Mountain fetipenem or its salts or esters for the treatment of mycobacterial infections

文档序号:1408796 发布日期:2020-03-06 浏览:28次 中文

阅读说明:本技术 用于治疗分枝杆菌感染的山费培南或其盐或酯 (Mountain fetipenem or its salts or esters for the treatment of mycobacterial infections ) 是由 D.巴罗斯阿吉雷 R.H.贝茨 R.冈扎勒兹德尔里奥 A.蒙多扎罗萨纳 S.拉蒙加西亚 于 2018-05-04 设计创作,主要内容包括:本发明涉及式(I)或其药学上可接受的盐或酯前药,其用于治疗分枝杆菌感染或由分枝杆菌感染引起的疾病,如结核病。<Image he="434" wi="681" file="DDA0002263834270000011.GIF" imgContent="drawing" imgFormat="GIF" orientation="portrait" inline="no"></Image>(The present invention relates to prodrugs of formula (I) or a pharmaceutically acceptable salt or ester thereof, which are useful for treating mycobacterial infections or diseases caused by mycobacterial infections, such as tuberculosis.)

1. A compound of the formula:

Figure FDA0002263834240000011

or a pharmaceutically acceptable salt or ester prodrug thereof, for use in the treatment of a disease caused by a mycobacterial infection.

2. A compound for use according to claim 1, or a pharmaceutically acceptable salt or ester prodrug thereof, wherein the disease is tuberculosis.

3. A compound of the formula:

or a pharmaceutically acceptable salt or ester prodrug thereof, for use in the treatment of a mycobacterial infection.

4. A compound or a pharmaceutically acceptable salt or ester prodrug thereof for use according to claim 3, wherein the mycobacterium infection is a mycobacterium tuberculosis infection.

5. A compound of the formula:

Figure FDA0002263834240000013

or a pharmaceutically acceptable salt or ester prodrug thereof, for use in the treatment of tuberculosis.

6. The compound for use according to any one of claims 1 to 5, wherein the compound is an ester prodrug of a compound of the formula:

Figure FDA0002263834240000021

7. the compound for use according to claim 6, wherein the ester prodrug is:

Figure FDA0002263834240000022

8. the compound for use according to claim 6 or 7, wherein the prodrug is administered orally.

9. The compound for use according to any one of claims 1 to 5, wherein the compound of formula:

Figure FDA0002263834240000023

10. a method of treating a disease caused by a mycobacterial infection in a patient in need thereof, comprising administering to said patient a therapeutically effective amount of a compound of the formula:

Figure FDA0002263834240000024

or a pharmaceutically acceptable salt or ester prodrug thereof.

11. A method according to claim 10, wherein the disease is tuberculosis.

12. A method of treating a mycobacterial infection in a patient in need thereof, comprising administering to said patient a therapeutically effective amount of a compound of the formula:

or a pharmaceutically acceptable salt or ester prodrug thereof.

13. The method according to claim 12, wherein the mycobacterial infection is a mycobacterium tuberculosis infection.

14. A method of treating tuberculosis in a patient in need thereof, comprising administering to said patient a therapeutically effective amount of a compound of the formula:

Figure FDA0002263834240000032

or a pharmaceutically acceptable salt or ester prodrug thereof.

15. A method according to any one of claims 10 to 14 which comprises administering an ester prodrug of a compound of the formula:

Figure FDA0002263834240000033

16. the method according to claim 15, wherein said ester prodrug is a compound of the formula:

Figure FDA0002263834240000034

17. a process according to any one of claims 10 to 14 wherein the compound of the formula:

Figure FDA0002263834240000041

18. the method of any one of claims 10 to 17, wherein the patient is a human.

19. Use of a compound of the formula:

Figure FDA0002263834240000042

20. a pharmaceutical composition comprising (a) a compound of the formula:

Figure FDA0002263834240000043

or a pharmaceutically acceptable salt or ester prodrug thereof; and (b) a pharmaceutically acceptable excipient for use in the treatment of tuberculosis, a mycobacterial infection, or a disease caused by a mycobacterial infection.

21. A combination of (a) a compound of the formula:

Figure FDA0002263834240000044

or a pharmaceutically acceptable salt or ester prodrug thereof; and (b) an additional anti-tuberculosis agent for use in the treatment of a mycobacterial infection, a disease caused by a mycobacterial infection, or tuberculosis.

22. A combination for use according to claim 21, wherein the additional anti-tuberculosis agent (b) is selected from isoniazid, rifampin, pyrazinamide, ethambutol, moxifloxacin, rifapentine, clofazimine, ethionamide, prothiocyponamide, pentoxyphenylthiourea, thiosemicarbazide, rifabutin, diarylquinolines such as bedaquiline (TMC207) or TBAJ-587, nitroimidazoxazine PA-824, delamanide (OPC-67683), oxazolidinones such as linezolid, tedizozolid, reed azolil, sudilazolil (PNU-100480), petazolil (AZD-5847) or TBI-223, EMB analogue SQ109, OPC-167832, GSK3036656 (also known as GSK070), GSK2556286, GSK3211830, benzothiazinones such as BTZ043 or PBTZ, azaindoles such as TBA-631, dinitrobenzene 3036656 (also known as gmk 070), GSK2556286, GSK3211830, benzothiazinone such as e-e.

23. A combination for use according to claim 21 or 22, wherein the additional anti-tuberculosis agent (b) is AUGMENTIN (amoxicillin clavulanate).

24. The combination for use according to claim 21 or 22, further comprising an antiviral agent, including an antiretroviral agent.

25. The combination for use according to claim 24, wherein the antiretroviral agent is selected from zidovudine, didanosine, lamivudine, zalcitabine, abacavir, stavudine, adefovir dipivoxil, fozivudine, todoxil, emtricitabine, alovudine, amdoxovir, elvucitabine, nevirapine, delavirdine, efavirenz, loveamine, cloviramine, valmazone, oltipraz, capravirin, rivvirine, GSK2248761, TMC-278, TMC-125, etravirine, saquinavir, ritonavir, indinavir, nelfinavir, amprenavir, fosamprenavir, breinavir, darunavir, atazanavir, tipinavir, tipranavir, enparib, T-20, T-9, PRO-12475, tnvavir-BMS-806-BMS 806, BMS-663068and BMS-626529, 5-Helix, raltegravir, Etegravir, GSK1349572, GSK1265744, Vilivir (Sch-C), Sch-D, TAK779, maraviroc, TAK449, didanosine, tenofovir, lopinavir or darunavir.

26. A combination of (a) a compound according to any one of claims 1 to 9, or a pharmaceutically acceptable salt or ester prodrug thereof, and (b) β -lactamase inhibitor.

27. The combination according to claim 26, wherein the β -lactamase inhibitor is clavulanate or clavulanic acid.

Technical Field

The present invention relates to sanfetrinem (sanfetrinem), or a pharmaceutically acceptable salt or ester prodrug thereof, and its use to treat mycobacterial infections or diseases caused by mycobacterial infections. More specifically, the invention relates to the use of sanfetida or a pharmaceutically acceptable salt or ester prodrug thereof for the treatment of tuberculosis. In particular, the invention relates to prodrugs, sanfetrine ciloxentin, and their use in the treatment of tuberculosis.

Background

According to reports issued by the world health organization in 2014, nearly ten million people are infected with Tuberculosis (TB) every year, resulting in 150 million deaths each year. Despite the availability of tuberculosis therapies, the global disease burden remains a major problem because the causative agent of tuberculosis is mycobacterium tuberculosis, which is resistant to many therapies.

Although tuberculosis is caused by bacterial infection, the use of β -lactam, the most predominant antibiotic, has been largely ignored, despite the fact that the dozens of approved β -lactam drugs account for approximately two thirds of the global antibiotic market, their assessment of resistance to tuberculosis is limited by early failure of clinical trials, and the inability of the lipophilic mycobacterial cell wall to penetrate this highly polar molecule is believed.

