<<

(2001) 15, 875–890  2001 Nature Publishing Group All rights reserved 0887-6924/01 $15.00 www.nature.com/leu REVIEW

Nucleoside analogues: mechanisms of drug resistance and reversal strategies CM Galmarini1, JR Mackey2 and C Dumontet1,3

1Unite´ INSERM 453, Laboratoire de Cytologie Analytique, Faculte´ de Me´de´cine Rockefeller, Lyon, France; 2Medical and Experimental Oncology, Department of Oncology, University of Alberta, Edmonton, Alberta, Canada; and 3Service d’He´matologie, Centre Hospitalier Lyon Sud, France

Nucleoside analogues (NA) are essential components of AML ter drugs have mostly been used in the treatment of low-grade induction therapy ( arabinoside), effective treatments hematological malignancies.8,9 of lymphoproliferative disorders (fludarabine, ) and are also used in the treatment of some solid tumors The importance of NA chemotherapeutic agents has (). These important compounds share some gen- recently increased as a result of the introduction of new com- eral common characteristics, namely in terms of requiring pounds into clinical use and the expansion of indications into transport by specific membrane transporters, and the field of solid tumors. During the same period, there has interaction with intracellular targets. However, these com- been rapid progress in the understanding of mechanisms of pounds differ in regard to the types of transporters that most drug resistance to NA. In this review, we highlight the current efficiently transport a given compound, and their preferential interaction with certain targets which may explain why some knowledge concerning the cellular mechanisms of resistance compounds are more effective against rapidly proliferating to NA and possible strategies that may be used to overcome tumors and others on neoplasia with a more protracted evol- such resistance. ution. In this review, we analyze the available data concerning mechanisms of action of and resistance to NA, with particular emphasis on recent advances in the characterization of nucleo- Normal nucleoside transport and phosphorylation side transporters and on the potential role of activating or inac- tivating in the induction of clinical resistance to these compounds. We performed an extensive search of published Because physiologic are generally hydrophilic in vitro and clinical data in which the levels of expression of and do not readily permeate the plasma membrane, their nucleoside-activating or inactivating enzymes have been corre- cellular uptake occurs primarily via nucleoside-specific mem- lated with tumor response or patient outcome. Strategies aim- brane transport carriers (NT). Of the seven distinct NT activi- ing to increase the intracellular concentrations of active com- ties observed in human cells, only four have been defined pounds are presented. Leukemia (2001) 15, 875–890. 9 in molecular terms. These are classified into two categories: Keywords: nucleoside analogues; nucleoside transporters; 5 - 10,11 ; drug resistance equilibrative (ENT) or concentrative (CNT). Human equil- ibrative NTs (hENTs) are found in virtually all studied cell types and have broad permeant selectivity, accepting both Introduction and nucleosides as substrates. The two cloned hENTs differ in their sensitivity to inhibition by nano- Nucleoside analogues (NA) constitute an important class of molar concentrations of nitrobenzylmercaptopurine ribonu- used in the treatment of hematological malig- cleoside (NBMPR): hENT1 has es activity (equilibrative and 1 NBMPR-sensitive) while hENT2 possesses ei (equilibrative and nancies and, more recently, in solid tumors. These thera- 12–14 peutic compounds mimic physiological nucleosides in terms NBMPR-insensitive) nucleoside transport activity. The of uptake and metabolism and are incorporated into newly human concentrative nucleoside transporters (hCNT) are synthesized DNA resulting in synthesis inhibition and chain inwardly-directed transporters that are capable of transporting nucleosides against a concentration gradient by utilizing the termination. Some of these drugs also inhibit key enzymes 15–17 involved in the generation of the purine and pyrimidine transmembrane sodium concentration gradient. The and RNA synthesis, and directly activate the hCNT1 protein has greater affinity for pyrimidine nucleosides but also transports , while the hCNT2 protein trans- caspase cascade. All of these effects may lead to cell death. 18 The NA family includes various pyrimidine and purine ana- ports purine nucleosides and . In all cases, human logues. Among the pyrimidine analogues cytosine arabinoside NTs accept only dephosphorylated compounds. (ara-C, ) is extensively used in the treatment of triphosphate (dNTPs) pools present acute leukemia, while gemcitabine has more recently demon- within cells come from two sources, the de novo pathway strated activity in pancreatic, breast and lung .2–4 The which is specifically activated in replicating cells, and the sal- vage pathway, which is the main source of nucleotides in two first purine analogues to have been described were the 19,20 6 (6-MP) and 6-thioguanine (6- quiescent cells. The key step in the de novo pathway is TG). These compounds could more appropriately be desig- the conversion of diphosphates into deoxyri- nated as ‘’. Other new purine analogues are 2- bonucleoside diphosphates by reductase (RR). chlorodeoxyadenosine (2-CdA) and fludarabine.5–7 These lat- Replicating hematopoietic cells are heavily dependent on the de novo pathway. Conversely in resting cells the salvage path- way, which recycles bases and nucleosides derived from DNA or RNA catabolism, is the unique provider of dNTPs. These Correspondence: CM Galmarini, Laboratoire de Cytologie Analytique, Faculte´ de Me´decine Rockefeller, 8 Avenue Rockefeller, 69373, Lyon compounds are first phosphorylated by nucleoside Cedex 08, France; Fax: 33 4 78 95 35 05 such as (dCK), kinase 1 and Received 21 September 2000; accepted 1 February 2001 2, or kinase (dGK). This initial phosphoryl- Nucleoside analogues: mechanisms of drug resistance and reversal strategies CM Galmarini et al 876 Table 1 Human nucleoside transporters (NT) mediating uptake of nucleoside analogues

NT ara-Ca Gemcitabine 2-CdA Ref.

hENT1 ++++ ++++ +++ ++++ 16, 96, 242, 95, 243 hENT2 ND ++ ++ ++ 11, 54, 81 hCNT1 + ++++ − − 17, 54, 81 hCNT2 ND −−− 18, 81

ND, not determined. aDegree of transport: (+), low transported; (++), moderate transported; (+++/++++), highly transported; (−), not transported.

ation step constitutes the key step in the salvage pathway that to ‘self-potentiate’ their own cytotoxic effects. However each concludes with the formation of triphosphate or deoxytri- of these compounds also possesses specific properties in terms derivatives. There are thus two dNTP pools: one of drug–target interactions which may explain their differences derived from the de novo pathway that is predominantly in activity in various diseases. In particular the cytotoxic directed into replicating DNA and a second derived from effects of the purine analogues fludarabine and 2-CdA on salvage synthesis used for DNA repair.21,22 quiescent cells may be explained by interaction with targets involving DNA repair rather than replication, and direct or indirect effects on mitochondria. Mechanisms of action of nucleoside analogues and Ara-C All of the NA share common characteristics including active transport by membrane transporters (Table 1), activation by Ara-C (1-β-D-arabinofuranosylcytosine, cytosine arabinoside, kinases such as dCK allowing retention of the monophosphate cytarabine) is an structural analogue of deoxycytidine (dCyd) residues in the cell and the formation of the active triphos- (Figure 2) used for the treatment of acute and lym- phates metabolites, and dephosphorylation by 59-nucleotidase phomas. Ara-C differs from dCyd by the presence of a (59-NU) (Figure 1). Moreover, most of them have the ability hydroxyl group in the β-configuration at the 29-position of the sugar moiety. Intracellular penetration of ara-C depends on plasma ara-C concentrations.23–26 Standard-dose ara-C (SD ara-C; 100–200 mg/m2) achieves steady-state plasma levels of 0.5–1 µM.27,28 At these concentrations the expression of the hENT1 protein is the rate-limiting factor in ara-C uptake. In the presence of plasma concentrations greater than 50 µM, such as those reached with high-dose ara-C (HD ara-C; 2–3 g/m2), simple inward diffusion rates exceed those of pump- mediated transport.29 Once inside the cell, ara-C is phosphorylated by dCK and pyrimidine kinases to the active 59-triphosphate derivative ara- CTP.30,31 The catabolism of ara-C results from rapid deamin- ation by deaminase (CDD) to the non-toxic metab- olite arabinoside uridine while ara-CMP is dephosphorylated by the action of cytoplasmic 59-nucleotidase (59-NU).32 Ara- C cytotoxicity is believed to result from a combination of DNA polymerase inhibition and from incorporation of ara-CTP into

Figure 1 Representation of the metabolism and drug target interac- tions of deoxynucleoside analogues in proliferating cells (NA). NA enters cells via specific nucleoside transporters. Once inside the cell, NA are phosphorylated by (DCK), NMPK and NDPK to the active 59-triphosphate derivatives. NA catabolism can result from rapid deamination by to non-toxic metabolites. Cytoplasmic 59-nucleotidase (59Nucl) activity opposes that of DCK by dephosphorylating 59-monophosphate derivatives, thereby preventing the production of the active form. NA exerts their action by incorporation into newly synthesized DNA resulting in chain termination and cell death. Also, some NA indirectly block DNA replication by inhibiting ribonucleotide reductase (RR) , that in turn inhibit reduction of diphosphate (NDPs) to diphosphate (dNDPs). The decrease of deox- yribonucleotides triphosphate (dNTPs) pools favors incorporation of NA active 59-triphosphate derivatives into DNA. Figure 2 Chemical structure of pyrimidine analogues.

Leukemia Nucleoside analogues: mechanisms of drug resistance and reversal strategies CM Galmarini et al 877 DNA, in competition with deoxycytidine triphosphate (dCTP). This incorporation causes chain termination, resulting in a block of DNA synthesis.33–35 Sustained high cellular concen- trations of ara-CTP relative to that of dCTP are thought to favor drug incorporation into replicating DNA, thereby initiating the leukemic cell death associated with therapeutic response.36 Low incorporation of ara-CTP into the DNA of blast cells in vitro has been shown to be predictive of an adverse outcome in AML patients receiving ara-C-based therapy.37,38

Gemcitabine

Gemcitabine (difluorodeoxycytidine, dFdC) (Figure 2) is a new with promising activity in solid tumors.39,40 This compound is a dCyd analogue with two flu- orine substituted for the two atoms in the 29 position of the sugar.41 After the initial phosphorylation into dFd-CMP by dCK,42 gemcitabine is subsequently phos- phorylated by pyrimidine kinases to the active 59-diphosphate (dFd-CDP) and triphosphate (dFd-CTP) derivatives.43 Inacti- Figure 3 Chemical structure of purine analogues. vation of gemcitabine can occur by deamination into its inac- tive metabolite dFd-U by CDD or dephosphorylation of dFd- CMP by 59-NU.44,45 malignancies, in particular hairy-cell leukemia.51–53 2-CdA dFd-CTP is incorporated into DNA by replication synthesis enters cells via NT54 and is phosphorylated to 2-CdATP by in the C sites of the growing DNA strand.46 Once incorporated dCK, AMP kinase, and nucleoside diphosphate kinase. Mito- into the DNA strand, an additional natural nucleoside is chondrial dGK has also been identified as a 2-CdA phos- added, masking gemcitabine and preventing DNA repair by phorylating enzyme.55 2-CdAMP can be dephosphorylated by pair excision. Thereafter, the DNA polymerases are 59-NU. The net accumulation of the triphosphate derivative unable to proceed,47 a process designated as ‘masked DNA has been shown to depend on the relative concentrations of chain termination’. The active diphosphate metabolite of gem- dCK and 59-NU.56 citabine also inhibits DNA synthesis indirectly through inhi- 2-CdA is cytotoxic both to dividing and resting cells.57,58 bition of RR.47 This effect blocks the de novo DNA synthesis In dividing cells, this compound inhibits DNA synthesis by pathway and self-potentiates gemcitabine activity by decreas- incorporation of its triphosphate metabolite into DNA.59,60 ing intracellular concentrations of normal dNTPs (particularly Once incorporated 2-CdATP is capable of terminating chain dCTP). Reduction in cellular dCTP results in increased gemcit- elongation mediated by DNA polymerases, inducing an S abine incorporation into DNA and increased for- phase-specific apoptosis.61 2-CdA also inhibits DNA repli- mation of active gemcitabine di- and triphosphates, since dCK cation indirectly through its inhibitory action on RR,62 causing activity is down-regulated by high cellular dCTP levels. Other a subsequent reduction of the dNTPs pool required for DNA self-potentiating mechanisms of gemcitabine include a synthesis.59,63 The reduction of intracellular dATP results in decreased elimination of gemcitabine nucleotides by direct self-potentiation of 2-CdA as it enhances the incorporation of inhibition of CDD.48 Gemcitabine is not only incorporated triphosphate metabolites into DNA. Moreover, the decrease into DNA, but also into RNA.49 in dCTP could result in an increased rate of phosphorylation The spectrum of activity of gemcitabine in solid tumors may of 2-CdA and other NA, since dCK activity is regulated by be due to different characteristics. Compared with ara-C, gem- cellular dCTP levels. An original mechanism of 2-CdA cyto- citabine serves as a better transport substrate for membrane toxicity is related to the inhibition of ADA and S-adenosylho- pumps, is phosphorylated more efficiently, and is eliminated mocysteine hydrolase (SAHH).64 Inhibition of ADA could lead more slowly, favoring a longer retention time of the active to increased levels of Ado and dAdo, thereby causing the metabolite in tumor cells.48,50 Moreover, gemcitabine cytotox- inactivation of SAHH and reduced homocysteine formation. icity is enhanced by a number of unique self-potentiating This might result in altered reactions and mechanisms that contribute to maintaining high intracellular indirectly inhibit nucleotide synthesis and cellular prolifer- concentrations of the active metabolites. This prolonged pres- ation.65,66 ence of active gemcitabine derivatives may explain part of its Different mechanisms have been proposed to explain why effect on slowly dividing tumor cells. These differences, 2-CdA is toxic to a cell that is not undergoing replicative DNA together with greater lipophilicity, masked chain termination, synthesis. First, 2-CdA-induced killing in non-dividing cells RNA incorporation and RR inhibition, which are not observed occurs by inhibition of cellular DNA repair processes. Incor- with ara-C, may explain why gemcitabine is, and ara-C is not, poration of 2-CdATP into DNA by the repair machinery ter- active in solid tumors. minates the nucleotide excision repair process leading to the progressive accumulation of DNA single-strand breaks which eventually initiate apoptosis by p53-dependent or p53-inde- 2-CdA pendent pathways.67,68 p53 activation subsequently regulates the levels of expression of p53-dependent proteins such as 2-CdA (Figure 3) is an analogue of (dAdo) Bcl-2 and Bax leading to the activation of the caspase path- resistant to deamination by (ADA). 2- way.69 Second, incorporation of 2-CdA into DNA may alter CdA is commonly used in the treatment of indolent lymphoid gene with a consequent depletion of proteins

