throbber
United States
`(19)
`a2) Patent Application Publication 0) Pub. No.: US 2008/0057086 Al
`Etter
`(43) Pub. Date:
`Mar.6, 2008
`
`
`US 20080057086A1
`
`(54) COLON-TARGETED ORAL FORMULATIONS
`OF CYTIDINE ANALOGS
`Inventor:
`Jeffrey B. Etter, Boulder, CO
`(US)
`
`(75)
`
`Publication Classification
`
`(51)
`
`Int. Cl.
`AGLK 9/00
`AG6IK 31/7068
`A6IP 43/00
`
`(2006.01)
`(2006.01)
`(2006.01)
`
`Correspondence Address:
`SWANSON & BRATSCHUN, L.L.C.
`8210 SOUTHPARK TERRACE
`LITTLETON, CO 80120
`
`(73) Assignee:
`
`PHARMION CORPORATION,
`Boulder, CO (US)
`
`11/849,958
`
`(21) Appl. No.:
`;
`Filed:
`
`(22)
`
`Sep. 4, 2007
`+
`as
`Related U.S. Application Data
`(60) Provisional application No. 60/824,320,filed on Sep.
`1, 2006.
`
`(52) U.S. Cheects ereneeiens 424/400; 514/49
`
`(57)
`
`ABSTRACT
`:
`.
`.
`.
`The present invention provides an oral formulation of a
`cytidine analog, including, 5-azacytidine, for delivery to the
`lower gastrointestinal tract, including, the large intestine;
`methods to treat diseases associated with abnormal cell
`proliferation by treatment with the oral formulations of the
`present invention; and methodsto increase the bioavailabil-
`ity of a cytidine analog upon administration to a patient by
`providing an oral formulation of the present invention.
`
`(M10°crn/sec}
`
`
`Pano
`
`feum
`
`Colon
`
`intestinal Segment
`
`CELGENE 2124
`APOTEX v. CELGENE
`IPR2023-00512
`
`

`

`Patent Application Publication Mar. 6, 2008 Sheet 1 of 6
`
`US 2008/0057086 Al
`
`
`
`re
`
`ob
`ho
`[L,
`
`Colon
`
`es
`
`aa 5
`
`& &
`ae
`“2
`8
`=
`
`aa2
`
`=5
`
`= 3a“
`
`
`
`Ea
`
`oO
`
`Pe
`
`16
`
`LO
`at
`Oy
`foospue OLPy
`
`od
`
`aa
`
`cae)
`
`

`

`Patent Application Publication Mar. 6, 2008 Sheet 2 of 6
`
`US 2008/0057086 Al
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`Colon
`
`lleum
`
`
`
`IntestinalSegment
`
`Jejunum
`
`
`
`
`
`
`
`Fig.2
`
`ao
`TT
`
`t+

`Tr oT
`x
`cr
`
`N
`rT
`2
`c
`
`FF
`
`DD
`oO
`
`O©
`oO
`
`TFT
`oO
`
`AN
`oO
`
`CO
`
`d |Ojous}y/eulpiioezy
`
`

`

`Patent Application Publication
`
`Mar. 6, 2008 Sheet 3 of 6
`
`US 2008/0057086 Al
`
`
`
`
`
`
` >4ou0gO
`GJOuNG
`
`
`
`
`
`
`
`
`
` ol
`
`%30'0e100%100'0%S0'°O%10'°0%100'0JOJUOD
`
`
`
`
`
`
`YEAQe]IWEsAQEIesqE]SDdLSDdLSDSdL
`
`
`
`€“SI
`
`yuayu0g
`
`(9eS/Wd ,OL x) 44q
`
`

`

`Patent Application Publication
`
`Mar. 6, 2008 Sheet 4 of 6
`
`US 2008/0057086 Al
`
`
`
`
`
`
`GJ0Uu0g&yJouogO
`
`
`
`
`
`%G0'0*lLOO%lO0OO %G0'0%10'0%t00'O=|O4JU0D
`
`
`
`IWEAGe]=JEIQE7]—IJEAQE"]SOdLSDOdLSOdL
`
`
`
`
`
`
`}Ua}U0D
`
`pSI
`
`ot
`
`one
`
`80
`
`dde
`
`9°0
`
`vO
`
`¢0
`
`d OjOUus}V7-oUIpIIoezy
`
`

