throbber
Review of the clinical experience with 5-azacytidine and 5-aza-2'-
`deoxycytidine in solid tumors
`Ana Aparicio* & Jeffrey S Weber
`Address
`Division of Medical Oncology
`University of Southern California
`Norris Comprehensive Cancer Center
`1441 Eastlake Avenue
`Los Angeles
`CA 90033
`USA
`Email: aaparici@usc.edu
`
`627
`
`mature oocytes and sperm are wiped out with a genome-
`wide wave of demethylation. Then, selective de novo
`methylation takes place, sparing the housekeeping genes
`and those that need to be active during embryogenesis.
`Finally,
`tissue-specific
`genes
`are
`demethylated in
`of
`association with the onset
`their
`activity. which
`presumably
`leads
`to
`tissue
`differentiation. DNA
`methylation is also involved in the inactivation of the X
`chromosome in females and in parental imprinting, and it
`probablycontributes to the aging process [5,6].
`
`Changes in methylation are among the most common
`genomic alterations found in neoplasia. On one hand, there
`is global hypomethylation
`of
`the DNA leading
`to
`chromosomal
`instability and an increased rate of genetic
`mutations [7e]. On the other, there is hypermethylation of
`CpGislands located in the promoters of tumor suppressor
`genes such as p16, p13. VHL and Rb. that renders them silent
`
`and provides a growth advantage for the cell
`[8]. In this
`regard,
`it
`is interesting to note that
`the establishment of
`immortal cell
`lines in vitro is also associated with de novo
`
`methylation of CpG islands [9¢,10]. Other genes knownto
`contribute to tumorigenesis such as the DNA-repair gene
`MLHI {1i], E-cadherin [12]. cyclooxygenase-2 [13.14] and
`estrogen
`receptor
`o
`[15.16,17]
`are
`also
`silenced
`by
`methylation of CpG islands in their upstream promoters.
`Additionally, it appears that resistance to chemotherapeutic
`agents may be mediated by methylation of genes in the
`apoptotic pathway. For example,
`the restoration of Apaf-1
`expression in highly chemoresistant melanoma cell
`lines
`after treatment with 5-aza-2'-deoxycytidine, led to a marked
`enhancement in their sensitivity to adriamycin and a rescue
`of the apoptotic defects associated with Apaf-I silencing
`[18ee].
`If these epigenetic changes could be reversecl, we
`would potentially be able to re-establish antiproliferation,
`differentiation and chemotherapy sensitivity pathways in
`malignant cells.
`
`*To whom correspondence should be addressed
`
`Current Opinion in Investigational Drugs 2002 3(4):627-633
`© PharmaPress Ltd ISSN 1472-4472
`
`in
`changes
`importance of epigenetic
`the
`In recent years
`carcinogenesis has been unfolding. It is now clear that the fifth
`base of the genome. methylcytosine. plays a critical role in the
`control of gene expression during normal development and
`carcinogenesis. Efforts to decrease methylation in neoplasias as a
`therapeutic
`strategy have
`been productive
`in
`hematologic
`malignancies but disappointing in solid tumors. The following is a
`review ofthe clinical experience with the agents 5-azacytidine and
`5-aza-2'-deoxycytidine in solid malignancies and a discussion of
`the difficulties encountered.
`
`Keywords 5-Azacytidine, 5-aza-2-deoxycytidine, clinical trials,
`DNAmethylation, solid tumor
`
`DNA methylation
`Methylation occurs after DNA replication by the addition
`of a methyl group from S-adenosylmethionine (SAM)
`to
`the 5'-position of cytosine residues. Approximately 3 to 4%
`of cytosines in mammalian DNA are methylated. Most of
`the 5-methylcytosine residues in eukaryotic DNA are
`found in the dinucleotide sequence 9'-CpG-3'
`[le]. CpG
`dinucleotides are scarce throughout mammalian DNA,
`except in the so-called CpG islands, where their frequency
`is
`normal
`or
`higher
`than
`expected.
`Spontaneous
`deamination of 5-methylcytosine leads to thymine and
`thus methylated CpG sites are highly mutagenic. In fact,
`although CpG dinucleotides are only found at one-fifth of
`the expected frequency in human DNA. more than 30%of
`all known disease-related point mutations are found at
`these sites [2°,3e].
`
`
`
`
`
`
`
` AOVCUUELUPHUY
`
`
`
`AEOSEADIDIED)|
`
` —
`
`(decitabine;
` 5-aza-2'-deoxycytidine
`and
`5-Azacylidine
`SuperGen Inc; Figure 1) are pyrimidine analogs that result
`from substituting nitrogen at the fifth carbon pesition ofthe
`nucleosides
`cytidine
`and 2'-deoxeytidine,
`respectively.
