throbber
j631
`
`25
`Clofarabine: From Design to Approval
`John A. Secrist III, Jaideep V. Thottassery, and William B. Parker
`
`25.1
`Introduction
`
`Many different classes of compounds have been found to have utility in treating a
`variety of different types of cancers. These compounds act through a variety of
`different mechanisms, in many cases targeting metabolic differences between
`normal and cancerous cells, and more recently targeting cancer-specific targets and
`pathways. An examination of the drugs that have been approved by governments
`worldwide demonstrates that antimetabolites – compounds that affect the pathways
`leading to nucleic acids – represent a rich source of anticancer drugs. Almost all of
`those approved drugs are either nucleosides or compounds that are converted to
`nucleosides or nucleotides after administration to patients. In the United States, the
`list of FDA-approved antimetabolites of this type includes 5-fluorouracil (colorectal,
`breast, stomach, and pancreatic carcinomas), 6-thioguanine (acute non-lymphocytic
`leukemias), 1-b-D-arabinofuranosylcytosine [acute lymphocytic leukemia (ALL), and
`0
`acute myelocytic leukemia (AML)], 5-fluoro-2
`-deoxyuridine (metastatic colon cancer),
`fludarabine phosphate [chronic lymphocytic leukemia (CLL)], 2-deoxycoformycin
`(hairy cell leukemia), cladribine (hairy cell leukemia), gemcitabine [pancreatic cancer,
`non-small cell lung cancer (NSCLC)], capecitabine (metastatic colorectal and breast
`cancer), nelarabine (T-cell acute lymphoblastic leukemia and lymphoma), decitabine
`(myelodysplastic syndrome), and clofarabine (pediatric ALL). Both fludarabine phos-
`phate andclofarabine were discovered andpushed forwardpreclinically attheSouthern
`Research Institute, and both are products of our preclinical optimization process for
`nucleoside analogues.
`This chapter will focus on the development of clofarabine, and will present that
`development from our viewpoint as preclinical scientists. A recent review covering
`many aspects of the development of clofarabine is recommended to the reader for
`additional details [1].
`
`Modified Nucleosides: in Biochemistry, Biotechnology and Medicine. Edited by Piet Herdewijn
`Copyright Ó 2008 WILEY-VCH Verlag GmbH & Co. KGaA, Weinheim
`ISBN: 978-3-527-31820-9
`
`1
`
`GIL2006
`I-MAK, INC. V GILEAD PHARMASSET LLC
`IPR2018-00122
`
`

`

`632j 25 Clofarabine: From Design to Approval
`
`25.2
`Clofarabine: The Background
`
`Before examining the chronology of the discovery and development of clofar-
`abine, it is important first to identify its current and potential future uses, as well
`as the companies involved. Clofarabine was approved for the treatment of
`pediatric ALL in the US in December 2004, and in Europe in May 2006. It has
`been granted orphan drug status in both the US and Europe. Early efforts toward
`pushing the drug into clinical trials, beginning in 1992, were initiated at the M.
`D. Anderson Cancer Center (MDACC) in Houston, Texas, and included clin-
`icians Drs. J. Freireich, M. Keating and H. Kantarjian, as well as pharmacologists
`Drs. W. Plunkett and V. Gandhi. The initial licensing of the drug by Southern
`Research Institute was to the Eurobiotech Group in 1998, which utilized MDACC
`for the clinical trials. The lag time evident in the above dates is truly unfortunate
`from the standpoint of cancer patients who might have benefited from the drug.
`The difficulties in licensing clofarabine stemmed from two views prevalent in the
`pharmaceutical industry at the time: (i) clofarabine was just another fludarabine,
`and had little chance of making a mark on its own; and (ii) future cancer drugs
`needed to focus on solid tumors such as colorectal, breast, prostate and lung cancer,
`and nucleosides were of little interest in that regard. Moreover, even if they
`had some activity, the market size was too small to be of interest to the larger
`companies. The CEO of Eurobiotech (most recently called Bioenvision), Dr.
`Christopher Wood, understood the properties of fludarabine phosphate, and
`believed that clofarabine had properties that might make it significantly better,
`and on that basis he was eager to proceed. Similarly, the team at MDACC also
`believed that clofarabine had the potential to take its own place among anticancer
`treatments. The eventual approval of clofarabine was aided immeasurably by the
`commitment of these people.
`
`25.3
`The Beginnings
`
`During the early 1980s, two types of nucleoside were found to have very promising
`0
`selectivity in animal models. These compounds were the 2-halo-2
`-deoxyadenosines
`(1), with the halogen being fluorine, chlorine or bromine, and the corresponding b-D-
`0
`arabinofuranosyl analogues (2). The 2
`-deoxy compounds had been prepared in
`several different laboratories [2–4] and examined in various cell lines as potential
`0
`anticancer drugs. In our laboratories, we examined all three of the 2
`-deoxy com-
`pounds in a series of experiments in the then-standard L1210 mouse leukemia model
`system. Interestingly, all three compounds had excellent selectivity, and a summary
`of that previously unpublished data is shown in Table 25.1 [5]. Although it is easy to
`see that all three had some selectivity, the chloro and bromo analogues appeared to be
`the most promising, with some cures seen, and these results warranted further
`investigation.
`
`2
`
`

