US20100204178A1 - Novel parenteral carbamazepine formulation - Google Patents

Novel parenteral carbamazepine formulation Download PDF

Info

Publication number
US20100204178A1
US20100204178A1 US12/571,039 US57103909A US2010204178A1 US 20100204178 A1 US20100204178 A1 US 20100204178A1 US 57103909 A US57103909 A US 57103909A US 2010204178 A1 US2010204178 A1 US 2010204178A1
Authority
US
United States
Prior art keywords
carbamazepine
cyclodextrin
cbz
complex
oral
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.)
Abandoned
Application number
US12/571,039
Inventor
James Cloyd
Angela Birnbaum
Ilo Leppik
Stephen D. Collins
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.)
Lundbeck LLC
Original Assignee
Lundbeck 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
Priority claimed from PCT/US2006/038508 external-priority patent/WO2007041524A2/en
Priority to US12/571,039 priority Critical patent/US20100204178A1/en
Application filed by Lundbeck LLC filed Critical Lundbeck LLC
Publication of US20100204178A1 publication Critical patent/US20100204178A1/en
Assigned to OVATION PHARMACEUTICALS reassignment OVATION PHARMACEUTICALS ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: BIRNBAUM, ANGELA, CLOYD, JAMES, LEPPIK, ILO, COLLINS, STEPHEN D.
Assigned to LUNDBECK INC. reassignment LUNDBECK INC. CHANGE OF NAME (SEE DOCUMENT FOR DETAILS). Assignors: OVATION PHARMACEUTICALS, INC.
Priority to US13/547,866 priority patent/US9629797B2/en
Assigned to LUNDBECK LLC reassignment LUNDBECK LLC MERGER (SEE DOCUMENT FOR DETAILS). Assignors: LUNDBECK INC.
Priority to US13/679,715 priority patent/US20130072476A1/en
Priority to US14/051,938 priority patent/US9770407B2/en
Priority to US15/657,470 priority patent/US20180153797A1/en
Priority to US16/939,390 priority patent/US20210196623A1/en
Priority to US17/501,287 priority patent/US20220265547A1/en
Abandoned legal-status Critical Current

Links

Images

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0019Injectable compositions; Intramuscular, intravenous, arterial, subcutaneous administration; Compositions to be administered through the skin in an invasive manner
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/55Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having seven-membered rings, e.g. azelastine, pentylenetetrazole
    • 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
    • A61K31/715Polysaccharides, i.e. having more than five saccharide radicals attached to each other by glycosidic linkages; Derivatives thereof, e.g. ethers, esters
    • A61K31/716Glucans
    • A61K31/724Cyclodextrins
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/36Polysaccharides; Derivatives thereof, e.g. gums, starch, alginate, dextrin, hyaluronic acid, chitosan, inulin, agar or pectin
    • A61K47/40Cyclodextrins; Derivatives thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P25/00Drugs for disorders of the nervous system
    • A61P25/08Antiepileptics; Anticonvulsants
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08BPOLYSACCHARIDES; DERIVATIVES THEREOF
    • C08B37/00Preparation of polysaccharides not provided for in groups C08B1/00 - C08B35/00; Derivatives thereof
    • C08B37/0006Homoglycans, i.e. polysaccharides having a main chain consisting of one single sugar, e.g. colominic acid
    • C08B37/0009Homoglycans, i.e. polysaccharides having a main chain consisting of one single sugar, e.g. colominic acid alpha-D-Glucans, e.g. polydextrose, alternan, glycogen; (alpha-1,4)(alpha-1,6)-D-Glucans; (alpha-1,3)(alpha-1,4)-D-Glucans, e.g. isolichenan or nigeran; (alpha-1,4)-D-Glucans; (alpha-1,3)-D-Glucans, e.g. pseudonigeran; Derivatives thereof
    • C08B37/0012Cyclodextrin [CD], e.g. cycle with 6 units (alpha), with 7 units (beta) and with 8 units (gamma), large-ring cyclodextrin or cycloamylose with 9 units or more; Derivatives thereof
    • CCHEMISTRY; METALLURGY
    • C08ORGANIC MACROMOLECULAR COMPOUNDS; THEIR PREPARATION OR CHEMICAL WORKING-UP; COMPOSITIONS BASED THEREON
    • C08BPOLYSACCHARIDES; DERIVATIVES THEREOF
    • C08B37/00Preparation of polysaccharides not provided for in groups C08B1/00 - C08B35/00; Derivatives thereof
    • C08B37/0006Homoglycans, i.e. polysaccharides having a main chain consisting of one single sugar, e.g. colominic acid
    • C08B37/0009Homoglycans, i.e. polysaccharides having a main chain consisting of one single sugar, e.g. colominic acid alpha-D-Glucans, e.g. polydextrose, alternan, glycogen; (alpha-1,4)(alpha-1,6)-D-Glucans; (alpha-1,3)(alpha-1,4)-D-Glucans, e.g. isolichenan or nigeran; (alpha-1,4)-D-Glucans; (alpha-1,3)-D-Glucans, e.g. pseudonigeran; Derivatives thereof
    • C08B37/0012Cyclodextrin [CD], e.g. cycle with 6 units (alpha), with 7 units (beta) and with 8 units (gamma), large-ring cyclodextrin or cycloamylose with 9 units or more; Derivatives thereof
    • C08B37/0015Inclusion compounds, i.e. host-guest compounds, e.g. polyrotaxanes

