throbber

`

`WO 2009/139888
`
`PCT/0S2009/002999
`
`1
`
`ORAL FORMULATIONS OF CYTIDINE ANALOGS
`AND METHODS OF USE THEREOF
`
`I.
`
`CROSS-REFERENCE TO RELATED APPLICATIONS
`
`(0001]
`
`This application claims priority to U.S. Provisional Patent Application Nos.
`
`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.
`
`II.
`
`FIELD
`
`(0002]
`
`Provided herein are pharmaceutical formulations 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 formulations and methods for using the
`
`formulations to treat diseases and disorders including cancer, disorders related to abnormal
`
`cell proliferation, hematologic disorders, and immune disorders, among others.
`
`III.
`
`BACKGROUND
`
`(0003]
`
`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.
`
`(0004] 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
`
`

`

`WO 2009/139888
`
`PCT/0S2009/002999
`
`2
`
`production. See, e.g., The Merck Manual 953 (17th ed. 1999); List et al., J Clin. Oneal.
`
`8:1424 (1990).
`
`[0005) MDS are 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 more of the three
`
`lineages. Patients 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 programmed cell death (apoptosis). As the disease
`
`progresses and converts into leukemia, a proliferation of leukemic cells 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 MDS eventually experience bone
`
`marrow failure. Up to 50% ofMDS patients succumb to complications, such as infection or
`
`bleeding, before progressing to AML.
`
`[0006)
`
`Primary and secondary MDS are defined by taking into account patients' 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 groups is the complexity of abnormal
`
`karyotypes; single chromosome aberrations 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 1 lq23 and 21q22. The
`
`genetic changes in the malignant cells of MDS result mainly in the loss of genetic material,
`
`including probable tumor suppressor genes.
`
`[0007)
`
`An international 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., et al.,
`
`Ann. Intern. Med., I 03( 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 found in all subtypes. Information is available regarding the pathobiology of
`
`MDS, certain MDS classification systems, and particular methods of treating and managing
`
`MDS. See, e.g., U.S. Patent No. 7,189,740 (issued March 13, 2007), which is incorporated by
`
`reference herein in its entirety.
`
`

`

`WO 2009/139888
`
`PCT/0S2009/002999
`
`3
`
`Nucleoside analogs have been used clinically for the treatment of viral infections
`
`[0008]
`and cancer. Most nucleoside analogs are classified as anti-metabolites. After they enter the
`
`cell, nucleoside analogs are successively phosphorylated to nucleoside 5'-mono-phosphates,
`
`di-phosphates, and tri-phosphates.
`
`[0009]
`
`5-Azacytidine (National Service Center designation NSC-102816; CAS Registry
`
`Number 320-67-2), also known as azacitidine, AZA, or 4-amino-l-~-D-ribofuranosyl-1,3,5-
`
`triazin-2(1H)-one, is currently marketed as the drug product VIDAZA ®. 5-Azacytidine is a
`
`nucleoside analog, more specifically a cytidine analog. 5-Azacytidine is an antagonist of its
`
`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 of the cytosine ring in place of a carbon. 5-Azacytidine may be
`
`defined as having the molecular formula CsH12N4Os, a molecular weight of 244.21 grams per
`
`mole, and the following structure:
`
`NH2
`
`N~N
`
`llN~O
`HO'l..-~oJ
`tf-il
`
`OH OH
`
`5-Azacytidine.
`
`[0010]
`
`Other members of the class of cytidine analogs include, for example: 1-~-D-
`
`arabinofuranosylcytosine (Cytarabine or ara-C); 5-aza-2'-deoxycytidine (Decitabine or 5-aza(cid:173)
`
`CdR); pseudoisocytidine (psi ICR); 5-fluoro-2'-deoxycytidine (FCdR); 2'-deoxy-2',2'(cid:173)
`
`difluorocytidine (Gemcitabine); 5-aza-2'-deoxy-2',2'-difluorocytidine; 5-aza-2'-deoxy-2'(cid:173)
`
`fluorocytidine; l-~-D-ribofuranosyl-2(1H)-pyrimidinone (Zebularine); 2',3'-dideoxy-5-fluoro-
`
`3'-thiacytidine (Emtriva); 2'-cyclocytidine (Ancitabine); l-~-D-arabinofuranosyl-5-
`
`azacytosine (Fazarabine or ara-AC); 6-azacytidine (6-aza-CR); 5,6-dihydro-5-azacytidine
`(dH-aza-CR); N4-pentyloxycarbonyl-5'-deoxy-5-fluorocytidine (Capecitabine); N4-octadecyl(cid:173)
`cytarabine; and elaidic acid cytarabine.
`
`

