throbber
Review
`
`Annals of Oncology 6'. 871-881, 1995.
`O 1995 Kluwer Academic Publishers. Primed in the Netherlands.
`
`Folate-based thymidylate synthase inhibitors as anticancer drugs
`
`A. L. Jackman' & A. H. Calvertz
`
`‘The CRC Centre for Cancer Therapeutics at the Institute of Cancer Research, Sutton, Surrey; ’I7re Cancer Research Unit, University of
`Newcastle upon Tyne, Newcastle upon Tyne, UK.
`
`Summary
`
`The enzyme, thymidylate synthase (TS) is considered an
`important
`target for the development of new anticancer
`agents. Moreover, the folate—binding site in TS is believed to
`offer better opportunities for the design of highly specific
`inhibitors than the pyrimidine (dUMP) binding site. This
`belief led to the design of N '°-propargyl-5,8-dideazafolic
`acid (CB3717), a quinazoline-based drug which had anti-
`tumour activity in clinical studies. Occasional, but serious
`nephrotoxicity led to the withdrawal of CB37l7 from further
`clinical
`study. More water-soluble and non-nephrotoxic
`analogues were developed with an interesting diversity in bio-
`chemical profile, particularly with respect to interactions
`with the reduced-folate cell membrane carrier (RFC) and
`folylpolyglutamate synthetase (FPGS). An example of a com-
`pound that uses both of these processes well is the quinazo-
`line, ZD1694 (Tomudex), a drug which is about to complete
`phase II] evaluation for colorectal cancer. High chain length
`polyglutamates are formed that are up to 70-fold more
`potent TS inhibitors than the parent drug (Ki tetraglutamate
`-= 1 nM). Furthermore they are retained in cells/tissues for a
`prolonged period. A number of other novel folate-based TS
`inhibitors are currently in pre-clinical or clinical study. For
`
`example, LY2315l4 is a pyrrolopyrirnidine analogue in
`phase I study and, although less potent as a T8 inhibitor, has
`biochemical properties similar to ZDI694. Another com-
`pound
`in
`phase
`I
`study
`is
`the benzoquinazoline,
`BW]843U89 which has somewhat different properties. It is
`a very potent TS inhibitor (Ki = 0.09 nM) and an excellent
`substrate for the RFC (human) and FPGS, but polyglutama-
`tion proceeds to diglutamate only and is not accompanied by
`increased TS inhibition. Another highly water-soluble com-
`pound in pre-clinical development is ZD9331 which was
`specifically designed to use the RFC but not be a substrate
`for FPGS. Potent TS inhibition (Ki = 0.4 nM) was achieved
`through a rational programme of computerised molecular
`modelling of the active site of “IS and a large database of
`structure-activity relationships. Two lipophilic compounds
`were designed to be devoid of interactions with either the
`RFC or FPGS. High resolution crystal complexes of E. coli
`TS were central to obtaining potent TS inhibitors and both
`AG337 (Ki human recombinant TS - 16 nM) and AG331
`(Ki - 12 nM) are in clinical studies. This portfolio of novel
`compounds therefore comprehensively addresses the poten-
`tial of TS as a target for cancer chemotherapy.
`
`Key words: thymidylate synthase, antifolates
`
`Introduction
`
`Thymidylate synthase (TS) catalyses the methylation of
`dUMP to give TMP, which after metabolism to TTP is
`exclusively incorporated into DNA (Fig. 1). This fact
`alone makes TS an attractive target for the develop-
`ment of an anticancer agent. The co-substrate for the
`reaction is the reduced-folate cofactor, 5,10-methylene
`tetrahydrofolate (CHZFI-I4) which becomes oxidised to
`dihydrofolate (Fl-I2) during this reductive methylation
`reaction. Methotrexate (MTX; Fig. 2a) and 5-t]uorou-
`racil (FU; Fig. 2b) were amongst the earliest anticancer
`drugs developed and both partly act through inhibition
`of TS (Fig. 1). MTX primarily inhibits dihydrofolate
`reductase (Dl-IFR), an enzyme which functions to re-
`generate FH, (produced in the TS reaction) to the fully
`reduced tetrahydrofolate (FH,) form (reviewed in [1]).
