throbber
Oncogene (2000) 19, 6574 ± 6583
`ã 2000 Macmillan Publishers Ltd All rights reserved 0950 ± 9232/00 $15.00
`
`www.nature.com/onc
`
`From oncogene to drug: development of small molecule tyrosine kinase
`inhibitors as anti-tumor and anti-angiogenic agents
`
`Michael J Morin*,1
`
`1P®zer Global R&D, Groton, Connecticut, CT 06340, USA
`
`The con¯uence of two distinct but related activities in the
`past 10 years has dramatically accelerated e€orts
`towards the discovery and development of novel drugs
`to treat
`cancer. The ®rst
`is a rapidly emerging
`understanding that a number of distinct tyrosine kinases
`play roles in diverse but fundamentally important aspects
`of tumor progression (growth, survival, metastasis and
`angiogenesis). The second is the discovery that small
`molecule compounds have the capacity to potently and
`selectively inhibit the biochemical function of tyrosine
`kinases by competing for ATP binding at the enzyme
`catalytic site. These observations have been conjoined in
`major e€orts to bring forward into clinical development
`novel cancer drugs with the potential to provide both
`clinical ecacy and improved tolerability. The focus of
`this review is on the development of small molecule
`tyrosine kinase inhibitors, and does not extend to other
`approaches that could be applied to disrupt the same
`pathways in clinical
`tumors (receptor and/or ligand-
`competitive antibodies, intrabodies, antisense ribonucleo-
`tides, ribozymes, phosphatase inhibitors or SH2/SH3-
`directed agents). Selected tyrosine kinase inhibitors,
`known or believed to be in development
`in cancer
`treatment trials, are summarized as are some of the
`key issues that must be addressed if these compounds are
`to be developed into clinically useful cancer chemother-
`apeutic agents. Oncogene (2000) 19, 6574 ± 6583.
`
`Keywords: tyrosine kinase inhibitors; anti-tumor; anti-
`angiogenesis
`
`Origin of species ± brief overview of substrate-based
`inhibitors of protein tyrosine kinases
`
`Among all non-traditional (non-DNA-directed) cancer
`targets
`for which pharmacological
`intervention is
`feasible, there are none that have generated as much
`widespread interest, and have
`invoked as much
`resource investment in both the public and private
`sectors in the past 7 years, as have the tyrosine kinases.
`Several excellent recent reviews have described the
`functions of various
`tyrosine kinases
`in the key
`pathways that drive tumor progression,
`from ®rst
`genetic insult to disseminated disease (Hanahan and
`Weinberg, 2000; Hunter, 2000; Gibbs, 2000). Key
`among these are the receptor tyrosine kinases which
`initiate signal transduction in tumor cells or endothelial
`cells following the binding of the growth factors EGF,
`PDGF and VEGF. There are also several excellent
`reviews that provide detailed overviews of the work
`
`*Correspondence: MJ Morin
`
`accomplished to date to understand the molecular
`pharmacology of small molecule inhibitors of receptor
`tyrosine kinases (Sedlacek, 2000; Fry, 2000; Bridges,
`1999; Levitzki, 1999; Lawrence and Niu, 1998). With-
`out summarizing each of these important reviews, they
`provide an appropriate context for understanding the
`obstacles and triumphs that have led, very recently, to
`the ®rst reproducible, objective clinical responses in
`cancer patients treated with tyrosine kinase inhibitors.
`The catalytic function of protein tyrosine kinases
`involves the simple transfer of the gamma phosphate of
`ATP to hydroxyl group of a tyrosine residue of
`proteins (or peptides) encompassing a diversity of
`primary sequences and tertiary structures (Songyang
`and Cantley, 1998). Each of
`the substrates in the
`phosphotransfer reaction, the tyrosine hydroxy group
`and ATP, represent reasonable pharmacological start-
`ing points for the design of substrate analogs and
`competitive inhibitors of tyrosine kinases. A diverse set
`of pharmacophores, including natural products (laven-
`dustins and erbstatins) and synthetic tyrosine mimetics,
`have all been characterized on the basis of their ability
`to competitively inhibit
`tyrosine kinase
`function
`(Levitzki, 1999). These compounds tended to have
`poor potency (particularly in cells), to yield relatively
`¯at structure-activity relationships, and to be some-
`what non-speci®c in their kinase inhibition (Fry, 2000).
