Substituted 1,2,3-triazol-1-yl-methyl-2,3-dihydro-2-methyl-6-nitroimidazo[2,1-b]oxazo- Les As Anti-mycobacterial Agents And A Process For The Preparation Thereof

YEMPALLA; Kushalava Reddy ;   et al.

Patent Application Summary

U.S. patent application number 15/520799 was filed with the patent office on 2017-11-02 for substituted 1,2,3-triazol-1-yl-methyl-2,3-dihydro-2-methyl-6-nitroimidazo[2,1-b]oxazo- les as anti-mycobacterial agents and a process for the preparation thereof. This patent application is currently assigned to COUNCIL OF SCIENTIFIC & INDUSTRIAL RESEARCH. The applicant listed for this patent is COUNCIL OF SCIENTIFIC & INDUSTRIAL RESEARCH. Invention is credited to Inshad Ali KHAN, Gurunadham MUNAGALA, Sumit SHARMA, Parvinder Pal SINGH, Samsher SINGH, Ram Asrey VISHWAKARMA, Kushalava Reddy YEMPALLA.

Application Number20170313721 15/520799
Document ID /
Family ID54361129
Filed Date2017-11-02

United States Patent Application 20170313721
Kind Code A1
YEMPALLA; Kushalava Reddy ;   et al. November 2, 2017

SUBSTITUTED 1,2,3-TRIAZOL-1-YL-METHYL-2,3-DIHYDRO-2-METHYL-6-NITROIMIDAZO[2,1-B]OXAZO- LES AS ANTI-MYCOBACTERIAL AGENTS AND A PROCESS FOR THE PREPARATION THEREOF

Abstract

The present invention relates to new generation of triazole functionality containing 6-nitro-2,3-dihydroimidazooxazole agents for general formula I, their method of preparation and their use as drugs for treatment of tuberculosis, MDR-TB and XDR-TB either alone or in combination with other anti-tubercular agents. In general formula 1, X is selected from a group (CH.sub.2).sub.n or a direct bond, where n is any number from 1-6, Y is selected from O, S or direct bond, R.sub.1 is selected from the group consisting of H, alkyl, substituted alkyl, aryl, substituted aryl, biaryl, substituted biaryl, heterocyclic and substituted heterocyclic, wherein the substituted heterocyclic is selected from any of the following rings consisting of piperazinyl, morpholinyl, piperidyl, pyridyl, triazolyl, triazinyl, pyrimidinyl, pyridazinyl, oxazolyl, furanyl, benzofuranyl, thiophenyl, pyrrolyl, imidazoyl, thiazoyl, quinolinyl, isoquinolinyl, benzooxazolyl and benzothiazolyl and the substitution on aryl and biaryl is selected from the group consisting of F, CI, Br, I, CF.sub.3, OCF.sub.3, OR.sub.11, NO.sub.2 and alkyl chain from C1 to C14, wherein R.sub.11 is selected from the group consisting of H, alkyl, phenyl and substituted phenyl. ##STR00001##


Inventors: YEMPALLA; Kushalava Reddy; (Jammu, IN) ; MUNAGALA; Gurunadham; (Jammu, IN) ; SINGH; Samsher; (Jammu, IN) ; SHARMA; Sumit; (Jammu, IN) ; KHAN; Inshad Ali; (Jammu, IN) ; VISHWAKARMA; Ram Asrey; (Jammu, IN) ; SINGH; Parvinder Pal; (Jammu, IN)
Applicant:
Name City State Country Type

COUNCIL OF SCIENTIFIC & INDUSTRIAL RESEARCH

New Delhi

IN
Assignee: COUNCIL OF SCIENTIFIC & INDUSTRIAL RESEARCH
New Delhi
IN

Family ID: 54361129
Appl. No.: 15/520799
Filed: September 9, 2015
PCT Filed: September 9, 2015
PCT NO: PCT/IN2015/050111
371 Date: April 20, 2017

Current U.S. Class: 1/1
Current CPC Class: C07D 498/04 20130101; A61P 31/06 20180101
International Class: C07D 498/04 20060101 C07D498/04

Foreign Application Data

Date Code Application Number
Oct 21, 2014 IN 3009/DEL/2014

Claims



1. A compound of general formula I, ##STR00056## wherein `X` is selected from a group (CH.sub.2).sub.n or a direct bond, where n is any number from 0, 1, 2 to 6, `Y` is selected from a group O, S or direct bond, R.sub.I is selected from the group consisting of H, alkyl, substituted alkyl, aryl, substituted aryl, biaryl, substituted biaryl, heterocyclic and substituted heterocyclic, wherein the substituted heterocyclic is selected from any of the following rings consisting of piperazinyl, morpholinyl, piperidyl, pyridyl, triazolyl, triazinyl, pyrimidinyl, pyridazinyl, oxazolyl, furanyl, benzofuranyl, thiophenyl, pyrrolyl, imidazoyl, thiazoyl, quinolinyl, isoquinolinyl, benzooxazolyl and benzothiazolyl and the substitution on aryl and biaryl is selected from the group consisting of F, Cl, Br, I, CF.sub.3, OCF.sub.3, OR.sub.I1, NO.sub.2 and alkyl chain from C1 to C14, wherein R.sub.I1 is selected from the group consisting of H, alkyl, phenyl, substituted phenyl.

2. The compound of formula I as claimed in claim 1, wherein the compound is selected from the group consisting of the following compounds: (R)-2-{[4-(4-trifluoromethoxyphenyl)-1H-1,2,3-triazol-1-yl]methyl}-2,3-di- hydro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.1) (R)-2-{[4-(4-trifluoromethylphenyl)-1H-1,2,3-triazol-1-yl]methyl}-2,3-dih- ydro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.2) (R)-2-{[4-(4-fluorophenyl)-1H-1,2,3-triazol-1-yl]methyl}-2,3-dihydro-2-me- thyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.3) (R)-2-{[4-(4-fluoro-3-methylphenyl)-1H-1,2,3-triazol-1-yl]methyl}-2,3-dih- ydro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.4) (R)-2-{[4-(2,4-difluorophenyl)-1H-1,2,3-triazol-1-yl]methyl}-2,3-dihydro-- 2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.5) (R)-2-{[4-(4-phenoxyphenyl)-1H-1,2,3-triazol-1-yl]methyl}-2,3-dihydro-2-m- ethyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.6) (R)-2-[(4-pentyl-1H-1,2,3-triazol-1-yl)methyl]-2,3-dihydro-2-methyl-6-nit- roimidazo[2,1-b]oxazole (compound I.sub.7) (R)-2-{[4-(4-trifluoromethoxyphenoxy)methyl)-1H-1,2,3-triazol-1-yl]methyl- }-2,3-dihydro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.8) (R)-2-{[4-(4-trifluoromethylphenoxy)methyl)-1H-1,2,3-triazol-1-yl]methyl}- -2,3-dihydro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.9) (R)-2-{[4-(3-chlorophenoxy)methyl)-1H-1,2,3-triazol-1-yl]methyl}-2,3-dihy- dro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.10) (R)-2-{[4-(4-bromophenoxy)methyl)-1H-1,2,3-triazol-1-yl]methyl}-2,3-dihyd- ro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.11) (R)-2-{[4-(4-methylphenoxy)methyl)-1H-1,2,3-triazol-1-yl]methyl}-2,3-dihy- dro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.12) (R)-2-{[4-(3-methylphenoxy)methyl)-1H-1,2,3-triazol-1-yl]methyl}-2,3-dihy- dro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.13) (R)-2-{[4-(2-methylphenoxy)methyl)-1H-1,2,3-triazol-1-yl]methyl}-2,3-dihy- dro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.14) (R)-2-{[4-(4-ethylphenoxy)methyl)-1H-1,2,3-triazol-1-yl]methyl}-2,3-dihyd- ro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.15) (R)-2-{[4-(3-fluorophenoxy)methyl)-1H-1,2,3-triazol-1-yl]methyl}-2,3-dihy- dro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.16) (R)-2-{[4-(2-fluorophenoxy)methyl)-1H-1,2,3-triazol-1-yl]methyl}-2,3-dihy- dro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.17) (R)-2-{[4-(4-isopropylphenoxy)methyl)-1H-1,2,3-triazol-1-yl]methyl}-2,3-d- ihydro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.18) (R)-2-methyl-6-nitro-2-((4-((pyridin-2-yloxy)methyl)-1H-1,2,3-triazol-1-y- l)methyl)-2,3-dihydroimidazo[2,1-b]oxazole (compound I.sub.19) (R)-2-methyl-6-nitro-2-((4-((p-tolylthio)methyl)-1H-1,2,3-triazol-1-yl)me- thyl)-2,3-dihydroimidazo[2,1-b]oxazole (compound I.sub.20) (R)-2-methyl-6-nitro-2-((4-(2-(p-tolyloxy)ethyl)-1H-1,2,3-triazol-1-yl)me- thyl)-2,3-dihydroimidazo[2,1-b]oxazole (compound I.sub.21) (R)-2-methyl-2-((4-(morpholinomethyl)-1H-1,2,3-triazol-1-yl)methyl)-6-nit- ro-2,3-dihydroimidazo[2,1-b]oxazole (compound I.sub.22)

3. The compound of general formula I as claimed in claim 1, for use in the treatment of tuberculosis.

4. The compound of formula I as claimed in claim 1, wherein said compound exhibits an in vitro anti-tuberculosis activity against replicating and non-replicating stages of Mycobacterium tuberculosis with MIC values in the range of 0.12 to 32 .mu.g/ml.

