NZ753713B2 - Novel tebipenem pivoxil immediate and modified release oral dosage forms - Google Patents

Novel tebipenem pivoxil immediate and modified release oral dosage forms Download PDF

Info

Publication number
NZ753713B2
NZ753713B2 NZ753713A NZ75371317A NZ753713B2 NZ 753713 B2 NZ753713 B2 NZ 753713B2 NZ 753713 A NZ753713 A NZ 753713A NZ 75371317 A NZ75371317 A NZ 75371317A NZ 753713 B2 NZ753713 B2 NZ 753713B2
Authority
NZ
New Zealand
Prior art keywords
dosage form
tebipenem pivoxil
core
tebipenem
release
Prior art date
Application number
NZ753713A
Other versions
NZ753713A (en
Inventor
Grazyna Fraczkiewicz
Akash Jain
Timothy Keutzer
Shoufeng Li
Enxian Lu
Shaoqiong Lyu
Joyce Macwan
Luke Utley
Original Assignee
Spero Therapeutics Inc
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 Spero Therapeutics Inc filed Critical Spero Therapeutics Inc
Priority claimed from PCT/US2017/066729 external-priority patent/WO2018112372A1/en
Publication of NZ753713A publication Critical patent/NZ753713A/en
Publication of NZ753713B2 publication Critical patent/NZ753713B2/en

Links

Abstract

Immediate and modified release oral dosage forms of tebipenem pivoxil including dosage form cores are provided. The dosage form core includes tebipenem pivoxil in free base or salt form, and excipients including a binder, a lubricant, optionally a diluent, and optional additional excipients. The weight to weight ratio of tebipenem pivoxil to the excipients in the dosage form core is from 30:60 to 60:30. The disclosure includes methods of treating bacterial infections including complicated urinary tract infections. ght to weight ratio of tebipenem pivoxil to the excipients in the dosage form core is from 30:60 to 60:30. The disclosure includes methods of treating bacterial infections including complicated urinary tract infections.

