throbber
Section 4
`Cancer and Infectious Diseases
`
`Editor: Jacob J. Plattner
`Anacor Pharmaceuticals
`Palo Alto
`California
`
`Sandoz Inc. IPR2016-00318
`Sandoz v. Eli Lilly, Exhibit 1080-0001
`
`

`
`Sandoz Inc. IPR2016-00318
`Sandoz Inc. IPR2016-00318
`Sandoz v. Eli Lilly, Exhibit 1080-0002
`Sandoz V. Eli Lilly, Exhibit 1080-0002
`
`

`
`Recent Advances in Antimetabolite
`Cancer Chemotherapies
`
`James R. Henry and Mary M. Mader
`
`Lilly Research Laboratories, Indianapolis, IN 46285, USA
`
`Contents
`1. Introduction
`2. Mechanisms of antimetabolite cancer chemotherapy
`2.1. Thymidine biosynthesis
`2.2. Purine nucleotide synthesis
`2.3. Ribonucleoside reductase
`2.4. Intracellular transformations of antimetabolites
`3. Antimetabolites
`4. Clinical advances with combinations of antimetabolites
`5. Conclusions
`References
`
`1. INTRODUCTION
`
`161
`161
`161
`162
`163
`164
`166
`168
`170
`170
`
`An antimetabolite interferes with the formation or utilization of a normal cellular
`metabolite. Most antimetabolites interfere with the enzymes involved in the synthesis of
`new DNA, are incorporated into the newly formed DNA, or in some cases both processes
`are important to an agent's efficacy. As a result, many antimetabolites are derivatives of
`the building blocks of DNA itself, such as the nucleoside based inhibitors, or analogs of
`critical cofactors such as the antifolates. A variety of key cellular pathways have been
`disrupted with antimetabolite therapy, including inhibition of the thymidine and purine
`nucleotide biosynthesis pathway, and the inhibition of ribonucleoside reductase. Given
`their mechanism of action, it is not surprising that the observed benefits of
`antimetabolites are often accompanied by significant toxicity, due to the fact that the
`affected cellular metabolites are critical to both normal and cancer cells. Single
`antimetabolite agents can act on a single pathway, or on multiple pathways at once, but in
`either instance, they are often used in combination with other therapies in the clinic.
`
`2. MECHANISMS OF ANTIMETABOLITE CANCER
`CHEMOTHERAPY
`
`2.1. Thymidine biosynthesis
`
`Critical to the cell's process of replication is its ability to synthesize thymidine.
`This process involves several key enzymes including thymidylate synthase (TS),
`
`ANNUAL REPORTS IN MEDICINAL CHEMISTRY. VOLUME 39
`ISSN: 0065-7743 DO! IO.IOI6/S0065-7743(04)39013-5
`
`© 2004 Elsevier Inc.
`Al! rights reserved
`
`Sandoz Inc. IPR2016-00318
`Sandoz v. Eli Lilly, Exhibit 1080-0003
`
`

`
`162
`
`J.R. Henry and M.M. Mader
`
`HN:SM'
`oAN
`
`o
`
`I
`R
`
`dTMP
`
`N5,N10-CHr THF
`
`Glycine ....J SHMT
`
`7,8-DHF
`
`NADPH+H+
`
`DHFR
`
`PLP ) - - THF : {
`
`Serine
`
`NADP+
`
`o
`II
`HO-7-o~0"-lBase
`OH ~
`
`R'
`
`OH
`
`Fig. 1. Key steps in thymidine biosynthesis.
`
`dihydrofolate reductase (DHFR), and serine hydroxymethyl transferase (SHMT)
`(Fig. 1).
`The methylation of deoxyuridine 5' monophosphate (dUMP) to produce deoxythymi(cid:173)
`dine 5' monophosphate (dTMP) is mediated by TS [1]. The methyl group for dTMP is
`provided by N5,N1O-methylene tetrahydrofolate (N5,N1O-CHrTHF) through its
`conversion to 7,8-dihydrofolate (7,8-DHF). The 7,8-DHF must then be converted to
`tetrahydrofolate (THF) by DHFR [2], followed by further transformation back to N5,N1O(cid:173)
`CHr THF through the action of SHMT [3]. Therefore, inhibition of TS, DHFR, or SHMT
`with an appropriate agent would interrupt the process of thymidine biosynthesis. Low
`thymidine levels cause defects in DNA which in turn activates stress response elements,
`such as the Fas ligand/Fas death pathway leading to apoptosis [4]. It has also been
`proposed that defects in this Fas-dependent apoptotic signaling pathway are one cause of
`cellular resistance to drugs.
`
`2.2. Purine nucleotide synthesis
`
`The cell's ability to provide the needed purine nucleotides for DNA and RNA synthesis is
`also critical to its survival. The de novo biosynthesis of purine nucleotides involves 10
`
`Sandoz Inc. IPR2016-00318
`Sandoz v. Eli Lilly, Exhibit 1080-0004
`
`

`
`Recent Advances in Antimetabolite Cancer Chemotherapies
`
`163
`
`AICAR
`
`Inosinic acid
`
`AMP
`
`GMP
`
`10-CHO-THF
`
`THF
`
`GR
`
`formylglycinamide
`ribon ucleotide
`
`Fig. 2. Key step in purine biosynthesis.
