EP1940417A2 - Méthode de traitement d'une diarrhée associée à clostridium difficile - Google Patents

Méthode de traitement d'une diarrhée associée à clostridium difficile

Info

Publication number
EP1940417A2
EP1940417A2 EP06826544A EP06826544A EP1940417A2 EP 1940417 A2 EP1940417 A2 EP 1940417A2 EP 06826544 A EP06826544 A EP 06826544A EP 06826544 A EP06826544 A EP 06826544A EP 1940417 A2 EP1940417 A2 EP 1940417A2
Authority
EP
European Patent Office
Prior art keywords
mixture
weight
tiacumicin
mcc
compound
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Withdrawn
Application number
EP06826544A
Other languages
German (de)
English (en)
Other versions
EP1940417A4 (fr
Inventor
Pamela Sears
Youe-Kong Shue
Starr Louise Miller-Shangle
Robert Brian Walsh
Farah Babakhani
Yu-Hung Chiu
Alex Romero
Sherwood Gorbach
Thomas John Louie
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Merck Sharp and Dohme LLC
Original Assignee
Optimer Pharmaceuticals LLC
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by Optimer Pharmaceuticals LLC filed Critical Optimer Pharmaceuticals LLC
Publication of EP1940417A2 publication Critical patent/EP1940417A2/fr
Publication of EP1940417A4 publication Critical patent/EP1940417A4/fr
Withdrawn legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/70Carbohydrates; Sugars; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/04Drugs for disorders of the alimentary tract or the digestive system for ulcers, gastritis or reflux esophagitis, e.g. antacids, inhibitors of acid secretion, mucosal protectants
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P1/00Drugs for disorders of the alimentary tract or the digestive system
    • A61P1/12Antidiarrhoeals
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P31/00Antiinfectives, i.e. antibiotics, antiseptics, chemotherapeutics
    • A61P31/04Antibacterial agents