Description

NOVEL TEBIPENEM PIVOXIL IMMEDIATE AND MODIFIED RELEASE ORAL DOSAGE FORMS CROSS REFERENCE TO D APPLICATION This application claims priority to US. Provisional Application No. 62/434,643, filed December 15, 2016, and US. Provisional Application No. 62/438,071, filed December 22, 2016, both of which are hereby incorporated by reference in their entirety.
FIELD OF THE DISCLOSURE Immediate and modified release tebipenem pivoxil oral dosage form cores and oral dosage forms are provided by this disclosure. Methods of treating a urinary tract infection in a patient with an immediate or modified release oral dosage form of this disclosure are also included.
BACKGROUND Tebipenem pivoxil is an —administered pivaloyloxymethyl prodrug of nem, an antibiotic from the carbapenem subgroup of B—lactam antibiotics: tebipenem pivoxil Tebipenem pivoxil has been developed to treat severe bacterial infections that have acquired antibiotic ance to commonly used anti—infective medicines. Following oral stration, the ester and acetal bonds of the prodrug are cleaved to release active tebipenem.
While the prodrug has high lity in gastrointestinal (GI) ted fluids, it readily decomposes at pH 1 and pH 9 and is ive to oxidative conditions.
Thus, stable tebipenem pivoxil dosage forms having consistent and desirable physical and chemical properties are needed. This sure provides dosage forms that meet this need and have onal advantages which are described in the disclosure.
SUMMARY This disclosure provides immediate and modified release oral dosage forms of tebipenem pivoxil. The disclosure provides immediate and ed release solid oral dosage form comprising a dosage form core, wherein the dosage form core comprises: tebipenem pivoxil or a pharmaceutically able salt thereof and excipients including a binder, a lubricant, an optionally a diluent.
The disclosure provides an immediate release solid oral tebipenem pivoxil dosage form, which provides an in vitro release of greater than 85 % of the tebipenem l in 15 minutes when measured by the USP Paddle method at 50 rpm in 900 mL of 50 mM acetate buffer, pH 5.0 at 37 0C.
The disclosure also provides a modified release solid oral tebipenem pivoxil dosage form, which provides an in vitro release of NMT (not more than) 50% of the tebipenem pivoxil after 30 minutes when measured by the USP Paddle method at 50 rpm in 900 mL of 50 mM acetate buffer, pH 5.0 at 37 0C. In certain embodiments this modified release dosage form releases releases NLT (not less than) 85% of the total tebipenem pivoxil after 4 hours, when measured by the USP Paddle method at 50 rpm in 900 mL of 50 mM acetate buffer, pH 5.0 at 37 This disclosure also provides a short modified release solid oral tebipenem pivoxil dosage form, which es an in vitro release of NMT (not more than) 50% of the tebipenem pivoxil after 30 minutes when measured by the USP Paddle method at 50 rpm in 900 mL of 50 mM e buffer, pH 5.0 at 37 0C. In certain embodiments this modified release dosage form releases releases NLT (not less than) 85% of the total tebipenem pivoxil after 2 hours, when measured by the USP Paddle method at 50 rpm in 900 mL of 50 mM acetate , pH 5.0 at 37 The disclosure also provides a method of treating a ial infection in a patient comprising administering an oral dosage form of the disclosure to the patient. In certain embodiments the bacterial infection is a urinary tract infection, including a complicated urinary tract infection. [0011a] In a first aspect of the disclosure, there is provided an immediate or modified release solid oral dosage form comprising a dosage form core, wherein the dosage form core comprises: nem pivoxil HBr and excipients, wherein the weight to weight ratio of tebipenem pivoxil as a free base to the excipients in the core is from 1:2 to 2:1, and the excipients include 5-40 % (w/w%) of a binder, 0.1-5% of a lubricant, and optionally 5-40% of a diluent. [0011b] In a second aspect of the disclosure, there is provided a use of an oral dosage form according the first aspect of the disclosure in the manufacture of a medicament for treating urinary tract infection. [0011c] In a third aspect of the disclosure, there is ed a use of an oral dosage form according to the first aspect of the sure in the manufacture of a medicament for ng a bacterial infection. [0011d] In a fourth aspect of the disclosure, there is provided a use of an oral dosage form according to the first aspect of the disclosure in the manufacture of a medicament for treating a Mycobacterial tuberculosis infection.
BRIEF DESCRIPTION OF THE DRAWINGS The above and other aspects and features of the t disclosure will become more apparent by describing exemplified embodiments thereof in detail with reference to the attached drawings, in which: shows a dissolution profile of a 500 mg immediate release tebipenem pivoxil uncoated tablet core at pH 5. The formulation of the tablet core is ed in Example 1. shows a dissolution profile of a 500 mg nem pivoxil ed release uncoated tablet core at pH 5. The formulation of the modified release dosage form is provided in Example 3. shows the ution profiles of a 500 mg enteric coated immediate release tablet at pH 6.8. The composition of the tablet core is provided in Example 1 and the composition of the enteric g is provided at Example 2, Table 3. is a graph of concentration (milligrams per liter, ug/mL) and mass grams, mg) versus simulation time (hours, h). The graph is a simulated pharmacokinetic profile of the human -administered (PO) dose of a 500 mg tebipenem pivoxil tablet. is a comparison of the dissolution profiles of the modified release tebipenem pivoxil HBr salt (Table 12A) and modified release tebipenem l free base (Table 7) at pH 5.0. is a ison of the dissolution profiles of the modified release tebipenem pivoxil HBr salt (Table 12A) and modified release tebipenem pivoxil free base (Table 7) at pH 6.8. shows a dissolution profile of a 300 mg tebipenem pivoxil immediate release uncoated tablet core at pH 5. The formulation of the modified release dosage form is provided in Table l1B. shows a ution profile of a 300 mg tebipenem pivoxil modified release uncoated tablet core at pH 5. The formulation of the modified release dosage form is provided in Table 12B.
[Followed by page 4] shows a dissolution profile of a 300 mg nem pivoxil short modified release uncoated tablet core at pH 5. The formulation of the modified release dosage form is provided in Table 13.
DETAILED DESCRIPTION Recitation of ranges of values are merely intended to serve as a shorthand method of referring individually to each separate value falling within the range, unless otherwise indicated herein, and each separate value is incorporated into the specification as if it were individually recited herein. The endpoints of all ranges are included within the range and independently combinable. All methods described herein can be performed in a suitable order unless otherwise ted herein or ise clearly contradicted by context. The use of any and all examples, or language indicating an example (e.g., "such as"), is intended merely for illustration and does not pose a limitation on the scope of the invention unless otherwise claimed.
No language in the specification should be construed as indicating any non—claimed element as ial to the practice of the invention.
The terms "a" and "an" do not denote a tion of quantity, but rather denote the presence of at least one of the referenced item.
The term " is used synonymously with the term "approximately." As one of ordinary skill in the art would understand, the exact boundary of "about" will depend on the component of the composition. Illustratively, the use of the term "about" indicates that values slightly outside the cited values, i.e., plus or minus 0.1% to 10%, which are also effective and safe are ed in the value. Thus compositions slightly outside the cited ranges are also encompassed by the scope of the t claims.
The terms "comprising," ding," and "containing" are non—limiting. Other non—recited elements may be present in embodiments claimed by these transitional phrases.
Where "comprising,:9 6‘containing," or "including" are used as transitional s other elements may be included and still form an embodiment within the scope of the claim. The open—ended transitional phrase "comprising" encompasses the intermediate transitional phrase sting essentially of" and the close—ended phrase "consisting of." The term "immediate release" refers to a solid dosage form that release the active ingredient, tebipenem pivoxil, immediately after oral administration.
The term "modified release" refers to a solid oral dosage form that permits the release of the active ient, tebipenem pivoxil, over an extended period of time relative to the time for the immediate release formulation to release the active agent, to maintain therapeutically effective plasma levels. The modified release formulation may be a controlled release ation, one that exhibits substantially zero order release cs. It may also be a sustained e formulation, which exhibits first order cs.