`
`separate enzyme catalyzed reactions starting with 5-phosphoribosyl-l-pyrophosphate
`and leading to inosinic acid [5]. Both adenosine monophosphate (AMP) and guanine
`monophosphate (GMP) are then derived from inosinic acid (Fig. 2). The third step in this
`process is the biosynthesis of formylglycinamide ribonucleotide from glycinamide
`ribonucleotide (GR) via glycinamide ribonucleotide formyltransferase (GARFT). The
`last two steps in the synthesis of inosinic acid occur via a bifunctional enzyme having
`both aminoimidazolecarboxamide ribonucleotide formyltransferase (AlCARFT) and
`inosine monophosphate cyclohydrolase (IMPCH) activity. This enzyme has been shown
`to be made up of a 39 kDa carboxy-terminal AICARFT active fragment along with a
`25 kDa amino-terminal IMPCH active fragment [6]. Both GARFT and AICARFT
`catalyze the transfer of a formyl group from lO-CHO-tetrahydrofolate (lO-CHO-THF) to
`GR or aminoimidazolecarboxamide ribonucleotide (AICAR) respectively, returning
`THF as the second product of the reaction.
`
`2.3. Ribonucleoside reductase
`
`The synthesis of new DNA within a cell requires the production of deoxynucleotides.
`The four required deoxynucleotides (adenosine, guanosine, cytidine, and thymidine) are
`produced as by reduction of the appropriate ribonucleotide substrate with ribonucleoside
`reductase, also referred to a nucleoside diphosphate reductase (NDPR) [7]. The resulting
`oxidized form of NDPR can then be reduced back to NDPR by the action of
`glutaredoxin, which is in tnrn oxidized to thioredoxin [8]. NDPR is a dimer, with each
`monomer made up of two subunits: a larger (Ml) and a smaller (M2) subunit. The Ml
`subunit contains two allosteric sites involved in regulation of the overall activity of the
`enzyme and the enzyme's substrate specificity. The deoxynucleoside triphosphates bind
`to this allosteric site, and regulate their own synthesis. The M2 subunit is responsible for
`the key reduction reaction, carrying a tightly bound iron atom that stabilizes the tyrosyl
`free radical critical to reduction. Deoxynucleotide pools in proliferating cells are
`
`Sandoz Inc. IPR2016-00318
`Sandoz v. Eli Lilly, Exhibit 1080-0005
`
`

`
`164
`
`J.R. Henry and M.M. Mader
`
`sufficient for only a few minutes of DNA synthesis without regeneration, thus making
`NDPR inhibition an attractive candidate for cancer chemotherapy [9].
`o
`II
`
`Ho-ro-rOtt
`
`0
`II
`
`o
`0
`II
`II
`HO -r~o-r~O~o"se
`)-l
`
`NDPR
`-------I.~
`
`HO
`
`OH
`
`HO
`
`2.4. Intracellular transformations of antimetabolites
`
`Most antimetabolites must undergo modification within the cell before they are active
`agents, and so in essence are prodrugs. Methotrexate (MTX, 1), an antifolate agent
`targeting DHFR, and pemetrexed (Alimta, 2), a multi-targeted antifolate, exert much
`of their pharmacological effects as a polyglutamate, as do most classical antifolates
`[10]. This transformation is carried out by the enzyme folylpolyglutamate synthetase
`(FPGS). Formation of the polyglutamate of antifolate drugs can cause a dramatic
`increase in the activity of the agent toward its intended target. Further, polyglutamates
`(above diglutamate) are less susceptible to cellular efflux, thus providing a long-lived
`pool of drug within the cell [11]. Most natural folate cofactors exist as polyglutamates,
`and so the beneficial action of FPGS on antifolate drugs is not surprising. It also
`follows that any cellular change leading to decreased FPGS activity could lead to
`antifolate resistance.
`
`o
`
`C02H
`
`N~C02H
`
`C02Na
`N~C02Na
`
`H
`
`/
`
`H
`
`"
`
`site of polyglutamate
`formation
`
`Many nucleoside derived antimetabolite analogs also undergo intracellular trans(cid:173)
`formations to become active agents (Fig. 3). The earliest of these agents, 5-fluorouracil
`(5-FU, 3) is converted into three major metabolites that are responsible for its activity.
`5-FU is changed into 5-fluoro-2'-deoxyuridine monophosphate (FdUMP, 4) that acts as a
`mimic for the natnral substrate of TS, dUMP, thus inhibiting TS activity. 5-FU can also
`be tranformed to 5-fluoro-2'-deoxyuridine triphosphate, which is eventually incorporated
`into DNA causing DNA damage, and finally to 5-fluorouridine triphosphate (5), which is
`incorporated into RNA leading to impaired RNA function [12]. Deoxyribonucleoside
`
`Sandoz Inc. IPR2016-00318
`Sandoz v. Eli Lilly, Exhibit 1080-0006
`
`

`
`Recent Advances in Antimetabolite Cancer Chemotherapies
`
`165
`
`cellular
`
`Hi
`
`:Y F
`
`o
`I metabolism.