Leukemia Nucleoside analogues: mechanisms of drug resistance and reversal strategies CM Galmarini et al 878 required for cell survival. It has been demonstrated that when sine triphosphate (dATP) for incorporation into the A sites of 2-CdA metabolites were present in one or both DNA strands, elongating DNA by DNA polymerases.84 Once incorporated it the yield of full-length transcripts was reduced.70 Moreover, is capable of terminating chain elongation mediated by DNA RNA polymerase did not bind as well to 2-CdATP-containing polymerases α (F-ara-ATP).85 RR inhibition causes a sub- promoters. In addition, on binding to the substituted promoter, sequent reduction of dNTPs pools.63,86 Similarly to 2-CdA, the RNA polymerase had an altered conformation that led to decrease of cellular dATP and dCTP results in a self-potenti- enhanced proteolytic clipping by endoproteinase Glu-C.70 ation of the drug and other NA. Other enzyme targets of F- Alternatively, it has been shown that 2-CdA cytotoxicity ara-ATP in dividing cells include DNA , DNA poly- may involve alterations of mitochondrial function or integrity. merase α, DNA ligase and topoisomerase II.84 Thus, the mech- 2-CdA has been shown to disrupt the integrity of mitochondria anism of action of F-ara-A in proliferating cells is mainly cell in CLL cells.69 The nucleoside-induced damage leads to the cycle-specific, and incorporation of F-ara-A into DNA during release of the pro-apoptotic mitochondrial protein cyto- is required for the induction of apoptosis.87 chrome c, thereby initiating the caspase proteolytic cascade. In non-dividing cells, the inhibition of cellular DNA repair Moreover, 2-CdATP can cooperate with cytochrome c and processes appears to be the major mechanism of cytotoxicity Apaf-1 to activate caspase-3 and trigger a caspase path- of F-ara-A. Incorporation of F-ara-AMP into DNA by the repair way.71,72 2-CdA also interferes either directly or indirectly machinery terminates the polymerization step of the nucleo- with mitochondrial transcription that will eventually reduce tide excision repair process causing irreversible damage the levels of mitochondrial proteins necessary for electron recognized by the cells, which would signal for p53-mediated transport and oxidative phosphorylation. In vitro studies have or PARP-mediated apoptosis.20 As occurs after exposure to 2- demonstrated that 2-CdATP is incorporated into mitochon- CdA, fludarabine may induce PARP activation resulting in drial DNA by mitochondrial DNA polymerase.62 This incor- consumption of NAD and depletion of total nucleo- poration may decrease transcription by mitochondrial RNA tides with subsequent cell death.88 polymerase. On the other hand, high intracellular 2-CdATP Other mechanisms of action of F-ara-A may explain its cyto- levels could indirectly repress transcription of mitochondrial toxicity in non-cycling cells.82 First, F-ara-AMP and F-ara-ATP genes by inhibiting reduction of ADP to dADP.73 This leads incorporation into RNA results in premature termination of the to high ADP concentrations that cause a net influx of adenine RNA transcript, impairing its function as a template for protein nucleotides into mitochondria and an increase in the intrami- synthesis.89 F-ara-ATP also inhibits RNA synthesis by sup- tochondrial ATP concentration.59,74 High intramitochondrial pressing the activity of RNA polymerase II.89 Inhibition of RNA ATP levels in turn inhibit mitochondrial RNA polymerase transcription correlates with the cytotoxic action of fludarab- activity.74 ine in CLL cells.90 Secondly, F-ara-ATP is the most potent PARP activation has been suggested to be involved in p53- nucleotide activator of APAF-1, with the consequent acti- independent mechanisms of cell killing by 2-CdA.57 In vation of the caspase-9 and caspase-3 pathways.71 Moreover, addition to being a target for proteolytic cleavage during F-ara-A has been reported to downregulate Bcl-2 mRNA apoptosis, PARP is thought to play a role in DNA repair by expression, thereby favoring apoptosis.91 Finally, F-ara-A signaling genomic damage.75 PARP binds to DNA breaks, affects mitochondria but only as a late event, suggesting an becomes activated and, using NAD as a substrate, adds ADP- indirect mechanism related to the p53-apoptotic pathway.69 polymers to a range of nuclear proteins including itself, The different effects of F-ara-A and 2-CdA on mitochondria histones and DNA repair enzymes.76,77 These and other find- integrity may be related to the fact that 2-CdATP may accumu- ings have led to a proposed model of 2-CdA action involving late more rapidly than F-ara-ATP in the mitochondria because the following sequence of events: inhibition of ongoing DNA it is converted with higher efficiency by mitochondrial dGK.92 repair by 2-CdA with accumulation of DNA breaks, PARP It has also been stated that 2-chloro derivatives are slightly activation with resultant consumption of NAD, and depletion more hydrophobic than the corresponding 2-fluoro derivatives of total adenine nucleotides.57,76–78 Given that the cell mem- and therefore may be able to insert more easily into the mito- brane requires ATP for many of its functions, it seems likely chondrial membrane, thereby affecting their integrity.69 In that PARP-mediated ATP depletion might contribute to the summary inhibition of DNA repair, termination of mRNA tran- cell membrane disruption that occurs during apoptosis.79 scription and the consequent depletion of proteins required for cell survival, as well as the capacity of F-ara-ATP to acti- vate the apoptosome pathway appear to contribute to the Fludarabine cytotoxicity of fludarabine in non-dividing cells.

β 9- -D-arabinosyl-2-fluoroadenine (F-ara-A) (Figure 3) is an Mechanisms of resistance to nucleoside analogues analogue of adenosine (Ado) which is resistant to deamination by adenosine deaminase (ADA).80 F-ara-A is administered as Three general mechanisms of resistance to NA have been the 59-monophosphate form (fludarabine). The phosphate described in cell lines and in clinical samples. A primary group on the 59- position of the has the sole func- mechanism of resistance to NA arise from an insufficient intra- tion of making the clinical formulation -soluble. Before cellular concentration of NA triphosphates, which may result entering cells, fludarabine is rapidly dephosphorylated by from inefficient cellular uptake, reduced levels of activating membrane CD73 into F-ara-A which is transported inside the enzymes, increased NA degradation by increased 59-NU or cell via hNTs.81 Like other NA, it requires phosphorylation to CDD activity, or expansion of the dNTP pools.93 A second its triphosphate form, F-ara-ATP for cytotoxic activity.82,83 The type of resistance mechanism may be due to the inability to initial step in this activation process is performed by dCK. achieve sufficient alterations in DNA strands or dNTP pools, Like 2-CdA, F-ara-A is cytotoxic both against dividing and either by altered interaction with DNA polymerases, by lack resting cells. In dividing cells, this compound inhibits DNA of inhibition of RR, or because of inadequate p53 exonuclease synthesis and RR.82 DNA synthesis inhibition is mediated by activity. Finally, drug resistance may be a consequence of a competition of the triphosphate metabolite with deoxyadeno- defective induction of apoptosis.

Leukemia Nucleoside analogues: mechanisms of drug resistance and reversal strategies CM Galmarini et al 879 Membrane transporters decrease in the activity level of this enzyme as one of the mechanisms responsible for clinical resistance to ara-C, flud- In vitro studies have demonstrated that NT-deficient cells are arabine or 2-CdA. Conversely, other authors reported no sig- highly resistant to NA.94 Several cell lines exhibit cross-resist- nificant relationship between dCK activity and clinical out- ance to a spectrum of NA because of reduced uptake.11,54,95 come to NA in these diseases.120,121 In isolated leukemic blasts, sensitivity to ara-C, fludarabine Structural analyses of the dCK gene have show inactivating and 2-CdA correlated with the cellular abundance of and deletions as a cause of dCK deficiency in vitro. hENT196,97 determined by cellular binding of NBMPR. In Owens and co-workers122 claimed that to acquire a drug- these cells, the number of such binding sites ranged from 500 resistant phenotype, cells required two independent mutations to 27600 sites/cell.11,26 As initial rates of cellular drug uptake that markedly disrupt the catalytic activity of the enzyme. are, in general, proportional to hENT1 abundance, this wide However, these cDNA mutations rarely occur in vivo and variation suggests that transport-deficient cells would display therefore may not constitute a major mechanism of clinical a drug-resistant phenotype. In AML patients, Wiley et al98 NA resistance.115,116,123 Moreover, it seems that most of the observed that low transport rates correlated with poor clinical aminoacid replacements did not occur in a functionally rel- responsiveness to ara-C therapy. Although the measurement evant region of the enzyme.124 More recently alternatively of hENT1 expression by a quantitative flow cytometric assay spliced forms of dCK mRNA were detected in leukemic blasts with the hENT1 , SAENTA fluorescein, shows promise from patients with clinically resistant AML but not in leukemic in identifying transport-deficient leukemic cells, the clinical blasts from patients with sensitive AML.125 These alternatively significance of NT deficiency to in vivo resistance is not yet spliced forms of dCK mRNA were shown to be inactive in an well established. This is due to the difficulty of assessing drug in vitro dCK activity assay. These data indicate that the pres- transport in clinical samples, the contribution of transporters ence of dCK splice products may contribute to the occurrence other than hENT1, and the technical difficulties of quantifying of clinical ara-C resistance. NTs in neoplastic clones admixed with normal cells.11 Structural alterations of the dCK gene complex other than mutations may play a role in drug resistance to NA in vivo.126 Downregulation of dCK activity in cytarabine-resistant cells Deoxycytidine kinase has been shown to correlate with hypermethylation of the pro- moter region of the gene.127,128 Decreased levels of dCK Deoxycytidine kinase is the rate-limiting enzyme that cata- mRNA has been shown to be induced under various con- lyzes the initial step of the 59-phosphorylation of dCyd to ditions in vitro.127,129 Finally dCK activity may be regulated dCMP and of many NA to their corresponding monophos- post-translationally, since this enzyme is feedback-inhibited phates.99–101 The dCK activity is high in quiescent cells in by dNTPs (particularly dCTP) and by some phosphorylated which it phosphorylates nucleosides necessary for DNA repair analogues such as ara-CTP.130 and, depending on the cell type, may increase several-fold when the cells enter the S-phase of the .102–105 Deoxycytidine kinase activity has been shown to be Deoxyguanosine kinase decreased or absent in different cell lines resistant to NA.106–109 Transfection of the dCK gene in dCK-deficient tumor cell lines Deoxyguanosine kinase (dGK) is another enzyme responsible restores in vitro sensitivity to ara-C.110,111 Moreover, in vitro for the phosphorylation of purine deoxynucleosides, in mam- models have shown crossresistance between 2-CdA, gemcita- malian cells. This enzyme showed a broad substrate speci- bine, fludarabine and ara-C with reduced dCK activity as the ficity towards natural purine deoxynucleosides, as well as underlying determinant of resistance.109,112 It is noteworthy towards nucleoside analogues such as ara-G and 2- that in vitro only significant reductions in the levels of dCK CdA.55,73,92 dGK is located predominantly in the mitochon- activity (10% or less) are associated with a resistant pheno- dria.131 However, Petrakis et al132 described a novel amino- type, suggesting that under baseline conditions dCK is present terminally truncated isoform that corresponds to about 14% of in excess. the total dGK mRNA population in mouse spleen. This isoform In the clinical setting, it is important to note that bone mar- cannot translocate into the mitochondria and thus may rep- row and lymphoid tissues exhibit the highest activities of dCK resent a cytoplasmic enzyme. resulting in the clinical success observed when treating these Mitochondrial dGK is responsible for phosphorylation of target tissues with NA.113,114 However, the relevance of dCK purine and their analogues in the mito- deficiency in clinical resistance to NA is controversial (Table chondrial matrix providing the dNTPs necessary for mitochon- 2). In childhood acute lymphoblastic leukemias (ALL),115,116 drial DNA synthesis.133–136 Cytoplasmic dGK may also con- AML117,118 and lymphoproliferative disorders119 some authors tribute to the supply of purine deoxynucleotides for nuclear observed decreased expression of the dCK gene or significant DNA replication and repair.132 In this way, nucleoside ana-

Table 2 Correlation between low levels of expression of dCK gene or low dCK activity and drug resistance in hematological malignancies

Author Disease n Drug Technique Resistance Ref.

Tattersal AML 30 ara-C EA assay Yes 117 Stammler ALL 107 ara-C rt-PCR Yes 116 Albertioni CLL 17 2-CdA EA assay No 120 Kawasaki HCL/CLL 7/25 2-CdA IA Yes 119 Leiby NHL 25 Fludarabine EA assay No 121

N, number of patients; EA, enzyme activity; IA, immunoassay; rt-PCR, polymerase chain reaction.

Leukemia Nucleoside analogues: mechanisms of drug resistance and reversal strategies CM Galmarini et al 880 Table 3 Human 59-nucleotidase (59-NU) activities involved in NA resistance

9 Enzyme activity Cytosolic low Km Cytosolic high Km Cytosolic Ecto-5 -NU 59-NU 59-NU 59(39)-nucleotidase (CD73)

Ref. 244, 245 131, 140, 244 244 131, 246 NMM (kDa) 143 210 45 120–160 SMM (kDa) 76 53 22 60–80 Preferred substrates CMP.dUMP.G DIMP.dGMP dIMP.dUMP.dT AMP MP.AMP MP.dGMP Optimum pH 7.4–9 6.5 6.0–6.5 7.0–8.0 Stimulation dATP, GTP, 2, 3 DPG ATP, ADP Inhibition ADP, ATP(dADP, dATP, PI) Pi, orthophosphate (Pi)

NMM, native molecular mass; SMM, subunit molecular mass.

logues phosphorylated by dGK may exert their cytotoxic patients with no expression.144 However, due to the multi- effects by interference with both mitochondrial and nuclear plicity of 59-NU activities, and the limited ability of classical DNA synthesis. Whether inactivation of dGK is involved in biochemical methodology to differentiate between these resistance to NA is not presently known. activities, the particular 59-NU enzyme(s) with the largest impact on clinical resistance to NA have not yet been convincingly identified.145 59-Nucleotidase Cytidine deaminase 59-NU dephosphorylates nucleoside monophosphates and cytotoxic mononucleotides through its hydrolysis of the ester- Cytidine deaminase catalyses the inactivation of cytidine and bond between the 59-carbon and the phosphate group. 59-NU dCyd to uridine and , respectively. CDD also activity therefore opposes that of dCK. 59-NUs comprise a deaminates ara-C and gemcitabine.146 Several lines of evi- large and complex group of enzyme activities differing in dence have indicated a role for increased levels of CDD in cellular localization, pH sensitivity, and inhibition or stimu- the development of resistance to ara-C.147–149 Different studies lation by ATP (Table 3). Two broad classes of 59-NU are impli- have revealed that transfection of the human CDD gene into cated in drug resistance to NA: cytosolic and membrane- murine fibroblasts and hematopoietic cells confers drug bound 59-NUs. Membrane-bound or ‘ecto-59-NU’ is also resistance to ara-C and gemcitabine.44,150,151 known as CD73, and is frequently expressed in acute leuke- However, the in vivo correlation between CDD activity and mias.137,138 The physiological function of CD73 is the salvage ara-C resistance remains controversial and the relative contri- of extracellular nucleotides into nucleosides capable of cellu- bution of CDD to drug resistance has not yet been fully eluci- lar entry by means of nucleoside transport proteins. In con- dated (Table 4). The clinical relevance of a high level of CDD trast, several intracellular 59-NU activities have been ident- activity as a major cause of ara-C resistance in AML patients ified and cloned to date.139–141 The ‘low-Michaelis-Menten has been emphasized by several investigators.118,152–154 Con- 9 117 constant’ (Km)5-NU is specific for pyrimidine monophos- versely, Tattersal et al did not find a relationship between phates at low micromolar Km values and is inhibited by ATP. increased CDD activity and resistance to therapy in AML 9 The other, high Km 5 -NU is purine selective at high micromo- patients. 142,143 lar Km values, and it is activated by ATP. In vitro, Structural analyses of the CDD gene correlated polymor- increased 59-NU activity has been consistently associated with phism at codon 27 to substantially different deamination rates nucleoside drug resistance.45,58,109 of ara-C in vitro.155 This structural aberration did not seem to In the clinic, levels of cyto-59-NU activity have been corre- represent a major cause of the observed differences of CDD lated to clinical response in CLL, HCL and AML (Table 4). In activities between ara-C-sensitive and ara-C-resistant patients. CLL and HCL, Kawasaki et al119 demonstrated that pretreat- Schro¨der and co-workers154 demonstrated a significant corre- 9 ment levels of high Km 5 -NU allowed prediction of 2-CdA lation of the amount of CDD mRNA with CDD enzyme activi- responsiveness, since 59-NU levels were significantly lower in ties in AML blasts suggesting that variations in CDD activity responders than in non-responding patients. In AML, we have result from differences in gene expression. It therefore appears 9 reported that patients whose blasts expressed high Km 5 -NU that CDD activity in vivo is correlated with transcriptional mRNA had shorter time-to-relapse and overall survival than regulation rather than with CDD gene aberrations.

Table 4 59-NU and CDD enzyme activity measurements in hematological malignancies

Author Disease Drug Enzyme Results Ref.

Galmarini AML ara-C 59-NU High mRNA levels correlates with DR 144 Kawasaki HCL/CLL 2-CdA 59-NU Increased EA correlates with DR 119 Colly AML ara-C CDD Increased EA correlates with DR 118 Jahns AML ara-C CDD Increased EA correlates with DR 153 Schro¨der AML ara-C CDD Increased EA correlates with DR 154 Tattersal AML ara-C CDD No relationship to DR 117

Tech, technique; EA, enzyme activity; DR, drug resistance.

Leukemia Nucleoside analogues: mechanisms of drug resistance and reversal strategies CM Galmarini et al 881 Levels of dCTP resistance to NA.68,188 p53 appears to have a specific role in resistance to NA since it possesses exonuclease activity, and Various cell lines and AML blasts containing high levels of may therefore be able to excise illegally DNA-incorporated dCTP have been found to be resistant to ara-C.126,156,157 dCTP NA. Recently, Feng et al reported that although the 39-59 levels regulate ara-C metabolism at three levels: by feedback exonuclease of wild-type p53 (wt-p53) protein was able to inhibition of dCK activity with the consequent decrease in ara- bind and excise gemcitabine residues from DNA in vitro, C phosphorylation;117 by allosteric activation of the catabolic removal of the drug from cellular DNA was slow enzyme, CDD;126 and by competing with ara-CTP for incor- in sensitive cells containing wt-p53, and undetectable in drug- poration into DNA.158,159 Moreover, Ohno and coworkers160 resistant mut-p53 cells. Collectively, these data imply that have shown that dCTP in sufficiently high concentrations can molecular regulators of apoptosis should be taken into con- overcome ara-C DNA strand termination so that strand elong- sideration when analyzing factors associated with resistance ation continues past the ara-C incorporation site. dFd-CTP to NA. also compete with dCTP for DNA polymerase and for incor- poration into DNA.46 Thus, elevated levels of dCTP will favor the survival of cells exposed to these NA. Reversal strategies

It is now widely accepted that the antileukemic effect of NA Ribonucleotide reductase and DNA polymerases requires sufficiently high intracellular concentrations of tri- phosphate analogues in tumor cells.38,82,189 One approach has RR consists of two non-identical proteins called M1 and M2, been to develop compounds which are insensitive to degra- both of which are required for activity. Protein M1 contains dation. Fludarabine and cladribine are resistant to the effect the substrate and effector binding sites and is present through- of ADA. Another approach could consist in the use of specific out the cell cycle in proliferating cells. Protein M2 is a dimer inhibitors of degradative enzymes, such as 59-NU. Combi- that contains stoichiometric amounts of non-heme iron and a nations of a 59-NU inhibitor with a NA could be expected to unique tyrosyl free radical essential for reductase activity. Its potentiate the cytotoxicity of the NA. However, there is still expression is specific of S phase and in quiescent cells M1 uncertainty regarding the relevant 59-NU enzyme(s) that and M2 mRNA and proteins are not detectable.161 Clinically mediate NA resistance, and there are currently no clinically useful NA target RR, altering dNTPs concentrations.62,162 available inhibitors of 59-NU activity. Because many potential Recently, Goan et al163 reported a human KB cancer cell line inhibitors of 59-NU are themselves nucleoside analogues, resistant to gemcitabine with overexpression of the M2 sub- competition with cytotoxic NA for membrane transporter sites unit as the sole mechanism explaining this resistance.163 may be problematic. Regulation of dCK activity could also be Most of the NA inhibit DNA polymerase activity by compet- a target for modulation. It has been shown that dCK activity ing with dNTPs,164 thereby contributing to the inhibition of is stimulated after treatment with 2-CdA, as well as with flud- DNA synthesis.165 The degree of inhibition of DNA poly- arabine and ara-C in normal human lymphocytes and various merases is not the same for all the NA. For example, DNA leukemic cell lines.190,191 The increase of dCK activity polymerase α has been shown to be more sensitive to inhi- induced by NA was considered to result from post- bition by ara-CTP than DNA polymerase β.166 F-ara-ATP is translational modifications of dCK during inhibition of DNA not a potent inhibitor of DNA polymerases β and γ or DNA synthesis.192,193 primase.84,162 In vitro, overexpression of DNA polymerases The importance of the ratio of nucleoside analogues to nor- may be a significant factor in the development of drug resist- mal nucleosides has been recognized more recently. Reduc- ance.167 Tanaka et al168 found that DNA polymerase α sensi- ing the intracellular pools of normal nucleotides therefore tivity to ara-CTP and F-ara-ATP was lower in blast cells appears to be a therapeutic alternative to increasing the con- obtained from ALL patients than in cells obtained from AML centrations of cytotoxic NA triphosphates. In practice, given patients, and concluded that DNA polymerase α from ALL the feedback inhibitory effects of dNTPs on NA activation, blast cells has a decreased affinity for NA.168 these two goals may be attained simultaneously. Various bio- chemical modulation strategies, based on NA combinations, have been suggested to increase the concentration of NA Defective cell death pathways and p53 exonuclease triphosphate in leukemic cells. activity

DNA damage caused by NA induces expression of p53, lead- Inhibition of ribonucleotide reductase ing to the induction of pro-apototic molecules such as Bax and to down-regulation of anti-apoptotic proteins, such as Large amounts of dNTPs generated by RR are directed into Bcl-2.169–171 These events lead to induction of apoptosis.172,173 replicating DNA, a process that effectively excludes NA from Various investigators have shown that B-CLL and AML replicating DNA synthesis.194 Furthermore intracellular dNTP patients with mutations in the p53 gene displayed resistance pools inhibit dCK activity, thereby reducing NA activation. to fludarabine, 2-CdA or ara-C and/or poorer response and Thus, drugs capable of depleting or reducing normal dNTPs shorter remission and/or overall survival174–177 (Table 5). Con- by inhibiting RR might be expected to potentiate cytotoxicity versely, other authors failed to find any relationship between of different NA. Sequential incubation of K562 human leuke- p53 abnormalities and resistance to NA in these dis- mia cells with F-ara-A followed by ara-C enhanced ara-CTP eases.178,179 Other genes regulating apoptosis, such as Bcl-2, accumulation, thereby suggesting possible therapeutic syner- Bcl-X/L, Mcl-1 or Bag-1, have been suggested to influence gism between ara-C and fludarabine.86 Subsequent clinical sensitivity to NA, although the results remain contro- studies have demonstrated the effectiveness of this strat- versial.180–187 egy.195,196 Lymphocytes isolated from patients with CLL and Alterations in p53 function have been related to in vitro incubated with ara-C after in vitro F-ara-A incubation or a

Leukemia Nucleoside analogues: mechanisms of drug resistance and reversal strategies CM Galmarini et al 882 Table 5 Involvement of p53 protein in drug resistance to NA

Author Disease Drug Tech Results Ref.

El-Rouby CLL Flud SSCP DR in patients with mutations 174 Wattel CLL/AML Flud/ara-C SSCP DR in patients with mutations 175 Johnston CLL Flud/2-CdA SSCP, IB DR in patients with mutations 176 Lens CLL Flud SB, ICC No correlation with DR 178 Dohner CLL Flud FISH, SSCP DR in patients with mutations 177 Kasimir AML ara-C SSCP No correlation with DR 179

Tech, technique; Flud, fludarabine; DR, drug resistance; IB, immunoblot; SB, Southern blot; ICC, immunocytochemistry.

therapeutic infusion of fludarabine (25 or 30 mg/m2) displayed higher concentrations of ara-CTP, greater ratios of analogue an increase in ara-CTP accumulation.197 The same phenom- triphosphates to normal dNTPs and tandem incorporation of enon was observed in leukemic lymphocytes and AML 2-Cl-dATP and ara-CTP in growing DNA strands. blasts.198,199 This increase was caused by a higher rate of Although potentially interesting, the usefulness of NA com- anabolism rather than by a slower rate of catabolism of ara- binations in the clinic remains to be determined. In particular CTP. More recently, Seymour et al200 have shown that F-ara- the toxicity to normal cells due to the increase of NA accumu- ATP concentrations (30–50 µM, achieved with usual clinical lation appears to be significant, casting doubt on an improve- doses of fludarabine), enhances both intracellular ara-CTP ment of the therapeutic index in comparison to single agent accumulation and incorporation into cellular DNA at concen- NA therapy. A potential consequence related to NA modu- trations of ara-C which are clinically achievable. Such drug lation therapy may be the increased risk of secondary malig- combinations have induced responses in untreated and nancies in a group of patients whose disease already places refractory patients with acute leukemias.201,202 them at a great risk of second , and dose-related effects This modulatory effect of fludarabine can be extended to like severe neurotoxicity.210,211 Apparently, the frequency and other NA such as 9-β-D-arabinofuranosylguanine (ara-G).203 severity of neurotoxicity may be greater with the combination In vitro studies using human leukemia cell lines and primary therapy than with either drug used alone. leukemia cells obtained from patients established that the effectiveness of ara-G is due to intracellular accumulation of ara-GTP. The rate-limiting step of ara-GTP accumulation is the Modulation of dCK and 59-NU activitity with initial phosphorylation performed both by dGK and dCK. In bryostatin vivo, Gandhi and coworkers demonstrated in a phase I clini- cal trial that patients who responded to therapy with the Bryostatin, a macrocyclic lactone and protein kinase C acti- water-soluble ara-G prodrug GW506U achieved a signifi- vator, has been shown to increase the ratio of dCK/59-NU cantly greater concentration of ara-GTP in their circulating activity and thereby increase sensitivity to fludarabine and cla- cells than patients who did not respond.204,205 In this setting, dribine in CLL cells, both in vitro and in vivo.212,213 Phase I the combination with fludarabine could increase the intra- studies of single agent bryostatin have been completed and cellular accumulation of ara-GTP in target cells and improve this compound is now in clinical trials in combination with response rates in patients.205 NA.214 Other inhibitors of RR can be used for the modulation of ara-C metabolism. Iwasaki and coworkers194 showed that inhibition of RR with gemcitabine and the consequent Use of hematopoietic growth factors decrease in cellular dCTP pools favored ara-C incorporation into replicating DNA. Colly et al206 and Bhalla et al207 tested Different in vitro studies have shown that pretreatment of AML the ability of the RR inhibitor hydroxyurea to enhance ara-C blasts by growth factors (GF) enhances ara-C mediated cyto- metabolism and cytotoxicity in drug-sensitive and -resistant toxicity against leukemic cells, possibly through their prolifer- cell lines. This combination significantly reduced dCTP pools, ation-inducing effects.215,216 GF may also enhance ara-C cyto- increased DNA incorporation of ara-C and enhanced drug toxicity by increasing hENT1 expression. High cellular cytotoxicity, even in dCK deficient ara-C-resistant cell lines. proliferation rates are associated with high levels of hENT1- Similarly, as 2-CdA inhibits the reduction of CDP and ADP, mediated activity that increase the nucleoside drug a decrease in the dCTP and dATP pools is expected when uptake.217,218 Wiley et al219 determined that in vitro treatment cells are exposed to this compound. The decrease of these of isolated human leukemic cells with GM-CSF, resulted in an pools would increase the ratios of 2-CdATP to dATP and ara- increase in hENT1 expression. Reuter et al220 established that CTP to dCTP. In vitro incubation of AML blasts with 2-CdA blasts from AML patients treated with GM-CSF displayed an followed by ara-C produced a higher rate of ara-CTP accumu- enhanced drug uptake after low or SDAC doses. In this study, lation than did ara-C alone.208 Gandhi and coworkers209 stud- GM-CSF also increased intracellular ara-CTP/dCTP pool ratios ied 17 patients with refractory AML receiving combination and enhanced ara-C incorporation into DNA by activation of with ara-C and 2-CdA. Seven of nine patients DNA polymerases. In vivo, GM-CSF priming of leukemic studied during therapy had an increased rate of ara-CTP for- blasts prior to induction therapy translates into a higher anti- mation and peak ara-CTP concentrations in leukemic blasts. leukemic activity as indicated by a prolonged survival of This modulation strategy increased the effective dose intensity treated mice with advanced AML and a high rate of complete of the active metabolite of ara-C by 40% in the tumor cells. remission in patients with AML.221,222 However, various ran- The co-administration of 2-CdA and ara-C resulted in domized clinical trials failed to demonstrate the effectiveness maximum inhibition of DNA synthesis, accumulation of of the GF-priming strategy and concluded that administration

