throbber
(19) United States
`(12) Patent Application Publication (io) Pub. No.: US 2004/0162263 Al
`Sands et al.
`(43) Pub. Date:
`Aug. 19,2004
`
`US 20040162263A1
`
`(54) PHARMACEUTICAL FORMULATIONS
`TARGETING SPECIFIC REGIONS OF THE
`GASTROINTESINAL TRACT
`
`(75)
`
`Inventors: Howard Sands, Wilmington, DE (US);
`Sanjeev Redkar, Union City, CA (US);
`Harish Ravivarapu, Union City, CA
`(US)
`
`Correspondence Address:
`WILSON SONSINI GOODRICH & ROSATI
`650 PAGE MILL ROAD
`PALO ALTO, CA 943041050
`
`(73) Assignee: SuperGen, Inc., a Delaware Corpora(cid:173)
`tion, Dublin, CA
`
`(21) Appl. No.:
`
`10/698,983
`
`(22) Filed:
`
`Oct. 31, 2003
`
`Related U.S. Application Data
`
`(60) Provisional application No. 60/423,179, filed on Oct.
`31, 2002.
`
`Publication Classification
`
`(51)
`
`Int. Cl.7
`
`(52) U.S. Cl
`
`A61K 31/7076; A61K 31/7072;
`A61K 9/24
`514/46; 514/50; 424/471
`
`(57)
`
`ABSTRACT
`
`Oral formulations ol pharmaceuticals are provided with
`enhanced bioavailability by targeting specific regions ol the
`gastrointestinal tract. Particularly, water soluble and acid-
`labile drugs such as cytidine analogs (e.g., decitabine) and
`2'-deoxyadenosine analogs (e.g., pentostatin) are formulated
`with pH-sensitive polymers so that these drugs are prefer(cid:173)
`ably absorbed in the upper regions of the small intestine,
`such as the jejunum. In addition, drugs with poor oral
`bioavailability such as camptothecin compounds (e.g., 9-ni-
`tro-camptothecin) can also be formulated using similar
`strategies in order to significantly improve their oral bio(cid:173)
`availability. These formulations can be used to treat a wide
`variety of diseases or conditions, such hematological disor(cid:173)
`ders, benign tumors, cancer, restenosis, inflammatory dis(cid:173)
`eases, and autoimmune diseases.
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1021-0001
`
`

`

`Patent Application Publication Aug. 19,2004 Sheet 1 of 15
`
`US 2004/0162263 Al
`
`DUODENUM i-
`
`TRANSVERSE:
`COLON
`
`ASCENDING
`COLON
`
`JEJUNUM
`
`CECUM
`
`STOMACH
`
`/PANCREAS
`
`LARGE
`INTESTINE
`SMALL
`INTESTINE
`
`DESCENDING
`COLON
`
`SIGMOID
`FLEXURE
`
`, l £ UM ^APPENDIX IN RECTUM
`
`HUMAN INTESTINES
`
`FIGURE 1
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1021-0002
`
`

`

`Patent Application Publication Aug. 19,2004 Sheet 2 of 15
`
`US 2004/0162263 Al
`
`FIGURE 2
`
`Pentostatin
`
`El Saline
`• Buffered
`
`feE IC
`
`IL
`IJ
`Intestinal Region
`
`t
`
`PO
`
`1.400
`
`1.200
`
`1.000
`
`+•>
`
`1 0.800
`'55
`% 0.600
`o
`S
`
`0.400
`
`0.200
`
`0.000
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1021-0003
`
`

`

`Patent Application Publication Aug. 19,2004 Sheet 3 of 15
`
`US 2004/0162263 Al
`
`10000.0
`
`1000.0
`
`•£• 100.0
`u
`c
`o
`o
`
`10.0
`
`1.0
`
`3000.0
`
`2500.0
`
`f
`
`^ 2000.0
`
`•S- 1500.0
`
`O 1000.0
`
`FIGURES
`
`Intravenous Administration
`
`-X— Systemic
`-O— Portal Vein
`
`200
`
`400
`
`600
`
`800
`
`Time (min)
`
`Intravenous Administration
`
`-X— Systemic
`-D— Portal Vein
`
`200
`
`400
`Time (min)
`
`-G-
`
`800
`
`600
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1021-0004
`
`

`

`Patent Application Publication Aug. 19,2004 Sheet 4 of 15
`
`US 2004/0162263 Al
`
`1000.0
`
`I- 100.0 ^
`D)
`c
`
`O
`
`10.0
`
`1.0
`
`250.0
`
`200.0
`
`150.0
`
`FIGURE 4
`
`Oral Administration
`
`Saline
`Buffer
`
`200
`
`400
`
`600
`
`800
`
`Time (min)
`
`Oral Administration
`
`-•— Saline
`-O-Buffer
`
`400
`
`600
`
`800
`
`Time (min)
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1021-0005
`
`

