throbber
COVER SHEET FOR PROVISIONAL APPLICATION FOR PATENT
`
`Commissioner for Patents
`P.O. Box [450
`Alexandria, VA 22313-1450
`Sir:
`
`Docket Number
`
`INVENTOR(s) APPLICANT(s)
`
`
`
`LAST NAME MIDDLE INITIAL|RESIDENCE (CITY AND EITHER STATE OR FOREIGN COUNTRY)FIRST NAME
`
`
`
`This is a request for filing a PROVISIONAL APPLICATIONunder 37 CFR 1.53(c).
`
`
`95 16-825-888
`inside this box 6
`Type a plus sign (+)
`
`
`
`
`
`
`
`
`
`Backstrom
`Etter
`Lai
`
`Jay
`Jeff
`Mei
`
`T
`B
`
`Boulder, CO
`Boulder, CO
`Longmont, CO
`
`
`TITLE OF THE INVENTION (280 characters max)
`
`ORAL DOSAGE FORMS OF CYTIDINE ANALOGS AND METHODSOF USE THEREOF
`
`JONES DAY
`CORRESPONDENCE ADDRESS
`
`ENCLOSED APPLICATION PARTS(check all that apply)
`
` EX Specification
`(J Applicant claims small entity status, see 37 CFR §1.27
`Number ofPages
`
`Ed Drawing(s)
`
`Number ofSheets ([]_ Other(specity)
`
`
`EXTRA
`EXTRA RATE
`TYPE
`NOQ. FILED
`
`
`
`
`Total Numberof
`
`96 * 0.75 = 72
`Pages w/ drawings
`
`Basic Filing Fee
`
`
`
`
`
` Total Filing Fee
`220,00
`
`[J] A check or moneyorderis enclosed to cover the Provisionalfiling fees.
`
`$270.00 for each 50 pages
`over 100
`
`$
`
`FEE
`
`Applicant qualifies for the 50% Reduction for Independent inventor, Nonprofit Organization or Small Business
`Concem
`
`Total
`
`0.00
`220.00
`
`220.00
`
`
`
`&] The abovecalculation is an estimate ofthe fees due. The fees will be paid via EFS-Web. Please charge any
`additional fees to Jones Day Deposit Account No. 50-3013, if required.
`
`The invention was made by an agency of the United States Government or under a contract with an agency of the United States Government.
`BI No.
`[] Yes, the name ofthe U.S. Government agency and the Government contract numberare:
`
`Respecifully submitted,
`
`Signature
`
`
`
`/
`
`Robert’ Chang
`3.753) REGISTRATION NO.
`for Anthony M.Insog
`(ifappropriate}
`
`JONES DAY
`
`ao
`
`5
`35,203
`
`Dat
`ate
`
`March 5, 2009
`
`
`
`[J Additional inventors are being named on separately numbered sheets attached hereto.
`
`Total number of cover sheet pages.
`PROVISIONAL APPLICATION FILING ONLY
`
`LAI-3007509v1
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1050-0001
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1050-0001
`
`