5. The compound of formula I as claimed in claim 1, wherein said compound exhibits an in vitro anti-tuberculosis activity against XDR Mycobacterium tuberculosis (resistant to isoniazid, rifampicin, amikacin and moxifloxacin), MDR-TB (resistant to isoniazid and rifampicin) with MIC values in the range of 0.12 to 32 .mu.g/ml and the compound does not exhibit any cytotoxicity up to 40 .mu.g/ml in macrophage J774 cell line.

6. A process for preparation of the compound of formula I as claimed in claim 1 wherein the said process comprising the steps: i) reacting a compound of formula 8 in an organic solvent and in the presence of an azide source at a temperature in the range of 25.degree. C. to 80.degree. C. for a period ranging between 1 h to 3 h to obtain a compound of formula 9; ##STR00057## ii) reacting the compound of formula 9 in an organic solvent and in the presence of a base at a temperature in the range of 10.degree. C.-25.degree. C. for a period of 1 h to 12 h to obtain a compound of formula 10; ##STR00058## iii) reacting the compound of formula 10 with a compound of formula selected from the group consisting of formula 13 (a-k) or 14 (a-g) or 15 (a-d) in 1:1 tert-BuOH/H.sub.2O mixture in the presence of sodiumascorbate and CuSO.sub.4 at a temperature in the range of 10.degree. C. to 25.degree. C. for a period of 1 h to 12 h to obtain the compound of formula I. ##STR00059##

7. The process as claimed in step i) of claim 6, wherein the organic solvent is selected from toluene, acetonitrile, DMF, dichloroethane or any combination thereof.

8. The process as claimed in step i) of claim 6, wherein the azide source is selected from sodiumazide, trimethylsilylazide, tetrabutyl ammonium bromide or any combination thereof.

9. The process as claimed in step ii) of claim 6, wherein the base is selected from sodium hydride, cesium carbonate, potassium carbonate or any combination thereof.

10. The process as claimed in step ii) of claim 6, wherein the organic solvent is selected from toluene, acetonitrile, DMF, tetrahydrofuran or any combination thereof.

11. The compound of formula I as claimed in claim 2, wherein said compound exhibits an in vitro anti-tuberculosis activity against replicating and non-replicating stages of Mycobacterium tuberculosis with MIC values in the range of 0.12 to 32 .mu.g/ml.

12. The compound of formula I as claimed in claim 2, wherein said compound exhibits an in vitro anti-tuberculosis activity against XDR Mycobacterium tuberculosis (resistant to isoniazid, rifampicin, amikacin and moxifloxacin), MDR-TB (resistant to isoniazid and rifampicin) with MIC values in the range of 0.12 to 32 .mu.g/ml and the compound does not exhibit any cytotoxicity up to 40 .mu.g/ml in macrophage J774 cell line.
Description



FIELD OF THE INVENTION

[0001] The present invention relates to substituted 1,2,3-triazol-1-yl-methyl-2,3-dihydro-2-methyl-6-nitroimidazo[2,1-b]oxazo- lesas anti-mycobacterial agents. The present invention particularly relates to the compounds of 6-nitro-2,3-dihydroimidazooxazole scaffold that have been designed, synthesized and their biological evaluation result for anti-tuberculosis are presented. The present invention relates to novel compounds of general formula I, their method of preparations, and their use as drugs for treatment of tuberculosis.

BACKGROUND OF THE INVENTION

[0002] Tuberculosis remains a leading infectious cause of death worldwide and infects about one-third of the world's population. The World Health Organization (WHO) has estimated that if the present conditions remain unchanged, more than 30 million lives will be claimed by TB between 2000 and 2020. In 2012, an estimated 8.6 million people developed TB and 1.3 million died from the disease (including 320 000 deaths among HIV-positive people). TB has also been declared as a global health emergency because of the increase in secondary infections and/or co-infection in cancer and immunocompromised patients (such as those infected with human immunodeficiency virus). The existing lengthy TB therapy and emergence of multidrug resistant TB (MDR-TB) and extensively drug resistant TB (XDR-TB), [BemerMelchior, P.; Bryskier, A.; Drugeon, H. B. J. Antimicrob. Chemother. 2000, 46, 571; Abubaker, J.; Schraufnagel, D. J. Am. Med. Assoc. 2000, 283, 54; Dye. C.; Scheele, S.; Dolin, P.; Pathania, V.; Raviglione, M. C. J. Am. Med. Assoc. 1999, 282, 677] necessitates the development of new and potent anti-tuberculosis agents.

[0003] In this direction, we have initiated a medicinal chemistry programme on 6-nitro-2,3-dihydroimidazooxazole scaffold and discovered new potent structures (2954/DEL/2013) and in continuation, in the present invention, new generation triazole functionality containing 6-nitro-2, 3-dihydroimidazooxazole is synthesized and screened for anti-TB activity.

OBJECTIVE OF THE INVENTION

[0004] The main objective of the present invention is to provide new generation triazole functionality containing 6-nitro-2, 3-dihydroimidazooxazoles agents for treatment of tuberculosis.

[0005] Still another object of the present invention is to provide a process for the preparation of triazole functionality containing 6-nitro-2, 3-dihydroimidazooxazoles.

[0006] Still another object of the present invention is to provide treatment against multi-drug resistant (MDR) and extensive drug resistant (XDR) tuberculosis.

SUMMARY OF THE INVENTION

[0007] The present invention relates to new generation of triazole functionality containing 6-nitro-2, 3-dihydroimidazooxazole agents, their method of preparation and their use as drugs for treatment of tuberculosis.

[0008] Accordingly the present invention provides a compound having a general structure of formula I,