`

`25.3 The Beginningsj633
`
`Over the next few years, the 2-chloro compound was examined in further detail
`through a collaboration between John Montgomery at Southern Research Institute
`and Dennis Carson at the University of California at San Diego, while the 2-bromo
`compound was further examined by Raymond Blakley and his colleagues at St. Jude
`Children’s Hospital. As events unfolded, the 2-chloro compound (cladribine) was
`eventually approved in 1992 by the FDA for the treatment of hairy cell leukemia.
`In the 2-halo-ara-A series, the fluoro, chloro, and bromo compounds were prepared
`at Southern Research Institute and elsewhere [2, 6–10], and all three had some
`activity [11, 12]. The data demonstrated that the fluoro compound was significantly
`better than the other two, and consequently it was carried forward, eventually being
`0
`approved in 1991 as the 5
`-phosphate (fludarabine phosphate), a prodrug form
`developed to aid solubility, for the treatment of CLL.
`During the first half of the 1980s, some preliminary data were acquired on these
`compounds which, when combined with previous information regarding the physi-
`cal properties of the two series, suggested that some improvements in the structures
`could be made that might have a significant effect on their potential clinical utility. At
`the time, it was of course not known whether any of these compounds would become
`approved, and the quest was to prepare compounds that would have enhanced
`properties that might either achieve approval if the earlier compounds did not, or
`might be next-generation compounds with more attractive properties than any earlier
`compounds that did achieve approval.
`
`Table 25.1 Response of intraperitoneally (i.p.) implanted L1210
`0
`-deoxyadenosines.
`leukemia to 2-halo-2
`
`Compound
`
`Optimal
`i.p. dose
`(LD10, mg
`
`1 dose1)a)
`kg
`
`Total dose
`1)
`(mg kg
`
`Median %
`ILSb) (dying
`mice only)
`
`Net log10
`cell killc)
`
`Tumor-free
`survivors/
`total
`
`F-dAdo
`25
`600
`Cl-dAdo
`20
`480
`Br-dAdo
`40
`960
`a) Treatment schedule was q3 h  8, Days 1, 5, and 9.
`b) Median day of death of tumored control mice (105 cells) was 8 days. ILS, increase in lifespan.
`c) Net log10 reduction in the tumor cell population between the beginning and the end of therapy,
`based on the median day of death of the mice that died.
`
`0/10
`5/10
`3/6
`
`þ118
`þ150
`þ125
`
`þ0.5
`þ2.9
`þ1.1
`
`3
`
`