`Attacking this
`reaction from the other
`side, by
`identifying compounds that mimic ATP, was originally
`thought
`to be even less tractable. As reviewed by
`Lawrence and Niu (1998), the theoretical obstacles
`were immense. First, the primary sequence of the ATP-
`binding pocket of all kinases is highly conserved, and
`therefore selectivity,
`if not speci®city, represents a
`signi®cant technical challenge. Secondly, the intracel-
`lular
`concentration of ATP can exceed 5 mM,
`particularly in tumor cells, while the Km for ATP in
`most kinase active sites is in the micromolar range,
`thus ensuring full-time saturation by ATP. ATP-
`competitive inhibitors would need to exhibit at least
`nanomolar inhibitory kinetic constants to e€ectively
`compete in this circumstance (Lawrence and Niu,
`1998). Finally,
`there are multiple non-kinase ATP-
`dependent enzymes important to normal physiology,
`and so an indiscriminant ATP mimetic would likely
`have
`toxicities
`that were pharmacologically
`and
`medically unacceptable.
`This theoretical
`logjam was broken in convincing
`fashion when the tyrosine kinase inhibitory activities of
`anilinoquinazolines were ®rst described in 1994 by
`three separate groups (Fry et al., 1994; Ward et al.,
`1994; Osherov and Levitzki, 1994). For example, the
`work of Fry et al. (1994) at Warner Lambert revealed
`that 4-anilinoquinazolines were potent (nM) inhibitors
`
`1 of 10
`
`BI Exhibit 1093
`
`

`

`Tyrosine kinase inhibitors in cancer treatment trials
`MJ Morin
`
`6575
`
`of the EGFR tyrosine kinase with good cell activity
`and profound biochemical selectivity relative to other
`kinases within the tyrosine kinase family. Further
`elaboration of structure-activity relationships rich in
`new possibilities resulted in ATP-competitive inhibitors
`of the EGFR tyrosine kinase with Ki values in the
`single digit picomolar range. It is interesting to note
`that the Michaelis-Menten equation could not be used
`to derive the Ki values of these molecules. So avid was
`the binding of compound to the ATP site,
`the
`conventional approximation that total and free enzyme
`concentrations were equivalent did not apply under
`these conditions. These accomplishments, which may
`be among the most important in pharmacology for the
`last 10 years, were largely achieved by empirical
`screening and iterative medicinal chemistry. Even more
`new chemotypes may emerge as structure-based design
`becomes more commonly applied to the identi®cation
`of both active site- and allosteric site-directed inhibitors
`for an ever-widening slate of tyrosine kinase targets.
`While these early lead molecules had biopharmaceu-
`tical properties which were by-and-large incompatible
`with oral bioavailability and good duration of exposure
`in vivo, the results spurred on a number of groups,
`which have since identi®ed and developed tyrosine
`kinase inhibitors with signi®cant potential
`to treat
`clinical cancer.
`
`Selected development candidates ± updates
`
`PDGFR inhibitors: STI 571 and SU101
`
`STI 571 (CGP57148B) Among all of the candidates
`currently in clinical development, perhaps none has
`provided as much `proof of concept' for the clinical
`ecacy and tolerability of small molecule tyrosine
`kinase inhibitors as has STI 571. Originally disclosed by
`Novartis as a multitrophic tyrosine kinase inhibitor,
`STI 571 was described by Druker et al. (1996); and
`Druker and Lydon (2000) as having potent activity vs
`the translocation product bcr-abl,
`the transforming
`tyrosine kinase found in virtually all CML cells
`expressing the Philadelphia chromosome (Kurzrock et
`al., 1988; Kelliher et al., 1990). The inhibition of v-abl,
`bcr-abl and PDGFR autophosphorylation by the 2-
`phenylaminopyrimidine STI 571 (Figure 1) at nanomo-
`lar concentrations was found to translate to both in
`vivo anti-tumor activity, and to the inhibition of
`clonogenicity of blasts from CML patients (le Coutre
`et al., 1999; Druker et al., 1996). The results of a
`clinical trial
`in which STI 571 was administered to
`CML and ALL patients expressing bcr-abl
`in their
`leukemic blasts were most recently summarized in May
`2000 (Talpaz et al., 2000). STI 571 was used to treat 33
`acute leukemia patients, which included 21 myeloid
`blast crisis CML patients and 12 bcr-abl-positive ALL
`or
`lymphoid blast
`crisis CML patients. Clinical
`responses, as de®ned by a decrease in the percentage
`of patients achieving reduction in bone marrow blasts
`to 15% of pre-treatment levels, were observed in 55%
`of myeloid blast crisis patients, with complete responses
`in 22% of these patients. The response rates in patients
`with bcr-abl positive ALL and lymphoid blast crisis of
`CML were higher (82% with 55% complete responses),
`but all of
`the patients with lymphoid leukemias
`
`relapsed on drug between 45 and 81 days. Of 19
`responding patients, 10 experienced Grade 3 ± 4 neu-
`tropenia. This response rate, and the incidence of
`Grade 3 ± 4 toxicity, compares very favorably to the
`standard of care cytotoxic chemotherapies for CML.