Definitions

  • This invention relates to the treatment of a disease caused by the presence of a bacterium selected from the group consisting of Clostridium species, Staphylococcus species, and Enterococcus species and combinations thereof, in particular a disease caused by the presence of a bacterium selected from the group consisting of Clostridium difficile ("C. difficile”) , Clostridium perfringens ("C. perfringens”) , Staphylococcus aureus (“S. aureus”) and combinations thereof, more particular a disease caused by the presence of C. difficile.
  • the disease may be colitis, pseudomembranous colitis, or diarrhea.
  • Antibiotic-associated diarrhea is caused by toxin producing strains of C. difficile, S. aureus including methicillin-resistant Staphylococcus aureus
  • MRSA Clostridium perfringens
  • C. perfringens Clostridium perfringens
  • AAD represents a major economic burden to the healthcare system that is conservatively estimated at $3-6 billion per year in excess hospital costs in the U.S. alone.
  • AAD is a significant problem in hospitals and long- term care facilities and in the community.
  • C. difficile is the most common cause of AAD in the hospital setting, accounting for approximately 20% of cases of AAD and the majority of cases of antibiotic-associated colitis (AAC) .
  • AAC antibiotic-associated colitis
  • CDAD C. difficile associated diarrhea
  • CDAD C. difficile associated diarrhea
  • PMC pseudomembranous colitis
  • CDAD pseudomembranous colitis
  • the overall mortality rate from CDAD is low, but is much greater in patients who develop severe colitis or systemic toxicity.
  • a recent study has shown that even when death is not directly attributable to C. difficile, the rate of mortality in CDAD patients as compared to case-matched controls is much greater.
  • Diarrhea and colitis are caused by the elaboration of one or more C. difficile toxins . The organism proliferates in the colon in patients who have been given broad-spectrum antibiotics or, less commonly, cancer chemotherapy.
  • CDAD is diagnosed in approximately 20% of hospitalized patients who develop diarrhea after treatment with such agents .
  • Current therapy for AAD or CDAD includes discontinuation of implicated antimicrobial or chemotherapy agents, nonspecific supportive measures, and treatment with antibiotics directed against C. difficile .
  • the most common antimicrobial treatment options include vancomycin, and Metronidazole.
  • Treatment of CDAD with antibiotics is associated with clinical relapse of the disease. Frequency of relapse is reported to be 5-50%, with a 20-30% recurrence rate being the most commonly quoted figure. Relapse occurs with nearly equal frequency regardless of the drug, dose, or duration of primary treatment with any of the antibiotics listed above.
  • the major challenge in therapy is in the management of patients with multiple relapses, where antibiotic control is problematic.
  • Vancomycin is the only drug approved by the FDA for this indication.
  • Vancomycin is not recommended for first-line treatment of CDAD mainly because it is the only antibiotic active against some serious life-threatening multi-drug resistant bacteria. Therefore, in an effort to minimize the emergence of vancomycin-resistant Enterococcus (VRE) or vancomycin-resistant Staphylococcus aureus (VRSA) , the medical community discourages the use of this drug except when absolutely necessary.
  • VRE vancomycin-resistant Enterococcus
  • VRSA vancomycin-resistant Staphylococcus aureus
  • Metronidazole is recommended as initial therapy out of concern for the promotion and selection of vancomycin resistant gut flora, especially enterococci. Despite reports that the frequency of C.
  • metronidazole remains nearly as effective as vancomycin, is considerably less expensive, and can be used either orally or intravenously. Metronidazole is associated with significant adverse effects including nausea, neuropathy, leukopenia, seizures, and a toxic reaction to alcohol. Furthermore, it is not safe for use in children or pregnant women.
  • the present invention provides a method of treating a disease or disorder caused by the presence of a bacterium selected from the group consisting of Clostridium species, Staphylococcus species, Enterococcus species and combinations thereof comprising administering to a patient in need an effective amount of a mixture.
  • the mixture comprises an effective amount of tiacumicin B and an additional macrocycle selected from the group consisting of:
  • the mixture comprises about 0.1 to about 5% compound of formula XIV.
  • the mixture comprises at least 90% tiacumicin B by weight. More preferably, the mixture comprises at least 95% tiacumicin B by weight.
  • the mixture comprises at least 1%, more preferably, from about 2% to about 5%, of additional macrocycle by weight.
  • the mixture comprises about 0.1% to about 5%, more preferably 0.3% to 3%, in particular 0.3% to 1.5%, especially about 1%, lipiarmycin A4 by weight.
  • the mixture also comprises at least one of the following compounds :
  • the mixture exhibits an HPLC profile substantially depicted at Figure 5
  • the disease or disorder treated in accordance with the present invention is associated with C. difficile, C. perfringens, S. aureus, and combinations thereof. More preferably, the disease or disorder treated in accordance with the present invention is associated with C. difficile .
  • the disease treated in accordance with the present invention is diarrhea or colitis, in particular diarrhea, more particularly CDAD.
  • the mixture in accordance with the present invention is prepared by a process comprising: culturing a microorganism in a nutrient medium to accumulate the mixture in the nutrient medium; and isolating the mixture from the nutrient medium; the nutrient medium comprises an adsorbent to adsorb the mixture.