`

`634j 25 Clofarabine: From Design to Approval
`
`Thus, attention was focused on three properties while seeking to improve on the two
`seriespresentedabove.Itiseasiesttoconsiderthecharacteristicsthatmightbeimproved
`by focusing on the two compounds that were eventually approved, fludarabine and
`cladribine. In the case of fludarabine phosphate, when administered to an animal it is
`rapidly cleaved to fludarabine, which enters the cells and is further metabolized [13].
`0
`In the case of cladribine, it is well known that 2
`-deoxy compounds in the purine
`series are susceptible to chemical cleavage at low pH, and thus a loss of potency
`through hydrolytic cleavage is clearly an issue with cladribine. In addition, cleavage of
`the glycosidic bond by phosphorylases is another means of loss of potency. With both
`types of cleavage, 2-chloroadenine would be generated, which is a compound with
`only modest toxicity.
`For fludarabine, the chemical hydrolysis of the glycosidic bond is not a significant
`0
`problem as the presence of the 2
`-hydroxyl group provides significant stability,
`although there is a loss of potency through some phosphorylase cleavage [13–16].
`In the case of fludarabine, this process generates 2-fluoroadenine, which is a highly
`undesirable metabolite. This purine is readily metabolized up to 2-fluoro-ATP,
`which is an extremely toxic but unselective compound, and so its systemic generation
`could present a concern. In recent years, attention has been focused on utilizing
`gene therapy approaches to generate this toxin in tumor cells in a selective manner [17].
`The other mechanism of loss of potency that can occur with adenine derivatives is
`enzymic deamination, which is carried out by adenosine deaminase at the nucleoside
`level, and by AMP deaminase at the monophosphate level. The incorporation of a
`2-halogen into the adenine ring of a nucleoside confers significant resistance to
`deamination as compared to the parent adenine compounds [11, 12]. Many evalua-
`tions have been carried out examining the ability of the 2-haloadenine nucleosides to
`serve as substrates for adenosine deaminase, and although they are highly resistant to
`deamination, all have at least some substrate activity. The order of deamination is
`F > Cl > Br, and within that order the 2-fluoro compounds are significantly more
`susceptible to deamination than the other two. Thus, in the case of fludarabine there
`is a minor loss of potency through some deamination [13–16], a pathway that is not a
`significant problem with cladribine.
`The other key issue with regard to nucleoside analogues is their activation to an
`active metabolite which, in the vast majority of cases, is the nucleoside triphosphate
`(NTP). In general, nucleosides exert their effects on the biosynthetic pathway leading
`0
`to DNA, and thus, analogues of 2
`-deoxynucleosides are typically of more interest
`than the building blocks of RNA, although ribonucleoside analogues were also
`prepared. It was determined that the addition of a 2-halogen did not prevent the
`0
`phosphorylation of some 2
`-deoxynucleoside analogues, and many laboratories have
`determined that the major enzyme carrying out the initial phosphorylation is
`generally deoxycytidine kinase [18–20]. Another key observation was that nucleosides
`with an arabino configuration often were also substrates of deoxycytidine kinase.
`The other enzymes carrying out conversion of monophosphates to their di- and
`triphosphate metabolites were in general less discriminating, and the majority of
`0
`nucleosides that could be metabolized to the 5
`-monophosphate were converted at a
`meaningful rate to the triphosphate.
`
`4
`
`

`

`25.4 The Next Generation of Compoundsj635
`
`The above-described information relates to the situation during the early to mid-
`1980s as ways were sought to improve on the activity of this class of potential
`anticancer nucleosides. The set of simple conclusions drawn from the above
`information can be summarized as follows:
`
`. A 2-halogen in an adenine ring analogue dramatically reduces deamination,
`but in general will allow phosphorylation, depending upon the carbohydrate
`attached.
`. A 2-chloro or 2-bromoadenine ring is more desirable than a 2-fluoro, based upon
`the high toxicity of any 2-fluoroadenine that may be generated, and also based upon
`its increased ability of 2-fluoroadenine-containing nucleosides to serve as sub-
`strates for adenosine deaminase.
`0
`in the arabino configuration – one that will significantly
`. A stabilizing group at C-2
`reduce both phosphorylase cleavage and hydrolytic cleavage of the glycosidic
`bond – is highly desirable.
`
`25.4
`The Next Generation of Compounds
`
`Over the years, a highly efficient system was developed for the rapid examination of
`new compounds in our anticancer drug discovery program, which was strongly
`supported by the US National Institutes of Health. Whenever a new compound had
`been prepared and properly characterized, it was submitted for an evaluation of its
`cytotoxicity in a small series of cancer cell lines, with generally six or seven such lines
`being derived from various types of human tumors. Typically, the results were
`available in a few weeks. In all of these cases, the corresponding human tumor
`xenograft mouse model was available if a compound exhibited significant cytotoxicity.
`The main challenge was to prepare sufficient material for evaluation in a mouse
`model, once it had been learned that such an examination was warranted based upon
`the cytotoxicity profile. When sufficient material became available for such an initial
`evaluation, the compound was submitted and generally placed into a test system
`within a month. In parallel, the mechanistic evaluation of compounds of potential
`interest was started in our biochemistry laboratories. Together, this research effort
`provided us with the basic information on the activation of new nucleosides to the
`various phosphorylated metabolites, their effects on DNA, RNA and protein synthe-
`sis, and also specific information on key enzymes. Feedback from both the in-vitro
`and in-vivo evaluations was thus rapidly available, and we were able quickly to adjust
`our target structure list based upon this iterative feedback. This simple system
`prevented us from spending too much time on the synthesis of series of compounds
`that did not show promise as anticancer drugs.
`By utilizing this efficient system, a variety of compounds was evaluated relatively
`rapidly. The major efforts revolved around carbohydrate modifications with the
`2-haloadenines as the bases, and on similar compounds with the nitrogen base
`somewhat altered.
`
`5
`
`