Leukemia Nucleoside analogues: mechanisms of drug resistance and reversal strategies CM Galmarini et al 883 of GF during and after induction chemotherapy does not improve the clinical outcome of ara-C-treated AML patients.223,224

DNA repair induction

Another way of enhancing incorporation of NA into DNA consists of using the DNA repair pathway after combined exposure of tumor cells to NA and DNA damaging agents.225 The recognition and repair of the damaged strand caused by the DNA damaging agent allow resynthesis using the opposite strand as a template. This creates an opportunity for the inser- Figure 4 Chemical structure of troxacitabine. tion of the NA instead of its normal counterpart into the DNA repair patch. The incorporated NA is relatively resistant to repair excision and causes irreversible damage recognized by 4) is a stereochemically synthetic that the cell. Alternatively inhibition of DNA repair by NA may has potent antitumor activity in preclinical models.235–237 It is increase the accumulation and slow the removal of DNA phosphorylated in vitro by deoxycytidine kinase (dCK) with a lesions induced by DNA damaging agents, thereby potentiat- Km similar to its natural substrate, dCyd, but unlike gemcitab- ing the cytotoxicity of these latter compounds. In vitro, combi- ine and ara-C, it is not a substrate of CDD. Troxacitabine tri- nation of gemcitabine and fludarabine with gener- phosphate can be incorporated into cellular DNA,235 which ated synergistic cytotoxicity.225 Van Den Neste et al226 causes rapid chain termination. However, neither ribonucleo- recently reported the in vitro synergistic cytotoxicity of 2-CdA tide reductase nor mitochondrial DNA synthesis are inhibited and in B-CLL cells isolated from patients by troxacitabine in cell culture. Because of its broad preclini- pretreated with alkylating agents. cal spectrum with cytotoxicity both against leukemic and epi- In vivo, the combination of a DNA damaging agent and a thelial malignancies, and promising phase I activity demon- NA may prove to be particularly useful to destroy cancer cells strated in acute leukemia,238 phase II testing is now being without significant DNA replication activity such as CLL and carried out in several diseases. indolent . The combination of fludarabine with and prednisone is clinically active against CLL.227 Fludarabine and ara-C with or without cisplatin, have been 2-amino-9-β-D-arabinosyl-6-methoxy-9H- administered to patients with CLL with cytoreductive activity.228 Results of clinical studies using In 1983, Cohen et al239reported that the deoxyguanosine (which induces protein-associated DNA strand breaks), fluda- derivative ara-G (9-β-D-arabinofuranosylguanine) (Figure 5) rabine, and dexamethasone demonstrated a high response was resistant to cleavage by purine nucleoside rate, especially in follicular lymphomas, with a number of (PNP) and was toxic to T-lymphocytes. The devolopment of patients achieving complete remission.229,230 A combination this drug was limited by water insolubility. However, 2-amino- of mitoxantrone and ara-C for the treatment of acute leukemia 9-β-D-arabinosyl-6-methoxy-9H-guanine (GW506U78) is 10- has been encouraging.231,232 Moreover, clinical trials evaluat- fold more soluble, and is rapidly converted to ara-G by ing NA in combination with DNA damaging agents have plasma adenosine deaminase activity. shown effectiveness against solid tumors with low growth In phase I clinical testing, this agent has shown particular fractions.233,234 promise in the therapy of T cell malignancies,240 where 54% of patients achieved partial or complete remissions after one to two cycles of drug treatment. Of particular interest, neuro- New nucleoside analogues toxicity was dose limiting, with little clinical myelosuppres- sion.241 This raises the possibility of successful combination The broadening of the spectrum of nucleoside analog activity therapy with other active agents, including other hematolog- to solid tumors has triggered renewed interest in this class of ically active NA. Although no formal studies have been perfor- antimetabolites. Many new nucleoside drugs have been syn- med to demonstrate mechanisms of ara-G resistance, accumu- thesized and studied over the past decade, with several that lation of ara-GTP in leukemic blasts correlated with the are in early phase clinical trials, and others that have become standardly prescribed agents (gemcitabine and ). We will review the most promising of these agents: troxacitab- ine, and the GW506U78, an ara-G prodrug. We have excluded from this discussion 5-FU prodrugs such as capecita- bine since their mechanisms of action and resistance mech- anisms differ substantially from those of other anticancer nucleosides.

Troxacitabine

Troxacitabine (BCH-4556; (−)-2-(S)-hydroxymethyl-4-(s)- (cytosin-19-yl)-1,3-dioxolane; β-L-dioxalane cytidine) (Figure Figure 5 Chemical structure of ara-G.

Leukemia Nucleoside analogues: mechanisms of drug resistance and reversal strategies CM Galmarini et al 884 cytotoxic activity against malignant cells.241 This suggests that amic and pharmacokinetic properties and therapeutic potential in the early steps of uptake and metabolism of ara-G may be haematological malignancies. Drugs 1993; 46: 872–894. major determinants of cellular drug sensitivity, as for other 6 Robertson LE, Huh YO, Butler JJ, Pugh WC, Hirsch-Ginsberg C, Stass S, Kantarjian H, Keating MJ. Response assessment in chronic NA. lymphocytic leukemia after fludarabine plus prednisone: clinical, pathologic, immunophenotypic, and molecular analysis. Blood 1992; 80: 29–36. Conclusions 7 Ross SR, McTavish D, Faulds D. Fludarabine. A review of its phar- macological properties and therapeutic potential in malignancy. During the past decade there has been dramatic progress in Drugs 1993; 45: 737–759. 8 Juliusson G, Liliemark J. Long-term survival following cladribine the understanding of the individual mechanisms of resistance (2-chlorodeoxyadenosine) therapy in previously treated patients to nucleoside analogues, including the role of cellular drug with chronic lymphocytic leukemia. Ann Oncol 1996; 7: 373– uptake, drug metabolism, interaction with cellar targets, and 379. the resulting apoptotic cascade. However, most studies have 9 Sorensen JM, Vena DA, Fallavollita A, Chun HG, Cheson BD. focused on a single mechanism of resistance, thereby failing Treatment of refractory chronic lymphocytic leukemia with fluda- to provide the relative importance of the different known rabine phosphate via the group C protocol mechanism of the mechanisms of resistance. It is clear that malignancies are het- National Cancer Institute: five-year follow-up report. J Clin Oncol 1997; 15: 458–465. erogeneous in terms of mechanisms of resistance, and that the 10 Boleti H, Coe IR, Baldwin SA, Young JD, Cass CE. Molecular clinical benefit of resistance-reversal strategies will only be identification of the equilibrative NBMPR-sensitive (es) nucleoside demonstrated when targeted to the appropriate subgroup of transporter and demonstration of an equilibrative NBMPR-insensi- patients. tive (ei) transport activity in human erythroleukemia (K562) cells. The ultimate goal of research into anticancer therapy Neuropharmacology 1997; 36: 1167–1179. remains improving patient survival. However, in diseases such 11 Baldwin SA, Mackey JR, Cass CE, Young JD. Nucleoside trans- porters: molecular and implications for therapeutic devel- as CLL or indolent in which several treatments are opment. Mol Med Today 1999; 5: 216–224. effective but not curative, a realistic goal would be the identi- 12 Crawford CR, Patel DH, Naeve C, Belt JA. Cloning of the human fication of tumor cell traits which could allow tailored or tar- equilibrative, nitrobenzylmercaptopurine riboside (NBMPR)- getted therapy. In such patients, a predictive assay to deter- insensitive ei by functional expression in a mine a phenotype of probable resistance to certain NA would transport-deficient cell line. J Biol Chem 1998; 273: 5288–5293. be of great value in the choice of optimal therapy, and the 13 Griffiths M, Beaumont N, Yao SY, Sundaram M, Boumah CE, Dav- avoidance of ineffective but toxic treatments. ies A, Kwong FY, Coe I, Cass CE, Young JD, Baldwin SA. Cloning of a human nucleoside transporter implicated in the cellular In diseases such as AML in which curative drug combi- uptake of adenosine and chemotherapeutic drugs. Nat Med 1997; nations exist, the objective is to optimize the efficacy of cur- 3: 89–93. rently available NA and to develop newer, more effective ana- 14 Griffiths M, Yao SY, Abidi F, Phillips SE, Cass CE, Young JD, Bald- logues that are less susceptible to the resistance mechanisms win SA. Molecular cloning and characterization of a nitrobenzyl- described above. Efforts to increase intracellular levels and thioinosine-insensitive (ei) equilibrative nucleoside transporter DNA incorporation of phosphorylated NA are very promising. from human placenta. Biochem J 1997; 328: 739–743. 15 Crawford CR, Ng CY, Noel LD, Belt JA. Nucleoside transport in NA therapy combined with agents modulating apototic L1210 murine leukemia cells. Evidence for three transporters. J responses are expected to provide additional benefit. In the Biol Chem 1990; 265: 9732–9736. same way that combination chemotherapy has provided cura- 16 Belt JA, Marina NM, Phelps DA, Crawford CR. Nucleoside trans- tive treatment of certain cancers, a multifactorial approach of port in normal and neoplastic cells. Adv Enzyme Regul 1993; 33: drug resistance should allow significant progress in the treat- 235–252. ment of currently chemoresistant disease. 17 Graham KA, Leithoff J, Coe IR, Mowles D, Mackey JR, Young JD, Cass CE. Differential transport of cytosine-containing nucleosides by recombinant human concentrative nucleoside transporter pro- tein hCNT1. Nucleosides Nucleotides Nucleic Acids 2000; 19: Acknowledgements 415–434. 18 Schaner ME, Wang J, Zhang L, Su SF, Gerstin KM, Giacomini KM. Carlos Galmarini is a recipient of the ‘Michel Clavel’ grant. Functional characterization of a human purine-selective, Na+- The authors acknowledge the support of the Ligue Contre le dependent nucleoside transporter (hSPNT1) in a mammalian Cancer du Rhoˆne and the Alberta Cancer Foundation and expression system. J Pharmacol Exp Ther 1999; 289: 1487–1491. 19 Kufe DW, Weichselbaum R, Egan EM, Dahlberg W, Fram RJ. National Cancer Institute of Canada. Lethal effects of 1-beta-D-arabinofuranosylcytosine incorporation into deoxyribonucleic acid during repair. Mol Pharma- col 1984; 25: 322–326. References 20 Sandoval A, Consoli U, Plunkett W. Fludarabine-mediated inhi- bition of nucleotide excision repair induces apoptosis in quiescent 1 Cheson BD. New antimetabolites in the treatment of human malig- human lymphocytes. Clin Cancer Res 1996; 2: 1731–1741. nancies. Semin Oncol 1992; 19: 695–706. 21 Nicander B, Reichard P. Relations between synthesis of deoxyri- 2 Rustum YM, Raymakers RA. 1-Beta-arabinofuranosylcytosine in bonucleotides and DNA replication in 3T6 fibroblasts. J Biol Chem therapy of leukemia: preclinical and clinical overview. Pharmacol 1985; 260: 5376–5381. Ther 1992; 56: 307–321. 22 Xu YZ, Huang P, Plunkett W. Functional compartmentation of 3 Burris HA III, Moore MJ, Andersen J, Green MR, Rothenberg ML, dCTP pools. Preferential utilization of salvaged deoxycytidine for Modiano MR, Cripps MC, Portenoy RK, Storniolo AM, Tarassoff DNA repair in human lymphoblasts. J Biol Chem 1995; 270: P, Nelson R, Dorr FA, Stephens CD, Von Hoff DD. Improvements 631–637. in survival and clinical benefit with gemcitabine as first-line ther- 23 Shrecker AW, Urshel MJ. Metabolism of 1-beta-D-arabinofurano- apy for patients with advanced pancreas cancer: a randomized sylcytosine in leukemia L1210: studies with intact cells. Cancer trial. J Clin Oncol 1997; 15: 2403–2413. Res 1968; 28: 793–801. 4 Kaye SB. Gemcitabine: current status of phase I and II trials 24 Plagemann PG, Marz R, Wohlhueter RM. Transport and metab- (editorial). J Clin Oncol 1994; 12: 1527–1531. olism of deoxycytidine and 1-beta-D-arabinofuranosylcytosine 5 Bryson HM, Sorkin EM. Cladribine. A review of its pharmacodyn- into cultured Novikoff rat hepatoma cells, relationship to phos-