`

`Patent Application Publication Mar. 6, 2008 Sheet 5 of 6
`
`US 2008/0057086 Al
`
`1000
`
`ae
`
`= 100
`6
`
`©5
`
`@ SC (75 mg/m?)
`
`HE Po 60 mg (101)
`
`Ae PO 80 mg (102)
`
`PO 80 mg(201)
`
`
`5 080mg (202)
`PO 80mg(202
`Cc
`oO
`
`10
`
`0
`
`1
`
`2
`
`3
`
`4
`
`5
`
`Time After Dosing (h)
`
`FIG. 5
`
`O®
`
`o £oS
`
`ON <
`
`€
`

`

`Patent Application Publication Mar. 6, 2008 Sheet 6 of 6
`
`US 2008/0057086 Al
`
`
`
`
`
`AzacitidineConcentration(ng/mL)
`
`400
`
`@ SC (75 mgim2)
`
`EPO 60 mg (101)
`
`~@ PO mean 80 mg
`
`0
`
`1
`
`2
`
`3
`
`4
`
`5
`
`Time After Dosing (h)
`
`FIG. 6
`
`

`

`US 2008/0057086 Al
`
`Mar.6, 2008
`
`COLON-TARGETED ORAL FORMULATIONS
`OF CYTIDINE ANALOGS
`
`RELATED APPLICATIONS
`
`[0001] This application is a non-provisional of U.S. Patent
`Application Ser. No. 60/824,320, filed Sep. 1, 2006, entitled
`“Oral Formulations of Cytidine Analogs”, which is incor-
`porated by reference herein in its entirety.
`
`BACKGROUND OF THE INVENTION
`
`[0002] Cellular proliferative disorders are responsible for
`numerousdiseases resulting in major morbidity and mortal-
`ity 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.
`[0003] Nucleoside analogs have been used clinically for
`the treatment of viral infections and proliferative disorders
`for decades. Most of the nucleoside analog drugs are clas-
`sified 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 incorpo-
`ration of modified nucleotides into DNA or RNA sequences.
`Nucleosides mayact also as their diphosphates.
`[0004]
`5-Azacytidine (also known as azacitidine and
`4-amino-1-(-D-ribofuranosyl-1 ,3,5-triazin-2(1H)-one;
`Nation Service Center designation NSC-102816; CAS Reg-
`istry 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 Silverman et al., J. Clin. Oncol. 20(10): 2429-2440
`(2002). 5-Azacytidine maybe defined as having a molecular
`formula of CgH,,N,O;, a relative molecular weight of
`244.21 and a structure of:
`
`
`
`[0005] Azacitidine (also referred to herein as 5-azacyti-
`dine herein) is a nucleoside analog, more specifically a
`cytidine analog. 5-azacytidine is an antagonistofits related
`natural nucleoside, cytidine. 5-azacytidine, as well as decit-
`abine,i.e., 5-aza-2'-deoxycytidine, are antagonists of decit-
`abine’s related natural nucleoside, deoxycytidine. The only
`structural difference between the analogs and their related
`natural nucleosides is the presence of nitrogen at position 5
`of the cytosine ring in place of oxygen.
`[0006] Other members of the class of deoxycytidine and
`cytidine analogs include arabinosylcytosine (Cytarabine),
`
`5-aza-2'-
`(Gemcitabine),
`2'-deoxy-2',2'-difluorocytidine
`deoxycytidine (Decitabine), 2(1H) pyrimidine riboside (Ze-
`bularine), 2',3'-dideoxy-5-fluoro-3'thiacytidine (Emtriva),
`N*-pentyloxycarbonyl-5'-deoxy-5-fluorocytidine (Capecit-
`abine),
`2'-cyclocytidine,
`arabinofuanosyl-5-azacytidine,
`dihydro-5-azacytidine, N*-octadecyl-cytarabine,
