<<

Leukemia (1997) 11, 175–180  1997 Stockton Press All rights reserved 0887-6924/97 $12.00

REVIEW Pharmacological approach for optimization of the dose schedule of 5-Aza-2’- deoxycytidine (Decitabine) for the therapy of leukemia

RL Momparler, S Coˆte´ and N Eliopoulos

De´partement de Pharmacologie, Universite´ de Montre´al; and Centre de Recherche Pe´diatrique, Hoˆpital Ste-Justine, Montre´al, Que´bec, Canada

5-Aza-2Ј-deoxycytidine (5-Aza-CdR; Decitabine) is an active drome (MDS).8 One problem in evaluating the clinical effi- antineoplastic agent in patients with leukemia. Since 5-Aza- cacy of 5-Aza-CdR in these studies is that most of the leu- CdR is an S phase specific agent and has a short plasma half- life, its antileukemic activity is dose schedule-dependent. Leu- kemic patients received prior therapy with Ara-C. Since both kemia patients who are candidates for 5-Aza-CdR therapy fol- 5-Aza-CdR and Ara-C are metabolized by the same enzymes, lowing relapse after therapy with arabinoside are at the possibility exists that subpopulations of drug-resistant leu- greater risk for the problem of drug resistance since these cyto- kemic cells were present at the time of administration of the sine analogues are metabolized by the same first course of 5-Aza-CdR therapy. enzymes. Due to its unique mechanism of action of demethylat- ing DNA, 5-Aza-CdR has the potential to activate tumor (growth) suppressor and differentiation genes that have been accidentally silenced by DNA methylation in leukemic cells. All Drug resistance to cytosine nucleoside analogues these factors should be taken into account in the design of the optimal dose schedule of this analogue. The optimal dose The major mechanisms of resistance to these cytosine nucleo- schedule of 5-Aza-CdR should be based on the kinetic para- side analogues are summarized in Table 1. Both 5-Aza-CdR meters of , its pharmacokinetics, its and Ara-C must first be phosphorylated by deoxycytidine kin- effects on DNA methylation and the cell cycle parameters of ase to be active inhibitors. Cells lacking this enzyme are com- the leukemic cells and the normal hematopoietic stem cells. 9,10 Since granulocytopenia is the major toxic effect produced by pletely resistant to these analogues. Cells with an increased 5-Aza-CdR, the use of hematopoietic growth factors to shorten pool of dCTP also show drug resistance to these analogues the duration of leukopenia should be investigated. Another since dCTP is a feedback inhibitor of deoxycytidine kinase.9 approach which we are investigating is to use the methods of In addition, dCTP reduces the incorporation of 5-Aza-CdR and gene therapy to insert the deaminase gene into normal Ara-C into DNA by competition for DNA polymerase with the hematopoietic progenitor cells so as to make them drug resist- 9,11 ant to 5-Aza-CdR. The use of other agents that can induce the triphosphate forms of these analogues. Finally, cells with differentiation of leukemic cells in combination with 5-Aza-CdR increased cytidine deaminase show drug resistance since may have the potential to increase the clinical effectiveness of deamination of these analogues results in a loss of antineo- this analogue for the therapy of leukemia. plastic activity.12–14 Keywords: 5-Aza-2Ј-deoxycytidine; DNA methylation; chemo- therapy Activation of tumor (growth) suppressor genes by 5-AZA- CdR Introduction The methylation in DNA of cytosine to 5-methylcytosine in 5-Aza-2Ј-deoxycytidine (5-Aza-CdR; Decitabine) was first the promoter region of a gene can suppress its expression.15 shown to be an active antileukemic agent in a mouse model As shown in Figure 1 the incorporation of 5-Aza-CdR into by Sorm and Vesely.1 In the mouse L1210 leukemia model, DNA can block DNA methylation11,16 which can result in 5-Aza-CdR was demonstrated to be much more effective than gene activation and the induction of differentiation.16–18 cytosine arabinoside (Ara-C),2 one of the most effective agents Recent investigations have shown that the lack of for the treatment of acute myeloid leukemia.3 In vitro studies expression of several types of tumor (growth) suppressor genes showed that 5-Aza-CdR is also a more potent antineoplastic is due to aberrant DNA methylation.19 As summarized in agent against human myeloid leukemic cells than Ara-C.4 Table 2, 5-Aza-CdR has been demonstrated to activate the Phase I–II studies in Canada and Europe on 5-Aza-CdR expression of tumor suppressor genes: p16,20,21 p15,22 VHL23 showed that this cytosine analogue could induce complete and retinoic acid receptor ␤ (RAR␤)24 that have been silenced remission in patients with acute leukemia who had relapsed by DNA methylation. Recent reports support the hypothesis 5–7 on conventional . In addition, 5-Aza-CdR has that RAR␤ is a tumor suppressor gene.25,26 shown clinical activity in patients with myelodysplastic syn- The role of most tumor suppressor genes in the pathogenesis