Definitions

  • Carbamazepine or 5H-dibenz[b,f]azepine-5-carboxamide, is a widely used antiepileptic agent. It is available in the U.S. as Tegretol' brand chewable tablets of 100 mg, tablets of 200 mg and suspension of 100 mg/5 mL, intended for oral administration as a treatment for epilepsy or as a specific analgesic for trigeminal neuralgia. Other brand names include Equetro, Carbatrol, Tegretol XR and Epitol. Generic versions of these oral dosage forms are also available. Dosage forms include Carbatrol available in 100, 200, and 300 mg strengths; and Tegretol XR, available in 100, 200, and 400 mg strengths.
  • recommended maintenance dosage levels in adults and children over 12 years of age are 800-1200 mg daily, although up to 2400 mg daily have been used in adults.
  • the maintenance dosage level is usually 20-30 mg/kg/d and in children less than 6 years old the maintenance dosage level is usually 10-20 mg/kg/d.
  • carbamazepine is a widely used anticonvulsant drug. It is also of proven efficacy in the treatment of generalized tonic-clonic (grand mal) seizures. Carbamazepine has also been used in treating simple partial (focal or Jacksonian) seizures and in patients with mixed seizure patterns which include the above, or other partial or generalized seizures. It is not used in the treatment of absence seizures (petit mal).
  • carbamazepine has, in many respects, a more favorable profile in terms of the incidence and severity of side-effects than other anticonvulsants.
  • carbamazepine is less sedating and causes less intellectual function impairment than other antiepileptic drugs such as phenobarbital, primidone and phenyloin.
  • carbamazepine does not precipitate gingival hypertrophy, hirsutism, acne or other undesired effects associated with phenyloin. These attributes have helped to make carbamazepine the drug of choice in women and children.
  • CBZ carbamazepine
  • Cyclodextrins sometimes referred to as Schardinger's dextrins, were first isolated by V Amsterdam in 1891 as a digest of Bacillus amylobacter on potato starch. The foundations of cyclodextrin chemistry were laid down by Schardinger in the period 1903-1911. Until 1970, however, only small amounts of cyclodextrins could be produced in the laboratory and the high. production cost prevented the usage of cyclodextrins in industry. In recent years, dramatic improvements in cyclodextrin production and purification have been achieved and cyclodextrins have become much less expensive, thereby making the industrial application of cyclodextrins possible.
  • Cyclodextrins are cyclic oligosaccharides with hydroxyl groups on the outer surface and a void cavity in the center. Their outer surface is hydrophilic, and therefore they are usually soluble in water, but the cavity has a lipophilic character.
  • the most common cyclodextrins are ⁇ -cyclodextrin, ⁇ -cyclodextrin and ⁇ -cyclodextrin, consisting of 6, 7 and 8 ⁇ -1,4-linked glucose units, respectively. The number of these units determines the size of the cavity.
  • Cyclodextrins are capable of forming inclusion complexes with a wide variety of hydrophobic molecules by taking up a whole molecule (a “guest molecule”), or some part of it, into the void cavity. The stability of the resulting complex depends on how well the guest molecule fits into the cyclodextrin cavity.
  • Common cyclodextrin derivatives are formed by alkylation (e.g., methyl-and-ethyl ⁇ -cyclodextrin) or hydroxyalkylation of the hydroxyethyl-derivatives of ⁇ -, ⁇ -, and ⁇ -cyclodextrin) or by substituting the primary hydroxyl groups with saccharides (e.g., glucosyl- and maltosyl- ⁇ -cyclodextrin).
  • cyclodextrins have been used to increase the solubility, dissolution rate and/or stability of a great many compounds, it is also known there are many drugs for which cyclodextrin complexation either is not possible or yields no advantages. See J. Szejtli, Cyclodextrins in Drug Formulations: Part II, Pharmaceutical Technology, 24-38, August, 1991. Despite this potential pharmaceutical utility, certain cyclodextrins are have limitations.
  • Cyclodextrins and their derivatives are mostly crystalline solids. Concentration of some cyclodextrins in the renal tissue is followed by crystal formation causing necrotic damage to the cells. Despite forming water soluble clathrate complexes, the crystalline cyclodextrin drug complexes have generally been limited in their utility to sublingual or topical administration.
  • U.S. Pat. Nos. 5,134,127 and 5,376,645, whose disclosures are incorporated herein by reference, are directed to novel cyclodextrin derivatives, in particular sulfoalkyl cyclodextrin derivatives, that overcome the limitations of other cyclodextrins.
  • the sulfoalkyl cyclodextrin derivatives disclosed therein exhibit lower nephrotoxicity while exhibiting high aqueous solubility.
  • the present invention is based, inter alia, on the determination that carbamazepine stable inclusion complexes with cyclodextrins are highly water soluble relative to the non-complexed drug.
  • the carbamazepine-cyclodextrin inclusion complexes of the invention result in an injectable formulation that provides significant benefits and advantages over other carbamazepine formulations.
  • the carbamazepine-cyclodextrin inclusion complexes of the present invention are completely bioavailable, delivering 100% of the dose to the bloodstream in a consistent and predictable manner which is not the case with solid oral dosage forms.
  • the carbamazepine-cyclodextrin inclusion complexes of the present invention can be administered to a patient suffering from a generalized tonic-clonic or other acute seizure via a peripheral rather than oral route.
  • the carbamazepine-cyclodextrin inclusion complexes of the present invention satisfy a significant unmet medical need for a stable injectable formulation of carbamazepine that overcomes the limitations of poorly soluble and variably absorbed oral formulations.
  • the present invention contemplates a carbamazepine-cyclodextrin inclusion complex useful for the parenteral administration of carbamazepine comprising a carbamazepine complexed with a modified cyclodextrin.
  • the modified cyclodextrin is a sulfoalkyl-cyclodextrin.
  • a preferred modified cyclodextrin is sulfobutylether-7-beta-cyclodextrin.
  • the inclusion complex preferably has a concentration of about 5 to about 50 mg/ml carbamazepine, and more preferably a concentration of about 10 mg/ml carbamazepine.
  • a carbamazepine-cyclodextrin inclusion complex useful for the parenteral administration of carbamazepine in which dosing is about 30% to about 100% of oral maintenance doses, or preferably about 65% to 75% of oral maintenance doses.
  • the present invention provides a carbamazepine-cyclodextrin inclusion complex useful for the parenteral administration of carbamazepine having a half-life of about 8 to about 65 hours, and more preferably having a half-life of about 24 hours.
  • the present invention contemplates a carbamazepine-cyclodextrin inclusion complex useful for the parenteral administration of carbamazepine having an area under the plasma concentration-time curve (AUC) of about 70% to about 130% of the AUC for an oral carbamazepine dosage form, and more preferably having an AUC of about 80% to about 125% of the AUC for an oral carbamazepine dosage form.
  • AUC plasma concentration-time curve
  • the present invention contemplates a carbamazepine-cyclodextrin inclusion complex useful for the parenteral administration of carbamazepine having a minimum plasma concentration (Cmin) of about. 70% to about 130% of the Cmin for an oral carbamazepine dosage form, and more preferably having a Cmin of about 80% to about 125% of the Cmin for an oral carbamazepine dosage form.
  • Cmin minimum plasma concentration
  • the present invention provides a carbamazepine-cyclodextrin inclusion complex useful for the parenteral administration of carbamazepine having an intravenous dosing interval of every four to twelve hours, more preferably having an intravenous dosing interval of every six hours, and still more preferably having an intravenous dosing interval of every eight hours.
  • the present invention provides a method of administering a carbamazepine-cyclodextrin inclusion complex useful for the parenteral administration of carbamazepine comprising: 1) providing a carbamazepine-cyclodextrin inclusion complex; and 2) infusing the complex intravenously to a patient in need thereof every four to twelve hours.
  • the period of infusing occurs over about 5 to about 60 minutes, more preferably over 30 minutes and still more preferably over 5 minutes.
  • the infusing is done every six hours, or in another aspect every eight hours.
  • the present invention provides a method of preparing a carbamazepine-cyclodextrin inclusion complex by admixing a modified cyclodextrin and carbamazepine in a physiologically acceptable fluid to form a carbamazepine-cyclodextrin inclusion complex.
  • the method further includes the step of sterilizing the carbamazepine-cyclodextrin inclusion complex.
  • the physiologically acceptable fluid is isotonic.
  • the modified cyclodextrin is a sulfoalkyl-cyclodextrin.
  • the modified cyclodextrin is more preferably sulfobutylether-7-beta-cyclodextrin.
  • FIG. 1 shows carbamazepine phase solubility as a function of cyclodextrin concentration at ambient laboratory temperature.
  • FIG. 1A represents the compiled solubility data.
  • FIG. 1B represents the averaged solubility data.
  • FIG. 2 shows the DSC/TGA overlay of Carbamazepine Orgamol batch #899954.
  • FIG. 3 shows the DSC/TGA overlay of Carbamazepine Spectrum batch #SA0491.
  • FIG. 4 shows the observed and predicted plasma concentration-time profiles following intravenous administration of 100 mg of carbamazepine using a 3-compartment PK model.
  • FIG. 5 shows the simulated plasma concentration-time profiles of carbamazepine following different infusion durations.
  • Model parameters are the typical values parameters from the three-compartment model.
  • FIG. 6 shows the effect of infusion duration on Cmax,ss following IV administration of carbamazepine.
  • the present invention is directed to a carbamazepine-cyclodextrin inclusion complex useful for the parenteral administration of carbamazepine.
  • parenteral refers to administration by injection, infusion or implantation. Injection and infusion include administration into a vein (intravenous), into an artery (intraarterial), into a muscle (intramuscular), under the skin (subcutaneous), and into the peritoneum (intraperitoneal). Intrapulmonary (administration within the lungs or its bronchi) and nasal (administration into the nose or by way of the nose) is also contemplated.
  • the carbamazepine-cyclodextrin inclusion complex is prepared by the admixture of a modified cyclodextrin and carbamazepine in a physiologically acceptable fluid.
  • Modified cyclodextrins include 2-hydroxypropyl-beta-cyclodextrin and sulfoalkyl cyclodextrins. More particularly, the sulfoalkyl cyclodextrins are those described and disclosed in U.S. Pat. Nos. 5,134,127 and 5,376,645.
  • a physiologically acceptable fluid includes sterile isotonic water, Ringer's lactate, D5W (5% dextrose in water), physiological saline, and similar fluids suitable for parenteral administration.
  • the admixture can be sterilized. Sterilization can be by methods well known to those of ordinary skill in the art, such as by autoclaving or by sterile filtration such as passage through a 0.22 micron filter.
  • the carbamazepine-cyclodextrin inclusion complex can be directly packaged into sterile ampoules, containers for fluids suitable for intravenous administration, or the complex can be lyophilized for prolonged storage according to techniques well known in the art.
  • the carbamazepine-cyclodextrin inclusion complex can be prepared so that the concentration of carbamazepine ranges from 1 mg/ml to 50 mg/ml, more preferably from 1 mg/ml to 10 mg/ml, and most preferably about 10 mg/ml. Variations in the carbamazepine concentration in the carbamazepine-cyclodextrin inclusion complex of the present invention is conventionally accomplished by varying the amount of carbamazepine used in the preparation of that inclusion complex, as described elsewhere herein.
  • the carbamazepine-cyclodextrin inclusion complex can be administered parenterally in a single dose of up to 1600 mg, or preferably up to 500 mg, more preferably divided doses from 20 to 500 mg, and most preferably divided doses from 75 to 400 mg. Dosing is dependent upon the indication of the patient being treated, as well as interactions with other drugs that the patient can be taking, and other clinical considerations well within the skill of the attending physician.
  • the carbamazepine-cyclodextrin inclusion complex of the present invention has a bioavailability of 100% and a half-life of about 24 hours. Resultant plasma concentrations after intravenous administration are reasonably predictable with every 1 mg/kg dose producing an increase in CBZ concentration of 0.75.+ ⁇ .0.2 mg/L. Given an average oral bioavailability of 65-75%, the initial IV replacement dose will be 65-75% of a patient's maintenance dose although some adjustment in subsequent IV doses may be necessary depending on a patient's actual oral CBZ bioavailability. This dosing regimen is selected in order to ensure that trough CBZ concentrations remain within the therapeutic range, while minimizing the risk of adverse events associated with elevated, end-of-infusion of CBZ concentrations.
  • the carbamazepine-cyclodextrin inclusion complex of the present invention preferably has certain pharmacokinetic parameters statistically similar to those of oral CBZ dosage forms.
  • the carbamazepine-cyclodextrin inclusion complex preferably has a minimum plasma concentration (Cmin) of about 70% to about 130% of the Cmin of an oral CBZ dosage form, and more preferably from about 80% to about 125% of the Cmin of an oral CBZ dosage form.
  • the carbamazepine-cyclodextrin inclusion complex has an area under the plasma concentration-time curve (AUC) of about 70% to about 130% of the AUC of an oral CBZ dosage form, and more preferably from about 80% to about 125% of the AUC of an oral CBZ dosage form.
  • AUC plasma concentration-time curve
  • the total daily intravenous (IV) dose can be administered as four equal doses every six hours, infused over up to 60 minutes, or preferably over 30 minutes, or more preferably over 15 minutes.
  • the infusion duration and dosing interval can be adjusted depending upon clinical considerations within the skill of the attending physician. For example, in a situation where rapid return to steady-state levels of CBZ is desired, the infusion duration can be as short as 2-5 minutes via IV push or IV bolus administration to a patient in need thereof.
  • the total daily IV dose can be administered as three equal doses every eight hours, infused over up to 60 minutes, or preferably over 30 minutes, or more preferably over 15 minutes.
  • administration can be continuous, or can be administered using a patient controlled device that permits controlled dosing on an as needed basis.
  • Other dosing schedules are well known in the art, and can be readily determined by pharmacists and physicians skilled in the art based upon considerations of, for example, age of the patient, indication, divided dose and total daily dosage.
  • the carbamazepine-cyclodextrin inclusion complexes of the present invention can be administered via rectal, oral or nasal routes for those patients who either cannot tolerate parenteral administration or who are so young that parenteral administration is not practical.
  • those patients who can receive the formulation of the present invention via an enteral route will obtain the benefits of substantially complete bioavailability over present sold oral dosage forms.
  • Enteral administration does not require a change in the formulation of the present invention, as those carbamazepine-cyclodextrin inclusion complexes can be directly delivered enterally.
  • Taste masking formulations well known in the art, can be used to modify formulations designed to be administered orally to eliminate any unpleasant taste. Taste masking is, however, related to patient compliance rather than related to efficacy of the present invention for enteral administration.
  • the carbamazepine-cyclodextrin inclusion complexes of the present invention can be administered to a mammal in need of CBZ treatment.