`As such, more de®nitive trials assessing the ecacy and
`safety of STI 571 are ongoing in CML.
`It is interesting to speculate as to the biochemical
`basis for both the ecacy and the toleration pro®le of
`STI 571. Two other tyrosine kinases potently inhibited
`by STI 571, c-kit and PDGFR, are both believed to play
`important roles in maintaining bone marrow stroma ±
`progenitor cell interactions (Ashman, 1999; Sungaran et
`al., 2000). Thus, inhibition of c-kit and PDGFR could
`also account for some of the compelling clinical activity
`of STI 571 in CML, as well as for its toxicity pro®le
`(neutropenia). Treatment of a c-kit expressing a human
`myeloid leukemia cell line, M-07e, with STI 571 before
`stimulation with kit ligand inhibited c-kit autopho-
`sphorylation, activation of mitogen-activated protein
`(MAP) kinase, and activation of Akt, with an IC50 of
`100 nM (Heinrich et al., 2000). STI 571 was even more
`potent in a human mast cell leukemia cell line (HMC-1)
`expressing an activated mutant form of c-kit. Similar
`results have also recently been reported in non-
`hematopoietic tumor cells (Wang et al., 2000). The
`ecacy and safety hypotheses for inhibition of c-abl in
`CML may perhaps only be addressed with a more
`selective abl
`tyrosine kinase inhibitor. Given the
`apparent therapeutic bene®t of STI 571, this may be
`largely an academic question, but one with important
`implications as one tries to rationalize the desired
`selectivity pro®les of tyrosine kinase inhibitors most
`likely to generate both ecacy and safety in humans.
`
`SU101 (le¯unomide; HWA 486) Le¯unomide was
`originally described and developed as an inhibitor of
`dihydroorotate dehydrogenase, a key enzyme in the de
`novo synthesis of pyrimidines, for use as an immuno-
`suppressive or
`anti-arthritic
`agent
`(Bartlett
`and
`Schleyerbach, 1985; Kuo et al., 1996). Le¯unomide
`has
`shown signi®cant activity as a treatment
`for
`rheumatoid arthritis (Smolen and Emery, 2000; Cohen
`et al., 2000b), and was launched by Aventis as Arava2
`in the US and elsewhere beginning in 1998. Extending
`the work of others (Mattar et al., 1993; Xu et al.,
`1995), Shawver and co-workers reported that micro-
`molar concentrations of
`le¯unomide inhibited the
`autophosphorylation of the tyrosine kinase receptors
`for PDGF and VEGF (Shawver et al., 1997). The
`compound was also e€ective at blocking mitogenesis
`stimulated by both PDGF and EGF, but exogenous
`uridine could not reverse the e€ect of le¯unomide on
`PDGF mitogenesis, suggesting that inhibition of the
`receptor
`tyrosine kinase, and not
`inhibition of
`pyrimidine pools, was a key pharmacological activity.
`The inhibition of EGF-induced mitogenesis by le¯u-
`nomide was reversed in part by uridine (Shawver et al.,
`1997), despite the fact that le¯unomide and close-in
`analogs also have inhibitory activity vs the EGFR
`tyrosine kinase (Ghosh et al., 1999).