`This in turn accepts a 1 carbon unit from a donor such
`as serine allowing it to function once more in 1 carbon
`transfer in various folate-dependent reactions such as
`
`TS and the enzymes involved in de novo purine syn-
`thesis (Fig. 1). FU is metabolised intracellularly to pro-
`duce a number of anabolites. One of these, 5-fluoro-
`deoxyuridine monophosphate (FdUMP) binds tightly
`to the dUMP binding site of 'IS [2]. However other
`anabolites of FU are believed, at least in some tumours
`and in some administration protocols, to have other
`effects, particularly after incorporation of the base into
`RNA [3,4].
`MTX and FU have each found roles in the treatment
`
`of certain tumour types, alone or in combination with
`other drugs. For example, MTX is still an important
`single agent treatment for low risk choriocarcinoma [S],
`and, when combined with other drugs, for the treat-
`ment of childhood acute lymphoblastic leukemia
`(maintenance therapy) and non-Hodgkins lymphoma
`[6]. MTX and/or 5-FU, when used in combination with
`other agents, have a place in the treatment of the com-
`mon solid tumours e.g. CMF (cyclophosphamide/
`MTX/5-FU) therapy for breast cancer [7]. Bolus 5-FU
`
`Lilly Ex. 2073
`Sandoz V. Lilly IPR2016-00318
`
`Lilly Ex. 2073
`Sandoz v. Lilly IPR2016-00318
`
`

`
`RFC modabd
`:0 lD1694 (gluln
`D1094
`LY231614 ——F l.Y23181((glu)n
`BW1843UI9 *5 BW1843Ul9[¢|u)2
`
`5.10-CHQFH4
`
`dUMP
`
`IE1
`
`RNA
`
`DNA
`
`Fig. I. Thymidylate synthase as a target for antifolates and 5-
`fluorouracil.
`
`as a single agent had, for many years, been the standard
`drug for the treatment of colorectal cancer, albeit with
`very limited response rates (~ 10%) and no marked
`improvement on the survival of treated patients (re-
`viewed in [8, 9]). More recently the use of an adjuvant
`F U + Levarnisole regimen for Dukes C colorectal can-
`cer has been associated with a marked improvement in
`survival [10], suggesting a potentially curative role for
`drugs of this type (data from studies randomised
`against FU alone are not available). It is thus clear that
`the place of these drugs in clinical protocols has estab-
`lished the principle that some solid tumours may be
`antimetabolite-sensitive.
`The biochemical pharmacology of both MTX (re-
`viewed in [l]) and FU (reviewed in [9]) has been very
`widely investigated and understood, and it may be
`argued that the vast amount of knowledge accrued is
`out of proportion to the clinical usefulness of either
`drug. However such fundamental research has led to
`the design of alternative administration protocols, drug
`combinations, modulatory and rescue agents that im-
`proved response rates and overcame resistance in par-
`ticular tumour types [8, 9]. Examples include the use of
`high-dose MTX followed by Leucovorin (LV) rescue
`for the treatment of osteogenic sarcomas (11) and the
`sequential dosing of MTX followed by FU for colo-
`rectal cancer (reviewed in l8,9]). The most notable
`example in recent years relating to the use of FU was
`the discovery that the folate cofactor for the TS reac-
`tion can be low and limiting for the formation of the
`stable ternary complex in which FdUMP binds (re-
`viewed in [9]). The provision of LV to elevate the co-
`factor pool forms the basis for the now commonly used
`
`FU/LV clinical protocols for the treatment of advanced
`colorectal cancer [8, 9] as well as for adjuvant therapy.
`However, it is probably fair to conclude that increased
`response rates, in the order of 25%-30%, are seldom
`accompanied by a significant improvement in patient
`survival [8, 9].
`Other antimetabolites have also found roles in can-
`
`cer therapy (particularly the leukemias) but are not
`generally considered particularly active
`in
`solid
`tumours. It may be pertinent to question whether the
`concept of using antimetabolite therapy to treat the
`common (and usually slowly proliferating) cancers is
`flawed, whether we are approaching the wrong targets
`or whether the targets are right but we are just using
`inadequate drugs. Thymidylate synthase is probably
`one of the best examples of a target where, in a few
`years, these answers should be available. This is for a
`number of reasons. First, there has been the rational
`design of specific, folate-based inhibitors of TS in the
`last 15-20 years. Commitment to the rapid develop-
`ment of TS inhibitors for clinical study initially lay with
`the Institute of Cancer Research and their collabora-
`
`tors, ICI Pharmaceuticals (Zeneca Pharmaceuticals)
`which led to the discovery and clinical development of
`CB3717 (Fig. 2c) in 1979 and 1981, respectively [12-
`17}. Since then at least three other drug companies
`(usually with external academic collaborators) have
`been actively involved in this area. It was soon realised
`that the scope for novel chemical modifications was
`large, and that factors other than potent TS inhibition
`were generally important
`for
`the phannacological
`activity of novel compounds. For one of the companies,
`Agouron Pharmaceuticals, the approach to the design
`of novel chemical entities was somewhat different.