`

`636j 25 Clofarabine: From Design to Approval
`
`Examples of ring-altered compounds included 2-fluoro-8-azaadenine nucleosides
`such as 3 [21], and a ring-fluorinated analogue 4 of formycin A [22]. Unfortunately,
`however,thetwocompounds3and4hadcharacteristicsthatpreventedthemfrombeing
`of therapeutic use. For example, compound 3 was not significantly cytotoxic, presum-
`ably because it was not amenable to initial phosphorylation, while 4 was converted to
`the monophosphate, but not further converted to the di- and triphosphate levels.
`
`In the case of carbohydrate-modified nucleosides, the initial focus – as noted above
`0
`– was at the 2
`-position. It was felt that building in halogen atoms as well as certain
`0
`other groups with significant electronegativity at the 2
`-position would accomplish
`several goals. First, these compounds should impart significant hydrolytic stability of
`the glycosidic bond. Second, this alteration might well reduce the ability of these
`compounds to serve as substrates for phosphorylases. Therefore, attention was
`0
`redirected towards preparing compound series 5, which incorporated a 2
`-bromine,
`chlorine, fluorine, azido or amino group. Another related compound, 6, which also
`has anticancer activity, has been prepared by the Matsuda group and includes a
`0
`2
`-cyano group and the arabino configuration [23].
`
`Hence, compounds were prepared that incorporated all of these groups at the
`0
`2
`-position and also contained a 2-haloadenine moiety [24]. At that time, the synthetic
`0
`routes involved the displacement of a leaving group at C-2
`of a nucleoside with
`inversion from the ribo to the arabino configuration, as had been accomplished by
`Ueda [25]. However, the cytotoxicity results from this series of compounds, with the
`0
`exception of those incorporating a 2
`-fluorine, were not impressive [24], and thus
`0
`attention was re-focused on compounds with a 2
`-fluorine, in turn leading to the
`preparation of the series of compounds 7. The incorporation of a fluorine atom has
`always been attractive, because the size of the atom causes the least disruption, and is
`the closest to hydrogen. A fluorine also often significantly alters the biochemical
`
`6
`
`

`

`25.4 The Next Generation of Compoundsj637
`
`properties of a molecule. Initially, the synthetic route was quite laborious because the
`fluorocarbohydrate precursor required a lengthy synthetic route. However, starting
`0
`in the 1970s the group of Fox and Watanabe was also preparing nucleosides with a 2
`-
`fluorine [26–28], though with a major focus on pyrimidine nucleosides as antiviral
`agents, and we were able to take advantage of their synthetic procedures. The initial
`route (see below) [29, 30] yielded predominantly the desired b nucleoside, but
`separation of the anomers was necessary. The first synthesis in this target series
`was completed during the mid-1980s. By utilizing different bases for the coupling, all
`three compounds 7 were prepared. Based upon the pursuit of one of the pyrimidine
`nucleosides of antiviral interest, an improved synthesis of the fluorocarbohydrate
`precursor was developed and published [31], and this route proved to be a very useful
`0
`advance. Quite recently, another important advance relative to the a/b ratio at C-1
`has been made by utilizing a three-solvent mix for the coupling reaction, which is
`carried out with 2-chloroadenine rather than 2,6-dichloropurine [32]. This improve-
`ment was made during the optimization of the synthesis of clofarabine (7) for
`manufacturing purposes.
`
`7
`
`

