WO2003028624A2 - Levothyroxine compositions and methods - Google Patents

Levothyroxine compositions and methods Download PDF

Info

Publication number
WO2003028624A2
WO2003028624A2 PCT/US2002/025752 US0225752W WO03028624A2 WO 2003028624 A2 WO2003028624 A2 WO 2003028624A2 US 0225752 W US0225752 W US 0225752W WO 03028624 A2 WO03028624 A2 WO 03028624A2
Authority
WO
WIPO (PCT)
Prior art keywords
levothyroxine
composition
standard
cmax
test
Prior art date
Application number
PCT/US2002/025752
Other languages
French (fr)
Other versions
WO2003028624A3 (en
Inventor
G. Andrew Franz
Elaine A. Strauss
Phillip A. Dimenna
Rocco L. Gemma
Original Assignee
King Pharmaceuticals, Inc
Priority date (The priority date is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the date listed.)
Filing date
Publication date
Application filed by King Pharmaceuticals, Inc filed Critical King Pharmaceuticals, Inc
Priority to AU2002362468A priority Critical patent/AU2002362468A1/en
Publication of WO2003028624A2 publication Critical patent/WO2003028624A2/en
Publication of WO2003028624A3 publication Critical patent/WO2003028624A3/en

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2004Excipients; Inactive ingredients
    • A61K9/2022Organic macromolecular compounds
    • A61K9/205Polysaccharides, e.g. alginate, gums; Cyclodextrin
    • A61K9/2054Cellulose; Cellulose derivatives, e.g. hydroxypropyl methylcellulose
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/185Acids; Anhydrides, halides or salts thereof, e.g. sulfur acids, imidic, hydrazonic or hydroximic acids
    • A61K31/19Carboxylic acids, e.g. valproic acid
    • A61K31/195Carboxylic acids, e.g. valproic acid having an amino group
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/20Pills, tablets, discs, rods
    • A61K9/2072Pills, tablets, discs, rods characterised by shape, structure or size; Tablets with holes, special break lines or identification marks; Partially coated tablets; Disintegrating flat shaped forms
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P5/00Drugs for disorders of the endocrine system
    • A61P5/14Drugs for disorders of the endocrine system of the thyroid hormones, e.g. T3, T4