  • the nutrient medium preferably comprises 0.5-15% of the adsorbent by weight.
  • the absorbent is preferably an adsorbent resin. More preferably, the adsorbent resin is selected from the group consisting of Amberlite® XAD16, XAD16HP, XAD2, XAD7HP, XAD1180, XAD1600, IRC50, and Duolite® XAD761.
  • the microorganism is preferably Dactylosporangium aurantiacum subspecies hamdenensis .
  • the nutrient medium comprises, based on weight, 0.2% to 10% of glucose, 0.02% to 0.5% of K 2 HPO 4 , 0.02% to 0.5% of MgS0 4 -7H 2 0, 0.01 % to 0.3% of KCl, 0.1% to 2% of CaCO 3 , 0.05% to 2% of casamino acid, 0.05% to 2% of yeast extract, and 0.5% to 15% of XAD-16 resin.
  • the culturing step is preferably conducted at a temperature from about 25 to about 35 0 C and at a pH from about 6.0 to about 8.0.
  • the disease treated in accordance with the present invention is associated with the use of antibiotics or cancer chemotherapies or antiviral therapy.
  • the mixture is administered in an amount of about 50 mg to 1000 mg, more preferably 100 mg to 400 mg, in particular 200 mg, one to three times daily, more preferably once or twice daily, in particular twice daily, within three to fifteen days, in particular around ten days. Oral administration is preferred.
  • the treatment of the present invention may allow for the effective treatment of diarrhea diseases associated with enterotoxigenic strains of C. difficile, S. aureus, and C. perffingens without compromising systemic antibiotics and without increasing vancomycin resistant enterococci (VRE) in the gut.
  • the present invention also reduces the presence of VRE in the gut.
  • Figure 1 shows the Phase IB-MD Dosing schedule.
  • Figure 2 shows the bacteroides count following treatment. Pairs signed-ranks test, 2 tailed. For counts
  • Figure 3 shows the effect of Vancomycin therapy vs
  • FIG. 4 shows the quantitative reduction of C. difficile vegetative counts after treatment with MCC.
  • Figure 5 is a typical HPLC profile of the mixture, which may be used in the method of the present invention. 7
  • AAD antibiotic-associated diarrhea
  • ATCC American Type Culture Collection 13
  • C carbon 13 CO2 - carbon dioxide
  • N 2 nitrogen
  • H 2 hydrogen
  • TAPS N-Tris (hydroxymethyl)methyl-3- aminopropanesulfonic acid
  • MOPS 3- (N-Morpholino) propanesulfonic acid
  • CDAD Clostridium difficile-associated diarrhea
  • MIC 50 minimum inhibitory concentration to inhibit 50% of bacterial strains tested
  • MIC 90 minimum inhibitory concentration to inhibit 90% of bacterial strains tested
  • MRSA methicillin-resistant Staphylococcus aureus
  • antibiotic-associated condition refers to a condition resulting when antibiotic therapy- disturbs the balance of the microbial flora of the gut, allowing pathogenic organisms such as enterotoxin producing strains of C. difficile, S. aureus and C. perfringens to flourish. These organisms can cause diarrhea, pseudomembranous colitis, and colitis and are manifested by diarrhea, urgency, abdominal cramps, tenesmus, and fever among other symptoms. Diarrhea, when severe, causes dehydration and the medical complications associated with dehydration.
  • MCC refers to a preparation primarily- containing tiacumicin B with respect to the whole antibiotic substance (e.g., at least 90%, preferably 95%- 98% by HPLC assay) . MCC also comprise a small amount (e.g., at least 1%, preferably 2%-5%) of tiacumicin B related compounds, i.e., lipiarmycin A4 and at least one of compound of formula III-XIV shown above.
  • PCT application PCT/US03/21977 having an international publication number of WO 2004/014295 A2 , provides a process of making a mixture comprising tiacumicin B. The entire content of this PCT application is incorporated herein as reference.
  • excipient refers to an inert substance added to a pharmacological composition to further facilitate administration of a compound.
  • excipients include but are not limited to, calcium carbonate, calcium phosphate, various sugars and types of starch, cellulose derivatives, gelatin, vegetable oils and polyethylene glycols .
  • halogen includes F, Cl, Br and I.
  • macrocycles refers to organic molecules with large ring structures usually containing over 10 atoms .
  • 18-membered macrocycles refers to organic molecules with ring structures containing 18 atoms .
  • membered ring can embrace any cyclic structure, including carbocycles and heterocycles as described above.
  • membered is meant to denote the number of skeletal atoms that constitute the ring.
  • pyridine, pyran and thiopyran are 6 membered rings and pyrrole, furan, and thiophene are 5 membered rings.
  • MIC or “minimum inhibitory concentration” refers to the lowest concentration of an antibiotic that is needed to inhibit growth of a bacterial isolate in vitro.
  • a common method for determining the MIC of an antibiotic is to prepare several tubes containing serial dilutions of the antibiotic, that are then inoculated with the bacterial isolate of interest. Following incubation at appropriate atmosphere and temperature, the MIC of an antibiotic can be determined from the tube with the lowest concentration that shows no turbidity (no growth) .
  • MIC 50 refers to the lowest concentration of antibiotic required to inhibit the growth of 50% of the bacterial strains tested within a given bacterial species .
  • ⁇ MIC 9 o refers to the lowest concentration of antibiotic required to inhibit the growth of 90% of the bacterial strains tested within a given bacterial species .
  • patient refers to a human or animal in need of medical treatment. For the purposes of this invention, human patients are typically institutionalized in a primary medical care facility such as a hospital or nursing home.
