either in the presence of the compound, or the DMSO vehicle (the controls). Of the 12 assay wells on each manifold, 3 randomized wells acted as controls, 3 wells received 1 μg/ml of compound, 3 received 10 μg/ml and 3 received 25 μg/ml. After 24 hours, cells and media were removed from the lower wells and replaced by phosphate buffered saline plus 2 mM EDTA. This solution was used to remove all cells from the lower well, and this wash was added to the recovered cells plus medium from the same well. These samples were then collected onto a polylysine containing polycarbonate membrane containing 3 μm pores using a Dot-Blot manifold system. These collection filters were then fixed in methanol, and stained using Wright's stain (Leukostat staining kit) in situ. The filters were then mounted on microscope slides with microscope immersion oil (which reduced the light refraction from the pores) and 5 microscopic fields were counted to calculate the number of cells which invaded through the membrane over the 24 hour period. These numbers were then statistically analyzed as discussed below.
    TABLE V
    INHIBITORY ACTIVITY OF MATRIX
    METALLOPROTEINASES (MMPs)1
    COMPOUND NO. MMP-1 MMP-2 MMP-3 MMP-7 MMP-9 MMP-14
    1 267 >700 258 815 43 86
    2 479 80 348 >1000 117 119
    3 >1000 >1000 304 >1000 136 216
    4 409 421 219 278 36 61
    5 647 86 237 441 194 447
    6 394 135 241 >1000 83 56
    7 R(5) >1000 300 335 64 >1000
    8 >1000 599 232 388 >1000 >1000
    9 372 337 154 507 28 29
    10 499 484  96 >1000 41 53
    11 427 >1000 270 >1000 96 46
    12 >1000 >1000 3902 503 270 >1000
    13 126 48 206 >1000 64 14
    14 279 R >1000  R R 386
    15 >1000 234 738 716 402 768
    16 452 215 >1000  569 215 715
    17 122 >1000 350 110 98
    18 123 233 335 131 102
    19 270 925 394 98 90
    20 429 511 281 162 266
    21 293 >1000 >1000  79 115
    22 396 254 462 341 459
    23 >1000 306 >1000  >1000 >1000
    24 405 724 191 127 160
    25 974 397 303 148 185
    26 >1000 413 >1000  384 586
    27 498 963 149 101 92
    28 >1000 >1000 231 125 49
    29 895 983 213 172 98
    30 576 >1000 938 >1000 >1000
    31 621 258 >1000  311 593
    32 601 236 244 214 197
    33 295 263 205 142 94
    34 242 242 136 308 197
    35 947 249 218 397 108
    36 539 >1000 283 >1000 608
    37 279 >1000 274 445 41
    38 230 175  95 215 69
    39 860 186 682 114 83
    40 560 362  91 R 29
    41 478 889  54 250 403
    42 R 606 1557  405 428
    43 600 2482 174 319
    44 1438 1468 224 591
    45 384 2446 194 6626
    46 194 3803  42 59 90
    47 318 117  68 88 60
    48 12831 1929 506 424 354
    49 372 1572 141 489 87
    50 211 2533  48 34 40
    51
    52 >1000 250 >1000  >1000 32 68
    53
    HUMAN CELL LINE EVALUATED AND PERCENT INHIBITION
    Multiple
    Breast Melanoma Myeloma Prostrate
    COMPOUND NO. MDA-MB-231 C8161 RPMI-8226 PC-3 ML
    1  7B  20B  5B  5  20  35B  20  10  15  2  47B  60B
     25B  25B  32B  40B  49B  56B  21B  21B  29B  13B  24B  24B
    2  20  15  30B  20B  25B  27B  +8 +10B +11  20  8  15
     1B  +4B  7B
    3  +8  +2  12  30B  22B  15B  +7 +23B  27B  3  +1  25B
    +22  +3 +24B
    4  5  6  2  27B  24B  17  +4  +5 +14  10  +8  3
    5  6  +4  46B  15  27B  35B  6  4  36  44B  35  46B
     17  37B  42B
    6  +5 +17B  27B  18B  33B  32B  +6 +25B  +5  21  26  41B
     24  44B  42B  8  21B  33B  12B  10  5  13  32B  30B
    7 +18 +41B +27  7  14  16 +14B  12  +2  0  0  3
    8 +10  +5 +13  13B  41B  40B  11  3  +2  0  +4  0
    9
    10  9  27B  37B  21  22  11  3  5  29B  40B  50B  65B
     +5  0  8  7  28B  30B  9  16  5
    11  6  15  36B  16  46B  41B  +3  0  36B
    12 +11  +13  +12
    13  4  0  +9  6  12  14 +11 +13 +12  8  23B  13
    14
    15
    16
    17
    18
    19
    20
    21
    22
    23
    24
    25
    26
    27  15  35  72B  0  +1  73  24B  6  42B  17B  35B  70
     5  17  48B  11  +38B  +51B  20  9  11  9  56B  70
    28 +24 +36B  54B +20  34B  40B  4 +10  34B  7 +11B  80
     +7  +15  45B +25B  +53B  19  5  +3  62B  35B  28B  61
    29 +41B  7  59B  12  +20  61B  13  12  8  28B  50B  58
    +13 +25B  17 +10  +14B   1  29B  13  35B  8  8  69
    30
    31
    32
    33
    34
    35
    36
    37  +6  1  46B  +1  20  69B  10  5  46B  11  26  36
    38  +7  37B  70B  3   1  66B  33B  13 +11  37B  41B  41
    +14 +14  37 +28  +26  39  4 +49B  +8  38B  27B  52
    39  18B  29B  76B  10  11  24  5  10  46B  44B  67B  69B
    40  9  15  20B  +1  +5  +4  16  11  9  +1  +7  +4
    41  +8  3  2  2   8   6  7  +6  14  +4  4  4
    42
    43
    44
    45
    46  20B  +5  +1  21   7  10  7 +11  12  20  16  27B