`

`638j 25 Clofarabine: From Design to Approval
`
`Table 25.2 Anticancer activity of clofarabine against human tumor xenografts in mice.
`
`Tumor
`
`COLO 205 colon
`DLD-1 colon
`HCC-2998 colon
`HCT-15 colon
`HCT-116 colon
`HT29 colon
`KM20L2 colon
`SW-620 colon
`A498 renal
`CAKI-1 renal
`RXF 393 renal
`SN12C renel
`
`Activitya)

`A594 lung
`
`NCI-H23 lung
`
`NCI-H322M lung

`NCI-H460 lung

`DU-145 prostate

`LNCAP prostate
`þþ
`PC-3 prostate
`þþþ
`HL-60 leukemia

`CCRF-CEM leukemia
`þþ
`K-562 leukemia
`–
`MOLT-4 leukemia
`
`AS283 lymphoma
`þþ
`RL lymphoma
`a) – inactive;  marginal; þ minimal; þ þ good (tumor regressions); þ þ þ excellent (cures).
`
`Tumor
`
`Activitya)
`þþ

`þþþ
`þþ

`þþþ
`þþ
`þþ
`þþ
`þþþ
`–
`þþ
`
`0
`-fluorine in the arabino
`All three of the 2-haloadenine nucleosides with a 2
`configuration were significantly cytotoxic in multiple cancer cell lines [29, 30], in
`contrast to the other compounds mentioned above, and also in contrast to the
`0
`0
`0
`2
`-fluoro compound with the ribo configuration or the 2
`,2
`-difluoro analogue [33].
`This result encouraged us to move in the direction of human tumor xenograft
`experiments, and also to embark upon biochemical studies to determine the
`compound’s mechanism of action. The initial animal data strongly suggested that
`the 2-chloro compound had the most activity, and it was therefore chosen for further
`experiments. Although the 2-fluoro compound was also of some interest, it was more
`difficult to synthesize, and even a small amount of glycosidic cleavage yielding 2-
`fluoroadenine (see above) would be undesirable.
`Mechanistic information presenting the unique profile of clofarabine is summa-
`rized in the following section. With regard to further animal studies, many different
`human tumor xenograft models were employed, and much of the data obtained have
`been presented in one report [34]. A concise summary of clofarabine activity in
`human tumor models in mice is provided in Table 25.2. The conclusion to be drawn
`from these animal studies was that clofarabine had significant and often curative
`activity in a number of systems across a broad spectrum of human tumor types. This
`type of profile is exactly what is required for a new drug to have a strong chance of
`success in clinical trials. It is known that there is no direct correlation between
`outstanding activity in a particular human tumor type in a xenograft model and
`clinical success with that agent in treating the same type of tumor. In our experience,
`however, robust, broad-spectrum activity against a variety of human tumors in these
`models is a good indication that a compound will find clinical utility. Clofarabine
`clearly had such a profile, and we were eager for it to have the opportunity to move to a
`clinical trial.
`
`8
`
`