The shanfeng west ester is an experimental antibiotic produced in the 90 s of the 20 th century and is associated with infections caused by various bacterial substances, but does not include mycobacteria.

Due to the growing multidrug resistant strains of mycobacterium tuberculosis and the continuing high prevalence of tuberculosis, there is an urgent need to provide further pharmaceutical compounds to treat tuberculosis.

Disclosure of Invention

In a first aspect of the invention, there is provided a compound of the formula:

it has the name (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazetidino [2,1-a ]]Isoindole-4-carboxylic acid, or a pharmaceutically acceptable salt or ester prodrug thereof, for use in treating a disease caused by a mycobacterial infection.

In a second aspect of the invention, there is provided a compound of the formula:

Figure BDA0002263834250000022

it has the name (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazetidino [2,1-a ]]Isoindole-4-carboxylic acid, or a pharmaceutically acceptable salt or ester prodrug thereof, for use in treating a mycobacterial infection.

In a third aspect of the invention, there is provided a compound of the formula:

Figure BDA0002263834250000023

it has the name (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazetidino [2,1-a ]]Isoindole-4-carboxylic acid, or a pharmaceutically acceptable salt or ester prodrug thereof, for use in the treatment of tuberculosis.

In a fourth aspect of the invention, there is provided a method of treating a disease caused by a mycobacterial infection in a patient in need thereof, comprising administering to said patient a therapeutically effective amount of a compound of the formula:

Figure BDA0002263834250000031

it has the name (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazetidino [2,1-a ]]Isoindole-4-carboxylic acid, or a pharmaceutically acceptable salt or ester prodrug thereof.

In a fifth aspect of the invention, there is provided a method of treating a mycobacterial infection in a patient in need thereof, comprising administering to said patient a therapeutically effective amount of a compound of the formula:

Figure BDA0002263834250000032

it has the name (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazetidino [2,1-a ]]Isoindole-4-carboxylic acid, or a pharmaceutically acceptable salt or ester prodrug thereof.

In a sixth aspect of the invention, there is provided a method of treating tuberculosis in a patient in need thereof, comprising administering a therapeutically effective amount of a compound of the formula:

Figure BDA0002263834250000033

it has the name (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazetidino [2,1-a ]]Isoindole-4-carboxylic acid, or a pharmaceutically acceptable salt or ester prodrug thereof.

In a seventh aspect of the present invention there is provided the use of a compound of the formula:

Figure BDA0002263834250000041

it has a nameWeighing (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazetidino [2,1-a ]]Isoindole-4-carboxylic acid.

In an eighth aspect of the invention, there is provided a pharmaceutical composition comprising (a) a compound of the formula:

it has the name (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazetidino [2,1-a ]]Isoindole-4-carboxylic acid, or a pharmaceutically acceptable salt or ester prodrug thereof; and (b) a pharmaceutically acceptable excipient, for use in the treatment of tuberculosis, mycobacterial infection or disease caused by mycobacterial infection.

In a ninth aspect of the invention, there is provided (a) a compound of the formula:

Figure BDA0002263834250000043

it has the name (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazetidino [2,1-a ]]Isoindole-4-carboxylic acid, or a pharmaceutically acceptable salt or ester prodrug thereof; and (b) another other anti-tuberculosis agent for use in the treatment of a mycobacterial infection, a disease caused by a mycobacterial infection, or tuberculosis.

In a tenth aspect of the present invention, there is provided (a) a compound of the formula:

Figure BDA0002263834250000051

it has the name (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazetidino [2,1-a ]]A combination of isoindole-4-carboxylic acid, or a pharmaceutically acceptable salt or ester prodrug thereof, and (b) β -lactamase inhibitor.

Drawings

Figure 1 shows the results of a mouse model used to evaluate the anti-tubercular activity in vivo of sanfetida and sanfetida west ester.

FIG. 2 is a MIC profile of mountain fetipenem (sodium salt) against a panel of laboratory strains and clinical isolates.

Figure 3 shows the dose-response curves of the combination of sanfetilin alone, sanfetilin + amoxicillin, and sanfetilin + amoxicillin + clavulanic acid at the time of use, the% growth as a function of concentration (concentration values of amoxicillin in μ g/mL).

Figure 4 shows the dose-response curve,% growth as a function of concentration, of amoxicillin alone and amoxicillin + clavulanate in combination, when used.

Fig. 5 shows dose-response curves of mountain fetilin alone and mountain fetilin + delamani combination.

Fig. 6 shows dose-response curves of sanfetilin alone and in combination with sanfetilin + rifampin.

Fig. 7 shows the dose-response curves of sanfetilin alone and in combination with sanfetilin + ethambutol.

Fig. 8 shows the dose-response curves of sanfetilide alone and sanfetilide + amoxicillin combination.

Detailed Description

The present invention relates to a compound having the following structure (hereinafter also referred to as compound a):

Figure BDA0002263834250000052

or a pharmaceutically acceptable salt or ester prodrug thereof, for use in the treatment of a disease caused by a mycobacterial infection. A mycobacterial infection is an infection caused by an infection with mycobacteria.

The name of compound a is (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazetidino [2,1-a ] isoindole-4-carboxylic acid.

The mycobacterium may be a member of one of the following mycobacterial groups: mycobacterium Tuberculosis Complex (MTC), Mycobacterium Avium Complex (MAC), Mycobacterium gordonae complex (Mycobacterium gordonae complex), Mycobacterium kansasii complex (Mycobacterium kansaii complex), Mycobacterium chelonii complex (Mycobacterium chelonii complex), Mycobacterium fortuitum complex (Mycobacterium fortuitum complex), Mycobacterium parafortuitum complex (Mycobacterium paraquat complex), or Mycobacterium vaccae complex (Mycobacterium vaccae). The Mycobacterium may also be Mycobacterium ulcerosa (mycobacterulluraces) or Mycobacterium leprae (Mycobacterium leprae).

Members of the Mycobacterium Tuberculosis Complex (MTC) include Mycobacterium tuberculosis, Mycobacterium africanum (M. africanum), Mycobacterium bovis (M. bovis), Mycobacterium bovis BCG (M. bovis BCG), Mycobacterium cassiae (M. canitti), Mycobacterium capriae (M. caprae), Mycobacterium microti (M. cinerea) and Mycobacterium marinum (M. pinipedii). These mycobacteria are the causative agents of tuberculosis in humans and animals. Mycobacterium tuberculosis is the leading cause of tuberculosis in humans.

In one embodiment, the infection is caused by an infection with a mycobacterium that is a member of the Mycobacterium Tuberculosis Complex (MTC).

In one embodiment, the infection is a mycobacterium tuberculosis infection. In other words, the mycobacterial infection is caused by infection with mycobacterium tuberculosis.

Members of the avian Mycobacterium complex (MAC) include Mycobacterium avium, Mycobacterium paratuberculosis (Mycobacterium avium paratuberculosis), Mycobacterium silaticum, Mycobacterium hominis, Mycobacterium columbiense, and Mycobacterium indicus pranii.

Members of the Mycobacterium gordoniae clade include Mycobacterium asia (Mycobacterium astricium) and Mycobacterium gordoniae (Mycobacterium gordonae).

Members of the Mycobacterium kansasii clade include Mycobacterium gastri (Mycobacterium gasti) and Mycobacterium kansasii (Mycobacterium kansasii).

Members of the Mycobacterium cheloni clade include Mycobacterium abscessus (Mycobacterium abscessus), Mycobacterium boletii, and Mycobacterium cheloni (Mycobacterium chelonae).

