CEPHALOSPORIN COMPOUND

The cephalosporin compound of formula (I) is disclosed, which exhibits antibiotic activity against Gram-negative (e.g., Pseudomonas aeruginosa) and Gram-positive (e.g., methicillin-resistant Staphylococcus aureus) bacteria. Methods of manufacturing the compound of formula (I), and uses of the compound in the preparation of pharmaceutical compositions and antibacterial applications are also disclosed.

Skip to: Description  ·  Claims  · Patent History  ·  Patent History
Description
RELATED APPLICATIONS

The present application is a continuation application of U.S. application Ser. No. 14/020,230, filed Sep. 6, 2013, which claims priority to U.S. Provisional Application No. 61/698,241, filed on Sep. 7, 2012. The entire contents of these applications are incorporated herein by reference in their entirety.

TECHNICAL FIELD

This disclosure is directed to a cephalosporin compound which is useful as an antibiotic, as well as pharmaceutical compositions comprising the compound, methods of using the compound as an antibacterial agent, and processes and intermediates for preparing the compound.

BACKGROUND

A variety of cephalosporin derivative compounds with various substitutions on a beta-lactam core have antibacterial activity. Cephalosporin compounds with a quaternary ammonium group at the 3-position of the cephalosporin beta lactam core structure and an aminothiazole/oxime structure at the 7-position can provide antibacterial activity against multiple types of bacteria, including Gram-negative bacteria Pseudomonas aeruginosa (Pa). For example, ceftazidime and cefpirome, which include the cephalosporin core with an aminothiazolyl group at the 7-position and a quaternary salt substituent at the 3-position, have antibacterial activity against a wide spectrum of bacteria from Gram-positive bacteria in addition to Pseudomonas aeruginosa. However, even compounds such as ceftazidime and cefpirome may not be satisfactory in the antibacterial activity against both Pseudomonas aeruginosa and methicillin-resistant Staphylococcus aureus (MRSA). In addition, infectious diseases caused by methicillin-resistant Staphylococcus aureus (MRSA) continue to present significant clinical challenges. There remains a need for novel cephalosporin antibiotics which have improved antibacterial activity also against these and other bacteria.

SUMMARY

A cephalosporin compound with antibacterial activity against Gram-negative and Gram-positive bacteria, including Pseudomonas aeruginosa and methicillin-resistant Staphylococcus aureus (MRSA), is disclosed. In particular, this disclosure provides a cephalosporin compound of formula (I) having antibacterial activity against both Pseudomonas aeruginosa and methicillin-resistant Staphylococcus aureus (MRSA), for example as measured according to Minimum Inhibitory Concentrations (MICs) as measured by the method of Examples 3-5.

Provided herein is a cephalosporin compound having the structure:

The cephalosporin compound of formula (I) has antibacterial activity against both Gram-negative bacteria and Gram-positive bacteria and is useful in treating a bacterial infection in a host, such as a human or other mammal. For example, pharmaceutical compositions comprising formula (I), or a pharmacologically acceptable salts thereof, can be independently effective against both Gram-negative bacteria such as Pseudomonas aeruginosa and Gram-positive bacteria such as Staphylococcus aureus—including MRSA—with a MIC of not more than about 4 micrograms/mL (as measured according to the method of Example 3). Methods of treating bacterial infections can include administering to an infected host a pharmaceutical composition comprising an antibacterially effective amount of the compound of formula (I), or a pharmacologically acceptable salt thereof.

The compound of formula (I) can be prepared by a variety of synthetic routes, including synthetic schemes described herein. These synthetic routes can be applied to large scale synthesis with appropriate adjustment of reaction sequence, reaction conditions, isolation/purification methods and choice of solvents which are environmentally friendly and cost-effective.

The term “therapeutically effective amount,” as used herein refers to a total administered amount of an antibacterial compound that is effective to perform the function being sought by the researcher or clinician without unduly harming the tissues of the subject to which the agent is administered.

The term “subject,” as used herein, refers to an animal, a plant, or a cell culture. In one embodiment, a subject is a human or other animal patient in need of antibacterial treatment.

The following abbreviations have the following meanings unless otherwise indicated. Abbreviations not defined below have their generally accepted meaning.

Boc=tert-butoxycarbonyl

Boc2O=di-tert-butyldicarbonate

CDI=1,1′-Carbonyldiimidazole

CFU=colony-forming units

CLSI=Clinical Laboratory Standards Institute

cSSSI=complicated skin and skin structure infections

DCC=N,N′-Dicyclohexylcarbodiimide

DCM=dichloromethane

DIPEA=diisopropylethylamine

DMF=N,N-dimethylformamide

DMSO=dimethyl sulfoxide

DPPA=diphenylphosphoryl azide

EDCl=1-ethyl-3-(3′-dimethylaminopropyl)carbodiimide

HCl=hydrochloride

EtOAc=ethyl acetate

ESI-MS=Electrospray ionization mass spectrometry

HAP=Hospital-Acquired Pneumonia

HATU=2-(7-Aza-1H-benzotriazole-1-yl)-1,1,3,3-tetramethyluronium hexafluorophosphate

HCl=hydrochloride

HOBT=1-hydroxybenzotrizole

hrs=hours

HPLC=High performance liquid chromatography

Hunig's base=N,N-Diisopropylethylamine

mCPBA=meta-Chloroperoxybenzoic acid

MIC=Minimum inhibitory concentration

mL=milliliter

MS=Mass spectrometry

MRSA=Methicillin-resistant Staphylococcus aureus

NCS=N-Chlorosuccinimide

NMR=nuclear magnetic resonance

Pa=Pseudomonas aeruginosa

Pd2(dba)3=Tris(dibenzylideneacetone)dipalladium(0).

