throbber
[CANCER RESEARCH 46, 5511-5517, November 1986]
`Differences
`in DNA Damage Produced by Incorporation of 5-Aza-2'-deoxycytidine
`
`or 5,6-Dihydro-5-azacytidine
`
`into DNA of Mammalian Cells
`
`Joseph M. Covey,1 Maurizio D'Incaici,2 Eugene J. Tilchen, Daniel S. Zaharko, and Kurt W. Kohn
`
`Laboratory of Molecular Pharmacology [J. M. C., M. D'I, E. J. T., K. W. K.], and Developmental Therapeutics Program, Division of Cancer Treatment [J. M. C.,
`M. D'I., E. J. T., D. S. Z., K. W. K.], National Cancer Institute, NIH, Bethesda, Maryland 20892
`
`ABSTRACT
`(aza-dCyd) and 5,6-dihydro-5-
`The effects of 5-aza-2'-deoxycytidine
`azacytidine (H2-aza-Cyd) on the integrity of DNA from several mam
`malian cell
`lines were compared using the alkaline elution technique.
`While both compounds have been shown to inhibit DNA methylation, a
`direct comparison of their effects on DNA structure has not previously
`been reported. Exposure of I 121(1cells to ll,-:i/a-( yd (1-100 fig/ml)
`and simultaneous labeling with [I4C|thymidine for 24 h resulted in the
`production of single-strand breaks
`in DNA, which were significantly
`repaired when cells were incubated in drug-free medium for an additional
`24 h. This differed from our previous findings for aza-dCyd, confirmed
`here in parallel experiments, which showed that this compound produces
`alkali-labile lesions that persist for 48 h. The DNA effects of both drugs
`were significantly reduced when cells were prelabeled with [l4C]thymidine,
`indicating that production of DNA lesions requires incorporation of the
`anomalous base. Studies utilizing pulse-labeled DNA indicated that aza-
`dCyd has little effect on the rate of DNA elongation, whereas H2-aza-
`Cyd produced a complete inhibition for at least 6 h after drug removal.
`The contrasting pattern of DNA damage induced by these compounds in
`LI 210 was also observed in two human lymphoblastoid cell lines, one of
`which was derived from a patient with xeroderma pigmentosum. We had
`previously concluded that alkali-labile sites in DNA from aza-dCyd-
`treated cells probably arise due to the chemical instability of aza-dCyd.
`In contrast,
`incorporated H2-aza-Cyd is chemically stable. The single-
`strand breaks produced in H2-aza-Cyd treated cells were not of the alkali-
`labile type, and may represent an accumulation of DNA replication
`fragments and/or intermediates
`in an excision repair process. Thus,
`the
`DNA lesions produced by the two drugs have markedly different char
`acteristics, and H2-aza-Cyd should not be considered to be merely a stable
`pharmacological congener of aza-dCyd.
`
`by cytidine kinase from
`(15). This compound is phosphorylated
`L1210 and HeLa cells, although less efficiently than is azacy
`tidine (16, 17), yet is incorporated into LI210 nuclear RNA at
`levels equal
`to or exceeding those of azacytidine (18). Studies
`demonstrating
`effects of H2-aza-dCyd on gene expression sug
`gest that
`it is also incorporated
`into DNA and inhibits DNA
`methylation ( 19), although it may be less active than aza-dCyd.
`While recent
`reports have demonstrated
`a correlation
`be
`tween inhibition of DNA methylation and aza-dCyd cytotox-
`icity (20), it is not certain that
`this is the unique mechanism of
`action for these compounds. We have shown that aza-dCyd
`treatment
`of LI210
`leukemia cells /// vitro results
`in alkali-
`labile sites in DNA as demonstrated
`by the alkaline elution
`technique
`(21). These DNA lesions increase during 24-48 h
`following drug removal and are then slowly removed over the
`next 48 h. It was proposed that
`this DNA damage results from
`the chemical
`instability of aza-dCyd incorporated
`into DNA.
`In this paper we compare
`and contrast
`the DNA lesions
`produced by aza-dCyd and the stable analogue H2-aza-Cyd in
`LI210 leukemia and in two human lymphoblastoid cell lines as
`measured by alkaline elution. We report here differences
`in
`both the nature of the lesions induced by these two drugs and
`in their persistence in DNA. It appears
`therefore that H2-aza-
`Cyd should not be considered a pharmacological
`analogue of
`aza-dCyd.
`
`MATERIALS AND METHODS
`
`INTRODUCTION
`aza-dCyd3 ( 1) has been shown to have cytotoxic and antineo-
`plastic activities
`in a number of experimental
`tumor
`systems
`(2-5) and has undergone clinical
`trial (6). aza-dCyd is anabol-
`ized to the nucleoside triphosphate
`(7) and is incorporated into
`DNA (8-10) but not into RNA (9). The presence of azacytosine
`moieties in DNA (even a small percentage substitution of DNA
`cytosines)
`results in inactivation of DNA-cytosine-methyltrans-
`ferase, an enzyme which catalyzes
`the transfer of a methyl
`group to the 5-position
`of cytosines
`in DNA (10-12). The
`hypomethylation
`of DNA produced by aza-dCyd and related
`compounds
`have been shown to result
`in activation of latent
`cellular genes (13).