  • treatment of a disease associated with the use of antibiotics or cancer chemotherapies or antiviral therapies can occur on an outpatient basis, upon discharge from a primary care facility, or can be prescribed by a physician for home-care, not in association with a primary medical care facility. Animals in need of medical treatment are typically in the care of a veterinarian.
  • pharmaceutically acceptable carrier refers to a carrier or diluent that is pharmaceutically acceptable .
  • salts derived from appropriate bases include alkali metal (e.g., sodium or potasium) , alkaline earth metal (e.g., magnesium), ammonium and N(C 1 -C 4 alkyl) 4 + salts, and the like.
  • alkali metal e.g., sodium or potasium
  • alkaline earth metal e.g., magnesium
  • ammonium and N(C 1 -C 4 alkyl) 4 + salts and the like.
  • Illustrative examples of some of these include sodium hydroxide, potassium hydroxide, choline hydroxide, sodium carbonate, and the like.
  • composition refers to a mixture of one or more of the Tiacumicins described herein, or physiologically acceptable salts thereof, with other chemical components, such as physiologically acceptable carriers and/or excipients.
  • physiologically acceptable carrier refers to a carrier or diluent that does not cause significant irritation to an organism and does not abrogate the biological activity and properties of the administered compound.
  • pseudomembranous colitis or "enteritis” refers to the formation of pseudomembranous material (i.e., material composed of fibrin, mucous, necrotic epithelial cells and leukocytes) due to inflammation of the mucous membrane of both the small and large intestine .
  • Teiacumicin refers to a family of compounds all of which comprise the 18- membered macrocycle shown below in Formula I :
  • Tiacumicin B refers to the 18-membered macrocycle shown below in Formula II:
  • lipiarmycin A4 refers to the 18-membered macrocycle shown below in Formula XIV:
  • recurrence of C. difficile -associated diarrhea can be inhibited in a patient by administering MCC in an amount and for a duration effective to inhibit recurrence of C. difficile but with a lack of effect on normal gut flora in the patient.
  • the daily oral dosage of MCC for CDAD will range from between about 50 mg to about 1.0 grams of active agent per day, preferably, from between about 100 mg to about 600 milligrams per day. Generally, treatment will be continued for a time period ranging from between about 3 to about 15 days. Greater or lesser amounts of drug and treatment intervals may be utilized as required. For example, according to the results of a clinical study hereinafter reported, a dosage of about 100-400 milligrams of MCC per day, over the course of from about ten days, proved effective in treating CDAD with minimal clinical recurrence.
  • the mixture can be made by the following general process.
  • MCC-producing bacteria was grown in vessels ranging from shake flasks to large "batch" fermenters .
  • submerged aerobic fermentation in tanks is utilized.
  • small amounts may be obtained by shake-flask culture.
  • tank fermentation it is preferable to use a vegetative inoculum.
  • the vegetative inoculum is prepared by inoculating a small volume of culture medium with the spore form, mycelial fragments, or a lyophilized pellet of the organism to obtain a fresh, actively growing culture of the organism.
  • the vegetative inoculum is then transferred to a larger tank where, after a suitable incubation time, the MCC antibiotic is produced in much improved yield.
  • the production proceeds in a control medium with other additives/ingredients to improve the production.
  • a liquid-submerged, stirred-culture process is used for the production of MCC. Fermentation is carried out at a temperature range of 25 0 C to 37 0 C. The consumption of the carbon source is carefully monitored and an additional amount of carbon source is added as needed.
  • the pH of the fermentation is preferably maintained between about 6.0 to about 8.0. MCC is produced and accumulated between 3 to 15 days after inoculation of the fermentation.
  • MCC adsorbent resin
  • exceptional yield > 100 mg/L broth
  • adsorbent resins such as Amberlite resin (XAD-16)
  • XAD-16 Amberlite resin
  • the solid mass is separated from the broth by sieving. The solid mass are eluted with ethyl acetate then concentrated under reduced pressure .
  • the solid mass (including the adsorbent resin) is separated from the broth by sieving.
  • MCC is eluted from the resin with organic solvents such as ethyl acetate, methanol, acetonitrile or a mixture of two or more organic solvents.
  • organic solvents such as ethyl acetate, methanol, acetonitrile or a mixture of two or more organic solvents.
  • the extract is then concentrated under reduced pressure.
  • This residue is further purified by trituration with low polarity solvents such as hexanes, heptanes, methylcyclohexane, or by partitioning between two phase solvent systems such as: ethyl acetate/water; ethyl acetate/aqueous sodium chloride solution; methanol/hexane, acetonitrile/hexane or other mixtures of two or more solvents in various ratios and combinations or by column chromatography eluting with an appropriate organic solvent system.
  • low polarity solvents such as hexanes, heptanes, methylcyclohexane
  • the current purification process of MCC is based on medium-pressure reverse-phase (C-18) column using 50:50:1 CH 3 CN/H 2 O/AcOH or 70:30:1, MeOH/H 2 O/AcOH as eluent.
  • the fractions contain desired MCC were washed with brine and the concentrated.
  • the residue was dissolved in ethyl acetate and washed with water and organic layer was evaporated to dryness to provide a pale yellow foam which was again washed with isopropyl alcohol and dried under reduced pressure to yield a white powder.