`~
`0 :
`
`F
`
`Hi
`
`F
`
`Hi
`
`~O
`:yo
`O~N + 1. O~N
`I
`I
`
`H,opO ~ HO,P J ~~
`
`HO
`
`OH
`HO
`40 5
`
`NH2
`
`NH2
`
`HN~O~
`F~N
`lN~O
`H'C~
`
`OH
`
`HO
`
`6
`
`HO
`
`F
`
`7
`
`HO
`
`OH
`
`8
`
`Fig. 3. Metabolism of 5-FU 3 and other nucleoside antimetabolites.
`
`analogs such as cytarabine (6) and gemcitabine (7) are converted to their triphosphate
`derivatives in vivo before showing their effects [l3,14] , while capecitabine (8) is
`metabolized in vivo to 3 (Fig. 4). 8 is readily absorbed in the gastrointestinal tract, then
`passes intact through the intestinal mucosa. It is subsequently converted to 5'(cid:173)
`deoxycytidine 9 by carboxylesterase in the liver, 5'-deoxy-5-fluorouridine 10 by cytidine
`deaminase, and finally to 5-FU 3 by thymidine phosphorylase.
`
`NH-Z.
`
`o
`F~N OR
`H3CHN~O
`
`carboXYleste~se
`
`NH
`
`0
`
`H
`
`F~N' F 'n
`H3CHN~O H3CHN~O F~O
`
`cytidine
`deaminase.
`
`thymine
`phosphoryl,;,e
`
`1/
`NH
`I N~
`
`HO OH
`8
`
`HO OH
`
`9
`
`HO OH
`10
`
`0
`
`H
`3
`
`Fig. 4. Metabolism of capecitabine to 5-FU.
`
`Sandoz Inc. IPR2016-00318
`Sandoz v. Eli Lilly, Exhibit 1080-0007
`
`

`
`166
`
`J.R. Henry and M.M. Mader
`
`3. ANTIMETABOLITES
`
`Historically, antimetabolites have been useful agents in hematopoietic therapy. However,
`in recent years, they have been shown to be effective in the treatment of solid tumors
`(Table 1). Three antimetabolites have been approved for clinical use since 1996:
`gemcitabine, capecitabine and pemetrexed. A new method for the delivery of cytarabine,
`DepoCyt, was given accelerated approval in 1999. Additional oncolytic indications for
`some of the approved antimetabolites are in clinical trials, and other antimetabolites,
`including nolatrexed and decitabine, are currently undergoing Phase III study. An
`antifolate, raltitrexed, has been approved for use outside of the US, but its development as
`a single agent, as well as that of the nucleoside analog eniluracil has been discontinued in
`the USA [15,16].
`Although 5-FU is widely used, its pharmacokinetic profile is not ideal. Its optimal
`method of delivery is by continuous intravenous infusion, as its bioavailability after oral
`administration is variable. 5-FU is rapidly metabolized, with a mean half-life of
`elimination of approximately 16 min. Within 3 h, no intact drug can be detected in
`plasma. 5-FU is more effective when co-administered with leucovorin, a prodrug of
`5, lO-CHr THF. Inhibition of TS by FdUMP is dependent on the cofactor 5, lO-CHr THF,
`which combines with TS and FdUMP to form a covalent teruary complex. Excess
`cofactor decreases the dissociation rate of this complex, and consequently addition of
`
`Table 1. Selected oncolytic antimetabolites in clinical use
`
`Clinical agent
`
`Trade name
`
`Year approved
`(USA)
`
`Indication
`
`Ref.
`
`Folate Antagonists
`
`Methotrexate (MTX)
`Pemetrexed
`Nolatrexed
`Nucleoside analogs
`
`5-Fluorouracil
`Cytarabine
`Fludarabine
`Pentostatin
`Cladribine
`Gemcitabine
`
`Cytarabine
`lyposomal
`Capecitabine
`
`Decitabine
`
`Alimta®
`Thymitaq®
`
`1953, 1959, 1971
`2004
`(Phase III)
`
`Leukemia
`Mesothelioma
`Liver cancer
`
`Adrucil®
`Cytosar-U®
`Fludara®
`Nipent®
`Leustatin®
`Gemzar®
`
`DepoCyt®
`
`Xeloda®
`
`Dacogen®
`
`1962
`1969
`1991
`1991
`1993
`1996
`1998
`
`1999
`
`1998
`200l
`(Phase III)
`
`Colorectal cancer
`Leukemia
`Leukemia
`Leukemia
`Leukemia
`Pancreatic cancer
`Non-small cell
`lung cancer
`Lymphomatous
`meningitis
`Breast cancer
`Colorectal cancer
`Leukemia
`
`[18]
`[19]
`[20]
`
`[21]
`[22]
`[23]
`[24]
`[25]
`[26]
`[27]
`
`[28]
`
`[29]
`[30]
`[31]
`
`Sandoz Inc. IPR2016-00318
`Sandoz v. Eli Lilly, Exhibit 1080-0008
`
`

`
`Recent Advances in Antimetabolite Cancer Chemotherapies
`
`167
`
`leucovorin increases the cytotoxicity of 5-FU. The major toxicities of 5-FU are to bone
`marrow and mucous membranes [17].
