WO2004005298A1 - Derivatives of rifabutine useful as antimicrobial agents - Google Patents

Derivatives of rifabutine useful as antimicrobial agents Download PDF

Info

Publication number
WO2004005298A1
WO2004005298A1 PCT/PT2003/000009 PT0300009W WO2004005298A1 WO 2004005298 A1 WO2004005298 A1 WO 2004005298A1 PT 0300009 W PT0300009 W PT 0300009W WO 2004005298 A1 WO2004005298 A1 WO 2004005298A1
Authority
WO
WIPO (PCT)
Prior art keywords
rifabutin
oxide
compound according
compound
reaction
Prior art date
Application number
PCT/PT2003/000009
Other languages
French (fr)
Inventor
Maria Medeiros
Maria Costa
Ricardo Figueiredo
Maria Rosa
Maria Curto
Lina Santos
Maria Cruz
Maria Gaspar
Sónia FEIO
Original Assignee
Instituto Nacional De Engenharia E Tecnologia Industrial
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Instituto Nacional De Engenharia E Tecnologia Industrial filed Critical Instituto Nacional De Engenharia E Tecnologia Industrial
Priority to AU2003245183A priority Critical patent/AU2003245183A1/en
Publication of WO2004005298A1 publication Critical patent/WO2004005298A1/en

Links

Classifications

    • CCHEMISTRY; METALLURGY
    • C07ORGANIC CHEMISTRY
    • C07DHETEROCYCLIC COMPOUNDS
    • C07D498/00Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms
    • C07D498/22Heterocyclic compounds containing in the condensed system at least one hetero ring having nitrogen and oxygen atoms as the only ring hetero atoms in which the condensed system contains four or more hetero rings
    • 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

Definitions

  • Rifabutin la is a known derivative of rifamycin S active against mycobateria including atypic organisms like Mycoba terium avium and Mycobacterium intracell ulare . It is also active against rifampicin resistant Mycoba cteri um tuberculosis isolates, and has comparable activity to rifampicin against Staphylococcus aureus, Haemophil us ducreyi e Chlamydia trachoma tis (cf . R.N. Brogden and A. Fitton, "Rifabutin A Review of its Antimicrobial Activity, Pharmacokinetic Properties and Therapeutic Efficacy", Drugs, 47(6): 983-1009, 1994).
  • Rifabutin is the generic name of the chemical compound 4-deoxo-3, - [2-spiro (N-isobutyl-4-piperidyl) - 2, 5-dihydro-lH-imidazo] -rifamicin S.
  • Rifabutin la The preparation of Rifabutin la is reported in U.S. Patent No. 4.219.478 published on 26 August 1980.
  • Rifabutin is a red-violet coloured amorphous powder freely soluble in chloroform and methanol, slightly soluble in water, melting point 148-156°C (with decomposition) .
  • Rifabutin la is positioned as a especially relevant antibiotic in therapy.
  • Rifabutin la was the only studied antimycobacterial agent which was effective in placebo controlled prophylactic clinical tests.
  • Mycobutin R Upjohn & Pharmacia
  • Mycobutin R Upjohn & Pharmacia
  • MAC Mycobacterium avium complex
  • PCT/US93/00335 equivalent to WO 93/13774. 1993-07-21.
  • Rifabutin la is not effective for the treatment of influenza or common colds, although it may be useful in the establishment of a prophylactic regimen for multiple opportunist pathogens
  • MDR Mycobacteri um tuberculosis
  • any pharmaceutical candidate may present secondary undesirable effects and, in the case of Rifabutin la, uveite has been associated (an inflammatory condition of the eye with possible unpaired vision) since the first reported studies on the tolerance to dose-limit (e.g.: Siegal, F.; Eilbott, D.; Burger, H.; Gehan, K. ; Davidson, B., et al . , Dose-limiting toxicity of rifabutin in AIDS- related complex: syndrome of arthralgia/arthritis, AIDS, 4:433-441, 1990; Shafran, S.; Deschenes, J. ; Miller, M.
  • dose-limit e.g.: Siegal, F.; Eilbott, D.; Burger, H.; Gehan, K. ; Davidson, B., et al . , Dose-limiting toxicity of rifabutin in AIDS- related complex: syndrome of arthralgia/arthritis, AIDS
  • This invention relates to new compounds derived from Rifabutin la with relevant antibiotic activity.
  • Such derivatives are selected from a group consisting of compounds of general formula I
  • R 1 is hydrogen or -COR 2 , where R 2 is independently selected from straight, branched or cyclic C ⁇ -C 20 alkyl, saturated or unsaturated chain;
  • R 3 is oxygen or hydroxyl
  • R 4 is a nitrogen atom or an N-oxide group, and their pharmaceutically acceptable salts and solvates, including hydrates .
  • Oxone ® DuPont
  • aqueous solutions of Oxone may be useful to carry out oxidations either in homogeneous conditions, or in biphasic systems with an immiscible co-solvent and a phase transfer catalyst.
  • the heteroatoms are good acceptors of oxygen, the latter through the side presenting these unpaired electron pairs, the dimethyldioxirane behaving as an electrophilic oxidant able to selectively oxidize the tertiary amine function of Rifabutin la (See: Boyd, D. R. ; Coulter, P. B.; McGuckin, M. R.; Sharma, N. D.; Jennings, W. B. and Wilson, V. E., J. Chem . Soc . Perkin Trans . I, pp. 301- 306, 1990) and analogous molecules prepared in the field of present invention, producing further preferred compounds of this invention, in particular N-oxides of general Formula II
  • R 1 is hydrogen or -COR , where R is independently selected from hydrogen or straight, branched or cyclic C ⁇ C 2 rj alkyl, saturated or unsaturated chain;
  • R J is oxygen or hydroxyl; and their pharmaceutically acceptable salts and solvates, including hydrates.
  • Another embodiment of the present invention is the preparation of more preferred N-acylated derivatives of Rifabutin la with relevant antibiotic activity, using conventional techniques for selective acylation of the tertiary amine known to those skilled in the art thereby producing compounds of formula III
  • R 1 is hydrogen or -COR 2 , where R 2 is independently selected from straight, branched or cyclic C ⁇ -C 20 alkyl, saturated or unsaturated chain;
  • R 3 is oxygen or hydroxyl
  • R 4 is a nitrogen atom or an N-oxide group, and their pharmaceutically acceptable salts and solvates, including hydrates .
  • the mass spectra were obtained in a Brucker Daltonics Esquire 3000 spectrometer, with an ESI source/interface detecting negative ions; the concentration of the analysed solutions was: 1 ⁇ g of
  • M is the molecular ion
  • Ch is the chromophore (molecule core without the ansa chain)
  • a, b, and c are ions (cf. Studies of Vekey, K. ; Edwards, D. M. F. and Zerilli L. F. ; Journal of Chroma tography, 474, 317-327, 1989).
  • the infrared spectra were obtained in a Perkin-Elmer FT-IR 1725X spectrometer, as film.
  • the dichloromethane used was previously purified (cf. Perrin D.D., Armarego L.F., "Purification of laboratory chemicals", 2nd Edition, Oxford, Pergamon Press, 1980) .
  • the exact mass was determined by mass spectrometry operating with a laser ionisation-desorption source, at 70 eV and close to 200°C, with negative charged ions detection.
  • the reaction was complete after ca. 28 h 30 min.
  • the aqueous and organic layers were separated and the aqueous layer extracted with dichloromethane. The collected organic layers were washed with water, dried over anhydrous sodium sulfate, filtered and the solvent evaporated at reduced pressure.
  • agar 7H10 enriched with OADC from Difco the molecules derived from Rifabutin la of this invention were added at the final concentrations 0.1. 0.2 and 0.4 ⁇ g/mL and also Rifabutin la was added at final concentrations 0.1 and 0.2 ⁇ g/mL.
  • Plates containing Rifabutin la or its derivatives of this invention were inoculated with a strain of M. avium 1581 at dilutions 10 5 and 10 "4 . Plates without antibiotic were inoculated with a strain of M. avium at dilutions 10 "7 ' 10 "6, 10 "5 e 10 ⁇ 4 .
  • Table 1- Results from assays with Myc . avium 1581
  • Disc diffusion disc method was used according to the procedure described by Hong and Song, 2001 (see: Hong-Xi and Song, F. Lee, (2001) "Activity of plant flavonoids against antibiotic-resistant bacteria", Phytot er. Res . , 15. 39-43) as a diagnostic assay for antibacterial activity.
  • Petri dishes were prepared with an adequate culture medium and containing the microorganism beeing tested at the concentration 10 8 cfu mL -1 .
  • the compounds were dissolved in dimethylsulfoxide (DMSO) at the concentration lmg.mL "1 and applied in Oxoid discs at the ratio 20 ⁇ g/disc After evaporation of the solvent, the compounds of the invention were applied over each Petri dish medium prepared as indicated above and incubated at 37 °C, followed by determination of the diameters of the inhibition zones 24h later. Discs containing DMSO, without any compound of the invention added, were used as control, and did not show any inhibition of colonies. Rifampicin (Sigma) was used as Reference for the evaluation of the activity of the compounds to be tested against Gram-positive and Gram-negative bacteria.
  • DMSO dimethylsulfoxide
  • the Minimum Inhibitory Concentration (MIC) of the active compounds provided by the present invention was determined through the medium dilution method as described by Muroi and Kubo, 1996 (see: Muroi, H and Kubo, I, "Antibacterial activity of anacardic acid and totarol, alone and in combination with methicillin, against methicillin-resistant Staphylococcus aureus” , J. Appl . Bacteriol . 80. 387-394 (1996)). After incubation during 24h, the bacteria growth was examined and the value for the MIC defined as the minimum concentration of the tested compound, from which there is a null result after 24h incubation at 37°C. The minimum inhibitory concentration of each compound derived from Rifabutin provided by the present invention was determined at least in duplicate assays.
  • Tabela 2 Antibacterial Activity of Test Compounds determined by the Method of Diffusion with Discs
  • N-oxide of Rifabutin Ila inhibited the development of Gram-positive bacteria, and also the growth of Gram-negative bacteria, E. coli e S . enteri tidis .
  • the N-oxide of Rifabutinol lib and the N'- Acetyl-Rifabutinol Illb did not promote any inhibition of the growth of Gram-negative bacteria.
  • the compounds provided by the present invention which showed any antibacterial activity using the diffusion disc method were also tested by the liquid medium method using successive dilutions, for the determination of the minimum inhibitory concentration (MIC) .
  • Rifabutin Ila presented a weak activity against Gram-negative bacteria, with MIC values much higher (5 and 10 ⁇ g mL -1 ) for E. coli e S . enteri tidis than Rifabutin la and N ' -Acetil-Rifabutin Ilia.
  • Rifabutin la is bactericide against S . enteri tidis while N' -Acetyl-Rifabutin Ilia and rifampicin are bacteriostatic compounds.
  • Rifabutin la and N' -Acetyl-Rifabutin Ilia showed a more efficient inhibition of the growing of P.
  • aeruginosa than rifampicin, presenting MIC values of 6 ⁇ g mL- 1 .
  • Rifampicin demonstrated to be more active against Gram-positive bacteria than Rifabutin la and N ' -Acetyl-Rifabutin Ilia.
  • the bacteriostatic compounds are bactericides at superior concentrations.