    47 +17 +16B  0 +30  +4  +2  28  18  60B  3  3  5
    48
    49  6  +5  +1  27B   7  30B  11  +2  15  4  +5  6
    50  7  0  2  5  +2   1 +14  +8 +13 +14 +17 +10
    51 +15B +18  25B  +9  +31B +122B  14 +26B  +2  +4 +13  4
    52  5  12  52B  5  15  30  2  26B  68B  52B  68B  70B
     19B  33B  46B  31B  48B  63B
    53  9  49B 83B +40B +158B  83B +95B  18  84B  29B  21  45B
    INHIBITORY ACTIVITY OF MATRIX
    METALLOPROTEINASES (mmPs)1
    COMPOUND NO. MMP-1 MMP-2 MMP-3 MMP-7 MMP-9 MMP-14
    1 267 >700 258 815 43 86
    2 479 80 348 >1000 117 119
    3 >1000 >1000 304 >1000 136 216
    5 647 86 237 441 194 447
    6 394 135 241 <1000 83 56
    9 372 337 154 507 28 98
    10 499 484 96 >1000 41 53
    13 126 48 206 >1000 64 14
    CELL LINE EVALUATED AND PERCENT INHIBITIONA
    COMPOUND NO. RPMI-8226 ARP-1 ARK
    1  20  10  15  +9 +13  1  9 +9  +8
     21B  21B  29B  13  8  17B
    2  +8 +10B +11  10  7  6  20B  9B  10
     1B  +4B  7B
    3  +7 +23B +27B +13  +1 +16 +11 +6 +15
    +22  +3 +24B
    5  6  4  36 +25B  1  4B  +7 22B  21B
     17  37B  42B
    6  +6 +25B  +5 NE  12  0  8
     12B  10  5
    9  21B  24B  28B +14 +21B  7  17B 24B  20B
    10  3  5  29B NE  6 +7  1
    13 +11 +13 +12 NE  2 +7  1
    Key to Table V:
    Compound Substance
     1: 9-nitro-6-deoxy-5-hydroxytetracycline H2SO4
     2: 9-amino-6-deoxy-5-hydroxytetracycline H2SO4
     3: 9-Isopropylamino-6-deoxy-5-hydroxytetracycline H2SO4
     4: 7-Dimethylamino-6-demethyl-6-deoxy-9-nitrotetracycline H2SO4
     5: Same as 2
     6: 9-Azido-6-deoxy-5-hydroxytetracycline H2SO4
     7: 9-Amino-7-dimethylamino-6-demethyl-6-deoxytetracycline H2SO4
     8: 9-Acetamido-7-dimethylamino-6-demethyl-6-deoxytetracycline H2SO4
     9: 7-dimethylamino-6-demethyl-6-deoxytetracycline-9-diazonium H2SO4 HCL
    10: 9-Azido-7-demethylamino-6-demethyl-6-deoxytetracycline H2SO4
    11: 6-Deoxy-5-hydroxytetracycline-9-diazonium H2SO4
    12: 7-Amino-6-deoxy-5-hydroxytetracycline H2SO4
    13: 7,9-Dibromo-6-demethyl-6-deoxytetracycline H2SO4
    14: 7-Amino-6-deoxy-5-hydroxy-9-nitrotetracycline H2SO4
    15: 7-Dimethylamino-6-deoxy-5-hydroxy-tetracycline H2SO4
    16: 9-Acetamido-6-deoxy-5-hydroxytetracycline H2SO4
    17: 7-Di-n-Butylamino-6-deoxy-5-hydroxytetracycline H2SO4
    18: 7-Di-n-Hexylamino-6-deoxy-5-hydroxytetracycline H2SO4
    19: 7-Di-(3,3-dimethylbutyl)amino-6-deoxy-5-hydroxytetracycline H2SO4
    20: 7-Azido-6-deoxy-5-hydroxy-tetracycline H2SO4
    21: 6-Deoxy-5-hydroxytetracycline-7-diazonium H2SO4
    22: 7-Acetamido-9-nitro-6-deoxy-5-hydroxytetracycline H2SO4
    23: 7-Acetamido-6-deoxy-5-hydroxytetracycline H2SO4
    24: 7-Di-n-propylamino-6-deoxy-5-hydroxytetracycline H2SO4
    25: 7-Isobutylamino-6-deoxy-5-hydroxytetracycline H2SO4
    26: 7-Acetamido-9-amino-6-deoxy-5-hydroxytetracycline H2SO4
    27: 7-Isobutylmethylamino-6-deoxy-5-hydroxytetracycline H2SO4
    28: 6-Deoxy-5-acetoxy-tetracycline H2SO4
    29: 7-Acetylisobutylamino-6-deoxy-5-hydroxytetracycline H2SO4
    30: 7-Acetamido-6-deoxy-5-hydroxytetracycline-9-diazonium H2SO4
    31: 7-Dimethylamino-6-deoxy-5-hydroxytetracycline-9-diazonium H2SO4
    32: 7-Cyclobutylamino-6-deoxy-5-hydroxytetracycline H2SO4
    33: 7-Cyclobutylmethylamino-6-deoxy-5-hydroxytetracycline H2SO4
    34: 4-Dedimethylamino-6-deoxy-5-hydroxytetracycline
    35: 4-Dedimethylamino-6-deoxy-5-hydroxy-9-nitrotetracycline
    36: 9-animo-4-dedimethylamino-6-deoxy-5-hydroxytetracycline sulfate
    37: 4-Dedimethylamino-6-deoxy-5-hydroxytetracycline-9-diazonium sulfate
    38: 7-nitro-9-tertbutyl-6-deoxy-5-hydroxytetracycline H2SO4
    39: 9-tertbutyl-6-deoxy-5-hydroxytetracycline H2SO4
    40: 7-Nitro-9-tertbutyl-4-dedimethylamino-6-deoxy-5-hydroxytetracycline H2SO4
    41: 4-Dedimethylamino-7-dimethylamino-6-demethyl-6-deoxytetracycline H2SO4
    42: 7-Acetylamino-9-azido-6-deoxy-5-hydroxy-tetracycline
    43: 9-tertbutyl-6-deoxy-5-hydroxytetracycline-7-diazonium H2SO4
    44: 7-Amino-9-tertbutyl-6-deoxy-5-hydroxytetracycline H2SO4
    45: 7-Azido-9-tertbutyl-6-deoxy-5-hydroxytetracycline H2SO4
    46: 6-deoxy-5-hydroxytetracycline-4-Methiodide
    47: 7-(1,2-benzylcarboxyhydrazine)-6-deoxy-5-hydroxy-tetracycline HCl
    48: 4-Dedimethylamino-7-amino-6-deoxy-5-hydroxyltetracycline H2SO4
    49: 7-Amino-9-tertbutyl-4-dedimethyl-6-deoxy-5-hydroxytetracycline- H2SO4
    50: 4-Dedimethylamino-9-tertbutyl-6-deoxy-5-hydroxytetracycline-7-diazonium
    H2SO4
    51: 9-(4-fluorophenyl)-6-deoxy-5-hydroxytetracycline
    52: 4-Epi-7-Chlorotetracycline hydrochloride
    53: 6-Demethyl-6-deoxytetracycline HCl

