throbber
Hindawi Publishing Corporation
`Chemotherapy Research and Practice
`Volume 2011, Article ID 965826, 9 pages
`doi: 10.1155/2011/965826
`
`Research Article
`
`Development of an Oral Form of Azacytidine:
`2'3'5' Triacetyl-5-Azacytidine
`
`Amy Ziemba,' Eugene Hayes,' Burgess B. Freeman III,” Tao Ye,’ and Giuseppe Pizzorno!
`
`! Nevada Cancer Institute, One Breakthough Way, Las Vegas, NV 89135, USA
`*St. Jude Children’s Research Hospital, Memphis, TN 38105, USA
`>The Hong Kong Polytechnic University, Kowloon, Hong Kong
`
`Correspondence should be addressed to Giuseppe Pizzorno, gpizzorno@nvcancer.org
`
`Received 12 July 2011; Revised 14 September 2011; Accepted 21 September 2011
`
`Academic Editor: G. J. Peters
`
`Copyright © 2011 Amy Ziembaetal. ‘his is an open accessarticle distributed under the Creative CommonsAttribution License,
`which permits unrestricted use, distribution, and reproduction in any medium, provided the original work is properlycited.
`
`Myelodysplastic syndromes (MD&Ss) represent a group of incurable stem-cell malignancics which are predominantly treated by
`supportive care. Epigenetic silencing through promoter methylation of a number of genes is present in poor-risk subtypes of
`MDS and often predicts transformation to acute myelogenous leukemia (AML). Azacitidine and decitabine, two FDA-approved
`DNA methyltransferase (DNMT) inhibitors, are able to improve overall response although their oral bioavailability complicates
`their clinical use. This study evaluated 2', 3’, 5'-triacetyl-5-azacitidine (TAC) as a potential prodrug for azacitidine. The prodrug
`demonstrated significant pharmacokinetic improvements in bioavailability, solubility, and stability over the parent compound.
`In vivo analyses indicated a lack of general toxicity coupled with significantly improved survival. Pharmacodynamic analyses
`confirmed its ability to suppress global methylation in vivo. These data indicate that esterified nucleoside derivatives may be
`effective prodrugs for azacitidine and encourages further investigation of TAC into its metabolism, activity, and possible clinical
`evaluation.
`
`1. Introduction
`
`Currently, it is estimated that between 12,000 and 20,000 new
`cases of MDS are diagnosed each year in the United States.
`Although MDS can affect all ages, the highest prevalence
`occurs in those over 60 years of age [1, 2]. Much of the pop-
`ulation has indolent forms of MDS, making it one of the
`most prevalent hematologic malignanciesof older adults.
`MDS represents a heterogeneous group of hematopoietic
`disorders which are derived from an abnormal multipotent
`progenitor cell and are characterized by hyperprolifera-
`tive bone marrow, cellular dysplasia, and ineffective hem-
`atopoiesis [3]. Morbidity and mortality result from anemia,
`bleeding, and infection, along with transformation to acute
`myelogenous leukemia (AML) in approximately one-third
`of patients
`[4, 5]. The basis of therapy is supportive
`care, including red blood cell or platelet transfusions and
`treatment of infections. Stem-cell transplantation remains
`the only chance for cure, but it is associated with significant
`treatment-related morbidity and mortality and is generally
`
`restricted to patients <60 years of age [6]. Similar limitations
`exist for the use of high dose chemotherapy. Given the
`limitations of existing therapies, there is a clear need for
`additional therapeutic options for patients with MDS.
`Cancer cells are characterized by abnormal DNA methy-
`lation patterns, and DNA hypermethylation is suspected
`of being involved in MDS progression and leukernoge-
`nesis. Therefore,
`inhibitors of DNA methylation repre-
`sent a useful approach to revert
`these epigenetic chan-
`ges. 5-azacitidine (Vidaza) and its derivative 5-aza-2’-
`deoxycytidine/decitabine (Dacogen) are nucleoside analogs
`with DNA hypomethylating activity that have been FDA-
`approved during the past 5-6 years for MDS treatment
`(7, 8]. hey appear to induce re-expression of key tumor
`suppressor genes in MDS [9]. Compared to supportivecare,
`both agents show an improved overall response (60% versus
`5%), a longertime to progression to AML ordeath, but still
`with limited overall survival advantage [10]. Azacitidine has
`been developed for the treatment of acute leukemia and is
`currently being evaluated in a variety of other disorders [11].