`Since azacytidine and aza-dCyd spontaneously decompose in
`aqueous solution (14) leading to difficulties in pharmaceutical
`formulation,
`the stable analogue H2-aza-dCyd was synthesized
`
`accepted 8/6/86.
`revised 4/24/86;
`Received 6/3/85;
`The costs of publication of this article were defrayed in part by the payment
`of page charges. This article must
`therefore be hereby marked advertisement
`in
`accordance with 18 U.S.C. Section 1734 solely to indicate this fact.
`1To whom requests for reprints should be addressed, at Laboratory of Molec
`ular Pharmacology, Developmental Therapeutics Program, National Cancer In
`stitute, NIH, Building 37, Room 5A19, Bethesda, MD 20892.
`2On leave from Istituto di Ricerche Farmacologiche
`"Mario Negri," Milan
`Italy.
`3The abbreviations used are: aza-dCyd, 5-aza-2'-deoxycytidine; H2-aza-Cyd,
`5,6,-dihydro-5-azacytidine;
`ara-C,
`l-/3-D-arabinofuranosylcytosine;
`XP, xero
`derma pigmentosum.
`
`Cell Culture. Mouse 11210 leukemia cells were grown at 37*C in
`suspension culture in RPMI 1630 (HEM Research, Inc.) supplemented
`with 20% heat-inactivated horse serum (GIBCO, Grand Island, NY),
`0.84 HIML-glutamine, penicillin (82 units/ml),
`streptomycin (82 ng/
`ml) (all from the Media Unit, NIH), and SO/<\i mercaptoethanol
`(Sigma
`Chemical Co., St. Louis, MO). Stock cultures were maintained
`in
`exponential growth at a density between 0.5 x 10s and 1 x 10' cells/
`ml.
`Two human lymphoblastoid cell lines were also used in these studies
`(Institute for Medical Research, Camden, NJ). One line (GM 3714)
`was derived from a clinically normal
`female, while the other
`(GM
`2345A) was obtained from an 8-year-old female with xeroderma pig
`mentosum (complementation group A). Both lines were maintained in
`vitro at 37°Cin RPMI 1630 medium (GIBCO) supplemented with 15%
`heat-inactivated fetal calf serum.
`Labeling. In most experiments cells were labeled for 24 h concurrent
`with drug treatment using [2-14C]thymidine (specific activity, 52-58
`mCi/mmol; New England Nuclear) at a concentration of 0.02 uC\/m\.
`For some experiments, LI210 cells were labeled for 24 h ([2-uC]-
`thymidine, 0.02 ¿iCi/ml),washed 2 times with fresh medium, and then
`treated with H2-aza-Cyd or aza-dCyd for 24 h.
`Drug Treatment. aza-dCyd was provided by Dr. D. DeVos, Phar-
`machemie B. V. (Haarlem, Holland), and by the Drug Synthesis and
`Chemistry Branch, National Cancer
`Institute (Bethesda, MD), which
`also provided H2-aza-Cyd. aza-dCyd is unstable in aqueous solution
`and decomposes in a temperature-
`and pH-dependent manner
`(14) to
`form several noncytotoxic products (4). In RPMI 1630 medium at 37"C
`aza-dCyd decomposed in a first-order
`fashion with a fi/2of 17.5 h. H2-
`aza-Cyd has been shown to be stable for weeks in aqueous solution at
`room temperature (15). For in vitro experiments, both compounds were
`CELGENE 2074
`5511
`APOTEX v. CELGENE
`IPR2023-00512
`
`

`

`DNA DAMAGE PRODUCED BY Aza-dCyd AND H2-aza-Cyd
`
`pH 12.1
`
`1.0
`0.9
`0.8
`0.7
`0.6
`
`0.5
`
`0.4
`
`0.3
`
`0.2
`
`0.1
`0'.9
`
`OO t
`
`oc
`I
`
`0.8
`9
`=* 0.7
`H,
`O 0.6
`
`5
`
`0.5
`
`0.4
`
`0.3
`
`0.2
`
`pH 12.1
`24 Ht posi drug incubation
`
`pH 12.6
`24 Hi posi drug ncubation
`
`\ W•i
`
`300 Radi
`
`0.1
`O
`
`5.0
`
`10
`
`5.0
`
`10
`
`15
`
`O
`15
`TIME OF ELUTION (Hr)
`Fig. 1. Alkaline elution profiles of [14C]DNA from L1210 cells treated for 24
`h with 11;-a/;i i 'yd at the indicated concentrations
`(/tg/ml). Elutions were carried
`out at pH 12.1 (I and Q or pH 12.6 (B and /') either
`immediately after drug
`washout (A and ÃŒI)or following 24 h postincubation in drug-free medium (C and
`D). Labeling with ["CJthymidine
`(0.02 /iCi/ml) and treatment with H2-aza-Cyd
`were simultaneous. Points, mean of two to three values; bars, range;
`, elution
`of [14C]DNA from untreated L1210 after exposure to 300 rads in the cold.