` elaidic
`acid cytarabine, and cytosine 1-$-D-arabinofuranoside (ara-
`C).
`In general, oral delivery of membersofthis class of
`[0007]
`compounds has proven difficult due to combinations of
`chemical instability, enzymatic instability, and/or poortissue
`permeability. For example, these compounds are known to
`be acid labile and thus unstable in the acidic gastric envi-
`ronment. In the case of 5-azacytidine, ara-C, decitabine and
`gemcitabine, an enzyme thought to be responsible for a
`significant portion of drug metabolism is cytidine deami-
`nase. Strategies to improve the oral bioavailability of this
`drug class have included the use of prodrugs to modify
`chemical and enzymatic instability, and/or the use of enzy-
`matic inhibitors.
`
`[0008] For example, DeSimoneet al describe theability of
`5-azacytidine to induce fetal hemoglobin production in
`baboons when administered via the intravenous (IV), sub-
`cutaneous (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, however no specific data is provided
`on the doses or responses observed without THU. 5-azacy-
`tidine doses ranged from 0.25 mg/kg/d to 8 mg/kg/d with
`co-adiministration of 20 mg/kg/d THU. Administration of
`THUalone was shownto result in a significant decrease in
`peripheral cytidine deaminase activity.
`[0009] Neil, et al describe the effect of THU on the
`pharmacokinetics and pharmacodynamicsofinter peritoneal
`(1.P.) and peri oral (P.O.) 5-azacytidine when administered to
`leukemic mice. Pharmacokinetic parameters were deter-
`mined using a bioassay that did not discriminate between
`5-azacytidine and its degradation and metabolism products.
`Inclusion of THU with IP administration hadlittle effect on
`the clearance or degradation of 5-azacytidine. Inclusion of
`THU with PO administration significantly increased both
`Cand t,,.. In both acute and chronic IP dosing the
`inclusion of THU did not influence the pharmacodymamic
`effects of 5-azacytidine except at the highest chronic dose
`which was toxic. Conversely, co-administration of THU
`with PO 5-azacytidine resulted in increased efficacy at all
`doses exceptthe highest chronic dose which was again toxic.
`[0010] Dunbar, et al describe the administration of 5-aza-
`cytidine via IV and PO routes for increased production of
`total 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 not result in increased
`
`hemoglobin levels.
`[0011] Dover, et al describe administration of 5-azacyti-
`dine via the SC and PO routes for increased production of
`total hemoglobin,fetal hemoglobin andF cells in sicklecell
`patients. 5-azacytidine oral bioavailability was assessed by
`clinical response only. Dover reports that oral doses of
`5-azacytidine (2 mg/kg/d) alone or THU (200 mg/d) alone
`did not result in increased F reticulocyte production. How-
`ever oral 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 5-aza-
`
`