##STR00002##

[0009] wherein, [0010] `X` is selected from a group (CH.sub.2).sub.n or a direct bond, where n is any number from 0, 1, 2 to 6, [0011] `Y` is selected from a group O, S or direct bond, [0012] R.sub.I is selected from the group consisting of H, alkyl, substituted alkyl, aryl, substituted aryl, biaryl, substituted biaryl, heterocyclic and substituted heterocyclic, wherein the substituted heterocyclic is selected from any of the following rings consisting of piperazinyl, morpholinyl, piperidyl, pyridyl, triazolyl, triazinyl, pyrimidinyl, pyridazinyl, oxazolyl, furanyl, benzofuranyl, thiophenyl, pyrrolyl, imidazoyl, thiazoyl, quinolinyl, isoquinolinyl, benzooxazolyl and benzothiazolyl and the substitution on aryl and biaryl is selected from the group consisting of F, Cl, Br, I, CF.sub.3, OCF.sub.3, OR.sub.I1, NO.sub.2 and alkyl chain from C1 to C14. [0013] wherein R.sub.I1 is selected from the group consisting of H, alkyl, phenyl, substituted phenyl. [0014] In an embodiment of the present invention, the representative compound of general formula I comprising: [0015] (R)-2-{[4-(4-trifluoromethoxyphenyl)-1H-1,2,3-triazol-1-yl] methyl}-2,3-dihydro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.1, Table 1) [0016] (R)-2-{[4-(4-trifluoromethylphenyl)-1H-1,2,3-triazol-1-yl] methyl}-2,3-dihydro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.2, Table 1) [0017] (R)-2-{[4-(4-fluorophenyl)-1H-1,2,3-triazol-1-yl]methyl}-2,3-dihydro-2-me- thyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.3, Table 1) [0018] (R)-2-{[4-(4-fluoro-3-methylphenyl)-1H-1,2,3-triazol-1-yl]methyl}-2,3-dih- ydro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.4, Table 1) [0019] (R)-2-{[4-(2,4-difluorophenyl)-1H-1,2,3-triazol-1-yl]methyl}-2,3-d- ihydro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.5, Table 1) [0020] (R)-2-{[4-(4-phenoxyphenyl)-1H-1,2,3-triazol-1-yl]methyl}-2,3-dihy- dro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.6, Table 1) [0021] (R)-2-[(4-pentyl-1H-1,2,3-triazol-1-yl)methyl]-2,3-dihydro-2-methy- l-6-nitroimidazo[2,1-b]oxazole (compound I.sub.7, Table 1) [0022] (R)-2-{[4-(4-trifluoromethoxyphenoxy)methyl)-1H-1,2,3-triazol-1-yl]methyl- }-2,3-dihydro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.8, Table 1) [0023] (R)-2-{[4-(4-trifluoromethylphenoxy)methyl)-1H-1,2,3-triazol-1-yl]methyl}- -2,3-dihydro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.9, Table 1) [0024] (R)-2-{[4-(3-chlorophenoxy)methyl)-1H-1,2,3-triazol-1-yl]methyl}-2,3-dihy- dro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.10, Table 1) [0025] (R)-2-{[4-(4-bromophenoxy)methyl)-1H-1,2,3-triazol-1-yl]methyl}-2,- 3-dihydro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.11, Table 1) [0026] (R)-2-{[4-(4-methylphenoxy)methyl)-1H-1,2,3-triazol-1-yl]methyl- }-2,3-dihydro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.12, Table 1) [0027] (R)-2-{[4-(3-methylphenoxy)methyl)-1H-1,2,3-triazol-1-yl]methyl}-2,3-dihy- dro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.13, Table 1) [0028] (R)-2-{[4-(2-methylphenoxy)methyl)-1H-1,2,3-triazol-1-yl]methyl}-2- ,3-dihydro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.14, Table 1) [0029] (R)-2-{[4-(4-ethylphenoxy)methyl)-1H-1,2,3-triazol-1-yl]methyl}- -2,3-dihydro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.15, Table 1) [0030] (R)-2-{[4-(3-fluorophenoxy)methyl)-1H-1,2,3-triazol-1-yl]methyl}-2,3-dihy- dro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.16, Table 1) [0031] (R)-2-{[4-(2-fluorophenoxy)methyl)-1H-1,2,3-triazol-1-yl]methyl}-2- ,3-dihydro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.17, Table 1) [0032] (R)-2-{[4-(4-isopropylphenoxy)methyl)-1H-1,2,3-triazol-1-yl]met- hyl}-2,3-dihydro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.18, Table 1) [0033] (R)-2-methyl-6-nitro-2-((4-((pyridin-2-yloxy)methyl)-1H-1,2,3-triazol-1-y- l)methyl)-2,3-dihydroimidazo[2,1-b]oxazole (compound I.sub.19, Table 1) [0034] (R)-2-methyl-6-nitro-2-((4-((p-tolylthio)methyl)-1H-1,2,3-triazol-- 1-yl)methyl)-2,3-dihydroimidazo[2,1-b]oxazole (compound I.sub.20, Table 1) [0035] (R)-2-methyl-6-nitro-2-((4-(2-(p-tolyloxy)ethyl)-1H-1,2,3-triazol-- 1-yl)methyl)-2,3-dihydroimidazo[2,1-b]oxazole (compound I.sub.21, Table 1) [0036] (R)-2-methyl-2-((4-(morpholinomethyl)-1H-1,2,3-triazol-1-yl)methyl- )-6-nitro-2,3-dihydroimidazo[2,1-b]oxazole (compound I.sub.22, Table 1)

[0037] In an embodiment of the invention wherein the compound of general formula I, for use in treatment of tuberculosis.

[0038] In yet another embodiment of the invention of general formula I, wherein said compound exhibits an in-vitro anti-tuberculosis activity against H.sub.37Rv Mycobacterium tuberculosis, MDR-TB (resistant to isoniazid and rifampicin) with MIC values in the range of 0.12 to 32 .mu.g/ml.

[0039] In still another embodiment of the invention of general formula I, wherein said compound exhibits an in vitro anti-tuberculosis activity against XDR-TB (resistant to isoniazid, rifampicin, amikacin and moxifloxacin) with MIC values in the range of 0.12 to 32 (.mu.g/ml).

[0040] In another embodiment of the invention of general formula I, wherein said compound does not exhibit any cytotoxicity upto 40 .mu.g/ml in macrophage J774 cell line.

[0041] The compound of formula I exhibits an in vitro anti-tuberculosis activity against replicating and non-replicating stages of Mycobacterium tuberculosis with MIC values in the range of 0.12 to 32 .mu.g/ml.

[0042] The compound of formula I exhibits an in vitro anti-tuberculosis activity against XDR Mycobacterium tuberculosis (resistant to isoniazid, rifampicin, amikacin and moxifloxacin), MDR-TB (resistant to isoniazid and rifampicin) with MIC values in the range of 0.12 to 32 .mu.g/ml and the compound does not exhibit any cytotoxicity upto 40 .mu.g/ml in macrophage J774 cell line.

[0043] The present invention also provides a process for preparation of the compound of formula I wherein the said process comprising the steps: [0044] i) reacting a compound of formula 8 in an organic solvent selected from group consisting of toluene, acetonotrile, DMF, dichloroethane or any combination thereof in the presence of an azide source selected from sodiumazide, trimethylsilylazide and tetrabutyl ammonium bromide at a temperature in the range of 25.degree. C. to 80.degree. C. for a period ranging between 1 h to 3 h to obtain the desired compound of formula 9.

[0044] ##STR00003## [0045] ii) reacting the compound of formula 9 with a base selected from a group consisting of sodium hydride, cesium carbonate, potassium carbonate or any combination thereof in an organic solvent selected from a group consisting of toluene, acetonotrile, DMF, tetrahydrofuran or any combination thereof in the presence of at a temperature in the range of 10.degree. C.-25.degree. C. for a period of 1 h to 12 h to obtain a desired compound of formula 10.

[0045] ##STR00004## [0046] iii) reacting the compound of formula 10 with a compound of formula selected from the group consisting of formula 13(a-k) or 14(a-g) or 15(a-d) in 1:1 tert-BuOH/H.sub.2O mixture in the presence of sodiumascorbate and CuSO.sub.4 at a temperature in the range of 10.degree. C. to 25.degree. C. for a period of 1 h to 12 h to obtain the desired compound of formula I.

##STR00005##

[0046] List of Abbreviations:

[0047] ATCC: american type culture collection [0048] AcOH: acetic acid [0049] CFU: colony forming units [0050] DMAP: 4-dimethylaminopyridine [0051] DCM: dichloromethane [0052] d: doublet [0053] dd: doublet of doublet [0054] Et: ethyl [0055] ESI: electron spray ionisation [0056] FCS: fetal calf serum [0057] H.sub.37Rv: a well characterised virulent strain of Mycobacterium tuberculosis [0058] h: hours [0059] IC.sub.50: half maximal inhibitory concentration [0060] J: coupling constant [0061] MIC: minimum inhibitory concentration [0062] MS: mass spectrometry [0063] ml: millilitre [0064] MHz: mega hertz [0065] m: multiplet [0066] MDR-TB: Multi drug resistant tuberculosis [0067] Me: methyl [0068] min: minutes [0069] m/z: mass to charge ratio [0070] MTB: Mycobacterium tuberculosis [0071] NMP: N-methylpyrrolidinone [0072] Rif.sup.R: rifampicin resistant tuberculosis [0073] RPMI: rosewell park memorial institute medium [0074] R.sub.f: retention factor [0075] s: singlet [0076] TFA: trifluoroacetic acid [0077] TLC: thin layer chromatography [0078] TB: Tuberculosis [0079] TDR-TB: Total drug resistant tuberculosis [0080] t: triplet [0081] tert: tertiary [0082] WHO: world health organization [0083] XDR-TB: Extensive drug resistant tuberculosis [0084] .mu.g: microgram [0085] .sup.1H NMR: proton nuclear magnetic resonance

DETAILED DESCRIPTION OF THE INVENTION

[0086] The present invention relates to new generation of triazole functionality containing 6-nitro-2, 3-dihydroimidazooxazole agents, their method of preparation and their use as drugs for treatment of tuberculosis.