Landscapes

  • Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Health & Medical Sciences (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Oncology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Communicable Diseases (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Health & Medical Sciences (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Nitrogen And Oxygen Or Sulfur-Condensed Heterocyclic Ring Systems (AREA)

Abstract

This invention relates to new derivatives of rifabutine of general formula I, wherein R1 is hydrogen or COR2 in which R2 is independently selected from saturated or insaturated linear, branched or cyclic alkyl C1-C20; R3 is oxygen or hydroxyl; R4 is a nitrogen atom or N-oxide, and their acceptable salts and solvates, including hydrates. These derivatives are useful as anti-infectious agents, namely against Mycobacterium avium, and Mycobacterium tuberculosis. (I)

Description

FIELD OF THE INVENTION
DERIVATIVES OF RIFABUTINE USEFUL AS ANTIMICROBIAL AGENTS
BACKGROUND OF THE INVENTION
Rifabutin la is a known derivative of rifamycin S active against mycobateria including atypic organisms like Mycoba terium avium and Mycobacterium intracell ulare . It is also active against rifampicin resistant Mycoba cteri um tuberculosis isolates, and has comparable activity to rifampicin against Staphylococcus aureus, Haemophil us ducreyi e Chlamydia trachoma tis (cf . R.N. Brogden and A. Fitton, "Rifabutin A Review of its Antimicrobial Activity, Pharmacokinetic Properties and Therapeutic Efficacy", Drugs, 47(6): 983-1009, 1994).
Rifabutin is the generic name of the chemical compound 4-deoxo-3, - [2-spiro (N-isobutyl-4-piperidyl) - 2, 5-dihydro-lH-imidazo] -rifamicin S. Depending on the nomenclature system utilized it may also be identified as 6, 9-dihydro-5,17, 19, 21-tetrahydroxy-8, 9- [2-spiro- (N- isobutyl-4-piperidyl) -2, 5-dihydro-lH-imidazo] -23-metoxi- 2, 4, 12, 16, 18, 20, 22-heptamethyl-6-oxo-2, 7-epoxypentadeca- [1, 11, 13]-trienimine-2H-furo[2',3',7,8]nafto[l,2- d] imidazolo-2, 4 ' -piperidine-5, 10,26. (3H, 9H) -trione-16- acetate. Rifabutin is a compound of formula la:
Figure imgf000004_0001
la
Molecular Formula: C46H62 40n Molecular Weight: 847.12
The preparation of Rifabutin la is reported in U.S. Patent No. 4.219.478 published on 26 August 1980. Rifabutin is a red-violet coloured amorphous powder freely soluble in chloroform and methanol, slightly soluble in water, melting point 148-156°C (with decomposition) . Rifabutin la is positioned as a especially relevant antibiotic in therapy. In 1994 Rifabutin la was the only studied antimycobacterial agent which was effective in placebo controlled prophylactic clinical tests. It is commercially known as MycobutinR (Upjohn & Pharmacia) and it is prescribed daily in 300 mg dosage units for prophylaxis of infections caused by disseminated Mycobacterium avium complex (MAC) in AIDS patients ( see : PCT/US93/00335 equivalent to WO 93/13774. 1993-07-21. BANKS PHILLIP L.,(US); WOLGEMUTH RICHARD L.,(US); WYNNE BEVERLEY A., Applicant ERBAMONT INC (US)) and also more recently as prophylactic agent in HIV and tuberculosis infected patients (Narita, M.; Stambaugh, J. ; Hollender, S.; Jones, D. ; Pitchenik, E.; Ashkin, D. , Clin . Infect . Dis . , 30(5), 779-783. 2000). Rifabutin la is not effective for the treatment of influenza or common colds, although it may be useful in the establishment of a prophylactic regimen for multiple opportunist pathogens
(see: Benson, A.; Williams, L.; Cohn, L.; Becker, S . ; Hojczyk, P.; Nevin, T . ; Korvick, A.; Heifets, L. ; Child, C; Lederman, M.; Reichman, C; Powderly, G.; Notario, F.; Wynne, A.; Hafner, R. , J. Infect . Dis . , 181(4), 1289- 1297, 2000; and, Cirioni, 0.; Giacometti, A.; Barchiesi, F.; Fortuna, M.; Scalise, ' G. , J. Antimicrob . Chemother. , 44(5), 653-659, 1999) and against Mycobacteri um tuberculosis (MTB) isolates and multiresistances (MDR) (see, e.g., Chien, H.-P.; Yu, M.-C; Ong, T.-F.; Lin, T.- P.; Luh, K.-T., J. Formosan Med. Assoc , 99(5), 408-411, 2000) . Rifabutin la was also recently reported for the treatment of Chron's disease (see: US Patent No. 6297015. published 2 October 2001. by Shafran I. (US) untitled Chron 's disease diagnostic and trea tmen t methods and composi tions) and for treatment of patients where the eradication of infections caused by Helicobacter pylori had not occurred after triple reference therapies based on inhibitors of the proton pump (see: Patent WO9702039 untitled Anti-jbacterial synergistic composi tions comprising rifabutin, Date of publication: 1997-01-23. Inventor (s): ROSSI R. ; JABES D. ; CASTELLANI P. and applied by PHARMACIA & UPJOHN SPA (IT)).
Once approved as a medicine for a specific use, any pharmaceutical candidate may present secondary undesirable effects and, in the case of Rifabutin la, uveite has been associated (an inflammatory condition of the eye with possible unpaired vision) since the first reported studies on the tolerance to dose-limit (e.g.: Siegal, F.; Eilbott, D.; Burger, H.; Gehan, K. ; Davidson, B., et al . , Dose-limiting toxicity of rifabutin in AIDS- related complex: syndrome of arthralgia/arthritis, AIDS, 4:433-441, 1990; Shafran, S.; Deschenes, J. ; Miller, M. ; Phillips, P., Toma, E., Uveitis and pseudojaundice during a regimen of clarithromycin, rifabutin, and ethambutol. Correspondence. New England Journal of Medicine, 330:438- 439, 1994) .
The authors of the present invention became interested in the exceptional biological profile of Rifabutin la and considered an important challenge the optimization of its therapeutic activity. Accordingly, a first step was trying to understand the structure- activity relationships of Rifabutin la and related molecules through a few studies of conformational analysis using nuclear magnetic resonance spectroscopy techniques which were published in speciality journals (See: Santos, L.; Fant F. ; Medeiros, M. A.; Borremans, F. A. M.; Costa, M. C. and Curto, M. J. M., "Structural Characterization by NMR of Rifabutinol, A Derivative of Rifabutin," Magn . Reson . Chem . , 38: 937-945, 2000; Santos, L.; Medeiros, M. A.; Santos, S.; Costa, M. C; Tavares, R. and Curto, M. J. M. , "NMR STUDIES of Some Rifamycins," Journal of Molecular Structure, 563-564, 61- 78, 2001) . As a result of the above preliminary studies, active derivatives of Rifabutin were synthesized for the first time possessing high antibacterial activity, in particular against Mycobacterium avi um . Such compounds offered new advantageous perspectives for the therapy of diseases where Rifabutin la has been currently indicated and administered.
DETAILED DESCRIPTION OF THE INVENTION
This invention relates to new compounds derived from Rifabutin la with relevant antibiotic activity. Such derivatives are selected from a group consisting of compounds of general formula I
Figure imgf000008_0001
wherein: R1 is hydrogen or -COR2, where R2 is independently selected from straight, branched or cyclic Cι-C20 alkyl, saturated or unsaturated chain;
R3 is oxygen or hydroxyl;
R4 is a nitrogen atom or an N-oxide group, and their pharmaceutically acceptable salts and solvates, including hydrates .
Such compounds are obtained as a powder having a colour ranging from orange-reddish to red-violet, they are soluble in polar organic solvents and water insoluble. New compounds provided by this invention may be obtained by reaction of Rifabutin la above with well known mild oxidative reagents, for example Oxone® (DuPont) , which is a stable and convenient source of potassium monoperoxosulfate (caroate) , and may be used for oxidation purposes in a great variety of conditions known to one skilled in the art. Thus, aqueous solutions of Oxone may be useful to carry out oxidations either in homogeneous conditions, or in biphasic systems with an immiscible co-solvent and a phase transfer catalyst. In a similar manner to other currently used oxidation procedures, there is an in si tu formation and reaction of a secondary reactant in the particular reaction conditions, because in reactions involving aqueous Oxone- ketone systems, dioxirane production occurs as an active intermediate (See, e.g.: Burke S. D. e Danheiser R. L.; "Handbook of reagents for organic synthesis - oxidizing and reducing agents," John Wiley & Sons, 2000; Yang, D. ; Yip, Y. C. and Wong, M. K. , J. Org. Chem . , 64. 1635-1639, 1999) . Owing to the presence of available electron pairs, the heteroatoms are good acceptors of oxygen, the latter through the side presenting these unpaired electron pairs, the dimethyldioxirane behaving as an electrophilic oxidant able to selectively oxidize the tertiary amine function of Rifabutin la (See: Boyd, D. R. ; Coulter, P. B.; McGuckin, M. R.; Sharma, N. D.; Jennings, W. B. and Wilson, V. E., J. Chem . Soc . Perkin Trans . I, pp. 301- 306, 1990) and analogous molecules prepared in the field of present invention, producing further preferred compounds of this invention, in particular N-oxides of general Formula II
