throbber
8208264
`
`March 18, 2022
`
`THIS IS TO CERTIFY THAT ANNEXED IS A TRUE COPY FROM THE
`RECORDS OF THIS OFFICE OF THE FILE WRAPPER AND CONTENTS
`OF:
`
`APPLICATION NUMBER: 12/466,213
`FILING DATE: May 14, 2009
`PATENT NUMBER: 8846628
`ISSUE DATE: September 30, 2014
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1022-0001
`
`

`

`PTO/SB/05 (07-07
`Approved for use through 06/30/2010. B 0651-0032
`Patent and Trademark Office: U.S. DEPARTMENT OF COMMERCE
`Under the Paperwork Reduction Act of 1995, no persons are required to respond to a collection of information unlessit displays a valid OMB control
`number,
`
`
`
`UTILITY
`PATENT APPLICATION
`TRANSMITTAL
`(Onlyfor new nonprovisional applications under 37 CFR 1.33(bj)
`
`1. &
`
`48)
`
`(For information on the preferred arrangement, see MPEP 608.01 (aj)
`Drawing(s) (35 USC 13)
`[Total Sheets 20 }
`
`10.0
`1.0
`12.00
`
`Total Pages
`95 16-847-999
`Attorney DocketNo.
`
`First Named Inventor
`Jeffrey B. Etter
`Title
`ORAL FORMULATIONS OF CYTIDINE ANALOGS AND
`
`METHODS OF USE THEREOF
`Express Mail Label No.
`ELECTRONIC FILING
`
` Commissioner for Patents
`
`APPLICATION ELEMENTS
`
`Address to
`P.O. Box 1450
`
`See MPEP chapters 600 concerning utility patent application contents.
`
`Alexandria, VA 22313-1450
`
`Fee Transmittal Form
`
`
`(Submit an original, and a duplicate forfee processing)
`ACCOMPANYING APPLICATION PARTS
`
`
`[Total Sheets
`1 |
`20 9. C1_—Assignment Papers (cover sheet & document(s)Applicant claims Small Entity status, see 37 C.FLR. § 1.27
`
`
`Specification
`[Total Pages 103 |
`3.2
`Nameof Assignee
`Both the claims and abstract must start on a new page
`37 CFR 3.73(b) Statement
`O Powerof Attomey
`(whenthere is an assignee)
`English Translation Document(ifapplicable}
`Information Disclosure Statement (PTO/SB/08 or PTO-
`1449)
`OO Copiesof citations attached
`Preliminary Amendment
`
`| Total Sheets
`
`2 ]
`
`3.0
`
`5.2
`
`8.0
`
`Oath or Declaration
`a. BJ Unexecuted
`b. 1 A copy from aprior application (37 CFR 1.63(d))
`(for continuation/divisional with Box 18 completed)
`i. (1 DELETION OF INVENTOR(S)
`Signed statementattached deleting inventor(s) name in the prior
`application, sec 37 CFR 1.63(d)(2) and 1.33(b).
`ee
`6 Application Data Sheet. See 37 CFR 1.76
`[Total Sheets
`70
`CD-ROM or CD-R in duplicate, large table or Computer Program
`(Appendix)
`OO Landscape Table on CD
`Nucleotide and/or Amino Acid Sequence Submission
`(ifapplicable, items a- c are required)
`a. LJComputer Readable Form (CRF)
`
`
`
`}
`
`144.0
`
`is
`
`16.0]
`
`17.0
`
`Retum Receipt Postcard (MPEP 503)
`(Should be specifically itemized)
`Certified Copy of Priority Document(s)
`(ifforeign priority is claimed)
`
`Nonpublication Request under 35 U.S.C. 122 (b)(2){i).
`Applicant must attach form PTO/SB/35 or it’s equivalent
`Other:
`
`b. (Specification Sequence Listing on:
`i. [] CD-ROM or CD-R (2 copies); or
`ii. [paper
`c. [JStatementverifying identity of above copies
`18. Ifa CONTINUING APPLICATION, check appropriate box and supply the requisite information below andina preliminary amendment, or in an Application
`Data Sheet under 37 CFR 1.76:
`
`
`
`filed
`[1] Continuation-in-part (CIP)
`(1 Divisional
`O Continuation
`Group Art Unit:
`Examiner:
`Prior application information:
`For CONTINUATION OR DIVISIONAL APPSonly: Theentire disclosure of the prior application, from which an oath or declaration is supplied under Box
`5b, is considered a part of the disclosure of the accompanying continuation or divisional application and is hereby incorporated by reference. The
`incorporation can only be relied upoa whena portion has been inadvertently omitted from the submitted application parts.
`19. CORRESPONDENCE ADDRESS
`
`ofprior application No.:
`
`tyS Laoot a
`
`_] Correspondence address below
`.
`|
`& Customer Number
`|oeneeeenceeerteeernereinsertCustomerNow5
`
`
`2 =
`
`NAME
`
`‘This formis estimated to take 0.2 hours to complete. Time will vary depending upon the needs of the individual case. Any comments on the amount of time you are
`Burden Hour Statement:
`required to complete this form should be sent to the Chief Information Officer, Patent and Trademark Office, Washington, DC 20231. DO NOT SEND FEES OR COMPLETED FORMS TO
`THIS ADDRESS, SEND TO: Commissioner for Patents, P.O. Box 1450. Alexandria, VA 22313-1450,
`
`NYI-4183306v1
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1022-0002
`
`
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1022-0002
`
`