"Pharmaceutically acceptable salts" includes derivatives of tebipenem pivoxil, wherein the nem pivoxil is modified by making acid or base addition salts thereof, and further refers to pharmaceutically acceptable solvates, including hydrates, and co—crystals of such compounds and such salts. Examples of pharmaceutically acceptable salts include, but are not limited to, mineral or organic acid addition salts of basic residues such as amines; alkali or organic addition salts of acidic residues; and the like, and combinations comprising one or more of the ing salts. The pharmaceutically acceptable salts include non—toxic salts and the quaternary ammonium salts of the active agent, tebipenem pivoxil. For example, non—toxic acid salts include those derived from inorganic acids such as hydrochloric, hydrobromic, sulfuric, sulfamic, phosphoric, nitric and the like; other acceptable inorganic salts include metal salts such as sodium salt, potassium salt, cesium salt, and the like; and alkaline earth metal salts, such as calcium salt, magnesium salt, and the like, and combinations comprising one or more of the foregoing salts. Pharmaceutically acceptable organic salts includes salts prepared from organic acids such as acetic, propionic, succinic, glycolic, c, lactic, malic, tartaric, citric, ascorbic, pamoic, , hydroxymaleic, acetic, glutamic, benzoic, salicylic, mesylic, esylic, besylic, sulfanilic, 2—acetoxybenzoic, fumaric, toluenesulfonic, methanesulfonic, ethane disulfonic, oxalic, isethionic, HOOC—(CH2)n—COOH Where n is 0—4, and the like; organic amine salts such as triethylamine salt, pyridine salt, ne salt, ethanolamine salt, anolamine salt, dicyclohexylamine salt, N,N'—dibenzylethylenediamine salt, and the like; and amino acid salts such as arginate, asparginate, glutamate, and the like; and ations sing one or more of the foregoing salts; organic amine salts such as triethylamine salt, ne salt, picoline salt, ethanolamine salt, triethanolamine salt, dicyclohexylamine salt, N,N' dibenzylethylenediamine salt, and the like; and amino acid salts such as arginate, asparginate, glutamate, and the like; and combinations comprising one or more of the foregoing salts. All forms of nem pivoxil are included in the disclosure, including all crystalline, amorphous, and rph forms.
When a weight of tebipenem pivoxil that may be in a salt form is given the value refers to the amount of tebipenem pivoxil rather than the weight of tebipenem pivoxil salt. For example, 500 mg of tebipenem pivoxil, where the tebipenem pivoxil is a hydrobromide salt, indicates that 581.2 g of tebipenem pivoxil hydrobromide are present. Unless it is clear from the context that tebipenem l free base is intended or tebipenem l free base is explicitly specified the term "tebipenem pivoxil" es tebipenem pivoxil free base and the pharmaceutically acceptable salts of tebipenem pivoxil, for e tebipenem pivoxil HBr.
DOSAGE FORM COMPOSITION New tebipenem pivoxil immediate and modified release solid dosage forms have been developed. The dosage forms may be a capsule, a granule, a tablet core, a powder, but are not limited thereto. The dosage forms have a core including tebipenem pivoxil as an active ingredient and one or more excipients. While the nem pivoxil dosage forms may contain a range of tebipenem pivoxil, for example, from 100 to 1,200 milligrams (mg) or 100 to 600 mg of tebipenem pivoxil, tablets containing 100 mg, 300mg, and 500 mg of tebipenem pivoxil are exemplified.
The disclosure provides ate or modified release solid oral dosage forms sing a dosage form core, wherein the dosage form core comprises: tebipenem l or a pharmaceutically acceptable salt thereof and excipients including a binder, a lubricant, an optionally a diluent. The weight to weight ratio of nem pivoxil (as free base) to the excipients in the core is from 30:60 to 60:30, or in some embodiments from 30:45 to 45:30, or about 50:50, or about 30:40.
The disclosure includes tebipenem pivoxil immediate and modified release oral dosage forms, such as a tablet dosage forms.
The tebipenem pivoxil dosage forms of this disclosure have a number of desirable properties. For e certain of the new modified release dosage forms are effective for at least 6 hours, ting less frequent administration than immediate release form and in some embodiment permitting twice a day administration. The modified release tebipenem pivoxil dosage exhibit a lower plasma Cmax and higher plasma Cmm than previous dosage forms resulting in an increased duration of the effective concentration, increased efficacy, increased duration of effect as compared to immediate dosage forms. Once released, the modified release dosage form provides high absorbtion of the tebipenem pivoxil from the digestive tract. The modified release tebipenem pivoxil dosage forms of this disclosure reaches plasma Cum 1 to 4 hours after oral stration, in certain embodiments the modified e tebipenem pivoxil dosage forms of the disclosure reach plasma Cum 2 to 4 hours after oral administration, and in other embodiments the modified release tebipenem pivoxil dosage forms of this disclosure reaches plasma Cum 1 to 2.5 hours after oral administration.
The new immediate release tebipenem pivoxil dosage form is very quickly soluble in water or an aqueous solution and also provides high absorbtion of the nem pivoxil from the digestive tract. Thus, the immediate release dosage form provides rapid onset of drug action and rapid therapeutic benefit. The immediate release tebipenem pivoxil dosage form of this disclosure reaches plasma Cmax within 0.5—2 hours or 0.5 to 1.5 hours after oral administration. When the HBr salt is used the immediate release dosage form of this disclosure reaches plasma Cum in less than 1.5 hours.
The immediate and modified release tebipenem pivoxil dosage form has a shelf stability of at least 12 months and ably at least 24 months at room ature.
The immediate and modified release tebipenem pivoxil dosage forms have dissolution profiles that are independent of dosage th from 100 to 1000 mg of tebipenem In n embodiments the modified release tebipenem pivoxil dosage form provides an in vivo plasma TmX of 3 to 8, 3 to 6, or 3 to 4 hours after administration. In certain embodiments the short modified release tebipenem pivoxil dosage form provides an in vivo plasma Tum of 0.5 to 4, 1 to 3, or 1 to 2.5 hours after administration.
The disclosure es tebipenem pivoxil tablet formulations in which at least %, at least 40%, at least 45%, at least 50%, at least 55%, at least 60%, or at least 65% of the tablet by weight is tebipenem pivoxil. The disclosure includes tebipenem pivoxil dosage forms in which nem pivoxil is 30 % to 70% or 40% to 60% by weight of the tablet core and the excipients include 70% to 30% or 60% to 40% by weight of the tablet core. In n embodiments the tebipenem pivoxil includes 45% to 55% by weight of the tablet core and the excipients include 55% to 45% by weight of the tablet core. In some other embodiments, tebipenem pivoxil es 50% of the tablet core and the excipients include 50% of the tablet core. In some embodiments the nem pivoxil es 40% of the tablet core.
The excipients include, but are not limited to, one or more of a binder, a release control agent, a disintegrant, a diluent, or a lubricant. The excipients may further include a glidant, a solvent, a viscosity agent, an emulsifier, a , a bulking agent, a coloring agent, a taste—improving agent, a flow agent, an absorbent, but are not limited thereto.
The sure includes dosage cores and coated tablets containing a binder. For example, the binder may be microcrystalline cellulose, silicified microcrystalline cellulose, ethyl cellulose, lactose, starch, n, or any combination of any two of the foregoing. The binder may be present in the tablet core or coated tablet in an amount (%w/w) from 1% to 50% or from % to 40%, or from 10% to 20%, or 15%. In certain embodiments, Avicel PH102, a type of microcrystalline cellulose, is the binder.
The disclosure includes modified release oral dosage forms, including dosage cores and coated tablets containing a release control agent. A release control agent is an agent that assists in delivery of the active ingredient over a desired period of time. The release control agent may be present in the tablet core or coated tablet in an amount (%w/w) from 1% to 50% or from 5% to 40%, or from 10% to 20%, or 15%. In certain embodiments hydroxypropyl methylcellulose (HPMC) is included as a e l agent. K grade HPMC products have a methoxyl degree of substitution of 1.4, approximately 22% methoxyl content, and ypropyl content of imately 8.1%. E grade HPMC products have a methoxyl degree of substitution of 1.9, approximately 29% methoxyl content, and approximately 8.5% hydroxypropyl t.
HPMC K4M is used as a release l agent in some modified release tebipenem pivoxil dosage forms of this disclosure. In certain embodiments the HPMC K4M is a hydroxypropyl methyl cellulose with a particle size range of 170 to 250 micrometers and a ity range of 2700 to 5050 cps at 40 0C or about 4000 mPa-s at 2% in water at 20 OC . HPMC E50 LV and HPMC E3 are also used as a release control agents in some embodiments of this disclosure.
HPMC E50 LV has viscosity of 35—65 cps at 40 0C or about 50 mPa-s at 2% in water at 20 OC, and HPMC E3 and has a viscosity of about 3 mPa-s at 2% in water at 20 0C. In certain embodiments such as the 2 hr modified release dosage form the suitable release control agent is polyvinylpyrrolidone such as PVP K12, which has an average molecular weight of 3500— 6000 and is slightly (5%) water soluble. Other suitable release control agents include hydroxypropyl methylcellulose, hydroxypropyl cellulose, hypromellose acetate succinate, polyVinylpyrrolidone, and copovidone.
The disclosure includes dosage cores and coated tablets containing a diluent. For example, the t may be starch, sucrose, monohydrate lactose, mannitol, ol, or any combination of any two of the foregoing. The t may be present in the tablet core or coated tablet in an amount (%w/w) from 1% to 50% or from 5% to 40%, or from 10% to 20%, or 15%.
In some embodiments the diluents are monohydrate lactose and mannitol.
The disclosure includes dosage forms, including dosage form cores and coated tablets containing a disintegrant. For example, the disintegrant may be croscarmellose sodium, crospovidone (also called copovidone), sodium starch glycolate, or any combination of any two or more of the foregoing. The egrant may be present in the tablet core or coated tablet in an amount (%w/w) from 1% to 50% or from 5% to 40%, or from 10% to 20%, or 15%. In some ments PVPP XL—lO, a type of crospovidone, is the disintegrant. PVPP XL—lO is a crosslinked polyVinylpyrrolidone haVing an average particle size of 30 um. Other suitable disintegrants include croscarmellose , pregelatinized starch, sodium starch glycolate, mannitol, maize starch, potato starch, alginic acid, and wheat starch.