`Their starting point was the X-ray crystal structure of
`bacterial TS co—crystallised with FdUMP and CB3717
`[18]. These different approaches have led to an interest-
`ing diversity in chemical structures and biochemical
`profiles. A second reason why elucidation of the useful-
`ness of TS as a target may be forthcoming, is the wealth
`of knowledge available on the mechanisms involved in
`novel compound activity and potential mechanisms by
`which cells may be, or become, resistant. Much of this
`has been the direct result of the large amount of fun-
`damental research into antifolates and fluorinated pyri-
`midines that has been performed by academic organi-
`sations over several decades. Undoubtedly this has
`aided ‘fast-track’, pre-clinical development of new
`folatc-based TS inhibitors and specific examples will be
`highlighted below.
`
`CB3717: The first clinically evaluated folate-based TS
`inhibitor
`
`CB 3717, the first folate-based TS inhibitor to be devel-
`oped clinically, was developed following the discovery
`that certain 2-amino-4-hydroxy quinazoline analogues
`of folic acid both inhibited TS and had antitumour
`
`Lilly Ex. 2073
`Sandoz V. Lilly IPR2016-00318
`
`Lilly Ex. 2073
`Sandoz v. Lilly IPR2016-00318
`
`

`
`NH;
`
`H
`

`
`F
`
`H2NJ\\N N’
`23. MTX
`
`O H
`
`'
`
`coo“
`
`°J\'.'
`
`I
`
`0
`
`HN
`
`.,.x~. 0
`
`2h. BW 1B43U89
`
`Fig. 2. Structures of some folate-based thymidylatc synthase inhibitors.
`
`activity [16]. The observations both of clinical activity
`and toxicity of CB3717 (Fig. 2c) are salient to the
`development of subsequent analogues and are outlined
`in the context of knowledge of TS inhibitors. The
`potency of CB3717 as a TS inhibitor (Ki ~3 nM) was
`central to its further development as it represented a
`major advance in the search for a specific, potent,
`folate-based inhibitor of this enzyme [12, 19, 20].
`CB3717 was shown to have slightly better folylpoly-
`glutamate synthetase (FPGS) substrate activity than
`MTX and polyglutamate metabolites were formed
`intracellularly [20,21]. These polyglutamates, particu-
`larly those of higher chain length, were shown to be ex-
`quisitely potent TS inhibitors with estimated Ki values
`of ~30 pM [22,23]. Furthermore,
`these polygluta-
`mates were retained inside cells [21]. Later, the rate of
`this polyglutamation was demonstrated to be relatively
`slow compared with certain other quinazoline ana-
`logues, a property assigned to the poor affinity of
`CB3717 for the reduced-folate cell membrane carrier
`
`(RFC) and its moderate affinity for FPGS [24, 25].
`Although many of the finer points of CB3717 prop-
`erties were unknown at the time of its early develop-
`ment, it represented a major advance in this field. The
`problems of testing inhibitors of TS in rodent anti-
`tumour and toxicity models were beginning to be re-
`alised (see below). Nevertheless CB3717 had antitu-
`mour activity attributable to TS inhibition and it was
`therefore advanced to phase I clinical study. At this
`time the development of CB3717 became a joint ven-
`ture with [Cl Pharmaceuticals and a phase I study com-
`
`menced in 1981 at the Royal Marsden Hospital/lnsti-
`tute of Cancer Research. Antitumour activity was
`demonstrated in this and further phase I/ll studies,
`particularly in breast,
`liver and platinum refractory
`ovarian cancer [13, 17, 24, 26, 27]. No activity was
`demonstrated in colorectal cancer [28]. Transient rises
`in liver transaminases were noted but, more seriously,
`sporadic life-threatening toxicity consisting of nephro-
`toxicity coupled with myelosuppression was seen (un-
`published data). A decision was therefore made that,
`although the concept of TS as an antitumour target was
`a good one, CB3717 itself would not be developed
`further. However, an ongoing biological and chemical
`synthetic programme at the Institute of Cancer Re-
`search had identified the poor aqueous solubility of
`CB3717 at acid pH as the cause of the nephrotoxicity,
`and had designed and synthesised a more water-soluble
`analogue (desamino-CB3717) devoid of this toxicity in
`mice [20, 29]. The discovery of desamino-CB3717 was
`the impetus for a new and very vigorous collaboration
`between the Institute of Cancer Research and ICI Phar-
`
`maceuticals which led to the synthesis and evaluation
`of >3,000 quinazoline analogues, most of which had
`"IS as the intracellular locus of action. The compounds
`fall into four broad classes, each with distinct intra-
`cellular biochemical and phannacological properties.