Table 1 Drug resistance to 5-Aza-CdR and Ara-C Correspondence: RL Momparler, Centre de Recherche Pe´diatrique, Hoˆpital Ste-Justine, 3175 Chemin Coˆte Ste-Catherine, Montre´al, Que´- bec H3T 1C5 Canada 1 Deficiency in deoxycytidine kinase This paper will also appear in a supplementary monograph of the 2 Increase intracellular pool of dCTP journal Leukemia, Workshop on Clinical Results with Decitabine (5- (a) Inhibition of deoxycytidine kinase AZA-2Ј-deoxycytidine) in Hematological Malignancies, Guest Editor (b) Competition for DNA polymerase Dr Hagop M Kantarjian, 1997; 11 3 Increase in cytidine deaminase Received 1 July 1996; accepted 16 October 1996 Review RL Momparler et al 176 action is also schedule-dependent. At diagnosis or relapse, the leukemic patient may have 109 to 1010 leukemic stem cells that should be eradicated by treatment in order to produce curative therapy. To accomplish this difficult task, every para- meter that can influence the response to the 5-Aza-CdR should be evaluated. In Table 3 are summarized the major parameters that should be taken into account in the design of the optimal dose schedule for 5-Aza-CdR. Chemotherapy studies in mice with leukemia showed that 5-Aza-CdR treatment produces ‘cures’ only at the higher dose levels.31 Extrapolation to the clinical therapy of leukemia, the higher dose ranges of 5-Aza-CdR should be given a priority. The steady state plasma concentration of 5-Aza-CdR during a Figure 1 Inhibition of methylation of cytosine by the incorporation continuous i.v. infusion should be above its minimal cytotoxic of 5-Aza-CdR (AZA) into DNA. After DNA replication, methylation of the newly synthesized DNA strand takes place at complementary sites concentration as determined by in vitro colony assays on determined by presence of methyl groups (CH ) in parental DNA human leukemic cells. Since the minimal cytotoxic concen- 3 − strand in order to maintain a specific pattern of DNA methylation. The tration for 5-Aza-CdR is estimated to be about 10 7 M,32 the incorporation of AZA in place of cytosine at a specific methylation steady state plasma concentration of this analogue should be − site produces an inactivation of DNA methylase resulting in a region above 10 7 M. This would ensure that adequate cytotoxic con- of hypomethylation. centrations of 5-Aza-CdR will be obtained in all anatomical compartments. In certain anatomical sites the concentration of 5-Aza-CdR will be less than that of the plasma concen- Table 2 Activation of tumor suppressor genes by 5-Aza-CdR tration. For example, the concentration of 5-Aza-CdR in the Tumor suppressor gene Ref. cerebral spinal fluid is about 50% lower than the concen- tration in the plasma.33 Due to the high level of cytidine

INK4B1 22 deaminase, the enzyme that inactivates 5-Aza-CdR, in liver p15 Herman et al 34 p16INK4 Otterson et al20 and spleen, it is possible that the concentration of this ana- Merlo et al21 logue could fall below the minimal cytotoxic concentration Retinoic acid receptor ␤ Coˆte´ and Momparler24 creating a biochemical sanctuary for leukemic cells in these von Hippel-Lindau (VHL) Herman et al23 organs. Serious consideration should be given to the use of cytidine deaminase inhibitors to overcome this problem.35 What is the optimal plasma concentration of 5-Aza-CdR? Clinical studies on Ara-C have shown that it is possible to use of leukemia is unclear at this time since DNA methylation very high doses with cytosine nucleoside analogues for the studies have been performed primarily on tumor cells. The therapy of leukemia.36 However, very high doses can produce tumor suppressor gene p16 does not appear to be modified unexpected side-effects such as the neurological toxicity that by methylation in leukemia.22 However, the p15 tumor sup- sometimes is produced by high dose Ara-C therapy.37 Since pressor gene was demonstrated to be hypermethylated in leu- intermediate dose Ara-C therapy can produce similar kemic cells in six of seven patients with AML and four of eight responses as high-dose therapy,3 it is reasonable to also favor patients with ALL.22 The p15 gene is a cyclin-dependent kin- the use of intermediate dose 5-Aza-CdR for the therapy of leu- ase inhibitor.27 Cyclin-dependent kinases play an important kemia. The optimal plasma concentration for 5-Aza-CdR is