[0087] The present invention describes a compound having general structure of formula I

##STR00006##

[0088] wherein [0089] `X` is selected from a group (CH.sub.2).sub.n or a direct bond, where n is any number from 0, 1, 2 to 6, [0090] `Y` is selected from a group O, S or direct bond, [0091] RI is selected from the group consisting of H, alkyl, substituted alkyl, aryl, substituted aryl, biaryl, substituted biaryl, heterocyclic and substituted heterocyclic, wherein the substituted heterocyclic is selected from any of the following rings consisting of piperazinyl, morpholinyl, piperidyl, pyridyl, triazolyl, triazinyl, pyrimidinyl, pyridazinyl, oxazolyl, furanyl, benzofuranyl, thiophenyl, pyrrolyl, imidazoyl, thiazoyl, quinolinyl, isoquinolinyl, benzooxazolyl and benzothiazolyl and the substitution on aryl and biaryl is selected from the group consisting of F, Cl, Br, I, CF3, OCF3, ORI1, NO2 and alkyl chain from C1 to C14. [0092] wherein R.sub.I1 is selected from the group consisting of H, alkyl, phenyl, substituted phenyl.

[0093] The most preferred compounds of general formula I are: [0094] (R)-2-{[4-(4-trifluoromethoxyphenyl)-1H-1,2,3-triazol-1-yl]methyl}-2,3-di- hydro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.1, Table 1) [0095] (R)-2-{[4-(4-trifluoromethylphenyl)-1H-1,2,3-triazol-1-yl]methyl}-- 2,3-dihydro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.2, Table 1) [0096] (R)-2-{[4-(4-fluorophenyl)-1H-1,2,3-triazol-1-yl]methyl}-2,3-di- hydro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.3, Table 1) [0097] (R)-2-{[4-(4-fluoro-3-methylphenyl)-1H-1,2,3-triazol-1-yl]methyl}-- 2,3-dihydro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.4, Table 1) [0098] (R)-2-{[4-(2,4-difluorophenyl)-1H-1,2,3-triazol-1-yl]methyl}-2,- 3-dihydro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.5, Table 1) [0099] (R)-2-{[4-(4-phenoxyphenyl)-1H-1,2,3-triazol-1-yl]methyl}-2,3-d- ihydro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.6, Table 1) [0100] (R)-2-[(4-pentyl-1H-1,2,3-triazol-1-yl)methyl]-2,3-dihydro-2-methy- l-6-nitroimidazo[2,1-b]oxazole (compound I.sub.7, Table 1) [0101] (R)-2-{[4-(4-trifluoromethoxyphenoxy)methyl)-1H-1,2,3-triazol-1-yl]methyl- }-2,3-dihydro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.8, Table 1) [0102] (R)-2-{[4-(4-trifluoromethylphenoxy)methyl)-1H-1,2,3-triazol-1-yl]methyl}- -2,3-dihydro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.9, Table 1) [0103] (R)-2-{[4-(3-chlorophenoxy)methyl)-1H-1,2,3-triazol-1-yl]methyl}-2,3-dihy- dro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.10, Table 1) [0104] (R)-2-{[4-(4-bromophenoxy)methyl)-1H-1,2,3-triazol-1-yl]methyl}-2,- 3-dihydro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.ii, Table 1) [0105] (R)-2-{[4-(4-methylphenoxy)methyl)-1H-1,2,3-triazol-1-yl]methyl- }-2,3-dihydro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.12, Table 1) [0106] (R)-2-{[4-(3-methylphenoxy)methyl)-1H-1,2,3-triazol-1-yl]methyl}-2,3-dihy- dro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.13, Table 1) [0107] (R)-2-{[4-(2-methylphenoxy)methyl)-1H-1,2,3-triazol-1-yl]methyl}-2- ,3-dihydro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.14, Table 1) [0108] (R)-2-{[4-(4-ethylphenoxy)methyl)-1H-1,2,3-triazol-1-yl]methyl}- -2,3-dihydro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.15, Table 1) [0109] (R)-2-{[4-(3-fluorophenoxy)methyl)-1H-1,2,3-triazol-1-yl]methyl}-2,3-dihy- dro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.16, Table 1) [0110] (R)-2-{[4-(2-fluorophenoxy)methyl)-1H-1,2,3-triazol-1-yl]methyl}-2- ,3-dihydro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.17, Table 1) [0111] (R)-2-{[4-(4-isopropylphenoxy)methyl)-1H-1,2,3-triazol-1-yl]met- hyl}-2,3-dihydro-2-methyl-6-nitroimidazo[2,1-b]oxazole (compound I.sub.18, Table 1) [0112] (R)-2-methyl-6-nitro-2-((4-((pyridin-2-yloxy)methyl)-1H-1,2,3-triazol-1-y- l)methyl)-2,3-dihydroimidazo[2,1-b]oxazole (compound I.sub.19, Table 1) [0113] (R)-2-methyl-6-nitro-2-((4-((p-tolylthio)methyl)-1H-1,2,3-triazol-- 1-yl)methyl)-2,3-dihydroimidazo[2,1-b]oxazole (compound I.sub.20, Table 1) [0114] (R)-2-methyl-6-nitro-2-((4-(2-(p-tolyloxy)ethyl)-1H-1,2,3-triazol-- 1-yl)methyl)-2,3-dihydroimidazo[2,1-b]oxazole (compound I.sub.21, Table 1) [0115] (R)-2-methyl-2-((4-(morpholinomethyl)-1H-1,2,3-triazol-1-yl)methyl- )-6-nitro-2,3-dihydroimidazo[2,1-b]oxazole (compound I.sub.22, Table 1)

[0116] The compounds of general formula I, are useful in treatment of tuberculosis. The compound of general formula I, exhibits an in vitro anti-tuberculosis activity against H.sub.37Rv Mycobacterium tuberculosis, MDR-TB (resistant to isoniazid and rifampicin) with MIC values in the range of 0.12 to 32 .mu.g/ml.

[0117] The compound of general formula I, exhibits an in vitro anti-tuberculosis activity against XDR-TB (resistant to isoniazid, rifampicin, amikacin and moxifloxacin) with MIC values in the range of 0.12 to 32 (.mu.g/ml).

[0118] The compound of general formula I does not exhibit any cytotoxicity upto 40 .mu.g/ml in macrophage J774 cell line.

[0119] An embodiment of the invention; provides a process for the preparation of a compound of formula 9, wherein the process step comprising of the reaction of compound of formula 8 in an organic solvent selected from toluene, acetonotrile, DMF, dichloroethaneor any combination thereof in the presence of azide source selected from sodiumazide, trimethylsilylazide, tetrabutyl ammonium bromide or any combination thereof at a temperature in the range of 25.degree. C. to 80.degree. C. for a period of 1 h to 3 h to obtain the desired compound of formula 9.

[0120] In another embodiment of the invention a process for the preparation of the compound of formula 10, wherein the process step comprising of the reaction of compound of formula 9 in an organic solvent selected from toluene, acetonotrile, DMF, tetrahydrofuran or any combination thereof in the presence of base selected from sodium hydride, cesium carbonate, potassium carbonate or any combination thereof at a temperature in the range of 10.degree. C. to 25.degree. C. for a period of 1 h to 12 h to obtain the desired compound of formula 10.

[0121] In another embodiment of the invention, a process for the preparation of the compound of general formula I, wherein the process step comprising the reacting compound of formula 10 with a compound of formula selected from the group consisting of formula 13 or 14 or 15 in 1:1 tert-BuOH/H.sub.2O mixture in the presence of sodium ascorbate and CuSO.sub.4 at a temperature in the range of 10.degree. C. to 25.degree. C. for a period of 1 h to 12 h to obtain the desired compound of general formula I.

The present invention discloses process for synthesis of the fragments A and B which in turn can be used for synthesis of compounds of general formula I. The entire synthesis of compound I is illustrated by Reaction Schemes 1 to 4 as given below: --

##STR00007##

##STR00008##

##STR00009##

##STR00010##

##STR00011##

EXAMPLES

Synthesis of Compounds:

[0122] The following examples are given by way of illustrating the present invention and should not be construed to limit the scope of the invention:

R)-3-(2-Chloro-4-nitro-1H-imidazol-1-yl)-2-hydroxy-2-methylpropylmethanesu- lfonate (8)

[0123] The synthesis of compound 8 was successfully synthesized from a starting material 4-Nitroimidiazole 1 as shown in scheme 1 by following known procedure (Sasaki, H.; Haraguchi, Y.; Itotani, M.; Kuroda, H.; Hashizume, H.; Tomishige, T.; Kawasaki, M.; Matsumoto, M.; Komatsu, M.; Tsubouchi, H. J. Med. Chem. 2006, 49, 7854.