`These properties include different interactions with the
`RFC and FPGS. ZD1694 (Tomudex; in clinical devel-
`opment), ZD933l
`(in preclinical development) and
`further
`lead compounds with different
`features
`emerged from this portfolio of compounds.
`
`Lilly Ex. 2073
`Sandoz V. Lilly IPR2016-00318
`
`Lilly Ex. 2073
`Sandoz v. Lilly IPR2016-00318
`
`

`
`874
`
`Other phannaceutical companies (and their aca-
`demic collaborators) which have actively sought folate-
`based TS inhibitors have now reached the stage of pre-
`clinical or clinical evaluation. These will be discussed in
`further detail below.
`
`Second generation analogue design
`
`The first compound to emerge from new analogue syn-
`thesis, which was widely investigated, was the 2-desa-
`mino-2-methyl-analogue of CB3717 (ICI 198583;
`MPPDF) [30]. [Cl 198583 had good water-solubility,
`no detectable nephrotoxicity in mice and improved in
`vivo antitumour potency (L1210:lCR) over CB3717
`and desamino-CB3717 [31]. This compound repre-
`sents a class of highly active compounds displaying
`high affinities for the RFC and FPGS as well as for the
`target enzyme, TS. Results indicated that although
`there was no direct correlation between TS inhibition
`
`and cytotoxic potency there was a good correlation
`between the extent of polyglutamation and cytotoxicity
`[24,25]. Furthermore, the rapid polyglutamation of
`some analogues and the increased potency of the poly-
`glutamate metabolites towards TS result in their cel-
`lular pharrnacokinetics/dynamics being different from
`those of the DHFR inhibitor, MTX. Another feature of
`this class of compound is their activity by bolus admini-
`stration in mice, not necessarily predicted from their
`rapid plasma clearance, but consistent with a high
`degree of polyglutamate formation [20,31]. Although
`ease of administration and cytotoxic potency were con-
`siderations that favoured the development of this class
`of TS inhibitor, a positive decision was made to develop
`a drug that took advantage of the fact that FPGS was
`reported to be expressed relatively highly in some
`tumours and believed to explain some of the selective
`activity of MTX in mice ([32,33] and reviewed in [1]).
`A more recent report by Rumberger et al. endorses this
`further [34].
`A general theme in the synthesis and development of
`TS inhibitors, whatever their structural class (several of
`which are described later) is the elegant manner with
`which they can be evaluated in vitro because of the
`array of biological models and assays developed
`[2 5, 35]. However, it is their in vivo evaluation that pro-
`vides the most challenge to their development as drugs.
`One major problem is the high plasma thymidine
`(dThd) level in rodents relative to man (at least 10-fold
`higher) which, through the activity of the dThd salvage
`pathway, is able largely to circumvent any TS inhibition
`[36]. This accounts for the generally low activity of TS
`inhibitors in many mouse tumours and human tumour
`xenografts. Similarly their toxicity to normal proliferat-
`ing tissues is low, particularly in non-chronic admin-
`istration schedules, and therefore not necessarily pre-
`dictive of that in man. Test systems were introduced
`that at least, in part, address their in vivo activity. In-
`hibition of TS by polyglutamatable species in tumour
`
`cells that have been removed at time intervals after
`
`drug injection can serve as a pharmacodynarnic meas-
`urement [20, 31, 37]. Thymidine kinase deficient tu-
`mours such as the L5178Y TK-/- mouse lymphoma
`generally respond to single bolus therapy (polygluta-
`matable compounds) but normal proliferating tissues
`remain unaffected [38,39]. TK competent tumours,
`such as the L5178Y TK+/- [38,39], the L1210:ICR
`leukemia [20,31,37] or some human tumour xeno-
`grafts ]39,40], may be sensitive to TS inhibitors if
`administered for prolonged periods. This is probably,
`in part, the result of a fall in plasma dThd that occurs
`after administration of a TS inhibitor ([36] and unpub-
`lished data). Prolonged administration also gives drug-
`induced nonnal tissue toxicity. Taking these problems
`into account, it emerged that one compound had a bet-
`ter therapeutic index than the others in a short-list of
`active compounds (manuscript in preparation). This
`compound was ZD1694 (lCl D1694; Tomudex; Fig.