`Le¯unomide/SU101 is clearly a tyrosine kinase
`inhibitor with multiple biochemical e€ects, and readily
`generates a predominant active metabolite (SU0020 or
`A771726; Figure 1)
`that has a complex inhibitory
`pro®le of its own (Hamilton et al., 1999). SU 101 was,
`
`Oncogene
`
`2 of 10
`
`BI Exhibit 1093
`
`

`

`6576
`
`Tyrosine kinase inhibitors in cancer treatment trials
`MJ Morin
`
`Figure 1 Structures of selected tyrosine kinase inhibitors in clinical development for cancer
`
`nonetheless, progressed into clinical trials by SUGEN
`(now part of Pharmacia). A Phase I study in cancer
`patients revealed that SU 101 was well-tolerated as a
`24 h continuous i.v. infusion at doses up to 443 mg/m2/
`wk. At this dose, the plasma concentration of the
`active metabolite was maintained at levels sucient to
`block both PDGFR and EGFR signaling, as well as
`pyrimidine biosynthesis (Eckhardt et al., 1999). Toxi-
`cities were
`relatively minor
`(Grade 1 ± 2 nausea,
`vomiting and fever
`in approximately 20% of all
`courses given). Surprisingly, hematopoietic toxicities
`and hemolysis, which had been noted in the preclinical
`experience with SU 101, were not seen in this Phase I
`population. One partial
`response was
`seen in 26
`patients receiving an average of two courses each; the
`responding patient received 13 courses (52 infusions) to
`treat an anaplastic astrocytoma, and had a notable
`(450%) reduction in one measurable lesion (Eckhardt
`et al., 1999). SU 101 has been reported to be in
`advanced trials for multiple solid tumor types, but
`recent disclosures (Garber, 2000) indicate that Phase
`III trials in at least one tumor type (glioblastoma) have
`been abandoned. The status of other trials (ongoing
`Phase II trials for ovarian and NSCLC; planned Phase
`III trials for prostate, colon and NSCLC) is uncertain
`at the present time.
`
`EGFR inhibitors: Iressa2 (ZD1839), OSI-774
`(CP-358,774) and CI-1033 (PD183805)
`
`Iressa2 (ZD1839) While STI 571 has provided no-
`table clinical proof-of-concept for the clinical ecacy
`and safety of
`tyrosine kinase inhibitors,
`the early
`
`clinical ®ndings with AstraZeneca's ZD1839 (Iressa2)
`have been equally compelling. The pharmacological
`characteristics of Iressa2 were ®rst described in 1996
`(Wakeling et al., 1996; Woodburn et al., 1997) as a
`potent and selective inhibitor of the EGFR tyrosine
`kinase. This quinazoline-based compound (Figure 1) is
`an ATP-competitive inhibitor of the EGFR tyrosine
`kinase (IC50 25 nM) with 50-fold selectivity relative to
`closely homologous erbB family members (IC50 for
`erbB2 1 ± 3 mM) and even greater selectivity for more
`divergent
`tyrosine kinases.
`It demonstrates good
`cellular potency (80 nM IC50 for inhibition of EGF-
`dependent mitogenesis) and robust, dose-dependent
`anti-tumor ecacy in a variety of human tumor
`xenografts (Woodburn et al., 1997). These results have
`been most recently extended to show that Iressa2 has
`in vivo ecacy in a diverse human tumor xenograft
`models both with (Ciardello et al., 2000) and without
`(Sirotnak et al., 2000) highly activated EGFR signaling
`pathways. Of equal interest are the observations that
`Iressa2 combines with standard cytotoxic agents
`(platinums, taxanes, topoisomerase I inhibitors, etc.)
`to produce additive or
`supra-additive anti-tumor
`ecacy in vivo without exacerbation of the toxicity of
`the co-administered cytotoxics. The ®ndings that tumor
`EGFR density does not predict ecacy when the
`compound is used in conjunction with cytotoxic agents
`have signi®cantly impacted the development strategy
`employed by AstraZeneca as Iressa2 moves towards
`pivotal clinical trials.
`trials with Iressa2 have been
`Multiple Phase I
`summarized, and the
`results
`revealed reasonable
`pharmacokinetics, good toleration and the ®rst signs
`
`Oncogene
`
`3 of 10
`
`BI Exhibit 1093
`
`

`

`Tyrosine kinase inhibitors in cancer treatment trials
`MJ Morin
`
`6577
`
`of clinical ecacy when used as a single agent in
`patients with advanced disease (Ferry et al., 2000;
`Baselga et al., 2000; Kelly et al., 2000). Following oral
`administration of a single dose (50 mg), maximum
`plasma drug concentrations (mean 45 ng/ml) occurred
`1 ± 5 h post-dose. The mean terminal t1/2 was 34 h.