`

`
`
`23
`
`PATENT APPLICATION FEE VALUE
`
`
` NO, FILED
`
`EXTRA RATE
`
`
`ee $220.00 each
`|TotalClaimssiClaims
`
`
`
`$52.00 each
`3692.00
`
`Independent
`Total Numberof Pages w/
`
`
`$270.00 for
`drawings (excluding
`electronically filed sequence
`each 50 pages
`over 100
`270.00
`[oF
`or computer codelisting)
`
`
`
`
`
`330.00
`Basic Filing Fee
`
`220.00
`
`
`
`
`
`ia)
`
`
`
`
`
`
`JONES DAY
`COUNSELORS AT LAW
`222 East 41st Street
`NewYork, New York 10017-6702
`
`ELECTRONIC FILING
`
`ATTORNEY DOCKETNO. 9516-847-999
`Commissioner for Patents
`P.O. Box 1450
`Alexandria, VA 223 13-1450
`
`Sir:
`
`Date: May 14, 2009
`
`The following utility patent application is enclosed for filing:
`
`Applicant(s):
`
`Jeffrey B. Etter
`Mei Lai
`Jay Thomas Backstrom
`
`Executed on:
`
`unexecuted
`
`Title of Invention:
`
`ORAL FORMULATIONS OF CYTIDINE ANALOGS AND METHODSOF USE
`THEREOF
`
`
` -20
`
`Examination Fee ($220.00)
`
`ed
`
`Search Fee ($540.00)
`
`Multiple Dependency Fee
`If Applicable ($390.00)
`Total
`
`540.00
`
`390.00
`
`5662.00
`
`Applicant qualifies for the 50%Reduction for Independent Inventor, Nonprofit Organization
`or Small Business Concern.
`
`Applicant qualifies for an additional $75.00 reduction in Basic Filing Fee for Independent
`Inventor, Nonprofit Organization or Small Business Concern Filing Electronically.
`
`[x]
`
`5662.00
`Total Filing Fee
`Priority of application nos. 61/053,609 filed on May 15, 2008, 61/201,145 filed on December 5, 2008,
`and 61/157,875 filed on March 5, 2009, is claimed under 35 U.S.C. § 119.
`
`The above calculation is an estimate of the fees due.
`Deposit Account 50-3013.
`
`Please charge the required fees to Jones Day
`
`Reypectfully submitted,
`Andrew V. xfe oF
`foe
`flathony Mag
`
`
`oo Reg No.
`59,239
`
`For: Anthony M. Insogna (Reg No. 35,203)
`JONES DAY
`
`Enclosure
`
`This form is not for use with continuation, divisional, re-issue, design or plant patent applications.
`NYT-4 1 83304vI
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1022-0003
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1022-0003
`
`