    For Table V:

    Aconcentations of 1, 10 and 25 ug/mL were used

    Bsignificant, i.e., P < 0.05

    1IC50 in μM unless specified otherwise

    2Predicted

    += increased invasiveness

    NE = not evaluated

    R = to be repeated (number times repeated)
  • While the present invention has been described with respect to particular embodiment thereof, it is apparent that numerous other forms and modifications of the invention will be obvious to those skilled in the art. The appended claims and this invention generally should be construed to cover all such obvious forms and modifications, which are within the true spirit and scope of the present invention.

Claims (6)

1. A treatment for inhibiting microbial or tumor growth which comprises adding to a subject in need of said treatment an effective amount of a tetracycline composition or salt thereof, comprising a general formula:
Figure US20050245491A9-20051103-C00013
wherein R is selected from CH2N(RaRb) or CH2N
Figure US20050245491A9-20051103-C00014
RaRb are C1 to C6 alkyl;
Rc and Rd are (CH2), CHRe, n is 0 or 1 and Re is H, C1 to C6 alkyl, or NH2, and X is NH, S, or CH2;
R1 is H or OH;
R2 is H, OH, ═O, or OCOR8, where R8 is C1 to C6 alkyl, or alternatively, R2 is N(R9)2, where R9 is hydrogen or C1 to C6 lower alkyl, or R9 CO, with the proviso that when R2 is keto or N(R9)2, then R3 and R4 are H or CH3, and R3 and R4 are not both CH3 or H;
5a hydrogen is α or β;
R3 and R4 are H, CH3 or F, with the proviso that when R3 is CH3, then R4 is H or F, and when R4 is CH3, then R3 is H or F and R4 and R5 are not both F;
R5 and R7 are halogen, H, NO2, N3, N2 +, C2H5OC(S)S—, CN, NR10R11, where R10 and R11 are H, C1 to C10 alkyl, and R12(CH2)nCO—, where n is 0 to 5 and R9 is H, NH2, mono or disubstituted amino selected from straight, branched or cyclized C1 to C10 alkyl groups, with the proviso that
R10 and R11 are not both R12(CH2)nCO—;
R7, alternatively, is tertiary butyl; and
R6 is halogen, acetylene, R13—C6H5, where R13 is NO2, halogen, acetylamino, amino, phenyl, alkyl, or alkoxy.
2. A treatment for inhibiting microbial or tumor growth which comprises adding to a subject in need of said treatment an effective amount of a tetracycline composition or salt thereof, comprising a general formula:
Figure US20050245491A9-20051103-C00015
wherein R is selected from CH2N(RaRb) or CH2N
Figure US20050245491A9-20051103-C00016
where RaRb are C1 to C6 alkyl;
Rc and Rd are (CH2)n CHRe, n is 0 or 1 and Re is H, C1 to C6 alkyl, or NH2, and X is NH, S, or CH2;
R1 is H or OH;
R2 is H or OH;
R3 is H or CH3;
R4 and R6 are halogen, H, NO2, N3, N2 +, C2H5OC(S)S—, CN, NR7R8, where R7 and R8 H, C1 to C10 alkyl, and R9(CH2)nCO—, where n is 0 to 5 and R9 is H, NH2, mono or disubstituted amino selected from straight, branched or cyclic C1-C10 alkyl, with the proviso that R7 and R8 are not both R9(CH2)nCO—; and
R6 is H or halogen, acetylene, R10—C6H5, where R10 is NO2, halogen, acetylamino, amino, phenyl, alkyl, or alkoxy.
3. A treatment for inhibiting microbial or tumor growth which comprises adding to a subject in need of said treatment an effective amount of a tetracycline composition or salt thereof, comprising a general formula:
Figure US20050245491A9-20051103-C00017
wherein R is selected from CH2N(RaRb) or CH2N
Figure US20050245491A9-20051103-C00018
where RaRb are C1-C6 alkyl;
Rc and Rd are (CH2), CHRe, n is 0 or 1 and Re is H, C1 to C6 alkyl, or NH2, and X is NH, S, or CH2;
R1 and R2 are H or N(R9)2, where R9 is H or C1 to C6 alkyl with the proviso that R1 and R2 are not both N(R9)2, R1 and R2 are also N(R10)3 I, where R10 is C1 to C6 alkyl, with the proviso that R1 and R1 are not both N(R10)3 I;
R3 is H;
R4 and R5 are H, CH3 or F, with the proviso that when R4 is CH3, then R5 is H or F, and when R5 is CH3, then R4 is H or F, and R4 and R5 are not both F;
R6 and R8 are halogen, H, NO2, N3, N2 +, C2H5OC(S)S—, CN, NR11R12, where R11 and R12 are H, C1 to C10 alkyl, and R13(CH2)nCO—, where n is 0 to 5 and R13 is H, NH2, mono or disubstituted amino selected from straight, branched or cyclized C1-C10 alkyl groups, with the proviso that R10 and R11 are not both R12 (CH2)nCO—; and