Leukemia Nucleoside analogues: mechanisms of drug resistance and reversal strategies CM Galmarini et al 885 phorylation, and regulation of triphosphate synthesis. Cancer Res 45 Schirmer M, Stegmann AP, Geisen F, Konwalinka G. Lack of cross- 1978; 38: 978–989. resistance with gemcitabine and cytarabine in cladribine-resistant 25 Capizzi RL, Yang JL, Rathmell JP, White JC, Cheng E, Cheng YC, HL60 cells with elevated 59-nucleotidase activity. Exp Hematol Kute T. Dose-related pharmacologic effects of high-dose ara-C and 1998; 26: 1223–1228. its self-potentiation. Semin Oncol 1985; 12: 65–74. 46 Huang P, Chubb S, Hertel LW, Grindey GB, Plunkett W. Action 26 Wiley JS, Taupin J, Jamieson GP, Snook M, Sawyer WH, Finch of 29,29-difluorodeoxycytidine on DNA synthesis. Cancer Res LR. Cytosine arabinoside transport and metabolism in acute leuke- 1991; 51: 6110–6117. mias and T cell lymphoblastic lymphoma. J Clin Invest 1985; 75: 47 Huang P, Plunkett W. Induction of apoptosis by gemcitabine. 632–642. Semin Oncol 1995; 22: 19–25. 27 Weinstein HJ, Griffin TW, Feeney J, Cohen HJ, Propper RD, Sallan 48 Plunkett W, Huang P, Gandhi V. Preclinical characteristics of SE. of continuous intravenous and subcutane- gemcitabine. Anticancer Drugs 1995; 6 (Suppl. 6): 7–13. ous infusions of cytosine arabinoside. Blood 1982; 59: 1351– 49 Ruiz van Haperen VW, Veerman G, Vermorken JB, Peters GJ. 1353. 29,29-Difluoro-deoxycytidine (gemcitabine) incorporation into 28 Ho DH, Frei E. Clinical pharmacology of 1-beta-d-arabinofurano- RNA and DNA of tumour cell lines. Biochem Pharmacol 1993; syl cytosine. Clin Pharmacol Ther 1971; 12: 944–954. 46: 762–766. 29 Capizzi RL, Yang JL, Cheng E, Bjornsson T, Sahasrabudhe D, Tan 50 Cartei G, Sacco C, Sibau A, Pella N, Iop A, Tabaro G. Cisplatin RS, Cheng YC. Alteration of the pharmacokinetics of high-dose and gemcitabine in non-small-cell lung cancer. Ann Oncol 1999; ara-C by its metabolite, high ara-U in patients with acute leuke- 10: S57–S62. mia. J Clin Oncol 1983; 1: 763–771. 51 Hoffman MA, Janson D, Rose E, Rai KR. Treatment of hairy-cell 30 Plunkett W, Liliemark JO, Estey E, Keating MJ. Saturation of ara- leukemia with cladribine: response, toxicity, and long-term fol- CTP accumulation during high-dose ara-C therapy: pharmacologic low-up. J Clin Oncol 1997; 15: 1138–1142. rationale for intermediate-dose ara-C. Semin Oncol 1987; 14: 52 Jehn U, Bartl R, Dietzfelbinger H, Vehling-Kaiser U, Wolf-Hor- 159–166. nung B, Hill W, Heinemann V. Long-term outcome of hairy cell 31 Liliemark JO, Plunkett W, Dixon DO. Relationship of 1-beta-D- leukemia treated with 2-chlorodeoxyadenosine. Ann Hematol arabinofuranosylcytosine in plasma to 1-beta-D-arabinofuranosyl- 1999; 78: 139–144. 9 cytosine 5 -triphosphate levels in leukemic cells during treatment 53 Lauria F, Rondelli D, Zinzani PL, Bocchia M, Marotta G, Salvucci with high-dose 1-beta-D-arabinofuranosylcytosine. Cancer Res M, Raspadori D, Ventura MA, Birtolo S, Forconi F, Tura S. Long- 1985; 45: 5952–5957. lasting complete remission in patients with 32 Dumontet C, Fabianowska-Majewska K, Mantincic D, Callet Bau- treated with 2-CdA: a 5-year survey. Leukemia 1997; 11: 629– chu E, Tigaud I, Gandhi V, Lepoivre M, Peters GJ, Rolland MO, 632. Wyczechowska D, Fang X, Gazzo S, Voorn DA, Vanier-Viornery 54 King KM. Membrane transport of 29-chloro-29-deoxyadenosine A, Mackey JR. Common resistance mechanisms to deoxynucleo- and 2-chloro-29arabinofluoro-29-deoxyadenosine is required for side analogues in variants of the human erythroleukaemic line cytotoxicity. Proc Am Assoc Cancer Res 1994; 35: A3436. K562. Br J Haematol 1999; 106: 78–85. 55 Wang L, Karlsson A, Arner ES, Eriksson S. Substrate specificity of 33 Major PP, Egan EM, Beardsley GP, Minden MD, Kufe DW. Lethal- mitochondrial 29-deoxyguanosine kinase. Efficient phosphoryl- ity of human myeloblasts correlates with the incorporation of arab- ation of 2-chlorodeoxyadenosine. J Biol Chem 1993; 268: inofuranosylcytosine into DNA. Proc Natl Acad Sci USA 1981; 22847–22852. 78: 3235–3239. 56 Carson DA, Kaye J, Wasson DB. The potential importance of sol- 34 Major PP, Egan EM, Herrick DJ, Kufe DW. Effect of ARA-C incor- uble deoxynucleotidase activity in mediating deoxyadenosine tox- poration on deoxyribonucleic acid synthesis in cells. Biochem icity in human lymphoblasts. J Immunol 1981; 126: 348–352. Pharmacol 1982; 31: 2937–2940. 57 Seto S, Carrera CJ, Kubota M, Wasson DB, Carson DA. Mechanism 35 Kufe DW, Major PP, Egan EM, Beardsley GP. Correlation of cyto- of deoxyadenosine and 2-chlorodeoxyadenosine toxicity to non- toxicity with incorporation of ara-C into DNA. J Biol Chem 1980; dividing human lymphocytes. J Clin Invest 1985; 75: 377–383. 255: 8997–8900. 58 Carson DA, Wasson DB, Taetle R, Yu A. Specific toxicity of 2- 36 Gunji H, Kharbanda S, Kufe D. Induction of internucleosomal chlorodeoxyadenosine toward resting and proliferating human DNA fragmentation in human myeloid leukemia cells by 1-beta- lymphocytes. Blood 1983; 62: 737–743. D-arabinofuranosylcytosine. Cancer Res 1991; 51: 741–743. 59 Griffig J, Koob R, Blakley RL. Mechanisms of inhibition of DNA 37 Raza A, Gezer S, Anderson J, Lykins J, Bennett J, Browman G, synthesis by 2-chlorodeoxyadenosine in human lymphoblastic Goldberg J, Larson R, Vogler R, Preisler HD. Relationship of cells. Cancer Res 1989; 49: 6923–6928. [3H]Ara-C incorporation and response to therapy with high-dose 60 Hentosh P, Koob R, Blakley RL. Incorporation of 2-halogeno-29- Ara-C in AML patients: a Leukemia Intergroup study. Exp Hematol deoxyadenosine 5-triphosphates into DNA during replication by 1992; 20: 1194–1200. 38 Estey E, Plunkett W, Dixon D, Keating M, McCredie K, Freireich human polymerases alpha and beta. J Biol Chem 1990; 265: EJ. Variables predicting response to high dose cytosine arabinoside 4033–4040. 61 Lassota P, Kazimierczuk Z, Darzynkiewicz Z. Apoptotic death of therapy in patients with refractory acute leukemia. Leukemia 9 1987; 1: 580–583. lymphocytes upon treatment with 2-chloro-2 -deoxyadenosine (2- 39 Kaye SB. New antimetabolites in cancer chemotherapy and their CdA). Arch Immunol Ther Exp 1994; 42: 17–23. clinical impact. Br J Cancer 1998; 78: 1–7. 62 Parker WB, Bapat AR, Shen JX, Townsend AJ, Cheng YC. Interac- 40 Possinger K. Gemcitabine in advanced breast cancer. Anticancer tion of 2-halogenated dATP analogs (F, Cl, and Br) with human Drugs 1995; 6 (Suppl. 6): 55–59. DNA polymerases, DNA primase, and ribonucleotide reductase. 41 Baker CH, Banzon J, Bollinger JM, Stubbe J, Samano V, Robins Mol Pharmacol 1988; 34: 485–491. MJ, Lippert B, Jarvi E, Resvick R. 29-Deoxy-29-methylenecytidine 63 Plunkett W, Huang P, Gandhi V. Metabolism and action of fluda- and 29-deoxy-29,29-difluorocytidine 59-diphosphates: potent rabine phosphate. Semin Oncol 1990; 17: 3–17. mechanism-based inhibitors of ribonucleotide reductase. J Med 64 Fabianowska-Majewska K, Wasiak TJ, Warzocha K, Marlewski M, Chem 1991; 34: 1879–1884. Fairbanks L, Smolenski RT, Duley J, Simmonds A. A new mech- 42 Abbruzzese JL, Grunewald R, Weeks EA, Gravel D, Adams T, anism of toxicity of 2-chlorodeoxyadenosine (2CdA). Adv Exp Nowak B, Mineishi S, Tarassoff P, Satterlee W, Raber MN et al. Med Biol 1994; 370: 125–128. A phase I clinical, plasma, and cellular pharmacology study of 65 Abeles RH, Tashjian AH Jr, Fish S. The mechanism of inactivation gemcitabine. J Clin Oncol 1991; 9: 491–498. of S-adenosylhomocysteinase by 29-deoxyadenosine. Biochem 43 Plunkett W, Huang P, Xu YZ, Heinemann V, Grunewald R, Biophys Res Commun 1980; 95: 612–617. Gandhi V. Gemcitabine: metabolism, mechanisms of action, and 66 Wolos JA, Frondorf KA, Davis GF, Jarvi ET, McCarthy JR, Bowlin self-potentiation. Semin Oncol 1995; 22: 3–10. TL. Selective inhibition of T cell activation by an inhibitor of S- 44 Neff T, Blau CA. Forced expression of cytidine deaminase confers adenosyl- L-homocysteine hydrolase. J Immunol 1993; 150: resistance to cytosine arabinoside and gemcitabine. Exp Hematol 3264–3273. 1996; 24: 1340–1346. 67 Pettitt AR, Clarke AR, Cawley JC, Griffiths SD. Purine analogues

Leukemia Nucleoside analogues: mechanisms of drug resistance and reversal strategies CM Galmarini et al 886 kill resting lymphocytes by p53-dependent and -independent B and normal T cells in chronic lymphocytic leukemia. Blood mechanisms. Br J Haematol 1999; 105: 986–988. 1998; 91: 1742–1748. 68 Pettitt AR, Sherrington PD, Cawley JC. The effect of p53 dysfunc- 88 Pettitt AR, Sherrington PD, Cawley JC. Role of poly(ADP- tion on cytotoxicity in chronic lymphocytic leu- ribosyl)ation in the killing of chronic lymphocytic leukemia cells kaemia. Br J Haematol 1999; 106: 1049–1051. by purine analogues. Cancer Res 2000; 60: 4187–4193. 69 Genini D, Adachi S, Chao Q, Rose DW, Carrera CJ, Cottam HB, 89 Huang P, Plunkett W. Action of 9-beta-D-arabinofuranosyl-2- Carson DA, Leoni LM. Deoxyadenosine analogs induce pro- fluoroadenine on RNA metabolism. Mol Pharmacol 1991; 39: grammed cell death in chronic lymphocytic leukemia cells by 449–455. damaging the DNA and by directly affecting the mitochondria. 90 Huang P, Sandoval A, Van Den Neste E, Keating MJ, Plunkett Blood 2000; 96: 3537–3543. W. Inhibition of RNA transcription: a biochemical mechanism of 70 Hentosh P, Tibudan M. In vitro transcription of DNA containing 2- action against chronic lymphocytic leukemia cells by fludarab- chloro-29-deoxyadenosine monophosphate. Mol Pharmacol 1995; ine. Leukemia 2000; 14: 1405–1413. 48: 897–904. 91 Gottardi D, De Leo AM, Alfarano A, Stacchini A, Circosta P, Gre- 71 Genini D, Budihardjo I, Plunkett W, Wang X, Carrera CJ, Cottam goretti MG, Bergui L, Aragno M, Caligaris-Cappio F. Fludarabine HB, Carson DA, Leoni LM. Nucleotide requirements for the in ability to down-regulate Bcl-2 gene product in CD5+ leukaemic vitro activation of the apoptosis protein-activating factor-1- B cells: in vitro/in vivo correlations. Br J Haematol 1997; 99: mediated caspase pathway. J Biol Chem 2000; 275: 29–34. 147–157. 72 Leoni LM, Chao Q, Cottam HB, Genini D, Rosenbach M, Carrera 92 Sjoberg AH, Wang L, Eriksson S. Substrate specificity of human CJ, Budihardjo I, Wang X, Carson DA. Induction of an apoptotic recombinant mitochondrial deoxyguanosine kinase with cyto- program in cell-free extracts by 2-chloro-29-deoxyadenosine 59- static and antiviral purine and pyrimidine analogs. Mol Pharma- triphosphate and cytochrome c. Proc Natl Acad Sci USA 1998; col 1998; 53: 270–273. 95: 9567–9571. 93 Galmarini CM, Thomas X, Calvo F, Rousselot P, Dumontet C. 73 Hentosh P, Tibudan M. 2-Chloro-29-deoxyadenosine, an antileu- Mechanisms of resistance to cytarabine in relapsing acute kemic drug, has an early effect on cellular mitochondrial function. myeloid leukemia (AML) patients. Blood 1999; 94: 1249A. Mol Pharmacol 1997; 51: 613–619. 94 White JC, Rathmell JP, Capizzi RL. Membrane transport influ- 74 Enriquez JA, Fernandez-Silva P, Perez-Martos A, Lopez-Perez MJ, ences the rate of accumulation of cytosine arabinoside in human Montoya J. The synthesis of mRNA in isolated mitochondria can leukemia cells. J Clin Invest 1987; 79: 380–387. be maintained for several hours and is inhibited by high levels of 95 Mackey JR, Mani RS, Selner M, Mowles D, Young JD, Belt JA, ATP. Eur J Biochem 1996; 237: 601–610. Crawford CR, Cass CE. Functional nucleoside transporters are 75 de Murcia G, Menissier de Murcia J. Poly(ADP-ribose) poly- required for gemcitabine influx and manifestation of toxicity in merase: a molecular nick-sensor (published erratum appears in cancer cell lines. Cancer Res 1998; 58: 4349–4357. Trends Biochem Sci 1994; 19: 250). Trends Biochem Sci 1994; 96 Gati WP, Paterson AR, Larratt LM, Turner AR, Belch AR. Sensi- 19: 172–176. tivity of acute leukemia cells to cytarabine is a correlate of cellu- 76 D’Amours D, Desnoyers S, D’Silva I, Poirier GG. Poly(ADP- lar es nucleoside transporter site content measured by flow cyto- ribosyl)ation reactions in the regulation of nuclear functions. metry with SAENTA-fluorescein. Blood 1997; 90: 346–353. Biochem J 1999; 342: 249–268. 97 Gati WP, Paterson AR, Belch AR, Chlumecky V, Larratt LM, Mant 77 Althaus FR, Richter C. ADP-ribosylation of proteins. Enzymology MJ, Turner AR. Es nucleoside transporter content of acute leuke- and biological significance. Mol Biol Biochem Biophys 1987; 37: mia cells: role in cell sensitivity to cytarabine (araC). Leuk Lym- 1–237. phoma 1998; 32: 45–54. 78 Carson DA, Carrera CJ, Wasson DB, Yamanaka H. Programmed 98 Wiley JS, Jones SP, Sawyer WH, Paterson AR. Cytosine arabino- cell death and adenine deoxynucleotide metabolism in human side influx and nucleoside transport sites in acute leukemia. J lymphocytes. Adv Enzyme Regul 1988; 27: 395–404. Clin Invest 1982; 69: 479–489. 79 Zamzami N, Marchetti P, Castedo M, Decaudin D, Macho A, 99 Coleman CN, Stoller RG, Drake JC, Chabner BA. Deoxycytidine Hirsch T, Susin SA, Petit PX, Mignotte B, Kroemer G. Sequential kinase: properties of the enzyme from human leukemic granulo- reduction of mitochondrial transmembrane potential and gener- cytes. Blood 1975; 46: 791–803. ation of reactive species in early programmed cell death. 100 Carson DA, Wasson DB, Kaye J, Ullman B, Martin DW Jr, Robins J Exp Med 1995; 182: 367–377. RK, Montgomery JA. Deoxycytidine kinase-mediated toxicity of 80 Brockman RW, Schabel FM Jr, Montgomery JA. Biologic activity deoxyadenosine analogs toward malignant human lymphoblasts of 9-beta-D-arabinofuranosyl-2-fluoroadenine, a metabolically in vitro and toward murine L1210 leukemia in vivo. Proc Natl stable analog of 9-beta-D-arabinofuranosyladenine. Biochem Acad Sci USA 1980; 77: 6865–6869. Pharmacol 1977; 26: 2193–2196. 101 Heinemann V, Hertel LW, Grindey GB, Plunkett W. Comparison 81 Mackey JR, Baldwin SA, Young JD, Cass CE. The role of nucleo- of the cellular pharmacokinetics and toxicity of 29,29-difluoro- side transport in anticancer drug resistance. Drug Resistance deoxycytidine and 1-beta-D-arabinofuranosylcytosine. Cancer Updates 1998; 1: 310–324. Res 1988; 48: 4024–4031. 82 Plunkett W, Gandhi V, Huang P, Robertson LE, Yang LY, Gregoire 102 Wan CW, Mak TW. Deoxycytidine kinase and cytosine nucleo- V, Estey E, Keating MJ. Fludarabine: pharmacokinetics, mech- side deaminase activities in synchronized cultures of normal rat anisms of action, and rationales for combination therapies. Semin kidney cells. Cancer Res 1978; 38: 2768–2772. Oncol 1993; 20: 2–12. 103 Arner ES, Flygar M, Bohman C, Wallstrom B, Eriksson S. Deoxy- 83 Plunkett W, Saunders PP. Metabolism and action of purine cytidine kinase is constitutively expressed in human lympho- nucleoside analogs. Pharmacol Ther 1991; 49: 239–268. cytes: consequences for compartmentation effects, unscheduled 84 Tseng WC, Derse D, Cheng YC, Brockman RW, Bennett LL Jr. In DNA synthesis, and viral replication in resting cells. Exp Cell Res vitro biological activity of 9-beta-D-arabinofuranosyl-2-fluoroad- 1988; 178: 335–342. enine and the biochemical actions of its triphosphate on DNA 104 Terai C, Wasson DB, Carrera CJ, Carson DA. Dependence of cell polymerases and ribonucleotide reductase from HeLa cells. Mol survival on DNA repair in human mononuclear phagocytes. J Pharmacol 1982; 21: 474–477. Immunol 1991; 147: 4302–4306. 85 Huang P, Chubb S, Plunkett W. Termination of DNA synthesis 105 Hengstschlager M, Denk C, Wawra E. Cell cycle regulation of by 9-beta-D-arabinofuranosyl-2-fluoroadenine. A mechanism for deoxycytidine kinase. Evidence for post-transcriptional control. cytotoxicity. J Biol Chem 1990; 265: 16617–16625. FEBS Lett 1993; 321: 237–240. 86 Gandhi V, Plunkett W. Modulation of arabinosylnucleoside 106 Verhoef V, Sarup J, Fridland A. Identification of the mechanism metabolism by arabinosylnucleotides in human leukemia cells. of activation of 9-beta-D-arabinofuranosyladenine in human Cancer Res 1988; 48: 329–334. lymphoid cells using mutants deficient in nucleoside kinases. 87 Consoli U, El-Tounsi I, Sandoval A, Snell V, Kleine HD, Brown W, Cancer Res 1981; 41: 4478–4483. Robinson JR, DiRaimondo F, Plunkett W, Andreeff M. Differential 107 Bhalla K, Nayak R, Grant S. Isolation and characterization of a induction of apoptosis by fludarabine monophosphate in leukemic deoxycytidine kinase-deficient human promyelocytic leukemic