Members of Mycobacterium fortuitum include Mycobacterium polynicotinae (Mycobacterium boisenei), Mycobacterium brisket (Mycobacterium brisbanense), Mycobacterium aesthetic (Mycobacterium cosmeticum), Mycobacterium fortuitum, Mycobacterium aceti-formis (Mycobacterium fortuitum), Mycobacterium houston (Mycobacterium homeoense), Mycobacterium margaricum (Mycobacterium macroense), Mycobacterium neoaerosol (Mycobacterium newbereneum), Mycobacterium exocomplex (Mycobacterium perensense), Mycobacterium suis (Mycobacterium tuberculosis), Mycobacterium endogamum (Mycobacterium senense), and Mycobacterium haemophilum (Mycobacterium septicum).

Members of the Mycobacterium paraguatum clade include Mycobacterium africanum (Mycobacterium australiana), Mycobacterium dychii (Mycobacterium dianhofer), Mycobacterium perogerberensis (Mycobacterium frederi), Mycobacterium hodeloides (Mycobacterium hodleri), Mycobacterium neoaurum (Mycobacterium neoaurum) and Mycobacterium paraguatum (Mycobacterium parafortuitum).

Thus, a mycobacterial infection may be caused by an infection with a mycobacterium selected from the group consisting of: mycobacterium tuberculosis, Mycobacterium africanum, Mycobacterium bovis BCG, Mycobacterium cassettii, Mycobacterium caprine, Mycobacterium microti, Mycobacterium seal, Mycobacterium avium, Mycobacterium paratuberculosis, Mycobacterium sellaticum, Mycobacterium avium, Mycobacterium hominisis, Mycobacterium columbiense, Mycobacterium indicus pranii, Mycobacterium asia, Mycobacterium gordonii, Mycobacterium gastri, Mycobacterium kansasii, Mycobacterium abscessus, Mycobacterium bolletii, Mycobacterium bellertii, Mycobacterium boidinii including Mycobacterium polynicoides, Mycobacterium brisketchuensis, Mycobacterium beautifulli, Mycobacterium fortuitum subsp Mycobacterium disi, Mycobacterium perogeri, Mycobacterium hodelum, Mycobacterium neogold, Mycobacterium paraguatum, Mycobacterium ulcerosa and Mycobacterium leprae.

Diseases caused by mycobacterial infection include, but are not limited to, tuberculosis (e.g., from mycobacterium tuberculosis), leprosy (e.g., from mycobacterium leprae), johns disease (Johne's disease) (e.g., from mycobacterium paratuberculosis subspecies avium), brudri (Buruli) or bantzda (Bairnsdale) ulcers (e.g., from mycobacterium ulcerosa), crohn's disease (e.g., from mycobacterium paratuberculosis subspecies), cystic fibrosis (e.g., from non-tuberculosis mycobacteria such as intracellular avian-type mycobacterial complex and mycobacterium abscessus), pulmonary disease or lung infection, pneumonia, synovial sac, synovium, tendon sheath, topical abscess, lymphadenitis, skin and soft tissue infection, wendemal man syndrome (Lady winddermeremer syndrome) (e.g., from avian-type mycobacterial complex (MAC)), MAC lung disease, disseminated avian complex (DMAC), DMAC, and c, Disseminated intracellular Mycobacterium avium complex (DMAIC), heat bath lung disease (e.g., from Mycobacterium avium complex), MAC mastitis, MAC pyomyositis, or granulomatous disease.

In one embodiment, the disease caused by a mycobacterium infection is tuberculosis, such that the present invention relates to compound a, or a pharmaceutically acceptable salt or ester prodrug thereof, for use in the treatment of tuberculosis.

The invention also relates to compound a, or a pharmaceutically acceptable salt or ester prodrug thereof, for use in the treatment of a mycobacterial infection. In a specific embodiment, the mycobacterium infection is a mycobacterium tuberculosis infection.

More particularly, the present invention relates to compound a, or a pharmaceutically acceptable salt or ester prodrug thereof, for use in the treatment of tuberculosis. In one embodiment, the treatment of tuberculosis may involve treating multidrug-resistant tuberculosis, extensively drug-resistant tuberculosis, or drug-sensitive tuberculosis.

In one embodiment, the treatment of tuberculosis involves multi-drug resistant tuberculosis or extensively drug resistant tuberculosis.

Furthermore, the treatment may involve pulmonary and/or extrapulmonary tuberculosis. The treatment may also involve the treatment of latent tuberculosis.

Compound a is also known as mountain fetilin or GV 104326. The sodium salt of sanfetilin is called sanfetilin sodium. The potassium salt of sanfetilin is called sanfetilin potassium, and so on.

More specifically, the present invention relates to ester prodrugs of compound a useful for treating mycobacterial infections or diseases caused by mycobacterial infections, wherein said prodrugs have the following structure

Figure BDA0002263834250000081

The ester prodrug has the name (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazepino [2,1-a ] isoindole-4-carboxylic acid 1- (((cyclohexyloxy) carbonyl) oxy) ethyl ester. The prodrug is also known as sanfetida or GV 118819X.

Thus, the present invention also relates to (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazepino [2,1-a ] isoindole-4-carboxylic acid 1- (((cyclohexyloxy) carbonyl) oxy) ethyl ester, i.e., ethyl ester

Figure BDA0002263834250000091

It can be used for treating tuberculosis.

It is understood that the ester prodrug, 1- (((cyclohexyloxy) carbonyl) oxy) ethyl (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazepino [2,1-a ] isoindole-4-carboxylate (described above) is a mixture of diastereomers, also known aS epimers. Diastereomers may be present in the mixture in equal amounts (1:1 mixture) or in unequal amounts. Alternatively, the compound may exist as one diastereomer. The individual diastereomers are shown below.

The single diastereomer can be obtained by separating a mixture of diastereomers.

The above prodrug can be advantageously used for the treatment of tuberculosis, since it can be administered orally to patients.

An alternative prodrug is (S) -1- ((ethoxycarbonyl) oxy) ethyl (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazepino [2,1-a ] isoindole-4-carboxylate, having the structure,

Figure BDA0002263834250000101

the prodrug can be prepared according to, for example, the procedure described in WO94/21637 or similar methods.

All chemical structures have been named using ChemBioDraw Ultra version 12.0 (by converting the structure to a name).

It will be understood that reference herein to compound a or a salt thereof includes compound a as the free acid, or as a pharmaceutically acceptable salt thereof. Thus, in one embodiment, the invention relates to compound a. In another embodiment, the invention relates to a pharmaceutically acceptable salt of compound a. In another embodiment, the invention relates to ester prodrugs of compound a.

The term "ester prodrug" refers to compound a in which an ester has been formed on or using a free carboxylic acid moiety present. In other words, "ester prodrug" means that an ester has been formed using the free acid available in the structure below.

Figure BDA0002263834250000102

Examples of such ester prodrugs are compounds having the structure

Figure BDA0002263834250000103

The term "pharmaceutically acceptable" refers to those compounds (including salts), materials, compositions, and dosage forms which are, within the scope of sound medical judgment, suitable for use in contact with the tissues of human beings and animals without excessive toxicity, irritation, or other problem or complication, commensurate with a reasonable benefit/risk ratio.

Pharmaceutically acceptable Salts include, inter alia, those described in Berge, j.pharm.sci., 1977, 66, 1-19 or P H Stahl and C G Wermuth, editors, Pharmaceutical Salts; properties, Selection and use, second edition, Stahl/Wermuth: Wiley-VCH/VHCA, 2011 (see http:// www.wiley.com/WileyCDA/WileyTitle/productCd-3906390519. html).

Suitable pharmaceutically acceptable salts may include base addition salts.

The base addition salts may be formed by reaction of (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazepino [2,1-a ] isoindole-4-carboxylic acid with a suitable base, optionally in a suitable solvent such aS an organic solvent, to give a salt which may be isolated by a variety of methods including crystallization and filtration.