`
`(cid:38)(cid:40)(cid:47)(cid:42)(cid:40)(cid:49)(cid:40)(cid:3)(cid:21)(cid:20)(cid:22)(cid:21)
`CELGENE 2132
`(cid:36)(cid:51)(cid:50)(cid:55)(cid:40)(cid:59)(cid:3)(cid:89)(cid:17)(cid:3)(cid:38)(cid:40)(cid:47)(cid:42)(cid:40)(cid:49)(cid:40)
`APOTEX v. CELGENE
`(cid:44)(cid:51)(cid:53)(cid:21)(cid:19)(cid:21)(cid:22)(cid:16)(cid:19)(cid:19)(cid:24)(cid:20)(cid:21)
`IPR2023-00512
`
`

`

`DNA methylation results in the addition of a methyl
`group at the carbon 5 position of the cytosine ring in CpG
`dinucleotides, which is critical to chromatin structure and
`genomic stability [12]. Since 5-methyl cytosine can be deam-
`inated to thymidine, DNA hypermethylation also facilitates
`gene mutations in human cancers [13]. The transferring
`of methyl groups from S-adenosylmethionine to cytosine
`is catalyzed by DNA methyltransferases, the best known of
`which is DNMT1. The inactivation of DNM1Ts has been
`shown to be the most effective method of inhibiting DNA
`methylation although it is recognized that this approach
`lacks specificity. However, inhibiting the activity of DNMTs
`has resulted in the abrogation of tumorigenicity in murine
`cancer models [14].
`DNMT-inhibiting nucleoside analogs require metabo-
`lism by kinases into nucleotides before their incorporation
`into DNA and/or RNA in order to inhibit DNA methylation.
`‘The modification at C5 prevents the release of DNMITs by
`forming a covalent complex, preventing further DNA meth-
`ylation and consequently the DNA of the progenycells is not
`methylated [15]. Azacitidine and Decitabine are extremely
`potent in inhibiting DNA methylation, but their short half-
`lives in aqueous solutions [16, 17] and low oral bivavail-
`ability complicate their delivery. For years,
`these drugs
`used as antineoplastic agents in leukemias were escalated to
`maximum tolerated doses (MTD)
`[18]; however, recent
`clinical trials have confirmed that low-dose exposures lead
`to greater responses and are associated with less toxicity
`[19, 20].
`In an effort to overcome the stability and pharmacoki-
`netic limitations, a numberof groups are working to develop
`oral DNMTinhibitors, including oral forms of decitabine
`and azacitidine. As a proof of principle we have synthesized
`an acetylated derivative of azacitidine, 2’,3’,5'-triacetyl-5-
`azacitidine (TAC)
`to evaluate as an oral prodrug. While
`this is not a totally new approach, it was never utilized
`for this derivative and in this set of diseases. Our cur-
`rent study demonstrates that 2’,3',5’-triacetyl-5-azacitidine
`(TAC) effectively inhibits methylation and improves life
`expectancies in murine models while demonstrating lower
`toxicity when comparedto its parent compound,azacitidine.
`
`2. Materials and Methods
`
`2.1. Preparation of 2',3',5'-'lriacetyl-5-Azacitidine (IAC)
`[21]. TAC was prepared through condensation of trimeth-
`ylsilylated-5-azacytosine
`and
`1,2,3,3-tetra-~O-acetyl-B-D-
`ribofuranose. In a 150 mL, 3-necked flask, a mixture of 5-
`azacytosine, hexamethyldisilazane, and ammonium sulfate
`was heated at reflux for 2 hours. A fresh amount of
`ammonium sulfate was added, and the reflux was continued
`for 6 hours. The excess hexamethyldisilazane was removed
`under
`reduced vacuum to afford trimethylsilylated 5-
`azacytosine as an off-white residuc.
`‘letra-O-acetyl-D-ribofuranose was prepared by adding
`acetic anhydride dropwise to D-ribose dissolved in dry
`pyridine. The solvent was removed by heating the solution
`at 50°C for 15 minutes, and tetra-O-acetyl-D-ribofuranose
`wascrystallized from isopropanol |22].
`
`Chemotherapy Research and Practice
`
`Trimethylsilylated 5-azacytosine and 1,2,3,5-Tetra-O-
`acetyl-$-D-ribofuranose were dissolved in acetonitrile, and
`the mixture was cooled at 0°C in an ice-water bath. Tri-
`methylsily] trifluoromethanesulfonate (TMSOT) was added
`slowly at 0°C then stirred at room temperature for 3
`hours. The reaction mixture was poured into a solution
`of NazCO; and NaHCO; in ice water and then extracted
`with dichloromethane. The combined organic layer was
`washed with cold H20, cold brine, and dried over anhydrous
`NazSO,. The residue, after removal of the solvent, was re-
`crystallized from a mixture of dichloromethane and hexane
`to provide the desired compound with an 85%yield.