`

`Patent Application Publication Aug. 19, 2004 Sheet 5 of 15
`
`US 2004/0162263 Al
`
`FIGURES
`
`Jejuna! Administration
`
`1000.0 -I
`
`M\
`r "^-^ ^i;^
`
`- •- Saline
`—TT— Rirffcir
`i—i uLnrer
`
`-
`
`- ^T
`^~^^-_id
`
`5
`
`U
`c
`o
`O
`
`100.0 -
`
`^/^K
`
`\
`
`1
`10 0 i
`J
`
`1 0 -
`c
`)
`
`200
`
`400
`
`600
`
`800
`
`Time (min)
`
`Jejunal Administration
`
`450.0
`400.0
`350.0
`=* 300.0
`"I 250.0
`li 200.0
`O 150.0
`100.0
`50.0
`0.0
`
`-•—Saline
`-Q-Buffer
`
`200
`
`400
`Time (min)
`
`600
`
`800
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1021-0006
`
`

`

`Patent Application Publication Aug. 19,2004 Sheet 6 of 15
`
`US 2004/0162263 Al
`
`1000.0
`
`I 1000
`
`1.0
`
`300.0 i
`
`250.0
`
`FIGURE 6
`
`lleum Administration
`
`-•—Saline
`•O—Buffer
`
`200
`
`400
`Time (min)
`
`600
`
`800
`
`lleum Administration
`
`-•—Saline
`•O— Buffer
`
`200
`
`400
`Time (min)
`
`600
`
`800
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1021-0007
`
`

`

`Patent Application Publication Aug. 19,2004 Sheet 7 of 15
`
`US 2004/0162263 Al
`
`FIGURE?
`
`Colon Administration
`
`-A—Saline
`-&-Buffer
`
`200
`
`400
`
`600
`
`800
`
`Time (min)
`
`Colon Administration
`
`—A—Saline
`- A- Buffer
`
`1000.0
`
`=* 100.0
`
`1.0
`
`aKn n _
`
`300.0 •
`
`r
`1
`
`_ 250.0 -
`
`~5> 200.0 -
`
`I'sn n •
`
`ti
`o
`
`50.0 '
`
`^r
`
`--
`_
`
`—-
`
`K
`
`\
`
`K^ N i — ^ * _ __
`
`0.0 L
`\
`0
`
`.
`200
`
`m—i
`400
`
`Time (min)
`
`—i
`600
`
`%J
`
`1
`800
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1021-0008
`
`

`

`Patent Application Publication Aug. 19,2004 Sheet 8 of 15
`
`US 2004/0162263 Al
`
`FIGURE 8
`
`IV Dose Response
`
`1400
`
`B ^i
`
`iByt,.
`
`Wfg?=;o*wm ft
`^ i^gaBfr^j
`
`1 2 00
`0
`5 1000
`.2 800
`1 600
`w
`>
`W
`
`400
`
`200
`
`3?
`
`•fgryiV
`
`^^f^
`
`it J*
`
`H
`
`J ^ - - fe
`
`JV
`
`^ i ;^
`
`^ H fv -
`
`»-r V ^ ^?
`
`F*SE
`
`XM
`
`•
`
`AUC
`
`if
`
`.
`
`Linear (AUC)
`
`S U li
`
`0.5
`
`1
`
`1.5
`
`2.5
`
`Dose (mg/kg)
`
`FIGURE 9
`
`Low IV dose (0.75 mg/kg)
`
`1000
`
`-jE 800
`
`£*- ^MT^
`
`r cr
`
`S ys
`
`• PV
`
`a p - i 'v •>«
`
`' ?-
`
`*
`
`"if"
`
`f I if £4
`
`'
`
`• a1"
`
`"?»
`
`"•
`
`50
`
`100
`
`150
`
`200
`
`250
`
`300
`
`Time (min)
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1021-0009
`
`

`

`Patent Application Publication Aug. 19,2004 Sheet 9 of 15
`
`US 2004/0162263 Al
`
`FIGURE 10
`
`Medium IV dose (1.5 mg/kg)
`
`1400
`
`•Sys
`
`• PV
`
`-t
`
`""rvv
`
`50
`
`100
`
`150
`Time (min)
`
`200
`
`250
`
`300
`
`FIGURE 11
`
`High dose IV dose (2.5 mg/kg)
`
`2500
`
`--*« * " »/* ^zr.
`
`Jjj 2000
`c
`~ 1500
`
`'
`
`iggj. - - r - ^ s-
`'•4» ^
`f
`\}
`,d"'3ffl
`^4!
`
`^ -m- sys «•;% -^ ' v
`. ^m:
`" ^ %•, -, rj»i -
`^
`,. 4" ?
`t i t"
`
`iS^i'
`
`50
`
`100
`
`150
`
`200
`
`250
`
`300
`
`Time (min)
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1021-0010
`
`