`

`25.5 Mechanism of Action of Clofarabinej639
`
`25.5
`Mechanism of Action of Clofarabine
`
`Generally speaking, all nucleoside analogues used in the treatment of cancer,
`including clofarabine, have a similar mechanism of action, namely that they are
`0
`converted to their respective 5
`-triphosphates and inhibit DNA synthesis. However, it
`is clear from the varying clinical activities of these agents that subtle quantitative and
`qualitative differences in the metabolism of these agents and their interactions with
`target enzymes can have a profound impact on their antitumor activity. Thus, a precise
`understanding of the mechanism of action of each of these compounds is important in
`order to determine those biochemical actions which are most important to antitumor
`activity and might aid the rational design and development of new agents.
`
`25.5.1
`Transport and Metabolism to Active Metabolites
`
`Clofarabine is efficiently transported into cells [35] by both the human equilibrative
`nucleoside transporters (hENT1, hENT2) and the human concentrative nucleoside
`transporters (hCNT2, hCNT3). As noted earlier, the primary enzyme involved in the
`activation of clofarabine in tumor cells is deoxycytidine kinase [36–38]. Clofarabine is
`a very good substrate for this enzyme, with Km and Vmax values similar to those of
`deoxycytidine. The structure of deoxycytidine kinase with clofarabine in the active site
`has recently been determined [39]. The results indicated that the conformation of the
`enzyme/clofarabine complex was similar to structures of the pyrimidine-bound
`complexes, and that interactions between the 2-Cl group and its surrounding
`hydrophobic residues contributed to the high catalytic efficiency of deoxycytidine
`kinase with clofarabine. Clofarabine is also a good substrate for deoxyguanosine
`kinase [40], an enzyme that is expressed in mitochondria. The contribution of
`deoxyguanosine kinase to the phosphorylation of clofarabine in cells is low due to
`the much higher expression of deoxycytidine kinase activity in most cell types [41],
`although this may be important in cells that express low activities of deoxycytidine
`kinase.
`Similar intracellular concentrations of clofarabine-5
`-monophosphate (the product
`0
`-triphosp-
`of the reaction of clofarabine with deoxycytidine kinase) and clofarabine-5
`hate (clofarabine-TP) accumulate in tumor cells treated with clofarabine [37]. This
`indicates that phosphorylation of the monophosphate of clofarabine is the rate-
`limiting step in its activation to the triphosphate, and that the monophosphate kinase
`does not easily tolerate substitutions at the 2-position of a purine nucleoside as large
`as a chlorine atom. Nucleoside kinases are usually the rate-limiting enzymes in the
`activation of anticancer nucleoside analogues. Even though clofarabine is a relatively
`poor substrate for the monophosphate kinase, clofarabine-TP accumulates to high
`levels in cancer cells treated with clofarabine. Clinical studies have indicated that the
`concentration of clofarabine-TP in blasts cells is 10-fold greater than the plasma
`concentration achieved after a 1-h infusion [42].
`
`0
`
`9
`
`

`

`640j 25 Clofarabine: From Design to Approval
`
`Phosphorylated metabolites of nucleoside analogues do not readily cross cell
`membranes, and are therefore trapped in the cell in which they were created; this
`contrasts with the nucleosides themselves, which will freely distribute across the
`cell membrane. Consequently, the antitumor activity of these agents can extend
`well beyond the time that the parent drug circulates in the plasma, because the
`active metabolite is maintained in tumor cells long after the drug has disappeared
`from the plasma. The initial half-life for the clearance of clofarabine-TP from
`cultured tumor cells (CEM) is approximately 2 h [33, 37]. In samples taken from
`patients treated with clofarabine, more than 50% of the clofarabine-TP was still
`present in circulating leukemic cells 24 h after the completion of transfusion [42].
`The long retention time of clofarabine-TP is believed to be a major contributory
`factor to the high activity demonstrated by clofarabine in solid tumor xenografts in
`mice [34].
`
`25.5.2
`Inhibition of DNA Synthesis
`
`As with other anticancer nucleoside analogues, the primary activity of clofarabine
`that is responsible for its antitumor activity is the inhibition of DNA synthesis [33, 36].
`RNA and protein synthesis are inhibited by clofarabine only at high concentrations.
`Clofarabine-TP is a potent inhibitor of ribonucleotide reductase [36, 43], a critical
`enzyme involved in the de-novo synthesis of deoxynucleotides [44]. The activity of
`ribonucleotide reductase in cells is tightly controlled by the natural deoxynucleoside
`triphosphates to ensure that the cell has all of the deoxynucleotides needed for DNA
`synthesis, and in the correct concentrations. dATP is a potent regulator of ribonu-
`cleotide reductase activity, and inhibits the reduction of ADP, UDP, and CDP [45].
`The effect of clofarabine on intracellular nucleotide pools suggest that clofarabine-TP
`interacts with ribonucleotide reductase in the allosteric binding site as an analogue of
`dATP. The inhibition of ribonucleotide reductase activity results in decreases in the
`levels of deoxynucleotide triphosphates, which are required for the synthesis of DNA.
`Clearly, decreasing the natural deoxynucleotide pools is sufficient to cause an
`inhibition of DNA synthesis; this is also the mechanism of action of hydroxyurea,
`another useful anticancer agent.
`Clofarabine is readily incorporated into DNA, although a small amount of drug is
`also detected in RNA [37]. At low concentrations, most of the clofarabine detected in
`DNA is incorporated into internal sites in the DNA of CEM cells, which indicates that
`the elongation of DNA synthesis is not prevented by the drug’s incorporation into
`DNA. The immediate inhibition of DNA synthesis in cells treated with clofarabine
`(even when most of the clofarabine is in internal positions in the DNA), however,
`indicates that inhibition of the DNA replication complex is the primary action of
`clofarabine that results in the death of the tumor cell. The disruption of DNA function
`due to the incorporation of clofarabine into daughter strands is of secondary
`importance to the activity of clofarabine.
`Clofarabine-TP is a good substrate and inhibitor of DNA polymerases a and e, two
`important enzymes involved in the replication of chromosomal DNA [36, 37].
`
`10
`
`