`

`ORAL DOSAGE FORMSOF CYTIDINE ANALOGS
`AND METHODS OF USE THEREOF
`
`I. FIELD
`
`[0001]
`
`Provided herein are pharmaceutical dosage forms comprising cytidine
`
`analogs, or their salts, solvates, hydrates, precursors, and/or derivatives thereof, for oral
`
`administration in subjects. Also provided are methods for making the dosage forms and
`
`for using the dosage formsto treat cancer and disorders related to abnormalcell
`
`proliferation.
`
`il. BACKGROUND
`
`[8002]
`
`Cancer is a major worldwide public health problem; in the United States
`
`alone, approximately 570,000 cancer-related deaths were expected in 2005. See, e.g.,
`
`Jemal et al., CA Cancer J. Clin. 55(1):10-30 (2005). Many types of cancer have been
`
`described in the medical literature. Examples include cancer of the blood, bone, lung
`
`(e.g., non-small-cell lung cancer and small-cell lung cancer), colon, breast, prostate,
`
`ovary, brain, and intestine. The incidence of cancer continues to climb as the general
`
`population ages and as new forms of cancer develop. A continuing need exists for
`
`effective therapies to treat subjects with cancer.
`
`{0003}
`
`Myelodysplastic syndromes (MDS)refers to a diverse group of
`
`hematopoietic stem cell disorders. MDS affects approximately 40,000-50,000 people in
`
`the U.S. and 75,000-85,000 subjects in Europe. MDS may be characterized by a cellular
`
`marrow with impaired morphology and maturation (dysmyelopoiesis), peripheral blood
`
`cytopenias, and a variable risk of progression to acute leukemia, resulting from
`
`ineffective blood cell production. See, e.g., The Merck Manual 953 (17th ed. 1999); List
`
`et al., J. Clin. Oncol. 8:1424 (1990).
`
`[0004]
`
`MDSare grouped together because of the presence of dysplastic changes in
`
`one or more ofthe hematopoietic lineages including dysplastic changes in the myeloid,
`
`erythroid, and megakaryocytic series. These changes result in cytopenias in one or more
`
`of the three lineages. Subjects afflicted with MDS may develop complications related to
`
`anemia, neutropenia (infections), and/or thrombocytopenia (bleeding). From about 10%
`
`to about 70% of patients with MDS may develop acute leukemia. In the early stages of
`
`MDS,the main cause of cytopenias is increased programmedcell death (apoptosis). As
`
`the disease progresses and converts into leukemia, a proliferation of leukemic cells
`
`NY1-4164551v1
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1050-0002
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1050-0002
`
`

`

`overwhelms the healthy marrow. The disease course differs, with some cases behaving
`as an indolent disease and others behaving aggressively with a very short clinical course
`
`that converts into an acute form of leukemia. The majority of people with higher risk
`
`MDSeventually experience bone marrow failure. Up to 50% of MDSpatients succumb
`
`to complications, such as infection or bleeding, before progressing to AML.
`[0005]
`Aninternational group of hematologists, the French-American-British (FAB)
`Cooperative Group, classified MDS into five subgroups, differentiating them from acute
`
`myeloid leukemia. See, e.g., The Merck Manual 954 (17th ed. 1999); Bennett J. M., e¢
`
`al., Ann. Intern. Med., 103(4): 620-5 (1985); and Besa E. C., Med. Clin. North Am.
`
`76(3): 599-617 (1992). An underlying trilineage dysplastic change in the bone marrow
`
`cells of the patients is foundin all subtypes. Information is available regarding the
`pathobiology of MDS,certain MDSclassification systems, and particular methods of
`treating and managing MDS. See, e.g., U.S. Patent No. 7,189,740 (issued March13,
`
`2007), which is incorporated by reference herein in its entirety.
`
`[0006]
`
`Nucleoside analogs have been used clinically for the treatmentofviral
`
`infections and cancer. Most nucleoside analogsare classified as anti-metabolites. After
`
`they enter the cell, nucleoside analogs are successively phosphorylated to nucleoside 5’-
`
`mono-phosphates, di-phosphates, and tri-phosphates. In most cases, nucleosidetri-
`phosphatesare the chemical entities that inhibit DNA or RNA synthesis, either through
`competitive inhibition of polymerases or through incorporation of the modified
`
`nucleotides into DNA or RNA sequences. Nucleosides mayalso act as di-phosphates.
`[0007]
`5-Azacytidine (National Service Center designation NSC-102816; CAS
`
`Registry Number 320-67-2), also known as azacitidine, AZA, or 4-amino-I-B-D-
`ribofuranosyl-1,3,5-triazin-2(1H)-one, is currently marketed as the drug product
`VIDAZA®, 5-Azacytidineis a nucleoside analog, more specifically a cytidine analog.
`5-Azacytidine is an antagonistofits related natural nucleoside, cytidine. 5-Azacytidine
`and 5-aza-2'-deoxycytidine (also known as decitabine, an analog of deoxycytidine) are
`also antagonists of deoxycytidine. A structural difference between these cytidine
`analogsandtheir related natural nucleoside is the presence ofa nitrogen at position 5 of
`the cytosine ring in place of a carbon. 5-Azacytidine may be defined as having the
`molecular formula CgHi2N4Qs, a molecular weight of 244.21 grams per mole, and the
`following structure:
`
`NYI-4164551v1
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1050-0003
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1050-0003
`
`

`

`HO
`
`OH
`
`OH
`
`5-Azacytidine.
`
`[0008]
`
`Other membersofthe class of cytidine analogs include, for example: 1-B-D-
`
`arabinofuranosylcytosine (Cytarabine or ara-C); 5-aza-2'-deoxycytidine (Decitabine or
`
`5-aza-CdR); pseudoisocytidine (psi ICR); 5-fluoro-2'-deoxycytidine (FCdR); 2'-deoxy-
`
`2',2'-difluorocytidine (Gemcitabine); 5-aza-2'-deoxy-2',2'-difluorocytidine; 5-aza-2'-
`
`deoxy-2'-fluorocytidine; 1-B-D-ribofuranosyl-2(1 H)-pyrimidinone (Zebularine); 2',3'-
`
`dideoxy-5-fluoro-3'-thiacytidine (Emtriva); 2'-cyclocytidine (Ancitabine); 1-B-D-
`
`arabinofuranosyl-5-azacytosine (Fazarabine or ara-AC); 6-azacytidine (6-aza-CR); 5,6-
`dihydro-5-azacytidine (dH-aza-CR); N‘-pentyloxycarbony]-5'-deoxy-5-fluorocytidine
`(Capecitabine); N*-octadecyl-cytarabine; and elaidic acid cytarabine.
`[0009]
`After its incorporation into replicating DNA, 5-azacytidine or 5-aza-2'-
`
`deoxycytidine forms a covalent complex with DNA methyltransferase. DNA
`
`methyltransferase is responsible for reproducing the methylation patterns in the daughter
`
`strands. Inhibition of DNA methyltransferase by a cytidine analog leads to DNA
`
`hypomethylation, thereby reestablishing the anti-proliferative signals extinguished by
`
`DNAhypermethylation in malignant cells, such as morphologically dysplastic and
`
`immature hematopoietic cells. The cytotoxic effects of these cytidine analogs cause the
`
`death of rapidly dividing cells, including cancercells, that are no longer responsive to
`
`normal cell growth control mechanisms.
`
`[0610]
`
`5-Azacytidine and 5-aza-2'-deoxycytidine have been tested in clinicaltrials
`
`and showedsignificant anti-tumor activity, such as, for example, in the treatment
`
`myelodysplastic syndromes (MDS), acute myelogenous leukemia (AML), chronic
`
`myelogenous leukemia (CML), acute lymphocytic leukemia (ALL), and non Hodgkin’s
`
`lymphoma (NHL). See, e.g., Aparicio ef al., Curr. Opin. Invest. Drugs 3(4): 627-33
`
`(2002). 5-Azacytidine has undergone NCI-sponsoredtrials for the treatment of MDS
`
`and has been approved fortreating all FAB subtypes of MDS. See, ¢.g., Kornblith e¢ a/.,
`
`J Clin. Oncol. 20(10): 2441-2452 (2002); Silverman ef al., J. Clin. Oncol. 20(10): 2429-
`
`NYL-4164551v1
`
`-3-
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1050-0004
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1050-0004
`
`

`

`2440 (2002). 5-Azacytidine may alter the natural course of MDS bydiminishing the
`
`transformation to AMLthrough its cytotoxic activity and its inhibition of DNA
`
`methyltransferase.
`
`In a Phase III study, 5-azacytidine significantly prolonged survival
`
`and time to AMLtransformation or death in elderly subjects. See, e.g., Silverman et al,
`
`Blood 106(11): Abstract 2526 (2005).
`
`[OOL1]
`
`5-Azacytidine and other cytidine analogs are approved for subcutaneous (SC)
`
`or intravenous (1V) administration to treat various proliferative disorders. Oral dosing of
`
`cytidine analogs would be more desirable and convenient for patients and doctors, e.g..
`
`byeliminating injection-site reactions that may occur with SC administration and/or by
`
`permitting improved patient compliance. However, oral delivery of cytidine analogs has
`
`proven difficult due to combinations of chemical instability, enzymatic instability,
`
`and/or poor permeability. For example, cytidine analogs have been considered acid
`
`labile and unstable in the acidic gastric environment. Previous attempts to develop oral
`
`dosage forms of cytidine analogs have required enteric coating of the drug core to
`
`protect the active pharmaceutical ingredient (API) from what was understood and
`
`accepted to be therapeutically unacceptable hydrolysis in the stomach, such that the drug
`
`is preferably absorbed in specific regions of the lower gastrointestinal tract, such as the
`
`jejunum in the small intestine. See, e.g., Sands, et al., U.S. Patent Publication No.
`
`2004/0162263 (App. No. 10/698,983). In addition, a generally accepted beliefin the art
`
`has been that water leads to detrimental hydrolytic degradation of cytidine analogs
`
`during formulation, subsequently affecting the stability of the API in the dosage form.
`
`As a result, coatings applied to the drug core for prospective oral delivery of cytidine
`
`analogs have previously been limited to organic solvent-based systems to minimize
`
`exposure of the API to water.
`
`[0012]
`
`A great need remainsfor oral formulations and dosage forms of cytidine
`
`analogs, such as, ¢.g., 5-azacytidine, to potentially permit, inter alia, more advantageous
`
`dosing amounts or dosing periods; improved pharmacokinetic profiles,
`
`pharmacodynamicprofiles, or safety profiles; evaluation of the benefits of long-term or
`
`maintenance therapies; development of improved treatment regimens that maximize
`
`demethylation or gene re-expression; use of cytidine analogs for treating new diseases or
`
`disorders; and/or other potential advantageous benefits.
`
`NY1-4164551vI
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1050-0005
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1050-0005
`
`

`

`LL SUMMARY
`
`{0013}
`
`Provided herein are pharmaceutical formulations comprising cytidine
`
`analogs, wherein the formulations release the API substantially in the stomach uponoral
`
`administration. Also provided are methods for making the formulations, and methods
`
`for using the formulations to treat cancer and disorders related to abnormalcell
`
`proliferation.
`
`[0014]
`
`In certain embodiments, the cytidine analog is 5-azacytidine.
`
`In other
`
`embodiments, the cytidine analog is 5-aza-2'-deoxycytidine (decitabine or 5-aza-CdR).
`
`In yet other embodiments, the cytidine analog is, for example: 1-B-D-
`
`arabinofuranosylcytosine (Cytarabine or ara-C); pseudoisocytidine (psi ICR); 5-fluoro-
`
`2'-deoxycytidine (FCdR); 2'-deoxy-2',2'-difluorocytidine (Gemcitabine); 5-aza-2'-deoxy-
`
`2',2'-difluorocytidine; 5-aza-2'-deoxy-2'-fluorocytidine; |-B-D-ribofuranosyl-2(1 H)-
`
`pyrimidinone (Zebularine); 2',3'-dideoxy-5-fluoro-3'-thiacytidine (Emtriva); 2'-
`
`cyclocytidine (Ancitabine); 1-B-D-arabinofuranosyl-5-azacytosine (Fazarabine or ara-
`
`AC); 6-azacytidine (6-aza-CR); 5,6-dihydro-5-azacytidine (dH-aza-CR);
`N‘-pentyloxycarbonyl-5'-deoxy-5-fluorocytidine (Capecitabine); N*-octadecyl-
`
`cytarabine; elaidic acid cytarabine; or their derivatives or related analogs.
`
`(0015)
`
`Certain embodiments herein provide formulations that are single unit dosage
`
`forms comprising a cytidine analog. Certain embodiments herein provide formulations
`
`that are tablets comprising a cytidine analog. Certain embodiments herein provide
`
`formulations that are capsules comprising a cytidine analog. The capsules may be,e.v.,
`
`a hard gelatin capsule or a soft gelatin capsule. In certain embodiments, the single unit
`
`dosage formsoptionally further contain one or more excipients.
`
`In certain
`
`embodiments, the tablets optionally further contain one or more excipients. In other
`
`embodiments, the capsules optionally further contain one or more excipients.
`
`In certain
`
`embodiments, the formulation is a tablet that effects an immediate release of the API
`
`upon oral administration.
`
`In other embodiments, the formulation is a tablet that effects a
`
`controlled release of the API substantially in the stomach. In certain embodiments, the
`
`formulation is a capsule that effects an immediate release of the API upon oral
`
`administration. In other embodiments, the formulation is a capsule that effects a
`
`controlled release of the API substantially in the stomach. In particular embodiments,
`
`the tablet contains a drug core that comprises a cytidine analog, and optionally further
`
`contains a coating of the drug core, wherein the coating is applied to the drug core using
`
`an aqueous solvent, such as, for example, water.
`
`NYI-4164551v1
`
`-5-
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1050-0006
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1050-0006
`
`