Figure imgf000010_0001
II
wherein:
R1 is hydrogen or -COR , where R is independently selected from hydrogen or straight, branched or cyclic Cι~ C2rj alkyl, saturated or unsaturated chain;
RJ is oxygen or hydroxyl; and their pharmaceutically acceptable salts and solvates, including hydrates. Another embodiment of the present invention is the preparation of more preferred N-acylated derivatives of Rifabutin la with relevant antibiotic activity, using conventional techniques for selective acylation of the tertiary amine known to those skilled in the art thereby producing compounds of formula III
Figure imgf000011_0001
12'
III
wherein:
R1 is hydrogen or -COR2, where R2 is independently selected from straight, branched or cyclic Cι-C20 alkyl, saturated or unsaturated chain;
R3 is oxygen or hydroxyl;
R4 is a nitrogen atom or an N-oxide group, and their pharmaceutically acceptable salts and solvates, including hydrates . EXPERIMENTAL
All the reagents and the solvents used were analytical pure products. The reactions were followed by thin layer chromatography (t.l.c.) on Merck 60 F254 (0.2 mm), silica gel pre-prepared plates. The isolation of the products was performed by column chromatography using silica gel Merck 60 (230-400 mesh, 0.04-0.063 mm), at normal atmospheric pressure or flash with nitrogen pressure, or also by preparative thin layer chromatography on silica gel Merck 60 F254 (0.5 mm) plates. In the case of 25-hydroxy-Rifabutin-N-oxide lie, column chromatography purification took place with RP-18 silica of Fluka (>400 mesh, 0.015-0.035 mm).
The mass spectra were obtained in a Brucker Daltonics Esquire 3000 spectrometer, with an ESI source/interface detecting negative ions; the concentration of the analysed solutions was: 1 μg of
'substrate/ mL of 1:1 solution of 0.1% acetic acid/acetonitrile. "M" is the molecular ion, "Ch" is the chromophore (molecule core without the ansa chain) and a, b, and c are ions (cf. Studies of Vekey, K. ; Edwards, D. M. F. and Zerilli L. F. ; Journal of Chroma tography, 474, 317-327, 1989).
With the exception of the Rifabutin-N-oxide Ila and 25-hydroxy-Rifabutin-N-oxide lie, nuclear magnetic resonance spectra were registered in a General Electric GE-NMR spectrometer, with a proton frequency of 300.65 MHz and a carbon-13 frequency of 75.6 MHz, all the other spectra were registere in a Brucker spectrometer with a proton frequency of 400.13 MHz and a carbon-13 frequency of 100.62 MHz. With the exception of Rifabutinol N-oxide lib that was dissolved in deuterated methanol, all the samples were dissolved in 99% deuterated chloroform, with tetramethylsilane as internal standard. The obtained results are presented in the following order: solvent, chemical shift (δ in ppm) ; relative intensity (as number of protons, H) ; multiplicity (s-singlet, bs-broad singlet, d-duplet, dd-double duplet, bd-broad duplet, t- triplet, q-quartet, m-multiplet) ; coupling constant J in Hz; and location in the molecule structure.
The infrared spectra were obtained in a Perkin-Elmer FT-IR 1725X spectrometer, as film. The dichloromethane used was previously purified (cf. Perrin D.D., Armarego L.F., "Purification of laboratory chemicals", 2nd Edition, Oxford, Pergamon Press, 1980) . The exact mass was determined by mass spectrometry operating with a laser ionisation-desorption source, at 70 eV and close to 200°C, with negative charged ions detection.
Example 1. Synthesis of Rifabutinol-N-oxide lib 1.1. Synthesis of Rifabutinol lb
Figure imgf000014_0001
To Rifabutin la (300.0 mg; 0.354 mmol) previously dissolved in THF (9.2 mL) in a round-bottomed flask, was added Pt02 (10.7 mg; 0.047 mmol) and sodium borohydrate (278.1 mg; 7.35 mmol) dissolved in ethanol (6.5 mL) dropwise. The reaction mixture was maintained under stirring at room temperature, and the reaction was followed by t.l.c. using a mixture of petroleum ether/diethyl acetate/methanol (55:35:10) as eluant, till completion (ca. 4 h 05 min). The reaction mixture was filtered through Celite, and the Celite washed with diethyl ether until the absence of colour was noted. A few mL of distilled water were added and the resulting solution transferred to a separatory funnel. A few extractions of the aqueous layer with diethyl ether were made until the ether layer was discoloured, and collected in a Erlenmeyer, dried over NaS04, filtered to produce a crude red coloured solid product dried in vacuum (282.9 mg; η=94.2%), and further purified by Flash Column Chromatography, using mixtures of increasing polarity: Petroleum ether/ethyl acetate/methanol (62.5:35:2.5) and Petroleum ether/ethyl acetate/methanol (60:35:5), to give Rifabutinol lb (143.1 mg; η=47.6%). Rifabutinol lb :
^-N.M.R. (CDCI3) δ(ppm) : 0.04 (3H, d, J34/26= 7.2Hz, Me- 34); 0.61 (3H, d, J33/24= 6.8Hz, Me-33) ; 0.85 (3H, d, J3ι/20= 7.0Hz, Me-31); 0.93 (6H, d, Jιr/ιo= Ji2'/ιo= 6.4Hz, Me-11' e Me-12') ; 1.04 (3H, d, J32 22= 7.0Hz, Me-32); 1.52 (1H, ddd, J26/25=9.9Hz, J26/27=2.5Hz, J26/34=7.3. H-26) ; 1.67- 1.85 (4H, m, H-24/H-22/H-10' ) ; 1.94 (3H, s, Me-13) ; 2.00 (4H, m, CH2-4'/CH2-8' ) ; 2.06 (6H, s, Me-30 e Me-36) ; 2.22 (3H, s, Me-14) ; 2.23 (2H, d, J9-/ιo' = 6.8Hz, CH2-9' ) ; 2.38 (1H, m, H-20) ; 2.69-2.79 (4H, m, CH2-5'e CH2-7'); 3.03 (1H, m, H-23); 3.06 (3H, s, Me-37); 3.48 (1H, m, H-27); 3.72 (1H, s, 21-OH); 3.79 (1H, bd, J2ι/20= 9.9Hz, H-21) ; 4.08 (1H, d, JOH/23= 4.5Hz, 23-OH) ; 4.95 (1H, dd, J28 27= 4.2Hz/ J28/29= 12.3Hz, H-28); 5.05 (1H, d, J25/26= 10.1Hz, H- 25); 5.55 (1H, s, H-ll) ; 5.98 (1H, dd, J29/28= 12.3Hz, J29 27= 1.2Hz H-29) ; 6.15 (1H, dd, Jι9/ι8= 15.7Hz, Jι9 20= 6.8Hz, H-19); 6.25 (1H, bd, Jι7/ι8= 10.8Hz, H-17); 6.43 (1H, dd, Ji8/ι7= 10.9Hz, Jι8/ι9= 15.8Hz, H-18) ; 6.80 (1H, s, 11-OH) ; 8.12 (1H, bs, NH' ) ; 8.33 (1H, s, NH) ; 13.70 (1H, s, 8-OH)
13C-N.M.R. (CDC13) δ(ppm) : 8.2 (C-14); 8.7 (C-33) ; 8.8 (C- 34); 11.2 (C-32) ; 17.7 (C-31); 20.5 (C-30) ; 20.8 (C- ll'/C-12') ; 20.9 (C-36) ; 25.1 (C-13) ; 25.8 (C-10' ) ; 33.3 (C-22) ; 36.3 (C-4')*; 36.4 (C-8')*; 37.2 (C-24); 37.4 (C- 20) ; 39.0 (C-26); 51.5 (C-5'/C-7'); 57.5 (C-37) ; 66.4 (C- 9'); 73.2 (C-21); 74.2 (C-25) ; 76.6 (C-ll) ; 77.2 (C-23) ; 77.5 (C-27); 92.9 (C-2'); 105.8 (C-3) ; 108.3 (C-9) ; 111.5 (C-28) ; 111.8 (C-12); 113.1 (C-7) ; 120.0 (C-5) ; 120.7 (C- 10); 124.0 (C-18) ; 130.3 (C-16) ; 133.7 (C-17); 140.8 (C- 2); 141.7 (C-19); 142.4 (C-29) ; 159.0 (C-4); 159.9 (C-6) ; 163.2 (C-8); 168.6 (C-15) ; 172.7 (C-35) ; 182.1 (C-l). * - interchangeable values
I.V. fum (NaCl) ,vmέx(cm_1) : 3252 (N-H) ; 2964. 2821 (C-H) 1714 (C35=0) ;1674 (Cι5=0) ; 1645 (C4=N) ; 1616. 1574 (C=C) 1505 δ(N-H);1417 (C8-0) e δ(O-H); 1384 δ(C-H); 1344 (C-N) 1283 δ(O-H); 1262 (C35-0-C25) ; 1149 (C-N); 1063 (C-O) e (C35-0-C25) ; 974 γ(C-H); 769 (chromophore ' s vibration).
Mass/ESI (m/z) : 847 [M-H] •"; 755 [ (M-H) -CH3C00H-CH30H] ■"; 729 [M- iC4H9-N(CH3)=CH2)-H20] ■" ; 545 [(M-H)-c] -~;423[Ch- H] •"; 323[Ch-(iC4H9-N(CH3)=CH2) ] •"
Exact Mass (m/z)- C46H64 40ιι (M)" obtained: 848.45769/ (M) " caic = 848.45771.
1.2. Synthesis of Rif abutinol-N-oxide lib
Figure imgf000016_0001
To a round-bottomed flask with Rifabutinol lb (181.1 mg; 0.213 mmol) in dichloromethane (9.1 mL) , was added benzyl triethylammonium chloride (74.0 mg; 0.325 mmol), sodium hydrogenocarbonate (12.1 mg; 0.144 mmol), Oxone