`Inter-subject variability in exposure was signi®cant
`following single and multiple administration (up to
`sevenfold at each dose level), but exposure increased
`proportionally with dose, with no apparent change in
`terminal t1/2 across the dose range tested (Kelly et al.,
`2000). In a larger dose-escalation trial, Ferry and
`collaborators administered Iressa2 at doses of 50 ±
`700 mg once daily, given orally for 14 days followed by
`14 days of observation (Ferry et al., 2000). In total, 64
`patients with advanced disease, who had each
`progressed while on prior chemotherapy, completed
`145 cycles. Cmax and AUC0-24h were proportional
`across
`the entire dose range (mean values 113 ±
`2255 ng/ml and 1.8 ± 38.5 mg.h/ml, respectively). As
`in single dose studies, Iressa2 showed a long terminal
`elimination half-life (mean of 46 h). Iressa2 was very
`well-tolerated in this study; the most common adverse
`events were diarrhea and acne-like skin rash (Grade 1 ±
`2). Acne-like skin rashes have emerged as a common,
`mechanism-based adverse event for EGFR inhibitors,
`but the speci®c toxicological e€ect in the skin is not yet
`well understood. Grade 3 ± 4 adverse events were
`shown to be rare with Iressa2 treatment, and were
`generally ascribed to disease progression. The dose-
`limiting toxicity, de®ned at the 700 mg dose level, was
`Grade 3 diarrhea (Ferry et al., 2000).
`A compelling level of ecacy was also revealed in
`these early trials (Ferry et al., 2000). Anti-tumor
`responses were most evident among the 16 NSCLC
`patients treated with Iressa2 ± two had an objective
`partial response, two patients had signi®cant regression
`of disease and two patients had stable disease. Similar
`pharmacokinetic and safety pro®les were noted in a
`separate study (Baselga et al., 2000), one that also
`revealed the potential
`for ecacy from Iressa2 in
`patients with advanced prostatic
`and head-neck
`cancers. These early results added importantly to the
`proof-of-concept that selective tyrosine kinase inhibi-
`tors could have signi®cant single agent ecacy, as
`measured by objective tumor regressions,
`in patients
`with advanced disease. The clinical observations have
`therefore recapitulated the pre-clinical data showing
`that Iressa2 increased apoptosis and regressions in
`human tumor xenograft models (Ciardello et al., 2000).
`The Iressa2 data indicate that the ecacy of these
`agents can be measured using more classically de®ned
`clinical endpoints. There will undoubtedly be signi®-
`cant value in the use of pharmacodynamic and
`surrogate endpoints to guide dose-intensi®cation or to
`pre-select patients for whom other tyrosine kinase
`inhibitors might represent the most promising treat-
`ment option. Pharmacodynamic endpoints have not
`played a major role in the early development of EGFR
`tyrosine kinase inhibitors, despite the fact that several
`reasonable options
`exist,
`including both invasive
`techniques (direct measurement of tumor-derived or
`normal tissue-derived EGFR phosphotyrosine, phos-
`phorylation of down-stream signaling molecules;
`apoptosis markers) and non-invasive techniques such
`as PET imaging of metabolically modulated tumors
`
`(Pollack et al., 1999; Goss et al., 2000; Allen et al.,
`2000). Given the overall safety and toleration pro®le of
`Iressa2, AstraZeneca has committed to an aggressive
`development strategy, which includes two large Phase
`III studies to assess the use of Iressa2 in combination
`with cis- or
`carbo-platinum plus
`a
`taxane or
`gemcitabine in ®rst-line therapy for NSCLC (trials 14
`and 17), as well as a Phase II trial (trial 16) to con®rm
`the single agent activity of Iressa2 in patients with
`advanced NSCLC (Kelly et al., 2000). It is important
`to note that these trials do not call for a prospective
`selection for patients with tumors with some pre-
`de®ned level of EGFR over-expression. All epithelial
`tumors express some EGFR, and in the disease target
`here, NSCLC,
`tumors often present with a high
`proportion of EGFR over-expression (up to 80 ± 90%
`in advanced disease). The strategy is also consistent
`with pre-clinical data suggesting that ecacy in drug
`combinations may not be determined in large part by
`the level of EGFR over-expression in tumors (Sirotnak
`et al., 2000). Results are expected from these pivotal
`trials in a late-2001 or early-2002 timeframe.