  • the mixture comprising tiacumicin B as major components ranged from 90% to 99%, lipiarmycin A4 (0.1 % to 5%), and at least one or more of the macrocycles of formula III-XIV described above.
  • EXAMPLES [0062] The following examples are provided by way of describing specific embodiments of the present invention without intending to limit the scope of the invention in any way. [0063] The mixture used in the following examples is prepared in accordance with the process of making described above. The following table shows composition of several exemplary mixtures made in accordance with the present invention.
  • HPLC assay is conducted in accordance with the following procedure.
  • Mobile Phase A Add 2.0 mL of trifluoroacetic acid to 2L of HPLC water, filter and degas.
  • Mobile Phase B Add 1.0 mL of trifluoroacetic acid to 2L of Acetonitrile, filter and degas.
  • Retention time of the mixture must be within 8-12 minutes .
  • Standard Preparation Accurately weigh about 20 mg of the mixture into a 100 mL volumetric flask, dissolve in and dilute to volume with Diluent.
  • Sample Preparation Accurately weight about 20 mg of the mixture in a 100 mL volumetric flask. Add about 60 mL Diluent and vortex to dissolve. Dilute to volume with Diluent and mix.
  • R u tiacumicin B peak area obtained from the assay preparation.
  • R s tiacumicin B peak area obtained from the Standard preparation.
  • P Purity of Reference standard, including water factor.
  • W std Standard weight (mg)
  • StdDil Standard dilution (mL)
  • W smp Sample weight (mg)
  • WF Sample water factor.
  • R u tiacumicin B peak area obtained from the Sample Preparation.
  • Example 1 Effect of Inoculum, pH, and Cations on the In Vitro Activity of MCC Vs .
  • Clostridium, difficile [0068] The MIC values measured for many antibiotics are known to be affected by environmental variables such as pH, the concentration of divalent cations such as calcium and magnesium, and the bacterial density. The dependence of the antibacterial activity on these factors is an important consideration, particularly for an antibiotic that targets bacteria in the gut, where these parameters can vary greatly with the diet and disease state, m
  • the sensitivity of the MIC to these environmental variables may also be an important factor to consider when designing methodology for future in vitro testing.
  • CLSI Clinical and Laboratory Standards Institute
  • CLSI (formerly NCCLS) recommends using Brucella agar supplemented with vitamin Ki and hemin for Minimal Inhibitory Concentration (MIC) determination for anaerobes .
  • the level of divalent cations in this medium is not standardized.
  • the pH of the media used under anaerobic glove box may also vary under different gas mixtures. Anaerobes are typically incubated in a mixture of nitrogen, hydrogen, and carbon dioxide, and the presence of CO 2 will acidify the medium and can be a significant source of variability.
  • the inoculum size may also be difficult to standardize given the variety of atmospheric conditions available for anaerobic susceptibility testing (H 2 /CO 2 generator, evacuation/replacement method, or anaerobic chamber) .
  • the anaerobic conditions available to each lab will determine the duration of organism exposure to aerobic atmosphere during bench top manipulations and anaerobic equilibration, and thus affect culture viability and experimental result.
  • the inocula were prepared by first making a suspension of ⁇ 10 8 cfu/mL and then serially diluting the suspension by 10-fold factors to obtain a culture density range between 10 5 - 10 8 cfu/mL, to give spot densities of 10 2 - 10 5 cfu/spot. pH effect on MIC values :
  • anaerobic equilibration of media in the glove box environment (10% H 2 / 5% CO 2 / 85% N 2 ) lowered the pH of the media, resulting in an anaerobic pH range from 5 - 7.5 (as tested using a portable pH meter with a flat-bottomed pH probe calibrated with buffer standards outside the glove box, then transferred inside) .
  • buffer was added to media to resist pH shifts caused by anaerobic equilibration.
  • Divalent cation concentration effect on MIC values [0078] The agar dilution method was used to determine the effect of calcium and magnesium ion concentrations on susceptibility of C. difficile strains to MCC. The level of divalent cations in the Brucella broth as acquired from the manufacturer were determined by the Laboratory Specialists, Inc. Additional amounts of divalent cations were added (in the form of calcium or magnesium chloride) in order to give calcium ion concentrations of 2.1, 3.0 and 5.7 mg/dL and magnesium ion concentrations of 3.3, 4.5, and 7.5 mg/dL.
  • Tables 1 and 2 demonstrate the effect of inoculum density on the MIC of MCC and vancomycin against two strains of C. difficile (ATCC 9689 and ATCC 700057) . Susceptibility of both C. difficile strains to MCC was unaffected by inoculum concentration from 10 5 - 10 8 cfu/ml (10 2 - 10 s CFU/spot) , as shown by identical MIC values obtained for all inoculum concentrations tested. The MIC of vancomycin, however, increased progressively with increasing inoculum concentration, with the highest inoculum density showing a fourfold increase in MIC over the lowest inoculum density. These results demonstrate that inoculum density is not a significant factor affecting the outcome of MCC susceptibility testing of C. difficile.
  • Table 3 depicts the effect of various pH values on susceptibility of C. difficile to MCC as measured by agar dilution method on two separate days .
  • the highest pH treatment pH 7.9
  • the lowest pH treatment pH 7.9
  • the MIC value remained high for both strains.
  • No increase in MCC MIC was observed between pH 6.2 and pH 7 for either strain.
