CA2819234A1 - Pharmaceutical compositions of iron for oral administration - Google Patents

Pharmaceutical compositions of iron for oral administration Download PDF

Info

Publication number
CA2819234A1
CA2819234A1 CA2819234A CA2819234A CA2819234A1 CA 2819234 A1 CA2819234 A1 CA 2819234A1 CA 2819234 A CA2819234 A CA 2819234A CA 2819234 A CA2819234 A CA 2819234A CA 2819234 A1 CA2819234 A1 CA 2819234A1
Authority
CA
Canada
Prior art keywords
iron
dosage form
oral dosage
pharmaceutical composition
subject
Prior art date
Legal status (The legal status is an assumption and is not a legal conclusion. Google has not performed a legal analysis and makes no representation as to the accuracy of the status listed.)
Abandoned
Application number
CA2819234A
Other languages
French (fr)
Inventor
Alan Cullen
Edel O'toole
David C. Coughlan
Kishore Babu Chalasani
Thomas W. Leonard
Current Assignee (The listed assignees may be inaccurate. Google has not performed a legal analysis and makes no representation or warranty as to the accuracy of the list.)
Merrion Research Ill Ltd
Original Assignee
Merrion Research Ill Ltd
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 Merrion Research Ill Ltd filed Critical Merrion Research Ill Ltd
Publication of CA2819234A1 publication Critical patent/CA2819234A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K33/00Medicinal preparations containing inorganic active ingredients
    • A61K33/24Heavy metals; Compounds thereof
    • A61K33/26Iron; Compounds thereof
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/52Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an inorganic compound, e.g. an inorganic ion that is complexed with the active ingredient
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/045Hydroxy compounds, e.g. alcohols; Salts thereof, e.g. alcoholates
    • A61K31/07Retinol compounds, e.g. vitamin A
    • 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
    • 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
    • A61K31/192Carboxylic acids, e.g. valproic acid having aromatic groups, e.g. sulindac, 2-aryl-propionic acids, ethacrynic acid 
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/335Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin
    • A61K31/35Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom
    • A61K31/352Heterocyclic compounds having oxygen as the only ring hetero atom, e.g. fungichromin having six-membered rings with one oxygen as the only ring hetero atom condensed with carbocyclic rings, e.g. methantheline 
    • A61K31/3533,4-Dihydrobenzopyrans, e.g. chroman, catechin
    • A61K31/355Tocopherols, e.g. vitamin E
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K45/00Medicinal preparations containing active ingredients not provided for in groups A61K31/00 - A61K41/00
    • A61K45/06Mixtures of active ingredients without chemical characterisation, e.g. antiphlogistics and cardiaca
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/50Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates
    • A61K47/51Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent
    • A61K47/54Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient the non-active ingredient being chemically bound to the active ingredient, e.g. polymer-drug conjugates the non-active ingredient being a modifying agent the modifying agent being an organic compound
    • A61K47/547Chelates, e.g. Gd-DOTA or Zinc-amino acid chelates; Chelate-forming compounds, e.g. DOTA or ethylenediamine being covalently linked or complexed to the pharmacologically- or therapeutically-active agent
    • 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
    • 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
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • 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
    • A61K9/2013Organic compounds, e.g. phospholipids, fats
    • A61K9/2018Sugars, or sugar alcohols, e.g. lactose, mannitol; Derivatives thereof, e.g. polysorbates
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61PSPECIFIC THERAPEUTIC ACTIVITY OF CHEMICAL COMPOUNDS OR MEDICINAL PREPARATIONS
    • A61P7/00Drugs for disorders of the blood or the extracellular fluid
    • A61P7/06Antianaemics

Abstract

The present invention generally relates to orally administered pharmaceutical compositions of iron compounds with medium chain fatty acid salts. The invention further relates to methods of using the pharmaceutical compositions to treat iron deficiency and related disorders.

Description

Pharmaceutical Compositions of Iron for Oral Administration Alan Cullen, Edel O'Toole, David Coughlan, Kishore Chalasani, Thomas Leonard Related Applications [0001] This application claims the benefit under 35 U.S.C. 119(e) of United States Provisional Patent Application Serial Number 61/430,575, filed January 7, 2011, the disclosure of which is incorporated herein by reference in its entirety.
Field of the Invention [0002] The present invention generally relates to orally administered pharmaceutical compositions of iron compounds with medium chain fatty acid salts. The invention further relates to methods of using the pharmaceutical compositions to treat iron deficiency and related disorders.
Background of the Invention [0003] Iron in a variety of forms is administered for the treatment of iron deficiency and related disorders (e.g., anemia) as well as prophylactically to supply the minimum daily recommended allowance. A variety of iron compounds have been administered, including ferric and ferrous forms of elemental iron as salts, complexes, hydrates, chelates, and bound to polymer. Current oral iron preparations have low bioavailability and substantial side effects associated with them. Numerous formulation attempts have been made to create dosage forms that provide adequate iron absorption to treat deficiencies with less side effects. The side effects from oral administration primarily are a result of the large doses necessary to promote adequate absorption. It is likely that the presence of unabsorbed iron that remains in the gastrointestinal (GI) tract also significantly contributes to irritation, therefore side effects are also related to the poor bioavailability. The side effects include abdominal pain, heartburn, constipation, diarrhea, nausea, and vomiting. The inadequate absorption by oral delivery combined with poor compliance from the adverse effects on the GI tract means that in cases of substantial iron deficiencies, therapy must be carried out by parenteral injection, either intravenously or intramuscularly. Parenteral therapy is also associated with substantial side effects, including anaphylactic shock, injection site issues, hypotension, muscle cramps, dizziness, headache, graft complications, hypertension, chest pain, dysguesia, ear pain, and peripheral edema, as well as the cost and discomfort associated with injectable medication.
[0004] There is a need for improved oral formulations that achieve much better bioavailability. Such a formulation allows therapeutic doses to be given by a non-parenteral route while providing decreased side effects compared to present formulations.
Such an improved formulation can be used to replace parenteral therapy, and further improve patient quality of life. By improving the oral bioavailability, the total dose and the residual amounts of iron in the GI tract will be lowered which will improve the side effect profile, improve compliance, and allow a therapeutic effect approaching that of the injectable products to be achieved. Oral doses can then be used to achieve the required blood concentrations and body loads which now can only be achieved via parenteral doses to improve hemoglobin and ferritin levels, e.g., in severely anemic patient.
[0005] Rapid release oral iron formulations are preferred since iron is best absorbed in the upper small intestine. Controlled and extended release formulations are not effective for iron since there is minimal lower intestinal and colonic absorption. The ideal site for absorption is the upper small intestine, where active transport/divalent metal transporters and favorable pH promote absorption.
[0006] WO 2005/041928 describes a composition comprising iron and a transport moiety, such as a fatty acid. The composition is prepared in a manner such that the iron and the transport moiety form a tight ion binding pair, i.e., a salt. Creation of new salt forms for iron has inherent issues associated with it, including the need to establish safety, stability, and commercial manufacturing procedures. Development of a formulation that can work with existing salts and/or chelates is much preferred.
[00071 There is a continuing need for development of novel pharmaceutical formulations of iron suitable for oral administration, which not only offer the convenience of oral dosing, but also provide increased bioavailability of iron without excessive side effects.
Summary of the Invention [0008] The present invention relates to pharmaceutical compositions and oral dosage forms of iron and an absorption enhancer. The compositions provide increased bioavailability of iron. The compositions also overcome the negative feedback mechanism that limits iron absorption.
[0009] In one aspect, the invention provides a pharmaceutical composition for oral administration comprising, consisting essentially of, or consisting of an iron compound and an absorption enhancer, wherein the absorption enhancer is a medium chain fatty acid salt having a carbon chain length of from about 4 to about 20 carbon atoms. In some aspects, the composition does not involve chemical modification of the iron compound. In other aspects, the iron compound and the absorption enhancer are not part of a tight ion binding pair. The invention allows the use of conventional iron compounds (e.g., salts of iron, such as ferrous sulfate, or chelates and complexes of iron, such as EDTA and pyrophosphate complexes) to be used in oral dosage forms containing medium chain fatty acid salts. The oral dosage forms include solid dosage forms such as tablets, capsules, and powders and liquid dosage forms such as solutions, suspensions, elixirs, and syrups.
[0010] In a further aspect, the invention relates to a pharmaceutical composition for oral administration comprising an iron compound and an absorption enhancer, wherein the composition provides a bioavailability of iron that is at least 1.5 times greater than the bioavailability provided by a composition comprising ferrous sulfate that does not contain an absorption enhancer.
[0011] In another aspect, the invention relates to an oral dosage form (e.g., a solid, semi-solid, or liquid oral dosage form) comprising, consisting essentially of, or consisting of the pharmaceutical composition of the invention.
[0012] In another aspect, the invention relates to a method of orally delivering iron to a subject, comprising administering to the subject the oral dosage form of the invention.
[0013] In another aspect, the invention relates to a method for increasing the level of iron in a subject in need thereof, e.g., in the blood of a subject in need thereof, comprising administering to the subject the oral dosage form of the invention.
[0014] In another aspect, the invention relates to a method for increasing the level of hemoglobin in the blood of a subject in need thereof, comprising administering to the subject the oral dosage form of the invention.
[0015] In another aspect, the invention relates to a method for treating an iron deficiency in a subject in need thereof, comprising administering to the subject the oral dosage form of the invention.
[0016] In another aspect, the invention relates to a method for maintaining iron indices in a subject to ensure maximum efficacy of erythropoietin and/or other erythropoiesis-stimulating agents, comprising administering to the subject the oral dosage form of the invention.
[0017] In another aspect, the invention relates to a method for treating a disease or disorder characterized by an iron deficiency in a subject in need thereof, comprising administering to the subject the oral dosage form of the invention.

Brief Description of the Drawings [0018] Figure 1 shows the plasma iron concentration vs. time profiles of the test formulations after period 1 and period 2 dosing in dogs.
[0019] Figure 2 shows the dissolution profiles of C10 and Fe for enhanced tablets containing ferric pyrophosphate (FPP) soluble and different solubilizing excipients.
[0020] Figure 3 shows the dissolution profiles of C10 and Fe for enhanced tablets containing FPP soluble without any solubilizing excipients (no citric acid).
[0021] Figure 4 shows the dissolution profiles of C10 and Fe for enhanced tablets containing FPP soluble with citric acid as solubilizing aid.
[0022] Figure 5 shows the dissolution profiles of C10 and Fe for enhanced tablets containing FPP soluble and both citric acid plus sorbitol.
[0023] Figure 6 shows the dissolution profiles of C10 and Fe for enhanced tablets containing FPP soluble and high concentration of citric acid (148 mg tablet). Iron content normalized forl 00%.
[0024] Figure 7 shows the dissolution profiles of C10 and Fe for enhanced tablets containing FPP soluble and sodium citrate (148mg tablet) from n = 2 tablets.
[0025] Figure 8 shows the dissolution profiles of Fe for enhanced tablets containing FPP soluble and different solubilizing excipients.
[0026] Figure 9 shows the dissolution profiles of Fe and C10 for enhanced tablets containing FPP soluble and ascorbic acid and placebo C10 tablets.
[0027] Figure 10 shows the profiles of Fe and C10 normalized for enhanced tablets containing FPP soluble and ascorbic acid.
[0028] Figure 11 shows the dissolution profiles of Fe and C10 for enhanced tablets containing FPP soluble and different solubilizing excipients.
[0029] Figure 12 shows the dissolution profiles of Fe and C10 for enhanced iron tablets containing varying concentrations of citric acid (n = 2 tablets).
[0030] Figure 13 shows the dissolution profiles of Fe and C10 for enhanced iron tablets containing FPP soluble and citric acid prepared to evaluate C10 recovery and co release (EXP 1642).
[0031] Figure 14 shows the dissolution profiles of Fe and C10 for enhanced iron tablets containing FPP soluble and citric acid prepared to evaluate C10 recovery and co release (EXP 1649).

[0032] Figure 15 shows the dissolution profiles of Fe and C10 for enhanced iron tablets containing FPP soluble and ascorbic acid prepared to evaluate C10 recovery and co release (EXP 1650).
[0033] Figure 16 shows the dissolution profiles of C10 for enhanced iron tablets containing FPP/FCC and different levels of solubilizing excipients.
[0034] Figure 17 shows the comparative dissolution profiles of Fe with various enhanced iron tablets.
[0035] Figure 18 shows the comparative dissolution profiles of C10 with various enhanced iron tablets.
[0036] Figure 19 shows the mean plasma concentration of baseline corrected plasma iron vs. time (6 hours) profiles of the test items after administration to different groups at different periods in female beagle dogs (n =12).
[0037] Figure 20 shows plasma iron levels at key stages of the study.
[0038] Figure 21 shows a plot of AUC vs. predose plasma iron concentration for ferrous sulfate.
[0039] Figure 22 shows a plot of AUC vs. predose plasma iron concentration for iron Form B.
Detailed Description [0040] The foregoing and other aspects of the present invention will now be described in more detail with respect to the description and methodologies provided herein. It should be appreciated that the invention can be embodied in different forms and should not be construed as limited to the embodiments set forth herein.
Rather, these embodiments are provided so that this disclosure will be thorough and complete, and will fully convey the scope of the invention to those skilled in the art.
[0041] All patents, patent applications and publications referred to herein are incorporated by reference in their entirety. In case of a conflict in terminology, the present specification is controlling.
[0042] The terminology used in the description of the invention herein is for the purpose of describing particular embodiments only and is not intended to be limiting of the invention.
[0043] As used in the description of the embodiments of the invention and the appended claims, the singular forms "a", "an" and "the" are intended to include the plural forms as well, unless the context clearly indicates otherwise.
[0044] As used herein, "and/or" refers to and encompasses any and all possible combinations of one or more of the associated listed items.
[0045] The term "about," as used herein when referring to a measurable value such as an amount of a compound, dose, time, temperature, and the like, is meant to encompass variations of 20%, 10%, 5%, 1%, 0.5%, or even 0.1% of the specified amount.
[0046] The terms "comprises" and/or "comprising," when used in this specification, specify the presence of stated features, integers, steps, operations, elements, and/or components, but do not preclude the presence or addition of one or more other features, integers, steps, operations, elements, components, and/or groups thereof.
[0047] The term "consists essentially of' (and grammatical variants), as applied to the compositions of this invention, means the composition can contain additional components as long as the additional components do not materially alter the composition.
The term "materially altered," as applied to a composition, refers to an increase or decrease in the therapeutic effectiveness of the composition of at least about 20% or more as compared to the effectiveness of a composition consisting of the recited components.
[0048] Unless the context indicates otherwise, it is specifically intended that the various features of the invention described herein can be used in any combination.
[0049] Moreover, the present invention also contemplates that in some embodiments of the invention, any feature or combination of features set forth herein can be excluded or omitted.
[0050] The term "tablet" as used herein includes, but is not limited to, immediate release (IR) tablets, sustained release (SR) tablets, matrix tablets, multilayer tablets, multilayer matrix tablets, extended release tablets, delayed release tablets and pulsed release tablets, any or all of which may optionally be coated with one or more coating materials, including polymer coating materials, such as enteric coatings, rate-controlling coatings, semi-permeable coatings and the like. The term "tablet" also includes osmotic delivery systems in which a drug compound is combined with an osmagent (and optionally other excipients) and coated with a semi-permeable membrane, the semi-permeable membrane defining an orifice through which the drug compound may be released. Tablet solid oral dosage forms that may be useful in the practice of the invention include those selected from the group consisting of IR tablets, SR
tablets, coated IR tablets, coated SR tablets, matrix tablets, coated matrix tablets, multilayer tablets, coated multilayer tablets, multilayer matrix tablets and coated multilayer matrix tablets. In some embodiments, a tablet dosage form is an enteric-coated tablet dosage form. In some embodiments, a tablet dosage form is an enteric-coated rapid onset tablet dosage form.

[0051] The term "capsule" as used herein includes, but is not limited to, IR
capsules, SR capsules, coated IR capsules, and coated SR capsules including delayed release capsules. Capsules may be filled with powders, granules, multiparticulates, tablets, semi-solids, or liquids. In some embodiments, a capsule dosage form is an enteric-coated capsule dosage form. In some embodiments, a capsule dosage form is an enteric-coated rapid onset capsule dosage form. Capsules may be made of hard gelatin, soft gelatin, starch, cellulose polymers, or other materials as known to the art.
[0052] The term "multiparticulate" as used herein means a plurality of discrete particles, pellets, mini-tablets and mixtures or combinations thereof. If the oral form is a multiparticulate capsule, hard or soft gelatin capsules or capsules of other materials can suitably be used to contain the multiparticulate. In some embodiments, a sachet can suitably be used to contain the multiparticulate. In some embodiments, the multiparticulate may be coated with a layer containing rate controlling polymer material.
In some embodiments, a multiparticulate oral dosage form according to the invention may comprise a blend of two or more populations of particles, pellets, or mini-tablets having different in vitro and/or in vivo release characteristics. For example, a multiparticulate oral dosage form may comprise a blend of an instant release component and a delayed release component contained in a suitable capsule.
[0053] In some embodiments, the multiparticulate and one or more auxiliary excipient materials can be compressed into tablet form such as a multilayer tablet. In some embodiments, a multilayer tablet may comprise two layers containing the same or different levels of the same active ingredient having the same or different release characteristics. In some embodiments, a multilayer tablet may contain different active ingredient in each layer. Such a tablet, either single layered or multilayered, can optionally be coated with a controlled release polymer so as to provide additional controlled release properties. In some embodiments, a multiparticulate dosage form comprises a capsule containing delayed release rapid onset minitablets. In some embodiments, a multiparticulate dosage form comprises a delayed release capsule comprising instant release minitablets. In some embodiments, a multiparticulate dosage form comprises a capsule comprising delayed release granules. In some embodiments, a multiparticulate dosage form comprises a delayed release capsule comprising instant release granules. In some embodiments the particulates may all be of uniform composition. In some embodiments, the particulates may vary in composition.
[0054] By the terms "treat," "treating," or "treatment of' (and grammatical variations thereof) it is meant that the severity of the subject's condition is reduced, at least partially improved, or stabilized and/or that some alleviation, mitigation, decrease, or stabilization in at least one clinical symptom and/or parameter is achieved and/or there is a delay in the progression of the disease or disorder.
[0055] The terms "prevent," "preventing," and "prevention" (and grammatical variations thereof) refer to avoidance, prevention and/or delay of the onset of a disease, disorder and/or a clinical symptom(s) in a subject and/or a reduction in the severity of the onset of the disease, disorder and/or clinical symptom(s) relative to what would occur in the absence of the methods of the invention. The prevention can be complete, e.g., the total absence of the disease, disorder and/or clinical symptom(s). The prevention can also be partial, such that the occurrence of the disease, disorder and/or clinical symptom(s) in the subject and/or the severity of onset is less than what would occur in the absence of the present invention.
[0056] An "effective amount," as used herein, refers to an amount that imparts a desired effect, which is optionally a therapeutic or prophylactic effect.
[0057] A "treatment effective" amount, as used herein, is an amount that is sufficient to provide some improvement or benefit to the subject.
Alternatively stated, a "treatment effective" amount is an amount that will provide some alleviation, mitigation, decrease, or stabilization in at least one clinical symptom in the subject.
Those skilled in the art will appreciate that the therapeutic effects need not be complete or curative, as long as some benefit is provided to the subject.
[0058] A "prevention effective" amount, as used herein, is an amount that is sufficient to prevent and/or delay the onset of a disease, disorder and/or clinical symptoms in a subject and/or to reduce and/or delay the severity of the onset of a disease, disorder and/or clinical symptoms in a subject relative to what would occur in the absence of the methods of the invention. Those skilled in the art will appreciate that the level of prevention need not be complete, as long as some benefit is provided to the subject.
[0059] As used herein, a "therapeutically effective" or "therapeutically acceptable" amount refers to an amount that will elicit a therapeutically useful response in a subject. The therapeutically useful response may provide some alleviation, mitigation, or decrease in at least one clinical symptom in the subject. The terms also include an amount that will prevent or delay at least one clinical symptom in the subject and/or reduce and/or delay the severity of the onset of a clinical symptom in a subject relative to what would occur in the absence of the methods of the invention. Those skilled in the art will appreciate that the therapeutically useful response need not be complete or curative or prevent permanently, as long as some benefit is provided to the subject.