`

`Patent Application Publication Aug. 19,2004 Sheet 10 of 15
`
`US 2004/0162263 Al
`
`FIGURE 12
`
`PV infusion (2.5 mg/kg)
`
`SYS
`
`^36. J »
`
`-VH
`
`&xtm
`
`ml
`
`50
`
`100
`
`150
`
`200
`
`Time (min)
`
`^ J
`250
`
`300
`
`FIGURE 13
`
`PO dose 2.5 mg/kg
`
`2000
`
`1500
`
`1000
`

`
`500
`
`1000
`

`
`800
`
`600
`
`400
`
`200
`
`u
`

`2
`
`50
`
`100
`
`150
`
`200
`
`250
`
`300
`
`Time (min)
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1021-0011
`
`

`

`Patent Application Publication Aug. 19,2004 Sheet 11 of 15
`
`US 2004/0162263 Al
`
`FIGURE 14
`
`USI infusion 2.5 mg/kg
`
`J? "r a *
`
`T-**
`
`, S « i«
`
`• py
`
`i C *i
`
`50
`
`100
`
`150
`
`200
`
`250
`
`300
`
`Time (min)
`
`FIGURE 15
`
`LSI infusion 2.5 mg/kg
`
`1000
`
`^
`$sf
`?&«r*
`
`• - PV
`
`^
`Pfc
`
`*
`
`^ <%&
`
`100
`
`150
`
`200
`
`250
`
`Time (min)
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1021-0012
`
`

`

`Patent Application Publication Aug. 19,2004 Sheet 12 of 15
`
`US 2004/0162263 Al
`
`FIGURE 16
`
`Colon infusion 2.5 mg/kg
`RS-44
`
`80
`60
`Time (min)
`
`140
`
`FIGURE 17
`
`Colon infusion 2.5 mg/kg
`RS-45
`
`1000
`
`f" -<>•< > > 41 ^ f «V
`80
`60
`Time (min)
`
`140
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1021-0013
`
`

`

`Patent Application Publication Aug. 19,2004 Sheet 13 of 15
`
`US 2004/0162263 Al
`
`FIGURE 18
`
`10 z
`
`1
`
`0.1
`
`~
`c
`o
`ra
`i_
`c
`0)
`o
`c
`o
`o
`ro E
`w
`i5
`CL
`
`0.01
`
`JEJUNUM Administration
`
`9-NC systemic
`9-NC portal vein
`9-AC systemic
`9-AC portal vein
`
`10
`
`15
`
`20
`
`25
`
`Time (hr)
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1021-0014
`
`

`

`Patent Application Publication Aug. 19,2004 Sheet 14 of 15
`
`US 2004/0162263 Al
`
`FIGURE 19
`
`£>
`c o
`CD 4= c
`O c o o
`
`CD
`
`ro
`w
`i5
`
`10 i
`
`1
`
`0.1
`
`0.01
`
`ILEUM Administration
`•— 9-NC systemic
`o— 9-NC portal vein
`•— 9-AC systemic
`9-AC portal vein
`
`10
`
`15
`
`Time (hr)
`
`25
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1021-0015
`
`

`

`Patent Application Publication Aug. 19,2004 Sheet 15 of 15
`
`US 2004/0162263 Al
`
`FIGURE 20
`
`COLON Administration
`
`•— 9-NC systemic
`o— 9-NC portal vein
`•— 9-AC systemic
`v— 9-AC portal vein
`
`10 q
`
`c
`c
`o
`
`•J=>
`
`c
`a>
`o
`c
`o
`o
`ro
`E
`
`0.1
`
`0.01
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1021-0016
`
`