  • the increase in MIC values with pH did not consistently correlate with increased growth, thus the effect of pH on MIC did not appear to be merely due to the enhanced viability of the organism at higher pH.
  • the pH 7 treatment had less dense organism spot growth relative to the pH 6.2 and pH 7.9 treatments.
  • Table 4 represents MIC data from the broth microdilution susceptibility method performed on three separate days with pH ranges from 5 to 8.1.
  • the MIC of MCC at pH 7.5 was 8x greater than the MIC at pH 5.9 for both C. difficile strains (Table 4) .
  • the MIC at pH 5 could not be determined, because the organism failed to grow at this pH.
  • the buffered (10 inM) pH 7.6 treatment showed 8-fold and 16-fold increases in MCC MIC over the pH 6 treatment for C. difficile ATCC 9689 & ATCC 700057, respectively (Table 5) .
  • MCC MIC values with different commercial lots of media [0087] Three different lots of supplemented Brucella agar media were used on three separate days to compare the activity of MCC against C. difficile strains.
  • the MIC assays were controlled by testing the activity of the QC organism, Eubacterium lentum vs. clindamycin which was within the CLSI (NCCLS) acceptable ranges, i.e. 0.06 - 0.25 ⁇ g/mL.
  • Another control step for the MIC assays was to include metronidazole and monitor its activity vs. C . difficile strains, which in our laboratory has been shown to have MIC values ranging between 0.25 - 0.5 ⁇ g/mL.
  • the activity of MCC vs. C. difficile was not affected by different lots of supplemeted Brucella agar. All controls demonstrated activities within established ranges .
  • 10 C. difficile was unaffected by inoculum concentrations, in the range of 10 2 - 10 5 cfu/spot.
  • the MIC values for both MCC and vancomycin increased with increasing pH over a pH range of 6 - 8.
  • the high MIC values at basic pH may be due to
  • Organism density generally increased with increasing pH/ the dependence of growth density, but not MIC, on pH was reduced in the presence of buffering agents. Though organism density was positively correlated with basicity in the absence of buffer, it is unlikely that MIC trends are the result of the effect of pH on organism density alone. This is because the same relationship between MIC and pH was observed in buffered experiments where organism density was more equivalent across pH treatments, presumably due to the differential effect of buffer type on organism growth.
  • Subjects were admitted to the research unit on Day 0 and again on Day 9 of the 10-day dosing period, and stayed for up to 48 hours after each admission. Subjects were discharged on Day 2 and Day 11 after completing the scheduled events and procedures. During the outpatient period, subjects reported daily to the research unit for dosing and stayed for 3 hours under observation.
  • Time to resolution of diarrhea was defined as time (in days) from the first dose of study medication to the resolution of diarrhea; time to resolution of diarrhea was compared among the three treatment groups .
  • the cessation day of diarrhea was defined as the first day that ⁇ 3 unformed stools (watery or loose) within a 24 hour period occurred and was sustained for the duration of treatment up to study Day 10. Resolution of diarrhea was assessed during a 10 to 12 day period utilizing the subject diary data.
  • Table 11 Summary demographics for the Phase 2A study; demographics for the 48 subjects in the population evaluable for safety are shown.
  • Black 1 (6. 3%) 1 (6.3%) 0 (0.0%) 2 (4. 2%) Asian 0 (0. 0%) 0 (0.0%) 1 (6.3%) 1 (2. 1%) Hispanic 0 (0. 0%) 0 (0.0%) 0 (0.0%) 0 (0. 0%) Other a 1 (6. 3%) 0 (0.0%) 1 (6.3%) 2 (4. 2%)
  • the time to resolution of diarrhea was defined as the time for the patient to resolve to less than 3 unformed stools per day, according to the patient's diary card.
  • the median time to relief was 5.5 days, 3.5 days, and 3.0 days for the MCC 100 mg/day, 200 mg/day and 400 mg/day treatment groups, respectively.
  • the mean time to resolution of diarrhea in days was 6.3+3.66 days in 100 mg/day-treated subjects, 4.8 ⁇ 3.56 days in 200 mg/day-treated subjects, and 3.6+2.03 in 400 mg/day-treated subjects.
  • Cardiac failure congestive 1 (6.3) 0 (0.0 0 (0.0)
  • Chest pain 1 (6.3) 1 (6.3 0 (0.0)
  • Urinary tract infection 1 (6.3) 0 (0.0 0 (0.0)
  • Musculoskeletal and connective 1 ( 6.3) 0 (0.0 0 (0.0) tissue disorders )
  • Nervous system disorders 0 (0.0) 1 (6.3 0 (0.0)
  • Renal and urinary disorders 1 (6.3) 0 (0.0 0 (0.0)
  • Nephrolithiasis 1 (6.3) 0 (0.0 0 (0.0)
  • Vascular disorders 2 (12.5 0 (0.0 0 (0.0)
  • Observable MCC concentrations ranged from 9.45 to 12.3 ng/mL in the MCC 100 mg/day treatment group, 5.12 to 93.7 ng/mL in the MCC 200 mg/day treatment group, and 5.32 to 84.9 ng/mL in the MCC 400 mg/day treatment group.
  • Table 17 shows fecal concentrations from the IB-MD study, normalized to the 150 mg dose; fecal MCC averaged 916.0 ⁇ g/g (138.4-1768.9 ⁇ g/g) .
  • Bacteroides group organisms are considered to be uniformly present in subjects and in high counts, and is likely one of the major components of the normal flora conferring Colonization resistance' , this group was used as an index of suppression of the anaerobic fecal flora.
  • subsequent samples were processed to document time of return of this group. If samples were not immediately processed, aliquots were frozen at -80 degrees C with 15% glycerol / Brain Heart Infusion Broth for subsequent processing.
  • C. difficile counts were determined by dilution of the sample 10 ⁇ 2,4,6,8 / g ra m stool wet weight on CCFA agar. Spore counts were determined by treating an aliquot of stool with an equal volume of 100% ethyl alcohol x 1 hour, centrifuged, washed twice and resuspended for quantitative counts.
  • vancomycin fecal filtrate concentrations were determined in triplicate by bioassay using a C. perfringens as the indicator organism.