[0060] "Subjects" according to the present invention include mammals, avians, reptiles, amphibians, and fish. In some embodiments, the subjects are mammalian subjects including, but not limited to, humans, non-human mammals, non-human primates (e.g., monkeys, chimpanzees, baboons, etc.), dogs, cats, mice, hamsters, rats, horses, cows, pigs, rabbits, sheep, and goats. Avian subjects include, but are not limited to, chickens, turkeys, ducks, geese, quail, and pheasant, and birds kept as pets (e.g., parakeets, parrots, macaws, cockatoos, and the like). In particular embodiments, the subject is from an endangered species. In particular embodiments, the subject is a laboratory animal. Human subjects include neonates, infants, juveniles, adults, and geriatric subjects. In certain embodiments, the subject is in need of the methods of the present invention, e.g., has an iron deficiency. In other embodiments, the subject has, may have, or is at risk for an iron deficiency.
[0061] The phrase "the composition does not involve chemical modification of the iron compound" refers to the fact that the iron compound is not chemically modified by or chemically interact with (i.e., form covalent or ionic bonds) any component in the composition during preparation, storage, and use of the composition.
[0062] A "tight ion binding pair," as used herein, refers to a pair of ions that are, at physiological pH and in an aqueous environment, not readily interchangeable with other loosely paired or free ions that may be present in the environment of the tight ion binding pair. A tight ion binding pair can be experimentally detected by noting the absence of an interchange of a member of the pair with another ion, at physiologic pH
and in an aqueous environment, using isotopic labeling and NMR or mass spectroscopy.
Tight ion binding pairs also can be found experimentally by noting the lack of separation of the ion pair, at physiologic pH and in an aqueous environment, using reverse phase HPLC.
[0063] An "iron compound," as used herein, refers to a complex comprising elemental iron and an additional atom, ion, or molecule, and includes iron salts, iron chelates, iron complexes, and polymer-bound iron.
[0064] An "iron complex," as used herein, refers to elemental iron in neutral or cationic form covalently or electrostatically linked to an additional atom, ion, or molecule.
[0065] An "iron chelate," as used herein, refers to an iron cation and anions that surround the iron cation and are joined to it by electrostatic bonds.
[0066] An "active agent," as used herein, refers to a compound or molecule that has a therapeutic, prophylactic, or nutritive effect when delivered to a subject.
[0067] A "disease or disorder characterized by an iron deficiency," as used herein, refers to any disease or disorder in which whole body stores of iron are less than desired.
Low body stores may be indicated by various symptoms including a blood level of iron that is below normal, low ferritin levels, and/or low hemoglobin levels.
Exemplary low levels may be a cause and/or symptom of the disease or disorder and includes any disease or disorder in which elevating iron levels in the subject, e.g., in the blood, treats and/or prevents one or more symptoms of the disease or disorder, or where maintenance of iron indices is required for effectiveness of another agent, e.g., erythropoiesis-stimulating agents. The normal serum iron level for human adults is considered to be about 50 to about 170 Itg/dL.
[0068] The present invention provides a pharmaceutical composition for oral administration comprising, consisting essentially of, or consisting of an iron compound (e.g., an iron salt, chelate, complex, or polymer bound) and an absorption enhancer, wherein the absorption enhancer is a medium chain fatty acid salt having a carbon chain length of from about 4 to about 20 carbon atoms. In some embodiments, the iron compound is an iron chelate or iron complex. In some embodiments, the composition does not involve chemical modification of the iron compound. In some embodiments, the iron compound and the absorption enhancer are not part of a tight ion binding pair. In some embodiments, the use of an iron chelate or iron complex in which iron is tightly bound to another moiety prevents the formation of a tight ion binding pair between the iron and the fatty acid. In certain embodiments, the iron compound is present in the composition in a treatment effective amount. In other embodiments, the iron compound is present in the composition in a prevention effective amount. In certain embodiments, the pharmaceutical composition comprises, consists essentially of, or consists of one or more auxiliary excipients in addition to the iron compound and absorption enhancer.
[0069] In a further aspect, the invention relates to a pharmaceutical composition for oral administration comprising, consisting essentially of, or consisting of an ,iron compound (e.g., an iron salt, chelate, complex, or polymer bound) and an absorption enhancer, wherein the composition provides a bioavailability of iron that is at least 1.5 times greater than the bioavailability provided by conventional oral iron preparations (e.g., a composition comprising ferrous sulfate (e.g., a ferrous sulfate tablet) or any other currently available iron dosage form), e.g., one that does not contain an absorption enhancer. In certain embodiments, the bioavailability of iron is at least 1.5, 2, 3, 4, 5, 6, 7, 8, 9, or 10 times greater or more than the bioavailability provided by conventional oral iron preparations.
[0070] In some embodiments, the iron compound is an iron salt (e.g., a ferrous salt or a ferric salt), an iron chelate, an iron complex, or polymer bound iron. In certain embodiments, the iron compound is a ferric compound, as it may produce less side effects than ferrous compounds.
[0071] Iron salts include, but are not limited to, ferrous sulfate, ferrous gluconate, ferrous fumarate, ferric hypophosphite, ferric albuminate, ferric chloride, ferric citrate, ferric oxide saccharate, ferric ammonium citrate, ferrous chloride, ferrous iodide, ferrous lactate, ferric trisglycinate, ferrous bisglycinate, ferric nitrate, ferrous hydroxide saccharate, ferric sulfate, ferric gluconate, ferric aspartate, ferrous sulfate heptahydrate, ferrous phosphate, ferric ascorbate, ferrous formate, ferrous acetate, ferrous malate, ferrous glutamate, ferrous cholinisocitrate, ferroglycine sulfate, ferric oxide hydrate, ferric pyrophosphate soluble, ferric hydroxide saccharate, ferric manganese saccharate, ferric subsulfate, ferric ammonium sulfate, ferrous ammonium sulfate, ferrous ethylenediammonium sulfate tetrahydrate, ferric sesquichloride, ferric choline citrate, ferric manganese citrate, ferric quinine citrate, ferric sodium citrate, ferric sodium edetate, ferric formate, ferric ammonium oxalate, ferric potassium oxalate, ferric sodium oxalate, ferric peptonate, ferric manganese peptonate, ferric acetate, ferric fluoride, ferric phosphate, ferric pyrophosphate, ferrous pyrophosphate, ferrous carbonate saccharate, ferrous carbonate mass, ferrous succinate, ferrous citrate, ferrous tartrate, ferric fumarate, ferric succinate, ferrous hydroxide, ferrous nitrate, ferrous carbonate, ferric sodium pyrophosphate, ferric tartrate, ferric potassium tartrate, ferric subcarbonate, ferric glycerophosphate, ferric saccharate, ferric hydroxide saccharate, ferric manganese saccharate, ferrous ammonium sulfate, ferric sodium pyrophosphate, ferrous carbonate, ferric hydroxide, ferrous oxide, ferric oxyhydroxide, ferrous oxalate, and/or combinations thereof.
[0072] Iron chelates and complexes include, but are not limited to, ferric pyrophosphate, soluble ferric pyrophosphate, iron polysaccharide, iron bis glycinate, iron proteinate, methylidine-iron complex, EDTA-iron complex, phenanthrolene iron complex, p-toluidine iron complex, ferrous saccharate complex, ferrlecit, ferrous gluconate complex, ferrum vitis, ferrous hydroxide saccharate complex, iron-arene sandwich complexes, acetylacetone iron complex salt, iron-dextran complex, iron-dextrin complex, iron-maltodextrin complex, iron-sorbitol-citric acid complex, saccharated iron oxide, ferrous fumarate complex, iron porphyrin complex, iron phtalocyamine complex, iron cyclam complex, dithiocarboxy-iron complex, desferrioxamine-iron complex, bleomycin-iron complex, ferrozine-iron complex, iron perhaloporphyrin complex, alkylenediamine-N,N'-disuccinic acid iron(III) complex, hydroxypyridone-iron(III) complex, aminoglycoside-iron complex, transferrin-iron complex, iron thiocyanate complex, iron complex cyanides, porphyrinato iron(III) complex, polyaminopolycarbonate iron complexes, dithiocarbamate iron complex, adriamycin iron complex, anthracycline-iron complex, MGD-iron complex, ferrioxamine B, ferrous citrate complex, ferrous sulfate complex, ferric gluconate complex, ferrous succinate complex, polyglucopyranosyl iron complex, polyaminodisuccinic acid iron complex, biliverdin-iron complex, deferiprone iron complex, ferric oxyhydride-dextran complex, dinitrosyl dithiolato iron complex, iron lactoferrin complexes, 1,3-PDTA ferric complex salts, diethylenetriaminepentaacetic acid iron complex salts, cyclohexanediaminetetraacetic acid iron complex salts, methyliminodiacetic acid iron complex salts, glycol ether diaminetetraacetic acid iron complex salts, ferric hydroxypyrone complexes, ferric succinate complex, ferric chloride complex, ferric glycine sulfate complex, ferric aspartate complex, sodium ferrous gluconate complex, ferrous hydroxide polymaltose complex, and/or combinations thereof.
[0073] Suitable iron compounds for use in the pharmaceutical compositions of the invention can be determined using techniques well known in the art and described herein. For example, the centrifugation techniques described in Example 2 below can be used to evaluate the compatibility of iron compounds and enhancers.
Additionally, compatibility can be evaluated by determining the ability of an iron compound and an enhancer to form mixed micelles.
[0074] In certain embodiments of the invention, iron compound is the only active agent in the composition. In other embodiments, the composition comprises additional active agents. In one example, the additional active agents are useful for treating iron deficiency or disorders associated with iron deficiency. In other examples, the additional active agents are agents that are beneficially administered with iron, e.g., erythropoiesis-stimulating agents such as erythropoietin, epoetin alfa (PROCRIT/EPOGEN), epoetin beta (NEORECORMON), darbepoetin alfa (ARANESP), and methoxy polyethylene glycol-epoetin beta (MIRCERA). In other examples, the additional active agents are nutrients (e.g., vitamins and/or minerals) that provide the daily recommended amounts of nutrients and/or disorder treatment and/or prevention effective amounts. In some embodiments, the additional active agent is selected from the group consisting of folic acid, vitamin A, vitamin B (all series, including Bl, B2, B3, B5, B6, B9, B12), vitamin C, vitamin D, vitamin E, calcium, chromium, copper, magnesium, manganese, potassium, selenium, zinc, phosphorus, iodine, biotin. inositol, para-amino benzoic acid, choline, and any combination thereof. As used herein, the term "vitamin Bl" refers to thiamine. As used herein, the term "vitamin B2" refers to riboflavin. As used herein, the term "vitamin B3" refers to niacin and nicotinic acid. As used herein, the term "vitamin B5"
refers to pantothenic acid. The term "vitamin B6" refers to pyridoxal, pyridoxamine and pyridoxine compounds. As used herein, the term "vitamin B9" refers to folic acid. The term "vitamin B12" refers to all forms of cobalamin including, without limitation, hydroxocobalamin, cyanocobalamin and methylcobalamin. The term "vitamin C" is used herein to refer to any form of vitamin C, including ascorbate and L-threonate.
The term "vitamin D" is used to refer to both cholecalciferol (vitamin D3) and ergocalciferol (vitamin D2). The term "vitamin E" is used herein to refer to alpha-tocopherol, d-alpha-tocopherol, d-alpha-tocopheryl succinate (or acetate), dl-alpha-tocopherol, dl-alpha-tocopheryl acetate (or succinate), gamma tocopherol, mixed tocopherols, and dl-alpha tocopherol nicotinate. The term "calcium" is used herein to refer to any form of calcium including calcium carbonate, phosphate, lactate, gluconate, citrate and combinations thereof. The term "magnesium" is used herein to refer to any form of magnesium, including magnesium oxide, magnesium chloride, magnesium lactate, magnesium sulfate and magnesium gluconate.
[0075] In some embodiments, the enhancer is a medium chain fatty acid salt which has a carbon chain length of from about 4 to about 20 carbon atoms, e.g., from 6 to 20 carbon atoms, e.g., from 8 to 14 carbon atoms. In some embodiments, the enhancer is solid at room temperature. In certain embodiments, the medium chain fatty acid salt is the only absorption enhancer in the oral dosage form. In additional embodiments, the pharmaceutical composition comprises more than one medium chain fatty acid salt, e.g., 2, 3, 4, or more medium chain fatty acid salts. In other embodiments, the enhancer is a sodium salt of a medium chain fatty acid. In some embodiments, the enhancer is selected from the group consisting of sodium caprylate, sodium caprate and sodium laurate.
Exemplary enhancers are further described in U.S. Patent Nos. 7,658,938 and 7,670,626 and U.S. Published Application Nos. 2003/0091623 and 2007/0238707, which are incorporated by reference in their entirety. In some embodiments, the absorption enhancer is present in a ratio of from 1:100,000 to 10:1 (iron:enhancer).
[00761 One aspect of the invention relates to a solid oral dosage form comprising, consisting essentially of, or consisting of the pharmaceutical composition of the invention.
The dosage form can comprise an amount of iron that is treatment effective (e.g., for treatment of iron deficiency and/or a disease or disorder characterized by iron deficiency) or prevention effective (e.g., to provide the daily recommended requirement of iron and/or prevent the onset of disease or disorders characterized by iron deficiency). In one embodiment, the solid oral dosage form comprises about 1 mg to about 200 mg iron, e.g., about 1, 2, 3, 4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, or 200 mg or more of iron or any range therein. The amount of iron in the composition refers to the weight of the elemental iron, not the iron compound, present in the composition.
[0077] In certain embodiments, the dosage form is a solid oral dosage form (e.g., a tablet, capsule, multiparticulate, or powder dosage form) or a liquid oral dosage form (e.g., a solution, suspension, emulsion, syrup, or elixir). In some embodiments, the dosage form is a delayed release or controlled release dosage form. For example, the dosage form can be a tablet or other form comprising a rate controlling polymer material, e.g., hydroxypropyl methylcellulose, a polymer of acrylic or methacrylic acid or their respective esters, or copolymers of acrylic or methacrylic acid or their respective esters.
[0078] In some embodiments, the iron compound, absorption enhancer, and at least one auxiliary excipient are compressed into tablet form prior to coating with a rate controlling polymer. In some embodiments, the iron compound, absorption enhancer, and at least one auxiliary excipient are compressed into tablet form prior to coating with a delayed release polymer. In some embodiments, the iron compound, absorption enhancer, rate controlling polymer, and at least one auxiliary excipient are compressed to form a controlled release matrix tablet. In some embodiments, the controlled release matrix tablet is coated with a rate-controlling polymer. In some embodiments, the controlled release matrix is coated with a delayed release polymer. In some embodiments, the iron compound, absorption enhancer, and at least one auxiliary excipient are compressed into the form of a multilayer tablet prior to coating with a rate controlling-polymer. In some embodiments, the iron compound, absorption enhancer, and at least one auxiliary excipient are compressed into the form of a multilayer tablet prior to coating with a delayed release polymer. Yet, in another embodiment, the iron compound and absorption enhancer are dispersed in the rate-controlling polymer material and compressed into the form of a multilayer tablet. In some embodiments, the multilayer tablet is coated with a rate-controlling polymer. In some embodiments, the multilayer tablet is coated with a delayed release polymer.
[0079] In certain embodiments, the iron compound, absorption enhancer, at least one auxiliary excipient, and the rate-controlling polymer material are combined into a multiparticulate form. In some embodiments, the multiparticulate form comprises discrete particles, pellets, minitablets, or combinations thereof. In some embodiments, the dosage form of the present invention comprises a blend of two or more populations of particles, pellets or mini-tablets having different in vitro or in vivo release characteristics.
In some embodiments, the multiparticulate is encapsulated in capsules, e.g., hard or soft gelatin capsules. In another embodiment, the capsule is coated with a rate-controlling polymer. In some embodiments, the capsule is coated with a delayed release polymer. In some embodiments, the multiparticulate is incorporated into a sachet.
[0080] In some embodiments, the discrete particles or pellets are compressed into tablet form. In some embodiments, the tablet form is coated with a rate controlling polymer material. Yet, in another embodiment, the tablet form is coated with a delayed release polymer. In some embodiments, the discrete particles or pellets are compressed into a multilayer tablet. In some embodiments, the multilayer tablet is coated with a rate controlling material. In some embodiments, the multilayer tablet is coated with a delayed release polymer.
[0081] In any of the above-mentioned embodiments, a controlled release coating (e.g., an enteric coating) may be applied to the final dosage form (capsule, tablet, multilayer tablet, etc.). The controlled release coating may typically comprise a rate controlling polymer material as defined above. The dissolution characteristics of such a coating material may be pH dependent or independent of pH.
[0082] The pharmaceutical compositions and oral dosage forms of the invention can comprise one or more auxiliary excipients, such as for example rate-controlling polymeric materials, solubilizers, diluents, lubricants, disintegrants, plasticizers, anti-tack agents, opacifying agents, glidants, pigments, flavorings, and the like. As will be appreciated by those skilled in the art, the exact choice of excipients and their relative amounts will depend to some extent on the final dosage form.
[0083] One excipient that can be included in the composition is one or more saccharides. Any suitable saccharide may be used in the composition of the present invention. As used herein, the "saccharides" used in the invention include, without limitation, sugar alcohols, monosaccharides, disaccharides, and oligosaccharides.
Exemplary sugar alcohols include, but are not limited to, xylitol, mannitol, sorbitol, erythritol, lactitol, pentitol, and hexitol. Exemplary monosaccharides include, but are not limited to, glucose, fructose, aldose and ketose. Exemplary disaccharides include, but are not limited to, sucrose, isomalt, lactose, trehalose, and maltose. Exemplary oligosaccharides include, but are not limited to, fructo-oligosaccharides, inulin, galacto-ologosaccharides, and mannan-oligosaccharides. In some embodiments, the saccharide is sorbitol, mannitol, or xylitol. In some embodiments, the saccharide is sorbitol. In some embodiments, the saccharide is sucrose.
[0084] Any suitable amounts of saccharide may be added in the compositions of the present invention. In some embodiments of the present invention, the ratio of the enhancer and saccharide may be adjusted to achieve a desired dissolution rate and/or compressibility of the resulting pharmaceutical composition. In some embodiments, the ratio of the enhancer and saccharide is 2:1 to 20:1. According to some embodiments, the ratio of the enhancer and saccharide is about 4:1 to 6:1. In another embodiment, the ratio of the enhancer and saccharide is about 5:1.
[0085] Any suitable grade of saccharide may be used in the composition of the present invention. However, in some embodiments, the selection of the grade of saccharide may be dependent upon the particle size distribution (PSD) of a specific grade of saccharide. Further, in another embodiment, the specific grade of the saccharide may affect the characteristics of the resulting pharmaceutical composition such as dissolution rate and/or compressibility. In some embodiments, the selection of the grade of saccharide is dependent upon the PSD of other excipients and the therapeutically active ingredient. In some embodiments, the saccharide is Parteck SI 150 (Merck KGaA, Darmstadt, Germany), a directly compressible sorbitol. In other embodiments, the saccharide is Parteck SI 400 (Merck KGaA, Darmstadt, Germany).
[0086] Suitable diluents include, for example, pharmaceutically acceptable inert fillers such as microcrystalline cellulose, lactose, dibasic calcium phosphate, saccharides, and/or mixtures of any of the foregoing. Examples of diluents include microcrystalline cellulose such as that sold under the Trademark Avicel (FMC Corp., Philadelphia, Pa.), for example, AvicelTM pH101, AvicelTM pH102 and AvicelTM pH112; lactose such as lactose monohydrate, lactose anhydrous and Pharmatose DCL21; dibasic calcium phosphate such as Emcompress; mannitol; starch; sorbitol; sucrose; glucose;
and combinations and mixtures thereof.
[0087] Suitable lubricants, including agents that act on the flowability of the powder to be compressed are, for example, colloidal silicon dioxide such as AerosilTM
200; talc; stearic acid; magnesium stearate; calcium stearate; and combinations and mixtures thereof.
[0088] Suitable disintegrants include, for example, lightly crosslinked polyvinyl pyrrolidone, corn starch, potato starch, maize starch and modified starches, croscarmellose sodium, crospovidone, sodium starch glycolate, and corn binations and mixtures thereof.
[0089] The term "rate controlling polymer material" as used herein includes hydrophilic polymers, hydrophobic polymers and mixtures of hydrophilic and/or hydrophobic polymers that are capable of controlling or retarding the release of the peptide or protein from a solid oral dosage form of the present invention.
Suitable rate controlling polymer materials include those selected from the group consisting of hydroxyalkyl cellulose such as hydroxypropyl cellulose and hydroxypropyl methyl cellulose; poly(ethylene) oxide; alkyl cellulose such as ethyl cellulose and methyl cellulose; carboxymethyl cellulose; hydrophilic cellulose derivatives;
polyethylene glycol; polyvinylpyrrolidone; cellulose acetate; cellulose acetate butyrate;
cellulose acetate phthalate; cellulose acetate trimellitate; polyvinyl acetate phthalate;
hydroxypropylmethyl cellulose phthalate; hydroxypropylmethyl cellulose acetate succinate; polyvinyl acetaldiethylamino acetate; poly(alkylmethacrylate) and poly (vinyl acetate). Other suitable hydrophobic polymers include polymers and/or copolymers derived from acrylic or methacrylic acid and their respective esters, zein, waxes, shellac and hydrogenated vegetable oils. Particularly useful in the practice of the present invention are poly acrylic acid, poly acrylate, poly methacrylic acid and poly methacrylate polymers such as those sold under the Eudragit tradename (Rohm GmbH, Darmstadt, Germany) specifically Eudragit L, Eudragit S, Eudragit RL, and Eudragit RS coating materials and mixtures thereof. Some of these polymers can be used as delayed release polymers to control the site where the drug is released. They include poly methacrylate polymers such as those sold under the Eudragit tradename (Rohm GmbH, Darmstadt, Germany) specifically Eudragit L, Eudragit S, Eudragit RL, and Eudragit RS coating materials and mixtures thereof.
[0090] The pharmaceutical composition can further comprise a solubilizer as an excipient. The term "solubilizer," as used herein, refers to any compound that improves the solubility of iron compounds in vitro, and includes organic chelation agents such as, without limitation, citric acid or salts thereof, ascorbic acid and salts thereof, EDTA, or any combination thereof. In one embodiment, the solubilizer is citric acid and/or sodium citrate. The solubilizer can be present in the pharmaceutical composition in an amount sufficient to increase the solubility of the iron compound and/or the absorption enhancer.
In some embodiments, the solubilizer is present in an amount of about 2% to about 25%
by weight, e.g., about 5% to about 20%, e.g., about 2, 3, 4, 5, 6, 7, 8, 9, 10, 11, 12, 13, 14, 15, 16, 17, 18, 19, 20, 21, 22, 23, 24, or 25%.
[0091] Without being limited to a specific mechanism, it is believed that the presence of a solubilizer improves the dissolution rate of both the iron compound and the enhancer in the compositions of the invention. However, as the ratio of solubilizer to iron compound increases past a certain point, the dissolution rate of the iron compound and the enhancer start to diverge such that the desired co-dissolution of the iron compound and the enhancer is no longer present. This is due to the continued increase in dissolution rate of iron compound as the ratio increases while the dissolution rate of the enhancer states to slow down and then decrease as the ratio increase. The ideal ratio will be different for each iron compound and solubilizer. The differential effect of solubilizer on the dissolution rates of the iron compound and the enhancer is surprising and unexpected.
Thus, in certain embodiments, solubilizer is present in the pharmaceutical compositions of the invention in an amount sufficient to improve the dissolution rate of both the iron compound and the enhancer yet still maintain a substantially similar release of the iron compound and the enhancer.
[0092] As used herein, the term "substantially similar release" is defined as a ratio of the time for a percentage of the iron compound to be released from a dosage form without coating to the time for the same percentage of the enhancer to be released in the range of about 2.0 to about 0.5, e.g., about 1.3 to about 0.7, e.g., about 1.1 to about 0.9.
In other embodiments, the term "substantially similar release" is defined as a ratio of the time for a percentage of the iron compound to be released from a dosage form with a coating (e.g., an enteric coating or other type of delayed release or sustained release coating) to the time for the same percentage of the enhancer to be released in the range of about 1.3 to about 0.7. To be considered substantially similar, the ratio must fall within the range at least two different time points, e.g., at least 3, 4, or 5 different time points. In one embodiment, the dissolution is carried out in 900 mL pH 6.8 phosphate buffer at 37 C
with a USP Paddle Apparatus at 50 rpm. In one embodiment, the dissolution assay includes a preliminary step of acid treatment (e.g., 2 hrs in 0.1 N HCI). For example, if the iron compound has a dissolution of 80% in about 20 minutes, sodium caprate (enhancer) must have a dissolution of 80% in the range of about 14 minutes to 26 minutes to be substantially similar. In one embodiment, the ratio is in the range of about 1.1 to about 0.9. For example, if the iron compound has a dissolution of 80% in about minutes, sodium caprate (enhancer) must have a dissolution of 80% in the range of about 18 minutes to about 22 minutes.
[0093] In some embodiments, the solubilizer is present in the pharmaceutical composition in an amount sufficient to increase the dissolution rate of at least one of the iron compound and the enhancer by at least about 5%, e.g., at least about 10%, 15% , 20%, or more. In other embodiments, the solubilizer is present in the pharmaceutical composition in an amount such that the iron compound and the enhancer have a substantially similar release. In certain embodiments, the solubilizer is present in the pharmaceutical composition in an amount sufficient to enhance the dissolution rate such that both the iron compound and the enhancer achieve at least about 80%
dissolution in 3 hours, e.g., at least about 80% dissolution in 2.5,2, 1.5, or 1 hour.
[0094] The pharmaceutical compositions and oral dosage forms of the invention can comprise liquid oral dosage forms (solutions, syrups, suspensions, elixirs, emulsions, etc.) or powder oral dosage forms (either for reconstitution or ingestion). As will be appreciated by those skilled in the art, the exact choice of excipients and their relative amounts will depend to some extent on the final dosage form.
[0095] It is well known that iron absorption occurs predominantly via active transport in the duodenum and upper jejunum. A feedback mechanism exists that increases iron absorption in subjects that are iron deficient. In subjects with iron overload, this feedback mechanism dampens iron absorption.
[0096] Data collected in Example 12 were evaluated to determine the relationship between iron absorption and the iron levels at baseline for each of the study periods. It was found in this analysis that the amount of iron absorbed from the unenhanced formulation decreased as the iron levels at baseline increased. Fig. 21 demonstrates this relationship. The unenhanced AUCs vs. predose iron levels displays a clear relationship between predose iron levels and absorbed iron. Animals with higher predose iron levels absorbed less iron from the unenhanced test formulation. This fits with the theory that iron is absorbed by active transport and there is a feedback mechanism.
[0097] In contrast, for the enhanced formulation, this decrease in absorption with increased baseline iron levels was not similarly seen. In fact, the dog with the third highest baseline level was the second highest absorbing subject. Fig. 22, the plot of enhanced iron AUCs vs. predose iron levels, demonstrates this. This is likely a result of paracellular and transcellular absorption of iron promoted by the enhanced formulation caused by the formation of mixed micelles of the iron and the enhancer system.
Hence, while absorption of iron by active transport is suppressed when predose iron levels rise, absorption by paracellular and transcellular absorption promoted by enhancers of the invention is not affected by negative feedback. Without being limited to a specific mechanism, it may be that the ability of the compositions of the invention to overcome the negative feedback mechanism for iron absorption is at least in part responsible for the enhanced bioavailability of iron.
[0098] One aspect of the present invention relates to a method of orally delivering elemental iron contained in an iron compound to a subject, comprising administering to the subject the oral dosage form of the invention. Another aspect of the present invention relates to a method for increasing the level of iron in a subject, e.g., in the blood and/or stored as ferritin in bone marrow, liver, and/or spleen, comprising administering to the subject the oral dosage form of the invention. Another aspect of the present invention relates to a method for increasing the level of hemoglobin in the blood of a subject, comprising administering to the subject the oral dosage form of the invention.
The subject can be a subject in need thereof, e.g., a subject having an iron deficiency, a subject at risk of an iron deficiency, or a subject that desires to maintain normal blood and/or tissue levels of iron. In some embodiments, the methods of the invention can increase the level of iron in the blood by at least about 5 WA e.g., at least about 10, 20, 30, 40, 50, 60, 70, 80, 90, or 100 midi or more. In some embodiments, the methods of the invention can increase the level of hemoglobin in the blood by at least about 0.5 g/dl, e.g., at least about 1, 2, 3, 4, or 5 g/dl or more. Methods for measuring the level of iron and hemoglobin in the blood are well known in the art.
[0099] One aspect of the present invention relates to a method for treating or preventing an iron deficiency in a subject in need thereof, comprising administering to the subject the oral dosage form of the invention. Another aspect of the present invention relates to a method for treating or preventing a disease or disorder characterized by an iron deficiency in a subject in need thereof, comprising administering to the subject the oral dosage form of the invention.
[0100] In certain embodiments, the method treats or prevents various anemic states. In some embodiments, the anemia is an iron deficiency anemia, such as that associated with chronic blood loss, acute blood loss, pregnancy, childbirth, childhood development, psychomotor and cognitive development in children, breath holding spells, heavy uterine bleeding, menstruation, chronic recurrent hemoptysis, idiopathic pulmonary siderosis, chronic internal bleeding, gastrointestinal bleeding, parasitic infections, chronic kidney disease, dialysis, surgery or acute trauma, and chronic ingestion of alcohol, chronic ingestion of salicylates, chronic ingestion of steroids, chronic ingestion of non-steroidal anti-inflammatory agents, or chronic ingestion of erythropoiesis stimulating agents. In some aspects, the anemia is anemia of chronic disease, such as rheumatoid arthritis, cancer, Hodgkin's lymphoma, non-Hodgkin's lymphoma, cancer chemotherapy, inflammatory bowel disease, ulcerative colitis, thyroiditis, hepatitis, systemic lupus erythematosus, polymyalgia rheumatica, sclerodenna, mixed connective tissue disease, Sjogren's syndrome, congestive heart failure/cardiomyopathy, or idiopathic geriatric anemia. In some embodiments, the anemia is due to impaired iron absorption or poor nutrition, such as anemia associated with Crohn's disease, gastric surgery, ingestion of drug products that inhibit iron absorption, and chronic use of calcium. In some embodiments, the disorder is a functional iron deficiency as can occur where there is a failure to release iron rapidly enough to keep pace with the demands of the bone marrow for erythropoiesis (e.g., subjects on erythropoietin therapy). In other embodiments, the method treats restless leg syndrome, blood donation, Parkinson's disease, hair loss, or attention deficit disorder.
[0101] Identification of subjects in need of treatment can be carried out by methods known to those of skill in the art. For example, need can be assessed by monitoring a subject's iron status. The diagnosis of iron deficiency can be based on appropriate laboratory tests, for example, hemoglobin (Hb), serum ferritin, serum iron, transferrin saturation (TfS), and hypochromic red cells. Additional techniques for measuring iron levels are disclosed in U.S. Patent Nos. 7,659,074, 7,609,369, 7,601,684, 7,412,275, 7,361,512, and 7,361,510, each herein incorporated by reference in its entirety.
[0102] In another aspect, the invention relates to a method for maintaining iron indices in a subject to ensure maximum efficacy of erythropoietin and/or other erythropoiesis-stimulating agents (ESA), comprising administering to the subject the oral dosage form of the invention. The subject may be one that is currently being administered ESA or will be administered ESA. In some embodiments, the oral dosage form of the invention is administered to the subject before, during, and/or after the administration of ESA. The oral dosage form may be administered in an amount to prevent a decrease in blood levels of iron during ESA administration and/or to increase blood levels of iron to the level that existed in the subject prior to administration of ESA.
[0103] In one embodiment of the invention, the oral dosage form of the invention is administered to the subject as needed to raise serum iron levels and/or treat and/or prevent a disorder. The dosage form can be administered continuously or intermittently.
In one embodiment, the dosage form is administered to the subject more than once a day, e.g., 2, 3, 4, or more times a day or once every 1, 2, 3, 4, 5, 6, or 7 days.
In another embodiment, the dosage form is administered to the subject no more than once a week, e.g., no more than once every two weeks, once a month, once every two months, once every three months, once every four months, once every five months, once every six months, or longer. In a further embodiment, the dosage form is administered using two or more different schedules, e.g., more frequently initially (for example to build up to a certain level, e.g., once a day or more) and then less frequently (e.g., once a week or less).
In other embodiments, the dosage form can be administered by any discontinuous administration regimen. In one example, the dosage form can be administered not more than once every two days, every three days, every four days, every five days, every six days, every seven days, every eight days, every nine days, or every ten days, or longer.
The administration can continue for one, two, three, or four weeks or one, two, or three months, or longer. Optionally, after a period of rest, the dosage form can be administered under the same or a different schedule. The period of rest can be one, two, three, or four weeks, or longer, according to the pharmacodynamic effects of the dosage form on the subject.
[0104] The oral dosage form is delivered to the subject at a dose that is effective to raise serum iron levels, raise Hb levels, and/or treat and/or prevent a disorder. The effective dosage will depend on many factors including the gender, age, weight, and general physical condition of the subject, the severity of the disorder, the particular composition being administered, the duration of the treatment, the nature of any concurrent treatment, the carrier used, and like factors within the knowledge and expertise of those skilled in the art. As appropriate, a treatment effective amount in any individual case can be determined by one of skill in the art by reference to the pertinent texts and literature and/or by using routine experimentation (see, e.g., Remington, The Science and Practice of Pharmacy (21st ed. 2005)). In one embodiment, the dosage form is administered at a dose of about 1 to about 200 mg iron, e.g., about 1, 2, 3,4, 5, 6, 7, 8, 9, 10, 15, 20, 25, 30, 35, 40, 45, 50, 55, 60, 65, 70, 75, 80, 85, 90, 95, 100, 110, 120, 130, 140, 150, 160, 170, 180, 190, or 200 mg or more of iron. In some instances, the dose can be even lower, e.g., as low as 0.001 or 0. 1 mg or lower. In some instances, the dose can be even higher, e.g., as high as 500 or 1000 mg or higher. The present invention encompasses every sub-range within the cited ranges and amounts.
[0105] Another aspect of the present invention provides a process for manufacturing a solid oral dosage form of a pharmaceutical composition comprising the steps of: a) blending iron with an absorption enhancer, and optionally auxiliary excipients to form a blend; wherein the enhancer is a medium chain fatty acid salt having a carbon chain length of from about 4 to about 20 carbon atoms; and b) forming a solid oral dosage from the blend by i) directly compressing the blend to form the solid oral dosage form, or ii) granulating the blend to form a granulate for incorporation into the solid oral dosage form, or iii) spray drying the blend to form a multiparticulate for incorporation into the solid oral dosage form. In some embodiments, the iron and the enhancer are blended in a ratio of from 1:100,000 to 10:1 (iron:enhancer).
[0106] The present invention will now be described in more detail with reference to the following examples. However, these examples are given for the purpose of illustration and are not to be construed as limiting the scope of the invention.
[0107] In the examples, it can be seen that formulations of the present invention can achieve a bioavailability increase for sodium iron EDTA and soluble iron pyrophosphate of more than five-fold over unenhanced formulations.

Pharmacokinetic studies of sodium iron EDTA complex [0108] Pre-formulation experiments indicated no gross incompatibility between the proposed sodium caprate formulation (55 mg/mL) and the sodium EDTA iron chelate (Table 1). Visual observation for two weeks at ambient temperature indicated good solution stability for all formulations, except sample 5 and 6 (5.43 and 8.14 mg elemental iron), where some precipitation was noticed. The data also indicated that this iron compound is amenable to an enhanced iron formulation with an acceptable dose solubility of elemental iron concentration required for clinical use. In-house analytical methods were not carried out to assay iron content but the development was based on label claim elemental iron content. The final formulations dosed in dogs were assayed for iron content carried out by a contract laboratory.
Table 1: Solution stability of sodium EDTA Fe+3 chelate with sodium caprate Concentration mg/mL
Sample No Sodium caprate EDTA Salt Elemental Iron 1 55 3.26 0.47 2 55 6.50 0.93 3 55 9.77 1.40 4 55 19.00 2.71 55 38.00 5.43 6 55 57.00 8.14 [0109] Based on the above results, the following formulations of iron complex, ethylenediaminetetraacetic acid ferric sodium (or an EDTA chelate) were prepared prior to in vivo dosing. Iron concentration was based on label claim of elemental iron (Product No E6760, batch No. 0191(01541) and was assumed to be pure. The test items were stored in glass containers. The formulations were shipped at ambient temperature to contract analytical centre for the assay of iron content.
1. Control unenhanced formulation: Aqueous formulation of iron complex alone (0.783 mg/mL elemental iron) in de-ionized water.
2. Enhanced iron test formulation A: Aqueous formulation of iron complex (0.783 mg/mL elemental iron) in 55 mg/mL sodium caprate aqueous solution.
[0110] A two-way crossover pre-clinical study was carried out in an iron deficient model of beagle dogs. Eight adult female non-naïve intra-duodenally (ID) cannulated beagle dogs (5.3-7.2 kg) of 23-40 months were divided into two groups (Group A and B) of four animals each (n=4). An iron deficient state was induced by phlebotomy, where a plasma iron drop >25% and 20% drop in hematocrit were deemed to be adequate. For the purpose of dose administration, the dogs used in this study were surgically implanted with vascular access ports (VAPs). Each VAP is connected to a cannula which is inserted into the duodenum. The liquid test formulations were administered to all subjects intra-duodenally through the VAP (at a dose of 7.83 mg elemental iron/animal) in two phases with a wash-out period in between (Table 2).
Blood samples were withdrawn at scheduled intervals to estimate plasma iron levels and were used to calculate PK parameters. Body weights were recorded prior to each dosing and throughout the study. Prior to dose administration, dogs were fasted for at least 12 hours and food was returned ¨4 hours post dosing. Data collected prior to the end of the fast was evaluated for demonstration of iron absorption, as the diet supplied contained additional iron. The 10 mL volume of test items 1 and 2 were administered through the VAPs connected to a cannula into the duodenum. Following dose administration, the devices were flushed with 0.9% saline water to ensure that the entire test item was delivered into the duodenum. Blood was collected at scheduled intervals to estimate plasma drug levels.
Table 2: Experimental schedule of the test formulations, and dosing details Animals Day Period Formulation No. Group(n=4 of Route Formulation d Dose (mg/dose*) Dose Coe 1 A 8 ID* (Solution) TIL UC1 51.5 Iron Chelate control unenhanced TI2: 51.5 Iron Chelate 1B 8 ID* (Solution) enhanced EF1 550 mg Sodium formulation I Caprate TI2: 51.5 Iron Chelate 2 A 14 ID* (Solution) enhanced EF1 550 mg Sodium formulation I Caprate 2 TIl:
14 ID* (Solution) UC1 51.5 Iron Chelate control unenhanced ID = Intraduodenal, Each mL contains 5.15mg Iron Chelate (0.783 mg elemental iron) and 550 mg Sodium Caprate (TI2).
[0111] On the days of dosing, each dog was housed individually for observation and was returned to group housing after the 24 hour blood collection time point of each period involving administration of respective test item. Teklad 21% Lab Dog Diet (W) 8755 was fed once per day and tap water was provided ad libitum except as indicated during dosing. Environmental controls were set to maintain temperatures from and humidity from 30-70%. Light source was fluorescent lighting on a 12 hr/12 hr on/off cycle except as required for specimen collection.
[0112] Prior to study initiation, each dog was confirmed to be healthy. Dogs were observed at least once daily 7 days prior to dosing and for the duration of the study.
The clinical observations were recorded once daily and any other times when a clinically significant change was noted. In-life phase assessments included but were not limited to assessment of activity, posture, respiration, hydration status, and overall body condition.
Animals were returned to stock following the end of the live period portion of this study.
[0113] Blood (-1.5 ml) was collected, via cephalic or jugular vein, into heparinized blood collection tubes at pre-dose (TO), 1, 2, 4, 6, 8, 10, 12, 14, 16, 20, 24 and 48 hours following dose administration. Care was taken to avoid hemolysis during the blood collection procedure. The blood was kept on ice for a maximum of 20 minutes until centrifuged. The centrifugation procedure was carried out at 4 C and 3000 rpm for minutes. Immediately after centrifugation, all plasma was transferred to appropriate labeled vials, frozen (at -70 C) and was shipped to the bio-analytical centre for the assay of total plasma iron content.
[0114] At the bio-analytical centre, plasma samples were assayed using a method for plasma iron content (Roche Diagnostics for the Hitachi 917 chemistry analyzer). Any remaining plasma was disposed of by incineration at the test facility, following the issuing of the final report for this study.
[0115] Upon receipt of bio-analytical data, the individual animal plasma concentration vs. time data was loaded into an Excel spread sheet (Microsoft office 2007). The PK parameters including AUC04, maximum plasma concentrations (Cmax) and time to achieve Cmax (Tmax) were calculated using macros written for MS Excel by Usansky et al. The above plasma data were used to calculate mean PK
parameters, standard deviations, and SE (mean Standard errors). The relative bioavailability was expressed as fold increase relative to the un-enhanced formulation. Data were evaluated as reported, and with a baseline correction, where the value of the 0-hour sample for each dog in each period was subtracted from the subsequent levels for that dog during that period. Statistical significance between groups was analyzed using a two tail student T
test assuming equal variance. The data is expressed as mean SD or mean SE, Only data collected during the fasting period were used in the analysis of absorption, as the diet fed to the dogs included iron.
[0116] The live phase portion of this study was 16 days in duration, which includes the induction of an iron deficiency state utilizing a phlebotomy procedure and crossover dosing of two test items in two phases with a washout period of six days. The dogs were made iron deficient by collecting an estimated 20% of the dog's blood volume from the jugular vein on days 0, 1, 2 for all dogs and 10% volume on Day 4 from dogs 4, 6, 7 and 8. The iron deficient state was deemed to be adequate when the mean hematocrit, or packed cell volume (PCV) was reduced to % of initial and the target mean plasma iron was reduced to < 25% of normal range noticed during pre-phlebotomy.
Although mean hematocrit reached required levels on day 2 itself, plasma iron levels were higher on day 2 compared to pre-phlebotomy levels (day 0). Refer to Tables 3 and 4 for plasma iron and hematocrit values during first and second period dosing.
[0117] First period dosing was carried out in respective groups (Table 2) on day 8 followed by the scheduled blood collection time points. Based on plasma iron content, it was decided that no phlebotomy was required prior to period 2 dosing, which was carried out after a six day wash out period after the first period of dosing.
Table 3: Plasma iron and hematocrit levels before periodl dosing of test items Induction phase % reduction prior Group Phasel % Hematocrit (plasma Iron litg/dL) to phasel No Animal ID Day() Dayl Day2 Day3 Day4 Day7 Day 8 PCV% Plasma 07dec 08dec 9dec 10dec lldec 14dec 15dec Day8 Di raoyn8 (1135491) (129) (169) (62) (93) (118) (87) 39 (1142837) (117) (143) (55) (65) (113) (60) 35 (1144694)) (134) (188) (203) (186) (99) (68) 40 (1146522) (109) (114) (78) (137) (151) (78) 42 (1138520) (161) (127) (61) (71) (113) (66) 37 B (1141482) (177) (283) (264) (208) (160) (132) 35 (1144546) (115) (210) (224) (139) (300) (66) 40 (1145526) (141) (181) (90) (109) (93) (108) 33 Plasma iron protocol range = 145-153 ug/dL, Target = -25%, * mean reductions Group A 40% 11, Group B 38% 17, Hematocrit -Normal = 40-55 %, Group A 39%+3, Group B 36% 3 Table 4: Plasma iron and hematocrit levels before period 2 dosing of test items Induction phase Phase2 Group Animal ID % Hematocrit %reduction prior to phase2*
TO
(plasma Iron itg/dL) Day0 Day 8 Day 11 Day14 Hematocrit Plasma iron 07dec 15dec 18dec 21dec Day14*
Day14 (1135491) (129) (87) 36 (93) (1142837) (117) (60) 38 (78) A

(1144694)) (134) (68) 34 (104) (1146522) (109) (78) 37 (73) (1138520) (161) (66) 35 (120) B (1141482) (177) (132) 37 (118) (1144546) (115) (66) 38 (75) (1145526) (141) (108) 35 (74) Plasma iron, Target = -25%, * mean reductions Group A 29% 5, Group B 35%
9: Hematocrit -Normal = 40-55 %, Group A 21% 4, Group B 22% 4 [0118] In a two-way crossover design, the relative oral absorption of enhanced iron formulation A was compared with an unenhanced control formulation. In each phase, both liquid formulations were administered to each group of four dogs (n=4) in two phases at an elemental iron dose of 7.83 mg/subject (Table 2).
[0119] Total plasma iron concentrations after oral administration of test formulations are shown in Table 5. The raw data for individual animals is shown in Table 6.
Table 5: Plasma iron concentrations of test formulations in periods 1 and period 2 Enhanced formulation Group A Group B
Time (hrs) 1135491 1142837 1144694 1146522 1138520 1141482 1144546 1145526 Mean 1 36 115 11 22 21 73 48 29 44.375 2 49 169 14 29 29 123 71 37 65.125 4 42 128 15 30 44 164 79 35 67.125 Control formulation Group A Group B
Time (hrs) 1135491 1142837 1144694 1146522 1138520 1141482 1144546 1145526 Mean 1 9 9 11 8 12 23 17 0 11.125 4 25 9 26 19 18 35 20 5 19.625 Attorney DocKet No. 9 /u1-D.5 wu Table 6: Plasma iron concentrations of test formulations for individual animals _______________________________________________________________________________ ______________________________________ tµ.) % o Change Change t-.) -a 5 in Fe in Fe Animal Base- pre- from from ID Group line dose baseline 1 hr 2 hr 4 hr 6 hr 8 hr 10 hr 12 hr 14 hr 16 hr 20 hr 24 hr 48 hr Baseline 1135491 A 129 87 -32.56% 96 93 112 169 218 280 319 403 402 262 158 86 -33.33%
1142837 A 117 60 -48.72% 69 65 69 100 134 147 179 286 325 320 110 57 -51.28%
1144694 A 134 68 -49.25% 79 87 94 145 196 278 362 347 337 113 46 59 -55.97%
1146522 A 109 78 -28.44% 86 80 97 142 184 202 226 268 314 211 79 71 -34.86%
Mean 122.3 73.3 -39.7% 82.5 81.3 93.0 139.0 183.0 226.8 271.5 326.0 344.5 226.5 98.3 68.3 -43.9%
SD 11.4 11.8 10.8% 11.4 12.1 17.8 28.7 35.6 64.4 83.8 61.5 39.5 87.8 47.6 13.4 11.5%
c?.
1138520 B 161 66 -59.01% 87 95 110 145 165 136 105 175 353 307 122 97 -39.75% g 1141482 B 177 132 -25.42% 205 255 296 343 352 337 328 333 339 333 189 104 -41.24% E.
1144546 B 115 66 -42.61% 114 137 145 178 204 207 189 198 297 205 113 81 -29.57%
1145526 B 141 108 -23.40% 137 145 143 188 282 366 365 349 359 269 73 54 -61.70%
Mean 148.5 93.0 -37.6% 135.8 158.0 173.5 213.5 250.8 261.5 246.8 263.8 337.0 278.5 124.3 84.0 -43.1%
SD 26.8 32.7 16.7% 50.5 68.3 83.2 88.3 83.2 108.5 121.1 89.9 28.0 55.6 48.1 22.2 13.5%
A=TIl: Control Unenhanced, B= T12: Enhanced Formulation I
_______________________________________________________________________________ ________________________ %
Change Change 'A
in Fe in Fe 1-3 Animal Base- pre- from from ID Group line dose baseline 1 hr 2 hr 4 hr 6 hr 8 hr 10 hr 12 hr 14 hr 16 hr 20 hr 24 hr 48 hr Baseline 1135491 A 129 93 -27.91% 129 142 135 159 198 221 283 439 405 274 164 95 -26.36% tcil 1142837 A 117 78 -33.33% 193 247 206 188 194 202 220 249 348 333 173 62 -47.01% Z, oe Auorney IJOCKel IVO. /V1-33 1144694 A 134 104 -22.39% 115 118 119 144 174 178 210 309 358 292 158 41 -69.40% g 1146522 A 109 73 -33.03% 95 102 103 126 151 167 199 285 315 263 90 75 -31.19%
Mean 122.3 87.0 -29.2%
133.0 152.3 140.8 154.3 179.3 192.0 228.0 320.5 356.5 290.5 146.3 68.3 -43.5% -a-, SD 11.4 14.2 5.2%
42.4 65.3 45.4 26.2 21.6 24.2 37.7 82.8 37.2 30.8 38.0 22.7 19.4% ,u4 oT:
1138520 B 161 120 -25A7% 132 127 138 184 218 225 328 358 367 285 76 39 -75.78%

1141482 B 177 118 -33.33% 150 155 153 174 170 167 167 200 288 378 257 215 21.47%
1144546 B 115 75 -34.78% 92 95 95 131 162 206 283 379 373 233 102 97 -15.65%
1145526 B 141 74 -47.52% 65 82 79 127 246 380 381 374 366 277 93 64 -54.61%
Mean 148.5 96.8 -35.3% 109.8 114.8 116.3 154.0 199.0 244.5 289.8 327.8 348.5 293.3 132.0 103.8 -31.1%
SD 26.8 25.7 9.1% 38.4 32.8 34.9 29.2 39.9 93.5 91.1 85.6 40.5 61.0 84.0 77.9 43.0%
A=TI2: Enhanced Formulation I, TIl: Control c?.
Unenhanced oe [0120] Relative bioavailability was analysed using AUC0-4 hrs significant improvement was noted with the enhanced iron formulation (Fig. 1).
[0121] The extent of absorption is 5.8 fold higher for the baseline corrected iron data set. Analysis of both phases of dosing (n=8, groups A and B), demonstrate that enhanced iron form A shows significant improvement (P < 0.02) in the relative oral bioavailability (5.48-fold) of iron absorption (Table 7). Over all, the in vivo crossover analysis indicates a potential of enhanced iron form A to meet the objective(s) of oral bioavailability enhancement of iron.
Table 7: Summary of relative oral bioavailability of enhanced Iron A
Grou Relative bioavailability 0 - 4 hrs p Total Iron Absorbed Iron O 1.55 0.24 5.48 2.09 verall ( n=8**) (0.05) (0.02) Conclusion [0122] This study shows the relative performance of an enhanced iron formulation on the oral absorption of elemental iron compared to an unenhanced control.
The enhanced iron formulation showed superior rate and extent of absorption compared with control formulation. As the animals were fed a standard meal containing iron after 4 hours, only these data were used to calculate relative bioavailability. The analysis demonstrates a significant improvement in the rate and extent oral absorption of iron (i.e., 5.5-fold higher relative bioavailability).
Therefore, the absorption enhancement technology has a potential utility to replace (1) currently available parenteral formulations and (2) oral formulations to preclude associated GI side effects.

Evaluation of forms of iron with enhancer systems [0123] This study developed procedures for evaluation of existing and potential salts and chelates of iron for use in oral enhanced formulations. Iron compounds are screened for compatibility with sodium caprate (C10). Appropriate formulations with optimized dissolution profiles are developed. Based on the procedures developed, a representative iron formulation in solution was developed. This procedure can be used to screen other salts and chelates and to develop appropriate enhanced iron formulations.
Solubility and compatibility (Pre-formulation) Experiments [0124] A test for precipitation was carried out. The solution formulations were centrifuged initially at 1000 rpm for 5 minutes. No precipitation was noted.
They were further centrifuged at 5000 rpm for 5 minutes. No precipitation was noted.
Hence this iron compound was considered compatible with C10. Refer to Table 8 for results.
Discussion [0125] Aqueous solubility studies of ferric pyrophosphate soluble and ferric pyrophosphate were carried out up to 50 mg/mL in purified water. C10 compatibility was tested by dissolving incremental amounts of these iron compounds (up to 50 mg/m1) in 55 mg/mL sodium caprate.
[0126] Ferric pyrophosphate (CAS No 10058-44-3) is practically insoluble in water, and C10 did not change its aqueous solubility (Table 8). Ferric pyrophosphate soluble (CAS No 85338-24-5) is available as a complex mixture of ferric pyrophosphate (46%) and sodium citrate (54%), and hence has no specific molecular weight, however it is expected to be a low molecular weight compound (<1000 Daltons).
[0127] Ferric pyrophosphate soluble has good water solubility up to the tested range (50 mg/mL, green color solution). Up to the tested range (5 mg/mL), this compound has shown solution compatibility with C10 (55 mg/mL), which was a hazy yellowish orange solution. There was no precipitation demonstrated by centrifugation indicating that iron is dispersed/solubilized, and probably exists as a micelle solution.
Table 8: Pre-formulation with iron chelates Material spec Observations Material Aqueous Fe Spec COA Sodium caprate solution compatibility solubility Light yellow solution at 5mg/mL and Ferric 50mg/mL soluble, 10mg/m1 in 55mg/mL C10 pyrophosphate Min 11 A
soluble 11.32 green color At higher concentrations (up to 50mg/mL) , N 101578 solution. the solution was turbid yellow but not precipitated(centrifugation test) Ferric 10.5-pyrophosphate** 12.5% 11.1%# Insoluble in water Not soluble even in 55mg/mL sodium caprate ** Molecular weight 745.21 for Fe4 (P207)3, discrepancy noted as the iron content needs to be above 20-30%.
[0128] Aqueous solubility studies of ferrous asparto glycinate and ferrous bis glycinate were carried out at 10-50 mg/mL in purified water. C10 compatibility was tested by dissolving incremental amounts of these iron compounds (up to 10 mg/ml) in 55 mg/mL C10. Refer to Table 9 for results.
[0129] Amino acid based iron chelates were found to be incompatible with C10 despite good aqueous solubility (Table 9). Since the iron compound contained in the Ferrochel0 product( Albion; Ferrous/Ferric bisglycinate) was reported to be soluble in the acidic and alkaline environments, this compound was tested for solubility in purified water, phosphate buffer pH 6.8, and a solution of C10 (Table 10). Despite good aqueous solubility, Ferrochel0 is deemed not suitable for enhancer development as this compound precipitates in the presence of C10. Ferrous gluconate also showed C10 incompatibility (refer to Table 10).
Table 9: Pre-formulation with Iron chelates (EXP-NB GIPET Iron 1- 23*) Observations Material Aqueous solubility Sodium caprate solution compatibility Solution turned black to grayish black colour, with severe Ferrous Asparto 10mg/mL soluble, Light precipitation glycinate green solution Despite good aqueous solubility, It does not have compatibility with C10 Ferrous bis 50mg/mL soluble, Clear Also, precipitation noted at 5mg/mL
in 55mg/mL C10, glycinate solution 50% dilution did not improve solubility.
Table 10: Pre-formulation with iron compounds Material* Observations Aqueous solubility Sodium caprate solution compatibility At 10mg/mL in 55mg/m1 C10, severe precipitation observed, same was noted at 50% dilution (i.e. 27.5mg/mL C10 and 5mg/mL API) Dilution up to 90% with DW did not 50mg/mL soluble, Light green solubilize the above precipitate.
Ferrous Bisglycinate solution (Ferrochel) Not soluble at 2mg/mL API in 55mg/mL
50mg/mL soluble in phosphate sodium caprate buffer pH 6.8 300mg API and 550mg sodium caprate cannot be co-solubilized in 250mL water.
Despite good aqueous solubility (also in pH
6.8), this compound is not compatible with C10.
Ferrous Gluconate 85g/L ( MSDS) Severe precipitation noted at 5mg/mL in FCC/USP* 50mg/mL soluble, Clear solution 55mg/mL
sodium caprate, Ferrous GluconateSevere precipitation noted at 5mg/mL in Solubility was tested at 50mg/mL, FCC/USP* 55mg/mL sodium caprate, Clear solution Low HM
Preparation of tablets with saccharide or citric acid [0130] Ferric pyrophosphate soluble was used to prepare tablets with approximately 33 mg of elemental iron and different solubilizing excipients, and was tested for dissolution of both elemental iron and C10. All ingredients were weighed into weigh boats and transferred into a 250 mL Brawn PP bottle. Mixing was carried out manually for 3-5 minutes in the bottle. The mixing times includes one minute final mixing with stearic acid. The blend was carefully transferred into a weigh boat and used for tablet compression. A slight excess (2-5 mg) of blend relative to the target weight was weighed for compression to avoid transfer losses. Compression was carried out using a Globe Pharma, MTCM1; compression tooling used was 18x8 mm (SN
426539931995-04 for both upper, lower punch and die), and compression was carried out at 1500-2000 psi.
[0131] Tablets were characterised for average weights and hardness. Also, tablets were evaluated for dissolution profiles of both elemental iron and C10. In some experiments tablet disintegration time in purified water (800 mL) was tested.
Formulations prepared and results are given in Tables 11-13 and Fig. 2. The following observations were made.
1. Tablets prepared without any saccharide excipient showed very slow dissolution for both iron and C10 with 76-77% in 3 hours, and complete dissolution and/or plateau occurring in 5.5 hours.
2. Tablets prepared with saccharide (sorbitol) improved dissolution profile with 78%
iron and 74% C10 released in 2 hours, followed by complete release found in 5 hours.
3. Tablets prepared with citric acid as solubilizing aid significantly improved dissolution profile of both Fe and C10 with complete release (>97%) occurring in 3 hours.
[0132] These experiments demonstrate that citric acid agents (or similar functional agents) are necessary to formulate ferric pyrophosphate soluble as enhanced iron tablets to achieve acceptable dissolution.
Table 11: FPP soluble tablets with and without sorbitol Tablets without sorbitol Tablets with sorbitol Material Iron 1 - 27 EXP 1560A Iron 1-27 EXP 1560B
Amount / tablet(mg) % Amount/ tablet (mg) %
FPP soluble 290 34.41 290 30.38 Sodium caprate 550 65.18 550 57.54 Partek 0.00 0.00 112 11.72 Stearic acid 3.5 0.41 3.5 0.37 Total 844 100 956 100 Characterization 846.41 2.45mg (n=6) 846.41 2.45mg (n=6) Ave .weights(mg) Hardness 126 7N Hardness 126 7N
Ave. Hardness N
Blended for 5 minutes, which includes 1 minute blending with stearic acid and compressed at -1500 psi Table 12: FPP soluble tablets with citric acid Citric acid tablets EXP 1563 Material Amount / tablet(mg) %
FPP soluble 290 30.27 Sodium caprate 550 57.41 Citric acid 115 11.95 Stearic acid 3.5 0.37 Total 958 100 Tablet weights Average tablet weight 960mg(n=25) Average tablet Hardness 117N(n=3) blended all ingredients except stearic acid for 5 minutes, and was blended further 2 minutes after addition of stearic acid. Compression was carried out at 100 bar(-1450 psi) pressure.
Table 13: Dissolution profiles of Fe and C10 with various ferric pyrophosphate soluble tablets contain different excipients (n = 2 tablets) Tablets without sorbitol Tablets with sorbitol Citric acid tablets Time % Iron % C-10 % Iron % C-10 % Iron* % C-10 0 0.0 0.0 0.0 0.0 0.0 0.0 24.8 45.2 37.4 48.9 28.9 23.4 36.3 52.2 49.8 57.0 43.9 35.1 41.0 55.1 53.5 59.2 54.2 43.7 60 50.0 60.6 64.2 66.1 74.2 63.3 90 59.6 65.1 71.8 69.3 87.1 77.5 120 69.2 69.8 78.2 74.0 95.1 87.9 150 72.5 73.3 82.1 78.5 98.2 95.0 180 77.5 76.6 84.8 79.6 99.0 97.3 210 82.3 79.6 87.8 82.1 100.2 99.6 240 86.0 82.8 90.0 84.6 99.6 100.4 270 92.3 86.1 93.6 85.4 101,0 n/a 300 97.2 90.8 96,8 88.2 101.2 n/a 330 98.7 90.5 97.7 89.2 100.8 n/a *% iron release reached a maximum of 107%, hence the profile was normalized for 100%
Example 4 Preparation of tablets containing 65 mg of elemental iron and citric acid [0133] FPP soluble tablets containing 65 mg elemental iron were prepared similarly to those in Example 3. Table 14 contains details of the tablets.
These tablets were evaluated for dissolution profiles (Figs. 3 and 4). As noticed previously, tablets without citric acid showed very slow dissolution profiles as complete release was not achieved even after 3 hours; only 77% Fe and 82% C10 was dissolved (Table 15).
On the other hand, tablets containing citric acid (9%) showed good dissolution profiles with >97% Fe release occuring in 2 hours. The release of C10 was not complete, however it reached plateau (84%) at 2 hours.
Table 14: FPP soluble tablets containing 65 mg elemental iron Material Tablets Without citric acid Tablets with Citric acid EXP 1574 Sample A EXP 1574 Sample B
Amount / tablet(mg % Amount/ tablet (mg) FPP soluble 574 50.91 574 46.22 Sodium caprate 550 48.78 550 44.28 Citric acid 114.5 9.22 Stearic acid 3.5 0.31 3.5 0.28 Total 1128 100 1242 100 Average Tablet wt 1132mg(n=10) Average Tablet wt 1247mg(n=10) Average Hardness 146N(n=3), Average Hardness 168N (n=3), Tablet Dimensions length 18mm, width 8mm, Dimensions length 18mm, width 8mm, and characterization and thickness 8.2mm thickness 9mm Disintegration time 19` tablet 18min40sec, Disintegration time 1st tablet 15min56sec, last last tablet 23 min lOsec tablet 19 min 12sec All materials were dispensed, screened thorough 355mesh, and blended for 3minutes, 18 x 8 mm tool Compression force 2000psi.
Table 15: Dissolution profiles of Fe and C10 with FPP soluble enhanced tablets with and without citric acid (n = 2 tablets) % dissolution No citric acid % dissolution Citric acid Time Iron C-10 Iron C-10 0 0.0 0.0 0.0 0.0 18.4 29.3 40.7 18.3 28.0 35.1 56.4 32.5 34.5 39.1 64.3 41.8 60 45.6 47.4 85.2 65.2 90 54.0 53.9 92.8 77.1 120 61.9 61.6 97.6 83.5 150 69.8 68.4 ND ND
180 77.3 81.7 98.3 85.6 240 98.2 84.1 Example 5 Tablets containing 65 mg of elemental iron with citric acid and with/without saccharide [0134] Tablets with 65 mg elemental iron were prepared with citric acid in the tablet cores according to the formulae in Table 16 using the procedure in Example 3.
Tablets were prepared with and without saccharide. The following points summarize the results, which are given in Tables 16 and 17.
1) High concentration of citric acid (11.6%) (148 mg/tablet). Complete dissolution of Fe was observed at 60 minutes, C10 dissolution was not complete however, it reached plateau at 90 minutes (Fig. 5).
2) Tablets containing both citric acid and sorbitol (8.4% each). Complete dissolution of iron was noticed at 90 minutes, C10 dissolution was not complete however, it reached plateau at 90 minutes (Fig. 6).
Table 16: Iron tablets of FPP soluble containing 65 mg elemental iron and varying concentrations of solubilizing excipients Sorbitol + Citric acid High Concentration of Citric acid NB Iron 1 ¨ Page 37 NB Iron 1 ¨Page 38 Material EXP 1579 Sample A EXP 1580 Sample B
Qty per tablet(mg) Qty per tablet (mg) FPP soluble 574 42.31 574 44.98 Sodium caprate 550 40.55 550 43.10 Citric acid 115 8.48 148 11.60 Sorbitol 115 8.48 0 0 Stearic acid 3.5 0.26 3.5 0.27 Total 1357 100 1276 100 Ave. Tablet wt 1278mg(n=25) Ave. Tablet wt 1359mg(n=25) Hardness 180N(n=3), Hardness 158N(n=3) Tabletization Dimensions length 18mm, width 8mm, Dimensions length 18mm, width 8mm, and character and thickness 9.7mm thickness 9.1mm Disintegration time Pt tablet Disintegration time 1st tablet 13min40sec, 13min24sec, last tablet 17 min 44sec last tablet 16 min 57sec All materials were dispensed, screened thorough 355 mesh, blended for 3 minutes, 18x8 mm tool Compression force 2000 psi.
Table 17: Dissolution profiles FPP soluble tablets containing varying quantities of solubilizing excipients (n = 2 tablets) % dissolution % dissolution citric acid plus sorbitol High concentration of Citric acid Time *Iron C-10 *Iron C-10 0 0.0 0.0 0.0 0.0 45.3 17.3 58.7 20.4 65.0 34.3 81.8 39.8 76.5 48.5 90.8 52.7 60 93.7 69.6 100 64.5 90 100.7 81.1 100.1 73.9 120 99.7 85.7 99.4 75.7 180 100.4 86.8 100.9 78.0 240 100.0 83.8 100.0 79.4 *% Iron reached a maximum of approx. 114%, hence has been normalized for 100%
release Conclusions [0135] Tablets with citric acid (12% or 148 mg/tablet) were preferred. These experiments' reveal that citric acid (or similar functional agents) are necessary to formulate ferric pyrophosphate soluble iron tablets to achieve an acceptable dissolution profile. The target dissolution profile is rapid, providing co dissolution of both iron and enhancer.
Example 6 Preparation of tablets with various solubilizing excipients [0136] The following experiments were performed to investigate citric acid, sodium citrate or ascorbic acid as solubilizing excipients.
[0137] Evaluation systems were made as in Example 2. Citric acid and sodium citrate were evaluated as solubilizers with soluble iron pyrophosphate (7.83 mg Fe), along with the relevant concentration of citric acid or sodium citrate (15 mg) in 55 mg/mL
solution of sodium caprate.
[0138] With citric acid as the solubilizer (Table 18), precipitation was noticed up to 17 mg of Fe, and a hazy orange dispersion was noticed from 21 mg Fe onwards. On the other hand, sodium citrate as the solubilization aid (Table 19) was found to be relatively superior as dispersion characteristics were observed from 7.6 mg Fe onwards.
Table 18: Solution compatibility of FPP soluble (API)with sodium caprate in the presence of varying concentrations of citric acid Elemental Iron(mg) Iron compound concentration(mg) Increments of Planned Actual Total API
API added Observation 62.2 Cloudy white ppt#(C10) at the bottom 7.83 7.62 5.1 67.3 Soluble, pH 8.45 8.93 11.6 78.90 ppt as above 12 16.93 70.6 149.50 ppt as above 20.57 32.17 181.67 Hazy Light orange dispersion 32.27 103.3 284.97 Clear colloidal 8.06 NC* 47.27 132.45 417.42 As above 65 66.00 165,58 583 As above # ppt: precipitate; *NC Not calculated Table 19: Solution compatibility of FPP soluble with sodium caprate in the presence of varying concentrations of sodium citrate Elemental Iron(mg) Iron compound concentration (mg) Increments of API
Planned Actual added Total API Observation 62.2 Fine white PPT at bottom 7.83 7.98 8.3 70.5 Soluble pH 8.45 10 10.45 21.8 92.3 12 11,40 8.4 100.7 15 15.06 32.3 133 20 20.16 45 178 Clear colloidal 8.15 26.35 54.7 232.7 30.59 37.4 270.1 40.88 90.9 361 51.07 90 451 65 66.00 132 583 # ppt: precipitate Example 7 Preparation of tablets with sodium citrate [0139] Iron tablets were made using sodium citrate in place of citric acid according to the procedure used in Example 3. Details of the tablets are shown in Table 20. Dissolution of both Fe and C10 was slow, however complete release of Fe and >80%
C10 was achieved in 4 hours (Fig. 7).
[0140] The comparative dissolution profiles of Fe from citric acid and sodium citrate is depicted in Fig. 8. The results indicate citric acid is the better solubilizer.
Table 20: Iron tablets of FPP soluble containing 65mg elemental iron and sodium citrate as solubilizing excipient FPP soluble tablets with sodium citrate Material EXP 1598 Amount tablet(mg) FPPsoluble 574 44.98 Sodium caprate 550 43.10 Sodium citrate 148.4 11.64 Stearic acid 3,5 0.28 Total 1276 100.00 Tablet characterization 1278.90 1.35 Average weight(mg SD; n=6) Hardness 131 6N
Average Hardness(N SD; n=4) Blended for 5 minutes and compression was carried out at 1500psi.
Example 8 Preparation of iron tablets containing ascorbic acid [0141] Because ascorbic acid is known to be an oral absorption promoter of iron, tablets were manufactured using ascorbic acid in place of citric acid according to the procedure in Example 2. Details of the tablets are shown in Table 21. The results indicate Fe was dissolved in 60 minutes and C10 dissolution reached a plateau with 80%
release (Fig. 9). A control experiment carried out with C10 alone (exact tablet composition: 550 mg C10, 3.5 mg stearic acid, 115 mg sorbitol) showed 100%
release.
The data were replotted by normalizing C10 and showed that 97% C10 had dissolved after 90 minutes and reached a plateau (Fig. 10). Comparative dissolution profiles of citric acid and ascorbic acid tablets are depicted in Fig. 11, and results indicate rapid and complete release with citric acid. Ascorbic acid tablets show a relatively superior co-release of both Fe and C10, however complete release was not achieved.
Table 21: Iron tablets of FPP soluble containing 65 mg elemental iron and ascorbic acid as solubilizing excipient Ascorbic acid tablets EXP 1615 Material Amount / tablet(mg) FPP soluble 574 44.98 Sodium caprate 550 43.10 Ascorbic acid 148.4 11.64 Stearic acid 3.5 0.28 Total 1276 100.00 Tablet characterization 1279.27 2.20 Average weight(mg SD; n=6) Average Hardness(N SD; n=4) Hardness 135 6N
blended for 5 minutes, and compressed at 1500 psi.
Example 9 Preparation of iron tablets with varying concentrations of citric acid [0142] Additional batches of enhanced tablets were prepared with varying concentrations of citric acid. These concentrations were 12%, 15%, and 21%
according to the procedure in Example 3 The 15% tablets had a reduced concentration of iron.
Details of the tablets are shown in Table 22).
[0143] Fe release was rapid and complete with all the batches (Fig. 12).
Tablets with 12% and 15% (API load 50%) showed release of C10 in 120 minutes. These results demonstrate 100% recovery of C10 can be achieved.
Table 22: Iron tablets of FPP soluble containing 65 mg elemental iron and ascorbic acid as solubilizing excipient Target tablet 50% Iron concentration Very high citric acid Material Qty/ tablet Qty/ tablet Qty/ tablet (mg) (mg) (mg) FPP soluble 574 44.98 287 29.02 574 40.31 Sodium caprate 550 43.10 550 55.62 550 38.62 Citric acid 148.4 11.63 148.4 15.00 296.8 20.84 Stearic acid 3.5 0.27 3.5 0.35 3.5 0.25 Total 1276 100 989 100 1424 100 Tablet Ave. Tablet wt Ave. Tablet wt Ave. Tablet wt characterization 1282mg(n=20) 989mg(n=20) 1428mg(n=20) All material were dispensed, screened thorough 355mesh, blended for 3minutes, 18x8mm tool Compression force 2000psi.
Example 10 Replicate Batches of Iron Tablets [0144] Additional batches of iron tablets were prepared according to the procedure in Example 3 to evaluate the reproducibility of manufacture and performance.
Details of the tablet batches are shown in Table 23. The results indicate rapid and complete release with citric acid. Ascorbic acid tablets showed a relatively superior co-release of both iron and C10, but complete release was not achieved (Table 24 and Figs.
13-15).
Table 23: Iron tablets of FPP soluble containing 65 mg elemental iron and solubilizing excipients Operator 1/ citric Operator2/citric Operator 2/ascorbic Material Qty/ tablet Qty/ tablet Qty/ tablet Vo 0/0 %
(mg) (mg) (mg) FPP soluble 574 44.98 580 45.24 580 45.24 Sodium caprate 550 43.10 550 42.90 550 42.90 Citric acid 148.4 11.63 148.4 11.58 0 0 Ascorbic Acid 0 0 0 0 148.4 11.58 Stearic acid 3.5 0.27 3.5 0.27 3.5 0.27 Total 1276 100 1282 100 1282 100 1275.07 0.91 Tablet characterization 1282.15 0.93 1283.75 3.28 Average weight(mg SD; n=6) Tablet hardness not Average Hardness(N SD; n=4) tested 130 6N

All material were dispensed and blended for 5 -6 minutes, 18x8mm tool Compression force 1500 -2000psi.
Table 24: Iron tablets of FPP soluble containing 65 mg elemental iron and solubilizing excipients EXP 1642 (Ope 1 citric) EXP 1649 (Ope 2 citric) EXP
1650(ascorbic) Time %dissolved (n = 2 tablets) %dissolved (n = 6 tablets) %dissolved (n = 6 tablets) % %
C10 Iron C10 Iron C-10 Iron RSD RSD RSD RSD
0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 0.0 22.2 50.5 15.0 21.0 23.2 16.4 7.4 11.7 11.6 7.3 43.6 74.4 28.1 13.9 39.1 9.3 11.6 8.0 18.7 8.8 62.4 89.4 41.2 10.6 53.6 7.0 15.9 7.5 25.4 9.9 60 88.2 96.9 75.8 6.3 84.6 4.2 27.9 11.5 41.9 112 90 92.6 96.8 94.7 13 96.3 1.7 38.8 10.3 56.0 10.7 120 93.6 96.7 99.1 1.7 98.6 0.7 48.2 10.1 , 67.3 9.6 180 94.8 97.3 99.9 1.1 98.8 0.5 61.9 8.2 81.7 7.2 240 95,2 96.7 99.4 1.2 99.6 0.5 71.5 5.3 90.8 4.1 Example 11 Iron tablets prepared with varying levels of citric acid and sodium citrate [0145] Iron tablets containing ferric pyrophosphate (Food Chemicals Codex (FCC)) were manufactured with varying levels of citric acid and sodium citrate were prepared according to the procedure in Example 3. Details of these tablets are shown in Tables 25 and 26.
[0146] The dissolution of sodium caprate was rapid in the presence of 15%
citric acid or sodium citrate. Within 2 hours, C10 dissolution reached plateau with 80-84%
release (Fig. 16). Increasing citric acid content in the tablets decreased C10 dissolution with <60% release with 30% citric acid and <40% release with 36% citric acid.
None of these tablets showed Fe dissolution (or solubilization), hence neither ferric pyrophosphate alone nor in combination with sodium citrate at a level similar to the soluble version is suitable. These data confirm the conclusions obtained in Example 2 relative to ferric pyrophosphate versus soluble ferric pyrophosphate.
Table 25: Iron tablets of FPP/FCC containing 65 mg elemental iron and citric acid as solubilizing excipient 15% citric acid 36% citric acid 30% citric acid Material Qty/ tablet Qty/ tablet Qty/ tablet (mg) (mg) (mg) FPP FCC 260 27.01 260 20.51 260 22.35 Sodium caprate 550 57.19 550 43.42 550 47.32 Citric acid 148.4 15.43 453 35.79 349 30.03 Stearic acid 3.5 0.36 3.5 0.28 3.5 0.30 Total 962 100 1267 100 1162 100 Tablet characterization 962.59 1.39mg 1267.31 0.72mg 1162.93 0.52mg Average weight(mg SD; n=6) Average Hardness(NISD; n=4) 140 6 N 142 5 N 141 5 N
All material were dispensed and blended for 5 minutes, 18x8mm tool Compression force 1000-2000psi.(Average-1500 psi) Table 26: Iron tablets of FPP/FCC containing 65 mg elemental iron and citric acid plus sodium citrate as solubilizing excipient FPP FCC tablets with sodium citrate and citric acid.

Material Qty/ tablet (mg) FPP FCC 260 19.84 Sodium caprate 550 42.01 Sodium citrate 347 26.54 Citric acid 148.4 11.34 Stearic acid 3.5 0.27 Total 1309 100 Tablet characterization 1309.70 1.26mg Average weight(mg SD; n=6) Average Hardness(N SD; n=4) 14214 N
All material were dispensed and blended for 5minutes, 18x8mm tool Compression force 1500-2000psi Pharmacokinetic study of dosage forms of iron pyrophosphate [0147] A two way cross over preclinical study was undertaken to evaluate the relative fasted oral bioavailability of iron (soluble ferric pyrophosphate) formulation versus an unenhanced control formulation of ferrous sulfate in an iron deficient intraduodenally cannulated beagle dog model. Twelve female dogs were included in the study and were divided into two groups of six animals each (Group A (n=6) and Group B
(n=6). These dogs were made iron deficient by phlebotomy prior to dosing of the test formulations. A drop in plasma iron levels of ¨25% and/or a drop of ¨20% in hematocrit (measured as % packed cell volume (PCV) relative to pre-phlebotomy values was considered as iron deficiency.
Experimental Details [0148] The live phase portion of this study was 18 days in duration (including the iron deficiency induction); the live-phase began on day -3 with clinical observation and live-phase ended after the 24 hour blood collection in Period 2.
[0149] The induction phase of the study to induce the iron deficient state was designated as Days 0-10. Iron deficiency was induced by collecting an estimated 20% of the dog's blood volume from the jugular vein, on Days 0, 1, 4, 5, 8 and 11.
[0150] Phlebotomy blood samples taken during the anemia induction phase were used for the measurement of iron levels and PCV. Animals were allocated to two groups (Group A and B) by stratified randomization based on pre-study body weights.
[0151] On day lithe iron deficiency target was reached. Both formulations were administered to each group in a crossover design. The washout period between each dose was 24 hours (Refer Table 27). Blood samples were collected and analyzed for plasma iron levels at pre-dose (TO), 0.5, 1, 2, 4, 6, and 24 hours following test article administration. Oral bioavailability of enhanced iron TI2 was estimated relative to the control formulation ferrous sulfate alone unenhanced TI1 .
Table 27: Study design for crossover dosing of test items Animals Day Formulation Dose Formulation Blood Period Group of Route Code Collection No. name (mg/dog**) (n=6) Dose Batch No Time Points Ferrous TO(Pre-dose) , 12- ID* unenhanced UC2**
1 A sulfate 0.5, 1, 2, 4, 13 (Solution) TI1 EXP1667 A
¨15mgFe/dog and, 24hrs Ferric pyrophosphate 12- ID Form B EF2*** TO(Pre-dose) , 1 B Iron TI2 0,5, 1, 2, 4, 13 (Solution) ¨15mgFe and EXP1668A
and, 24hrs 550 mg enhancer/dog Ferric pyrophosphate TO(Pre-dose) , 13- ID form B EF2 2 A Iron TI2 0.5, 1, 2, 4, 14 (Solution) ¨15mgFe and EXP1668B
and, 24hrs 550 mg enhancer/dog Ferrous TO(Pre-dose) , 13- ID unenhanced UC2 2 B sulfate 0.5, 1, 2, 4, 14 (Solution) TI1 EXP1667 B
¨15mgFe/dog and, 24hrs ID = Intraduodenal , ** UC2: Un-enhanced Control formulation 2; Each mL contains 1.5 mg elemental iron *** EF2: Enhancer formulation 2. Each mL contains 1.5 mg elemental iron, and 55mg Sodium Caprate, and 3.43 mg citric acid.
Data Analysis [0152] Upon receipt of bio-analytical data, the individual animal plasma iron concentration data were loaded into an Excel spreadsheet (Microsoft office Excel 2003).
PK parameters including AUCo_t, Cmax, and Tmax, were calculated using macros written for MS Excel by Usansky et al.
[0153] Because systemic iron levels comprise both the endogenous iron pool and exogenously administered iron absorbed from the formulation, the following analysis was carried out: 1) Evaluation of total plasma iron vs. time profile; and 2) Evaluation of plasma iron corrected for base line levels (CT-TO), obtained by subtracting the total plasma iron of each animal prior to dose administration (TO) from total plasma iron at each time point (CT). Both total plasma iron vs. time and plasma iron corrected for base line vs. time data were used to calculate the PK parameters. For plasma iron corrected for base line, negative values were considered as zero for AUC calculations.
[0154] Only AUC0,6 was used to evaluate the relative oral bioavailability (Frei), which is presented as the fold increase of iron T12 relative to the ferrous sulfate formulation. The pre-dose data generated from phlebotomy experiments, plasma iron levels and %PCV, are also summarized.
Results [0155] This summary includes 1) pharmacokinetic (PK) data analysis and 2) main observations of iron deficiency induction.
Summary of pharmacokinetic (PK) parameters [0156] Individual animal baseline corrected plasma iron concentration for the unenhanced TI1 and iron TI2 are presented in Tables 28 and 29 and plotted in Fig. 19.
[0157] PK parameters were estimated using plasma iron levels and plasma iron levels corrected for base line data (predose plasma iron concentration).
Baseline corrected data was used as TO plasma iron varied by animal and by period values, which can be presumed to be reflective of variations of the endogenous iron pool.
For baseline corrected plasma iron levels, the negative values were considered as zero for AUC
calculation. The mean PK parameters of the test and reference items based on total plasma iron and baseline corrected iron are summarized in Table 30.
[0158] The relative oral bioavailability (Fret) of iron TI2 was calculated using extent of absorption until 6 hours (AUC04 as the animals were fed a normal iron content meal after 6 hours. The AUC0_6h and the Crnax ratios demonstrate that iron TI2 showed superior oral absorption of iron to the ferrous sulfate solution (TI1). iron TI2 showed a 3.45-fold increase (n=11) in relative oral bioavailability compared to TI1 (unenhanced oral ferrous sulfate) (Table 31).
[0159] The difference in magnitude of absorption in group B for the two formulations (ref Table 32) was relatively low. This may be attributable to predose iron levels being lower prior to dosing the reference formulation than prior to dosing the iron formulation (66 10 [tg/dL vs. 83 18 [tg/dL).
Table 28: Ferrous Sulphate unenhanced TII Individual animal's baseline corrected plasma iron concentration [tg/dL and AUCs Plasma Iron conc. (tig/dL) AUC
Dog Time(hr) (0-6hr) Period Test Item ID 0 0.5 1 2 4 6 Dog 1 1 TI1 0 0 0 0 0 0 _ Dog2 1 TI1 0 149 123.5 78.5 53.5 0.5 392.38 Dog 3 1 TI1 0 22 52.5 4 0 0 56.38 Dog 4 1 TI! 0 44.5 43.5 30.5 2.5 3.5 109.13 Dog 5 1 TI1 0 56.5 39.5 20.5 0 0 88.63 Dog 6 1 TI1 0 135.5 158.5 94.5 7.5 0 343.38 Dog 7 2 TI1 0 52 46 0 0 0 60.5 Dog 8 2 TI! 0 57 72 35 7 2 151 Dog 9 2 TI1 0 132 89 40 15 11 233.75 Dog 2 TI1 0 177 154 69 35 27 404.5 Dog 2 TI1 0 185 183 113 14 0 427.25 Dog Table 29: Iron TI2 Individual animal's baseline corrected plasma iron concentration pz/dL, AUCs, and Frei (fold v control) Frel Plasma Iron conc. (ttg/dL) AUC (fold v Dog Perio Test Time(hr) (0-6hr) control) ID d Item 0 0.5 1 2 4 6 Dog 1 2 T12 0 171 142 100 25 0 392.00 -Dog2 2 T12 0 245 173 68 0 0 354.25 0.90 Dog 3 2 T12 0 219 173 149 41 0 544.75 9.66 Dog 4 2 T12 0 231 238 243 105 31 899.50 8.24 Dog 5 2 T12 0 238 166 101 14 0 423.00 4.77 Dog 6 2 T12 0 313 325 224 63 7 869.25 2.53 1 TI2 236. 164. 108.
Dog 7 0 5 5 5 23.5 11.5 _ 462.88 7.65 1 TI2 187.
Dog 8 0 5 155 81 5 0 341.50 2.26 1 TI2 132.
Dog 9 0 5 81 0 0 0 127.00 0.54 1 TI2 172.
Dog 10 0 5 87 11.5 0 0 168.75 0.42 1 TI2 157.
Dog 11 _______________ 0 5 124 58 0 0 258.75 0,61 1 TI2 116.
Dog 12 0 5 66.5 0 0 0 108.13 0.37 , Table 30: Mean pharmacokinetics of the test items (n=12 or 11) Unenhanced (TI1) Iron (TI2) PK parameter Base line Base line Total plasma Iron corrected* Total plasma Iron corrected*

626.90143.43 213.49144.57 874.13177.48 412.48174.47 ug/dl*h (%RSD) (24.00) (72.31) (30.70) (62.54) FRe0-6) Fold Vs 1.4710.17 3.45 1.07**
Unenhanced _____________________________________ (40.48) (102.44) Cmax 179.17112.63 108.64115.75** 289.71 14.75 203.67116.87 ug/dI
(%RSD) (24.43) (50.21) (17.64) (28.69) Tmax 0.6310.09 0.6810.07** 0.6710.13 0.6710.13 Hrs (%RSD) (49.73) (37.00) (66.57) (66.57) Ratio of Cmax 1.7110.16 2.59 0.51 Vs Unenhanced (31.92) (65.73) * Negative values were set to zero for the purposes of AUC calculation as this represents no/or negligible absorption **Calculated T0-6 hrs interval, **n=11 as one animal in unenhanced group had no values greater than baseline, hence cannot used be for calculation of Frel.
Dog 1 was omitted from analysis.
Table 31: Summary of the relative oral bioavailability of the test items dosed at different intervals in different groups Relative oral bioavailability (Frel Fold Increase SEM) (Cmax Ratio) Groups Base line corrected Total plasma Iron Plasma Iron 1.47 0.17 3.45 1.07*
Overall (n=12) (1.7110.16) (2.59 10.51) 1.86 0.25 5.22 1.66 (2.01 0.23) (3.5111.44) Group A
1.08 0.09 1.97 1.17 Group B (1.4110.14) (1.8310.61) *n=11, Frel was assessed for each dosing period since the iron load at baseline will impact iron absorption.
Summary of induction of iron deficiency [0160] The animals reached an iron deficient state as defined in the protocol (reduction of plasma iron levels of ¨25% and/or a drop-20% in hematocrit) (Table 32, Fig. 20) prior to the initial dosing.
[0161] The plasma iron levels were analyzed prior to dose administration on Days 0, 5, 11 and 12 and were reduced by at least 25% relative pre-induction phase (Day 0) for all animals, except one subject, Dog 3. However, this animal was also included in the study analysis as hematocrit levels were within the acceptable target (Refer to Tables 33 and 34).
[0162] At the end of the 24 hours wash-out following the Period 1 dose administration, iron levels were still deficient for both groups (82.5 ug/dL
Group A and 66.0 ug/dL Group B).
Table 32: Summary of plasma iron and hematocrit (%PCV) levels at key stages (Mean +
SD, n=6) Group A Group B
Period of Testing Plasma Iron Plasma Iron % PCV %PCV
(.tg/dL) ( g/dL) Pre Induction 52 2 165 69 51 4 147 28 Pre-Period 1 Dosing 35 2 93 55 32 2 90130 Pre-Period 2 Dosing 83144 66 24 Table 33: Plasma iron levels of individual animals during the induction of iron deficient state Dog No %
Day 0 Day 5 Day 11 Day 12 Reduction (ID) Dog 1 293 412 87 147.5 50 (1437284) Dog 2 128 270 94 55.5 57 (1544676) Dog 3 121 134 111 176.5 -46 (1454138) Dog 4 118 243 133 69.5 41 (1544749) Dog 5 n/a* 196 83 71.5 64 (1175018) Dog 6 135 203 41 38.5 71 (1451716) Dog 7 149 138 71 57.5 61 (1544765) Dog 8 (1439686) Dog 9 152 258 183 88 42 (1444264) Dog 10 196 219 132 144.5 26 (1544382) Dog 11 (1436946) Dog 12 139 178 70 94.5 32 (1439163) * Data not available at this point since this dog was added to the study at a later date.
Table 34: Hematocrit levels (%PCV) of the animals during the induction of iron deficient state Hematocrit levels (%PCV) Dog No Day Day Day Day Day Day Day Day Day (ID) 0 1 4 5 6 7 8 11 12 Reduction Dog 1 (1437284) Dog 2 (1544676) 54 42 54 38 40 40 34 44 34 37 Dog 3 (1454138) Dog 4 (1544749) Dog 5 (1175018) n/a* n/a* n/a* 54 44 40 34 36 34 37 Dog 6 (1451716) Dog 7 (1544765) Dog 8 (1439686) Dog 9 (1444264) Dog 10 (1544382) Dog 11 (1436946) Dog 12 (1439163) * Data not available during these points since this dog was added to the study at a later date.
Administration Effects/Potential issues [0163] A summary of the clinical observations noted after test item dosing are detailed in Table 35 below. As expected, dogs had varied plasma iron and %PCV
levels at the completion of the anemia induction period. Iron deficiency criteria were met in all animals and were deemed sufficient for the purposes of the study.

Table 35. Summary of clinical observations after dosing of each Test Item Study Period Test Item Clinical Observations*
Test Item 1 No adverse clinical observations Ferrous sulfate Dog7 (1544765) vomited 5mL within lh post administration.
Oedema/swelling was noticed at surgical port site within ¨2hours ( noticed 4 hr 13 min) Period 1 Test Item 2 Dogll (1436946) vomited 20mL within 8 minutes post Iron administration.
Dog12 (1439163) had severe vomiting post administration at following intervals.
20mL within 4minutes + 10mL 9minutes later+ 10mL 7 minutes later+5mL 1 hr 13minutes later+ 6mL 1 hr 38 minutes later Dogl (1437284) vomited 8mL within 10 minutes post administration and another 10mL 53minutes later, and also had Test Item 2 diarrhoea 2 minutes later (1 hour 5 minutes post administration) Iron Dog3 (1454138) had diarrhoea at 1 hour 2mintutes post dose administration.
Period 2 T 1 Dog7 (1544765) has Oedema/swelling and was noticed at surgical est Item port site prior to dose administration.
Ferrous sulfate No adverse clinical observations for other dogs * Only subjects (dogs) showing significant signs of side effects were included, all other animals used in the study had no apparent signs of side effects.
Conclusions [0164] This study summarizes relative performance of an enhanced iron formulation on the oral absorption of iron compared to an unenhanced formulation of ferrous sulfate in an iron deficient dog model designed for this study. To evaluate relative oral bioavailability, the PK parameters were calculated based on fasting plasma iron levels (animals were fed 6 hours post dosing) using measured levels, and levels corrected for baseline levels (obtained immediately prior to dosing). Iron deficiency state was deemed to be adequate for the purposes of the study as the chosen plasma iron and hematocrit targets were achieved prior to dose administration in each period.
The results of the study demonstrate that iron TI2 showed higher systemic absorption of iron compared to ferrous sulfate TI1 in different groups and different dosing periods of this two way crossover study. The relative oral bioavailability (Frei) of iron TI2 calculated using extent of absorption until 6 hours (AUC0_6) showed a 3.45-fold increase (n=11) in relative oral bioavailability compared to an intraduodenally administered ferrous sulfate solution (both solutions contained equal amounts of elemental iron. Twelve iron deficient female beagle dogs administered two single intraduodenal doses of TIl (Ferrous Sulfate Formulation) and iron T12 (Enhanced Formulation) did not exhibit death, morbidity, or severe adverse clinical signs. Clinical signs associated with dose administration of iron T12 Formulation were mild and limited to vomiting and diarrhea.
Individual Animal Raw Data [0165] Tables 36-47 show additional mean pharmacokinetics values and individual animal data.
Table 36: Mean plasma concentration vs. time profiles of the test items dosed at different periods in different groups Plasma Total Concentrations; tig/dL Mean SEM, n=6 (*plasma iron corrected for base line Mean I SEM, n=6) Time Period 1 Period 2 (ars) Group A Group B
Group B Group A
Unenhanced Unenhanced Iron TI2 Iron TI2 TIl TIl (0+0) (0 0) (0 0) (0 0) 0.5 (60130) (167117) (236119) (109126) (58133) (113117) (203128) (110121) (24126) (42119) (148129) (57117) (-14120) (-26115) (41116) (11 8) (-28116) (-34116) (-419) (318) (-11114) (-24120) (84124) (96134) *Plasma iron corrected for baseline(CT-T0); calculated by subtracting baseline plasma iron value (TO).
i.e., CT plasma concentration at each interval ,TO Plasma concentration prior to dose administration.
Table 37: Mean pharmacokinetics of the test items based on total plasma iron in different groups Period 1 Period 2 PK
Group A Group B Group A Group B
parameter#
Unenhanced Iron Form B Iron Form B Unenhanced 611.04+77.03 683.13+5230 1065.13+95.31 642.75+47.61 pg/dl*h (30.88) (18.75) (21.92) (18.14) (%RSD) FRel(0-6) Fold vs. 1.08+0.09** 1.86+0.25*
**
Unenhanced (19.96) (32.30) Cmax 170.00+18.98 256.75+17.14 322.67115.15 188.33+17.56 1.1g/d1 (27.35) (16.35) (11.50) (22.84) (%RSD) Tmax 0.58+0.15 0.50+0.00 0,83+0.25 0.67+0.11 Hrs (64.52) (0.00) (72.66) (38.73) (%RSD) Ratio of C13-1.41+0.14** 2.01+0.23* **
vs. Unenhanced # Mean+ SEM (n=6). * Group A comparison; ** Group B comparison.
Table 38: Mean phannacokineties of the test items based on plasma iron corrected for baseline Period 1 Period 2 PK parameter Group A Group B Group A Group B
Unenhanced Iron Form B Iron Form B Unenhanced 164.98+66.21 244.50+56.40 580.46+99.66 262.00+58.43 Itg/dl*h (98.30) (56.51) (42.05) (54.63) (%RSD) FRel(0-6) Fold vs. 1.97+1.17** 5.22+1.66*#
**
Unenhanced (145.47) (70.96) Cmax 92.90125.25 167.17+17.43 240.17+20.38 122.33+22.01 l_tg/d1 (61.23) (25.54) (20.79) (44.06) (%RSD) Tm 8X 0.67+0.11 0.50+0.00 0.83+0.25 0.67+0.11 Hrs (38.73) (0.00) (72.66) (38.73) (%RSD) Ratio of Cmax vs.
1.83+0.61** 3.51+1.44*# **
Unenhanced # Mean+ SEM (n=6). *#n=5 * Group A comparison; ** Group B comparison Table 39: Individual dog plasma iron concentrations and PK data after dose administration of Unenhanced-TI1(Ferrous Sulfate) at Period 1 in Group A
animals Total Plasma Iron concentration ng/mL
Dog 1 Dog 2 Dog 3 Dog 4 Dog 5 Dog 6 %RS
ID ID ID ID ID ID Mean SEM D
TIME 1437284 1544676 1454138 1544749 1175018 1451716 (n=6) (n=6) 0 147.5 55.5 176.5 69.5 71.5 38.5 93.17122.61 59.44 0.5 99.0 204.5 198.5 114.0 128.0 174.0 153.00118.46 29.56 1 76.0 179.0 229.0 113.0 111.0 197.0 150.83124.26 39.39 2 61.0 134.0 180.5 100.0 92.0 133.0 116.75116.96 35.58 4 59.0 109.0 124.0 72.0 63.0 46.0 78.83112.54 38.97 6 64.0 56.0 101.0 73.0 67.0 31.0 65.3319.31 34.91 24 104.0 52.0 136.0 54.0 122.0 27.0 82.50117.99 53.41 AUC0_6 416.88 725.38 934.88 526.13 496.13 566.88 611.04177.03 30.88 (ng/dl*h) Cmax 147.5 204.5 229.0 114.0 128.0 197.0 170.00118.98 27.35 ( g/d1) Tmax (Hrs) 0.0 0.5 1.0 0.5 0.5 1.0 0.5810.15 64.52 Table 40: Individual plasma iron concentrations and PK data after dose administration of iron TI2 (Ferric Pyrophosphate soluble) at Period 1 in Group B animals Total Plasma Iron concentration Itg/mL
Dog 7 Dog 8 Dog 9 Dog 10 Dog 11 Dog 12 %RS
ID ID ID ID ID ID Mean+SEM D
TIME 1544765 1439686 1444264 1544382 1436946 1439163 (n=6) (n=6) 0 57.5 73.0 88.0 144.5 80.0 94.5 89.58 12.16 33.26 0.5 294.0 260.5 220.5 317.0 237.5 211.0 256.75 17.14 16.35 1 222.0 228.0 169.0 231.5 204.0 161.0 202.58 12.54 15.16 2 166.0 154.0 83.0 156.0 138.0 92.0 131.50 14.44 26.89 4 81.0 78.0 46.0 64.0 62.0 51.0 63.67 5.72 22.00 6 69.0 73.0 45.0 48.0 54.0 46.0 55.83 4.99 21.89 24 99.0 87.0 58.0 49.0 68.0 35.0 66.00 9.74 36.16 AUC0.6 (p,g/d1*h) 807,88 779.50 520.50 778.25 676.75 535.88 683.13 52.30 18.75 FRel(0-6) Fold vs. 1.42 1.16 0.89 1.11 0.81 1.06 1.0810.09 19.96 Unenhanced Cmax 294.0 260.5 220.5 317.0 237.5 211.0 256.75 17.14 16.35 (m/d1) Tmax 0.5 0.5 0.5 0.5 0.5 0.5 0.50 0.00 0.00 (Firs) Table 41: Total plasma iron concentration and PK data of individual dogs after dose administration of Unenhanced TI1 (Ferrous Sulfate) at Period 2 Group B animals Total Plasma Iron concentration g/mL
Dog 7 Dog 8 Dog 9 Dog 10 Dog 11 Dog 12 %RS
ID ID ID ID ID ID Mean SEM D
TIME 1544765 1439686 1444264 1544382 1436946 1439163 (n=6) (n=6) 0 99.0 87.0 58.0 49.0 68.0 35.0 66.00+9.74 36.16 0.5 151.0 144.0 190.0 226.0 253.0 86.0 175.00+24.74 34.63 1 145.0 159.0 147.0 203.0 251.0 151.0 176.00+17.39 24.20 2 92.0 122.0 98.0 118.0 181.0 124.0 122.50+12.88 25.75 4 75.0 94.0 73.0 84.0 82.0 54.0 77.00+5.51 17.54 6 70.0 89.0 69.0 76.0 64.0 46.0 69.00+5.79 20.54 24 1510 136.0 123.0 314.0 151.0 98.0 162.33+ 31.42 47.41 AUCo-6 (ng/dL*h) 567.00 673.00 581.75 698.50 831.25 505.00 642.75+47.61 18.14 Cmax ( g/d1) 151.0# 159.0 190.0 226.0# 253.0 151.0 188.33117.56 22.84 Tmax (Hrs) 0.5# 1.0 0.5 0.5# 0.5 1.0 0.67+0.11 38.73 # based on 0-6 interval Table 42: Total plasma iron concentration and PK data of individual dogs after dose administration of iron T12 (Ferric pyrophosphate soluble) at Period 2 Group A
animals Total Plasma Iron concentration ftg/mL
Dog 1 Dog 2 Dog 3 Dog 4 Dog 5 Dog 6 %RS
ID ID ID ID ID ID Mean SEM D
TIME 1437284 1544676 1454138 1544749 1175018 1451716 (n=6) (n=6) 0 104.0 510 136.0 54.0 122.0 27.0 82.50+17.99 53.41 0.5 275.0 297.0 355.0 285.0 360.0 340.0 318,67+15.27 11.74 1 246.0 225.0 309.0 292.0 288.0 352.0 285.33+18.48 15.86 2 204.0 120.0 285.0 297.0 223.0 251.0 230.00+26.32 28,03 4 129.0 510 177.0 159.0 136.0 90.0 123.83+18.75 37.08 6 104.0 43.0 112.0 85.0 93.0 34,0 78,50+13.25 41.34 24 300.0 131.0 166.0 149.0 184.0 71.0 166.83130.99 45.50 AUCo-6 (p.g/dL*h) 1016.00 657.25 1336.75 1223,50 1126.00 1031,25 1065.13+95,31 21.92 FRel(0-6) Fold vs.
Unenhanced 2.44 0.91 1.43 2.33 2.27 1.82 1.86+0,25 32,30 Cinax (tg/d1) 275.011 297.0 355.0 297,0 360.0 352.0 322.67+15.15 11,50 Tmax (Hrs) 0.5# 0.5 0.5 2.0 0.5 1.0 0.83+0.25 72.66 # based on 0-6 interval Table 43: Baseline corrected plasma iron concentration and PK data of individual dogs after dose administration of Unenhanced TI1 (Ferrous Sulfate) at Period 1 in Group A
animals Absorbed iron concentration p,g/mL*( TO-CT) Dog 1 Dog 2 Dog 3 Dog 4 Dog 5 Dog 6 %RSD
TIME ID ID ID ID ID ID Mean SEM (n=6) 1437284 1544676 1454138 1544749 1175018 1451716 (n=6) 0 0.0 0.0 0.0 0.0 0.0 0.0 0.00+0.00 0.00 0.5 -48.5 149.0 22.0 44.5 56.5 135.5 59.83 30.05 123.02 1 -71.5 123.5 52.5 43.5 39.5 158.5 57.67+32.53 138.17 2 -86.5 78.5 4.0 30.5 20.5 94.5 23.58+26.21 272.20 4 -88.5 53.5 -52.5 2.5 -8.5 7.5 -14.33+20.29 -346.78 6 -83.5 0.5 -75.5 3.5 -4.5 -7.5 -27.83+16.45 -144,73 24 -43,5 -3.5 -40.5 -15.5 50.5 -11.5 -10.67+13.89 -318.88 AUCo-6 (p.g/d1*h) 0.00 392.38 56.38 109,13 88.63 343.38 164.98+66.21 98.30 C..:
(fig/d1) -48.5** 149.0 52.5 44.5 56.5 158.5 92.90 25.25# 61.23#
T,,,,.
(Hrsµ) 0.5** 0.5 1.0 0.5 0.5 1.0 0.67+0.11 38.73 *TO Plasma Iron concentration, CT Plasma Iron concentration at each interval of time after dose administration. All negative values set at zero for purposes of calculating AUC values.
**Calculated over 0-6 hour period. #n5 Table 44: Baseline corrected plasma iron concentration and PK data of individual dogs after dose administration of iron TI2 (Ferric pyrophosphate soluble) at Period 1 in Group B animals Absorbed iron concentration vig/mL*( TO-CT) Dog 7 Dog 8 Dog 9 Dog 10 Dog 11 Dog 12 ID ID ID
ID ID ID Mean SEM %RSD
TIME 1544765 1439686 1444264 1544382 1436946 1439163 (n=6) (n=6) 0 0.0 0.0 0.0 0.0 0.0 0.0 0.00+0.00 0.00 0.5 236.5 187.5 132.5 172.5 157.5 116.5 167.17+17.43 25.54 1 164.5 155.0 81.0 87.0 124.0 66.5 113,00+16.73 36.27 2 108.5 81.0 -5.0 11.5 58.0 -2.5 41.92+19.42 113.51 4 23.5 5.0 -42.0 -80.5 -18.0 -43.5 -25.92+15.28 -144.45 6 11.5 0.0 -43,0 -96.5 -26.0 -48.5 -33.75+15.80 -114.66 24 41.5 14.0 -30.0 -95.5 -12.0 -59.5 -23,58+20,24 -210.19 AUCo-6 ( g/d1*h) 462.88 341.50 127.00 168.75 258.75 108.13 244.50+56.40 56.51 FRel(0-6) Fold vs.
Unenhanced 7.65 2.26 0.54 0.42 0.61 0.37 1.9711.17 145.47 Cm , (llgiaclx1) 236.5 187.5 132.5 1715 157.5 116.5 167.17+17.43 25.54 Tnia:
(Hrsµ) 0.5 0.5 0.5 0.5 0.5 0.5 0.50+0.00 0.00 *TO Plasma Iron concentration, CT Plasma Iron concentration at each interval of time after dose administration. All negative values set at zero for purposes of calculating AUC
values.**Calculated over 0-6 hour period.
Table 45: Baseline corrected plasma iron concentration and PK data of individual dogs after dose administration of TI1 (Ferrous Sulfate) at Period 2 in Group B animals Total Plasma Iron concentration lig/mL
Dog 7 Dog 8 Dog 9 Dog 10 Dog 11 Dog 12 ID ID ID ID ID ID Mean+SEM %RSD
TIME 1544765 1439686 1444264 1544382 1436946 1439163 (n=6) (n=6) 0 0.0 0.0 0.0 0.0 0.0 0.0 0.00+0.00 0.00 0.5 52.0 57.0 132.0 177.0 185.0 51.0 109.00+25.98 58.38 _ 1 46.0 72.0 89.0 154.0 183.0 116.0 110.00+21.04 46.85 2 -7.0 35.0 40.0 69.0 113.0 89.0 56.50+17.48 75.80 4 -24.0 7.0 15.0 35.0 14.0 19.0 11.00+7.97 177.49 6 -29.0 2.0 11.0 27.0 -4.0 11.0 3.00+7.70 628.58 24 53.0 49.0 65.0 265.0 83.0 63.0 96.33+34.08 86.65 AUCo-6 ( g/dL*h) 60.50 151.00 233.75 404.50 427.25 295.00 262.00 58.43 54.63 Cmax WA 52.0# 72.0 132.0 177.01/ 185.0 116.0 122.33+22.01 44.06 Tmax (Hrs) 0.5# 1 0.5 0,5# 0.5 1 0.6710.11 38.73 # based on 0-6 interval Table 46: Baseline corrected plasma iron concentration vs. time profiles and PK data of individual dogs after dose administration of T12 (iron Form B) at Period 2 Group A animals Total Plasma Iron concentration vtg/mL
Dog 1 Dog 2 Dog 3 Dog 4 Dog 5 Dog 6 ID ID ID ID ID ID Mean SEM %RSD
TIME 1437284 1544676 1454138 1544749 1175018 1451716 (n=6) (n=6) 0 0.0 0,0 0.0 0.0 0.0 0.0 0.00+0.00 0.00 0,5 171.0 245.0 219.0 231.0 238.0 313.0 236.17118.76 19.45 1 142.0 173.0 173.0 238.0 166.0 325.0 202.83127.69 33.44 2 100.0 68.0 149.0 243.0 101,0 224.0 147.50129.28 48.62 4 25.0 0.0 41.0 105.0 14.0 63.0 41.33115.54 92.10 6 0.0 -9.0 -24.0 -31.0 -29.0 7.0 -4.00+8,97 -549.09 24 196.0 79.0 30,0 95.0 62.0 44.0 84.33124.28 70.53 ( g/dL*h) 392.00 354.25 544.75 899.50 423.00 869.25 580.46 99.66 42.05 FRel(0-6) Fold VS.
Unenhanced #DIV/0! 0.90 9.66 8.24 4.77 2.53 5.22 1,51 70.96 Cmax (4/d1) 171.0#
245.0 219.0 243.0 238.0 325.0 240.17 20.38 20.79 Tmax (Hrs) 0.5# 0.5 0.5 2.0 0.5 1.0 0.8310.25 72,66 # based on 0-6 interval Table 47: Mean overall plasma concentration (ug/dL - Mean SEM, n=12) vs.
time profiles of the test items Time Unenhanced Iron Form B
Total plasma Base line Total plasma Base line Iron corrected* Iron corrected*
0 79.58112.43 0.0010.00 86.04110.41 0.0010.00 0.5 164.00115.09 84.42120.34 287.71114.38 201.67116.04 1 163.42114.73 83.83120,08 243.96116.40 157.92120.53 2 119.63110.19 40.04115.82 180.75120.62 94.71123,11 4 77.9216.54 -1.67111,07 93.75113.02 7.71114.52 6 67.1715.26 -12.4219.83 67.1717.56 -18.88 9.75 24 122.42121.04 42.83123.83 116.42121.70 30.38122.18 *Amount plasma iron corrected for baseline (CT-T0); calculated by subtracting baseline plasma iron value (TO). i.e. CT plasma concentration at each interval ,TO Plasma concentration prior to dose administration.

Effect of dosage forms on iron feedback mechanism [0166] Data collected in Example 12 were evaluated to determine the relationship between iron absorption and the iron levels at baseline for each of the study periods. It was found in this analysis that the amount of iron absorbed from the unenhanced formulation decreased as the iron levels at baseline increased. Fig. 21 demonstrates this relationship. The unenhanced AUCs vs. predose iron levels displays a clear relationship between predose iron levels and absorbed iron. Animals with higher predose iron levels absorbed less iron from the unenhanced test formulation. This fits with the theory that iron is absorbed by active transport and there is a feedback mechanism.
[0167] In contrast, for the enhanced formulation, this decrease in absorption with increased baseline iron levels was not similarly seen. In fact, the dog with the third highest baseline level was the second highest absorbing subject. Fig. 22, the plot of enhanced iron AUCs vs. predose iron levels, demonstrates this. This is likely a result of paracellular and transcellular absorption of iron promoted by the enhanced formulation caused by the formation of mixed micelles of the iron and the enhancer system.
Hence, while absorption of iron by active transport is suppressed when predose iron levels rige, absorption by paracellular and transcellular absorption promoted by enhancers of the invention is not affected by negative feedback.
[0168] The foregoing is illustrative of the present invention and is not to be construed as limiting thereof. Although a few exemplary embodiments of this invention have been described, those skilled in the art will readily appreciate that many modifications are possible in the exemplary embodiments without materially departing from the novel teachings and advantages of this invention. Accordingly, all such modifications are intended to be included within the scope of this invention as defined in the claims. Therefore, it is to be understood that the foregoing is illustrative of the present invention and is not to be construed as limited to the specific embodiments disclosed, and that modifications to the disclosed embodiments, as well as other embodiments, are intended to be included within the scope of the appended claims. The invention is defined by the following claims, with equivalents of the claims to be included therein.

Claims (54)

1. A pharmaceutical composition comprising an iron compound and an absorption enhancer, wherein the absorption enhancer is a medium chain fatty acid salt having a carbon chain length of from about 4 to about 20 carbon atoms, and wherein the composition does not involve chemical modification of the iron compound.
2. A pharmaceutical composition comprising an iron compound and an absorption enhancer, wherein the composition provides a bioavailability of iron that is at least 1.5 times greater than the bioavailability provided by a composition comprising ferrous sulfate that does not contain an absorption enhancer.
3. The composition of claim 2, wherein the composition provides a bioavailability of iron that is at least four times greater than the bioavailability provided by a ferrous sulfate tablet that does not contain an absorption enhancer.
4. The pharmaceutical composition of any one of claims 1-3, wherein the iron compound is an iron salt, an iron chelate, or an iron complex.
5. The pharmaceutical composition of claim 4, wherein the iron compound is a ferric complex.
6. The pharmaceutical composition of claim 4, wherein the iron compound is a complex of iron with EDTA and sodium.
7. The pharmaceutical composition of claim 4, wherein the iron compound is a ferric chelate.
8. The pharmaceutical composition of claim 4, wherein the iron compound is ferric pyrophosphate.
9. The pharmaceutical composition of claim 4, wherein the iron compound is soluble ferric pyrophosphate.
10. The pharmaceutical composition of claim 1, wherein the medium chain fatty acid salt is solid at room temperature.
11. The pharmaceutical composition of claim 1, wherein the carbon chain length is from 8 to 14 carbon atoms.
12. The pharmaceutical composition of claim 1, wherein the absorption enhancer is a sodium salt of a medium chain fatty acid.
13. The pharmaceutical composition of claim 1, wherein the absorption enhancer is selected from the group consisting of sodium caprylate, sodium caprate, and sodium laurate.
14. The pharmaceutical composition of claim 1, wherein the iron compound and the absorption enhancer are present in a ratio of from 1:100,000 to 10:1 iron compound:enhancer.
15. The pharmaceutical composition of claim 1, wherein the medium chain fatty acid salt is the only absorption enhancer in the composition.
16. The pharmaceutical composition of claim 1, wherein the pharmaceutical composition comprises more than one medium chain fatty acid salt.
17. The pharmaceutical composition of claim 1, wherein the iron compound is the only active agent in the composition.
18. The pharmaceutical composition of claim 1, wherein the composition further comprises an additional active agent.
19. The pharmaceutical composition of claim 18, wherein the additional active agent is selected from the group consisting of folic acid, vitamin A, vitamin B (all series), vitamin C, vitamin D, vitamin E, calcium, chromium, copper, magnesium, manganese, potassium, selenium, zinc, phosphorus, iodine, biotin. inositol, para-amino benzoic acid, choline, and any combination thereof.
20. The pharmaceutical composition of claim 1, wherein the composition further comprises an auxiliary excipient.
21. The pharmaceutical composition of claim 20, wherein the excipient is a solubilizer.
22. The pharmaceutical composition of claim 21, wherein the solubilizer is an organic chelation agent,
23. The pharmaceutical composition of claim 21, wherein the solubilizer is citric acid, a salt of citric acid, ascorbic acid, or a salt of ascorbic acid.
24. The pharmaceutical composition of claim 21, wherein the solubilizer is present in an amount sufficient to increase the dissolution rate of the iron compound and the enhancer by about 5% and achieve at least about 80% dissolution of both the iron compound and the enhancer in 3 hours.
25. An oral dosage form comprising the pharmaceutical composition of claim 1 or 1
26. The oral dosage form of claim 25, which is a liquid oral dosage form.
27. The oral dosage form of claim 25, which is a solid oral dosage form.
28. The oral dosage form of claim 25, comprising about 1 mg to about 200 mg elemental iron.
29. The solid oral dosage form of claim 27, wherein the dosage form is a tablet, a capsule, a multiparticulate, or a powder dosage form.
30. The solid oral dosage form of claim 25, wherein the dosage form is a controlled release dosage form.
31. The solid oral dosage form of claim 25, wherein the dosage form is a delayed release dosage form.
32. The solid oral dosage form of claim 25, wherein the dosage form further comprises a rate controlling polymer material.
33. The solid oral dosage form of claim 27, wherein the dosage form is an enteric coated dosage form.
34. The solid oral dosage form of claim 32, wherein the rate-controlling polymer is hydroxypropyl methylcellulose.
35. The solid oral dosage form of claim 32, wherein the rate-controlling polymer is a polymer of acrylic or methacrylic acid or their respective esters or copolymers of acrylic or methacrylic acid or their respective esters.
36. The solid oral dosage form of claim 27, wherein the iron compound, the absorption enhancer, and at least one auxiliary excipient are compressed into tablet form prior to coating with a rate controlling polymer or a delayed release polymer.
37. The solid oral dosage form of claim 32, wherein the iron compound, the absorption enhancer, the rate controlling polymer and at least one auxiliary excipient are compressed to form a controlled release matrix tablet and optionally coated with a rate-controlling polymer or a delayed release polymer.
38. The solid oral dosage form of claim 27, wherein the iron compound, the absorption enhancer, and at least one auxiliary excipient are compressed into the form of a multilayer tablet prior to coating with a rate controlling-polymer or a delayed release polymer.
39. The solid oral dosage form of claim 27, wherein the iron compound and the absorption enhancer are dispersed in the rate-controlling polymer material and compressed into the form of a multilayer tablet and optionally coated with a rate-controlling polymer or a delayed release polymer.
40. The solid oral dosage form according to claim 27, wherein the iron compound, the absorption enhancer, at least one auxiliary excipient, and the rate-controlling polymer material are combined into a multiparticulate form.
41. The solid oral dosage form according to claim 40, wherein the multiparticulate form comprises discrete particles, pellets, minitablets, or combinations thereof.
42. The solid oral dosage form according to claim 40, comprising a blend of two or more populations of particles, pellets or mini-tablets having different in vitro or in vivo release characteristics.
43. The solid oral dosage form according to claim 40, wherein the multiparticulate is encapsulated in capsules and optionally coated with a rate-controlling polymer or a delayed release polymer.
44. The solid oral dosage form according to claim 40, wherein the multiparticulate is incorporated into a sachet.
45. The solid oral dosage form according to claim 41, wherein the discrete particles or pellets are compressed into tablet form and optionally coated with a rate-controlling polymer or a delayed release polymer.
46. The solid oral dosage form according to claim 41, wherein the discrete particles or pellets are compressed into a multilayer tablet and optionally coated with a rate-controlling polymer or a delayed release polymer.
47. A method of orally delivering iron to a subject, comprising administering to the subject the oral dosage form of claim 25.
48. A method for increasing the level of iron in the blood of a subject in need thereof, comprising administering to the subject the oral dosage form of claim 25.
49. A method for increasing the level of hemoglobin in the blood of a subject in need thereof, comprising administering to the subject the oral dosage form of claim 25.
50. A method for treating an iron deficiency in a subject in need thereof, comprising administering to the subject the oral dosage form of claim 25.
51. A method for treating a disease or disorder characterized by an iron deficiency in a subject in need thereof, comprising administering to the subject the oral dosage form of claim 25.
52. A method for increasing the efficacy of an erythropoiesis-stimulating agent in a subject in need thereof, comprising administering to the subject the oral dosage form of claim 25.
53. A method for reducing the dose of an erythropoiesis-stimulating agent necessary for a therapeutic effect in a subject, comprising administering to the subject the oral dosage form of claim 25.
54. A method for delaying the need for an erythropoiesis-stimulating agent in a subject, comprising administering to the subject the oral dosage form of claim 25.
CA2819234A 2011-01-07 2012-01-06 Pharmaceutical compositions of iron for oral administration Abandoned CA2819234A1 (en)

Applications Claiming Priority (3)

Application Number Priority Date Filing Date Title
US201161430575P 2011-01-07 2011-01-07
US61/430,575 2011-01-07
PCT/US2012/020487 WO2012094598A2 (en) 2011-01-07 2012-01-06 Pharmaceutical compositions of iron for oral administration

Publications (1)

Publication Number Publication Date
CA2819234A1 true CA2819234A1 (en) 2012-07-12

Family

ID=46457982

Family Applications (1)

Application Number Title Priority Date Filing Date
CA2819234A Abandoned CA2819234A1 (en) 2011-01-07 2012-01-06 Pharmaceutical compositions of iron for oral administration

Country Status (9)

Country Link
US (1) US8802114B2 (en)
EP (1) EP2661273A4 (en)
JP (1) JP2014501784A (en)
KR (1) KR20140026354A (en)
CN (1) CN103476419A (en)
AU (1) AU2012204213A1 (en)
BR (1) BR112013017169A2 (en)
CA (1) CA2819234A1 (en)
WO (1) WO2012094598A2 (en)

Cited By (1)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019082128A1 (en) * 2017-10-25 2019-05-02 Apotex Inc. Delayed release deferiprone tablets and methods of using the same

Families Citing this family (19)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
US7658938B2 (en) 1999-02-22 2010-02-09 Merrion Reasearch III Limited Solid oral dosage form containing an enhancer
BRPI0912384A2 (en) * 2008-05-07 2015-10-13 Merrion Res Iii Ltd composition and method for preparing a composition
US20100215743A1 (en) * 2009-02-25 2010-08-26 Leonard Thomas W Composition and drug delivery of bisphosphonates
US20110182985A1 (en) * 2010-01-28 2011-07-28 Coughlan David C Solid Pharmaceutical Composition with Enhancers and Methods of Preparing thereof
WO2011120033A1 (en) * 2010-03-26 2011-09-29 Merrion Research Iii Limited Pharmaceutical compositions of selective factor xa inhibitors for oral administration
CA2802184C (en) 2010-06-09 2019-01-08 Emisphere Technologies, Inc. Oral iron deficiency therapy
CA2900043A1 (en) * 2013-02-01 2014-08-07 Charak Llc Methods of treating iron deficiency with soluble ferric pyrophosphate
CN105517454B (en) * 2013-08-28 2018-07-03 帝斯曼知识产权资产管理有限公司 The iron supplement of meat soup concentrate
US20160213044A1 (en) * 2013-08-28 2016-07-28 Dsm Ip Assets B.V. Iron supplementation of a bouillon concentrate
JP6578293B2 (en) * 2013-11-05 2019-09-18 ロックウェル メディカル,インコーポレイテッド Methods for reducing doses of erythropoietin stimulants in patients with reduced responsiveness
US9492421B1 (en) * 2013-11-14 2016-11-15 Argent Development Group, Llc Nutritional supplements for treatment of iron deficiency anemia
WO2016120378A1 (en) 2015-01-29 2016-08-04 Novo Nordisk A/S Tablets comprising glp-1 agonist and enteric coating
GB201515387D0 (en) 2015-08-28 2015-10-14 Amazentis Sa Compositions
CN106344534B (en) * 2016-09-30 2019-07-19 广西科技大学 The preparation method of ferrous sulfate enteric coated drop pills
KR102084489B1 (en) * 2018-05-18 2020-03-04 강원대학교산학협력단 Iron nano colloidal dispersion produced by hot-melting extrusion method and uses thereof
WO2020049074A1 (en) * 2018-09-05 2020-03-12 Renapharma AB An iron containing composition and use thereof
TWI710370B (en) * 2018-12-20 2020-11-21 普惠德生技股份有限公司 Use of composition containing ferroamino acid chelate particles for preparing medicines for treating or slowing down Alzheimer's disease or Parkinson's disease
WO2020230090A1 (en) * 2019-05-14 2020-11-19 Nokha Trading Llp Ferrous sulphate oral compositions
CN111714470B (en) * 2020-05-27 2022-04-08 天津瑞益瑞美生物技术有限公司 Enteric-coated sodium iron ethylene diamine tetraacetate premix for livestock and preparation method thereof

Family Cites Families (148)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
GB953626A (en) 1961-04-25 1964-03-25 Meito Sangyo Kk Enteric-coated tablets of dextran sulphate ester and method of preparation thereof
US4525339A (en) 1982-10-15 1985-06-25 Hoffmann-La Roche Inc. Enteric coated oral dosage form
WO1984004674A1 (en) 1983-05-31 1984-12-06 Jang Choong Gook Dry direct compression compositions for controlled release dosage forms
DE3331009A1 (en) 1983-08-27 1985-03-14 Basf Ag, 6700 Ludwigshafen METHOD FOR INCREASING THE ENTERAL RESORBABILITY OF HEPARIN OR. HEPARINOIDS AND THE SO AVAILABLE HEPARIN OR HEPARINOID PREPARATION
US4590062A (en) 1984-04-16 1986-05-20 Tech Trade Corp. Dry direct compression compositions for controlled release dosage forms
US4654155A (en) 1985-03-29 1987-03-31 Reynolds Metals Company Microemulsion lubricant
US5288497A (en) 1985-05-01 1994-02-22 The University Of Utah Compositions of oral dissolvable medicaments
US4764375A (en) 1985-09-11 1988-08-16 Kv Pharmaceutical Company Sachet drug delivery system
US4789547A (en) 1987-06-17 1988-12-06 Warner-Lambert Company Transdermal matrix system
IL87710A (en) 1987-09-18 1992-06-21 Ciba Geigy Ag Covered floating retard form for controlled release in gastric juice
US5221734A (en) 1987-10-01 1993-06-22 Ciba-Geigy Corporation Process for preparing a polypeptide growth factor for milk
GB2212396A (en) 1987-12-18 1989-07-26 Procter & Gamble Dietary supplement comprising calcium and delayed release coated iron
NO179479C (en) 1988-03-11 1996-10-16 Teikoku Seiyaku Kk Process for the preparation of an intravaginal pharmaceutical preparation
US5169933A (en) 1988-08-15 1992-12-08 Neorx Corporation Covalently-linked complexes and methods for enhanced cytotoxicity and imaging
US5190748A (en) 1988-11-22 1993-03-02 Hoffmann-La Roche Inc. Absorption enhancement of antibiotics
ZA898331B (en) 1988-11-22 1990-07-25 Hoffmann La Roche Pharmaceutical compositions
IL92537A (en) 1988-12-15 1994-04-12 Riker Laboratories Inc Topical formulations and transdermal delivery systems containing- isobutyl-1H-imidazo [4,5-c] quinolin-4-amine
GB2244015B (en) 1990-03-14 1993-10-27 Hadley Sections Limited Tube and method of forming same
US5541155A (en) 1994-04-22 1996-07-30 Emisphere Technologies, Inc. Acids and acid salts and their use in delivery systems
US5110606A (en) 1990-11-13 1992-05-05 Affinity Biotech, Inc. Non-aqueous microemulsions for drug delivery
IT1245761B (en) 1991-01-30 1994-10-14 Alfa Wassermann Spa PHARMACEUTICAL FORMULATIONS CONTAINING GLYCOSAMINOGLICANS ABSORBABLE ORALLY.
US5744450A (en) 1991-03-14 1998-04-28 The Salk Institute For Biological Studies GnRH analogs
JPH06507172A (en) 1991-04-19 1994-08-11 アフィニティー バイオテック,インコーポレイテッド Convertible microemulsion formulation
US5688761A (en) 1991-04-19 1997-11-18 Lds Technologies, Inc. Convertible microemulsion formulations
CA2070061C (en) 1991-06-07 2004-02-10 Shigeyuki Takama Physiologically active polypeptide-containing pharmaceutical composition
AU661080B2 (en) 1991-11-22 1995-07-13 Warner Chilcott Company, Llc Risedronate delayed-release compositions
US5229130A (en) 1991-12-20 1993-07-20 Cygnus Therapeutics Systems Vegetable oil-based skin permeation enhancer compositions, and associated methods and systems
SE501389C2 (en) 1992-04-24 1995-01-30 Leiras Oy Pharmaceutical preparation and process for its preparation
DE4317458A1 (en) 1992-06-11 1993-12-16 Bayer Ag Use of cyclic depsipeptides with 18 ring atoms for the control of endoparasites, new cyclic depsipeptides with 18 ring atoms and process for their preparation
JP3190441B2 (en) 1992-07-20 2001-07-23 エーザイ株式会社 Stable formulation containing azelastine hydrochloride
RU2068689C1 (en) 1992-09-24 1996-11-10 Товарищество с ограниченной ответственностью "Лекрон" Method of paracetamol tablet producing
EP0667148B1 (en) 1992-11-06 2002-07-03 Hisamitsu Pharmaceutical Co., Inc. Peroral pharmaceutical preparation releasable in lower digestive tract
US5346701A (en) 1993-02-22 1994-09-13 Theratech, Inc. Transmucosal delivery of macromolecular drugs
SE9302135D0 (en) 1993-06-18 1993-06-18 Kabi Pharmacia Ab NEW PHARMACEUTICAL COMPOSITION
TW402506B (en) 1993-06-24 2000-08-21 Astra Ab Therapeutic preparation for inhalation
ES2068762B1 (en) 1993-07-21 1995-12-01 Lipotec Sa A NEW PHARMACEUTICAL PREPARATION TO IMPROVE THE BIOAVAILABILITY OF DRUGS OF DIFFICULT ABSORPTION AND PROCEDURE FOR THEIR OBTAINING.
DE69434626D1 (en) 1993-11-17 2006-04-20 Athena Neurosciences Inc TRANSPARENT LIQUID FOR THE ADMINISTRATION OF CAPTIVATED MEDICAMENTS
EP0744941B1 (en) 1994-02-16 2003-06-04 Abbott Laboratories Process for preparing fine particle pharmaceutical formulations
US5506207A (en) 1994-03-18 1996-04-09 The Salk Institute For Biological Studies GNRH antagonists XIII
US5639469A (en) 1994-06-15 1997-06-17 Minnesota Mining And Manufacturing Company Transmucosal delivery system
IL110024A (en) 1994-06-15 1998-04-05 Yissum Res Dev Co Controlled release oral drug delivery system containing hydrogel- forming polymer
GB9414318D0 (en) 1994-07-15 1994-09-07 Dowelanco Ltd Preparation of aqueous emulsions
PT742011E (en) 1994-11-17 2002-10-31 Toray Industries PREPARATION THAT MAY BE ABSORBED BY PERCUTANEOUS VIA
US6524557B1 (en) 1994-12-22 2003-02-25 Astrazeneca Ab Aerosol formulations of peptides and proteins
US5650386A (en) 1995-03-31 1997-07-22 Emisphere Technologies, Inc. Compositions for oral delivery of active agents
EP0827402A2 (en) 1995-05-17 1998-03-11 Cedars-Sinai Medical Center Compositions containing fatty acids for improving digestion and absorption in the small intestine
US5631347A (en) 1995-06-07 1997-05-20 Eli Lilly And Company Reducing gelation of a fatty acid-acylated protein
US6572879B1 (en) 1995-06-07 2003-06-03 Alza Corporation Formulations for transdermal delivery of pergolide
GB9516268D0 (en) 1995-08-08 1995-10-11 Danbiosyst Uk Compositiion for enhanced uptake of polar drugs from the colon
US5626884A (en) * 1995-08-18 1997-05-06 Lockett; Curtis G. Treatment of sickle cell disease
US5766620A (en) 1995-10-23 1998-06-16 Theratech, Inc. Buccal delivery of glucagon-like insulinotropic peptides
US5807983A (en) 1995-12-28 1998-09-15 The Salk Institute For Biological Studies GNRH antagonist betides
US6147088A (en) 1996-05-20 2000-11-14 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6150522A (en) 1996-05-20 2000-11-21 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6150352A (en) 1996-05-20 2000-11-21 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US5932547A (en) 1996-07-03 1999-08-03 Alza Corporation Non-aqueous polar aprotic peptide formulations
US5916582A (en) 1996-07-03 1999-06-29 Alza Corporation Aqueous formulations of peptides
GB9614235D0 (en) 1996-07-06 1996-09-04 Danbiosyst Uk Composition for enhanced uptake of polar drugs from mucosal surfaces
AU718811B2 (en) 1996-10-30 2000-04-20 Theratech, Inc. Fatty acid esters of glycolic acid and its salts as permeation enhancers
US5821230A (en) 1997-04-11 1998-10-13 Ferring Bv GnRH antagonist decapeptides
US5925730A (en) 1997-04-11 1999-07-20 Ferring Bv GnRH antagonists
US6156767A (en) 1997-06-05 2000-12-05 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6156772A (en) 1997-06-05 2000-12-05 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US5981550A (en) 1997-06-05 1999-11-09 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6004984A (en) 1997-06-05 1999-12-21 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
EP1012331B1 (en) 1997-07-01 2006-03-29 Isis Pharmaceuticals, Inc. Compositions and methods for the delivery of oligonucleotides via the alimentary canal
IL121269A0 (en) 1997-07-09 1998-01-04 Dpharm Ltd Compositions and methods for reversibly increasing permeability of biomembranes
IL121268A0 (en) 1997-07-09 1998-01-04 Dpharm Ltd Branched chain fatty acids their derivatives and use in the treatment of central nervous system disorders
SE9703691D0 (en) 1997-10-10 1997-10-10 Astra Ab Pharmaceutical compositions
US6017944A (en) 1997-10-28 2000-01-25 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6326360B1 (en) 1998-03-11 2001-12-04 Grelan Pharmaceuticals Co., Ltd. Bubbling enteric coated preparations
US6025366A (en) 1998-04-02 2000-02-15 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6077847A (en) 1998-04-02 2000-06-20 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US5998432A (en) 1998-04-02 1999-12-07 Merck & Co., Inc. Antagonists of gonadotropin releasing hormone
US6270804B1 (en) 1998-04-03 2001-08-07 Biovail Technologies Ltd. Sachet formulations
GB2336311A (en) 1998-04-15 1999-10-20 Merck & Co Inc Bisphosphonate Dosing Regimen
US6656922B2 (en) 1998-05-28 2003-12-02 Mediplex Corporation, Korea Oral delivery of macromolecules
JP3853985B2 (en) 1998-08-03 2006-12-06 有限会社 健康百二十才 Disinfectant containing iron ions
AU1071200A (en) 1998-10-19 2000-05-08 Biotech Australia Pty Limited Systems for oral delivery
JP2002537321A (en) 1999-02-22 2002-11-05 エラン コーポレイション ピーエルスィー Solid oral dosage form containing enhancer
US7658938B2 (en) 1999-02-22 2010-02-09 Merrion Reasearch III Limited Solid oral dosage form containing an enhancer
US20070148228A1 (en) 1999-02-22 2007-06-28 Merrion Research I Limited Solid oral dosage form containing an enhancer
US8119159B2 (en) 1999-02-22 2012-02-21 Merrion Research Iii Limited Solid oral dosage form containing an enhancer
SE9901272D0 (en) 1999-04-09 1999-04-09 Astra Ab New improved formulation
JP2001081031A (en) 1999-08-30 2001-03-27 Schering Ag Benzamide derivative-containing preparation having improved solubility and oral adsorption
DE60011678T2 (en) 1999-12-08 2005-07-14 Xcyte Therapies, Inc., Seattle DEPSIPEPTIDES AND ITS ANALOGS FOR USE AS IMMUNOSUP PRESSIVES
AU2270101A (en) 1999-12-20 2001-07-03 Merck & Co., Inc. Pharmaceutical kit
US20020002140A1 (en) 2000-01-14 2002-01-03 Holick Michael F. Novel bisphosphonates and uses thereof
EP1267890A2 (en) 2000-04-07 2003-01-02 The Board of Regents of The University of Texas System Unique compositions of zwitterionic phospholipids and bisphosphonates and use of the compositions as bisphosphate delivery systems with reduced gi toxicity
US6468559B1 (en) 2000-04-28 2002-10-22 Lipocine, Inc. Enteric coated formulation of bishosphonic acid compounds and associated therapeutic methods
IT1318539B1 (en) 2000-05-26 2003-08-27 Italfarmaco Spa PROLONGED RELEASE PHARMACEUTICAL COMPOSITIONS FOR THE PARENTERAL ADMINISTRATION OF BIOLOGICALLY HYDROPHILE SUBSTANCES
CN1141974C (en) 2000-06-07 2004-03-17 张昊 Colon-releasing oral biological preparation
HU228400B1 (en) 2000-06-20 2013-03-28 Novartis Ag The use of bisphosphonates for treatment of osteoporosis
WO2002055017A2 (en) 2000-11-21 2002-07-18 Wake Forest University Method of treating autoimmune diseases
GB0029111D0 (en) 2000-11-29 2001-01-10 Novartis Ag Organic compounds
US6379960B1 (en) 2000-12-06 2002-04-30 Isis Pharmaceuticals, Inc. Antisense modulation of damage-specific DNA binding protein 2, p48 expression
US20020103131A1 (en) 2001-01-26 2002-08-01 Jacobson Jill D. Prevention of diabetes by administration of GnRH antagonists
WO2002064148A2 (en) 2001-02-16 2002-08-22 Shimizu Pharmaceutical Co., Ltd. Oral formulations containing mucopolysaccharide for small intestine delivery and their use in the treatment circulatory disorders
NZ527157A (en) 2001-03-01 2005-04-29 Emisphere Tech Inc Compositions for delivering bisphosphonates
CN1277545C (en) 2001-05-02 2006-10-04 诺瓦提斯公司 Use of bisphosphonates in the treatment of bone metastasis associated with prostate cancer
CA2446622C (en) 2001-05-11 2012-08-14 Elan Corporation, Plc Isostearic acid salts as permeation enhancers
WO2003003999A2 (en) 2001-07-02 2003-01-16 Elan Corporation, Plc. Delivery of a bioactive material
US20030031757A1 (en) * 2001-08-03 2003-02-13 Kraft Food Holdings, Inc. Stable and bioavailable iron fortified beverages
US7098305B2 (en) 2001-09-06 2006-08-29 Ardana Bioscience Limited Sustained release of microcrystalline peptide suspensions
DE10157628A1 (en) 2001-11-26 2003-06-12 Zentaris Ag Solution for injection of an LHRH antagonist
US7214662B2 (en) 2001-11-27 2007-05-08 Zentaris Gmbh Injectable solution of an LHRH antagonist
US8535650B2 (en) 2001-12-03 2013-09-17 Soligenix, Inc. Stabilized reverse micelle compositions and uses thereof
WO2003051373A1 (en) 2001-12-13 2003-06-26 Merck & Co., Inc. Liquid bisphosphonate formulations for bone disorders
KR20040066177A (en) 2001-12-19 2004-07-23 알자 코포레이션 Formulation and dosage form for increasing oral bioavailability of hydrophilic macromolecules
AU2003211474A1 (en) 2002-02-28 2003-09-09 Nipro Corporation Stabilized albumin preparaions
US7148257B2 (en) 2002-03-04 2006-12-12 Merck Hdac Research, Llc Methods of treating mesothelioma with suberoylanilide hydroxamic acid
NZ535705A (en) 2002-05-10 2007-08-31 Hoffmann La Roche Ibandronic acid for the treatment and prevention of osteoporosis
US7154002B1 (en) 2002-10-08 2006-12-26 Takeda San Diego, Inc. Histone deacetylase inhibitors
AR047938A1 (en) 2003-08-25 2006-03-15 Combinatorx Inc FORMULATIONS, CONJUGATES AND COMBINATIONS OF PHARMACOS FOR THE TREATMENT OF NEOPLASMS
JP2007505886A (en) 2003-09-19 2007-03-15 ファイザー・プロダクツ・インク Pharmaceutical compositions and methods comprising combinations of 2-methylene-19-nor-vitamin D derivatives and bisphosphonates
WO2005041925A2 (en) * 2003-10-31 2005-05-12 Alza Corporation Compositions and dosage forms for enhanced absorption
US8987322B2 (en) 2003-11-04 2015-03-24 Circ Pharma Research And Development Limited Pharmaceutical formulations for carrier-mediated transport statins and uses thereof
DE10358525A1 (en) 2003-12-13 2005-07-07 Bayer Healthcare Ag Endoparasiticides Means for topical application
AR046773A1 (en) 2003-12-23 2005-12-21 Novartis Ag PHARMACEUTICAL FORMULATIONS OF BISPHOSPHONATES
WO2005062041A1 (en) 2003-12-24 2005-07-07 Astrazeneca Ab Pharmaceutical dissolution testing using a non-ionic surfactant
US7606313B2 (en) 2004-01-15 2009-10-20 Ittiam Systems (P) Ltd. System, method, and apparatus for error concealment in coded video signals
WO2005072747A1 (en) 2004-02-02 2005-08-11 Ono Pharmaceutical Co., Ltd. Bone resorption inhibitors
US20070219131A1 (en) 2004-04-15 2007-09-20 Ben-Sasson Shmuel A Compositions capable of facilitating penetration across a biological barrier
US8241670B2 (en) 2004-04-15 2012-08-14 Chiasma Inc. Compositions capable of facilitating penetration across a biological barrier
US20070212395A1 (en) 2006-03-08 2007-09-13 Allergan, Inc. Ocular therapy using sirtuin-activating agents
ATE475412T1 (en) 2004-05-24 2010-08-15 Warner Chilcott Co Llc ENTERIC SOLID ORAL DOSAGE FORM OF A BISPHOSPHONATE WITH A CHELATE
EP1778278B1 (en) 2004-07-16 2014-05-21 Oakwood Laboratories L.L.C. Gonadotropin releasing hormone antagonists
ES2618028T3 (en) 2004-07-19 2017-06-20 Biocon Limited Insulin-oligomer conjugates, formulations and uses thereof.
US8710181B2 (en) 2004-08-31 2014-04-29 Novo Nordisk A/S Use of tris(hydroxymethyl) aminomethane for the stabilization of peptides, polypeptides and proteins
EP1674082A1 (en) 2004-12-22 2006-06-28 Zentaris GmbH Process for the manufacture of sterile suspensions or lyophilisates of low-soluble basic peptide complexes, pharmaceutical formulations comprising these complexes and their use as medicament
NZ561666A (en) 2005-03-17 2010-05-28 Elan Pharma Int Ltd Nanoparticulate biphosphonate compositions
TWI362392B (en) 2005-03-18 2012-04-21 Novo Nordisk As Acylated glp-1 compounds
IL174387A0 (en) 2005-03-31 2008-01-20 Dexcel Pharma Technologies Ltd A solid composition for intra-oral delivery of insulin
US8968781B2 (en) 2005-04-29 2015-03-03 Cubist Pharmaceuticals, Inc. Therapeutic compositions
JP2008542235A (en) 2005-05-25 2008-11-27 ノボ・ノルデイスク・エー/エス Stabilized polypeptide preparation
TW200716140A (en) 2005-06-17 2007-05-01 Dynamis Therapeutics Inc Treatment of inflammatory conditions
EP1906974A4 (en) 2005-07-22 2010-06-23 Univ California Heparin compostions and selectin inhibition
US20070077313A1 (en) * 2005-10-04 2007-04-05 U.S. Pharmaceutical Corporation Toleration iron supplement compositions
CN101437522A (en) 2006-04-07 2009-05-20 默里昂研究Ⅲ有限公司 Solid oral dosage form containing an enhancer
WO2007146234A2 (en) 2006-06-09 2007-12-21 Merrion Research Ii Limited Solid oral dosage form containing an enhancer
US20090004281A1 (en) 2007-06-26 2009-01-01 Biovail Laboratories International S.R.L. Multiparticulate osmotic delivery system
CN101125132A (en) 2007-07-25 2008-02-20 司炳奎 Oral insulin enteric capsule and its preparation method
BRPI0912384A2 (en) 2008-05-07 2015-10-13 Merrion Res Iii Ltd composition and method for preparing a composition
EP3210474B1 (en) 2008-09-17 2020-11-18 Chiasma, Inc. Pharmaceutical compositions comprising polypeptides and related methods of delivery
US20100215743A1 (en) 2009-02-25 2010-08-26 Leonard Thomas W Composition and drug delivery of bisphosphonates
US20110182985A1 (en) 2010-01-28 2011-07-28 Coughlan David C Solid Pharmaceutical Composition with Enhancers and Methods of Preparing thereof
WO2011120033A1 (en) 2010-03-26 2011-09-29 Merrion Research Iii Limited Pharmaceutical compositions of selective factor xa inhibitors for oral administration
CN103370051A (en) 2010-12-15 2013-10-23 迈瑞昂研究第三有限公司 Pharmaceutical compositions of selective factor xa inhibitors for oral administration

Cited By (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2019082128A1 (en) * 2017-10-25 2019-05-02 Apotex Inc. Delayed release deferiprone tablets and methods of using the same
US10780055B2 (en) 2017-10-25 2020-09-22 Chiesi Farmaceutici S.P.A. Delayed release deferiprone tablets and methods of using the same
US10940115B2 (en) 2017-10-25 2021-03-09 Chiesi Farmaceutici S.P.A. Delayed release deferiprone tablets and methods of using the same
US10940116B2 (en) 2017-10-25 2021-03-09 Chiesi Farmaceutici S.P.A. Delayed release deferiprone tablets and methods of using the same
US11357731B2 (en) 2017-10-25 2022-06-14 Chiesi Farmaceutici S.P.A. Delayed release deferiprone tablets and methods of using the same
US11458103B2 (en) 2017-10-25 2022-10-04 Chiesi Farmaceutici S.P.A. Delayed release deferiprone tablets and methods of using the same
US11607389B2 (en) 2017-10-25 2023-03-21 Chiesi Farmaceutici S.P.A. Delayed release deferiprone tablets and methods of using the same
US11723874B2 (en) 2017-10-25 2023-08-15 Chiesi Farmaceutici S.P.A. Delayed release deferiprone tablets and methods of using the same

Also Published As

Publication number Publication date
US20120189692A1 (en) 2012-07-26
AU2012204213A1 (en) 2013-06-13
CN103476419A (en) 2013-12-25
WO2012094598A2 (en) 2012-07-12
BR112013017169A2 (en) 2016-10-04
KR20140026354A (en) 2014-03-05
EP2661273A4 (en) 2014-06-04
US8802114B2 (en) 2014-08-12
JP2014501784A (en) 2014-01-23
WO2012094598A3 (en) 2012-10-26
EP2661273A2 (en) 2013-11-13

Similar Documents

Publication Publication Date Title
US8802114B2 (en) Pharmaceutical compositions of iron for oral administration
EP0792154B1 (en) Composition of cisplatin in combination with 2,2&#39;-dithio-bis(ethanesulfonate) (dimesna)
TWI410258B (en) Improved stability in vitamin and mineral supplements
Emmett et al. Calcium acetate control of serum phosphorus in hemodialysis patients
US11278591B2 (en) Compositions comprising choline
US20090028962A1 (en) Methods and Compositions for Enhancing Iron Absorption
US20090124572A1 (en) Iron-containing nutritional supplement
US20120288531A1 (en) pharmaceutical compositions for delivery of ferric iron compounds, and methods of use thereof
AU2008231033A1 (en) Anti-inflammatory compositions comprising tetracyclines and use thereof in therapy
UA123852C2 (en) A pharmaceutical composition for anaemia
KR20160105499A (en) Dosage regimen of ferric trimaltol
US10786514B2 (en) Dosage regiment of ferric maltol
Funk et al. Interactions Between Iron (III)-hydroxide Polymaltose Complex and Commonly Used Medications
EP0427078A1 (en) Folinic acid-cyclodextrin inclusion compound
WO2017120311A1 (en) Composition therapy with an iron compound and a citrate compound
US20150313939A1 (en) Oral iron (iii) based phosphate adsorbent for treating iron-deficiency anemia in cats with chronic kidney disease
KR101324425B1 (en) Bisphosphonate composition having enhanced oral bioavailability
KR20190008291A (en) Ready-to-use injectable compositions of iron dextran complexes containing vitamins for the prevention and treatment of anemia
Bereda Anaemia and Antianaemic Medications

Legal Events

Date Code Title Description
FZDE Discontinued

Effective date: 20180108