`
`2.2. LC/MS/MS Analysis of TAC and Its Metabolites Mono-
`and Di-Acetyl-5-Azacitidine and ACT. To monitor the in
`vivo absorption, distribution, and metabolism of TAC, a
`highly sensitive LC/MS/MSanalytical method was developed
`that allows the quantitation of concentrations as low as
`10 ng/mL of TAC andits metabolites. The samples were pre-
`pared by adding 200uL of plasma to 400uL of cold
`acetonitrile, vortexed, and centrifuged at 14,000 rpm for 10
`minutes. The supernatant was placed into a 10 x 75 mm glass
`test tube and evaporated to 50uL under nitrogen at 37°C.
`The residue was then diluted with 1mL of 2% phosphoric
`acid and vortexed for 10 seconds at 3,000 rpm.
`Solid phase extraction was conducted using a Varian
`Bond Elute PLEXA PCX 1 mLcolumnsplaced on a vacuum
`manifold. Columns were activated by adding 1 mL of MeOH
`followed by |1mL H.O and ImLof 2% phosphoric acid.
`All samples were allowed to drip by gravity flow. ‘he
`extraction columns were then washed sequentially with 1 mL
`2% phosphoric acid, 1mL H2O, and 1mL MeOH/AcCN
`(1/1) and eluted into a 10 x 75 mmglasstest tube containing
`50uL of 25% formic acid in MeOH with 2 x 600uL of
`2% NH4OH in MeOH/AcCN (1/1). The liquid was then
`evaporated to dryness with nitrogen at 37°C, and 100 uL of
`mobile phase A was added and vortexed for 10 seconds at
`3,000 rpm and transferred to LC autosamplervials at 4°C.
`HPLC separation was conducted on a reverse phase
`C18 column (Varian Pursuit C18, 3 micron particle size;
`100mm x 2.0mm) using a Shimadzu SIL-HTc controller
`with dual LC-20AD pumps and DGU-20A3 degasser. The
`separation was achieved using a gradient of Mobile Phase A:
`0.1% Formic Acid and Mobile Phase B: 0.1% Formic Acid,
`90% MeOH with a flow rate of 400 ¢L/min at 40°C over
`10 minutes. MS/MS fragmentation was performed on an
`Applied Biosystems API 3200 instrument with a Turbo V Ion
`Spray ion source and Positive ion polarity.
`
`2.3. Drug Stability. ACY and ‘TAC stabilities were determined
`after incubation at pH1 (2% Phosphoric acid), pH3 (0.1%
`Formic Acid), pH5 (K3PO,4), and pH7.4 (Dulbecco’s PBS).
`100 ng of each compound was resuspended in 1.2 mL total
`volume, and 200 pL samples taken at 1, 3, 6, 29, and 48 hr.
`Each pH point was repeated 6 times in 2 dram vials with
`‘Teflon-lined screw caps at room temperature. Samples were
`extracted by solid-phase extraction using 100ng decitabine
`as an internal standard and analyzed by tandem mass spec-
`troscopy as described above.
`
`

`

`Chemotherapy Research and Practice
`
`2.4, Log P Determination. The OSIRIS Property Explorer
`software was used to generate initial cLogP values. The par-
`tition coefficient was determined by using an analyte concen-
`tration near the solubility limit for the compound in water
`and incubating in a water: octanol mix ranging from 0.5: 1,
`1:1, to 2:1. The pH of the water was 2 units above the pKa
`of the compound, preventing ionization. The experiment
`was conducted using the shake-flask method, after allowing
`octanol and water to equilibrate for 24 hours at 25°C.
`The initial drug concentration in water was 0.2mM. Tubes
`were rotated at a 45 degree angle for 10 minutes with a
`3 second rotation. The test was performed in a constant
`environment of 20°C-25°C. Tubes were centrifuged at 1000 ¢
`for 10 minutes, and the octanol phase was extracted. The
`UV/Visible absorption spectrum was evaluated from 550 nm
`to 210 nm to determine the absorption maximum. The ana-
`lyte concentration was determined and the LogP calculated:
`Log = log(concentration in octanol/concentration in water).
`
`2.5. Permeability across Caco-2 Monolayers. Caco-2 cells
`(ATCC) were maintained under standard conditions in Dul-
`becco’s modified Eagle’s medium (DMEM). Approximately 1
`x 10° cells (passage number 25-35) were seeded onto poly-
`carbonate cell culture inserts (Transwell, 0.45 um, 12mm
`diameter). The cells were allowed to growand differentiate
`for three weeks changing the medium every 2-3 days.
`Lucifer yellow was used to ensure appropriate paracellular
`permeability levels [23].