`2d) and was selected for pre-clinical and clinical devel-
`opment. The model systems used in the development
`of the other TS inhibitors in clinical study will be dealt
`with under the appropriate section.
`
`ZD1694 (Tomudex), a highly polyglutamatable TS
`inhibitor
`
`ZD1694 (Fig. 2d) has a Ki for isolated mouse and
`human TS of ~60 nM [37,41]. This 20-fold loss in
`inhibition compared with CB3717 is compensated for
`by a ~500-fold increase in cytotoxic potency [37, 42].
`This latter activity was prevented by co-incubation with
`dThd alone indicating that TS is the target enzyme for
`ZD1694 [37,42]. ZD1694 is internalised into cells via
`the RFC (Ki for the inhibition of MTX influx -2 p.M)
`and has a very low Km for mouse liver and human
`FPGS (1.3 p.M) [25, 37, 43]. The corresponding values
`for CB3717 are -40 pM for both proteins and for
`MTX are ~4 and —-166 p.M for the RFC and FPGS,
`respectively [21,25]. ZD1694 is almost completely
`metabolised to polyglutamate forms (tetra and penta-
`glutamates usually predominate) in a variety of tumour
`cells in culture [37, 44] and in certain normal tissues in
`mice such as liver, kidney and gut epithelium [42]. The
`higher polyglutamate forms (triglu and above) have Ki
`values for TS of ~1 nM and are not readily effluxed
`from the cell [37,41]. lt is concluded that ZD1694 is
`active as the polyglutamate metabolites and indeed
`structural modifications that enhance TS inhibition but
`
`largely prevent polyglutamation (for example C7
`methylation), give compounds that are -100-fold less
`potent as antitumour agents
`[24]. Furthennore a
`mechanism of acquired resistance to ZD 1694 is defec-
`tive polyglutamation (at least partly due to decreased
`FPGS activity) which illustrates the importance of this
`metabolising enzyme for drug activity [45]. The con-
`sequence of the formation of slowly effluxable polyglu-
`tamates is that short incubation periods with ZD1694
`
`Lilly Ex. 2073
`Sandoz V. Lilly IPR2016-00318
`
`Lilly Ex. 2073
`Sandoz v. Lilly IPR2016-00318
`
`

`
`(e.g. 4 h) will give a high level of cytotoxicity which is
`not seen with analogues that carmot be polyglutamated
`[25]. Indeed TTP pools in tumour cells deplete rapidly
`during incubation with the drug and recover very slow-
`ly after resuspension of cells into drug-free medium
`[46].
`After a single i.p. bolus injection of ZD1694 to mice,
`rapid clearance of the drug from the plasma occurs
`(t./B of ~30 min) [47]. A third, much slower phase of
`elrinination is then measured, giving a persistently low,
`but possibly significant, drug level. Some of the anti-
`tumour and toxicological properties of ZD1694 are to
`be found in the literature and will be the subject of
`further communications so that only a very brief sum-
`mary is given here [39, 42, 47-49]. The problems of
`testing TS inhibitors in rodents (high dThd) means that
`very little useful data can be gained as to their thera-
`peutic index and the dose and schedule applicable to
`humans. Nevertheless pharmacodynamic measure-
`ments, such as inhibition of TS in tumour cells in vivo,
`and the use of a TK deficient tumour (L5178Y TK-)
`demonstrated that a single bolus injection of ~ 10 mg/
`kg was very active, consistent with the formation of
`highly retained polyglutamates [37, 39, 50]. Repeat
`daily dosing in TK competent tumour-bearing mice
`gave some antitumour activity (including human tu-
`mour xenografts) and normal tissue toxicity (mainly
`gastrointestinal) [37, 39, 40, 48]. Co-administration of
`dThd prevented the antitumour activity (L1210:lCR)
`and toxicity of ZD1694 which confirmed that, in vivo,
`T8 was the antitumour locus of action and toxicity was
`mechanism related [37]. Consistent with this was the
`fact
`that
`the non-mechanism related nephrotoxicity
`associated with the original compound, CB3717, was
`not observed [39,48]. Antitumour activity in mice was
`also prevented by co-administration of LV which was
`explained by a series of in vitro experiments demon-
`strating competition for cellular uptake and polygluta-
`mation [24, 37].