Representative pharmaceutically acceptable base addition salts include, but are not limited to, aluminum salts, 2-amino-2- (hydroxymethyl) -1, 3-propanediol (TRIS, tromethamine) salts, arginine salts, benzphetamine (N-benzylphenethylamine) salts, benzathine (N, N '-dibenzylethylenediamine) salts, bis- (2-hydroxyethyl) amine salts, bismuth salts, calcium salts, chloroprocaine salts, choline salts, clemizole (1-p-chlorobenzyl-2-pyrrolidin-1' -ylmethylbenzimidazole) salts, cyclohexylamine salts, dibenzylethylenediamine salts, diethylamine salts, diethyltriamine salts, dimethylamine salts, dimethylethanolamine salts, dopamine salts, ethanolamine salts, ethylenediamine salts, L-histidine salts, iron salts, isoquinoline salts, luridine salts, Lithium salt, lysine salt, magnesium salt, meglumine (N-methylglucamine) salt, piperazine salt, piperidine salt, potassium salt, procaine salt, quinine salt, quinoline salt, sodium salt, strontium salt, tert-butylamine salt and zinc salt.

Specific pharmaceutically acceptable salts according to the invention are the sodium and potassium salts of compound a, such that the compounds administered to a patient for treatment are potassium (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazepino [2,1-a ] isoindole-4-carboxylate or sodium (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazepino [2,1-a ] isoindole-4-carboxylate, are respectively shown as follows

Figure BDA0002263834250000121

In one embodiment, the pharmaceutically acceptable salt of compound a is a sodium salt.

It is understood that the compound (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazetidino [2,1-a ] isoindole-4-carboxylic acid or a pharmaceutically acceptable salt thereof, or a prodrug thereof, (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazepino [2,1-a ] isoindole-4-carboxylic acid 1- (((cyclohexyloxy) carbonyl) oxy) ethyl ester can be in any suitable solvated (e.g., hydrated) and/or polymorphic form thereof.

The compound (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazetidino [2,1-a ] isoindole-4-carboxylic acid or a pharmaceutically acceptable salt thereof, or a prodrug thereof, (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazetidino [2,1-a ] isoindole-4-carboxylic acid 1- (((cyclohexyloxy) carbonyl) oxy) ethyl ester can be prepared according to the procedures described in EP0416953 and WO94/21637 or by analogous methods.

Specifically, (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazetidino [2,1-a ] isoindole-4-carboxylic acid or a salt thereof can be prepared according to the procedures described in examples 4 and 5 of WO 94/21637.

In particular, the prodrug 1- (((cyclohexyloxy) carbonyl) oxy) ethyl (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazepino [2,1-a ] isoindole-4-carboxylate can be prepared according to the procedure described in example 1 of WO 92/03437. Other ester prodrugs can be prepared according to analogous methods and methods known to those skilled in the art.

In another aspect of the invention, there is provided a method of treating a disease caused by a mycobacterium infection in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazetidino [2,1-a ] isoindole-4-carboxylic acid or a pharmaceutically acceptable salt or ester prodrug thereof.

The mycobacterial infection may be caused by one of the mycobacteria selected from the list above, such as mycobacterium tuberculosis.

In one embodiment, the disease to be treated is tuberculosis. Thus, in one embodiment, the invention also relates to a method of treating tuberculosis in a patient in need thereof, comprising administering to said patient a therapeutically effective amount of (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazetidino [2,1-a ] isoindole-4-carboxylic acid or a pharmaceutically acceptable salt or ester prodrug thereof.

In another aspect, there is provided a method of treating a mycobacterium infection in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazepino [2,1-a ] isoindole-4-carboxylic acid, or a pharmaceutically acceptable salt or ester prodrug thereof.

The mycobacterial infection may be caused by one of the mycobacteria selected from the list above. In one embodiment, the mycobacterium infection is a mycobacterium tuberculosis infection, and the present invention relates to methods of treating a mycobacterium tuberculosis infection.

In another aspect, there is provided a method of treating tuberculosis in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazepino [2,1-a ] isoindole-4-carboxylic acid, or a pharmaceutically acceptable salt or ester prodrug thereof.

In one embodiment, there is provided a method of treating tuberculosis in a patient in need thereof, comprising administering to the patient a therapeutically effective amount of an ester prodrug of compound a. In particular, the ester prodrug is (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazepino [2,1-a ] isoindole-4-carboxylic acid 1- (((cyclohexyloxy) carbonyl) oxy) ethyl ester, which has the structure shown above.

In another embodiment, there is provided a method of treating tuberculosis in a patient in need thereof, comprising administering to said patient a therapeutically effective amount of a pharmaceutically acceptable salt of compound a. In particular, the pharmaceutically acceptable salt is the sodium salt, such that the drug administered to the patient is sodium (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazetidino [2,1-a ] isoindole-4-carboxylate.

In one embodiment, the patient in need thereof is a human patient. The term patient is intended to mean a person suffering from or infected with a mycobacterial infection, a disease caused by a mycobacterial infection or tuberculosis.

Also provided is the use of the compound (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazepino [2,1-a ] isoindole-4-carboxylic acid, or a pharmaceutically acceptable salt or ester prodrug thereof, in the manufacture of a medicament for the treatment of a disease caused by or for the treatment of a mycobacterial infection. In particular, the invention relates to the use of the compound (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazepino [2,1-a ] isoindole-4-carboxylic acid, or a pharmaceutically acceptable salt or ester prodrug thereof, for the manufacture of a medicament for the treatment of tuberculosis.

In particular, (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazepino [2,1-a ] isoindole-4-carboxylic acid or (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a, use of 1- (((cyclohexyloxy) carbonyl) oxy) ethyl 8 b-octahydroazetidino [2,1-a ] isoindole-4-carboxylate in the preparation of a medicament for the treatment of tuberculosis.

As used herein, the term "therapeutically effective amount" refers to any amount that results in improved treatment, cure, prevention, or amelioration of a disease, disorder, or side effect, or any amount that reduces the rate of progression of a disease or disorder, as compared to a corresponding subject that does not receive that amount.

The appropriate "therapeutically effective amount" will depend upon a number of factors including, for example, the age and weight of the subject, the exact condition to be treated and its severity, the nature of the formulation and the route of administration, and will ultimately be at the discretion of the attendant physician.

Those skilled in the art will appreciate that reference herein to treatment refers to treatment of established conditions, including, for example, mycobacterial infection, disease caused by mycobacterial infection, and/or tuberculosis. However, depending on the condition, (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazepino [2,1-a ] isoindole-4-carboxylic acid and pharmaceutically acceptable salts or ester prodrugs thereof can also be used for the prevention of mycobacterial infections, diseases caused by mycobacterial infections and/or tuberculosis. Thus, in one embodiment, treatment or prevention of a disease is provided. In another embodiment, treatment of a disease is provided. In another embodiment, prevention of a disease is provided.

Although it is possible that for the treatment of a mycobacterial infection, a disease caused by a mycobacterial infection or tuberculosis, (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazepino [2,1-a ] isoindole-4-carboxylic acid and pharmaceutically acceptable salts or ester prodrugs thereof can be administered alone, it is usually present aS an active ingredient of a pharmaceutical composition comprising one or more pharmaceutically acceptable excipients.

Thus, in one embodiment, there is also provided a pharmaceutical composition comprising (a) sodium (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazepino [2,1-a ] isoindole-4-carboxylate or prodrug 1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazepino [2,1-a ] isoindole-4-carboxylic acid 1- (((cyclohexyloxy) carbonyl) oxy) ethyl ester, and (b) a pharmaceutically acceptable excipient for use in the treatment of a mycobacterial infection, a disease caused by a mycobacterial infection, or tuberculosis.

The pharmaceutical compositions may be administered by any suitable route, for example by the oral (including buccal or sublingual), inhalation, intranasal, topical (including buccal, sublingual or transdermal) or parenteral (including subcutaneous, intramuscular, intravenous or intradermal) route.