`Capecitabine (8) is an orally administered fluoropyrimidine carbamate that is
`metabolized in vivo to 5-FU (3) [32] . Pharmacokinetic studies in patients showed rapid
`gastrointestinal absorption of capecitabine, followed by extensive conversion to 10, with
`only low systemic levels of 5-FU [33]. Significantly, capecitabine appears to deliver drug
`selectively to tumors. Analysis of the tumor:plasma AUC ratios of capecitabine vs. 5-FU
`in four human tumor xenografts in mice (HCTlI6, CXF280, COL0205, and WiDr) at the
`maximum tolerated dose (p.o.) showed that although the half-life of 5-FU was similar in
`all four tumors, the tumor:plasma AUC ratio of 5-FU was significantly higher for animals
`dosed with capecitabine. For example, in HCTI16, 5-FU exposure was 127-fold higher in
`tumor than plasma in animals treated with capecitabine, and 209-fold higher in CXF280
`[34]. In the clinic, the efficacy of capecitabine equals or exceeds 5-FU, although its
`pharmacology in the murine models might have predicted consistently superior efficacy.
`Capecitabine is approved for the treatment of breast and colorectal cancer, and its ease of
`dosing by mouth is attractive to patients.
`Gemcitabine (7) is a nucleoside analog that exhibits cell phase specificity, primarily
`killing cells undergoing DNA synthesis (S-phase) and also blocking the progression of
`cells through the Gl/S-phase boundary. The cytotoxic effect of gemcitabine is attributed
`to a combination of two actions of the diphosphate and the triphosphate nucleosides,
`which leads to inhibition of DNA synthesis. First, gemcitabine diphosphate inhibits
`NDPR, which causes a reduction in the concentrations of deoxynucleotides, including
`deoxycytidine triphosphate (dCTP) [35] . Second, gemcitabine triphosphate competes
`with dCTP for incorporation into DNA. The reduction in the intracellular concentration
`of dCTP (by the action of the diphosphate) enhances the incorporation of gemcitabine
`triphosphate into DNA (self-potentiation). After the gemcitabine nucleotide is
`incorporated into DNA, only one additional nucleotide is added to the growing DNA
`strands. After this addition, there is inhibition of further DNA synthesis. Because of the
`addition of this final nucleotide, DNA polymerase epsilon is unable to remove the
`gemcitabine nucleotide and repair the growing DNA strands (masked chain termination).
`First approved by the FDA in 1996, 7 was demonstrated to have a significant clinical
`benefit response in advanced pancreatic cancer patients compared to 5-FU, with a
`survival advantage of 5.6 months vs. 4.4 months in the 5-FU-treated patients [26]. The
`clinical benefit was measured as improvement in three symptoms present in most
`pancreatic cancer patients: pain, functional impairment and weight loss. Gemcitabine has
`become accepted as the standard of care for the treatment of advanced pancreatic cancer,
`and in 1998 the FDA approved its combination with cisplatin for the treatment of non(cid:173)
`small cell lung cancer (NSCLC).
`DepoCyt is an injectable, sustained release form of cytarabine (ara-C; 6), for the
`treatment of antineoplastic meningitis (NM) arising from lymphoma (lymphomatous
`meningitis) [28]. Ara-C acts by inhibiting DNA polymerase as well as through
`incorporation of its triphosphate into DNA. A phase III trial of DepoCyt in
`lymphomatous NM showed it to be more convenient and associated with a higher
`positive response rate than ara-C. The DepoCyt formulation of ara-C is encapsulated in
`the aqueous chambers of a spherical 20 fLM matrix comprised of lipids biochemically
`similar to normal human cell membranes (phospholipids, triglycerides, and cholesterol).
`
`Sandoz Inc. IPR2016-00318
`Sandoz v. Eli Lilly, Exhibit 1080-0009
`
`

`
`168
`
`J.R. Henry and M.M. Mader
`
`When injected into the cerebral spinal fluid (CSF) at room temperature, the particles
`spread throughout the neuroaxis and slowly release ara-C. A single injection of free
`unencapsulated ara-C maintains cytotoxic concentrations in the CSF for <24 h [36] ,
`whereas a single injection of 50 mg of DepoCyt maintains cytotoxic concentrations of
`ara-C in most patients in the CSF for> 14 days [37]. As cytotoxicity is a function of both
`drug concentration and duration of exposure, this formulation maintains high
`concentrations of ara-C in the cancer cell for prolonged periods of time and increases
`the efficacy of the agent.