`

`WO 2009/139888
`
`PCT/0S2009/002999
`
`4
`
`[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 DNA strands ofreplicating 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 normal cell cycle
`
`regulation, differentiation and death. The cytotoxic effects of these cytidine analogs cause
`
`the death of rapidly dividing cells, including cancer cells, that are no longer responsive to
`
`normal cell 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, RNAand protein synthesis, incorporation into
`
`RNA and DNA, and activation of DNA damage pathways.
`
`[0012]
`
`5-Azacytidine and 5-aza-2'-deoxycytidine have been tested in clinical trials and
`
`showed significant anti-tumor activity, 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-sponsored trials for the treatment of MDS and has been
`approved for treating all F AB subtypes of MDS. See, e.g., Komblith et al., J. Clin. Oncol.
`
`20(10): 2441-2452 (2002); Silverman et al., J. Clin. Oncol. 20(10): 2429-2440 (2002). 5-
`
`Azacytidine may alter the natural course of MDS by diminishing the transformation to AML
`
`through its cytotoxic activity and its inhibition of DNA methyltransferase. In a Phase III
`
`study, 5-azacytidine administered subcutaneously significantly prolonged survival and time
`
`to AML transformation or death in subjects with higher-risk MDS. See, e.g., P. Fenaux et 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 convenient for 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
`
`proven difficult due to combinations of chemical instability, enzymatic instability, and/or
`
`poor permeability. For example, cytidine analogs have been considered acid labile and
`
`

`

`WO 2009/139888
`
`PCT/0S2009/002999
`
`5
`
`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 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. 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.
`
`[0014)
`
`A great need remains for oral formulations and dosage forms of cytidine analogs,
`
`such as, e.g., 5-azacytidine, to potentially permit, inter alia, more advantageous dosing
`
`amounts or dosing periods; improved pharmacokinetic profiles, pharmacodynamic profiles,
`
`or safety profiles; evaluation of the benefits oflong-term or maintenance therapies;
`
`development of improved treatment regimens that maximize biologic activity; use of cytidine
`
`analogs for 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
`
`abnormal cell proliferation, and hematologic disorders, among others.
`
`[0016)
`
`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-P-D-arabinofuranosylcytosine
`
`(Cytarabine or ara-C); pseudoisocytidine (psi ICR); 5-fluoro-2'-deoxycytidine (FCdR); 2'(cid:173)
`
`deoxy-2',2'-difluorocytidine (Gemcitabine ); 5-aza-2'-deoxy-2' ,2'-difluorocytidine; 5-aza-2'(cid:173)
`
`deoxy-2'-fluorocytidine; l-P-D-ribofuranosyl-2(1H)-pyrimidinone (Zebularine); 2',3'-dideoxy-
`
`5-fluoro-3'-thiacytidine (Emtriva); 2'-cyclocytidine (Ancitabine); l-P-D-arabinofuranosyl-5-
`
`azacytosine (Fazarabine or ara-AC); 6-azacytidine (6-aza-CR); 5,6-dihydro-5-azacytidine
`
`

`

`WO 2009/139888
`
`PCT/0S2009/002999
`
`6
`(dH-aza-CR); N4-pentyloxycarbonyl-5'-deoxy-5-fluorocytidine (Capecitabine); N4-octadecyl(cid:173)
`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 compositions that are
`
`capsules comprising a cytidine analog. The capsules may be, e.g., a hard gelatin capsule or a
`
`soft gelatin capsule; particular embodiments provide hydroxypropyl 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 composition is a tablet that effects an immediate
`
`release of the API upon oral administration. In other embodiments, the composition is 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-aqueous solvent, 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 indicates that
`
`the formulation is for the treatment of certain diseases or disorders including, e.g., a cancer, a
`
`disorder related to abnormal cell proliferation, a hematologic disorder, or an immune
`
`disorder.
`
`[0019]
`
`Certain embodiments herein provide methods of using the formulations provided
`
`herein to treat diseases or disorders including, e.g., cancer, disorders related to abnormal cell
`
`proliferation, hematologic disorders, or immune disorders, among others. In certain
`
`embodiments, the formulations of cytidine analogs are 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,
`
`melanoma, carcinoma, or cancer of the colon, breast, ovary, gastrointestinal system, kidney,
`
`