Leukemia Nucleoside analogues: mechanisms of drug resistance and reversal strategies CM Galmarini et al 887 cell line highly resistant to 1-beta-D-arabinofuranosylcytosine. kinase in patients with resistant . Blood Cancer Res 1984; 44: 5029–5037. 2000; 96: 1517–1524. 108 Stegmann AP, Honders MW, Kester MG, Landegent JE, Willemze 126 Chiba P, Tihan T, Szekeres T, Salamon J, Kraupp M, Eher R, R. Role of deoxycytidine kinase in an in vitro model for AraC- Koller U, Knapp W. Concordant changes of pyrimidine metab- and DAC-resistance: substrate-enzyme interactions with deoxy- olism in blasts of two cases of acute myeloid leukemia after cytidine, 1-beta-D-arabinofuranosylcytosine and 5-aza-29-deoxy- repeated treatment with ara-C in vivo. Leukemia 1990; 4: cytidine. Leukemia 1993; 7: 1005–1011. 761–765. 109 Dumontet C, Fabianowska-Majewska K, Mantincic D, Callet 127 Chottiner EG, Shewach DS, Datta NS, Ashcraft E, Gribbin D, Bauchu E, Tigaud I, Gandhi V, Lepoivre M, Peters GJ, Rolland Ginsburg D, Fox IH, Mitchell BS. Cloning and expression of MO, Wyczechowska D, Fang X, Gazzo S, Voorn DA, Vanier- human deoxycytidine kinase cDNA. Proc Natl Acad Sci USA Viornery A, MacKey J. Common resistance mechanisms to 1991; 88: 1531–1535. deoxynucleoside analogues in variants of the human erythroleu- 128 Nyce J, Liu L, Jones PA. Variable effects of DNA-synthesis inhibi- kaemic line K562. Br J Haematol 1999; 106: 78–85. tors upon DNA methylation in mammalian cells. Nucleic Acids 110 Stegmann AP, Honders WH, Willemze R, Ruiz van Haperen VW, Res 1986; 14: 4353–4367. Landegent JE. Transfection of wild-type deoxycytidine kinase 129 Antonsson BE, Avramis VI, Nyce J, Holcenberg JS. Effect of 5- (dck) cDNA into an AraC- and DAC-resistant rat leukemic cell azacytidine and congeners on DNA methylation and expression line of clonal origin fully restores drug sensitivity. Blood 1995; of deoxycytidine kinase in the human lymphoid cell lines 85: 1188–1194. CCRF/CEM/0 and CCRF/CEM/dCk-1. Cancer Res 1987; 47: 111 Hapke DM, Stegmann AP, Mitchell BS. Retroviral transfer of 3672–3678. deoxycytidine kinase into tumor cell lines enhances nucleoside 130 Harris AL, Grahame-Smith DG. Cytosine arabinoside triphosph- toxicity. Cancer Res 1996; 56: 2343–2347. ate production in human leukaemic myeloblasts: interactions 112 Orr RM, Talbot DC, Aherne WG, Fisher TC, Serafinowski P, Har- with deoxycytidine. Cancer Chemother Pharmacol 1981; 5: 9 rap KR. 2 -Deoxycytidine kinase deficiency is a major determi- 185–192. 9 nant of 2-chloro-2 -deoxyadenosine resistance in lymphoid cell 131 Arner ES, Eriksson S. Mammalian kinases. lines. Clin Cancer Res 1995; 1: 391–398. Pharmacol Ther 1995; 67: 155–186. 113 Ho DH. Distribution of kinase and deaminase of 1-beta-D-arabin- 132 Petrakis TG, Ktistaki E, Wang L, Eriksson S, Talianidis I. Cloning ofuranosylcytosine in tissues of man and mouse. Cancer Res and characterization of mouse deoxyguanosine kinase. Evidence 1973; 33: 2816–2820. for a cytoplasmic isoform. J Biol Chem 1999; 274: 24726–24730. 114 Spasokoukotskaja T, Arner ES, Brosjo O, Gunven P, Juliusson G, 133 Johansson M, Karlsson A. Cloning and expression of human Liliemark J, Eriksson S. Expression of deoxycytidine kinase and deoxyguanosine kinase cDNA. Proc Natl Acad Sci USA 1996; phosphorylation of 2-chlorodeoxyadenosine in human normal 93: 7258–7262. and tumour cells and tissues. Eur J Cancer 1995; 2: 202–208. 134 Johansson M, Karlsson A. Cloning of the cDNA and chromosome 115 Kakihara T, Fukuda T, Tanaka A, Emura I, Kishi K, Asami K, Uchi- localization of the gene for human thymidine kinase 2. J Biol yama M. Expression of deoxycytidine kinase (dCK) gene in leu- Chem 1997; 272: 8454–8458. kemic cells in childhood: decreased expression of dCK gene in 135 Wang L, Hellman U, Eriksson S. Cloning and expression of relapsed leukemia. Leuk Lymphoma 1998; 31: 405–409. human mitochondrial deoxyguanosine kinase cDNA. FEBS Lett 116 Stammler G, Zintl F, Sauerbrey A, Volm M. Deoxycytidine kinase 1996; 390: 39–43. mRNA expression in childhood acute lymphoblastic leukemia. 136 Wang L, Munch-Petersen B, Herrstrom Sjoberg A, Hellman U, Anticancer Drugs 1997; 8: 517–521. Bergman T, Jornvall H, Eriksson S. Human thymidine kinase 2: 117 Tattersall MH, Ganeshaguru K, Hoffbrand AV. Mechanisms of molecular cloning and characterisation of the enzyme activity resistance of human acute leukaemia cells to cytosine arabino- with antiviral and cytostatic nucleoside substrates. FEBS Lett side. Br J Haematol 1974; 27: 39–46. 1999; 443: 170–174. 118 Colly LP, Peters WG, Richel D, Arentsen-Honders MW, Starren- 137 Resta R, Yamashita Y, Thompson LF. Ecto-enzyme and signaling burg CW, Willemze R. Deoxycytidine kinase and deoxycytidine functions of lymphocyte CD73. Immunol Rev 1998; 161: 95– deaminase values correspond closely to clinical response to cyto- 109. sine arabinoside remission induction therapy in patients with 138 Pieters R, Thompson LF, Broekema GJ, Huismans DR, Peters GJ, acute myelogenous leukemia. Semin Oncol 1987; 14: 257–261. Pals ST, Horst E, Hahlen K, Veerman AJ. Expression of 59-nucleo- 119 Kawasaki H, Carrera CJ, Piro LD, Saven A, Kipps TJ, Carson DA. tidase (CD73) related to other differentiation antigens in leuke- Relationship of deoxycytidine kinase and cytoplasmic 59-nucleo- mias of B-cell lineage. Blood 1991; 78: 488–492. tidase to the chemotherapeutic efficacy of 2-chlorodeoxyadeno- 139 Spychala J, Mitchell BS. Regulation of low Km (ecto-) 59-nucleo- sine. Blood 1993; 81: 597–601. tidase gene expression in leukemic cells. Adv Exp Med Biol 120 Albertioni F, Lindemalm S, Reichelova V, Pettersson B, Eriksson S, Juliusson G, Liliemark J. Pharmacokinetics of cladribine in 1994; 370: 683–687. 9 9 140 Rampazzo C, Gazziola C, Ferraro P, Gallinaro L, Johansson M, plasma and its 5 -monophosphate and 5 -triphosphate in leu- 9 kemic cells of patients with chronic lymphocytic leukemia. Clin Reichard P, Bianchi V. Human high-Km 5 -nucleotidase effects Cancer Res 1998; 4: 653–658. of overexpression of the cloned cDNA in cultured human cells. 121 Leiby JM, Snider KM, Kraut EH, Metz EN, Malspeis L, Grever Eur J Biochem 1999; 261: 689–697. 141 Rampazzo C, Johansson M, Gallinaro L, Ferraro P, Hellman U, MR. Phase II trial of 9-beta-D-arabinofuranosyl-2-fluoroadenine 9 9 59-monophosphate in non-Hodgkin’s lymphoma: prospective Karlsson A, Reichard P, Bianchi V. Mammalian 5 (3 )-deoxyri- comparison of response with deoxycytidine kinase activity. Can- bonucleotidase, cDNA cloning, and overexpression of the cer Res 1987; 47: 2719–2722. enzyme in Escherichia coli and mammalian cells. J Biol Chem 122 Owens JK, Shewach DS, Ullman B, Mitchell BS. Resistance to 2000; 275: 5409–5415. 1-beta-D-arabinofuranosylcytosine in human T-lymphoblasts 142 Spychala J, Madrid-Marina V, Fox IH. Evidence for ‘low Km’ and 9 mediated by mutations within the deoxycytidine kinase gene. ‘high Km’ soluble 5 - in human tissues and rat liver. Cancer Res 1992; 52: 2389–2393. Adv Exp Med Biol 1989; 253: 129–134. 123 Momparler RL, Cote S, Eliopoulos N. Pharmacological approach 143 Madrid-Marina V, Lestan B, Nowak PJ, Fox IH, Spychala J. Alt- for optimization of the dose schedule of 5-Aza-29-deoxycytidine ered properties of human T-lymphoblast soluble low Km 59- () for the therapy of leukemia. Leukemia 1997; 11 nucleotidase: comparison with B-lymphoblast enzyme. Leuk Res (Suppl. 1): S1–S6. 1993; 17: 231–240. 124 Flasshove M, Strumberg D, Ayscue L, Mitchell BS, Tirier C, Heit 144 Galmarini CM, Thomas X, Calvo F, Rousselot P, El Jaffari A, Cros W, Seeber S, Schutte J. Structural analysis of the deoxycytidine E, Dumontet C. Expression of cytoplasmic 59-nucleotidase in leu- kinase gene in patients with acute myeloid leukemia and resist- kemic blasts is an adverse prognostic factor in AML patients ance to cytosine arabinoside. Leukemia 1994; 8: 780–785. treated with cytarabine. Blood 2000; 96: 101a. 125 Veuger MJ, Honders MW, Landegent JE, Willemze R, Barge RM. 145 Mansson E, Spasokoukotskaja T, Sallstrom J, Eriksson S, Alber- High incidence of alternatively spliced forms of deoxycytidine tioni F. Molecular and biochemical mechanisms of fludarabine