role in the regulation of cellular progression into the S phase probably in the range of the Km value of deoxycytidine kinase, 32 and the control of cell proliferation. the rate limiting enzyme that activates this analogue. The Km In order for tumor suppressor genes to participate in cellular is the concentration of drug at which the enzyme reaction transformation, both alleles must be inactivated. Inactivation takes place at 50% of its maximum velocity. For deoxycytid-

of tumor suppressor genes can occur by chromosomal ine kinase the Km of 5-Aza-CdR is in the range of approxi- − deletion or rearrangement, mutation and DNA methylation. In mately 10 5 M.32 In order to obtain a plasma concentration of evaluation of the clinical response of leukemic patients to 5- this range, 5-Aza-CdR should be infused at a rate of about 1– Aza-CdR, the prior chemotherapy should be examined. As 2 mg/kg/h or 30–60 mg/m2/h.5,6 The pharmacokinetics of 5- illustrated in Figure 2, prior treatment with mutagenic or DNA Aza-CdR are also important. Since 5-Aza-CdR has a short damaging anticancer drugs have the potential to produce plasma half life of 15–20 min,6 a constant continuous i.v. mutations and chromosomal translocations28 that can inacti- infusion of this analogue is the best way to maintain a steady − vate tumor suppressor genes and possibly decrease the state level of approximately 10 5 M in the plasma. response to subsequent therapy with 5-Aza-CdR. The duration of the 5-Aza-CdR therapy will depend on the cell cycle parameters of the leukemic cells and the normal hematopoietic stem cells. Recent investigations on the cell Optimization of dose schedule of 5-Aza-CdR kinetics of myeloid leukemic cells in man gave an estimate of the length of the cell cycle with a mean value of 59.8 h (range The classical investigation by Skipper et al29 demonstrated 18–211 h).38 From these data the estimated optimal duration that the antileukemic activity of the S phase specific agent, of 5-Aza-CdR therapy is probably in the range of 90 h (only Ara-C, was markedly schedule-dependent in murine leukemia six of 45 patients showed a cell cycle time Ͼ90 h). models. These data plus additional data on the pharmacology The 5-Aza-CdR therapy will also induce terminal differen- of Ara-C were used by Momparler30 to propose a model for tiation of the dividing hematopoietic progenitor cells. There- the therapy of leukemia with this analogue. Since 5-Aza-CdR fore, the success of the therapy depends on the survival of the is also an S phase specific agent, its in vivo antileukemic resting hematopoietic stem cells. If prior therapy with other Review RL Momparler et al 177

Figure 2 Tumor (growth) suppressor gene can be inactivated by either mutation (M), chromosomal deletion or DNA methylation (CH3). Both alleles of the tumor suppressor gene must be inactivated to result in a complete loss of expression. Prior treatment with mutagenic or DNA damaging drugs by inactivation of both alleles can reduce the clinical response to 5-Aza-CdR.

Table 3 Parameters to consider in the design of optimal dose- gation in our laboratory is to use gene therapy to protect the schedule of 5-Aza-CdR for the therapy of leukemia normal hematopoietic stem cells from drug-induced toxicity. The enzyme cytidine deaminase, as discussed above, inacti- 1 Curative therapy of murine leukemia occurs at high vates cytosine nucleoside analogues such as 5-Aza-CdR and dose. 2 Minimal cytotoxic concentration for human leukemic Ara-C by deamination. We have cloned the human cytidine 40 cells. deaminase cDNA and used a retroviral vector to insert and 3 Inactivation by cytidine deaminase in liver and spleen. express this gene in murine fibroblast cells. We have observed 4 Pharmacokinetics. that these transduced cells are drug resistant to both 5-Aza- 14,41,42 5 Km value of deoxycytidine kinase. CdR and Ara-C. Recently, we have inserted the cytidine 6 Cell cycle parameters of leukemic stem cells. deaminase gene into normal murine hematopoietic cells and 7 Cell cycle parameters of normal hematopoietic