Example 1

(R)-1-Azido-3-(2-chloro-4-nitro-1H-imidazol-1-yl)-2-methylpropan-2-ol (9)

[0124] To a solution of (R)-3-(2-Chloro-4-nitro-1H-imidazol-1-yl)-2-hydroxy-2-methylpropyl methanesulfonate (8) (10 mmol) in DMF (20 mL) was added sodium azide (30 mmol) and tetrabutyl ammonium bromide (1 mmol). After the solution was stirred at 80.degree. C. for 3 h, the reaction mixture was extracted with ethyl acetate twice, and the combined organic layer was washed with brine, dried over sodium sulphate, and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography in 5% DCM/Ethyl acetate to give the compound 9 as yellow colour solid. The compound 9 is also prepared using the process as described above using different solvents such as toluene, acetonitrile or dichloroethane at 60-80.degree. C. for a period of 2-4 hr as given in the following table.

TABLE-US-00001 Reactant Solvent Temp .degree. C. Time Reagents Product (R)-3-(2-Chloro-4- Toluene 60 2 Sodium azide (R)-1-Azido-3-(2- nitro-1H-imidazol-1- chloro-4-nitro-1H- yl)-2-hydroxy-2- imidazol-1-y1)-2- methylpropyl methylpropan-2-ol (9) methanesulfonate (8) (R)-3-(2-Chloro-4- Acetonitrile 60 4 Trimethylsily (R)-1-Azido-3-(2- nitro-1H-imidazol-1- lazide chloro-4-nitro-1H- y1)-2-hydroxy -2- imidazol-1-y1)-2- methylpropyl methylpropan-2-ol (9) methanesulfonate (8) (R)-3-(2-Chloro-4- dichloroethane 80 2 Sodium azide (R)-1-Azido-3-(2- nitro-1H-imidazol-1- chloro-4-nitro-1H- yl)-2-hydroxy-2- imidazol-1-y1)-2- methylpropyl methylpropan-2-ol (9) methanesulfonate (8)

Example 2

(R)-2-(Azidomethyl)-2-methyl-6-nitro-2,3-dihydroimidazo[2,1-b]oxazole (10)

[0125] To a solution of (R)-1-Azido-3-(2-chloro-4-nitro-1H-imidazol-1-yl)-2-methylpropan-2-ol 9)(10 mmol) in DMF (20 mL) was added cesium carbonate (30 mmol) at below 15.degree. C. portion wise, After the solution was stirred for 12 h at 25.degree. C., The reaction mixture was poured into the ice water and extracted with ethyl acetate twice, and the combined organic layer was washed with brine, dried over sodium sulphate, and filtered. The filtrate was concentrated under reduced pressure. The residue was purified by silica gel column chromatography to give the compound 10 as yellow colour solid.

[0126] The reaction is also carried out by using the different solvent such as toluene, acetonitrile or tetrahydrofurane at 25.degree. C. for a period of 6-12 hr. to produce the compound 10.

TABLE-US-00002 Reactant Solvent Tem. .degree. C. Time Reagents Product (R)-1-Azido-3-(2- DMF 25 6 Sodium (R)-2-(Azidomethyl)- chloro-4-nitro-1H- hydride 2-methy1-6-nitro-2,3- imidazol-1-y1)-2- dihydroimidazo [2,1- methylpropan-2-ol (9) b] oxazole (10) (R)-1-Azido-3-(2- Toluene 25 6 Sodium (R)-2-(Azidomethyl)- chloro-4-nitro-1H- hydride 2-methy1-6-nitro-2,3- imidazol-1-y1)-2- dihydroimidazo [2,1- methylpropan-2-ol (9) b] oxazole (10) (R)-1-Azido-3-(2- Acetonitrile 25 12 Potassium (R)-2-(Azidomethyl)- chloro-4-nitro-1H- carbonate 2-methy1-6-nitro-2,3- imidazol-1-y1)-2- dihydroimidazo [2,1- methylpropan-2-ol (9) b] oxazole (10) (R)-1-Azido-3-(2- tetrahydro- 25 12 Sodium (R)-2-(Azidomethyl)- chloro-4-nitro-1H- furan carbonate 2-methy1-6-nitro-2,3- imidazol-1-y1)-2- dihydroimidazo [2,1- methylpropan-2-ol (9) b] oxazole (10)

Example 3

[0127] General Procedure for the Preparation of Compounds (I.sub.1 to I.sub.22):

[0128] (R)-2-(Azidomethyl)-2-methyl-6-nitro-2,3-dihydroimidazo[2,1-b]oxazo- le (10) (1 mmol) and 13 (a-k) or 14 (a-g) or 15 (a-d) suspended in 6 mL of a 1:1 tert-BuOH/H.sub.2O mixture. While the mixture was being stirred, sodium ascorbate (0.1 mmol) was added, followed by CuSO.sub.4 pentahydrate (0.02 mmol). Left stirring for 12 h at 25.degree. C., after which time it was diluted with water, and the solid was filtered off. The crude was purified by silica gel column chromatography to give the compounds I.sub.1 to I.sub.22.

(R)-2-Methyl-6-nitro-2-((4-(4-(trifluoromethoxy)phenyl)-1H-1,2,3-triazol-1- -yl)methyl)-2,3-dihydroimidazo[2,1-b]oxazole (I.sub.1, Table 1, Scheme 3)

##STR00012##

[0130] TLC (EtOAc:DCM 1:9): R.sub.f=0.3; .sup.1H NMR (400 MHz, Acetone) .delta. 8.50 (s, 2H), 8.01 (t, J=4.5 Hz, 7H), 7.39 (d, J=8.0 Hz, 5H), 5.10 (q, J=14.9 Hz, 6H), 4.66 (d, J=11.2 Hz, 3H), 4.42 (d, J=11.2 Hz, 3H), 1.79 (s, 7H); MS (ESI+): m\z 410.0950.

(R)-2-Methyl-6-nitro-2-((4-(4-(trifluoromethyl)phenyl)-1H-1,2,3-triazol-1-- yl)methyl)-2,3-dihydroimidazo[2,1-b]oxazole (I.sub.2, Table 1, Scheme 3)

##STR00013##

[0132] TLC (EtOAc:DCM 1:9): R.sub.f=0.45; .sup.1H NMR (400 MHz, DMSO) .delta. 8.74 (s, 1H), 8.07 (t, J=4.0 Hz, 3H), 7.81 (d, J=8.3 Hz, 2H), 5.05 (d, J=14.8 Hz, 1H), 4.98 (d, J=14.8 Hz, 1H), 4.44 (d, J=11.3 Hz, 1H), 4.26 (d, J=11.3 Hz, 1H), 1.63 (s, 3H); MS (ESI+): m\z 394.1001.

(R)-2-((4-(4-Fluorophenyl)-1H-1,2,3-triazol-1-yl)methyl)-2-methyl-6nitro-2- ,3-dihydro imidazo[2,1-b]oxazole (I.sub.3, Table 1, Scheme 3)

##STR00014##

[0134] TLC (EtOAc:DCM 1:9): R.sub.f=0.35; .sup.1H NMR (400 MHz, DMSO) .delta. 8.54 (s, 1H), 8.05 (s, 1H), 7.87 (dd, J=8.7, 5.5 Hz, 2H), 7.28 (t, J=8.9 Hz, 2H), 5.01 (d, J=14.8 Hz, 1H), 4.95 (d, J=14.8 Hz, 1H), 4.42 (d, J=11.3 Hz, 1H), 4.24 (d, J=11.2 Hz, 1H), 1.63 (s, 3H); MS (ESI+): m\z 344.1033.