`

`Atty Docket No.
`501872-999847/95 16-847-999
`
`ORAL FORMULATIONS OF CYTIDINE ANALOGS
`AND METHODSOF USE THEREOF
`
`I.
`
`CROSS-REFERENCE TO RELATED APPLICATIONS
`
`This application claimspriority to U.S. Provisional Patent Application Nos.
`{0001}
`61/053,609, filed May 15, 2008; 61/201,145, filed December 5, 2008; and 61/157,875, filed
`
`March 5, 2009, the contents of each of which are incorporated by reference herein in their
`entireties.
`
`Il.
`
`FIELD
`
`Provided herein are pharmaceutical formulations comprising cytidine analogs, or
`[0002]
`their salts, solvates, hydrates, precursors, and/or derivatives thereof, for oral administration in
`
`subjects. Also provided are methods for making the formulations and methodsfor using the
`formulations to treat diseases and disorders including cancer, disorders related to abnormal
`cell proliferation, hematologic disorders, and immune disorders, amongothers.
`
`i.
`
`BACKGROUND
`
`Cancer is a major worldwide public health problem; in the United States alone,
`[0003]
`approximately 570,000 cancer-related deaths were expected in 2005. See, e.g., Jemalet 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 (¢.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,
`
`Myelodysplastic syndromes (MDS)refers to a diverse group of hematopoietic
`[0004]
`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
`
`NYI-4182720vI
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1022-0004
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1022-0004
`
`

`

`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).
`[0005]
`MDSare grouped together because of the presence of dysplastic changes in one or
`more of the hematopoietic lineages including dysplastic changes in the myeloid, erythroid,
`and megakaryocytic series. These changes result in cytopenias in one or moreofthe three
`lineages. Patients afflicted with MDS may develop complicationsrelated 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 overwhelmsthe
`healthy marrow. The disease course differs, with some cases behaving as an indolent disease
`and others behaving aggressively with a veryshort clinical course that converts into an acute
`form of leukemia. The majority of people with higher risk MDS eventually experience bone
`marrow failure. Up to 50% of MDS patients succumb to complications, such as infection or
`
`bleeding, before progressing to AML.
`[0006]
`Primary and secondary MDS are defined by taking into accountpatients’ prior
`history: previous treatments with chemotherapy, radiotherapy or professional exposure to
`toxic substances are factors delineating secondary MDS (sMDS)from primary MDS.
`Cytogenetically, one difference between the two groupsis the complexity of abnormal
`karyotypes; single chromosomeaberrations are typical for primary MDS, while multiple
`changes are more frequently seen in secondary disorders. Some drugs may have specific
`targets such as hydroxurea for 17p and topoisomerases inhibitors for 11q23 and 21q22. The
`genetic changesin the malignant cells of MDSresult mainly in the loss ofgenetic material,
`
`including probable tumor suppressor genes.
`[0007]
`An international group of hematologists, the French-American-British (FAB)
`Cooperative Group,classified MDSinto five subgroups, differentiating them from acute
`myeloid leukemia.See, e.g., The Merck Manual 954 (17th ed. 1999); Bennett J. M., et al.,
`Ann. Intern. Med., 103(4): 620-5 (1985); and Besa E. C., Med. Clin. North Am. 76(3): 599-
`617 (1992). An underlyingtrilineage dysplastic change in the bone marrowcells of the
`patients is found in all subtypes. Information is available regarding the pathobiology of
`MDS, certain MDSclassification systems, and particular methodsoftreating and managing
`
`
`
`NYI-4182720vI
`
`-2-
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1022-0005
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1022-0005
`
`