The disclosure includes dosage cores and coated tablets containing a lubricant.
The kind and amount of lubricant are not limited so long as within the ranges acceptable for pharmaceutical use. For example, the lubricant may be a fatty acid ester, stearic acid, magnesium stearate, glyceryl behenate, calcium stearate, sodium stearyl fumarate, sodium lauryl sulfate, magnesium lauryl sulfate, sodium benzoate, talc, hydrogenated oil, Carnauba wax, or a combination of any two of the foregoing. The lubricant may be present in the tablet core or coated tablet in an amount (%w/w) from 0.1% to 5%, 0.1% to 2.0%, or from 0.1% to 1.0%, or from 0.25% to 0.5%, or 0.375%. In n embodiments the lubricant is magnesium te.
In an embodiment, the tablet core may be a modified release tablet core, which includes tebipenem pivoxil as an active ingredient, hydroxypropyl methyl cellulose as a release control agent, mannitol as a diluent, microcrystalline cellulose as a binder, and magnesium stearate as a lubricant. For e, the table core may e 40 to 60 weight t of tebipenem l, 10 to 30 weight percent hydroxypropyl methyl cellulose, 10 to 20 weight percent mannitol, 10 to 20 weight percent microcrystalline ose, and 0.1 to 5 weight percent magnesium stearate.
The immediate release nem l tablet core can release at least 80%, at least 85%, at least 90%, or at least 95% of the total amount of tebipenem pivoxil in a pH 5.0 buffer at 37°C with a paddle speed of 50 revolutions per minute (rpm) in 30 minutes, in 15 minutes, or in 10 minutes. In certain embodiments the immediate release tebipenem pivoxil tablet core can release at least 95% of the total amount of tebipenem pivoxil in a pH 5.0 buffer at 37°C with a paddle speed of 50 revolutions per minute (rpm) in 15 minutes. The immediate e tebipenem l tablet core may release tebipenem pivoxil substantially as shown in the dissolution profile of of FIG 7, when the tebipenem pivoxil is in the form of tebipenem pivoxil HBr.
The modified release nem pivoxil tablet core can release n 30% and 50%, or between 35% and 45%, or NMT than 50% of the total amount of tebipenem pivoxil after s in a pH 5.0 buffer at 37°C with a paddle speed of 50 tions per minute (rpm).
The modified release tebipenem pivoxil tablet core can release between 50% and 70%, or between 55% and 65%, of the total amount of tebipenem pivoxil after 60 minutes in a pH 5.0 buffer at 37°C with a paddle speed of 50 revolutions per minute (rpm). The tebipenem pivoxil tablet core may also release NLT 85%, or NLT 90%, of the total amount of tebipenem pivoxil after 4 hours in a pH 5.0 buffer at 37°C with a paddle speed of 50 rpm. The ed release tablet core may release tebipenem l ntially as shown in the dissolution profile of of FIG 8 (for the HBr salt formulation disclosed in Table 12B).
COATINGS The dosage form may further include a coating.
In certain embodiments the coating is an enteric coating. Such coatings are ble at the acidic pH of gastric fluid, for example less than pH 3, but readily soluble at the neutral or nearly neutral pH of the gastrointestinal tract, for example at pH 6.8 to pH 7.4.
The tebipenem pivoxil dosage form core may be coated with an enteric coating, such as an immediate release or modified release enteric coating, to form an enteric coated dosage form. An enteric coating is designed to be resistant to stomach acid, and will not dissolve until the tablet reaches the small intestine. The pH dependent coating can protect an acid— sensitive tebipenem pivoxil from the low pH of gastric fluid. When the enteric coated tebipenem pivoxil dosage form reaches the neutral environment of the intestine, its active ingredients are then dissolved and become available for absorption into the bloodstream. In an embodiment, an enteric g may be a polymer applied on the tebipenem pivoxil core. The polymer may be selected from a poly(acrylate), a poly(methacrylate), a methyl acrylate—methacrylic acid copolymer, a methyl methacrylate—methacrylic acid copolymer, shellac, cellulose acetate phthalate (CAP), cellulose acetate succinate, hydroxypropyl methyl cellulose ate, hydroxypropyl methyl cellulose e succinate (hypermellose acetate succinate), polyvinyl acetate phthalate (PVAP), cellulose acetate trimellitate, sodium alginate, zein, and a combination thereof. The enteric coating may further include a base such as an alkali metal hydroxide, an alkali metal carbonate, an alkali metal hydrocarbonate, a ation thereof, but is not limited thereto.
In certain embodiments the immediate release c coating comprises a poly(methacrylate) and a base. In an embodiment, the base can be a metal hydroxide, for example, sodium hydroxide or potassium hydroxide. In another embodiment, the base can be a metal carbonate, for example, sodium carbonate or potassium carbonate, or a metal hydrocarbonate, for example, sodium arbonate or potassium hydrocarbonate. Other suitable coatings include polyvinyl alcohol, n grades of hydroxylpropyl cellulose, and mixtures of HPMC and polyethylene glycol (PEG). The coating may additionally include a plasticizer. r, the immediate release coating is not limited thereto and any conventional coating that is insoluble at low pH (pH less than about 3) but that will immediately releasing the active ingredient at neutral or basic pH can also be used. The ate release enteric coating may have a thickness of about 20 micrometers to about 100 micrometers. The weight gain of the coating can be from about 0.1 to 100% weight gain based on the weight of the tablet core, or about 2 to about 75%, about 2 to about 50%, about 2% to about 15%, about 2% to about 10%, or about 4% to about 8%.
The coating can be an ate release coating or in some embodiments the coating is a ed release coating. Suitable coatings include hypromellose or hypromellose USP also known as hydroxypropyl methylcellulose (HPMC). HPMC is a water soluble excipient that may be used film coating agent, although certain grades of HPMC are more commonly used as binders. PHARMACOA’I' tsn Chemical-(30., Ltd). PHARMACOAT 603 is a hypennellose grade having snbstitntinn type 2910 and a labeled viscosity of 3 mPa—s that is suitable for use as a film coating. The ()PADRY tradenarne is. used for a variety of pharmaceutical coatings, and includes immediate release coatings enmprisedprimarily 0f HPMC (hydroxypropyl methyl cellulose or hypromellose) or PVA (polyvinyl alcohol) and PEG thylene glycol), PEG may be used as a plasticizer in OPADRY coatings. EUDRAGITs, such as IT L l00-—55, are another suitable class of coatings. EUDRAGI’I‘S are copolymers derived from esters of acrylic and methacrylic acid, with additional functional groups in some cases.
The coating may include additional excipients such as film formers, printing inks, buffering agents, pH adjusters, vatives, dyes, and flavors. In some instances, a single material will include any two or more of any of the foregoing excipients.
An amount of tebipenem pivoxil in the uncoated tablet may be 100 mg and the uncoated tablet weight may be 200 mg. In another example, an amount of tebipenem pivoxil in the uncoated tablet may be 500 mg and the uncoated tablet weight may be 1,000 mg. In another example the weight of tebipenem l HBr in the uncoated tablet may be 348.9 mg, the weight of tebipenem pivoxil in the uncoated tablet may be 300 mg, and the total weight of the uncoated tablet may be 750 mg. A thickness of the 500 mg tablet may be from 5.00 to 8.00 millimeters (mm), or from 6.00 to 7.00 mm, or from 6.50 to 7.00 mm, or from 6.75 to 7.00 mm.
An amount of the coating of the tablet is from 1 to 5 weight percent of the total weight of the tablet, for example, from 2 to 3 weight percent of the total weight of the tablet.
Embodiments may be combined only so long as a stable solid formulation or its core results. "A ation of any of the foregoing" only includes ations that results in a stable tablet or tablet core.
METHODS OF TREATMENT The disclosure includes a method of ng a bacterial ion in a subject by administering a dosage form the disclosure to a subject at risk for a ial infection or suffering from a bacterial infection.
Treatment of human patients is particularly contemplated. However, treatment of non—human subjects, such as livestock or companion animals, is within the scope of the disclosure.
In some ments, the bacterial infection or antibiotic—tolerant or antibiotic— resistant infection is caused by a egative bacterium.
The extended release tebipenem pivoxil oral dosage form of this disclosure may be administered alone, so that tebipenem pivoxil is the only active agent administered to the t or the dosage form may be administered in combination with one or more onal active agents. Combination administration es concurrent or sequential administration of active agents.
In an embodiment of any of the s of this disclosure, the microbial infection is the result of a enic bacterial infection. Examples of pathogenic bacteria include, Without limitation, bacteria Within the genera Aerobacter, Aeromonas, Acinetobacter, cterium, Bacillus, Bacteroides, Bartonella, Bordetella, Brucella, Burkholderia, Calymmatobacterium, Campylobacter, acter, Clostridium, Corynebacterium, bacter, Enterococcus, Escherichia, Francisella, Fusobacterium, Haemophilus, Hafnia, Helicobacter, Klebsiella, Legionella, Listeria, ella, Moraxella, Porphyromonas, Proteus, Providencia, Pseudomonas, Salmonella, Serratia, Shigella, Staphylococcus, Streptococcus, Treponema, Xanthomonas, Vibrio, and Yersinia. Specific examples ofsuch bacteria include Vibrio harveyi, Vibrio cholerae, Vibrio parahemolyticus, Vibrio alginolyticus, Pseudomonas phosphoreum, Pseudomonas aeruginosa, Yersinia enterocolitica, Escherichia coli, Salmonella typhimurium, Haemophilus influenzae, Helicobacter pylori, us subtilis, Borrelia burgdorferi, Neisseria meningitidis, Neisseria gonorrhoeae, Yersinia pestis, Campylobacter jejuni, Mycobacterium tuberculosis, Enterococcus faecalis, Streptococcus pneumoniae, Streptococcus pyogenes, Klebsiella pneumoniae, Burkholderia cepacia, Acinetobacter baumannii, Staphylococcus epidermidis, and Staphylococcus aureus.