Leukemia Nucleoside analogues: mechanisms of drug resistance and reversal strategies CM Galmarini et al 888 and cladribine resistance in a human promyelocytic cell line. inhibition of DNA polymerase by 1-beta-D-arabinofuranosylcyto- Cancer Res 1999; 59: 5956–5963. sine 59-triphosphate. Cancer Res 1970; 30: 2636–2644. 146 Laliberte J, Momparler RL. Human cytidine deaminase: purifi- 165 Allaudeen HS, Kozarich JW, Sartorelli AC. Comparative effects cation of enzyme, cloning, and expression of its complementary of the 59-triphosphates of 9-beta-(29-azido-29-deoxy-D-arabino- DNA. Cancer Res 1994; 54: 5401–5407. furanosyl)adenine and 9-beta-D-arabinofuranosyladenine on 147 Capizzi RL, White JC, Powell BL, Perrino F. Effect of dose on the DNA polymerases from L1210 leukemia cells. Nucleic Acids Res pharmacokinetic and pharmacodynamic effects of cytarabine. 1982; 10: 1379–1387. Semin Hematol 1991; 28: 54–69. 166 Yoshida S, Yamada M, Masaki S. Inhibition of DNA polymerase- 148 Honma Y, Onozuka Y, Okabe-Kado J, Kasukabe T, Hozumi M. alpha and -beta of calf thymus by 1-beta-D-arabinofuranosylcyto- Hemin enhances the sensitivity of erythroleukemia cells to 1- sine-59-triphosphate. Biochim Biophys Acta 1977; 477: 144– beta-D-arabinofuranosylcytosine by both activation of deoxycyti- 150. dine kinase and reduction of cytidine deaminase activity. Cancer 167 Higashigawa M, Ido M, Nagao Y, Kuwabara H, Hori H, Ohkubo Res 1991; 51: 4535–4538. T, Kawasaki H, Sakurai M. Decreased DNA polymerase sensi- 149 Momparler RL, Laliberte J. Induction of cytidine deaminase in tivity to 1-beta-D-arabinofuranosylcytosine 59-triphosphate in HL-60 myeloid leukemic cells by 5-aza-29-deoxycytidine. Leuk P388 murine leukemic cells resistant to . Leuk Res Res 1990; 14: 751–754. 1991; 15: 675–681. 150 Momparler RL, Eliopoulos N, Bovenzi V, Letourneau S, Green- 168 Tanaka M, Yoshida S. Altered sensitivity to 1-beta-D-arabinofur- baum M, Cournoyer D. Resistance to cytosine arabinoside by anosylcytosine 59-triphosphate of DNA polymerase alpha from retrovirally mediated gene transfer of human cytidine deaminase leukemic blasts of acute lymphoblastic leukemia. Cancer Res into murine fibroblast and hematopoietic cells. Cancer Gene 1982; 42: 649–653. Ther 1996; 3: 331–338. 169 Reed JC. Bcl-2 and the regulation of programmed cell death. J 151 Schroder JK, Kirch C, Flasshove M, Kalweit H, Seidelmann M, Cell Biol 1994; 124: 1–6. Hilger R, Seeber S, Schutte J. Constitutive overexpression of the 170 Miyashita T, Harigai M, Hanada M, Reed JC. Identification of a cytidine deaminase gene confers resistance to cytosine arabino- p53-dependent negative response element in the bcl-2 gene. side in vitro. Leukemia 1996; 10: 1919–1924. Cancer Res 1994; 54: 3131–3135. 152 Steuart CD, Burke PJ. Cytidine deaminase and the development 171 Miyashita T, Reed JC. Tumor suppressor p53 is a direct transcrip- of resistance to arabinosyl cytosine. Nat New Biol 1971; 233: tional activator of the human bax gene. Cell 1995; 80: 293–299. 109–110. 172 Avramis VI, Nandy P, Kwock R, Solorzano MM, Mukherjee SK, 153 Jahns-Streubel G, Reuter C, Auf der Landwehr U, Unterhalt M, Danenberg P, Cohen LJ. Increased p21/WAF-1 and p53 protein Schleyer E, Wormann B, Buchner T, Hiddemann W. Activity of levels following sequential three drug combination regimen of thymidine kinase and of polymerase alpha, as well as activity fludarabine, cytarabine and induces apoptosis in and gene expression of deoxycytidine deaminase in leukemic human leukemia cells. Anticancer Res 1998; 18: 2327–2338. blasts are correlated with clinical response in the setting of gra- 173 Gartenhaus RB, Wang P, Hoffman M, Janson D, Rai KR. The nulocyte–macrophage colony-stimulating factor-based priming induction of p53 and WAF1/CIP1 in chronic lymphocytic leuke- before and during TAD-9 induction therapy in acute myeloid leu- mia cells treated with 2-chlorodeoxyadenosine. J Mol Med 1996; kemia. Blood 1997; 90: 1968–1976. 74: 143–147. 154 Schro¨der JK, Kirch C, Seeber S, Schutte J. Structural and func- 174 el Rouby S, Thomas A, Costin D, Rosenberg CR, Potmesil M, tional analysis of the cytidine deaminase gene in patients with Silber R, Newcomb EW. p53 gene in B-cell chronic acute myeloid leukaemia. Br J Haematol 1998; 103: 1096–1103. lymphocytic leukemia is associated with drug resistance and is 155 Kirch HC, Schroder J, Hoppe H, Esche H, Seeber S, Schutte J. independent of MDR1/MDR3 gene expression. Blood 1993; 82: 3452–3459. Recombinant gene products of two natural variants of the human 175 Wattel E, Preudhomme C, Hecquet B, Vanrumbeke M, Quesnel cytidine deaminase gene confer different deamination rates of B, Dervite I, Morel P, Fenaux P. p53 mutations are associated cytarabine in vitro. Exp Hematol 1998; 26: 421–425. with resistance to chemotherapy and short survival in hemato- 156 Chabner BA, Hande KR, Drake JC. Ara-C metabolism: impli- logic malignancies. Blood 1994; 84: 3148–3157. cations for drug resistance and drug interactions. Bull Cancer 176 Johnston JB, Daeninck P, Verburg L, Lee K, Williams G, Israels 1979; 66: 89–92. LG, Mowat MR, Begleiter A. P53, MDM-2, BAX and BCL-2 and 157 Liliemark JO, Plunkett W. Regulation of 1-beta-D-arabinofuranos- drug resistance in chronic lymphocytic leukemia. Leuk Lym- ylcytosine 59-triphosphate accumulation in human leukemia phoma 1997; 26: 435–449. cells by deoxycytidine 59-triphosphate. Cancer Res 1986; 46: 177 Dohner H, Fischer K, Bentz M, Hansen K, Benner A, Cabot G, 1079–1083. Diehl D, Schlenk R, Coy J, Stilgenbauer S et al. p53 gene deletion 158 Kunz BA. Mutagenesis and deoxyribonucleotide pool imbalance. predicts for poor survival and non-response to therapy with Mutat Res 1988; 200: 133–147. purine analogs in chronic B-cell leukemias. Blood 1995; 85: 159 Meuth M. The molecular basis of mutations induced by deoxyri- 1580–1589. bonucleoside triphosphate pool imbalances in mammalian cells. 178 Lens D, Dyer MJ, Garcia-Marco JM, De Schouwer PJ, Hamoudi Exp Cell Res 1989; 181: 305–316. RA, Jones D, Farahat N, Matutes E, Catovsky D. p53 abnormali- 160 Ohno Y, Spriggs D, Matsukage A, Ohno T, Kufe D. Effects of 1- ties in CLL are associated with excess of prolymphocytes and beta-D-arabinofuranosylcytosine incorporation on elongation of poor prognosis. Br J Haematol 1997; 99: 848–857. specific DNA sequences by DNA polymerase beta. Cancer Res 179 Kasimir-Bauer S, Ottinger H, Meusers P, Beelen DW, Brittinger 1988; 48: 1494–1498. G, Seeber S, Scheulen ME. In acute myeloid leukemia, coex- 161 Bjorklund S, Skog S, Tribukait B, Thelander L. S-phase-specific pression of at least two proteins, including P-glycoprotein, the expression of mammalian ribonucleotide reductase R1 and R2 multidrug resistance-related protein, bcl-2, mutant p53, and subunit mRNAs. 1990; 29: 5452–5458. heat-shock protein 27, is predictive of the response to induction 162 Parker WB, Shaddix SC, Chang CH, White EL, Rose LM, Brock- chemotherapy. Exp Hematol 1998; 26: 1111–1117. man RW, Shortnacy AT, Montgomery JA, Secrist JAd, Bennett LL, 180 Kitada S, Andersen J, Akar S, Zapata JM, Takayama S, Krajewski Jr. Effects of 2-chloro-9-(2-deoxy-2-fluoro-beta-D-arabinofuranosyl) S, Wang HG, Zhang X, Bullrich F, Croce CM, Rai K, Hines J, adenine on K562 cellular metabolism and the inhibition of Reed JC. Expression of apoptosis-regulating proteins in chronic human ribonucleotide reductase and DNA polymerases by its 59- lymphocytic leukemia: correlations with in vitro and in vivo triphosphate. Cancer Res 1991; 51: 2386–2394. chemoresponses. Blood 1998; 91: 3379–3389. 163 Goan YG, Zhou B, Hu E, Mi S, Yen Y. Overexpression of ribonu- 181 Lomo J, Smeland EB, Krajewski S, Reed JC, Blomhoff HK. cleotide reductase as a mechanism of resistance to 2,2-difluoro- Expression of the Bcl-2 homologue Mcl-1 correlates with survival deoxycytidine in the human KB cancer cell line. Cancer Res of peripheral blood B lymphocytes. Cancer Res 1996; 56: 40–43. 1999; 59: 4204–4207. 182 Gottardi D, Alfarano A, De Leo AM, Stacchini A, Aragno M, Rigo 164 Graham FL, Whitmore GF. Studies in mouse L-cells on the incor- A, Veneri D, Zanotti R, Pizzolo G, Caligaris-Cappio F. In leu- poration of 1-beta-D-arabinofuranosylcytosine into DNA and on kaemic CD5+ B cells the expression of BCL-2 gene family is