In one embodiment the pharmaceutical composition is administered by the oral route of administration. When the pharmaceutical composition is for oral administration, the ester prodrug 1- (((cyclohexyloxy) carbonyl) oxy) ethyl (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazepino [2,1-a ] isoindole-4-carboxylate is used in particular aS an active pharmaceutical ingredient.

In one embodiment the pharmaceutical composition is administered by the intravenous route of administration. When the pharmaceutical composition is for intravenous administration, the sodium or potassium salt of the compound a is specifically used as an active pharmaceutical ingredient.

Suitable pharmaceutically acceptable excipients include the following types of excipients: carriers, diluents, fillers, binders, disintegrants, lubricants, glidants, granulating agents, coating agents, wetting agents, solvents, co-solvents, suspending agents, emulsifiers, sweeteners, flavoring agents, taste masking agents, colorants, anti-caking agents, wetting agents, chelating agents, plasticizers, viscosity increasing agents, antioxidants, preservatives, stabilizers, surfactants, and buffers.

Preparation of (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazetidino [2,1-a ]]Suitable methods for isoindole-4-carboxylic acid or a pharmaceutically acceptable salt or ester prodrug thereof include those familiar to those skilled in the art, which are described in Remington: the Science and practice of Pharmacy, 21stEdition 2006。

The pharmaceutical compositions may be presented in unit dosage forms containing a predetermined amount of the active ingredient per unit dose. Preferred unit dosage compositions are those containing a daily dose or sub-dose, or an appropriate fraction thereof, of the active ingredient. Thus, such unit doses may be administered more than once per day. Preferred unit dosage compositions are those containing a daily dose or sub-dose (for more than one administration per day), or an appropriate fraction thereof, of the active ingredient as described above.

For the administration of the active ingredient, e.g. ester prodrug (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazepino [2,1-a ] isoindole-4-carboxylic acid 1- (((cyclohexyloxy) carbonyl) oxy) ethyl ester, the dose can be from 100mg to 2000mg or from 100mg to 1000 mg. For example, the dose may be 250mg to 500 mg. In particular, the total dose may be 250mg to 1000mg, administered twice daily, to provide a total daily dose of 500mg to 2000 mg. The total dose may be administered once daily. The total amount of active ingredient administered per day may be 500, 1000 or 2000 mg.

For intravenous administration, for example, (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazepino [2,1-a ] isoindole-4-carboxylic acid, or a pharmaceutically acceptable salt thereof, can be administered in a dose of up to 1g three times per day, providing a total daily dose of 3 g. Alternatively, the total daily dose may be 1.5g or 2g per day.

The compounds may be administered in a single dose per day or in a number (e.g. two, three, four, five or six) of sub-doses per day, such that the total daily dose is the same. An effective amount of a pharmaceutically acceptable salt or ester prodrug thereof may be determined in proportion to an effective amount of the compound (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazepino [2,1-a ] isoindole-4-carboxylic acid or a pharmaceutically acceptable salt thereof itself.

Examples of oral tablet formulations that can be used are described in EP502465, as shown below.

Figure BDA0002263834250000161

The active ingredient and lactose can be mixed together and then granulated with water as the granulation liquid. The dried granules are then mixed with ethylcellulose, sodium lauryl sulfate and magnesium stearate and the tablet cores are formed using a suitable punch. The tablets may then be coated (e.g., with an enteric coating) using conventional techniques and coatings.

For use in the present invention, the compound (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazepino [2,1-a ] isoindole-4-carboxylic acid, or a pharmaceutically acceptable salt or ester prodrug thereof, can be used alone or in combination with other therapeutic agents. In particular, the compound (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazetidino [2,1-a ] isoindole-4-carboxylic acid, or a pharmaceutically acceptable salt or ester prodrug thereof, can be used in combination with another anti-tuberculosis and/or anti-viral agent, including anti-retroviral agents.

For example, also disclosed herein is a combination of (a) compound a, or a pharmaceutically acceptable salt or ester prodrug thereof, and (b) an additional anti-tuberculosis agent.

Accordingly, the invention also includes a combination of (a) compound a, or a pharmaceutically acceptable salt or ester prodrug thereof, and (b) an additional anti-tuberculosis agent, for use in the treatment of a mycobacterial infection, a disease caused by a mycobacterial infection, or tuberculosis. In one embodiment, the combination is for use in the treatment of tuberculosis.

In one embodiment, the combination may comprise 2, 3, 4,5, 6 or 7 additional anti-tuberculosis agents. For example, in the treatment of multi-drug resistant tuberculosis, combinations of four or more drugs are typically administered to a patient. For example, in the treatment of drug-sensitive tuberculosis, a combination of three or four drugs is usually administered to a patient.

The additional anti-tuberculosis agent may be a drug under development, approved or recommended for use in the treatment of tuberculosis.

In one embodiment, the anti-tuberculosis agent may be selected from isoniazid (isoniazid), rifampin (rifampin), pyrazinamide (pyrazinamide), ethambutol (ethambutol), moxifloxacin (moxifloxacin), rifapentine (rifapentine), clofazimine (clofazimine), ethionamide (ethionamide), prothioconazole (prothionamide), pentoxyphenylthiourea (isoxyl), thiosemicarbazide (thiacetone), rifabutin (rifabutin), diarylquinolines such as bedaquiline (TMC207) or TBAJ-587, nitroimidazoxazine-824 (tetramanid), delamazone (delamanidid) (67683), oxazolidine linezolid ketones such as linezolid (, tezoledrine (texiletine), dinitro-1833, such as buprofezin (SAE), or buprofezin (BToxazid) (BToxazid-100480), such as buprofezin (BToxazid), thiofenadine (BToxazid-368, TME) -or a (BToxazidine) (BToxazid-100480), such as BToxazid) (BToxazid-100480), or BToxazid) (BToxapenem-100480), or BTE-368 (BToxapenem-100480).

In one embodiment, the anti-tuberculosis agent may be AUGMENTIN, amoxicillin-clavulanate. Thus, also disclosed herein is a combination of compound a, or a pharmaceutically acceptable salt or ester prodrug thereof, and AUGMENTIN.

In another embodiment, the anti-tuberculosis agent may be selected from the group consisting of delamanic, rifampin, and ethambutol. Thus, also disclosed herein is a combination of compound a, or a pharmaceutically acceptable salt or ester prodrug thereof, and at least one of delamanib, rifampin, and ethambutol.

The combination used according to the invention may further comprise an antiviral agent, including an antiretroviral agent.

The antiretroviral agent may be selected from zidovudine (zidovudine), didanosine (didanosine), lamivudine (lamivudine), zalcitabine (zalcitabine), abacavir (abacavir), stavudine (stavudine), adefovir (adefovir), adefovir dipivoxil (adefovir dipivoxil), fuzivudine (fozivudine), todoxil, emtricitabine (emtricitabine), alovudine (alovudine), amdoxovir (amdoxovivir), elvucitabine (elvucitabine), nevirapine (nevirapine), delavirdine (delpravir), efavirenz (efavirenz), lovirdine (loviride), valvirucine (elvucinacavir), valviravirenz (ritonavir), nelfinavir (valviravir), thalivir (valavir-125), quinavir (tmavir), nevirane (tmcivir), nevirane (valvirine), nevira (valvirine), valvirine (tmcivir), valvirine (valvirine), valvirine (valviravir (valvirine), valvirine (valvirine), valvirine, Darunavir (daronavir), atazanavir (atazanavir), tipranavir (tipranavir), palinavir (palinavir), lacinavir (lasinavir), enfuvirdine (enfuvirtide), T-20, T-1249, PRO-542, PRO-140, TNX-355, BMS-806, BMS-663068andBMS-626529, 5-heix, raltegravir (raltegravir), erigerovir (elvitegravir), GSK1349572, GSK1265744, viriviroc (schcriviroc), Sch-D, TAK779, maraviroc (maravavir), TAK449, dihydroxyl (didanosine), inosine (tenofovir), lotrinavirus (lotavirus), or pinavir (pinavir).