stem 42 cells. demonstrated that they are drug resistant to Ara-C. We are 8 Stage of leukemic disease. currently investigating if these hematopoietic cells are also cross-resistant to 5-Aza-CdR. One problem to overcome before this type of gene therapy can be used clinically is the accidental insertion of the cytid- drugs or disease progression produces a marked reduction in ine deaminase gene into a few leukemic cells that may con- the number of these resting stem cells, the 5-Aza-CdR will taminate a normal marrow aspirate. Future technological produce a prolonged granulocytopenia. Under these con- advances in the cloning of normal hematopoietic stem cells ditions a more conservative approach to therapy with 5-Aza- can overcome this problem. In addition, if the leukemic cells CdR should be used. have an identifiable genetic marker that can be detected by the polymerase chain reaction, the possible contamination by leukemic cells can be verified prior to transplantation of the Approaches to overcome the problem of hematopoietic normal stem cells. toxicity

Hematopoietic growth factors, such as G-CSF or GM-CSF, are In vitro assays to evaluate clinical response currently used to reduce the duration of leukopenia following therapy with cytotoxic anticancer agents.39 We have observed Since a conventional clinical trial on an experimental therapy a gradual reduction in the white blood cell count (WBC) fol- for leukemia can take up to 5 years and a significant number lowing therapy with 5-Aza-CdR,5,6 which one would expect of patients for complete evaluation, in practice only a very for an agent that induces cellular differentiation. The terminal small number of trials can be performed. This poses a serious differentiation of the dividing hematopoietic progenitor cells problem for an agent such as 5-Aza-CdR whose clinical effi- induced by 5-Aza-CdR is a slow process requiring many days cacy is dose-schedule dependent. One approach to overcome and thus the feedback mechanism to activate the recruitment this problem is to perform a small series of pilot studies in of resting hematopoietic stem cells into the proliferation com- which in vitro tests before and after therapy are used to evalu- partment may take place much later in time than produced ate efficacy in conjunction with the conventional clinical by cytolytic drugs. If these conditions exist, treatment with response end points. For example, we have used an in vitro G-CSF or GM-CSF after 5-Aza-CdR therapy may reduce the drug sensitivity test to evaluate the response of leukemic duration of granulocytopenia. patients to 5-Aza-CdR. We observed in some patients that the Another approach to overcome the problem of hematopo- leukemic cells that survived the 5-Aza-CdR treatment showed ietic toxicity produced by 5-Aza-CdR which is under investi- drug resistance.5,6,13 One possible interpretation of these data Review RL Momparler et al 178 is that the dose schedule of 5-Aza-CdR used was capable of kemic blasts without any sign of drug toxicity (Momparler RL, eradicating all of the drug-sensitive leukemic blasts, but not Rivard GE, unpublished observation). In accordance with this the small fraction of drug-resistant leukemic cells. One observation is our mouse study, using L1210 leukemic cells important conclusion in this type of analysis is that only an that are drug resistant due to the deficiency in deoxycytidine ‘optimal’ dose schedule of 5-Aza-CdR can eliminate all the kinase, in which we demonstrated that 3-DU markedly drug-sensitive leukemic blasts and permit the survival of the increased the effectiveness of therapy with 5-Aza-CdR.10 drug-resistant cells. Using this approach it should be possible to evaluate different dose schedules of 5-Aza-CdR in a period of months instead of years by the conventional method and it Differentiation therapy of leukemia will also require fewer patients. The spontaneous frequency of mutation in mammalian cells One important aspect of the action of 5-Aza-CdR is its for a single copy gene is about 10−5.9 Since a cell contains capacity to induce leukemic cell differentiation.17,18 Since a two copies of the autosomal gene, deoxycytidine kinase, the very large number of leukemic cells must be eradicated by theoretical predicted frequency of drug resistance due to a the 5-Aza-CdR therapy, the possibility exists that a very small complete loss of enzyme activity due to a mutation in each fraction of the