  • the word “mammal” is given its ordinary and customary meaning in the art, and includes human beings. Accordingly, the carbamazepine-cyclodextrin inclusion complexes of the present invention can be used in veterinary applications as well as the treatment of human conditions. With respect to human treatment, the carbamazepine-cyclodextrin inclusion complexes of the present invention is particularly well suited for pediatric administration, because the instant formulation does not require a peroral route of administration.
  • the carbamazepine-cyclodextrin inclusion complexes of the present invention can be used for any indication for which CBZ is used.
  • CBZ is indicated for seizure disorders such as partial seizures with complex symptoms (psychomotor, temporal lobe epilepsy), generalized tonic-clonic (grand mal) seizures, mixed seizure patterns or other partial or generalized seizures.
  • CBZ is also indicated for trigeminal neuralgia (tic douloureux) such as treatment of pain associated with true trigeminal neuralgia and bipolar disorders.
  • CBZ is also beneficial in glossopharyngeal neuralgia.
  • neurogenic diabetes insipidus include schizoaffective illness, depression, agitation, behavioral disturbances related to dementia, resistant schizophrenia, and dyscontrol syndrome associated with limbic system dysfunction; alcohol withdrawal; fibromyalgia; neuropathy; status epilipticus; and refractory seizure disorders.
  • certain psychiatric disorders including schizoaffective illness, depression, agitation, behavioral disturbances related to dementia, resistant schizophrenia, and dyscontrol syndrome associated with limbic system dysfunction
  • alcohol withdrawal fibromyalgia
  • neuropathy status epilipticus
  • refractory seizure disorders include treatment of neurogenic diabetes insipidus; certain psychiatric disorders, including schizoaffective illness, depression, agitation, behavioral disturbances related to dementia, resistant schizophrenia, and dyscontrol syndrome associated with limbic system dysfunction; alcohol withdrawal; fibromyalgia; neuropathy; status epilipticus; and refractory seizure disorders.
  • carbamazepine-cyclodextrin inclusion complexes of the present invention provide reduced toxicity and 100% bioavailability compared to other parenteral carbamazepine formulations such as a PEG400 formulation. Moreover, the complexes of the present invention are less nephrotoxic while providing similar solubilities and dissolution rates of carbamazepine-cyclodextrin complexes.
  • Indwelling catheters were placed into the arms of test subjects. A single 100 mg dose of stable-labeled (non-radioactive) CBZ (SL-CBZ) was then infused over 10 minutes. At the end of the infusion, the subject's usual morning dose of oral CBZ, less 100 mg, was administered. Blood pressure, heart rate and rhythm, and infusion site discomfort were monitored during and for an hour after the infusion. A single blood sample was collected prior to the infusion and 12 samples were collected over the ensuing 96 hours.
  • SL-CBZ stable-labeled (non-radioactive) CBZ
  • CBZ concentration-time data were analyzed using a non-compartmental approach with the pharmacokinetic software, WinNONLIN.
  • CBZ-d 10 C/D/N Isotopes, Quebec, Canada
  • CBZ-d 10 C/D/N Isotopes, Quebec, Canada
  • CBZ was assayed similar to that described by Osterloh and Bertilsson. (Osterloh J, Bertilsson L. The absence of isotopic effect during the elimination of deuterium labeled carbamazepine in the rat. (Life Sci. 1978; 23:83-7.)
  • To obtain a standard for CBZ-glucuronide it was isolated from the urine of patients on CBZ monotherapy using a procedure similar to that previously published.
  • Unbound drug was separated from the bound fraction by ultrafiltration.
  • the mobile phase consists of 50% 0.05 M ammonium acetate buffer, pH 4.7, 50% MeOH at a flow rate of 0.4 ml/min, on a reverse phase C-18 column.
  • SIM selected ion monitoring
  • signals at m/z 237 (CBZ), 239 ( 13 C 15 N 2 -CBZ), 253 (CBZ-epoxide), 255 13 C 15 N 2 —CBZ-epoxide) and 247 (CBZ-d 10 ) were measured with a PC-based Hewlett-Packard Chem-Station®. software.
  • the lower limit of detection is 0.05 micrograms/ml for CBZ.
  • Table 3 provides the pharmacokinetic parameters for 76 subjects.
  • the range of oral CBZ daily dose ranged from 100 mg to 2400 mg for subjects in this study.
  • the absolute bioavailability of oral CBZ during maintenance therapy centers around 70-75% with 30 of the 56 subjects having bioavailabilities below 70%.
  • the variability of bioavailability is also substantial with a range of 0.35 to 1.65.
  • the highly variable bioavailability could be indicative of delayed release of drug from the various immediate and extended release oral formulations of CBZ or from continued absorption from an extended release dosage form into a subsequent dosing interval.
  • the distribution volume at steady state (VSS) is 1.24 ⁇ 0.439 L/kg. This is a previously unknown value (due to the lack of an IV formulation) the now permits precise dosing of an IV formulation to attain a targeted plasma CBZ concentrations as might be required when initiating therapy in patients whose oral therapy has been interrupted for 12 or more hours.
  • the present study utilized the 2-hydroxypropyl- ⁇ -cyclodextrin formulation for a drug solubilizing agent of SL-CBZ.
  • the pharmacokinetic data obtained from this study while not specifically designed to determine a full replacement IV dosing regimen, can assist in establishing the target dose for this study.
  • the results from the present study demonstrate a wide range of CBZ bioavailability values among subjects, several of which had calculated bioavailability values greater than 100%.
  • the justification for using the median bioavailability value for correcting the dose for IV administration of CBZ in the current study assumes that the true bioavailability in subjects should not be greater than 100% especially at steady-state dosing levels.
  • the value of 70% is between the calculated mean F value of 75% and the true rank order median value of 67%, and allows for an appropriate, calculable IV dose adjustment.
  • the typical subject should then receive a daily IV dose that is 70% of that individual's daily oral dose. This can produce CBZ plasma concentrations from the IV dose that are comparable to those concentrations resulting from oral administration in the majority of subjects.
  • the results of the present study provide initial safety and tolerability of a carbamazepine-cyclodextrin combination in subjects along with providing data that can be used to calculate the appropriate dose and dosing interval for replacement IV therapy.
  • the percent weight/volume cyclodextrin solutions were prepared by adding the appropriate amount of cyclodextrin to a flask and filling to the desired total volume (10 mL) with water, as shown in the following Table 4.
  • Cyclodextrin 0.0 0.5 1.0 1.5 2.0 2.5 3.0 3.5 4.0 4.5 5.0 6.0 7.0 8.0 9.0 Cyclodextrin 0 5 10 15 20 25 30 35 40 45 50 60 70 80 90 in Water % (w/v)
  • the flasks were graduated cylinders.
  • the cyclodextrin was first dissolved in a small volume of water, quantitatively transferred to a volumetric flask which was then brought to volume with water.
  • the solubility of CBZ was determined at ambient laboratory temperatures in various concentrations of aqueous Captisol and aqueous Cavitron (brand names of modified cyclodextrins useful in the present invention).
  • the drug substance was added to a microcentrifuge tube and the appropriate vehicle was added. Periodically, the samples were centrifuged and then an aliquot was removed from the supernatant, diluted as necessary and assayed by HPLC to determine the concentration.
  • the phase solubility was evaluated at least three times during each experiment to insure that the mixture achieved equilibrium. In general, the early concentration determination data (obtained approximately 2 hours after mixing the CBZ and the vehicle) were omitted because CBZ appeared to form supersaturated solutions initially before equilibration.
  • the solubilities reported in Table 5 are the average of two or three concentration determinations obtained during the respective time course.
  • FIG. 1A shows a graphical representation of the compiled solubility data. From these data, it appears that the CBZ solubility at most cyclodextrin concentrations is marginally improved in Cavitron, as compared to Captisol.
  • FIG. 1B shows the averaged solubility data with the associated standard deviations. The latter Figure also provides the trend lines for the averaged data.
  • phase solubility data were expressed in terms of molarity and the equations of the lines were:
  • Captisol:CBZ and Cavitron:CBZ were found to be 974 and 677 M ⁇ 1 , respectively. These relatively weak associations are within the range (100-20,000 M ⁇ 1 ) of those commonly seen with drug: cyclodextrin complexes (Crit. Rev. Ther. Drug Carrier Systems, 14 (1): 1-104, 1997). Stella et al. simulated drug release from cyclodextrins upon dilution (Advanced Drug Del. Rev.
  • the minimal volume of distribution of a drug administered intravenously is based on the plasma volume, which is approximately 5% of the body weight. Therefore, in a 70 kg subject, the plasma volume is approximately 3.5 L. Alternatively, one could assume that the volume of distribution is extracellular water, accounting for approximately 30% of the total body weight, in which case the volume of distribution is about 21 L.
  • Table 6 shows the theoretical dilutions that would result from a 25 mg/mL formulation administered at different dose volumes. These calculations do not assume that any endogenous compounds might displace CBZ, and so they can be considered to be very conservative estimations. If a 20 mL dose were administered, the dilution ranges from 175-1,050-fold.
  • phase solubility data from the first set of assays performed using the two cyclodextrins appear to be higher than all of the subsequent phase solubility experiments (data not shown).
  • the cyclodextrin solutions were prepared by weighing the appropriate amount of cyclodextrin and adding it to a graduated cylinder and mixing to dissolve the solid.
  • the appropriate amount of cyclodextrin was added to a vial, dissolved, quantitatively transferred to a volumetric flask, filled to volume with water and mixed.
  • This second method of vehicle preparation is more accurate and apparently significantly influenced the resulting phase solubility data.
  • the most relevant factor in considering the dose adjustment for intravenously administered CBZ is for treatment centers to maintain plasma concentrations of CBZ above the therapeutic threshold. Following administration of 100 mg of IV CBZ, observed plasma concentrations followed a tri-exponential decay indicating a very fast distribution to tissues followed by a slower elimination of drug out of the body. Steady-state plasma concentrations of IV CBZ were predicted using the method of superposition. Plasma concentrations following a single dose of IV CBZ were scaled-up to steady-state conditions, assuming linear pharmacokinetics, using an accumulation ratio, a mean F of 0.7, and correcting for dose (see Equations 1 and 2). As a result, this calculation allows for a comparison of steady-state trough plasma concentrations following oral (C 0 and IV (C 6hr ) administration of CBZ respectively assuming a dosing interval of once every six hours for the IV formulation.
  • C pss is the plasma concentration (C) at steady state for IV administration
  • C pSD is the plasma concentration after a single dose
  • Int is the y-intercept resulting from linear regression of the elimination phase
  • is the terminal elimination rate constant
  • D po is the total oral daily dose
  • D IV is the single IV dose
  • F is the absolute bioavailability
  • T is the dosing interval
  • t is the time of each observed concentration.
  • the dosing frequency of every six hours following IV administration of CBZ outlined is appropriate to maintain plasma concentrations above this threshold and be comparable to trough levels following oral administration.
  • CBZ concentration-time profiles following IV administration will differ from oral administration to the greatest extent for subjects on ER products, since these products provide formulation dependent control of the concentration-time profile.
  • Subjects on extended release formulations experience the least fluctuation in concentrations and will maintain higher relative trough concentrations compared to any non-extended release product.
  • the analysis in this subset gives a conservative assessment of potential differences in peak and trough exposures following IV administration.
  • This analysis included calculations of predicted C max , C min , and AUC ss values of CBZ for subjects predicted to be at steady-state on IV therapy.
  • the same procedure for scaling plasma concentrations of CBZ as stated previously was implemented using Equations 1 and 2.
  • Linear pharmacokinetics were assumed and plasma concentrations following a single 100 mg dose of IV CBZ were scaled-up to steady-state conditions using an accumulation ratio (determined using each individual's terminal elimination rate constant), and assuming a mean F of 0.7 for computation of the IV dose administered.
  • the scaled-up steady-state CBZ concentrations were predicted based upon a 100 mg single IV dose infused over 10 minutes.
  • the mean steady-state IV C max , value was 11.75 ⁇ g/mL, a plasma level that is higher compared to the mean oral C max at steady-state (see Table 9).
  • F values mean steady-state IV C max
  • subjects taking high doses of oral CBZ and subjects at the extreme lower end of bioavailability may experience maximum plasma concentrations of CBZ that are in excess of the reported therapeutic range for this compound (see Table 9).
  • Subjects taking high doses of oral CBZ compounded with an inherent low bioavailability could be at the greatest risk for adverse effects due to elevated CBZ plasma levels.
  • the 70% dose adjustment would be the standard across all subjects administered replacement IV CBZ therapy to ensure the majority of subjects stay above the minimum (trough) therapeutic threshold and thus preventing seizures. If a subject's true bioavailability is less than the F value used for dosing, drug accumulation will occur when the inclusion complex is administered intravenously.
  • CBZ bioavailability is extremely variable among subjects and complicating factors such as formulations with different release rates, doses, and dosing intervals add to an already complex pharmacokinetic profile.
  • Systemic exposures after IV administration of CBZ will not vary amongst subjects to the extent that systemic exposures vary after oral administration, since formulation characteristics and bioavailability are excluded as sources of variability following IV administration.
  • Dose adjustments in subjects for IV CBZ replacement therapy must protect against low plasma concentration levels possibly leading to an increased risk in break through seizures.
  • the effects of increased transient CBZ exposures can occur in some subjects at the extreme low end of oral bioavailability or distribution volume.
  • the scaled IV CBZ concentration-time curves from the pharmacokinetic data of subjects dosed over 10 minutes reveal that the frequency distribution of C max values is unequal with a skewed tail at the extreme high end of C max values.
  • the mean population C max value was 11.75 ⁇ g/mL with a median value of 9.93 ⁇ g/mL.
  • the 95th percentile C max value was 24.14 ⁇ g/mL with a range of C max values from 3.17 ⁇ g/mL to 47.00 ⁇ g/mL (See Table 9, “Summary Statistics of Predicted Steady-State Parameters Following Administration of IV or Oral Carbamazepine”).
  • the mean, modeled C max,ss value after a 60 minute infusion was 10.68 ⁇ g/mL compared to 10.04 ⁇ g/mL after a 30 minute infusion compared to 11.69 ⁇ g/mL after a 15 minute infusion.
  • the peak concentration from the typical value simulation (150 mg IV dose) shown in FIG. 5 is comparable but differed slightly to that of the scaled, observed IV C max,ss reported in Table 9 (approximately 9.50 ⁇ g/mL and 11.75 ⁇ g/mL respectively).
  • the model dependent prediction ( FIG. 6 ) more accurately captures the time point of the true maximum concentration, whereas the scaled, observed value (Table 9) is dependent upon the time of collection.
  • the median (maximum) elapsed time between the end of infusion and the next time of collection was 5.3 (38.0) minutes. With t1/2 ( ⁇ ) of 2.2 minutes, considerable decay in concentration will occur during this time.
  • the actual median (maximum) time to observed C max,ss after the end of infusion was 5.8 (240) minutes. Additionally, all modeled infusions were precisely of 15 minutes duration. If actual infusion durations were greater than 15 minutes, C max,ss would be lower than predicted by the model.