`The transport experiments were performed in Hanks’
`balancedsalt solution (HBSS) containing 25 mMHepes (pH
`7.4) as thoroughly described [24]. The drugs were dissolved
`in transport buffer to a final concentration of 0.2 mM. The
`monolayers were washed in prewarmed transport buffer for
`30min. The drug solutions were added to the donor side of
`the monolayers, and fresh transport buffer was added to the
`receiver side. The plates were gently shaken (50-100 rpm)
`at 37°C. At time intervals ranging from 5 minutes up to
`2h, 0.2mL samples were collected from the receiver cham-
`bers and replenished with fresh transport buffer. Sarnples
`containing the transported drug was collected for LC/MS
`analysis by immediately freezing at -80°C and subsequently
`extracted 2:1 with acetonitrile and analyzed as indicated in
`the LC/MS/MS methodologysection.
`
`‘To
`2.6. Pharmacokinetics and Metabolism of TAC in Mice.
`evaluate the in vivo pharmacokinetic properties of TAC,
`C57/BL6 female mice of 20g body weight were dosed with
`38 mg/kg ‘LAC p.o. (equimolar dose to 25 mg/kg of ACI’) or
`25 mg/kg ACT iv. via tail vein injection. TAC and ACT were
`solubilized in LX PBS immediately before administration and
`dosed at 0.01 ml/g fasted body weight. Blood samples (two
`time points per animal, three animals per time point) were
`obtained via retro-orbital bleeding with Natelson pipettes
`at 0.5, 1, 2, 4, 8, and 24 hours after drug administration.
`The collected blood was then centrifuged for 4 minutes
`at 13,000g. Plasma was immediately extracted with cold
`acetonitrile and stored at ~ 80°C until LC/MS/MSanalysis.
`
`Noncompartmental plasma pharmacokinetic parameters
`for TAC prodrug and ACT after oral TAC or iv ACT
`administrations were estimated using the sparse sampling
`option in WinNonlin Software version 4.1 (Pharsight Corp.,
`Mountain View, Calif, USA). Briefly, the WinNonlin sparse
`sampling option applies the linear trapezoidal rule for area
`under the concentration-time curve (AUC) calculations and
`generates a standard error for the mean AUC estimate
`that accounts for correlations in the data resulting from
`repeated sampling of individual mice. Parameters of interest
`included the AUC from time 0 to the last measurable
`concentration (AUCs), observed peak concentration after
`oral administration (Cmax), estimated peak concentration
`after iv. administration (Co), mean residence time to the
`last measurable concentration (MRT), and the half-life of
`the terminal phase (T1/.). To assess the multiphasic decline
`in plasma concentrations, an alpha (T12.) half-life was also
`estimated using a log-linear regression of the mean ACT
`concentrations through 2 hoursafter dosing.
`
`2.7, Global Methylation Detection Assay. 24 hours follow-
`ing drug administration, snap-frozen mouse tissues were
`homogenized and genomic DNA isolated using Promega’s
`Wizard Genomic DNA Purification Kit. 100ng of EcoRI
`linearized genomic DNA wasspotted onto PVDF membrane.
`‘The membrane wasair-dried and repermeabilized according
`to manufacturer’s instructions. Nonspecific binding was
`blocked using 5% milk in PBS-T and washed twice in
`PBS-T for 5 minutes.he membrane was incubated with
`primary antibody (1: 1000 anti-5-methylcytosine in PBS-T,
`Calbiochem) for 1 hour then washed four timesfor five min-
`utes each with PBS-T. The membrane was incubated with the
`HRP-conjugated secondary antibody for 1 hour and washed
`four times for five minutes each with PBS-T. Signalintensity
`following ECL detection was quantitated using KODAK
`Image Station software. Assay results were confirmed using
`EpiGentek’s Methylamp Global DNA Methylation Quantifi-
`cation Kit per manufacturer’s instructions.
`
`2.8 pISINK4B Methylation Status. Mouse leukemia cells
`(L1210) were purchased from ATCC. The published [C50
`value for ACT (0.2uM) was first confirmed using the
`Promega CellTiter MTS assay. Cells were treated with ACT
`(0.1 or 1 uM) or TAC (1 or 100 uM) tor 48 hours, with tresh
`drug added daily. Genomic DNAwasisolated after 48 hours
`using the Sigma GenElute Genomic DNA Kit, including
`the RNase treatment step to ensure no RNA carryover. The
`methylation status of plISINK4B was determined using a
`predesigned primer set and the EpiTect Methyl qPCR Assays
`MethylScreen technology (Qiagen, Frederick, Md, USA).
`
`2.9. Antileukemic Activity of Triacetyl-5-Azacitidine. The in
`vivo L1210 lymphocytic leukemia model
`in C57BL/6 x
`DBA/2 FI(BDF1) female mice was used to evaluate the
`in vivo antiproliferative effect of TAC. L1210 cells were
`carried in BDF1 mice by weekly ip. (intraperitoneal) pas-
`sages. Leukemia cells from ascites fluid were diluted ap-
`propriately in RPMI
`1610 medium and injected ip.