`

`MDS. See, e.g., U.S. Patent No. 7,189,740 (issued March 13, 2007), which is incorporated by
`reference herein in its entirety.
`
`Nucleoside analogs have been usedclinically for the treatment ofviral infections
`[0008]
`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.
`
`5-Azacytidine (National Service Center designation NSC-102816; CAS Registry
`[0009]
`Number 320-67-2), also known as azacitidine, AZA, or 4-amino-|-$-D-ribofuranosyl-1,3,5-
`triazin-2(LH)-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 analogs and their related natural nucleoside is the presence
`of a nitrogen at position 5 ofthe cytosine ring in place of a carbon. 5-Azacytidine may be
`defined as having the molecular formula CgH)2N4Os, a molecular weight of 244.21 grams per
`mole, and the following structure:
`
`NH»
`
`wan
`LAs
`
`HO
`
`H
`
`OH
`
`H
`OH
`
`5-Azacytidine.
`
`Other membersof the class of cytidine analogs include, for example: 1-B-D-
`[0010]
`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; |-B-D-riboturanosyl-2(1 H)-pyrimidinone (Zebularine); 2',3'-dideoxy-5-fluoro-
`3'-thiacytidine (Emtriva); 2'-cyclocytidine (Ancitabine); 1-B-D-arabinofuranosyl-5-
`azacytosine (Fazarabineor ara-AC); 6-azacytidine (6-aza-CR); 5,6-dihydro-5-azacytidine
`
`NYI-4182720v1
`
`-3-
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1022-0006
`
`
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1022-0006
`
`

`

`(dH-aza-CR); N*-pentyloxycarbonyl-5'-deoxy-5-fluorocytidine (Capecitabine); N‘-octadecyl-
`cytarabine; and elaidic acid cytarabine.
`[0011]
`After its incorporation into replicating DNA, 5-azacytidine or 5-aza-2'-
`deoxycytidine forms a covalent complex with DNA methyltransferases. DNA
`methyltransferases are responsible for de novo DNA methylation and for reproducing
`established methylation patterns in daughter DNAstrandsofreplicating DNA. Inhibition of
`DNA methyltransferases by 5-azacytidine or 5-aza-2'-deoxycytidine leads to DNA
`hypomethylation, thereby restoring normal functions to morphologically dysplastic, immature
`hematopoietic cells and cancer cells by re-expression of genes involved in normalcell cycle
`regulation, differentiation and death. The cytotoxic effects of these cytidine analogs cause
`the death of rapidly dividingcells, including cancer cells, that are no longer responsive to
`normalcell growth control mechanisms. 5-azacytidine, unlike 5-aza-2'-deoxycytidine, also
`incorporates into RNA. The cytotoxic effects of azacitidine may result from multiple
`mechanisms,including inhibition of DNA, RNAandprotein synthesis, incorporation into
`
`RNAand DNA,and activation of DNA damage pathways.
`[0012]
`5-Azacytidine and 5-aza-2'-deoxycytidine have beentested in clinical trials and
`showedsignificant anti-tumoractivity, such as, for example, in the treatment of
`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 et al., Curr. Opin. Invest. Drugs 3(4): 627-33 (2002).
`5-Azacytidine has undergone NCI-sponsoredtrials for the treatment of MDSand has been
`approvedfor treating all FAB subtypes of MDS. See,e.g., Kornblith et al., J. Clin. Oncol.
`20(10): 2441-2452 (2002); Silvermanet al., J. Clin. Oncol. 20(10): 2429-2440 (2002). 5-
`Azacytidine mayalter the natural course of MDS by diminishing the transformation to AML
`through its cytotoxic activity and its inhibition of DNA methyltransferase.
`In a PhaseII]
`study, 5-azacytidine administered subcutaneously significantly prolonged survival and time
`to AMLtransformation or death in subjects with higher-risk MDS. See, e.g., P. Fenaux ef al,
`Lancet Oncol., 2009, 10(3):223-32; Silverman et al., Blood 106(11): Abstract 2526 (2005).
`[0013}
`5-Azacytidine and other cytidine analogs are approved for subcutaneous (SC) or
`intravenous (IV) administration to treat various proliferative disorders. Oral dosing of
`cytidine analogs would be more desirable and convenientfor patients and doctors, e.g., by
`eliminating injection-site reactions that may occur with SC administration and/or by
`permitting improved patient compliance. However, oral delivery of cytidine analogs has
`
`NYI-4182720v1
`
`-~-4-
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1022-0007
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1022-0007
`
`