`

`[0016]
`
`Certain embodiments herein provide methods of making the formulations of
`
`cytidine analogs intended for oral delivery. Further provided are articles of manufacture
`
`containing packaging material, an oral formulation ofa cytidine analog, and a label that
`
`indicates that the formulation is for the treatment of certain disorders related to abnormal
`
`cell proliferation, such as, for example, a cancer.
`
`[0017]
`
`Certain embodiments herein provide methods of using the formulations
`
`provided herein to treat cancers or other disorders related to abnormal cell proliferation.
`
`In certain embodiments, the formulations of cytidine analogs are orally administered to
`
`subjects in need thereof to treat a cancer, including, but not limited to, a hematological
`
`disorder, such as, for example, MDS, AML, ALL, CML, NHL, leukemia, or lymphoma;
`
`or a solid tumor, such as, for example, sarcoma, melanoma, carcinoma, or cancer of the
`
`colon, breast, ovary, gastrointestinal system, kidney, lung (e.g., non-small-cell lung
`
`cancer and small-cell lung cancer), testicle, prostate, pancreas or bone. In certain
`
`embodiments, the oral formulations provided herein are co-administered with one or
`
`more therapeutic agents to provide a synergistic therapeutic effect in subjects in need
`
`thereof. The co-administered agents may be a cancer therapeutic agent, as described
`
`herein. In certain embodiments, the co-administered agent(s) may be dosed,e.g., orally
`
`or by injection.
`
`[0018]
`
`In particular embodiments, provided herein are tablets containing 5-
`
`azacytidine and methods for making and using the tablets to treat cancer and disorders
`
`related to abnormal cell proliferation. In certain embodiments, the tablets optionally
`
`further contain one or more excipients such as, for example, glidants, diluents,
`
`lubricants, colorants, disintegrants, granulating agents, binding agents, polymers, and/or
`
`coating agents. Examples of ingredients useful in preparing certain formulations
`
`provided herein are described in, e.g., Etter et al., U.S. Patent Application Publication
`
`No. 2008/0057086 (App. No. 11/849,958), which is incorporated herein by reference in
`
`its entirety.
`
`[0019]
`
`Specific embodiments herein provide, inter alia, pharmaceutical
`
`compositions comprising a therapeutically effective amount of 5-azacytidine, wherein
`
`the composition releases the 5-azacytidine substantially in the stomach following oral
`
`administration to a subject. Further embodiments provide the aforementioned
`
`compositions, which: are immediate release compositions; do not have an enteric
`
`coating; are tablets; are capsules; further comprise an excipient selected from any
`
`excipient disclosed herein; further comprise a permeation enhancer; further comprise d-
`
`NYI-416455l1v1
`
`-6-
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1050-0007
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1050-0007
`
`

`

`
`
`alpha-tocophery! polyethylene glycol 1000 succinate; further comprise a permeation
`
`enhancer in the formulation at about 2% by weight relative to the total weight of the
`
`formulation; are essentially free of a cytidine deaminase inhibitor; are essentially free of
`
`tetrahydrouridine; have an amount of 5-azacytidine ofat least about 40 mg; have an
`
`amount of 5-azacytidine ofat least about 400 mg; have an amount of 5-azacytidine ofat
`
`least about 1000 mg; achieve an area-under-the-curve value of at least about 200 ng-
`
`hr/mL following oral administration to a subject; achieve an area-under-the-curve value
`
`of at least about 400 ng-hr/mL following oral administration to a subject; achieve a
`
`maximumplasma concentration ofat least about 100 ng/mL following oral
`
`administration to a subject; achieve a maximum plasma concentration of at least about
`
`200 ng/mL following oral administration to a subject; achieve a time to maximum
`
`plasma concentration of less than about 90 minutes following oral administration to a
`
`subject; and/or achieve a time to maximum plasma concentration of less than about 60
`
`minutes following oral administration to a subject.
`
`[0020]
`
`Specific embodiments herein provide a pharmaceutical composition for oral
`
`administration comprising a therapeutically effective amount of 5-azacytidine, which
`
`releases the 5-azacytidine substantially in the stomach and achieves an area-under-the-
`
`curve value of at least about 200 ng-hr/mL following oral administration.
`
`[0021]
`
`Specific embodiments herein provide a pharmaceutical composition for oral
`
`administration comprising a therapeutically effective amount of 5-azacytidine, which
`
`releases the 5-azacytidine substantially in the stomach and achieves an area-under-the-
`
`curve value of at least about 400 ng-hr/mL following oral administration.
`
`[0022]
`
`Specific embodiments herein provide a pharmaceutical composition for oral
`
`administration comprising a therapeutically effective amount of 5-azacytidine, which
`
`releases the 5-azacytidine substantially in the stomach and achieves a maximum plasma
`
`concentration of at least about 100 ng/mL following oral administration.
`
`[0023]
`
`Specitic embodiments herein provide a pharmaceutical composition for oral
`
`administration comprising a therapeutically effective amount of 5-azacytidine, which
`
`releases the 5-azacytidine substantially in the stomach and achieves a maximum plasma
`
`concentration of at least about 200 ng/mL following oral administration.
`
`[0024]
`
`Specific embodiments herein provide a pharmaceutical composition for oral
`
`administration comprising a therapeutically effective amount of 5-azacytidine, which
`
`releases the 5-azacytidine substantially in the stomach and achieves a time to maximum
`
`plasmaconcentration of less than about 90 minutes following oral administration.
`
`NYI-4164551v1
`
`-7-
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1050-0008
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1050-0008
`
`

`

`
`
`[0025]
`
`Specific embodiments herein provide a pharmaceutical composition for oral
`
`administration comprising a therapeutically effective amount of 5-azacytidine, which
`
`releases the 5-azacytidine substantially in the stomach and achieves a time to maximum
`
`plasma concentration of less than about 60 minutes following oral administration.
`
`[0026]
`
`Specific embodiments herein provide any ofthe aforementioned
`
`compositions, as single unit dosage forms, tablets, or capsules.
`
`[0027]
`
`Specific embodiments herein provide, inter alia, methodsfor treating a
`
`subject having a disease associated with abnormalcell proliferation, comprising orally
`
`administering to the subject a pharmaceutical composition comprising a therapeutically
`
`effective amount of 5-azacytidine, wherein the composition releases the 5-azacytidine
`
`substantially in the stomach following oral administration to the subject. Further
`
`embodiments herein provide the aforementioned methods, in which:
`
`the disease is
`
`myelodysplastic syndrome; the disease is acute myelogenous leukemia; the method
`
`further comprises co-administering to the subject in need thereof an additional
`
`therapeutic agent selected from any additional therapeutic agent disclosed herein: the
`
`composition is an immediate release composition; the composition does not have an
`
`enteric coating; the composition further comprises a permeation enhancer; the
`
`composition further comprises the permeation enhancer d-alpha-tocopheryl polyethylene
`
`glycol 1000 succinate; the composition further comprises d-alpha-tocopheryl
`
`polyethylene glycol 1000 succinate in the formulation at about 2% by weightrelative to
`the total weight of the formulation; the method further comprises not co-administering a
`cytidine deaminase inhibitor with the cytidine analog; the composition is a single unit
`
`dosage form; the compositionis a tablet; the composition is a capsule; the composition
`
`further comprises an excipient selected from any excipient disclosed herein; the amount
`
`of 5-azacytidineis at least about 40 mg; the amountof5-azacytidine is at least about 400
`
`mg; the amountof 5-azacytidine is at least about 1000 mg; the method achieves an area-
`
`under-the-curve value of at least about 200 ng-hr/mL following oral administration to
`
`the subject; the method achieves an area-under-the-curve value ofat least about 400 ng-
`hr/mL following oral administration to the subject; the method achieves a maximum
`
`plasma concentration of at least about 100 ng/mL following oral administration to the
`
`subject; the method achieves a maximum plasma concentration ofat least about 200
`
`ng/mL following oral administration to the subject; the method achieves a time to
`
`maximum plasma concentration ofless than about 90 minutes following oral
`
`NY1-4164551v1
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1050-0009
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1050-0009
`
`