(263.6 mg; 0.429 mmol) in acetone (1.8 mL; 24.5 mmol), distilled water (9.2 mL) and a Metrohm pH=7 buffer (12.7 mL) , stirring at room temperature. The reaction was followed by t.l.c, using petroleum ether/ethyl acetate/methanol (55:35:10) as eluant; 1 h 15 min after the beginning of the reaction it was progressively added
2.25 eq. more of Oxone (593.4 mg; 0.965 mmol) and buffer
(2.5 mL) to adjust the medium pH. The reaction was followed by t.l.c. and it was complete after 26 h 30 min. The flask content was transferred to a separatory funnel, and extracted with dichloromethane (2x25 mL) ; the combined organic layers were collected in a Erlenmeyer, transferred to a new separatory funnel, washed with distilled water and the resulting organic layers were collected and dried over anhydrous sodium sulfate. The resulting organic layers were filtered, the solvent evaporated at reduced pressure, and a crude red coloured solid produced was dried in vacuum (152.1 mg; η=82.4%) and further purified by Flash Column Chromatography, using mixtures of increasing polarity: Petroleum ether/ethyl acetate/methanol (65:25:10), Petroleum ether/ethyl acetate/methanol (55:35:10), Petroleum ether/ethyl acetate/methanol (50:35:15), Petroleum ether/ethyl acetate/methanol (45:30:25), ethyl acetate/methanol (40:60), ethyl acetate/methanol (30:70), ethyl acetate/methanol (20:80) and ethyl acetate/methanol
(10:90), to give Rifabutinol-N-oxide lib (94.2 mg; η=51.0%) . Rifabutinol-N-oxide lib:
^-N.M.R. (MeOD) δ(ppm) : -0.04 (3H, d, J34/26= 6.6Hz, Me- 34) ; 0.56 (3H, d, J33 24=6.6Hz, Me-33) ; 0.84 (3H, d, J3ι/20= 6.6Hz, Me-31) ; 0.95 (3H, d,J32 22= 6.8Hz, Me-32) ; 1.13 (6H, d, Jιi'/ιo= Ji2'/ιo= 6.6Hz, Me-11' e Me-12' ) ; 1.21-1.28 (IH, m, H-26) ; 1.62 (IH, m, H-24) ; 1.72 (IH, m, H-22) ; 1.85 (3H, s, Me-13) ; 1.94 (3H, s, Me-36) ; 2.03 (3H, s, Me-30) ; 2.08 (3H, s, Me-14) ; 2.32 (IH, m, H-20) 2.40 (IH, m, H-
10') ; 2.98 (3H, s, Me-37) ; 3.04 (1. m, H-23) ; 3.32 (2H, m, CH2-9' ) ; 3.38 (IH, m, H-27) 1.3-3.8 (m, CH2-7'/ CH2-5' /
CH2-4'/ CH2-8'); 3.85 (IH, bd, J2ι/20= 9.6Hz, H-21) ; 4.89
(IH, m, H-28) ; 5.18 (IH, d, J25/26= 10.0Hz, H-25) ; 5.59 (IH, s, H-ll) ; 5.93 (IH, d, J29/28= 12.4Hz, H-29) ; 6.02 (IH, dd, Ji9 i8= 15.6Hz, Jι9 20= 6.8Hz, H-19) ; 6.27 (IH, bd, Ji7/ιβ= 10.4Hz, H-17); 6.59 (IH, dd, Jι8/ι7= 11.0Hz, Jι8/ιg= 15.6Hz, H-18) ; 13C-N.M.R. (MeOD) δ(ppm) : 8.4 (C-14); 9.5 (C-33) ; 9.7 (C- 34) ; 11.3 (C-32) ; 18.0 (C-31); 20.8 (C-30) ; 21.0 (C-36) ; 23.5 (C-ll'/C-12' ) ; 24.6 (C-10' ) ; 25.3 (C-13); 31.7 (C- 4' /C-8') ; 34.3 (C-22); 39.0 (C-20/C-24); 40.7 (C-26) ; 57.5 (C-37) ; 63.7 (C-5' ) ; 64.0 (C-7' ) ; 74.6 (C-21) ; 74.9 (C-25) ; 77.6 (C-ll); 78.1 (C-23) ; 78.2 (C-27); 79.5 (C- 9' ); 91.5 (C-2' ) ; 105.4 (C-3) ; 110.2 (C-9) ; 113.0 (C-12) 114.2 (C-7) ; 114.8 (C-28) ; 121.3 (C-5); 121.9 (C-10) 127.1 (C-18); 132.0 (C-16) ; 134.6 (C-17) ; 140.9 (C-19) 143.4 (C-29) ; 145.1 (C-2); 160.8 (C-4) ; 161.9 (C-6) ; 164.1 (C-8) ; 169.3 (C-15) ; 172.8 (C-35) ; 184.9(C-1) . I.V. n, (NaCl) vmέx(cm_1): 3418 (N-H) ; 3252 (O-H) ; 2964.
2938. 2880 (C-H) ; 1730 (Cu=0) ; 1673 (Cι5=0) ; 1645 (C4=N) ;
1633 (Cι=0) ; 1616. 1605. 1582 (C=C) ; 1520 δ(N-H); 1418
(C8-0) e δ(O-H); 1374 δ(C-H); 1285 δ(O-H); 1256 (C35-0-C25) ;
1149 (C-N); 1078 (N-O); 1062 (C-O) e (C35-0-C25) ; 974 γ(C-
H) ; 768 (chromophore's vibration) .
Mass/ESI (m/z): 864 [M] •"; 848 [M-O] '"; 820 [ (M-H) -C3H8] •";
735 [ (M-H) -iC4H9N(CH3)=CH2] •"; 703 [ (Ch-H) - (iC4H9N (CH3) =CH2) -
CH3OH]-~; 543 [ (M-3H) -O-c] •"; 422 [ (Ch-2H) -O] •";
Exact Mass (m/z ) C46H64N42 (M) " obtained : 848 . 45779 (M-O) " .
Example 2 . Synthesis of 25-hydroxy-Rif abutin N-oxide l ie
2 . 1 . Synthesis of 25-hydroxy-Rif abutin Ic
Figure imgf000019_0001
To a solution of Rifabutin la (300.1 mg; 0.355 mmol) in methanol (15 mL) in a round-bottomed flask, a solution of NaOH (0.184M in MeOH/H20 1:1) was added followed by an aqueous solution of ZnCl2 (48.7 mg; 0.357 mmol). The extraction mixture was stirred at room temperature and followed by t.l.c, and ^ eq. more of ZnCl2 (24.4 mg; 0.179 mmol) was added and NaOH (27.2 mg) until complete conversion of starting material. After completion of the reaction (ca. 6 h 30 min) the mixture was extracted with ethyl acetate (5x25 mL) and the organic layers washed with water (30 mL) . Drying agent (MgS04) was added, the solution filtered, evaporated in a rotary evaporator and dried in vacuum, to give a chromatographic homogeneous 25-hydroxy-Rifabutin Ic, as a purple coloured solid (284.4 mg; η=99.8%) .
25-hydroxy-Rifabutin Ic:
^-N.M.R. (CDC13) δ(ppm) : -0.12 (3H, d, J34/26= 6.8Hz, Me- 34) ; 0.56 (3H, d, J33/24= 6.8Hz, Me-33) ; 0.84 (3H, d, J3ι 2o= 6.8Hz, Me-31) ; 0.95 (6H, d, Jιr/ιo= Ji2'/ιo= 6.8Hz, Me-11' e Me-12'); 1.02 (IH, m, H-24); 1.09 (3H, d, J32/22= 6.8Hz, Me-32); 1.74 (3H, s, Me-13) ; 1.7-1.8 (IH, m, H-22) ; 1.8- 1.9 (5H, m, H-10'/H-26/CH2-8'*) ; 2.05 (3H, bs, Me-30) ; 2.0-2.1 (2H, m, CH2-4')*; 2.30 (3H, s, Me-14); 2.32 (2H, m, CH2-9' ) ; 2.43 (IH, m, J20/3ι=6.8Hz, H-20) ; 2.67 (2H, m, CH2-5')*; 2.96-2.99 (4H, m, 21-OH/23-OH/CH2-7' *) ; 3.18 (3H, s, Me-37); 3.32 (IH, bd, J25/26= 10.4Hz, H-25) ; 3.40 (IH, dd, J27 26=3.8Hz/ J27/28=10.1Hz, H-27) ; 3.55 (IH, t, J23/23OH=8.3HZ, H-23) ; 3.72 (IH, d, J2ι/20= 9.7Hz, H-21) ; 4.20 (IH, s, 25-OH) ; 5.20 (IH, dd, J28 27= 10.3Hz/J28 29= 12.6Hz, H-28); 5.91 (IH, dd, Jι9/ι8= 14.9Hz, Jι9/20= 6.0Hz, H-19) ; 6.24-6.32 (2H, m, H-18/H-17) ; 6.36 (IH, bd, J29 28= 12.8Hz, H-29); 8.29 (IH, s, NH) ; 9.71 (IH, s, NH' ) ; 14.60 (IH, s, 8-OH) . 13C-R.M.C. (CDCI3) δ(ppm) : 7.7 (C-14) ; 8.3 (C-33) ; 10.8 (C- 32) ; 12.1 (C-34); 17.1 (C-31) ; 20.0 (C-30); 20.8 (C- ll'/C-12'); 22.8 (C-13) ; 25.8 (C-10'); 32.7 (C-22) ; 35.2 (C-4')*; 36,0 (C-8')*; 37.7 (C-26) ; 38.8 (C-24); 39.4 (C- 20); 51.5 (C-5' /C-7') ; 56.0 (C-37) ; 66.3 (C-9' ) ; 70.8 (C- 25); 71.5 (C-21) ; 76.3 (C-23) ; 85.4 (C-27); 94.7 (C-2' ) ; 104.2 (C-3) ; 108.0 (C-12) ; 109.4 (C-10) ; 111.4 (C-9) ; 114.6 (C-7/C-28); 123.2 (C-18) ; 124.8 (C-5) ; 132.4 (C- 16); 132.8 (C-17); 140.9 (C-19) ; 141.5 (C-2) ; 147.4 (C-29) ; 154.9 (C-4); 168.1 (C-8); 168.2 (C-15) ; 170.9 (C- 6) ; 180.3(C-1) ; 192.0 (C-ll) . * - interchangeable values
I.V. fun, (NaCl) vmέx(cm_1) : 3493 (O-H) ; 3360 (N-H) ; 2960. 2824 (C-H) ; 1728 (Cu=0) ; 1674 (Cι5=0) ; 1646 (C4=N) ; 1602. 1562 (C=C); 1515 δ(N-H); 1455. 1422 (C8-0) e δ(O-H); 1372 δ(C-H) ; 1342 (C-N) ; 1294 δ(O-H) ; 1157 (C-N); 1071 (C-O); 981 γ(C-H); 766 (chromophore ' s vibration) .
Mass/ESI (m/z) : 804 [M] •" ; 771 [(M-H) - CH3OH] •" ; 753 [(M-H) - CH3OH - H20]-"; 571 [(M-H) - b] '"; 543 [(M-H) - c]-"; 421 [Ch-H] ■"; 322 [Ch - (iC H9-N (CH3) =CH2) ] '" Exact Mass (m/z) C 4H60N4O10 (M-H)" obtained: 803. 2332/ (M- H)" caι = 803.423670.
2.2. Synthesis of 25-hydroxy-Rifabutin-N-oxide lie
Figure imgf000022_0001
25-hydroxy-Rifabutin Ic 25-hydroxy-Rifabutin N-oxide lie M=804 , 98g/mol M=820, 98g/mol
In a round-bottomed flask containing 25-hydroxy- Rifabutin Ic (150.0 mg; 0.186 mmol) in dichloromethane (7.9 mL) the following reactants were added with stirring at room temperature: benzyl triethylammonium chloride (70.0 mg; 0.307 mmol); NaHC03 (13.5 mg; 0.161 mmol), distilled water (7.9 mL) , pH=7 buffer (10 mL) and Oxone (179.1 mg; 0.291 mmol). The Oxone was slowly added to the reactional mixture during a period ca. 3 h 15 min of the reaction.
The reaction was followed by t.l.c. (petroleum ether/ethyl acetate/methanol - 45:35:20); after ca. 19 h of reaction, more Oxone was added (282.0 mg; 0.458 mmol), as well as pH=7 buffer in order to maintain the pH ca . 7. The reaction was complete after ca. 28 h 30 min. The aqueous and organic layers were separated and the aqueous layer extracted with dichloromethane. The collected organic layers were washed with water, dried over anhydrous sodium sulfate, filtered and the solvent evaporated at reduced pressure. A crude product was produced after drying in vacuum (102.5 mg; η=67.0%), which was further purified by Flash column chromatography, using reverse phase silica with the petroleum ether/methanol (2:98) eluant, to give 25- hydroxy-Rifabutin-N-oxide lie (29.3 mg; η=19.1%).