`

`US 2008/0057086 Al
`
`Mar.6, 2008
`
`cytidine was co-administered with THU good clinical
`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 5-azacytidine 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
`compounds have also been described in, for example, U.S.
`Patent Application Publication No. 2004/0162263 (Sands, et
`al.) In this publication, delivery of 5-azacytidine 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. All U.S. patents and patent
`publications referenced herein are incorporated by reference
`herein in their entireties.
`[0013] Despite these efforts, a need remains for more
`effective methods and compositions which increase oral
`bioavailability of this class of compounds.
`
`BRIEF DESCRIPTION OF THE DRAWINGS
`
`represents a graph showing Absolute
`1
`FIG.
`[0014]
`Mucosal to Serosal Permeability of 5-azacytidine in Human
`Intestinal Tissue with and without Enzymatic Inhibition.
`[0015]
`FIG. 2 represents a graph showing Relative
`Mucosal to Serosal Permeability of 5-azacytidine in Human
`Intestinal Tissue with and without Enzymatic Inhibition with
`Respect to Atenolol.
`[0016]
`FIG. 3
`represents a graph showing Absolute
`Mucosal to Serosal Permeability of 5-azacytidine in Human
`Colonic Tissue with Various Concentrations of TPGS or
`
`Labrafil without Enzymatic Inhibition.
`[0017]
`FIG. 4 represents a graph showing Relative
`Mucosal to Serosal Permeability of 5-azacytidine in Human
`Colonic Tissue with Various Concentrations of TPGS or
`Labrafil without Enzymatic Inhibition.
`[0018]
`FIG. 5 shows concentration vs time profiles of
`individual subjects administered an oral formulation of the
`present invention.
`[0019]
`FIG. 6 shows concentration vs time profiles for the
`60 mg dose and the mean of the three 80 mg doses for
`individual subjects administered an oral formulation of the
`present invention.
`
`SUMMARY OF THE INVENTION
`
`In a first embodiment, the present invention com-
`[0020]
`prises a controlled release pharmaceutical composition for
`oral administration for enhanced systemic delivery of a
`cytidine analog comprising a therapeutically effective
`amount of a cytidine analog and a drugrelease controlling
`component which is capable of providing release of the
`cytidine analog primarily in the large intestine. After inges-
`tion by a patient, the cytidine analog is released primarily in
`the large intestine.
`invention
`the present
`[0021]
`In another embodiment,
`includes a method for treating a patient having a disease
`associated with abnormal cell proliferation. The method
`includes orally administering to the patient a controlled
`release pharmaceutical composition, comprising a therapeu-
`tically effective amount of 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.
`invention
`the present
`[0022]
`In another embodiment,
`includes a method of 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 amount of 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 and
`increases the bioavailability of the cytidine analog.
`[0023]
`In one embodiment, a condition to treat using the
`present invention is a myelodysplastic syndrome. In one
`embodiment, the cytidine analog is 5-azacytidine. In one
`embodiment, the drug release controlling componentis an
`enteric coating.
`
`DETAILED DESCRIPTION OF ‘THE
`INVENTION
`
`[0024] The present invention is based on the surprising
`discovery that 5-azacytidine and related compoundsare best
`absorbed in the lower gastrointestinal tract, i.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 found that in the case for cytidine analogs,
`absorption is greatest and most consistent between patients
`in colonic tissue. Accordingly, the present invention dem-
`onstrates the preparation of a solid oral dosage form of a
`cytidine analog, such as 5-azacytidine, using common phar-
`maceutical excipients designed for delivering pharmaceuti-
`cal compositions to the large intestine and colon. The term
`“absorb”, “absorption”, “absorbed” andthe like are used to
`indicate transfer of a cytidine analog across a relevant tissue,
`such as, for example,
`intestinal tissue. In some embodi-
`ments, absorbed cytidine analogs are taken up by the blood
`stream making the cytidine analog availableat least partially
`systemically. In some embodiments, absorption occurs with-
`out
`substantive degradation (i.e., undesirable chemical
`modification of) of the cytidine analog.
`[0025]
`Furthermore, the inventors have demonstrated that
`inclusion of THU (taught by others as a requirement to
`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, for-
`mulations of the present invention obviate the need to utilize
`enzymatic inhibitors such as THU in formulations to
`increase bioavailability of cytidine analogs. Avoidance of
`enzymatic inhibitorsis a desirable attribute for a therapeutic
`dosage form since such inclusion increases the formulation
`cost and complexity, and mayresult in instability, or unde-
`sirable, pharmacological,
`toxicological or other effects.
`Accordingly, oral delivery of 5-azacytidine without inclu-
`sion 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 5-azacytidine 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
`
`