`

`25.5 Mechanism of Action of Clofarabinej641
`
`Clofarabine-TP is utilized by DNA polymerase a, with Km and Vmax values similar to
`those of dATP (the natural substrate). Once clofarabine-TP is incorporated into the
`growing chain, however, the ability of the polymerase to add new nucleotides is
`significantly less than after dATP incorporation. The inhibition of DNA polymerase a
`by clofarabine-TP is similar to that of fludarabine-TP [36]. The effect of clofarabine-TP
`on DNA polymerase e activity was similar to that seen with DNA polymerase a, except
`that clofarabine-TP more effectively inhibited chain elongation by DNA polymerase
`e [43]. This enhanced inhibition of chain elongation resulted in a significant
`inhibition of DNA synthesis at clofarabine-TP concentrations that were only 3% of
`the dATP levels used in the experiment.
`0
`The removal of clofarabine from the 3
`-end of DNA chains is an important part of
`the mechanism of action of nucleoside analogues that has not yet been evaluated. If
`0
`clofarabine can be quickly removed from the 3
`-terminus of the DNA chain, then
`DNA synthesis could continue normally; however, if it is not removed, then it will
`have a lasting effect on the ability of the DNA polymerase to elongate the DNA chain.
`The incorporation of two or more clofarabine residues sequentially in the DNA [36]
`may be harder to repair than single incorporations, and may represent a greater block
`to DNA synthesis. The continued inhibition of DNA synthesis is another attribute of
`nucleoside analogues that is a major contributor to their mechanisms of action. A
`short exposure of tumor cells to circulating drug may have lasting effects on the ability
`of the tumor cell to replicate its DNA.
`The results of the above-mentioned studies concluded that DNA synthesis is
`inhibited in cells treated with clofarabine by two distinct, but complementary, actions:
`(i) the inhibition of ribonucleotide reductase; and (ii) the inhibition of DNA
`polymerase activities. The potent inhibition of ribonucleotide reductase activity by
`clofarabine-TP enhances its inhibition of the replicative DNA polymerases by
`decreasing the intracellular concentration of the natural substrate, dATP, which in
`turn competes with clofarabine-TP for use as a substrate by these enzymes. With
`respect to the inhibition of ribonucleotide reductase and DNA polymerases, clofa-
`rabine combines the features of cladribine (potent inhibition of ribonucleotide
`reductase) and fludarabine (potent inhibition of DNA polymerase) into one molecule.
`
`25.5.3
`Induction of Apoptosis
`
`The inhibition of DNA synthesis by clofarabine is responsible for the induction of the
`apoptotic response in replicating cells. Inhibition of DNA replication in cells
`normally leads to the “turning on” of replication checkpoint pathways. Stalled
`replication forks can threaten DNA replication fidelity, and cells respond to replica-
`tion blocks by triggering checkpoint pathways that monitor replication fork progres-
`sion [46]. This monitoring of DNA synthesis operates normally during low-intensity
`replication stress, and is required for tumor cells to resume cell-cycle progression.
`However, during chronic or high-intensity replication stress, stalled forks do not
`restart after removal of the stressor, and this results in irreversible S-phase arrest,
`possibly mitotic catastrophe, and cell death.
`
`11
`
`