25-hydroxy-Rifabutin-N-oxide lie:
^-N.M.R. (CDC13) δ(ppm) : -0.16 (3H, d, J34/26= 6.8Hz, Me-
34) ; 0.53 (3H, d, J33/24= 6.8Hz, Me-33) ; 0.82 (3H, d, J3ι/2o= 6.8Hz, Me-31); 1.16 (6H, d, Jιr/ιo= Jι2'/ιo= 6.8Hz,
Me-11' e Me-12' ) ; 1.04 (3H, d, J32/22= 6.8Hz, Me-32);
1.16 (IH, m, H-24) ; 1.24 (IH, m, H-10' ) ; 1.64 (IH, m, H-
26) ; 1.73 (3H, s, Me-13) ; 1.7-1.8 (IH, m, H-22); 2.06
(3H, bs, Me-30) ; 2.28 (3H, s, Me-14); 2.36 (IH, m, H- 20) ; 3.16 (3H, s, Me-37); 3.32 (2H, m, CH2-9' ) ; 3.3-3.6 (3H, m, H-23/H-25/H-27) ; 3.73 (IH, d, J2ι/20= 9.3Hz, H-21) ; 4.20 (IH, s, 25-OH) ; 5.11 (IH, dd, J28 27= 8.7Hz/ J28 29= 12.4Hz, H-28) ; 5.91 (IH, dd, Jι9/ι8= 14.6Hz, Jι9 20= 6.2Hz, H-19); 6.22 (IH, bd, J29 28= 12.7Hz, H-29) ; 6.29-6.36 (2H, m, H-18/H-17) .
13C-N.M.R. (CDC13) δ(ppm) : 7.7 (C-14) ; 8.4 (C-33) ; 11.0 (C- 32); 11.2 (C-34); 17.4 (C-31) ; 20.2 (C-30); 22.7 (C-13) ; 23.4 (C-ll'/C-12') ; 23.6 (C-10'); 32.8 (C-22) ; 38.6 (C- 24) ; 39.1 (C-20); 56.5 (C-37); 62.8 (C-5' /C-7' ) ; 70.4 (C- 25) ; 72.0 (C-21) ; 77.3 (C-23) ; 79.2 (C-9') ; 91.6 (C-2') ; 105.1 (C-3) ; 107.8 (C-12) ; 109.2 (C-10); 111.6 (C-9); 114.7 (C-7/C-28) ; 123.9 (C-18) ; 124.8 (C-5) ; 132.2 (C- 16) ; 132.7 (C-17) ; 140.5 (C-19) ; 156.1 (C-4); 168.1 (C- 8) ; 168.3 (C-15) ; 171.2 (C-6) ; 181.8 (C-l) ; 192.6 (C-ll) . I.V. fiim (NaCl) vmέx(cm"1) : 3402 (O-H) ; 2965. 2934 (C-H) ; 1724 (Cu=0); 1672 (C15=0) ; 1602. 1568 (C=C) ; 1521 δ(N-H) ; 1422 (C8-0) e δ(O-H) ; 1374 δ(C-H); 1328 (C-N) ; 1295 δ(0- H) ; 1160 (C-N) ; 1083 (N-O); 1059 (C-O); 978 γ(C-H) ; 770 (chromophore' s vibration)
Mass/ESI (m/z) : 819 [M-H]-" ; 803 [ (M-H) -O] '"; 323 [Ch- iC4H9N(CH3)=CH2] •"] .
Example 3. Synthesis of Rifabutin-N-oxide Ila
Figure imgf000024_0001
To a round-bottomed flask containing Rifabutin la (100.0 mg; 0.118 mmol) in dichloromethane (4 mL) , was added benzyl triethylammonium chloride (40.9 mg; 0.180 mmol), sodium hydrogenocarbonate (6.7 mg; 0.080 mmol); Oxone (145.5 mg; 0.237 mmol) in acetone (1.0 mL) , distilled water (5 mL) and a Metrohm pH=7 buffer (7.0 mL) , in permanent agitation and at room temperature. The reaction was followed by t.l.c, using Petroleum ether/ethyl acetate/methanol (55:35:10) as eluant; after ca . 17 h it was progressively added 1 eq. more of Oxone (146.0 mg; 0.237 mmol) and buffer (0.5 mL) to adjust the pH of the medium. The reaction was complete after ca. 26 h 30 min. The flask content was transferred to a separatory funnel, and 3 extractions with dichloromethane were followed by 2 washings with water. The combined organic layers were collected in an Erlenmeyer, dried over anhydrous sodium sulfate, and filtered. The solvent was evaporated at reduced pressure, and the crude red coloured solid obtained was dried in vacuum (152.1 mg; η=82.4%) and purified by Flash Column Chromatography, using as eluant: Petroleum ether/ethyl acetate/methanol (55:35:10), giving Rifabutin-N-oxide Ila (94.2 mg; η=51.0%) .
Rif abutin-N-oxide Ila:
^-N.M.R. (CDC13) δ(ppm) : -0.08 (3H, d, J34/26= 7.0Hz, Me- 34); 0.61 (3H, d, J33/24=6.9Hz, Me-33) ; 0.86 (3H, d, J3ι 20= 6.9Hz, Me-31); 1.03 (3H, d, J32/22= 6.9Hz, Me-32); 1.1-1.7 (m, CH2-4' /CH2-8' ) 1.21 (6H, d, Jn-/10= Jι2'/ιo= 6.6Hz, Me- 11' e Me-12') ; 1.42 (IH, m, H-24) ; 1.68-1.82 (2H, m, H- 22/H-26) ; 1.76 (3H, s, Me-13) ; 1.98 (3H, s, Me-36) ; 2.06 (3H, s, Me-30) ; 2.35 (3H, s, Me-14) ; 2.30-2.41 (IH, m,
H-20); 2.50 (IH, m, H-10' ) ; 3.01 (1. m, H-23) ; 3.09 (3H, s, Me-37); 3.34 (IH, m, H-27); 3.40 (IH, bs, 21-OH) ; 3.48 (2H, m, CH2-9' ) ; 3.58 (IH, d, 23-OH) ; 3.69 (IH, bd, J2ι/20= 9.9Hz, H-21) ; 3.2-4.2 (m, CH2-7'/ CH2-5' ) ; 4.81 (IH, bd, J25/26=9.9Hz, H-25) ; 5.12 (IH, dd, J28 27= 7.2Hz /J28/29= 12.3Hz, H-28) ; ) ; 6.00 (IH, dd, Jι9 ι8= 15.3Hz, Jι9/20= 6.9Hz, H-19) ; 6.14 (IH, d, J29/28= 12.6Hz, H-29) ; 6.29 (IH, bd, Ji7/i8= 10.2Hz, H-17); 6.39 (IH, dd, Jι8/ι7= 10.2Hz, 15.3Hz, H-18); 8.43 (IH, s, NH) ; 8.74 (IH, s, NH' ) ; 14.54 (IH, bs, 8-OH) . 13C-N.M.R. (MeOD) δ(ppm) : 7.6 (C-14) ; 8.8 (C-33) ; 10.7 (C- 34); 11.1 (C-32) ; 17.3 (C-31); 20.3 (C-30); 21.0 (C-36) ; 21.8 (C-13) ; 23.2 (C-ll' /C-12' ) ; 23.7 (C-10' ) ; 31.6 (C- 4') ; 31.9 (C-8'); 32.9 (C-22) ; 37.6 (C-24/C-26) ; 38.4 (C- 20) ; 56.9 (C-37); 62.8 (C-5' /C-7' ) ; 72.5 (C-21) ; 73.0 (C- 25) ; 76.9 (C-23) ; 78.4 (C-9' ) ; 80.5 (C-27); 91.3 (C-2') 105.4 (C-3); 107.3 (C-12); 108.8 (C-10); 111.8 (C-9) 114.8 (C-7) ; 115.4 (C-28) ; 124.2 (C-18) ; 124.7 (C-5) 131.0 (C-16) ; 133.2 (C-17) ; 140.8 (C-19) ; 141.8 (C-2) 144.0 (C-29) ; 156.2 (C-4); 168.1 (C-8) ; 168.3 (C-15) 171.6 (C-6); 172.1 (C-35) ; 181.9(C-1) .
I.V. film (NaCl) vmόx(cm_1) : 3423 (O-H) ; 2965. 2931 (C-H) ; 1725 (Cu=0) ; 1672 (C15=0) ; 1602. 1562 (C=C) ; 1524 δ(N-H) ; 1422 (Cβ-O) e δ(O-H) ; 1376 δ(C-H) ; 1295 δ(O-H) ; 1248 (C35- 0-C25) ; 1161 (C-N); 1085 (N-O); 1062 (C-O) e (C35-0-C25) ; 976 γ(C-H) ; 770 (chromophore ' s vibration) .
Mass/ESI (m/z) : 861 [M-H]"" ; 845 [(M-H)-O]"" ; 785 [ (M-H) - O-CH3COOH] '"; 753 [ (M-H) -O-CH3COOH-CH3OH] ■"; 709 [(M-H)-O- CH3COOH-CH3OH-C3H8] •"; 651 [ (M-H) -0-CH3COOH-CH3OH-C3H8-
C3H8N]-~; 571 [ (M-3H) -O-b) ] '"; 543 [ (M-3H) - c] •"; 421 [Ch- H] •";
Exact Mass (m/z) - C46H62N4θι2 (M(-H)-O)" obtained: 846.44133 ((M-H)-O)" Example 4. Synthesis of N' -acetyl-Rifabutin Ilia
Figure imgf000027_0001
To a round-bottomed flask containing a solution of Rifabutin la (500 mg; 0.59 mmol) in tetrahydrofuran (15 mL) , triethylamine (248 μL; 1.9 mmol) was added dropwise, at 0°C, with stirring, followed by an acetyl chloride solution (129 μL; 1.9 mmol) in tetrahydrofuran (10 mL) , and the resulting mixture was left stirred till it reached room temperature. The reaction was complete 5 minutes later, the solvent removed under nitrogen flux, and the diluted mixture in ethyl acetate, filtered and evaporated to dryness in a rotary evaporator and at high vacuum, to originate N' -Acetyl-Rifabutin Ilia, as a chromatographic homogeneous orange-coloured solid in 95% yield.
N' -acetyl-Rifabutin Ilia: ^-N.M.R. (CDC13) δ(ppm): -0.07 (3H, bs, Me-34); 0.59 (3H, d, J33/24=6.0Hz, Me-33); 0.86 (3H, d, J3ι/20= 6.0Hz, Me-31); 0.99 (6H, d, JUM2 IO'= 6.3Hz, Me-11' e Me-12' ) ; 1.03 (3H, d, J32/22= 7.5 Hz, Me-32); 1.6 (IH, m, H-24); 1.6 (IH, m, H-26); 1.77 (3H, s, Me-13) ; 1.8 (IH, m, H-22) ; 1.91 (IH, , Jιi',i2Vio-= 6.4Hz, H-10' ) ; 1.7* (2H, m, CH2-8') ; 1.7*
(2H, m, CH2-4') ; 2.02 (3H, s, Me-36) ; 2.05 (3H, s, Me-30) ;
2.44 (3H, bs, Me-39); 2.4 (2H, m, CH2-9') ; 2.4 (IH, m, H- 20); 2.44 (3H, s, Me-14); 2.9* (2H, m, CH2-5') ; 2.9* (2H, m, CH2-7'); 2.99 (IH, m, H-23) ; 3.01 (IH, bs, H-21); 3.09
(3H, s, Me-37) ; 3.41 (IH, m, H-27); 3.77 (IH, bs, 23-OH)
3.87 (IH, d, 21-OH); 4.97 (IH, bd, J25 26=10.2Hz, H-25)
5.19 (IH, bs, H-28); 6.07 (IH, bd, Jι9 20= 6.3Hz, H-19) 6.09 (IH, m, H-29) ; 6.23 (IH, bd,
Figure imgf000028_0001
10.8Hz, H-17)
6.72 (IH, m, H-18); 8.21 (IH, s, NH) ; 13.89 (IH, s, 8-
OH) .
13 C-N.M.R. (CDC13) δ(ppm) : 7.36 (C-14); 8.44 (C-33) ; 9.44
(C-34) 10.78 (C-32); 17.39 (C-31) ; 20.45 (C-30); 20.71 ((CC--3366)) ; 20.87 (C-ll'); 20.87 (C-12') ; 21.22 (C-13) ; 25.24 (C-39) 25.88 (C-10'); 32.97 (C-22) ; 35.34 (C-4') ; 35.34 (C-8') 37.30 (C-24); 38.16 (C-26) ; 38.23 (C-20); 51.13* (C-7') 51.29* (C-5') ; 57.20 (C-37) ; 65.92 (C-9') ; 72.90 (C-21) 73.81 (C-25); 77.11 (C-23) ; 79.1 (C-27) ; 96.38 ((CC--22'')); 106.86 (C-3) ; 108.93 (C-12); 110.97 (C-10)
113.34 (C-9); 113.77 (C-7) ; 116.73 (C-28) ; 125.67 (C-18)
126.41 (C-5); 129.5 (C-16) ; 134.95 (C-17) ; 140.77 (C-19)
140.77 (C-2); 142.37 (C-29) ; 153.92 (C-4) ; 166.58 (C-8)
166.66 (C-38); 167.61 (C-15) ; 172.08 (C-6) ; 177.22(C-35) 186.86 (C-l); 191.97 (C-ll) .
Mass/FAB (m/z) : 888 [M]+
Example 5. Synthesis of N' -acetyl-Rifabutinol Illb
Figure imgf000029_0001
To a round-bottomed flask containing a solution of Rifabutinol lb (300 mg; 0.35 mmol) in tetrahydrofuran (9 mL) , triethylamine (149 μL; 1.08 mmol) was added dropwise, at 0°C, with stirring, followed by an acetyl chloride solution (77.1 μL; 1.08 mmol) in tetrahydrofuran (6 mL) , and the resulting mixture was left stirred till it reached room temperature. The reaction was completed 5 minutes later, the solvent removed under nitrogen flux, and the diluted mixture in ethyl acetate, filtered and evaporated to dryness in a rotary evaporator and at high vacuum, to originate the chromatographic homogeneous N'- Acetyl-Rifabutinol Illb, in a 95% yield, identified by 1ti- N.M.R., 13C-N.M.R. and Mass/FAB (m/z): 888 [M]+.