The combination may conveniently be presented for use in the form of a pharmaceutical composition or formulation. Accordingly, also contemplated herein are pharmaceutical compositions comprising (a) compound a, or a pharmaceutically acceptable salt or ester prodrug thereof, as described herein, in combination with (b) another anti-tubercular drug and (c) optionally an antiviral agent, including an antiretroviral agent, and (d) one or more pharmaceutically acceptable excipients.

The compound (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazetidino [2,1-a ] isoindole-4-carboxylic acid, or a pharmaceutically acceptable salt or ester prodrug thereof, and the additional therapeutically active agent (such aS an anti-tuberculosis agent and an antiviral agent) can be administered together or separately, and when administered separately, can be administered separately or sequentially in any order (by the same or different routes of administration).

In one embodiment, compound a, or a pharmaceutically acceptable salt or ester prodrug thereof, is co-administered with an additional anti-tuberculosis agent. The term "co-administration" refers to the simultaneous administration or separate administration in any way of compound a or a pharmaceutically acceptable salt or ester prodrug thereof and an additional anti-tuberculosis agent which may be used for the treatment of a mycobacterial infection, a disease caused by a mycobacterial infection or tuberculosis, in particular tuberculosis.

The amounts of (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazetidino [2,1-a ] isoindole-4-carboxylic acid, or a pharmaceutically acceptable salt or ester prodrug thereof, and the additional therapeutically active agent, and the relative times of administration, will be selected to achieve the desired combined therapeutic effect.

The compound (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazetidino [2,1-a ] isoindole-4-carboxylic acid, or a pharmaceutically acceptable salt or ester prodrug thereof, can be administered in combination with an β -lactamase inhibitor.

Thus, the present invention also relates to the combination of the ester prodrug of sanfetilin (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazepino [2,1-a ] isoindole-4-carboxylic acid 1- (((cyclohexyloxy) carbonyl) oxy) ethyl ester and β -lactamase inhibitor.

The invention further relates to the combination of sodium salt of sanfetida (1S,5S,8aS,8bR) -1- ((R) -1-hydroxyethyl) -5-methoxy-2-oxo-1, 2,5,6,7,8,8a,8 b-octahydroazepino [2,1-a ] isoindole-4-carboxylic acid and β -lactamase inhibitor.

In one embodiment, the β -lactamase inhibitor is selected from clavulanic acid or a water-soluble salt thereof (e.g., potassium clavulanate), tazobactam, avibactam, and sulbactam.

In one embodiment, the β -lactamase inhibitor is clavulanic acid or a water soluble salt thereof, such as potassium clavulanate.

In one embodiment, the β -lactamase inhibitor may be provided as a combination of amoxicillin and potassium clavulanate (referred to as co-amoxiclav) (also referred to, for example, as AUGMENTIN.) the co-amoxiclav may be administered as a 375mg or 625mg tablet, where the potassium clavulanate is present in an amount equivalent to 125mg clavulanic acid.

The invention will now be described with reference to the following examples.

Examples

The sodium salt of sanfetepenem was tested in the following extracellular and intracellular viability assays some other β -lactam drugs-meropenem, tebipenem, ertapenem and faropenem were also tested in the same assay.

The preparation of mountain fetipenem is carried out according to one of the processes described in EP0416953 and WO94/21637 (see for example examples 4 and 5). In the assay described below, the sodium salt of mountain fetilin was used.

Faropenem sodium was purchased from AOKChem-CN (Ref A6030).

Meropenem was purchased from Combi Blocks, USA (Ref ST-9229).

Tebipenem is available from chemexpress (shanghai haoyuan) co., Ltd. (Ref HY-a 0076).

Ertapenem was purchased from Amatek (Ref DM-0004).

Potassium clavulanate was used as the source of clavulanic acid and is referred to in the table below as "clav".

List of abbreviations

DMSO, DMSO: dimethyl sulfoxide

ADC: albumin/dextrose/catalase

CFU: colony forming units

FBS: fetal bovine serum

Mtb: mycobacterium tuberculosis

RPMI: rosevir Park Memorial Park (Roswell Park Memorial Institute)

PBS: phosphate buffered saline

MIC assay

The Minimum Inhibitory Concentration (MIC) of each compound against mycobacterium tuberculosis H37Rv was tested in 96-well flat-bottomed polystyrene microtiter plates in a final volume of 200 μ L.

Test compounds were subjected to two-fold drug dilutions starting at 80 μ M in pure DMSO, from column 1 to column 10 for 10 aliquots. Moxifloxacin (MX) was used as a dose-responsive compound control, with a 2-fold dilution of MX starting at 1 μ g/ml in column 11. Rifampicin was dispensed at 1. mu.g/ml into G-12 to H-12 as a non-growth control. DMSO was dispensed into a12 to F12 as growth control.

Another plate with the same layout was also prepared, but 4. mu.g/ml potassium clavulanate (Fluka reference: 33454) was added to all the plates to test the change in MIC in the presence of the β -lactamase inhibitor.

Standardizing inoculum to about 1X107cfu/ml, and in Middlebrook 7H9 broth supplemented with adc (difco) at a ratio of 1: dilution at a ratio of 200. The inoculum (200. mu.L and 10. mu.L)4cfu/well) was added to the entire plate.

All plates were placed in a closed box to prevent drying of the peripheral wells and incubated at 37 ℃ for six days without shaking.

A resazurin solution was prepared by dissolving one tablet of resazurin (a resazurin tablet for cow milk test; reference 330884Y' vw international Ltd) in 30ml sterile PBS (phosphate buffered saline). To each well 25. mu.L of this solution was added.

Fluorescence (Spectramax M5Molecular Devices, excitation 530nm, emission 590nm, cut-off 570nm) was measured after 48 hours to determine MIC values.

Extracellular MIC values for sanfetilin (including sanfetilin pivoxil) and other representative β -lactams (meropenem, tebipenem, faropenem, and ertapenem) are reported in table 1 below.

Figure BDA0002263834250000211

TABLE 1

Intracellular viability assay

The antitubercular activity of the compounds against Mycobacterium tuberculosis grown in human THP-1 monocytes was determined using Mycobacterium tuberculosis H37Rv containing the firefly luciferase gene.

The THP-1 monocytes were kept in suspension in RPMI-1640 medium containing 10% FBS, 1mM pyruvate, 2mM L-glutamine, and 5% CO2Incubate at 37 ℃.

Monocyte growth to sub-confluency (5X 10)5Cells/ml) and infected in a cell roller bottle for 4 hours, with a multiplicity of infection (MOI) of 1, wherein sterile glass beads were dispersed in a bacterial suspension in RPMI-0.05% tween 80. Excess bacteria were removed by washing five times (1500rpm, 5 min) in RPMI medium.

Partitioning of infected cells into cells containing compound 1: 2 serial dilutions in a 96-well white plate (50,000 cells/well). The percentage of DMSO must be less than 0.5%.

Luminescence was measured after 5 days in a Victor 1420 system using the Steady-Glo Promega kit.

Results were processed using Grafit software. MIC90 values were calculated from dose response curves by nonlinear regression analysis.

Intracellular MIC50 values for sanfetida (including sanfetida west ester) and other representative β -lactams (meropenem, faropenem, tebipenem, and ertapenem) are reported in table 2 below the intracellular MIC90 values for the same compounds are reported in table 3.

Figure BDA0002263834250000221

TABLE 2

Figure BDA0002263834250000222

TABLE 3

In vivo experiments

To evaluate the in vivo antitubercular activity of sanfetida, the previous anti-tubercular antibodies previously described in anitirob agentschemither.2015aug; 59(8) 4997-9. However, different background strains were used for DHP-1 knockout mice: the C57Bl/6 background strain described in this article was replaced by a 129sv background strain.