`
`OSI-774 (CP-358,774) CP-358,774 is also a potent
`and selective quinazoline-based inhibitor of the EGFR
`function (Figure 1). This compound is a reversible,
`ATP-competitive inhibitor (IC50 of 2 nM) of the EGFR
`tyrosine kinase, with greater than 500-fold selectivity
`against other tyrosine kinases, such as the closely
`related erbB2 kinase, as well as v-src, c-abl and the
`insulin and IGF-1 receptors, (Moyer et al., 1997). CP-
`358,774 inhibits the autophosphorylation of the EGF
`receptor in a variety of EGFR over-expressing tumor
`cells (IC50=20 nM), and produces cell cycle arrest and
`apoptosis in multiple cell types (Moyer et al., 1997;
`Barbacci et al., 1997; Iwata et al., 1997). In vivo, CP-
`358,774 e€ectively inhibits EGFR-speci®c
`tyrosine
`phosphorylation in human tumor xenografts (ED50 of
`10 mg/kg p.o. when given as a single dose) with
`signi®cant duration of action; daily dosing produces
`substantial growth inhibition and regressions in human
`tumor xenografts (Pollack et al., 1999). Moreover, the
`dose-response for tumor growth inhibition shows good
`agreement with the dose-response for inhibition of
`EGFR-phosphotyrosine in tumors from treated ani-
`mals. As with Iressa2, CP-358,774 was
`found to
`generate additive anti-tumor activity when used in
`combination with cis-platinum and other cytotoxic
`agents, without exacerbating the toxicities of the other
`chemotherapeutants (Pollack et al., 1999).
`Clinical studies with CP-358,774 have revealed that
`the agent is well-tolerated at oral doses that achieve
`plasma concentrations projected to be required for
`anti-tumor ecacy in humans (400 ± 500 ng/ml). In one
`study, escalating doses were administered orally once
`every week (Karp et al., 1999). Eighteen patients with
`advanced solid tumors were treated at ®ve doses (100 ±
`1000 mg)
`for a maximum period of 24 weeks.
`Toxicities were observed only at doses higher than
`200 mg/week, and included mild fatigue, Grade 2
`maculopapular (acneiform) rash, Grade 2 nausea, and
`Grade 2 diarrhea. Like Iressa2, CP-358,774 exhibited
`intra- and inter-subject variability in exposure, but
`dose-proportional increases in exposure were observed
`throughout
`the 100 ± 1000 mg weekly dose range.
`During the ®rst 24 h following a single dose, the Cavg
`
`Oncogene
`
`4 of 10
`
`BI Exhibit 1093
`
`

`

`Tyrosine kinase inhibitors in cancer treatment trials
`MJ Morin
`
`(0.9 ± 4.8 mg/ml for 100 ± 1000 mg doses, respectively)
`was
`some
`two-
`to 10-fold above
`the projected
`ecacious plasma concentration. No maximally toler-
`ated dose or dose-limiting toxicity was discerned in this
`study. In a second Phase I study (Siu et al., 1999),
`patients were given CP-358,774 tablets in a variety of
`dose schedules, culminating in daily dosing at
`the
`maximally tolerated dose. The target Cavg of 400 ±
`500 ng/ml was achievable at doses at and above
`100 mg/day on a well-tolerated schedule (Cavg values
`following continuous daily dosing at the 50, 100 and
`200 mg/day levels were 432, 973 and 2120 ng/ml,
`respectively). Dose-limiting diarrhea was encountered
`at
`the 200 mg/day level. An intermediate dose of
`150 mg/day was subsequently de®ned as the maximally
`tolerated dose (two of three patients had Grade 1
`diarrhea with loperamide support).
`Siu and co-workers also made e€orts to understand
`the `characteristic' Grade 1 ± 2 acneiform rash seen in
`patients treated with CP-358,774, which was limited to
`regions of the upper body where adolescent acne is
`usually manifest (face, back and scalp). Histopathology
`of
`skin biopsies showed subepidermal neutrophilic
`in®ltration and epidermal hyperproliferation (Siu et
`al., 1999). While the precise cytopathic basis for the
`acneiform rash has not yet been determined,
`the
`consistent clinical observations with three di€erent
`agents targeting EGFR function (CP-358,774, Iressa2
`and Imclone's C-225 antibody) suggest that this is a
`mechanism-based ®nding (Siu et al., 1999; Ferry et al.,
`2000; Cohen et al., 2000b). Skin changes are consis-
`tently noted in preclinical studies with rodents exposed
`to CP-358,774 for extended dosing periods, and these
`toxicological results are analogous to the skin changes
`seen in the waved-2 mouse, which has a mutated and
`marginally functional EGFR tyrosine kinase (Luetteke
`et al., 1994).