`

`WO 2009/139888
`
`PCT/0S2009/002999
`
`7
`
`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 analogs are orally
`
`administered to subjects in need thereof to treat an immune disorder. 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
`
`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.
`
`[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 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 of at least about 40 mg; have an amount of 5-azacytidine of
`
`at least about 400 mg; have an amount of 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 of at least about 400 ng-hr/mL following oral
`
`administration to a subject; achieve a maximum plasma concentration of at least about 100
`
`ng/mL following oral administration to a subject; achieve a maximum plasma concentration
`
`

`

`WO 2009/139888
`
`PCT/0S2009/002999
`
`8
`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.
`
`[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 maximum plasma concentration
`
`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 5-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.
`
`

`

`WO 2009/139888
`
`PCT/0S2009/002999
`
`9
`
`[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, methods for treating a subject
`
`having a disease associated with abnormal cell 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 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 of 5-azacytidine is at least
`
`about 40 mg; the amount of 5-azacytidine is at least about 400 mg; the amount of 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 of at 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 of at least about 200 ng/mL following oral administration to the subject; the
`
`method achieves a time to maximum plasma concentration of less than about 90 minutes
`
`following oral administration to the subject; and/or the method achieves a time to maximum
`
`plasma concentration of less than about 60 minutes following oral administration to the
`
`subject
`
`[0030)
`
`Specific embodiments herein provide, inter alia, pharmaceutical compositions
`
`comprising a therapeutically effective amount of 5-azacytidine, wherein the compositions are
`
`for treating a disease or disorder associated with abnormal cell proliferation, wherein the
`
`

`

`WO 2009/139888
`
`PCT/0S2009/002999
`
`10
`
`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 of at least about 200 ng-hr/mL or 400 ng-hr/mL following oral administration; are
`
`prepared to achieve a maximum plasma concentration of at 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 I 000 succinate;
`
`are single unit dosage forms; are tablets or capsules; and/or further comprise an excipient
`
`selected from any excipient disclosed herein.
`
`[0031]
`
`Specific embodiments herein provide, inter alia, uses of 5-azacytidine for the
`
`preparation of a pharmaceutical composition for treating a disease associated with abnormal
`
`cell 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 amount disclosed herein; and/or the composition is prepared for immediate
`
`release. Further embodiments provide, inter alia, methods for treating a subject having a
`
`disease or disorder provided herein by administering a pharmaceutical compositions provided
`
`herein, wherein the treatment results in improved survival of the subject.
`
`V.
`
`BRIEF DESCRIPTION OF THE DRAWINGS
`
`[0032]
`
`Figure 1 represents processes and steps that may be used to make particular tablets
`
`comprising azacitidine for oral dosing; in specific embodiments, one or more steps may be
`
`optionally omitted.
`Figure 2 represents human PK profiles following 75 mg/m2 SC dosing of
`[0033]
`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).
`
`