`

`
`
`administration to the subject; and/or the method achieves a time to maximum plasma
`concentration ofless than about 60 minutes following oral administration to the subject
`
`[0028]
`
`Specific embodiments herein provide, inter alia, pharmaccutical
`
`compositions comprising a therapeutically effective amount of 5-azacytidine, wherein
`
`the compositions are for treating a disease or disorder associated with abnormalcell
`
`proliferation, wherein the compositions are prepared for oral administration, and
`
`wherein the compositions are prepared for release of the 5-azacytidine substantially in
`the stomach. Further embodiments herein provide the aforementioned compositions,
`
`which: have an amount of5-azacytidine of about 40 mg, about 400 mg, or about 1000
`
`mg; are prepared to achieve an area-under-the-curve value of at least about 200 ng-
`hr/mL or 400 ng-hr/mL following oral administration: are prepared to achieve a
`
`maximum plasma concentration ofat least about 100 ng/mL or 200 ng/mL following
`oral administration; are prepared to achieve a time to maximum plasma concentration of
`
`less than about 60 minutes or 90 minutes after being administered; are prepared in the
`
`form of an immediate release composition; are prepared for oral administration in
`
`combination with an additional therapeutic agent selected from any additional
`
`therapeutic agent disclosed herein; are for treating myelodysplastic syndromeor acute
`
`myelogenous leukemia; further comprise a permeation enhancer; which further comprise
`the permeation enhancer d-alpha-tocopheryl polyethylene glycol 1000 succinate; are
`
`single unit dosage forms: are tablets or capsules; and/or further comprise an excipient
`
`selected from any excipient disclosed herein.
`
`[0029]
`Specific embodiments herein provide, inter alia, uses of 5-azacytidine and,
`optionally, a permeation enhancer, for the preparation of a pharmaceutical composition
`for treating a disease associated with abnormalcell proliferation, wherein the
`
`composition is prepared for oral administration, and wherein the composition is prepared
`for release of the 5-azacytidine substantially in the stomach. Further embodiments
`
`herein provide the aforementioned uses, in which:
`
`the disease is myelodysplastic
`
`syndrome or acute myelogenous leukemia; the amount of 5-azacytidine is selected from
`
`any amountdisclosed herein; and/or the composition is prepared for immediate release.
`
`NY1I-4164551 v1
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1050-0010
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1050-0010
`
`

`

`IV. BRIEF DESCRIPTION OF THE DRAWINGS
`
`[0030]
`
`Figure 1 represents processes and steps that may be used to make particular
`
`tablets comprising azacitidine for oral dosing.
`[0031]
`Figure 2 represents human PK profiles following 75 mg/m* SC dosing of
`azacitidine on Days 1 and 7 in a multiple dose escalation study (n = 18). The X-axis
`
`represents time; the Y-axis represents azacitidine plasma concentrations (mean + SD).
`|0032|
`Figure 3 represents human PK profiles following SC (75 mg/m’) and PO
`(240 mg, 300 mg, and 360 mg) dosing ofazacitidine in a multiple dose escalation study.
`
`The azacitidine plasma PK profiles are compared among various doses. The X-axis
`
`represents time; the Y-axis represents azacitidine plasma concentrations (mean + SD).
`
`[0033]
`
`Figure 4 represents PD data from anindividual patient (Subject 02008, 80
`
`year old male, RAEB-1) collected during a multiple dose escalation study. The patient
`
`was dosed with azacitidine Formulation #3, 240 mg. Platelets (K/uL), Hgb (g/dL), ANC
`
`(K/uL), and Relative BM Blast (%) are plotted versus sampling dates over the course of
`
`the study.
`
`[0034]
`
`Figure 5 represents PD data from an individual patient (Subject 02007, 76
`
`year old male, CMML)collected during a multiple dose escalation study. The patient
`
`was dosed with azacitidine Formulation #3, 240 mg. Platelets (K/uL), Hgb (g/dL), ANC
`
`(K/L), and Relative BM Blast (°%) are plotted versus sampling dates over the course of
`
`the study.
`
`[0035]
`
`Figure 6 represents PD data froman individual patient (Subject 02004, 61
`
`year old male, MDS, MDACC)collected during a multiple dose escalation study. The
`
`patient was dosed with azacitidine Formulation 1, 120 mg. Platelets (K/uL), Hgb
`
`(g/dL), ANC (K/pL), and Relative BM Blast (%) are plotted versus sampling dates over
`
`the course of the study.
`
`[0036]
`
`Figure 7 represents a study design of a Rapid Aza Clinical Evaluation
`
`(RACE) study. Doses given on various days within a treatment cycle are depicted.
`
`Dose may be administered + 1 day, as long as there is at least 48 hours between doses.
`
`Figure 8 represents azacitidine human PKprofiles from an individual patient
`[0037]
`(Subject 106003, N = 1) following SC (124 mg, 75 mg/m’) and PO (180 mg, 360 mg,
`1,200 mg, Formulation 4) dosing of azacitidine from a RACEclinical study. AUC(0-t)
`
`values for the SC and PO doses are depicted.
`
`Figure 9 represents azacitidine human PK profiles from an individual patient
`[0038]
`(Subject 106004, N = 1) following SC (120 mg, 75 mg/m’) and PO (180 mg, 360 mg,&
`
`NYI-4164551v1
`
`-10-
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1050-0011
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1050-0011
`
`