`from ongoing Phase II
`Early ecacy readouts
`clinical trials with CP-358,774 have been compelling.
`The agent appears to have a broad potential to treat a
`variety of human solid tumors,
`including NSCLC,
`breast, ovarian and squamous head and neck tumors
`(Bonomi et al., 2000; Allen et al., 2000; Siu et al., 2000;
`Hammond et al., 2000). For example, in 34 NSCLC
`patients who had failed prior chemotherapy, daily oral
`doses of 150 mg CP-358,774 were well-tolerated, with a
`maculopapular
`(acneiform)
`rash being the most
`common adverse event reported. In 56 total patients
`evaluable for tumor response,
`there have been six
`partial responses in the lung and/or liver at 8 weeks
`and several patients with stable disease (Bonomi et al.,
`2000).
`In 71 patients with refractory squamous
`carcinomas of the head and neck, CP-358,774 was
`again found to cause a reversible acneiform rash and
`Grade 1 ± 2 diarrhea. Of 78 patients evaluable for
`response,
`there have been at
`least eight con®rmed
`partial responses and 23 patients with stable disease
`(Siu et al., 2000). These preliminary results indicate
`that CP-358,774 is generally well-tolerated and demon-
`strates evidence of single agent anti-tumor activity in
`patients with recurrent head and neck cancer, as well
`as in treatment-refractory NSCLC.
`Due to signi®cant interests in both CP-358,774 and
`CI-1033, P®zer was directed to divest one of these two
`agents as a condition of their acquisition of Warner
`Lambert in 2000. As such, Oncogene Science (OSIP)
`
`has taken over complete responsibility for the devel-
`opment of CP-358,774, which is now formally referred
`to as OSI-774.
`
`CI-1033 (PD183805) As described above, the selec-
`tive and reversible inhibitors of the EGFR tyrosine
`kinase appear to o€er the promise of
`therapeutic
`ecacy coupled to reasonable
`tolerability.
`It
`is
`important to note, however, that the therapeutic index
`of neither Iressa2 nor CP-358,774 has yet to be fully
`elaborated, and that there may be signi®cant proximity
`between the maximally tolerated doses and the
`ecacious doses
`for both agents. Moreover,
`the
`ecacy of neither agent has yet to be established in a
`blinded, placebo controlled study. As
`such,
`there
`continues to be an opportunity to discover and develop
`distinctly di€erent EGFR tyrosine kinase inhibitors
`with even greater potential for ecacy and a broader
`spectrum of activity. CI-1033 is one such distinctly
`di€erent development candidate. As recently reviewed
`by David Fry of
`the
`former Warner Lambert
`organization, signaling through the erbB family of
`tyrosine kinase receptors often involves complex cross-
`talk among the members of that receptor family (Fry,
`2000). The four family members (EGFR or erbB;
`erbB2, erbB3 and erbB4) are known to intensify their
`kinase-dependent transforming signals via the forma-
`tion of heterodimers with each other (Tzahar et al.,
`1996). There is, therefore, a compelling rationale to
`consider the potential utility of nonspeci®c but selective
`inhibitors that e€ectively block the function of the erbB
`family but do not inhibit more structurally diverse
`tyrosine kinases.
`to consider
`There
`is also a strong rationale
`irreversible tyrosine kinase inhibitors. The reversible
`inhibitors have apparently generated clinical ecacy
`with dosing regimens designed to maintain plasma
`concentrations at fairly high levels for extended periods
`of
`time. The optimal dosing paradigm for an
`irreversible inhibitor would be less likely to require
`prolonged exposure. Moreover, the `absolute ®nality'
`(Fry, 2000) of the irreversible inhibitors could con-
`ceivably provide signi®cant advantages in terms of
`antitumor ecacy. To be balanced, a multi-tropic and
`irreversible inhibitor would also have the potential to
`generate a toxicity pro®le that was di€erent and,
`perhaps, without advantages
`relative to the more
`selective, reversible inhibitors. Preclinical data suggest
`that irreversible EGFR tyrosine kinase inhibitors can
`generate
`signi®cant
`ecacy with good toleration
`(Vincent et al., 1999), but the ultimate utility of these
`agents can only be determined in clinical trials.