`

`proven difficult due to combinations of chemicalinstability, 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
`
`absorbedin specific regions of the lower gastrointestinal tract, such as the jejunum in the
`
`small intestine. See, e.g., Sands, ef al., U.S. Patent Publication No. 2004/0162263 (App. No.
`
`10/698,983). In addition, a generally accepted belief in 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. Asa 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.
`
`A great need remains for oral formulations and dosage formsof cytidine analogs,
`[0014]
`suchas, e.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 maintenancetherapies;
`
`development of improved treatment regimens that maximize biologic activity; use of cytidine
`analogsfor treating new diseases or disorders; and/or other potential advantageous benefits.
`
`IV.
`
`SUMMARY
`
`[0015]
`
`Provided herein are pharmaceutical compositions comprising cytidine analogs,
`
`wherein the compositions release the API substantially in the stomach upon oral
`
`administration. Also provided are methods for making the compositions, and methods for
`
`using the compositions to treat diseases and disorders including cancer, disorders related to
`
`abnormalcell proliferation, and hematologic disorders, among others.
`
`[0016]
`
`In certain embodiments, the cytidine analog is 5-azacytidine.
`
`In other
`
`In yet
`embodiments, the cytidine analog is 5-aza-2'-deoxycytidine (decitabine or 5-aza-CdR).
`other embodiments, the cytidine analog is, for example: 1-f-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; 1-B-D-ribofuranosyl-2(1H)-pyrimidinone (Zebularine); 2',3'-dideoxy-
`5-fluoro-3'-thiacytidine (Emtriva); 2'-cyclocytidine (Ancitabine); |-8-D-arabinofuranosy]-5-
`
`NYT-4182720v1
`
`~5-
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1022-0008
`
`
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1022-0008
`
`

`

`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.
`[0017]
`Certain embodiments herein provide compositions that are single unit dosage
`forms comprising a cytidine analog. Certain embodiments herein provide compositions that
`are non-enteric-coated. Certain embodiments herein provide compositions that are tablets
`
`comprising a cytidine analog. Certain embodiments herein provide compositionsthat are
`capsules comprising a cytidine analog. The capsules maybe, e.g., a hard gelatin capsule or a
`soft gelatin capsule; particular embodiments provide hydroxypropy! methylcellulose (HPMC)
`capsules. In certain embodiments, the single unit dosage forms optionally 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 compositionis a tablet that effects an immediate
`release of the API upon oral administration. In other embodiments, the compositionis a
`
`tablet that effects a controlled release of the API substantially in the stomach. In certain
`
`embodiments, the composition is a capsule that effects an immediate release of the API upon
`
`oral administration.
`
`In other embodiments, the composition 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, or non-aqueoussolvent, such as, for example ethanol.
`[0018]
`Certain embodiments herein provide methods of making formulations of cytidine
`analogs intended for oral delivery. Further provided are articles of manufacture containing
`packaging material, an oral formulation of a cytidine analog, and a label that indicatesthat
`the formulation is for the treatment of certain diseases or disorders including,e.g., a cancer, a
`
`disorder related to abnormalcell proliferation, a hematologic disorder, or an immune
`
`disorder.
`
`Certain embodiments herein provide methods of using the formulations provided
`[0019]
`herein to treat diseases or disorders including, ¢.g., cancer, disorders related to abnormalcell
`
`In certain
`proliferation, hematologic disorders, or immunedisorders, among others.
`embodiments, the formulations ofcytidine analogsare orally administered to subjects in need
`
`thereof to treat a cancer or a hematological disorder, such as, for example, MDS, AML, ALL,
`
`CML, NHL,leukemia, or lymphoma; or a solid tumor, such as, for example, sarcoma,
`
`NY1-4182720v1
`
`-6-
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1022-0009
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1022-0009
`
`