Definitions

  • 60/312,273 is entitled Levothyroxine Compositions Having Unique Tmax Properties
  • Serial No. 60/312,206 is entitled Levothyroxine Compositions Having Unique Triiodothyronine Tmax Properties
  • Serial No. 60/312,289 is entitled Unique Levothyroxine Aqueous Materials.
  • the invention generally relates to stable pharmaceutical compositions, and methods of making and administering such compositions.
  • the invention features stabilized pharmaceutical compositions that include pharmaceutically active ingredients such as levothyroxine (T4) sodium and liothyronine (T3) sodium (thyroid hormone drugs), preferably in an immediate release solid- dosage form.
  • pharmaceutically active ingredients such as levothyroxine (T4) sodium and liothyronine (T3) sodium (thyroid hormone drugs)
  • T3 thyroid hormone drugs
  • Thyroid hormone preparations of levothyroxine sodium and liothyronine sodium are pharmaceutical preparations useful to the treatment of hypothyroidism and. thyroid hormone replacement therapy in mammals, for example, humans and dogs.
  • Thyroid hormone preparations are used to treat reduced or absent thyroid function of any etiology, including human or animal ailments such as myxedema, cretinism and obesity. Hypothyroidism is a common condition. It has been reported in the United States Federal
  • hypothyroidism has a prevalence of 0.5 percent to 1.3 percent in adults, h people over 60, the prevalence of primary hypothyroidism increases to 2.7 percent in men and 7.1 percent in women. Because congenital hypothyroidism may result in irreversible mental retardation, which can be avoided with early diagnosis and treatment, newborn screening for this disorder is mandatory in North America. Europe, and Japan.
  • Thyroid hormone replacement therapy can be a chronic, lifetime endeavor.
  • the dosage is established for each patient Individually. Generally, the initial dose is small. The amount is increased gradually until clinical evaluation and laboratory tests indicate that an optimal response has been achieved. The dose required to maintain this response is then continued.
  • the age and general physical condition ofthe patient and the severity and duration of hypothyroid symptoms determine the initial dosage and the rate at which the dosage may be increased to the eventual maintenance level. It has been reported that the dosage increase should be very gradual in patients with myxedema or cardiovascular disease to prevent precipitation of angina, myocardial infarction, or stroke. It is important that thyroid hormone treatment have the correct dosage. Both under- treatment and over-treatment can have deleterious health impacts.
  • under-treatment In the case of under-treatment, a sub-optimal response and hypothyroidism could result. Under-treatment has also been reported to be a potential factor in decreased cardiac contractility and increased risk of coronary artery disease. Conversely, over-treatment may result in toxic manifestations of hyperthyroidism such as cardiac pain, palpitations, or cardiac arrhythmia's. In patients with coronary heart disease, even a small increase in the dose of levothyroxine sodium may be hazardous in a particular. Hyperthyroidism is a known risk factor fbr osteoporosis. Several studies suggest that sub clinical hyperthyroidism in premenopausal women receiving thyroid hormone drugs for replacement or suppressive therapy is associated with bone loss.
  • the dose be kept to the lowest effective dose. Because of the risks associated with over-treatment or under-treatment with levothyroxine sodium, there is a need for thyroid hormone products that are consistent in potency and bioavailability. Such consistency is best accomplished by manufacturing techniques that maintain consistent amounts ofthe active moiety during tablet manufacture.
  • Thyroid hormone drugs are natural or synthetic preparations containing tetraiodothyronine (T , levothyroxine) or triiodothyronine (T __, liothyronine) or both, usually as their pharmaceutically acceptable (e.g. sodium) salts.
  • T 4 and T 3 are produced in the human thyroid gland by the iodination and coupling ofthe amino acid tyrosine.
  • T 4 contains four iodine atoms and is formed by the coupling of two molecules of diiodotyrosine (DIT).
  • T 3 contains three atoms of iodine and is formed by the coupling of one molecule of DIT with one molecule of monoiodotyrosine (MIT). Both hormones are stored in the thyroid colloid as thyroglobulin.
  • Thyroid hormone preparations belong to two categories: (1) natural hormonal preparations derived from animal thyroid, and (2) synthetic preparations. Natural preparations include desiccated thyroid and thyroglobulin.
  • Desiccated thyroid is derived from domesticated animals that are used for food by man (either beef or hog thyroid), and thyroglobulin is derived from thyroid glands of the hog.
  • the United States Pharmacopoeia (USP) has standardized the total iodine content of natural preparations.
  • Thyroid USP contains not less than (NLT) 0.17 percent and not more than (NMT) 0.23 percent iodine, and thyroglobulin contains not less than (NLT) 0.7 percent of organically bound iodine.
  • Iodine content is only an indirect indicator of true hormonal biologic activity. Synthetic forms for both T 4 and T 3 thyroid hormone are available from a number of producers.
  • liothyronine sodium (T 3 ) tablets are available under the trademark Cytomel ® from King Pharmaceuticals, Inc., St. Louis, Missouri.
  • Levothyroxine sodium (T ) is available under the tradename Levoxyl ® from King Pharmaceuticals, Inc., under the tradename Synthroid ® from Knoll Pharmaceutical, Mt. Olive, New Jersey, and under the tradename Unithroid ® from Jerome Stevens Pharmaceuticals, Bohemia, New York.
  • a veterinarian preparation of levothyroxine so ⁇ Tum is available under the tradename Soloxine ® from King Pharmaceuticals, Inc.
  • Levoxyl ® (levothyroxine sodium tablets,USP) contain synthetic crystalline L-3,3',5,5 '- tetraiodothyronine sodium salt [levothyroxine (T 4 ) sodium].
  • T 4 tetraiodothyronine sodium salt
  • the levothyroxine (T 4 ) sodium in Levoxyl ® has an empirical formula of C ⁇ 5 H 10 L; NNaO 4 • H 2 O, molecular weight of 798.86 g/mol (anhydrous), and a structural formula as shown:
  • thyroid hormone drugs are quite poor. They are hygroscopic and degrade in the presence of moisture or light, and under conditions of high temperature. The instability is especially notable in the presence of pharmaceutical excipients. such as carbohydrates, including lactose, sucrose, dextrose and starch, as well as certain dyes.
  • pharmaceutical excipients such as carbohydrates, including lactose, sucrose, dextrose and starch, as well as certain dyes.
  • the critical nature of the dosage requirements, and the lack of stability of the active ingredients in the popular pharmaceutical formulations, have led to a crisis which has adversely effected the most prescribed thyroid drug products. See, e.g., 62 Fed. Reg. 43535 (Aug. 14, 1997).
  • U.S. Patent No. 5,22 5,204 (the '204 patent) is directed to improving the stability of levothyroxine sodium.
  • stabilized levothyroxine sodium was prepared in a dry state by mixing levothyroxine sodium with a cellulose tableting agent using geometric dilution and subsequently combining this mixture with the same or a second cellulose tableting agent, such as microcrystalline cellulose.
  • Other tableting aids or excipients can be used in this formulation.
  • This '204 patent is incorporated by reference herein, in its entirety.
  • the microcrystalline cellulose disclosed In '204 is AVICEL 101 ® , AVICEL 102 ® ,
  • microcrystalline cellulose products used in the '105 and '975 patents were also the ⁇ - form Avicel microcrystalline cellulose products.
  • U. S. Patents 5,955,105 and 6,056,975 are incorporated by reference herein, in their entirety.
  • Another microcrystalline cellulose product is a ⁇ - sheet form microcrystalline cellulose having a flat needle shape, marketed under the trademark CEOLUS KG801 ® by FMC Company of Newark, Delaware.
  • the Ceolus ® product has different morphology, and different performance characteristics, than those of the Avicel product.
  • the ⁇ - sheet microcrystalline cellulose of the present invention is disclosed in U.S. Patent 5,574,150, which is hereby incorporated by reference. Further disclosure relating to ⁇ - sheet microcrystalline cellulose is found in InternationalJournal of Pharmaceutics 182 (199) 155 which is hereby incorporated by reference.
  • the Ceolus ® product ( ⁇ - sheet microcrystalline cellulose) is disclosed by FMC, in its product bulletin dated October 1997, as being suitable for "smaller size tablets” and "exceptional drug carrying capacity.”
  • the Ceolus ® product was said to provide superior compressibility and drug loading capacity, that still exhibited effective flowability.
  • the examples given in the Ceolus ® bulletin were of vitamin C combined with Ceolus ® microcrystalline cellulose at levels of from 30 to 45 weight % Ceolus ® product in the form of a tablet.
  • Ceolus ® product there have been problems using the Ceolus ® product. For example, at higher levels of Ceolus ® product concentration, flow problems were encountered in the process of compressing tablets, and the Ceolus ® product was considered unsuitable for compression at higher concentrations than about 45 weight %.
  • T4 and T3 thyroid hormone drugs
  • T4 and T3 thyroid hormone drugs
  • T4 and T3 in an immediate release solid dosage form
  • T4 and T3 in the form of their sodium salts that are relatively stable.
  • T4 and T3 in the form of their sodium salts that are relatively stable.
  • T4 and T3 in the form of their sodium salts that are relatively stable.
  • T4 and T3 in the form of their sodium salts that are relatively stable.
  • T4 and T3 in the form of their sodium salts that are relatively stable.
  • T4 and T3 in the form of their sodium salts that are relatively stable
  • the present invention overcomes and alleviates the above-mentioned drawbacks and disadvantages in the thyroid drug art through the discovery of novel oral levothyroxine (T4) and/or liothyronine (T3) (thyroid hormone drags) pharmaceutical compositions and methods.
  • T4 oral levothyroxine
  • T3 liothyronine
  • the present invention relates to stabilized solid as levothyroxine (T4) sodium and/or liothyronine (T3) sodium (thyroid hormone drugs) pharmaceutical compositions and in particular, immediate release, stabilized pharmaceutical compositions that include pharmaceutically active ingredients such as levothyroxine (T4) sodium and/or liothyronine (T3) sodium (thyroid hormone drugs).
  • the novel pharmaceutical compositions are provided in a solid dosage form, such as a tablet.
  • compositions of the present invention are useful for, among other things, as replacement or supplemental therapy in hypothyroidism of any etiology, except transient hypothyroidism during the recovery phase of subacute thyroiditis, suppression of pituitary TSH secretion in the treatment or prevention of various types of euthyroid goiters, including thyroid nodules, Hashimoto's thyroiditis, multinodular goiter and, as adjunctive therapy in the management of thyrotropin-dependent well-differentiated thyroid cancer in warm- . blooded animals, especially humans including pediatrics.
  • the present invention also provides methods for making such immediate release and stabilized levothyroxine (T4) sodium and/or liothyronine (T3) sodium (thyroid hormone drugs) pharmaceutical compositions.
  • T4 immediate release and stabilized levothyroxine
  • T3 liothyronine
  • a method of administration to children and patients who have difficulty taking pills wherein the solid composition having the appropriate dosage is simply put in an aqueous fluid, e.g., juice, where it dissolves in a matter of 1-3 minutes, and the patient can then ingest the fluid, and receive the appropriate dosage, is now made practical.
  • an aqueous fluid e.g., juice
  • the present invention has a wide range of important uses including providing pharmaceutically active levothyroxine compositions with enhanced bioavailability, improved shelf life, and more reliable potency.
  • immediate release pharmaceutical compositions that include as pharmaceutically active ingredients at least one of levothyroxine and liothyronine, preferably at least one levothyroxine salt, as the major active ingredient.
  • Such preferred immediate release compositions desirably provide at least about 85% (w/v) dissolution of the levothyroxine salt in less than about 20 minutes as determined by standard assays disclosed herein.
  • compositions so that the ingredients are released almost immediately after ingestion or contact with an aqueous solution, e.g., in a matter of minutes.
  • Preferred invention compositions are stable and provide better shelf life and potency characteristics than prior pharmaceutical compositions.
  • the immediate release pharmaceutical compositions of the invention provide important uses and advantages.
  • a major advantage is the stability of the active ingredients in the composition.
  • prior formulations with sugars, starches, and various types of celluloses, including micro-cellular celluloses such as the Avicel products have experienced substantial degradation ofthe active ingredients, e.g. T4 sodium.
  • pharmaceutical manufacturers have over-formulated the T4-containing pharmaceutical compositions containing such active ingredients, so that the patient can obtain at least the prescribed dosage despite the carbohydrate-induced instability of the active ingredient.
  • the patient who obtains the pharmaceutical immediately after it is made receives an over-dosage of the active compound; whereas, the patient who has received the pharmaceutical after it has sat on the pharmacy shelf for an extended period, will receive an under-dosage ofthe active ingredient. In either case, the patient receives the wrong dosage, with possible serious consequences.
  • the use of the ⁇ -sheet microcrystalline cellulose in the compositions of the present invention substantially increase the stability of the thyroid hormone drugs, so that the patient obtains consistent potency over an extended shelf life, compared to prior thyroid hormone drug products.
  • the term "stabilized", as applied to levothyroxine and/or liothyronine means that the loss of potency over the shelf life ofthe product is less than about 0.7 % potency per month, for at least about 18 months.
  • Preferred compositions have a loss of potency of less than about 0.5% per month for sucn a period, and more preierre ⁇ compositions nave a loss ol potency ot less than about 0.3% per month for such a period.
  • compositions of the invention provide favorable pharmacokinetic characteristics when compared to prior formulations.
  • the immediate release pharmaceutical compositions that include levothyroxine salt have are more quickly available for absorption by the gastrointestinal (GI) tract faster and are absorbed more completely than has heretofore been possible.
  • This invention feature substantially enhances levothyroxine bioavailability, thereby improving efficacy and reliability of many standard thyroid hormone replacement strategies.
  • the desirable immediate release characteristics of the present invention facilitate dosing of patients who may be generally adverse to thyroid hormone replacement strategies involving solid dosing.
  • immediate release pharmaceutical compositions disclosed herein can be rapidly dissolved in an appropriate aqueous solution (e.g., water, saline, juice) or colloidal suspension (e.g., baby formula or milk) for convenient administration to such patients.
  • aqueous solution e.g., water, saline, juice
  • colloidal suspension e.g., baby formula or milk
  • infants, children, and adults who may experience swallowing difficulties e.g., infants, children, and adults who may experience swallowing difficulties.
  • the invention thus makes standard thyroid hormone replacement strategies more flexible and reliable for such patients.
  • the invention features an immediate release pharmaceutical composition
  • a levothyroxine salt preferably one of such a salt.
  • At least about 80% ofthe levothyroxine dissolves in aqueous solution in less than about 20 minutes as determined by a standard assay, disclosed herein.
  • at least about 80% of the levothyroxine is dissolved in the aqueous solution by about 15 minutes from the time that the composition, in pill form, is placed in the aqueous solution.
  • at least about 85% ofthe levothyroxine is released to the aqueous solution by about 10 minutes, most preferably by about 5 minutes after exposure of the composition to the aqueous solution.
  • compositions in accordance with the present invention can be formulated to release 85% of the levothyroxine within 2-3 minutes after exposure to the aqueous solution.
  • compositions with favorable immediate release characteristics it is believed that by combining one or more of the pharmaceutically active agents with ⁇ -form microcrystalline cellulose, it is possible to produce compositions with favorable immediate release characteristics. Without wishing to be bound to theory, it is believed that the agents do not bind well to certain grades of the ⁇ - sheet form microcrystalline cellulose. More o ⁇ me agent is thus available for immediate release, m contrast, it is believed that many prior formulations have active agents that bind cellulose additives, making less available. The release characteristics ofthe compositions ofthe invention are also improved by the use of other agents, as discussed further below.
  • the present invention relates to a stabilized pharmaceutical composition
  • a stabilized pharmaceutical composition comprising a pharmaceutically active ingredient, such as levothyroxine, and the ⁇ - sheet form of microcrystalline cellulose, in the form of a solid dosage.
  • a pharmaceutically active ingredient such as levothyroxine sodium and/or liothyronine sodium, at least about 50 weight % ofthe dosage weight composed ofthe ⁇ - sheet form of microcrystalline cellulose, and, optionally, additional excipients, in a solid dosage form.
  • the invention provides an aqueous solution or colloidal suspension that includes at least one ofthe compositions of this invention, preferably between from about one to about five of same, more preferably about one of such compositions. It has also been found that ⁇ - sheet microcrystalline cellulose grades having preferred bulk densities provide for more compact processing than use of other celluloses. That is, use of the ⁇ - sheet microcrystalline cellulose having bulk densities in accord with this invention helps to provide for higher compression ratios (initial volume/final volume). As discussed below, other invention aspects help reduce or avoid production of damaging compression heat that has damaged prior formulations made from high compression ratios. The compositions of the present invention generally also require less compressional force to form the tablets.
  • the invention also provides methods for making an immediate release pharmaceutical composition comprising at least one levothyroxine salt, preferably one of such a salt.
  • the method includes at least one and preferably all of the following steps: a) mixing a levothyroxine salt with microcrystalline ⁇ -cellulose and preferably a crosscarmellose salt to make a blend; and
  • the method involves preparing an oral dosage form of a pharmaceutically active ingredient comprising dry blending the pharmaceutically active ingredient and at least about 50 weight % ofthe ⁇ - sheet form of microcrystalline cellulose, and compressing the blend to form a solid dosage.
  • Figures 1A - 1C illustrate various solid dosage forms such as cylindrical tablets and raised violin shaped tablets;
  • Figure 2 illustrates a tableting die pair
  • Figure 3 pair is graphical depiction of comparative dissolution data of various strengths of Levoxyl ® tablets made in accordance with the invention.
  • Figure 4A is an HPLC chromatogram showing a levothryoxine and liothyronine standards.
  • Figure 4B is an HPLC chromatograph showing results of levothyroxine sodium sample made in accordance with the present invention.
  • Figure 5A is a chromatogram showing various levothryoxine impurity standards.
  • Figure 5B is a chromatograph showing results of levothyroxine sodium sample made in accordance with the present invention.
  • T4 novel oral levothyroxine
  • T3 liothyronine
  • the invention relates to immediate release solid pharmaceutical compositions such as stabilized pharmaceutical compositions that include pharmaceutically active ingredients such as levothyroxine (T4) sodium and liothyronine (T3) sodium (thyroid hormone drugs), preferably in a solid dosage form. Also provided are methods for making such immediate release and stabilized compositions.
  • pharmaceutically active ingredients such as levothyroxine (T4) sodium and liothyronine (T3) sodium (thyroid hormone drugs)
  • T3 thyroid hormone drugs
  • immediate release a pharmaceutical composition in which one or more active agents therein demonstrates at least about 80% (w/v) dissolution, preferably between from about 90% (w/v) to about 95% (w/v), more preferably about 95% (w/v) to about
  • a pharmaceutical composition of the invention is “stable” or “stabilized” if one or more ofthe active agents therein exhibit good stability as determined by a standard potency test. More specifically, such compositions exhibit a potency loss of less than about 15%, preferably less than about 10%, more preferably less than about 1% to about 5% as determined by the test.
  • Potency can be evaluated by one or a combination of strategies known in the field. See the USP.
  • a preferred potency test compares loss or conversion of the active agent in the presence
  • the pharmaceutical compositions of the invention include, as active agent, levothyroxine (T4), preferably a salt thereof such as levothyroxine sodium USP.
  • T4 levothyroxine
  • Such compositions typically exhibit a levothyroxine (T4) plasma Cmax of between from about 12 ⁇ g/dl to about 16 ⁇ g/dl, preferably as determined by the standard Cmax test.
  • the In(Cmax) ofthe levothyroxine (T4) plasma level is between from about 1 to about 3.
  • the standard Cmax test can be performed by one or a combination of strategies known in the field. See e.g., the USP.
  • a preferred Cmax test is disclosed below in Examples 8 and 9.
  • compositions in accord with the invention provide a triiodothyronine (T3) plasma Cmax of between from about 0.1 ng/ml to about lOng/ml, preferably 0.5 ng/ml to about 2ng/ml, as determined by the standard Cmax test.
  • T3 triiodothyronine
  • the In(Cmax) is between from about 0.01 to about 5.
  • Further preferred compositions exhibit a levothyroxine (T4) plasma Tmax of between from about 0.5 hours to about 5 hours, preferably as determined by a standard Tmax test.
  • the standard Tmax test can be performed by procedures generally known in the field. See e.g., the USP. A preferred Tmax test is disclosed below in Examples 8 and 9.
  • compositions of the invention exhibit a triiodothyronine (T3) plasma Tmax of between from about 10 hours to about 20 hours, preferably about 12 to about 16 hours as determined by the standard Tmax test.
  • T3 plasma Tmax of between from about 10 hours to about 20 hours, preferably about 12 to about 16 hours as determined by the standard Tmax test.
  • compositions feature a levothyroxine (T4) plasma AUC (0-t) of between from about 450 ⁇ g-hour/dl to about 600 ⁇ g-hour/dl, preferably 500 ⁇ g-hour/dl to about 550 ⁇ g-hour/dl as determined by a standard AUC (0-t) test.
  • T4 plasma AUC (0-t) of between from about 450 ⁇ g-hour/dl to about 600 ⁇ g-hour/dl, preferably 500 ⁇ g-hour/dl to about 550 ⁇ g-hour/dl as determined by a standard AUC (0-t) test.
  • the In[AUC(0- t)] is between from about 1 to about 10.
  • compositions feature a triiodothyronine (T3) AUC (0-t) of between from about 10 ng-hour/ml to about 100 ng-hour/ml, preferably 20 ng-hour/ml to about 60 ng-hour/ml, as determined by the standard AUC (0-t) test.
  • T3 AUC triiodothyronine
  • the In[AUC(0-t)] is between from about 1 to about 5.
  • compositions of the invention are provided in which the active material is a non-granulated material.
  • Prior levothyroxine compositions have been granulated in various size reduction machines to grains of less than, e.g., 5 - 20 microns average particle size in order to be effectively incorporated into the administrable pharmaceutical composition.
  • the granulation process subjects the active material to degrading heat, which can have adverse effects on the active material, as well as reducing the activity level.
  • Prior manufacturers purchase micronized levothyroxine manufactured under DMF No. 4789, and then granulate it before incorporating it into the levothyroxine pharmaceutical product.
  • the raw material is not granulated before incorporation into the pharmaceutical composition. Rather, the ingredients of the preferred pharmaceutical are mixed and the mixture is subjected to direct compression to form the pharmaceutical tablets of appropriate dosage. As a result, the activity of the active ingredient is not degraded prior to the direct compression step.
  • Bulk levothyroxine is obtained in a fine powdered form, preferably from Biochemie GmbH, A-6250 Kundl, Austria. More importantly, the use of the preferred process results in a product which is immediately dispersible in aqueous solution, to make the active ingredient available for absorption in the body.
  • non-granulated means that the bulk USP compound is used without subjecting it to granulators or similar high energy size reduction equipment before being mixed with the other pharmaceutical components and formed into the appropriate pill.
  • the bulk active ingredient is mixed with the appropriate amounts of other ingredients and directly compressed into pill form. Since it is not necessary to granulate the material, it is not necessary to subject it to degrading temperatures in the process of forming the pharmaceutical compositions containing the active materials. In the present process we start with micronized active material, which merely needs to be blended with the B and other materials and then compressed. Others have to be granulated, and then dried, which steps interfere with the dissolution of the active material.
  • the drying temperatures employed in manufacturing other active ingredients can cause degradation of the levothyroxine, as experienced in other available thyroxine. It has been found that providing the invention compositions in a non-granulated format helps to reduce or eliminate active agent degradation, presumably by facilitating a reduction in friction, and thus degrading heat, during compression ofthe compositions into pills.
  • the ⁇ -fo ⁇ n microcrystalline cellulose has a bulk density of between from about 0.10 g/cm 3 to about 0.35 g/cm 3 , more preferably between from about 0.15 g/cm 3 to about
  • 0.25 g/cm 3 still more preferably between from about 0.17 g/cm 3 to about 0.23 g/cm 3 , most preferably between from about 0.19 g/cm to about 0.21 g/cm .
  • the ⁇ -form microcrystalline cellulose are substantially non- conductive.
  • the ⁇ -form microcrystalline cellulose has a conductivity of less than about 200 ⁇ S/cm, more preferably, less than about 75 ⁇ S/cm, still more preferably between from about 0.5 ⁇ S/cm to 50 ⁇ S/cm, most preferably between from about 15 ⁇ S/cm to 30 ⁇ S/cm.
  • a specifically preferred ⁇ -form microcrystalline cellulose is sold by Asahi Chemical Industry Co., Ltd (Tokyo, Japan) as Ceolus (Type KG-801 and or KG-802).
  • compositions of the invention have a post-packaging potency of between from about 95% to about 120%, preferably 98% to about 110% as determined by the standard potency test.
  • the present invention is a pharmaceutical product that is in the form of a solid dosage, such as a sublingual lozenge, buccal tablet, oral lozenge, suppository or a compressed tablet.
  • the pharmaceutically active ingredient is dry mixed with the ⁇ -form of the microcrystalline cellulose, optionally with additional excipients, and formed into a suitable solid dosage.
  • Preferred tablets according to the invention have a total hardness of between from about 1 to about 30 KP, preferably about 6 to about 14 KP as determined by a standard hardness test. Methods for determining tablet hardness are generally known in the field. See e.g., the USP. A preferred standard hardness test is disclosed below in Example 4. Additionally preferred pharmaceutical compositions including those in tablet format preferably include less than about 10% total impurities, more preferably less than about 5% of same as determined by a standard impurity test.
  • Reference herein to the "standard impurity test” means a USP recognized assay for detecting and preferably quantitating active drug degradation products.
  • such products include, but are not limited to, at least one of diiodothyronine (T2), triiodothyronine (T3), levothyroxine, t ⁇ iodothyroacetic aci " amide, tri ⁇ odothyroethyiamine, triiodothyroacetic acid, triiodothyroethyl alcohol, tetraiodothyroacetic acid amide, tetraiodothyroacetic acid, triiodothyroethane, and tetraiodothyroethane.
  • T2 diiodothyronine
  • T3 triiodothyronine
  • T3 triiodothyroacetic acid
  • tetraiodothyroacetic acid impurities A preferred impurity test for monitoring levothyroxine and liothyronine breakdown products involves liquid chromatography (LC) separation and detection, more preferably HPLC. Specifically preferred impurity tests are provided below in Examples 5 and 6
  • compositions in accord with the invention include one or more standard disintegrating agents, preferably crosscarmellose, more preferably a salt of same. Still further preferred compositions include a pharmaceutically acceptable additive or excipient such as a magnesium salt.
  • the present invention can be prepared as a direct compression formula, dry granulation formula, or as a wet granulation formula, with or without preblending of the drug, although preferably with preblending.
  • the pharmaceutically active ingredient can be any type of medication which acts locally in the mouth or systemically, which is the case of the latter, can be administered orally to transmit the active medicament into the gastrointestinal tract and into the blood, fluids and tissues of the body.
  • the medicament can be of any type of medication which acts through the buccal tissues of the mouth to transmit the active ingredient directly into the blood stream thus avoiding first liver metabolism and by the gastric and intestinal fluids which often have an adverse inactivating or destructive action on many active ingredients unless they are specially protected against such fluids as by means of an enteric coating or the like.
  • the active ingredient can also be of a type of medication which can be transmitted into the blood circulation through the rectal tissues.
  • Representative active medicaments include antacids, antimicrobials, coronary dilators, peripheral vasodilators, anti psychotropics, antimanics, stimulants, antihistamines, laxatives, decongestants, vitamins, gastrosedatives, antidiarrheal preparations, vasodilators, antiarrythmics, vasoconstrictors and migraine treatments, anticoagulants and antithrombotic drugs, analgesics, antihypnotics, sedatives, anticonvulsants, neuromuscular drugs, hyper and hypoglycemic agents, thyroid and antithyroid preparations, diuretics, antispasmodics, uterine relaxants, mineral and nutritional additives, antiobesity drugs, anabolic drugs, erythropoietic drugs, antiasthematics, expectorants, cough suppressants, mucolytics, antiuncemic drugs