R8 can also be tertiary butyl and R7 is H or halogen.
4. A treatment for inhibiting microbial or tumor growth which comprises adding to a subject in need of said treatment an effective amount of a tetracycline composition or salt thereof, comprising a general formula:
Figure US20050245491A9-20051103-C00019
wherein R is selected from CH2N(RaRb) or CH2N
Figure US20050245491A9-20051103-C00020
Ra Rb are C1-C6;
Rc and Rd are (CH2)n CHRe, n is 0 or 1, Re is H, alkyl (C1 to C6), NH2, and X is NH, S, or CH2;
R1 and R2 are H or N(R9)2 where R9 is hydrogen or lower alkyl (C1 to C6 ), with the proviso that R1 and R2 can not both be N(R9)2.
R3 is H, OH, ═O, OCOR10, where R10 is C1 to C6, or alternatively, R3 is N(R9)2 where R9 is hydrogen or C1 to C6, with the proviso that when R23 is ═O or N(R9)2, then R4 and R5 are H or CH3 and R4 and R5 are not both CH3 or H;
5a hydrogen is α or β;
R4 and R5 are H, CH3 or F, with the proviso that when R4 is CH3, then R5 is H or F, and when R5 is CH3 then R4 is H or F and R4 and R5 are not both F;
R6 and R8 are halogen, H, NO2, N3, N2 +, C2H5OC(S)S—, CN, NR10R11, where R10 and R11 are H, alkyl (C1 to C10), and R12 (CH2)nCO—, where n is 0 to 5 and R12 is H, NH2, mono or disubstituted amino selected from straight, branched or cyclized C1 to C10 groups, with the proviso that R10 and R11 are both R12 (CH2)nCO—, or, alternatively, R8 is tertiary butyl;
R7 is H or halogen, acetylene, R12—C6H5, where R12 is NO2, halogen, acetylamino, amino, phenyl, alkyl, or alkoxy.
5. A treatment for inhibiting microbial or tumor growth which comprises adding to a subject in need of said treatment an effective amount of a tetracycline composition or salt thereof, comprising a general formula:
Figure US20050245491A9-20051103-C00021
wherein R1 is H or N(CH3)2; R2 is H, CH3 or OCOCH3; R3 is H or CH3; R4 is H; R5 is H, N(CH3)2, NH2, N(C4H9)2, N(C6H13)2, N(3,3-dimethylbutyl)2, N3, N2 +, NO2, NHCOCH3, NH(m-propyl)2, NH-isobutyl, NH-isobutylmethyl, NH(cyclobutyl), NH(cyclobutyl methyl); and R6 is H, NO2, NH2, (CH3)2 CHNH, N3, NHCOCH3, N2 +, N3 or (CH3)3C.
6. A treatment for inhibiting microbial or tumor growth which comprises adding to a subject in need of said treatment an effective amount of a tetracycline composition or salt thereof, comprising a general formula:
Figure US20050245491A9-20051103-C00022
wherein R1 is H or N(CH3)2; R2 is H, OH or OCOCH3; R3 is H or CH3; R4 is H; R5 is H, N(CH3)2; N(C4H9)2, N(C6H13)2, N(3,3-dimethylbutyl)2; N3, N2 +, NO2, NHCOCH3, NH(n-propyl)2, NH-isobutyl, NH-isobutylmethyl, N(CH3CO)(isobutyl), NH(cyclobutyl), NH(cyclobutyl methyl), or NH2; and R6 is H, NO2, NH2, (CH3)2 CHNH, N3, NHCOCH3, N2 +, or (CH3)3C.
US10/264,454 2001-10-05 2002-10-04 Tetracycline derivatives and methods of use thereof Expired - Fee Related US7214669B2 (en)

Priority Applications (4)

Application Number Priority Date Filing Date Title
US10/264,454 US7214669B2 (en) 2001-10-05 2002-10-04 Tetracycline derivatives and methods of use thereof
US11/076,276 US20050267079A1 (en) 2001-10-05 2005-03-09 Tetracycline derivatives and methods of use thereof
US11/673,589 US20070142338A1 (en) 2001-10-05 2007-02-11 Tetracycline Derivatives and Methods of Use Thereof
US11/679,744 US20070149488A1 (en) 2001-10-05 2007-02-27 Tetracycline Derivatives and Methods of Use Thereof

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
US32750201P 2001-10-05 2001-10-05
US10/264,454 US7214669B2 (en) 2001-10-05 2002-10-04 Tetracycline derivatives and methods of use thereof

Related Child Applications (3)

Application Number Title Priority Date Filing Date
US11/076,276 Division US20050267079A1 (en) 2001-10-05 2005-03-09 Tetracycline derivatives and methods of use thereof
US11/673,589 Division US20070142338A1 (en) 2001-10-05 2007-02-11 Tetracycline Derivatives and Methods of Use Thereof
US11/679,744 Division US20070149488A1 (en) 2001-10-05 2007-02-27 Tetracycline Derivatives and Methods of Use Thereof

Publications (3)

Publication Number Publication Date
US20040067912A1 US20040067912A1 (en) 2004-04-08
US20050245491A9 true US20050245491A9 (en) 2005-11-03
US7214669B2 US7214669B2 (en) 2007-05-08

Family

ID=23276795

Family Applications (4)