PdCl2(dppf)=Pd[1,1-bis (diphenylphosphino) ferrocene]dichloropalladium(II)

PFPOH=pentafluorophenol

PMB=para-methoxybenzyl

Prep=Preparative

ppm=parts per million

rt=room temperature

THF=tetrahydrofuran

TFA=trifluoroacetic acid

TMS=tetramethylsilane

TLC=thin layer chromatography

VAP=Ventilator-Associated Pneumonia

Xantphos=4,5-Bis(diphenylphosphino)-9,9-dimethylxanthene

BRIEF DESCRIPTION OF THE DRAWINGS

FIG. 1 depicts the efficacy of the compound of formula (I) against MRSA #712 thigh infection compared with vancomycin in neutropenic mice.

FIG. 2 depicts the efficacy of the compound of formula (I) against ceftazidime-susceptible pseudomonas aeruginosa #44 (ATCC#27853) lung infection compared with ceftazidime and primaxin in neutropenic mice.

FIG. 3 depicts the efficacy of the compound of formula (I) against ceftazidime-susceptible pseudomonas aeruginosa #2245 lung infection compared with ceftazidime and primaxin in neutropenic mice.

FIG. 4 depicts the efficacy of the compound of formula (I) against ceftazidime-resistant pseudomonas aeruginosa #2545 lung infection compared with ceftazidime and primaxin in neutropenic mice.

FIG. 5 depicts the synthesis of the compound of formula (I).

FIG. 6 depicts the distribution of MICs of the compound of formula (I) against MRSA and Pa strains with comparators.

DETAILED DESCRIPTION

Provided herein is a compound of formula (I), or a pharmaceutically acceptable salt thereof, which is useful for treating bacterial infections. In particular, pharmaceutical compositions comprising the cephalosporin compound of formula (I) have a surprisingly broad spectrum of antibacterial activity and are useful to inhibit or kill various bacteria. For example, the antibacterial compound disclosed herein can be used in the manufacture of antibacterial medicaments and in methods for the treatment of bacterial infections, including treatment of both certain Gram-positive and Gram-negative bacterial infections. The cephalosporin compound of formula (I), or a pharmacologically acceptable salt thereof has the following structure:

The compound of formula (I) has a MIC (measured by the method of Example 3) against Pseudomonas aeruginosa and methicillin-resistant Staphylococcus aureus (MRSA) of less than or equal to about 1-2 micrograms/mL.

Also included in formula (I) are zwitterionic forms containing an anionic carboxylic moiety and the cationic nitrogen in the pyridinium moiety. The imino group in the C-7 side chain position of the cephalosporin fused ring structure of formula (I) are shown in the “syn” (Z) configuration. The compound of formula (I) are preferably prepared as the syn-isomer (Z-isomer), or in mixtures comprising an amount of the syn-isomer (e.g., at least 90%) to provide a desired level of antibacterial activity, including compound of formula (I) that are syn isomers which are essentially free of the corresponding anti imino isomers.

The antibacterial properties of this compound based on minimum inhibitory concentrations (MICs) against certain Staphylococcus and Pseudomonas strains is listed in micrograms/mL which can be compared to comparable MICs measured for the compounds of Table 1.

The compound of formula (I) can be prepared from cephalosporin intermediate compound 4a as described in the Examples, or following literature procedures. For similar literature procedures, see, e.g., (a) Lattrell, R.; Blumbach, J.; Duerckheimer, W.; Fehlhaber, H. W.; Fleischmann, K.; Kirrstetter, R.; Mencke, B.; Scheunemann, K. H.; Schrinner, E.; et al. J. of Antibiotics 1988, 41, 1374. (b) Lattrell, R.; Blumbach, J.; Duerckheimer, W.; Fleischmann, K.; Kirrstetter, R.; Klesel, N.; Mencke, B.; Scheunemann, K. H.; Schwab, W.; et al. J. of Antibiotics 1988, 41, 1395. (c) Hanaki, H.; Yamazaki, H.; Harada, H.; Kubo, R.; Kobayashi, T.; Atsuda, K.; Sunakawa, K. J. of Antibiotics 2005, 58, 69-73; (d) Long, D. D.; Aggen, J. B.; Chinn, J.; Choi, S.-K.; Christensen, B. G.; Fatheree, P. R.; Green, D.; Hegde, S. S.; Judice, J. K.; Kaniga, K.; Krause, K. M.; Leadbetter, M.; Linsell, M. S.; Marquess, D. G.; Moran, E. J.; Nodwell, M. B.; Pace, J. L.; Trapp, S. G.; Turner, S. D. J. of Antibiotics 2008, 61, 603-614, all of which are incorporated by reference in their entireties.

The antibiotic activity of the compound of formula (I) was measured against a variety of pathogenic microorganisms including Gram-positive and Gram-negative bacteria, including against Staphylococcus and Pseudomonas bacteria. Minimum inhibitory concentrations (MICs) for the compound of formula (I) against certain Staphylococcus, Pseudomonas and pneumoniae strains are listed in micrograms/mL in Table 2. The MICs were determined as described in Example 4. Accordingly, the compound of formula (I) can be included in pharmaceutical antibacterial compositions and are useful both in the manufacture of medicaments for treating bacteria or bacterial infections, and in methods of treating conditions caused by bacteria such as infections.