] In some ments, the Gram—negative bacterium is a Pseudomonas, e. g., P. aeruginosa, Burkholderia or an obacter, e. g., A. nii.
In some embodiments, the Gram—negative bacterium is an Enterobacteriaceae, e.g., Klebsiella pneumonia, e. g., Escherichia coli, e.g., Enterobacter cloacae, e.g., Serratia marcescens, e. g., Salmonella typhimurium, e. g., Shigella dysenteriae, e. g., Proteus mirabilis, e.g., Citrobacterfreundii, e.g., ia pestis.
In some embodiments, the ion is a polymicrobial infection, e.g., an infection comprising more than one organism. In some embodiments, the infection ses at least one of the organisms listed above, e.g., one or more of Pseudomonas, e. g., P. aeruginosa, Klebsiella, e. g., Klebsiella pneumoniae, and/or Acinetobacter, e.g., A. baumannii.
In some ments, the methods further include administering an additional active agent in combination the modified release solid dosage form the disclosure, such as an 2017/066729 antibiotic selected from the group consisting of but not limited to: beta—lactams such as penicillins, cephalosporins, carbacephems, ycins, carbapenems, monobactams, quinolones including fluoroquinolones and similar DNA synthesis inhibitors, tetracyclines, aminoglycosides, macrolides, glycopeptides, chloramphenicols, glycylcyclines, amides, lipopeptides, lipodepsipeptides, such as daptomycin, and oxazolidinones.
In some embodiments, the bacterial infection is an upper and lower respiratory tract infection, pneumonia, bacteremia, a systemic infection, sepsis and septic shock, a urinary tract infection, a gastrointestinal infection, endocarditis, a bone infection, central nervous system infections such as meningitis, or an infection of the skin and soft tissue. The disclosure includes a method of treating C. difi‘icile by administering a dosage form of the disclosure to a patient in need of such treatment. The disclosure particularly includes a method of treating a cated urinary tract infection in a patient. A "complicated urinary tract infection" is a clinical syndrome characterized by pyuria and a documented microbial pathogen on culture of urine blood, accompanied by local and systemic signs and symptoms, including fever (i.e., oral or tympanic ature greater than 38 degrees Celsius), chills, malaise, flank pain, back pain, and/or costo— vertebral angle pain or tenderness, that occur in the presence of a functional or anatomical abnormality of the urinary tract or in the presence of catheterization. Patients with pyelonephritis, regardless of underlying abnormalities of the y tract, are considered a subset of patients with cUTIs.
In some embodiments, the t is a mammal, e.g., a human or man mammal. In some embodiments, the methods include treating one or more cells, e.g., cells in a culture dish.
In one aspect, the present sure features a method of treating a Gram— negative infection in a subject, the method comprising stering to said subject in need of such treatment a therapeutically ive amount of a nd described herein.
In some embodiments, the Gram—negative infection is caused by monas aeruginosa.
In other ments the disclosure includes treating an infection caused by Gram— positive bacteria, such as Staphylococcus epidermidis and Staphylococcus aureus.
In some embodiments, the subject is a trauma patient or a burn t suffering from a burn or skin wound.
In a further aspect, the present disclosure features a method of reducing ial tolerance in a subject, the method comprising administering to said subject a therapeutically effective amount of a nd described herein.
In some embodiments, the method further includes identifying said subject suffering from an ion with bacteria resistant to antimicrobial therapy.
EXAMPLES TABLET MANUFACTURE The tebipenem pivoxil s use a common blend that is manufactured via dry granulation such as roller compaction ed by milling. Conventional blending/milling and ing equipment and processes are used. Conventional film coating equipment and ses can be used to coat the tablet cores.
Tebipenem pivoxil blends can be prepared by a variety of granulation processes, including spray drying, t wet granulation, aqueous wet granulation and dry granulation using roller tion; however, dry granulation using roller compaction efficiently produces tebipenem pivoxil tablet blends with desirable bulk density and flow properties.
The manufacturing process of nem pivoxil tablets is optimized through evaluation of blend and tablet physical properties including bulk and tap density measurement, flow analysis, screen analysis, and uniformity of the blend; weight, thickness, hardness, friability, potency, disintegration, dissolution and content uniformity tests on the tablet cores and tablets.
EXAMPLE 1. TEBIPENEM PIVOXIL IMMEDIATE RELEASE TABLET CORES Tebipenem pivoxil, crospovidone (PVPP XL—lO), monohydrate lactose, and microcrystalline cellulose l PH102) are weighed and placed into a container. These ingredients are then sieved through a mesh 30 sieve.
The sieved ingredients are blended for 10 minutes at 20 rpm using a single arm fixed hopper mixing machine.
To the resulting mixture of the blended ingredients, magnesium stearate is added in the mixing machine, the mixture is further blended for 5 minutes at 20 rpm.
The mixture is compressed into tablets with 9 mm diameter round punches using single—punch tablet press. 2017/066729 The compositions of the 500 mg and 100 mg immediate release tablet cores are listed in Tables 1 and 2, respectively. The dissolution profile of the 500 mg immediate release tebipenem pivoxil tablet is at pH 5.0 is shown in TABLE 1 Components on Quantity per 500 mg tablet (mg) tebipenem pivoxil Active 50.0 500.0 (free base) Crospovidone (PVPP Disintegrant 5.0 50.0 XL—10) Monohydrate Lactose Diluent 21.5 215.0 rystalline Binder 21.5 215 .0 cellulose (Avicel PH102) ium Stearate Lubricant 2.0 20.0 Total 100.00 1,000 TABLE 2 Components Function Quantity per 100 mg tablet (mg) tebipenem pivoxil Active 50.0 100.0 (free base) Crospovidone (PVPP Disintegrant 5.0 10.0 XL—10) Monohydrate Lactose Diluent 21.0 42.0 Microcrystalline Binder 21.0 42.0 cellulose (Avicel PH102) Magnesium Stearate Lubricant 2.0 4.0 SiOz Filler 1 .0 2.0 Table Core Total 100.00 200 Film Coating (see below) 7.0 14.0 Tablet Weight 107.0 214.0 EXAMPLE 2. IMMEDIATE RELEASE COATING ATION AND PROCESS Immediate release nem pivoxil table cores, such as those disclosed in Tables 1 and 2, can be coated with coating formulations listed in Tables 3 and 4. The ution profile of a 500 mg enteric coated immediate release tebipenem pivoxil is provided in The formulation of the tablet core is provided in Example 1 and the formulation of the enteric coating is provided in Table 3. The resulting enteric coated dosage form can release 100% of the tebipenem pivoxil at pH 6.8 in 45 minutes, but releases less than 5% of the tebipenem pivoxil at pH 1.2 in 45 minutes. The following abbreviations are used: GMS (glycerol monostearate) and TEC (Triethyl citrate).
Eudragit Dispersion (a) Eudragit Dispersion (b) The it Dispersion (a) is prepared as follows. Eudragit L100—55 is weighed and added slowly into the water under stirring that forms a vortex. The mixture is d for 10 minutes to make the polymer wet tely. An aqueous 4% sodium hydroxide solution is added during a 5 minute period to the ing dispersion by drops, and the resulting mixture is further stirred for 30 minutes.
The Eudragit Dispersion (b) is prepared as follows. A mixture of Tween 80, TEC, and GMS is added to water heated to 80 OC, and the ing mixture was homogenized for 10 minutes at a speed of 5,000 rpm. Additional water (25% of the original ) is added to the dispersion under stirring. The resulting mixture is cooled to ambient temperature. The prepared dispersion is then added to Eudragit Dispersion (a) ed as described above under stirring. After 10 minutes of stirring, the resulting mixture is passed through a mesh 80 sieve.
The resulting dispersion can be used to coat tebipenem pivoxil immediate release tablet cores (weight gain 7%) ing to the parameters listed in Table 5. The dissolution profiles of uncoated tebipenem pivoxil immediate release tablet core (formulation in Example 1) and enteric coated tebipenem pivoxil.
Temperature (inlet) 45—55 °C Temperature (material) 33-35 °C Fan frequency (inlet) 900 Hz Fan ncy(outlet) l,500 Hz Flow rate 2—5 g/min Atomization pressure 2.5 bar Pan speed 7—l5 rpm EXAMPLE 3. TEBIPENEM L MODIFIED RELEASE TABLET CORE ITION Modified release tebipenem pivoxil tablet cores of Tables 6 and 7 are manufactured as follows. First, a half portion of hydroxypropyl methyl cellulose (HPMC E50 LV) is charged into a blender, such as a V—blender or bin blender, followed by addition of tebipenem pivoxil and then the ing half portion of hydroxypropyl methyl cellulose, and then blending the als.
The ium stearate may be screened to break up any agglomerates such as through a 20 mesh screen. The screened magnesium stearate is added to the blender containing the tebipenem pivoxil/release control agent blend and blended for several minutes. The nem pivoxil/ release control agent / magnesium stearate blend from the blender is discharged.