`Pemetrexed is an antifolate that inhibits multiple folate-requiring enzymes including
`TS, DHFR, GARFT and to a lesser extent, AICARFT [38,39] . Having multiple sites of
`inhibition results in an activity profile that differs from the TS inhibitor, 5-FU, or the
`DHFR inhibitor, MTX. Folic acid and vitamin B-12 supplementation modulate
`pemetrexed's overall toxicity while enhancing its cytotoxic effects, and pre-treatment
`with folic acid is a component of the clinical regimen [40] . In a Phase II study of
`pemetrexed as a single agent in patients with malignant pleural mesothelioma (MPM),
`a 17% response rate was observed (9 of 64 patients). In combination with cisplatin,
`however, a Phase III trial found a response rate of 41.3% and a median survival of 12.3
`months [19] . The control arm of the study received cisplatin monotherapy, and the
`response rate in these patients was 16.7% with median survival of9.3 months [41]. Based
`on these findings, the pemetrexed/cisplatin combination was approved by the FDA in
`February 2004 as a treatment for MPM. Clinical trials of pemetrexed are underway as a
`therapy for solid tumors including non-small cell lung, pancreatic, metastatic breast,
`colorectal, and gastric cancers [42].
`
`4. CLINICAL ADVANCES WITH COMBINATIONS
`OF ANTIMETABOLITES
`
`Anti-cancer agents are rarely given singly, as combinations of drugs have proven to be
`far superior to single agent therapy for a variety of cancers. Antimetabolites are no
`exception; they have been combined with other antimetabolites and with other
`chemotherapeutic agents. The goal of combination therapy is to find agents whose
`activities are synergistic, i.e., a regime where the combined effect is greater than what
`would be expected from the sum of the two individual agent's activities, and have non(cid:173)
`overlapping toxicities. Since most antimetabolites interfere with the process of DNA
`synthesis or growth, many combinations with drugs that react with DNA have been used
`[43]. An illustration of this is the combination of pemetrexed with cisplatin. Other
`potential combinations would be with compounds targeted towards inducing apoptosis,
`preventing angiogenesis, or with antimetabolites targeting different enzymes in the same
`pathway. An example is the combination of cyclophosphamide (a DNA alkylating agent),
`MTX, and 5-FU. This regimen, referred to as CMF, was an early standard of care in the
`treatment of metastatic breast cancer [44]. 5-FU targets the thymine biosynthesis
`pathway by inhibiting TS, while MTX targets the same general pathway by inhibiting
`DHFR. Combinations are first studied preclinically in cellular and animal models, but
`application of the models to patients in the clinical setting is complicated by ADMET
`phenomena that are not well predicted by the models [45] . Clinical success for therapy
`
`Sandoz Inc. IPR2016-00318
`Sandoz v. Eli Lilly, Exhibit 1080-0010
`
`

`
`Recent Advances in Antimetabolite Cancer Chemotherapies
`
`169
`
`(combination or single agent) is assessed by a variety of measures relative to the standard
`of care for the indication, including toxicities, survival benefit, response rate, median
`survival, and survival to a determined point in time (e.g. 12 months). Our focus will be on
`recent clinical outcomes with combinations of antimetabolites and the biochemical
`rationale behind them.
`Thymidylate synthase inhibitors including 5-FU, capecitabine, and raltitrexed have
`been tested in the clinic in combination with gemcitabine. Gemcitabine and the TS
`inhibitors inhibit DNA and RNA synthesis by different mechanisms and possess almost
`no overlapping toxicity profiles. Gemcitabine inhibits NDPR, depleting cellular dUMP
`pools, thereby decreasing the dUMP competition with 5-FdUMP at TS. Raltitrexed
`prevents binding of the folate cofactor on TS, and also has little overlapping toxicity with
`gemcitabine. At least additive, and possibly synergistic cytotoxic effects for
`gemcitabine/5-FU were anticipated based on preclinical results with the combination
`in HT29 colon cancer cells [46] . To date, several Phase III trials in pancreatic cancer have
`found gemcitabine/5-FU regimens to be tolerable in terms of toxicities, but an optimal
`dosing schedule has yet to be found which improves the median survival of patients with
`advanced pancreatic carcinoma compared with single-agent gemcitabine [47]. A Phase II
`trial of bi-weekly high dose gemcitabine plus capecitabine in pancreatic cancer similarly
`found a good therapeutic index for the combination, but no advantage was observed
`in terms of efficacy parameters compared to mono therapy with gemcitabine [48,49] .
`The combination of gemcitabine with raltitrexed has been investigated in a Phase II trial
`involving pancreatic cancer patients, and was found to be convenient with little
`symptomatic toxicity. However, like the combinations with the nucleoside TS inhibitors,
`the dual therapy of gemcitabine with the antifolate TS inhibitor was no more efficacious
`in terms of survival benefit than single gemcitabine monotherapy [50].