`

`melanoma,carcinoma,or cancerof 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 formulations of cytidine analogsare orally
`
`administered to subjects in need thereof to treat an immunedisorder. 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.
`
`In
`
`certain embodiments, the oral formulations provided herein are co-administered with one or
`
`more therapeutic agents to provide a resensitization 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.
`
`[0020]
`
`In particular embodiments, provided herein are tablets containing 5-azacytidine
`
`and methods for making and using the tablets to treat cancer, disorders related to abnormal
`
`cell proliferation, or hematologic disorders. 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
`
`describedin, 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.
`
`[0021]
`
`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 (i.e., are non-enteric-coated);
`
`are tablets; are capsules; further comprise an excipient selected from any excipient disclosed
`
`herein; further comprise a permeation enhancer; further comprise d-alpha-tocopheryl
`
`polyethylene glycol 1000 succinate; further comprise a permeation enhancerin 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 amountof5-azacytidine of at least about 40 mg; have an amountof 5-azacytidine of
`
`at least about 400 mg; have an amountof 5-azacytidine of at 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 ofat least about 400 ng-hr/mL following oral
`
`administration to a subject; achieve a maximum plasma concentration ofat least about 100
`
`NYI-4182720v1
`
`-7-
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1022-0010
`
`
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1022-0010
`
`

`

`ng/mL following oral administration to a subject; achieve a maximum plasmaconcentration
`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.
`[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 an area-under-the-curve value of
`
`at least about 200 ng-hr/mL following oral administration.
`[0023]
`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.
`[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 maximum plasma concentration
`
`of at least about 100 ng/mL following oral administration.
`
`Specific embodiments herein provide a pharmaceutical composition for oral
`[0025]
`administration comprising a therapeutically effective amount of 5-azacytidine, which releases
`
`the 5-azacytidine substantially in the stomach and achieves a maximum plasmaconcentration
`
`of at least about 200 ng/mL following oral administration.
`[0026]
`Specific embodiments herein provide a pharmaceutical composition for oral
`administration comprising a therapeutically effective amount of S-azacytidine, which releases
`
`the 5-azacytidine substantially in the stomach and achieves a time to maximum plasma
`
`concentration of, e.g., less than about 6 hr, less than about 5 hr, less than about 4 hr, less than
`
`about 3 hr, less than about 2.5 hr, less than about 2 hr, less than about 1.5 hr, less than about 1
`
`hr, less than about 45 min, or less than about 30 min following oral administration.
`
`In
`
`specific embodiments, the presence of food may affect (e.g., extend) the total exposure and/or
`
`time to maximum plasma concentration.
`[0027]
`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.
`
`NY1-4182720v 1
`
`-§-
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1022-0011
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1022-0011
`
`

`

`[0028]
`
`Specific embodiments herein provide any of the aforementioned compositions, as
`
`single unit dosage forms, tablets, or capsules.
`
`[0029]
`
`Specific embodiments herein provide, inter alia, methodsfortreating 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
`
`stomachfollowing 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 thereofan 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 weight relative 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
`
`composition is a tablet; the composition is a capsule; the composition further comprises an
`
`excipient selected from any excipient disclosed herein; the amount of5-azacytidineis at least
`
`about 40 mg; the amount of5-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 ofat 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 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
`
`[0030]
`
`Specific embodiments herein provide, inter alia, pharmaceutical compositions
`
`comprising a therapeutically effective amount of5-azacytidine, wherein the compositions are
`
`for treating a disease or disorder associated with abnormalcell proliferation, wherein the
`
`NYT-4182720v1
`
`-9.
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1022-0012
`
`Apotex v. Cellgene - IPR2023-00512
`Petitioner Apotex Exhibit 1022-0012
`
`

`

`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 of 5-azacytidine of about
`40 mg, about 400 mg, or about 1000 mg; are prepared to achieve an area-under-the-curve
`value ofat 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 syndrome or
`
`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.
`
`Specific embodiments herein provide, inter alia, uses of 5-azacytidine for the
`[0031)
`preparation of a pharmac

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