leukemic cell population will not reach the allele of this gene is 10−10 (10−5 × 10−5). This means that theor- threshold level of gene expression so as to undergo irrevers- etically one cell in about 10 billion leukemic stem cells would ible terminal differentiation. This subpopulation of leukemic be drug resistant due to the complete loss of deoxycytidine cells may survive the 5-Aza-CdR treatment. One approach to kinase activity. Therefore, the 5-Aza-CdR therapy would have overcome this problem is to use other differentiation agents to produce about a 10-log reduction in leukemic stem cells in combination with 5-Aza-CdR. The remarkable observation in order to permit survival of a leukemic cell with this type of that retinoic acid can induce complete remission of patients drug resistance. Only the ‘optimal’ dose schedule of 5-Aza- with acute promyelocytic leukemia48,49 suggests that this vit- CdR can produce a reduction in the number of leukemic stem amin A metabolite may be a good agent to test in combination cells of such a magnitude. If prior to 5-Aza-CdR treatment, the with 5-Aza-CdR. In vitro studies have shown an additive anti- leukemic patient is treated with a mutagenic drug, one would neoplastic effect of retinoic acid in combination with 5-Aza- 50 predict an increase in the frequency of drug resistance. CdR on human myeloid leukemic cells. Vitamin D3 which As discussed earlier, lack of response to 5-Aza-CdR therapy can also induce differentiation of leukemic cells was demon- can also be due to deletions, mutations or DNA hypermethyl- strated to produce a synergistic antineoplastic effect in combi- ation in tumor suppressor genes. In vitro tests, such as by nation with 5-Aza-CdR on human myeloid leukemic cells.51 Southern blotting with specific DNA probes to the region of Vitamin D analogues which have reduced calcium toxicity the first exon after digestion of genomic DNA with methyl- may be interesting agents to use in combination with 5-Aza- ation-sensitive and insensitive restriction enzymes can be used CdR in phase I studies in leukemia.52 Interferon-␣ also has to detect tumor suppressor genes that have been silenced by some potential to induce leukemic cell differentiation and has DNA hypermethylation.20–23 In addition, novel techniques shown an interesting interaction with 5-Aza-CdR on human have been developed to identify the specific sites of cytosine myeloid leukemic cells.53 An interesting differentiating agent methylation by DNA sequencing.43–45 These in vitro tests to investigate for use in combination with 5-Aza-CdR is which can be performed on the leukemic cells before and hexamethylene bisacetamide which has been evaluated after treatment with 5-Aza-CdR will assist in the evaluation of in patients with myeloid leukemia and myeodysplastic the clinical response to 5-Aza-CdR. syndrome.54 The action of steroids on leukemic cells is also an interest- ing area of investigation. It should be pointed out that 5-Aza- Approaches to overcome the problem of drug resistance CdR treatment of a steroid-resistant T lymphoid leukemic cell line produced an increased sensitivity to the cytolytic effects The use of other antileukemic agents in combination with the of dexamethasone,55 suggesting that this cytosine analogue 5-Aza-CdR therapy is the conventional way to overcome the may be an effective therapeutic agent for the treatment of ster- problem of drug resistance to this cytosine analogue. The oid-resistant lymphoid leukemias. Remarkably, estrogen choice of drug and its time of administration should be care- receptor expression is silenced by DNA methylation in most fully chosen. As discussed above, the use of a mutagenic or hematological neoplasms suggesting that modification of its DNA damaging antileukemic drug prior to or simultaneously gene may be one of the early events in the pathogenesis of with the 5-Aza-CdR therapy is not recommended. leukemia and suggesting a possible therapeutic role of de- Interesting experimental agents that can be used in combi- methylating agents.56 In this regard, it is interesting to note nation with 5-Aza-CdR are 3-deazauridine (3-DU) and cyclo- that 5-Aza-CdR was demonstrated to activate estrogen recep- pentyl-cytosine (CPC). 3-DU and CPC are biochemical modu- tor (ER) expression in ER-negative breast cancer cells by de- lators which inhibit CTP synthetase resulting in a secondary methylation of its gene.57 reduction in the intracellular pool of dCTP.46,47 As discussed above, one mechanism of resistance to 5-Aza-CdR is an increased pool of dCPT. Using in vitro studies with leukemic Conclusions cells we demonstrated a synergistic antineoplastic action when either 3-DU or CPC were used in combination with 5- The novel mechanism of action of 5-Aza-CdR in the activation Aza-CdR. One very interesting aspect about 3-DU is that cells of tumor (growth) suppressor and differentiation genes and its that are lacking deoxycytidine kinase activity are very sensi- potent antileukemic activity in animal models make it a very tive to the cytotoxic action of this analogue. In one terminal interesting agent to investigate in the therapy of leukemia. leukemic patient that was treated with 5-Aza-CdR, we Since it is an S-phase specific agent and its antileukemic detected drug resistance due to a deficiency in deoxycytidine activity is dose-schedule dependent, a careful analysis of its kinase; 3-DU therapy completely eliminated the blood leu- pharmacology is essential in order to design its optimal dose Review RL Momparler et al 179 schedule. This analysis should include a clinical history of 15 Tate PH, Bird AP. Effects of DNA methylation on DNA-binding prior therapy used on patients, in vitro tests for drug resistance proteins and gene expression. Curr Opin Genet Dev 1993; 3: and in vitro genetic tests of specific suppressor and differen- 226–231. 16 Jones PA, Taylor SM. Cellular differentiation, cytidine analogs and tiation genes. Current analysis favors the clinical use of 5-Aza- DNA methylation. Cell 1980; 20: 85–93. CdR in the initial treatment of leukemia. The use of experi- 17 Pinto A, Attadia V, Fusco IA, Ferrara F, Spada OA, DiFiore P. 5- mental therapy in place of conventional therapy in leukemia Aza-2Ј-deoxycytidine induces terminal differentiation of leukemic is a complex ethical and scientific issue. Perhaps leukemia blasts from patients with acute myeloid leukemia. Blood 1984; patients that have been identified as very high risk could be 64: 922–929. candidates to receive initial therapy with 5-Aza-CdR. Another 18 Momparler RL, Bouchard J, Samson J. Induction of differentiation and inhibition of DNA methylation in HL-60 myeloid leukemic interesting class of patients to investigate are those with sec- cells by 5-Aza-2Ј-deoxycytidine. Leukemia Res 1985; 9: 1361– 28 ondary leukemia, a disease that responds poorly to conven- 1366. tional chemotherapy. Since the methodology is available to 19 Little M, Wainwright B. Methylation and p16: suppressing the sup- make early diagnosis of secondary leukemia and the patient pressor. Nature New Med 1995; 1: 633–634. population at risk is known,28 it should be possible to initiate 20 Otterson GA, Khleif SN, Chen W, Coxon AB, Kaye FJ. CDKN2 gene silencing in lung cancer by DNA hypermethylation and kin- therapy with 5-Aza-CdR at the early stages of the disease per- Ј mitting an in depth evaluation of its clinical potential. etics of p16INK4 protein induction by 5-Aza 2 deoxycytidine. Oncogene 1995; 11: 1211–1216. 21 Merlo A, Herman JG, Mao L, Lee DJ, Gabielson E, Burger PC, Baylin SB, Sidransky D. 5ЈCpG island methylation is associated References with transcriptional silencing of the tumor suppressor p16/CDKN2/MTS1 in human cancers. Nature Med 1995; 1: 686–692. 1 Sorm F, Vesely J. Effect of 5-aza-2Ј-deoxycytidine against leukemic 22 Herman JG, Jen J, Merlo A, Baylin SB. Hypermethylation-associa- and hemopoietic tissues in AKR mice. Neoplasma 1968; 15: ted inactivation indicates a tumor suppressor role for p15INK4B1. 339–343. Cancer Res 1996; 56: 722–727. 2 Momparler RL, Momparler LF, Samson J. Comparison of the anti- 23 Herman JG, Latif F, Weng Y, Lerman MI, Zbar B, Liu S, Samid D, leukemic activity of 5-Aza-2Ј-deoxycytidine, 1-␤-D-arabinofurano- Duand SR, Gnarra JR, Linehan WM, Baylin SB. Silencing of the syl-cytosine and 5-azacytidine against L1210 leukemia. Leukemia VHL tumor-suppressor gene by DNA methylation in renal carci- Res 1984; 8: 1043–1049. noma. Proc Natl Acad Sci USA 1994; 91: 9700–9704. 3 Estey E, Keating MJ, Plunkett W, McCredie KB, Freireich EJ. Con- 24 Coˆte´ S, Momparler RL. Antineoplastic action of all-trans retinoic tinuous infusion of high-dose cytosine arabinoside without anthra- acid and 5-aza-2Ј-deoxycytidine on human DLD-1 colon carci- cyclines as induction and intensification therapy in adults under noma cells. Cell Pharmacol 1995; 2: 221–228. age 50 with newly diagnosed acute myelogenous leukemia. Semin 25 Houle B, Rochette-Egly C, Bradley WEC. Tumor-suppressive effect Oncol 1987; 14 (Suppl. 