`

`US 2004/0162263 Al
`
`Aug. 19, 2004
`
`PHARMACEUTICAL FORMULATIONS
`TARGETING SPECIFIC REGIONS OF THE
`GASTROINTESINAL TRACT
`
`CROSS-REFERENCE TO RELATED
`APPLICATION
`
`[0001] This application claims priority to U.S. Provisional
`Application No. 60/423,179, filed Oct. 31, 2002, which is
`hereby incorporated by reference herein.
`
`BACKGROUND OF THE INVENTION
`
`[0002] 1. Field of the Invention
`[0003] This invention relates to pharmaceutical formula(cid:173)
`tions for oral delivery of drugs to specific regions of the
`gastrointestinal tract for enhanced bioavailability, and more
`particularly relates to oral formulations of water-soluble,
`acid-labile drugs such as cytidine analogs (e.g., decitabine)
`and 2'-deoxyadenosine analogs (e.g., pentostatin), as well as
`drugs with poor bioavailability such as camptothecin com(cid:173)
`pounds.
`
`[0004] 2. Description of the Related Art
`[0005] 1. Decitabine
`[0006] Decitabine, 5-aza-2'-deoxycytidine, is an antago(cid:173)
`nist of its related natural nucleoside, deoxycytidine. The
`only structural difference between these two compounds is
`the presence of a nitrogen at position 5 of the cytosine ring
`in decitabine as compared to a carbon at this position for
`deoxycytidine. Two isomeric forms of decitabine can be
`distinguished. The P-anomer is the active form. The modes
`of decomposition of decitabine in aqueous solution are (a)
`conversion of the active P-anomer to the inactive a-anomer
`(Pompon et al. (1987) J. Chromat. 388:113-122); (b) ring
`cleavage of the aza-pyrimidine ring to form N-(formylami-
`dino)-N'-P-D-2'-deoxy-(ribofuranosy)-urea
`(Mojaverian
`and Repta (1984) J. Pharm. Pharmacol. 36:728-733); and (c)
`subsequent forming of guanidine compounds (Kissinger and
`Stemm (1986) J. Chromat. 353:309-318).
`
`[0007] Decitabine possesses multiple pharmacological
`characteristics. At a molecular level, it is S-phase dependent
`for incorporation into DNA. At a cellular level, decitabine
`can induce cell differentiation and exert hematological tox-
`icity. Despite having a short half life in vivo, decitabine has
`an excellent tissue distribution.
`
`[0008] The most prominent function of decitabine is its
`ability to specifically and potently inhibit DNAmethylation.
`Methylation of cytosine to 5-methylcytosine occurs at the
`level of DNA. Inside the cell, decitabine is first converted
`into its active form, the phosphorylated 5-aza-deoxycyti-
`dine, by deoxycytidine kinase which is primarily synthe(cid:173)
`sized during the S phase of the cell cycle. The affinity of
`decitabine for the catalytical site of deoxycytidine kinase is
`similar to the natural substrate, deoxycytidine. Momparler et
`al. (1985) 30:287-299. After conversion to its triphosphate
`form by deoxycytidine kinase, decitabine is incorporated
`into replicating DNA at a rate similar to that of the natural
`substrate, dCTP Bouchard and Momparler (1983) Mol.
`Pharmacol. 24:109-114.
`
`[0009]
`Incorporation of decitabine into the DNA strand
`has a hypomethylation effect. Each class of differentiated
`cells has its own distinct methylation pattern. After chro(cid:173)
`
`mosomal duplication, in order to conserve this pattern of
`methylation, the 5-methylcytosine on the parental strand
`serves to direct methylation on the complementary daughter
`DNA strand. Substituting the carbon at the 5 position of the
`cytosine for a nitrogen interferes with this normal process of
`DNA methylation. The replacement of 5-methylcytosine
`with decitabine at a specific site of methylation produces an
`irreversible inactivation of DNAmethyltransferase, presum(cid:173)
`ably due to formation of a covalent bond between the
`enzyme and decitabine. Juttermann et al. (1994) Proc. Natl.
`Acad. Sci. USA 91:11797-11801. By specifically inhibiting
`DNA methyltransferase, the enzyme required for methyla(cid:173)
`tion, the aberrant methylation of the tumor suppressor genes
`can be prevented.
`[0010] Decitabine is commonly supplied as a sterile lyo-
`philized powder for injection, together with buffering salt,
`such as potassium dihydrogen phosphate, and pH modifier,
`such as sodium hydroxide. For example, decitabine is sup(cid:173)
`plied by SuperGen, Inc., as lyophilized powder packed in 20
`mL glass vials, containing 50 mg of decitabine, monobasic
`potassium dihydrogen phosphate, and sodium hydroxide.
`When reconstituted with 10 mL of sterile water for injection,
`each mL contain 5 mg of decitabine, 6.8 mg of KH2P04, and
`approximately 1.1 mg NaOH. The pH of the resulting
`solution is 6.5-7.5. The reconstituted solution can be further
`diluted to a concentration of 1.0 or 0.1 mg/mL in cold
`infusion fluids, i.e., 0.9% Sodium Chloride; or 5% Dextrose;
`or 5% Glucose; or Lactated Ringer's. The unopened vials
`are typically stored under refrigeration (2-8° C; 36-46° R),
`in the original package.
`
`[0011] Decitabine
`to
`typically administrated
`is most
`patients by injection, such as by a bolus I.V. injection,
`continuous I.V. infusion, or I.V. infusion. The length of I.V.
`infusion is limited by decitabine's decomposition in aqueous
`solutions.
`[0012] It has been found that when 5-azacytidine (azaC) is
`orally administered (8 mg/kg) to repeatedly phlebotomized
`baboon (PCV less than 20%) there is no elevation in the fetal
`hemoglobin levels (Hb F), indicating very minimal oral
`bioavailability. DeSimone et al (1985) American. J. of Hem.
`18:283-288. AzaC is more active when administered
`parenterally than orally in the treatment of L1210 leukemic
`mice due to poor bioavailability. Neil at al (1975) Cancer
`Chemother. Rep. 59:459-465. In L1210 leukemic mice,
`peroral doses of cytarabine (cytosine arbinaoside) required
`to elicit an anti-tumor effect are about 3 to 10 times those
`required when administered parenterally. Neil et al. (1970)
`Cancer Research 30:2166-2172. The poor bioavailability of
`such cytidine analogs is presumably due to the degradation
`of the cytidine analog by cytidine deaminases as well as their
`inherent chemical instability in the acidic gastric environ(cid:173)
`ment.
`[0013] 2. 2'-Deoxyadenosine Analogs
`[0014] Certain 2'-deoxyadenosine analogs have been
`found to have very useful clinical pharmacological benefits.
`These include, but are not limited to, 2'-deoxycoformycin
`(also referred to as dCF, pentostatin, or NIPENT®), an
`inhibitor of adenosine deaminase; fludarabine monophos-
`phate (FLU), a fluorinated analogue of adenine that is
`relatively resistant to adenosine-deaminase and 2-chloro-2'-
`deoxyadenosine (also known as cladribine or 2CDA) a drug
`also resistant to adenosine deaminase through introduction
`of a chlorine at the 2 carbon.
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1021-0017
`
`