`
`

`

`4
`
`Chemotherapy Research and Practice
`
`TaBLeE1: Stability of TAC and its metabolites at various pH.
`
`Sample
`TAC (%)
`DAC (%) MAC(%)
`ACT (%)
`
`Control
`100
`
`0
`0
`1
`99
`pH i—1hr
`0
`4
`22
`74
`pH 1—3hr
`0
`Al
`41
`38
`pH 1—6hr
`46
`3]
`22
`1
`pH 1—29 hr
`0
`0
`3
`97
`pH 3—1 hr
`0
`0
`4
`96
`pH 3—3hr
`0
`2
`6
`92
`pH 3—6 hr
`1
`2
`4
`93
`pH 3—29 hr
`0
`0
`2
`98
`pH 5—1 hr
`0
`0
`6
`94
`pH 5—3hr
`0
`0
`2
`98
`pH 5—6 hr
`1
`1
`4
`94
`pH 5—29 hr
`0
`0
`4
`96
`pH 7.4—1 hr
`0
`2
`7
`91
`pH 7.4—3 hr
`0
`3
`12
`85
`pH7.4—6 hr
`1
`13
`Vw
`69
`pH 7.4—29 hr
`TAC:tri-acetyl-azacitidine; DAC: di-acetyl-azacitidine; MAC: mono-acetyl-
`azacitidine; ACT:azacitidine.
`
`TAC (26%) to the mono- and diesters in the first 3 hours
`of incubation increasing to 60% after 6 hrs. However, after
`24, 99% of TAC was hydrolyzed, including 46% to the parent
`compound, ACT.
`A similar experiment to establish the stability of azaci-
`tidine in solution resulted in significant degradation of the
`nucleoside analog throughout the pH range with maximum
`stability at pH 1. As previously determined,
`[25]
`the
`initial degradation to N-formylguanylribosylurea followed
`bythe irreversible formation of guanylribosylurea resulted in
`two derivatives with no pharmacological activity (data not
`shown).
`
`3.2. LogP Determination. The logP value of a compound is
`a well established measure of a compound’s hydrophilicity.
`The theoretical physical-chemical properties of TAC were
`evaluated by measuring the partition coefficient of the un-
`ionized molecules in two immiscible phases of water and
`octanol. Azacitidine resulted in an experimental LogP value
`of —2.32 compared to the calculated cLogP values of —2.17
`while the changes to the molecule by acetylation resulted in
`an improved LogP value for TAC of —0.61 versus a cLogP of
`—0.85.
`
`3.3. Permeability across Caco-2 Monolayers. The human
`colon carcinoma Caco-2 cell model was used to evaluate the
`oral absorption of the prodrug across membranes anddeter-
`minethe permeability coefficient (absorption rate constant).
`A sigmoidal relationship exists between drug absorption
`rates as measured with the in vitro Caco-2 model and human
`absorption [26]. Permeability coefficients typically range
`from 5 x 10°* to 5 x 107° cm/s. Drugsthat are well absorbed
`in humans have permeability coefficients greater
`than
`
`QL
`
`NAN
`aaisenyy?OIMS
`H
`1
`
`H2NV
`
`ZN
`
`ey °
`‘ Y 1
`Q
`<4 Tt
`>Q_p NEN
`y-6 a
`oO
`2 T 5
`oO
`TMSOT£ MeCN
`‘-5 a
`o ;
`
`Ficure 1: Chemical synthesis of 2’,3’,5’-triacetyl-5-Azacytidine.
`Results of analytical IH NMR (CDCI3, 500 MHz): d 8.18{s, 1H),
`7.69(s, 1H), 6.33(s, 1H), 5.82(d, 1H, J = 3.0Hz), 5.54(t, 1H, J =
`4.0 Hz), 5.41(t, 1H, J = 6.0Hz), 4.30-4.41(m, 3H), 2.11{s, 3H),
`2.10(s, 3H), and 2.08(s, 3H) ppm. 13C NMR (CDCI3, 125 MHz):
`d 170.3, 169.6, 169.5, 166.0, 156.0, 153.1, 89.7, 79.9, 73.7, 69.9, 62.8,
`20.7, 20.4, and 20.3 ppm.
`
`(110° cells/0.1 mL/mouse) into recipient mice. A daily oral
`administration of TAC (38 mg/kg via oral gavage) or ACT
`(25 mg/kg administered i.p.) diluted in PBS was given to
`leukemic BDF1 mice starting 24 hours from the inoculation
`of L1210 cells for a total of 5 days. Compounds were prepared
`fresh daily. A group of untreated leukemic mice received
`sterile water for injections and served as controls.