`

`642j 25 Clofarabine: From Design to Approval
`
`It has been found that replication stress induced by the treatment of CEM cells with
`clofarabine leads to Chk1 phosphorylation, Chk1 down-regulation, concomitant
`apoptosis, and cell death (unpublished data). Consistent with our findings on Chk1
`activation, it has been found that Cdc25A is down-regulated upon low-dose clofa-
`rabine treatment in CEM cells, a consequence of its phosphorylation by Chk1, which
`is accompanied by accumulation of cells in G1/S and S [47]. Chk1 and its upstream
`activator, ATR, phosphorylate a host of substrates involved in the control of the firing
`of additional replication origins. In addition, it is known that phosphorylation of
`Chk1 on Ser345 triggers its ubiquitylation, resulting in the proteasomal degradation
`of Chk1 [48].
`The induction of apoptosis in response to replication stress can also be mediated by
`the tumor suppressor/transcription factor p53. Recently, it was demonstrated that
`0
`two deoxycytidine analogues – gemcitabine and 4
`-thio-araC – can induce the
`stabilization of p73, a p53 paralogue in both p53 null cancer cells and in cells with
`wild-type p53 [49]. The p73 protein shares significant sequence similarity with p53,
`and can transactivate proapoptotic p53 target genes such as Bax and PUMA. In turn,
`these proteins can induce the release of cytochrome c and other apoptogenic
`molecules from the mitochondrial outer membrane. It has been found that the
`incubation of CEM cells (in which p53 is inactive) with clofarabine also robustly
`induced p73 levels (unpublished data).
`
`25.5.4
`Activity against Non-Proliferating Cancer Cells
`
`Clofarabine is also active against non-replicating cells [50], and has been shown to
`interfere with mitochondrial integrity and function in primary CLL cells [51], causing
`the release of cytochrome c and apoptotic inducing factor (AIF-1). This has led to the
`suggestion that clofarabine induces cell death by initiating the apoptotic cascade in
`these cells, although the precise mechanism for such activity has not yet been
`elucidated. Other adenine nucleosides such as cladribine and fludarabine have been
`shown to induce the intrinsic apoptotic pathway, which causes the release of
`apoptogenic molecules, such as cytochrome c into the cytosol; this leads to the
`cleavage and activation of caspase 9 (an initiator caspase) and subsequently caspase 3
`(an executioner caspase) [52, 53]. The present authors and others have shown that
`clofarabine induces the depleti

This document is available on Docket Alarm but you must sign up to view it.


Or .

Accessing this document will incur an additional charge of $.

After purchase, you can access this document again without charge.

Accept $ Charge
throbber

Still Working On It

This document is taking longer than usual to download. This can happen if we need to contact the court directly to obtain the document and their servers are running slowly.

Give it another minute or two to complete, and then try the refresh button.

throbber

A few More Minutes ... Still Working

It can take up to 5 minutes for us to download a document if the court servers are running slowly.

Thank you for your continued patience.

This document could not be displayed.

We could not find this document within its docket. Please go back to the docket page and check the link. If that does not work, go back to the docket and refresh it to pull the newest information.

Your account does not support viewing this document.

You need a Paid Account to view this document. Click here to change your account type.

Your account does not support viewing this document.

Set your membership status to view this document.

With a Docket Alarm membership, you'll get a whole lot more, including:

  • Up-to-date information for this case.
  • Email alerts whenever there is an update.
  • Full text search for other cases.
  • Get email alerts whenever a new case matches your search.

Become a Member

One Moment Please

The filing “” is large (MB) and is being downloaded.

Please refresh this page in a few minutes to see if the filing has been downloaded. The filing will also be emailed to you when the download completes.

Your document is on its way!

If you do not receive the document in five minutes, contact support at support@docketalarm.com.

Sealed Document

We are unable to display this document, it may be under a court ordered seal.

If you have proper credentials to access the file, you may proceed directly to the court's system using your government issued username and password.


Access Government Site

We are redirecting you
to a mobile optimized page.





Document Unreadable or Corrupt

Refresh this Document
Go to the Docket

We are unable to display this document.

Refresh this Document
Go to the Docket