Abstract

The present invention is directed to a carbamazepine-cyclodextrin inclusion complex useful for the parenteral administration of carbamazepine. The carbamazepine-cyclodextrin inclusion complex is prepared by the admixture of a modified cyclodextrin and carbamazepine in a physiologically acceptable fluid. Modified cyclodextrins include 2-hydroxypropyl-beta-cyclodextrin and sulfoalkyl cyclodextrins. More particularly, the sulfoalkyl cyclodextrins are those described and disclosed in U.S. Pat. Nos. 5,134,127 and 5,376,645. A physiologically acceptable fluid includes sterile isotonic water, Ringer's lactate, D5W (5% dextrose in water), physiological saline, and similar fluids suitable for parenteral administration.

Description

    RELATED APPLICATIONS
  • This application claims priority from U.S. provisional patent application 60/722,284 filed Sep. 30, 2005.
  • BACKGROUND OF THE INVENTION
  • Carbamazepine, or 5H-dibenz[b,f]azepine-5-carboxamide, is a widely used antiepileptic agent. It is available in the U.S. as Tegretol' brand chewable tablets of 100 mg, tablets of 200 mg and suspension of 100 mg/5 mL, intended for oral administration as a treatment for epilepsy or as a specific analgesic for trigeminal neuralgia. Other brand names include Equetro, Carbatrol, Tegretol XR and Epitol. Generic versions of these oral dosage forms are also available. Dosage forms include Carbatrol available in 100, 200, and 300 mg strengths; and Tegretol XR, available in 100, 200, and 400 mg strengths.
  • As shown in Table 1, recommended maintenance dosage levels in adults and children over 12 years of age are 800-1200 mg daily, although up to 2400 mg daily have been used in adults. In children of 6 to 12 years of age, the maintenance dosage level is usually 20-30 mg/kg/d and in children less than 6 years old the maintenance dosage level is usually 10-20 mg/kg/d.
  • TABLE 1
    Labeled dosage for carbamazepine oral dosage forms.
    Recommended Dosing Frequency
    Daily (for IR
    Age Maintenance Dose formulations)
    <6 yrs 10-20 mg/kg 2-4 doses/day
    6-12 yrs 20-30 mg/kg 2-4 doses/day
    max dose 1000 mg
    Children >12 yrs 400-1200 mg 2-4 doses/day
    1600-2400 mg
    Adults-epilepsy 800-1200 mg 3-4 divided doses
    some pts require
    1600-2400 mg
    Adults- 400-800 mg 2 doses/day
    trigeminal max dose 1200 mg
    neuralgia
    Adults-bipolar Doses greater 2 doses/day
    disorder than 1600 mg
    have not been
    studied
    Note:
    only Equetro, an extended release formulation is approved for bipolar disorder.
  • For complex partial seizures (temporal lobe, psychomotor), carbamazepine is a widely used anticonvulsant drug. It is also of proven efficacy in the treatment of generalized tonic-clonic (grand mal) seizures. Carbamazepine has also been used in treating simple partial (focal or Jacksonian) seizures and in patients with mixed seizure patterns which include the above, or other partial or generalized seizures. It is not used in the treatment of absence seizures (petit mal).
  • In addition to its proven effectiveness, carbamazepine has, in many respects, a more favorable profile in terms of the incidence and severity of side-effects than other anticonvulsants. Thus, carbamazepine is less sedating and causes less intellectual function impairment than other antiepileptic drugs such as phenobarbital, primidone and phenyloin. Furthermore, carbamazepine does not precipitate gingival hypertrophy, hirsutism, acne or other undesired effects associated with phenyloin. These attributes have helped to make carbamazepine the drug of choice in women and children.
  • Use of carbamazepine is complicated by incomplete, slow and variable absorption; extensive protein binding; and induction of its own metabolism. From Spina E Chapter 21 in Antiepileptic Drugs 5th edition. Lippincott, Williams & Wilkins, Philadelphia, 2002 pp 236-246 and references cited therein. The absolute bioavailability (the percentage of a dose that reaches the bloodstream) for the immediate release and extended release tablets has previously been estimated to range from 75-85 although the absence of an intravenous formulation has precluded systematic study of the extent and inter-patient variability in absorption.
  • U.S. Pat. No. 5,231,089 to Bodor mentions the lack of an injectable formulation for carbamazepine, noting that therefore there has not been precise information relating to the drug's absolute bioavailability. In addition, the lack of an injectable formulation for carbamazepine means that there is no method for providing emergent carbamazepine therapy to a patient in need thereof, as occurs when patients are undergoing surgery, have certain gastro-intestinal diseases, are unconscious or have seizures that preclude oral drug administration, or that require rapid re-establishment of steady state plasma levels.
  • The absence of an intravenous formulation places patients treated with carbamazepine (sometimes referred to herein as CBZ) at substantial medical risk. Sudden discontinuation of CBZ therapy for whatever reason, can expose an individual to potentially life threatening seizure emergencies. The only alternative is to give the patient a different drug that is available as an intravenous formulation. Exposure to a new medications exposes the patient to adverse reactions and unknown efficacy.
  • Cyclodextrins, sometimes referred to as Schardinger's dextrins, were first isolated by Villiers in 1891 as a digest of Bacillus amylobacter on potato starch. The foundations of cyclodextrin chemistry were laid down by Schardinger in the period 1903-1911. Until 1970, however, only small amounts of cyclodextrins could be produced in the laboratory and the high. production cost prevented the usage of cyclodextrins in industry. In recent years, dramatic improvements in cyclodextrin production and purification have been achieved and cyclodextrins have become much less expensive, thereby making the industrial application of cyclodextrins possible.
  • Cyclodextrins are cyclic oligosaccharides with hydroxyl groups on the outer surface and a void cavity in the center. Their outer surface is hydrophilic, and therefore they are usually soluble in water, but the cavity has a lipophilic character. The most common cyclodextrins are α-cyclodextrin, β-cyclodextrin and γ-cyclodextrin, consisting of 6, 7 and 8 α-1,4-linked glucose units, respectively. The number of these units determines the size of the cavity.
  • Cyclodextrins are capable of forming inclusion complexes with a wide variety of hydrophobic molecules by taking up a whole molecule (a “guest molecule”), or some part of it, into the void cavity. The stability of the resulting complex depends on how well the guest molecule fits into the cyclodextrin cavity. Common cyclodextrin derivatives are formed by alkylation (e.g., methyl-and-ethylβ-cyclodextrin) or hydroxyalkylation of the hydroxyethyl-derivatives of α-, β-, and γ-cyclodextrin) or by substituting the primary hydroxyl groups with saccharides (e.g., glucosyl- and maltosyl-β-cyclodextrin). Hydroxypropyl-β-cyclodextrin and its preparation by propylene oxide addition to β-cyclodextrin, and hydroxyethyl-β-cyclodextrin and its preparation by ethylene oxide addition to β-cyclodextrin, were described in a patent of Gramera et al. (U.S. Pat. No. 3,459,731, issued August 1969) over 35 years ago.
  • Although cyclodextrins have been used to increase the solubility, dissolution rate and/or stability of a great many compounds, it is also known there are many drugs for which cyclodextrin complexation either is not possible or yields no advantages. See J. Szejtli, Cyclodextrins in Drug Formulations: Part II, Pharmaceutical Technology, 24-38, August, 1991. Despite this potential pharmaceutical utility, certain cyclodextrins are have limitations.
  • Cyclodextrins and their derivatives are mostly crystalline solids. Concentration of some cyclodextrins in the renal tissue is followed by crystal formation causing necrotic damage to the cells. Despite forming water soluble clathrate complexes, the crystalline cyclodextrin drug complexes have generally been limited in their utility to sublingual or topical administration.
  • U.S. Pat. Nos. 5,134,127 and 5,376,645, whose disclosures are incorporated herein by reference, are directed to novel cyclodextrin derivatives, in particular sulfoalkyl cyclodextrin derivatives, that overcome the limitations of other cyclodextrins. In particular, the sulfoalkyl cyclodextrin derivatives disclosed therein exhibit lower nephrotoxicity while exhibiting high aqueous solubility.
  • The present invention is based, inter alia, on the determination that carbamazepine stable inclusion complexes with cyclodextrins are highly water soluble relative to the non-complexed drug. Surprisingly and unexpectedly, the carbamazepine-cyclodextrin inclusion complexes of the invention result in an injectable formulation that provides significant benefits and advantages over other carbamazepine formulations. For example, the carbamazepine-cyclodextrin inclusion complexes of the present invention are completely bioavailable, delivering 100% of the dose to the bloodstream in a consistent and predictable manner which is not the case with solid oral dosage forms. Also, unlike solid oral dosage forms, the carbamazepine-cyclodextrin inclusion complexes of the present invention can be administered to a patient suffering from a generalized tonic-clonic or other acute seizure via a peripheral rather than oral route. The carbamazepine-cyclodextrin inclusion complexes of the present invention satisfy a significant unmet medical need for a stable injectable formulation of carbamazepine that overcomes the limitations of poorly soluble and variably absorbed oral formulations.
  • SUMMARY OF THE INVENTION
  • In one aspect, the present invention contemplates a carbamazepine-cyclodextrin inclusion complex useful for the parenteral administration of carbamazepine comprising a carbamazepine complexed with a modified cyclodextrin. Preferably, the modified cyclodextrin is a sulfoalkyl-cyclodextrin. A preferred modified cyclodextrin is sulfobutylether-7-beta-cyclodextrin. The inclusion complex preferably has a concentration of about 5 to about 50 mg/ml carbamazepine, and more preferably a concentration of about 10 mg/ml carbamazepine.
  • In another aspect, the present invention, there is provided a carbamazepine-cyclodextrin inclusion complex useful for the parenteral administration of carbamazepine in which dosing is about 30% to about 100% of oral maintenance doses, or preferably about 65% to 75% of oral maintenance doses.
  • In a further aspect, the present invention provides a carbamazepine-cyclodextrin inclusion complex useful for the parenteral administration of carbamazepine having a half-life of about 8 to about 65 hours, and more preferably having a half-life of about 24 hours. In another embodiment, the present invention contemplates a carbamazepine-cyclodextrin inclusion complex useful for the parenteral administration of carbamazepine having an area under the plasma concentration-time curve (AUC) of about 70% to about 130% of the AUC for an oral carbamazepine dosage form, and more preferably having an AUC of about 80% to about 125% of the AUC for an oral carbamazepine dosage form. In a further embodiment, the present invention contemplates a carbamazepine-cyclodextrin inclusion complex useful for the parenteral administration of carbamazepine having a minimum plasma concentration (Cmin) of about. 70% to about 130% of the Cmin for an oral carbamazepine dosage form, and more preferably having a Cmin of about 80% to about 125% of the Cmin for an oral carbamazepine dosage form.
  • In a still further aspect, the present invention provides a carbamazepine-cyclodextrin inclusion complex useful for the parenteral administration of carbamazepine having an intravenous dosing interval of every four to twelve hours, more preferably having an intravenous dosing interval of every six hours, and still more preferably having an intravenous dosing interval of every eight hours.
  • In another embodiment the present invention provides a method of administering a carbamazepine-cyclodextrin inclusion complex useful for the parenteral administration of carbamazepine comprising: 1) providing a carbamazepine-cyclodextrin inclusion complex; and 2) infusing the complex intravenously to a patient in need thereof every four to twelve hours.
  • Preferably, the period of infusing occurs over about 5 to about 60 minutes, more preferably over 30 minutes and still more preferably over 5 minutes. Preferably, the infusing is done every six hours, or in another aspect every eight hours.
  • In a still further embodiment, the present invention provides a method of preparing a carbamazepine-cyclodextrin inclusion complex by admixing a modified cyclodextrin and carbamazepine in a physiologically acceptable fluid to form a carbamazepine-cyclodextrin inclusion complex. In another aspect, the method further includes the step of sterilizing the carbamazepine-cyclodextrin inclusion complex. Preferably, the physiologically acceptable fluid is isotonic. Preferably, the modified cyclodextrin is a sulfoalkyl-cyclodextrin. The modified cyclodextrin is more preferably sulfobutylether-7-beta-cyclodextrin.
  • BRIEF DESCRIPTION OF THE DRAWINGS
  • FIG. 1 shows carbamazepine phase solubility as a function of cyclodextrin concentration at ambient laboratory temperature. FIG. 1A represents the compiled solubility data. FIG. 1B represents the averaged solubility data.
  • FIG. 2 shows the DSC/TGA overlay of Carbamazepine Orgamol batch #899954.
  • FIG. 3 shows the DSC/TGA overlay of Carbamazepine Spectrum batch #SA0491.
  • FIG. 4 shows the observed and predicted plasma concentration-time profiles following intravenous administration of 100 mg of carbamazepine using a 3-compartment PK model.
  • FIG. 5 shows the simulated plasma concentration-time profiles of carbamazepine following different infusion durations. The mean IV dose=150 mg, the average adjusted IV dose assuming F=0.7. Model parameters are the typical values parameters from the three-compartment model.
  • FIG. 6 shows the effect of infusion duration on Cmax,ss following IV administration of carbamazepine.
  • DETAILED DESCRIPTION OF THE INVENTION
  • The present invention is directed to a carbamazepine-cyclodextrin inclusion complex useful for the parenteral administration of carbamazepine.
  • As used herein, the term “parenteral” is given its ordinary and customary meaning in the field of pharmaceutical drug routes of administration. According to the Food and Drug Administration's Center for Drug Evaluation and Research Data Standards Manual (CDER Data Element Number C-DRG-00301; Data Element Name: Route of Administration) “parenteral” refers to administration by injection, infusion or implantation. Injection and infusion include administration into a vein (intravenous), into an artery (intraarterial), into a muscle (intramuscular), under the skin (subcutaneous), and into the peritoneum (intraperitoneal). Intrapulmonary (administration within the lungs or its bronchi) and nasal (administration into the nose or by way of the nose) is also contemplated. Any appropriate route of administration set forth in the above-referenced Food and Drug Administration document is specifically included within the scope of the instant invention, and nothing herein shall be construed to limit in any way those routes of administration that would be useful in connection with the carbamazepine-cyclodextrin inclusion complex of the present invention.
  • In one embodiment, the carbamazepine-cyclodextrin inclusion complex is prepared by the admixture of a modified cyclodextrin and carbamazepine in a physiologically acceptable fluid. Modified cyclodextrins include 2-hydroxypropyl-beta-cyclodextrin and sulfoalkyl cyclodextrins. More particularly, the sulfoalkyl cyclodextrins are those described and disclosed in U.S. Pat. Nos. 5,134,127 and 5,376,645. A physiologically acceptable fluid includes sterile isotonic water, Ringer's lactate, D5W (5% dextrose in water), physiological saline, and similar fluids suitable for parenteral administration.
  • After an admixture of the modified cyclodextrin and carbamazepine is prepared, the admixture can be sterilized. Sterilization can be by methods well known to those of ordinary skill in the art, such as by autoclaving or by sterile filtration such as passage through a 0.22 micron filter. After-sterilization, the carbamazepine-cyclodextrin inclusion complex can be directly packaged into sterile ampoules, containers for fluids suitable for intravenous administration, or the complex can be lyophilized for prolonged storage according to techniques well known in the art.