Biological activity screening tests
Microbiology Assay for the Evaluation of Mycobacterium avium 1581 Susceptibility to Rifabutin and Derivatives .
To the culture medium agar 7H10 enriched with OADC from Difco the molecules derived from Rifabutin la of this invention were added at the final concentrations 0.1. 0.2 and 0.4 μg/mL and also Rifabutin la was added at final concentrations 0.1 and 0.2 μg/mL. Plates containing Rifabutin la or its derivatives of this invention were inoculated with a strain of M. avium 1581 at dilutions 105 and 10"4. Plates without antibiotic were inoculated with a strain of M. avium at dilutions 10"7' 10"6, 10"5 e 10~4. Plates were then incubated 15 days in an oven at 37°C in the presence of 5% C02. By comparing with control plates (inoculated with the strain M. avium but in the absence of antibiotic) the concentration of the Rifabutin derivative which is related with the strain whose colonies number is smaller than 1% in relation to the colonies number in control plates. The results obtained while testing the molecules of this invention are summarized in Table 1. Rifabutin la produced an inhibition greater than 99% in the tested concentrations (0.15 and 0.2 μg/mL). Rifabutinol lb and 25-hydroxy-Rifabutin Ic showed inibitions from 86 to 91%, respectively, at a concentration 0.2 μg/mL. The N-oxide of Rifabutin Ila, and N-oxide of 25-hydroxy-rifabutin lie were inactives at the tested concentrations (0.2 and 0.4 μg/mL). N' -acetyl-rifabutinol Illb and N-oxide of Rifabutinol lib showed inhibitions between 44 and 54% at concentration 0.2 μg/mL and the derivative N'-acetyl- rifabutin Ilia presented an activity similar to Rifabutin la. Table 1- Results from assays with Myc . avium 1581
Figure imgf000031_0001
Evaluation of the activity against Gram-positive and Gram-negative bacteria MATERIALS AND METHODS
Compounds tested: N-oxide of Rifabutin Ila N-oxide of Rifabutinol lib Rifabutin la
N' -acetyl-Rifabutin Ilia N' -acetyl-rifabutinol Illb Rifampicin
Microorganisms
Staphylococcus aureus CCMI 335; Streptococcus faecalis CCMI 338. Escherichia coli CCMI 270. Pseudomonas aeruginosa CCMI 331. Salmonella enteri tidis CCMI 859.
Culture Media
S . aureus e S . faecium were incubated in brain and heart infusion (BHI) (Merck) . Escherichia coli , Pseudomonas aeruginosa and Salmonella enteri tidis were incubated in nutritive agar (Na) (Oxoid) .
General Methods Qualitative Microbiological Assays
Disc diffusion disc method was used according to the procedure described by Hong and Song, 2001 (see: Hong-Xi and Song, F. Lee, (2001) "Activity of plant flavonoids against antibiotic-resistant bacteria", Phytot er. Res . , 15. 39-43) as a diagnostic assay for antibacterial activity. Petri dishes were prepared with an adequate culture medium and containing the microorganism beeing tested at the concentration 108 cfu mL-1. The compounds were dissolved in dimethylsulfoxide (DMSO) at the concentration lmg.mL"1 and applied in Oxoid discs at the ratio 20 μg/disc After evaporation of the solvent, the compounds of the invention were applied over each Petri dish medium prepared as indicated above and incubated at 37 °C, followed by determination of the diameters of the inhibition zones 24h later. Discs containing DMSO, without any compound of the invention added, were used as control, and did not show any inhibition of colonies. Rifampicin (Sigma) was used as Reference for the evaluation of the activity of the compounds to be tested against Gram-positive and Gram-negative bacteria.
Quantitative Microbiological Assays
The Minimum Inhibitory Concentration (MIC) of the active compounds provided by the present invention was determined through the medium dilution method as described by Muroi and Kubo, 1996 (see: Muroi, H and Kubo, I, "Antibacterial activity of anacardic acid and totarol, alone and in combination with methicillin, against methicillin-resistant Staphylococcus aureus" , J. Appl . Bacteriol . 80. 387-394 (1996)). After incubation during 24h, the bacteria growth was examined and the value for the MIC defined as the minimum concentration of the tested compound, from which there is a null result after 24h incubation at 37°C. The minimum inhibitory concentration of each compound derived from Rifabutin provided by the present invention was determined at least in duplicate assays.
Bacteriostatic and Bactericide Activities
From the samples used for the MIC determination, where there was not consequently any observed bacteria growth, Petri dishes were inoculated with the strains in a BHI agar medium, and they were incubated for 24h at 37°C; after this period the plates were observed. Those where bacteria growth was observed were considered as bacteriostatic and conversely those where the bacteria growth was absent corresponded to the bactericide activities . RESULTS
The results from the evaluation of the antibacterial activity for compounds derived from Rifabutin provided this invention and tested against S. aureus, S. faecalis, E. coli, P. aeruginosa e S. enteritidis using the diffusion disc method are presented in Table 2.
Tabela 2: Antibacterial Activity of Test Compounds determined by the Method of Diffusion with Discs
Compounds S. S. E. P. S.
(lmg/mL) aureus faecalis coli aeruginosa enteretidis
Ila 22 15 10 ± 8 lib 20 13 ± - ± la 26 18 15 7 14
Ila + la 22 16 8 8 10 lib + la 23 18 8 6 10
Ila + 21 17 8 10 11
Rifampicin lib + 20 16 6 ± 9
Rifampicin
He + 24 19 10 9 13
Rifampicin
Ilia 30 16 25 16 19
Illb 25 10 - - -
Rifampicin 25 20 7 7 12
±: reduction of microorganism growth The N-oxide of Rifabutin Ila inhibited the development of Gram-positive bacteria, and also the growth of Gram-negative bacteria, E. coli e S . enteri tidis . The N-oxide of Rifabutinol lib and the N'- Acetyl-Rifabutinol Illb did not promote any inhibition of the growth of Gram-negative bacteria. The results suggested that N-oxide of Rifabutinol lib is especially active against Gram-positive bacteria. This compound did not show activity against P. aeruginosa and only partially inhibited the growth of E. coli and S . enteri tidis . N ' -Acetyl-Rifabutin Ilia as well as Rifabutin la inhibited the growth of all microorganisms tested, presenting large activity spectra. None of the mixtures of the compounds tested showed any synergistic effect.
The compounds provided by the present invention which showed any antibacterial activity using the diffusion disc method were also tested by the liquid medium method using successive dilutions, for the determination of the minimum inhibitory concentration (MIC) .
The results on the MICs for Rifabutin and derivatives are presented in Table 3. Table 3 : Determination of MICs for Rifabutin la and some derivatives
MIC (μg . mL-1 )
Compounds S. S. E. P. S. aureus faecalis coli aeruginosa enteretidis
Rifabutin
N-oxide Ila 0.1* 5* 5" n.t. 10* Rifabutinol -
N-oxide lib 0.1* 2.5* n.t. - n.t. Rifabutin la 0.003* 0.25* 1* 0.6* 1.0* N' -Acetyl- Rifabutin 0.006* 0.25* 1* 0.6* 1.0* Ilia
N'-Acetyl- rifabutinol 0.03* 1.0* n.t. - n.t. Illb
Rifampicin <0.001 0.06* 1* 2.5* 1.0* n.t.: not tested *: bactericide activity; * : bacteriostatic activity.
The N-oxide of Rifabutin Ila presented a weak activity against Gram-negative bacteria, with MIC values much higher (5 and 10 μg mL-1) for E. coli e S . enteri tidis than Rifabutin la and N ' -Acetil-Rifabutin Ilia. At the same MIC, Rifabutin la is bactericide against S . enteri tidis while N' -Acetyl-Rifabutin Ilia and rifampicin are bacteriostatic compounds. Rifabutin la and N' -Acetyl-Rifabutin Ilia showed a more efficient inhibition of the growing of P. aeruginosa than rifampicin, presenting MIC values of 6 μg mL-1. Rifampicin demonstrated to be more active against Gram-positive bacteria than Rifabutin la and N ' -Acetyl-Rifabutin Ilia. The bacteriostatic compounds are bactericides at superior concentrations.