Leukemia Nucleoside analogues: mechanisms of drug resistance and reversal strategies CM Galmarini et al 889 shifted toward protection from apoptosis. Br J Haematol 1996; nosylcytosine pharmacology. Cancer Chemother Pharmacol 94: 612–618. 1994; 34: 30–36. 183 Thomas A, El Rouby S, Reed JC, Krajewski S, Silber R, Potmesil 200 Seymour JF, Huang P, Plunkett W, Gandhi V. Influence of fluda- M, Newcomb EW. Drug-induced apoptosis in B-cell chronic rabine on pharmacokinetics and pharmacodynamics of cytarab- lymphocytic leukemia: relationship between p53 gene mutation ine: implications for a continuous infusion schedule. Clin Cancer and bcl-2/bax proteins in drug resistance. Oncogene 1996; 12: Res 1996; 2: 653–658. 1055–1062. 201 Keating MJ, Estey E, O’Brien S, Kantarjian H, Robertson LE, Plun- 184 McConkey DJ, Chandra J, Wright S, Plunkett W, McDonnell TJ, kett W. Clinical experience with fludarabine in leukaemia. Drugs Reed JC, Keating M. Apoptosis sensitivity in chronic lymphocytic 1994; 47: 39–49. leukemia is determined by endogenous endonuclease content 202 Avramis VI, Wiersma S, Krailo MD, Ramilo-Torno LV, Sharpe A, and relative expression of BCL-2 and BAX. J Immunol 1996; 156: Liu-Mares W, Kowck R, Reaman GH, Sato JK. Pharmacokinetic 2624–2630. and pharmacodynamic studies of fludarabine and cytosine arabi- 185 Bromidge TJ, Turner DL, Howe DJ, Johnson SA, Rule SA. In vitro noside administered as loading boluses followed by continuous chemosensitivity of chronic lymphocytic leukaemia to purine infusions after a phase I/II study in pediatric patients with analogues – correlation with clinical course. Leukemia 1998; 12: relapsed leukemias. The Children’s Cancer Group. Clin Cancer 1230–1235. Res 1998; 4: 45–52. 186 Zaja F, Di Loreto C, Amoroso V, Salmaso F, Russo D, Silvestri 203 Shewach DS, Mitchell BS. Differential metabolism of 9-beta-D- F, Fanin R, Damiani D, Infanti L, Mariuzzi L, Beltrami CA, Bacca- arabinofuranosylguanine in human leukemic cells. Cancer Res rani M. BCL-2 immunohistochemical evaluation in B-cell 1989; 49: 6498–6502. chronic lymphocytic leukemia and hairy cell leukemia before 204 Gandhi V, Kisor D, Rodriguez Jr C, Mitchell B, Kurtzberg J, Keat- treatment with fludarabine and 2-chloro-deoxy-adenosine. Leuk ing M, Plunkett W. Pharmacokinetics of arabinosylguanine (ara- Lymphoma 1998; 28: 567–572. G) and its triphosphate (ara-GTP) during a phase I trial of com- 187 Morabito F, Filangeri M, Callea I, Sculli G, Callea V, Fracchiolla pound GW506U in refractory hematological malignancies: cor- NS, Neri A, Brugiatelli M. Bcl-2 protein expression and p53 gene relation with response. Blood 1996; 88: 670a. mutation in chronic lymphocytic leukemia: correlation with in 205 Rodriguez CO Jr, Legha JK, Estey E, Keating MJ, Gandhi V. Phar- vitro sensitivity to and purine analogs. Haemato- macological and biochemical strategies to increase the accumu- logica 1997; 82: 16–20. lation of arabinofuranosylguanine triphosphatein primary human 188 Feng L, Achanta G, Pelicano H, Zhang W, Plunkett W, Huang P. leukemia cells. Clin Cancer Res 1997; 3: 2107–2113. Role of p53 in cellular response to anticancer nucleoside analog- 206 Colly LP, Richel DJ, Arentsen-Honders MW, Kester MG, ter Riet induced DNA damage. Int J Mol Med 2000; 5: 597–604. PM, Willemze R. Increase in Ara-C sensitivity in Ara-C-sensitive 189 Plunkett W, Iacoboni S, Estey E, Danhauser L, Liliemark JO, Keat- and -resistant leukemia by stimulation of the salvage and inhi- ing MJ. Pharmacologically directed ara-C therapy for refractory bition of the de novo pathway. Ann Hematol 1992; 65: 26–32. leukemia. Semin Oncol 1985; 12: 20–30. 207 Bhalla K, Swerdlow P, Grant S. Effects of thymidine and hydrox- 190 Sasvari-Szekely M, Spasokoukotskaja T, Szoke M, Csapo Z, Turi yurea on the metabolism and cytotoxicity of 1-B-D-arabinofur- A, Szanto I, Eriksson S, Staub M. Activation of deoxycytidine kin- anosylcytosine in highly resistant human leukemia cells. Blood ase during inhibition of DNA synthesis by 2-chloro-29-deoxyad- 1991; 78: 2937–2944. enosine (Cladribine) in human lymphocytes. Biochem Pharmacol 208 Gandhi V, Estey E, Keating MJ, Plunkett W. Biochemical modu- 1998; 56: 1175–1179. lation of arabinosylcytosine for therapy of leukemias. Leuk Lym- 191 Spasokoukotskaja T, Sasvari-Szekely M, Keszler G, Albertioni F, phoma 1993; 10: 109–114. Eriksson S, Staub M. Treatment of normal and malignant cells 209 Gandhi V, Estey E, Keating MJ, Chucrallah A, Plunkett W. Chlor- with nucleoside analogues and enhances deoxycytid- odeoxyadenosine and arabinosylcytosine in patients with acute ine kinase activity. Eur J Cancer 1999; 35: 1862–1867. myelogenous leukemia: pharmacokinetic, pharmacodynamic, 192 Sasvari-Szekely M, Csapo Z, Spasokoukotskaja T, Eriksson S, and molecular interactions. Blood 1996; 87: 256–264. Staub M. Activation of deoxycytidine kinase during inhibition of 210 Baker WJ, Royer GL Jr, Weiss RB. Cytarabine and neurologic DNA synthesis in human lymphocytes. Adv Exp Med Biol 1998; toxicity. J Clin Oncol 1991; 9: 679–693. 431: 519–523. 211 Cheson BD, Vena DA, Foss FM, Sorensen JM. Neurotoxicity of 193 Spasokoukotskaja T, Sasvari-Szekely M, Hullan L, Albertioni F, purine analogs: a review. J Clin Oncol 1994; 12: 2216–2228. Eriksson S, Staub M. Activation of deoxycytidine kinase by vari- 212 Mohammad RM, Beck FW, Katato K, Hamdy N, Wall N, Al-Katib ous nucleoside analogues. Adv Exp Med Biol 1998; 431: 641– A. Potentiation of 2-chlorodeoxyadenosine activity by bryostatin 645. 1 in the resistant chronic lymphocytic leukemia cell line (WSU- 194 Iwasaki H, Huang P, Keating MJ, Plunkett W. Differential incor- CLL): association with increased ratios of dCK/59-NT and poration of ara-C, gemcitabine, and fludarabine into replicating Bax/Bcl-2. Biol Chem 1998; 379: 1253–1261. and repairing DNA in proliferating human leukemia cells. Blood 213 Mohammad RM, Limvarapuss C, Hamdy N, Dutcher BS, Beck 1997; 90: 270–278. FW, Wall NR, Al-Katib AM. Treatment of a de novo fludarabine 195 Gandhi V, Kemena A, Keating MJ, Plunkett W. Fludarabine resistant-CLL xenograft model with bryostatin 1 followed by flu- infusion potentiates arabinosylcytosine metabolism in lympho- darabine. Int J Oncol 1999; 14: 945–950. cytes of patients with chronic lymphocytic leukemia. Cancer Res 214 Varterasian ML, Mohammad RM, Eilender DS, Hulburd K, Rodri- 1992; 52: 897–903. guez DH, Pemberton PA, Pluda JM, Dan MD, Pettit GR, Chen 196 Suki S, Kantarjian H, Gandhi V, Estey E, O’Brien S, Beran M, BD, Al-Katib AM. Phase I study of bryostatin 1 in patients with Rios MB, Plunkett W, Keating M. Fludarabine and cytosine arabi- relapsed non-Hodgkin’s lymphoma and chronic lymphocytic noside in the treatment of refractory or relapsed acute lympho- leukemia. J Clin Oncol 1998; 16: 56–62. cytic leukemia. Cancer 1993; 72: 2155–2160. 215 Miyauchi J, Kelleher CA, Wang C, Minkin S, McCulloch EA. 197 Gandhi V, Nowak B, Keating MJ, Plunkett W. Modulation of ara- Growth factors influence the sensitivity of leukemic stem cells to binosylcytosine metabolism by arabinosyl-2-fluoroadenine in cytosine arabinoside in culture. Blood 1989; 73: 1272–1278. lymphocytes from patients with chronic lymphocytic leukemia: 216 Butturini A, Santucci MA, Gale RP, Perocco P, Tura S. GM-CSF implications for combination therapy. Blood 1989; 74: 2070– incubation prior to treatment with cytarabine or doxorubicin 2075. enhances drug activity against AML cells in vitro: a model for 198 Gandhi V, Estey E, Keating MJ, Plunkett W. Fludarabine leukemia chemotherapy. Leuk Res 1990; 14: 743–749. potentiates metabolism of cytarabine in patients with acute mye- 217 Wiley JS, Snook MB, Jamieson GP. Nucleoside transport in acute logenous leukemia during therapy. J Clin Oncol 1993; 11: leukaemia and lymphoma: close relation to proliferative rate. Br 116–124. J Haematol 1989; 71: 203–207. 199 Gandhi V, Robertson LE, Keating MJ, Plunkett W. Combination 218 Petersen AJ, Brown RD, Pope BB, Jamieson GP, Paterson AR, of fludarabine and arabinosylcytosine for treatment of chronic Gibson J, Wiley JS, Joshua DE. Multiple myeloma: expression lymphocytic leukemia: clinical efficacy and modulation of arabi- of nucleoside transporters on malignant plasma cells and their

Leukemia Nucleoside analogues: mechanisms of drug resistance and reversal strategies CM Galmarini et al 890 relationship to cellular proliferation. Leuk Lymphoma 1994; 13: dose cytosine arabinoside for the treatment of refractory acute 491–499. lymphocytic leukemia. Cancer 1990; 65: 5–8. 219 Wiley JS, Cebon JS, Jamieson GP, Szer J, Gibson J, Woodruff RK, 232 Heinemann V, Murray D, Walters R, Meyn RE, Plunkett W. McKendrick JJ, Sheridan WP, Biggs JC, Snook MB et al. Assess- Mitoxantrone-induced DNA damage in leukemia cells is ment of proliferative responses to granulocyte–macrophage col- enhanced by treatment with high-dose arabinosylcytosine. Can- ony-stimulating factor (GM-CSF) in acute myeloid leukaemia cer Chemother Pharmacol 1988; 22: 205–210. using a fluorescent ligand for the nucleoside transporter. Leuke- 233 Crino L, Scagliotti G, Marangolo M, Figoli F, Clerici M, De Mar- mia 1994; 8: 181–185. inis F, Salvati F, Cruciani G, Dogliotti L, Pucci F, Paccagnella A, 220 Reuter C, Auf der Landwehr U, Schleyer E, Zuhlsdorf M, Ameling Adamo V, Altavilla G, Incoronato P, Trippetti M, Mosconi AM, C, Rolf C, Wormann B, Buchner T, Hiddemann W. Modulation Santucci A, Sorbolini S, Oliva C, Tonato M. Cisplatin–gemcitab- of intracellular metabolism of cytosine arabinoside in acute ine combination in advanced non-small-cell lung cancer: a myeloid leukemia by granulocyte–macrophage colony-stimulat- phase II study. J Clin Oncol 1997; 15: 297–303. ing factor. Leukemia 1994; 8: 217–225. 234 Mosconi AM, Crino L, Tonato M. Combination therapy with 221 Ben-Ishay Z, Prindull G, Sharon S. Improved prognosis in mice gemcitabine in non-small cell lung cancer. Eur J Cancer 1997; with advanced myeloid leukemia following administration of 33 (Suppl. 1): S14–S17. GM-CSF and cytosine arabinoside. Leuk Res 1991; 15: 321–325. 235 Grove KL, Guo X, Liu SH, Gao Z, Chu CK, Cheng YC. Anticancer 222 Bettelheim P, Valent P, Andreeff M, Tafuri A, Haimi J, Gorischek activity of beta-L-dioxolane-cytidine, a novel nucleoside ana- C, Muhm M, Sillaber C, Haas O, Vieder L et al. Recombinant logue with the unnatural L configuration. Cancer Res 1995; 55: human granulocyte–macrophage colony-stimulating factor in 3008–3011. combination with standard induction chemotherapy in de novo 236 Grove KL, Cheng YC. Uptake and metabolism of the new acute myeloid leukemia. Blood 1991; 77: 700–711. anticancer compound beta-L-(−)-dioxolane-cytidine in human 223 Lowenberg B, Suciu S, Archimbaud E, Ossenkoppele G, Verhoef prostate carcinoma DU-145 cells. Cancer Res 1996; 56: 4187– GE, Vellenga E, Wijermans P, Berneman Z, Dekker AW, Stryck- 4191. mans P, Schouten H, Jehn U, Muus P, Sonneveld P, Dardenne 237 Kadhim SA, Bowlin TL, Waud WR, Angers EG, Bibeau L, M, Zittoun R. Use of recombinant GM-CSF during and after DeMuys JM, Bednarski K, Cimpoia A, Attardo G. Potent anti- remission induction chemotherapy in patients aged 61 years and tumor activity of a novel nucleoside analogue, BCH-4556 (beta- older with acute myeloid leukemia: final report of AML-11, a L-dioxolane-cytidine), in human renal cell carcinoma xenograft phase III randomized study of the Leukemia Cooperative Group tumor models. Cancer Res 1997; 57: 4803–4810. of European Organisation for the Research and Treatment of Can- 238 Giles F, Cortes J, Thomas DA, Koller C, Beran M, Proulx L, Jolivet cer and the Dutch Belgian Hemato-Oncology Cooperative J, Freireich E, Bivins CA, Estey E, Kantarjian HM. Troxacitabine, Group. Blood 1997; 90: 2952–2961. (BCH-4556), a novel dioxolane nucleoside analog, has anti-leu- 224 Ohno R, Naoe T, Kanamaru A, Yoshida M, Hiraoka A, Kobayashi kemic activity. Proc Am Soc Hemat 1999; 94: 4231A. T, Ueda T, Minami S, Morishima Y, Saito Y et al. A double-blind 239 Cohen A, Lee JW, Gelfand EW. Selective toxicity of deoxyguano- controlled study of granulocyte colony-stimulating factor started sine and arabinosyl guanine for T-leukemic cells. Blood 1983; two days before induction chemotherapy in refractory acute 61: 660–666. myeloid leukemia. Kohseisho Leukemia Study Group. Blood 240 Aguayo A, Cortes JE, Kantarjian HM, Beran M, Gandhi V, Plunk- 1994; 83: 2086–2092. ett W, Kurtzberg J, Keating MJ. Complete hematologic and cyto- 225 Yang LY, Li L, Keating MJ, Plunkett W. Arabinosyl-2-fluoroaden- genetic response to 2-amino-9-beta-D-arabinosyl-6-methoxy-9H- ine augments cisplatin cytotoxicity and inhibits cisplatin-DNA guanine in a patient with chronic myelogenous leukemia in T- cross-link repair. Mol Pharmacol 1995; 47: 1072–1079. cell blastic phase: a case report and review of the literature. Can- 226 Van Den Neste E, Bontemps F, Delacauw A, Cardoen S, Lou- cer 1999; 85: 58–64. viaux I, Scheiff JM, Gillis E, Leveugle P, Deneys V, Ferrant A, 241 Kurtzberg J, Ernst T, Keating M, Gandhi V, Hodge J, Kisor D, Van den Berghe G. Potentiation of antitumor effects of cyclopho- Therriault R, Stephens C, Levin J, Krenitsky T, Elion G, Mitchell sphamide derivatives in B-chronic lymphocytic leukemia cells B. A phase I study of 2-amino-9-B-D-arabinosyl-6-methoxy-9H- by 2-chloro-29-deoxyadenosine. Leukemia 1999; 13: 918–925. purine (506U78) administered on a consecutive five-day sched- 227 Robertson LE, O’Brien S, Kantarjian H, Koller C, Beran M, And- ule in children and adults with refractory hematologic malig- reeff M, Lerner S, Keating MJ. Fludarabine plus doxorubicin in nancies. Proc Am Soc Hematol 1999; 94: 2794A. previously treated chronic lymphocytic leukemia. Leukemia 242 Jamieson GP, Snook MB, Bradley TR, Bertoncello I, Wiley JS. 1995; 9: 943–945. Transport and metabolism of 1-beta-D-arabinofuranosylcytosine 228 Keating MJ, O’Brien S, Robertson LE, Kantarjian H, Dimopoulos in human ovarian adenocarcinoma cells. Cancer Res 1989; 49: M, McLaughlin P, Cabanillas F, Gregoire V, Li YY, Gandhi V et 309–313. al. The expanding role of fludarabine in hematologic malig- 243 Avery TL, Rehg JE, Lumm WC, Harwood FC, Santana VM, Blak- nancies. Leuk Lymphoma 1994; 14: 11–16. ley RL. Biochemical pharmacology of 2-chlorodeoxyadenosine 229 McLaughlin P, Hagemeister FB, Swan F Jr, Cabanillas F, Pate O, in malignant human hematopoietic cell lines and therapeutic Romaguera JE, Rodriguez MA, Redman JR, Keating M. Phase I effects of 2-bromodeoxyadenosine in drug combinations in mice. study of the combination of fludarabine, mitoxantrone, and Cancer Res 1989; 49: 4972–4978. dexamethasone in low-grade lymphoma. J Clin Oncol 1994; 12: 244 Hoglund L, Reichard P. Cytoplasmic 59(39)-nucleotidase from 575–579. human placenta. J Biol Chem 1990; 265: 6589–6595. 230 McLaughlin P, Hagemeister FB, Romaguera JE, Sarris AH, Pate 245 Misumi Y, Ogata S, Ohkubo K, Hirose S, Ikehara Y. Primary O, Younes A, Swan F, Keating M, Cabanillas F. Fludarabine, structure of human placental 59-nucleotidase and identification mitoxantrone, and dexamethasone: an effective new regimen for of the glycolipid anchor in the mature form. Eur J Biochem 1990; indolent lymphoma. J Clin Oncol 1996; 14: 1262–1268. 191: 563–569. 231 Kantarjian HM, Walters RL, Keating MJ, Estey EH, O’Brien S, 246 Zimmermann H. 59-Nucleotidase: molecular structure and func- Schachner J, McCredie KB, Freireich EJ. Mitoxantrone and high- tional aspects. Biochem J 1992; 285: 345–365.

Leukemia