Application Number Title Priority Date Filing Date
US10/264,454 Expired - Fee Related US7214669B2 (en) 2001-10-05 2002-10-04 Tetracycline derivatives and methods of use thereof
US11/076,276 Abandoned US20050267079A1 (en) 2001-10-05 2005-03-09 Tetracycline derivatives and methods of use thereof
US11/673,589 Abandoned US20070142338A1 (en) 2001-10-05 2007-02-11 Tetracycline Derivatives and Methods of Use Thereof
US11/679,744 Abandoned US20070149488A1 (en) 2001-10-05 2007-02-27 Tetracycline Derivatives and Methods of Use Thereof

Family Applications After (3)

Application Number Title Priority Date Filing Date
US11/076,276 Abandoned US20050267079A1 (en) 2001-10-05 2005-03-09 Tetracycline derivatives and methods of use thereof
US11/673,589 Abandoned US20070142338A1 (en) 2001-10-05 2007-02-11 Tetracycline Derivatives and Methods of Use Thereof
US11/679,744 Abandoned US20070149488A1 (en) 2001-10-05 2007-02-27 Tetracycline Derivatives and Methods of Use Thereof

Country Status (10)

Country Link
US (4) US7214669B2 (en)
EP (1) EP1439843A4 (en)
JP (1) JP2005505588A (en)
CN (2) CN1961885A (en)
AU (1) AU2002341970B2 (en)
CA (1) CA2462572C (en)
MY (1) MY133403A (en)
NO (1) NO20041272L (en)
RU (1) RU2300380C2 (en)
WO (1) WO2003030819A2 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US20060084634A1 (en) * 2001-03-13 2006-04-20 Paratek Pharmaceuticals, Inc. 7-Pyrollyl tetracycline compounds and methods of use thereof
US7696188B2 (en) 2000-07-07 2010-04-13 Trustees Of Tufts College 7,8 and 9-substituted tetracycline compounds
US7851460B2 (en) 2000-06-16 2010-12-14 Trustees Of Tufts College 7-phenyl-substituted tetracycline compounds
US7858601B2 (en) 2004-10-25 2010-12-28 Paratek Pharmaceuticals, Inc. 4-substituted tetracyclines and methods of use thereof
US8088755B2 (en) 2005-02-04 2012-01-03 Paratek Pharmaceuticals, Inc. 11a, 12-derivatives of tetracycline compounds
US9533943B2 (en) 2003-07-09 2017-01-03 Paratek Pharmaceuticals, Inc. Substituted tetracycline compounds

Families Citing this family (25)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2301914A1 (en) * 2001-03-13 2011-03-30 Paratek Pharmaceuticals, Inc. 7,9-Substituted tetracycline compounds
JP2005505588A (en) * 2001-10-05 2005-02-24 テトラジェネックス ファーマスーティカルズ インク Tetracycline derivative and method of using the same
US20050026833A1 (en) * 2001-11-08 2005-02-03 Rabbani Shafaat Ahmed PSP-94: use for treatment of hypercalcemia and bone metastasis
US7056902B2 (en) * 2002-01-08 2006-06-06 Paratek Pharmaceuticals, Inc. 4-dedimethylamino tetracycline compounds
WO2004038000A2 (en) * 2002-10-24 2004-05-06 Paratek Pharmaceuticals, Inc. Methods of using substituted tetracycline compounds to modulate rna
US20060287283A1 (en) * 2003-07-09 2006-12-21 Paratek Pharmaceuticals, Inc. Prodrugs of 9-aminomethyl tetracycline compounds
US8012950B2 (en) * 2003-08-29 2011-09-06 Wisconsin Alumni Research Foundation Method to diagnose and treat degenerative joint disease
EP2332904A3 (en) * 2004-01-15 2012-04-11 Paratek Pharmaceuticals, Inc. Derivatives of tetracycline compounds
CA2767912C (en) 2004-05-21 2015-08-11 President And Fellows Of Harvard College Synthesis of tetracyclines and analogues thereof
AR057324A1 (en) * 2005-05-27 2007-11-28 Wyeth Corp TIGECICLINE AND METHODS TO PREPARE 9-AMINOMINOCICLINE
AR057033A1 (en) * 2005-05-27 2007-11-14 Wyeth Corp TIGECICLINE AND METHODS TO PREPARE 9-NITROMINOCICLINE
AR057032A1 (en) * 2005-05-27 2007-11-14 Wyeth Corp TIGECICLINE AND PREPARATION METHODS
JP5335664B2 (en) * 2006-04-07 2013-11-06 プレジデント アンド フェロウズ オブ ハーバード カレッジ Synthesis of tetracycline and its analogs.
JP2009543544A (en) * 2006-05-15 2009-12-10 パラテック ファーマシューティカルズ インコーポレイテッド Methods for modulating gene or gene product expression using substituted tetracycline compounds
WO2008127361A2 (en) 2006-10-11 2008-10-23 President And Fellows Of Harvard College Synthesis of enone intermediate
WO2008045507A2 (en) 2006-10-11 2008-04-17 Paratek Pharmaceuticals, Inc. Substituted tetracycline compounds for treatment of bacillus anthracis infections
US9073829B2 (en) 2009-04-30 2015-07-07 President And Fellows Of Harvard College Synthesis of tetracyclines and intermediates thereto
PL2327676T3 (en) * 2009-11-26 2014-08-29 Sandoz Ag Reaction of organic compounds with low amounts of hydrogen
EP2544692A4 (en) * 2010-03-10 2013-08-07 Univ Health Network Use of tigecycline for treatment of cancer
CN102631356B (en) * 2012-04-11 2014-03-12 广州市东来生物科技有限公司 Application of 4-dedimethyl tetracycline derivatives in aspect of resisting Helicobacter pylori
EP2852383A4 (en) * 2012-05-21 2016-03-16 Dcb Usa Llc Methods for drug screen using zebrafish model and the compounds screened thereform
MA40836A (en) 2014-10-23 2017-08-29 Tetraphase Pharmaceuticals Inc SEMI-SYNTHESIS PROCEDURES
WO2018193114A1 (en) 2017-04-20 2018-10-25 Novintum Biotechnology Gmbh Triphenylphosphonium-tethered tetracycyclines for use in treating cancer
CN110156624B (en) * 2019-05-29 2022-03-08 河北冀衡药业股份有限公司 Method for synthesizing minocycline and derivatives thereof
KR20230154834A (en) 2021-02-03 2023-11-09 스카이바이오 엘엘씨 Chemically coupled carrier for low-hydrophobic bioactive drugs into the central nervous system