Landscapes

  • Health & Medical Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Veterinary Medicine (AREA)
  • Public Health (AREA)
  • Chemical & Material Sciences (AREA)
  • General Health & Medical Sciences (AREA)
  • Medicinal Chemistry (AREA)
  • Animal Behavior & Ethology (AREA)
  • General Chemical & Material Sciences (AREA)
  • Organic Chemistry (AREA)
  • Nuclear Medicine, Radiotherapy & Molecular Imaging (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Engineering & Computer Science (AREA)
  • Bioinformatics & Cheminformatics (AREA)
  • Molecular Biology (AREA)
  • Epidemiology (AREA)
  • Oncology (AREA)
  • Communicable Diseases (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)

Abstract

La présente invention a pour objet une méthode de traitement d'une maladie ou d'un trouble provoqué(e) par la présence d'une bactérie sélectionnée au sein du groupe constitué par l'espèce Clostridium, l’espèce Staphylococcus, l’espèce Enterococcus et les combinaisons de ces bactéries, la méthode comprenant l'administration à un patient la nécessitant d'une quantité active d'un mélange comprenant de la tiacumicine B, de la lipiarmycine A4 et au moins un autre composé macrocyclique.
EP06826544A 2005-10-21 2006-10-23 Méthode de traitement d'une diarrhée associée à clostridium difficile Withdrawn EP1940417A4 (fr)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US72913505P 2005-10-21 2005-10-21
US74964105P 2005-12-12 2005-12-12
PCT/US2006/041436 WO2007048059A2 (fr) 2005-10-21 2006-10-23 Méthode de traitement d'une diarrhée associée à clostridium difficile

Publications (2)

Publication Number Publication Date
EP1940417A2 true EP1940417A2 (fr) 2008-07-09
EP1940417A4 EP1940417A4 (fr) 2010-07-07

Family

ID=37963388

Family Applications (1)

Application Number Title Priority Date Filing Date
EP06826544A Withdrawn EP1940417A4 (fr) 2005-10-21 2006-10-23 Méthode de traitement d'une diarrhée associée à clostridium difficile

Country Status (8)

Country Link
EP (1) EP1940417A4 (fr)
JP (1) JP2009512691A (fr)
KR (1) KR101399621B1 (fr)
CN (2) CN102503994A (fr)
AU (1) AU2006304868B2 (fr)
CA (1) CA2626698C (fr)
HK (1) HK1126410A1 (fr)
WO (1) WO2007048059A2 (fr)

Families Citing this family (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7906489B2 (en) 2004-05-14 2011-03-15 Optimer Pharmaceuticals, Inc. 18-membered macrocycles and analogs thereof
US7378508B2 (en) 2007-01-22 2008-05-27 Optimer Pharmaceuticals, Inc. Polymorphic crystalline forms of tiacumicin B
TWI523654B (zh) 2007-11-27 2016-03-01 默沙東藥廠 抗生素巨環化合物以及其製造及使用之方法
JO3464B1 (ar) 2013-01-15 2020-07-05 Astellas Pharma Europe Ltd التركيبات الخاصة بمركبات التياكوميسين
CN104846044B (zh) * 2014-02-17 2019-02-01 上海医药工业研究院 一种提高非达霉素产量的发酵培养基
CN105237599B (zh) * 2015-10-09 2018-10-02 华北制药集团新药研究开发有限责任公司 闰年霉素a4晶体及其制备方法
CN107970253B (zh) * 2017-10-25 2020-05-26 中山大学 台勾霉素衍生物在制备治疗寨卡病毒感染引起的相关疾病和/或症状的药物中的应用
CN107714713B (zh) * 2017-10-25 2020-05-26 中山大学 台勾霉素衍生物在制备治疗登革病毒感染引起的相关疾病和/或症状的药物中的应用
JP7397071B2 (ja) * 2018-06-07 2023-12-12 アルチュジェン セラピューティクス リミテッド C.ディフィシル治療方法及び組成物

Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5583115A (en) * 1995-05-09 1996-12-10 Abbott Laboratories Dialkyltiacumicin compounds
WO2005112990A2 (fr) * 2004-05-14 2005-12-01 Optimer Pharmaceuticals, Inc. Traitement de maladies consecutives a un traitement antibiotique
WO2006085838A1 (fr) * 2005-01-31 2006-08-17 Optimer Pharmaceuticals, Inc. Macrocycles a 18 chainons et leurs analogues

Family Cites Families (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US4918174A (en) 1986-09-26 1990-04-17 Abbott Laboratories Tiacumicin compounds
US7507564B2 (en) * 2002-07-29 2009-03-24 Optimer Pharmaceuticals, Inc. Tiacumicin production