  • The carbamazepine-cyclodextrin inclusion complex can be prepared so that the concentration of carbamazepine ranges from 1 mg/ml to 50 mg/ml, more preferably from 1 mg/ml to 10 mg/ml, and most preferably about 10 mg/ml. Variations in the carbamazepine concentration in the carbamazepine-cyclodextrin inclusion complex of the present invention is conventionally accomplished by varying the amount of carbamazepine used in the preparation of that inclusion complex, as described elsewhere herein.
  • The carbamazepine-cyclodextrin inclusion complex can be administered parenterally in a single dose of up to 1600 mg, or preferably up to 500 mg, more preferably divided doses from 20 to 500 mg, and most preferably divided doses from 75 to 400 mg. Dosing is dependent upon the indication of the patient being treated, as well as interactions with other drugs that the patient can be taking, and other clinical considerations well within the skill of the attending physician.
  • The carbamazepine-cyclodextrin inclusion complex of the present invention has a bioavailability of 100% and a half-life of about 24 hours. Resultant plasma concentrations after intravenous administration are reasonably predictable with every 1 mg/kg dose producing an increase in CBZ concentration of 0.75.+−.0.2 mg/L. Given an average oral bioavailability of 65-75%, the initial IV replacement dose will be 65-75% of a patient's maintenance dose although some adjustment in subsequent IV doses may be necessary depending on a patient's actual oral CBZ bioavailability. This dosing regimen is selected in order to ensure that trough CBZ concentrations remain within the therapeutic range, while minimizing the risk of adverse events associated with elevated, end-of-infusion of CBZ concentrations.
  • The carbamazepine-cyclodextrin inclusion complex of the present invention preferably has certain pharmacokinetic parameters statistically similar to those of oral CBZ dosage forms. For example, the carbamazepine-cyclodextrin inclusion complex preferably has a minimum plasma concentration (Cmin) of about 70% to about 130% of the Cmin of an oral CBZ dosage form, and more preferably from about 80% to about 125% of the Cmin of an oral CBZ dosage form. Similarly, the carbamazepine-cyclodextrin inclusion complex has an area under the plasma concentration-time curve (AUC) of about 70% to about 130% of the AUC of an oral CBZ dosage form, and more preferably from about 80% to about 125% of the AUC of an oral CBZ dosage form. Workers of ordinary skill in the art of pharmaceutical formulation are well acquainted with these concepts, which are further explained in the Food and Drug Administrations' Guidance for Industry entitled “Statistical Approaches to Establishing Bioequivalence” of January 2001 (see the world wide web fda.gov/cder/guidance/3616fnl.htm.)
  • The total daily intravenous (IV) dose can be administered as four equal doses every six hours, infused over up to 60 minutes, or preferably over 30 minutes, or more preferably over 15 minutes. As is well known in the art, the infusion duration and dosing interval can be adjusted depending upon clinical considerations within the skill of the attending physician. For example, in a situation where rapid return to steady-state levels of CBZ is desired, the infusion duration can be as short as 2-5 minutes via IV push or IV bolus administration to a patient in need thereof. In other embodiments, the total daily IV dose can be administered as three equal doses every eight hours, infused over up to 60 minutes, or preferably over 30 minutes, or more preferably over 15 minutes. In further embodiments, administration can be continuous, or can be administered using a patient controlled device that permits controlled dosing on an as needed basis. Other dosing schedules are well known in the art, and can be readily determined by pharmacists and physicians skilled in the art based upon considerations of, for example, age of the patient, indication, divided dose and total daily dosage.
  • In other embodiments, the carbamazepine-cyclodextrin inclusion complexes of the present invention can be administered via rectal, oral or nasal routes for those patients who either cannot tolerate parenteral administration or who are so young that parenteral administration is not practical. In addition, those patients who can receive the formulation of the present invention via an enteral route will obtain the benefits of substantially complete bioavailability over present sold oral dosage forms. Enteral administration does not require a change in the formulation of the present invention, as those carbamazepine-cyclodextrin inclusion complexes can be directly delivered enterally. Taste masking formulations, well known in the art, can be used to modify formulations designed to be administered orally to eliminate any unpleasant taste. Taste masking is, however, related to patient compliance rather than related to efficacy of the present invention for enteral administration.
  • The carbamazepine-cyclodextrin inclusion complexes of the present invention can be administered to a mammal in need of CBZ treatment. The word “mammal” is given its ordinary and customary meaning in the art, and includes human beings. Accordingly, the carbamazepine-cyclodextrin inclusion complexes of the present invention can be used in veterinary applications as well as the treatment of human conditions. With respect to human treatment, the carbamazepine-cyclodextrin inclusion complexes of the present invention is particularly well suited for pediatric administration, because the instant formulation does not require a peroral route of administration.
  • The carbamazepine-cyclodextrin inclusion complexes of the present invention can be used for any indication for which CBZ is used. For example, CBZ is indicated for seizure disorders such as partial seizures with complex symptoms (psychomotor, temporal lobe epilepsy), generalized tonic-clonic (grand mal) seizures, mixed seizure patterns or other partial or generalized seizures. CBZ is also indicated for trigeminal neuralgia (tic douloureux) such as treatment of pain associated with true trigeminal neuralgia and bipolar disorders. CBZ is also beneficial in glossopharyngeal neuralgia. Other uses include treatment of neurogenic diabetes insipidus; certain psychiatric disorders, including schizoaffective illness, depression, agitation, behavioral disturbances related to dementia, resistant schizophrenia, and dyscontrol syndrome associated with limbic system dysfunction; alcohol withdrawal; fibromyalgia; neuropathy; status epilipticus; and refractory seizure disorders.
  • While the carbamazepine-cyclodextrin inclusion complexes of the present invention provide reduced toxicity and 100% bioavailability compared to other parenteral carbamazepine formulations such as a PEG400 formulation. Moreover, the complexes of the present invention are less nephrotoxic while providing similar solubilities and dissolution rates of carbamazepine-cyclodextrin complexes.
  • Further details of the preferred embodiments of the present invention are illustrated in the following examples, which are understood to be non-limiting.
  • EXAMPLES Example 1 Preparation of Carbamazepine-Cyclodextrin Inclusion Complex
  • 450 Grams of hydroxypropyl-beta-cyclodextrin (HPBCD) was dissolved in 2.0 L of deionized water to generate a 22.50 w/v solution. 13C, 15N-labeled carbamazepine (CBZ) [purchased from Cambridge Isotope Laboratories (CIL), 50 Frontage Road, Andover, Mass. 01810], 20 grams, was added to this solution. The resulting admixture was stirred for 24 hours at room temperature (20-25° C.). After 24 hours, the solution was sterile filtered through a sterile 0.22 micron Durapore filter into a sterile receiver. Previously sterilized ampoules were then filled and sealed under a nitrogen flush. The filled ampoules were stored at 2-8° C. The resulting inclusion complex had a CBZ concentration of approximately 10 mg/ml.
  • Example 2 Stability Testing
  • Ampoules containing 10.1 mg/ml carbamazepine-cyclodextrin inclusion complex were placed on room temperature stability studies and sampled every six months. CBZ was detected by HPLC using UV detection at 215 nm. Results are presented in Table 2.
  • TABLE 2
    Stability of Intravenous, Stable-labeled Carbamazepine Solution
    Initial Degradation
    Testing Concentration % Product- CBZ:
    Date in Vial Recovery iminostilbine
    May 31, 2005 10.1 mg/ml 104.65% not detected
    Nov. 10, 2004 10.1 mg/ml 97.07% not detected
    May 2, 2004 10.1 mg/ml 96.67% not detected
  • Example 3 Pharmacokinetics of Intravenous and Oral Carbamazepine in Patients on Maintenance Therapy
  • Indwelling catheters were placed into the arms of test subjects. A single 100 mg dose of stable-labeled (non-radioactive) CBZ (SL-CBZ) was then infused over 10 minutes. At the end of the infusion, the subject's usual morning dose of oral CBZ, less 100 mg, was administered. Blood pressure, heart rate and rhythm, and infusion site discomfort were monitored during and for an hour after the infusion. A single blood sample was collected prior to the infusion and 12 samples were collected over the ensuing 96 hours. Plasma was separated from blood and analyzed, using a LC-MS assay, for CBZ and CBZ-epoxide, an active metabolite, and glucuronidated metabolite that is inactive Unbound CBZ was measured following ultrafiltration. CBZ concentration-time data were analyzed using a non-compartmental approach with the pharmacokinetic software, WinNONLIN.
  • A validated LC-MS assay for SL-CBZ, CBZ and their respective epoxide metabolites was used. Carbamazepine-d.sub.10 (CBZ-d10, C/D/N Isotopes, Quebec, Canada) was used as the internal standard. CBZ was assayed similar to that described by Osterloh and Bertilsson. (Osterloh J, Bertilsson L. The absence of isotopic effect during the elimination of deuterium labeled carbamazepine in the rat. (Life Sci. 1978; 23:83-7.) To obtain a standard for CBZ-glucuronide, it was isolated from the urine of patients on CBZ monotherapy using a procedure similar to that previously published. (Sinz M W, Remmel R P. Analysis of lamotrigine and lamotrigine 2-N-glucuronide in guinea pig blood and urine by reserved-phase ion-pairing liquid chromatography. J Chromatogr. 1991; 571:217-30) A 0.5 ml aliquot of patient plasma and 10 microliters of internal standard was added to blank plasma and extracted with 3 volumes of ethyl acetate. After shaking and centrifugation, the organic layer was removed and evaporated under nitrogen gas to dryness. Each sample was then redissolved with the addition of 25 microliters of ethyl acetate. Plasma samples were measured for unbound and total CBZ, CBZ glucuronide and CBZ-E by LCMS. Unbound drug was separated from the bound fraction by ultrafiltration. The mobile phase consists of 50% 0.05 M ammonium acetate buffer, pH 4.7, 50% MeOH at a flow rate of 0.4 ml/min, on a reverse phase C-18 column. For selected ion monitoring (SIM), signals at m/z 237 (CBZ), 239 (13C15N2-CBZ), 253 (CBZ-epoxide), 255 13C15N2—CBZ-epoxide) and 247 (CBZ-d10) were measured with a PC-based Hewlett-Packard Chem-Station®. software. The lower limit of detection is 0.05 micrograms/ml for CBZ. LC-MS method has been validated for the determination of [13C,15N]-carbamazepine, carbamazepine, and their 10,11-epoxide metabolites in human plasma. Over a concentration range of 1.5 to 12 micrograms/ml the percent coefficient of variance was ≦5%.
  • Table 3 provides the pharmacokinetic parameters for 76 subjects. The range of oral CBZ daily dose ranged from 100 mg to 2400 mg for subjects in this study. As can be seen, the absolute bioavailability of oral CBZ during maintenance therapy centers around 70-75% with 30 of the 56 subjects having bioavailabilities below 70%. The variability of bioavailability is also substantial with a range of 0.35 to 1.65. The highly variable bioavailability could be indicative of delayed release of drug from the various immediate and extended release oral formulations of CBZ or from continued absorption from an extended release dosage form into a subsequent dosing interval. The distribution volume at steady state (VSS) is 1.24±0.439 L/kg. This is a previously unknown value (due to the lack of an IV formulation) the now permits precise dosing of an IV formulation to attain a targeted plasma CBZ concentrations as might be required when initiating therapy in patients whose oral therapy has been interrupted for 12 or more hours.
  • Another clinically important observation is the prolonged CBZ elimination half-life under steady-state conditions. The mean value was determined to be 25.8 hours (range from 8.79 to 64.6 hours) in contrast with the reported range of 12 to 17 hours in the package insert for Tegretol° and Carbatrol°. The most likely explanation for this difference is the use of an SL-IV CBZ solution in the present study that permitted rigorous characterization of elimination for 2-3 half-lives while subjects continued to take their oral CBZ doses as prescribed. The extended half-lives observed in the adult subjects will limit the fluctuation of CBZ concentrations following IV administration every six hours, further reducing the risk of sub-therapeutic CBZ concentrations.
  • TABLE 3
    Carbamazepine Summary Pharmacokinetics
    AUCss AUC
    Body (po) 0-°o
    Age Weight (μg · (μg · VSS CL CLSS/F
    (yr) (kg) F (hr) hr/mL) hr/mL) (L/kg) (L/hr/kg) (L/hr/kg)
    N 76 76 70 76 70 76 76 76 70
    Mean 46.9 81.7 0.742 25.8 89.5 38.2 1.24 0.040 0.045
    (SD) (15.7) (17.7) (0.29) (11.2) (29.3) (15.3) (0.439) (0.015) (0.024)
    Min 19.0 48.0 0.348 8.79 28.5 14.9 0.612 0.013 0.0087
    Median 45.0 82.0 0.670 24.6 87.2 36.5 1.13 0.036 0.041
    Max 87.0 151 1.65 64.6 168 94.5 3.17 0.089 0.122
    Note:
    The values of F, AUCss (po), and CLss/F included only 70 subjects due to 6 subjects only receiving the morning IV dose of CBZ. The range of oral CBZ daily dose ranged from 100 mg to 2400 mg.
  • The present study utilized the 2-hydroxypropyl-β-cyclodextrin formulation for a drug solubilizing agent of SL-CBZ. The pharmacokinetic data obtained from this study, while not specifically designed to determine a full replacement IV dosing regimen, can assist in establishing the target dose for this study. The results from the present study demonstrate a wide range of CBZ bioavailability values among subjects, several of which had calculated bioavailability values greater than 100%. As a result, a bioavailability value of 70% has been chosen for use when dosing subjects in this sequential study. This value is similar to the median F value (67%) determined from subjects administered IV CBZ in the present study (n=76). The justification for using the median bioavailability value for correcting the dose for IV administration of CBZ in the current study assumes that the true bioavailability in subjects should not be greater than 100% especially at steady-state dosing levels. The value of 70% is between the calculated mean F value of 75% and the true rank order median value of 67%, and allows for an appropriate, calculable IV dose adjustment. The typical subject should then receive a daily IV dose that is 70% of that individual's daily oral dose. This can produce CBZ plasma concentrations from the IV dose that are comparable to those concentrations resulting from oral administration in the majority of subjects. Thus, the results of the present study provide initial safety and tolerability of a carbamazepine-cyclodextrin combination in subjects along with providing data that can be used to calculate the appropriate dose and dosing interval for replacement IV therapy.
  • Example 4 Preparation of CBZ Standard Solution Equipment
  • Shimadzu HPLC systems with autosampler, pumps, degassers, UV detector, column oven, system controllers, and Shimadzu Class VP system Software
  • Cahn Microbalance
  • Shimadzu AY-120 Analytical balance
    pH meter, Orion pH/ISE Model # 420 A
  • Labnet VX100 Vortex Eppendorf Centrifuge 5415 D Fisher Scientific FS30 Sonicator LabQuake Shaker Materials
  • Material Manufacturer Lot No.
    Carbamazepine Spectrum SA0491
    Carbamazepine Orgamol 899954
    Cavitron Cargill H3M134P
    82004
    Captisol Cydex CY03A020535
    Acetonitrile Fisher 031168
    Purified
    water