`The clinical evaluation of ZD1694 began in Europe
`in February 1991, recruitment being principally at the
`Royal Marsden Hospital/Institute of Cancer Research,
`one of the institutes of the co-discovers of the drug [17,
`49, 51]. The starting dose was 0.1 mg/m2 (15 min i.v.
`infusion) every three weeks, which was predicted to be
`a safe starting dose from studies in dogs (data of Zene-
`ca Pharmaceuticals). Dose escalation continued up to
`3.5 mg/m2 and included 61 patients with a range of
`solid tumours
`[51]. Dose-limiting toxicities were
`malaise, gastrointestinal and haematological (leuco-
`penia or thrombocytopenia). Other toxicities which
`were encountered included reversible rises in liver
`
`transaminases, skin rash and anorexia. Three objective
`partial responses were seen (2.6 and 3 mg/m’) in pre-
`viously treated patients with ovarian and breast cancer
`and adenocarcinoma of unknown origin [51]. Clear-
`ance from the plasma was triphasic and the half lives
`for the beta and gamma phases were ~2 and 75 h,
`respectively [51]. No relationship was observed
`
`875
`
`between various pharmacokinetic parameters and re-
`sponse or toxicity. A dose of 3 mg/m’ was recommend-
`ed as the phase 1] dose. Another phase I was per-
`formed by the NCI in the U.S.A. (using the same proto-
`col but in a mainly pre-treated colorectal patient popu-
`lation (76%)), the results of which led to their recom-
`mendation of a phase II dose of 4 mg/ml [52]. How-
`ever the first phase II studies were performed at 3 mg/
`m2 and the general experience seen in the phase I was
`reproduced in these multicentred trials. Objective re-
`sponses (CR and PR) were seen at interim analysis in
`breast (25%), platinum resistant ovarian (8.5%), non-
`small cell lung (10%), and pancreatic carcinomas (14%)
`[53]. The most exciting response rate was seen in colo-
`rectal cancer where, in a study of 176 patients with ad-
`vanced disease, 26% (95% C1. 19°/o—33°/o) had objec-
`tive responses which included 4 complete and 41 par-
`tial responses [54]. A further 30% of patients had
`minor responses. The median time to progression was
`'18 weeks and median survival was 42 weeks. Grade 1]]
`
`and IV toxicities included asthenia (12%), nausea and
`vomiting (11%), diarrhoea (10%) and leucopenia (6%).
`Overall, toxicity was considered acceptable and man-
`ageable. The colorectal response rate is comparable to
`many of the reported FU/LV studies (23%—30%).
`These facts, taken together with the relative ease of
`ZD1694 administration, encouraged the iriitiation of
`the European phase II] study in 1993 (3 mg/m2, 15
`min infusion every three weeks), randomised against
`FU and LV given as 5 daily iv. bolus injections (425
`mg/m2 FU, 20 mg/m2 LV) repeated weeks 4, 8 and
`then 5 weekly (Mayo Regimen). The first results of this
`study (439 patients) at a median follow-up of 5.3
`months are now published [55]. 20% of all patients
`receiving ZD1694, compared with 13% receiving FU/
`LV, had objective partial or complete responses
`(p = 0.059; odds ratio 1.7). In addition, a further 9.4%
`of patients receiving ZD1694 had a 40%—50% reduc-
`tion in measurable lesion size (3% in FU/LV group).
`There was less grade [I] and IV leucopenia and muco-
`sitis in the ZD1694 arm of the study (p<0.001).
`Asymptomatic and reversible rises in transarninases
`were seen more frequently in patients
`receiving
`ZD1694. Overall the authors concluded that ZD1694
`
`compares favourably with FU/LV, with relatively good
`antitumour activity and an acceptable toxicity profile.