`

`US 2008/0057086 Al
`
`Mar.6, 2008
`
`colon may be achieved with commercially available and
`pharmaceutically acceptable coatings such as, for example,
`enteric coatings.
`[0026]
`Furthermore, the inventors have demonstrated the
`preparation of solid oral dosage forms containing excipients
`and coatings which possess acceptable production and sta-
`bility characteristics for use as a pharmaceutical dosage
`form.
`
`In one embodiment, the present invention includes
`[0027]
`a controlled release pharmaceutical composition for oral
`administration comprising a) a therapeutically effective
`amountof a cytidine analog and b) a drugrelease controlling
`component for providing the release of the cytidine analog
`primarily in the large intestine. The controlled release phar-
`maceutical compositionsof the present invention will in one
`embodiment lack THU.
`
`In one embodiment, the cytidine analog useful in
`[0028]
`the present invention includes any moiety which1s struc-
`turally related to cytidine or deoxycytidine and functionally
`mimics and/or antagonizes the action of cytidine or deoxy-
`cytidine. These analogs mayalso be called cytidine deriva-
`tives 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'-difluorocy-
`tidine, 5-aza-2'-deoxy-2'-fluorocytidine, 2'-deoxy-2',2'-dif-
`luorocytidine (also called gemcitabine), or cytosine 1-B-D-
`arabinofuranoside (also called ara-C), 2(1H) pyrimidine
`riboside (also called zebularine), 2'-cyclocytidine, arabino-
`fuanosyl-5-azacytidine, dihydro-5-azacytidine, N*-octade-
`cyl-cytarabine, and elaidic acid cytarabine. In one embodi-
`ment,
`is 5-azacytidine and 5-aza-2'-deoxycytidine The
`definition of cytidine analog used herein also includes
`mixtures of cytidine analogs.
`[0029] Cytidine analogs useful in the present invention
`maybe manufactured by any methods knownin theart. In
`one embodiment, methods to manufacture include methods
`as disclosed in U.S. Ser. No. 10/390,526 (U.S. Pat. No.
`7,038,038); U.S. Ser. No. 10/390,578 (U.S. Pat. No. 6,887,
`855); U.S. Ser. No. 11/052,615 (U.S. Pat. No. 7,078,518);
`US. Ser. No. 10,390,530 (U.S. Pat. No. 6,943,249); and
`USS. Ser. No. 10/823,394, all
`incorporated by reference
`herein in their entireties.
`
`the amounts of a cytidine
`In one embodiment,
`[0030]
`analog to use in methodsof the present invention and in the
`oral formulations of the present invention include a thera-
`peutically effective amount. Therapeutic indications are dis-
`cussed more fully herein below. Precise amounts for thera-
`peutically 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/m? (based on patient body
`surface area) or about 4 mg/kg (based on patient body
`weight) as single or divided (2-3) daily doses.
`[0031] The controlled release pharmaceutical composi-
`tions of the present invention include a drug release con-
`trolling component. The drug release controlling component
`is adjusted suchthatthe release of the cytidine analog occurs
`primarily in the large intestine. In one embodiment,atleast
`about 95% of the cytidine analog is 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 analog is released in the large
`
`least about 70% of the cytidine analog is
`intestine, at
`released in the large intestine, at least about 60% of the
`cytidine analog is 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
`intestinesis at least about 40%,at least about 30%,orat least
`about 20% of the cytidine analog. The term “release” refers
`to the process whereby the 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.
`[0032] 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 comprisea plural-
`ity of granules, beads, powders or pellets that may or may
`not be encapsulated. Such formats mayalso be referred to as
`the “drug core” which contains the cytidine analog. Such
`dosage forms are prepared using conventional methods
`known to those in the field of pharmaceutical formulation
`and are describedin the pertinent texts, e.g., in REMINGTON:
`THE SCIENCE AND PRACTICE OF PHARMACY, 20th Edition,
`Lippincott Williams & Wilkins, 2000).
`[0033]
`‘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 process-
`ing procedures and equipment. One method for forming
`tablets is by direct compression of a powdered, crystalline or
`granular composition containing the cytidine analog, alone
`or in combination with one or morecarriers, additives, or the
`like. As an alternative to direct compression, tablets can be
`prepared using wet-granulation or dry-granulation pro-
`cesses. Tablets may also be moldedrather than compressed,
`starting with a moist or otherwise tractable material; par-
`ticularly, compression and granulation techniques are used
`in one embodiment.
`
`In another embodiment, capsules may be used. Soft
`[0034]
`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 andthe 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, corn 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 composi-
`tion may additionally include colorings,
`flavorings and
`opacifiers as required.
`[0035] The cytidine analog in one embodimentis prepared
`as a controlled release tablet or capsule which includes a
`drug core comprising the pharmaceutical composition and
`optional excipients (described elsewhere herein). Option-
`ally, a “seal coat”, described elsewhere herein, is applied to
`the drug core before addition of the drug release component.
`The drug release component is formulated to provide for
`release of the cytidine analog primarily in the large intestine
`(colon). In one embodiment, minimalrelease of the cytidine
`analog occurs in the upper reaches of the gastrointestinal
`tract, e.g., the stomach and small intestine.
`[0036] The small intestine extends from the pylorusto the
`colic valve where it ends in the large intestine. The small
`
`