`

`US 2004/0162263 Al
`
`Aug. 19, 2004
`
`[0015] In humans, these compounds are assumed to act
`through a number of adenosine related pathways, particu(cid:173)
`larly the adenosine deaminase (ADA) pathway. A genetic
`deficiency of ADA may cause severe combined immunode(cid:173)
`ficiency. Dighiero, G., "Adverse and beneficial immunologi-
`cal effects of purine nucleoside analogues,"//emato/ Cell
`Ther, 38:575-581 (1996).
`
`dosage forms. However, the art recognized serious problems
`with the development of an oral dosage form. Chief among
`these is that adenosine analogs have been known for years
`to be susceptible to acid-catalyzed glycosidic cleavage.
`Therefore, one of skill in the art would expect that an orally
`administered adenosine analog would be cleaved in the
`stomach, and rendered inactive.
`
`[0016] While the exact nature of the ADA pathway inter(cid:173)
`vention seems unclear, it may be that analogs of adenosine
`resistant to cellular deamination might mimic the ADA-
`deficient state. Lack of ADA seems to lead to a build up of
`deoxyadenosine and adenosine triphosphate in the cell, thus
`fatally accelerating DNA strand breaks in the cell. Under
`normal conditions, cells are continuously breaking and
`rejoining DNA. When this physiological process is acceler(cid:173)
`ated by the effect of excess adenosine triphosphate, it leads
`to consumption of NAD for poly-ADP-ribose synthesis.
`This polymer is produced from nicotinamide adenosine
`dinucleotides (NAD) in a reaction catalyzed by the chroma-
`tin-associated poly(ADP-ribose) synthetase, leading to a
`depletion of the NAD content of the cell. This depletion
`induces a profound alteration of cellular reducing power,
`because of lethal ADP and ATP depletion.
`
`[0017] The result is programmed cell death through acti(cid:173)
`vation of a Ca2+, Mg2+-dependent endonuclease. Hence, it
`appears that nucleoside analogs according to the invention
`can act on cells, with preferential lymphocytic activity, via
`an apoptotic process. The fact that supplementation of a cell
`medium with the NAD precursor of nicotinamide or 3-ami-
`nobenzamide, an inhibitor of poly (ADP-ribose) synthetase,
`prevented NAD depletion and reduces 2CDA toxicity, tends
`to support this hypothesis.
`
`[0018] The various 2'-deoxyadenosine analogs affect the
`ADA pathway in different manners. DCF, for example, has
`been shown to be an quasi-irreversible inhibitor of ADA. By
`favoring the predominance of deoxycytidine kinase (DCK)
`over the dephosphorylating enzyme 5-nucleotidase in lym(cid:173)
`phocytes it induces a preferential accumulation of deoxy-
`adenosine-5'-triphosphate (dATP). By comparison, FLU and
`2CDA are rather resistant to the enzyme. Both drugs are
`initially phosphorylated by DCK and contribute to the
`accumulation of cellular adenosine triphosphate surrogates.
`As noted above, the accumulation of adenosine triphosphate,
`whether by the presumed DCF mechanism, or the FLU or
`2CDA mechanism, promotes the apoptotic death of the cell.
`
`[0019] A problem with administering these 2-deoxyad-
`enosine analogs is their dosage form. Currently, these ana(cid:173)
`logs are available only in an intravenous (IV) dosage form.
`While this dosage form is customary, especially for use in
`oncology indications, it is limiting in a variety of ways. For
`example, IV dosing is expensive. It requires a highly trained
`medical professional to administer the IV dose. The dosing
`involves expensive equipment and materials. Additionally,
`IV dosing presents increased possibilities of infection,