`

`1,200 mg, Formulation 6) dosing of azacitidine from a RACEclinical study. AUC(0-0)
`
`values for the SC and PO doses are depicted.
`
`[0039]
`
`Figure 10 represents human PK profiles (linear scale) following SC and oral
`
`administration of azacitidine in clinical studies.
`
`[0040]
`
`Figure 11 represents human PK profiles (semi-log scale) following SC and
`
`oral administration of azacitidine in clinical studies.
`
`[0041]
`
`Figure 12 represents human AUC values following SC dosing of azacitidine
`
`and oral dosing ofazacitidine with Formulations #3, #4, and #6 at various dosage levels
`
`in clinical studies.
`
`[0042]
`
`Figure 13 represents human Cmax valuesin patients following SC dosing of
`
`azacitidine and oral dosing of azacitidine with Formulations #3, #4, and #6 at various
`
`dosage levels in clinical studies.
`
`[0043]
`
`Figure 14 represents relative oral bioavailability in humans following oral
`
`dosing of azacitidine with Formulations #3, #4, and #6 at various dosage levels.
`
`[0044]
`
`Figure 15 represents percent exposure in humansrelative to SC
`
`administration following oral dosing of azacitidine with Formulations #3, #4, and #6 at
`
`various dosage levels.
`
`[0045]
`
`Figure 16 represents a plot of percent methylated CpG loci in the blood DNA
`
`from an individual patient dosed with azacitidine in a MTD study.
`
`[0046]
`
`Figure 17 represents a plot of percent highly methylated CpG loci in the
`
`blood DNA from an individual patient dosed with azacitidine in a MTD study.
`
`Vv. DETAILED DESCRIPTION
`
`[0047]
`
`Unless defined otherwise, all technical and scientific terms used herein have
`
`the same meaning as commonly understood byone ofordinary skill in the art. All
`
`publications and patents referred to herein are incorporated by reference herein in their
`
`entireties.
`
`5.1 Definitions
`
`[0048]
`
`As used in the specification and the accompanying claims, the indefinite
`
`articles “‘a” and “an” and the definite article “the” include plural as well as singular
`
`referents, unless the context clearly dictates otherwise.
`
`[0049]
`
`The term “about” or “approximately” means an acceptable error for a
`
`particular value as determined byone of ordinary skill in the art, which depends in part
`
`on how the value is measured or determined. In certain embodiments, the term “about”
`
`NY1-4164551v1
`
`Apotexv.Cellgene -IPR2023-00512
`Petitioner Apotex Exhibit 1050-0012
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1050-0012
`
`

`

`or “approximately” means within 1, 2, 3, or 4 standard deviations. In certain
`
`embodiments, the term “about” or “approximately” means within 30%, 25%, 20%, 15%,
`
`10%, 9%, 8%, 7%, 6%, 5%, 4%, 3%, 2%, 1%, 0.5%, 0.1%, or 0.05% of a given value or
`
`range.
`
`As used herein, and unless otherwise specified, the terms “treat,” “treating”
`[0050]
`and “treatment” refer to the eradication or amelioration of a disease or disorder, or of
`
`one or more symptomsassociated with the disease or disorder. In certain embodiments,
`
`the terms refer to minimizing the spread or worsening ofthe disease or disorder resulting
`from the administration of one or more prophylactic or therapeutic agents to a subject
`with such a disease or disorder. In some embodiments, the terms refer to the
`
`administration of a compound or dosage form provided herein, with or without one or
`
`more additional active agent(s), a

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