Citations (29)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3029284A (en) * 1960-09-14 1962-04-10 Pfizer & Co C Nu-dealkylation of carboxamido-nu-alkyltetracycline and its analogs
US3145228A (en) * 1962-09-06 1964-08-18 Pfizer & Co C 5-alkoxy-and 5-benzyloxy-tetracycline, derivatives and analogues thereof
US3265732A (en) * 1964-02-28 1966-08-09 American Cyanamid Co Novel 5alpha, 6-anhydrotetracyclines
US3338963A (en) * 1960-10-28 1967-08-29 American Cyanamid Co Tetracycline compounds
US3345370A (en) * 1965-03-16 1967-10-03 American Cyanamid Co 4-(n-mono-substituted) hydrazones of 4-dedimethylamino-4-oxo-tetracycline
US3345410A (en) * 1966-12-01 1967-10-03 American Cyanamid Co Substituted 7- and/or 9-amino tetracyclines
US3388162A (en) * 1966-03-14 1968-06-11 American Cyanamid Co 7-and/or 9-nitro or amino tetracyclines
US3502696A (en) * 1961-08-18 1970-03-24 Pfizer & Co C Antibacterial agents
US3502660A (en) * 1967-01-12 1970-03-24 Pfizer & Co C 2-((5'-(3' and/or 4'-substituted)isoxazoyl) methylidene)-3,4,10-trioxo-1,2,3,4,4a,9,9a,10-octahydroanthracenes
US3509184A (en) * 1961-08-18 1970-04-28 Pfizer & Co C Anthracyclidine-acetic acid derivatives
US3515731A (en) * 1966-06-17 1970-06-02 Pfizer & Co C Antibacterial agents
US3609188A (en) * 1964-10-29 1971-09-28 American Cyanamid Co 4-dedimethylamino-4-substituted-amino-6-demethyltetracyclines
US3622627A (en) * 1967-09-13 1971-11-23 Pfizer 4-dedimethylaminatetracycline and 5a, 6-anhydro derivatives thereof
US3772363A (en) * 1961-08-18 1973-11-13 Pfizer 3,4,10-trioxo octahydroanthracene-2-aminoacetic acids and derivatives thereof
US3824285A (en) * 1967-09-13 1974-07-16 Pfizer 4-oxo-4-dedimethylaminotetracycline-4,6-hemiketals
US3829453A (en) * 1961-08-18 1974-08-13 Pfizer Octahydroanthracene-2-aminoacetic acids and esters and mixed anhydrides thereof
US3849493A (en) * 1966-08-01 1974-11-19 Pfizer D-ring substituted 6-deoxytetracyclines
US4666897A (en) * 1983-12-29 1987-05-19 Research Foundation Of State University Inhibition of mammalian collagenolytic enzymes by tetracyclines
US4704383A (en) * 1983-12-29 1987-11-03 The Research Foundation Of State University Of New York Non-antibacterial tetracycline compositions possessing anti-collagenolytic properties and methods of preparing and using same
US4935412A (en) * 1983-12-29 1990-06-19 The Research Foundation Of State University Of New York Non-antibacterial tetracycline compositions possessing anti-collagenolytic properties and methods of preparing and using same
US4935411A (en) * 1983-12-29 1990-06-19 The Research Foundation Of State University Of New York Non-antibacterial tetracycline compositions possessing anti-collagenolytic properties and methods of preparing and using same
US5045538A (en) * 1990-06-28 1991-09-03 The Research Foundation Of State University Of New York Inhibition of wasting and protein degradation of mammalian muscle by tetracyclines
US5122519A (en) * 1989-06-27 1992-06-16 American Cyanamid Company Stable, cosmetically acceptable topical gel formulation and method of treatment for acne
US5495030A (en) * 1992-08-13 1996-02-27 American Cyanamid Company 9-[(substituted glycyl)amido)]-6-demethyl-6-deoxytetracyclines
US5494903A (en) * 1991-10-04 1996-02-27 American Cyanamid Company 7-substituted-9-substituted amino-6-demethyl-6-deoxytetracyclines
US5532227A (en) * 1992-11-17 1996-07-02 The Research Foundation Of State University Of New York Tetracyclines including non-antimicrobial chemically-modified tetracyclines inhibit excessive glycosylation of different types of collagen and other proteins during diabetes
US5776898A (en) * 1991-05-14 1998-07-07 Dana-Farber Cancer Institute Method for treating a tumor with a chemotherapeutic agent
US6506740B1 (en) * 1998-11-18 2003-01-14 Robert A. Ashley 4-dedimethylaminotetracycline derivatives
US6818634B2 (en) * 2000-03-31 2004-11-16 Paratek Pharmaceuticals, Inc. 7-and 9-carbamate, urea, thiourea, thiocarbamate, and heteroaryl-amino substituted tetracycline compounds