Patent Citations (3)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5583115A (en) * 1995-05-09 1996-12-10 Abbott Laboratories Dialkyltiacumicin compounds
WO2005112990A2 (fr) * 2004-05-14 2005-12-01 Optimer Pharmaceuticals, Inc. Traitement de maladies consecutives a un traitement antibiotique
WO2006085838A1 (fr) * 2005-01-31 2006-08-17 Optimer Pharmaceuticals, Inc. Macrocycles a 18 chainons et leurs analogues

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
See also references of WO2007048059A2 *

Also Published As

Publication number Publication date
CN101340919B (zh) 2011-12-07
JP2009512691A (ja) 2009-03-26
AU2006304868B2 (en) 2013-02-21
KR20080064177A (ko) 2008-07-08
EP1940417A4 (fr) 2010-07-07
HK1126410A1 (en) 2009-09-04
AU2006304868A1 (en) 2007-04-26
CA2626698A1 (fr) 2007-04-26
CN102503994A (zh) 2012-06-20
KR101399621B1 (ko) 2014-06-18
CA2626698C (fr) 2015-12-01
CN101340919A (zh) 2009-01-07
WO2007048059A3 (fr) 2007-05-31
WO2007048059A2 (fr) 2007-04-26

Similar Documents

Publication Publication Date Title
CA2626698C (fr) Methode de traitement d'une diarrhee associee a clostridium difficile
US20140107054A1 (en) Method of treating clostridium difficile-associated diarrhea
EP2826473B1 (fr) Utilisation antibactérienne du patchoulol
EP2305244B1 (fr) Traitement de maladies consecutives a un traitement antibiotique
CN102215858A (zh) 使用单剂量奥利万星的治疗方法
Raasch Anidulafungin: review of a new echinocandin antifungal agent
KR0145553B1 (ko) 클로스트리듐 디피실레 하리증 및 위막성 대장염의 예방 및 치료용 의약조성물
Naber et al. Gatifloxacin 400 mg as a single shot or 200 mg once daily for 3 days is as effective as ciprofloxacin 250 mg twice daily for the treatment of patients with uncomplicated urinary tract infections
CN111419829B (zh) 和厚朴酚在抑制猪链球菌或其生物被膜中的用途
WO2012050826A1 (fr) Procédés de traitement d'infections par clostridium difficile
Yanagihara et al. Susceptibility of Clostridium species isolated in Japan to fidaxomicin and its major metabolite OP-1118
CN111249264A (zh) 没食子酸在逆转猪链球菌对抗生素耐药性中的用途
CN113318149B (zh) 一种素馨花提取物及其制备方法和应用
CN109678917A (zh) 艾美克勒霉素及其制备方法和应用
Alonso et al. Plasma and peritoneal ceftriaxone concentrations after intraperitoneal administration in horses with septic peritonitis
CN110141569B (zh) 抗牛多杀性巴氏杆菌的药物组合物
CN114907287B (zh) 一种用于治疗肾损伤的化合物
US11806342B2 (en) Diiodohydroxyquinoline for the treatment of clostridium difficile infection
CN109553565B (zh) 两种新生物碱及其制备方法和在制备抗mrsa的抗菌剂中的应用
CN109568494B (zh) 一种白及醇提物在制备多粘菌素抗菌增效剂中的应用
CN107951878A (zh) 螺缩酮多炔类化合物在制备抗外泵耐药金葡菌增敏药物中的用途
Aleem et al. Drugs in therapeutic application of dogs and cats
CN111249296A (zh) 一种抗真菌的药物组合物及互动抗真菌活性测定方法
CN103690526A (zh) 一种阿扎霉素f衍生物和维生素k3的抗菌药物组合物
JPH1029947A (ja) 生体内毒素型細菌性腸管感染症治療剤

Legal Events

Date Code Title Description
PUAI Public reference made under article 153(3) epc to a published international application that has entered the european phase

Free format text: ORIGINAL CODE: 0009012

17P Request for examination filed

Effective date: 20080425

AK Designated contracting states

Kind code of ref document: A2

Designated state(s): AT BE BG CH CY CZ DE DK EE ES FI FR GB GR HU IE IS IT LI LT LU LV MC NL PL PT RO SE SI SK TR

A4 Supplementary search report drawn up and despatched

Effective date: 20100609

RIC1 Information provided on ipc code assigned before grant

Ipc: A61P 31/04 20060101ALI20100602BHEP

Ipc: A61K 45/06 20060101ALI20100602BHEP

Ipc: A61K 31/70 20060101AFI20080428BHEP

17Q First examination report despatched

Effective date: 20110216

17Q First examination report despatched

Effective date: 20110302

DAX Request for extension of the european patent (deleted)
RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: OPTIMER PHARMACEUTICALS, INC.

RAP1 Party data changed (applicant data changed or rights of an application transferred)

Owner name: MERCK SHARP & DOHME CORP.

STAA Information on the status of an ep patent application or granted ep patent

Free format text: STATUS: THE APPLICATION IS DEEMED TO BE WITHDRAWN

18D Application deemed to be withdrawn

Effective date: 20160202