`through use of contaminated equipment or accidental con(cid:173)
`tamination, for example. This is a special concern in health
`care settings where increased incidences of antibiotic resis(cid:173)
`tant bacteria are being noted.
`
`[0020] A seemingly natural solution to the IV dosage
`problem is the development of an oral dosage form. Such a
`dosage form alleviates most, if not all, of the above-men(cid:173)
`tioned problems associated with IV or other parenteral
`
`[0021] For example, investigators studying 2'-deoxycofor-
`mycin have not considered oral administration of the drug
`worth studying because of its known acid lability. Marvin
`M. Chassin et al. Biochemical Pharmacology 28:1849-1855
`(1979). Likewise, other researchers have reported on the
`acid lability of 2'-deoxycoformycin. L. A. al-Razzak et al.
`7:452-460 (1990).
`
`[0022] Other adenosine analogs may be expected to have
`similar acid lability characteristics. A. Tarasiuk et al. Arch.
`Immunol. Ther. Exp. (Warsz) 42:13-15 (1994); T. Ono
`Nucleic Acids Res. 25:4581-4588 (1997).
`[0023] 3. Camptothecin Compounds
`[0024] The original Camptothecin was isolated from the
`plant, Camptotheca acuminata, in the 1960's (Wall, M. et al.
`(1966) J. Am. Chem. Soc. 88: 3888-3890). Camptothecin
`has a pentacyclic ring system with only one asymmetric
`center in ring E with a 20(S)-configuration. The pentacyclic
`ring system includes a pyrrole quinoline moiety (rings A, B
`and C), a conjugated pyridone (ring D), and a six-membered
`lactone (ring E) with an a-hydoxyl group.
`
`[0025] Camptothecin itself is highly lipophilic and poorly
`water-soluble. Sodium camptothecin that is solubilized by
`sodium hydroxide in water was used in clinical trials in the
`early 70's and found to have antitumor activity. However,
`this formulation of camptothecin administered via i.v.
`caused unpredictable side effects such as myelosuppression
`and hemorrhagic cystitis. Clinical trials with sodium camp(cid:173)
`tothecin were eventually discontinued because of these
`toxicities and the lack of consistent antitumor activity.
`
`[0026] Continued evaluation of this agent showed that the
`sodium carboxylate salt is only 10% as potent as the native
`camptothecin with the closed lactone ring intact (Wall et al.
`in (1969) "International Symposium on Biochemistry and
`Physiology of the Alkaloids, Mothes et al. eds. Academic
`Verlag, Berlin, 77; Giovanella et al. (1991) Cancer Res.
`51:3052). Studies also showed that camptothecin and its
`derivatives undergo an alkaline hydrolysis of the E-ring
`lactone, resulting in a carboxylate form of camptothecin. At
`pH levels below 7.0, the lactone E-ring form of camptoth(cid:173)
`ecin predominates. However, intact lactone ring E and
`a-hydoxyl group have been shown to be essential for
`antitumor activity of camptothecin and its derivatives.
`
`[0027] Camptothecin and its derivatives have been shown
`to inhibit DNA topoisomerase I by stabilizing the covalent
`complex ("cleavable complex") of enzyme and strand-
`cleaved DNA. Inhibition of topoisomerase I by camptoth(cid:173)
`ecin induces protein-associated DNA single-stran breaks
`which occur during the S-phase of the cell cycle. Since the
`S-phase is relatively short compared to other phases of the
`cell cycle, longer exposure to camptothecin should result in
`increased cytotoxicity of tumor cells. Studies indicate that
`only the closed lactone form of the drug helps stabilize the
`cleavable complex, leading to inhibition of the cell cycle and
`apoptosis.
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1021-0018
`
`