Pharmaceutical antibacterial compositions can be formed by combining the compound of formula (I) or a pharmacologically acceptable salt thereof with a pharmacologically acceptable carrier suitable for delivery to a recipient subject (e.g., a human) in accordance with known methods of drug delivery. Antibacterial pharmaceutical compositions suitable for administration of one or more compound of formula (I) can be formulated. The compound of formula (I), and/or pharmacologically acceptable salts of formula (I) can be included in a pharmaceutical antibacterial composition along with one or more carriers.

Pharmaceutical compositions can be formed by combining the compound of formula (I) or a pharmacologically acceptable salt thereof with a pharmacologically acceptable carrier suitable for delivery to a recipient subject (e.g., a human) in accordance with known methods of drug delivery. Antibacterial pharmaceutical compositions suitable for administration of one or more compound of formula (I) can be formulated. The compound of formula (I), and/or pharmacologically acceptable salts of formula (I) can be included in a pharmaceutical antibacterial composition along with one or more carriers. Pharmaceutical preparations can be prepared in accordance with standard procedures and are administered at dosages that are selected to treat infection (see, e.g., Remington's Pharmaceutical Sciences, Mack Publishing Company, Easton, Pa. and Goodman and Gilman's “The Pharmaceutical Basis of Therapeutics,” Pergamon Press, New York, N.Y., the contents of which are incorporated herein by reference, for a general description of the methods for administering various antimicrobial agents for human therapy).

The compound of formula (I) can be formulated as a variety of salts to improve stability or toxicological properties of the compound, increase or decrease solubility, improve pharmacokinetic performance of the compound (e.g., Cmax or AUC measurements) or improve storage properties (e.g., to reduce hygroscopicity) of a pharmaceutical composition. As used herein, the term “pharmaceutically-acceptable salt” refers to pharmaceutical salts that are, within the scope of sound medical judgment, suitable for use in contact with the tissues of humans and lower animals without undue toxicity, irritation, and allergic response, and are commensurate with a reasonable benefit/risk ratio. Pharmaceutically-acceptable salts are well known in the art. For example, S. M. Berge, et al. describes pharmacologically acceptable salts in J. Pharm. Sci., 1977, 66:1-19. Pharmaceutically-acceptable salts of compound of formula (I) may be prepared by conventional means from the corresponding compound of the invention by treating, for example, the compound with the appropriate acid or base. Examples of publications describing the selection and formation of pharmacologically acceptable salts of medicinal compounds include Haynes, Delia A., et al., “Occurrence of Pharmacologically acceptable Anions and Cations in the Cambridge Structural Database,” Journal of Pharmaceutical Sciences, v. 94, no. 10, 2111-2120 (October 2005), and Stahl, P H, et al., Eds., “Handbook of Pharmaceutical Salts: Properties, Selection and Use,” Weinheim/Zurich, Wiley-VCH/VHCA.

A pharmaceutical composition can include a pharmaceutically-acceptable carrier and the compound of formula (I) and/or salts of formula (I). As used herein, the phrase “pharmaceutically-acceptable carrier” refers generally to solvents, dispersion media, excipients, coatings, matrices, stabilizers, buffers, absorption enhancers, adjuvents, controlled release media, and the like, that are compatible with an intended use, such as pharmaceutical administration. The use of such carriers for pharmaceutically active substances is well known in the art. The pharmaceutical compositions can be formulated for parenteral delivery, including intravenous, intramuscular, intraperetoneal, subcutaneous, intraocular, intrathecal, intra-articular, intra-synovial, cisternal, intrahepatic, intralesional and intracranial injection, infusion, and/or inhaled routes of administration for the therapeutic treatment of medical conditions, such as bacterial infections.

Pharmaceutical compositions for parenteral injection can comprise pharmaceutically-acceptable aqueous or nonaqueous solutions of antibacterial compound of formula (I) in addition to one or more of the following: pH buffered solutions, adjuvants (e.g. preservatives, wetting agents, emulsifying agents, and dispersing agents), liposomal formulations, nanoparticles, dispersions, suspensions or emulsions as well as sterile powders for reconstitution into sterile injectable solutions or dispersions just prior to use. For intravenous (IV) use, the pharmaceutical composition can include any of the commonly used intravenous fluids and administered by infusion, such as physiological saline or Ringer's solution.

The antibacterial compound of formula (I) disclosed herein, and pharmacologically acceptable salts thereof, including pharmaceutical compositions comprising these compounds, is useful in the manufacture of antibacterial pharmaceutical compositions, and treatment of bacteria. Significantly, the antibacterial compounds are useful in treating and eliminating a broad spectrum of bacterial pathogens, including both gram negative and gram positive bacterial infections. The antibacterial compound of formula (I) can be used in vivo, for example, to treat bacterial infections in a subject, as well as in vitro, for example to treat cells (e.g., bacteria) in culture to eliminate or reduce the level of bacterial contamination of a cell culture. In one embodiment, the compound of formula (I), or a pharmaceutical composition thereof, is administered to a cell culture, such as by administering in a nutrient medium. Methods of treating bacterial infections in subjects (e.g., humans and animals) can include the administration of a therapeutically effective amount of the compound of formula (I) or a pharmacologically acceptable salt thereof.

Methods of treatment of such infections include administering to a subject in need thereof a therapeutically effective amount of the compound of formula (I). The compound can be parenterally administered to a subject having or suspected to have a bacterial infection, such as a gram negative infection.

The antibacterial compound of formula (I) is preferably used in vivo to treat an infection in a subject by administering a therapeutically effective amount of the compound of formula (I) in a pharmaceutical composition. The method can comprise parenterally administering to a subject in need thereof a pharmaceutical composition comprising a therapeutically effective dose of at least one compound of formula (I). Pharmaceutical compositions include compositions comprising compound(s) of formula (I) in a dose sufficient to achieve the intended purpose, i.e., the treatment or prevent of infectious diseases. The amount and concentration of antibacterial compound of formula (I) in the pharmaceutical composition, as well as the quantity of the pharmaceutical composition administered to a subject, can be selected based on clinically relevant factors, such as medically relevant characteristics of the subject (e.g., age, weight, gender, other medical conditions, and the like), the solubility of the antibacterial compound in the pharmaceutical composition, the potency and activity of the antibacterial compound, any toxicity associated with the pharmaceutical composition dose and method of administration, and the manner of administration of the pharmaceutical composition.

A pharmaceutical composition comprising a therapeutically effective dose of the compound of formula (I) can be administered intravenously to a patient for treatment of gram negative infections in a clinically safe and effective manner, including one or more separate administrations of the composition. The total daily dose of the compound of formula (I) can be about 2.0 mg/kg/day to about 50 mg/kg/day of all compound of formula (I) administered intravenously to a subject one to three times a day (e.g., QD, BID or TID). The amount per administered dose or the total amount administered will depend on such factors as the nature and severity of the infection, the age and general health of the patient, the tolerance of the patient to the compound and the microorganism or microorganisms involved in the infection. The total amount of the pharmaceutical composition administered can be selected to be therapeutically effective.

In particular, the pharmaceutical compositions comprising the compound of formula (I) can be used to treat a subject having a bacterial infection in which the infection is caused or exacerbated by a gram-negative bacteria. A method of treating a bacterial infection in a host can include administering to an infected host a pharmaceutical composition comprising a therapeutically effective dose (e.g., an antibacterially effective amount) of the compound of formula (I), or a pharmacologically acceptable salt thereof.

The pharmaceutical composition and/or compound(s) of formula (I) can be administered to treat a bacterial infection or population in vitro (e.g., a bacterial colony on a surface outside the body) or in vivo (e.g., within an infected host). The bacterial infection or population can include Gram-negative and/or Gram-positive bacteria. For example, the Gram-negative bacteria can comprise Pseudomonas aeruginosa and the Gram-positive bacteria can include Staphylococcus aureus. The bacteria can form an infection present in vitro (e.g., a bacterial colony or sample), or in vivo (e.g., an infected host subject). For instance, the bacterial infection can be identified as a methicillin-resistant Staphylococcal infection. Preferably, the antibacterially effective dose and therapeutically effective amount of the compound(s) of Formula (I) can be effective in killing both a Pseudomonas aeruginosa bacteria and a MRSA bacteria with a MIC that is less than the comparable value for a compound in Table 1, and/or independent MIC values measured according to Example 3 that is not more than about 4 micrograms/mL (e.g., including values of about 3.5, 3.0, 2.5, 2.0, 1.5, 1.0, 0.5 micrograms/mL or lower) independently measured against each of Pseudomonas aeruginosa bacteria and methicillin-resistant Staphylococcal bacteria.

A pharmaceutical composition containing a therapeutically effective amount of the compound of formula (I) can be administered to a patient presenting symptoms of an infection. The compounds of this invention can be used to treat infections of cSSSI and HAP/VAP in adults or children. In particular methicillin resistant Staphylococcus aureus infections and/or Pseudomonas infections may be treated with the compound of formula (I). Other representative types of infections or bacteria-related medical conditions which can be treated or prevented with pharmaceutical compositions comprising antibacterial compound of formula (I) include, but are not limited to, skin and skin structure infections, urinary tract infections, pneumonia, endocarditis, catheter-related blood stream infections, osteomyelitis, and the like. In treating such conditions, the patient may treated with the pharmaceutical compositions comprising the compound of formula (I) upon presenting symptoms consistent with a bacterial infection. The pharmaceutical compositions may be administered prior to or after identifying the types of bacteria present in the patient.

EXAMPLES

The specific examples which follow illustrate the synthesis of certain compounds used in the preparation of the compound of formula (I). Further, the disclosure includes variations of the methods described herein to produce the compound of formula (I) that would be understood by one skilled in the art based on the instant disclosure.

All temperatures are understood to be in Centigrade (C) when not specified. The nuclear magnetic resonance (NMR) spectral characteristics refer to chemical shifts (γ) expressed in parts per million (ppm) versus tetramethylsilane (TMS) as reference standard. The relative area reported for the various shifts in the proton NMR spectral data corresponds to the number of hydrogen atoms of a particular functional type in the molecule. The nature of the shifts as to multiplicity is reported as broad singlet (br s), broad doublet (br d), singlet (s), multiplet (m), doublet (d), quartet (q), doublet of doublet (dd), doublet of triplet (dt), and doublet of quartet (dq). The solvents employed for taking NMR spectra are DMSO-d6 (perdeuterodimethysulfoxide), D2O (deuterated water), CDCl3 (deuterochloroform) and other conventional deuterated solvents. The prep-HPLC conditions are: Waters SunFire® C18 (30×100 mm, 5 μm) column; flow rate: 50 mL/min, UV or Mass-triggered fraction collection; sample loading: each injection loading varied from 30-80 mg depending for different crude samples depending on their solubility and purity profiles; solvent system: solvent A: water with 0.5% formic acid, solvent B: acetonitrile with 0.5% formic acid.

Example 1 Synthesis of (6R,7R)-4-methoxybenzyl 7-((Z)-2-(2-amino-5-chlorothiazol-4-yl)-2-(methoxyimino)acetamido)-3-(chloromethyl)-8-oxo-5-thia-1-azabicyclo[4.2.0]oct-2-ene-2-carboxylate (intermediate compound 4a)

Step 1: Preparation of (6R,7R)-4-methoxybenzyl 7-amino-3-(chloromethyl)-8-oxo-5-thia-1-azabicyclo[4.2.0]oct-2-ene-2-carboxylate

To a suspension of PCl5 (3.21 g) in dry DCM (50 mL) at 0° C. was added anhydrous pyridine (1.24 mL) and the resulting white suspension was stirred for 0.5 h. 7-Phenylacetamido-3-chloromethyl-3-cephem-4-carboxylic acid p-methoxy benzyl ester (5.0 g) was added. After stirring at 0° C. for additional 2 h, the reaction mixture was cooled to −40° C. and methanol (15 mL) was added dropwise. The stirring was continued at −30° C. for additional 0.5 h, and the reaction mixture was then concentrated. To the residue were added water (5 mL), EtOAc (20 mL) and diethyl ether (200 mL), and the mixture was stirred at 0° C. until precipitation formed. The yellow precipitate was collected by filtration, rinsed with diethyl ether and then 20% DCM in diethyl ether, and dried under high vacuum. The resulting light yellow solid was used directly in step 3. ESI-MS (EI+, m/z): 369.05.

Step 2: Synthesis of (Z)-2-(2-amino-5-chlorothiazol-4-yl)-2-(methoxyimino)acetic acid

To a solution of (Z)-2-(2-aminothiazol-4-yl)-2-(methoxyimino)acetic acid (15.0 g) in anhydrous DMF (150 mL) was added NCS (11.95 g) and the mixture was stirred at rt for 2-3 hrs. The reaction mixture was concentrated to remove most of the DMF. The resulting oil was added dropwise to DCM (˜600 mL) at 0° C. with stirring. Solid precipitation was collected by filtration, rinsed with DCM and dried under high vacuum to provide a brownish solid (11.8 g, 67%). ESI-MS (EL, m/z): 236.0. 1H NMR (300 MHz, DMSO-d6) δ 7.60 (br s, 2H), 3.88 (s, 3H).

Step 3: Synthesis of Compound 4a via EDCI Coupling

To a solution of (6R,7R)-4-methoxybenzyl 7-amino-3-(chloromethyl)-8-oxo-5-thia-1-azabicyclo[4.2.0]oct-2-ene-2-carboxylate (6.8 g) in dry DMF (60 mL) was added (Z)-2-(2-amino-5-chlorothiazol-4-yl)-2-(methoxyimino)acetic acid (4.17 g). The mixture was cooled to 0° C., then 2,4,6-collidine (2.3 g) and EDCI (3.4 g) were added sequentially. After 2 hrs, the solution was poured into cold water and filtered. The solid was rinsed with water, dried under high vacuum and purified by silica gel chromatography (DCM:EtOAc=3:1) to afford 4a (5.0 g, 57%). ESI-MS (EL, m/z): 586.04; 1H NMR (400 MHz, D2O) δ 9.60 (d, J=8.8 Hz, 1H), 7.39 (br s, 2H), 7.36 (d, J=8.4 Hz, 2H), 6.93 (d, J=8.4 Hz, 2H), 5.83 (dd, J=8.8, 5.2 Hz, 1H), 5.24 (d, J=12.0 Hz, 1H), 5.19 (d, J=5.2 Hz, 1H), 5.18 (d, J=12.0 Hz, 1H), 4.54 (d, J=11.4 Hz, 1H), 4.46 (d, J=11.4 Hz, 1H), 3.86 (s, 3H), 3.76 (s, 3H), 3.70 (d, J=18.0 Hz, 1H), 3.54 (d, J=18.0 Hz, 1H).

Example 2 Synthesis of tert-butyl 1-(pyridin-4-yl)hydrazinecarboxylate

A mixture of 4-bromo-pyridine hydrochloride salt (8.0 g), tert-butyl hydrazinecarboxylate (10.87 g), CuI (784 mg), L-4-hydroxyproline (1.08 g) and Cs2CO3 (33.50 g) in dry DMSO (41 mL) under N2 was heated at 80° C. with efficient stirring for 5-6 hrs. After cooled to rt, the reaction mixture was diluted with EtOAc (150 mL) and saturated NH4Cl (150 mL). The organic layer was separated and the aqueous was extracted with EtOAc (3×150 mL). The combined organic layers were washed with brine (50 mL), dried (Na2SO4) and concentrated. The residue was purified by silica gel column chromatography (DCM to 10% MeOH/DCM gradient) to provide the title compound (3.4 g, 40%). ESI-MS (EL, m/z): 210.0 [M+H]+. 1H NMR (300 MHz, DMSO) δ 8.38 (d, J=4.9 Hz, 2H), 7.64 (d, J=6.4 Hz, 2H), 5.12 (s, 2H), 3.34 (s, 1H), 1.51 (s, 9H).

Example 3 Biological Activity Assay (A)

As shown in Table 1, the antibacterial activity of the comparator compounds was demonstrated by the minimum inhibitory concentrations (MIC) of the compounds against various bacteria measured by using the broth microdilution method performed according to Clinical and Laboratory Standards Institute (CLSI) guidelines with modifications as described below (CLSI guidelines can be derived from the CLSI document M7-A8 published in January 2009: “Methods for Dilution Antimicrobial Susceptibility Tests for Bacteria That Grow Aerobically; Approved Standard-Eighth Edition”).