  • Typical active medicaments include gastrointestinal sedatives such as metoclopramide and propantheline bromide, antacids such as aluminum trisilicate, aluminum hydroxide and cimetidine, asprin-like drugs such as phenylbutazone, indomethacin, and naproxen.
  • gastrointestinal sedatives such as metoclopramide and propantheline bromide
  • antacids such as aluminum trisilicate, aluminum hydroxide and cimetidine
  • asprin-like drugs such as phenylbutazone, indomethacin, and naproxen.
  • ibuprofen ibuprofen, flurbiprofen, diclofenac, dexamethasone, prednisone and prednisolone, coronary vasodialator drugs such as glyceryl trinitrate, isosorbide dinitrate and pentaerythritol tetranitrate, peripheral and cerebral vasodilators such as soloctidilum, vincamine, naftidrofuryl oxalate, comesylate, cyclandelate, papaverine and nicotinic acid, antimicrobials, such as erythromycin stearate, cephalexin, nalidixic acid, tetracycline hydrochloride, ampicillin, flucolaxacillin sodium, hexamine mandelate and hexamine hippurate, neuroleptic drugs such as fluazepam, diazepam, temazepam, amitryptyline, doxe
  • diltiazem diltiazem, procainamide, disopyramide, bretylium tosylate, quinidine sulfate and quinidine gluconate
  • drags used in the treatment of hypertension such as propranolol hydrochloride, guanethidine monosulphate, methyldopa, oxprenolol hydrochloride, captopril, Actace and hydralazine
  • drags used in the treatment of migraine such as ergotamine
  • drags effecting coagulability of blood such as epsilon aminocaproic acid and protamine sulfate
  • analgesic drags such as acetylsalicyclic acid, acetaminophen, codeine phosphate, codeine sulfate, oxycodone, dihydrocodeine tartrate, oxydodeinone, morphine, heroin, nalbuphine, buto ⁇
  • the amount of pharmaceutically active ingredient in the present composition can vary widely, as desired.
  • the active ingredient is present in a composition of the present invention in an effective dosage amount.
  • Exemplary of a range that the active ingredient may be present in a composition in accordance with the present invention is from about 0.000001 to about 10 weight %. More preferably, the amount of active ingredient is present in the range of about 0.001 to 5 weight %.
  • any suitable pharmaceutically acceptable form of the active ingredient can be employed in the compositions of the present invention, i.e., the free base or a pharmaceutically acceptable salt thereof, e.g., levothyroxine sodium salt, etc.
  • the preferred amount ofthe active moiety in the composition is present in the range of about 0.00005 to about 5 weight %.
  • the more preferred range is from about 0.001 to about 1.0 weight %, and the most preferred range is from about 0.002 to about 0.6 weight % levothyroxine.
  • the minimum amount of levothyroxine can vary, so long as an effective amount is utilized to cause the desired pharmacological effect.
  • the dosage forms have a content of levothyroxine in the range of about 25 to 300 micrograms per 145 milligram pill for human applications, and about 100 to 800 micrograms per 145 mg pill for veterinary applications.
  • a goal of levothyroxine replacement therapy is to achieve and maintain a clinical and biochemical euthyroid state, whereas a goal of suppressive therapy is to inhibit growth and/or function of abnormal thyroid tissue.
  • a dose of levothyroxine that is adequate to achieve these goals depends of course on a variety of factors including the patient's age, body weight, cardiovascular status, concomitant medical conditions, including pregnancy, concomitant medications, and the specific nature ofthe condition being treated. Hence, the following recommendations serve only as dosing guidelines. It should be understood by those versed in this art that dosing should be individualized and adjustments made based on periodic assessment of a patient's clinical response and laboratory parameters.
  • levothyroxine when using levothyroxine to treat, it should be taken in the morning on an empty stomach, at least one-half hour before any food is eaten.
  • levothyroxine is preferably taken at least about 4 hours apart from drags that are known to interfere with its absorption.
  • the peak therapeutic effect at a given dose of levothyroxine sodium may not be attained for about 4 to about 6 weeks.
  • the average full replacement dose of levothyroxine sodium is approximately 1.7 mcg/kg/day (e.g., about 100 to about 125 meg/day for a 70 kg adult). Older patients may require less than 1 mcg/kg/day. Levothyroxine sodium doses greater than about 200 meg/day may or may not be required.
  • an initial starting dose of about 25 to about 50 meg/day of levothyroxine sodium is recommended, with gradual increments in dose at about 6 to about 8 week intervals, as needed.
  • the recommended starting dose of levothyroxine sodium in elderly patients with cardiac disease is about 12.5 to about 25 meg/day, with gradual dose increments at about 4 to about 6 week intervals.
  • the levothyroxine sodium dose is generally adjusted in about 12.5 to about 25 meg increments until the patient with primary hypothyroidism is clinically euthyroid and the serum TSH has normalized.
  • the recommended initial levothyroxine sodium dose is about 12.5 to about 25 meg/day with increases of about 25 meg/day about every 2 to about 4 weeks, accompanied by clinical and laboratory assessment, until the TSH level is normalized.
  • the levothyroxine sodium dose should be titrated until the patient is clinically euthyroid and the serum free-T 4 level is restored to the upper half of the normal range.
  • levothyroxine therapy may be instituted at full replacement doses as soon as possible.
  • Levothyroxine compositions ofthe present invention may be administered to infants and children who cannot swallow intact tablets by crashing the tablet and suspending the freshly crushed tablet in a small amount (5-10 mL or 1-2 teaspoons) of water. This suspension can be administered by spoon or dropper. Foods that decrease abso ⁇ tion of levothyroxine, such as soybean infant formula, should not be used for administering levothyroxine sodium tablets.
  • a recommended starting dose of levothyroxine sodium in newborn infants is about 10 to about 15 mcg/kg/day.
  • a lower starting dose (e.g., about 25 meg/day) may be considered in infants at risk for cardiac failure, and the dose should be increased in 4-6 weeks as needed based on clinical and laboratory response to treatment, hi infants with very low ( ⁇ about 5 mcg/dL) or undetectable serum T 4 concentrations, a recommended initial starting dose is about 50 meg/day of levothyroxine sodium.
  • levothyroxine therapy is usually initiated at full replacement doses, with the recommended dose per body weight decreasing with age (see Dose Table below).
  • an initial dose of about 25 meg/day of levothyroxine sodium is recommended with increments of 25 meg every 2-4 weeks until the desired effect is achieved.
  • Hyperactivity in an older child may be minimized if the starting dose is one-fourth ofthe recommended full replacement dose, and the dose is then increased on a weekly basis by an amount equal to one-fourth the full-recommended replacement dose until the full recommended replacement dose is reached.
  • Levothyroxine sodium tablets, USP, in accordance with the present invention may be supplied as oval or violin-shaped, color-coded, potency marked tablets in, for example, 12 strengths as indicated in the Strength Table
  • the preferred amount of the active moiety in the composition is present in the range of about 0.000005 to 0.5 weight %.
  • the more preferred range is from about 0.00001 to 0.1 weight %, and the most preferred range is from about 0.00004 to about 0.002 weight % liothyronine.
  • the minimum amount of lyothyronine can vary, so long as an effective amount is utilized to cause the desired pharmacological effect.
  • the dosage forms have a content of levothyroxine in the range of about 5 to 50 micrograms per 145 milligram pill for human applications.
  • the ⁇ -form microcrystalline cellulose product of the present invention is prepared by forming a wet cake, drying the cake with a dram dryer, then passing the dried product through a screen or mill tor sizing which produces a ⁇ -sheet microcrystalline cellulose which has a flat needle shape, as disclosed in U.S. Patent 5,574,150.
  • ⁇ -sheet microcrystalline product is available from Asahi Chemical of Japan and/or marketed by FMC Company of Newark, Del, under the trademark Ceolus®.
  • the mo ⁇ hology and performance characteristics of the Ceolus® product are different from those of ⁇ - form microcellulose products (for example, Avicel® and Emcocel®), and are suitable for preparing the present stabilized pharmaceutical composition.
  • the amount of ⁇ -form microcrystalline product used in the present composition is at least 50 weight % ofthe final composition.
  • the amount of ⁇ -fonn microcrystalline product is in the range of about 50 to 99 weight %.
  • the amount of ⁇ -fonn microcrystalline product is in the range of about 60 to 90 weight % ofthe final composition.
  • suitable excipients for the present invention include fillers such as starch, alkaline inorganic salts such as trisodium phosphate, tricalcium phosphate, calcium sulfate and sodium or magnesium carbonate.
  • the fillers can be present in the present composition in the range of about 0 to 50 weight %.
  • Suitable disintegrating agents include corn starch, cross-linked sodium carboxymethylcellulose (crosscarmellose) and cross-linked polyvinyipyrrolidone (crospovidone).
  • a preferred disintegrating agent is crosscarmellose.
  • the amount of disintegrating agent used is in the range of about 0 to 50 weight %.
  • the disintegrating agent is in the range of about 5 to 40 weight %, more preferably about 10 to about 30 weight %.
  • the recommended loading of crosscarmellose is 0.5 to about 2%> by weight.
  • the higher loadings of the disintegrating agents substantially improves the ability ofthe product to disperse in aqueous media.
  • Suitable gildents for use in the present invention include colloidal silicon dioxide and talc.
  • the amount of gildent in the present composition is from about 0 to 5 weight %, and the preferred amount is about 0 to 2 weight %.
  • Suitable lubricants include magnesium and zinc stearate. sodium stearate fumarate and sodium and magnesium lauryl sulfate. A preferred lubricant is magnesium stearate.
  • the amount of lubricant is typically in the range of about 0 to 5 weight %>, preferably in the range of about 0.1 to 3 weight %.
  • the oral pharmaceutical product is prepared by thoroughly mtermixing the active moiety and the ⁇ -form of microcrystalline cellulose, along with other excipients to form the oral dosage. Food grade dyes can also be added. For example, it is common to distinguish dosages of various potency by the color characteristics of such dyes.
  • a preferred immediate release pharmaceutical composition in tablet form includes levothyroxine sodium, hi a preferred embodiment, the composition includes at least one of, preferably all ofthe following:
  • NF (Ceolus) having a bulk density of between from about 0.10 g/cm 3 to about 0.35 g/cm 3 , c) between from about 25mg/tablet to about 50mg/tablet of crosscarmellose sodium, NF (Ac-di-sol); and d) between from about 0.5 mg/tablet to about 5mg/tablet of magnesium stearate, NF.
  • the composition further comprises at least one pharmaceutically acceptable coloring agent.
  • compositions having less than about 5% total impurities as determined by the standard impurity test Preferably, the method further comprises forming a tablet, particularly those tablets having a raised violin configuration.
  • the stabilized oral dosages of thyroid hormone are prepared by forming a trituration of the active moiety (i.e. levothyroxine sodium and/or liothyronine sodium) and ⁇ -form microcrystalline cellulose. The trituration is blended with ⁇ -form microcrystalline cellulose and additional excipients and compressed into oral dosages.
  • the formulation batches are a blend of solid compositions of various shapes and sizes. Blending is used to achieve a measure of homogeneity.
  • the active thyroid moiety is desired to be evenly distributed throughout the batch, hi a typical 410 kg batch, the amount of active moiety represents less than 1 kg of the total weight. For example, when producing 145 mg tablets with a 300 meg dosage, approximately 0.8 kg of a 410 kg batch is the active moiety.
  • each tablet is formulated to contain 100% label claim potency. It is typical for compressible medicament tablets to be formed using a 2:1 fill to compression ratio. However, for medicament tablets formed using the present invention a fill to compression ratio from 3.3:1 to 4:1 is needed to obtain desired tablet density.
  • the ⁇ -form microcrystalline cellulose has a lower bulk density, as compared to other excipients.
  • Tableting machines are commonly known to practitioners in the art and include those available from Manesty and Stokes. It has been found that making such adjustments to the compression ratio results in poor tablet surface finish as well as inconsistent tablet weights. Instead, the design of the tableting dies should be adjusted. It has been determined that during the filling ofthe tableting dies, a minimum of 5-6mm die overfill. In most cases this requires replacement of the usual tableting dies with dies which are an additional 2-3 mm deep.
  • the shape of the tablet is configured to increase heat transfer away from the tablet. More preferred tablets have a surface area per tablet of between from about 0.9 in. 2 to about 0.15 in. 2 , preferably about 0.115in. 2 , to assist such heat transfer. Additional tablet configurations are contemplated e.g., tablets that are beveled and/or include a notch.
  • a preferred tablet shape is a raised violin configuration, as shown in Figure lC.
  • Example 1 tablets comprise the ⁇ -f ⁇ rm microcrystalline cellulose while Control 1 tablets comprise the traditional -for m microcrystalline cellulose.
  • the stability of pharmaceutical formulations of the present invention is improved significantly by the use of the ⁇ - sheet microcrystalline cellulose.
  • Potency loss of the present invention after 15 months is 3.5 %, versus 16.0 %> potency loss experienced in a similar formulation with the ⁇ -form microcrystalline cellulose.
  • the average loss in potency per month in the case of the compositions of the present invention was only about 0.2 % per month, as compared to over 1% per month for the T4 products which included ⁇ -form microcrystalline cellulose, thus demonstrating a stability which is about 3 to 4 times better than the T4 products which utilized ⁇ -form microcrystalline cellulose.
  • Tableting testing was performed on the formulation for Example 1 tablets. Initial results with standard die depths provided a relative standard deviation of 2.2 to 3.5% tablet weight. With the use of the herein described extra deep tablet dies, the- relative standard deviation is 1.2%. Testing was performed on a Manesty tableting machine with compression ratios of from 3.3:1 to 4.0:1.
  • Tablet quality is also dependent upon the storage of the ⁇ -sheet microcrystalline cellulose. Best results are achieved when the cellulose is received in drums or portable containers instead of bags. The bag form suffers from compression during transportation from raw material suppliers. Test results for tableting are presented in attached Exhibit A.
  • Table 5 shows drag stability data for a number ofthe above formulations:
  • formulations of the present invention provide extreme stability for the levothyroxine activity over an extended shelf life for these pharmaceutical products.
  • Example 2
  • Table 8 shows comparative dissolution data for all strengths of Levoxyl ® tablets.
  • the appropriate dosage for the patient in question, in an immediate release pill made in accordance with the present invention is simply mixed with a suitable amount, e.g. 50 - 200 ml, of aqueous fluid, such as water, soft drinks, juice, milk, etc.
  • aqueous fluid such as water, soft drinks, juice, milk, etc.
  • the immediate release pill is easily dissoluted in the fluid, optionally with stirring or shaking, and simply admimstered to the patient.
  • the tablet potency can be tested according to method AM-021.
  • Method number: AM-021 is the same as method number: AM-003, except the tablets are dissolved whole without first grinding the tablets into a powder, as with method number: AM-003.
  • Chromatograph 5 replicate injections of the standard preparation. Record the peak responses as directed under "Procedure”.
  • T 3 / T working standard Dilute to volume with mobile phase and mix well. Label this standard as T 3 / T working standard.
  • concentration of the working standard should be about 0.2 ⁇ g/ml T 3 and 10.0 ⁇ g/ml T 4 .
  • Figures 5A and 5B show HPLC chromatograms of levothyroxine and liothyronine controls (T3/T4 working standard, shown in Figure 5A) and an experimental sample made in accordance with the present invention as described above.( Figure 5B).
  • the peaks in both chromatograms in the area of 1.325 to 3.1 correspond to materials in the solvent.
  • the peak at about 7.2 in Fig. 5 A shows the presence of T3.
  • Fig. 5B shows the absence of T3, as well as the absence of other related products or degradation products of levothyroxine.
  • the hardness of the pills lies between about 6.0 and about 14.0 kiloponds.
  • the pill hardness is from about 9 to about 13 kiloponds.
  • Typical results of products made in accordance with the present invention are about 9.3, 11.3, 9.8, 10.2, 12.3, etc.
  • Pharmaceutical tablets which inco ⁇ orate granulated active ingredient are typically much higher in hardness, which may add to the difficulty of dissolving or dissoluting them. Pills which are lower in hardness generally present more problems of pill fragmentation during handling and storage.
  • HPLC grade water Store in a plastic container.
  • Solvent 1 To 100.0 ml of 0.1 N Sodium Hydroxide solution add a 1:1 V/V mixture of methanol and water to make 1000 ml.
  • Solvent 2 77:23:0.1 H2): CANACN: H3P04; Degassed and filtered; mobile phase composition a may be altered to achieve a satisfactory resolution factor.
  • Extraction solution Pipette 50 ml of solvent 1 into a 1000 ml volumetric flask dilute to volume with solvent 2, stopper and mix well.
  • Injection Volume 100 up: next injection after approx. 40 min.
  • Chromatograph 5 replicate injections ofthe Reference I Standard preparation, chromatograpri 2 replicate injections ofthe Reference II Standard. Record the peak responses as directed under "Procedure”. An extraction blank is to be run after the standards.
  • the RSD must not be greater than 2.0% for each ofthe impurities in the standard reference solution I.
  • the resolution factor between liothyronine and levothyroxine in thestandard reference solution I must not be less than 5.0.
  • the Signal to Noise ratio must not be less than 5/1 for levothyroxine and impurities in the chromatogram obtained with standard reference solution II.
  • a peak of monochlorotriiodothyronine may occur just before the levothyroxine peak. Make sure that the degree of separation between this peak and of levothyroxine is at least sufficient to permit separate evaluations.
  • Monochlorotriiodothyronine reference material is not available to be purchase by any vendor. Any calculation of monochlorotriiodothyronine impurity will be done by its retention time.
  • Standard Reference Solution Preparation: Accurately weigh 10 mg +/- 0.1 mg of each Diiodothyronine, Liothyronine, Levothyroxine, Triiodothyroacetic acid and Tetraiodthyroacetic acid reference standards into a 100ml volumetric flask. Dissolve in Solvent 1 and dilute to volume, stopper and mix well. The concentration of each component will be approximately lOOmcg/mlL.
  • Test Preparation Crush not less than 20 tablets. Tare a 250 ml Erlenmeyer flask. Accurately weigh to the nearest 0.1 mg an equivalent of 500 meg of levothyroxine sodium (+/- 10%) into a 250 ml Erlenmeyer flask. Pipette 100.0 meg of the Extraction solution into the flask cover the flask with parafilm, sonicate, vortex and then centrifuge the solution for 1 minute each. The final concentration ofthe sample will be approximately 5 meg/ml of levothyroxine.
  • Procedure 1. Separately inject lOO ⁇ l ofthe sample preparation onto the column. Record the response ofthe analyte peaks and the calculate % w/w using the equations below.
  • the chromatogram may need to be reprocessed to obtain optimal integration.
  • a copy ofthe sample chromatograph is to be attached to the analytical packet.
  • Triiodothyroacetic Acid Sample area X Std Cone, (meg) X 100ml X 100% % w/w
  • Area rs - is the average area ofthe levothyroxine in the Standard reference solution II
  • Wt. the initial weight ofthe levothyroxine USP standard in mg
  • P the purity ofthe levothyroxine Na USP standard (%purity/100%)
  • 1.0283 conversion of levothyroxine into levothyroxine sodium
  • Area i mpu ri ty is the area of the greatest unknown impurity in the test solution with an area greater than the theoretical area for 0.05% ofthe levothyroxine Na taken into account.
  • Area ref std I is the area ofthe levothyroxine in the standard reference solution
  • Sum area impurity is the sum ofthe areas of all the other unknown impurities in the test solution
  • Area ref std I is the area ofthe levothyroxine in the standard reference solution I
  • A is the initial weight of levothyroxine Na in mg represented by the sample weight.
  • 0.1450gX 100 mcg/mg 2000 is the dilution ofthe reference solution.
  • Results of the test are shown in Figures 6 A and 6B.
  • Figure 6A shows an example of a chromatogram of Standard Reference Solution II, with exemplary peaks at about 5.4 for diiodo-1- thyronine, 8.4 for Hotliryonine, 12.8 for levothyroxine, 19.3 for triiodo thyroacetic acid, and 21.9 for tetraiodo thyroacetic acid.
  • Figure 6B shows results of an experimental sample of levothyroxine sodium, made in accordance with this invention. As can be seen, the sample had substantially only levothyroxine, with insignificant impurities.
  • composition may be altered to achieve a satisfactory resolution factor.
  • the R value must be greater than or equal to proceed.
  • Standard Preparation Accurately weigh 25 mg of USP Levothyroxine RS and transfer to a clear 250-mlL volumetric flask. Pipette 87.5 ml minute of acetonitrile in the flask. Swirl and then sonicate for less than a minute. Add portions of HPLC grade water to the flask with swirling and sonicating until the material has gone into solution. Be sure that there is no particulate material present. Do not dilute to volume at this point. The solution may be cold. Place into a room temperature water bath for ten minutes to allow the sample to warm to ambient temperature. Dilute to volume with HPLC grade water. Mix well. Label this solution as stock T 4 . The concentration of T 4 is about 100 ⁇ g/ml.
  • T 3 /T 4 working standard Dilute to volume with mobile phase and mix well. Label this standard as T 3 /T 4 working standard.
  • concentration ofthe working standard should be about 0.2 ⁇ g/ml T 3 and 10.0 ⁇ g/ml T 4 .
  • Assay Preparation Weigh and crash not less than the specified tablet quantity and calculate the average tablet weight. Tare a polypropylene weigh boat.
  • Procedure Separately inject 100 ⁇ l ofthe sample onto the column. Record the responses ofthe analyte peak.
  • NGT 2.0% liothyronine calculated as follows:
  • METHOD B Content Uniformity as Determined by Direct Assay of Active Ingredient: For Levothyroxine Sodium tablets the following procedure is followed. Individually weigh 10 tablets. Place the 10 individual tablets into round bottomed test tubes or flasks ofthe appropriate size as outlined in the chart below. Add the appropriate volume of extraction mobile comprised of water, acetonitrile, and phosphoric acid (65:35::0.05) to each test tube or flask as indicated in the chart below. Note: All test tubes are to be capped with screw on caps and all flasks are to be covered with parafilm as soon as mobile phase is added. Allow to stand at room temperature until the tablet completely crumbles. Secure all samples in a wrist action shaker. Test tubes are to be secured horizontally.
  • Erlenmeyer flasks are to be secured vertically. Set the wrist shaker to the setting specified in the table. Shake sample for 3 minutes. Transfer about 10 ml ofthe sample preparation (or the entirety of smaller samples) to a centrifuge tube. Centrifuge samples for 1 minute at about 3000 rpm. Transfer samples to autosampler vials using disposable Pasteur pipettes. Utilize the HPLC Method for levothyroxine separation (AM-003) for obtaining dosage uniformity, sample area, and standard area results.
  • AM-003 HPLC Method for levothyroxine separation
  • CALCULATIONS Dosage Uniformity Result (% Label Claim)
  • the results confirm an extremely low amount of variability in active material content between the 120 pills tested.
  • the variability for a 120 pill sample should be between about 90 and about 110 % of claimed activity, preferably between about 95 % and about 105%.
  • the RSD for a 120 pill sample should not be greater than 5%, and preferably is less than 3%.
  • the objective of the study was to determine the bioavailability of Levoxyl ® relative to a reference (oral solution) under fasting conditions.
  • the objective of the study was to determine the dosage-form bioequivalence between three different strengths of Levoxyl ® tablets (low, middle and high range).
  • Test Product Dose. Duration. Mode of Administration, and Batch Number:
  • test product was levothyroxine sodium (Levoxyl ® ) 2 X 0.3mg tablets admimstered as a single oral dose.
  • the batch number utilized in this study was TT26.
  • the reference product was levothyroxine sodium (Synthroid ® ) 2 X 500 ⁇ g injection vials
  • Pharmacokinetic assessment consisted ofthe determination of total (bound + free) T4 and T3 concentrations in serum at specified time points following drug administration. From the serum data, the parameters AUC(O-t), Cmax, and Tmax were calculated. Safety:
  • Safety assessment included vital signs, clinical laboratory evaluation (including TSH), physical examination, and adverse events (AEs) assessment.
  • TSH clinical laboratory evaluation
  • AEs adverse events
  • Descriptive statistics (arithmetic mean, standard deviation (SD), coefficient of variation (CV), standard error ofthe mean (SE), sample size (N), mi-oimum, and maximum) were provided for all pharmacokinetic parameters.
  • the effects of baseline and baseline-by treatment interaction were evaluated using a parametric (normal-theory) general linear model (ANCOVA) with treatment, period, sequence, subject within sequence, In(baseline), and interaction between In (baseline) and treatment as factors, applied to the In-transformed pharmacokinetic parameters and Cmax. hi the absence of significant In(baseline) and interaction between In(baseline) and treatment, these parameters were removed from the model.
  • the two one-sided hypotheses were tested at the 5% level of significance for In[AUC(0-t)] and In(Cmax) by constructing 90% confidence intervals for the ratio of Treatment A to Treatment Safety:
  • Frequency counts of all subjects enrolled in the study, completing the study, and discontinuing early were tabulated. Descriptive statistics were calculated for continuous demographic variables, and frequency counts were tabulated for categorical demographic variables for each gender and overall.
  • AEs were coded using the 5 th Edition of the COSTART dictionary. AEs were summarized by the number and percentage of subjects experiencing each coded event. A summary of the total number of each coded event and as a percentage of total AEs was also provided.
  • Laboratory summary tables included descriptive statistics for continuous serum chemistry and hematology results at each time point. Out-of-range values were listed by subject for each laboratory parameter. Descriptive statistics tor vital sign measurements at each time point and change from baseline to each time point were calculated by treatment group. Shifts from screening to post study results for physical examinations were tabulated.
  • Test Product Dose. Duration. Mode of Administration, and Batch Number: Subjects randomized to Treatment A received a single oral dose of 12 X 50 meg levothyroxine sodium (Levoxyl ® ) tablets, Lot No. TT24. Subjects randomized to Treatment B received 6 X 100 meg levothyroxine sodium (Levoxyl ® ) tablets, Lot No.TT25. Subjects randomized to Treatment C received 2 X 300 meg levothyroxine sodium (Levoxyl ® ) tablets, Lot No. TT26. Test products were manufactured by JMI-Daniels, a subsidiary of Jones Pharma Inco ⁇ orated.
  • Pharmacokinetics consisted ofthe detennination of total (bound + free) T4 and
  • T3 concentrations in serum at specified time points following drag administration were calculated.
  • Safety assessment included monitoring of sitting vital signs, clinical aboratory measurements, thyroid-stimulating hormone (TSH), physical examination, electrocardiogram (ECG), and adverse events (AEs).
  • TSH thyroid-stimulating hormone
  • ECG electrocardiogram
  • AEs adverse events
  • Descriptive statistics (arithmetic mean, standard deviation (SD), coefficient of variation (CN), standard error of the mean (SEM), sample size ( ⁇ ), minimum, and maximum) were provided for all pharmacokinetic parameters.
  • SD standard deviation
  • CN coefficient of variation
  • SEM standard error of the mean
  • minimum, and maximum
  • Frequency counts of all subjects enrolled in the study, completing the study, and discontinuing early were tabulated. Descriptive statistics were calculated for continuous demographic variables, and frequency counts were tabulated for categorical demographic variables for each gender and overall.
  • AEs were coded using the 5 th Edition of the COSTART dictionary. AEs were summarized by the number and percentage of subjects experiencing each coded event. A summary of the total number of each coded event and as a percentage of total AEs was also provided. Laboratory summary tables included descriptive statistics for continuous serum chemistry and hematology results at each time point. Out-of-range values were listed by subject for each laboratory parameter. Descriptive statistics for vital sign measurements at each time point and change from baseline to each time point were calculated by treatment group. Shifts from screening to post study results for physical examinations were tabulated.
  • Pharmacokinetic Resulta- T4 The arithmetic means of serum T4 pharmacokinetic parameters for Treatments A and B and the statistical comparison for the In-transformed parameters are summarized in the following table.
  • Pharmacokinetic Results - T3 The arithmetic means of serum T3 pharmacokinetic parameters for Treatments A and B and the statistical comparison for the In-transformed parameters are summarized in the following table. Summary ofthe EarmacoKinetic Parameters of Serum T3 for Treatments A and B
  • the 90% confidence intervals for the comparisons of In(Cmax) and In[AUC(0-t)] for T4 and T3 were within the 80% to 125%) range required for bioequivalence.
  • Comparison of total T4 and T3 pharmacokinetics following administration of 12 X 50 meg Levoxyl ® tablets (Treatment A) and 2 X 300 meg Levoxyl ® tablets (Treatment C) indicated that the two formulations met the requirements for bioequivalence.
  • the 90% confidence intervals for the comparisons of In(Cmax) and In[AUC(0-t)] for T4 and T3 were within the 80% to 125% range required for bioequivalence.
  • test formulations appear to be safe and generally well tolerated when given to healthy adult volunteers.