2): 58–63. of retinoic acid receptor ␤ in human epidermoid lung cancer cells. 4 Momparler RL, Onetto-Pothier N, Momparler LF. Comparison of antineoplastic activity of cytosine arabinoside and 5-aza-2Ј-deoxy- Proc Natl Acad Sci USA 1993; 90: 985–989. 26 Seewaldt VL, Johnson BS, Parker MB, Collins SJ, Swisshelm K. cytidine against human leukemic cells of different phenotype. Leu- ␤ kemia Res 1990; 14: 755–760. Expression of retinoic acid receptor mediates retinoic acid- 5 Rivard GE, Momparler RL, Demers J, Benoit P, Raymond R, Lin induced growth arrest and apoptosis in breast cancer cells. Cell KT, Momparler LF. Phase I study on 5-Aza-2Ј-deoxycytidine in Growth Diff 1995; 6: 1077–1088. children with acute leukemia. Leukemia Res 1981; 5: 453–462. 27 Serrano M, Hannon GJ, Beach D. A new regulatory motif in cell- 6 Momparler RL, Rivard GE, Gyger M. Clinical trial on 5-Aza-2- cycle control causing specific inhibition of cyclin D/CDK4. Nature deoxycytidine in patients with acute leukemia. Pharmacol Ther (London) 1993; 366: 704–707. 1986; 30: 277–286. 28 Smith M, McCaffrey RP, Karp JE. The secondary leukemias: chal- 7 Richel DJ, Colly LP, Kluin-Nelemans JC, Willemze R. The antileu- lenges and research directions. J Natl Cancer Inst 1996; 88: kaemic activity of 5-Aza-2Ј-deoxycytidine (Aza-dC) in patients 407–418. with relapsed and resistant leukaemia. Br J Cancer 1991; 64: 29 Skipper HE, Schabel FM, Mellet LG, Montgomery JA, Wilkoff LJ, 144–148. Lloyd HH, Brockman RW. Implications of biochemical, cyto- 8 Zagonel V, Giovanni LR, Maotta G, Babare R, Sardeo G, Gattie kinetic, pharmacologic and toxicologic relationships in design of V, De Angelis V, Monfardini S, Pinto A. 5-Aza-2-deoxycytidine optimal dose schedules. Cancer Chemother 1970; 54: 431–450. (Decitabine) induces trilineage response in unfavorable myelo- 30 Momparler RL. Model for the chemotherapy of acute leukemia ␤ dysplastic syndromes. Leukemia 1993; 7 (Suppl. 1): 30–35. with 1- -arabinofuranosylcytosine. Cancer Res 1974; 34: 1775– 9 Momparler RL, Onetto N. Drug resistance to cytosine arabinoside. 1787. In: D Kessel (ed.). Resistance to Antineoplastic Drugs. CRC Press: 31 Wilson VL, Jones PA, Momparler RL. Inhibition of DNA methyl- Ј Boca Raton, 1988, pp 354–367. ation in L1210 leukemic cells by 5-aza-2 -deoxycytidine as a 10 Momparler RL, Momparler LF. Chemotherapy of L1210 and possible mechanism for chemotherapeutic action. Cancer Res L1210/ARA-C leukemia with 5-aza-2Ј-deoxycytidine and 3- 1983; 43: 3493–3497. deazauridine. Cancer Chemother Pharmacol 1989; 25: 51–54. 32 Momparler RL. Molecular, cellular and animal pharmacology of 11 Bouchard J, Momparler RL. Incorporation of 5-aza-2Ј-deoxycytid- 5-Aza-2Ј-deoxycytidine. Pharmacol Ther 1985; 30: 287–299. ine 5Ј-triphosphate into DNA interactions with mammalian DNA 33 Chabot G, Rivard GE, Momparler RL. Plasma and cerebrospinal polymerase and DNA methylase. Mol Pharmacol 1983; 24: fluid pharmacokinetics of 5-Aza-2Ј-deoxycytidine in rabbits and 109–114. dogs. Cancer Res 1983; 43: 592–597. ␤ 12 Mu¨ller WEG, Zahn RK. Metabolism of 1-␤-D-arabinofuranosylura- 34 Ho DHW. Distribution of kinase and deaminase of 1- -arabinofu- cil in mouse L5178Y cells. Cancer Res 1979; 39: 1102–1107. ranosylcytosine in tissues of man and mouse. Cancer Res 1973; 13 Onetto N, Momparler RL, Momparler LF, Gyger M. In vitro bio- 33: 2816–2820. chemical tests to evaluate the response to therapy of acute leuke- 35 Laliberte´ J, Marquez VE, Momparler RL. Potent inhibitors for the mia with cytosine arabinoside or 5-aza-2Ј-deoxycytidine. Semin deamination of cytosine arabinoside and 5-Aza-2Ј-deoxycytidine Oncol 1987; 14: 231–237. by human cytidine deaminase. Cancer Chemother Pharmacol 14 Eliopoulos N, Bovenzi V, Momparler LF, Cournoyer D, Momparler 1992; 30: 7–11. RL. Gene transfer of human cytidine deaminase cDNA into murine 36 Wolff SN, Herzig RH, Phillips GL, Lazarus HM, Greer JP, Stein cells confers resistance to cytosine arabinoside, 5-aza-2Ј-deoxycy- IRS, Ray WA, Herzig GP. High-dose cytosine arabinoside and tidine and 2,2Ј-difluorodeoxycytidine. 9th NCI–EORTC sym- daunomycin as consolidation therapy for acute nonlymphocytic posium on new drugs in cancer therapy. Ann Oncol 1996; 7 leukemia in first remission: an update. Semin Oncol 1987; 14: (Suppl. 1): 58. 12–17. Review RL Momparler et al 180 37 Herzig RH, Herzig GP, Wolff SN, Hines JD, Fay JW, Phillips GL. 47 Bouffard DY, Momparler LF, Momparler RL. Enhancement of the Central nervous system effects of high-dose cytosine arabinoside. antileukemic activity of 5-Aza-2Ј-deoxycytidine by cyclopentenyl Semin Oncol 1987; 14: 21–24. cytosine in HL-60 leukemic cell line. Anti-Cancer Drugs 1994; 5: 38 Raza A, Preisler HD, Day R, Yasin Z, White M, Lykins J, Kukla 223–228. C, Barcos M, Bennett J, Browman G, Goldberg J, Grunwald H, 48 Huang ME, Ye UC, Chen SR, Chai JR, Lu JX, Zhoa L, Gu LJ, Wang Larson R, Vardiman J, Vogler R. Direct relationship between ZY. Use of all-trans retinoic acid in the treatment of acute promye- remission duration in acute myeloid leukemia and cell cycle kin- locytic leukemia. Blood 1988; 72: 567–572. etics: a leukemia intergroup study. Blood 1990; 11: 2191–2197. 49 Castaigne S, Chomienne C, Daniel MT, Ballerini P, Berger R, 39 Crawford J, Ozer H, Stoller R, Johnson D, Lyman G, Tabbara I, Fenaux P, Degos L. All-trans retinoic acid as a differentiation ther- Kris M, Grous J, Picozzi V, Rausch G, Smith R, Gradishar W, Yah- apy for acute promyelocytic leukemia: I. Clinical results. Blood anda A, Vincent M, Stewart M, Glaspy J. Reduction by granulo- 1990; 76: 1704–1709. cyte-stimulating factor of fever and neutropenia induced by 50 Momparler RL, Dore´ BT, Momparler LF. Effect of 5-Aza-2Ј-deoxy- chemotherapy in patients with small-cell lung cancer. New Engl cytidine and retinoic acid on differentiation and c-myc expression J Med 1991; 315: 164–170. in HL-60 myeloid leukemic cells. Cancer Lett 1990; 54: 21–28. 40 Laliberte´ J, Momparler RL. Human cytidine deaminase: purifi- 51 Momparler RL, Dore´ BT, Laliberte´ J, Momparler LF. Interaction of cation of enzyme, cloning and expression of its cDNA. Cancer 5-Aza-2Ј-deoxycytidine with amsacrine or 1,25-dihydroxyvitamin

Res 1994; 54: 5401–5407. D3 on HL-60 myeloid leukemic cells and inhibitors of cytidine 41 Momparler RL, Laliberte´ J, Eliopoulos N, Beause´jour C, Cournoyer deaminase. Leukemia 1993; 7: 17–20. D. Transfection of murine fibroblast cells with human cytidine 52 Dore´ BT, Uskokovic MR, Momparler RL. Increased sensitivity to deaminase cDNA confers resistance to cytosine arabinoside. a Vitamin D3 analog in HL-60 myeloid leukemic cells resistant to Anticancer Drugs 1996; 7: 266–274. all-trans retinoic acid. Leukemia 1994; 8: 2179–2182. 42 Momparler RL, Eliopoulos N, Bovenzi V, Le´tourneau S, Green- 53 Dore´ B, Momparler RL. Effect of 5-Aza-2Ј-deoxycytidine and inter- baum M, Cournoyer D. Resistance to cytosine arabinoside by feron-␣ on differentiation and oncogene expression in HL-60 retroviral-mediated gene transfer of human cytidine deaminase myeloid leukemic cells. Anti-Cancer Drugs 1992; 3: 281–287. into murine fibroblast and hematopoietic cells. Cancer Gene Ther 54 Andreeff M, Stone R, Michaeli J, Young CW, Tong WP, Sogoloff 1996; 3: 331–338. H, Kufe D, Rifkind RA, Marks PA. Hexamethylene bisacetamide 43 Frommer M, McDonald LE, Millar DS, Collis CM, Watt F, Grigg in myelodysplastic syndrome and acute myelogenous leukemia: GW, Molloy PL, Paul CL. A genomic sequencing protocol that a phase II clinical trial with a differentiation-inducing agent. Blood yields a positive display of 5-methylcytosine residues in individual 1992; 80: 2604–2609. DNA strands. Proc Natl Acad Sci USA 1992; 89: 1827–1831. 55 Gasson JC, Ryden T, Bourgeois S. Role of de novo DNA methyl- 44 Clark SJ, Harrison J, Paul CL, Frommer M. High sensitivity map- ation in the glucocorticoid resistance of a T-lymphoid cell line. ping of methylated . Nucleic Acids Res 1994; 22: Nature 1983; 302: 621–623. 2990–2997. 56 Issa J-PP, Zehnbauer BA, Civin CI, Collector MI, Sharkis SJ, David- 45 Paul CL, Clark SJ. Cytosine methylation: quantitation by auto- son N, Kaufmann SH, Baylin SB. The estrogen receptor CpG island mated genomic sequencing and GENESCAN analysis. Bio Tech- is methylated in most hematopoietic neoplasms. Cancer Res 1966; niques 1996; 21: 126–133. 56: 973–977. 46 Momparler RL, Vesely JK, Momparler LF, Rivard GE. Synergistic 57 Ferguson AT, Lapidus, RG, Baylin SB, Davidson NE. Demethyl- action of 5-Aza-2Ј-deoxycytidine and 3-deazauridine on L1210 ation of the estrogen receptor gene in estrogen-negative breast leukemic cells and EMT tumor cells. Cancer Res 1979; 39: cancer cells can reactivate estrogen gene expression. Cancer Res 3822–3827. 1995; 55: 2279–2283.