    Preparation of CBZ Standard Solution (0.05 mg/L)
  • Weigh out 5 mg of CBZ and place in a 100 ml volumetric flask. Fill to volume with 60% aqueous acetonitrile.
  • Preparation of Cyclodextrin Vehicles
  • The percent weight/volume cyclodextrin solutions were prepared by adding the appropriate amount of cyclodextrin to a flask and filling to the desired total volume (10 mL) with water, as shown in the following Table 4.
  • TABLE 4
    Amount of Cyclodextrin (g)
    0.0 0.5 1.0 1.5 2.0 2.5 3.0 3.5 4.0 4.5 5.0 6.0 7.0 8.0 9.0
    Cyclodextrin 0 5 10 15 20 25 30 35 40 45 50 60 70 80 90
    in Water % (w/v)

    In initial assays, the flasks were graduated cylinders. In subsequent assays, the cyclodextrin was first dissolved in a small volume of water, quantitatively transferred to a volumetric flask which was then brought to volume with water.
  • Preparation of Samples for Phase Solubility
  • An excess of carbamazepine was added to each eppendorf tube. Appropriate vehicles were added to each tube and the final volumes were 1 mL.
  • Example 5 Phase Solubility
  • The solubility of CBZ was determined at ambient laboratory temperatures in various concentrations of aqueous Captisol and aqueous Cavitron (brand names of modified cyclodextrins useful in the present invention). The drug substance was added to a microcentrifuge tube and the appropriate vehicle was added. Periodically, the samples were centrifuged and then an aliquot was removed from the supernatant, diluted as necessary and assayed by HPLC to determine the concentration. The phase solubility was evaluated at least three times during each experiment to insure that the mixture achieved equilibrium. In general, the early concentration determination data (obtained approximately 2 hours after mixing the CBZ and the vehicle) were omitted because CBZ appeared to form supersaturated solutions initially before equilibration. The solubilities reported in Table 5 are the average of two or three concentration determinations obtained during the respective time course.
  • TABLE 5
    Phase solubility data for CBZ in the presence of varying cyclodextrin concentrations
    Cyclodextrin
    Captisol Cavitron
    CBZ source
    Spectrum Orgamol Orgamol Spectrum Orgamol Orgamol
    Cyclodextrin Measured CBZ conc % Measured CBZ conc %
    conc (% w/v) (mg/mL) Ave S.D. R.S.D. (mg/mL) Ave S.D. R.S.D.
    0 0.2 0.2 0.2 0.2 0.0 0.0 0.3 0.2 0.2 0.2 0.0 13.3
    5 2.4 2.2 2.4 2.3 0.1 4.9 3.8 2.7 3.0 3.2 0.6 18.0
    10 6.2 4.1 4.6 5.0 1.1 22.1 4.1 5.4 5.6 5.0 0.8 16.2
    15 6.9 6.4 7.7 7.0 0.7 9.4 12.3 8.2 8.4 9.6 2.3 24.0
    20 9.4 9.2 10.3 9.6 0.6 6.1 16.4 10.9 11.1 12.8 3.1 24.4
    25 11.8 11.4 12.3 11.8 0.5 3.8 14.8 14.0 14.3 14.4 0.4 2.8
    30 14.6 13.5 15.2 14.4 0.9 6.0 17.9 15.9 16.2 16.7 1.1 6.5
    35 16.3 15.9 18.4 16.9 1.3 8.0 20.7 18.1 19.9 19.6 1.3 6.8
    40 19.3 18.3 20.8 19.5 1.3 6.5 23.2 22.5 22.0 22.6 0.6 2.7
    45 21.8 20.6 22.5 21.6 1.0 4.4 27.3 25.5 25.7 26.2 1.0 3.8
    50 26.6 22.5 24.8 24.6 2.1 8.3 31.5 27.8 28.0 29.1 2.1 7.3

    FIG. 1A shows a graphical representation of the compiled solubility data. From these data, it appears that the CBZ solubility at most cyclodextrin concentrations is marginally improved in Cavitron, as compared to Captisol. FIG. 1B shows the averaged solubility data with the associated standard deviations. The latter Figure also provides the trend lines for the averaged data.
  • Binding Constant
  • Assuming a 1:1 complex forms, the binding constant K1:1 can be calculated, according to the relationship:

  • K 1:1=slope/[S 0(1−slope)]
  • where S.sub.0 is the intrinsic solubility. The phase solubility data were expressed in terms of molarity and the equations of the lines were:
  • Captisol: y=0.4379x+0.0008r2=0.9989
  • Cavitron: y=0.3515x+0.0008r2=0.9954
  • From these equations, the binding constants of Captisol:CBZ and Cavitron:CBZ were found to be 974 and 677 M−1, respectively. These relatively weak associations are within the range (100-20,000 M−1) of those commonly seen with drug: cyclodextrin complexes (Crit. Rev. Ther. Drug Carrier Systems, 14 (1): 1-104, 1997). Stella et al. simulated drug release from cyclodextrins upon dilution (Advanced Drug Del. Rev. 36, 3-16, 1999) and suggested that if complexes are diluted 100-fold in the absence of any endogenous competing agent, approximately 30% of the drug will remain complexed, and if they are diluted 1000-fold, approximately 5% of the drug will remain complexed.
  • The minimal volume of distribution of a drug administered intravenously is based on the plasma volume, which is approximately 5% of the body weight. Therefore, in a 70 kg subject, the plasma volume is approximately 3.5 L. Alternatively, one could assume that the volume of distribution is extracellular water, accounting for approximately 30% of the total body weight, in which case the volume of distribution is about 21 L.
  • Table 6 shows the theoretical dilutions that would result from a 25 mg/mL formulation administered at different dose volumes. These calculations do not assume that any endogenous compounds might displace CBZ, and so they can be considered to be very conservative estimations. If a 20 mL dose were administered, the dilution ranges from 175-1,050-fold.
  • Based on the simulations of dilution effects, it appears that 70-95% of the CBZ will immediately dissociate from the inclusion complex in the blood if one assumes no interaction from endogenous agents.
  • TABLE 6
    Theoretical extent of dilution for a 25 mg/mL CBZ formulation
    CBZ dose, mg 200 500 800
    Total dose 8 20 32
    volume, mL
    Extent of Dilution
    Plasma volume, 438 175 109
    3,500 ml.
    Extracellular 2,625 1,050 656
    volume, 21,000
    mL
  • Method of Vehicle Preparation
  • The phase solubility data from the first set of assays performed using the two cyclodextrins appear to be higher than all of the subsequent phase solubility experiments (data not shown). In this first assay, the cyclodextrin solutions were prepared by weighing the appropriate amount of cyclodextrin and adding it to a graduated cylinder and mixing to dissolve the solid. In all subsequent assays, the appropriate amount of cyclodextrin was added to a vial, dissolved, quantitatively transferred to a volumetric flask, filled to volume with water and mixed. This second method of vehicle preparation is more accurate and apparently significantly influenced the resulting phase solubility data. However, it is very difficult to prepare the cyclodextrin solutions volumetrically.
  • Effect of Cooling Saturated Solutions at 2-8° C.
  • The saturated CBZ/cyclodextrin solutions were placed in a 2-8° C. refrigerator and the appearances were recorded at various times. At 24 hours, all of the solutions showed a precipitate.
  • Example 6 Thermal Analysis
  • CBZ is known to exist in at least four different polymorphic states (J. Pharm. Sci. 90, 1106-1114, 1990). At one point in this study, it was suspected that the variability in the solubility data might be due to polymorphic differences. Thermal analyses using differential scanning calorimetry (DSC) and thermogravimetric analysis (TGA) were performed on the Spectrum CBZ and the Orgamol CBZ to determine if there were different polymorphs present in each product. Table 7 summarizes the experimental conditions and the thermal data.
  • TABLE 7
    Thermal transitions obtained for CBZ from two manufacturers.
    Thermogravimetric analysis Differential scanning calorimetry
    Ramp
    10° C./min from Ramp 5° C./min
    25° C. to 400° C. From 25° C. to 300° C.
    Vendor Initial % Sample Onset All Onset AH
    Lot-Batch weight weight weight peak 1 Peak 1 Peak 1 peak 2 Peak 2 Peak 2
    # (mg) loss (mg) (° C.) (° C.) (l/g) (° C.) (° C.) (l/g)
    Orgamol 9.687 87.25 3.246 161.92 165.25 14.03 183.22 185.70 117.2
    899954
    Spectrum 5.351 100.1 4.510 157.7 164.38 10.62 189.53 190.25 127.3
    SA0491
  • Example 7 Appropriate Dosing Interval Determination
  • The most relevant factor in considering the dose adjustment for intravenously administered CBZ is for treatment centers to maintain plasma concentrations of CBZ above the therapeutic threshold. Following administration of 100 mg of IV CBZ, observed plasma concentrations followed a tri-exponential decay indicating a very fast distribution to tissues followed by a slower elimination of drug out of the body. Steady-state plasma concentrations of IV CBZ were predicted using the method of superposition. Plasma concentrations following a single dose of IV CBZ were scaled-up to steady-state conditions, assuming linear pharmacokinetics, using an accumulation ratio, a mean F of 0.7, and correcting for dose (see Equations 1 and 2). As a result, this calculation allows for a comparison of steady-state trough plasma concentrations following oral (C0 and IV (C6hr) administration of CBZ respectively assuming a dosing interval of once every six hours for the IV formulation.
  • F = AUC r , SS ( Oral ) AUC 0 - ( I V ) × Dose ( I V ) Dose ( Oral ) Equation 1 C pSS ( I V ) = C pSD ( t ) ( I V ) + Int exp - λ T exp - λ T 1 - exp - λ T · ( D po F ) D I V 4 Equation 2
  • Where Cpss(IV) is the plasma concentration (C) at steady state for IV administration; CpSD is the plasma concentration after a single dose; Int is the y-intercept resulting from linear regression of the elimination phase; λ is the terminal elimination rate constant; Dpo is the total oral daily dose; DIV is the single IV dose; F is the absolute bioavailability; T is the dosing interval; and t is the time of each observed concentration.
  • The observed mean, oral steady-state trough (C0) plasma concentration of CBZ following twice daily dosing was 8.98 μg/mL (n=62 evaluable subjects) (see Table 8, “Statistical Comparison of Trough Steady-State Carbamazepine Concentrations Following Oral BID Dosing or Q6 IV Dosing Based on Mean F=0.70”). The mean steady-state plasma concentration at the 6-hour time-point following IV administration of CBZ was predicted to be 8.04 μg/mL. Statistical comparison of these two trough values following oral and IV administration of CBZ was found to be not statistically different (α=0.10; p=0.1931). Thus, the dosing frequency of every six hours following IV administration of CBZ outlined is appropriate to maintain plasma concentrations above this threshold and be comparable to trough levels following oral administration. Analysis of the same parameters at the 12-hour time-point, post-dose, indicated that the trough values were significantly different suggesting that an IV dosing regimen of every 12 hours would not be feasible to ensure the plasma concentration of CBZ does not fall below the therapeutic range.
  • TABLE 8
    Statistical Comparison of Trough Steady-State Carbamazepine Concentrations
    Following Oral BID Dosing or Q6 IV Dosing Based on Mean F = 0.70
    Difference SE for the 90% CI for the
    Carbamazepine Between Difference Difference in p-
    Concentration N Mean the Means in the Means the Means value
    C0 62 8.98 0.09456 0.7158 (−0.254, 2.15) 0.1931
    (oral only)
    C6 hr (IV only) - 62 8.04
    Mean F)
  • Example 8 Anticipated CBZ Maximum and Minimum Concentration Values During Intravenous Administration: Subjects at the Extreme of Carbamazepine Bioavailability
  • A subset analysis of the subject data (n=47), including only those subjects taking extended release (ER) formulations of CBZ twice daily (as indicated by the product label), was performed to compare predicted maximum and minimum exposures of CBZ at steady-state administered via IV infusion to those observed following oral administration. CBZ concentration-time profiles following IV administration will differ from oral administration to the greatest extent for subjects on ER products, since these products provide formulation dependent control of the concentration-time profile. Subjects on extended release formulations experience the least fluctuation in concentrations and will maintain higher relative trough concentrations compared to any non-extended release product. Thus, the analysis in this subset gives a conservative assessment of potential differences in peak and trough exposures following IV administration.
  • This analysis included calculations of predicted Cmax, Cmin, and AUCss values of CBZ for subjects predicted to be at steady-state on IV therapy. In order to predict steady-state plasma concentrations of IV CBZ, the same procedure for scaling plasma concentrations of CBZ as stated previously was implemented using Equations 1 and 2. Linear pharmacokinetics were assumed and plasma concentrations following a single 100 mg dose of IV CBZ were scaled-up to steady-state conditions using an accumulation ratio (determined using each individual's terminal elimination rate constant), and assuming a mean F of 0.7 for computation of the IV dose administered.
  • Following scale-up of the plasma concentrations of CBZ to steady-state, summary statistics of pharmacokinetic parameters were reported to compare the range of CBZ Cmax, Cmin, and AUC values between the oral (observed) and IV formulations of CBZ assuming a bioavailability (F) of 0.7 (See Table 9, “Summary Statistics of Predicted Steady-State Parameters Following Administration of IV or Oral Carbamazepine”). The steady-state PK parameters for the oral and IV formulations were obtained from data within the 12 hour dosing interval following administration of the ER products (Carbatrol.®. and Tegretol XR.®.) or the 6 hour interval after dosing of the IV product, respectively.
  • TABLE 9
    Summary Statistics of Predicted Steady-State Parameters
    Following Administration of IV or Oral Carbamazepine
    Mean ± 5th 95th
    PK SD Minimum Percentile Median Percentile Maximum
    Parameter N (μg/mL) (μg/mL) (μg/mL) (μg/mL) (μg/mL) (μg/mL)
    IV CmaxSS 47 11.75 ± 7.90  3.17 4.80 9.93 24.14 47.0
    (mean
    F = 0.7)
    IV CminSS 47 8.63 ± 6.85 1.65 2.48 6.50 18.64 41.04
    (mean
    F = 0.7)
    IV AUCss 47 121.83 ± 90.93  26.88 43.10 93.69 257.16 551.11
    (mean
    F = 0.7)
    Oral 47 9.92 ± 2.99 2.89 4.98 9.81 14.60 17.10
    Cmaxss*
    Oral 47 9.32 ± 2.93 2.89 4.32 9.44 14.55 16.69
    Cminss
    Oral AUCss 47 96.79 ± 28.08 28.48 50.11 96.01 145.51 167.58
    *Oral Cmax, ss estimate is based on highest observed concentration, and may not be indicative of the subjects true Cmax, ss value due to sparse sampling; this study was not designed to assess the Cmax, ss of oral CBZ.
    *Oral Cmax, ss estimate is based on highest observed concentration, and may not be indicative of the subjects true Cmax, ss value due to sparse sampling; this study was not designed to assess the Cmax, ss of oral CBZ.
  • The scaled-up steady-state CBZ concentrations were predicted based upon a 100 mg single IV dose infused over 10 minutes. The mean steady-state IV Cmax, value was 11.75 μg/mL, a plasma level that is higher compared to the mean oral Cmax at steady-state (see Table 9). Given the wide range of individual subject bioavailability values (F values) (see Table 3), subjects taking high doses of oral CBZ and subjects at the extreme lower end of bioavailability may experience maximum plasma concentrations of CBZ that are in excess of the reported therapeutic range for this compound (see Table 9). Subjects taking high doses of oral CBZ compounded with an inherent low bioavailability could be at the greatest risk for adverse effects due to elevated CBZ plasma levels. The 70% dose adjustment would be the standard across all subjects administered replacement IV CBZ therapy to ensure the majority of subjects stay above the minimum (trough) therapeutic threshold and thus preventing seizures. If a subject's true bioavailability is less than the F value used for dosing, drug accumulation will occur when the inclusion complex is administered intravenously.
  • Example 9 Modeling and Simulation to Assess the Effect of Infusion Duration on Cmax,ss Following Administration of Intravenous Carbamazepine
  • CBZ bioavailability is extremely variable among subjects and complicating factors such as formulations with different release rates, doses, and dosing intervals add to an already complex pharmacokinetic profile. Systemic exposures after IV administration of CBZ will not vary amongst subjects to the extent that systemic exposures vary after oral administration, since formulation characteristics and bioavailability are excluded as sources of variability following IV administration. Dose adjustments in subjects for IV CBZ replacement therapy must protect against low plasma concentration levels possibly leading to an increased risk in break through seizures. Concurrently, the effects of increased transient CBZ exposures can occur in some subjects at the extreme low end of oral bioavailability or distribution volume.
  • The scaled IV CBZ concentration-time curves from the pharmacokinetic data of subjects dosed over 10 minutes reveal that the frequency distribution of Cmax values is unequal with a skewed tail at the extreme high end of Cmax values. At a mean bioavailability value for the population assigned to 70% (as is appropriate for the dosing in the present invention), the mean population Cmax value was 11.75 μg/mL with a median value of 9.93 μg/mL. The 95th percentile Cmax value was 24.14 μg/mL with a range of Cmax values from 3.17 μg/mL to 47.00 μg/mL (See Table 9, “Summary Statistics of Predicted Steady-State Parameters Following Administration of IV or Oral Carbamazepine”).
  • To assess the effect of infusion duration on Cmax,ss modeling and simulation was performed using the observed plasma concentrations of CBZ following IV administration of the single 100 mg dose. Based on various diagnostic plots assessing goodness-of-fit of the PK model, plasma concentrations of CBZ were best described by a three-compartment model, displaying a very rapid tissue distribution phase indicative of highly perfused tissues such as the liver, lung and brain, a second distribution phase indicative of deeper tissue penetration such as adipose tissue, and a more prolonged elimination phase (FIG. 4). The modeling results revealed that the mean t1/2 (α) and t1/2 (β) were rapid (approximately 2 minutes and approximately 65 minutes, respectively), indicating that elevated plasma levels of CBZ will be short-lived once an infusion is stopped. Population mean terminal t1/2 (γ) is approximately 28 hours, which is comparable to the value based on noncompartmental analysis (Table 3). Assuming that the pharmacokinetics are linear and stationary, using each subject's predicted parameters from the PK model, simulations were performed at steady-state to determine the effect of infusion duration on CBZ C.sub.max,ss (FIG. 5). The results showed a decrease in the mean Cmax,ss as the infusion duration increased (based upon a mean IV dose of 150 mg when dosed under steady state conditions). The mean, modeled Cmax,ss value after a 60 minute infusion was 10.68 μg/mL compared to 10.04 μg/mL after a 30 minute infusion compared to 11.69 μg/mL after a 15 minute infusion.
  • There was only an approximate 135 decrease in the mean Cmax,ss value when the infusion duration was increased from 15 minutes to 1 hour.
  • Notably, the peak concentration from the typical value simulation (150 mg IV dose) shown in FIG. 5 is comparable but differed slightly to that of the scaled, observed IV Cmax,ss reported in Table 9 (approximately 9.50 μg/mL and 11.75 μg/mL respectively). The model dependent prediction (FIG. 6) more accurately captures the time point of the true maximum concentration, whereas the scaled, observed value (Table 9) is dependent upon the time of collection. The median (maximum) elapsed time between the end of infusion and the next time of collection was 5.3 (38.0) minutes. With t1/2 (α) of 2.2 minutes, considerable decay in concentration will occur during this time. For the full population, the actual median (maximum) time to observed Cmax,ss after the end of infusion was 5.8 (240) minutes. Additionally, all modeled infusions were precisely of 15 minutes duration. If actual infusion durations were greater than 15 minutes, Cmax,ss would be lower than predicted by the model. Finally, the typical value prediction modeled in FIG. 5 used each individual's set of model parameters and a typical dose (150 mg, the average adjusted IV dose assuming F=0.70), where the scaled observed values are based on the range of individual doses along with applying the superposition method at each observed concentration. As shown in FIG. 6, the average Cmax,ss values are similar with a 30 minute and 60 minute infusion
  • The invention may be embodied in other specific forms without departing from the spirit or essential characteristics thereof. The present embodiments are therefore to be considered in all respects as illustrative and not restrictive, the scope of the invention being indicated by the appended claims rather than by the foregoing description, and all the changes which come within the meaning and range of equivalency of the claims are therefore intended to be embraced therein.

Claims (27)

1. A carbamazepine-cyclodextrin inclusion complex useful for the parenteral administration of carbamazepine comprising a carbamazepine complexed with a modified cyclodextrin.
2. The complex of claim 1 wherein said modified cyclodextrin is a sulfoalkyl-cyclodextrin.
3. The complex of claim 1 or 2 wherein said modified cyclodextrin is sulfobutylether-7-beta-cyclodextrin.
4. The complex of claim 1 having a concentration of about 5 to about 50 mg/ml carbamazepine.
5. The complex of claim 1 having a concentration of about 10 mg/ml carbamazepine.
6. A carbamazepine-cyclodextrin inclusion complex useful for the parenteral administration of carbamazepine in which dosing is about 30% to about 100% of oral maintenance doses.
7. The complex of claim 6 wherein said dosing is about 65% to 75% of oral maintenance doses.
8. A carbamazepine-cyclodextrin inclusion complex useful for the parenteral administration of carbamazepine having a half-life of about 8 to about 65 hours.
9. The complex of claim 8 having a half-life of about 24 hours.
10. A carbamazepine-cyclodextrin inclusion complex useful for the parenteral administration of carbamazepine having an area under the plasma concentration-time curve (AUC) of about 70% to about 130% of the AUC for an oral carbamazepine dosage form.
11. The complex of claim 10 having an AUC of about 80% to about 125% of the AUC for an oral carbamazepine dosage form.
12. A carbamazepine-cyclodextrin inclusion complex useful for the parenteral administration of carbamazepine having a minimum plasma concentration (Cmin) of about. 70% to about 130% of the Cmin for an oral carbamazepine dosage form.
13. The complex of claim 12 having a Cmin of about 80% to about 125% of the Cmin for an oral carbamazepine dosage form.
14. A carbamazepine-cyclodextrin inclusion complex useful for the parenteral administration of carbamazepine having an intravenous dosing interval of every four to twelve hours.
15. The complex of claim 14 having an intravenous dosing interval of every six hours.
16. The complex of claim 14 having an intravenous dosing interval of every eight hours.
17. A method of administering a carbamazepine-cyclodextrin inclusion complex useful for the parenteral administration of carbamazepine comprising: 1) providing a carbamazepine-cyclodextrin inclusion complex; and 2) infusing said complex intravenously to a patient in need thereof every four to twelve hours.
18. The method of claim 17 wherein the period of said infusing occurs over about 5 to about 60 minutes.
19. The method of claim 17 wherein the period of said infusing occurs over 30 minutes.
20. The method of claim 17 wherein the period of said infusing occurs over 5 minutes.
21. The method of claim 17 wherein said infusing is done every six hours.
22. The method of claim 17 wherein said infusing is done every eight hours.
23. A method of preparing a carbamazepine-cyclodextrin inclusion complex by admixing a modified cyclodextrin and carbamazepine in a physiologically acceptable fluid to form a carbamazepine-cyclodextrin inclusion complex.
24. The method of claim 23 further including the step of sterilizing said carbamazepine-cyclodextrin inclusion complex.
25. The method of claim 23 wherein said physiologically acceptable fluid is isotonic.
26. The method of claim 23 wherein said modified cyclodextrin is a sulfoalkyl-cyclodextrin.
27. The method of claim 23 wherein said modified cyclodextrin is sulfobutylether-7-betacyclodextrin.
US12/571,039 2005-09-30 2009-09-30 Novel parenteral carbamazepine formulation Abandoned US20100204178A1 (en)

Priority Applications (7)