Abstract

The present invention generally relates to stable pharmaceutical compositions, and methods of making and administering such compositions. In one aspect, the invention features stabilized pharmaceutical compositions that include pharmaceutically active ingredients such as levothyroxine (T4) sodium and liothyronine (T3) sodium (thyroid hormone drugs), preferably in an immediate release solid dosage form. Also provided are methods for making and using such immediate release and stabilized compositions.

Description

LEVOTHYROXINE COMPOSITIONS AND METHODS
U.S. Patent Application
This application for U.S. patent is filed as a utility application under U.S.C., Title 35, §lll(a).
Related U.S. Patent Applications
This application for U.S. patent claims priority to the following U.S. provisional applications, each of which was filed on August 14, 2001: Serial No. 60/312,114 and is entitled Methods of Administering Levothyroxine Pharmaceutical Compositions; Serial No. 60/312,113 and is entitled Levothyroxine Compositions Having Uunique Triiodothyronine Cmax Properties; Serial No. 60/312,287 and is entitled Levothyroxine Compositions Having Unique Triidothyronine Plasma AUC Properties; Serial No. 60/312,184 and is entitled Levothyroxine Compositions Having Unique Cmax Properties; Serial No. 60/312,273 and is entitled Levothyroxine Compositions Having Unique Tmax Properties; Serial No. 60/312,206 and is entitled Levothyroxine Compositions Having Unique Triiodothyronine Tmax Properties; Serial No. 60/312,289 and is entitled Unique Levothyroxine Aqueous Materials.
Field of the Invention
The invention generally relates to stable pharmaceutical compositions, and methods of making and administering such compositions. In one aspect, the invention features stabilized pharmaceutical compositions that include pharmaceutically active ingredients such as levothyroxine (T4) sodium and liothyronine (T3) sodium (thyroid hormone drugs), preferably in an immediate release solid- dosage form. Also provided are methods for making and using such immediate release and stabilized compositions.
Background Thyroid hormone preparations of levothyroxine sodium and liothyronine sodium are pharmaceutical preparations useful to the treatment of hypothyroidism and. thyroid hormone replacement therapy in mammals, for example, humans and dogs.
Thyroid hormone preparations are used to treat reduced or absent thyroid function of any etiology, including human or animal ailments such as myxedema, cretinism and obesity. Hypothyroidism is a common condition. It has been reported in the United States Federal
Register that Hypothyroidism has a prevalence of 0.5 percent to 1.3 percent in adults, h people over 60, the prevalence of primary hypothyroidism increases to 2.7 percent in men and 7.1 percent in women. Because congenital hypothyroidism may result in irreversible mental retardation, which can be avoided with early diagnosis and treatment, newborn screening for this disorder is mandatory in North America. Europe, and Japan.
Thyroid hormone replacement therapy can be a chronic, lifetime endeavor. The dosage is established for each patient Individually. Generally, the initial dose is small. The amount is increased gradually until clinical evaluation and laboratory tests indicate that an optimal response has been achieved. The dose required to maintain this response is then continued. The age and general physical condition ofthe patient and the severity and duration of hypothyroid symptoms determine the initial dosage and the rate at which the dosage may be increased to the eventual maintenance level. It has been reported that the dosage increase should be very gradual in patients with myxedema or cardiovascular disease to prevent precipitation of angina, myocardial infarction, or stroke. It is important that thyroid hormone treatment have the correct dosage. Both under- treatment and over-treatment can have deleterious health impacts. In the case of under-treatment, a sub-optimal response and hypothyroidism could result. Under-treatment has also been reported to be a potential factor in decreased cardiac contractility and increased risk of coronary artery disease. Conversely, over-treatment may result in toxic manifestations of hyperthyroidism such as cardiac pain, palpitations, or cardiac arrhythmia's. In patients with coronary heart disease, even a small increase in the dose of levothyroxine sodium may be hazardous in a particular. Hyperthyroidism is a known risk factor fbr osteoporosis. Several studies suggest that sub clinical hyperthyroidism in premenopausal women receiving thyroid hormone drugs for replacement or suppressive therapy is associated with bone loss. To minimize the risk of osteoporosis, it is preferable that the dose be kept to the lowest effective dose. Because of the risks associated with over-treatment or under-treatment with levothyroxine sodium, there is a need for thyroid hormone products that are consistent in potency and bioavailability. Such consistency is best accomplished by manufacturing techniques that maintain consistent amounts ofthe active moiety during tablet manufacture.
Thyroid hormone drugs are natural or synthetic preparations containing tetraiodothyronine (T , levothyroxine) or triiodothyronine (T __, liothyronine) or both, usually as their pharmaceutically acceptable (e.g. sodium) salts. T 4 and T 3 are produced in the human thyroid gland by the iodination and coupling ofthe amino acid tyrosine. T 4 contains four iodine atoms and is formed by the coupling of two molecules of diiodotyrosine (DIT). T 3 contains three atoms of iodine and is formed by the coupling of one molecule of DIT with one molecule of monoiodotyrosine (MIT). Both hormones are stored in the thyroid colloid as thyroglobulin. Thyroid hormone preparations belong to two categories: (1) natural hormonal preparations derived from animal thyroid, and (2) synthetic preparations. Natural preparations include desiccated thyroid and thyroglobulin.
Desiccated thyroid is derived from domesticated animals that are used for food by man (either beef or hog thyroid), and thyroglobulin is derived from thyroid glands of the hog. The United States Pharmacopoeia (USP) has standardized the total iodine content of natural preparations. Thyroid USP contains not less than (NLT) 0.17 percent and not more than (NMT) 0.23 percent iodine, and thyroglobulin contains not less than (NLT) 0.7 percent of organically bound iodine. Iodine content is only an indirect indicator of true hormonal biologic activity. Synthetic forms for both T 4 and T 3 thyroid hormone are available from a number of producers. For example, liothyronine sodium (T 3) tablets are available under the trademark Cytomel® from King Pharmaceuticals, Inc., St. Louis, Missouri. Levothyroxine sodium (T ) is available under the tradename Levoxyl® from King Pharmaceuticals, Inc., under the tradename Synthroid® from Knoll Pharmaceutical, Mt. Olive, New Jersey, and under the tradename Unithroid® from Jerome Stevens Pharmaceuticals, Bohemia, New York. In addition a veterinarian preparation of levothyroxine soάTum is available under the tradename Soloxine® from King Pharmaceuticals, Inc.
Levoxyl® (levothyroxine sodium tablets,USP) contain synthetic crystalline L-3,3',5,5 '- tetraiodothyronine sodium salt [levothyroxine (T4) sodium]. As indicated above, the synthetic T4 in Levoxyl® is identical to that produced in the human thyroid gland. The levothyroxine (T4) sodium in Levoxyl® has an empirical formula of Cι5H10 L; NNaO4 • H2O, molecular weight of 798.86 g/mol (anhydrous), and a structural formula as shown:
Figure imgf000006_0001
It is well known that the stability of thyroid hormone drugs is quite poor. They are hygroscopic and degrade in the presence of moisture or light, and under conditions of high temperature. The instability is especially notable in the presence of pharmaceutical excipients. such as carbohydrates, including lactose, sucrose, dextrose and starch, as well as certain dyes. The critical nature of the dosage requirements, and the lack of stability of the active ingredients in the popular pharmaceutical formulations, have led to a crisis which has adversely effected the most prescribed thyroid drug products. See, e.g., 62 Fed. Reg. 43535 (Aug. 14, 1997).
It is desirable, therefore, to prepare a stabilized dosage of levothyroxine and liothyronine, which will have a longer shelf life that can be used in the treatment of human or animal thyroid hormone deficiency. U.S. Patent No. 5,22 5,204 (the '204 patent) is directed to improving the stability of levothyroxine sodium. In one embodiment disclosed by '204, stabilized levothyroxine sodium was prepared in a dry state by mixing levothyroxine sodium with a cellulose tableting agent using geometric dilution and subsequently combining this mixture with the same or a second cellulose tableting agent, such as microcrystalline cellulose. Other tableting aids or excipients can be used in this formulation. This '204 patent is incorporated by reference herein, in its entirety. The microcrystalline cellulose disclosed In '204 is AVICEL 101®, AVICEL 102®,
AVICEL 103®, AVICEL 105®, trademarks of FMC Company of Newark, DE., and Microcrystalline Cellulose NF, or EMCQCEL®, a trademark owned by Penwest Pharmaceuticals of Patterson, NY. These microcrystalline cellulose products are prepared by re-slurrylng the cellulose and spray drying the product. This produces an α - helix spherical microcrystalline cellulose product. U. S. Patents 5,955,105 and 6,056,975 (the continuation of '105) disclose pharmaceutical preparations of levothyroxine and microcrystalline cellulose, along with other excipients. The microcrystalline cellulose products used in the '105 and '975 patents were also the α - form Avicel microcrystalline cellulose products. U. S. Patents 5,955,105 and 6,056,975 are incorporated by reference herein, in their entirety. Another microcrystalline cellulose product is a β - sheet form microcrystalline cellulose having a flat needle shape, marketed under the trademark CEOLUS KG801® by FMC Company of Newark, Delaware. The Ceolus® product has different morphology, and different performance characteristics, than those of the Avicel product. The β - sheet microcrystalline cellulose of the present invention is disclosed in U.S. Patent 5,574,150, which is hereby incorporated by reference. Further disclosure relating to β - sheet microcrystalline cellulose is found in InternationalJournal of Pharmaceutics 182 (199) 155 which is hereby incorporated by reference.
The Ceolus® product (β - sheet microcrystalline cellulose) is disclosed by FMC, in its product bulletin dated October 1997, as being suitable for "smaller size tablets" and "exceptional drug carrying capacity." The Ceolus® product was said to provide superior compressibility and drug loading capacity, that still exhibited effective flowability. The examples given in the Ceolus® bulletin were of vitamin C combined with Ceolus® microcrystalline cellulose at levels of from 30 to 45 weight % Ceolus® product in the form of a tablet.
However, there have been problems using the Ceolus® product. For example, at higher levels of Ceolus® product concentration, flow problems were encountered in the process of compressing tablets, and the Ceolus® product was considered unsuitable for compression at higher concentrations than about 45 weight %.
The is a definite need for solid levothyroxine (T4) and/or liothyronine (T3) (thyroid hormone drugs) pharmaceutical compositions, preferably in an immediate release solid dosage form, with the T4 and T3 in the form of their sodium salts that are relatively stable. There is also a need for methods tor making such immediate release and stabilized solid levothyroxine (T4) and/or liothyronine (T3) (thyroid hormone drags) pharmaceutical compositions.
Summary ofthe Invention The present invention overcomes and alleviates the above-mentioned drawbacks and disadvantages in the thyroid drug art through the discovery of novel oral levothyroxine (T4) and/or liothyronine (T3) (thyroid hormone drags) pharmaceutical compositions and methods.
Generally speaking, the present invention relates to stabilized solid as levothyroxine (T4) sodium and/or liothyronine (T3) sodium (thyroid hormone drugs) pharmaceutical compositions and in particular, immediate release, stabilized pharmaceutical compositions that include pharmaceutically active ingredients such as levothyroxine (T4) sodium and/or liothyronine (T3) sodium (thyroid hormone drugs). Preferably but not necessarily, the novel pharmaceutical compositions are provided in a solid dosage form, such as a tablet.
The pharmaceutical compositions of the present invention are useful for, among other things, as replacement or supplemental therapy in hypothyroidism of any etiology, except transient hypothyroidism during the recovery phase of subacute thyroiditis, suppression of pituitary TSH secretion in the treatment or prevention of various types of euthyroid goiters, including thyroid nodules, Hashimoto's thyroiditis, multinodular goiter and, as adjunctive therapy in the management of thyrotropin-dependent well-differentiated thyroid cancer in warm- . blooded animals, especially humans including pediatrics.
The present invention also provides methods for making such immediate release and stabilized levothyroxine (T4) sodium and/or liothyronine (T3) sodium (thyroid hormone drugs) pharmaceutical compositions.
Also in accordance with the present invention, because of the extraordinary release characteristics ofthe preferred compositions, a method of administration to children and patients who have difficulty taking pills, wherein the solid composition having the appropriate dosage is simply put in an aqueous fluid, e.g., juice, where it dissolves in a matter of 1-3 minutes, and the patient can then ingest the fluid, and receive the appropriate dosage, is now made practical.
The present invention has a wide range of important uses including providing pharmaceutically active levothyroxine compositions with enhanced bioavailability, improved shelf life, and more reliable potency. We have discovered immediate release pharmaceutical compositions that include as pharmaceutically active ingredients at least one of levothyroxine and liothyronine, preferably at least one levothyroxine salt, as the major active ingredient. Such preferred immediate release compositions desirably provide at least about 85% (w/v) dissolution of the levothyroxine salt in less than about 20 minutes as determined by standard assays disclosed herein. Suφrisingly, it has been found that by combining the pharmaceutically active ingredients with specific additives in accordance with the invention, it is possible to formulate the compositions so that the ingredients are released almost immediately after ingestion or contact with an aqueous solution, e.g., in a matter of minutes. Preferred invention compositions are stable and provide better shelf life and potency characteristics than prior pharmaceutical compositions.
The immediate release pharmaceutical compositions of the invention provide important uses and advantages. A major advantage is the stability of the active ingredients in the composition. For example, while, as indicated above, prior formulations with sugars, starches, and various types of celluloses, including micro-cellular celluloses such as the Avicel products, have experienced substantial degradation ofthe active ingredients, e.g. T4 sodium. To deal with this problem, pharmaceutical manufacturers have over-formulated the T4-containing pharmaceutical compositions containing such active ingredients, so that the patient can obtain at least the prescribed dosage despite the carbohydrate-induced instability of the active ingredient. However, the patient who obtains the pharmaceutical immediately after it is made, receives an over-dosage of the active compound; whereas, the patient who has received the pharmaceutical after it has sat on the pharmacy shelf for an extended period, will receive an under-dosage ofthe active ingredient. In either case, the patient receives the wrong dosage, with possible serious consequences.
In sharp contrast, it has been surprisingly found that the use of the β-sheet microcrystalline cellulose in the compositions of the present invention substantially increase the stability of the thyroid hormone drugs, so that the patient obtains consistent potency over an extended shelf life, compared to prior thyroid hormone drug products. In this application, the term "stabilized", as applied to levothyroxine and/or liothyronine means that the loss of potency over the shelf life ofthe product is less than about 0.7 % potency per month, for at least about 18 months. Preferred compositions have a loss of potency of less than about 0.5% per month for sucn a period, and more preierreα compositions nave a loss ol potency ot less than about 0.3% per month for such a period.
Further, the compositions of the invention provide favorable pharmacokinetic characteristics when compared to prior formulations. In particular, the immediate release pharmaceutical compositions that include levothyroxine salt have are more quickly available for absorption by the gastrointestinal (GI) tract faster and are absorbed more completely than has heretofore been possible. This invention feature substantially enhances levothyroxine bioavailability, thereby improving efficacy and reliability of many standard thyroid hormone replacement strategies. Additionally, the desirable immediate release characteristics of the present invention facilitate dosing of patients who may be generally adverse to thyroid hormone replacement strategies involving solid dosing. More specifically, immediate release pharmaceutical compositions disclosed herein can be rapidly dissolved in an appropriate aqueous solution (e.g., water, saline, juice) or colloidal suspension (e.g., baby formula or milk) for convenient administration to such patients. Illustrative of such patients include infants, children, and adults who may experience swallowing difficulties. The invention thus makes standard thyroid hormone replacement strategies more flexible and reliable for such patients.
Accordingly, and in one embodiment, the invention features an immediate release pharmaceutical composition comprising at least one levothyroxine salt, preferably one of such a salt. At least about 80% ofthe levothyroxine dissolves in aqueous solution in less than about 20 minutes as determined by a standard assay, disclosed herein. Preferably, at least about 80% of the levothyroxine is dissolved in the aqueous solution by about 15 minutes from the time that the composition, in pill form, is placed in the aqueous solution. More preferably, at least about 85% ofthe levothyroxine is released to the aqueous solution by about 10 minutes, most preferably by about 5 minutes after exposure of the composition to the aqueous solution. As shown below, compositions in accordance with the present invention can be formulated to release 85% of the levothyroxine within 2-3 minutes after exposure to the aqueous solution.
It has been found that by combining one or more of the pharmaceutically active agents with β-form microcrystalline cellulose, it is possible to produce compositions with favorable immediate release characteristics. Without wishing to be bound to theory, it is believed that the agents do not bind well to certain grades of the β - sheet form microcrystalline cellulose. More o± me agent is thus available for immediate release, m contrast, it is believed that many prior formulations have active agents that bind cellulose additives, making less available. The release characteristics ofthe compositions ofthe invention are also improved by the use of other agents, as discussed further below. Thus in one embodiment, the present invention relates to a stabilized pharmaceutical composition comprising a pharmaceutically active ingredient, such as levothyroxine, and the β - sheet form of microcrystalline cellulose, in the form of a solid dosage. More specifically, the present invention relates to a stabilized pharmaceutical composition comprising a pharmaceutically active ingredient, such as levothyroxine sodium and/or liothyronine sodium, at least about 50 weight % ofthe dosage weight composed ofthe β - sheet form of microcrystalline cellulose, and, optionally, additional excipients, in a solid dosage form.
In another aspect, the invention provides an aqueous solution or colloidal suspension that includes at least one ofthe compositions of this invention, preferably between from about one to about five of same, more preferably about one of such compositions. It has also been found that β - sheet microcrystalline cellulose grades having preferred bulk densities provide for more compact processing than use of other celluloses. That is, use of the β - sheet microcrystalline cellulose having bulk densities in accord with this invention helps to provide for higher compression ratios (initial volume/final volume). As discussed below, other invention aspects help reduce or avoid production of damaging compression heat that has damaged prior formulations made from high compression ratios. The compositions of the present invention generally also require less compressional force to form the tablets.
Accordingly, the invention also provides methods for making an immediate release pharmaceutical composition comprising at least one levothyroxine salt, preferably one of such a salt. In one embodiment, the method includes at least one and preferably all of the following steps: a) mixing a levothyroxine salt with microcrystalline β-cellulose and preferably a crosscarmellose salt to make a blend; and
b) compressing the blend in a ratio of initial volume to final volume of between from about 2:1 to about 5:1 to make the composition, preferably about 4: 1. In one embodiment, the method involves preparing an oral dosage form of a pharmaceutically active ingredient comprising dry blending the pharmaceutically active ingredient and at least about 50 weight % ofthe β - sheet form of microcrystalline cellulose, and compressing the blend to form a solid dosage.
These and other objects, features, and advantages of the present invention may be better understood and appreciated from the following detailed description of the embodiments thereof, selected for purposes of illustration and shown in the accompanying figures and examples. It should therefore be understood that the particular embodiments illustrating the present invention are exemplary only and not to be regarded as limitations ofthe present invention.
Brief Description of the Figs.
The foregoing and other objects, advantages and features of the invention, and the manner in which the same are accomplished, will become more readily apparent upon consideration of the following detailed description of the invention taken in conjunction with the accompanying Figs., which illustrate a preferred and exemplary embodiment, wherein:
Figures 1A - 1C illustrate various solid dosage forms such as cylindrical tablets and raised violin shaped tablets;
Figure 2 illustrates a tableting die pair; Figure 3 pair; is graphical depiction of comparative dissolution data of various strengths of Levoxyl® tablets made in accordance with the invention.
Figure 4A is an HPLC chromatogram showing a levothryoxine and liothyronine standards.
Figure 4B is an HPLC chromatograph showing results of levothyroxine sodium sample made in accordance with the present invention.
Figure 5A is a chromatogram showing various levothryoxine impurity standards. Figure 5B is a chromatograph showing results of levothyroxine sodium sample made in accordance with the present invention. Detailed Description By way of illustrating and providing a more complete appreciation of the present invention and many of the attendant advantages thereof, the following detailed description is given concerning me novel oral levothyroxine (T4) and or liothyronine (T3) (thyroid hormone drugs) pharmaceutical compositions and methods for use in warm-blooded animals, especially humans and children.
As discussed, the invention relates to immediate release solid pharmaceutical compositions such as stabilized pharmaceutical compositions that include pharmaceutically active ingredients such as levothyroxine (T4) sodium and liothyronine (T3) sodium (thyroid hormone drugs), preferably in a solid dosage form. Also provided are methods for making such immediate release and stabilized compositions.
Aspects ofthe present invention have been disclosed in U.S. Provisional Application No. 60/269,089, entitled Stabilized Pharmaceutical and Thyroid Hormone Compositions and Method of Preparation and filed on February 15, 2001 by Franz, G.A. et al. The disclosure of said provisional application is incorporated herein by reference.
By the phrase "immediate release" is meant a pharmaceutical composition in which one or more active agents therein demonstrates at least about 80% (w/v) dissolution, preferably between from about 90% (w/v) to about 95% (w/v), more preferably about 95% (w/v) to about
99% (w/v) or more within 15 to 20 minutes as determined by a standard dissolution test.
Suitable standard dissolution tests are known in the field. See FDA, Center for Drug Research,
Guidance for Industry, In Vivo Pharmacokinetics and Bioavailability Studies and In Vitro
Dissolution Testing for Levothyroxine Sodium Tablets, available at www.fda.gov/cder/guidance/index.htm. A specifically preferred dissolution test is provided in
Example 2, below.
A pharmaceutical composition of the invention is "stable" or "stabilized" if one or more ofthe active agents therein exhibit good stability as determined by a standard potency test. More specifically, such compositions exhibit a potency loss of less than about 15%, preferably less than about 10%, more preferably less than about 1% to about 5% as determined by the test.
Potency can be evaluated by one or a combination of strategies known in the field. See the USP.
A preferred potency test compares loss or conversion of the active agent in the presence
(experimental) or absence (control) of a carrier or excipient. A specifically preferred potency test is provided in Examples 1 and 3, below. In preferred embodiments, the pharmaceutical compositions of the invention include, as active agent, levothyroxine (T4), preferably a salt thereof such as levothyroxine sodium USP. Such compositions typically exhibit a levothyroxine (T4) plasma Cmax of between from about 12 μg/dl to about 16 μg/dl, preferably as determined by the standard Cmax test. Preferably, the In(Cmax) ofthe levothyroxine (T4) plasma level is between from about 1 to about 3.
The standard Cmax test can be performed by one or a combination of strategies known in the field. See e.g., the USP. A preferred Cmax test is disclosed below in Examples 8 and 9.
Additionally preferred compositions in accord with the invention provide a triiodothyronine (T3) plasma Cmax of between from about 0.1 ng/ml to about lOng/ml, preferably 0.5 ng/ml to about 2ng/ml, as determined by the standard Cmax test. Typically, the In(Cmax) is between from about 0.01 to about 5. See Examples 8 and 9 for more information. Further preferred compositions exhibit a levothyroxine (T4) plasma Tmax of between from about 0.5 hours to about 5 hours, preferably as determined by a standard Tmax test. The standard Tmax test can be performed by procedures generally known in the field. See e.g., the USP. A preferred Tmax test is disclosed below in Examples 8 and 9.
Still further preferred compositions of the invention exhibit a triiodothyronine (T3) plasma Tmax of between from about 10 hours to about 20 hours, preferably about 12 to about 16 hours as determined by the standard Tmax test.
Additionally preferred invention compositions feature a levothyroxine (T4) plasma AUC (0-t) of between from about 450 μg-hour/dl to about 600 μg-hour/dl, preferably 500 μg-hour/dl to about 550 μg-hour/dl as determined by a standard AUC (0-t) test. Preferably, the In[AUC(0- t)] is between from about 1 to about 10.
Standard methods for performing AUC (0-t) test determinations are generally known in the field. See e.g., the USP. Examples 8 and 9 below provide a specifically preferred method of determining the AUC (0-t).
Further preferred invention compositions feature a triiodothyronine (T3) AUC (0-t) of between from about 10 ng-hour/ml to about 100 ng-hour/ml, preferably 20 ng-hour/ml to about 60 ng-hour/ml, as determined by the standard AUC (0-t) test. Preferably, the In[AUC(0-t)] is between from about 1 to about 5.
As will be appreciated, many prior pharmaceutical formulations include lactose or other sugars as a pharmaceutically acceptable carrier. It has been found however, that sugars such as lactose can react with active agents including the levothyroxine (T4) compositions ofthe present invention. For example, and without wishing to be bound to theory, it is believed that lactose is particularly damaging to T4 and T3 molecules via Schϊiϊ reactions. The invention address this problem by providing compositions that are essentially sugar-free. Particular invention compositions are essentially free of lactose.
Additionally, preferred pharmaceutical compositions of the invention are provided in which the active material is a non-granulated material. Prior levothyroxine compositions have been granulated in various size reduction machines to grains of less than, e.g., 5 - 20 microns average particle size in order to be effectively incorporated into the administrable pharmaceutical composition. The granulation process subjects the active material to degrading heat, which can have adverse effects on the active material, as well as reducing the activity level. Prior manufacturers purchase micronized levothyroxine manufactured under DMF No. 4789, and then granulate it before incorporating it into the levothyroxine pharmaceutical product.
In the preferred method ofthe present invention, the raw material is not granulated before incorporation into the pharmaceutical composition. Rather, the ingredients of the preferred pharmaceutical are mixed and the mixture is subjected to direct compression to form the pharmaceutical tablets of appropriate dosage. As a result, the activity of the active ingredient is not degraded prior to the direct compression step. Bulk levothyroxine is obtained in a fine powdered form, preferably from Biochemie GmbH, A-6250 Kundl, Austria. More importantly, the use of the preferred process results in a product which is immediately dispersible in aqueous solution, to make the active ingredient available for absorption in the body. As used in this application, "non-granulated" means that the bulk USP compound is used without subjecting it to granulators or similar high energy size reduction equipment before being mixed with the other pharmaceutical components and formed into the appropriate pill. Preferably, the bulk active ingredient is mixed with the appropriate amounts of other ingredients and directly compressed into pill form. Since it is not necessary to granulate the material, it is not necessary to subject it to degrading temperatures in the process of forming the pharmaceutical compositions containing the active materials. In the present process we start with micronized active material, which merely needs to be blended with the B and other materials and then compressed. Others have to be granulated, and then dried, which steps interfere with the dissolution of the active material. The drying temperatures employed in manufacturing other active ingredients can cause degradation of the levothyroxine, as experienced in other available thyroxine. It has been found that providing the invention compositions in a non-granulated format helps to reduce or eliminate active agent degradation, presumably by facilitating a reduction in friction, and thus degrading heat, during compression ofthe compositions into pills.
Practice of the invention is compatible with several β-form microcrystalline cellulose grades. Preferably, the β-foπn microcrystalline cellulose has a bulk density of between from about 0.10 g/cm3 to about 0.35 g/cm3, more preferably between from about 0.15 g/cm3 to about
0.25 g/cm3, still more preferably between from about 0.17 g/cm3 to about 0.23 g/cm3, most preferably between from about 0.19 g/cm to about 0.21 g/cm .
Further preferred grades of the β-form microcrystalline cellulose are substantially non- conductive. Preferably, the β-form microcrystalline cellulose has a conductivity of less than about 200 μS/cm, more preferably, less than about 75 μS/cm, still more preferably between from about 0.5 μS/cm to 50 μS/cm, most preferably between from about 15 μS/cm to 30 μS/cm.
A specifically preferred β-form microcrystalline cellulose is sold by Asahi Chemical Industry Co., Ltd (Tokyo, Japan) as Ceolus (Type KG-801 and or KG-802).
Additionally preferred compositions of the invention have a post-packaging potency of between from about 95% to about 120%, preferably 98% to about 110% as determined by the standard potency test.
The present invention is a pharmaceutical product that is in the form of a solid dosage, such as a sublingual lozenge, buccal tablet, oral lozenge, suppository or a compressed tablet. The pharmaceutically active ingredient is dry mixed with the β-form of the microcrystalline cellulose, optionally with additional excipients, and formed into a suitable solid dosage.
Preferred tablets according to the invention have a total hardness of between from about 1 to about 30 KP, preferably about 6 to about 14 KP as determined by a standard hardness test. Methods for determining tablet hardness are generally known in the field. See e.g., the USP. A preferred standard hardness test is disclosed below in Example 4. Additionally preferred pharmaceutical compositions including those in tablet format preferably include less than about 10% total impurities, more preferably less than about 5% of same as determined by a standard impurity test.
Reference herein to the "standard impurity test" means a USP recognized assay for detecting and preferably quantitating active drug degradation products. In embodiments in which levothyroxine or liothyronine break-downs are to be monitored, such products include, but are not limited to, at least one of diiodothyronine (T2), triiodothyronine (T3), levothyroxine, tπiodothyroacetic aci " amide, triϊodothyroethyiamine, triiodothyroacetic acid, triiodothyroethyl alcohol, tetraiodothyroacetic acid amide, tetraiodothyroacetic acid, triiodothyroethane, and tetraiodothyroethane. Of particular interest are diiodothyronine (T2), triiodothyronine (T3), triiodothyroacetic acid, and tetraiodothyroacetic acid impurities. A preferred impurity test for monitoring levothyroxine and liothyronine breakdown products involves liquid chromatography (LC) separation and detection, more preferably HPLC. Specifically preferred impurity tests are provided below in Examples 5 and 6
Further preferred compositions in accord with the invention include one or more standard disintegrating agents, preferably crosscarmellose, more preferably a salt of same. Still further preferred compositions include a pharmaceutically acceptable additive or excipient such as a magnesium salt.
The present invention can be prepared as a direct compression formula, dry granulation formula, or as a wet granulation formula, with or without preblending of the drug, although preferably with preblending. The pharmaceutically active ingredient can be any type of medication which acts locally in the mouth or systemically, which is the case of the latter, can be administered orally to transmit the active medicament into the gastrointestinal tract and into the blood, fluids and tissues of the body. Alternatively, the medicament can be of any type of medication which acts through the buccal tissues of the mouth to transmit the active ingredient directly into the blood stream thus avoiding first liver metabolism and by the gastric and intestinal fluids which often have an adverse inactivating or destructive action on many active ingredients unless they are specially protected against such fluids as by means of an enteric coating or the like. The active ingredient can also be of a type of medication which can be transmitted into the blood circulation through the rectal tissues. Representative active medicaments include antacids, antimicrobials, coronary dilators, peripheral vasodilators, anti psychotropics, antimanics, stimulants, antihistamines, laxatives, decongestants, vitamins, gastrosedatives, antidiarrheal preparations, vasodilators, antiarrythmics, vasoconstrictors and migraine treatments, anticoagulants and antithrombotic drugs, analgesics, antihypnotics, sedatives, anticonvulsants, neuromuscular drugs, hyper and hypoglycemic agents, thyroid and antithyroid preparations, diuretics, antispasmodics, uterine relaxants, mineral and nutritional additives, antiobesity drugs, anabolic drugs, erythropoietic drugs, antiasthematics, expectorants, cough suppressants, mucolytics, antiuncemic drugs, and- drugs or substances actmg locally in the mouth.
Typical active medicaments include gastrointestinal sedatives such as metoclopramide and propantheline bromide, antacids such as aluminum trisilicate, aluminum hydroxide and cimetidine, asprin-like drugs such as phenylbutazone, indomethacin, and naproxen. ibuprofen, flurbiprofen, diclofenac, dexamethasone, prednisone and prednisolone, coronary vasodialator drugs such as glyceryl trinitrate, isosorbide dinitrate and pentaerythritol tetranitrate, peripheral and cerebral vasodilators such as soloctidilum, vincamine, naftidrofuryl oxalate, comesylate, cyclandelate, papaverine and nicotinic acid, antimicrobials, such as erythromycin stearate, cephalexin, nalidixic acid, tetracycline hydrochloride, ampicillin, flucolaxacillin sodium, hexamine mandelate and hexamine hippurate, neuroleptic drugs such as fluazepam, diazepam, temazepam, amitryptyline, doxepin, lithium carbonate, lithium sulfate, chlorpromazine, thioridazine, trifluperazine, fluphenazine, piperothiazine, haloperidol, maprotiline hydrochloride, imipramine and desmethylimipramine, central nervous stimulants such as methylphenidate, ephedrine, epinephrine, isoproterenol, amphetamine sulfate and amphetamine hydrochloride, anitidrags such as diphenylhydramine, diphenylpyramine, chloφheniramine and brompheniramine, antidiarrheal drags such as bisacodyl and magnesium hydroxide, the laxative drag, dioctyl sodium sulfosuccinate, nutritional supplements such as ascorbic acid, alpha tocopherol, thiamine and pyridoxine, antispasmotics such as dicyclomine and diphenoxylate, drags effecting the rhythm of the heart such as verapamil, nifedepine. diltiazem, procainamide, disopyramide, bretylium tosylate, quinidine sulfate and quinidine gluconate, drags used in the treatment of hypertension such as propranolol hydrochloride, guanethidine monosulphate, methyldopa, oxprenolol hydrochloride, captopril, Actace and hydralazine, drags used in the treatment of migraine such as ergotamine, drags effecting coagulability of blood such as epsilon aminocaproic acid and protamine sulfate, analgesic drags such as acetylsalicyclic acid, acetaminophen, codeine phosphate, codeine sulfate, oxycodone, dihydrocodeine tartrate, oxydodeinone, morphine, heroin, nalbuphine, butoφhanol tartrate, pentazocine hydrochloride, cyclazacine, pethidine, buprenorphine, scopolamine and mefenamic acid, antidrugs such as phenytoin sodium and sodium valproate, neuromuscular drags such as dantrolene sodium, substances used in the treatment of diabetes, such as tolbutamide, diabenase glucagon and insulin, drags used in the treatment of thyroid gland dysfunction such as triiodothyronine, liothyronine sodium, levothyroxilne sodium and related compounds, and propylthiouracil, diuretic drugs, such as furosemide, chlorthalidone, hydrochlorthiazide, spironolactone and triampterene, the uterine relaxant drugritodrine, appetite suppressants such as fenfluramine hydrochloride, phentermine and diethylproprion hydrochloride,antidrags stimulants such as aminophylline, theophylline, salbutamol, orciprenaline sulphate and terbutaline sulphate, expectorant drag such as guaiphenesin, cough suppressants such as dextromethorphan and mescaline, mucolytic drugs such as carbocisteine, antiseptics such as cetylpyridinium chloride, tyrothricin and chlorhexidine, decongestant drags such as phenylpropanolamine and pseudoephedrine, hypnotic drags such as dichloralphenazone and nitrazepam,antidrags Hi blockers such as promethazine theociate, haemopoetic drags such as ferrous sulphate, folic acid and calcium gluconate, uricosuric drags such as sulphinpyrazine, allopurinol and probenecid and the like. It is understood that the invention is not restricted to the above medications.
The amount of pharmaceutically active ingredient in the present composition can vary widely, as desired. Preferably, the active ingredient is present in a composition of the present invention in an effective dosage amount. Exemplary of a range that the active ingredient may be present in a composition in accordance with the present invention is from about 0.000001 to about 10 weight %. More preferably, the amount of active ingredient is present in the range of about 0.001 to 5 weight %.
Of course, it should be understood that any suitable pharmaceutically acceptable form of the active ingredient can be employed in the compositions of the present invention, i.e., the free base or a pharmaceutically acceptable salt thereof, e.g., levothyroxine sodium salt, etc.
When the pharmaceutically active moiety is levothyroxine sodium, the preferred amount ofthe active moiety in the composition is present in the range of about 0.00005 to about 5 weight %. The more preferred range is from about 0.001 to about 1.0 weight %, and the most preferred range is from about 0.002 to about 0.6 weight % levothyroxine. The minimum amount of levothyroxine can vary, so long as an effective amount is utilized to cause the desired pharmacological effect. Typically, the dosage forms have a content of levothyroxine in the range of about 25 to 300 micrograms per 145 milligram pill for human applications, and about 100 to 800 micrograms per 145 mg pill for veterinary applications. In accordance with the present invention, a goal of levothyroxine replacement therapy is to achieve and maintain a clinical and biochemical euthyroid state, whereas a goal of suppressive therapy is to inhibit growth and/or function of abnormal thyroid tissue. A dose of levothyroxine that is adequate to achieve these goals depends of course on a variety of factors including the patient's age, body weight, cardiovascular status, concomitant medical conditions, including pregnancy, concomitant medications, and the specific nature ofthe condition being treated. Hence, the following recommendations serve only as dosing guidelines. It should be understood by those versed in this art that dosing should be individualized and adjustments made based on periodic assessment of a patient's clinical response and laboratory parameters.
Preferably, but not necessarily, when using levothyroxine to treat, it should be taken in the morning on an empty stomach, at least one-half hour before any food is eaten. In addition, levothyroxine is preferably taken at least about 4 hours apart from drags that are known to interfere with its absorption.
Due to the long half-life of levothyroxine, the peak therapeutic effect at a given dose of levothyroxine sodium may not be attained for about 4 to about 6 weeks.
In people older than 50 years, who have been recently treated for hyperthyroidism or who have been hypothyroid for only a short time (such as a few months), the average full replacement dose of levothyroxine sodium is approximately 1.7 mcg/kg/day (e.g., about 100 to about 125 meg/day for a 70 kg adult). Older patients may require less than 1 mcg/kg/day. Levothyroxine sodium doses greater than about 200 meg/day may or may not be required.
For most patients older than 50 years or for patients under 50 years of age with underlying cardiac disease, an initial starting dose of about 25 to about 50 meg/day of levothyroxine sodium is recommended, with gradual increments in dose at about 6 to about 8 week intervals, as needed. The recommended starting dose of levothyroxine sodium in elderly patients with cardiac disease is about 12.5 to about 25 meg/day, with gradual dose increments at about 4 to about 6 week intervals. The levothyroxine sodium dose is generally adjusted in about 12.5 to about 25 meg increments until the patient with primary hypothyroidism is clinically euthyroid and the serum TSH has normalized.
In patients with severe hypothyroidism, the recommended initial levothyroxine sodium dose is about 12.5 to about 25 meg/day with increases of about 25 meg/day about every 2 to about 4 weeks, accompanied by clinical and laboratory assessment, until the TSH level is normalized. In patients with secondary (pιtuιtary)or tertiary (hypothalamic) hypothyroidism, the levothyroxine sodium dose should be titrated until the patient is clinically euthyroid and the serum free-T4 level is restored to the upper half of the normal range.
In children, levothyroxine therapy may be instituted at full replacement doses as soon as possible. Levothyroxine compositions ofthe present invention may be administered to infants and children who cannot swallow intact tablets by crashing the tablet and suspending the freshly crushed tablet in a small amount (5-10 mL or 1-2 teaspoons) of water. This suspension can be administered by spoon or dropper. Foods that decrease absoφtion of levothyroxine, such as soybean infant formula, should not be used for administering levothyroxine sodium tablets. A recommended starting dose of levothyroxine sodium in newborn infants is about 10 to about 15 mcg/kg/day. A lower starting dose (e.g., about 25 meg/day) may be considered in infants at risk for cardiac failure, and the dose should be increased in 4-6 weeks as needed based on clinical and laboratory response to treatment, hi infants with very low (< about 5 mcg/dL) or undetectable serum T4 concentrations, a recommended initial starting dose is about 50 meg/day of levothyroxine sodium.
As indicated above, levothyroxine therapy is usually initiated at full replacement doses, with the recommended dose per body weight decreasing with age (see Dose Table below). However, in children with chronic or severe hypothyroidism, an initial dose of about 25 meg/day of levothyroxine sodium is recommended with increments of 25 meg every 2-4 weeks until the desired effect is achieved. Hyperactivity in an older child may be minimized if the starting dose is one-fourth ofthe recommended full replacement dose, and the dose is then increased on a weekly basis by an amount equal to one-fourth the full-recommended replacement dose until the full recommended replacement dose is reached.
Figure imgf000021_0001
Figure imgf000022_0001
Levothyroxine sodium tablets, USP, in accordance with the present invention may be supplied as oval or violin-shaped, color-coded, potency marked tablets in, for example, 12 strengths as indicated in the Strength Table
Figure imgf000022_0002
When the pharmaceutically active moiety is liothyronine sodium, the preferred amount of the active moiety in the composition is present in the range of about 0.000005 to 0.5 weight %. The more preferred range is from about 0.00001 to 0.1 weight %, and the most preferred range is from about 0.00004 to about 0.002 weight % liothyronine. The minimum amount of lyothyronine can vary, so long as an effective amount is utilized to cause the desired pharmacological effect. Typically, the dosage forms have a content of levothyroxine in the range of about 5 to 50 micrograms per 145 milligram pill for human applications.
The β-form microcrystalline cellulose product of the present invention is prepared by forming a wet cake, drying the cake with a dram dryer, then passing the dried product through a screen or mill tor sizing which produces a β-sheet microcrystalline cellulose which has a flat needle shape, as disclosed in U.S. Patent 5,574,150. Such β-sheet microcrystalline product is available from Asahi Chemical of Japan and/or marketed by FMC Company of Newark, Del, under the trademark Ceolus®. The moφhology and performance characteristics of the Ceolus® product are different from those of α - form microcellulose products (for example, Avicel® and Emcocel®), and are suitable for preparing the present stabilized pharmaceutical composition.
The amount of β-form microcrystalline product used in the present composition is at least 50 weight % ofthe final composition. Preferably, the amount of β-fonn microcrystalline product is in the range of about 50 to 99 weight %. Most preferably, the amount of β-fonn microcrystalline product is in the range of about 60 to 90 weight % ofthe final composition.
Other suitable excipients for the present invention include fillers such as starch, alkaline inorganic salts such as trisodium phosphate, tricalcium phosphate, calcium sulfate and sodium or magnesium carbonate. The fillers can be present in the present composition in the range of about 0 to 50 weight %. Suitable disintegrating agents include corn starch, cross-linked sodium carboxymethylcellulose (crosscarmellose) and cross-linked polyvinyipyrrolidone (crospovidone). A preferred disintegrating agent is crosscarmellose. The amount of disintegrating agent used is in the range of about 0 to 50 weight %. Preferably, the disintegrating agent is in the range of about 5 to 40 weight %, more preferably about 10 to about 30 weight %. This is in substantial excess of the recommended levels of such materials. For example, the recommended loading of crosscarmellose is 0.5 to about 2%> by weight. However, it has been found that the higher loadings of the disintegrating agents substantially improves the ability ofthe product to disperse in aqueous media.
Suitable gildents for use in the present invention include colloidal silicon dioxide and talc. The amount of gildent in the present composition is from about 0 to 5 weight %, and the preferred amount is about 0 to 2 weight %.
Suitable lubricants include magnesium and zinc stearate. sodium stearate fumarate and sodium and magnesium lauryl sulfate. A preferred lubricant is magnesium stearate. The amount of lubricant is typically in the range of about 0 to 5 weight %>, preferably in the range of about 0.1 to 3 weight %. The oral pharmaceutical product is prepared by thoroughly mtermixing the active moiety and the β-form of microcrystalline cellulose, along with other excipients to form the oral dosage. Food grade dyes can also be added. For example, it is common to distinguish dosages of various potency by the color characteristics of such dyes. As discussed, a preferred immediate release pharmaceutical composition in tablet form includes levothyroxine sodium, hi a preferred embodiment, the composition includes at least one of, preferably all ofthe following:
a) between from about 0.01 mg/tablet to about 500 mg/tablet levothyroxine sodium (USP), b) between from about 100 mg/tablet to about 110 mg/tablet of microcrystalline β-cellulose, NF (Ceolus) having a bulk density of between from about 0.10 g/cm3 to about 0.35 g/cm3, c) between from about 25mg/tablet to about 50mg/tablet of crosscarmellose sodium, NF (Ac-di-sol); and d) between from about 0.5 mg/tablet to about 5mg/tablet of magnesium stearate, NF.
Preferably, the composition further comprises at least one pharmaceutically acceptable coloring agent.
More particular methods according to the invention provide compositions having less than about 5% total impurities as determined by the standard impurity test. Preferably, the method further comprises forming a tablet, particularly those tablets having a raised violin configuration. The stabilized oral dosages of thyroid hormone are prepared by forming a trituration of the active moiety (i.e. levothyroxine sodium and/or liothyronine sodium) and β-form microcrystalline cellulose. The trituration is blended with β-form microcrystalline cellulose and additional excipients and compressed into oral dosages.
Design of the tableting apparatus is important, in order to maintain consistency from one oral dosage to the next. The formulation batches are a blend of solid compositions of various shapes and sizes. Blending is used to achieve a measure of homogeneity. In particular the active thyroid moiety is desired to be evenly distributed throughout the batch, hi a typical 410 kg batch, the amount of active moiety represents less than 1 kg of the total weight. For example, when producing 145 mg tablets with a 300 meg dosage, approximately 0.8 kg of a 410 kg batch is the active moiety. In addition each tablet is formulated to contain 100% label claim potency. It is typical for compressible medicament tablets to be formed using a 2:1 fill to compression ratio. However, for medicament tablets formed using the present invention a fill to compression ratio from 3.3:1 to 4:1 is needed to obtain desired tablet density. The β-form microcrystalline cellulose has a lower bulk density, as compared to other excipients.
Higher tablet density can be accomplished by adjusting a tableting machine to increase the compression ratio. Tableting machines are commonly known to practitioners in the art and include those available from Manesty and Stokes. It has been found that making such adjustments to the compression ratio results in poor tablet surface finish as well as inconsistent tablet weights. Instead, the design of the tableting dies should be adjusted. It has been determined that during the filling ofthe tableting dies, a minimum of 5-6mm die overfill. In most cases this requires replacement of the usual tableting dies with dies which are an additional 2-3 mm deep.
When using the extra-deep dies and a compression ratio of from 3.3:1 to 4.0:1, consistent weight tablets with good surface finish were produced.
Preferably, the shape of the tablet is configured to increase heat transfer away from the tablet. More preferred tablets have a surface area per tablet of between from about 0.9 in.2 to about 0.15 in.2, preferably about 0.115in.2, to assist such heat transfer. Additional tablet configurations are contemplated e.g., tablets that are beveled and/or include a notch. A preferred tablet shape is a raised violin configuration, as shown in Figure lC.
The following examples are given by way of illustration only and are not to be considered limitations of this invention or many apparent variations of which are possible without departing from the spirit or scope thereof. Example 1
Stability Tests Stability testing was performed on samples ofthe thyroid hormone drug formulation used in manufacturing tablets with an active moiety of levothyroxine sodium. Tests were performed on direct compression formulations for dosage strength of 25 meg. Example 1 tablets comprise the β-fόrm microcrystalline cellulose while Control 1 tablets comprise the traditional -for m microcrystalline cellulose. The composition of Example 1 and Control 1 tablets are presented in Table 1 and stability test results in Table 2:
Table 1 - Tablet Formulation for 25mcg Dosages of Levothyroxine Sodium
Figure imgf000026_0001
Table 2 - Stability Test - Potency at25 ° C- % Label Claim
Figure imgf000026_0002
As seen in Table 2, the stability of pharmaceutical formulations of the present invention is improved significantly by the use of the β - sheet microcrystalline cellulose. Potency loss of the present invention after 15 months is 3.5 %, versus 16.0 %> potency loss experienced in a similar formulation with the α-form microcrystalline cellulose. The average loss in potency per month in the case of the compositions of the present invention was only about 0.2 % per month, as compared to over 1% per month for the T4 products which included β-form microcrystalline cellulose, thus demonstrating a stability which is about 3 to 4 times better than the T4 products which utilized α-form microcrystalline cellulose.
Tableting testing was performed on the formulation for Example 1 tablets. Initial results with standard die depths provided a relative standard deviation of 2.2 to 3.5% tablet weight. With the use of the herein described extra deep tablet dies, the- relative standard deviation is 1.2%. Testing was performed on a Manesty tableting machine with compression ratios of from 3.3:1 to 4.0:1.
Tablet quality is also dependent upon the storage of the β-sheet microcrystalline cellulose. Best results are achieved when the cellulose is received in drums or portable containers instead of bags. The bag form suffers from compression during transportation from raw material suppliers. Test results for tableting are presented in attached Exhibit A.
Additional examples of solid dosage formulations are illustrated in Tables 3 and 4. Stability testing data of additional examples are illustrated in Table 5.
Table 3 - Tablet Formulation for Dosages of Levothyroxine Sodium (per tablet)
Figure imgf000027_0001
Table 4 - Tablet Formulation for Dosages of Levothyroxine Sodium (per tablet)
100 meg Dosage 112 meg Dosage 1300 meg Dosage Component
Figure imgf000028_0001
Table 5 shows drag stability data for a number ofthe above formulations:
Table 5 - Stability Test - Potency at25 ° C - % Label Claim
Figure imgf000028_0002
Thus the formulations of the present invention provide extreme stability for the levothyroxine activity over an extended shelf life for these pharmaceutical products. Example 2
Dissolution Tests
The following preferred method for testing potency will sometimes be referred to herein s method number: AM-004B
Table 6 - Dissolution Test Procedure
Figure imgf000029_0001
Figure imgf000030_0001
Calculations: Sample Area x 798.86 x Amt. Std. Injected x 100% = % Dissoluted
% Dissoluted Std. Area 776.87 Amt. Samp. Injected
Where 798.86 = molecular weight of Levothyroxine as Sodium Salt
776.87 = molecular weight of Levothyroxine (as Base)
Table 7— Acceptance Criteria
Figure imgf000030_0002
Table 8 shows comparative dissolution data for all strengths of Levoxyl® tablets.
Table 8 - Comparative Dissolution Data
Figure imgf000030_0003
300 meg Dτσ%~ 74.5% 92.1% 91.4% 87.9% 84.0%
Figure 4 depicts graphs showing the mean results for each of the tablet strengths of Levoxyl® tested. Each point is the mean of three dissolutions, testing 12 tablets per dissolution or n=36. The data is presented as percent of label claim dissoluted vs. dissolution time. The results demonstrate that the multi-point dissolution profiles for Levoxyl® tablets are similar across a wide variety of tablet strengths. Moreover, all strengths substantially exceed the requirements for immediate release oral dosage forms (i.e. at least 80% dissoluted with 15 - 20 minutes). In each dosage form, these pills were over 90% dissoluted within two and a half minutes. The extremely rapid dispersion rates for the tablets of the present invention make possible a simplified treatment method for infants or others who have difficulty swallowing pills. In this approach, the appropriate dosage for the patient in question, in an immediate release pill made in accordance with the present invention, is simply mixed with a suitable amount, e.g. 50 - 200 ml, of aqueous fluid, such as water, soft drinks, juice, milk, etc. The immediate release pill is easily dissoluted in the fluid, optionally with stirring or shaking, and simply admimstered to the patient.
Example 3
Potency Test The following method for testing potency of the tablets will sometimes be referred to herein as method number: AM-003. Alternatively, the tablet potency can be tested according to method AM-021. Method number: AM-021 is the same as method number: AM-003, except the tablets are dissolved whole without first grinding the tablets into a powder, as with method number: AM-003.
Method Reference:
USP 24 pp. 968-970
Chromatographic Conditions: Mobile Phase: 65:35:0.05 H20: CAN: H3P04 degassed and filtered; mobile phase composition may be altered to achieve a satisfactory resolution factor. Column:
ACN, 4.6 mm x 25 to 30 cm
Flow Rate:
1.5 ml/minute
Detector:
Deuterium, set at 225 nm
Injection Volume:
100 ml
System Suitability: Chromatograph 5 replicate injections of the standard preparation. Record the peak responses as directed under "Procedure".
1.0 RSD for the standard replicates must not be more than 2.0%o for T4. 2.0 Calculate the resolution factor R on one of the five replicates. The R- value must be greater than or equal to 5.0 to proceed. See Method QC-009.
Standard Preparation:
Accurately weight 25 mg of USP Levothyroxine RS and transfer to an amber 250-ml volumetric flask. Add approximately 50ml extraction mobile phase. Let stand for 20 minutes with occasional swirling. Sonicate for 30 seconds. Gradually add more extraction solution and repeat sonication until no undissolved particles are observed. Dilute to volume with extraction solution. Mix well. The concentration of T4 is about 100 μg/ml. Also dissolve an accurately weighed quantity of USP Liothyronine RS to yield about 100 mg/ml, done as above with USP Levothyroxine RS. Label this solution as stock T3-A. Stock Standard dilution:
1. Pipette 10.0 ml stock T3-A into a 500 ml Type A volumetric flask.
2. Dilute to volume with Mobile Phase for a concentration of about 2 μg/ml. Mix well and label this solution as std. T3-B.
3. Pipette 50.0 ml each from the T4 and T3-B stock standards and transfer into a 500-ml Type A volumetric flask
Dilute to volume with mobile phase and mix well. Label this standard as T3/ T working standard. The concentration of the working standard should be about 0.2 μg/ml T3 and 10.0 μg/ml T4.
Note:
Concentrations of Levothyroxine and Liothyronine require adjustments for water content.
Assay Preparation:
Weigh not less than the specified tablet quantity and calculate the average tablet weight. Crash tablets into a uniform fine powder with a mortar and pestle. Tare a polypropylene weigh boat.
Accurately weigh (to 0.1 mg) a portion of the powder into the tared weigh boat using a preconditioned stainless steel scoop or spatula (either Teflon coated or uncoated). The spatula or scoop is preconditioned by dipping it into the powder. Use the Sample Calculation below to achieve 50 ml of a 10 μg/ml assay solution.
Record the sample weight taken. Carefully transfer the sample into an Erlenmeyer flask, reweigh the weigh boat and subtract the residual weight from the weight taken to obtain the actual sample weight. Pipette 50 ml of mobile phase into the flask. Cover the flask with parafilm, sonicate for approximately 10 seconds and vortex for approximately 235 seconds at a speed of 6 or greater. Observe sample preparation, and if clumping is noted, repeat the sonication and/or vortex steps. Centrifuge (~3,000φm) for NLT 1 minute until a clear supernatant is achieved. Transfer a portion ofthe supernatant to an auto sampler vial.
Vox In-Process granulation analysis, use the theoretical tablet weight (0.1455 g) in place of (weight of tablets/number of tablets) in the formula below. Sample Calculation:
Weight of Tablets X lOμg X 50ml = Amount to Weight Out per Assay Number of Tablets ml Dose (μg)
Procedure:
Separately inject 100 μl of the sample onto the column. Record the responses of the analyte peak and calculate %> label claim as follows.
Calculations:
Sample Area x Std cone, (μg) x 50 ml x avg. tablet weight in g x 798.86 = μg/dose x 100 = % Label Claim
Standard Area (ml) Actual Sample wt in g 776.87 Label Claim
Where 798.86 = molecular weight of Levothyroxine as the Sodium Salt
776.87 = molecular weight of Levothyroxine Standard Base
Results:
Figures 5A and 5B show HPLC chromatograms of levothyroxine and liothyronine controls (T3/T4 working standard, shown in Figure 5A) and an experimental sample made in accordance with the present invention as described above.(Figure 5B). The peaks in both chromatograms in the area of 1.325 to 3.1 correspond to materials in the solvent. The peak at about 7.2 in Fig. 5 A shows the presence of T3. Fig. 5B shows the absence of T3, as well as the absence of other related products or degradation products of levothyroxine.
Example 4 Hardness Test
The following preferred method for testing tablet hardness will sometimes be referred to herein as method number: QC-005 Table 9- QC-O05 Hardness Test Procedure
Figure imgf000035_0001
Generally the hardness of the pills lies between about 6.0 and about 14.0 kiloponds. Preferably the pill hardness is from about 9 to about 13 kiloponds. Typical results of products made in accordance with the present invention are about 9.3, 11.3, 9.8, 10.2, 12.3, etc. Pharmaceutical tablets which incoφorate granulated active ingredient are typically much higher in hardness, which may add to the difficulty of dissolving or dissoluting them. Pills which are lower in hardness generally present more problems of pill fragmentation during handling and storage.
Example 5 Impurity Tests
The following preferred method for testing tablet impurities is sometimes referenced herein as method number: SA-004
Table 10 -SA 004 Impurity Test Procedure
Method Reference: Biochemie Method No. 1417-6, Report JMI-DP-002
Equipment: HPLC with a gradient system and a detector at a wavelength of 225 nm
Reagents: Acetonitrile, HPLC grade
Methanol, HPLC grade
Water, HPLC grade Sodium Hydroxide, ACS reagent grade
Sodium Hydroxide 0.1 solution: Dissolve 40g of NaOH pellets in 1000 ml
HPLC grade water. Store in a plastic container.
Phosphoric acid, 85% reagent grade
Diiodothyronine reference material
Liothyronine RS USP reference material
Levothyroxine RS USP reference material
Triiodothyroacetic acid reference material
Tetraiodothyroacetic acid reference material
Solvent 1: To 100.0 ml of 0.1 N Sodium Hydroxide solution add a 1:1 V/V mixture of methanol and water to make 1000 ml.
Solvent 2: 77:23:0.1 H2): CANACN: H3P04; Degassed and filtered; mobile phase composition a may be altered to achieve a satisfactory resolution factor.
Extraction solution: Pipette 50 ml of solvent 1 into a 1000 ml volumetric flask dilute to volume with solvent 2, stopper and mix well.
Chromatography Nucleosil 100-lOCN, 250 mm long, 4.6 mm internal diameter, at ambient
Column: temperature
System: Gradient Elution
Mobile phase A: 1000: 1 H20:H3P04 V/V
Mobile phase B: Acetonitrile
Gradient program:
Time min
% of mobile phase A
% of mobile phase B
0
77 23
13 77 23
15 65 35
24 65
35
26
77 23
Flow rate: 1.5 ml/min.
Injection Volume: 100 up: next injection after approx. 40 min.
Detector: UV, 225 nm
System Suitability: Chromatograph 5 replicate injections ofthe Reference I Standard preparation, chromatograpri 2 replicate injections ofthe Reference II Standard. Record the peak responses as directed under "Procedure". An extraction blank is to be run after the standards.
1. The RSD must not be greater than 2.0% for each ofthe impurities in the standard reference solution I.
2. The resolution factor between liothyronine and levothyroxine in thestandard reference solution I must not be less than 5.0.
3. The Signal to Noise ratio must not be less than 5/1 for levothyroxine and impurities in the chromatogram obtained with standard reference solution II.
4. A peak of monochlorotriiodothyronine may occur just before the levothyroxine peak. Make sure that the degree of separation between this peak and of levothyroxine is at least sufficient to permit separate evaluations. Monochlorotriiodothyronine reference material is not available to be purchase by any vendor. Any calculation of monochlorotriiodothyronine impurity will be done by its retention time.
Standards 1. Stock Standard Reference Solution: Preparation: Accurately weigh 10 mg +/- 0.1 mg of each Diiodothyronine, Liothyronine, Levothyroxine, Triiodothyroacetic acid and Tetraiodthyroacetic acid reference standards into a 100ml volumetric flask. Dissolve in Solvent 1 and dilute to volume, stopper and mix well. The concentration of each component will be approximately lOOmcg/mlL.
2. Standard Reference solution I:
Pipette 5.0 ml of Stock Standard Reference Solution into a 100 ml volumetric flask, dilute to volume with Solvent 2, stopper and mix well. The Final concentration of each component will be approximately 5mcg/mlL.
3. Standard Reference solution II (0.05%):
Pipette 2.0 ml of Standard Reference Solution I into a 100 ml volumetric flask, dilute to volume with Solvent 2, stopper and mix well. The final concentration of each component will be approximately 0.1 mcg/mlL.100
Test Preparation: Crush not less than 20 tablets. Tare a 250 ml Erlenmeyer flask. Accurately weigh to the nearest 0.1 mg an equivalent of 500 meg of levothyroxine sodium (+/- 10%) into a 250 ml Erlenmeyer flask. Pipette 100.0 meg of the Extraction solution into the flask cover the flask with parafilm, sonicate, vortex and then centrifuge the solution for 1 minute each. The final concentration ofthe sample will be approximately 5 meg/ml of levothyroxine.
To calculate the amount to weigh for the test preparation use the following equation:
500 meg X 0.1450g* = Amount to weight for the test prep tablet label claim (meg)
* where 0.1450 g = theoretical tablet weight
Note: Analyst must keep all materials use in performing this assay until the results are calculated, checked, and recorded and it is verified that the test is acceptable. This includes the crush, the Erlenmeyer flask with Extraction solution, the centrifuge tube and the auto-sampler vial. If the analysis is running overnight, these materials should be sealed with parafilm and saved until results are obtained and the results are deemed acceptable
Procedure: 1. Separately inject lOOμl ofthe sample preparation onto the column. Record the response ofthe analyte peaks and the calculate % w/w using the equations below.
2. The chromatogram may need to be reprocessed to obtain optimal integration. A copy ofthe sample chromatograph is to be attached to the analytical packet.
3. Peaks on the sample chromatograph with areas less than a signal ratio of 5/1 will be considered none detected.
Calculations:
Diiodothyronine :
Sample area X Std Cone, (meg) X 100ml X 100% X 1.11" = % w/w Std. Area ml Wsimpl (g) 1000000 mcg/g or
Sample area X Std. Cone, (meg) X (λ01 X 1.11* = %w/w
* where 1.11 is a correction factor
Triiodothyroacetic Acid: Sample area X Std Cone, (meg) X 100ml X 100% % w/w
Std. Area ml Wsimpl (g) 1000000 mcg/g or Sample area X Std. Cone, (meg) X 0.01 %w/w
Std area ml Wsimpl (g)
Tetraiodothyroacetic Acid:
Sample area X Std Cone, (meg) X 100ml X 100% x 1.16* = % w/w
Std. Area ml Wsimpl (g) 1000000 mcg/g or Sample area X StcT. Cone, ImcgJ D QT T.I.5*" = %w/w
Std area ml Wsimpl (g)
* where 1.16 is a correction factor
Limit of Detection (LOD) Values
Figure imgf000039_0001
Calculation of the theoretical area for 0.05% of levothyroxine sodium, based on the initial amount in mg of levathyroxine sodium in the whole sample weight.
(Arears II)(A)(10.0) = Theoretical area for 0.05% of levothyroxine Na, based (.05) (T4std st.)(P)(l .0283) on the actual weight
Where: Arears -is the average area ofthe levothyroxine in the Standard reference solution II A = is the initial weight of levothyroxine Na in mg represented by the sample weight. This is calculated by using this equation: = sample weight (g) X claim T^in meg
0.1450g 1000 mcg/mg 10.0 = theoretical initial weight ofthe Levothyroxine USP reference standard 0.500 = is the theoretical initial weight ofthe Levothyroxine NA to be tested, in mg T std. Wt. = the initial weight ofthe levothyroxine USP standard in mg P = the purity ofthe levothyroxine Na USP standard (%purity/100%) 1.0283 = conversion of levothyroxine into levothyroxine sodium Greatest unknown impurity (individually) : f Area imDUritv iT4std wt mg)( 1.0283) (P) (100) = impurity (%) (Area ref std I) "(A) (2000) Where: Area impurity is the area of the greatest unknown impurity in the test solution with an area greater than the theoretical area for 0.05% ofthe levothyroxine Na taken into account.
1.0283 = conversion of levothyroxine into levothyroxine sodium P = the purity ofthe levothyroxine Na USP standard (% purity/100%) 100 is the dilution of the test solution
Area ref std I is the area ofthe levothyroxine in the standard reference solution I A = is the initial weight of levothyroxine Na in mg represented by the sample weight. This is calculated by using this equation: = sample weight (g) X claim T4 in meg
0.1450g X 1000 mcg/mg
2000 is the dilution ofthe reference solution. Total of other Unknown Impurities: f Sum area impurities) (T4 std wt mg) (1.0283) (P) (100) = Total Unknown impurities (%) (are refstdI) (A) (2000)
Where: Sum area impurity is the sum ofthe areas of all the other unknown impurities in the test solution
(only areas that are greater than the theoretical area for 0.05% ofthe levothryoxine sodium taken into account)
T4 std. wt. = the initial weight ofthe levothyroxine USP standard in mg 1.0283 = conversion of levothyroxine into levothyroxine sodium
P = the purity ofthe levothyroxine Na USP standard (% pursity/100%)
100 is the dilution ofthe test solution
Area ref std I is the area ofthe levothyroxine in the standard reference solution I
A = is the initial weight of levothyroxine Na in mg represented by the sample weight.
This is calculated by using this equation: = sample weight (g) X claim T4 in meg
0.1450gX 100 mcg/mg 2000 is the dilution ofthe reference solution. Results of the test are shown in Figures 6 A and 6B. Figure 6A shows an example of a chromatogram of Standard Reference Solution II, with exemplary peaks at about 5.4 for diiodo-1- thyronine, 8.4 for Hotliryonine, 12.8 for levothyroxine, 19.3 for triiodo thyroacetic acid, and 21.9 for tetraiodo thyroacetic acid. Figure 6B shows results of an experimental sample of levothyroxine sodium, made in accordance with this invention. As can be seen, the sample had substantially only levothyroxine, with insignificant impurities.
Example 6
Liothyronine (T3) Tests The following preferred method for testing for Triiodothyronine is sometimes referenced herein as method number: QC-001
Table 11 - QC- 001 T3 Test Procedure
Method Reference USP 24 p. 968-970
Chromatographic 65:35:0.05 H20:CACN:H3P04 degassed and filtered; mobile phase
Conditions: composition may be altered to achieve a satisfactory resolution factor.
Mobile Phase:
Column: CN, 4.6 mm x 25 to 30 cm
Flow Rate: 2.0 minute/minute Detector: Deuterium, set at 225 nm Injection Volume: 100 μL System Suitability: Chromatograph 5 replicate injections ofthe standard preparation. Record the peak responses as directed under "Procedure".
1.0 RSD for the standard replicates must not be more than 2.0% for T
2.0 Calculate the resolution factor (R) on one ofthe five replicates.
The R value must be greater than or equal to proceed. See Method
QC-009.
Standard Preparation: Accurately weigh 25 mg of USP Levothyroxine RS and transfer to a clear 250-mlL volumetric flask. Pipette 87.5 ml minute of acetonitrile in the flask. Swirl and then sonicate for less than a minute. Add portions of HPLC grade water to the flask with swirling and sonicating until the material has gone into solution. Be sure that there is no particulate material present. Do not dilute to volume at this point. The solution may be cold. Place into a room temperature water bath for ten minutes to allow the sample to warm to ambient temperature. Dilute to volume with HPLC grade water. Mix well. Label this solution as stock T4. The concentration of T4is about 100 μg/ml.
Also dissolve an accurately weighed quantity of USP Liothyronine RS to yield about 100 μg/minute, done as above with USP Levothyroxine RS. Label this solution as stock T3-A.
Stock Standard dilution:
1. Pipette 10.0 ml stock T3-A into a 500-mlL Type A volumetric flask.
2. Dilute to volume with Mobile Phase for a concentration of about 2 μg/ml. Mix well and label this solution as stock std. c-B.
3. Pipette 50.0 ml each from the T4 and T3 stock standards and transfer into 500-mlL Type A volumetric flask.
Dilute to volume with mobile phase and mix well. Label this standard as T3/T4 working standard. The concentration ofthe working standard should be about 0.2 μg/ml T3 and 10.0 μg/ml T4.
Assay Preparation: Weigh and crash not less than the specified tablet quantity and calculate the average tablet weight. Tare a polypropylene weigh boat.
Accurately weigh (to 0.1 mg) a portion ofthe powder into the tared weigh boat using a preconditioned stainless steel scoop or spatula (either Teflon coated or uncoated). The spatula or scoop is preconditioned by dipping it into the power. Use the Sample Calculation below to achieve 50 ml of a 10 μg/ml assay solution.
Record the sample weight taken. Carefully transfer the sample into an Erlenmeyer flask, reweigh the weigh boat and subtract the residual weight from the weight taken to obtain the actual sample weight. Pipette 50 ml of mobile phase into the flask. Cover the flask with parafilm, sonicate for approximately 10 seconds and vortex for approximately 35 seconds at a speed of 6 or greater. Observe sample preparation, and if clumping is noted, repeat the sonication and/or vortex steps. Centrifuge (~3,000 rpm) for NLT 1 minute until a clear supernatant is achieved. Transfer a portion of the supernatant to an autosampler vial.
Ε ox In-Process granulation analysis, use the theoretical tablet weight (0.1455 g) in place of (weight of tablets/number of tablets) in the formula below.
Note Analyst must keep all materials used in performing this assay until the results are calculated, checked, and recorded, and it is verified that the test is acceptable. This includes the crush, the Erlenmeyer flask with Mobile Phase, the centrifuge tube and the autosampler vial. If the analysis is running overnight, these materials should be sealed with parafilm and saved until results are obtained and the result is deemed acceptable.
Sample Calculation: Weight of Tablets X 10 μg/ml X 5Q nl = Amount to Weigh Out per
Assay Number of Tablets Dose (μg)
Procedure: Separately inject 100 μl ofthe sample onto the column. Record the responses ofthe analyte peak.
Calculations: Calculate the content of liothyronine using the following formula:
Sample T? Area X Std T? cone, (μg) x 50 ml = μg T3 Standard T3 Area (ml)
The specification is NGT 2.0% liothyronine calculated as follows:
Amt T2 Assayed fug) x 100 = % LIOTHYRONINE
Amt T Assayed (ug)*~
*This number is calculated using the T potency results as follows:
Sample T4 Area X Std Td cone, (μg) x 50 ml x 798.86 = μg T., Standard T4 Area " (ml) 776.87 where 798.86 = molecular weight of Levothyroxine as the Sodium Salt 776.87 = molecular weight of Levothyroxine Standard Base
NOTE: If the single active ingredient comprises 50% or more, by weight, ofthe dosage unit, use Method A; otherwise use Method B.
METHOD: USP 24 <905> pp. 2000-2002. METHODA: Content Uniformity as Determined by Weight Variation: Weight accurately 10 tablets, individually. From the results ofthe average potency ofthe active ingredient determined for the product (using the assay methods as stated in the individual monograph) calculate the content of active ingredient in each ofthe 10 tablets. CALCULATIONS: Individual = (Avg. potency) (Individual Wt.) Potency Avg. tablet weight
NOTE: If the active ingredient(s) are less than 50% by weight ofthe tablet content, refer to the individual test method for potency for those products.
METHOD B: Content Uniformity as Determined by Direct Assay of Active Ingredient: For Levothyroxine Sodium tablets the following procedure is followed. Individually weigh 10 tablets. Place the 10 individual tablets into round bottomed test tubes or flasks ofthe appropriate size as outlined in the chart below. Add the appropriate volume of extraction mobile comprised of water, acetonitrile, and phosphoric acid (65:35::0.05) to each test tube or flask as indicated in the chart below. Note: All test tubes are to be capped with screw on caps and all flasks are to be covered with parafilm as soon as mobile phase is added. Allow to stand at room temperature until the tablet completely crumbles. Secure all samples in a wrist action shaker. Test tubes are to be secured horizontally. Erlenmeyer flasks are to be secured vertically. Set the wrist shaker to the setting specified in the table. Shake sample for 3 minutes. Transfer about 10 ml ofthe sample preparation (or the entirety of smaller samples) to a centrifuge tube. Centrifuge samples for 1 minute at about 3000 rpm. Transfer samples to autosampler vials using disposable Pasteur pipettes. Utilize the HPLC Method for levothyroxine separation (AM-003) for obtaining dosage uniformity, sample area, and standard area results.
CALCULATIONS: Dosage Uniformity Result (% Label Claim)
798.86 X Area of Sample X Cone, of Std. X 100 = % Potency
776.87 Area of Std. Cone. Of Sample (see chart below)
SPECIFICATIONS FOR METHOD A OR METHOD B
S-1 The % active ingredient for 10 tablets tested must fall in the range of 85.0%> - 115.0% and the RSD ofthe 10 tablets must not exceed 6.0%.
NOTE: If 1 unit in S-1 fails to meet either ofthe specifications, but is no outside the range of
75% - 125%, test 20 more units and proceed to S-2. S-2 When n = 30, NGT one unit outside 85.0-115.0%, none outside 75.0- 125.0% and RSD
NGT 7.80%.
Results:
Results for a variety of dosages, using a sample size of 120 pills, are shown in Table 12: Table 12 — Dosage Consistency - 120 pill samples
Figure imgf000043_0001
Figure imgf000044_0001
The results confirm an extremely low amount of variability in active material content between the 120 pills tested. Generally the variability for a 120 pill sample should be between about 90 and about 110 % of claimed activity, preferably between about 95 % and about 105%. The RSD for a 120 pill sample should not be greater than 5%, and preferably is less than 3%.
Example 7
Levothyroxine Sodium Release Specification and Analytical Methods
The specifications for levothyroxine sodium tablets are stated in: USP 24 page 969-970 and Supplement 1 page 2638. The additional requirements are in place to ensure the tablet appearance, for the individual tablet strengths, is correct and the physical characteristics ensure a quality tablet.
A. Analytical Methods:
All the test methods utilized in the testing of levothyroxine sodium meet USP system suitability requirements. All Levoxyl® batches are tested for conformance to the following specifications. The Table 13 below lists the test parameter, specification and the test method employed.
Table 13 - USP Specifications
Figure imgf000044_0002
Additional Requirements:
Figure imgf000045_0001
Example 8
Bioavailability determination of Two Levothyroxine formulations
The following example was performed along lines of a 1999 FDA publication entitled In- Vivo Pharmacokinetics and Bioavailability Studies and In-Vitro Dissolution Testing for Levothyroxine Sodium Tablets. The example includes the following two studies.
Study 1. Single-Dose Bioavailability Study
The objective of the study was to determine the bioavailability of Levoxyl® relative to a reference (oral solution) under fasting conditions.
Study 2: Dosage-Form Equivalence Study
The objective of the study was to determine the dosage-form bioequivalence between three different strengths of Levoxyl® tablets (low, middle and high range).
Study Objective: To determine the bioavailability of levothyroxine sodium (Levoxyl®) 0.3 mg tablets manufactured by JONES PHARMA INCORPORATED, relative to Knoll Pharmaceutical Company's levothyroxine sodium 200 g (Synthroid®) injection given as an oral solution following a single 0.6mg dose.
Study Methodology:
Single-dose, randomized, open-label, two-way crossover design Protocol Reference:
Guidance for Industry: In Vivo Pharmacokinetics and Bioavailability Studies and In Vitro Dissolution Testing for Levothyroxine Sodium Tablets (June 1999).
Number of Subjects:
A total of 30 subjects were enrolled in the study, and 27 subjects completed the study. All 30 subjects were included in the safety analysis and 27 subjects who completed the study were included in the pharmacokinetic analyses.
Diagnosis and Main Criteria for Inclusion:
All subjects enrolled in this study were judged by the investigator to be "healthy volunteers who met all inclusion and exclusion criteria.
Test Product. Dose. Duration. Mode of Administration, and Batch Number:
The test product was levothyroxine sodium (Levoxyl®) 2 X 0.3mg tablets admimstered as a single oral dose. The batch number utilized in this study was TT26.
Reference Product. Dose. Duration. Mode of Administration, and Batch Number:
The reference product was levothyroxine sodium (Synthroid®) 2 X 500 μg injection vials
(Knoll Pharmaceutical Company) reconstituted and 600 μg admimstered orally. The reference product used was the 500 μg injection instead of 200 μg due to the unavailability of sufficient quantities of 200 μg injection to conduct the study. The batch number utilized in this study was 80130028.
Criteria for Evaluation: Pharmacokinetics:
Pharmacokinetic assessment consisted ofthe determination of total (bound + free) T4 and T3 concentrations in serum at specified time points following drug administration. From the serum data, the parameters AUC(O-t), Cmax, and Tmax were calculated. Safety:
Safety assessment included vital signs, clinical laboratory evaluation (including TSH), physical examination, and adverse events (AEs) assessment.
Statistical Methods Pharmacokinetics :
Descriptive statistics (arithmetic mean, standard deviation (SD), coefficient of variation (CV), standard error ofthe mean (SE), sample size (N), mi-oimum, and maximum) were provided for all pharmacokinetic parameters. The effects of baseline and baseline-by treatment interaction were evaluated using a parametric (normal-theory) general linear model (ANCOVA) with treatment, period, sequence, subject within sequence, In(baseline), and interaction between In (baseline) and treatment as factors, applied to the In-transformed pharmacokinetic parameters and Cmax. hi the absence of significant In(baseline) and interaction between In(baseline) and treatment, these parameters were removed from the model. The two one-sided hypotheses were tested at the 5% level of significance for In[AUC(0-t)] and In(Cmax) by constructing 90% confidence intervals for the ratio of Treatment A to Treatment Safety:
Frequency counts of all subjects enrolled in the study, completing the study, and discontinuing early were tabulated. Descriptive statistics were calculated for continuous demographic variables, and frequency counts were tabulated for categorical demographic variables for each gender and overall.
AEs were coded using the 5th Edition of the COSTART dictionary. AEs were summarized by the number and percentage of subjects experiencing each coded event. A summary of the total number of each coded event and as a percentage of total AEs was also provided.
Laboratory summary tables included descriptive statistics for continuous serum chemistry and hematology results at each time point. Out-of-range values were listed by subject for each laboratory parameter. Descriptive statistics tor vital sign measurements at each time point and change from baseline to each time point were calculated by treatment group. Shifts from screening to post study results for physical examinations were tabulated.
Pharmacokinetic Results - T4:
ANCONA analyses indicated that the effects of In(baseline) and interaction between In(baseline) and treatment were not significant. Thus, these factors were removed from the general linear model and an AΝONA with treatment, period, sequence, and subject within sequence was applied to the In-transformed Cmax and AUC(O-t) parameters. The arithmetic means of serum T4 pharmacokinetic parameters for Treatments A and B and the statistical comparison for In-transformed parameters are summarized in the following table.
Summary of the Pharmacokinetic Parameters of Serum T4 for Treatments A and B
Figure imgf000048_0001
* Treatment A = 2 X 0.3mg Levoxyl® Tablets: test
**Treatment B = 0.6mg Synthroid Reconstitute Oral Solution: reference
Pharmacokinetics Results - T3:
AΝCONA analyses indicated that the effects of In(baseline) and interaction between
In(baseline) and treatment were not significant and were removed from the ANON A model, except for In(baseline) on In(Cmax) which was significant and was kept in the model. An
AΝONA with treatment, period, sequence, and subject within sequence, and In(baseline), when significant, was applied to the In-transformed Cmax and AUC(O-t) parameters. The arithmetic means of serum T3 pharmacokinetic parameters for Treatments A and B and the statistical comparison for In-transformed parameters are summarized in the following table. Summary of the Pharmacokinetic Parameters of Serum T3 for Treatments A and B
Figure imgf000049_0001
* Treatment A = 2 X 0.3mg Levoxyl® Tablets: test
**Treatment B = 0.6mg Synthroid Reconstitute Oral Solution: reference
Comparison of total T4 and T3 pharmacokinetics following administration of Levoxyl® (Treatment A, test formulation) and Synthroid (Treatment B, reference formulation) indicated that the test formulation met the requirements for bioequivalence with the reference formulation.
The 90% confidence intervals for the comparisons of In(Cmax) and In[AUC(0-t)] for T4 and T3 were within the 80% to 125% range required for bioequivalence.
In regard to subject safety, both treatments appeared to be equally safe and well tolerated.
Example 9
Bioavailability Study To Assess Single Dose Bioequivalence of Three Strengths of Levothyroxine
The following example was performed to determine the dosage-form bioequivalence between three different strengths of levothyroxine sodium (Levoxyl®) tablets following a single 600 meg dose.
Study Methodology:
Single-dose, randomized, open-label, three-way crossover design.
Protocol Reference: Guidance for Industry: In Vivo PharmacoBhefi-cs and BioavailabilTty Sfudϊes and in Nitro Dissolution Testing for Levothyroxine Sodium Tablets (June 1999). This protocol was submitted in IΝD 59,177.
Number of Subjects:
A total of 28 subjects were enrolled in the study, and 24 subjects completed the study. All 28 subjects were included in the safety analysis and 24 subjects who completed the study were included in the pharmacokinetic analyses.
Diagnosis and Main Criteria for Inclusion:
All subjects enrolled in this study were judged by the investigator to be healthy volunteers who met all inclusion and exclusion criteria.
Test Product. Dose. Duration. Mode of Administration, and Batch Number: Subjects randomized to Treatment A received a single oral dose of 12 X 50 meg levothyroxine sodium (Levoxyl®) tablets, Lot No. TT24. Subjects randomized to Treatment B received 6 X 100 meg levothyroxine sodium (Levoxyl®) tablets, Lot No.TT25. Subjects randomized to Treatment C received 2 X 300 meg levothyroxine sodium (Levoxyl®) tablets, Lot No. TT26. Test products were manufactured by JMI-Daniels, a subsidiary of Jones Pharma Incoφorated.
Pharmacokinetics : Pharmacokinetic assessment consisted ofthe detennination of total (bound + free) T4 and
T3 concentrations in serum at specified time points following drag administration. From the serum data, the parameters AUC(O-t), Cmax, and Tmax were calculated.
Safety: Safety assessment included monitoring of sitting vital signs, clinical aboratory measurements, thyroid-stimulating hormone (TSH), physical examination, electrocardiogram (ECG), and adverse events (AEs).
Statistical Methods. Pharmacokinetics :
Descriptive statistics (arithmetic mean, standard deviation (SD), coefficient of variation (CN), standard error of the mean (SEM), sample size (Ν), minimum, and maximum) were provided for all pharmacokinetic parameters. A parametric (normal-theory) general linear model with treatment, period, sequence, and subject within sequence as factors was applied to the In- transformed Cmax and AUC(O-t). The two one-sided hypotheses were tested at the 5% level of significance for In[AUC(0-t)] and In(Cmax) by constructing 90% confidence intervals for the ratios of Treatment A to Treatment B, Treatment A to Treatment C, and Treatment B to Treatment C.
Safety:
Frequency counts of all subjects enrolled in the study, completing the study, and discontinuing early were tabulated. Descriptive statistics were calculated for continuous demographic variables, and frequency counts were tabulated for categorical demographic variables for each gender and overall.
AEs were coded using the 5th Edition of the COSTART dictionary. AEs were summarized by the number and percentage of subjects experiencing each coded event. A summary of the total number of each coded event and as a percentage of total AEs was also provided. Laboratory summary tables included descriptive statistics for continuous serum chemistry and hematology results at each time point. Out-of-range values were listed by subject for each laboratory parameter. Descriptive statistics for vital sign measurements at each time point and change from baseline to each time point were calculated by treatment group. Shifts from screening to post study results for physical examinations were tabulated.
Pharmacokinetic Resulta- T4: The arithmetic means of serum T4 pharmacokinetic parameters for Treatments A and B and the statistical comparison for the In-transformed parameters are summarized in the following table.
Summary ofthe Pharmacokinetic Parameters of Serum T4 for Treatments A and B
Figure imgf000052_0001
* Treatment A = 12 X 50 eg Levoxyl® Tablets **Treatment B = 6 X 100 meg Levoxyl® Tablets
The arithmetic means of serum T4 pharmacokinetic parameters for Treatments A and C and the statistical comparison for the In-transformed parameters are summarized in the following table.
Summary of the Pharmacokinetic Parameters of Serum T4 for Treatments A and C
Figure imgf000052_0002
Figure imgf000053_0001
* Treatment A = 12 X 50 meg Levoxyl® Tablets **Treatment C = 2 X 300 meg Levoxyl® Tablets
The arithmetic means of serum T4 pharmacokinetic parameters for Treatments B and C and the statistical comparison for the In-transformed parameters are summarized in the following table.
Pharmacokinetic Results - T4 (Continued):
Summary of the Pharmacokinetic Parameters of Serum T4 for Treatments B and C
Figure imgf000053_0002
* Treatment B = 6 X 100 mcg Levoxyl® Tablets **Treatment C = 2 X 300 meg Levoxyl® Tablets
Pharmacokinetic Results - T3: The arithmetic means of serum T3 pharmacokinetic parameters for Treatments A and B and the statistical comparison for the In-transformed parameters are summarized in the following table. Summary ofthe EarmacoKinetic Parameters of Serum T3 for Treatments A and B
Figure imgf000054_0001
* Treatment A = 12 X 50 meg Levoxyl® Tablets **Treatment B = 6 X 100 meg Levoxyl® Tablets
The arithmetic means of serum T3 pharmacokinetic parameters for Treatments A and C and the statistical comparison for the In-transformed parameters are summarized in the following table. Pharmacokinetic Results - T3 (Continued):
Summary of the Pharmacokinetic Parameters of Serum T3 for Treatments A and C
Figure imgf000054_0002
* Treatment A = 12 X 50 meg Levoxyl® Tablets **Treatment C = 2 X 300 meg Levoxyl® Tablets
The arithmetic means of serum T3 pharmacokinetic parameters for Treatments B and C and the statistical comparison for the In-transformed parameters are summarized in the following table. Summary ofthe Pharmacokinetic Parameters of Serum T3 for Treatments B and C
Figure imgf000055_0001
* Treatment B = 6 X 100 meg Levoxyl® Tablets * treatment C = 2 X 300 meg Levoxyl® Tablets
Safety Results:
There was a total of 59 treatment-emergent AEs reported by 15 (54%) ofthe 28 subjects dosed with study treatment. Incidence of AEs was similar across treatments. Headache was the most frequently reported event. The majority ofthe AEs were mild in intensity. There was one subject who experienced a serious adverse event of chest pain, considered by the Investigator to be unrelated to treatment. No trends were noted in vital signs, clinical laboratory results, or ECGs to suggest treatment-related differences. Comparison of total T4 and T3 pharmacokinetics following administration of 12 X 50 meg Levoxyl® tablets (Treatment A) and 6 X 100 meg Levoxyl® tablets (Treatment B) indicated that the two formulations met the requirements for bioequivalence. The 90% confidence intervals for the comparisons of In(Cmax) and In[AUC(0-t)] for T4 and T3 were within the 80% to 125%) range required for bioequivalence. Comparison of total T4 and T3 pharmacokinetics following administration of 12 X 50 meg Levoxyl® tablets (Treatment A) and 2 X 300 meg Levoxyl® tablets (Treatment C) indicated that the two formulations met the requirements for bioequivalence. The 90% confidence intervals for the comparisons of In(Cmax) and In[AUC(0-t)] for T4 and T3 were within the 80% to 125% range required for bioequivalence. Comparison of total T4 and T3 pharmacokinetics following administration of 6 X 100 meg Levoxyl® tablets (Treatment B) and 2 X 300 meg Levoxyl® tablets (Treatment C) indicated that the two fδrmufations met the requirements for bioequivalence. The 90% confidence intervals for the comparisons of In(Cmax) and In[AUC(0-t)] for T4 and T3 were within the 80% to 125% range required for bioequivalence.
The test formulations appear to be safe and generally well tolerated when given to healthy adult volunteers.
Example 10
Levothyroxine sodium (Levoxyl®) Tablet Compositions
The following preferred levothroxine sodium compositions in tablet form were made along lines disclosed herein.
Levoxyl® 25 meg Tablets
Color: orange; Markings: (front) dp/25 (back) LEVOXYL®
Figure imgf000056_0001
Levoxyl® 50 meg Tablets
Color: white; Markings: (front) dp/50 (back) LEVOXYL®
Figure imgf000056_0002
Levoxyl® 75 meg Tablets
Color: purple; Markings: (front) dp/75 (back) LEVOXYL®
Figure imgf000056_0003
Figure imgf000057_0001
Levoxyl® 88 meg Tablets
Color: olive; Markings: (front) dp/88 (back) LEVOXYL®
Figure imgf000057_0002
Levoxyl® 100 meg Tablets
Color: yellow; Markings: (front) dp/100 (back) LEVOXYL®
Figure imgf000057_0003
Levoxyl® 112 meg Tablets
Color: rose; Markings: (front) dp/112 (back) LEVOXYL®
Figure imgf000057_0004
Figure imgf000058_0001
Levoxyl® 137 meg Tablets
Color: dark blue; Markings: (front) dp/137 (back) LEVOXYL®
Figure imgf000058_0002
Levoxyl® 150 meg Tablets
Color: blue; Markings: (front) dp/150 (back) LEVOXYL®
Figure imgf000058_0003
Levoxyl® 175 eg Tablets
Color: turquoise; Markings: (front) dp/175 (back) LEVOXYL®
Figure imgf000058_0004
Levoxyl® 200 meg Tablets
Color: pink; Markings: (front) dp/200 (back) LEVOXYL®
Figure imgf000058_0005
Figure imgf000059_0001
Levoxyl® 300 meg Tablets
Color: green; Markings: (front) dp/300 (back) LEVOXYL®
Figure imgf000059_0002
While the present invention has been described in the context of preferred embodiments and examples, it will be readily apparent to those skilled in the art that other modifications and variations can be made therein without departing from the spirit or scope of the present invention. For example, the active moiety levothyroxine sodium can be changed to liothyronine sodium and similar products and still be considered as part of the claimed invention.
Accordingly, it is not Intended that the present invention be limited to the specifics of the foregoing description of the preferred embodiments and examples, but rather as being limited only by the scope ofthe invention as defined In the claims appended hereto.
Having described our invention, we claim:

Claims

CLAIMS:
1. A method of administering a levothyroxine pharmaceutical composition to a patient, comprising placing an immediate release levothyroxine pharmaceutical tablet in an aqueous medium, dispersing the levothyroxine composition in the aqueous medium for less than ten minutes, and administering the aqueous medium to the patient.
2. The method of claim 1, wherein the aqueous medium is selected from water, saline, soft drinks and milk.
3. The method of claim 2, wherein the dispersion step is conducted for less than five minutes.
4. An immediate release pharmaceutical composition comprising a levothyroxine salt, wherein the composition exhibits a triiodothyronine (T3) plasma Cmax of between from about 0.1 ng/ml to about 10 ng/ml as determined by the standard Cmax test.
5. The composition of claim 4, wherein the composition exhibits a triiodothyronine (T3) plasma Cmax of between from about 0.5 ng/ml to about 2 ng/ml as determined by the standard Cmax test.
6. The composition of claims 4, wherein the In(Cmax) is between from about 0.01 to about 5.
7. The composition of claims 5, wherein the In(Cmax) is between from about 0.01 to about
5.
8. An immediate release pharmaceutical composition comprising a levothyroxine salt, wherein the composition features a triiodothyronine (T3) AUC (0-t) of between from about 10 ng-hour/ mlL to about 100 ng-hour/ mlL
9. The composition of claim 8, wherein the composition features a triiodothyronine (T3) AUC (0-t) of between from about 20 ng-hour/ml to about 60 ng-hour/ ml.
10. The composition of claims 8, wherein the In[AUC(0~t)] is between from about 1 to about 5.
11. The composition of claims 9, wherein the In[AUC(0-t)] is between from about 1 to about
5.
12. The composition of claims 9, wherein the In(Cmax) of the levothyroxine (T4) plasma level is between from about 1 to about 3.
13. An immediate release pharmaceutical composition comprising a levothyroxine salt, wherein the composition exhibits a levothyroxine (T4) plasma Cmax of between from about 10 μg/dl to about 20 μg/dl as determined by a standard Cmax test.
14. The composition of claim 13, wherein the composition exhibits a levothyroxine (T4) plasma Cmax of between from about 12 μg/dl to about 16 μg/dl as determined by the standard Cmax test.
15. The composition of claims 13, wherein the In(Cmax) of the levothyroxine (T4) plasma level is between from about 1 to about 3.
16. The composition of claims 14, wherein the In(Cmax) of the levothyroxine (T4) plasma level is between from about 1 to about 3.
17. An immediate release pharmaceutical composition comprising a levothyroxine salt, wherein the composition exhibits a levothyroxine (T4) plasma Tmax of between from about 0.5 hours to about 5 hours as determined by a standard Tmax test.
18. The composition of claim 17, wherein the composition exhibits a levothyroxine (T4) plasma Tmax of between from about 1 hour to about 3 hours as determined by the standard Tmax test.
19. An immediate release pharmaceutical composition comprising a levothyroxine salt, wherein the composition exhibits a triiodothyronine (T3) plasma Tmax of between from about 10 hours to about 20 hours as determined by the standard Tmax test.
20. The composition of claim 19, wherein the composition exhibits a triiodothyronine (T3) plasma Tmax of between from about 12 hours to about 16 hours as determined by the standard Tmax test.
21. A pharmaceutical composition, comprising the product of dissolution of a preparation comprising levothyroxine salt, cross-linked sodium carboxymethylcellulose, and β-form microcrystalline cellulose, in an aqueous medium.
22. The composition of claim 21, wherein the preparation comprises about 0.001 to 1% by weight levothyroxine, about 5 to about 40% by weight cross-linked sodium carboxymethylcellulose, and at least about 50% by weight of β-form microcrystalline cellulose.
PCT/US2002/025752 2001-08-14 2002-08-14 Levothyroxine compositions and methods WO2003028624A2 (en)

Priority Applications (1)

Application Number Priority Date Filing Date Title
AU2002362468A AU2002362468A1 (en) 2001-08-14 2002-08-14 Levothyroxine compositions and methods

Applications Claiming Priority (11)

Application Number Priority Date Filing Date Title
US31211401P 2001-08-14 2001-08-14
US31227301P 2001-08-14 2001-08-14
US31248301P 2001-08-14 2001-08-14
US31228701P 2001-08-14 2001-08-14
US31211301P 2001-08-14 2001-08-14
US31218401P 2001-08-14 2001-08-14
US60/312,113 2001-08-14
US60/312,114 2001-08-14
US60/312,273 2001-08-14
US60/312,287 2001-08-14
US60/312,184 2001-08-14

Publications (2)

Publication Number Publication Date
WO2003028624A2 true WO2003028624A2 (en) 2003-04-10
WO2003028624A3 WO2003028624A3 (en) 2003-11-06

Family

ID=27617967

Family Applications (2)

Application Number Title Priority Date Filing Date
PCT/US2002/025854 WO2003061557A2 (en) 2001-08-14 2002-08-14 Levothyroxine pharmaceutical compositions, methods of making and methods of administration
PCT/US2002/025752 WO2003028624A2 (en) 2001-08-14 2002-08-14 Levothyroxine compositions and methods

Family Applications Before (1)

Application Number Title Priority Date Filing Date
PCT/US2002/025854 WO2003061557A2 (en) 2001-08-14 2002-08-14 Levothyroxine pharmaceutical compositions, methods of making and methods of administration

Country Status (2)

Country Link
AU (1) AU2002341555A1 (en)
WO (2) WO2003061557A2 (en)

Cited By (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6645526B2 (en) 2001-11-13 2003-11-11 Mylan Pharmaceuticals, Inc. Storage stable thyroxine active drug formulations and methods for their production
US7052717B2 (en) 2001-11-13 2006-05-30 Mylan Pharmaceuticals Inc. Storage stable thyroxine active drug formulations and methods for their production

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
EP2428516A1 (en) 2003-11-19 2012-03-14 Metabasis Therapeutics, Inc. Novel phosphorus-containing thyromimetics
MX2007014502A (en) 2005-05-26 2008-02-07 Metabasis Therapeutics Inc Thyromimetics for the treatment of fatty liver diseases.
US11202789B2 (en) 2016-11-21 2021-12-21 Viking Therapeutics, Inc. Method of treating glycogen storage disease
BR112019025659A2 (en) 2017-06-05 2020-08-25 Viking Therapeutics, Inc. compositions for the treatment of fibrosis
KR20200138283A (en) 2018-03-22 2020-12-09 바이킹 테라퓨틱스 인코포레이티드 Crystalline forms, and methods of preparing compounds in crystalline form
US20230023032A1 (en) * 2020-01-13 2023-01-26 Amneal Complex Products Research Llc Sustained release compositions comprising liothyronine

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5753254A (en) * 1994-02-01 1998-05-19 Knoll Aktiengesellschaft Therapeutic agents containing thyroid hormones
US6190696B1 (en) * 1998-06-08 2001-02-20 Pieter J. Groenewoud Stabilized thyroxine medications

Family Cites Families (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5955105A (en) * 1995-11-14 1999-09-21 Knoll Pharmaceutical Company Stabilized thyroid hormone preparations and methods of making same

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US5753254A (en) * 1994-02-01 1998-05-19 Knoll Aktiengesellschaft Therapeutic agents containing thyroid hormones
US6190696B1 (en) * 1998-06-08 2001-02-20 Pieter J. Groenewoud Stabilized thyroxine medications

Cited By (4)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US6645526B2 (en) 2001-11-13 2003-11-11 Mylan Pharmaceuticals, Inc. Storage stable thyroxine active drug formulations and methods for their production
US6936274B2 (en) 2001-11-13 2005-08-30 Mylan Pharmaceuticals, Inc. Storage stable thyroxine active drug formulations and methods for their production
US7052717B2 (en) 2001-11-13 2006-05-30 Mylan Pharmaceuticals Inc. Storage stable thyroxine active drug formulations and methods for their production
US7195779B2 (en) 2001-11-13 2007-03-27 Mylan Pharmaceuticals Inc. Storage stable thyroxine active drug formulations and methods for their production

Also Published As

Publication number Publication date
WO2003028624A3 (en) 2003-11-06
AU2002341555A1 (en) 2003-09-02
WO2003061557A2 (en) 2003-07-31
WO2003061557A3 (en) 2003-11-06

Similar Documents

Publication Publication Date Title
US6555581B1 (en) Levothyroxine compositions and methods
US20040202716A1 (en) Composition
US20080003284A1 (en) Levothyroxine compositions and methos
US7067148B2 (en) Stabilized pharmaceutical and thyroid hormone compositions and method of preparation
US7101569B2 (en) Methods of administering levothyroxine pharmaceutical compositions
WO2003028624A2 (en) Levothyroxine compositions and methods
US20030195253A1 (en) Unadsorbed levothyroxine pharmaceutical compositions, methods of making and methods of administration
WO2003013441A2 (en) Levothyroxine compositions and methods
WO2003070217A1 (en) Levothyroxine compositions and methods
WO2004014318A2 (en) Levothyroxine compositions and methods
US20030199585A1 (en) Levothyroxine compositions and methods
US20030180353A1 (en) Stabilized pharmaceutical compositions
US20030194437A1 (en) Levothyroxine compositions having unique triiodothyronine Cmax properties
US20040043066A1 (en) Levothyroxine compositions having unique triiodothyronine Tmax properties
US20030198667A1 (en) Methods of producing dispersible pharmaceutical compositions
US20030185885A1 (en) Non-granulated levothyroxine pharmaceutical compositions
US20030199588A1 (en) Levothyroxine compositions and methods
US20030198672A1 (en) Levothyroxine compositions having unique triidothyronine plasma AUC properties
US20030190349A1 (en) Methods of stabilizing pharmaceutical compositions
US20030195254A1 (en) Levothyroxine compositions having unique triiodothyronine Tmax properties
US20030203967A1 (en) Levothyroxine compositions having unique Tmax properties
US20030198671A1 (en) Levothyroxine compositions having unique plasma AUC properties
US20030194436A1 (en) Immediate release pharmaceutical compositions
US20030199587A1 (en) Levothyroxine compositions having unique Cmax properties
US20030199586A1 (en) Unique levothyroxine aqueous materials

Legal Events

Date Code Title Description
AK Designated states

Kind code of ref document: A2

Designated state(s): AE AG AL AM AT AU AZ BA BB BG BY BZ CA CH CN CO CR CU CZ DE DM DZ EC EE ES FI GB GD GE GH HR HU ID IL IN IS JP KE KG KP KR LC LK LR LS LT LU LV MA MD MG MN MW MX MZ NO NZ OM PH PL PT RU SD SE SG SI SK SL TJ TM TN TR TZ UA UG US UZ VN YU ZA ZM

AL Designated countries for regional patents

Kind code of ref document: A2

Designated state(s): GH GM KE LS MW MZ SD SL SZ UG ZM ZW AM AZ BY KG KZ RU TJ TM AT BE BG CH CY CZ DK EE ES FI FR GB GR IE IT LU MC PT SE SK TR BF BJ CF CG CI GA GN GQ GW ML MR NE SN TD TG

121 Ep: the epo has been informed by wipo that ep was designated in this application
DFPE Request for preliminary examination filed prior to expiration of 19th month from priority date (pct application filed before 20040101)
REG Reference to national code

Ref country code: DE

Ref legal event code: 8642

122 Ep: pct application non-entry in european phase
NENP Non-entry into the national phase in:

Ref country code: JP

WWW Wipo information: withdrawn in national office

Country of ref document: JP