US20200206223A1 - Liquid dosage forms of imatinib - Google Patents

Liquid dosage forms of imatinib Download PDF

Info

Publication number
US20200206223A1
US20200206223A1 US16/633,048 US201816633048A US2020206223A1 US 20200206223 A1 US20200206223 A1 US 20200206223A1 US 201816633048 A US201816633048 A US 201816633048A US 2020206223 A1 US2020206223 A1 US 2020206223A1
Authority
US
United States
Prior art keywords
canceled
liquid dosage
imatinib
dosage form
sodium
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
US16/633,048
Other languages
English (en)
Inventor
Sandip Mehta
Vijay Patel
Manish Umrethia
Jayanta Kumar Mandal
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.)
FTF Pharma Pvt Ltd
Original Assignee
FTF Pharma Pvt 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 FTF Pharma Pvt Ltd filed Critical FTF Pharma Pvt Ltd
Assigned to FTF PHARMA PRIVATE LIMITED reassignment FTF PHARMA PRIVATE LIMITED ASSIGNMENT OF ASSIGNORS INTEREST (SEE DOCUMENT FOR DETAILS). Assignors: MANDAL, JAYANTA KUMAR, MEHTA, SANDIP, PATEL, VIJAY, UMRETHIA, Manish
Publication of US20200206223A1 publication Critical patent/US20200206223A1/en
Abandoned legal-status Critical Current

Links

Classifications

    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K31/00Medicinal preparations containing organic active ingredients
    • A61K31/33Heterocyclic compounds
    • A61K31/395Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins
    • A61K31/495Heterocyclic compounds having nitrogen as a ring hetero atom, e.g. guanethidine or rifamycins having six-membered rings with two or more nitrogen atoms as the only ring heteroatoms, e.g. piperazine or tetrazines
    • A61K31/505Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim
    • A61K31/506Pyrimidines; Hydrogenated pyrimidines, e.g. trimethoprim not condensed and containing further heterocyclic rings
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0087Galenical forms not covered by A61K9/02 - A61K9/7023
    • A61K9/0095Drinks; Beverages; Syrups; Compositions for reconstitution thereof, e.g. powders or tablets to be dispersed in a glass of water; Veterinary drenches
    • 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
    • A61K47/02Inorganic compounds
    • 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
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/10Alcohols; Phenols; Salts thereof, e.g. glycerol; Polyethylene glycols [PEG]; Poloxamers; PEG/POE alkyl ethers
    • 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
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/12Carboxylic acids; Salts or anhydrides 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
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/08Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite containing oxygen, e.g. ethers, acetals, ketones, quinones, aldehydes, peroxides
    • A61K47/14Esters of carboxylic acids, e.g. fatty acid monoglycerides, medium-chain triglycerides, parabens or PEG fatty acid esters
    • 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
    • A61K47/06Organic compounds, e.g. natural or synthetic hydrocarbons, polyolefins, mineral oil, petrolatum or ozokerite
    • A61K47/26Carbohydrates, e.g. sugar alcohols, amino sugars, nucleic acids, mono-, di- or oligo-saccharides; Derivatives thereof, e.g. polysorbates, sorbitan fatty acid esters or glycyrrhizin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/34Macromolecular compounds obtained otherwise than by reactions only involving carbon-to-carbon unsaturated bonds, e.g. polyesters, polyamino acids, polysiloxanes, polyphosphazines, copolymers of polyalkylene glycol or poloxamers
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K47/00Medicinal preparations characterised by the non-active ingredients used, e.g. carriers or inert additives; Targeting or modifying agents chemically bound to the active ingredient
    • A61K47/30Macromolecular organic or inorganic compounds, e.g. inorganic polyphosphates
    • A61K47/36Polysaccharides; Derivatives thereof, e.g. gums, starch, alginate, dextrin, hyaluronic acid, chitosan, inulin, agar or pectin
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/0012Galenical forms characterised by the site of application
    • A61K9/0053Mouth and digestive tract, i.e. intraoral and peroral administration
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/08Solutions
    • AHUMAN NECESSITIES
    • A61MEDICAL OR VETERINARY SCIENCE; HYGIENE
    • A61KPREPARATIONS FOR MEDICAL, DENTAL OR TOILETRY PURPOSES
    • A61K9/00Medicinal preparations characterised by special physical form
    • A61K9/10Dispersions; Emulsions

Definitions

  • the present invention relates, in general, to the pharmaceutical field, and more precisely it relates to the liquid dosage forms of protein-tyrosine kinase inhibitor such as Imatinib or pharmaceutically acceptable salt thereof.
  • the present invention relates to ready to use, liquid dosage forms of Imatinib or pharmaceutically acceptable salt thereof and to the processes for the preparation thereof.
  • Imatinib chemically known as N-(4-methyl-3-[4-(pyridin-3-yl)pyrimidin-2-yl]amino)phenyl)-4-[(4-methylpiperazin-1-yl)methyl]benzamide having an empirical formula C 29 H 31 N 7 O and a molecular weight of 493.6 gm/mol has a following structural formula:
  • Imatinib mesylate is a protein-tyrosine kinase inhibitor; it inhibits the abnormal functioning Bcr-Abl tyrosine kinase, which is produced by the Philadelphia chromosome abnormality, found in chronic myeloid leukemia (CML). Imatinib inhibits cell proliferation and induces apoptosis (programmed cell death) in the Bcr-Abl cell lines and in the leukemic cells generated by CML. Imatinib also inhibits proliferation and induces apoptosis in gastrointestinal stromal tumor (GIST) cells, which express an activating c-kit mutation. More recently, the drug has been approved for the treatment of mesenchymal cell neoplasms of the intestinal tract.
  • GIST gastrointestinal stromal tumor
  • Imatinib mesylate can be used as a treatment for patients suffering from hepatic fibrosis based on its ability to downregulate stellate cell activation in culture and in vivo.
  • Imatinib mesylate is well absorbed after oral administration with Cmax achieved within 2-4 hours post-dose. It was also reported that mean absolute bioavailability is 98%. Biotransformation of Imatinib mesylate is via hepatic metabolism and cytochrome P450 enzymes (especially CYP3A4). Imatinib mesylate is converted to its main circulating active metabolite, a N-desmethylated piperazine derivative. This derivative, in vitro, has potency similar to Imatinib mesylate and comprises about 15% of the AUC (area under the curve) for Imatinib mesylate.
  • the elimination half-lives of Imatinib mesylate and its major active metabolite, the N-desmethyl derivative are approximately 18 and 40 hours, respectively and the time to reach peak concentration is 2 to 4 hours.
  • Imatinib mesylate is presently available as tablet form of 100 mg and 400 mg.
  • the approved dosage range for imatinib mesylate in the treatment of CML is 400 mg to 800 mg (400 mg twice a day) and 600 mg per day for gastrointestinal stromal tumors (GIST).
  • amounts of imatinib mesylate effective to treat hepatic fibrosis would broadly range between about 50 mg and about 600 mg per day and preferably between about 50 mg and about 200 mg per day administered orally.
  • EP 1895984 and US 20060275372 describes a stable nanoparticulate composition of imatinib mesylate, or a salt thereof and at least one surface stabilizer.
  • EP 2009008 discloses a pharmaceutical composition comprising imatinib mesylate having less than about 0.09% area HPLC percent units of desmethyl-imatinib mesylate and at least one pharmaceutically acceptable cxcipients.
  • EP 2120877 and US 2010087444 describes a solid dispersion of imatinib mesylate comprising imatinib mesylate and a pharmaceutically acceptable carrier, wherein said carrier is a cellulose derivative.
  • US 2016143850 and EP 3019159 describes a granulate composition of imatinib mesylate comprising of imatinib mesylate, binder and of disintegrant.
  • US 2008119479 discloses a pharmaceutical composition which comprises ZD6474 or a pharmaceutically acceptable salt thereof, and imatinib, in association with a pharmaceutically acceptable cxcipient or carrier.
  • EP 2782560 and US 2015125534 discloses a pharmaceutical powder formulation comprising granules of a tyrosine kinase inhibitor, wherein the granules of the tyrosine kinase inhibitor are coated with an enteric coating, wherein the tyrosine kinase inhibitor is present in an amount of up to 23% by weight based on the total weight of the pharmaceutical powder formulation.
  • WO 2006132930 discloses a pharmaceutical combination comprising a pyrimidylaminobenzamide compound and Imatinib.
  • EP 1893213 and WO 2006132930 discloses a medicament for the treatment of gastrointestinal stromal tumours by use of a pharmaceutical combination comprising of 4-Methyl-3-[[4-(3-pyridinyl)-2-pyrimidinyl]amino]-N-[5-(4-methyl-1H-imidazol-1-yl)-3-(trifluoromethyl)phenyl]benzamide or a pharmaceutically acceptable salt thereof, and imatinib or a pharmaceutically acceptable salt thereof.
  • WO 2014041551 discloses oral aqueous solution comprising Imatinib or pharmaceutically acceptable acid addition salts or polymorphs thereof, process for preparing such solution and their use in the treatment of chronic myeloid leukemia, gastrointestinal stromal tumors.
  • the specification of '551 publication teaches oral aqueous solution which essentially comprises viscosity regulating agent that is used to stabilize the active ingredient (i.e. Imatinib mesylate) or increase the viscosity of the oral solution.
  • Preferred viscosity regulating agents according to the specification of '551 publication are polyvinyl pyrrolidone and hypromellose.
  • Such preferred viscosity regulating agent has not been used in the oral solutions of the present invention being the first major difference between the present invention and the invention disclosed in the specification of '551 publication.
  • the stability achieved by the oral solution of the present invention and results provided in the present specification are without using (i) viscosity regulating agents preferred in the specification of '551 publication, and (ii) any additional stabilizing agent.
  • Another major difference between the invention disclosed in the specification of '551 publication and the present invention is that the oral solution of Imatinib disclosed in the specification of '551 publication is shown supra-bioavailable (which means the AUC and Cmax of the oral solution disclosed in the specification of '551 publication is higher than either the acceptable values or values shown by Imatinib tablets) when tested in rats against Imatinib tablets, whereas the oral solution of the present invention has been found bioequivalent (which means all the pharmacokinetic parameters such as AUC, Cmax is within the acceptable limits when compared with Imatinib tablets) to the marketed Gleevec (Imatinib) tablets during clinical studies in humans.
  • supra-bioavailability may not be advantageous as sometimes it may lead to toxicity and/or other side effects.
  • oral solution disclosed in the specification of '551 publication is supra-bioavailable in rats does not necessarily mean that it can be administered to humans.
  • the oral solution of Imatinib according to the present invention is ready to use for human administration.
  • Imatinib Currently available preparations of Imatinib are solid oral preparations e.g. tablets, and capsules. These preparations have their own disadvantages and limitations, for example they are not suitable for all types of patient populations. Therefore there is an existing need for liquid dosage forms of Imatinib having prolonged stability and palatability.
  • liquid dosage forms including solutions, syrups, suspensions, elixirs, and concentrates offer unique advantages to many patients.
  • liquids may provide better patient compliance for those with swallowing difficulties and better dosage control versus a fixed tablet dose.
  • liquid dosage forms are generally formulated for use in geriatric and pediatric patients.
  • challenges there are also a number of “challenges” surrounding the formulation and development of these forms.
  • Gleevec® tablets can be dissolved in water or apple juice for patients having swallowing difficulty but in any treatment an important consideration is to ensure that the patient receives the correct dose of medicine.
  • Administration of Gleevec® tablets by dissolving in water or apple juice may not administer correct and consistent dose every time. Disadvantages associated with such an administration is that (i) Gleevec tablets take much longer time to get dispersed in water or apple juice and (ii) it leaves behind lots of residues in the container after administration resulting into the administration of incorrect dose. Further, Imatinib has bitter taste and administration with apple juice may mask its taste and increase the palatability and patient compliance.
  • liquid dosage forms provide assurance of dosage uniformity upon administration to patients and eliminates difficulty of administration.
  • Liquid dosage forms can also provide physicians more flexibility in designing dosage regimens for patients. Such liquid dosage forms are advantageous to pediatric patients, geriatric patients and those patients who are unable to take oral therapy.
  • liquid dosage forms represent an ideal dosage form for patients who have difficulty swallowing tablets or capsules. This factor is of particular importance in administration of drugs to children and aged patients. Further, as mentioned above, administration of Imatinib tablets by dispersing in water or apple juice is also not preferred because of administration of incorrect and inconsistent dose every time. It is therefore principal object of the present invention to provide liquid dosage forms of Imatinib or pharmaceutically acceptable salt thereof.
  • the liquid dosage forms of the present invention are useful for administering to pediatric, geriatric patients and other patients who are unable to take solid oral therapy.
  • the liquid dosage forms according to the present invention include liquids, liquid dispersions, suspensions, solutions, emulsions, sprays, spot-on, syrups, elixirs, drops, gels, solution-gels, concentrates and the like.
  • Liquid dosage forms are designed as ready to use liquids and as powder for reconstitution into liquid orals like syrups, solutions, suspensions and emulsions. Powder for reconstitution may require skills & expertise and needs to be prepared by a healthcare provider and may not be prepared by the patient or caregiver. The reconstitution process may also he a time consuming process and the patient cannot be benefited by the immediate dose of Imatinib as and when required. In such a situation, ready to use, liquid dosage forms of Imatinib may be very useful and the patients can be given required doses immediately using ready to use, liquid dosage forms of Imatinib. Therefore, a yet another object of the present invention is to provide ready to use, liquid dosage forms of Imatinib or pharmaceutically acceptable salt thereof.
  • the solution dosage form can be a viable alternative for patients who have problems in swallowing the tablet or capsule dosage form. It provides assurance of dosage uniformity upon administration to patients and eliminates difficulty of administration. A solution can also provide physicians more flexibility in designing dosage regimens for patients. Imatinib solution dosage form is suitable for administration to both pediatric and geriatric patients while also compensating for a good organoleptic properties and remaining suitably stable. Hence, the development of a liquid formulation is therefore desirable since it offers improved patient compliance. A yet another object of the present invention is therefore to develop solution dosage forms of Imatinib or pharmaceutically acceptable salt thereof.
  • the solution dosage forms according to the present invention comprises Imatinib or pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable excipients or additives selected from the group comprising of vehicles, solvents/co-solvents, solubilizers, surfactants, pH adjusting agents and/or pH1 modifying agents and/or buffering agents or any combination thereof.
  • the solution dosage forms according to the present invention may further comprise one or more agents selected from the group comprising of preservatives, sweetening agents, flavoring agents and coloring agents or any combination thereof.
  • Suspensions possess certain advantages over other liquid dosage forms. Some drugs are insoluble in all acceptable media and must, therefore, be administered as a tablet, capsule, or as a suspension. In addition, disagreeable tastes can be masked by a suspension of the drug or a derivative of the drug. Drugs in suspension are chemically more stable than in solution. In another object, the present invention therefore provides suspension dosage forms of imatinib or pharmaceutically acceptable salt thereof.
  • the suspension dosage forms according to the present invention comprises lmatinib or pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable excipients or additives selected from the group comprising of vehicles, solvents/co-solvents, solubilizers, suspending agents/thickening agents/viscosity modifying agents, anti-foaming agents, anti-caking agents, wetting agents, surfactants, pH adjusting agents and/or pH modifying agents and/or buffering agents or any combination thereof.
  • the suspension dosage forms according to the present invention may further comprise one or more agents selected from the group comprising of preservatives, sweetening agents, flavoring agents and coloring agents or any combination thereof.
  • a yet another object of the present invention is to provide liquid dosage forms of Imatinib or pharmaceutically acceptable salt thereof having palatability, prolonged stability and improved and/or comparable pharmacokinetic profile or bioavailability when compared to the known or marketed Imatinib formulations.
  • the liquid dosage forms of the present invention comprise sweetener(s) and flavoring agent(s) which masks the bitter taste of Imatinib and provides pleasant taste.
  • a yet another object of the present invention is to provide process for the preparation of liquid dosage forms of Imatinib or pharmaceutically acceptable salt thereof.
  • a yet another object of the present invention is to provide use of the liquid dosage forms of the present invention in the manufacture of a medicament.
  • a yet another object of the present invention is to provide liquid dosage forms of the present invention for use as a medicament.
  • a yet another object of the present invention is to provide method for the treatment of a disease or disorder that can be treated by inhibiting protein-tyrosine kinase comprising administering to a patient, such as human, an effective dosage amount of a liquid dosage form comprising Imatinib or pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable excipients or additives as disclosed and described herein.
  • a yet another object of the present invention is to provide method for the treatment of a tumor disease or cancer disease comprising administering to a patient, such as human, an effective dosage amount of a liquid dosage form comprising Imatinib or pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable excipients or additives as disclosed and described herein.
  • a yet another object of the present invention is to provide method for the treatment of at least one disease or condition selected from the group comprising of chronic myeloid leukemia, acute lymphoblastic leukemia, myeldysplastic diseases, mycloproliferative diseases, aggressive systemic mastocytosis, hypereosinophilic syndrome and/or chronic eosinophilic leukemia, unresectable, recurrent and/or metastatic dermatofibrosarcoma protuberans, and unresectable and/or metastatic malignant gastrointestinal stromal tumors comprising administering to a patient, such as human, an effective dosage amount of a liquid dosage form comprising Imatinib or pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable excipients or additives as disclosed and described herein.
  • a patient such as human
  • a yet another object of the present invention is to use the liquid dosage forms of the present invention for the treatment of a disease or disorder that can be treated by inhibiting protein-tyrosine kinase.
  • a yet another object of the present invention is to use the liquid dosage forms of the present invention for the treatment of a tumor disease or a cancer disease.
  • a yet another object of the present invention is to use the liquid dosage forms of the present invention for the treatment of at least one diseases or condition selected from the group comprising of chronic myeloid leukemia, acute lymphoblastic leukemia, myelodysplastic diseases, myeloproliferative diseases, aggressive systemic mastocytosis, hypereosinophilic syndrome and/or chronic cosinophilic leukemia, unrescctablc, recurrent and/or metastatic dermatofibrosarcoma protuberans, and unresectable and/or metastatic malignant gastrointestinal stromal tumors.
  • diseases or condition selected from the group comprising of chronic myeloid leukemia, acute lymphoblastic leukemia, myelodysplastic diseases, myeloproliferative diseases, aggressive systemic mastocytosis, hypereosinophilic syndrome and/or chronic cosinophilic leukemia, unrescctablc, recurrent and/or metastatic dermatofibrosarcoma protuberans,
  • Characteristics of an active drug are of major concern in developing an oral liquid dosage formulation.
  • the major challenges in developing oral liquid dosage forms are (i) the stability of a drug in aqueous solution, suspension or emulsion. (ii) the solubility of a drug at the required level, and (iii) an acceptable taste and (iv) to be bioavailable when taken orally. It is the effective use of excipients, which allows formulators overcome these challenges Additionally, an excipient's compatibility with a drug in the solid state cannot infer the same compatibility in liquid dosage.
  • the decision to develop a solution, syrup or a suspension of a drug is influenced by many factors like solubility, particle size and the desired release profile of the drug and properties of the base vehicle like surface tension, viscosity, boiling point, and specific heat of solution, all of which may be affected in various ways.
  • solubility particle size and the desired release profile of the drug and properties of the base vehicle like surface tension, viscosity, boiling point, and specific heat of solution, all of which may be affected in various ways.
  • lack of solubility of the drug in the base vehicle may demand the need for miscible pharmaceutical co-solvents.
  • a miscible solvent may be needed to decrease the solubility of the drug in a primary vehicle in formulating a suspension.
  • the therapeutic utility of drugs involves the application of dosage forms/delivery systems, which serve as carrier systems together with several excipients to deliver the active therapeutic agent to the site of action.
  • Suspensions are an important class of pharmaceutical dosage forms that may be given by many routes, including oral, topical, parenteral, and also used in the eye for ophthalmic purposes.
  • large proportions of new drug candidates that are emerging are predominantly water insoluble and, therefore, demonstrate poor bioavailability in the solution dosage form.
  • suspensions present a viable formulation option for many drugs, particularly for water insoluble, hydrophobic drug substances, there are certain criteria that a well-formulated suspension should meet.
  • the suspension dosage form has long been used for poorly soluble active ingredients for various therapeutic indications. Development of stable suspensions over the shelf life of the drug product continues to be a challenge on many fronts. Drugs from suspension formulations typically exhibit an improved bioavailability when compared to the same drug formulated as a tablet or capsule.
  • suspension dosage forms include effective dispensing of hydrophobic drugs; avoidance of the use of co-solvents; masking of unpleasant taste of certain ingredients; offering resistance to degradation of drugs due to hydrolysis, oxidation or microbial activity; easy swallowing for young or elderly patients; and efficient intramuscular depot therapy.
  • relatively higher concentration of drugs can be incorporated into suspension products.
  • a solution dosage form e.g. a simple mixture
  • drugs are sufficiently soluble to allow suitable strength solution medicines to be developed and manufactured with an acceptable shelf-life.
  • an alternative approach could be to develop a stable aqueous suspension that will allow consistent dosing of the patient.
  • Pharmaceutical suspensions have several advantages and disadvantages when compared to other dosage forms. Since suspensions are liquids, dose adjustment for patients with renal or hepatic impairment, or for pediatric or geriatric patients, may be more straightforward. This is an oversimplification of the development of a dosing strategy for a drug candidate. There are many other details that must be considered for a formulation development project to be successful, but it does provide a simple overview of some of the issues.
  • the suspension must be physically stable (no appreciable settling) for a sufficient time, chemically stable over the required time (shelf-life), possess a viscosity that allows it to be used for its intended purpose, be easily reconstituted by shaking, and be acceptable in use to the patient care-giver or other user.
  • Some materials may possess a combination of properties useful in the formulation and manufacture of stable, elegant pharmaceutical suspensions. Formulation scientists need to consider the totality of properties possessed by a particular excipient. Even though it is being added for one particular characteristic, the other properties will still be present, and will still influence the formulation.
  • active pharmaceutical ingredients are quite hydrophobic with limited solubility. They may also be quite distasteful. Other drugs may also have quite a high chemical degradation precluding them to be administered as aqueous solutions, and in this case, it may be possible to synthesize an insoluble derivative. In other cases, some drugs are required to be present in the gastrointestinal tract or in the pre-corneal pocket with long residence time. For such drugs, a suspension is an ideal delivery system as it provides better chemical stability and larger surface area and is often more bioavailable than aqueous solutions, tablets, and capsules.
  • Formulation of an elegant, stable, preserved, safe, and effective suspension is a technically challenging task compared aqueous solutions, tablets, and capsules.
  • Pharmaceutical suspensions are thermodynamically unstable systems. Thus, preparation of such systems is often associated with problems of physical stability, content uniformity, sedimentation, caking, re-suspendibility, and crystal growth. Furthermore, issues related to the masking of bitter taste and undesirable odor of the pharmaceutical ingredient must be taken into consideration.
  • a quick means to identify whether or not a drug may be more suitable for solution or suspension is to overlap the pH-stability profile with the pH-solubility profile. This overlap creates a window, which may suggest which dosage form might be most desirable and subsequently the type of excipients needed.
  • Oral liquid formulation needs a meticulous blend of ingredients to perform various functions like wetting and solubilization, stabilization and to impart suitable color, taste and viscosity.
  • the blend should be compatible, non-reactive and stable.
  • the common excipients generally required for any liquid formulation are vehicles (base), viscosity builders, stabilizers, preservatives, colors and flavors.
  • solubilizers are required in case of clear liquids, suspending agents are needed for suspensions and emulsifying agents for emulsions.
  • Imatinib is an inhibitor of protein-tyrosine kinase and commercially available as tablets (Gleevec®) in the United States since 2003 and is indicated for the treatment of chronic myelogenous leukemia (CML) and acute lymphocytic leukemia (ALL) that are Philadelphia chromosome-positive (Ph+) and certain types of gastrointestinal stromal tumors (GIST), systemic mastocytosis, and myelodysplastic syndrome.
  • CML chronic myelogenous leukemia
  • ALL acute lymphocytic leukemia
  • GIST gastrointestinal stromal tumors
  • GIST gastrointestinal stromal tumors
  • liquid dosage forms include, but not limited to, liquids, liquid dispersions, suspensions, solutions, emulsions, ointments, creams, sprays, spot-on, syrups, elixirs, drops, gels, solution-gels, concentrates and the like.
  • Such liquid dosage forms can be prepared using appropriate one or more pharmaceutically acceptable excipients or additives.
  • excipients or additives may be known to those skilled in the art.
  • the solution dosage form can be a viable alternative for patients who have problems in swallowing the tablet or capsule dosage form. It provides assurance of dosage uniformity upon administration to patients and eliminates difficulty of administration.
  • Imatinib solution dosage form is suitable for administration to both pediatric and geriatric patients while also compensating for a good organoleptic properties and remaining suitably stable. Hence, the development of a liquid formulation is therefore desirable since it offers improved patient compliance.
  • the present invention therefore provides solution dosage forms of Imatinib or pharmaceutically acceptable salt thereof.
  • Suspensions possess certain advantages over other liquid dosage forms. Some drugs are insoluble in all acceptable media and must, therefore, be administered as a tablet, capsule, or as a suspension. In addition, disagreeable tastes can be masked by a suspension of the drug or a derivative of the drug. Drugs in suspension are chemically more stable than in solution. Therefore, in one of the further aspects, the present invention provides suspension dosage forms of Imatinib or pharmaceutically acceptable salt thereof.
  • Liquid dosage forms are designed as ready to use liquids and as powder for reconstitution into liquid orals like syrups, solutions, suspensions and emulsions. Powder for reconstitution may require skills & expertise and needs to be prepared by a healthcare provider and may not be prepared by the patient or caregiver. The reconstitution process may also be a time consuming process and the patient cannot be benefited by the immediate dose of Imatinib as and when required. In such a situation, ready to use, liquid dosage forms of Imatinib may be very useful and the patients can be given required doses immediately using ready to use, liquid dosage forms of Imatinib. In one of the further aspects, the present invention therefore provides ready to use, liquid dosage forms of Imatinib or pharmaceutically acceptable salt thereof.
  • Liquid dosage forms of an active drug can be prepared using one or more pharmaceutically acceptable excipients or additives suitable for the preparation of liquid dosage forms.
  • the present invention provides liquid dosage forms of Imatinib or pharmaceutically acceptable salt thereof and one or more excipients or additives suitable for preparing liquid dosage forms.
  • pharmaceutically acceptable excipients or additives refers to such pharmaceutically acceptable excipients which are known to those skilled in the all for the purposes of preparing liquid dosage forms of the present invention.
  • Such pharmaceutically acceptable excipients include, vehicles, solvents/co-solvents, solubilizers, solubility enhancing agents, tonicity agents, permeation/penetration enhancers, mucoadhesives, suspending agents/thickening agents/viscosity modifying agents, bulking agents/auxiliary suspending agents, wetting agents, anti-foaming agents, anti-caking agents, stabilizing agents, anti-oxidants, chelating agents, buffering agents/pH modifying agents/pH adjusting agents, surfactants, preservatives, sweetening agents, flavouring agents and the like or any combination thereof.
  • Such pharmaceutically acceptable excipients can be used in an amount which provides the liquid dosage forms of the present invention desired property for which they are intended or desired to use.
  • the present invention provides liquid dosage forms of Imatinib in the form of solution dosage forms comprising Imatinib or pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable excipients or additives selected from the group comprising of vehicles, solvents/co-solvents and/or solubilizers. pH adjusting agents and/or pH modifying agents and/or buffering agents or any combination thereof. One or more surfactants may also be added in the solution dosage forms of the present invention.
  • the present invention provides liquid dosage forms of Imatinib in the form of suspension dosage forms comprising Imatinib or pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable excipients or additives selected from the group comprising of vehicles, solvents/co-solvents and/or solubilizers, suspending agents/thickening agents/viscosity modifying agents, anti-foaming agents, surfactants, antioxidants, pH adjusting agents and/or pH modifying agents and/or buffering agents or any combination thereof.
  • One or more anti-caking agents may also be added in the suspension dosage forms of the present invention.
  • the liquid dosage forms of the present invention may also comprise anti-microbial agents or preserving agents or preservatives.
  • the present invention therefore provides palatable liquid dosage forms comprising Imatinib or pharmaceutically acceptable salt thereof and at least one or both selected from sweeteners/sweetening agents and flavouring agents.
  • liquid dosage forms according to the present invention include, aqueous dosage forms, alcoholic and/or hydro-alcoholic dosage forms and non-aqueous dosage forms.
  • Aqueous dosage forms according to the present invention may also comprise one or more non-aqueous and/or organic solvents.
  • the present invention provides liquid dosage forms of Imatinib in the form of suspensions comprising Imatinib or pharmaceutically acceptable salt thereof, vehicle(s), solvent(s)/co-solvent(s), solubilizer(s), suspending agent(s)/thickening agent(s)/viscosity modifying agent(s), preservative(s), anti-foaming agent(s), wetting agent(s), surfactant(s), pH adjusting agent(s)/pH modifier(s) or buffering agent(s) or both, sweetener(s) and flavoring agent(s).
  • vehicle(s) solvent(s)/co-solvent(s), solubilizer(s), suspending agent(s)/thickening agent(s)/viscosity modifying agent(s), preservative(s), anti-foaming agent(s), wetting agent(s), surfactant(s), pH adjusting agent(s)/pH modifier(s) or buffering agent(s) or both, sweetener(s)
  • the present invention provides liquid dosage forms of Imatinib in the form of solutions comprising Imatinib or pharmaceutically acceptable salt thereof, vehicle(s), solvent(s)/co-solvent(s), solubilizer(s), preservative(s), surfactant(s), pH adjusting agent(s)/pH modifier(s) or buffering agent(s) or both, sweetener(s) and flavoring agent(s).
  • the liquid dosage forms of the invention may be administered orally or via the oral cavity.
  • the liquid dosage forms of the present invention may also be administered transmucosally, sublingually, via the buccal cavity, via mucosal membranes and/or through the gastrointestinal tract.
  • the liquid dosage forms of the present invention may be administered via pulmonary, intravenous, rectal, opththalmic, colonic, parenteral, intracisternal, intravaginal, intraperitoneal, local, or topical administration.
  • liquid dosage forms of the present invention can be given using oral syringe to those patients who cannot take medicine through mouth e.g. patients who have undergone surgery.
  • liquid dosage forms of the present invention are in the form of spray and may be administered by oral route or nasal route.
  • Sprays are known by various names such as aerosol sprays, liquid pump sprays, or activated mists etc.
  • the liquid dosage forms of the present invention are in the form of immediate release dosage forms or modified release dosage forms, such as extended release, controlled release, sustained release, prolonged release and delayed release.
  • the liquid dosage forms comprise Imatinib or pharmaceutically acceptable salt thereof one or more suitable excipients or additives for the preparation of modified release dosage forms such as rate controlling polymers.
  • the liquid dosage forms of the present invention may also be prepared by reconstitution of dry powder in suitable diluent or media such as water.
  • the dry powder for reconstitution may be in the form of immediate release forms and comprise Imatinib or pharmaceutically acceptable salt thereof and one or more suitable excipients selected form the group comprising of fillers, binders, diluents, disintegrants, pore formers, lubricants, glidants, sweeteners, stabilizing agents, antioxidants, flavoring agents, suspending agents/thickening agents/viscosity modifying agents, surfactants, preservatives and plasticizers.
  • the dry powder for reconstitution may also be in the form of modified release forms and comprise modified release pellets, granules or particles. Such modified release pellets, granules or particles comprise one or more suitable excipients such as rate controlling polymers.
  • liquid dosage forms of the invention are suitable for administration to all types of patients' population.
  • liquid dosage forms of the invention are suitable for pediatric and geriatric patients.
  • the liquid dosage forms of the invention are also useful for the patients who are unable to take solid oral therapy.
  • the pH of the liquid dosage forms of the present invention is between about 2.0 and about 11.0. In some of the aspects, the pH of the liquid dosage forms of the present invention is between about 2.0 and about 7.0. In some of the aspects, the pH of the liquid dosage forms of the present invention is between about 3.0 and about 9.0. In some of the aspects, the pH of the liquid dosage forms of the present invention is between about 4.0 and about 8.0. In some of the aspects, the pH of the liquid dosage forms of the present invention is between about 5.0 and about 7.0. In some of the aspects, the pH of the liquid dosage forms of the present invention is between about 5.5 and about 6.5. In some of the aspects, the pH of the liquid dosage forms of the present invention is between about 3.5 and about 5.0.
  • the pH of the liquid dosage forms of the present invention is such that prevents the formation of degradants in undesired amounts and provides the liquid dosage forms of the present invention increased stability when stored under storage conditions.
  • the pH of the liquid dosage forms of the present invention which prevents the formation of degradants in undesired amounts and provides the liquid dosage forms of the present invention increased stability when stored under storage conditions is in the range from about 2.0 to about 7.0.
  • the liquid dosage forms of the present invention are stable for prolonged time when stored under storage conditions.
  • Prolonged time indicates that the liquid dosage forms of the present invention are stable for at least 1 month, at least 3 months, at least 6 months or at least 12 months when stored under storage conditions.
  • stable or “stability” encompass any characteristic of the liquid dosage forms which may be affected by storage conditions including, without limitation, potency, total impurities, degradation products, specific optical rotation, optical purity, water content, appearance, viscosity, sterility, and colour and clarity.
  • the storage conditions which may affect stability include, for example, duration of storage, temperature, humidity, and/or light exposure.
  • “stable” or “storage stable”, or “stability” when used with reference to the liquid dosage forms of the present invention or when used “stable liquid dosage forms” or “stability of the liquid dosage forms” all these terms/phrases refer to dosage forms of the present invention which retain at least about 90%, or at least about 95%, or at least about 96%, or at least about 98%, of the labelled concentration of Imatinib or salt thereof contained in the said dosage form after storage under typical and/or accelerated conditions.
  • stable liquid dosage forms or stability of the liquid dosage forms refer to less than about 15% (area percent), or less than about 10% (area percent), or less than about 7% (area percent), or less than about 5% (area percent), or less than about 2% (area percent) of Imatinib-related impurities are present after storage under typical and/or accelerated conditions.
  • liquid dosage forms of the present invention contain no more than about 15% (area percent), or no more than about 10% (area percent), or no more than about 7% (area percent), or no more than about 5% (area percent), or no more than about 2% (area percent), or no more than about 1% (area percent), or no more than about 0.5% (area percent), or no more than about 0.2% (area percent), or no more than about 0.1% (area percent) any known or unknown single Imatinib-related impurity or other impurity after storage under typical and/or accelerated conditions.
  • liquid dosage forms of the present invention contain no more than about 15% (area percent), or no more than about 10% (area percent), or no more than about 7% (area percent), or no more than about 5% (area percent), or no more than about 2% (area percent), or no more than about 1% (area percent), or no more than about 0.5% (area percent), or no more than about 0.2% (area percent), or no more than about 0.1% (area percent) total Imatinib-related impurities or other impurities after storage under typical and/or accelerated conditions.
  • liquid dosage forms of the present invention may be packaged within any type of pharmaceutically-acceptable package, containers, pumps, bottles with spray pump, bottles with dropper assembly, bottles, collapsible tubes, glass ampoules, stoppered vials, pre-filled syringes, low-density polyethylene (LDPE), high-density polyethylene (HDPE), polyolefin, polypropylene containers/bottles depending upon the quantity of the final dosage form.
  • LDPE low-density polyethylene
  • HDPE high-density polyethylene
  • polypropylene containers/bottles depending upon the quantity of the final dosage form.
  • the bottles or containers without limitation include clear/transparent/opaque or amber colored glass bottles or containers and clear/transparent/opaque or amber colored plastic bottles or containers made from polyethylene, polyamide, polycarbonate, acrylic multipolymers, polypropylene, polyethylene terephthalate, polyvinyl chloride, polystyrene and the like.
  • closures may have different shapes and sizes.
  • the closure of the packaging material may be made from polyethylene, polyamide, polycarbonate, acrylic multipolymers, polypropylene, polyethylene tercphthalate, polyvinyl chloride, polystyrene and the like.
  • Liquid dosage forms of the present invention may be packaged in a sterile single use bottle/container that contains a unit dose for administration to a patient.
  • Suitable bottles/containers may contain volumes between 1-10 ml, 10-20 ml, 20-40 ml, and 40-100 ml, and even more.
  • the container may typically comprise Imatinib or pharmaceutically acceptable salt thereof in an amount of between 10-40 mg, between 40-80 mg, between 80-130 mg, and even more.
  • the container may be a multi-use container (i.e., retains at least one more unit dose after a first unit dose is dispensed).
  • excipients which may be used to prepare liquid dosage forms of the present invention. It is in no way the intention of the present inventor(s)/applicant(s) to limit the scope of the liquid dosage forms of the present invention by the description of following embodiments. Described embodiments are for illustrative purpose only and a skilled person may use other excipients from the same or different classes as well which may provide liquid dosage forms of the present invention same or improved physico-chemical properties, palatability, stability and the like and retain or increase patients' acceptability towards the therapy. Such other excipients, classes of excipients and compositions resulted therefrom are also part of the present invention and covered within the scope of the present invention.
  • Vehicles may be used in the liquid compositions of the present invention.
  • Vehicles are the liquid bases that carry drugs and other excipients in dissolved or dispersed state.
  • Vehicles may be aqueous or non-aqueous or mixture thereof.
  • Non-aqueous solvents/co-solvents may also be added in the liquid compositions of the present invention to increase the solubility of poorly soluble substances and enhance the chemical stability of a drug.
  • Suitable solvents/co-solvents, solubilizers or vehicles, that may be employed, in the liquid compositions of the invention include, but are not limited to, dichloromethane, acetonitrile, ethyl acetate, acetone, propylene carbonate, water, glycerine, coconut fatty acid diethanolamide, medium and/or long chain fatty acids or glycerides, monoglycerides, diglycerides, triglycerides, structured triglycerides, soybean oil, peanut oil, corn oil, corn oil monoglycerides, corn oil diglycerides, corn oil triglycerides, polyethylene glycol, caprylocaproylmacroglycerides, caproyl 90, propylene glycol, polyoxyethylenesorbitan fatty acid esters, polyoxyethylene castor oil derivatives, castor oil, cottonseed oil, olive oil, safflower oil, peppermint oil, coconut oil, palm seed oil, beeswax, oleic
  • the non-aqueous solvent is glycerin.
  • the glycerin may also act as a stabilizing agent when the liquid dosage forms of the present invention comprise water as vehicle and provide desired stability when stored under storage conditions.
  • the amount of glycerin used to provide desired stability to the liquid dosage forms of the present invention is at least about 25% or more, at least about 30% or more, at least about 40% or more, at least about 50% or more, or at least about 60% or more.
  • wetting agents as used herein are routinely used in pharmaceutical formulations, especially in liquid dosage forms to create a homogeneous dispersion of solid particles in a liquid vehicle. This process can be challenging due to a layer of adsorbed air on the particle's surface. Hence, even particles with a high density may float on the surface of the liquid until the air phase is displaced completely.
  • the use of a wetting agent allows removal of adsorbed air and easy penetration of the liquid vehicle into pores of the particle in a short period of time.
  • alcohol, glycerin, and PG are frequently used to facilitate the removal of adsorbed air from the surface of particles.
  • mineral oil is commonly used as a wetting agent.
  • Non-limiting examples of wetting agents are Benzalkonium chloride, Benzcehonium chloride, Cetylpyridinium chloride, Docusate sodium, Nonoxynol 9, Octoxynol, Poloxamer, Poloxamer 124. Poloxamer 188, 237, 338, 407, Polyoxyl 35 castor oil, Polyoxyl 40 hydrogenated castor oil. Polyoxyl 10 oleyl ether. Polyoxyl 20 cetylstearyl ether. Polyoxyl 40 stearate. Polysorbate 20, Polysorbate 40, Polysorbate 60, Polysorbate 80, Sodium lauryl sulfate. Sorbitan monolaurate, Sorbitan monooleate. Sorbitan monopalmitate. Sorbitan monostearate, Tyloxapol and the like or any combinations thereof.
  • Solubility enhancing agents may include, but are not limited to, DL-methionine, caffeine, nicotinamide, vanillin, benzyl alcohol, ethanol and diethylene glycol monoethyl ether and the like or combinations thereof.
  • Stabilizing agents may include, but are not limited to, sodium metabisulphite, sodium bisulphite, ethylene diamine tetraacetic acid (EDTA) or salts thereof, ascorbic acid and the like or combinations thereof.
  • EDTA ethylene diamine tetraacetic acid
  • Penetration/permeation enhancers may include, but are not limited to, nicotinamide, caffeine, peppermint oil, sodium glycocholate, phospholipids, alkyl saccharides, aprotinin, benzalkonium chloride, ceramides, cetylpyridinium chloride, chitosan, chitosan-4-thiobutylamidine, cyclodextrins, dextran sulfate, dodecyl azacycloheptyl-2-ketone, ether lipids (plasmologens), glycerol, glycosylated sphingosines, lauric acid, 23-lauryl ether, lysophosphatidyl choline, menthol, methoxysalicylate, phosphatidyl choline, 1-palmitoyl-2-glutaroyl-sn-glycero-3-phosphocholine, polycarbophil cysteine, poly-L-arg
  • EDTA sodium deoxycholate, sodium glycocholate, sodium glycodeoxycholate, sodiun lauryl sulfate, sodium salicylate, sodium taurocholate, sodium taurodeoxycholate, sodium taurodihydrofusidate, sphingolipids, sterols and the like or combinations thereof.
  • Mucoadhesives and/or suspending agents may also be added in the compositions of the present invention.
  • suitable mucoadhesives include, but are not limited to, hydroxypropyl cellulose, gelatin, crosslinked polyacrylic acid, polymethacrylic acid, polyhydroxyethyl methacrylic acid, hydroxypropyl methyl cellulose, polyethylene glycol, sodium carboxymethyl cellulose, hyaluronic acid, chitosan, polycarbophil, pectin, xanthan gum, alginate, copolymers of dextran, polyacrylamide, acacia, copolymer of caprolactone and ethylene oxide, carbopol 934, tragacanth, eudragit and the like or combinations thereof.
  • Suspending agents may be natural or synthetic gums or gum derivatives or compounds which swell in the presence of aqueous media.
  • Stabilizing agents may include, but are not limited to, sodium metabisulphite, sodium bisulphite, ethylene diamine tetraacetic acid (EDTA) or salts thereof, ascorbic acid and the like or combinations thereof.
  • EDTA ethylene diamine tetraacetic acid
  • the pH of an oral liquid formulation is a key point in many regards. Control of the formulation pH, could prevent large changes during storage. Therefore, most formulations utilize a buffer to control potential changes in the solution pH.
  • the amount of buffer capacity needed is generally between 0.01 and 0.1 M, and a concentration between 0.05 and 0.5 M is usually sufficient.
  • the selection of a suitable buffer should be based on (i) Whether the acid-base forms are listed for use in oral liquids, (ii) The stability of the drug and excipients in the buffer, and (iii) The compatibility between the buffer and container. A combination of buffers can also be used to gain a wider range of pH compared to the individual buffer alone. However, not all buffers are suitable for use in oral liquids.
  • a boric acid buffer may be used for optical and IV delivery but not in oral liquids because of its toxicity.
  • the stabilizing effect of buffers that have multiple charged species in solution could also determine the potential reaction between excipients and API.
  • buffers that use carbonates, citrate, tartrate, and various phosphate salts may precipitate with calcium ions by forming sparingly soluble salts. However, this precipitation is dependent upon the solution pH. The activity of phosphate ions may be lowered due to interactions with other solution components.
  • the solution pH such as temperature, ionic strength, dilution, and the amount and type of co-solvents present.
  • the pH of acetate buffers is known to increase with temperature, whereas the pH of boric acid buffers decreases with temperature.
  • the drug in solution may itself act as a buffer. If the drug is a weak electrolyte, such as salicylic acid or ephedrine, the addition of base or acid, respectively, will create a system in which the drug can act as a buffer.
  • Suspending agents impart viscosity, and thus retard particle sedimentation.
  • Other factors considered in the selection of the appropriate agent include desired rheological property, suspending ability in the system, chemical compatibility with other excipients, pH stability, length of time to hydrate, batch-to-batch reproducibility, and cost.
  • pH adjusting agents/modifiers and buffers are Acetic acid, Adipic acid, Ammonium carbonate, Ammonium hydroxide, Ammonium phosphate. Boric acid, Citric acid, Diethanolamine, Fumaric acid. Hydrochloric acid, Malic acid. Nitric acid. Propionic acid. Potassium acetate.
  • Potassium bicarbonate Potassium chloride, Potassium citrate, Potassium metaphosphate. Potassium phosphate, Sodium acetate. Sodium bicarbonate. Sodium borate, Sodium carbonate. Sodium chloride. Sodium citrate, Sodium glycolate, Sodium hydroxide, Sodium lactate, Sodium phosphate, Sodium proprionate, Succinic acid, Sulfuric acid. Tartaric acid. Triethylamine, Triethanolamine, Tromethamine. Trolamine and the like or any combinations thereof.
  • buffering agent or “buffering agents” may also interchangeably be used with the terms “pH adjusting agent or pH adjusting agents” or “pH modifying agent or pH modifying agents” and vice versa and represents the same meaning.
  • Suspending agents can be classified into cellulose derivatives, clays, natural gums, and synthetic gums. In many cases, these excipients are used in combination.
  • water soluble hydrocolloids that can act as suspending agents in the formulation of pharmaceutical suspensions. They can be of natural, semi-synthetic or synthetic origin.
  • Non-limiting examples of suspending agents are Acacia. Agar, Alginic acid. Carbomer. Carmellose sodium. Dextrin, Gelatin. Veegum or Gel white. Gellan gum, Sodium alginate. Methylcellulose, Hydroxyethyl cellulose, Hydroxypropyl cellulose. Hydroxypropylmethyl cellulose, Hydroxypropyl starch, Hypromellose. Maltodextrin.
  • Methylcellulose Modified starch. Pectin, Poloxamer, Polycarbophil, Polyethylene glycol. Polyvinyl acetate, Poly (vinyl alcohol), Potassium alginate. Polyvinyl pyrrolidone. Pregelatinized starch. Propylene glycol alginate. Sodium alginate. Carboxymethyl cellulose or an alkali metal salt thereof, Microcrysalline cellulose, gum Arabic, Karaya gum, Sterculia gum. Tragacanth, Xanthangumn, Bentonite, Carageenan. Guar gum. Colloidal silicon dioxide and the like or any combinations thereof.
  • the suspending agent is present in the liquid dosage forms of the present invention at a concentration which helps to achieve desired dissolution profile of the suspension dosage forms of the present invention.
  • preservatives are bacteriostatic rather than bacteriocidal, and consists of both acid and nonacid types.
  • acidic types are phenol, chlorocresol, 9-phenyl phenol, alkyl esters of para-hydroxybenzoic acid, benzoic acid, boric acid, and sorbic acid, and their respective salts. Therefore, the pH of solution, and the pKa of the preservative need to be carefully evaluated prior to selecting a preservative for a formulation.
  • Neutral preservatives include chlorobutanol, benzyl alcohol, and beta-phenylethyl alcohol. Under alkaline conditions, it is generally regarded that microbial growth is insignificant and at these pH values, the need for a preservative is not generally recommended.
  • preservatives listed in the FDA inactive ingredient guide for liquid dosage forms are not recommended for use in oral liquids and hence the choice of an acceptable preservative for an oral liquid formulation is limited.
  • solubility of many preservatives in aqueous system may not be high enough for effective antimicrobial activity.
  • bacteriostatic agents like para hydroxyl hbenzoic acids can partition between organic and aqueous phases in a heterogenous liquid formulations in such a way that their activity is significantly reduced.
  • Non-limiting examples of preservatives are Alcohol, Ethanol, Chlorobutanol, Phenoxyethanol, Potassium benzoate, Benzyl alcohol. Benzoic acid, Potassium sorbate.
  • Sorbic acid Benzalkonium chloride, Benzethonium chloride, Cetrimonium bromide, Cetylpyridinium chloride. Bronopol, Chlorbutol, Chlorocresol, Cresol Butylparaben, Methylparaben. Propylparaben. Ethylparaben. Phenol, Thymol. Phenylethanol. Sodium benzoate, Antimicrobial solvents like Propylene glycol. Glycerin. Chloroform and the like or any combinations thereof. In addition, some formulation ingredients like nonionic surfactants, quaternary ammonium compounds, gelatin, ferric salts, calcium salts and salts of heavy metals, including silver, lead, and mercury prevent microbial growth.
  • the liquid dosage forms of the present invention comprise one or more than one preservatives. In some of the further non-limiting aspects, the liquid dosage forms of the present invention comprise combination of two preservatives. In some of the further non-limiting aspects of the present invention, the preservatives are present in the liquid dosage forms of the present invention at a concentration which helps to prevent microbial growth in the liquid dosage forms when stored for prolonged time under storage conditions.
  • Antioxidants can be compounds that can reduce a drug that has been oxidized, or compounds that are more readily oxidized than the agents they are to protect (oxygen scavengers). Many of the lipid-soluble antioxidants act as scavengers. Antioxidants can also act as chain terminators, reacting with free radicals in solution to stop the free-radical propagation cycle. Mixtures of chelating agents and antioxidants are often used because there appears to be a synergistic effect. This occurs because many of the agents act at differing steps in the oxidative process.
  • rancidity refers to many typical off-flavors that result from autoxidation of unsaturated fatty acids that are present in oils and fats, and it affects many oils and fats.
  • the distinct rancid odour may result from short-chain, volatile monomers resulting from the cleavage of the longer chain, less volatile oils and fats.
  • Non-limiting examples of anti-oxidants are ⁇ -Tocopherol acetate. Ascorbic acid. Erythorbic acid. Butylated hydroxytoluene (BHT), d- ⁇ -Tocopherol natural. Monothioglycerol, Sodium bisulfite, Sodium sulfite. Sodium metabisulfite. Potassium metabisulfite, Acetone sodium bisulfite. Ascorbyl palmitate, Cysteine, d- ⁇ -tocopherol synthetic. Nordihydroguaiaretic acid, Sodium formaldehyde sulfoxylate, Sodium thiosulfate. Acetylcysteine, Ascorbyl palmitate, Butylated hydroxyanisole (BHA), Cysteine hydrochloride, Dithiothreitol, Propyl gallate, Thiourea and the like or any combinations thereof.
  • BHT Butylated hydroxytoluene
  • Monothioglycerol Sodium bis
  • bulking agents also known as auxiliary suspending agents are used.
  • Non-limiting examples of bulking agents are Calcium carbonate. Calcium hydroxide. Cellulose, Crospovidone, Dibasic calcium phosphate, Magnesium carbonate. Magnesium hydroxide, Microcrystalline cellulose, Silica (silicon dioxide), Titanium dioxide and the like or any combinations thereof.
  • Surfactant is a general name for materials that possess surface activity; in solution they tend to orient at the surface of the liquid.
  • Surfactants are amphiphilic molecules, i.e. part of the molecule is hydrophilic, and part is lipophilic. This combination of the two opposite affinities in the same molecule causes them to orient to the interface and thereby reduce the interfacial tension between the continuous and disperse phases, such as in emulsions and suspensions.
  • Ionic surfactants work primarily through electrostatic forces
  • non-ionic surfactants work primarily through steric forces.
  • Non-limiting examples of surfactants are Sodium lauryl sulfate.
  • Anti-foaming agents may be used in the preparation of the liquid pharmaceutical compositions of the present invention to lower the surface tension and cohesive binding of liquid phase.
  • Non-limiting examples of anti-foaming agents are simnethicone, organic phosphates, alcohols, paraffin oils, stearates, glycols and the like or any combinations thereof.
  • Chelating agents also known as sequestrants, are molecules that have the ability to form stable complexes with metal ions, particularly di-valent and tri-valent metal ions including trace metals and heavy metals. These metal ions are often implicated in API degradation by acting as catalysts, e.g. Mg 2+ will catalyze both ester hydrolysis and the Maillard interaction between primary or secondary amines and reducing sugars. Oxidative degradation is also often catalyzed by heavy metals. In addition, certain trace metals are required for microbial growth, and chelation (sequestration) to form complexes can help prevent microbial growth and spoilage, and thus allow lower levels of microbiocidal agents to be used. Non-limiting examples of chelating agents are Calcium disodium edetate, Disodium edetate, Edetic acid (also known as ethylenediaminetetraacetic acid/EDTA), Citric acid and the like or any combinations thereof.
  • Palatability of oral medicines is an important factor in compliance. There are several components to palatability including flavor, mouth-feel and sweetness. Most patients prefer medicines that are not too bitter but may be slightly “tart” (acidic). Most APIs are bitter. However, for bitterness to develop, the drug must be sufficiently soluble to interact with taste receptors on the tongue. For insoluble APIs in the form of suspensions, components of the suspension are also bitter, e.g. preservatives, or very salty, e.g. buffer systems. However, a slight saltiness and a slight bitterness are desirable for palatability.
  • sweetening agents are sugar alcohols (also known as polyhydric alcohols, polyols and hydrogenated sugars).
  • sugar alcohols also known as polyhydric alcohols, polyols and hydrogenated sugars.
  • ionic and have the potential to interact with other components of the suspension Some sweetening agents are more stable than others in aqueous solution. These will be important factors in the final selection of the sweetening agent.
  • Non-limiting examples of sweetening agents are Glucose, Sucralose, Trehalose, Fructose, Xylose, Dextrose, Galactose, Tagatose, Maltose, Sucrose, Glycerol, Dulcitol, Mannitol, Lactitol, Sorbitol, Xylitol, Saccharine or the corresponding sodium, potassium or calcium salt, Cyclamate or the corresponding sodium or calcium salt. Aspartame, or Acesulfame or the potassium salt thereof. Dulcin or Ammonium glycyrrhizinate, Alitame, Inulin, Isomalt, Ncohespcridin dihydrochalcone. Thaumatin and the like or any combinations thereof.
  • Flavors are used to improve the palatability of oral medicines.
  • One problem that can arise with oral suspensions is that the suspension may produce a “cloying” sensation in the mouth. While this is not the same as a bitter taste, it can nevertheless cause problems for the patient and affect compliance. This can be a particular problem with high levels of inorganic components. Flavors can help reduce this “cloying” taste and thereby improve palatability, and ultimately patient compliance.
  • flavor development and compounding is a specialist discipline. When deciding on which particular flavor is appropriate, the flavor specialist would benefit from knowledge of the other likely components in the suspension, just as the formulation scientist would benefit from knowledge of the components of the flavor.
  • Flavors can adsorb onto finely divided solids, thus reducing their effectiveness. They can also be absorbed by packaging. Flavor preferences vary with age, but the citrus flavors appear generally acceptable to most age groups.
  • Non-limiting examples of flavoring agents are synthetic flavor oils and flavoring aromatics and/or natural oils, extracts from plants leaves, flowers, fruits, and so forth and the like or any combinations thereof. These may include cinnamon oil, oil of wintergreen, peppermint oils, clove oil, bay oil, anise oil, eucalyptus, thyme oil, cedar leaf oil, oil of nutmeg, oil of sage, oil of bitter almonds, and cassia oil and the like or any combinations thereof.
  • flavors are vanilla, citrus oil, including lemon, orange, grape, lime and grapefruit, and fruit essences, including apple, banana, pear, peach, strawberry, raspberry, cherry, plum, pineapple, apricot, and so forth and the like or any combinations thereof.
  • Solid forms, such as spray dried forms of flavoring agents, may also be useful in the liquid dosage forms disclosed herein.
  • Coloring agents may also be used in the preparation of the liquid compositions of the present invention.
  • Pharmaceutical colors come in two types; soluble dyes and insoluble pigments.
  • soluble dyes are often used; however, pigments may also be used and would be part of the disperse phase.
  • Soluble dyes have the potential to interact with other components of the formulation.
  • the liquid dosage forms of the present invention are non-caking liquid dosage forms.
  • non-caking as used herein means that the liquid dosage form has a smooth consistency and doesn't contain any caking or clumping particles, by visual inspection.
  • the liquid dosage form in accordance with the present invention does not cake or clump during manufacture, i.e., when mixed with excipients. Nor does it cake or clump upon storage, even under relatively humid conditions, e.g., a relative humidity of about 75% or greater and when stored for relatively long periods such as about 6 months or longer and even at elevated temperatures of about 40° C. or greater, or at any combination of such humidity, time and temperature parameters.
  • Imatinib as used herein, unless the context requires otherwise, includes Imatinib, its pharmaceutically acceptable salts and chemical derivatives thereof such as polymorphs, solvates, hydrates, anhydrous forms, amorphous forms, prodrugs, chelates, and complexes. “Imatinib” as used herein also includes racemic or substantially pure forms.
  • Imatinib or pharmaceutically acceptable salt thereof used for the preparation of the liquid dosage forms of the present invention comprise particles of Imatinib or pharmaceutically acceptable salt thereof, wherein the d 90 of the particles of Imatinib or pharmaceutically acceptable salt thereof is less than about 1000 ⁇ m, or less than about 950 ⁇ m, or less than about 900 ⁇ m, or less than about 850 ⁇ m, or less than about 800 ⁇ m, or less than about 750 ⁇ m, or less than about 700 ⁇ m, or less than about 650 ⁇ m, or less than about 600 ⁇ m, or less than about 550 ⁇ m, or less than about 500 ⁇ m, or less than about 450 ⁇ m, or less than about 400 ⁇ m, or less than about 350 ⁇ m, or less than about 300 ⁇ m, or less than about 250 ⁇ m, or less than about 200 ⁇ m, or less than about 150 ⁇ m, or less than about 100 ⁇ m, or less than about 90 ⁇ m, or
  • the liquid dosage forms of the present invention comprise particles of Imatinib or pharmaceutically acceptable salt thereof, wherein the d 90 of the particles of Imatinib or pharmaceutically acceptable salt thereof is less than about 1000 ⁇ m, or less than about 950 ⁇ m, or less than about 900 ⁇ m, or less than about 850 ⁇ m, or less than about 800 ⁇ m, or less than about 750 ⁇ m, or less than about 700 ⁇ m, or less than about 650 ⁇ m, or less than about 600 ⁇ m, or less than about 550 ⁇ m, or less than about 500 ⁇ m, or less than about 450 ⁇ m, or less than about 400 ⁇ m, or less than about 350 ⁇ m, or less than about 300 ⁇ m, or less than about 250 ⁇ m, or less than about 200 ⁇ m, or less than about 150 ⁇ m, or less than about 100 ⁇ m, or less than about 90 ⁇ m, or less than about 80 ⁇ m, or less than about 70 ⁇ m
  • general formula of the liquid dosage forms according to the present invention may be provided as follows.
  • liquid dosage forms of the present invention may be prepared using suitable excipients or additives in any suitable amount.
  • the present invention provides process for the preparation of the liquid dosage forms of Imatinib or pharmaceutically acceptable salt thereof.
  • the liquid dosage forms of the present invention may also be prepared using processes generally known to those skilled in the art.
  • the processes for the preparation of liquid dosage forms of the present invention may vary depending upon the final dosage form. e.g. solution, suspension, etc.
  • the processes for the preparation of the liquid dosage forms of the present invention may comprise multiple steps. Such steps may include sequential addition of suitable excipients/additives. Such steps may also include physical processes for example mixing, stirring, agitation etc.
  • the liquid dosage forms of the present invention are suitable for administration to a subject to treat or prevent a disease or condition.
  • the subject is a mammal. More preferably, the mammal is a human.
  • the disease or condition is a disease or condition that is treatable by the administration of Imatinib or pharmaceutically acceptable salt thereof.
  • the present invention is directed to the method for the treatment of a disease or disorder or medical condition that can be treated by inhibiting protein-tyrosine kinase comprising administering to a patient, such as human, an effective dosage amount of a liquid dosage form comprising Imatinib or pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable excipients or additives as disclosed and described herein.
  • the present invention is directed to the method for the treatment of a tumor disease or cancer disease, including without limitation, colon cancer, thyroid cancer, ovarian cancer, breast cancer, lung cancer (non-small cell lung cancer), head and neck cancer, uterine cancer, non-hodgkin lymphoma, blood cancer, skin cancer, prostate cancer, kidney cancer, rectal cancer, peritoneal cavity cancer, brain cancer, gastric cancer, metastatic cancer, colorectal cancer, pancreatic cancer, endometrial cancer, stomach cancer, gastrointestinal cancer, bladder cancer, and the like comprising administering to a patient, such as human, an effective dosage amount of a liquid dosage form comprising Imatinib or pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable excipients or additives as disclosed and described herein.
  • a tumor disease or cancer disease including without limitation, colon cancer, thyroid cancer, ovarian cancer, breast cancer, lung cancer (non-small cell lung cancer), head and neck cancer, uterine cancer, non-hodgkin lymphoma, blood cancer
  • the present invention is directed to the method for the treatment of at least one disease or condition selected from the group comprising of chronic myeloid leukemia, acute lymphoblastic leukemia, myelodysplastic diseases, myeloproliferative diseases, aggressive systemic mastocytosis, hypereosinophilic syndrome and/or chronic cosinophilic leukemia, unresectable, recurrent and/or metastatic dermatofibrosarcomna protuberans, and unresectable and/or metastatic malignant gastrointestinal stromal tumors comprising administering to a patient, such as human, an effective dosage amount of a liquid dosage form comprising Imatinib or pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable excipicnts or additives as disclosed and described herein.
  • a patient such as human
  • an effective dosage amount of a liquid dosage form comprising Imatinib or pharmaceutically acceptable salt thereof and one or more pharmaceutically acceptable excipicnts or additives as disclosed and described herein.
  • Effective dosage amount as used herein with respect to, for example Imatinib liquid dosage forms shall mean that dosage that provides the specific pharmacological response for which Imatinib administered in a significant number of subjects in need of such treatment. It is emphasized that “effective dosage amount”, administered to a particular subject in a particular instance will not always be effective in treating the diseases described herein, even though such dosage is deemed a “effective dosage amount” by those skilled in the art.
  • the present invention is directed to use liquid dosage forms of the present invention for the treatment of a disease or disorder that can be treated by inhibiting protein-tyrosine kinase.
  • the present invention is directed to use liquid dosage forms of the present invention for the treatment of a tumor disease or a cancer disease, including without limitation, colon cancer, thyroid cancer, ovarian cancer, breast cancer, lung cancer (non-small cell lung cancer), head and neck cancer, uterine cancer, non-hodgkin lymphoma, blood cancer, skin cancer, prostate cancer, kidney cancer, rectal cancer, peritoneal cavity cancer, brain cancer, gastric cancer, metastatic cancer, colorectal cancer, pancreatic cancer, endometrial cancer, stomach cancer, gastrointestinal cancer, bladder cancer, and the like.
  • the present invention is directed to use liquid dosage forms of the present invention for the treatment of at least one diseases or condition selected from the group comprising of chronic myeloid leukemia, acute lymphoblastic leukemia, myelodysplastic diseases, myeloproliferative diseases, aggressive systemic mastocytosis, hypereosinophilic syndrome and/or chronic eosinophilic leukemia, unresectable, recurrent and/or metastatic dermatofibrosarcoma protuberans, and unresectable and/or metastatic malignant gastrointestinal stromal tumors.
  • diseases or condition selected from the group comprising of chronic myeloid leukemia, acute lymphoblastic leukemia, myelodysplastic diseases, myeloproliferative diseases, aggressive systemic mastocytosis, hypereosinophilic syndrome and/or chronic eosinophilic leukemia, unresectable, recurrent and/or metastatic dermatofibrosarcoma protuberans, and unresectable and/
  • the liquid dosage forms of the present invention are proposed to have unexpectedly dramatic dissolution profiles. Rapid dissolution of an administered active agent is preferable, as faster dissolution generally leads to greater bioavailability and faster onset of action. To improve the dissolution profile and bioavailability of Imatinib it would be useful to increase Imatinib's dissolution so that it could attain a level dose to 100% dissolution of the drug substance.
  • the liquid dosage forms of the present invention comprising Imatinib or a pharmaceutically acceptable salt thereof, exhibit improved or comparable pharmacokinetic profiles as compared to known Imatinib compositions, e.g. Gleevec®.
  • the Cmax and/or AUC of the liquid dosage forms of Imatinib of the present invention can be greater than or substantially equal to the Cmax and/or AUC for known Imatinib compositions administered at the same dose.
  • the Tmax of the liquid dosage forms of Imatinib of the present invention can be lower than or substantially equal to that obtained for a known Imatinib compositions, administered at the same dose.
  • liquid dosage forms of Imatinib of the invention may result in minimal different absorption levels when administered under fed as compared to fasting conditions.
  • a liquid dosage form comprising Imatinib or pharmaceutically acceptable salt thereof exhibits in comparative pharmacokinetic testing with an Imatinib marketed or known formulation, administered at the same dose, a Tmax not greater than about 90%, not greater than about 80%, not greater than about 70%, not greater than about 60%, not greater than about 50%, not greater than about 30%, not greater than about 25%, not greater than about 20%, not greater than about 15%, not greater than about 10%, or not greater than about 5% of the Tmax exhibited by the marketed or known Imatinib formulation.
  • the liquid dosage form comprising Imatinib or pharmaceutically acceptable salt thereof exhibits in comparative pharmacokinetic testing with an Imatinib marketed or known formulation, administered at the same dose, a Cmax which is at least about 50%, at least about 100%, or at least about 150%, greater than the Cmax exhibited by the marketed or known Imatinib formulation.
  • the liquid dosage form comprising Imatinib or pharmaceutically acceptable salt thereof exhibits in comparative pharmacokinetic testing with an Imatinib marketed or known formulation, administered at the same dose, a Cmax which is in the range between about 70% and about 150%.
  • the liquid dosage form comprising Imatinib or pharmaceutically acceptable salt thereof exhibits in comparative pharmacokinetic testing with an Imatinib marketed or known formulation, administered at the same dose, an AUC which is at least about 25%, at least about 50%, at least about 75%, at least about 100%, at least about 125%, at least about 150%, at least about 175%, or at least about 200%, greater than the AUC exhibited by the marketed or known Imatinib formulation.
  • the liquid dosage form comprising Imatinib or pharmaceutically acceptable salt thereof exhibits in comparative pharmacokinetic testing with an Imatinib marketed or known formulation, administered at the same dose, an AUC which is in the range between about 80% and about 125%.
  • the Tmax of Imatinib or salt thereof, when assayed in the plasma of the mammalian subject is less than about 6 to about 8 hours. In other aspects of the invention, the Tmax of Imatinib or salt thereof is less than about 6 hours, less than about 5 hours, less than about 4 hours, less than about 3 hours, less than about 2 hours, less than about 1 hour, or less than about 30 minutes after administration.
  • liquid dosage forms of Imatinib of the present invention exhibit improved or comparable bioavailability as compared to known Imatinib compositions, e.g. Gleevec®.
  • the present invention is further exemplified by the following non-limiting examples.
  • liquid dosage forms of the present invention are explained in more detail with reference to the following examples. These examples are provided by way of illustration only and should not be construed as to limit the scope or spirit of the claims in any manner.
  • Example 1 Initial 3.48 0.02 0.57 Do not freeze but dark 3 M 40° C./25% RH 3.47 0.02 0.57 brownish colour 3 M 25° C./40% RH 3.48 0.02 0.57 observed
  • Example 2 Initial 4.31 0.03 0.66 Product was 3 M 40° C./25% RH 4.31 0.03 0.69 precipitated out 3 M 25° C./40% RH 4.27 0.03 0.66
  • Example 4 Irnatinib solution dosage form (with glycerine)
  • Acesulfame potassium 1.0 Citric acid monohydrate Q.S. to desired pH Strawberry flavour 0.1
  • Irnatinib suspension dosage form Ingredients Quantity (mg/mL) Irnatinib or salt thereof 10-200 Ethanol (absolute) 0.001-0.5 (mL) Butylated hydroxyl toluene (BHT) 0.01-1.0 Sucralose 0.1-25 Colloidal silicon dioxide 1-50 Flavor 0.1-25 Caprylocaproyl macrogo 8 glycerides 1-200 Medium chain triglyceride Q.S. to 1 mL
  • Example 7 Bio-Equivalency Study of the Liquid Dosage Forms of the Present Invention Prepared According to Example 4
  • the liquid dosage forms prepared according to Example 4 of the present invention were tested for its bio-equivalence against the reference product, i.e. Gleevec® marketed tablets.
  • the results of the bio-equivalence study shows that the liquid dosage forms of the present invention have improved and/or comparable pharmacokinetic profile and/or bioavailability when compared against the known Imatinib formulations. The results are summarized in the table below.
  • liquid dosage form comprising only glycerin does not freeze after gone through freeze-thaw cycles whereas the liquid dosage forms comprising (i) mixture of glycerin and polyethylene glycol and (ii) only polyethylene glycol gets freezed after gone through freeze-thaw cycle (for further data see Table-10 below).
  • glycerin is the preferred solvent/co-solvent/solubilizer according to the present invention and at least 25% or more glycerin is required to achieve optimum results.
  • the inventors of the present invention had prepared two formulations as Test Formulation-1 and Test Formulation-2 according to the present invention containing 30% glycerin and 20% glycerin respectively.
  • the inventors have compared these two test formulations with reference formulation prepared according to WO 2014041551. From above table it can be seen that both the test formulations have total impurities less than the reference formulation.
  • liquid dosage forms of Imatinib prepared according to the present invention are suitable for use in the industry.

Landscapes

  • Health & Medical Sciences (AREA)
  • Chemical & Material Sciences (AREA)
  • Life Sciences & Earth Sciences (AREA)
  • Animal Behavior & Ethology (AREA)
  • Public Health (AREA)
  • Veterinary Medicine (AREA)
  • Medicinal Chemistry (AREA)
  • Pharmacology & Pharmacy (AREA)
  • Epidemiology (AREA)
  • General Health & Medical Sciences (AREA)
  • Engineering & Computer Science (AREA)
  • Chemical Kinetics & Catalysis (AREA)
  • Oil, Petroleum & Natural Gas (AREA)
  • General Chemical & Material Sciences (AREA)
  • Inorganic Chemistry (AREA)
  • Molecular Biology (AREA)
  • Biochemistry (AREA)
  • Proteomics, Peptides & Aminoacids (AREA)
  • Dispersion Chemistry (AREA)
  • Nutrition Science (AREA)
  • Physiology (AREA)
  • Medicinal Preparation (AREA)
  • Pharmaceuticals Containing Other Organic And Inorganic Compounds (AREA)
US16/633,048 2017-07-26 2018-07-26 Liquid dosage forms of imatinib Abandoned US20200206223A1 (en)

Applications Claiming Priority (5)

Application Number Priority Date Filing Date Title
IN201721026519 2017-07-26
IN201721026519 2017-07-26
IN201823010403 2018-03-21
IN201823010403 2018-03-21
PCT/IB2018/055583 WO2019021229A1 (fr) 2017-07-26 2018-07-26 Formes galéniques liquides d'imatinib

Publications (1)

Publication Number Publication Date
US20200206223A1 true US20200206223A1 (en) 2020-07-02

Family

ID=63517936

Family Applications (2)

Application Number Title Priority Date Filing Date
US16/633,048 Abandoned US20200206223A1 (en) 2017-07-26 2018-07-26 Liquid dosage forms of imatinib
US16/634,475 Active 2040-04-27 US11957681B2 (en) 2017-07-26 2018-07-26 Liquid dosage forms of Imatinib

Family Applications After (1)

Application Number Title Priority Date Filing Date
US16/634,475 Active 2040-04-27 US11957681B2 (en) 2017-07-26 2018-07-26 Liquid dosage forms of Imatinib

Country Status (7)

Country Link
US (2) US20200206223A1 (fr)
EP (1) EP3658190A1 (fr)
CN (1) CN111278466A (fr)
AU (1) AU2018306149B2 (fr)
RU (1) RU2020108342A (fr)
SG (1) SG11202001720RA (fr)
WO (1) WO2019021229A1 (fr)

Families Citing this family (6)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2021009686A1 (fr) 2019-07-15 2021-01-21 Intas Pharmaceuticals Ltd. Composition pharmaceutique d'imatinib
EP4171512A1 (fr) * 2020-06-24 2023-05-03 Novartis AG Formulations liquides orales de ruxolitinib
RU2755094C1 (ru) * 2020-11-12 2021-09-13 Общество С Ограниченной Ответственностью "Научно-Производственная Компания "Фармасофт" Лекарственная форма для инъекций в виде раствора и стабилизатор для его приготовления
CA3198046A1 (fr) * 2020-11-17 2022-05-27 David W. Osborne Compositions et procedes pour l'administration de medicaments par voie intradermique profonde
CN114159387B (zh) * 2021-12-10 2022-08-19 江苏汉晨药业有限公司 一种氢溴酸右美沙芬口服溶液
WO2024063770A1 (fr) * 2022-09-21 2024-03-28 Kadmon Corporation, Llc Formulation liquide à base de belumosudil

Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014041551A1 (fr) * 2012-09-14 2014-03-20 Natco Pharma Limited Formulation comprenant de l'imatinib sous la forme d'une solution orale
US20150337034A1 (en) * 2013-05-06 2015-11-26 Scholar Rock, Inc. Compositions and methods for growth factor modulation

Family Cites Families (8)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2006035204A2 (fr) 2004-09-27 2006-04-06 Astrazeneca Ab Polytherapie
US7767688B2 (en) 2005-06-03 2010-08-03 Novartis Ag Combination of pyrimidylaminobenzamide compounds and imatinib for treating gastrointestinal stromal tumours
EA015102B1 (ru) 2005-06-03 2011-06-30 Элан Фарма Интернэшнл Лтд. Препараты наночастиц мезилата иматиниба
EP2009008A1 (fr) 2006-10-26 2008-12-31 Sicor, Inc. Base d'imatinib, et mesylate d'imatinib et son procédé de préparation
CN101641345A (zh) * 2006-10-26 2010-02-03 西科尔公司 晶体和无定形伊马替尼碱、甲磺酸伊马替尼及其制备方法
US20100087444A1 (en) 2007-03-12 2010-04-08 Dr. Reddy's Laboratories Ltd. Imatinib mesylate
CA2856692C (fr) 2011-11-24 2016-06-28 Imuneks Farma Ilac Sanayi Ve Ticaret A.S. Formes posologiques solides a base d'imatinib reconstituees juste avant l'utilisation
EP3019159A4 (fr) 2013-07-09 2017-01-18 Shilpa Medicare Limited Compositions pharmaceutiques orales comprenant du mésylate d'imatinib

Patent Citations (2)

* Cited by examiner, † Cited by third party
Publication number Priority date Publication date Assignee Title
WO2014041551A1 (fr) * 2012-09-14 2014-03-20 Natco Pharma Limited Formulation comprenant de l'imatinib sous la forme d'une solution orale
US20150337034A1 (en) * 2013-05-06 2015-11-26 Scholar Rock, Inc. Compositions and methods for growth factor modulation

Also Published As

Publication number Publication date
SG11202001720RA (en) 2020-03-30
EP3658190A1 (fr) 2020-06-03
CN111278466A (zh) 2020-06-12
US11957681B2 (en) 2024-04-16
WO2019021229A1 (fr) 2019-01-31
AU2018306149B2 (en) 2021-09-16
RU2020108342A (ru) 2021-08-26
US20220160709A1 (en) 2022-05-26
AU2018306149A1 (en) 2020-03-12
RU2020108342A3 (fr) 2021-08-26

Similar Documents

Publication Publication Date Title
US11957681B2 (en) Liquid dosage forms of Imatinib
US11759468B2 (en) Liquid oral formulations for sildenafil
US8263652B2 (en) Stabilized pediatric suspension of carisbamate
WO2019186515A1 (fr) Compositions pharmaceutiques liquides de médicaments anti-épileptiques
US10959985B1 (en) Pharmaceutical compositions including carvedilol and methods of using the same
JP6410814B2 (ja) フェキソフェナジンを含む経口投与用液体医薬組成物
US20210030671A1 (en) Liquid dosage forms of cinacalcet or salt thereof
US20240216373A1 (en) Liquid dosage forms of imatinib
WO2019162756A2 (fr) Compositions pharmaceutiques liquides de médicaments anticancéreux

Legal Events

Date Code Title Description
AS Assignment

Owner name: FTF PHARMA PRIVATE LIMITED, INDIA

Free format text: ASSIGNMENT OF ASSIGNORS INTEREST;ASSIGNORS:MEHTA, SANDIP;PATEL, VIJAY;UMRETHIA, MANISH;AND OTHERS;REEL/FRAME:051666/0463

Effective date: 20200122

STPP Information on status: patent application and granting procedure in general

Free format text: APPLICATION DISPATCHED FROM PREEXAM, NOT YET DOCKETED

STPP Information on status: patent application and granting procedure in general

Free format text: DOCKETED NEW CASE - READY FOR EXAMINATION

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: NON FINAL ACTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: RESPONSE TO NON-FINAL OFFICE ACTION ENTERED AND FORWARDED TO EXAMINER

STPP Information on status: patent application and granting procedure in general

Free format text: FINAL REJECTION MAILED

STPP Information on status: patent application and granting procedure in general

Free format text: ADVISORY ACTION MAILED

STCB Information on status: application discontinuation

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