Family Cites Families (12)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
DE1063598B (en) * 1957-02-14 1959-08-20 Hoechst Ag Process for the preparation of water-soluble derivatives of the tetracyclines
DE1088481B (en) * 1958-01-29 1960-09-08 American Cyanamid Co Process for the preparation of new compounds of the tetracycline series
US2997471A (en) * 1958-08-18 1961-08-22 Bristol Myers Co Tetracycline derivatives
US3104240A (en) * 1958-08-18 1963-09-17 Bristol Myers Co Mannich bases of tetracycline antibiotics
US3026284A (en) * 1959-05-13 1962-03-20 Westinghouse Electric Corp Process of treating a thermoplastic amine-aldehyde resin with a thermosetting additive and product obtained therefrom
US3326761A (en) * 1964-11-16 1967-06-20 Pfizer & Co C 6-demethyl-6-deoxytetracycline, anti-tumor antibiotic
FR1520600A (en) * 1967-02-27 1968-04-12 Snecma Improvements to axial flow turbo-machines, and in particular to axial compressors with two nested counter-rotating rotors
USRE34656E (en) 1983-12-29 1994-07-05 The Research Foundation Of State University Of New York Use of tetracycline to enhance bone protein synthesis and/or treatment of bone deficiency
EP0733369A1 (en) * 1995-03-23 1996-09-25 Stichting REGA V.Z.W. Protease inhibitors, a DNA construct for the expression of a protease and a process for measuring proteases and/or protease inhibitors
US5837696A (en) * 1997-01-15 1998-11-17 The Research Foundation Of State University Of New York Method of inhibiting cancer growth
JP5060685B2 (en) * 1998-11-18 2012-10-31 ガルデマ ラボラトリーズ インコーポレイテッド Novel 4-dedimethylaminotetracycline derivative
JP2005505588A (en) * 2001-10-05 2005-02-24 テトラジェネックス ファーマスーティカルズ インク Tetracycline derivative and method of using the same

Patent Citations (30)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3029284A (en) * 1960-09-14 1962-04-10 Pfizer & Co C Nu-dealkylation of carboxamido-nu-alkyltetracycline and its analogs
US3338963A (en) * 1960-10-28 1967-08-29 American Cyanamid Co Tetracycline compounds
US3509184A (en) * 1961-08-18 1970-04-28 Pfizer & Co C Anthracyclidine-acetic acid derivatives
US3502696A (en) * 1961-08-18 1970-03-24 Pfizer & Co C Antibacterial agents
US3829453A (en) * 1961-08-18 1974-08-13 Pfizer Octahydroanthracene-2-aminoacetic acids and esters and mixed anhydrides thereof
US3772363A (en) * 1961-08-18 1973-11-13 Pfizer 3,4,10-trioxo octahydroanthracene-2-aminoacetic acids and derivatives thereof
US3145228A (en) * 1962-09-06 1964-08-18 Pfizer & Co C 5-alkoxy-and 5-benzyloxy-tetracycline, derivatives and analogues thereof
US3265732A (en) * 1964-02-28 1966-08-09 American Cyanamid Co Novel 5alpha, 6-anhydrotetracyclines
US3609188A (en) * 1964-10-29 1971-09-28 American Cyanamid Co 4-dedimethylamino-4-substituted-amino-6-demethyltetracyclines
US3345370A (en) * 1965-03-16 1967-10-03 American Cyanamid Co 4-(n-mono-substituted) hydrazones of 4-dedimethylamino-4-oxo-tetracycline
US3388162A (en) * 1966-03-14 1968-06-11 American Cyanamid Co 7-and/or 9-nitro or amino tetracyclines
US3515731A (en) * 1966-06-17 1970-06-02 Pfizer & Co C Antibacterial agents
US3849493A (en) * 1966-08-01 1974-11-19 Pfizer D-ring substituted 6-deoxytetracyclines
US3345410A (en) * 1966-12-01 1967-10-03 American Cyanamid Co Substituted 7- and/or 9-amino tetracyclines
US3502660A (en) * 1967-01-12 1970-03-24 Pfizer & Co C 2-((5'-(3' and/or 4'-substituted)isoxazoyl) methylidene)-3,4,10-trioxo-1,2,3,4,4a,9,9a,10-octahydroanthracenes
US3824285A (en) * 1967-09-13 1974-07-16 Pfizer 4-oxo-4-dedimethylaminotetracycline-4,6-hemiketals
US3622627A (en) * 1967-09-13 1971-11-23 Pfizer 4-dedimethylaminatetracycline and 5a, 6-anhydro derivatives thereof
US4666897A (en) * 1983-12-29 1987-05-19 Research Foundation Of State University Inhibition of mammalian collagenolytic enzymes by tetracyclines
US4704383A (en) * 1983-12-29 1987-11-03 The Research Foundation Of State University Of New York Non-antibacterial tetracycline compositions possessing anti-collagenolytic properties and methods of preparing and using same
US4935412A (en) * 1983-12-29 1990-06-19 The Research Foundation Of State University Of New York Non-antibacterial tetracycline compositions possessing anti-collagenolytic properties and methods of preparing and using same
US4935411A (en) * 1983-12-29 1990-06-19 The Research Foundation Of State University Of New York Non-antibacterial tetracycline compositions possessing anti-collagenolytic properties and methods of preparing and using same
US5122519A (en) * 1989-06-27 1992-06-16 American Cyanamid Company Stable, cosmetically acceptable topical gel formulation and method of treatment for acne
US5045538A (en) * 1990-06-28 1991-09-03 The Research Foundation Of State University Of New York Inhibition of wasting and protein degradation of mammalian muscle by tetracyclines
US5776898A (en) * 1991-05-14 1998-07-07 Dana-Farber Cancer Institute Method for treating a tumor with a chemotherapeutic agent
US5494903A (en) * 1991-10-04 1996-02-27 American Cyanamid Company 7-substituted-9-substituted amino-6-demethyl-6-deoxytetracyclines
US5495030A (en) * 1992-08-13 1996-02-27 American Cyanamid Company 9-[(substituted glycyl)amido)]-6-demethyl-6-deoxytetracyclines
US5532227A (en) * 1992-11-17 1996-07-02 The Research Foundation Of State University Of New York Tetracyclines including non-antimicrobial chemically-modified tetracyclines inhibit excessive glycosylation of different types of collagen and other proteins during diabetes
US6506740B1 (en) * 1998-11-18 2003-01-14 Robert A. Ashley 4-dedimethylaminotetracycline derivatives
US6894036B2 (en) * 1998-11-18 2005-05-17 Collagenex Pharmaceuticals, Incorporated 4-dedimethylaminotetracycline derivatives
US6818634B2 (en) * 2000-03-31 2004-11-16 Paratek Pharmaceuticals, Inc. 7-and 9-carbamate, urea, thiourea, thiocarbamate, and heteroaryl-amino substituted tetracycline compounds