`
`saline (0.15 M NaCl-10.6 min
`dissolved in sterile phosphate-buffered
`KH2PO4-5.6 HIMNa2HPO4) and then immediately added to cell sus
`pensions
`(~5.0 x IO5 cells/ml)
`to give a final drug concentration
`between 0.1 and 100 Me/ml. After drug treatment,
`the cells were washed
`twice by centrifugation and resuspended in fresh medium. Cell density
`was determined using a Coulter Counter
`(Model ZBI, Coulter Elec
`tronics, Hialeah, FL), and aliquots of cells were removed for alkaline
`elution analysis. In some experiments,
`remaining cells were diluted as
`necessary to maintain cell density below S x 10s cells/ml and incubated
`in drug-free medium for an additional 24 h. At the end of this period,
`appropriate aliquots were again removed for alkaline elution.
`Alkaline Elution. The method of alkaline elution has been reviewed
`in detail (22). In brief, approximately
`IO1'cells were resuspended in
`cold phosphate-buffered saline and layered on polycarbonate filters, 0.8
`ftm pore size, 25 mm diameter
`(Nuclepore Corp., Pleasanton, CA).
`Cells were then lysed with a solution containing 2% sodium dodecyl
`sulfate-0.02 M disodium EDTA-0.1 M glycine, pH 10.0 (lysis solution),
`which was allowed to flow through the filter by gravity. After connecting
`the outlet of the filter holders to the pumping system, 2 ml of proteinase
`K, 0.5 mg/ml (EM Labs, Darmstadt, West Germany), dissolved in the
`lysis solution, were added to a reservoir over the polycarbonate
`filters
`and pumped for approximately
`l h at 2 ml/h. DNA was eluted from
`the filters by pumping 0.02 M EDTA solution adjusted to pH 12.1 or
`12.6 with tetrapropylammonium
`hydroxide (RSA Corp., Elmsford,
`NY) containing 0.1% sodium dodecyl sulfate through the filters at
`approximately 2 ml/h. Fractions were collected at 3-h intervals with
`fractions and filters processed as previously described (22). In most
`experiments, L1210 cells labeled overnight with [mer/ry/-3H]thymidine
`(specific activity, 20 Ci/mmole; 0.1 ¿iCi/mlcells; New England Nuclear)
`and irradiated with 300 nuls of y-radiation were added to suspensions
`of l4C-labeled experimental cells to serve as internal elution standards.
`Elongation Experiments. Exponentially growing L1210 cells (5-6 x
`IO5cells/ml) were pulse labeled for 15 min with [iwe/A}'/-3H]thymidine
`(1.0 /iCi/ml cells), washed twice by centrifugation,
`and resuspended in
`warm RPMI 1630 medium. aza-dCyd, H2-aza-Cyd, or ara-C was added
`and the cells were incubated at 37°Cfor 1 h. Cells were then washed,
`resuspended, and maintained at 37°C.Aliquots were removed at various
`times and analyzed by alkaline elution as described above, except
`that
`[l4C]thymidine (0.02 ¿iCi/ml)-IabeIedcells were used as internal stand
`ard.
`Flow Cytometry. LI210 cells were treated with aza-dCyd or H2-aza-
`Cyd as described above. The analysis of DNA by flow cytometry was
`performed at the end of 24 h treatment and after 24 h postincubation
`in drug-free medium. Before analysis, cells were washed and suspended
`in Hanks' solution. Cells were stained by adding 3 ml propidium iodide
`(Calbiochem-Behring, San Diego, CA) (propidium iodide, 50 Mg/ml in
`0.1% sodium citrate plus 30 n\ Nonidet P-40) to 1(10/iI cell suspension
`and were stored at 4"C for 30 min before DNA analysis. Absence of
`aggregates
`and the suitability of the preparation were checked by
`fluorescence microscopy before the samples were run. DNA analysis
`was performed using a 30L cytofluorograph (Ortho Instruments, West-
`wood, MA). The fluorescence pulses were detected in a spectral range
`between 580 and 780 nm (to exclude the overlapping region of excita
`tion and emission spectra of unbound propidium iodide) and then
`integrated. Each cytofluorometric
`assay was performed with 5 x IO4
`cells. The percentage of cells in each cycle phase was calculated by the
`method of Maisch et al. (23).