`Gemcitabine has also been investigated with pemetrexed in pancreatic cancer and
`NSCLC. Pemetrexed depletes the intracellular supply of both purine and thymidine
`deoxynucleotides, while gemcitabine is incorporated into nascent DNA strands
`ultimately resulting in strand termination. Thus, the two agents together would
`interfere with DNA replication at both the nucleotide and strand synthesis level. Early
`in vitro cell assays and tumor xenograft models indicated that gemcitabine/pemetrexed
`would show synergism in vivo, but the degree of activity was dependent on cell type
`and dosing schedule [40,51,52]. Each drug is active as a single agent in pancreatic
`cancer (Table 2) [26,53] , and a Phase I trial of the gemcitabine/pemetrexed combination
`in advanced pancreatic cancer was initiated. In patients, the recommended schedule and
`doses were found to be gemcitabine on days 1 and 8 @ 1250 mg/m2 with pemetrexed
`on day 8 only @ 500 mg/m2 [51]. Phase II results for the gemcitabine/pemetrexed
`combination have been reported, with patients showing a 15% partial response rate,
`with 29% of the evaluable patients surviving for 12 months. As the three measures
`(partial response, median survival, and l-yr survival) for the combination showed
`improvement relative to therapy with gemcitabine or pemetrexed alone, a Phase III trial
`was initiated. Enrollment for the Phase III trial has concluded, and a final data analysis
`is anticipated in the coming year. The combination has demonstrated efficacy in an
`ongoing Phase II trial for NSCLC, employing the same schedule, with evidence of
`improved median survival and l-yr survival in comparison to the gemcitabine/cisplatin
`combination or single agent pemetrexed.
`
`Sandoz Inc. IPR2016-00318
`Sandoz v. Eli Lilly, Exhibit 1080-0011
`
`

`
`170
`
`J.R. Henry and M.M. Mader
`
`Table 2. Comparisons of selected anti-metabolite single agent and combination clinical
`trials
`
`Patients
`totalJeval.
`
`Ref.
`
`Treatment
`
`Agent
`
`PRa n (%)
`
`Median
`survival
`(mo)
`
`1-yr
`survival
`(%)
`
`Advanced pancreatic cancer
`
`[26]
`[53]
`[54]
`
`63/56
`42/35
`42/40
`
`Phase III
`Phase II
`Phase II
`
`Non-small cell lung cancer
`
`[55]
`
`3011288
`
`Phase III
`
`[56]
`[57]
`
`33/30
`60/54
`
`Phase II
`Phase II
`
`apR = partial response.
`b Objective response.
`
`5. CONCLUSIONS
`
`Gemcitabine
`Pemetrexed
`Gemcitabinel
`pemetrexed
`
`Gemcitabinel
`cisplatin
`Pemetrexed
`Gemcitabinel
`pemetrexed
`
`3 (5.4)
`2 (5.7)
`6 (15)
`
`60 (21)
`
`7 (23)
`9 (17)b
`
`5.7
`6.5
`6.5
`
`8.1
`
`9.2
`1l.3
`
`18
`28
`29
`
`36
`
`25
`46
`
`Much progress has been made in the last decade in the development and clinical use of
`antimetabolites as chemotherapeutics for the treatment of solid tumors. Both mono- and
`combination therapies have been found to be efficacious, and clinical trials are underway
`to determine efficacies against a greater variety of tumor types, and of regimens involving
`two, three and four-drug combinations.
`
`REFERENCES
`
`[1] D. V. Santi and P. V. Danenberg, Folates in pyrimidine nucleotide biosynthesis. in Fo/ates
`and Pterins (eds R. L. Blakely and S. J. Benkovic), Wiley, New York, 1984, p. 345.
`[2] R. L. Blakely, Dihydrofolate reductase. in Fo/ates and Pterins (eds R. L. Blakely and S. J.
`Benkovic), Wiley, New York, 1984, p. 191.
`[3] V. Schirch, Folates in serine and glycine metabolism. in Fo/ates and Pterins (eds R. L.
`Blakely and S. J. Benkovic), Wiley, New York, 1984, p. 399.
`[4] J. A. Houghton, F. G. Harwood and D. M. Tillman, Proc. Nat/. Acad. Sci., 1997,94, 8144.
`[5] P. B. Rowe, Folates in the biosynthesis and degradation of purines. in Fo/ates and Pterins (eds
`R. L. Blakely and S. J. Benkovic), Wiley, New York, 1984.
`[6] J. L. Kan and R. G. Moran, Nuci. Acids. Res., 1997,25, 3ll8.
`[7] J. G. Cory and A. H Cory (eds), Inhibitors ofRibonucieoside Diphosphate Reductase Activity,
`Pergamon, New York, 1989.
`[8] A. Holmgren, Annu. Rev. Biochem., 1985,54,237.
`[9] J. G. Cory and P. Chiba, Pharmaco/. Ther., 1985,29, lll.
`[10] L. H. Matherly, R. L. Seither and 1. D. Goldman, Pharmaco/. Ther., 1987,35,27.
`[ll] J. J. McGuire, Antifolate polyglutamation in preclinical and clinical antifolate resistance. in
`Antifo/ate Drugs in Cancer Therapy (ed. A. L. Jackman), Humana Press, Totowa, 1999, p. 339.