(R)-2-((4-(4-Fluoro-3-methylphenyl)-1H-1,2,3-triazol-1-yl)methyl)-2-methyl- -6-nitro-2,3-dihydroimidazo[2,1-b]oxazole (I.sub.4, Table 1, Scheme 3)

##STR00015##

[0136] TLC (EtOAc:DCM 1:9): R.sub.f=0.5; .sup.1H NMR (400 MHz, DMSO) .delta. 8.47 (s, 1H), 8.02 (s, 1H), 7.72 (d, J=7.0 Hz, 1H), 7.69-7.60 (m, 1H), 7.19 (t, J=9.1 Hz, 1H), 5.00 (d, J=14.8 Hz, 1H), 4.93 (d, J=14.8 Hz, 1H), 4.41 (d, J=11.3 Hz, 1H), 4.23 (d, J=11.3 Hz, 1H), 2.27 (s, 3H), 1.63 (s, 3H); MS (ESI+): m\z 358.1190.

(R)-2-((4-(2,4-Difluorophenyl)-1H-1,2,3-triazol-1-yl)methyl)-2-methyl-6-ni- tro-2,3-dihydroimidazo[2,1-b]oxazole (I.sub.5, Table 1, Scheme 3)

##STR00016##

[0138] TLC (EtOAc:DCM 1:9): R.sub.f=0.25; .sup.1H NMR (500 MHz, Acetone) .delta. 8.35 (d, J=3.6 Hz, 2H), 8.21 (dd, J=15.5, 8.8 Hz, 2H), 7.81 (s, 2H), 7.20-7.11 (m, 4H), 5.15 (q, J=14.9 Hz, 6H), 4.68 (d, J=11.1 Hz, 3H), 4.43 (d, J=11.1 Hz, 3H), 1.79 (s, 8H); MS (ESI+): m\z 362.0939.

(R)-2-Methyl-6-nitro-2-((4-(4-phenoxyphenyl)-1H-1,2,3-triazol-1-yl)methyl)- -2,3-dihydroimidazo[2,1-b]oxazole (I.sub.6, Table 1, Scheme 3)

##STR00017##

[0140] TLC (EtOAc:DCM 1:9): R.sub.f=0.25; .sup.1H NMR (400 MHz, DMSO) .delta. 8.47 (s, 1H), 8.02 (s, 1H), 7.82 (d, J=8.7 Hz, 2H), 7.41 (t, J=7.9 Hz, 2H), 7.16 (t, J=7.4 Hz, 1H), 7.05 (dd, J=8.1, 6.0 Hz, 4H), 5.00 (d, J=14.8 Hz, 1H), 4.93 (d, J=14.8 Hz, 1H), 4.42 (d, J=11.3 Hz, 1H), 4.24 (d, J=11.3 Hz, 1H), 1.63 (s, 3H); MS (ESI+): m\z 418.1390.

(R)-2-[(4-Pentyl-1H-1,2,3-triazol-1-yl)methyl]-2,3-dihydro-2-methyl-6-nitr- oimidazo[2,1-b]oxazole (I.sub.7, Table 1, Scheme 3)

##STR00018##

[0142] TLC (EtOAc:DCM 1:9): R.sub.f=0.5; .sup.1H NMR (400 MHz, Acetone) .delta. 8.18 (s, 1H), 7.80 (s, 1H), 4.62 (d, J=11.2 Hz, 2H), 4.40 (d, J=11.2 Hz, 2H), 2.25 (t, 2H), 1.75 (s, 3H), 1.21 (m, 4H), 1.15 (m, 2H), 0.91 (m, 3H); MS (ESI+): m\z 320.1579.

(R)-2-Methyl-6-nitro-2-((4-((4-(trifluoromethoxy)phenoxy)methyl)-1H-1,2,3-- triazol-1-yl)methyl)-2,3-dihydroimidazo[2,1-b]oxazole (I.sub.8, Table 1, Scheme 4)

##STR00019##

[0144] TLC (EtOAc:DCM 1:9): R.sub.f=0.45; .sup.1H NMR (400 MHz, Acetone) .delta. 8.18 (s, 1H), 7.79 (s, 1H), 7.28 (d, J=9.2 Hz, 2H), 7.14 (d, J=9.2 Hz, 2H), 5.22 (s, 2H), 5.08 (q, J=14.9 Hz, 2H), 4.65 (d, J=11.2 Hz, 1H), 4.42 (d, J=11.2 Hz, 1H), 1.78 (s, 3H); MS (ESI+): m\z 440.1056.

(R)-2-Methyl-6-nitro-2-((4-((4-(trifluoromethyl)phenoxy)methyl)-1H-1,2,3-t- riazol-1-yl)methyl)-2,3-dihydroimidazo[2,1-b]oxazole (I.sub.9, Table 1, Scheme 4)

##STR00020##

[0146] TLC (EtOAc:DCM 1:9): R.sub.f=0.15; .sup.1H NMR (400 MHz, DMSO) .delta. 8.23 (s, 1H), 8.05 (s, 1H), 7.65 (d, J=8.7 Hz, 2H), 7.20 (d, J=8.5 Hz, 2H), 5.23 (s, 2H), 4.97 (q, J=14.8 Hz, 2H), 4.39 (d, J=11.3 Hz, 1H), 4.23 (d, J=11.3 Hz, 1H), 1.59 (s, 3H); MS (ESI+): m\z 424.1107.

(R)-2-((4(3-Chlorophenoxy)methyl)-1H-1,2,3-triazol-1-yl)methyl)-2-methyl-6- -nitro-2,3-dihydroimidazo[2,1-b]oxazole (I.sub.10, Table 1, Scheme 4)

##STR00021##

[0148] TLC (EtOAc:DCM 1:9): R.sub.f=0.25; .sup.1H NMR (400 MHz, DMSO) .delta. 8.21 (s, 1H), 8.05 (s, 1H), 7.30 (t, J=8.2 Hz, 1H), 7.11 (t, J=2.0 Hz, 1H), 6.99 (ddd, J=10.4, 8.2, 1.6 Hz, 2H), 5.14 (s, 2H), 4.96 (q, J=14.8 Hz, 2H), 4.39 (d, J=11.3 Hz, 1H), 4.22 (d, J=11.3 Hz, 1H), 1.59 (s, 3H); MS (ESI+): m\z 390.0843.

(R)-2-(4-((4-Bromophenoxy)methyl)-1H-1,2,3-triazol-1-yl)methyl)-2-methyl-6- -nitro-2,3-dihydroimidazo[2,1-b]oxazole (I.sub.11, Table 1, Scheme 4)

##STR00022##

[0150] TLC (EtOAc:DCM 1:9): R.sub.f=0.35; .sup.1H NMR (400 MHz, DMSO) .delta. 8.20 (s, 1H), 8.04 (s, 1H), 7.44 (d, J=8.6 Hz, 2H), 6.98 (d, J=8.4 Hz, 2H), 5.11 (s, 2H), 4.96 (q, J=14.8 Hz, 2H), 4.38 (d, J=11.3 Hz, 1H), 4.22 (d, J=11.2 Hz, 1H), 1.58 (s, 3H); MS (ESI+): m\z 434.0338.

(R)-2-Methyl-6-nitro-2-((4-((p-tolyloxy)methyl)-1H-1,2,3-triazol-1-yl)meth- yl)-2,3-dihydroimidazo[2,1-b]oxazole (I.sub.12, Table 1, Scheme 4)

##STR00023##

[0152] TLC (EtOAc:DCM 1:9): R.sub.f=0.3; .sup.1H NMR (400 MHz, DMSO) .delta. 8.17 (s, 1H), 8.05 (s, 1H), 7.08 (d, J=8.2 Hz, 2H), 6.88 (d, J=8.4 Hz, 2H), 5.07 (s, 2H), 5.02-4.88 (m, 2H), 4.38 (d, J=11.3 Hz, 1H), 4.22 (d, J=11.3 Hz, 1H), 2.23 (s, 3H), 1.59 (s, 3H); MS (ESI+): m\z 370.1390.

(R)-2-Methyl-6-nitro-2-((4-((m-tolyloxy)methyl)-1H-1,2,3-triazol-1-yl)meth- yl)-2,3-dihydroimidazo[2,1-b]oxazole (I.sub.13, Table 1, Scheme 4)

##STR00024##

[0154] TLC (EtOAc:DCM 1:9): R.sub.f=0.3; .sup.1H NMR (400 MHz, DMSO) .delta. 8.17 (s, 1H), 8.03 (s, 1H), 7.15 (t, J=7.8 Hz, 1H), 6.77 (dd, J=16.5, 7.7 Hz, 3H), 5.07 (s, 2H), 4.95 (q, J=14.8 Hz, 2H), 4.38 (d, J=11.3 Hz, 1H), 4.22 (d, J=11.3 Hz, 1H), 2.26 (s, 3H), 1.58 (s, 3H); MS (ESI+): m\z 370.1390.