Claims

Claims
1. A compound of general formula I,
33 32 31
Figure imgf000039_0001
wherein :
R1 is hydrogen or -COR2, where R2 is independently selected from straight, branched or cyclic Cι-C20 alkyl, saturated or unsaturated chain;
R3 is oxygen or hydroxyl;
R4 is a nitrogen atom or an N-oxide group, and their pharmaceutically acceptable salts and solvates, including hydrates.
2. A compound according to claim 1 wherein R1 is an acetyl group and R4 is a nitrogen atom.
3. A compound according to claim 1 wherein R1 is an acyl group R2 is an hydrogen or optionally an alkyl or acyl group and R4 is N-oxide.
4. A process for preparing compounds according to claims 1-3 wherein R1 is an acyl group introduced in Rifabutin la, by reaction of an acylating agent in the presence of basic catalyst.
5. A process for preparing a compound according to claims 1 and 2 wherein R1 is an acyl group introduced in Rifabutin la, by reaction of acetyl chloride in the presence of triethylamine in the temperature range between 0 and 30°C.
6. A process for preparing a compound according to claims 1 to 3 wherein R4 is introduced in Rifabutin la by reaction with a proper oxidative reagent able to selectively form an N-oxide.
7. A process for preparing a compound according to claims 1 to 3 wherein the oxidative reagent is
Oxone in buffered reaction medium.
8. A pharmaceutical composition containing as active ingredient a compound according to claims 1, 2 or 3, in a mixture with a pharmaceutical acceptable carrier.
9. The use of compounds according to claims 1, 2 or 3 in the preparation of a pharmaceutical drug directed to the treatment of infectious related to the presence of microorganisms susceptible to any compound of this invention.
10. A pharmaceutical composition to the treatment of bacterial infection in a mammal, that comprises the adminsitration to the mammal of a therapeutical effective quantity of a compound of general formula I according to claim 1 and a pharmaceutical acceptable carrier.
11. A method for the treatment of a bacterial infection in a mammal that comprises the administration of a therapeutical effective quantity of any compound according to claim 1 to 3 to the refered animal.
12. The use of compounds according to claims 1, 2, or 3 for the preparation of a composition to be administered to an animal as growth promoter.
13. The use of compounds according to claims 1 , 2 or 3, in the human Acquired Immunodeficiency Syndrome prophylaxis.
PCT/PT2003/000009 2002-07-09 2003-07-09 Derivatives of rifabutine useful as antimicrobial agents WO2004005298A1 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2003245183A AU2003245183A1 (en) 2002-07-09 2003-07-09 Derivatives of rifabutine useful as antimicrobial agents