`
`LY231514, a pyn-olopyrimidine folate-based TS
`inhibitor
`
`LY231514, a pyrrolopyrimidine folate-based TS inhibi-
`tor (Fig. 2e) serendipitously arose from a chemical pro-
`gramme at Princeton University synthesising analogues
`of DDAT1-IF (Lometrexol), an inhibitor of GAR trans-
`forrnylase [56]. LY231514, further developed by Eli
`Lilly, is a relatively poor inhibitor of isolated recom-
`binant human TS (Ki - 0.34 p.M) but is highly cyto-
`toxic to cultured CCRF-CEM human or mouse leuke-
`
`Lilly Ex. 2073
`Sandoz V. Lilly IPR2016-00318
`
`Lilly Ex. 2073
`Sandoz v. Lilly IPR2016-00318
`
`

`
`876
`
`mia cells (20 nM) [56]. This seems to be due to its very
`efficient metabolism to polyglutarnates (mouse liver
`FPGS Km = 0.8 nM) with increased TS inhibitory
`potency (pentaglu estimated to be 3.4 nM assuming
`competitive inhibition) [5 6]. Thymidine reversed most
`of the cytotoxic activity, the degree depending on the
`cell line used, which suggests that "I8 is probably the
`primary locus of action although other some other
`folate target may also be involved [56]. In many re-
`spects this agent has biochemical properties similar to
`those of ZD 1694; although less active than ZD1694 as
`an inhibitor of isolated TS it is only marginally less
`active as an inhibitor of cell growth (~ 3-fold; L1210).
`Antitumour studies in vivo demonstrated complete
`inhibition of the growth of a TK- and HX- murine
`lymphoma model (L5178Y TK-/HX-) when admin-
`istered in an i.p. daily X 8 schedule of 12.5 mg/kg
`[56, 57]. Myelosuppression and gastrointestinal toxicity
`were dose-lirniting. LY231514 was also active against
`two human colon tumour xenografts (VRC5 and
`HXGC3) with > 80% tumour inhibition being observed
`at 25 and 50 mg/kg i.p. daily x 10 [56].
`A phase I clinical study (10 min infusion, weekly for
`4 weeks repeated every 42 days) started at 10 mg/m 1/
`week and escalated to the MTD of 40 mg/m2 with neu-
`tropenia being dose-lirniting [58]. Other toxicities in-
`cluded mild fatigue, anorexia and nausea. No objective
`responses were seen in the 24 patients treated. Another
`schedule investigated was a 10 min infusion daily for 5
`days repeated every 21 days [59]. At the starting dose
`of 0.2 mg/m’ some gastrointestinal toxicity was ob-
`served and dose-escalation continued. Another sched-
`
`ule recently reported was a 10 min infusion every 21
`days. An MTD was determined of 600 mg/m’ [60].
`Neutropenia was the dose-limiting toxicity and minor
`responses were observed. Phase 11 studies in colorectal
`cancer are planned I60].
`
`AG331 and AG337, lipophilic TS inhibitors in clinical
`study
`
`Agouron Pharmaceuticals, in their search for lipophilic
`TS inhibitors that interact with the folate cofactor bind-
`
`ing site, have developed two structurally dissimilar
`compounds for clinical study. The potential advantages
`of such inhibitors are their lack of a requirement for
`active uptake into cells or for polyglutamation. While
`these two processes may contribute to the antitumour
`selectivity of classical antifolates in tumours expressing
`high levels of the relevant proteins, they are also impli-
`cated in mechanisms of resistance (i.e., in tumours ex-
`pressing low levels). As a single protein interaction is
`required for their activity i.e. the target enzyme TS, the
`adopted drug design strategy was to use high resolution
`ternary crystal complexes of E. coli TS (originally with
`FdUMP and CB3717) to suggest compounds that may
`have a high binding affinity for T8. An iterative cycle of
`compound synthesis, modelling, and with certain lead
`
`compounds, new crystal complexes led to potent TS
`inhibitors with in vitro growth inhibitory potency
`similar to CB3717 (-1 nM). AG337 (compound 21 in
`[18]) is a quinazoline structure linked through C5 to a
`4-pyridylthio moiety (Fig. 2f) that was designed to
`access a hydrophobic cavity in the active site of the
`enzyme that nonnally associates with the para-amino-
`benzoyl portion of CB3717 or of the natural substrate,
`Cl-I,FH,. The reported Ki for E. coli TS is 49 nM and
`for recombinant human T8 16 nM. Curative anti-
`
`tumour activity was observed in the mouse i.m. or i.p.
`L5178Y TK- lymphoma (i.p. or oral drug administra-
`tion) and significant growth delay was seen against the
`human GC,M/TK- colon tumour xenograft [61].
`AG331 (Fig. 2g; compound 27 in [62]) is structural-
`ly unrelated to the folate cofactor and interesting
`because more conservative studies into structure-activ-
`
`ity relationships would be unlikely to reveal such a
`compound. The Ki for E. coli TS is 1800 nM and for
`human recombinant TS is 2 nM [62]. No explanation is
`provided as to why these values are so different for the
`'18 of different sources. Tumour cell growth inhibition
`(lC5o) falls in the 0.5-1 uM range. AG331 may have a
`second locus in some hepatoma cell lines which is
`hypothesised to be due to metabolism to another drug
`species [63]. Antitumour activity was observed against
`a mouse L5178Y TK- variant implanted i.p. in mice
`[64].
`Clinical studies are in progress for both drugs. A
`pharrnacokinetic and pharrnacodynamic
`study of
`AG337 (dihydrochloride fonn) given as a 24-h con-
`tinuous i.v. infusion (every three weeks) has been re-
`ported [65]. Rapid plasma clearance (t./I - <2 h) was
`observed once the infusion ceased. A dose of 1.35
`
`g/m’ was reached with no antiproliferative toxicity
`being observed [65]. However, plasma deoxyuridine
`(dUrd) was monitored and found to rise during the
`infusion (result of the elevation in dUMP that occurs
`when TS is inhibited) but normalising once the infusion
`was stopped. The rise was not dose-related above 900
`mg/m’. Antitumour studies in vitro and in vivo sug-
`gested that >24 h exposure was required to induce a
`significant cytotoxic effect [66].
`Five days of chronic dosing (75 mg/m’ 4 hourly)
`gave a median growth delay of 14 days against the Hela
`Bu25Tl(— human cervical cell line implanted in nude
`mice [66], while it was inactive when given over a
`shorter time period. This evidence led to the conduct
`of a phase I study of 5-day continuous i.v. infusion. A
`maximum tolerated dose of 1130 mg/m’/day has been
`established with dose limiting toxicities of myelosup-
`pression and mucositis [67]. The dose recommended
`for phase [1 studies is 1000 mg/mi/day.
`Preclinical studies with AG331 indicated that the
`
`plasma half life was considerably longer in dogs (13 h)
`than in rodents (2-4 h) [68]. This was confirmed as
`-20 h in humans in the phase I study. Eighteen pa-
`tients received AG331 (glucuronate salt) at doses es-
`calated between 12.5 and 225 mg/m2 (10 min i.v. infu-
`
`Lilly Ex. 2073
`Sandoz V. Lilly IPR2016-00318
`
`Lilly Ex. 2073
`Sandoz v. Lilly IPR2016-00318
`
`

`
`sion) with side—effects being observed at >130 mg/m2
`(moderate nausea and vomiting) [68]. At the highest
`dose given some mild flushing was observed which is
`believed to be due to histamine release. A similar effect
`
`was observed prec1inically.A phase I study involving a
`5-day continuous infusion has now been reported (25-
`800 mg/mz/day) [69]. Elevated liver function tests
`were evident by day 3 but were not dose-limiting. Phar-
`macokinetics suggested saturable clearance. Phase II
`studies are planned.
`
`BW1843U89, a benzoquinazoline in clinical study
`
`The Wellcome Research Laboratories have developed
`this benzoquinazoline TS inhibitor for clinical study.
`Their drug development programme originally concen-
`trated on the synthesis of lipophilic benzoquinazolines
`[70] lacking the para-aminobenzoyl glutamate chain of
`the natural
`folates, or certain antifolates such as
`CB3717 or ZD1694. Poor water-solubility and low ac-
`tivity of such analogues led to the re-introduction of
`this side chain, which in the case of BW1843U89
`(Fig. 2h), is an isoindolinone modified glutamate. This
`compound is the most potent TS inhibitor described i

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