Briefly, 8-10 week old female 129sv DHP-1KO pathogen free mice were purchased and acclimated for one week. Mice were infected intratracheally with approximately 10e5 CFU/mouse (M.tuberculosis H37 Rv). The sanfetiliam sodium salt compound (SNF), sanfetilim pivoxil (SNFc), Meropenem (MRP) and Clavulanate (CLV) were administered twice daily from day 9 to day 14 post-infection. MRP and SFN were injected subcutaneously. SFNc and CLV are administered by the oral route. Lungs were collected on day 9 or 15. All lung lobes were removed aseptically, homogenized and frozen. The homogenate was thawed and plated in 10% OADC-7H11 medium + 0.4% activated charcoal and held at 37 ℃ for 18 days.

The lung CFU count of untreated mice was 7.4 at day 9 and 9.0logCFU at day 15. The lung CFU of subcutaneous (sc) SFN, oral (po) SFNc and MRP-CLV treated mice were 7.3, 7.6 and 7.3, respectively (see fig. 1). In fig. 1, each dot represents data for one mouse. The average lung CFU count for each group is shown above the dots.

SFN and SFNc were equally effective against MRP-CLV in this assay, all three were able to prevent bacterial growth.

Even though no net killing effect was observed, this experiment provided evidence that both sanfetida and sanfetilide have an effect in vivo.

All animal studies were ethically reviewed and conducted in compliance with european directives 2010/63/EU and GSK for care, welfare and disposal policies for animals.

Conclusion

As demonstrated in tables 1-3, sanfetida has anti-mycobacterial activity, particularly anti-mycobacterium tuberculosis activity, in both intracellular and extracellular assays. Rifapenem and rifapenem pivoxil also have antitubercular activity in vivo.

In vitro activity of mountain fetipenem antituberculous mycobacterium clinical isolate

Tests have been performed against a panel of laboratory strains and clinical isolates of the mountain fetipenem (sodium salt), including drug sensitive and resistant strains. Table 4 below provides a detailed description of the drug resistance profile of the strains used.

Figure BDA0002263834250000241

Table 4DS, drug sensitive; RIF, rifampin; INH, isoniazid; MOX, moxifloxacin; STR, streptomycin; LZD, linezolid

Bacterial strains, general growth conditions and MGIT susceptibility assays

The strains were propagated in Middlebrook albumin-dextrose-catalase (ADC) (Difco) supplemented with 10% Middlebrook albumin, 0.2% glycerol and 0.05% (v/v) Middlebrook 7H9 broth (Difco) at 37 ℃. Sensitivity tests were performed using a BD BL MGIT tube supplemented with a BD BLMGIT OADC, using the MGIT960 system from Becton Dickinson (BD) Medical Technology. Briefly, to a 7mL MGIT tube was added 0.8mL OADC supplement, 0.1mL mountain fetipenem DMSO stock solution, and 0.1mL cells (final volume of MGIT tube was 8mL) to achieve a total of 10 per tube5Final cell density of individual cells. The Time To Positive (TTP) for the standard inoculum was 4 to 5 days. TTP is defined as the time required for a bacterial culture to reach a MGIT Growth Index (GI) above 75. The mountain fetipenem was assayed at 4 different concentrations (i.e., 0.5. mu.M, 1.25. mu.M, 5. mu.M and 20. mu.M). For each isolate, a strict MIC cut-off was defined as being able to inhibit (GI)<75) The bacteria were grown for at least a minimum concentration of 7 days. Rifampicin, isoniazid, moxifloxacin, linezolid and streptomycin were also used as internal controls against the activity of clinical isolates.

Activity of sanfetilin was tested against a panel of twenty-three mycobacterium tuberculosis strains, including clinical isolates with a single drug resistance pattern. Four different concentrations of mountain fetipenem (0.5. mu.M, 1.25. mu.M, 5. mu.M and 20. mu.M) were evaluated. The number of strains sharing the same MIC values is shown in figure 2. The most common MIC value is 5 μ M (15 out of 23). The MIC values for all four strains were below 5. mu.M, while the MIC values for the other four strains were higher. All strains were sensitive to the mountain fetipenem in the tested concentration range.

Combined experiments

Further experiments were performed with the combination of mountain fetipenem (sodium salt) with other drugs such as amoxicillin (and clavulanic acid), rifampin, ethambutol and delamnib.

Materials and methods-combination with amoxicillin and/or clavulanate

Checkerboard

Experiments were performed in 96-well plates. A checkerboard of mountain fetilin (sodium salt) and amoxicillin was prepared. Amoxicillin was diluted along the abscissa from column 1 to column 9 (from 80. mu.M to 0.3. mu.M) and mountain fetipenem was diluted along the ordinate from row A to row G (from 5. mu.M to 0.08. mu.M).

The following compositions were used: 2 dilution series, the resulting checkerboard plate comprising a combination of sanfetilim and amoxicillin, with the highest concentration of amoxicillin in column 1 and sanfetilim in row a.

To determine their respective MICs, column 10 contained only sanfetepenem and row H contained only amoxicillin.

Inocula with DMSO as positive growth control were dispensed to a12 to D12, and inocula +1 μ g/ml rifampicin (R3501_ Sigma) were dispensed to E12 to H12 (as inhibition control). Using these controls, Z' values and signal to noise ratios can be established as quality controls for the panels tested.

In column 11, moxifloxacin was assigned as a control of the test to a11(1 μ g/ml) to H11(0.008 μ g/ml) to provide a dose response curve.

Four identical plates were produced and potassium clavulanate (33454-. Experiments were performed with and without clavulanic acid.

A second set of four identically designed plates was also prepared, but with lower amoxicillin concentrations (from 5. mu.M to 0.02. mu.M in columns 1 to 9), to ensure that all relevant drug concentrations were studied.

Inoculum

The medium used was 7H 9-ADC-tyloxapol. 4.7g of Middlebrook 7H9 base broth was dissolved in 900mL of deionized water. Then 5mL of 10% w/v tyloxapol and 10% albumin-dextrose-catalase (ADC) concentrate (Becton Dickinson) were added. The strain used was Mycobacterium tuberculosis H37Rv, and the culture was standardized to approximately when the culture was in exponential growth phase1×107CFU/mL (OD600 ═ 0.125). The cultures were then grown in 7H 9-ADC-tyloxapol at 1: 200, and diluted at 5x104CFUs/ml 200 μ L of this inoculum was added to each well. The plates were incubated at 37 ℃ for 6 days.

Reading number

Resazurin was used as a reading. A piece of Resazurin (Ref R/0040/79_ Fisher Scientific) was dissolved in 30ml PBS. The solution was sterilized by filtration (0.22 μm). To each well 25. mu.l of sterile solution was added and the plates were incubated at 37 ℃ for an additional 48 hours. After incubation, fluorescence was measured in SpectraMax M5(Molecular Devices). The method comprises the following steps: excitation is carried out at 530 nm; emission 590nm (cut-off 570 nm).

Data analysis

Each assay plate contained one set of DMSO negative controls (corresponding to 100% bacterial growth), and one set of positive controls (1 μ g/ml rifampicin), where 100% bacterial growth inhibition was achieved. These controls were used to monitor the test quality by determining Z' and to normalize the data for each plate (% growth).

Each set of dose-response measurements (change in% growth versus [ mountain ferpenem ] ([ sanf ])) for mountain ferpenem was fitted to the following Hill type equation with four parameters (top, bottom, XC50 and Hill slope) under different conditions: monotherapy and in combination with amoxicillin (at different concentrations) and/or clavulanic acid:

growth% + bottom + (top-bottom)/(1 +10^ ((LogXC50-Log [ sanf ]) HillSlope))

The parameters for each case were calculated using the nonlinear regression curve fitting method available in GraphPad Prism 6 software.