`
`2.10. TAC Toxicology. The in vivo toxicity of TAC waseval-
`uated in CD-1 mice using 3 groups of 3 male and 3 female
`CD-1 mice per group. CD-1 is the classical mouse strain
`chosen for toxicology studies including safety and efficacy.
`On Days 1-5 and Days 8-12, animals were administered
`vehicle (sterile water for injection, Group 1), 38 mg/kg/day
`TAC (Group 2), or 76 mg/kg/day 'TAC (Group 3) via oral
`gavage. Animals were not dosed on Days 6 and 7 and were
`euthanized on Day 13 for necropsy. Doses were prepared
`fresh daily from the drug stock. Criteria for evaluation
`included clinical observations, body weights, limited serum
`chemistry and hematology parameters, and histopathology
`evaluation.
`
`3. Results
`
`3.1. Synthesis and In Vitro Characterization of Triacetyl-5-
`Azacitidine. 2',3',5'-triacetyl-5-azacitidine has been success-
`fully synthesized through condensation oftrimethylsilylated-
`5-azacytosine and 1,2,3,5-tetra-O-acetyl-$-D-ribofuranose
`(Figure 1). An HPLC detection assay was optimized for TAC
`and ACTas well as somepotential analytes.
`The solubility of TAC in comparison to ACT was
`evaluated at various pH. ‘TAC and ACT were equally soluble
`at pH1 (40 mg/mL) and pH3 (25 mg/mL), while TAC was
`more soluble than ACT at pH5 (30 mg/mL versus 15 mg/mL)
`and pH7 (30 mg/mLversus 10 mg/mL). Thestability of the
`prodrug across a range of pHs is indicated in Table 1. TAC
`shows remarkable stability at pH 3, 5, and 7.4 with no
`degradation to the parent compound and 30% hydrolysis to
`the mono- anddiesters only at pH 7.4 after a 29 hr incubation
`at 25°C. At pH 1, we observed a moderate hydrolysis of
`
`

`

`100
`
`— Oo
`
`
`
`Concentration(uM)
`
`0.1
`
`0
`
`6
`
`12
`
`18
`
`24
`
`Time(hours)
`
`38 mg/kg p.o.
` [tin|6s]2|NE
`
`(a)
`ACT
`25 mg/kg iv.
`
`TAC
`
`Chemotherapy Research and Practice
`
`1 x 107° cm/s, while drugs and peptides that are absorbed
`to less than 1% have permeability coefficients less than
`1 x 10°’ cm/s [27, 28]. Between these ranges, compounds
`are considered to have intermediate absorption rates. In
`these studies, ACT was determined to have a permeability
`coefficient between 1-3 x 10°°, compared to 5~7 x 107°
`for ‘TAC. This indicates a potential 2-3 fold improvement
`in intestinal absorption. The efflux ratio is defined as the
`quotient of the secretory permeability and the absorptive
`permeability. ‘The efflux ratio was near 1.0, indicating these
`compoundsare not beingactively effluxed. No hydrolysis of
`the prodrug to azacitidine was observed during the course of
`this experiment.
`
`3.4. Pharmacokinetics and Metabolism of TAC in Mice. TAC
`was administered at 38 mg/kg to C57/BL6 mice via oral gav-
`age, an equimolar dose equivalent to 25 mg/kg of azacitidine.
`TAC andits active metabolite ACT were detectable in plasma
`at 30 minutes after the oral administration (Figure 2(a)).
`'TAC appearedto be rapidly deacetylated leading to a minimal
`accumulation of the prodrug resulting in plasma concen-
`trations below the limit of quantitation (10 ng/mL) at time
`points beyond 1 hour after administration. T[AC-derived
`azacitidine reached a peak concentration of approximately
`5,000 ng/mL (~20 uM) at 30 minutes with a pharmacologi-
`cally relevant concentrationofat least 0.5 uM being sustained
`for 24 hours after oral 'TAC dosing. The mean TAC and
`‘TAC-derived ACT plasma concentrations versus time plots
`after oral administration of TAC prodrug are depicted in
`Figure 2(a) with the pharmacokinetic parameters reported in
`Figure 2(b). ACT was also administered i.v. as a control to
`emulate the standard meansof administration in the clinical
`setting. The 'T1/2 and MRT: values for 'TAC-derived azaciti-
`dine were 9.2 hours and 7.7 hours, respectively, whereas iv.
`administration of ACT resulted in Ty and MRTias values
`of 6.8 hours and 1.1 hours, respectively. The AUC;values
`for plasma azacitidine were 73.5 hr-uM and 126 hr-uM after
`oral administration of TAC prodrug or iv. administration
`of ACT, respectively. While the absolute bioavailability of
`derived azacitidine after oral TAC administration is 58%, the
`prolonged Ty/2 and MRTast values for azacitidine observed
`in plasmaafter oral TAC administration suggest a protracted
`absorption and conversion of the TAC prodrug at
`the
`gastrointestinal or presystemic level (Figure 2(b)). The log-
`linear regression analysis of ACTconcentration data through
`2 hours revealed an increased Ti.q value for ACT after p.o.