To prepare for MIC testing, individual colonies were isolated by streaking frozen glycerol material containing Staphylococcus or Pseudomonas spp. onto rich, non-selective, tryptic soy agar containing 5% sheep's blood (TSAB), and incubated at 37° C. for 18-24 hrs.

On the day of testing, primary cultures were started by scraping off 5-10 colonies from the TSAB plates. The material was suspended in ˜5 mL of cation adjusted Mueller Hinton Broth (CAMHB) in 14 mL culture tubes and incubated at 37° C. with aeration (200 rpm) for ˜2 hrs until the OD600 was ≧0.1.

Inoculum cultures were prepared by standardizing the primary cultures to OD600=0.1 and then adding 20 μL of the adjusted primary culture per 1 mL CAMHB for Pseudomonas and CAMHB plus 4% NaCl for MRSA so that the final inoculum density was ˜105 colony forming units per milliliter. Diluted inoculum cultures were used to inoculate 50 μL per well in 96 well broth microdilution assay plates. 50 μL of CAMHB that contained compound concentrations ranging from 64-0.06 μg/mL in two-fold dilutions was also added to the broth microdilution assay plates for a final volume 100 μL per well, therefore final culture OD600 was approximately 0.001 and the final NaCl concentration for the MRSA strain was 2%.

Plates were incubated for 18-20 hours at 37° C. with aeration (200 rpm). Following incubation, growth was confirmed visually placing plates over a viewing apparatus (stand with a mirror underneath) and then OD600 was measured using a SpectraMax 340PC384 plate reader (Molecular Devices, Sunnyvale, Calif.). Growth was defined as turbidity that could be detected with the naked eye or achieving minimum OD600 of 0.1. MIC values were defined as the lowest concentration producing no visible turbidity.

Example 4 Biological Activity Assay (B)

As shown in Table 2, the efficacy of the compound of formula (I) was evaluated in mouse septicemia, thigh and lung infection models against multiple strains of MRSA, ceftazidime-susceptible and -resistant Pseudomonas aeruginosa and ceftazidime-resistant Klebsiella pneumoniae and compared with standard antibiotic treatment.

I. Summary

    • The compound of formula (I) is a novel broad spectrum cephalosporin with potent in vitro activities against G(+) and G(−) bacteria including MRSA and Pseudomonas aeruginosa (Pa). In vivo efficacy of (I) was evaluated against strains of MRSA, and ceftazidime-resistant Pa and Klebsiella pneumonia (Kpn) in mouse septicemia, thigh and lung infections. Septicemia: CD-1 mice received lethal bacterial inocula IP, followed by 2 doses of antibiotics SC at 1 and 6 hrs post-inoculation. Dose calculated to protect 50% of the mice (PD50) was derived based on probit analysis of percentage survival. Thigh and lung infections: Thighs or lungs of neutropenic mice were inoculated with MRSA or Pa. Two treatment doses were given SC at 1 and 6 hrs post-inoculation. Tissues were harvested at ˜24 hrs post-inoculation and bacterial burdens quantified. Efficacious dose resulting in a 2-log bacterial CFU reduction (ED−2log) compared with vehicle-treated mice was calculated. (I) demonstrated potent in vivo activity against several drug-resistant strains. Its in vivo potency against a clinical isolate of MRSA was comparable to vancomycin and ceftaroline. (I) also had good activities against ceftazidime-resistant Pa and Kpn. (I) exhibits excellent in vivo efficacy against infections caused by MRSA, and ceftazidime-resistant Pa and Kpn in mice.

II. Methods

    • A. Animals: Six week-old, specific pathogen free, female CD-1 mice purchased from Charles River Laboratories (Wilmington, Mass.) were used for all studies after acclimation in Cubist animal facility. All animal procedures were approved by the Cubist Institutional Animal Care and Use Committee.
    • B. Bacteria and In Vitro Susceptibility Testing: MRSA #712, Pseudomonas aeruginosa #2245 and #2545, and Klebsiella pneumoniae #573 are clinical isolates; Pseudomonas aeruginosa #44 is an ATCC strain (ATCC#27853). MIC was determined in MHB by standard CLSI microdilution. MICs of individual strains are shown in Table 2 and FIGS. 1-4.
    • C. Comparator Antibiotics Used in Animal Studies: Vancomycin hydrochloride USP was purchased from Hospira (Lake Forest, Ill.); ceftazidime hydrate from Sigma; Primaxin (imipenem and cilastatin) from Merck; ceftaroline was synthesized at a CRO.
    • D. Mouse Septicemia: Mice received lethal inocula of bacteria in 6% hog gastric mucin intraperitoneally (IP), followed by two treatment doses of antibiotics via subcutaneous (SC) administration at 1 and 6 hrs post-inoculation. The survival of animals was recorded daily for 7 days. The antibiotic dose calculated to protect 50% of the mice (PD50) was derived based on probit analysis of percentage survival.
    • E. Mouse Thigh and Lung Infections: Mice were rendered neutropenic by two IP injections of cyclophosphamide 4 days (150 mg/kg) and 1 day (100 mg/kg) before bacterial inoculation. Thigh or lung infection was induced by an intramuscular injection of 0.2 mL or intranasal inoculation of 0.1 mL inoculum preparation from an overnight bacterial culture into left thigh or nares. Tissues from one group of mice receiving no treatment (No Rx) were harvested at 1 hr for determination of baseline of bacterial burden. The remaining mice received two treatment doses of vehicles or antibiotics given SC at 1 and 6 hrs post-inoculation. Tissues from the treated mice were harvested at ˜24 hrs post-inoculation. Tissue bacterial burdens were quantified by serial dilution and spiral plating of homogenates. Doses of (I) were confirmed analytically. The efficacious dose resulting in a 2-log or a 3-log bacterial CFU reduction (ED—log or ED−3log) as compared with vehicle-treated mice at 24 h was derived by linear regression analysis.