Cited By (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7851460B2 (en) 2000-06-16 2010-12-14 Trustees Of Tufts College 7-phenyl-substituted tetracycline compounds
US8168810B2 (en) 2000-06-16 2012-05-01 Trustees Of Tufts College 7-phenyl-substituted tetracycline compounds
US7875649B2 (en) 2000-07-07 2011-01-25 Trustees Of Tufts College 7, 8 and 9-substituted tetracycline compounds
US7696188B2 (en) 2000-07-07 2010-04-13 Trustees Of Tufts College 7,8 and 9-substituted tetracycline compounds
US8252777B2 (en) 2000-07-07 2012-08-28 Trustees Of Tufts College 7, 8 and 9-substituted tetracycline compounds
US7696358B2 (en) 2001-03-13 2010-04-13 Paratek Pharmaceuticals, Inc. Five-membered heterocyclyl tetracycline compounds and methods of use thereof
US20060084634A1 (en) * 2001-03-13 2006-04-20 Paratek Pharmaceuticals, Inc. 7-Pyrollyl tetracycline compounds and methods of use thereof
US7897784B2 (en) 2001-03-13 2011-03-01 Paratek Pharmaceuticals, Inc. Process for preparing five-membered heterocyclyl tetracycline compounds and methods of use thereof
US20110160165A1 (en) * 2001-03-13 2011-06-30 Paratek Pharmaceuticals, Inc. 7-Pyrazolyl Tetracycline Compounds and Methods of Use Thereof
US20100081826A1 (en) * 2001-03-13 2010-04-01 Paratek Pharmaceuticals, Inc. 7-pyrrolyl tetracycline compounds and methods of use thereof
US8304445B2 (en) 2001-03-13 2012-11-06 Paratek Pharmaceuticals, Inc. 7-pyrazolyl tetracycline compounds and methods of use thereof
US9533943B2 (en) 2003-07-09 2017-01-03 Paratek Pharmaceuticals, Inc. Substituted tetracycline compounds
US7858601B2 (en) 2004-10-25 2010-12-28 Paratek Pharmaceuticals, Inc. 4-substituted tetracyclines and methods of use thereof
US8088755B2 (en) 2005-02-04 2012-01-03 Paratek Pharmaceuticals, Inc. 11a, 12-derivatives of tetracycline compounds
US8470804B2 (en) 2005-02-04 2013-06-25 Paratek Pharmaceuticals, Inc. 11a, 12-derivatives of tetracycline compounds

Also Published As

Publication number Publication date
WO2003030819A3 (en) 2004-01-08
EP1439843A2 (en) 2004-07-28
US20050267079A1 (en) 2005-12-01
RU2300380C2 (en) 2007-06-10
JP2005505588A (en) 2005-02-24
US20070142338A1 (en) 2007-06-21
US20040067912A1 (en) 2004-04-08
RU2004109986A (en) 2005-02-20
MY133403A (en) 2007-11-30
CN1564690A (en) 2005-01-12
US7214669B2 (en) 2007-05-08
US20070149488A1 (en) 2007-06-28
AU2002341970B2 (en) 2008-02-14
NO20041272L (en) 2004-07-02
CN1961885A (en) 2007-05-16
WO2003030819A2 (en) 2003-04-17
CA2462572C (en) 2008-11-18
CA2462572A1 (en) 2003-04-17
EP1439843A4 (en) 2007-01-03

Similar Documents

Publication Publication Date Title
US7214669B2 (en) Tetracycline derivatives and methods of use thereof
AU2002341970A1 (en) Tetracycline derivatives and methods of use thereof
AU651734B2 (en) Novel 9-amino-7-(substituted)-6-demethyl-6-deoxytetracyclines
EP1137410B1 (en) Novel 4-dedimethyl aminotetracycline derivatives
US20050171068A1 (en) Novel 4-dedimethylaminotetracycline derivatives
CA2503446C (en) Methods of using substituted tetracycline compounds to modulate rna
US6946453B2 (en) 4-dedimethylaminotracycline derivatives
CA2404628A1 (en) 7-and 9-carbamate, urea, thiourea, thiocarbamate, and heteroaryl-amino substituted tetracycline compounds
WO2004064728A2 (en) Use of specific tetracycline compounds in therapy
EP2157085B1 (en) Oxazole, derivatives of tetracyclines
EP1894569A2 (en) Tetracycline derivatives for treating cancer

Legal Events

Date Code Title Description
AS Assignment

Owner name: INNAPHARMA, INC., NEW JERSEY

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:HLAVKA, JOSEPH J.;ABLIN, RICHARD J.;REEL/FRAME:013677/0136

Effective date: 20030108

AS Assignment

Owner name: TETRAGENEX PHARMACEUTICALS, INC., NEW JERSEY

Free format text: MERGER;ASSIGNOR:INNAPHARMA, INC.;REEL/FRAME:016522/0775

Effective date: 20041124

REMI Maintenance fee reminder mailed
LAPS Lapse for failure to pay maintenance fees
STCH Information on status: patent discontinuation

Free format text: PATENT EXPIRED DUE TO NONPAYMENT OF MAINTENANCE FEES UNDER 37 CFR 1.362

FP Lapsed due to failure to pay maintenance fee

Effective date: 20110508