`‘TAC administration compared with i.v. ACT administration
`(0.73 hr versus 0.32 hr, resp.), also suggesting protracted
`absorption and both presystemic and systemic conversion of
`TAC.
`
`3.5. Pharmacodynamic Effect of Triacetyl-5-Azacitidine. MTS
`assays (Promega, Madison, Wis, USA) were performed to
`determine the in vitro effects of ACT and TAC on L1210
`cell proliferation. TAC had no effect on L1210 cell viability
`even at doses 100-fold higher than the known IC50 of ACT
`(0.2 uM) [29], and this is an indication that this compound
`does notget activated in vitro. This result was predicted,since
`
`(b)
`
`Figure 2: Pharmacokinetics of oral TAC versusi.v. 5-azacitidine in
`C57BL/6 mice. (a) Mean plasma concentration versus time for tri-
`acetyl azacytidine (TAC) (4) and resultant ACT (*) in nontumor
`bearing mice after a 38mg/kg dose of oral TAC. Mean plasma
`concentration versus time for ACT in nontumor bearing mice after
`25 mg/kg ACTi.v. (c). Error bars indicate standard deviation. (b)
`Noncompartmental plasma pharmacokinetic parameters for azac-
`itidine (ACT) andtri-acetyl azacitidine (TAC). AUCys values are
`mean (standard error). NE—not estimated. NC—notcalculated.
`
`cells in culture lack the necessary esterase activity to convert
`the prodrug to ACT that remains below thelimit of detection
`using our LC-MS method.
`Gene-specific methylation PCR was performed in order
`to confirm whether TAC could affect the methylation level of
`a specific target even in the absenceofcellular toxicity. ACT,
`which is the active derivative of TAC, has been extensively
`studied by numerous research groups to determine specific
`cellular methylation targets and mechanism of action [30,
`31]. PISINK4B is a classic target known to be hyper-
`methylated in AML and MDSandattributes to a poor
`prognosis in those patients [32, 33]. 0.1uM and 10uM ACT
`decreased P15 promoter methylation by 39% (+3) and 49%
`(+17), respectively. In comparison, 1uM and 100 uM ‘TAC
`decreased P15 promoter methylation by only 19% (+8) and
`23% (+16), respectively.
`
`

`

`ACT
`
`TAC
`Average
`
`Gut.2
`100
`14
`30
`
`Gut.3
`100
`34
`41
`
`Gut.4
`100
`33
`25
`
`Average
`100
`29
`38
`
`Spleen.1
`100
`54
`48
`
`Spleen.2
`100
`71
`31
`
`Spleen.3
`100
`48
`62
`
`Spleen.4
`100
`29
`22
`
`100
`50
`41
`
`(b)
`
`Ficure 3: Effect of DNMT inhibitors on global DNA methylation
`in tissues of mice exposed for 5 days to ACT or TAC. (a) Dot blot
`of mouse tissues isolated 24 hours after treatment with 25 mg/kg
`iv. ACT or 38 mg/kg p.o. TAC. Groups were compared to untreated
`animals (unt). (b) Densitometry of methylation levels represented
`in (a).
`
`In an in vivo experiment designed to evaluate the effect
`ofa prolonged administration of TAC on global methylation,
`TAC (38me/kg p.o.) or ACT (25mg/kg ip.) diluted in
`PBS was administered to C57BL6 mice every 24 hours for
`5 consecutive days (four animals per group). Tissue methyla-
`tion levels are reported in Figure 3. A significant reduction
`in global DNA methylation was detected in the gut and
`spleen after the administration of TAC, comparable to the
`methylation decrease detected after the administration of
`ACT. We did not observe a significant change in global
`methylation status in kidneyorliver tissue (data not shown).
`The positive results in gut and spleen were confirmed
`using the EpiGentek’s Methylamp Global DNA Methylation
`Quantification Kit. This quantitative analysis confirmed a
`comparable 50%-60% decrease in global methylation by
`both TAC and ACT (data not shown).
`
`3.6. Antileukemic Activity of Triacetyl-5-Azacitidine. The
`L1210 lymphocytic leukemia model was used to determine
`the efficacy of TAC in an animal model. The L1210 model
`is a classical model used to predict for compounds that
`are effective against leukemias and lymphomas. A daily oral
`administration of TAC (38 mg/kg equimolar to 25 mg/kg
`of ACT and a higher dose of 50mg/kg) or azacitidine i.p.
`(25 mg/kg) was given to leukemic mice for a total of 5 days.
`The median survival time for the untreated control group
`was 8 days, while the median survival time for the TAC
`
`Chemotherapy Research and Practice
`
`— Control
`—#— 25 mg/kg AC
`—#- 38 mg/kg TAC
`
`Days
`
`(a)
`
`Toxicity curve
`
`120
`100
`
`Survivial
`
`
`
`Averageweight
`
`(%)
`(g)
`
`Days
`
`—e Control
`—=-— 25mg/kg ACT
`
`—*— 38mg/kg TAC
`—— 50me/kg TAC
`
`(b)
`
`Figure 4: (a) Antileukemic activity ofTAC and ACT in BDF1 mice
`bearing L1210 lymphocytic leukemiacells after 5 days of treatment
`with indicated concentrations of oral TAC or i-p. ACT. (b) Toxic
`effect as indicated by average weightloss of the same tumorbearing
`mice.
`
`treated groups was 12 days resulting in a 50% increased
`lifespan, demonstrating not only an epigenetic modulating
`effect, but also antileukemic activity. Azacitidine at
`the
`maximum tolerated dose of 25mg/kg administered i.p.
`resulted in a 17-day survival with a 112% increased lifespan
`(Figure 4(a)). The toxicity curve (Figure 4(b)) indicates a
`nonsignificant weight loss for the TAC-treated groups at day
`3 followed by a rapid recovery indicating the lack of toxicity
`of this prodrug coupled with a significant antileukemiceffect,
`while the azacitidine-treated animal showed a 16% weight
`reduction with a significant recovery once the treatment was
`completed.
`
`3.7. TAC Toxicology. The in vivo toxicity of TAC was eval-
`uated by orally administering repeated doses to CD-1 mice
`for two weeks. On Days 1-5 and Days 8-12, animals
`were administered vehicle, 38mg/kg/day TAC (Group 2,
`equivalent
`to 25mg/kg of ACT), or 76mg/kg/day 'TAC
`(Group 3) via oral gavage. Criteria for evaluation included
`clinical observations, body weights, limited serum chemistry
`
`

`

`Chemotherapy Research and Practice
`
`
`
`60 qr ee
`y
`
`
`
`Control(%)
`
`y
`4------- FA
`40
`39 J... 7 ee eee
`2004 _---- on toes
`-
`be
`:
`
`
`RTC WBC
`
`NEU
`
`@ 38 mg/kg TAC
`GO 76meg/kg TAC
`
`Figure 5: TAC-assuciated effect on hematological parameters
`following TAC administration to CD-1 mice. RTC: reticulocyte
`count, WBC: white blood cell count, NEU: neutrophil count, LYM:
`lymphocyte count, MNO: monocyte count, EOS: cosinophil count,
`LUC: large unstained cell count.
`
`and hematology parameters, and histopathologyevaluation
`of a limited number of tissues from vehicle-treated and
`76 mg/kg/day TAC-treated animals.
`There were no deaths and no notable 'TAC-associated
`body weight changes during the study. Additionally, there
`were no TAC-associated changes in liver or renal function
`markers, including alanine and aspartate aminotransferases,
`alkaline phosphatase, bilirubin, blood urea nitrogen, and
`creatinine.
`TAC-associated changes in various hematological param-
`eters were observed (Figure 5). This included a reduction in
`white blood cell counts, absolute lymphocytes, and absolute
`leukocytes by 41%, 31%, and 24%, respectively, for the
`38 mg/kg/day dosage group. The reduction values for the
`76 mg/kg/day group only slightly increased by 44%, 31%,
`and 34%,
`respectively. There were 'TAC-related findings
`in the bone marrow (hypocellularity) and in the lymph
`nodes (inactive germinal centers) which reflect the decreased
`overall white blood cell count reductions. An additional
`microscopic '[AC-related finding was duodenum apoptosis
`of the crypt epithelium in the 76 mg/kg/day-treated animals.
`Other observations included a decrease in myeloid andery-
`throid compartmentcellularity correlating with microscopic
`findings in varioustissues.
`
`4, Discussion
`
`Epigenetic modulators are appealing cancer therapeutics due
`to their potential reversibility and preference for highly
`proliferating cancer cells. Azacitidine and decitabine have
`demonstrated improved clinical success in hematologic ma-
`lignancies with longer or continuous infusion [34]. However,
`the aza-pyrimidine ring is unstable in aqueous solutions and
`has poor bioavailability, limiting the use of oral formulations
`
`s
`
`that would be more convenientfor thepatients, help in long-
`term dosing, and reduce local side effects when administered
`as subcutaneous depot.
`We have successfully synthesized an azacitidine pro-
`drug, 2',3’,5’ -triacetyl-5-azacitidine and shownits favorable
`physical-chemical characteristics. TAC

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