`Whenthey are incorporated into replicating DNA they form
`a covalent complex with DNA methyltransferase | (DNMTI,
`This raises the question of why the fifth base of the
`responsible for reproducing the methylation patterns in the
`genome
`is maintained. The
`essential
`function
`of
`daughter strands) and deplete the cell of this enzymatic
`methylated cytosine residues appears to be to modily
`activity leading to the synthesis of hypomethylated strands
`protein-DNA interactions and thereby suppress gene
`[19°]. At high concentrations these drugs are cytotoxic but at
`transcription. CpG islands are often located in the
`lowerconcentrations they induce differentiation of ccll lines
`promoter regions of genes and it has been shown that
`(20). It is believed that the changes observedincells treated
`methylation of their cytosine residues effectively switches
`with these drugs are a consequence of their induction of
`off the downstream gene [4ee]. Like genetic mutations, a
`DNA demethylation and ensuing activation of silent genes,
`methylation pattern is
`information that
`is stable and
`although some evidence suggests that covalent binding of
`reproduced with each round of cell division, but unlike
`the enzyme to the drug-substituted DNA is the primary
`genetic mutations,
`if
`can be readily reversed. This
`mechanism of
`drug-induced
`toxicity
`[Zle22e]. Both
`property makes DNA methylation an essential
`tool
`compounds have been tested in clinical trials and found to
`
`during Shortly—afterembryonic development.
`
`
`have significant antitumoractivity.
`fertilization,
`the methylation patterns observed in the
`CELGENE 2003
`CELGENE 2003
`APOTEX v. CELGENE
`APOTEX v. CELGENE
`IPR2023-00512
`IPR2023-00512
`
`jes1Gojo0uQ
`
`

`

`628 Current Opinion in Investigational Drugs 2002 Vol 3 No 4
`
`Figure 1. Structures of 5-azacytidine and 5-aza-2'-deoxycytidine.
`
`5-azacytidine
`
`5-aza-2'-deoxycytidine
`(SuperGen)
`
`
`
`Clinical experience in hematologic diseases
`Both 5-azacytidine and 5-aza-2'-deoxycytidine, at doses
`ranging from 500 to 1500 mg/m’, have proved effective in
`the treatment of relapsed or refractory acute leukemias and
`in the blast crisis of chronic mycloid leukemia. At low doses
`(50
`to
`150 mg/m’)
`they
`have
`shown
`activity
`in
`myelodysplastic syndrome (MDS),
`leading to trilineage
`responses in some patients [23]. A randomized phaseIII trial
`of
`low-dose S-azacytidine administered subcutaneously
`demonstrated a decreased probability of transformation to
`acute myeloid leukemia in high risk MDS patients, an
`improvement
`in quality of
`life and a
`trend towards
`improvedsurvival [24]. There have been several reports of
`increase in fetal hemoglobin in patients with severe [-
`thalassemia and sickle cell anemia treated with the cytidine
`analogs, although progress in non-malignant diseases has
`been hampered by the potential risk of carcinogenicity that
`these drugs carry [22¢,25¢|.
`
`Clinical experiencein solid tumors
`5-Azacytidine
`Clinical trials with 5-azacytidine are summarized in Table 1.
`An initial phase | trial was reported in 1972 by Weiss et al
`[26] using doses of 0.55 to 2.4 mg/kg/dayfor 10 to 15 days.
`Thirteen of thirty treated patients were reported to have had
`a partial response (two ofsix colon cancers. seven of eleven
`breast cancers and two offive melanomas amongst them).
`Remissions commonly occurred early in
`the treatment
`course and lasted an average of 6 weeks. Re-induction of
`remissions was possible in two patients with breast cancer
`who hadrelapses at 8 to 10 weeks. A maintenance regimen
`using 2.4 mg/kg twice a week was given to responding
`patients and two of these remained in remission for at least 6
`months.
`
`Following these encouraging results, several phase II trials
`were conducted. At
`the Mayo Clinic, 29 patients with
`advanced gastrointestinal cancer were treated with 500 to
`750 mg/m’ per course [27]. Nausea and vomiting were so
`severe,
`however,
`that
`three
`different
`administration
`schedules were tried in an attempt
`to decrease
`the
`symptoms. The drug was given once-daily for 5 days, once-
`daily for 10 days and twice-daily for 5 days. The latter was
`the
`better-tolerated
`regimen
`but was_
`still
`seriously
`emetogenic. Only one partial response lasting 5 weeks was
`observed. A cooperative phase II study in 31 patients with
`breast cancer using 60 mg/m‘/day of
`intravenous 5-
`azacytidine for 10 days (followed by a maintenance dose of
`100 mg/m’ twice weekly after bone marrow recovery) [28]
`yielded only four disease stabilizations and two partial
`
`responses lasting a mean of 5.5 months. Another small phase
`Ul trial was reported in 1982 [29], where eight patients with
`osteogenic sarcomas and seven with skeletal Ewing's
`sarcoma were treated with {50 to 200 mg/m’/day every8 h
`for 5 days: no objective responses were seen.
`
`large studies involving a spectrum of solid tumors
`Several
`have been published. The Southwest Oncology Grouptreated
`191
`patients with
`5-azacytidine
`intravenously
`(225
`mg/m’/day)
`for 5 days every 3 weeks [30]. Because of
`myelosuppression, however,
`this dose had to be reduced
`initially to 175 and then to 150 mg/m’. Two patients with
`adenocarcinoma of
`the lung, one with squamous
`cell
`carcinoma ofthe lung and two with embryonal carcinoma of
`the testicle had partial responses lasting from 28 to 77 days.
`Five
`renal, one breast,
`two colon and two_ pancreatic
`adenocarcinomas plus single malignancies arising from six
`other primarysites, were stabilized for 39 to 255 days. 6% Of
`patients died and 11% refused further therapy because ofthe
`drug's severe gastrointestinal toxicity. The Central Oncology
`Group administered 5-azacytidine (1.6 mg/kg/day) for 10
`days to 221 patients [31].
`In an attempt
`to decrease the
`uncontrollable nausea and vomiting induced on the day of
`treatment, 29 patients received the drug as an 18 to 24 h
`infusion. The degrees of leukopenia and thrombocytopenia
`were greater with the slow infusion. Stomatitis as well as an
`erythematous rash appeared, bul nausea and vomiting, were
`minimal. Although 19 partial responses were reported (one
`lung, six breast,
`three lymphoma and nine miscellaneous
`tumors), they were mainly of non-visceral disease and short-
`lived (mean of 5 weeks). The Southeastern Cancer Study
`Group[32] tried a biweekly regimenof 150 mg/m’ (50 mg/m’
`in lymphoma patients) of 5-azacytidine in 91 patients with
`disseminated malignancies, but only obtained two partial
`responses (one large cell carcinoma of the lung and one
`melanoma).
`In all of these trials, all of the patients that
`received the drug as an intravenous bolus suffered severe
`nausea and vomiting that was unresponsive to antiemetics,
`and frequently accompanied by diarrhea. Leukopenia and
`thrombocytopenia were dose-related and occurredlate in the
`course oftreatment.
`
`A small study comparing intravenous and subcutaneous
`administration of 5-azacytidine in humans had shownthat
`plasmalevels were similar after 1 h andthat the drug tended
`to concentrate in tumor tissue regardless of the route used
`{33]. Based on these results, another phase [
`trial was
`conducted
`using
`275
`to
`850 mg/m’
`administered
`subcutaneouslydaily for 10 days followed by 35 to 90 mg/m’
`weekly in those who responded [34].
`18 Patients were
`evaluable for toxicity. Nausea, vomiting and diarrhea were
`mild in this trial, however, severe hepatic toxicity occurred in
`five patients (all with significant hepatic metastatic disease), of
`whichthree died in hepatic coma. The platelet counts ofthree
`patients dropped to < 50,000/mm’, and two died as a direct
`consequence. Only two partial
`responses were observed
`whichlasted 2 and 3 months, respectively.
`
`One study investigated the benefits of administering 5-
`azacytidine with pyrazofurin (PF; an inhibitor of the enzyme
`orotidylate decarboxylase
`in
`the
`de nove pyrimidine
`biosynthesis pathway) after significant synergism ofthese
`drugs had been demonstrated in cell cultures
`[35].
`5-
`Azacytidine was given as a continuous intravenous infusion
`
`

`

`Clinical experience with 5-azacytidine and 5-aza-2'-deoxycytidine in solid tumors Aparicio & Weber 629
`
`Table 1. Clinical trials of 5-azacytidine in solid tumors.
`Reference |
`Objective
`Schedule
`‘Dose
`Study
`Numberof
`patients
`responses
`
`(Evaluable)
`:
`.
`Phase |: Breast, colon, melanoma,
`:
`30.
`0.55 to 2.4 mg/kg lv bolus:
`gd x 10 to 15 days
`43 PR
`[26]
`lung, soft tissue sarcomas, ovary,
`:
`pancreas, lymphoma
`
`Phase Il: Colon, one pancreas
`and_one gastric
`Phase |: Ovary, breast, tonsil,
`lung, hepatoma, renal, colon,
`melanoma, chordoma
`
`Phase |} Breast
`
`Phase |i: Solid tumors’
`
`:
`
`29
`
`18
`
`27
`
`167
`
`500 to 750 mg/miv bolus.
`(total dose per course)
`275 to 850 mg/m’ sc
`(M: 35 to 90 mg/m’)
`
`-qd’x 5 days; qd x 10
`days: bid x 5 days
`qd x 10 days
`(M: once a week)
`
`60 mg/m/day iv bolus
`(M: 400 mg/m’)
`
`gd x 10 days
`(M: biweekly)
`
`150 to 225mgimiv bolus.
`g
`
`qd x 5 days
`every 3 weeks
`
`1 PR
`
`:
`
`2PR
`
`2PR
`4SD
`
`5.PR
`- 16SD
`
`19PR
`
`2PR
`no responses
`See
`no responses
`
`[27]
`
`[34]
`
`[28]
`
`-[30]
`
`:
`
`[34]
`
`[32]
`-[35]
`:
`
`[29]
`
`qd x 10 days
`(M: biweekly)
`Biweekly x 6 days
`qd x 5 days”
`
`Every 8h x 5 days
`
`PhaseII: Solid tumors?
`
`1.6 mg/kg iv bolus/18 to...
`24-h infusion
`(M: 2.4 mg/kg)
`150 mg/miv bolus
`PhaseII: Solid tumors’
`30 to 60 mg/m’
`Phase |: In combination with
`continuous iv infusion
`pyrazofurin
`“Phase ll: Sarcomas of the bone a 150 mg/m’iv over 3 h
`
`177
`
`91
`6
`
`M maintenance, qd oncedaily, PR partial response, SD stable disease.
`lung (adenocarcinoma (12),
`‘Tumor and number of patients shown in parentheses: Pancreas (adenocarcinoma (6) andislet cell carcinoma (1));
`squamouscell carcinoma (14) and undifferentiated cell carcinoma (12)); colorectal (15); kidney (17); breast (14); testicle (embryonalcell carcinoma
`(2), teratocarcinoma (1), choriocarcinoma (1)); urinary bladder (2); primary liver carcinoma (4); palate adenocarcinoma(1); parotid adenocarcinoma
`(3); uterus adenocarcinoma (1); cervix squamouscell carcinoma (4); ovary (5); non-Hodgkin's lymphoma (4); sarcoma (7); melanoma (13); tongue
`squamous cell carcinoma (2); nasal antrum (1); esophagus squamous cell carcinoma (3); stomach adenocarcinoma (7); skin squamous cell
`carcinoma(1); larynx squamouscell carcinoma (1); gall bladder (1); unknown primary (adenocarcinoma (8) and undifferentiated cell carcinoma (4)).
`“Tumor and numberof patients shown in parentheses: Lung (24); breast (29); large intestine (26); melanoma (12); Hodgkin's disease (6);
`non-Hodgkin's lymphoma (8); miscellaneous (59).
`*Tumor and numberof patients shown in parentheses: Breast (6); renal (10); other urogenital (3); colon (7); pancreas (2); stomach (3);
`rectum (1); lung (unspecified (2), squamouscell (6), adenocarcinoma (11), undifferentiated (5), small cell (2), large cell (2)); melanoma (10);
`head and neck(14): soft tissue sarcomas (3); lymphomas(4).
`
`
`for 5 days immeciiately following the injection of PF. PF
`doses ranged from 50 to 100 mg/m’ and5-azacytidine doses
`ranged from 30 to 60 mg/m’. The most commonsideeffect
`was skin rash, which was dose-related, and when severe
`was accompanied by stomatitis, proctitis and cystitis. Six
`patients with solid tumors were enteredinto the trial but no
`objective responses were observed.
`
`5-Aza-2'-deoxycytidine
`Clinical trials with 5-aza-2'-deoxycytidine in solid tumors are
`summarized in Table 2. The first phase | trial conducted with
`3-aza-2'-deoxycytidine used a schedule consisting of three
`consecutive 1-h infusions separated by 7 h [36], a schedule
`dictated by
`the
`instability of 5-aza-2'-deoxycytidine
`in
`aqueous solution andits short half-life. The starting dose was
`25 mg/m’. The dose-limiting toxicity consisted of reversible
`myelosuppression, with the white blood cell count nadir
`delayed to days 22 to 33 of treatment. Platelet nadir was
`observed between days 14 and 22. The maximumtolerated
`dose was 100 mg/m’ x 3. One partial response was observed
`in a patient with a
`locally recurrent undifferentiated
`carcinoma of the ethmoid sinus who continued 5-aza-2'-
`
`deoxycytidine treatment every 5 to 6 weeks and, after surgery
`of a
`residual
`lymph node mcetastatasis, remained free of
`
`the EORTC
`disease at 14 months. Based on these results,
`conducted phase Il
`trials with 5-aza-2'-deoxycytidine in
`patients with melanoma and colorectal, renal and head and
`neck cancers, using the same schedule evaluated in the phase
`I study, at a dose of 75 mg/m’ [37]. Of 82 evaluablepatients,
`only one short-lived partial
`response was observed in a
`patient with malignant melanoma. Tumor stabilization was
`reported in 22%ofthe patients with melanoma, in 15%ofthe
`patients with head and neck cancer,
`in 14%of the patients
`with renal cell carcinoma and in 7% of the patients with
`colorectal carcinoma. Despite the lack ofsignificant activity in
`these studies, the EORTC used the same schedule and dose in
`14 patients with non-seminomatoustesticular cancer, because
`of the postulated activity of 5-aza-2'-deoxycytidine as a
`differentiating agent, but no objective responses were seen
`[38]. The same dose and schedule were also used in three
`other phaseII trials in patients with uterine cervical cancer (n
`= 14), ovarian cancer (n = 21) and prostate cancer (n = 12). No
`responses were seen in the uterine cervical cancer group but
`stabilization of disease was observed in two patients with
`ovarian cancer and twopatients with prostate cancer [39-41].
`The most common non-hematologic toxicity encounteredin
`all
`trials with this regimen was mild-to-moderate nausea
`and vomiting.
`
`uy
`
`fe)
`3ito
`QQ
`
`

`

`630 Current Opinion in investigational Drugs 2002 Vol 3 No 4
`
`Table 2. Clinicaltrials of 5-aza-2'-deoxycytidine in solid tumors.
`Objective
`Study
`Numberof
`Dose
`patients
`responses
`
`_(Evaluabie)
`i
`Phase|
`i
`20
`25 to 100 mg/m’
`:
`Every 3 to 6 weeks
`4.PR
`{36}
`:
`over Th every 8hx2te3
`
`Reference
`
`Schedule
`
`82
`s
`
`i. mg/m’ over 4h.
`every 8hAX3-
`
`Every 5 weeks
`
`1PRand13SD.
`:
`
`[87]
`
`Phase Il: Melanoma,colorectal,
`renal and squamouscell cancer
`of the head and neck
`Phase Il: Ovarian cancer
`e
`PhaseIl: Cancerofthe uterine
`cervix
`"PhaseIl:Non-seminomatous|
`testicular cancer
`Phase I/Il: Non-small cell fang :
`cancer
`Phase ll: Prostate cancer:
`¥:
`:
`;
`Phase
`
`Phase |I: Non-small cell jung
`cancer
`ins
`Phase
`
`
`
`2d
`
`14
`
`e
`
`:
`
`ce 14
`:
`9
`12
`
`21
`
`14
`19
`
`75 mg/m’ over1 A
`every 8h x3
`75 mgimover 4 he
`every 8h x3-
`75 mgimover 1h.
`every 8h x3
`:
`200 to 660 maim! x1
`over 8h
`75 inantover 1 h
`every 8hx3_
`45 to 120 mg/n*
`over 1h +33 mg/m’
`cisplatin every 24h x 3
`OL mit’ over th + 33
`“mg/mcisplatin every 24
`hx3
`20to40 atid
`: “over 72h
`
`
`Every 5 weeks
`:
`Every 6.weeks
`:
`
`Every 5'weeks
`j
`Every & to 7weeks
`oS
`‘Every 5 to8 weeks
`:
`‘
`Every 3 weeks
`
`:
`
`28D
`
`:
`
`no responses
`:
`no responses.
`:
`SA SD nt:
`28D
`
`2
`
`4PRand2
`minor responses
`
`Every 3 weeks
`_EverySweeks
`
`:
`
`3 minor
`responses
`shoresponses
`
`[40]
`
`[39]
`:
`[38]
`:
`{43}
`44]
`S
`
`47]
`
`:
`
`[47]
`polddle
`
`cs
`
`cell lung cancer used 5-aza-2'-deoxycytidine (67 mg/m’)
`the
`Both in vitro and in vive data [42] suggested that
`andcisplatin (33 mg/m’) on days 1 to 3. Only three short-
`cytotoxicity of 5-aza-2'-deoxycytidine was dose- and time-
`
`dependent. schedules—ofTherefore, alternative
`
`
`lasting minor regressions were observed[47].
`administration were explored.
`In 1997 Momparler et al
`conducted a phase I/II
`trial
`in previously untreated
`patients with stage IV non-small cell lung cancer, using an
`8-h intravenous infusion of 5-aza-2'-deoxycytidine at
`a
`dose of 200 to 660 mg/m’. They reported an increase in
`survival time with the number of cycles administered and
`one long-term survivor. They concluded that 5-aza-2’-
`deoxycytidine has a delayed action on tumor growth that
`may require several cycles of treatment before becoming
`evident |43]. Our group conducted a phase [ trial using a
`72-h continuous intravenous infusion of doses between 20
`and 40 mg/m’/day. All of
`the patients were heavily
`pretreated and only one ofthe 19 wasable to receive more
`than one cycle of treatment. No objective tumor responses
`were observed [44].
`
`Detection of DNA methylation
`in DNA
`To clinically assess
`the utility of alterations
`methylation,
`it
`is
`important
`to accurately measure the
`changes in methylation that occur after treatment with
`demethylating agents. Until
`recently,
`the
`techniques
`available for this have been difficult
`to apply to clinical
`samples.
`
`synergistic
`demonstrated
`have
`studies
`vitro
`In
`cytotoxicity using 5-aza-2'-deoxycytidine and cisplatin, 4-
`hydroperoxycyclophosphamide
`{a
`derivative
`of
`cyclophosphamide) and topotecan [45,46]. Based on these
`results, a phase | trial was conducted by Schwartsmannet
`al
`[47]
`using cisplatin
`(33 mg/m‘)
`plus
`5-aza-2’-
`deoxycytidine (45 to 120 mg/m’) as a 2-h infusion on
`days 1
`to 3. One partial response was observed in a
`patient with advanced cervical cancer. A follow-up early
`phase II evaluation in 14 patients with inoperable non-small
`
`Initial protocols to detect changes in DNA methylation
`employed digestion of genomic DNA with methylation-
`sensitive restriction enzymes (characterized bytheir inability
`to
`cleave
`sequences
`that
`contain methylated CpG
`dinucleotides) followed by Southern blot analysis [le], The
`main drawbacks to this method included a requirement for
`large amounts of DNA (> 5 ug) and that
`the extent of
`analysis was limited to the CpG sites present
`in the
`recognition sites of the available enzymes. Later on, PCR
`amplification was applied using primers that flanked the
`restriction sites that were methylation sensitive. Although
`the sensitivity of the assay increased, this method could still
`only be used to assess CpG methylation at methylation-
`sensitive restricion sites, and it had the potential of
`generating false positive results if
`the cleavage of
`the
`unmethylated DNA was incomplete [48].
`
`

`

`asosyuy|
`
`
`
`
`
`
`
` _Aomuueyurnuy|
`
`Clinical experience with 5-azacytidine and 5-aza-2'-deoxycytidine in solid tumors Aparicio & Weber 631
`
`then
`bisulfite conversion of the sample DNA, which is
`amplified
`by
`PCR using
`three methylation-specific
`oligonucleotides: a probe with a 5'-fluorescent reporter dye
`and a 3'-quencher dye, and two locus specific PCR primers
`that flank the probe. During the PCR reaction, the nuclease
`activity of the Tag DNA polymerase cleaves the probe and
`the reporter is released resulting in a fluorescent signal that
`is proportional to the amount of PCR product generated and
`immeasurable with a real-time fluorescence instrumentation.
`
`This technology has mace possible the rapid screening of
`large numbers of human tumors for the methylation state of
`a parlicular locus [53] but cannot offer information on longer
`regions of DNA.
`
`technique to analyze the biological activity of
`The ideal
`demethylating agents in clinical
`trials should be able to
`combine the high-resolution information of the methylation
`state of a
`large area of DNA sequence,
`like bisulfite
`sequencing. with the sensitivity, accuracy and efficiency of
`high-throughput technology.
`
`5-
`detect
`to
`used
`also
`sequencing was
`Genomic
`methylcytosine residues, identified as a lack of bands in all
`tracks of a sequencing gel. However,
`interpretation was
`frequently complicated by close spacing of the bands or by
`backgroundcleavage lacders [te]. In 1992, Frommer ef al
`[49] described the bisulfite genomic sequencing technique
`based on treatment of single stranded DNA with sodium
`bisulfite to deaminate cytosine to uracil muchfaster than 5-
`methylcytosine to thymine,
`so that methylated cytosine
`residues are left
`intact. The bisulfite-treated DNA was
`amplified by PCR and the products sequenced. The 5-
`methylcytosine residues on the original sample appeared as
`the only remaining cytosines on the sequencing gel, since
`unmethylated cytosines were transformed to uracil by the
`bisulfite. This approach could be applied to small amounts
`of DNA; even DNA obtained from paraffin-embedded
`tissue samples. However, without cloning the amplified
`roducts, a labor-intensive and time-consuming process,
`his method was less sensitive than Southern analysis.
`evertheless, bisulfite treatment of genomic DNA provided
`he basis for multiple new strategies for the assessment of
`DNA methylation.
`
`
`
`
`
`References
`
`th
`
`Razin A, Riggs AD: DNA methylation and gene function. Science
`(1980) 210:604-610.
`* This is an early paper that describes how methylated DNA was assayed
`and what was knownofits biological function.
`
`2.
`
`Rideout WM, Coetzee GA, Olumi AF, Jones PA: 5-Methylcytosine as
`an endogenous mutagen in the human LDL receptor and p53
`genes. Science (1990) 249:1288-1290.
`» This paper presents evidence suggesting that 5-methylcytosine functions as
`an endogenous mutagen because the presence of ihe methy! group increases
`the potential for mutation at cytosine resicues.
`
`3.
`
`Pfefer GP, Tang M, Denissenko MF: Mutation hotspets and DNA
`methylation. Curr Top Microbiol Immunol (2000) 249:1-19.
`e This review focuses on the regulation of matrix activities and their biological
`and pathological implications.
`
`[52] developed a high-throughput
`et al
`Finally, Eads
`quantitative assay for the analysis of DNA methylation,
`called MethyLight. This technique begins with sodium
`
`5.
`
`Razn A, Shemer R: DNA methylation in early development. Hum Mol
`Genet (1995) 4:1751-1755.
`* This paper summarizes the changes in DNA methylation that occur during
`embryonic development.
`
`Future directions
`role in the
`Since DNA methylation plays an important
`[50] described the methylation-specific PCR
`Herman et al
`expression,
`
`
`regulation of—gene modulating it
`
`protocol in which they used three sets of primers designed
`pharmacologically is a very attractive therapeutic target. 5-
`specifically to amplify three types of bisulfite-treated DNA:
`Azacytidine
`and
`5-aza-2'-deoxycytidine have not
`been
`methylated, unmethylated and DNA that had not been
`dramatically successful, and pose significant restriclions on
`modified,
`The
`PCR
`products were
`compared
`on
`dosing schedule adjustments because of their instability in
`polyacrylamide
`gels
`stained with
`ethidium bromice,
`aqueous solution andtheir side effects. Development of new
`providing semiquantitative results. This method was highly
`drugs without Uvese limitations is a subject of active research.
`sensitive, capable of detecting even 0.1%of methylated DNA
`Other approaches to inhibit DNMTI. the enzymeresponsible
`in a sample, and significantly less time consuming than
`for the replication of the DNA methylation pattern, are being
`genomic sequencing. Gonzalgo and Jones [48] developedthe
`explored, incluciing the use of modified oligonucleotides that
`methylation-sensitive single nucleotide primer extension
`directly
`anlagonize
`the
`enzyme
`[54].
`Antisense
`assay in which the PCR product from the bisulfite-convertec
`oligorucleotides have been tested in the laboratory [55} and
`DNA amplification was isolated from an agarose gel andused
`clinfeal
`trials have moved ahead [56e]. Combinalions of
`as a template for a second PCR reaction. The second PCR
`demethylating agents with synergistic drugs, such as the
`reaction utilized “P-labeled dNTPs and internal primers tha
`histone deacetylase inhibitors (trichostatin A [57] and 4-
`terminated immediately 5' of the single nucleotide ofinterest.
`phenylbutyrate [58]), or targeting specific signal transduction
`The radiolabeled products were electrophoresed resulting in
`pathwaysthat rendercells sensitive to chemotherapy agents,
`two bands: the C band represented the methylated cytosine
`are other strategies that shouldbe further investigated.
`residues and the T band represented the unmethylatec
`cytosine
`residues. Phosphorimage
`analysis
`allowed for
`quantitation of each band. At the same time, Xiong and Laird
`[51] reported a combinedbisulfite restriction analysis,
`that
`involved a standard sodiumbisulfite PCR treatment, followec
`by the digestion of
`the purified PCR products using a
`restriction enzyme with CpG in its recognition sequence. In
`this way, cleavage only occurred if the CpG sequence had
`been retained during the bisulfite conversion, ie, if the origina
`C residue was methylated. Different
`restriction enzymes
`detected different levels of DNA methylation, depending on
`the number of CpG dinucleotides containedin its recognitior
`sequence. Gel
`electrophoresis,
`oligo hybridization anc
`phosphorimage analysis allowed quantitation of the level of
`methylation present. Although these methods were rapid,
`offered quantitative results and were compatible with paraffin
`4.
`Jones PA, Laird PW: Cancer epigenetics comes of age. Nat Genet
`
`
`
`embedded they—stilltissues, required cumbersome
`
`(1999) 24:163-167.
`manipulations and were not easily applicable to large
`ee This paper reviews the mechanisms of DNA methylation in malignant
`transformation.
`numbers of specimens.
`
`
`
`
`
`leveesia)
`
`flex)
`
`

`

`632 Current Opinion in Investigational Drugs 2002 Vol 3 No 4
`
`6.
`
`issa JP, Ottaviano YL, Celano P, Hamilton SR, Davidson NE, Baylin SB:
`Methylation of the oestrogen receptor CpG island links ageing and
`neoplasia in human colon. Nat Genet (1994) 7:536-540
`
`7.
`
`Chen RZ, Pettersson U. Beard C, Jackson-Grusby L, Jaenisch R: DNA
`hypomethylation leads to elevated mutation rates. Nature (1993)
`395:89-93.
`« This paper presents evidence that supports a role for DNA methylation in
`maintaining genome stability.
`
`8.
`
`Baylin SB, Herman JG, Graff JR, Vertino PM, Issa JP: Alterations in
`DNA methylation: a fundamental aspect of neoplasia. Adv Cancer
`Res (1998) 72:141-196.
`
`Gs
`
`Jones PA, Wolkowicz MJ, Rideout WM, Gonzales FA, Marziasz CM,
`Coetzee GA, Tapscott SJ: De nove methylation of the MyoD1 CpG
`island during the establishment of immortal cell
`lines. Proc Nat!
`Acad Sci USA (1990) 87:6117-6121.
`¢ This article describes the changes in methylation patterns that take place
`whencells are immortalizedin vitro.
`
`10. Antequera F, Boyes J, Bird A: High levels of de novo methylation and
`altered chromatin structure at CpG islands in cell lines. Ce/f (1990)
`62:503-514.
`
`11. Herman JG, Umar A, Polyak K, Graff JR, Ahuja N, Issa JP, Markowitz
`S, Willson JK, Hamilton SR, Kinzlier KW. Kane MF, Kolodner RD,
`Vogelstein B, Kunkel TA, Baylin SB:
`Incidence and functional
`consequences of hMLH1 promoter hypermethylation in colorectal
`carcinoma. Proc Nat! Acad Sci USA (1998) 95:6870-6875.
`
`12, Yoshiura K, Kanai Y, Ochiai A, Shimoyama Y, Sugimura T, Hirchashi S:
`Silencing of the E-cadherin invasion-suppressor gene by CpG
`methylation in human carcinomas. Proc Nati Acad Sci USA (1995)
`92:7416-7419.
`
`13.
`
`14.
`
`15.
`
`16.
`
`Toyota M, Shen L, Ohe-Toyota M, Hamilton SR, Sinicrope FA, Issa JP:
`Aberrant methylation of the cyclooxygenase 2 CpG island in
`colorectal tumors. Cancer Res (2000) 60:4944-4048
`
`Song SH, Jong HS, Choi HH, Inoue H, Tanabe T, Kim NK, Bang YJ:
`Transcriptional
`silencing
`of
`cyclooxygenase-2
`by
` hyper-
`methylation of the 5' CpG island in human gastric carcinomacells.
`Cancer Res (2001) 61:4628-4635
`
`Ferguson AT, Lapidus RG, Baylin SB, Davidson NE: Demethylation of
`the estrogen receptor gene in estrogen receptor-negative breast
`cancer celis can reactivate estrogen receptor gene expression.
`Cancer Res (1998) 55:2279-2283
`
`Yoshida T, Eguchi H, Nakachi K, Tanimoto K, Higashi Y, Suemasu K,
`lino Y, Morishita Y, Hayashi S: Distinct mechanisms of loss of
`estrogen receptor a@ gene expression in human breast cancer:
`methylation of the gene and alteration of trans-acting factors.
`Carcinogenesis (2000) 21:2193-2201
`
`17. Nass SJ, Herman JG, Gabrielson E, Iversen PW, Parl FF, Davidsan NE,
`Graff JR: Aberrant methylation of the estrogen receptor and E-
`cadherin 5° CpG islands increases with malignant progression in
`human breast cancer. Cancer Res (2000) 60:4346-4348.
`
`18.
`
`Soengas MS, Capodieci P, Polsky D, Mora J, Esteller M, Opitz-Araya X,
`McCombie R, Herman JG. Gerald WL, Lazebnik YA, Cordon-Cardo C,
`Lowe SW: Inactivation of the apoptosis effector Apaf-1 in malignant
`melanoma. Nature (2001) 409:207-211
`ee This article shows that Apaf-1 (a cofactor of caspase-9 and downstream effector
`of p53) expression can be recovered in melanoma cell lines by treatment with 5-
`aza-2"-deoxycytidine and leads to an enhancementoftheir chemosensitivity.
`
`49.
`
`Jones PA, Taylor SM: Hemimethylated duplex DNAs prepared from
`5-azacytidine-treated cells. Nucleic Acids Res (1981) 9:2933-2947.
`e This paper explores the mechanism by which 5-azacytidine results in
`demethylation of DNA.
`
`20.
`
`Jones PA, Taylor SM: Cellular differentiation, cytidine analogs and
`DNA methylation. Cel/ (1980) 20:85-93.
`
`21.
`
`Juttermann R, Li E, Jaenisch R: Toxicity of 5-aza-2'-deoxycytidine to
`mammalian cells is mediated primarily by covalent trapping of DNA
`methyltransferase rather than DNA demethylation. Proc Nat! Acad
`Sci USA (1994) 94:11797-11801.
`* This article presents evidence supporting the idea that toxicity of 5

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