`Homology modeling of ATP binding to the pocket
`of EGFR suggested that the thiol of cys773 would be a
`key potential site for attack by a rationally designed
`irreversible ATP-mimetic. One compound containing
`an acrylamide functionality at the six position of the 4-
`anilinoquinazoline nucleus (Figure 1) was found to
`have a profoundly rapid onset and long-lasting
`inhibition of both EGFR and erbB2 in tumor cells,
`and to be selective relative to non-erbB tyrosine kinases
`(Fry et al., 1998). When compared to very closely
`related reversible analogs (in which the acrylamide
`double bond was reduced), the 6-substituted irrever-
`sible analogs were more potent
`in vitro and had
`signi®cantly greater ecacy in vivo. Further improve-
`
`6578
`
`Oncogene
`
`5 of 10
`
`BI Exhibit 1093
`
`

`

`Tyrosine kinase inhibitors in cancer treatment trials
`MJ Morin
`
`6579
`
`ments (addition of substitutions which also improved
`water-solubility) led to the elaboration of PD 183805/
`CI-1033 (Figure 1). Like its predecessors, this com-
`pound has excellent (low nM) potency against erbB2
`and EGFR in both enzyme- and cell-based assays
`(Sherwood et al., 1999). Consistent with a predicted
`advantage relative to reversible inhibitors, CI-1033
`potently inhibits human tumor xenografts when dosed
`as infrequently as once per week, and a single dose
`eliminated the level of EGFR phosphorylation in
`tumors for longer than 72 h (Vincent et al., 1999).
`Like CP-358,774, CI-1033 combines well
`in drug
`combinations with cytotoxic agents. Given 24 h after
`gemcitabine, CI-1033 produced a signi®cant increase in
`the apoptotic fraction in tumors over treatment with
`either drug alone (Nelson and Fry, 2000). CI-1033 also
`e€ectively decreased the clonogenicity of human tumor
`cells taken from patients (Medina et al., 2000), with
`notable responses seen in breast (67%), NSCLC (60%)
`and ovarian cancer specimens. CI-1033 Phase I clinical
`trials have
`recently been initiated, but data on
`pharmacokinetics or safety have not yet been disclosed.
`
`Small molecule tyrosine kinase inhibitors targeting
`angiogenesis pathways
`
`There are multiple tyrosine kinase receptors which
`appear to have key roles in the generation of new
`tumor blood vessels and, as such, represent reasonable
`targets for cancer chemotherapy (for excellent recent
`reviews, see Cherrington et al., 2000; Randal, 2000;
`Thompson et al., 1999; Hamby and Showalter, 1999).
`Included among the key tyrosine kinase targets that
`have generated the most interest in the scienti®c and
`patent (Connell, 2000) literature are PDGFR, VEGFR,
`FGFR and tie-2. The key development candidates
`targeting PDGFR, STI 571 and SU101, were described
`above, though neither compound is likely to reveal the
`clinical utility of PDGFR-directed inhibition of
`angiogenesis due to their multiple mechanisms of
`action. Agents that selectively target FGFR and tie-2
`are not known to be in development, though several
`drugs targeting VEGFR have inhibitory activity vs
`FGFR. As such, the focus of the remainder of this
`overview will be on the clinical candidates targeting
`VEGFR. Two high anity receptors for VEGF have
`been identi®ed and characterized on human endothelial
`cells, ¯t-1 and KDR. KDR appears to be expressed
`primarily on activated endothelial cells and is thought
`to be more of a key driver of mitogenic responses
`commonly found in neovascularizing tumors, while ¯t-
`1 is expressed on multiple other cell types (Plate et al.,
`1994; Wedge et al., 2000a). For the purposes of this
`review, the terms KDR and VEGFR will be used
`interchangeably, unless otherwise speci®ed.
`
`SU 5416 and SU 6668 The former SUGEN organiza-
`tion (now part of Pharmacia) has clearly set the early
`pace in the race to identify and develop inhibitors of
`the VEGFR tyrosine kinase. E€orts towards this end
`have initially focused on the indolin-2-one pharmaco-
`phore (Figure 1). Among the earliest compounds of
`this class was SU 5416, which was found to be a potent
`inhibitor of the kinase a

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