`

`US 2008/0057086 Al
`
`Mar.6, 2008
`
`intestine is about 6 meters long and is 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.
`Almostall 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.
`
`Indigestible food passesinto the large intestine. By
`[0037]
`the time ingested material leaves the small intestine, virtu-
`ally all nutrient absorption will have occurred. The large
`intestine extends from the end of the ileum (distal ileum) to
`the anus. Thelarge intestine is divided into the cecum,colon,
`rectum, and anal canal. The colon is divided into four parts:
`the ascending,
`transverse, descending, and sigmoid. The
`substantial release of the cytidine compoundofthe present
`invention may occur in any portion ofthe large intestine. In
`one embodiment, release primarily occurs at
`the upper
`regions of the large intestine, such as, for example, at the
`distal ileum, cecum, and/or the ascending colon.
`[0038]
`It is knownthat 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 ofthe material
`leaving the stomach to approximately pH 6.0 ta 6.5. The
`large intestine and the colon are about pH 6.4 to 7.0. The
`transit time through the small intestine is approximately
`three hours. In contrast, the transit time through the large
`intestine is approximately 35 hours.
`[0039] Methods by which to formulate compositions to
`target specific regions of the gastrointestinal tract are known
`in the art, described in numerous publications, and all
`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 pro-
`cess in the gastrointestinal tract. A drug release controlling
`component may take advantage of processes and/or condi-
`tions within the gastrointestinal tract and in specific regions
`of the gastrointestinal tract such as, for example, osmotic
`pressure, hydrodynamic pressure, vapor pressure, mechani-
`cal action, hydration status, pH, bacterial
`flora, and
`enzymes. Specific U.S. patents incorporated by reference
`herein include, amongothers, 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.
`[0040] Optionally, pharmaceutical compositions of the
`present invention including drug cores may further comprise
`a seal coating material that seals the drug to prevent decom-
`position due to exposure to moisture, such as hydroxypro-
`pylmethylcellulose. Accordingly, the drug core of the phar-
`maceutical composition (containing the cytidine analog)
`mayfirst be sealed with the seal coating material and then
`coated with the drug release controlling component
`to
`prevent decomposition of the cytidine analog by exposure to
`moisture. Seal coating materials include,
`in one embodi-
`
`ment, acetyltributyl citrate, acetyltriethyl citrate, calcium
`carbonate, carauba wax, cellulose acetate, cellulose acetate
`phthalate, cetyl alcohol, chitosan, ethylcellulose, fructose,
`gelatin, glycerin, glyceryl behenate, glyceryl palmitostear-
`ate, hydroxyethyl cellulose, hydroxyethylmethyl cellulose,
`hydroxypropyl
`cellulose,
`hypromellose,
`hypromellose
`phthalate,
`isomalt,
`latex particles, maltitol, maltodextrin,
`methylcellulose, microcrystalline wax, paraffin, poloxamer,
`polydextrose, polyethylene glycol, polyvinyl acetate phtha-
`late, polyvinyl alcohol, povidone,
`shellac, shellac with
`stearic acid, sodium carboxymethy] cellulose, sucrose, tita-
`nium oxide, tributyl citrate, triethyl citrate, vanillin, white
`wax, xylitol, yellow wax, and zein. Compositions of the
`present invention may also include film forming agents,
`which include, for example, ammonium alginate, calcium
`carbonate, chitosan, chlorpheniramine maleate, copovidone,
`dibutyl phthalate, dibutyl sebacate, diethyl phthalate, dim-
`ethyl phthalate, ethyl lactate, ethylcellulose, gelatin, hydrox-
`yyethyl cellulose, hydroxypropyl cellulose, hypromellose,
`hypromellose acetate succinate, maltodextrin, polydextrose,
`polyethylene glycol, polyethylene oxide, polymethylacry-
`lates, poly(methylviny] ether/maleic anhydride), polyviny-
`lacetate phthalate, triethyl citrate, and vanillin. The amount
`of seal coating will vary in accordance with factors known
`by those of skill in the art. The amountof seal coat is, in one
`embodiment, about 1% w/w ofthe drug core; about 2%, w/w
`of the drug core, about 3%, w/w, of the drug core, about 4%,
`wiw, ofthe drug core; about 5% w/w ofthe drug core; about
`6%, w/w of the drug core, about 7%, w/w, of the drug core,
`about 8%, w/w/, of the drug core; about 9% w/w ofthe drug
`core; about 10%, w/w ofthe drug core, about 11%, w/w, of
`the drug core, about 12%, w/w, of the drug core; about 14%
`w/w of the drug core; about 16%, w/w of the drug core,
`about 18%, w/w, of the drug core, about 20%, w/w, of the
`drug core; or more, if determined to be appropriate. Seal
`coats may also be applied at amounts between about 1% and
`about 10% w/w ofthe drug core, between about 2% and 9%
`w/w of the drug core, between about 3% and 8% w/w ofthe
`drug core, between about 4% and 7% w/w ofthe drug core,
`and between about 5% and about 6% w/wofthe drug core.
`[0041]
`In one embodiment, drug release controlling com-
`ponents 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 coat-
`ings of either the same or different types of coatings. In
`choosing an appropriate coating or mixture thereof,
`the
`formulations practitioner may consider a numberof vari-
`ables 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 (which is influ-
`enced by thickness of the coatings and/or additional com-
`ponents in the coatings); time of transit through the gas-
`trointestinal tract, and whether the coatings can be degraded
`by the patent’s digestive enzymes or require enzymes
`present only in bacteria residing in the lowerintestine. As an
`example of a combination drug release controlling compo-
`nent is, 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
`
`

`

`US 2008/0057086 Al
`
`Mar.6, 2008
`
`rate. A numberof specific formulationseffective 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.
`
`EUDRAGIT S. The EUDRAGITSfilms are colorless,
`component to delay drug release for a predetermined period.
`The thickness of the inner layer can be adjusted to determine
`transparent and brittle. In one embodiment,
`the enteric
`the lag time.
`coating comprises EUDRAGIT $100. The EUDRAGIT S
`coatings are insoluble in pure water,
`in buffer solutions
`[0042] Methods bywhich skilled practitioners can assess
`below a pH of 6.0 andalso in natural andartificial gastric
`where a drug is released in the gastrointestinal tract of either
`juices. They are slowly soluble in the region of the digestive
`animal models or human volunteers are known in theart,
`tract where the juices are neutral to weakly alkaline(i.e., the
`and include scintigraphic studies,
`testing in biorelevant
`large intestine and the colon) and in buffer solutions above
`medium which simulates the fluid in relevant portions of the
`a pH of 7.0. Mixtures of these various enteric polymers
`gastrointestinal tract, among others.
`recited above, can be used in the present invention. Further,
`[0043]
`In one embodiment, a drug release controlling
`the use of plasticizers is included in one embodiment with
`component may include an enteric coating. The term
`the enteric polymer coatings useful herein.
`“enteric coating” refers to a coating that allows a cytidine
`[0047] As knownin the art and discussed in sources such
`analog formulation to pass through the stomach substantially
`intact and subsequently disintegrate substantially in the
`as Patel et al. “Colon Specific Delivery” Drug Delivery
`intestines. In one embodiment, the disintegration occurs in
`Technology (2006) Vol. 6 62-71, and Khan etal., J. Con-
`the large intestine.
`trolled Release 1999; 58:215-222,the disintegration rates of
`enteric coated tablets are dependent on the polymer combi-
`[0044] The coating of pH-sensitive (enteric) polymers to
`nation used to coat the tablets, the pH of the disintegration
`tablets, capsules and other oral formulations of the present
`media, and the coating levelof the tablets(i.e., thickness of
`invention provided delayed release and protect the active
`the coating). The presenceofplasticizer and the nature of the
`drug fromgastric fluid. In general, enteric coatings should
`salts in the dissolution medium also influence the dissolution
`be able to withstand the lower pH values of the

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