III. Results

    • A. (I) displayed potent efficacy in all systemic infection models. Its PD50 value of 2.0±0.3 mg/kg against MRSA #712 septicemia was comparable to vancomycin and ceftaroline. (I) was significantly more potent in vivo than ceftazidime against the CLSI reference strain of Pseudomonas aeruginosa #44 (PD50: 5.3±0.7 vs. 33.9±7.4 mg/kg, respectively. (I) demonstrated good in vivo potency against ceftazidime-resistant Pseudomonas aeruginosa #2545 and Klebsiella pneumoniae #573 systemic infections (Table 1).
    • B. (I) led to a dose-dependent reduction of MRSA #712 burden in the thigh vs. vehicle controls with an ED−2log of 68.9 mg/kg (FIG. 1). In contrast, vancomycin at 110 mg/kg SC resulted in a smaller reduction in bacterial CFU (˜1.5 log). The vancomycin dose was chosen based on PK studies showing that the plasma exposure at this dose level in mice (data on file) was approximately equivalent to that in humans.
    • C. (I) showed marked efficacy against ceftazidime-susceptible Pseudomonas aeruginosa #44 and #2245 lung infections with ED−3log values of 4.1 and 5.2 mg/kg, respectively. Ceftazidime had higher ED−3log values of 50.4 and 27.2 mg/kg, respectively (FIGS. 2 and 3). Bacterial burden in the lung at the highest doses of (I) in these two studies approached the lower limit of detection of the assay.
    • D. (I) was efficacious against a ceftazidime-resistant Pseudomonas aeruginosa #2545 lung infection (FIG. 4). Its ED−3log value was 11.4 mg/kg. In contrast, ceftazidime up to144.5 mg/kg had minimal impact on the bacterial burden in the lung.

IV. Conclusions

    • A. (I) exhibited excellent efficacy against systemic and tissue infections caused by MRSA and ceftazidime-resistant Pseudomonas aeruginosa and Klebsiella pneumoniae in mice.
    • B. In vivo efficacy of (I) against a clinical isolate of MRSA was comparable to both vancomycin and ceftaroline in the thigh infection model in neutropenic mice.
    • C. (I) appeared more efficacious than ceftazidime in lung infections caused by ceftazidime-susceptible Pseudomonas strains in neutropenic mice.
    • D. (I) maintained potent activity vs. a mouse lung infection induced by a ceftazidime-resistant Pseudomonas isolate.

Example 5 Biological Activity Assay (C)

I. Summary

    • The 3rd and 4th generation cephalosporins bearing pyridinium or quaternary ammonium groups at the C-3′ position, such as ceftazidime (CAZ) and cefepime, have potent activity against Gram-negative bacteria including Pseudomonas aeruginosa (Pa). However, their activity against Gram-positive bacteria such as methicillin-resistant Staphylococcus aureus (MRSA) is weak, limiting their clinical usefulness for empiric treatment. Thus, a broad spectrum cephalosporin with improved activity against both Pa and MRSA would be of particular interest. The compound of formula (I) demonstrated potent in vitro activity and broad spectrum coverage against both Gram-positive and Gram-negative bacteria, including MRSA and Pa. The MIC90 of the compound of formula (I) is equivalent to that of CAZ against Pa, and comparable to those of ceftaroline and vancomycin against MRSA. The compound of formula (I) has potent antibacterial activity against serious bacterial pathogens such as Pa and MRSA.

II. Biological Activity Assay

    • A. The antibacterial activity of the compounds in Table 3 were demonstrated by the minimum inhibitory concentrations (MIC) using the broth microdilution method performed according to Clinical and Laboratory Standards Institute (CLSI) guidelines with minor modifications.
    • B. MIC values were defined as the lowest concentration producing no visible turbidity. MIC90 values were defined as the concentration required to inhibit growth of 90% of the strains tested.
    • C. SAR was performed using the following isolates: Sa399 (ATCC43300, MRSA), Sa1721 (NARSA NRS384, MRSA USA 300), Pa44 (ATCC27853), and 4 clinical isolates (Pa2241, Pa2086 (mucoid strain), Pa2694 and Pa2698).
    • D. The in vitro antibacterial activity of the compound of formula (I) was further evaluated in an expanded MRSA and Pa MIC panel (see Table 4 and FIG. 6).

III. Efficacy Study

    • A. A mouse systemic infection model was used to determine in vivo efficacy. An overnight culture of S. aureus 712 (MRSA, clinical isolate) or Pa44 (ATCC 27853) suspended in 6% hog gastric mucin to prepare the inoculum. Mice were challenged via intraperitoneal (IP) inoculation of the bacteria with a lethal dose of 2×107 CFU/mouse.
    • B. Mice were given subcutaneous (SC) doses of test compound or saline at 1 and 6 hours after bacterial inoculation. The dose that protected 50% of the mice (PD50) was calculated by probit analysis based on percentage of mice surviving for 7 days post-inoculation.
    • C. Compounds were formulated in a dosing solution of saline or D5W.