The material discharged from the blender is roller compacted to form roller compacted ribbons or ts. The roller compacted material is then passed through a mill, such as an oscillating mill, impact mill, or screening mill. For e, a QUADRO COMIL o Engineering, Ontario, Canada) equipped with a 20 mesh screen may be used. The milled material is collected and then charged into a blender. Microcrystalline cellulose (Avicel PH102) and mannitol (Mannitol 200 SD) are added and the materials are blended.
Magnesium te may be screened to break up any agglomerates such as through a 20 mesh screen into the blender and blended several minutes.
The contents of the blender are then discharged into an appropriately labeled container, double lined with polyethylene bags with a desiccant between the two layers of bags.
Tablet cores are then formed on a rotary tablet press. Tablet cores may be coated in a film coater.
The compositions of the 500 mg (total weight 1000 mg) and 100 mg (total weight 200 mg) modified release tablet cores are listed in Tables 6 and 7, respectively.
Components Function Quantity per 1,000 mg tablet (mg) nem l Active 50.0 HPMC E50 LV Release 20.0 Control Agent 14-0 rystalline Binder 14.0 cellulose (Avicel PH102) Magnesium Stearate Lubricant 2.0 10000 TABLE 7 Components Function Amount Quantity per (%) 200 mg tablet (mg) __--tebipenem pivoxil Active 50.0 100.0 Components Function Amount Quantity per (%) 200 mg tablet (mg) HPMC E50 LV Release Control 20.0 40 0 Agent Mannitol 200 SD Diluent .0 28 .0 Microcrystalline 14 .0 28 .0 cellulose (AVicel PH102) Magnesium Stearate ant 2.0 SiOz Filler 2.0 10000 200 The tablet was prepared by a direct compression. The tooling size was 18.9 by 11.0 mm. The thickness of the tablet was 6.83 to 6.90 mm.
E 4. DISSOLUTION The dissolution method parameters are listed in Table 8.
TABLE 8 Drug Product/Formulation tebipenem PiVOXfl Tablet: 1,000 mg total core weight API: 500 m_ Dissolution Media pH 5.0 acetate buffer Media size 900 mL Replacement Volume Paddle speed 50 rpm (3-4 h, 200 rpm) Sampling Time Points 0.5, 1, 2, 3, and 4 h Diluent for Standard pH 5.0 buffer solution Standard Concentration 100 ug/mL Sample concentration 11.11—111.11ug/mL(10%-100% of drug e) range Welch l, XB-C18, 250 mm X 4.6 mm, 5 pm; Lot Column No. 02; Part# 00210-31043; Serial# 211603926 50 mM Ammonium Acetate / Acetonitrile / Triethylamine Mobile Phase (5824022, V/V/V), pH 3.5 (adjusted by phosphoric acid) Flow Rate, mL/min UV Wavelength, nm Injection , uL 10 EXAMPLE 5. SIMULATED HUMAN TEBIPENEM PIVOXIL COKINETIC PARAMETERS FOR IMMEDIATE RELEASE DOSAGE FORM The pharmacokinetic ters shown in Tables 9 and 10 were Obtained from a simulated study of the human orally—administered (PO) dose of the 500 mg immediate release tebipenem pivoxil tablet. The formulation of this tablet core is provided in Example 1. The simulation assumes that in viva dissolution of Tebipenem Pivoxil tablet is r to the measured in vitro dissolution.
The simulated pharmacokinetic profile of the human orally—administered (PO) dose of 500 mg Tebipenem Pivoxil tablet is shown in The simulation shows the plasma concentration of tebipenem (free form) in the venous return, i.e. the plasma concentration of tebipenem in the venous tment or systemic circulation.
TABLE 9 Time (hours) Amount Dissolved (%) 0.08 86.4 0.17 98.7 0.25 98.5 0.33 98.9 0.50 98.6 0.75 97.7 EXAMPLE 6. IMMEDIATE RELEASE TEBIPENEM PIVOXIL HBR TABLETS Immediate release tebipenem pivoxil HBr tablets are manufactured by the method given in Example 1. Tebipenem pivoxil free base is substituted by the HBr salt. Less e and Avicel PH102 is used to account for the greater weight of the tebipenem pivoxil salt. The composition of uncoated immediate release tebipenem pivoxil HBr tablets is given in Tables 11A and 11B. The 750 mg (total weight) tablet core disclosed in Table 11B was found to e 95.0% of the tebipenem pivoxil in 5 minutes, and approximately 95.2% of the tebipenem pivoxil in 15 minutes when measured by the USP Paddle method at 50 rpm in 900 mL of 50 mM acetate buffer, pH 5.0 at 37 OC. The dissolution profile is provided at The tablet core in Table 11B contains 300 mg tebipenem pivoxil.
TABLE 11A Components Function ty per 100 mg tablet (mg) tebipenem pivoxil Active 58.2 116.3 Crospovidone (PVPP Disintegrant 5.0 10.0 XL-10) drate Lactose Diluent 16.9 33.8 Microcrystalline Binder 17.0 42.0 ose (Avicel PH102) Magnesium Stearate Lubricant 2.0 4.0 SiOz Filler 1 .0 2.0 Table Core Total 100.00 200 TABLE 1113 Components Function Amount ty per 300 (%) mg tablet (750 total weight) tebipenem pivoxil Active 46.5 348.9 HBr (40% free base) PVPP XL— 10 Disintegrant 5 .0 37.5 (crospovidone) 2017/066729 TABLE 11B Components Function Amount Quantity per 300 (%) mg tablet (750 total weight) Mannitol 200 SD Diluent 23.2 174.3 Microcrystalline Binder 23.2 174.3 cellulose (Avicel PH102) Magnesium Stearate Lubricant 1.0 7.5 SiOz Filler 1.0 7.5 Total 100.0 750.0 EXAMPLE 7. MODIFIED E TEBIPENEM P1vox1L HBR TABLETS Modified release tebipenem pivoxil HBr tablets are manufactured by the method given in Example 2. Tebipenem pivoxil free base is substituted by the HBr salt. Less lactose and Avicel PH102 is used to account for the greater weight of the tebipenem pivoxil salt. The composition of es of uncoated modified release tebipenem pivoxil HBr tablets is given in Tables 12A and 12 B. The dissolution profile of the uncoated modified release tebipenem pivoxil tablet in Table 12A at pH 5.0 and pH 6.8 is compared to that of the free base in and FIG 6. The HBr salt exhibited a slightly slower release than the free base at pH 5.0 and a slightly faster e than the free base at pH 6.8.
TABLE 12A Components Function Amount Quantity per (%) 200 mg tablet (mg) nem pivoxil Active 58.2 116.3 HPMC K4M Release l 20.0 40.0 Agent Mannitol 200 SD Diluent 14.0 19.9 Microcrystalline Binder 14.0 19.8 cellulose (Avicel PH102) ium Stearate Lubricant 1.0 2.0 SiOz Filler 1.0 2.0 100.00 200.00 TABLE 123 Components Function Quantity per 300 mg tablet (750 total weight) tebipenem pivoxil Active 46.5 HBr (40% free base) HPMC E3 35.0 Agent ol 200 SD 8.2 Microcrystalline Binder 8.2 cellulose (Avicel PH102) Magnesium Stearate 1.0 $102 1-0 Total — 100-00 The 750 mg tablet core disclosed in Table 12B was found to release 21.9% of the tebipenem pivoxil in 30 minutes, 40.9% of the tebipenem l in 1 hour, 70.7% of the of the tebipenem pivoxil in 2 hours, 88.7% of the tebipenem pivoxil in 3 hours, and 100% of the of the tebipenem pivoxil in 4 hours when ed by the USP Paddle method at 50 rpm in 900 mL of 50 mM e buffer, pH 5.0 at 37 OC. The dissolution profile is provided at EXAMPLE 8. SHORT MODIFIED RELEASE TEBIPENEM PIVOXIL HBR FORMULATION Short modified release tebipenem pivoxil HBr s are manufactured by the method given in Example 2. This formulation releases NLT 85% of the tebipenem pivoxil from the dosage form core in 2 hours when measured by the USP Paddle method at 50 rpm in 900 mL of 50 mM acetate , pH 5.0 at 37 OC. Tebipenem pivoxil free base is substituted by the HBr salt. The composition of examples of uncoated modified release tebipenem pivoxil HBr tablets is given in Table 13. The 750 mg tablet core disclosed in Table 13 was found to release 37.5% of the nem pivoxil in 30 minutes, 69.8% of the tebipenem pivoxil in 1 hour, 99.9% of the of the tebipenem pivoxil in 2 hours, and 100.0% of the tebipenem pivoxil in 3 hours when measured by the USP Paddle method at 50 rpm in 900 mL of 50 mM e buffer, pH 5.0 at 37 °C.The dissolution profile of the short modified release tebipenem pivoxil HBr tables is given in TABLE 13 Components Function Quantity per 300 mg tablet (750 total weight) tebipenem pivoxil Active 46.5 HBr (40% free base) PVP K12 30.0 Agent Mannitol 200 SD 10.7 rystalline Binder 10.7 cellulose (Avicel PH102) Magnesium Stearate 1.0 SiOz 1-0 Total — 100-00 EXAMPLE 9. PK PARAMETERS FOR ORAL ADMINISTRATION TO MONKEYS The 100 mg immediate release tebipenem pivoxil free base (Free Base IR) and HBr (HBr IR) tablets Examples 1 and 6 and the 100 mg ed release tebipenem pivoxil HBr tablet of Example 7 (HBr MR) were orally administered to Cynomolgus monkeys (N=4 animals/ tested dosage form). Each animal received a one week wash out prior to the next dose and each served as its own control. Blood samples were collected into KzEDTA tubes before processing to plasma. Samples were ed at 15 minutes, 30 minutes, 1 hours, 2 hours, 4 hours, 6 hours, 8 hours, 10 hours, 12 hours, 16 hours, and 24 hours post administration. Plasma samples were analyzed for the prodrug, tebipenem pivoxil, and tebipenem via a tandem LC—MS method. Non— compartmental analysis used to derive pharmacokinetic parameters of the composite time vs. concentration profiles were performed for each dose level using Phoenix Winnonlin v. 6.4.
Plasma concentration of tebipenem for each time point was determined by HPLC. The PK parameters for the immediate release and ed release dosage forms are listed in Table 14.
] Maximal trations of tebipenem were observed at approximately 1.5 —2 hours in the free base and HBr immediate release . Cum was d with the modified release formulation compared to the ate release (free base and HBr) formulation but AUCau was relatively constant between the formulations. AUCinf could not be calculated due to the variability in the terminal elimination profile. Using previously generated IV data (13.06 ug*hr/mL for 10 mg/kg dose) bioavailability of nem was estimated to range 39—50%.
Mean residence time was increased in the extended release formulation.
Dosage Form MRTlast (hr) Tum (hr) Cmax (ug/ C12h (ug/ AUC mL) (hr* Mg/ —---------- Free Base IR 1.0 163122 9.2 17.5 HBrIR 0.82 -m 13.6 9.2 19.1 HBrMR (4 hr)- 1.3 1.3 6.04 ---1.4 The t inventive t has been described in terms of exemplified principles and embodiments, but those skilled in the art Will recognize that variations may be made and equivalents substituted for What is described Without departing from the scope and spirit of the disclosure as defined by the following claims.

Claims (30)

What is claimed is:
1. An immediate or ed e solid oral dosage form comprising a dosage form core, wherein the dosage form core comprises: tebipenem pivoxil HBr and excipients, wherein the weight to weight ratio of tebipenem l as a free base to the excipients in the core is from 1:2 to 2:1, and the ents include 5-40 % (w/w%) of a binder, 0.1-5% of a lubricant, and optionally 5-40% of a diluent.
2. The dosage form of claim 1, wherein the dosage form is an immediate release dosage form and es more than 85 % of the tebipenem pivoxil HBr in 15 minutes when measured by the USP Paddle method at 50 rpm in 900 mL of 50 mM acetate buffer, pH 5.0 at 37 °C.
3. The immediate release dosage form of claim 2, wherein the excipients additionally comprise 1-50 wt% of a disintegrant.
4. The immediate release dosage form of claim 3, wherein the disintegrant is croscarmellose sodium, crospovidone, sodium starch glycolate, or any combination of any two or more of the foregoing.
5. The dosage form of claim 1, wherein the dosage form is a modified e dosage form and provides an in vitro release of not more than (NMT) 50 % of the tebipenem pivoxil HBr after 30 minutes when measured by the USP Paddle method at 50 rpm in 900 mL of 50 mM acetate buffer, pH 5.0 at 37 °C.
6. The modified release dosage form of claim 5, wherein not less than (NLT) 85% of the total amount of tebipenem pivoxil HBr is released after 4 hours, when measured by the USP Paddle method at 50 rpm in 900 mL of 50 mM acetate buffer, pH 5.0 at 37 °C.
7. The modified release dosage form of claim 5, wherein not less than (NLT) 85% of the total amount of tebipenem pivoxil HBr is released after 2 hours, when measured by the USP Paddle method at 50 rpm in 900 mL of 50 mM acetate buffer, pH 5.0 at 37 °C.
8. The modified release dosage form of any one of claims 5 to 7, wherein the excipients additionally comprise a release control agent.
9. The modified e dosage form of claim 8, wherein the release control agent is hydroxypropyl methylcellulose, hydroxypropyl cellulose, hypromellose acetate succinate, polyvinyl pyrrolidone, copovidone, or any combination of any two or more of the foregoing.
10. The dosage form of any one of claims 1 to 9, wherein the binder is microcrystalline cellulose, silicified microcrystalline cellulose, ethyl cellulose, lactose, or any ation of any two or more of the foregoing.
11. The dosage form of claim 10, wherein the binder is microcrystalline ose.
12. The dosage form of any one of claims 1 to 11, wherein the dosage form comprises a diluent and the diluent is starch, sucrose, monohydrate lactose, mannitol, sorbitol, or any combination of any two or more of the foregoing.
13. The dosage form of any one of claims 1 to 12, wherein the dosage form comprises a lubricant and the lubricant is a fatty acid ester, stearic acid, magnesium stearate, glyceryl behenate, calcium stearate, sodium stearyl fumarate, sodium lauryl sulfate, magnesium lauryl sulfate, sodium benzoate, talc, hydrogenated oil, Carnauba wax, or any combination of any two or more of the ing.
14. The ed release dosage form of any one of claims 5 to 7, wherein the dosage form core comprises 10% to 40% hydroxypropyl methylcellulose or polyvinylpyrrolidone, 5% to 20% ol, 5% to 20 % microcrystalline cellulose, and 0.1% to 5% magnesium stearate.
15. The immediate e dosage form of claim 2, wherein the dosage form core comprises 30 % to 60 % (w%/w) tebipenem pivoxil HBr, 1% to 10 % crospovidone, 15% to 25% monohydrate lactose or mannitol, 15 % to 25% microcrystalline cellulose, and 0.1% to 5% magnesium stearate.
16. The immediate release dosage form of claim 4 or claim 15, wherein the disintegrant is crospovidone and the weight to weight ratio of tebipenem pivoxil HBr to crospovidone is 20:1 to 5:1.
17. The immediate release dosage form of any one of claims 2 to 4 or claim 15 wherein the dosage form core does not include hydroxypropyl methylcellulose.
18. The dosage form of any one of claims 1 to 17, wherein the dosage form core comprises 100, 300, 500, 600, 900, or 1000 milligrams of nem pivoxil HBr.
19. The dosage form of any one of claims 1 to 18, wherein the dosage form core is a tablet core.
20. The dosage form of claim 19, wherein nem pivoxil as a free base comprises at least 55% by weight of the tablet core.
21. The dosage form of claim 20, wherein tebipenem pivoxil as a free base comprises at least 60% by weight of the tablet core.
22. The dosage form of any one of claims 1 to 18, wherein the dosage form core is a granule.
23. The dosage form of claim 22, wherein the es are in a capsule.
24. The dosage form of any one of claims 1 to 19, wherein the dosage form core further comprises a coating.
25. The dosage form of claim 19, wherein the tablet core is coated to form a coated tablet, and the coated tablet has a thickness of 6.00 to 7.00 millimeters.
26. The dosage form of claim 25, wherein the tablet core is coated with a coating that ses from 2% to 5% (w%/w) of the total weight of the coated tablet.
27. The dosage form of any one of claims 1 to 26, wherein the dosage form is ated for twice a day or three times a day administration.
28. Use of an oral dosage form of any one of claims 1 to 27 in the manufacture of a medicament for treating a urinary tract infection.
29. Use of an oral dosage form of any one of claims 1 to 27 in the manufacture of a medicament for treating a ial infection.
30. Use of an oral dosage form of any one of claims 1 to 27 in the manufacture of a medicament for treating a Mycobacterial tuberculosis infection. Spero Therapeutics, Inc. By the patent attorneys for the applicant SPRUSON & FERGUSON
NZ753713A 2016-12-15 2017-12-15 Novel tebipenem pivoxil immediate and modified release oral dosage forms NZ753713B2 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
US201662434643P 2016-12-15 2016-12-15
US62/434,643 2016-12-15
US201662438071P 2016-12-22 2016-12-22
US62/438,071 2016-12-22
PCT/US2017/066729 WO2018112372A1 (en) 2016-12-15 2017-12-15 Novel tebipenem pivoxil immediate and modified release oral dosage forms

Publications (2)

Publication Number Publication Date
NZ753713A NZ753713A (en) 2021-09-24
NZ753713B2 true NZ753713B2 (en) 2022-01-06

Family

ID=

Similar Documents

Publication Publication Date Title
AU2020281095B2 (en) Novel tebipenem pivoxil immediate and modified release oral dosage forms
AU2008309385B2 (en) Zibotentan composition containing mannitol and/or microcrystalline cellulose
US20070059365A1 (en) Novel formulation of ropinirole
US11529326B2 (en) Eflornithine and sulindac, fixed dose combination formulation
WO2011102506A1 (en) Sustained-release solid preparation for oral use
JP5749247B2 (en) Oral sustained-release solid preparation
GB2414668A (en) Sustained release delivery system for tetracycline compounds
BG107372A (en) Sustained-release preparations of quinolone antibiotics and method for preparation thereof
US20130344148A1 (en) Oral Composition
WO2021078835A1 (en) Pharmaceutical composition comprising selexipag
NZ753713B2 (en) Novel tebipenem pivoxil immediate and modified release oral dosage forms
US20080206329A1 (en) Modified Release Ciprofloxacin Compositions
EA044993B1 (en) IMMEDIATE RELEASE ORAL DOSAGE FORM CONTAINING TEBIPENEM PIVOXIL
TWI743059B (en) Eflornithine and sulindac, fixed dose combination formulation
US20210346303A1 (en) Coating method
WO2016166732A1 (en) Colchicine salicylate and uses thereof