`
`Sandoz Inc. IPR2016-00318
`Sandoz v. Eli Lilly, Exhibit 1080-0012
`
`

`
`Recent Advances in Antimetabolite Cancer Chemotherapies
`
`171
`
`[12] J. A. Montgomery, Antimetabolites. in Cancer Chemotherapeutic Agents (ed. W. O. Foye),
`American Chemical Society, Washington, DC, 1995, p. 49.
`[13] P. P. Major, E. M. Egan, G. P. Beardsley, M. D. Minden and D. W. Kufe, Proc. Nat/. Acad.
`Sci., 1981,78, 3235.
`[14] P. Huang, S. Chubb, L. W. Hertel, G. B. Grindey and W. Plunkett, Cancer Res., 1991,
`51,6110.
`[15] D. Cunningham, J. Zalcberg, J. Maroun, R. James, S. Clarke, T. S. Maughan, M. Vincent,
`J. Schulz, M. Gonzalez Baron and T. Facchini, Eur. 1. Cancer, 2002, 38, 478.
`[16] H. L. Kindler and R. L. Schilsky, Clinical trials of the eniluracil!5-fiuorouracil combination.
`in Fluoropyrimidines in Cancer Therapy (ed. Y. M. Rustum), Humana Press, Totowa, NJ,
`2003, p. 235.
`[17] M. J. Moore and C. Erlichman, Pharmacology of anticancer drugs. in The Basic Science of
`Oncology (eds1.F. TannockandR. P. Hill), 3rd edition, McGraw-Hill,New York, 1998,p. 370.
`[18] R. Gorlick and J. R. Bertino, Clinical pharmacology and resistance to dihydrofolate reductase
`inhibitors. in Antifolate Drugs in Cancer Therapy (ed. A. L. Jackman), Humana Press,
`Totowa, NJ, 1999, p. 37.
`[19] N. J. Vogelzang, J. J. Rusthoven, J. Symanowski, C. Denham, E. Kaukel, P. Ruffie,
`U. Gatzemeier, M. Boyer, S. Emri, C. Manegold, C. Niyikiza and P. Paoletti, 1. Clin. Onco/.,
`2003, 21, 2636.
`[20] K. Stuart, J. Tessitore, J. Rudy, N. Clendennin and A. Johnston, Cancer, 1999, 86, 410.
`[21] S. Ananthan, Fluoropyrimidines. in Cancer Chemotherapeutic Agents (ed. W. O. Foye),
`American Chemical Society, Washington, DC, 1995, p. 49.
`[22] J. A. Secrist, III, 2' -Deoxyribonucleoside analogs. in Cancer Chemotherapeutic Agents
`(ed. W. O. Foye), American Chemical Society, Washington, DC, 1995, p. 71.
`[23] J. C. Adkins, D. H. Peters and A. Markham, Drugs, 1997, 53, 1005.
`[24] C. E. Dearden, E. Matutes and D. Catovsky, Semin. Onco/., 2000, 27, 22.
`[25] M. S. Tallman and D. Hakimian, Sem. Hemato/., 1996,33,23.
`[26] H. A. Burris, III, M. J. Moore, J. Andersen, M. R. Green, M. L. Rothenberg, M. R. Modiano,
`M. C. Cripps, R. K. Portenoy, A. M. Storniolo, P. Tarassoff, R. Nelson, F. A. DOff, C. D.
`Stephens and D. D. Von Hoff, 1. Clin. Onco/., 1997, 15, 2403.
`[27] A. Sandler and D. S. Ettinger, Oncologist, 1999, 4, 241.
`[28] K. A. Jaeckle, S. Phuphanich, M. J. van den Bent, R. Aiken, T. Batchelor, T. Campbell,
`D. Fulton, M. Gilbert, D. Heros, S. J. O'Day, W. Akerley, J. Allen, S. Baidas, S. Z. Gertler,
`H. S. Greenberg, S. LaFollette, G. M. Lesser, W. L. Recht, E. Wong, M. C. Chamberlain,
`A. Cohn, M. J. Glantz, J. C. Gutheil, B. Maria, P. Moots, P. New, C. Russell, W. Shapiro,
`L. Swinnen and S. B. Howell, Br. 1. Cancer, 2001, 84, 157.
`[29] P. M. Hoff, R. Ansari, G. Batist, J. Cox, W. Kocha, M. Kuperminc, J. Maroun, D. Walde,
`C. Weaver, E. Harrison, H. U. Burger, B. Osterwalder, A. O. Wong and R. Wong, 1. Clin.
`Onco/., 2001, 19, 2282.
`[30] C. Twelves, Eur. 1. Cancer, 2002, 38 (Supp!. 2), 15.
`[31] J.-P. Issa, Curro Opin. Onco/., 2003, 15,446.
`[32] N. Shimma, 1. Umeda, M. Arasaki, C. Murasaki, K. Masubuchi, Y. Kohchi, M. Miwa, M. Ura,
`N. Sawada and H. Tahara, Bioorg. Med. Chem., 2000, 8, 1697.