`

`WO 2009/139888
`
`PCT/OS2009/002999
`
`[0034]
`
`11
`Figure 3 represents human PK profiles following SC (75 mg/m2
`300 mg, and 360 mg) dosing of azacitidine in a multiple dose escalation study. The
`
`) and PO (240 mg,
`
`azacitidine plasma PK profiles are compared among various doses. The X-axis represents
`
`time; the Y-axis represents azacitidine plasma concentrations (mean± SD).
`
`[0035]
`
`Figure 4 represents PD data from an individual 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/µL), Hgb (g/dL), ANC (K/µL), and
`
`Relative BM Blast(%) are plotted versus sampling dates over the course of the study.
`
`[0036]
`
`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/µL), Hgb (g/dL), ANC (K/µL), and
`
`Relative BM Blast(%) are plotted versus sampling dates over the course of the study.
`
`[0037]
`
`Figure 6 represents PD data from an 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/µL), Hgb (g/dL), ANC (K/µL),
`
`and Relative BM Blast(%) are plotted versus sampling dates over the course of the study.
`
`[0038]
`
`Figure 7 represents a study design of a Rapid Aza Clinical Evaluation (RACE)
`
`study CLOOS. 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.
`
`[0039]
`Figure 8 represents azacitidine human PK profiles from an individual patient
`(Subject 106003, N = 1) following SC (124 mg, 75 mg/m2
`) and PO (180 mg, 360 mg, 1,200
`mg, Formulation 4) dosing of azacitidine from a RACE clinical study. AUC(0-t) values for
`
`the SC and PO doses are depicted.
`
`[0040]
`Figure 9 represents azacitidine human PK profiles from an individual patient
`(Subject 106004, N = 1) following SC (120 mg, 75 mg/m2
`mg, Formulation 6) dosing of azacitidine from a RACE clinical study. AUC(0-oo) values for
`
`) and PO (180 mg, 360 mg, 1,200
`
`the SC and PO doses are depicted.
`
`[0041]
`
`Figure 10 represents human PK profiles (linear scale) following SC and oral
`
`administration of azacitidine in clinical studies.
`[0042]
`
`Figure 11 represents human PK profiles (semi-log scale) following SC and oral
`
`administration of azacitidine in clinical studies.
`
`[0043]
`
`Figure 12 represents human AUC values following SC dosing of azacitidine and
`
`oral dosing of azacitidine with Formulations #3, #4, and #6 at various dosage levels in
`
`

`

`WO 2009/139888
`
`PCT/0S2009/002999
`
`clinical studies (CLOOS and CLOOS).
`
`12
`
`[0044]
`
`Figure 13 represents human Cmax values in patients following SC dosing of
`
`azacitidine and oral dosing of azacitidine with Formulations #3, #4, and #6 at various dosage
`
`levels in clinical studies.
`
`[0045)
`
`Figure 14 represents relative oral bioavailability in humans following oral dosing
`
`of azacitidine with Formulations #3, #4, and #6 at various dosage levels.
`
`[0046)
`
`Figure 15 represents percent exposure in humans relative to SC administration
`
`following oral dosing of azacitidine with Formulations #3, #4, and #6 at various dosage
`
`levels.
`
`[0047)
`
`Figure 16 represents profiles of human plasma concentration versus time (linear
`
`scale) following oral dosing of azacitidine with Formulations #3 and #6 and 180 mg (n=6).
`
`[0048)
`
`Figure 17 represents linear scale profiles of human plasma concentration (ng/ml)
`
`versus time (hr) following oral dosing of azacitidine with Formulations #3 and #6 and 360
`
`mg (n=6).
`
`[0049)
`
`Figure 18 represents a plot of values for individual ("ind") and mean azacitidine
`
`ACU(O-inf) (ng*hr/ml) versus azacitidine dose (mg), with calculated linear regression lines
`
`for Formulations #3 and #6.
`
`[0050)
`
`Figure 19 represents a comparison of azacitidine relative oral bioavailability (%)
`
`(mean± SD) versus azacitidine dose (mg) following dosing with Formulation #3 or #6.
`
`[0051)
`
`Figure 20 represents a comparison of azacitidine exposure as compared to SC
`
`dose (mean± SD) versus azacitidine dose (mg) following oral administration of Formulation
`
`#3 or #6.
`
`VI.
`
`DETAILED DESCRIPTION
`
`[0052)
`
`Unless defined otherwise, all technical and scientific terms used herein have the
`
`same meaning as commonly understood by one of ordinary skill in the art. All publications
`
`and patents referred to herein are incorporated by reference herein in their entireties.
`
`A.
`
`Definitions
`
`[0053]
`
`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.
`
`[0054)
`
`The term "about" or "approximately" means an acceptable error for a particular
`
`value as determined by one of ordinary skill in the art, which depends in part on how the
`
`

`

`WO 2009/139888
`
`PCT/0S2009/002999
`
`13
`
`value is measured or determined. In certain embodiments, the term "about" 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.
`
`[0055]
`
`As used herein, and

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