Application Number Priority Date Filing Date Title
US12/571,039 US20100204178A1 (en) 2006-10-02 2009-09-30 Novel parenteral carbamazepine formulation
US13/547,866 US9629797B2 (en) 2005-09-30 2012-07-12 Parenteral carbamazepine formulation
US13/679,715 US20130072476A1 (en) 2005-09-30 2012-11-16 Novel parenteral carbamazepine formulation
US14/051,938 US9770407B2 (en) 2005-09-30 2013-10-11 Parenteral carbamazepine formulation
US15/657,470 US20180153797A1 (en) 2005-09-30 2017-07-24 Novel parenteral carbamazepine formulation
US16/939,390 US20210196623A1 (en) 2005-09-30 2020-07-27 Novel parenteral carbamazepine formulation
US17/501,287 US20220265547A1 (en) 2005-09-30 2021-10-14 Parenteral carbamazepine formulation

Applications Claiming Priority (4)

Application Number Priority Date Filing Date Title
PCT/US2006/038508 WO2007041524A2 (en) 2005-09-30 2006-10-02 Novel parenteral carbamazepine formulation
USPCT/US06/38508 2006-10-02
US11/542,520 US20070185054A1 (en) 2005-09-30 2006-10-02 Novel parenteral carbamazepine formulation
US12/571,039 US20100204178A1 (en) 2006-10-02 2009-09-30 Novel parenteral carbamazepine formulation

Related Parent Applications (1)

Application Number Title Priority Date Filing Date
US11/542,520 Continuation US20070185054A1 (en) 2005-09-30 2006-10-02 Novel parenteral carbamazepine formulation

Related Child Applications (2)

Application Number Title Priority Date Filing Date
US13/547,866 Continuation US9629797B2 (en) 2005-09-30 2012-07-12 Parenteral carbamazepine formulation
US14/051,938 Continuation US9770407B2 (en) 2005-09-30 2013-10-11 Parenteral carbamazepine formulation

Publications (1)

Publication Number Publication Date
US20100204178A1 true US20100204178A1 (en) 2010-08-12

Family

ID=42540930

Family Applications (6)

Application Number Title Priority Date Filing Date
US12/571,039 Abandoned US20100204178A1 (en) 2005-09-30 2009-09-30 Novel parenteral carbamazepine formulation
US13/547,866 Active 2028-11-10 US9629797B2 (en) 2005-09-30 2012-07-12 Parenteral carbamazepine formulation
US13/679,715 Abandoned US20130072476A1 (en) 2005-09-30 2012-11-16 Novel parenteral carbamazepine formulation
US14/051,938 Active 2028-11-20 US9770407B2 (en) 2005-09-30 2013-10-11 Parenteral carbamazepine formulation
US15/657,470 Abandoned US20180153797A1 (en) 2005-09-30 2017-07-24 Novel parenteral carbamazepine formulation
US16/939,390 Abandoned US20210196623A1 (en) 2005-09-30 2020-07-27 Novel parenteral carbamazepine formulation

Family Applications After (5)

Application Number Title Priority Date Filing Date
US13/547,866 Active 2028-11-10 US9629797B2 (en) 2005-09-30 2012-07-12 Parenteral carbamazepine formulation
US13/679,715 Abandoned US20130072476A1 (en) 2005-09-30 2012-11-16 Novel parenteral carbamazepine formulation
US14/051,938 Active 2028-11-20 US9770407B2 (en) 2005-09-30 2013-10-11 Parenteral carbamazepine formulation
US15/657,470 Abandoned US20180153797A1 (en) 2005-09-30 2017-07-24 Novel parenteral carbamazepine formulation
US16/939,390 Abandoned US20210196623A1 (en) 2005-09-30 2020-07-27 Novel parenteral carbamazepine formulation

Country Status (1)

Country Link
US (6) US20100204178A1 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3220917A4 (en) * 2014-11-18 2018-07-25 Pixarbio Corporation Compositions for treating acute, post-operative, or chronic pain and methods of using the same
WO2020157257A1 (en) * 2019-02-01 2020-08-06 H. Lundbeck A/S Injectable carbamazepine composition essentially free of 10-br-carbamazepine

Families Citing this family (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
PT3411047T (en) 2016-02-04 2021-07-06 Czap Res And Development Llc Controlled-release and stratified cyclodextrin inclusion complex vehicles
KR102605030B1 (en) * 2016-12-14 2023-11-23 에스케이바이오팜 주식회사 Parenteral liquid preparation containing carbamate compounds

Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3459731A (en) * 1966-12-16 1969-08-05 Corn Products Co Cyclodextrin polyethers and their production
US5134127A (en) * 1990-01-23 1992-07-28 University Of Kansas Derivatives of cyclodextrins exhibiting enhanced aqueous solubility and the use thereof
US5231089A (en) * 1991-12-02 1993-07-27 University Of Florida Method of improving oral bioavailability of carbamazepine
US5376645A (en) * 1990-01-23 1994-12-27 University Of Kansas Derivatives of cyclodextrins exhibiting enhanced aqueous solubility and the use thereof
US5646131A (en) * 1994-02-22 1997-07-08 The Arab Company For Drug Industries And Medical Applicances (Acdima) Method for solubilizing drugs using cyclodextrins and carboxylic acids
US6046177A (en) * 1997-05-05 2000-04-04 Cydex, Inc. Sulfoalkyl ether cyclodextrin based controlled release solid pharmaceutical formulations
US6218375B1 (en) * 1999-01-21 2001-04-17 Bristol-Myers Squibb Company Complex of ras-farnesyltransferase inhibitor and sulfobutylether-7-β-cyclodextrin or 2-hydroxypropyl-β-cyclodextrin and method
US6458770B1 (en) * 1997-10-09 2002-10-01 Novartis Ag Parenteral formulations comprising carbamazepine or its derivatives
US20040157796A1 (en) * 2002-11-27 2004-08-12 Yatin Gokarn Concentrated liquid valdecoxib composition

Family Cites Families (15)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6546131B1 (en) 1990-12-19 2003-04-08 Canon Kabushiki Kaisha Image processing method and apparatus for achieving tone reproduction suited to the image
FR2713934B1 (en) 1993-12-22 1996-01-12 Commissariat Energie Atomique Use of amino cyclodextrins for the aqueous solubilization of hydrophobic compounds, in particular of pharmaceutically active molecules.
UA57734C2 (en) 1996-05-07 2003-07-15 Пфайзер Інк. Arylheterocyclic inclusion complexes
GB9713149D0 (en) 1997-06-21 1997-08-27 Pfizer Ltd Pharmaceutical formulations
ES2257795T3 (en) 1997-07-01 2006-08-01 Pfizer Products Inc. PROCEDURE TO PRODUCE A CYCLODEXTRINE.
US6858584B2 (en) 2000-05-02 2005-02-22 Theravance, Inc. Pharmaceutical compositions containing a glycopeptide antibiotic and a cyclodextrin
EP1383445A4 (en) 2001-03-20 2005-04-13 Cydex Inc Formulations containing propofol and a sulfoalkyl ether cyclodextrin
WO2003033025A2 (en) 2001-10-18 2003-04-24 Decode Genetics Ehf Cyclodextrin complexes
KR20030041577A (en) 2001-11-20 2003-05-27 디디에스텍주식회사 Solid dispersions containing substituted cyclodextrin and insoluble drug and their preparations
US6869939B2 (en) 2002-05-04 2005-03-22 Cydex, Inc. Formulations containing amiodarone and sulfoalkyl ether cyclodextrin
DE60327225D1 (en) 2002-08-20 2009-05-28 Bristol Myers Squibb Co ARIPIPRAZOL COMPLEX FORMULATION AND METHOD
US20070020298A1 (en) 2003-12-31 2007-01-25 Pipkin James D Inhalant formulation containing sulfoalkyl ether gamma-cyclodextrin and corticosteroid
EP2444090B1 (en) 2005-09-30 2018-12-12 Lundbeck Pharmaceuticals LLC Novel parenteral carbamazepine formulation
US7629331B2 (en) 2005-10-26 2009-12-08 Cydex Pharmaceuticals, Inc. Sulfoalkyl ether cyclodextrin compositions and methods of preparation thereof
US7635773B2 (en) 2008-04-28 2009-12-22 Cydex Pharmaceuticals, Inc. Sulfoalkyl ether cyclodextrin compositions

Patent Citations (9)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US3459731A (en) * 1966-12-16 1969-08-05 Corn Products Co Cyclodextrin polyethers and their production
US5134127A (en) * 1990-01-23 1992-07-28 University Of Kansas Derivatives of cyclodextrins exhibiting enhanced aqueous solubility and the use thereof
US5376645A (en) * 1990-01-23 1994-12-27 University Of Kansas Derivatives of cyclodextrins exhibiting enhanced aqueous solubility and the use thereof
US5231089A (en) * 1991-12-02 1993-07-27 University Of Florida Method of improving oral bioavailability of carbamazepine
US5646131A (en) * 1994-02-22 1997-07-08 The Arab Company For Drug Industries And Medical Applicances (Acdima) Method for solubilizing drugs using cyclodextrins and carboxylic acids
US6046177A (en) * 1997-05-05 2000-04-04 Cydex, Inc. Sulfoalkyl ether cyclodextrin based controlled release solid pharmaceutical formulations
US6458770B1 (en) * 1997-10-09 2002-10-01 Novartis Ag Parenteral formulations comprising carbamazepine or its derivatives
US6218375B1 (en) * 1999-01-21 2001-04-17 Bristol-Myers Squibb Company Complex of ras-farnesyltransferase inhibitor and sulfobutylether-7-β-cyclodextrin or 2-hydroxypropyl-β-cyclodextrin and method
US20040157796A1 (en) * 2002-11-27 2004-08-12 Yatin Gokarn Concentrated liquid valdecoxib composition

Non-Patent Citations (1)

* Cited by examiner, † Cited by third party
Title
Rowe, R.C., Sheskey, P.J., Owen, S.C. (2006) "Cyclodextrins" in HANDBOOK OF PHARMACEUTICAL EXCIPIENTS. Published by the Pharmaceutical Press, p. 217-221. *

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP3220917A4 (en) * 2014-11-18 2018-07-25 Pixarbio Corporation Compositions for treating acute, post-operative, or chronic pain and methods of using the same
EP3220955A4 (en) * 2014-11-18 2018-07-25 Pixarbio Corporation Compositions for treating acute, post-operative, or chronic pain and methods of using the same
WO2020157257A1 (en) * 2019-02-01 2020-08-06 H. Lundbeck A/S Injectable carbamazepine composition essentially free of 10-br-carbamazepine
US11529357B2 (en) 2019-02-01 2022-12-20 H. Lundbeck A/S Injectable carbamazepine composition essentially free of 10-bromo-carbamazepine

Also Published As

Publication number Publication date
US9770407B2 (en) 2017-09-26
US20210196623A1 (en) 2021-07-01
US20180153797A1 (en) 2018-06-07
US20140080812A1 (en) 2014-03-20
US20120283246A1 (en) 2012-11-08
US9629797B2 (en) 2017-04-25
US20130072476A1 (en) 2013-03-21

Similar Documents

Publication Publication Date Title
US20220265547A1 (en) Parenteral carbamazepine formulation
US20210196623A1 (en) Novel parenteral carbamazepine formulation
US20220160878A1 (en) Topiramate compositions and methods of making and using the same
JP6496381B2 (en) Formulations containing clopidogrel and sulfoalkyl ether cyclodextrins and methods of use
Brewster et al. Cyclodextrins as pharmaceutical solubilizers
Arima et al. Comparative studies of the enhancing effects of cyclodextrins on the solubility and oral bioavailability of tacrolimus in rats
CN100503647C (en) Hydroxypropyl- sulfobutyl-beta- cyclodextrin and its preparation method, analytical method and pharmaceutical uses
US5231089A (en) Method of improving oral bioavailability of carbamazepine
KR101129816B1 (en) Oral formulations of cladribine
AU2013200204B9 (en) Novel parenteral carbamazepine formulation
Bekers et al. 2′, 3′-Dideoxyinosine (ddI): its chemical stability and cyclodextrin complexation in aqueous media
Patani Biomembrane permeation of a heptylene-linked bistacrine analog: Characterization and modulation

Legal Events

Date Code Title Description
AS Assignment

Owner name: LUNDBECK INC., ILLINOIS

Free format text: CHANGE OF NAME;ASSIGNOR:OVATION PHARMACEUTICALS, INC.;REEL/FRAME:026461/0141

Effective date: 20090515

AS Assignment

Owner name: LUNDBECK LLC, ILLINOIS

Free format text: MERGER;ASSIGNOR:LUNDBECK INC.;REEL/FRAME:028537/0210

Effective date: 20111228

STCB Information on status: application discontinuation

Free format text: ABANDONED -- FAILURE TO RESPOND TO AN OFFICE ACTION