Example 6 Synthesis of (I)

As shown in FIG. 5, cephalosporin core intermediate 6 was synthesized by amide coupling of 7-aminocephalosporin derivative 3 and chloroaminothiazole derivative 5. Urea tail 12 was prepared by CDI mediated urea formation of aniline derivative 8 and aminopyridine derivative 11. Coupling of intermediate 6 and urea tail 12, followed by global deprotection using TFA/anisol provided the final product (I), which was purified by reverse phase HPLC using 0.1% formic acid buffer in water/acetonitrile.

The examples and illustrative embodiments described herein are provided by way of illustration, and do not constitute additional limitations on the scope of the claims. While some embodiments have been shown and described in the instant specification, the specification as ready by one of ordinary skill in the relevant arts also discloses various modifications and substitutions of embodiments explicitly disclosed herein. The exemplary embodiments from the specification are not provided to read additional limitations into the claims.

TABLE 1 MICs (μg/mL) of Comparator Compounds Sa399 Pa44 Compound Description Structure (MRSA) (WT Pa) XX- 990,020 Ceftazidime C A XX- 184,831 Cefpirome B A Cefepime Cefepime C A 1-1 C A 1-2 A C 1-3 B C 1-4 B C (A: ≦ 4; B: 8-16; C: > 16)

TABLE 2 PD50 and ED-2log of (I) in Mouse Systemic, Thigh and Lung Infections Septicemia PD50 Thigh (T) and Bacterial (I) or (±SE, Lung (L) MIC Strain Comparator mg/kg) ED2log (mg/kg) (μg/ml) MRSA #712 (I) 2.0 ± 0.3 68.9 (T) 2 Ceftaroline 1.0 ± 0.1 ND 1 Vancomycin 2.0 ± 1.3 >110 (T) 1 Pa #44 (I) 5.3 ± 0.7 1.8 (L) 1 (ATCC27853) Ceftazidime 33.9 ± 7.4  25.6 (L) 1 Primaxin <2 <8 (L) 1 Pa #2545 (I) <10.4 6.7 (L) 2 Ceftazidime >140.5 >144.5 (L) 32-64 Primaxin >4 <8 (L) ND Kpn #573 (I) <1.3 ND 1 Ceftazidime 50.3 ± 8.4  ND 256 Primaxin 1.0 ± 0.6 ND 0.12

TABLE 3 In vitro activities of the compound of formula (I) and comparators Com- MIC (μg/ml) pound MRSA Pseudomonas (I) Sa399 Sa1721 Pa44 Pa2241 Pa2086 Pa2694 Pa2698 Ceftaro- 0.5 2 8 16 16 >64 >64 line Ceftazi- 64 >64 2 2 2 8 8 dime (I) 2 8 2 1 8 8 8

TABLE 4 MIC90 of the compound of formula (I) against MRSA and Pa with comparators MIC90 (μg/mL) MRSA Pseudomonas aeruginosa Compound (n = 21) (n = 100) Ceftazidime 8 Meropenem 4 Ceftaroline 1 Vancomycin 1 (I) 2 8

Claims

1-3. (canceled)

4. A method for treating an infectious disease comprising administering to a subject in need thereof a therapeutically effective amount of a composition comprising the compound of Formula (I):

or a pharmaceutically acceptable salt thereof.

5-9. (canceled)

10. The method of claim 4, wherein the infectious diseases is caused by a Gram-negative bacteria.

11. The method of claim 10, wherein the Gram-negative bacteria is Pseudomonas aeruginosa.

12. The method of claim 4, wherein the subject has hospital acquired pneumonia (HAP).

13. The method of claim 4, wherein the subject has ventilator acquired pneumonia (VAP).

14. A method of treating an infection caused by Pseudomonas aeruginosa, comprising parenterally administering to a patient in need thereof a pharmaceutical composition comprising a carrier and a therapeutically effective amount of a compound of Formula (I):

a physiologically hydrolyzable ester thereof, a solvate thereof, or a pharmacologically acceptable salt thereof.

15. The method of claim 14, wherein the therapeutically effective amount is effective against a Pseudomonas aeruginosa bacteria with a minimum inhibitory concentration (MIC) of not more than about 4 micrograms/mL.

16. The method of claim 15, wherein the minimum inhibitory concentration (MIC) is not more than about 2 micrograms/mL.

17. The method of claim 15, wherein the minimum inhibitory concentration (MIC) is not more than about 1 microgram/mL.

18. The method of claim 15, wherein the subject has hospital acquired pneumonia (HAP).

19. The method of claim 15, wherein the subject has ventilator acquired pneumonia (VAP).

20. A method of making the compound of Formula (I) comprising the step of reacting intermediate compound 6:

with an intermediate compound 12:
to produce the compound of Formula (I):
Patent History
Publication number: 20150025053
Type: Application
Filed: Jul 17, 2014
Publication Date: Jan 22, 2015
Applicant: CUBIST PHARMACEUTICALS, INC. (Lexington, MA)
Inventors: Yong He (Bedford, MA), Yu Gui Gu (Acton, MA), Ning Yin (Lexington, MA)
Application Number: 14/333,966
Classifications
Current U.S. Class: 3-position Substituent Contains Pyridine Ring (514/203); 7-amino Cephalosporanic Acid Per Se Or Salt Thereof (i.e., 7-aca Or Salt Thereof) (540/219)
International Classification: C07D 501/60 (20060101); C07D 501/04 (20060101);