`
`RESULTS
`
`strand breaks must occur in regions of DNA which were syn
`thesized while drug was available for incorporation. Production
`of single-strand breaks was concentration
`dependent up to 100
`tig/ml. Treatment with H2-aza-Cyd concentrations
`above this
`level caused increasing
`cell
`lysis during drug washout
`and
`resulted in elution curves with markedly reduced retention in
`the first fraction (data not shown). Incubation of cells in drug-
`free medium for 24 h resulted in some repair of the single-
`strand breaks, as evidenced by the reduced elution rates shown
`in Fig. 1, C and D. Repair was incomplete, with damage
`produced by the higher H2-aza-Cyd concentrations
`diminishing
`to near the level initially produced by 5 tig/ml. Single-strand
`breaks produced by the latter drug concentration were only
`slightly reduced after 24 h drug-free incubation. This pattern
`of repair
`is in contrast
`to the results obtained with aza-dCyd
`treated cells (Fig. 2); 24 h postdrug incubation resulted in either
`little change or an increased elution rate, with no significant
`repair at any drug concentration
`over this time period.
`In addition,
`the elution profiles demonstrated
`a fundamental
`difference in the nature of DNA damage produced by these two
`agents. The marked increase in elution rate with increasing pH
`following aza-dCyd treatment,
`coupled with the characteristic
`convex shape of the elution curves (21, 22), demonstrates
`the
`formation
`of alkali-labile
`sites in the DNA.
`In contrast,
`the
`elution curves obtained after H2-aza-Cyd treatment were essen
`tially linear and showed only a minimal pH dependency.
`The effects of aza-dCyd and H2-aza-Cyd on L1210 DNA
`
`elution profiles (at pH 12.1 and 12.6) of DNA
`Representative
`from LI210 cells treated for 24 h with H2-aza-Cyd (5-100 fig/
`ml) are shown in Fig. 1. The data in Fig. 1, A and B were
`obtained at the end of drug treatment, whereas those in Fig. 1,
`C and n were obtained following an additional 24 h incubation
`of the cells in drug-free medium. The increased elution rate of
`DNA from H2-aza-Cyd-treated
`cells is indicative of single-
`strand breaks in the DNA. Since cells were exposed to [14C]
`thymidine and H2-aza-Cyd simultaneously,
`the observed single-
`5512
`
`

`

`DNA DAMAGE PRODUCED BY Aza-dCyd AND H2-aza-Cyd
`
`0.9
`0.8
`0.7
`0.6
`
`0.5
`
`0.4
`
`0.3
`
`Å’ 0.2
`
`pH 12.1
`
`\ 300 Rad»
`
`pH 126
`
`0
`
`1.0
`
`10
`H a/a C („gml)
`
`100
`
`24+24 Hi, pH 12 6
`
`24+24 Hr, pH 12.1
`24 Hi. Pre-tabeted,
`pH 12.6
`24 Hi. Pre-latated.
`pH 12.1
`
`24+24 Hr, pH 12.6
`
`0.1
`
`0.9
`0.8
`0.7
`0.6
`
`0.5
`
`0.4
`
`0.3
`
`0.2
`
`24 Hi, pH 12.6
`
`/I
`
`/
`
`/
`
`/
`
`)<
`J
`
`'
`
`24+24 Hi, pH
`
`12.1
`
`24 Hi, pH 12.1
`
`24 Hi, Pre-labeted.
`pH 12.1
`
`pH 12.1
`24 HI pott-drug
`
`pH 12.6
`24 Ht pDrKkug
`
`0
`
`0.1
`
`1.0
`Aza-dC (fig/ml)
`
`10
`
`0.1
`
`5.0
`
`10
`
`0
`15
`5.0
`TIME
`OF ELUTION
`IHrl
`Fig. 2. Alkaline elution pronies of [UC]DNA from L1210 cells treated for 24
`h with aza-dCyd at the indicated concentrations
`(¿ig/ml).Elutions were carried
`out at pH 12.1 (I and C) and pH 12.6 (B and /') either immediately after drug
`washout
`(I and B) or following 24 h postincubation in drug-free medium (C and
`/»).Labeling with 0.02 ,,< i ml [>4C]thymidine and treatment with aza-dCyd were
`simultaneous. Points, mean of three values; bars, range;
`, elution of ['HJDNA
`from untreated LI210 after irradiation with 300 rads in the cold; 0.1 .if 'i |'l(|
`thymidine for 24 h was used to label these cells; points, mean of 25 values; bars,
`SE.
`
`10
`
`15
`
`Fig. 3. Effect of elution pH and treatment protocol on DNA elution kinetics
`from LI210 cells treated with Hj-aza-Cyd (//j-aza-C)
`(A) or aza-dCyd (Aza-dC)
`(B). —Log(retention)
`is proportional
`to DNA strand length, where (retention)
`represents
`the fraction of [MC]DNA retained on the filter at
`the time when
`internal
`standard (|'Il|l)NA)
`retention is equal
`to 0.2. The duration of drug
`treatment and simultaneous labeling with ["CJthymidine was 24 h and the elutions
`were carried out at pH 12.1 (O, •)or pH 12.6 (D. •)either
`immediately after
`drug washout (•,•)or after 24 h postincubation in drug-free medium (O, O). C,
`C,
`-log
`retention
`of [MC]DNA from cells that were prelabeled with [I4C]-
`thymidine,
`then exposed to drug for 24 h and eluted immediately. Points, mean
`of two to six values; bars, range, except in A (•,•)for which SE is shown.
`
`instability of the incorporated molecule
`quence of the chemical
`(aza-dCyd) or from an accumulation
`of strand scission inter
`mediates in an excision repair process (H2-aza-Cyd; see below).
`in Figs. 1 and 2 as well as data from additional
`depicted
`It might also be proposed that
`incorporation
`of anomalous
`experiments
`are summarized
`in Fig. 3, which shows the rela
`bases in the growing DNA strand produces an inhibition of
`tionship between drug concentration
`and the DNA elution rate
`further elongation (chain termination)
`and the resultant accu
`(plotted
`as the negative
`logarithm of the fraction of DNA
`mulation of small size DNA fragments. The latter mechanism
`retained
`on the filter at a fixed point of internal
`standard
`has been proposed to explain the DNA damage observed in
`elution). Both drugs caused a dose-dependent
`increase in DNA
`cells treated with ara-C (24).
`elution rate following a 24-h exposure with simultaneous
`label
`To investigate
`the effects of aza-dCyd and H2-aza-Cyd on
`ing. aza-dCyd-induced
`lesions demonstrated
`a large pH-de-
`DNA elongation, pulse-labeled cells were treated with drug for
`pendent
`increase (Fig. 3Ä),whereas there was much less pH
`1 h. After drug washout, DNA size was followed using alkaline
`dependence
`in the case of H2-aza-Cyd-treated
`cells (Fig. 3A).
`elution. The results of two separate representative
`experiments
`Repair of H2-aza-Cyd-induced
`lesions was evident, while 24 h
`are shown in Fig. 4. At the end of pulse labeling (r ,). control
`of postincubation
`following aza-dCyd treatment
`resulted in a
`DNA was small but increased towards normal
`length over 5-6
`clear increase in DNA damage for concentrations
`above 0.1 ¿¿g/
`h of incubation. Treatment
`of cells with aza-dCyd (1-25 fig/
`ml. We previously reported (21) that aza-dCyd lesions persist
`ml) resulted in only slight slowing of elongation during and
`for at least 48 h after drug removal with no evidence of repair.
`immediately following drug exposure; by 4 h, treated and con
`In contrast,
`the present data indicate that many of the H2-aza-
`trol DNA were nearly equivalent
`in size. In contrast, ara-C, a
`Cyd lesions are cleared from the DNA within 24 h, probably
`compound with known chain-terminating
`activity (25), pro
`by a repair process. When cells were labeled with [l4C]thymidine
`duced marked inhibition of elongation. Somewhat surprisingly,
`before aza-dCyd or H2-aza-Cyd exposure, only a small
`increase
`H2-aza-Cyd blocked further chain elongation completely and
`in DNA elution rate was produced by either compound (Fig.
`irreversibly for at
`least 6 h after drug removal. These data
`suggest
`that whereas
`aza-dCyd produces
`little or no chain
`3). This suggests
`that significant DNA damage is limited to
`termination, H2-aza-Cyd may induce failure of DNA elongation
`DNA strand segments synthesized in the presence of drug.
`The observed reductions
`in DNA strand length could result
`and/or
`ligation.
`from strand cleavage at sites of drug incorporation
`as a conse-
`The effects of aza-dCyd and H2-aza-Cyd on LI 210 cell cycle
`5513
`
`

`

`DNA DAMAGE PRODUCED BY Aza-dCyd AND H2-aza-Cyd
`
`late S phase and G2 + M was increased compared to control,
`whereas the percentage in G, was reduced. After an additional
`24 h incubation in drug-free medium,
`there appears
`to have
`been some renewed cell cycle progression at 0.1 jug/ml but not
`at
`the higher drug concentrations.
`In contrast, H2-aza-Cyd
`treatment produced no cell cycle effects during 24 h of exposure.
`After a 24-h drug-free period, control and H2-aza-Cyd treated
`cells showed an increase in (.;,. probably because a plateau
`density was achieved. Again, a tendency for 100 fig/ml and
`higher
`levels of H2-aza-Cyd to cause cell fragmentation was
`observed, yielding a nonevaluable DNA histogram.
`and repair
`In an attempt
`to further
`elucidate
`the nature
`mechanisms of H2-aza-Cyd- and aza-dCyd-induced DNA dam
`age, studies were done using two human lymphoblastoid
`cell
`lines. One (GM2345A) was derived from a patient with XP,
`while the other (GM3714) was from a clinically normal subject.
`It was anticipated
`that comparison
`of the DNA damage pro
`duced in these two cell
`lines would indicate
`the degree of
`dependency of H2-aza-Cyd- and aza-dCyd-associated DNA re
`pair on the excision repair mechanism absent
`in XP cells
`(complimentation
`group A). Fig. 5 presents elution profiles for
`DNA from the normal
`(GM3714)
`cell
`line. Fig. 5, A and C
`show the elution of DNA from H2-aza-Cyd-treated
`cells after
`24 h exposure
`(.-I) and after an additional
`24 h of drug-free
`incubation (("). Fig. 5, B and D present analogous data for aza-
`dCyd-treated
`cells. The characteristics
`of DNA damage pro-
`
`1.0
`0.9
`0.8
`0.7
`0.6
`
`0.5
`
`0.4
`
`0.3
`
`0.2
`
`0.1
`
`I.Or.
`0.9 P
`0.8
`0.7
`0.6
`
`0.5
`
`0.4
`
`0.3
`
`0.2
`
`AM OC
`24 Hr poet-dug
`
`f
`
`0
`0.4
`0.8
`1.2
`
`1.6
`
`2.0
`2.4
`
`0
`0.4
`0.8
`1.2
`1.6
`
`2.0
`2.4
`
`-101
`
`23456
`
`HOURS AFTER END OF DRUG TREATMENT
`
`Fig. 4. Effect of cytidine analogues on DNA elongation. LI210 cells were
`pulse labeled for 15 min, washed twice, and then incubated for l h with either no
`drug (•)or A: aza-dCyd, 1 Mg/ml (•);ara-C, 0.5 /ig/ml fT); H2-aza-Cyd, 50 »ig/
`ml (A); B: aza-dCyd, 10 »<g/ml(O); aza-dCyd, 25 i/g/ml (A); ara-C, 12 »ig/ml(V).
`Cells were then washed and incubated at 37'C. Aliquots were removed at
`the
`indicated times and analyzed by alkaline elution. —Log(retention)
`is proportional
`to DNA strand length, where (retention)
`is the fraction of ['I I|l >NA remaining
`on the fÃ-lterwhen internal
`standard retention ((UC]DNA)
`is equal
`to 0.2. The
`ordinale is such that average single-strand length increases with height. Points,
`mean of two determinations.
`
`Table 1 Cell cycle distribution of LI210 cells treated with aza-dCyd
`
`inConcentration
`
`% of cells
`
`S
`phase12
`
`S
`phase +
`G2-M30
`phase1316
`
`45
`44
`43
`
`18
`
`33
`43
`42
`
`29
`31
`24Early
`
`10
`6
`12Mid-S
`
`1922Late
`
`00
`
`.1
`1.0
`10.0G,45
`
`64
`
`38
`34
`32
`
`0.1
`1.0
`10.0
`
`16
`9
`10
`
`13
`14
`16
`
`(Mg/ml)After
`24 h treatment
`
`After an additional 24
`h postdrug inculi.!
`
`Table 2 Cell cycle distribution of LI 210 treated with H2-aza-Cyd
`
`inAfter
`
`24 h treatmentConcentrationGig/ml)0
`1050
`
`100G,45
`
`% of cells
`
`S
`phase1211
`
`S
`phase +
`G2-M30
`phase1315
`
`45
`51
`39Early
`
`64
`
`13
`25Mid-S
`
`1115Late
`
`29
`24
`22
`
`18
`
`After an additional
`24 h postdrug incu
`bation
`
`' NE, not évaluable.
`
`0.1
`
`10
`
`10
`
`15
`
`23
`12
`NE
`
`99
`
`NE
`
`89
`
`NE
`
`60
`70
`NE°
`
`10
`50
`100
`
`5.0
`0
`15
`TIME OF ELUTION (Hr)
`Fig. 5. Alkaline elution profiles of [UC]DNA from human lymphoblasts (GM
`3714) treated with H2-aza-Cyd (H2-aza-Q (A and Q or aza-dCyd (Aza-dQ (B
`and /') at the indicated concentrations Gig/ml). Elutions were carried out at pH
`12.6 either immediately after drug washout
`(I and B) or following 24 h postin
`progression were examined using flow cytometry. These results
`cubation in drug-free medium (C and D). Labeling and drug treatment were
`are tabulated in Tables 1 (aza-dCyd) and 2 (H2-aza-Cyd). Fol
`simultaneous. Points, mean of two values; bars, range;
`, elution of [WC]DNA
`lowing 24 h of aza-dCyd exposure,
`the percentage of cells in
`from untreated 11210 after exposure to 300 rads in the cold.
`5514
`
`

`

`DNA DAMAGE PRODUCED BY Aza-dCyd AND H2-aza-Cyd
`
`in each cell line. While these results do not exclude the possi
`bility that
`the H2-aza-Cyd-induced
`lesions were removed by
`excision repair,
`it appears
`that
`the specific mechanism that
`is
`deficient
`in XP cells plays no role in the observed repair of aza-
`dCyd- and H2-aza-Cyd-induced DNA damage.
`
`DISCUSSION
`
`duced in this cell line was similar to that seen with LI210 cells
`(Figs. 1-3). H2-aza-Cyd produced linear elution curves, whereas
`aza-dCyd curves were convex, again suggesting that
`the latter
`drug produces alkali-labile sites. It is noteworthy that H2-aza-
`Cyd treatment
`resulted in a reduced DNA retention in the first
`fraction (Fig. 5/4). This pattern is generally indicative of non
`specific cell death and lysis, suggesting that
`these human cell
`lines are more sensitive than L1210 cells to the postulated lytic
`effect of H2-aza-Cyd.
`aza-dCyd-induced DNA
`After 24 h of drug-free incubation,
`lesions showed evidence of repair only at the lowest dose tested
`(0.1 jig/ml).
`In contrast, H2-aza-Cyd-induced
`lesions were re
`paired even at drug doses up to 100 /¿g/ml.These results are
`summarized in Fig. 6A, which again relates drug concentration
`to a measure of DNA retention. The overall degree of DNA
`damage produced by H2-aza-Cyd and aza-dCyd in GM 3714
`was less than that produced in LI210 with identical concentra
`tions and exposure periods. The present experiments are unable
`to distinguish whether
`this difference results from more exten
`sive incorporation
`of the drugs
`into LI210 DNA (resulting
`from greater anabolic activity and/or cell cycle effects), differ
`ences in the kinetics of DNA repair
`in the two cell
`lines, or
`both.
`performed in the XP
`Experiments were also simultaneously
`cell line (GM2345A). Elution curves for both H2-aza-Cyd- and
`aza-dCyd-treated
`cells were essentially identical
`to those ob
`tained with the normal cell line. The results are summarized in
`Fig. 6/;. The quantitative
`similarity of DNA damage produced
`in the two cell lines is evident (Fig. 6, A versus B). DNA strand
`breaks produced by both drugs were repaired to a similar extent
`
`Human Lymphoblastoid
`Cell Line GM3714
`
`B
`
`Human Lymphoblastoid (XPI
`Celi Une GM2345A
`
`Aza-dC. 24 Hr
`Aza-dC. 24+24 HI
`H,-az»-C.
`
`24 Hf
`
`the DNA alterations
`In this report we have investigated
`produced by incorporation
`of the cytidine analogues aza-dCyd
`and H2-aza-Cyd into DNA of LI 210 murine leukemia and two
`human lymphoblastoid
`cell
`lines. When cells are exposed to
`[l4C]thymidine
`and H2-aza-Cyd simultaneously
`for 24 h, a
`significant frequency of DNA single-strand breaks was observed
`in the labeled DNA by means of the alkaline elution technique.
`The lack of dependence of the frequency of these lesions on the
`pH of the eluting solution (Fig. 3/1) suggests that
`they are true
`single-strand breaks and not a consequence of alkali-labile sites
`in the DNA.
`In contrast,
`the effect of aza-dCyd in simultane
`ously ['"CJthymidine-labeled
`cells was to produce an alkaline
`elution profile characteristic
`of alkali-labile
`lesions: a marked
`pH dependence of the elution rate and elution curves which
`bend downward (22). The markedly reduced amount of DNA
`damage seen in prelabeled cells suggests that
`lesions are pro
`duced only in DNA which was synthesized in the presence of
`aza-dCyd or H2-aza-Cyd and which has therefore incorporated
`the abnormal bases. Moreover,
`this observation eliminates
`the
`possibility that a significant
`fraction of the DNA damage seen
`in simultaneously
`labeled and treated cells resulted from non
`specific toxicity and cell lysis, which would be expected to also
`damage preexisting DNA.
`Glazer and Knode (9), using alkaline agarose gel electropho-
`resis, demonstrated
`that aza-dCyd exposure resulted in forma
`tion of low molecular weight
`fragments of newly synthesized
`DNA, whereas previously synthesized DNA remained intact.
`They postulated that aza-dCyd inhibited DNA elongation. Our
`studies suggest
`that aza-dCyd has little effect on DNA elonga
`tion (Fig. 4) and that
`the observed damage results
`from the
`presence of alkali-labile sites in the DNA which are converted
`to single-strand breaks under the conditions of elution or elec-
`•trophoresis.These effects of aza-dCyd are in contrast
`to those
`demonstrated
`for ara-C. ara-C has been shown to possess a
`partial, concentration-dependent
`DNA chain-terminating
`activ
`ity (25) and to produce alkaline elution profiles
`showing no
`evidence of alkali-labile sites (24). This is consistent with our
`demonstration
`that ara-C markedly slows chain elongation but
`that
`the block is incomplete
`and gradual growth to full size
`DNA can occur. When these observations
`are considered to
`gether with the finding that ara-C potently inhibits overall DNA
`synthesis
`(25), whereas aza-dCyd does not (2), it can be con
`cluded that
`these two compounds
`have contrasting
`cellular
`effects and produce DNA damage through different mecha
`nisms.
`Following a 1-h treatment of L1210 with H2-aza-Cyd (50 ng/
`ml), further DNA elongation was completely inhibited for at
`least 6 h (Fig. 4). This is somewhat
`surprising in light of the
`near normal
`elongation which occurred during the 1 h of
`exposure (f0in Fig. 4A). Moreover,
`it was demonstrated in Figs.
`1 and 3 that rapidly eluting DNA present after 24 h of simul
`taneous H2-aza-Cyd and label will increase in size if cells are
`incubated for an additional 24 h in the absence of drug. These
`observations are consistent with a model
`in which H2-aza-Cyd
`incorporation
`does not produce
`chain termination within a
`single replicón but prevents subsequent
`ligation of replicons to
`5515
`
`to -Log (retention)] of [MC]DNA from
`Fig. 6. Strand length [proportional
`normal human lymphoblasts
`(A) or xeroderma pigmentosum lymphoblasts (B) as
`a function of Hj-aza-Cyd (H2-aza-C) (•,O) or aza-dCyd (Aza-dC) (•.D) concen
`tration.
`(Retention),
`fraction of DNA retained on the filter after 15 h of elution
`(fifth fraction) considering the retention after collection of the first fraction to be
`1.0. Duration of drug treatment and simultaneous
`labeling with ["Cjthymidine
`was 24 h, with elutions carried out at pH 12.6 immediately after drug washout
`(•,•)or following 24 h postincubation
`in drug-free medium (O, D). Points,
`the
`mean of two values; bars, range.
`
`

`

`DNA DAMAGE PRODUCED BY Aza-dCyd AND H2-aza-Cyd
`
`analysis conditions
`
`in cell culture methods and/or
`differences
`were involved.
`of aza-dCyd and H2-aza-
`When the range of concentrations
`Cyd used for alkaline elution and flow cytometry studies are
`compared relative to effects on LI210 cell viability, a contrast
`between the two compounds
`is again observed. As measured by
`soft-agar colony forming ability of treated cells, 24 h exposure
`to aza-dCyd yields a 1-Iog cell kill at 0.1 tig/ml and a maximum
`of 3 logs at 1-100 ¿ig/ml(4). H2-aza-Cyd exposure at concen
`trations
`less than 5 /¿g/mlproduces
`little or no cytotoxicity,
`while 100 Mg/ml yields only a 1- to 1.5-log cell kill (27). Thus
`H2-aza-Cyd appears both less potent and less efficacious than
`aza-dCyd in vitro, a pattern which is repeated in the results of
`therapy studies with LI210 bearing mice in vivo (5, 27). While
`these findings are consistent with the lower potency of H2-aza-
`Cyd for producing DNA lesions and with the more extensive
`reversal of H2-aza-Cyd-induced
`damage,
`the exact relationship
`between the cytotoxicity
`of these compounds
`and the DNA
`damage produced remains uncertain.
`It has been reported that
`aza-dCyd cytotoxicity is also correlated with the inhibition of
`DNA methylation (20). Moreover, Jones and Taylor
`(28) have
`shown H2-aza-Cyd to be less active than aza-dCyd in inhibiting
`methylation. Since this process and the production
`of DNA
`lesions appear
`to require anabolism to the nucleotide triphos-
`phate and incorporation of the drugs into DNA (10), differences
`in cytotoxicity
`between H2-aza-Cyd and aza-dCyd may ulti
`mately be related to the extent of their metabolic activation.
`Nevertheless,
`the contrasting nature of DNA damage produced
`by these two compounds
`suggests that the biochemical basis for
`their cytotoxic activity may be quite different.
`
`ACKNOWLEDGMENTS
`The authors wish to thank Susan Hurst-Calderone for technical
`assistance, YvesPommier and Donna Kerrigan for valuablediscussions,
`and Madie Tyler for typing the manuscript.
`
`If the drug-free incubation time is
`form large DNA strands.
`sufficiently long,
`it appears
`that
`this block can be partially
`overcome.
`H2-aza-Cyd and aza-dCyd are related compounds differing in
`the sugar moiety, and in that
`the azapyrimidine
`ring of H2-aza-
`Cyd is 5,6-saturated. This latter
`feature confers stability to the
`H2-aza-Cyd molecule: aza-dCyd undergoes
`spontaneous
`ring
`opening in aqueous
`solution (fi/2 = 17 h at 37°C,pH 7.4)
`ultimately
`forming
`l-0-D-2'-deoxyribofuranosyl-3-guanylurea
`(4, 14), whereas H2-aza-Cyd demonstrates
`no decomposition
`under
`similar conditions
`(15). We have previously proposed
`(21) that
`the formation of DNA alkali-labile sites in aza-dCyd-
`treated cells is related to the incorporation
`of the chemically
`unstable azacytosine base into DNA. It is uncertain if aza-dCyd
`incorporated
`into DNA undergoes
`spontaneous
`ring

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