Applications Claiming Priority (2)

Application Number Priority Date Filing Date Title
PTPT102807 2002-07-09
PT102807A PT102807A (en) 2002-07-09 2002-07-09 N-SUBSTITUTED DERIVATIVES OF USEFUL RIFABUTIN AS ANTIMICROBIAL AGENTS, PROCESS FOR PREPARING AND USING THEM AS MEDICINES

Publications (1)

Publication Number Publication Date
WO2004005298A1 true WO2004005298A1 (en) 2004-01-15

Family

ID=30113461

Family Applications (1)

Application Number Title Priority Date Filing Date
PCT/PT2003/000009 WO2004005298A1 (en) 2002-07-09 2003-07-09 Derivatives of rifabutine useful as antimicrobial agents

Country Status (3)

Country Link
AU (1) AU2003245183A1 (en)
PT (1) PT102807A (en)
WO (1) WO2004005298A1 (en)

Cited By (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2009536634A (en) * 2006-05-11 2009-10-15 アストラゼネカ アクチボラグ Novel synergistic pharmaceutical composition
WO2015127089A1 (en) * 2014-02-19 2015-08-27 The Texas A&M University System Compositions for drug sensitization of parasites
US9134329B2 (en) 2012-08-30 2015-09-15 The Texas A&M University System Compositions and methods for drug-sensitization or inhibition of a cancer cell
CN114702510A (en) * 2022-05-05 2022-07-05 重庆华邦胜凯制药有限公司 Preparation method of rifabutin oxidized impurities
WO2023198597A1 (en) 2022-04-11 2023-10-19 BioVersys AG Rifabutin analogs for the treatment of disease
WO2024076693A1 (en) * 2022-10-05 2024-04-11 Regents Of The University Of Minnesota Rifamycins for nontuberculous mycobacteria

Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4164499A (en) * 1976-09-30 1979-08-14 Archifar Laboratori Chimico Farmachologici S.p.A. Rifamycin compounds

Patent Citations (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4164499A (en) * 1976-09-30 1979-08-14 Archifar Laboratori Chimico Farmachologici S.p.A. Rifamycin compounds

Non-Patent Citations (3)

* Cited by examiner, † Cited by third party
Title
COCCHIARA G ET AL: "URINARY METABOLITES OF RIFABUTIN A NEW ANTIMYCOBACTERIAL AGENT IN HUMAN VOLUNTEERS", XENOBIOTICA, vol. 19, no. 7, 1989, pages 769 - 780, XP009020040, ISSN: 0049-8254 *
KUNIN C M: "ANTIMICROBIAL ACTIVITY OF RIFABUTIN", CLINICAL INFECTIOUS DISEASES, THE UNIVERSITY OF CHICAGO PRESS, CHICAGO, IL, US, vol. 22, no. SUPPL 1, 1 April 1996 (1996-04-01), pages S03 - S14, XP000600925, ISSN: 1058-4838 *
SANTOS ET AL.: "NMR studies of some rifamycins", J.MOL.STRUCT., vol. 563-564, 2001, pages 61 - 78, XP002260023 *

Cited By (10)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
JP2009536634A (en) * 2006-05-11 2009-10-15 アストラゼネカ アクチボラグ Novel synergistic pharmaceutical composition
US9134329B2 (en) 2012-08-30 2015-09-15 The Texas A&M University System Compositions and methods for drug-sensitization or inhibition of a cancer cell
US9539237B2 (en) 2012-08-30 2017-01-10 The Texas A&M University System Compositions and methods for drug-sensitization or inhibition of a cancer cell
US9867807B2 (en) 2012-08-30 2018-01-16 The Texas A&M University System Compositions and methods for drug-sensitization or inhibition of a cancer cell
EP3560497A1 (en) * 2012-08-30 2019-10-30 The Texas A&M University System Compositions and methods for drug-sensitization or inhibition of a cancer cell
WO2015127089A1 (en) * 2014-02-19 2015-08-27 The Texas A&M University System Compositions for drug sensitization of parasites
WO2023198597A1 (en) 2022-04-11 2023-10-19 BioVersys AG Rifabutin analogs for the treatment of disease
CN114702510A (en) * 2022-05-05 2022-07-05 重庆华邦胜凯制药有限公司 Preparation method of rifabutin oxidized impurities
CN114702510B (en) * 2022-05-05 2023-06-30 重庆华邦胜凯制药有限公司 Preparation method of rifabutin oxidation impurity
WO2024076693A1 (en) * 2022-10-05 2024-04-11 Regents Of The University Of Minnesota Rifamycins for nontuberculous mycobacteria

Also Published As

Publication number Publication date
PT102807A (en) 2004-01-30
AU2003245183A1 (en) 2004-01-23

Similar Documents

Publication Publication Date Title
TWI780304B (en) Modulators of cystic fibrosis transmembrane conductance regulator, pharmaceutical compositions, methods of treatment, and process for making the modulators
Ignacimuthu et al. Antimycobacterial activity of two natural alkaloids, vasicine acetate and 2-acetyl benzylamine, isolated from Indian shrub Adhatoda vasica Ness. leaves
Hellwig et al. Altersetin, a new antibiotic from cultures of endophytic Alternaria spp. Taxonomy, fermentation, isolation, structure elucidation and biological activities
Hichri et al. Antibacterial activities of a few prepared derivatives of oleanolic acid and of other natural triterpenic compounds
FUJITA et al. Fungal metabolites. Part 12. Potent immunosuppressant, 14-deoxomyriogin,(2s, 3r, 4r-(e) 2-amino-3, 4-dihydroxy-2-hydroxymethyleicos-6-enoic acid and structure-activity relationships of myriocin derivatives
GOMI et al. Isolation and structure of a new antibiotic, indolizomycin, produced by a strain SK2-52 obtained by interspecies fusion treatment
Akhtar et al. Proceragenin, an antibacterial cardenolide from Calotropis procera
WO2017059411A1 (en) Inhibitors of menaquinone biosynthesis
EP1720886A1 (en) Macrolides and methods for producing same
Yazawa et al. Inactivation of rifampin by Nocardia brasiliensis
HUT70207A (en) 21-norrapamycin, pharmaceutical compositions containing it and process for preparing them
CN104418864A (en) Conjugates of dihydroartemisinin and quinolones compounds as well as preparation method and application thereof
WO2005084673A1 (en) Non-immunosuppressive immunophilin ligands as neuroprotective and/or neuroregenerative agents
Marchi et al. 4-Deoxypyrido [1', 2': 1, 2] imidazo [5, 4-c] rifamycin SV derivatives. A new series of semisynthetic rifamycins with high antibacterial activity and low gastroenteric absorption
HRP20110437A2 (en) Rifamycin derivatives
Lasisi et al. New triterpene isovanniloyl and antibacterial activity of constituents from the roots of Paullinia pinnata Linn (Sapindaceae)
WO2004005298A1 (en) Derivatives of rifabutine useful as antimicrobial agents
AU2015331904B2 (en) Scalable synthesis of reduced toxicity derivative of amphotericin B
Figueiredo et al. Synthesis and evaluation of rifabutin analogs against Mycobacterium avium and H37Rv, MDR and NRP Mycobacterium tuberculosis
Samoylenko et al. Bioactive (+)-manzamine A and (+)-8-hydroxymanzamine A tertiary bases and salts from Acanthostrongylophora ingens and their preparations
KR20170070197A (en) Nargenicin compounds and uses thereof as antibacterial agents
Bujnowski et al. Rifamycin antibiotics—new compounds and synthetic methods. Part 1: Study of the reaction of 3-formylrifamycin SV with primary alkylamines or ammonia
Masood-ur-Rahman et al. Synthesis and antimicrobial activity of triazolyl analogs of diosgenin
Wang et al. 31-Homorifamycin W, a novel metabolite from Amycolatopsis mediterranei
Dinda et al. Chemical constituents of Evolvulus nummularius

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A1

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BR BY BZ CA CH CN CO CR CU CZ DE DK DM DZ EC EE ES FI GB GD GE GH GM HR HU ID IL IN IS JP KE KG KP KR KZ LC LK LR LS LT LU LV MA MD MG MK MN MW MX MZ NI NO NZ OM PG PH PL PT RO RU SC SD SE SG SK SL SY TJ TM TN TR TT TZ UA UG US UZ VC VN YU ZA ZM ZW

AL Designated countries for regional patents

Kind code of ref document: A1

Designated state(s): GH GM KE LS MW MZ SD SL SZ TZ UG ZM ZW AM AZ BY KG KZ MD RU TJ TM AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IT LU MC NL PT RO SE SI SK TR BF BJ CF CG CI CM GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP