throbber
Docket No. PHAR.O5/PR
`
`APPLICATION DATA SHEET
`
`Publication Filing Type:
`Application Type:
`Title of Invention:
`
`New Provisional
`Provisional
`ORAL FORMULATIONS OF CYTIDINE ANALOGS
`
`Customer Number Attorney and Correspondence Address:
`
`25871
`
`INVENTORS:
`
`ETTER, Jeffrey B.
`1318 Deer Trail Rd.
`Boulder, CO 80302
`(US Citizen)
`
`ASSIGNEE:
`Pharmion Corporation
`2525 28th Street
`Boulder, Colorado 80301
`(Delaware Corporation)
`
`CELGENE 2123
`CELGENE 2123
`APOTEX v. CELGENE
`APOTEX v. CELGENE
`IPR2023-00512
`IPR2023-00512
`
`

`

`ORAL FORMULATIONSOF CYTIDINE ANALOGS
`
`Attorney Docket: PHARM.05/PR
`
`BACKGROUNDOF THE INVENTION
`
`[0001]
`
`Cellular proliferative disorders are responsible for numerousdiseases resulting in
`
`major morbidity and mortality and have been intensively investigated for decades. Cancer now is
`
`the second leading cause of death in the United States, and over 500,000 people die annually
`
`from this proliferative disorder.
`
`[0002]
`
`Nucleoside analogs have been used clinically for the treatment of viral infections
`
`and proliferative disorders for decades. Most of the nucleoside analog drugs are classified as
`
`antimetabolites. After they enter cells, nucleoside analogs are successively phosphorylated to
`
`nucleoside 5'-monophosphates, 5'-diphosphates, and 5'-triphosphates. In most cases, nucleoside
`
`triphosphates are the chemical entities that inhibit DNA or RNA synthesis, either through a
`
`competitive inhibition of polymerases or through incorporation of modified nucleotides into
`
`DNAor RNAsequences. Nucleosides may act also as their diphosphates.
`
`[0003]
`
`5-Azacytidine (also known as azacitidine and 4-amino-1-B-D-ribofuranosy]-1,3,5-
`
`triazin-2(1H)-one; Nation Service Center designation NSC-102816; CAS Registry Number 320-
`
`67-2) has undergone NCI-sponsoredtrials for the treatment of myelodysplastic syndromes
`
`(MDS). See Kornblith et al., J. Clin. Oncol. 20(10): 2441-2452 (2002) and Silvermanetal., J.
`
`Clin. Oncol. 20(10): 2429-2440 (2002). 5-Azacytidine may be defined as having a molecular
`
`formula of CgH,2N.Os, a relative molecular weight of 244.21 and a structure of:
`N
`
`H S
`
`y
`
`N
`
`NLA
`
` [0004]
`
`[0005]
`
`Azacitidine (also referred to herein as 5-azacytidine herein) is a nucleoside
`
`analog, more specifically a cytidine analog. Azacitidine is an antagonistofits related natural
`
`

`

`nucleoside, cytidine. Azacitidine, as well as decitabine, 1.e., 5-aza-2’-deoxycytidine, are
`
`antagonists of decitabine’s related natural nucleoside, deoxycytidine. The only structural
`
`difference between the analogs andtheir related natural nucleosidesis the presence of nitrogen at
`
`position 5 of the cytosine ring in place of oxygen.
`
`[0006]
`
`Other membersofthe class of deoxycytidine and cytidine analogs include
`
`arabinosylcytosine (Cytarabine), 2'-deoxy-2',2'-difluorocytidine (Gemcitabine), 5-aza-2’-
`
`deoxycytidine (Decitabine), 2(1H) pyrimidine riboside (Zebularine), 2’ ,3’-dideoxy-5-fluoro-
`3° thiacytidine (Emtriva), N*-pentyloxycarbonyl-5’-deoxy-5-fluorocytidine (Capecitabine), 2’-
`cyclocytidine, arabinofuanosyl-5-azacytidine, dihydro-5-azacytidine, N*-octadecyl-cytarabine,
`
`elaidic acid cytarabine, and cytosine 1-$-D-arabinofuranoside (ara-C).
`
`[0007]
`
`In general, oral delivery of members of this class of compounds has proven
`
`difficult due to combinations of chemicalinstability, enzymatic instability, and/or poortissue
`
`permeability. For example, these compounds are knownto be acid labile and thus unstable in the
`
`acidic gastric environment. In the case of azacitidine, ara-C, decitabine and gemcitabine, an
`
`enzymethoughtto be responsible for a significant portion of drug metabolism is cytidine
`
`deaminase. Strategies to improvethe oral bioavailability of this drug class have included the use
`
`of prodrugs to modify chemical and enzymatic instability, and/or the use of enzymatic inhibitors.
`
`[0008]
`
`For example, DeSimoneet al describe the ability of azacitidine to inducefetal
`
`hemoglobin production in baboons when administered via the intravenous (IV), subcutaneous
`
`(SC), or perioral (PO) route. In the case of PO administration the author states that co-
`
`administration of THU (tetrahydrouridine) was necessary to achieve fetal hemoglobin induction,
`
`howeverno specific data is provided on the doses or responses observed without THU.
`
`Azacitidine doses ranged from 0.25 mg/kg/d to 8 mg/kg/d with co-administration of 20 mg/kg/d
`
`THU. Administration of THU alone was shownto result in a significant decrease in peripheral
`
`cytidine deaminaseactivity.
`
`[0009]
`
`Neil, et al describe the effect of THU on the pharmacokinetics and
`
`pharmacodynamics ofinter peritoneal (1.P.) and peri oral (P.O.) azacitidine when administered to
`
`leukemic mice. Pharmacokinetic parameters were determined using a bioassay that did not
`
`discriminate between azacitidine andits degradation and metabolism products. Inclusion of
`
`THU with IP administration had little effect on the clearance or degradation of azacitidine.
`
`Inclusion of THU with PO administration significantly increased both C,,.x and ty. In both
`
`

`

`acute and chronic IP dosing the inclusion of THU did not influence the pharmacodymamic
`
`effects of azacitidine except at the highest chronic dose which was toxic. Conversely, co-
`
`administration of THU with PO azacitidine resulted in increased efficacy at all doses except the
`
`highest chronic dose which wasagain toxic.
`
`[0010]
`Dunbar, et al describe the administration of azacitidine via IV and PO routes for
`increased production oftotal hemoglobin in a B°-thalassemic patient. Doses of 2 mg/kg/d IV
`
`resulted in a measurable increase to hemoglobin levels. Administration of 2 mg/d tid (three
`
`times daily) PO with co-administration of THU did notresult in increased hemoglobinlevels.
`
`[0011]
`
`Dover, et al describe administration of azacitidine via the SC and PO routes for
`
`increased production of total hemoglobin, fetal hemoglobin and F cells in sickle cell patients.
`
`Azacitidine oral bioavailability was assessed byclinical response only. Dover reportsthat oral
`
`doses of azacitidine (2 mg/kg/d) alone or THU (200 mg/d) alone did not result in increased F
`
`reticulocyte production. Howeveroral doses of 200 mg/d of THU were observedto result in a
`
`significant suppression of peripheral cytidine deaminase activity for several days post
`
`administration. When azacitidine was co-administered with THU goodclinical response was
`
`observed as determined by total hemoglobin, fetal hemoglobin and F cell levels. In fact
`
`comparable clinical response was observed with doses of 2 mg/kg/d SC without THU versus 0.2
`
`mg/kg/d PO with co-administration of 200 mg/d THU. Oral doses of azacitidine and THU were
`
`prepared by encapsulation at the clinical site. No information was provided with respect to
`
`excipients.
`
`[0012]
`
`Efforts to increase bioavailability of this class of compoundshave also been
`
`described in, for example, U.S. Patent Application Publication No. 2004/0162263 (Sands, etal.)
`
`In this publication, delivery of azacitidine in an enteric-coated formulation are disclosed such
`
`that the drugs are preferably absorbed in the upper regions of the small intestine, such as the
`
`jejunum.
`
`[0013]
`
`Despite these efforts, a need remains for more effective methods and
`
`compositions which increaseoral bioavailability of this class of compounds.
`
`BRIEF DESCRIPTION OF THE DRAWINGS
`
`[0014]
`
`Figure | represents a graph showing Absolute Mucosal to Serosal Permeability of
`
`Azacitidine in Human Intestinal Tissue with and without Enzymatic Inhibition.
`
`

`

`[0015]
`
`Figure 2 represents a graph showing Relative Mucosal to Serosal Permeability of
`
`Azacitidine in Human Intestinal Tissue with and without Enzymatic Inhibition with Respect to
`
`Atenolol.
`
`[0016]
`
`Figure 3 represents a graph showing Absolute Mucosal to Serosal Permeability of
`
`Azacitidine in Human Colonic Tissue with Various Concentrations of TPGS or Labrafil without
`
`Enzymatic Inhibition.
`
`[0017]
`
`Figure 4 represents a graph showing Relative Mucosal to Serosal Permeability of
`
`Azacitidine in Human Colonic Tissue with Various Concentrations of TPGS or Labrafil without
`
`Enzymatic Inhibition.
`
`SUMMARYOF THE INVENTION
`
`[0018]
`
`In a first embodiment, the present invention comprises a controlled release
`
`pharmaceutical composition for oral administration for enhanced systemic delivery of a cytidine
`
`analog comprising a therapeutically effective amountof a cytidine analog and a drug release
`
`controlling component whichis capable of providing release of the cytidine analog primarily in
`
`the large intestine. After ingestion by a patient, the cytidine analog is released primarily in the
`
`large intestine.
`
`[0019]
`
`In another embodiment, the present invention includes a methodfortreating a
`
`patient having a disease associated with abnormalcell proliferation. The method includesorally
`
`administering to the patient a controlled release pharmaceutical composition, comprising a
`
`therapeutically effective amountof a cytidine analog and a drug release controlling component
`
`whichis capable of providing release of the cytidine analog primarily in the large intestine.
`
`After ingestion by a patient the cytidine analog is released primarily in the large intestine.
`
`[0020]
`
`In another embodiment, the present invention includes a methodof increasing the
`
`bioavailability of a cytidine analog upon administration to a patient, comprising the following
`
`steps. First, provided is a controlled release pharmaceutical composition, comprising a
`
`therapeutically effective amountof a cytidine analog and a drug release controlling component
`
`capable of providing release of the cytidine analog primarily in the large intestine. Second, the
`
`patient ingests the composition, whereupon the composition contacts the biological fluids of the
`
`patient's body andincreases the bioavailability of the cytidine analog.
`
`

`

`[0021]
`
`In one embodiment, a condition to treat using the present inventionis a
`
`myelodysplastic syndrome. In one embodiment, the cytidine analog is azacitidine. In one
`
`embodiment, the drug release controlling componentis an enteric coating.
`
`DETAILED DESCRIPTION OF THE INVENTION
`
`[0022]
`
`The present invention is based on the surprising discovery that azacitidine and
`
`related compoundsare best absorbed in the lower gastrointestinal tract, 1.e., the large intestine
`
`(colon). Conventionally, it is expected that the upper gastrointestinal tract is the more desirable
`
`location for absorption, due to greater surface area,relatively greater liquidity, and the fact that
`
`typically the greater part of absorption of nutrients takes place therein. However, the inventors
`
`have foundthat in the case for cytidine analogs, absorption is greatest and most consistent
`
`between patients in colonic tissue. Accordingly, the present invention demonstrates the
`
`preparation of a solid oral dosage form of a cytidine analog, such as azacitidine, using common
`
`pharmaceutical excipients designed for delivering pharmaceutical compositionsto the large
`
`intestine and colon. The term "absorb", "absorption", "absorbed" and the like are used to
`
`indicate transfer of a cytidine analog across a relevanttissue, such as, for example, intestinal
`
`tissue. In some embodiments, absorbed cytidine analogs are taken up by the blood stream
`
`making the cytidine analog available at least partially systemically. In some embodiments,
`
`absorption occurs without substantive degradation(i.e., undesirable chemical modification of) of
`
`the cytidine analog.
`
`[0023]
`
`Furthermore, the inventors have demonstrated that inclusion of THU (taught by
`
`others as a requirementto facilitate bioavailability of this drug class) is not necessary to achieve
`
`useful oral bioavailability of cytidine analogs via delivery in the large intestine and colon.
`
`Accordingly, formulations of the present invention obviate the need to utilize enzymatic
`
`inhibitors such as THUin formulationsto increase bioavailability of cytidine analogs.
`
`Avoidance of enzymatic inhibitors is a desirable attribute for a therapeutic dosage form since
`
`such inclusion increases the formulation cost and complexity, and mayresult in instability, or
`
`undesirable, pharmacological, toxicological or other effects. Accordingly, oral delivery of
`
`azacitidine without inclusion of an enzymatic inhibitor is possible when the target tissue to which
`
`the drug is delivered is the colon.
`
`In the case of PO delivery of azacitidine to humans, data
`
`suggests that delivery to the upper GI tract may well benefit from enzymatic inhibition, however
`
`

`

`delivery to the colon does not require the inclusion of such an inhibitor. Targeting to the colon
`
`may be achieved with commercially available and pharmaceutically acceptable coatings such as,
`
`for example, enteric coatings.
`
`[0024]
`
`Furthermore, the inventors have demonstrated the preparation of solid oral dosage
`
`forms containing excipients and coatings which possess acceptable production andstability
`
`characteristics for use as a pharmaceutical dosage form.
`
`[0025]
`
`In one embodiment, the present invention includes a controlled release
`
`pharmaceutical composition for oral administration comprising a) a therapeutically effective
`
`amount of a cytidine analog and b) a drug release controlling componentfor providing the
`
`release of the cytidine analog primarily in the large intestine. The controlled release
`
`pharmaceutical compositions of the present invention will in one embodiment lack THU.
`
`[0026]
`
`In one embodiment, the cytidine analog useful in the present invention includes
`
`any moiety whichisstructurally related to cytidine or deoxycytidine and functionally mimics
`
`and/or antagonizesthe action of cytidine or deoxycytidine. These analogs mayalso be called
`
`cytidine derivatives herein. In one embodiment, cytidine analogs to use with the present
`
`invention include 5-aza-2’-deoxycytidine (decitabine), 5-azacytidine, 5-aza-2'-deoxy-2',2’-
`
`difluorocytidine, 5-aza-2'-deoxy-2'-fluorocytidine, 2'-deoxy-2',2'-difluorocytidine (also called
`
`gemcitabine), or cytosine 1-B-D-arabinofuranoside (also called ara-C), 2(1H) pyrimidine
`
`riboside (also called zebularine), 2’-cyclocytidine, arabinofuanosyl-5-azacytidine, dihydro-5-
`azacytidine, N*-octadecyl-cytarabine, andelaidic acid cytarabine. In one embodiment,is 5-
`
`azacytidine and 5-aza-2’-deoxycytidine The definition of cytidine analog used herein also
`
`includes mixtures of cytidine analogs.
`
`[0027]
`
`Cytidine analogs useful in the present invention may be manufactured by any
`
`methods knownin the art. In one embodiment, methods to manufacture include methods as
`
`disclosed in U.S. Serial No. 10/390,526 (U.S. Patent No. 7,038,038); U.S. Serial No. 10/390,578
`
`(U.S. Patent No. 6,887,855); U.S. Serial No. 11/052615 (U.S. Patent No. 7,078,518); U.S. Serial
`
`No. 10390530 (U.S. Patent No. 6,943,249); and U.S. Serial No. 10/823,394.
`
`[0028]
`
`In one embodiment, the amounts of a cytidine analog to use in methodsofthe
`
`present invention andin the oral formulations of the present invention include a therapeutically
`
`effective amount. Therapeutic indications are discussed more fully herein below. Precise
`
`amounts for therapeutically effective amounts of the cytidine analog in the pharmaceutical
`
`

`

`compositions of the present invention will vary depending on the age, weight, disease and
`
`condition of the patient. For example, pharmaceutical compositions may contain sufficient
`quantities of a cytidine analog to provide a daily dosage of about 150 mg/m7(based onpatient
`
`body surface area) or about 4 mg/kg (based on patient body weight) as single or divided (2-3)
`
`daily doses.
`
`[0029]
`
`The controlled release pharmaceutical compositions of the present invention
`
`include a drug release controlling component. The drug release controlling componentis
`
`adjusted such that the release of the cytidine analog occursprimarily in the large intestine. In
`
`one embodiment, at least about 95% of the cytidine analogis released in the large intestine, or at
`
`least about 90% of the cytidine analog is released in the large intestine. In other embodiments, at
`
`least about 80% of the cytidine analogis released in the large intestine, at least about 70% of the
`
`cytidine analogis releasedin the large intestine, at least about 60% of the cytidine analogis
`
`released in the large intestine, or at least about 50% of the cytidine analog is released in the large
`
`intestine. In other embodiments, the amount released in the intestines is at least about 40%, at
`
`least about 30%, or at least about 20% ofthe cytidine analog. The term "release" refers to the
`
`process wherebythe cytidine analog is made available for uptake by or transport across the
`
`epithelial cells that line the large intestine and is made available to the body.
`
`[0030]
`
`The pharmaceutical compositions of the present invention are intended for oral
`
`delivery. Oral delivery includes formats such as tablets, capsules, caplets, solutions, suspensions
`
`and/or syrups, and may also comprise a plurality of granules, beads, powdersor pellets that may
`
`or may not be encapsulated. Such formats may also be referred to as the "drug core" which
`
`contains the cytidine analog. Such dosage forms are prepared using conventional methods
`
`knownto those in the field of pharmaceutical formulation and are described in the pertinent
`
`texts, e.g., in REMINGTON: THE SCIENCE AND PRACTICE OF PHARMACY,20th Edition, Lippincott
`
`Williams & Wilkins, 2000). Tablets and capsules represent the most convenient oral dosage
`
`forms, in which case solid pharmaceutical carriers are employed. Tablets are used in one
`
`embodiment. Tablets may be manufactured using standard tablet processing procedures and
`
`equipment. One methodfor forming tablets is by direct compression of a powdered,crystalline
`
`or granular composition containing the cytidine analog, alone or in combination with one or
`
`more carriers, additives, or the like. As an alternative to direct compression,tablets can be
`
`prepared using wet-granulation or dry-granulation processes. Tablets may also be moldedrather
`
`

`

`than compressed, starting with a moist or otherwise tractable material; particularly, compression
`
`and granulation techniques are used in one embodiment.
`
`[0031]
`
`In another embodiment, capsules may be used. Soft gelatin capsules may be
`
`prepared in which capsules contain a mixture of the active ingredient and vegetable oil or non-
`
`aqueous, water miscible materials such as, for example, polyethylene glycol and the like. Hard
`
`gelatin capsules may contain granules of the active ingredient in combination with a solid,
`
`pulverulent carrier, such as, for example, lactose, saccharose, sorbitol, mannitol, potato starch,
`
`com starch, amylopectin, cellulose derivatives, or gelatin. A hard gelatin capsule shell can be
`
`prepared from a capsule composition comprising gelatin and a small amountofplasticizer such
`
`as glycerol. As an alternative to gelatin, the capsule shell may be made of a carbohydrate
`
`material. The capsule composition may additionally include colorings, flavorings and opacifiers
`
`as required.
`
`[0032]
`
`The cytidine analog in one embodimentis preparedas a tablet or capsule which
`
`represents a drug core comprising the pharmaceutical composition and the pharmaceutical
`
`composition is a controlled release pharmaceutical composition, and as such, comprisesat least
`
`one additional component comprising a drug release controlling component. Accordingly, the
`
`formulations of the present invention are formulated with a drug release controlling component
`
`in order to provide for release of the cytidine analog primarily in the large intestine. In one
`
`embodiment, minimal release of the cytidine analog occurs in the upper reachesof the
`
`gastrointestinaltract, e.g., the stomach and small intestine.
`
`[0033]
`
`The small intestine extends from the pylorus to the colic valve where it ends in
`
`the large intestine. The small intestine is about 6 meters long andis divisible into three portions:
`
`the duodenum,the jejunum, and the ileum. The small intestine is especially adapted for transport
`
`and absorption of nutrients and other molecules from ingested material, passing through the
`
`lining of the small intestine into the blood. The surface cells of the small intestine are highly
`
`specialized for digestion and absorption of nutrients. Almost all the body's nutrient absorption
`
`occurs in the small intestine, along its three sub-divisions: the duodenum, jejunum, and ileum.
`
`Sites for absorption of specific nutrients (eg: iron, vitamin.B12) are located in these divisions,
`
`but most absorption occurs in the jeyunum (middle section). Specialized cells contain digestive
`
`enzymes, carrier proteins and other secretions. Blood vessels transport nutrients away from the
`
`intestine to the liver in the first instance.
`
`

`

`[0034]
`
`Indigestible food passes into the large intestine. By the time ingested material
`
`leaves the small intestine, virtually all nutrient absorption will have occurred. The large intestine
`
`extends from the end of the ileum (distal ileum) to the anus. The large intestine is divided into
`
`the cecum, colon, rectum, and anal canal. The colonis divided into four parts: the ascending,
`
`transverse, descending, and sigmoid. The substantial release of the cytidine compoundof the
`
`present invention mayoccurin any portion of the large intestine. In one embodiment, release
`
`primarily occurs at the upper regionsof the large intestine, such as, for example,at the distal
`
`ileum, cecum, and/or the ascending colon.
`
`[0035]
`
`It is known that there are major variations in acidity in the gastrointestinal tract.
`
`The stomachis a region of high acidity (about pH 1 to 3). Specific glands and organs emptying
`
`into the small intestine raise the pH of the material leaving the stomach to approximately pH 6.0
`
`to 6.5. The large intestine and the colon are about pH 6.4 to 7.0. Thetransit time through the
`
`small intestine is approximately three hours. In contrast, the transit time throughthe large
`
`intestine is approximately 35 hours.
`
`[0036]
`
`Methods by which to formulate compositionsto target specific regions of the
`
`gastrointestinal tract are knownin the art, described in numerous publications, andall references
`
`specifically cited within the present documentare incorporated by reference herein. For
`
`example, release of drug in the gastrointestinal tract may be accomplished by choosing a drug
`
`release controlling component to work together with some physical, chemical or biochemical
`
`process in the gastrointestinal tract. A drug release controlling component may take advantage
`
`of processes and/or conditions within the gastrointestinal tract and in specific regions of the
`
`gastrointestinal tract such as, for example, osmotic pressure, hydrodynamic pressure, vapor
`
`pressure, mechanicalaction, hydration status, pH, bacterial flora, and enzymes. Specific U.S.
`
`Patents incorporated by reference herein include, among others, U.S. Pat. No. 3,952,741, U.S.
`
`Pat. No. 5,464,633, U.S. Pat. No. 5,474,784, U.S. Pat. No. 5,112,621.
`
`[0037]
`
`Optionally, pharmaceutical compositions of the present invention may further
`
`comprise a seal coating material that seals the drug to prevent decomposition due to exposure to
`
`moisture, such as hydroxylpropylmethylcellulose. Accordingly, the core of the pharmaceutical
`
`composition (containing the cytidine analog) mayfirst be sealed with the seal coating material
`
`and then coating with the drug release controlling componentto prevent decomposition of the
`
`cytidine analog by exposure to moisture. Seal coating materials include, in one embodiment,
`
`

`

`acetyltributyl citrate, acetyltriethyl citrate, calcium carbonate, carauba wax, cellulose acetate,
`
`cellulose acetate phthalate, cetyl alcohol, chitosan, ethylcellulose, fructose, gelatin, glycerin,
`
`glyceryl behenate, glyceryl palmitostearate, hydroxyethyl cellulose, hydroxyethylmethyl
`
`cellulose, hydroxypropyl cellulose, hypromellose, hypromellose phthalate, isomalt, latex
`
`particles, maltitol, maltodextrin, methylcellulose, microcrystalline wax, paraffin, poloxamer,
`
`polydextrose, polyethylene glycol, polyvinyl acetate phthalate, polyvinyl alcohol, povidone,
`
`shellac, shellac with stearic acid, sodium carboxymethylcellulose, sucrose, titanium oxide,
`
`tributyl citrate, triethyl citrate, vanillin, white wax, xylitol, yellow wax, and zein. Compositions
`
`of the present invention mayalso include film forming agents, which include, for example,
`
`ammonium alginate, calctum carbonate, chitosan, chlorpheniramine maleate, copovidone,
`
`dibutyl phthalate, dibutyl sebacate, diethyl phthalate, dimethyl phthalate, ethyl lactate,
`
`ethylcellulose, gelatin, hydroxyyethyl cellulose, hydroxypropyl cellulose, hypromellose,
`
`hypromellose acetate succinate, maltodextrin, polydextrose, polyethylene glycol, polyethylene
`
`oxide, polymethylacrylates, poly(methylvinyl ether/maleic anhydride), polyvinylacetate
`
`phthalate, triethyl citrate, and vanillin.
`
`[0038]
`
`In one embodiment, drug release controlling components include, for example,
`
`coatings, matrices, or physical changes. Coatings are used in one embodiment. Coatings
`
`include, for example, enteric coatings, time delay coatings, bacterially degradable coatings, and
`
`mixtures thereof. The pharmaceutical composition may comprise multiple coatings of either the
`
`sameor different types of coatings. In choosing an appropriate coating or mixture thereof, the
`
`formulations practitioner may consider a numberofvariables influencing the location in which a
`
`drug will become available in the gastrointestinal tract, e.g., the pH at which coatings dissolve;
`
`the time of dissolution (whichis influenced by thickness of the coatings and/or additional
`
`components in the coatings); time of transit through the gastrointestinal tract, and whether the
`
`coatings can be degraded bythe patent’s digestive enzymesor require enzymespresentonly in
`
`bacteria residing in the lowerintestine. As an example of a combination drug release controlling
`
`componentis, for example, an inner core with two polymeric layers. The outer layer, an enteric
`
`coating, may be chosen to dissolve at a pH level above 5. The inner layer, may be made up of
`
`hydroxypropylmethylcellulose to act as a time delay componentto delay drug release for a
`
`predetermined period. The thickness of the inner layer can be adjusted to determine the lag time.
`
`10
`
`

`

`[0039]
`
`Methods by whichskilled practitioners can assess where a drugis released in the
`
`gastrointestinal tract of either animal models or human volunteers are knownin the art, and
`
`include scintigraphic studies, testing in biorelevant medium whichsimulates the fluid in relevant
`
`portions of the gastrointestinal tract, among others.
`
`[0040]
`
`In one embodiment, a drug release controlling component mayinclude an enteric
`
`coating. The term "enteric coating" refers to a coating that allows a cytidine analog formulation
`
`to pass through the stomachsubstantially intact and subsequently disintegrate substantially in the
`
`intestines. In one embodiment, the disintegration occursin the large intestine.
`
`[0041]
`
`The coating of pH-sensitive (enteric) polymers to tablets, capsules and otheroral
`
`formulations of the present invention provided delayed release and protect the active drug from
`
`gastric fluid. In general, enteric coatings should be able to withstand the lower pH values of the
`
`stomach and small intestine and be able to disintegrate at the neutral or slightly alkaline pH of
`
`the large intestine. Enteric coatings are a well known class of compounds. Coating
`
`pharmaceutically active compositions with enteric coatings is well knownin theart to enable
`
`pharmaceutical compositions to bypass the stomach andits low acidity. Enteric coatings
`
`generally refer to a class of compoundsthat dissolve at or above a particular pH andinclude a
`
`number of pH-sensitive polymers. The pH dependent coating polymer may be selected from
`
`those enteric coatings knownto those skilled in the art. Such polymers may be one or moreof
`
`the group comprising hydroxypropylmethylcellulose phthalate, polyvinyl acetate phthalate
`
`(PVAP), hydroxypropylmethylcellulose acetate succinate (HPMCAS), alginate, carbomer,
`
`carboxymethyl cellulose, methacrylic acid copolymer (such as, for example, a cationic
`
`copolymerof dimethyl aminoethyl methacrylate and neutral methacrylic esters), polyvinyl
`
`acetate phthalate, cellulose acetate trimellitate, shellac, cellulose acetate phthalate (CAP), starch
`
`glycolate, polacrylin, methyl cellulose acetate phthalate, hydroxylmethylcellulose phthalate,
`
`hydroxymethylmethylcellulose acetate succinate, hydroxypropylcellulose acetate phthalate,
`
`cellulose acetate terephthalate, cellulose acetate isophthalate, and includes the various grades of
`
`each polymer such as HPMCAS-LF, HPMCAS-MEFand HPMCAS-HG,or mixturesthereof.
`
`Other enteric coatings suitable for the present invention include acetyltributyl citrate, carbomers,
`
`guar gum, hypromellose acetate succinate, hypromellose phthalate, polymethacrylates, tributyl
`
`citrate, triethyl citrate, white wax, and zein.
`
`11
`
`

`

`[0042]
`
`In one embodiment, the pH dependent coating is selected from the group
`
`consisting of methacrylic acid copolymers of varying threshold pH (suchas, but not limited to
`
`EUDRAGITS 100 (a cationic copolymer of dimethyl aminoethyl methacrylate and neutral
`
`methacrylic acid esters manufactured by Rohm Pharma GmbH of Darmstadt, Germany)).
`
`[0043]
`
`Multiple coatings of enteric polymers maybeutilized. In one embodiment, the
`
`first coating (closest to the core) is an enteric coating that will survive until the dosage form
`
`arrives at the large intestine/colon. To target the large intestine, in one embodimentan enteric
`
`coating comprises a series of methacrylic acid anionic copolymers known as EUDRAGITS. The
`
`EUDRAGITS filmsare colorless, transparent and brittle. In one embodiment, the enteric
`
`coating comprises EUDRAGIT S100. The EUDRAGITS coatings are insoluble in pure water,
`
`in buffer solutions below a pH of 6.0 andalso in natural and artificial gastric juices. They are
`
`slowly soluble in the region of the digestive tract where the juices are neutral to weakly alkaline
`
`(i.e., the large intestine and the colon) and in buffer solutions above a pH of 7.0. Mixtures of
`
`these various enteric polymers recited above, can be used in the present invention. Further, the
`
`use of plasticizers is included in one embodiment with the enteric polymer coatings useful
`
`herein.
`
`[0044]
`
`As knownin the art and discussed in sources such as Patel et al. "Colon Specific
`
`Delivery" Drug Delivery Technology (2006) Vol. 6 62-71, and Khanetal., J. Controlled Release
`
`1999; 58:215-222, the disintegration rates of enteric coated tablets are dependent on the polymer
`
`combination usedto coatthe tablets, the pH of the disintegration media, and the coating level of
`
`the tablets (i.e., thickness of the coating). The presence of plasticizer and the nature of the salts
`
`in the dissolution medium also influence the dissolution rate. A numberof specific formulations
`
`effective for release in the colon in human volunteers, using in vivo scintigraphic studies, is
`
`disclosed in Patel et al., and are incorporated by reference herein.
`
`[0045]
`
`The enteric coating may also be modified through the inclusion of an edible acid
`
`to retard or slow the dissolution of the coating in the intestines. Any edible acid may be used.
`
`Representative edible acids include acetic acid, benzoic acid, fumaric acid, sorbic acid, propionic
`
`acid, hydrochloric acid, citric acid, malic acid, tartaric acid, isocitric acid, oxalic acid, lactic acid,
`
`the phosphoric acids and mixtures thereof. One embodimentincludes fumaric acid and malic
`
`acids. The weight percent of the edible acid in the enteric coating solution (polymer, plasticizer,
`
`anti-tack agents, water and the like) can range from about 5 to about 40%, with 10 to 30%
`
`12
`
`

`

`present in one embodimentand 10 to 25% in another embodiment. Thoseskilled in the art will
`
`readily be able to determine the exact amountof edible acid to include in the coating solution,
`
`depending upon the pKaof the particular edible acid and the desired delay in dissolution of the
`
`enteric coating. After application of the enteric coating solution, as further described below, the
`
`percent of edible acid in the coating will range from about 10 to about 80 weight % of the
`
`coating; 20 to 60% in one embodiment; and 25-50% in another.
`
`[0046]
`
`Enteric coatings can be obtained from a number of manufacturers, such as, for
`
`example, Rohm Pharma GmbH of Darmstadt, Germany (EUDRAGIT). Particular blends of pH
`
`sensitive polymers and types can be selected by oneofskill in the art. As an example, the
`
`manufacturer of EUDRAGITpolymers teaches that the EUDRAGITgradesfor sustained release
`
`formulations are based on cop

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket