throbber
(cid:5)(cid:6)(cid:7)(cid:8)(cid:9)(cid:10)(cid:9)(cid:11)(cid:7)(cid:12)(cid:13)
`
`Q
`Taylor & Francis
`~ Taylor&.hand$Croup
`
`(cid:14)(cid:15)(cid:15)(cid:16)(cid:17)(cid:3)(cid:18)(cid:19)(cid:19)(cid:20)(cid:4)(cid:21)(cid:21)(cid:20)(cid:22)(cid:3)(cid:23)(cid:24)(cid:25)(cid:7)(cid:10)(cid:11)(cid:26)(cid:3)(cid:18)(cid:19)(cid:19)(cid:20)(cid:4)(cid:21)(cid:27)(cid:28)(cid:29)(cid:3)(cid:23)(cid:30)(cid:10)(cid:31)(cid:7)(cid:10)(cid:9)(cid:26)(cid:3)(cid:32)(cid:33)(cid:34)(cid:25)(cid:10)(cid:35)(cid:31)(cid:3)(cid:36)(cid:33)(cid:37)(cid:9)(cid:6)(cid:35)(cid:8)(cid:9)(cid:17)(cid:3)(cid:36)(cid:11)(cid:11)(cid:6)(cid:13)(cid:17)(cid:38)(cid:38)(cid:39)(cid:39)(cid:39)(cid:40)(cid:11)(cid:35)(cid:10)(cid:41)(cid:42)(cid:33)(cid:10)(cid:31)(cid:7)(cid:10)(cid:9)(cid:40)(cid:12)(cid:33)(cid:37)(cid:38)(cid:31)(cid:33)(cid:7)(cid:38)(cid:43)(cid:9)(cid:6)(cid:7)(cid:21)(cid:28)
`
`(cid:44)(cid:9)(cid:35)(cid:12)(cid:11)(cid:7)(cid:45)(cid:35)(cid:11)(cid:7)(cid:33)(cid:10)(cid:3)(cid:33)(cid:42)(cid:3)(cid:5)(cid:6)(cid:7)(cid:8)(cid:9)(cid:10)(cid:9)(cid:11)(cid:7)(cid:12)(cid:35)(cid:31)(cid:31)(cid:46)(cid:3)(cid:15)(cid:7)(cid:31)(cid:9)(cid:10)(cid:12)(cid:9)(cid:41)(cid:3)(cid:47)(cid:9)(cid:10)(cid:9)(cid:13)(cid:3)(cid:48)(cid:46)
`(cid:49)(cid:16)(cid:50)(cid:3)(cid:51)(cid:9)(cid:11)(cid:36)(cid:46)(cid:31)(cid:11)(cid:25)(cid:35)(cid:10)(cid:13)(cid:42)(cid:9)(cid:25)(cid:35)(cid:13)(cid:9)(cid:3)(cid:14)(cid:10)(cid:36)(cid:7)(cid:48)(cid:7)(cid:11)(cid:33)(cid:25)(cid:13)(cid:17)(cid:3)(cid:52)(cid:35)(cid:13)(cid:7)(cid:12)(cid:3)(cid:53)(cid:33)(cid:10)(cid:12)(cid:9)(cid:6)(cid:11)(cid:13)
`(cid:35)(cid:10)(cid:41)(cid:3)(cid:53)(cid:31)(cid:7)(cid:10)(cid:7)(cid:12)(cid:35)(cid:31)(cid:3)(cid:50)(cid:6)(cid:6)(cid:31)(cid:7)(cid:12)(cid:35)(cid:11)(cid:7)(cid:33)(cid:10)(cid:13)
`
`(cid:53)(cid:33)(cid:25)(cid:35)(cid:3)(cid:51)(cid:34)(cid:10)(cid:41)(cid:54)(cid:3)(cid:52)(cid:33)(cid:41)(cid:33)(cid:3)(cid:52)(cid:25)(cid:34)(cid:9)(cid:12)(cid:43)(cid:10)(cid:9)(cid:25)(cid:3)(cid:55)(cid:3)(cid:56)(cid:25)(cid:35)(cid:10)(cid:43)(cid:3)(cid:57)(cid:46)(cid:43)(cid:33)
`
`(cid:58)(cid:33)(cid:3)(cid:12)(cid:7)(cid:11)(cid:9)(cid:3)(cid:11)(cid:36)(cid:7)(cid:13)(cid:3)(cid:35)(cid:25)(cid:11)(cid:7)(cid:12)(cid:31)(cid:9)(cid:17)(cid:3)(cid:5)(cid:6)(cid:7)(cid:8)(cid:3)(cid:9)(cid:10)(cid:11)(cid:12)(cid:13)(cid:3)(cid:14)(cid:6)(cid:12)(cid:6)(cid:3)(cid:14)(cid:7)(cid:10)(cid:15)(cid:16)(cid:17)(cid:11)(cid:15)(cid:7)(cid:3)(cid:18)(cid:3)(cid:19)(cid:7)(cid:8)(cid:11)(cid:17)(cid:3)(cid:20)(cid:21)(cid:17)(cid:6)(cid:3)(cid:22)(cid:23)(cid:24)(cid:24)(cid:25)(cid:26)(cid:3)(cid:27)(cid:15)(cid:8)(cid:16)(cid:28)(cid:29)(cid:30)(cid:8)(cid:28)(cid:29)(cid:6)(cid:11)(cid:3)(cid:6)(cid:31)
`(cid:32)(cid:33)(cid:29)(cid:34)(cid:15)(cid:11)(cid:15)(cid:28)(cid:29)(cid:16)(cid:8)(cid:35)(cid:35)(cid:21)(cid:3)(cid:36)(cid:29)(cid:35)(cid:15)(cid:11)(cid:16)(cid:15)(cid:12)(cid:3)(cid:37)(cid:15)(cid:11)(cid:15)(cid:38)(cid:3)(cid:39)(cid:21)(cid:3)(cid:40)(cid:41)(cid:42)(cid:3)(cid:9)(cid:15)(cid:28)(cid:43)(cid:21)(cid:35)(cid:28)(cid:7)(cid:8)(cid:11)(cid:38)(cid:31)(cid:15)(cid:7)(cid:8)(cid:38)(cid:15)(cid:3)(cid:44)(cid:11)(cid:43)(cid:29)(cid:39)(cid:29)(cid:28)(cid:6)(cid:7)(cid:38)(cid:45)(cid:3)(cid:14)(cid:8)(cid:38)(cid:29)(cid:16)(cid:3)(cid:5)(cid:6)(cid:11)(cid:16)(cid:15)(cid:33)(cid:28)(cid:38)(cid:3)(cid:8)(cid:11)(cid:12)(cid:3)(cid:5)(cid:35)(cid:29)(cid:11)(cid:29)(cid:16)(cid:8)(cid:35)
`(cid:42)(cid:33)(cid:33)(cid:35)(cid:29)(cid:16)(cid:8)(cid:28)(cid:29)(cid:6)(cid:11)(cid:38)(cid:13)(cid:3)(cid:32)(cid:33)(cid:29)(cid:34)(cid:15)(cid:11)(cid:15)(cid:28)(cid:29)(cid:16)(cid:38)(cid:13)(cid:3)(cid:46)(cid:45)(cid:46)(cid:13)(cid:3)(cid:47)(cid:4)(cid:46)(cid:48)(cid:13)(cid:3)(cid:40)(cid:49)(cid:44)(cid:45)(cid:3)(cid:46)(cid:24)(cid:50)(cid:48)(cid:46)(cid:25)(cid:46)(cid:51)(cid:15)(cid:33)(cid:29)(cid:50)(cid:46)(cid:50)(cid:46)(cid:50)(cid:23)(cid:52)(cid:53)(cid:54)
`
`(cid:58)(cid:33)(cid:3)(cid:31)(cid:7)(cid:10)(cid:43)(cid:3)(cid:11)(cid:33)(cid:3)(cid:11)(cid:36)(cid:7)(cid:13)(cid:3)(cid:35)(cid:25)(cid:11)(cid:7)(cid:12)(cid:31)(cid:9)(cid:17)(cid:3) (cid:5)(cid:6)(cid:6)(cid:7)(cid:8)(cid:9)(cid:10)(cid:10)(cid:11)(cid:12)(cid:13)(cid:14)(cid:12)(cid:15)(cid:16)(cid:10)(cid:17)(cid:18)(cid:14)(cid:19)(cid:17)(cid:20)(cid:17)(cid:10)(cid:21)(cid:7)(cid:13)(cid:14)(cid:17)(cid:14)(cid:17)(cid:14)(cid:22)(cid:23)(cid:24)(cid:25)
`
`(cid:26)(cid:27)(cid:28)(cid:29)(cid:13)(cid:8)(cid:5)(cid:21)(cid:11)(cid:3)(cid:12)(cid:30)(cid:29)(cid:13)(cid:30)(cid:21)(cid:9)(cid:3)(cid:17)(cid:18)(cid:3)(cid:31)(cid:32)(cid:15)(cid:3)(cid:22)(cid:18)(cid:18)(cid:20)(cid:14)
`
`(cid:33)(cid:27)(cid:28)(cid:34)(cid:13)(cid:6)(cid:3)(cid:35)(cid:12)(cid:27)(cid:15)(cid:3)(cid:32)(cid:15)(cid:6)(cid:13)(cid:36)(cid:29)(cid:21)(cid:3)(cid:6)(cid:12)(cid:3)(cid:6)(cid:5)(cid:13)(cid:8)(cid:3)(cid:37)(cid:12)(cid:27)(cid:15)(cid:30)(cid:32)(cid:29)(cid:3)
`
`(3'
`
`W!l
`
`(cid:38)(cid:15)(cid:6)(cid:13)(cid:36)(cid:29)(cid:21)(cid:3)(cid:39)(cid:13)(cid:21)(cid:40)(cid:8)(cid:9)(cid:3)(cid:20)(cid:18)(cid:18)
`
`(cid:41)(cid:13)(cid:21)(cid:40)(cid:3)(cid:15)(cid:21)(cid:29)(cid:32)(cid:6)(cid:21)(cid:11)(cid:3)(cid:32)(cid:15)(cid:6)(cid:13)(cid:36)(cid:29)(cid:21)(cid:8)(cid:3)
`
`(3'
`
`(cid:42)(cid:13)(cid:6)(cid:13)(cid:30)(cid:16)(cid:3)(cid:32)(cid:15)(cid:6)(cid:13)(cid:36)(cid:29)(cid:21)(cid:8)(cid:9)(cid:3)(cid:25)(cid:25)(cid:3)(cid:41)(cid:13)(cid:21)(cid:40)(cid:3)(cid:36)(cid:13)(cid:6)(cid:13)(cid:30)(cid:16)(cid:3)(cid:32)(cid:15)(cid:6)(cid:13)(cid:36)(cid:29)(cid:21)(cid:8)(cid:3)
`
`(3'
`
`CELGENE 2022
`APOTEX v. CELGENE
`IPR2023-00512
`
`(cid:43)(cid:27)(cid:29)(cid:29)(cid:3)(cid:44)(cid:21)(cid:15)(cid:34)(cid:8)(cid:3)(cid:45)(cid:3)(cid:42)(cid:12)(cid:30)(cid:11)(cid:13)(cid:6)(cid:13)(cid:12)(cid:30)(cid:8)(cid:3)(cid:12)(cid:46)(cid:3)(cid:32)(cid:36)(cid:36)(cid:21)(cid:8)(cid:8)(cid:3)(cid:32)(cid:30)(cid:11)(cid:3)(cid:27)(cid:8)(cid:21)(cid:3)(cid:36)(cid:32)(cid:30)(cid:3)(cid:28)(cid:21)(cid:3)(cid:46)(cid:12)(cid:27)(cid:30)(cid:11)(cid:3)(cid:32)(cid:6)
`(cid:5)(cid:6)(cid:6)(cid:7)(cid:8)(cid:9)(cid:10)(cid:10)(cid:40)(cid:40)(cid:40)(cid:14)(cid:6)(cid:32)(cid:30)(cid:11)(cid:46)(cid:12)(cid:30)(cid:29)(cid:13)(cid:30)(cid:21)(cid:14)(cid:36)(cid:12)(cid:34)(cid:10)(cid:32)(cid:36)(cid:6)(cid:13)(cid:12)(cid:30)(cid:10)(cid:37)(cid:12)(cid:27)(cid:15)(cid:30)(cid:32)(cid:29)(cid:47)(cid:30)(cid:46)(cid:12)(cid:15)(cid:34)(cid:32)(cid:6)(cid:13)(cid:12)(cid:30)(cid:48)(cid:37)(cid:12)(cid:27)(cid:15)(cid:30)(cid:32)(cid:29)(cid:42)(cid:12)(cid:11)(cid:21)(cid:49)(cid:50)(cid:21)(cid:7)(cid:13)(cid:22)(cid:18)
`
`

`

`[Epigenetics 1:1, 7-13, January/February/March 2006]; ©2006 Landes Bioscience
`
`Review
`Reactivation of Epigenetically Silenced Genes by DNA Methyltransferase
`Inhibitors
`Basic Concepts and Clinical Applications
`
`Cora Mund
`Bodo Brueckner
`Frank Lyko*
`
`ABSTRACT
`Hypermethylation of tumor suppressor genes is one of the most consistent hallmarks of
`human cancers. This epigenetic alteration has been associated with gene silencing and
`thus represents an important pathway for generating loss-of-function mutations. In this
`review, we survey the available literature on systematic, genome-wide approaches aimed
`at the identification of epigenetically silenced loci. These studies uncovered a variety of
`diverse genes, but a common signature for epigenetic reactivation has not been identified.
`Nevertheless, DNA methyltransferase inhibitors have shown significant clinical benefits,
`mostly in the therapy of leukemias. Recent analyses revealed substantial drug-induced
`methylation changes that can now be used as endpoints for the further refinement of clinical
`treatment schedules. Further optimization of epigenetic cancer therapies should be feasible
`through the use of novel DNA methyltransferase inhibitors with improved specificity.
`Rational design of epigenetic inhibitors might provide the foundation for a broader use
`of these drugs in the treatment of cancer.
`
`Epigenetic mechanisms play a fundamental role in the interpretation of genetic infor-
`mation.1 Depending on its particular epigenetic modification pattern, a gene can be
`expressed or silenced. Epigenetic modifications thus represent an integral mechanism for
`the control of complex gene expression patterns. One prominent example for an epigenetic
`control mechanism is the covalent modification of histones.2 Histones can be modified at
`various amino acid residues by acetylation, phosphorylation, methylation and ubiquitina-
`tion3 and it has been suggested that particular combinations of these modifications
`constitute an “epigenetic code“ for the regulation of gene expression.2 Another prominent
`epigenetic modification is the methylation of cytosine residues in genomic DNA.4 About
`4% of the cytosines are usually methylated in mammalian genomic DNA5 and it has been
`shown that this methylation is essential for mouse development.6 In addition, it has also
`been shown that DNA methylation plays an essential role in several epigenetic phenomena,
`including genomic imprinting,7 X-chromosome inactivation,8 and retroelement silencing.9
`CpG dinucleotides represent the consensus target sequences of DNA methylation in
`differentiated mammalian cells, but only a relatively small fraction of these sequences
`becomes methylated.10 CpG dinucleotides can be clustered in CpG islands11,12 which are
`often associated with promoter regions and remain unmethylated for most genes. The
`signals that determine whether a particular CpG-containing sequence becomes methylated
`have not been determined yet, but it has been suggested that protein-protein interactions
`between DNA methyltransferases and chromatin-associated proteins might play an important
`role.13 As a consequence, DNA methylation is not evenly distributed over the genome.
`While the majority of repetitive elements are usually heavily methylated, gene-specific
`methylation appears to be rather restricted.
`Genomic DNA methylation patterns can be drastically altered in human tumors.14
`Due to the selective forces driving carcinogenesis, tumor cells are characterized by specific
`epigenetic changes that promote uncontrolled cellular proliferation.15 This explains the
`remarkable consistency of gene hypermethylation across all forms of human cancers.16
`When hypermethylation affects the CpG islands of genes it can trigger their stable repres-
`sion and thus has consequences that are functionally equivalent to genetic mutations.17
`The acquisition of such an “epimutation“ can therefore make an important contribution
`to cellular transformation. Over the past few years, the number of known epimutations
`has vastly increased. Prominent examples for genes “hit“ by hypermethylation-induced
`silencing include the cell-cycle regulators p15 and p16, the mismatch repair gene MLH1,
`as well as the apoptosis effector gene Apaf-1.18-20 These epimutations constitute a tumor-
`specific epigenetic program that is reflected by the typical characteristics of human cancer
`cells, the lack of cell cycle control and aggressive growth.
`
`Division of Epigenetics; Deutsches Krebsforschungszentrum; Im Neuenheimer Feld
`580; Heidelberg, Germany
`
`*Correspondence to: Frank Lyko, Ph.D.; Deutsches Krebsforschungszentrum; Im
`Neuenheimer Feld 580; 69120 Heidelberg, Germany; Tel.: +49.6221.423800;
`Fax: +49.6221.423802; Email: f.lyko@dkfz.de
`
`Received 11/04/05; Accepted 11/29/05
`
`Previously published as an Epigenetics E-publication:
`http://www.landesbioscience.com/journals/epigenetics/abstract.php?id=2375
`
`KEY WORDS
`
`DNA methylation, DNA methyltransferase
`inhibitors, epigenetic therapy
`
`©2006 LANDES BIOSCIENCE. DO NOT DISTRIBUTE.
`
`www.landesbioscience.com
`
`Epigenetics
`
`7
`
`

`

`DNA Methyltransferase Inhibitors
`
`E
`
`CR?
`I promoter
`
`,,,
`
`promoter
`
`~
`F
`
`promoter
`
`DNA methylation is catalyzed by DNA methyltransferases, a
`family of enzymes that comprises DNMT1, DNMT2, DNMT3A
`and DNMT3B in human cells.21 To analyze the functional role of
`individual DNA methyltransferases in hypermethylation-induced
`gene silencing, homologous recombination has been used to disrupt
`DNMT1 and DNMT3B in the colorectal cancer cell line
`HCT116.22,23 Lack of DNMT1 or DNMT3B had little effect on
`the DNA methylation pattern. However, a double knockout cell line
`lacking both enzyme activities showed a very strong reduction in
`5-methylcytosine content.23 The effects of DNA methyltransferase
`knockouts on genomic DNA methylation patterns have been ana-
`lyzed by two approaches:24 Differential methylation hybridization,
`which detects methylated regions in the genome through a CpG
`island microarray, and amplification of inter-methylated sites, which
`amplifies anonymous DNA sequences with a differential methylation
`pattern. It was demonstrated that cells lacking both DNMT1 and
`DNMT3B undergo a substantial loss of DNA methylation in the
`promoter region of tumor suppressor genes.24 This effect could not
`be seen in single DNMT knockout cell lines, and it was therefore
`concluded that different DNA methyltransferases cooperate in the
`epigenetic regulation of gene silencing.
`
`DNA METHYLTRANSFERASE INHIBITORS AS DEMETHYLATING
`AGENTS
`Based on the rationale that hypermethylation-induced gene
`silencing could be uncovered by gene demethylation and reactivation
`(Fig. 1), many laboratories have analyzed gene expression patterns in
`human cancer cells with experimentally reduced DNA methylation
`levels. To this end, DNA methyltransferase inhibitors,25 like 5-aza-
`cytidine, 5-aza-2'-deoxycytidine and, to a lesser extent, zebularine,
`have found widespread use. All three compounds need to be metab-
`olized and phosphorylated to deoxynucleotide triphosphates in
`order to become incorporated into DNA. DNA methyltransferases
`recognize the modified bases as natural substrate, but fail to resolve
`a covalent reaction intermediate,26 which results in the degradation
`of the covalently trapped enzymes. DNA demethylation is a direct
`consequence of enzyme trapping because of ongoing DNA replication
`in the presence of diminished DNA methyltransferase levels. After
`several rounds of DNA replication, this becomes detectable as a
`substantial decrease in the genomic methylation level.
`5-azacytidine is one of the best-known DNA methyltransferase
`inhibitors and has been used both in the laboratory and in the clinical
`practice for more than 20 years. The compound also represents the
`first established epigenetic drug, since it gained FDA approval for
`the treatment of myelodysplastic syndrome in May 2004. It was
`originally developed as a cytotoxic agent27 and its demethylating
`activity was only discovered through its ability to influence cellular
`differentiation.28 5-azacytidine is a ribose nucleoside and thus needs
`to be metabolized into a deoxyribonucleoside-triphosphate before it
`can be incorporated into DNA. Indeed, a major fraction of the drug
`becomes incorporated into RNA and thereby interferes with protein
`biosynthesis.29 This effect most likely plays a major role in the
`cytotoxicity of 5-azacytidine.
`5-aza-2'-deoxycytidine is the deoxyribose analogue of 5-azacytidine
`and a promising new drug for the treatment of myelodysplastic
`syndrome and other leukemias. This compound is a more potent
`hypomethylating agent, but it does not become incorporated into
`RNA. However, high doses of 5-aza-2'-deoxycytidine still induce
`substantial cytotoxicity, which is attributable to various side
`
`exon 1
`
`exon 1
`
`9
`i DNA methylatlon
`~
`+ DNMT inhibitors
`9
`
`exon 1
`
`C
`0
`
`I-+- .E:i
`~
`...
`0..
`""'
`
`en
`
`I-+- ·~
`
`•t,;
`
`C
`.S!
`
`I-+-~ C,
`
`~
`
`Figure 1. Establishment and reversion of epigenetic mutations. In healthy cells,
`tumor suppressor genes are unmethylated and expressed at normal levels. In
`cancer cells, hypermethylation of the promoter (and/or exon 1) region leads
`to gene silencing. Treatment with DNA methyltransferase inhibitors (DNMT
`inhibitors) removes the majority of 5-methylcytosine (CH3), and can reactivate
`gene expression.
`
`effects.30 It was also shown that the differentiation inducing ability
`of decitabine in cultured fibroblasts had a narrow dose window,
`which provided a first indication that lower doses might induce
`demethylation with lower levels of overall cytotoxicity.28 Both 5-aza-
`cytidine and 5-aza-2'-deoxycytidine are unstable in aqueous solu-
`tions,31,32 which has limited their clinical application.
`The more recently discovered nucleoside inhibitor zebularine33 is
`a stable cytidine analog that has shown promising characteristics in
`several in vitro assays as well as in mouse models.34-36 Cancer cell
`lines that responded to zebularine by DNA demethylation showed a
`complete depletion of the DNMT1 methyltransferase and a partial
`depletion of DNMT3A and DNMT3B.36 Gene expression profiling
`revealed that zebularine specifically affected the gene expression
`patterns of human cancer cells, even if the number of genes upregu-
`lated by drug treatment was rather small (Table 1). Interestingly,
`zebularine was also shown to be active in mice when administered
`orally,34 which suggested novel treatment modalities for epigenetic
`therapies. However, a recent study has demonstrated very low oral
`bioavailability in monkeys37 and the clinical value of zebularine still
`remains to be determined.
`
`IDENTIFICATION OF NOVEL TUMOR SUPPRESSOR GENES
`BY EXPERIMENTAL DEMETHYLATION
`Because of the strong correlation between promoter hypermethy-
`lation and gene silencing, demethylating drugs have been repeatedly
`used for the identification of epigenetically silenced cancer genes.
`Transcriptional profiling in a variety of inhibitor-treated cancer cell
`lines revealed a relatively small number of genes that were significantly
`upor downregulated by demethylating drugs (Table. 1). Two studies
`suggested that genes in the interferon pathway might be induced by
`5-aza-2'-deoxcycytidine treatment,38,39 but, otherwise, no defined
`gene expression signatures have been identified. Based on the rationale
`that tumor suppressor genes become hypermethylated and silenced
`
`8
`
`Epigenetics
`
`2006; Vol. 1 Issue 1
`
`

`

`DNA Methyltransferase Inhibitors
`
`Table 1 Overview of gene expression changes induced by DNMT inhibitors
`
`Cell Line
`
`HT29
`
`T24
`
`LD419
`
`RKO
`
`KYSE30, KYSE410,
`KYSE520
`
`LNCaP
`
`DU145
`
`Drug
`
`No. of Genes
`Analyzed
`
`Genes
`Upreg. (%)
`
`Genes
`Downreg. (%)
`
`Reference
`
`5-aza-CdR
`
`5-aza-CdR
`
`5-aza-CdR
`
`5-aza-CdR
`
`5-aza-CdR
`
`5-aza-CdR
`
`5-aza-CdR
`
`4608
`
`6600
`
`6600
`
`10814
`
`12599
`
`1176
`
`1176
`
`0.4
`
`0.9
`
`0.5
`
`0.5
`
`1.0
`
`2.0
`
`1.6
`
`0.8
`
`n.d.
`
`0.03
`
`0.2
`
`n.d.
`
`n.d.
`
`2.0
`
`1.8
`
`0.6
`
`38
`
`39
`
`39
`
`41
`
`40
`
`77
`
`77
`
`43
`
`43
`
`36
`
`36
`
`44
`
`0.3
`
`0.05
`
`0.05
`
`0.4
`
`demethylation and were subsequently
`found to be hypermethylated in col-
`orectal cancers.41 More recently, it has
`been shown that epigenetic silencing of
`SFRP genes plays a functional role in
`the constitutive activation of the WNT
`signalling pathway, which further
`underscored the significance of these
`events for colorectal carcinogenesis.42
`To compare the effects of pharma-
`cological and genetic inactivation of
`DNA methyltransferases, transcriptional
`profiles of knockout cells were compared
`to those obtained from inhibitor-treated
`cells.43 Interestingly, the effects of
`5-aza-2'-deoxycytidine appeared fully
`established after 24 hours and more
`closely resembled those of histone
`deacetylase inhibitor (TSA) treatment
`than the effects of DNA methyltrans-
`ferase knockouts. These results cannot
`be explained by the covalent trapping
`of DNA methyltransferases, followed
`by passive demethylation of DNA.
`Rather, it seems likely that active
`demethylation is triggered by the
`pharmacological inhibitors through an
`upstream mechanism involved in the regulation of both DNA
`methylation and histone deacetylation. Another surprising finding
`was the substantial fraction of genes found to be downregulated after
`exposure to 5-aza-2'-deoxycytidine. In contrast to the prevailing
`view, this suggested that hypomethylation may also be associated
`with gene silencing. Consistently, demethylation of the APM2
`promoter region closely coincided with its transcriptional silencing.43
`The validity of the results obtained with cell lines was confirmed
`by a study that used primary cells from leukemia patients after treatment
`with 5-aza-2'-deoxycytidine ex vivo or in vivo.44 The transcriptional
`profiles obtained in these experiments were comparable to those
`obtained in cultured cell lines, which suggested that the molecular
`responses in cells and in patients might be similar. However, only
`half of the induced genes contained putative CpG islands in their 5'
`promoter region and only a fraction of those showed changes in their
`methylation patterns. This result provides a good starting point for
`a review of the complex data on drug-induced methylation changes
`in patient material.
`
`THE USE OF DEMETHYLATING DRUGS IN CLINICAL TRIALS
`Both 5-azacytidine and 5-aza-2'-deoxycytidine have been used in
`dozens of clinical trials for more than 20 years.45,46 In agreement
`with standard procedures, most early trials used the drugs at concen-
`trations that were close to the maximum tolerated dose. The clinical
`results were generally disappointing and included severe toxicities
`related to prolonged myelosuppression. More recently, treatment
`schedules have been adjusted to increase drug tolerance,47 which has
`allowed the completion of several effective low-dose trials and
`provided the foundation for an increased clinical interest in DNA
`methyltransferase inhibitors. A randomized, controlled phase III trial
`of low-dose 5-azacytidine vs. supportive care in myelodysplastic
`syndrome patients48 showed a significant clinical benefit in
`
`HCT116
`
`HCT116 (1KO)
`
`5-aza-CdR
`
`5-aza-CdR
`
`T24, HCT15, CFAPC-1
`
`zebularine
`
`LD98, LD419, T-1,
`CCD-1070K
`
`zebularine
`
`8000
`
`8000
`
`13300
`
`13300
`
`OCI-AML2
`
`5-aza-CdR
`
`22000
`
`5-aza-CdR: 5-aza-2’-deoxycytidine; n.d., not determined
`
`0.5
`
`0.1
`
`0.1
`
`0.4
`
`1.0
`
`0.8
`
`0.6
`
`5-azacytidine
`-
`- - supportive care
`
`Q)
`
`~ -..!.
`
`C
`Q)
`> Q)
`Cl
`.!=
`C ·ro
`E
`
`~ 0.4
`:0
`Cl)
`.0
`0
`C.
`
`0.0
`
`0
`
`6
`
`12
`
`36
`30
`24
`18
`time at risk [months]
`
`···-·-·· .................... ,_.,
`
`42
`
`48
`
`54
`
`~ 0.4 -0
`
`Figure 2. Clinical efficacy of DNA methyltransferase inhibitors in cancer
`patients. Survival of myelodysplastic syndrome patients treated with 5-azacy-
`tidine or supportive care, as determined by Kaplan-Meier analysis. Adapted
`from reference 48 with permission from the American Society of Clinical
`Oncology.
`
`in cancer cells, pharmacological inhibition of DNA methyltransferases
`was also used to uncover novel cancer-related genes. Transcriptional
`profiling of esophageal squamous cell carcinoma (ESCC) cell lines
`treated with 5-aza-2'-deoxycytidine identified ten putative tumor
`suppressor genes that were found to be methylated in primary tumor
`tissues.40 Three of these genes, CRIP-1, Apo D and NU were over-
`expressed in ESCC cells and found to strongly inhibit their ability to
`grow colonies.40 This provided an indication that all three genes
`might function as suppressors of tumor growth. In a similar study,
`the genes encoding secreted frizzled-related proteins (SFRPs) were
`identified through a genomic screen for genes upregulated by
`
`www.landesbioscience.com
`
`Epigenetics
`
`9
`
`

`

`DNA Methyltransferase Inhibitors
`
`diagnosis
`
`treatment
`
`monitoring
`
`I\
`sample O
`
`I\
`sample 2
`I\
`sample 3
`
`sample 1
`
`sample2
`
`sample 3
`
`sampleO
`
`drug-treated patients (Fig. 2). Similar results
`appear to have been obtained in a phase III
`clinical trial of 5-aza-2'-deoxycytidine.49 In
`addition, promising clinical responses have
`also been reported in phase I trials of low-
`dose 5-aza-2'-deoxycytidine in other leukemias,
`including AML and CML.50,51 These results
`suggest that the demethylation induced by
`azanucleoside drugs might be of general
`benefit for the reversion of epigenetic lesions
`in cancer patients.
`Drug-induced methylation changes have
`only recently been analyzed in the clinical
`setting. One of the first such studies focused
`on the methylation of the p15 tumor sup-
`pressor gene
`in the bone marrow of
`5-aza-2'-deoxycytidine-treated patients with
`myelodysplastic syndrome.52 The results
`suggested demethylation in nine out of 12
`patients and also provided evidence of p15
`reactivation by immunohistochemistry in
`four patients. It remains a possibility that
`these changes are at least partially attributable
`to the expansion of nonclonal cells in
`responding patients, but gene reactivation
`was also shown in morphologically dysplastic
`cells from patients that were not in complete
`remission. This indicated a significant poten-
`tial of the drug to activate gene expression in tumors and also
`suggested a direct involvement of DNA demethylation.52 However,
`there appeared to be no correlation between p15 demethylation and
`clinical response to decitabine in a larger phase I study of decitabine
`in MDS and AML patients.50 Overall similar results were also
`obtained in a phase I study of 5-azacytidine in Epstein-Barr
`virus-associated tumors.53 While these tumors showed substantial
`demethylation in the EBV promoter, reactivation of EBV expression
`could only be observed in one case and there appeared to be no
`detectable clinical response.
`Biological responses might be maximized by changes in the drug
`delivery schedule, and for this reason different schedules are currently
`being tested. In some of these clinical trials, the clinical responses are
`being analyzed in parallel with the changes in genomic DNA methy-
`lation patterns (Fig. 3). One such example is provided by a recent
`phase I study of continuous 5-aza-2'-deoxycytidine infusion in a
`small group of patients with refractory solid tumors.54 While the
`patient group was too small to assess the clinical benefit of the treat-
`ment schedule, all patients showed indications for demethylation in
`blood samples. This effect could be observed either by quantitative
`PCR-based methylation analysis of the MAGE-1 promoter or by
`HPLC analysis of genomic DNA. Importantly, it could also be
`shown that DNA methylation reverted to pretreatment levels within
`four weeks, which indicated that drug-induced demethylation is
`transient. These findings are in agreement with an analysis of drug-
`induced demethylation effects observed after 5-aza-2'-deoxycytidine
`treatment of MDS patients.55 Here, the analysis of global DNA
`methylation levels from serial bone marrow samples indicated strong
`demethylation after the fourth treatment cycle, while karyotype
`normalization had occurred after the second treatment cycle. This
`indicated an important role of drug-mediated cytotoxicity, followed
`by a substantial demethylation of nonclonal cells. Again, DNA
`
`Figure 3. Epigenetic cancer therapy. After diagnosis, cancer patients can be treated with DNA methyl-
`transferase and/or histone deacetylase inhibitors. Treatment response is being monitored at clinical
`and molecular levels by analysis of tumor and peripheral blood samples. Correlations between clinical
`and molecular parameters will be important for the optimization of clinical treatment schedules.
`
`methylation returned to pretreatment levels several weeks after the
`last treatment cycle, which confirmed the transient nature of the
`demethylating effect.
`The dynamics of drug-induced demethylation will be important
`for the design of combination trials that address the potential synergy
`between demethylating drugs and other chemotherapeutic agents.
`Experiments with mouse xenograft tumors have shown a strong
`chemosensitizing effect for 5-aza-2'-deoxycytidine when the drug
`was administered a few days before standard cytotoxic drugs.56 This
`effect is presumably due to the epigenetic reactivation of apoptosis
`effector and DNA repair genes. 5-aza-2'-deoxycytidine has also been
`shown to be effective in a clinical combination study with the tyrosine
`kinase inhibitor imatinib mesylate (Gleevec).51 A complete hemato-
`logical response after decitabine treatment was seen in up to 50% of
`the Gleevec-resistant patients in the chronic phase of the disease.
`Furthermore, the demethylating effect was assessed by methylation
`analysis of LINE-1 elements as well as p15 tumor suppressor gene.
`Surprisingly, the degree of LINE-1 hypomethylation at the end of
`therapy was higher in patients who did not subsequently respond to
`therapy. This has been interpreted to reflect the possibility that
`hypomethylated cells die more rapidly, while cells resistant to therapy
`can withstand higher degrees of hypomethylation.51
`Lastly, it will also be critical to design treatment schedules that
`maximize the reversion of epigenetic mutations with minimal influ-
`ences on epigenetic effects required for normal cellular functions.57
`This is an important aspect in light of the finding that strong and
`continuous demethylation in animal and cellular models have been
`shown to promote genome instability and thereby induce tumor
`formation.58,59 A recent analysis used Apcmin mice with three different
`levels of the DNA maintenance methyltransferase DNMT1 to
`model the degree of hypomethylation observed in tumors (Yamada
`intestinal tumorigenesis but promotes microadenoma formation in
`
`10
`
`Epigenetics
`
`2006; Vol. 1 Issue 1
`
`

`

`azanucleoside derivatives
`
`5-azacytidine (Vidaza)
`5-aza-2´-deoxycytidine
`(Decitabine, Dacogen)
`
`Covalent enzyme trapping (26)
`
`zebularine
`
`Covalent enzyme trapping (33)
`
`small-molecule compounds
`
`hydralazine
`
`noncovalent enzyme inhibition (68)
`
`procaine (procainamide)
`
`binding to CpG-rich DNA (65),
`competitive inhibition
`of DNMT1 (66)
`
`~ ~N
`d o
`
`(
`
`O~N~
`
`..,,.
`
`DNA Methyltransferase Inhibitors
`
`Table 2 Overview of DNA methyltransferase inhibitors
`
`Compound
`
`Proposed Inhibitory Mechanism
`
`Structure
`
`determined, it suggests that epige-
`netic therapies could benefit from a
`further refinement of treatment
`schedules and the use of more specific
`drugs.
`
`DEVELOPMENT OF NOVEL DNA
`METHYLTRANSFERASE
`INHIBITORS
`The apparent lack of specificity
`and the inherent toxicity of nucleo-
`side inhibitors have invigorated the
`search for alternative DNA methyl-
`transferase inhibitors.25 The aim has
`been to identify compounds that do
`not need to be incorporated into
`DNA and thus would circumvent
`the toxic side-effects caused by the
`formation of protein-DNA adducts.61
`Alternatively, it might also be feasible
`to reduce DNA methyltransferase
`activity though oligonucleotides,
`either by allosteric enzyme inhibi-
`tion62 or by siRNA-mediated mRNA
`degradation.63,64
`There are several small-molecule-
`like compounds that have been
`described to induce demethylation
`in human cancer cell lines (Table. 2).
`Interestingly, some of these represent
`established drugs for the treatment
`of diseases not related to cancer. For
`example, the local anaesthetic pro-
`caine and its derivative procainamide,
`an approved antiarrhythmic drug,
`have been described to cause signifi-
`cant demethylation of genomic DNA
`in human cancer cell lines.65,66 This
`was confirmed by drug-induced
`demethylation and reactivation of
`the hypermethylated RARβ2 tumor
`suppressor gene.65 To explain the
`demethylating effects, it was suggest-
`ed that procaine and procainamide
`associate with CpG-rich sequences
`in the genome and thereby interfere
`with the DNA methylation machin-
`ery.65 Additionally, it has also been
`suggested
`that
`procainamide
`decreases the affinity of DNMT1
`for both DNA and the methyl-
`group donor S-adenosyl-L-methio-
`nine.66 Inhibition of DNA methylation has also been reported for
`hydralazine, an established antihypertensive drug that has been
`described to have demethylating activity in human cancer cell
`lines.67 Experimental results derived from gene-specific and
`whole-genome methylation analyses
`suggested
`significant
`drug-induced demethylation that was accompanied by corresponding
`gene reactivation.67,68 Results from a recent phase I of hydralazine in
`
`OH
`
`0
`
`~ '"
`Y'OH
`
`OH
`
`(-)-epigallocatechin-3-gallate
`(EGCG)
`
`noncovalent enzyme
`inhibition (70)
`
`HO
`
`RG108
`
`noncovalent enzyme
`inhibition (73)
`
`the colon. In this context, demethylation of centromeric satellite
`elements might play an important role in chromosomal instability.14
`The analysis of DNA from decitabine-treated MDS patients after
`various treatment time points demonstrated that drug-induced
`hypomethylation also led to demethylation of chromosome 1 satellite
`2 sequences,55 which has been linked to chromosomal instability.60
`While the clinical significance of this observation remains to be
`
`www.landesbioscience.com
`
`Epigenetics
`
`11
`
`

`

`DNA Methyltransferase Inhibitors
`
`cervical cancer patients also indicated demethylation and concomitant
`reactivation of hypermethylated tumor suppressor genes in tumor
`samples.68 While these results seemed impressive at first, it needs

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