Note that in this equation XC50 represents the concentration at which half the value of% growth between the bottom and top is reached. The parameter of interest is the concentration of mountain fetipenem at which 90% inhibition (i.e. 10% growth) is achieved, denoted herein as IC 90. Using the above equation, this parameter can be calculated directly from the estimated parameter, as follows:

IC90 ═ XC50 [ ((10% -bottom)/(top-10%)) (1/HillSlope)

To characterize amoxicillin in monotherapy, IC80 was calculated instead because it did not reach 90% inhibition. This parameter is similarly calculated from estimated parameters derived from a non-linear fit of the corresponding dose response measurements: change in% growth versus [ amoxicillin ] ([ amox ]).

Results

The data are reported in table 5 and fig. 3 and 4.

Compound (I) IC90(μg/ml)
Mountain faropenem 1.22
Mountain fetipenem + Clav 0.55
Mountain fetipenem + amoxicillin (5 mug/ml) 0.91
Mountain fetipenem + amoxicillin (5 mu g/ml) + Clav 0.1
Mountain fetipenem + amoxicillin (10 mug/ml) 0.33
Mountain fetipenem + amoxicillin (10 mu g/ml) + Clav 0.1

TABLE 5

It should be noted that amoxicillin was tested alone and the observed IC80 (in μ g/mL) value was 42. A combination test was also performed on amoxicillin and potassium clavulanate and an IC80 (in μ g/mL) value of 3.9 was observed.

For 90% growth inhibition (by FICI)90Expressed) Fractional Inhibitory Concentration Index (FICI) (synergy, antagonism and their checkerboard with respect to each other. Odds, f.c.1, s.l., Journal of analytical chemistry, 2003, vol.52) based on the Loewe additivity model (What is synergy? The

Figure BDA0002263834250000271

Ingredient revisited. tang j., Wennerberg k. and attokallo t.181, s.l., Frontiers in Pharmacology,2015, vol.6) are used herein to evaluate the synergy of the combination in this in vitro in vivo assay for extracellular activity. FICI ≦ 0.5 is considered to indicate synergy. This characterization/quantification of synergy is exploratory and does not explain the actual synergy or antagonistic mechanism (as indicated by Tang j., Wennerberg k. and aittokoro t.).

FICI90The calculation of (a) requires IC90 of the individual mountain faropenem, 1.22. mu.g/ml; IC90, 0.55 μ g/ml in combination with clavulanate; IC90, 0.33 μ g/ml in combination with amoxicillin (10 μ g/ml); and IC90, 0.1 μ g/ml in combination with amoxicillin (5 μ g/ml) + clavulanate. FIC because clavulanate alone did not work and amoxicillin alone or in combination with clavulanate did not achieve 90% growth inhibition (FIG. 7: dose response to amoxicillin), the effect of the drug on FIC was not as good as that of amoxicillin alone or in combination with clavulanate90The corresponding fractional term in the calculation is zero:

mountain fetipenem + clavulanate:

FICI90=0.55/1.22=0.45

this is almost a dilution and therefore is not considered to represent a significant synergy.

Mountain fetipenem + amoxicillin (10 μ g/ml):

FICI90=0.33/1.22=0.27

the combination is believed to represent a synergistic effect.

Mountain fetipenem + amoxicillin (5 μ g/ml) + clavulanate:

FICI90=0.1/1.22=0.1

the combination is believed to represent a synergistic effect.

Materials and methods-mountain fetilin (sodium salt) with Rifampin (RIF), Ethambutol (EMB), delamanib (DLD) and combinations of Amoxycillin (AMX)

Bacterial strains and general growth conditions

The Mtb H37Rv strain was routinely propagated in Middlebrook 7H9 broth (Difco) supplemented with 10% Middlebrook albumin-dextrose-catalase (ADC) (Difco), 0.2% glycerol, and 0.05% (v/v) at 37 ℃.

Drug sensitivity assay

Compound stocks used in this study were always prepared fresh on the same day of plating. For use in 384-well plate format, compounds were dissolved in DMSO and dispensed at two-fold dilution using an HP D3000 digital dispenser and an HP T8 dispenser head box (reference number CV 081A).

The Minimum Inhibitory Concentration (MIC) was determined in a broth based on 7H 9. It was supplemented with 0.2% glycerol and 10% ADC and was free of tyloxapol. Growing the Mycobacterium cell to OD6000.5-0.8 and the stock was frozen at-80 ℃. After thawing, cells were diluted in assay medium to a final concentration of 105cells/mL and dispensed in 50 uL/well in the plate. MTT [3- (4, 5-dimethylthiazol-2-yl) -2, 5-diphenyltetrazolium bromide](stock solution concentration 5 mg/mLAcross Organics ref 15224654) and Bactiter-Glo luciferase assay system (Promega, Madison, Wis.) were used as indicators of cell growth of the Mtb H37Rv strain. Luminescence was measured in an Envision Multilabel plate reader (PerkinElmer) using a white opaque 384-plate (781075_ Greiner) ultrasensitive luminescence mode with a measurement time of 50 milliseconds per well for the Bactiter-Glo system and OD 580nm for MTT readout in a Spectramax M5(Molecular Devices) reader using a black 384-micro transparent plate (781091_ Greiner).

Plates were incubated for 7 days, and then either ATP production (according to manufacturer's instructions) or M was measuredTT to first

Figure BDA0002263834250000291

Transformation (25 uL/well from stock on day 6, 10% SDS at day 7 25%/well). The lowest drug concentration that inhibited 90% of MTT conversion or ATP production was used to define the MIC value (IC) compared to the internal control wells without drug addition (DMSO control)90)。

Checkerboard detection

Drug activity was determined in 384-well plate format using MTT or ATP assays, as described above. The Fractional Inhibitory Concentration (FIC) of each compound was calculated as follows: FICA(MIC of compound a in the presence of compound B)/(MIC of compound a alone). Similarly, the FIC for Compound B was calculated. The FIC index (FICI) is calculated by the formula: FICI ═ FICA+FICB]. FICI ≦ 0.5 is considered to indicate synergy.

A checkerboard of mountain fetilin (sodium salt) and rifampicin was prepared. Rifampicin was diluted from column 12 to column 3 (from 0.08. mu.M to 0.00015625. mu.M) along the abscissa, while Rispenem was diluted from row A to row G (from 12.8. mu.M to 0.4. mu.M) along the ordinate.

Column 2 and row H are used to calculate each MIC (respectively, mafetinan and rifampin). Equivalent positions in quadrants 2, 3 and 4 were used to calculate ethambutol, delamasib and amoxicillin.

Dilammanib was diluted from 40nM to 0.078nM and ethambutol from 32. mu.M to 0.06. mu.M. Amoxicillin was diluted from 128. mu.M to 0.25. mu.M. They are assigned to equivalent positions in quadrant 2 and quadrant 3, respectively.

Results

The data are recorded in table 6 and fig. 5 to 8. All MIC values in table 6 are reported in μ M.

Figure BDA0002263834250000301

TABLE 6

The following terms are used in table 6.

Comp compounds

MIC minimum inhibitory concentration,. mu.M

MICsyn in the Presence of other Compounds (B or A, respectively), synergistic MIC of the test Compound (A or B)

Fold MIC/MICsyn

FIC fractional inhibitory concentration. MIC of A or B in the presence of B or A/MIC of A or B alone, respectively

FICI graded inhibitory concentration index. FIC _ a + FIC _ B. A value of 0.5 or less is considered to be the presence of a synergistic interaction

35页详细技术资料下载
上一篇:一种医用注射器针头装配设备
下一篇:细胞凋亡信号调节激酶1抑制剂及其使用方法

网友询问留言

已有0条留言

还没有人留言评论。精彩留言会获得点赞!

精彩留言,会给你点赞!