`[33] M. Mackean, A. Planting, C. Twelves, J. Schellens, D. Allman, B. Osterwalder, B. Reigner,
`T. Griffin, S. Kaye and J. Verweij, 1. Clin. Onco/., 1998, 16, 2977.
`[34] T. Ishikawa, M. Utoh, N. Sawada, M. Nishida, Y. Fnkase, F. Sekiguchi and H. Ishitsuka,
`Biochem. Pharmaco/., 1998,55, 1091.
`[35] C. H. Baker, J. Banzon, J. M. Bollinger, J. Stubbe, V. Samano, M. J. Robins, B. Lippert,
`E. Jarvi and R. Resvick, 1. Med. Chem., 1991,34, 1879.
`[36] S. Zimm, J. M. Collins, J. Miser, D. Chatterji and D. G. Pop lack, Clin. Pharmaco/. Ther.,
`1984, 35, 826.
`[37] S. Kim, E. Chatelut, J. C. Kim, S. B. Howell, C. Cates, P. A. Kormanik and M. C.
`Chamberlain, 1. Clin. Onco/., 1993,11,2186.
`[38] E. C. Taylor, D. Kuhnt, C. Shih, S. M. Rinzel, G. B. Grindey, J. Barredo, M. Jannatipour and
`R. G. Moran, 1. Med. Chem., 1992,35,4450.
`
`Sandoz Inc. IPR2016-00318
`Sandoz v. Eli Lilly, Exhibit 1080-0013
`
`

`
`172
`
`J.R. Henry and M.M. Mader
`
`[39] C. Shih, V. J. Chen, L. S. Gossett, S. B. Gates, W. C. Mackellar, L. L. Habeck, K. A.
`Shackelford, L. G. Mendelsohn, D. J. Soose, V. F. Patel, S. L. Andis, J. R. Bewley, E. A. Rayl,
`B. A. Moroson, G. P. Beardsley, W. Kohler, M. Ratnam and R. M. Schultz, Cancer Res., 1997,
`57,1116.
`[40] J. F. Worzalla, C. Shih and R. M. Schultz, Anticancer Res., 1998, 18, 3235.
`[41] G. V. Scagliotti, D.-M. Shin, H. L. Kindler, M. J. Vasconcelles, U. Keppler, C. Manegold, H.
`Burris, U. Gatzemeier, J. Blatter, J. T. Symanowski and J. J. Rusthoven, 1. Clin. Onco/., 2003,
`21, 1556.
`[42] L. Paz-Ares, S. Bezares, J. M. Tabernero, D. Castellanos and H. Cortes-Funes, Cancer
`(Supplement), 2003, 97, 2056.
`[43] S. P. Ackland and R. Kimbell, Antifolates in combination therapy. in Antifolate Drugs in
`Cancer Therapy (ed. A. L. Jackman), Humana Press, Totowa, NJ, 1999, p. 365.
`[44] G. Bonadonna, P. Valagussa, A. Rossi, G. Tancini, C. Brambilla, M. Zambetti and
`U. Veronesi, Breast Cancer Res. Treat., 1985,5,95.
`[45] A. L. Jackman, R. Kimbell and H. E. R. Ford, Eur. 1. Cancer, 1999, 35, S3.
`[46] L. Schulz, A. Schalhorn, W. Wilmanns and V. Heinemann, Proc. Am. Soc. Clin. Onco/., 1998,
`abstract 965.
`[47] V. Heinemann, Semin. Onco/., 2002, 29, 25.
`[48] V. Hess, M. Salzberg, M. Borner, R. Morant, A. D. Roth, C. Ludwig and R. Herrmann, 1. Clin.
`Onco/., 2003, 21, 66.
`[49] W. Scheithauer, B. Schull, H. Ulrich-Pur, K. Schmid, M. Raderer, K. Haider, W. Kwasny,
`D. Depisch, B. Schneeweiss, F. Lang and G. Kornek, Ann. Onco/., 2003, 14, 97.
`[50] E. Kralidis, S. Aebi, H. Friess, M. W. Buchler and M. M. Borner, Ann. Onco/., 2003, 14, 574.
`[51] A. A. Adjei, C. Erlichman, J. A. Sloan, J. M. Reid, H. C. Pitot, R. M. Goldberg,
`P. Peethambaram, P. Atherton, L. J. Hanson, S. R. Alberts and J. Jett, 1. Clin. Onco/., 2002,
`18,1748.
`[52] B. A. Teicher, V. Chen, C. Shih, K. Menon, P. A. Forler, V. G. Phares and T. Amsrud, Clin.
`Cancer Res., 2000, 6, 1016.
`[53] K. D. Miller, J. Picus, C. Blanke, W. John, J. Clark, L. N. Shulman, D. Thornton, E. Rowinsky
`and P. J. S. Loehrer, Ann. Onco/., 2000, 11, 101.
`[54] H. L. Kindler, II, W. Dugan, H. Hochster, D. Strickland, A. Jacobs and W. J. John, Am. Soc.
`Clin. Onc. Orlando, FL,

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