(R)-2-Methyl-6-nitro-2-((4-((o-tolyloxy)methyl)-1H-1,2,3-triazol-1-yl)meth- yl)-2,3-dihydroimidazo[2,1-b]oxazole (I.sub.14, Table 1, Scheme 4)

##STR00025##

[0156] TLC (EtOAc:DCM 1:9): R.sub.f=0.35; .sup.1H NMR (400 MHz, Acetone) .delta. 8.14 (s, 7H), 7.78 (s, 6H), 7.12 (dt, J=22.2, 7.9 Hz, 26H), 6.85 (t, J=6.9 Hz, 8H), 5.17 (s, 21H), 5.07 (q, J=14.9 Hz, 27H), 4.62 (d, J=11.2 Hz, 12H), 4.40 (d, J=11.2 Hz, 12H), 2.13 (s, 20H), 1.77 (s, 36H); MS (ESI+): m\z 370.1390.

(R)-2-((4-((4-Ethylphenoxy)methyl)-1H-1,2,3-triazol-1-yl)methyl)-2-methyl-- 6-nitro-2,3-dihydroimidazo[2,1-b]oxazole (I.sub.15, Table 1, Scheme 4)

##STR00026##

[0158] TLC (EtOAc:DCM 1:9): R.sub.f=0.5; .sup.1H NMR (400 MHz, Acetone) .delta. 8.11 (s, 1H), 7.79 (s, 1H), 7.13 (d, J=8.4 Hz, 2H), 6.92 (d, J=8.5 Hz, 2H), 5.19-4.98 (m, 6H), 4.63 (d, J=11.2 Hz, 2H), 4.40 (d, J=11.1 Hz, 2H), 2.58 (dd, J=15.1, 7.5 Hz, 2H), 1.76 (s, 3H), 1.18 (t, J=7.6 Hz, 14H); MS (ESI+): m\z 384.1546.

(R)-2-((4-((3-Fluorophenoxy)methyl)-1H-1,2,3-triazol-1-yl)methyl)-2-methyl- -6-nitro-2,3-dihydroimidazo[2,1-b]oxazole (I.sub.16, Table 1, Scheme 4)

##STR00027##

[0160] TLC (EtOAc:DCM 1:9): R.sub.f=0.2; .sup.1H NMR (400 MHz, DMSO) .delta. 8.21 (s, 1H), 8.03 (s, 1H), 7.30 (dd, J=15.3, 7.7 Hz, 1H), 6.93-6.72 (m, 3H), 5.12 (s, 2H), 4.96 (q, J=14.7 Hz, 2H), 4.38 (d, J=11.1 Hz, 1H), 4.22 (d, J=11.2 Hz, 1H), 1.59 (s, 3H); MS (ESI+): m\z 374.1139.

(R)-2-((4-((2-Fluorophenoxy)methyl)-1H-1,2,3-triazol-1-yl)methyl)-2-methyl- -6-nitro-2,3-dihydroimidazo[2,1-b]oxazole (I.sub.17, Table 1, Scheme 4)

##STR00028##

[0162] TLC (EtOAc:DCM 1:9): R.sub.f=0.3; .sup.1H NMR (400 MHz, Acetone) .delta. 8.18 (s, 1H), 7.80 (s, 1H), 7.29 (td, J=8.6, 1.6 Hz, 1H), 7.17-7.09 (m, 2H), 6.96 (ddd, J=7.8, 7.0, 1.5 Hz, 1H), 5.26 (s, 3H), 5.07 (q, J=14.9 Hz, 4H), 4.62 (d, J=11.2 Hz, 2H), 4.40 (d, J=11.2 Hz, 2H), 1.75 (s, 5H); MS (ESI+): m\z 374.1139.

(R)-2-((4-((4-Isopropylphenoxy)methyl)-1H-1,2,3-triazol-1-yl)methyl)-2-met- hyl-6-nitro-2,3-dihydroimidazo[2,1-b]oxazole (I.sub.18, Table 1, Scheme 4)

##STR00029##

[0164] TLC (EtOAc:DCM 1:9): R.sub.f=0.35; .sup.1H NMR (400 MHz, DMSO) .delta. 8.18 (s, 1H), 8.07 (s, 1H), 7.14 (d, J=8.5 Hz, 2H), 6.91 (d, J=8.6 Hz, 2H), 5.07 (s, 2H), 4.96 (q, J=14.8 Hz, 2H), 4.39 (d, J=11.3 Hz, 1H), 4.22 (d, J=11.2 Hz, 1H), 2.83 (dt, J=13.8, 6.8 Hz, 1H), 1.58 (s, 3H), 1.17 (d, J=6.9 Hz, 6H); MS (ESI+): m\z 398.1703.

(R)-2-Methyl-6-nitro-2-((4-((pyridin-2-yloxy)methyl)-1H-1,2,3-triazol-1-yl- )methyl)-2,3-dihydroimidazo[2,1-b]oxazole (I.sub.19, Table 1, Scheme 5)

##STR00030##

[0166] TLC (EtOAc:DCM 1:9): R.sub.f=0.4; .sup.1H NMR (400 MHz, DMSO) .delta. 8.19 (s, 1H), 8.04 (s, 1H), 7.98 (m, 1H), 7.25-7.38 (m, 3H), 5.10 (s, 2H), 4.95 (q, J=14.8 Hz, 2H), 4.38 (d, J=11.3 Hz, 1H), 4.22 (d, J=11.3 Hz, 1H), 1.58 (s, 3H); MS (ESI+): m\z 357.1186.

(R)-2-Methyl-6-nitro-2-((4-((p-tolylthio)methyl)-1H-1,2,3-triazol-1-yl)met- hyl)-2,3-dihydroimidazo[2,1-b]oxazole (I.sub.20, Table 1, Scheme 5)

##STR00031##

[0168] TLC (EtOAc:DCM 1:9): R.sub.f=0.3; .sup.1H NMR (400 MHz, DMSO) .delta. 8.17 (s, 1H), 8.05 (s, 1H), 7.08 (d, J=8.2 Hz, 2H), 6.88 (d, J=8.4 Hz, 2H), 5.07 (s, 2H), 5.02-4.88 (m, 2H), 4.38 (d, J=11.3 Hz, 1H), 4.22 (d, J=11.3 Hz, 1H), 2.23 (s, 3H), 1.59 (s, 3H); MS (ESI-F): m\z 386.1161.

(R)-2-Methyl-6-nitro-2-((4-(2-(p-tolyloxy)ethyl)-1H-1,2,3-triazol-1-yl)met- hyl)-2,3-dihydroimidazo[2,1-b]oxazole (I.sub.21, Table 1, Scheme 5)

##STR00032##

[0170] TLC (EtOAc:DCM 1:9): R.sub.f=0.35; .sup.1H NMR (400 MHz, DMSO) .delta. 8.17 (s, 1H), 8.05 (s, 1H), 7.08 (d, J=8.2 Hz, 2H), 6.88 (d, J=8.4 Hz, 2H), 5.02-4.88 (m, 2H), 4.38 (d, J=11.3 Hz, 1H), 4.22 (d, J=11.3 Hz, 1H), 4.12 (d, J=4.8 Hz, 2H), 2.32 (d, J=4.8 Hz, 2H), 2.23 (s, 3H), 1.59 (s, 3H); MS (ESI+): m\z 384.1546.

(R)-2-Methyl-2-((4-(morpholinomethyl)-1H-1,2,3-triazol-1-yl)methyl)-6-nitr- o-2,3-dihydroimidazo[2,1-b]oxazole (I.sub.22, Table 1, Scheme 5)

##STR00033##

[0172] TLC (EtOAc:DCM 1:9): R.sub.f=0.25; .sup.1H NMR (400 MHz, DMSO) .delta. 8.16 (s, 1H), 8.06 (s, 1H), 5.04 (s, 2H), 5.02-4.88 (m, 2H), 4.38 (d, J=11.3 Hz, 1H), 4.22 (d, J=11.3 Hz, 1H), 4.09-4.18 (d, 4H), 3.36-3.48 (m, 4H), 1.58 (s, 3H); MS (ESI+): m\z 349.1499.