`

`US 2004/0162263 Al
`
`Aug. 19, 2004
`
`[0028] To preserve the lactone form of camptothecin,
`camptothecin and its water insoluble derivatives have been
`dissolved in N-methyl-2-pyrrolidinone in the presence of an
`acid (U.S. Pat. No. 5,859,023). Upon dilution with an
`acceptable parenteral vehicle, a stable solution of camptoth(cid:173)
`ecin was obtained. The concentrated solution of camptoth(cid:173)
`ecin was also filled in gel capsules for oral administration.
`It is believed that such formulations increase the amount of
`lipophilic lactone form of camptothecin that diffuse through
`the cellular and nuclear membranes in tumor cells.
`
`SUMMARY OF THE INVENTION
`
`[0029] The present invention provides innovative oral
`formulations of pharmaceuticals with enhanced bioavail-
`ability by targeting specific regions of the gastrointestinal
`tract. Particularly, water soluble and acid-labile drugs such
`as cytidine analogs (e.g., decitabine and 5'-azacytidine) and
`2'-deoxyadenosine analogs (e.g., pentostatin) are formulated
`with pH-sensitive polymers so that these drugs are prefer(cid:173)
`ably absorbed in the upper regions of the small intestine,
`such as the jejunum. In addition, drugs with poor oral
`bioavailability such as camptothecin compounds (e.g., 9-ni-
`tro-camptothecin) can also be formulated using similar
`strategies in order to significantly improve their oral bio(cid:173)
`availability.
`
`[0030] In one aspect of the invention, a pharmaceutical
`composition is provided. The pharmaceutical composition
`comprises: a water-soluble, acid-labile drug enteric-coated
`with a coating material that dissolves at pH above about 5.2.
`[0031] According to the invention, the solubility of the
`drug is preferably above 1 mg/ml in water or aqueous
`solution, more preferably above 5 mg/ml in water or aque(cid:173)
`ous solution, and most preferably above 10 mg/ml in water
`or aqueous solution.
`[0032] Also according to the invention, the drug is labile
`preferably at pH lower than 5.0, more preferably at pH lower
`than 4.0, and most preferably at pH lower than 2.0.
`[0033] Examples of the drug includes, but are not limited
`to, cytidine analogs or derivatives such as 5-azacytidine and
`5-aza-2'-deoxycytidine (or decitabine), and 2'-deoxyadenos-
`ine analogs and derivatives such as 2'-deoxycoformycin (or
`pentostatin), fludarabine monophosphate, and 2-chloro-2'-
`deoxyadenosine (or cladribine).
`
`[0034] The coating material for enteric-coating of the drug
`is pH-sensitive and preferably or selectively dissolves at a
`threshold pH above about 5.2, optionally at pH above about
`5.5, optionally at pH above about 5.8, optionally at pH above
`about 6.0, optionally at pH above about 6.2, optionally at pH
`above about 6.5, optionally at pH above about 6.5, and most
`preferably at pH above about 6.8, or optionally at pH above
`about 7.0. The pharmaceutical composition is preferred to
`substantially disintegrate in an aqueous medium at a pH
`equal or above the threshold pH within 3 hours, optionally
`within 2 hours, optionally within 1 hour, more preferably
`within 30 min, and most preferably within 15 mm.
`
`[0035] Examples of such a coating material include, but
`are not limited to, cellulose phthalates that selectively dis(cid:173)
`solve at pH above 5.6, the Eudragit® family of polymers
`(e.g., Eudragit L30D with threshold pH of 5.6, Eudragit L
`with threshold pH of 6.0, and Eudragit S with threshold pH
`of 6.8), Aquateric with threshold pH of 5.8, polyvinylacetate
`
`phthalate (PVAP) that releases drug at pH values above
`about 5.0, Shellac® that releases the drug at about pH7.0,
`and cellulose acetate phthalate (CAP) with threshold pH of
`6.0.
`[0036] In a preferred embodiment, the drug is enteric-
`coated with Eudragit L100 with the threshold pH of 6.0 or
`L100-55 with a threshold pH of 5.5.
`[0037] Also according to the invention, the pharmaceuti(cid:173)
`cal composition is preferred not to substantially disintegrate
`in an acidic, aqueous medium at pH 1.0-3.0 for at least 1
`hour, more preferred not to substantially disintegrate in an
`acidic, aqueous medium at pH 1.2-2.0 for at least 1 hour,
`more preferably for at least 2 hours, and most preferably for
`at least 3 hours. Optionally, the pharmaceutical formulation
`does not substantially disintegrate in an acidic, aqueous
`medium at pH 1.2-1.5 for at least 1 hour, more preferably for
`at least 2 hours, and most preferably for at least 3 hours. The
`composition is considered to be substantially disintegrated if
`at least 50% of the composition disintegrates, e.g., under(cid:173)
`goes rupture.
`
`[0038] In addition, the pharmaceutical composition pref(cid:173)
`erably disintegrates substantially in an aqueous medium at
`pH 5.2-7.5 within 1 hour, more preferably disintegrates
`substantially in an aqueous medium at pH 6.0-7.2 within 30
`minutes, and most preferably disintegrates substantially in
`an aqueous medium at pH 6.5-7.0 within 15 minutes.
`
`[0039] The amount of the enteric-coating material is pref(cid:173)
`erably 1-10% w/w in the composition, more preferably
`2-8% w/w in the composition, and most preferably 3-6%
`w/w in the composition.
`[0040] The pharmaceutical composition may be in a form
`of tablet or capsule. In a preferred embodiment, the com(cid:173)
`position is in a form of tablet. The hardness of the tablet
`without the enteric-coat is preferably at least 4 kp, more
`preferably at least 8 kp, and most preferably 10 kp. The size
`of the tablet is preferably 5-20 mm, more preferably 8-15
`mm, and most preferably 10-13 mm.
`
`[0041] In any of the above dosage forms, the concentra(cid:173)
`tion of the drug is preferably 0.1-20% w/w, optionally
`1-10% w/w, or optionally 2-5% w/w.
`[0042] Optionally, the pharmaceutical composition may
`further comprise a seal-coating material that seals the drug
`to prevent decomposition due to exposure to moisture, such
`as hydroxy propylmethylcellulose. Optionally, the pharma(cid:173)
`ceutical composition may further comprise buffer salt such
`as potassium or sodium phosphate in an amount sufhcient to
`maintain the pH of the local environment to be 5.2-7.0 when
`the pharmaceutical composition is dissolved in the GI tract.
`Examples of such buffer salts include, but are not limited to,
`KH2P04 and Na2HP04.
`
`[0043]
`In another aspect of the invention, a pharmaceuti(cid:173)
`cal composition for delivering a camptothecin compound in
`vivo is provided. The pharmaceutical composition com(cid:173)
`prises: a camptothecin compound enteric-coated with an
`enteric coating material that dissolves at pH above 5.2.
`
`[0044] The enteric coating material for enteric-coating of
`the camptothecin compound is pH-sensitive and preferably
`or selectively dissolves at pH above about 5.2, preferably at
`pH above about 5.8, more preferably at pH above about 6.0,
`and most preferably at pH above about 6.4.
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1021-0019
`
`