Biological Evaluation

Example 1

[0173] In Vitro Activity of Compounds I.sub.I to I.sub.22 Against M. tuberculosis H.sub.37Rv and Two Clinical Isolates (M. tuberculosis MDR & M. tuberculosis XDR)

[0174] MIC Determination:

[0175] MIC was determined by broth dilution method against M. tuberculosis H.sub.37Rv (ATCC 27294; American Type Culture Collection, Manassas, Va.), M. tuberculosis MDR (resistant to isoniazid and rifampicin) and M. tuberculosis XDR (resistant to isoniazid, rifampicin, amikacin and moxifloxacin). The bacterial strains were grown for 10 to 15 days in Middlebrook 7H9 broth (Difco Laboratories, Detroit, Mich.) supplemented with 0.5% (v/v) glycerol, 0.25% (v/v) Tween 80 (Himedia, Mumbai India), and 10% ADC (albumin dextrose catalase, Becton Dickinson, Sparks, Md.) under shaking conditions at 37.degree. C. in 5% CO.sub.2 to facilitate exponential-phase growth of the organism. Bacterial suspension was prepared by suspending M. tuberculosis growth in normal saline containing 0.5% tween 80 and turbidity was adjusted to 1 McFarland standard which is equivalent to 1.5.times.10.sup.7 CFU/ml. The 2-fold serial dilutions of compounds I.sub.1 to I.sub.22 were prepared in Middle brook 7H9 (Difco laboratories) for M. tuberculosis in 100 .mu.l per well in 96-well U bottom microtitre plates (Tarson, Mumbai, India). The above-mentioned bacterial suspension was further diluted in the growth media and 100 .mu.l volume of this diluted inoculum was added to each well of the plate resulting in the final inoculum of 5.times.10.sup.5 CFU/ml in the well and the final concentrations of compound I.sub.I to I.sub.22 ranged from 0.015 to 32 .mu.g/ml (0.015, 0.03, 0.06, 0.12, 0.25, 0.5, 1, 2, 4, 8, 16, 32). The plates were incubated at 37.degree. C. for 3-weeks in 5% CO.sub.2. The plates were read visually and the minimum concentration of the compound showing no turbidity was recorded as MIC.

Results:

[0176] i) The compounds of general formula I (compounds I.sub.1 to I.sub.22), were screened against both replicating & non-replicating stages of M. tuberculosis, wherein 7 compounds I.sub.10, I.sub.11, I.sub.13, I.sub.14, I.sub.17, I.sub.18 and I.sub.21 showed MIC value of <1.0 .mu.g/ml (results provided in Table 2). Three compounds I.sub.11, I.sub.17 and I.sub.21 showed very potent MIC of 0.12, 0.25 and 0.25 .mu.g/ml against replicating stages of M. Tuberculosis and MIC of 0.25, 0.5 and 0.25 against non-replicating stages of M. Tuberculosis. The results are given in Table 2. ii) The compounds of general formula I (compounds I.sub.1 to I.sub.22), were screened against both multi-drug and extensive-drug resistant strains of M. tuberculosis, wherein five compounds I.sub.11, I.sub.17, I.sub.19, I.sub.20 and I.sub.21 showed MIC value of <1.0 .mu.g/ml. Three compounds I.sub.11, I.sub.17 and I.sub.21 showed very potent MIC of 0.12, 0.5 and 0.25 .mu.g/ml against multi and extensive-drug resistant strains of M. Tuberculosis. The results are given in Table 2.

Example 2

[0177] Cytotoxicity Assay of Compounds I.sub.1 to I.sub.22:

Cell Culture:

[0178] The study was carried out using macrophage J774 cells line (ATCC-USA). Cells were grown in Rosewell Park Memorial Institute Medium (RPMI) containing 10% fetal calf serum (FCS) and supplemented with 75 mg/litre penicillin, 100 mg/litre streptomycin, 110 mg/litre Sodium pyruvate, 2.38 gm/litre HEPES, 0.05 mM 2 .beta.-mercaptoethanol, and 2 gm/litre NaHCO.sub.3, in a humidified atmosphere in 5% CO.sub.2 at 37.degree. C., and were sub-cultured at 1:4 ratio once a week.

Cell Treatment:

[0179] Cells were plated at a density of 3.times.10.sup.4 cells/cm.sup.2 and maintained in culture medium for 12 hours. Cells were seeded onto 96-well flat bottom plates and FCS was reduced to 5% for the experiment. Stock solutions of compounds 9 to 37 were prepared fresh to avoid oxidation. Cells were incubated with the compounds (40 .mu.g/ml) for 24 hrs.

Cytotoxicity Assays:

[0180] After the completion of incubation, the medium was removed and cell viability was evaluated by assaying for the ability of functional mitochondria to catalyze the reduction of 3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltetrazolium bromide (MTT) to form formazan salt by mitochondrial dehydrogenases, and determined by Elisa reader at 450 nm (Multiskan Spectrum; Thermo Electron Corporation, USA). Percentage cytotoxicity was calculated with respect to the untreated cells.

Results:

[0181] Compounds I.sub.1 to I.sub.22 were not toxic up to 40 .mu.g/ml concentration and the cytotoxicity assay results are shown in Table 2.

TABLE-US-00003 TABLE 1 Structure of representative compounds I.sub.1 to I.sub.22 of general formula I Entries Codes Structures 1 I.sub.1 ##STR00034## 2 I.sub.2 ##STR00035## 3 I.sub.3 ##STR00036## 4 I.sub.4 ##STR00037## 5 I.sub.5 ##STR00038## 6 I.sub.6 ##STR00039## 7 I.sub.7 ##STR00040## 8 I.sub.8 ##STR00041## 9 I.sub.9 ##STR00042## 10 I.sub.10 ##STR00043## 11 I.sub.11 ##STR00044## 12 I.sub.12 ##STR00045## 13 I.sub.13 ##STR00046## 14 I.sub.14 ##STR00047## 15 I.sub.15 ##STR00048## 16 I.sub.16 ##STR00049## 17 I.sub.17 ##STR00050## 18 I.sub.18 ##STR00051## 19 I.sub.19 ##STR00052## 20 I.sub.20 ##STR00053## 21 I.sub.21 ##STR00054## 22 I.sub.22 ##STR00055##

TABLE-US-00004 TABLE 2 Anti-tuberculosis activity and cytotoxicity of representative compounds of general formula I (I.sub.1 to I.sub.22) MIC (.mu.g/ml) Compound M. tb Non-replicating MIC Cytotoxicity S. No. code H.sub.37Rv strain (Rif.sup.R) MDR XDR (.mu.g/ml) 1. I.sub.1 2 4 2 2 1 >40 2. I.sub.2 2 8 4 2 2 >40 3. I.sub.3 1 2 2 2 2 >40 4. I.sub.4 2 4 1 4 2 >40 5. I.sub.5 1 8 4 1 4 >40 6. I.sub.6 2 2 1 1 1 >40 7. I.sub.7 2 4 8 2 8 >40 8. I.sub.8 2 4 2 2 2 >40 9. I.sub.9 1 2 4 4 4 >40 10. I.sub.10 0.5 1 2 2 4 >40 11. I.sub.11 0.12 0.25 0.12 0.12 0.25 >40 12. I.sub.12 1 2 8 2 8 >40 13. I.sub.13 0.5 0.5 1 1 2 >40 14. I.sub.14 0.5 2 1 1 1 >40 15. I.sub.15 2 2 4 2 4 >40 16. I.sub.16 1 2 2 2 2 >40 17. I.sub.17 0.25 0.5 1 0.5 1 >40 18. I.sub.18 0.5 1 2 1 4 >40 19. I.sub.19 1 2 0.5 0.25 0.5 >40 20. I.sub.20 2 1 0.25 0.5 2 >40 21. I.sub.21 0.25 0.25 1 0.25 0.25 >40 22. I.sub.22 2 1 2 2 2 >40

* * * * *


uspto.report is an independent third-party trademark research tool that is not affiliated, endorsed, or sponsored by the United States Patent and Trademark Office (USPTO) or any other governmental organization. The information provided by uspto.report is based on publicly available data at the time of writing and is intended for informational purposes only.

While we strive to provide accurate and up-to-date information, we do not guarantee the accuracy, completeness, reliability, or suitability of the information displayed on this site. The use of this site is at your own risk. Any reliance you place on such information is therefore strictly at your own risk.

All official trademark data, including owner information, should be verified by visiting the official USPTO website at www.uspto.gov. This site is not intended to replace professional legal advice and should not be used as a substitute for consulting with a legal professional who is knowledgeable about trademark law.

© 2024 USPTO.report | Privacy Policy | Resources | RSS Feed of Trademarks | Trademark Filings Twitter Feed