`

`US 2004/0162263 Al
`
`Aug. 19, 2004
`
`[0045] The enteric coating material for enteric-coating of
`the drug is pH-sensitive and preferably or selectively dis(cid:173)
`solves at a threshold pH above about 5.2, optionally at pH
`above about 5.5, optionally at pH above about 5.8, option(cid:173)
`ally at pH above about 6.0, optionally at pH above about 6.2,
`optionally at pH above about 6.5, optionally at pH above
`about 6.5, and most preferably at pH above about 6.8,
`optionally at pH above about 7.0, optionally at pH above
`about 7.2, or optionally at pH above about 7.5. The phar(cid:173)
`maceutical composition is preferred to substantially disin(cid:173)
`tegrate in an aqueous medium at a pH equal or above the
`threshold pH within 3 hours, optionally within 2 hours,
`optionally within 1 hour, more preferably within 30 min, and
`most preferably within 15 min.
`
`[0046] Examples of such a coating material include, but
`are not limited to, cellulose phthalates that selectively dis(cid:173)
`solve at pH above 5.6, the Eudragit® family of polymers
`(e.g., Eudragit L30D with threshold pH of 5.6, Eudragit L
`with threshold pH of 6.0, and Eudragit S with threshold pH
`of 6.8), Aquateric with threshold pH of 5.8, polyvinylacetate
`phthalate (PVAP) that releases drug at pH values above
`about 5.0, Shellac® that releases the drug at about pH7.0,
`and cellulose acetate phthalate (CAP) with threshold pH of
`6.0.
`
`[0047]
`In a preferred embodiment, the drug is enteric-
`coated with Eudragit L100 with the threshold pH of 6.0 or
`Eudragit L100-55 with a threshold pH of 5.5.
`[0048] Also according to the invention, the pharmaceuti(cid:173)
`cal composition is preferred not to substantially disintegrate
`in an acidic, aqueous medium at pH 1.0-3.0 for at least 1
`hour, more preferred not to substantially disintegrate in an
`acidic, aqueous medium at pH 1.2-2.0 for at least 1 hour,
`more preferably for at least 2 hours, and most preferably for
`at least 3 hours. Optionally, the pharmaceutical formulation
`does not substantially disintegrate in an acidic, aqueous
`medium at pH 1.2-1.5 for at least 1 hour, more preferably for
`at least 2 hours, and most preferably for at least 3 hours. The
`composition is considered to be substantially disintegrated if
`at least 50% of the composition disintegrates, e.g., under(cid:173)
`goes rupture.
`
`[0049]
`In addition, the pharmaceutical composition pref(cid:173)
`erably disintegrates substantially in an aqueous medium at
`pH 5.2-7.5 within 1 hour, more preferably disintegrates
`substantially in an aqueous medium at pH 6.0-7.2 within 30
`minutes, and most preferably disintegrates substantially in
`an aqueous medium at pH 6.5-7.0 within 15 minutes.
`
`[0050] The amount of the enteric-coating material is pref(cid:173)
`erably 1-10% w/w in the composition, more preferably
`2-8% w/w in the composition, and most preferably 3-6%
`w/w in the composition.
`
`[0051] The camptothecin compound may be the original
`20(S)-camptothecin isolated from the plant, Camptotheca
`acuminata, analogs of 20(S)-camptothecin, derivatives of
`20(S)-camptothecin, prodrugs of 20(S)-camptothecin, and
`pharmaceutically active metabolites of 20(S)-camptothecin.
`
`[0052] Examples of camptothecin derivatives include, but
`are not limited to, 9-nitro-20(S)-camptothecin, 9-amino-
`20(S)-camptothecin,
`9-methyl-camptothecin,
`9-chloro-
`camptothecin, 9-flouro-camptothecin, 7-ethyl camptothecin,
`10-methyl-camptothecin,
`10-chloro-camptothecin,
`10-bromo-camptothecin, 10-fluoro-ca

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