throbber
' peygpectgve
`§EflEE§
`
`arm targets For
`cancer therapy
`
`Wifiifiam G. Kaolin, _§r.,
`
`Eéwgr
`
`Anticancer’ drug targets: gro h factors and
`geo
`h factor signaling
`
`_l’acl<so:n B. Gibbs
`WP] 6-10 l Cancer Research, Department of Cancer Research, Merck Research Laboratories,
`Sumneytown Pike, West Point, Pennsylvania 19486, U .“i.
`Phone: (215) 652-5278; Fax: (215) 652--732.0; E-mailzg I gibbs@rnerctl<.corn.
`
`
`
`Signaling mechanisms that drive cell proliferation are
`closely associated with tumor malign ancy. Components
`of these pathways, encoded by some of the very first
`oncogenes identified, include the PDGF—lil<e ligand Sis,
`the tyrosine kin ases Src and H'ER~2/c—Neu {HER~2), and
`the (3’l‘P~binding switch Ras. The study of communica-
`tion by these oncoproteins has identified a complex
`array ofintracellular circuits. In some cancers, mutations
`in key components lead to constitutive activation of
`these pathways; this activation is associated with the pro~
`liferative properties of the tumor cells. In this Perspec-
`tive, I provide a broad overview ofa growth factor signal
`transduction system, with a focus on those points that
`have been translated to drugs or clinical candidates. Due
`to editorial restrictions limiting the number of reference
`citations, much of the clinical data gleaned from
`abstracts is not listed in the references. Instead, the read-
`er is directed to the 1999 Proceedings of the American
`Society ofClinical Oncology and the 1999 Proceedings
`ofthe AACR—NCl-EORTC International Conference.
`
`Signaling pathways are initiated, with the binding ofa
`ligand, such as PDGF, EGF, EGF-like ligands g., TGF
`and amphiregulin), or IGF, to its cognate transinembrane
`receptor I). Ligand binding induces the dimerization of
`receptor subunits, promoting autophosphoiylation of the
`receptor and recruiting a variety of intracellular doclzing
`proteins (such as Grb2, Shc, and Ncl<) to the plasma mem-
`brane. These d,ocl<in,g proteins create a molecular scaffold
`from which subsequent signals emanate. For example, the
`guanine nucleotide exchange factor Sos binds to Grb2,
`which in turn interacts with the Ras protein. Ras serves as
`a molecular switch in the plasma membrane that alter~
`nates between an inactive GDP—bound state and an active.
`
`GTE’-bound state. Normally, Ras is bound to GDP be
`cause of th e abundance ofG"l,"Pase~activatin,g protein and
`neurofibromin, which both suppress Ras function. How-
`ever, upon recruitment ofSos to the membrane, Sos binds
`Ras~GDP and facilitates release of GDP. In cells, the
`nucleotide CETP is about l0-fold more abundant than
`
`GDP; GTP binds to Ras by mass action. Ras—GTP adopts
`a conformation that permits interaction with down-
`stream targets called, effector molecules. These effectors
`include the protein kinase Raf, which activates the MAP
`kinase cascade; GTPase-activating protein, which links
`Ras to the Rho/Rae pathway; and phosphoinositicle (PI)
`3’-l<inase and Ral—guan,ine nu cleotide dissoci ati on stirnu~
`lator (Ral-GDS), which activate lipid pathways (2). The
`dysregulation ofthese signals in tumor cells leads to mul~
`tiple cellular changes, including altera.tion,s in DNA syn~
`thesis, lipidmetabolism, cellular morphology, cell adhe-
`sion properties, and gene expression.
`
`In the broadest sense, the study of signaling mecha-
`nisms has already yielded therapeutic agents in the treat~
`ment ofcancer, as evidenced by antiestrogen an tiandro~
`gens, agonists of gonadotropin-releasing hormone, and
`stem cell growth factors, for example. However, research
`into oncoproteins that function within the signal trans~
`duction system is only beginning to be applied in the clin-
`ic. Therapeutic approaches of interest include tools such
`as mAbs against the extracellular domain of receptors,
`oligonucleotides that are an tisense to key target proteins,
`and small molecule inhibitors of enzymes (Table 1).
`
`Growth factor receptors
`Efforts to inhibit HER-2 yielded the first cancer thera~
`peu tic agent based on research in growth factor signal-
`ing. Unlike other members ofthe EGF receptor family,
`I:'ll3lR~2 has no known ligand (3).
`':IlE3l"~i~2 expression, is
`upregulated in approximately 25-30% of human breast
`cancers; this upregulation is believed to promote HER—2
`heterodimerization with other it embers of the EGF
`
`receptor family, as well as HER.-2 homodimerization,
`which results in a constitutively active tyrosine kir ase.
`In creased expression of HERJ generally correlates with
`the severity of disease, and expression is consistently
`higher in tumor tissue than in normal tissue, making the
`tumor more prone to antibody therapy.
`Genentech Inc. developed the mAb trastuzuinab,
`which is directed, against the extracellular domain, of
`l"IER.~2
`Use of this drug requires genotyping patient
`tumor samples for the expression of HER~2. it is
`thought that trastuzumab inhibits the proliferation of
`breast cancer cells by several mechanisms (5). First,
`binding of trastuzumab is associated with u pregula~
`tion of the p271‘-5? inhibitor ofsome cyclin-dependent
`kinases. Second, this agent accelerates the internali;.:a-
`tion and degradation of HER-2, reducing the cellular
`level of activated tyrosine protein kinase. Third,
`trastuzumab may induce immune—mecliated effects,
`including cell~mediated cytotoxicity and complement
`fixation. in combination with cisplatin, doxorubicin,
`and especially paclitaxel, trastuzumab shows enhanced
`an,ti~tnnior activity in preclinical models
`Trastu~
`zurnab has also proved its value in the clinic and is par-
`ticularly effective in combination with p&1,ClIlC£L\L£‘l 7, 8).
`The combination of trastuzuinab with doxorubicin
`
`also appears to be effective, but may have higher car-
`diotoxicity than trastuzumab alone (8, 9).
`From the perspective ofpharmaceutical development,
`it is interesting to note that the time from the discovery
`of the HERE/'c'—neu oncogene in i985 and the associa~
`tion of HER—2 amplification in human breast cancer in
`
`’I'b.e_lournal ofClinical Investigation
`
`1
`
`January 2000
`
`] Volume 1.05
`
`] Numberl
`
`APOTEX EX. 1010-001
`
`

`
`Tobie 1
`
`Examples ofinhibitors ofgrowth factor signaling for cancer treatment
`
`
`Developrnent status
`
`Launched as Herceptinlwl
`Phase HE
`
`"_”3C'i”lCal
`
`pmcggngcaa
`
`Pha
`Preclinical
`Phase H
`
`
`
`Compound
`
`Trastuzumab
`C225
`E7.6.3
`Z [34 8.39
`CP—358,774
`PD—l68,393
`SU-‘l Ul
`AS ODN
`ISIS--25"’
`R115
`SCH 66I;:':6
`L—778,l 23
`BMS—2’£4662
`|SlS—Sl32
`ZM 336372
`l_.--
`-9450
`PD--184352
`UOl26
`ISIS?) 521
`CCP /-H251
`LlCN—0'l
`LY294002
`
`
`
`
`
`larger
`
`HER2/c-rieu
`EGF receptor‘
`
`PDCF receptor
`lCiFR
`Ras
`
`Raf
`
`MEK
`
`PKC
`
`Pl 3"-kinase
`
`residue near the ATP binding site;
`its irreversible binding may afford
`improved anti-tumor activity. it will
`be interesting to monitor the devel-
`opment of this class of inhibitor:
`such reactive molecules are often
`
`dismissed as drugs, because of their
`potential for nonspecific interac-
`tions, but if they are sufficiently
`selective for their targets, reactivity
`need not be seen as a negative trait.
`Aspirin, for example,
`is an irre-
`versible inhibitor of cyclooxygen as-
`es.
`
`pmclinicag
`Preclinical
`P'!'fhCl‘”‘CHal
`H
`
`2
`Phas
`Preclinical
`
`SU-101, an inhibitor of PDGF
`receptor ltinase activity (15, 16), is
`currently in phase II development
`for treating glioblastonias. Another
`receptor tyrosine ltinase that has
`been explored with increasing atten-
`tiori as a drug target is the E61?’ type
`l
`(IGF-l) receptor (17, 18). This
`receptor activates cell proliferation,
`but its role as an antiapoptotic sig-
`nal may be more significant. Initial
`evidence from preclinical studies of an antisense
`oligonucleotide suggests that TGF-l receptor inhibition
`can promote tumor apoptosis (17).
`
`Targeting a CTPase switch
`The ms gene, discovered in 1978, has attracted a great
`deal of attention because it was the among the first
`oncogenes associated with human cancer, and studies
`of Ras function have helped to elucidate many of the
`mitogenic cell signaling pa,tl1wa.ys (l Mutated forms
`of Kirsten-ms (Kzlms) and N—ms are found in solid tumors
`(lung, colon, pancreas, and brain) and leuliemias, where-
`as inutant Harvey-ms Hal--ms) alleles are found in only a
`small subset of bladder, head, and rieclc: tumors. The
`agents currently in clinical trials that are based on this
`area ofresearch act either by regulating ms gene expres-
`sion or by inhibiting protein farnesylation. An an tisen,se
`oligonucleotide (lSlS~2S03) directed against H/.Z—1‘;sZ5
`expression (20) displayed significant anti-turnor activi-
`ty against a variety of human tumor cell lines in pre-
`clinical mouse tumor xenograft studies. ISIS-2503
`appears to act against tumors whether or not they have
`suffered mutations in Ha--ms, but the basis ofthis broad
`activity is unclear. lSlS-2503 has completed phase I eval-
`uation; an initial report noted some disease sta.bili;:a-
`tion when this agent was administered by continuous
`intravenous infusion (20).
`A second approach for inhibiting Ras function has
`attracted broad attention within the pharmaceutical
`industry. Ras proteins carry an essential lipid moiety — a
`farnesyl group
`at their COOH termini. Genetic data
`indicate that inhibition of Ras farnesylation blocks Ras
`localization to the plasma membrane. ‘X/’ithc>ut this mem-
`brane localization, Ras fails to interact with critical regu-
`latory and effector molecules (19), and is transformation
`defective. Hence, farnesyl-protein transferase inhibitors
`(FT1s) are predicted to block cellular transformation.
`
`l987 to FDA approval of trastuzurnab in l998 was a rel-
`atively short period. This rapid progress reflects an
`understanding of the underlying science, as well
`the
`fact that trastuzumab is a biological agent. In general,
`biological agents may be developed more quicltly than
`are chemical entities.
`
`Tlierapeu tic antibodies have also been developed
`against
`the EGF receptor. C223, a human/mouse
`chimeric a itibody (10), and E/.613, a fully human anti-
`body (1 1), bind to the EGF receptor extracellular domain
`and block EGF ligand binding. These antibodies block
`the liganddependent proliferation of breast cancer cell
`lines in cell culture, and can induce tumor regression in
`mouse xenograft tumor
`Like trastuzumab, C225
`appears to be especially effective in combination with
`doxorubicin or paclita:<el (10). C225 is currently under-
`going clinical evaluation. in preliminary trial results,
`complete responses were noted in head and neck cancers
`when C225 was combined with radiotherapy.
`The EGF receptor is also the target for the develop-
`ment ofinhibitors of the intracellular tyrosine l(lI‘1£i,SC
`domain. ZD-l839 and CP-358,774, competitive in-
`hibitors ofATP binding to the receptors active site, are
`currently in clinical trials (12, 13). Their mechanism of
`action has led to some concern about safety, given the
`variety and physiological significance of protein l<inas—
`es and other enzymes that bind ATP. However, these
`compounds appear to have good, anti-cancer activity in
`preclinical models, with an acceptable therapeutic
`index, particularly in patients with non—srnall cell lung
`cancer. The dermatological toxicity observed for these
`drugs is most liltely mechanism based, arising
`a con-
`sequence of their intended biochemical activities.
`More recently, highly potent and selective irreversible
`inhibitors of the EGF receptor ltinase have been
`reported, such as PD-ltS8,593 (3,4). This compound
`appears to bind specifically to an active—site cysteine
`
`‘E0
`
`Tl)('E_l()‘lH‘t1E1l,()f.Cl,il]ié223,l Investigation
`
`E
`
`J;muary 2000
`
`E Volurne 105
`
`E Numberl
`
`APOTEX EX. 1010-002
`
`

`
`However, the transferase reaction is essential not only to
`the function of Ras, but also to the function of at least 20
`other farnesyl proteins. Thu FTl.s are not truly Ras-spe»
`cific inhibitors. Nevertheless, a number ofFTls have been
`developed as potential anti-cancer drugs (21, 22).
`Potent FTIS of diverse chemical structu res inhibit
`
`tumor growth in both nude mouse xenograft models and
`avariety of transgenic mouse tumor models — including
`those that overexpress Ha--ms, K2’--ms, or N--ms
`The
`similar: effects of structurally distinct l?’Tl.s, and their
`effectiveness at doses that bloclz substrate protein farne-
`sylation, confirm that these compounds achieve the
`desired anti-tumor activity by inhibiting farnesyl-protein
`transferase. Unlike cytotoxic an ti-turnor agents, .F7Tls
`appear to act without overt systemic toxicity. Since FTls
`were origin ally thought to be cytostatic agents, it was sur-
`prising to observe in preclinical tissue culture and trans-
`genic tumor models that they induce apoptosis in tumor
`cells. The induction of apoptosis occurs by caspase-3 acti-
`vation and is in dependent ofwild-type p53 function (21,
`23) — an important finding given the usual association
`of loss of p53 function with resistance to chemotherapy
`(see Sellers and Fisher in this Perspective series).
`in 3.997 and 19.98, nearly 20 years after the discove.ry of
`Elias and about 9 yea rs after the discovery of Ras fa.rnesy-
`lation, clinical trials began with FT1s (22). At least 4 dif-
`ferent FTls are currently undergoing evaluation:
`Rl l5777; EiCl:'l 66335; l./,"78,l23; and Bl»/l5}-2 l.-4662 (Z4)
`(Tablel). R1 ES777 and SCH 66336 are administered by
`the oral route, L-778,123 is given by continuous infu-
`sion, and EMS-2 l 4662 is administered either orally or
`in tra.venousl.y. The more advanced trials with l1{l.l5777
`and SCH 66356 have reported dose-limiting toxicities
`involving bone marrow and the gastrointestinal tract,
`indicating that at high enough con centrations, .FTls can
`have general antiproliferative effects on normal tissues.
`The doses achieved in the clinic so far with L-778,123
`and SCH 66336 were sufficient to inhibit protein farn e-
`sylation. in readily obtainable tissues such as white blood
`cells and cells of the buccal mucosa. Reports on the effi-
`cacy of FTls are anxiously awaited. Based upon preclini-
`cal data, it is anticipated that .F7Tls will also be used in.
`combination. with other treatments, such as paclitaxel,
`vincristine, cisplatin, 5-fluorouracil, gemcitabine, cyclo-
`phosphamide, or radiation (25
`
`inhibiting protein kinase effectors
`A series of protein phosphorylation events within the cell
`en sue upon Ras activation. The first key step is the direct
`binding of the Raf protein l<inase to Ras-CETP (1, 2). Raf
`in turn phosphorylates and activates MAP,/.3rl< kinase
`(l\/IEK), which in turn phosphoiylates and actvates MAP
`kin ase. The key role of this pathway in. Ras-mediated cel-
`lular transformation has inspired several efforts to devel-
`op inhibitors of these protein ltinase reactions (Table
`ISIS-5132, an antisense oligonucleotide directed
`against
`is in phase ll. clinical development 20). This
`compound causes a dose-dependent reduction of c-Raf
`mRNA levels in preclinical tumor models. This pharma-
`codynarnic monitoring h also been performed in the
`clinic using peripheral blood mononuclear cells from
`treated patients as a tissue source. in a phase ii trial, the
`
`median reduction of Raf mRNA was 42% at 48 hours,
`with significant inhibitions observed up to 15 days,
`although this decrease did not appear to be close depend-
`ent. Of the 65 patients evaluated in these initial reports,
`4 patients with ovarian, pancreatic, renal, and colon can-
`cer have seen their disease. remain stable for up to 10
`months. Interestingly, in. Z of the other patients, disease
`progression coincided with the loss of suppression of
`RafmRNA levels (20).
`Raf protein kinase inhibitors remain at an earlier stage
`of development. The most extensive analysis is from Hall-
`jacltson et al. (29, 30), who characterized the biological
`effects ofboth a direct Raf l{il1ELS€ inhibitor, ZM 3367372,
`and a p58 kinase inhibitor, SB 203580, which weakly
`inhibits Rafkinase activity. Cells treated with ZM 336372
`or SB 203580 exhibit a paradoxical increase in Rafactiv-
`ity measured ex vi vo, indicating that these compound.s do
`not inhibit Raf signaling pathways. ZM 336372 does not
`inhibit R.as- or Raf-mediated cellular transformation, but
`a preliminary report by Heimbrook et al. (31) indicates
`that the triarylimida.zole derivative L-779,450, which
`inhibits Raf protein kinase activity in vitro, blocks intra-
`cellular signaling by Ki-Ras and Ha-Ras.
`Two groups have recently described novel MEK in-
`hibitors (Table
`.Parl<e-"Davis Pharrnaceutical. Re-
`search, which described the first l‘vlEl{ inhibitor, PD-
`098059,
`identified a more potent and selective
`compound (PD-184552) from a coupled biochemical
`screen that included GST-MEK, MAP ltinase, and the
`MAE“-’ kinase substrate myelin basic protein (32). DuPont
`Pharmaceuticals Co. identified U01Z6 in a cell-based
`
`assay that rnonitored AP-3. response elements, and they
`subsequently found that this compound inhibits MEK
`activity
`Neither PD-‘E84-352 nor U0126 compete
`for binding to ,1-‘Cl"l’ or protein substrates, suggesting
`that these compounds function as allosteric inhibitors
`of MEK. Both compounds blocl: MAP lzinase phospho-
`rylation in cells, and at doses that abolish intracellular
`lx/l.EK activity, PD-3.84352 inhibits the anch.orage.-inde-
`pendent growth of several human tumor cell lines and
`causes cells to adopt a flattened morphology. At similar
`doses, Pl)-184352 also inhibited tumor growth in
`mouse tumor xenograft models (32). The correlation.
`between this surrogate biochemical endpoint and bio-
`logical activity provides strong evidence for mechanism-
`based anti-tumor activity, but lVl.l3ll< inhibitors remain
`at the preclinical development stage.
`
`Blocking lipid-mediated signaling
`Activation, of growth factor receptors is also associated
`with changes in phospholipid metabolism (1-3, 18). in 1
`pathway, the phosphorylated residues on the intracellu-
`lair domain of these receptors bind phospholipase C,
`which then cleaves membrane phospholipids. One of
`these breakdown products, diacylglycerol, can activate
`some forms ofprotein ltinase C (PKC), such as PKG-oi,
`which has been implicated in cell proliferative processes
`and tumorigenesis (34). PKC-oe. expression has been
`found in some human breast tumors to be elevated rela-
`
`tive to surrounding norrna.l tissue. Both antisense in.-
`hibitors to l."l<;C-oz (ISIS-352i.) and inhibitors of PKC
`l(i11£LSE3 activity (CGP 41251 and UCN-01) are in clinical
`
`Th.e_lournal ofClinical lnvestigation
`
`I
`
`January 2000
`
`] Volume 105
`
`] Numberl
`
`11
`
`APOTEX EX. 1010-003
`
`

`
`lar rational molecular approaches for anti—cancer thera~
`pies may also be developed to control cell cycle regula~
`tion and cell cycle checkpoints see Shapiro in this Per-
`spective series), apoptosis {_Sellers and Fisher, this series),
`telomere biology, and an giogenesis Keshet, this series).
`How these different therapeutic strategies can best be
`combined remains an open question. ‘Will it be better to
`have multiple inhibitors targeting different steps of
`growth factor signaling pathways? Or will agents direct-
`ed at tliffer‘eii.t fundamental aspects of a cancer cell prove
`the most effective. combination?
`
`Finally, it is interesting to note that surrogate phar-
`macodynamic endpoints are beginning to be used for
`the development of signal transduction inhibitors (see
`Dru her and Lydon in this Perspective series). In preclin~
`ical animal models, the biological efficacy ofFTIs was
`monitored in relation to inhibition of protein farnesy~
`lation and inhibition of downstream pathways such as
`MAP lzinase and p70 kinase. Lilzewise, inhibition of
`MAP kinase phosphorylation showed a positive corre-
`lation with the an ti~tu mor activity of the M EEK inhibitor
`PD~l843S2. Development of the ISIS antisense com-
`pounds has also been lin l<ed with a reduction in the tar-
`get mRNA levels. This approach has also been carried
`into the clinical development of some of these com-
`pounds, such as has been reported for SCH 66336, L~
`778,123, and ISlS~5 132. Given the genetic complexities
`of cancer, it will be important to analyze whether mon~
`itoring these pharmacodynamic endpoints provides
`useful clinical information, particularly for compounds
`that do not have clearly defined dose—limiting toxicities.
`After all, this is what some believe to be the ultimate
`promise of these agents: lethality to tumors without
`overt systemic toxicity.
`
`pincott—Raven Pu ,
`Channing, I. 15798.
`
`
`
`
`
`
`A
`
`.
`
`
`5... ..vl<o L
`Lg the m
`anism '. ”..
`L 26;6'(?——7Cl.
`2
`‘...l., and Mendel:~:ohn,j. 1998.
`6.Baselga,,., Nortc.
`R .ombinant hum
`in) en hances the
`
`
`I'{El'{2/rieu over-
`.
`.
`, .umc-r activity‘ ofpac taxel
`expressing human breast cancer 2;enografts. C41fi"!C.’,’;'”R:“S. 532825 3834..
`
`
`7.Haselga,>l., et al. 1999. PL...
`‘ Il SLL V ofweekly intravenous trast..zurn~
`
`' g metastatic
`ab (Irlerceptin‘; in pati
`5: wi
`I
`l
`'
`breast cancer. Semm. ()m(;Z. 26..
`8.Sl'\al<, S. 1999, Over '
`monoclonal an tibod,
`tic hr ast cancer '
`
`
`
`
`
`‘
`’
`]., and Bc":nja.n1in,
`
`
`
`
`..1rytox~
`stuzumah (Ilerceptin): p.
`ipatients rece. ing tr‘
`'4: or :;e.rJ,uen1:ial. stress, or surveillance artifact?‘ S./rm!’/i. O"/(col
`
`
`
`
`
`C7‘
`1 1. Ya
`
`mots by a fully
`..
`human monoclonal a
`‘. ‘actor receptor
`,he epiderm .
`,
`'
`
`’ .-.36— 1243.
`notherap CmzcerRe.
`without concomit.
`
`et gal. 13. .
`.
`..
`, an epidermal growth I‘ -tor tyrov
`
`sine lzinase ii abitor selected for clinical development. Proceedings offbe
`Ameiicmi Asraczlii afar (‘miter Resmrcb Amman’ ll/1'eel2':4,g. 58: 33.
`l3.Mo
`‘
`9
`duction 0" goptosis and
`cy e arrest by
`
`
`
`.
`7. owth factor receptor tyrcr.
`"
`
`trials {Table 1). The lzinase inhibitors, both of which are
`derivatives ofstaurosporin e, potently inhibit PKC activi-
`ty and are active in mouse tumor xenograft niotlels
`CGP r1251 also inhibits the P—glycoprotein transporter,
`which mediates the multidrug resistance of many
`atlvancetl tumors. The toxicities noted for UCN-(ll and
`
`CGP 4-125i in the clinic are so far not remarkable, but
`this may be related to the high capacity of these corn-
`pounc s to bind plasma proteins
`a characteristic that
`might also be expected to blunt their anti-ttnnor activity
`(34). The antisense compound ISIS—352.l exhibits an
`acceptable safety profile. Its side effects
`fatigue, fever,
`and thrombocytopenia
`are typical ofphosphoroth-
`ioate~based antisense compounds (20). lSl.S-352 I. is being
`tested in combination with carboplatin and paclitaxel in
`patients with non---small cell lung cancer; preliminary
`data indicate partial, responses in 5 of 8 patients treated.
`In a second pathway, activation of Ras directly activates
`PI 3’-kinase. The product of this reaction is then able to
`activate the protein kinase Al<t, which is a suppressor of
`apoptosis (2). Inhibition of.PI 3’-lc:ina.se activity would
`then be predicted to inactivate Akt activity and subse-
`quently activate apoptotic pathways in tumors. In pre~
`clinical studies, LY 294002 potently inhibited Pl 3’~
`ltinase. This compound inhibits lipid signaling by
`growth factor receptors. In combination with an FTI, it
`was shown to induce apoptosis in attached tumor cells,
`which normally do not respond to FTI. alone (35 This
`result raises the interesting; possibility that inhibitors of
`different steps ofthe signaling pathways may be ofgreat—
`est benefit when used in combin ation.
`
`Conclusions
`
`Growth factor---regulated proliferation pathways eluci~
`dated over the last 2 decades are finally reaching the cl,in~
`ic to be tested. So far, just 1 product, trastuzumab, has
`emerged, but
`its apparent success provides much
`encouragement. This product shows the therapeutic
`value of a treatment based upon a fundamental genetic
`defect in a cancer and raises hopes for other agents, such
`as those summarized in Table 1. It is interesting to note
`how our thinking h changed as the basic research find-
`ings ofgrowth factor signaling have been translated into
`pharmaceutical entities.
`First, it has become clear that these compounds do not
`act solely on tumor tissue. Each agent has a particular
`toxicity that must be managed. In some cases, as with
`EGF receptor inhibitors or FTls, these effects are mech~
`anisin based, but the undesirable consequences ofother
`agents, including phosphorothioate antisense oligonu-
`cleotide compounds, are structure. based. In either event,
`therapies developed on growth signaling pathways offer
`new mechanisms to attaclc: cancer, but they do not nec-
`essarily provide a true cure for cancer.
`Second, we have come to appreciate the value of com-
`bining these new inhibitors with existing therapeutic
`regimens. This realigzation. reinforces the notion that can~
`cer is a disease of multiple and changing genetic alter-
`ations that must be attacked with therapies having dill
`ferent mechanisms of action. Therapies designed based
`on knowl edge of signal transduction, pathways represent
`just I approach to developing new agents. Clearly, simi-
`
`‘E2
`
`Tliejonrual. of"Cl.inical Investigation
`
`E
`
`J;muary 2000
`
`E Volurne 105
`
`E Nurnberl
`
`APOTEX EX. 1010-004
`
`

`
`kihétfir‘
`;‘te7‘1’\./5. 57:48"P»~4848.
`
`Sir1gh,j 1 er al. 1997’. S
`cLur+:—’1;1sed design ofa potem, s;e1ecm":,;1r
`
`
`
`
`irrevctr ible mm‘
`its
`fzhez catalvtic do.
`1.111 ofthe erbi’; mcepr.-Jr sub-
`sen 1{ivn:y 1:0 tazml an-J’, eipiiihli
`
`
`
`
`369- 1571-}.
`
`family 0fpt'oI.ein L‘/I‘
`'
`’ Z‘\/fed. Clyem. 4431130 1135.
`"<:khar<“
`'
`
`
`
`1998.
`
`
`
`
`
`
`
`
`
`
`if
`6 36, an am. y bi0a.vai]--
`n trzmsferase, in :;L1}'E]21I'1 Lurnc-r
`'c m;7<:<.>. Cmxc./31* R125. 53:4S747r—4S756.
` rC()Tl"
`ass ofn-;m—L‘
`the Amemrzn Axsacizzt‘
`p
`
`
`Kl‘3.US[=‘I’2L‘3:3 and gemn
`, T:{-311g-3, A., E‘..11E:-
`FIE "
`C £lC'j€I]
`[S ..
`get'any1I.t'ansfer .
`1: com
`tic:-n .. ‘rapy v./iu.
`
`
`
`
`
`_.
`Cm2cerR:‘=s. 59:491..
`in, Taxol, and gemcitzx
`L\‘,x5
`. Gtaff, I\..)i;1r2g. W., Yang, J, and [5*]()rit1i,). 1'09. Varizzticm
`. H all :1 ackscm, (LA, at :1]. 199‘). Parzzdoxi-:31‘. zzctivaiion (1J‘}l;1.fb}/ 9. novel
`Rafiuhibiz-Jr. Chem. Biol. 6:55.‘?
`'
`
`
`in the Jgnaling paxhways by which IC-
`
`30. }{;z]1J.L- \S(3Tl, (LA.
`53 703580 on thc.
`1320
`
`
`
`
`em. 62:851—891.
`
`
`and Don‘, F./\. 1999. Toward
`, Mania, BR, Kw-ah, '
`olmlund,
`zmLis;€nse ()]1gcmuc1e-uide Lr2er;1pyfot'cancet‘: $18 cc:-mpounds in -:hni—
`
`
`cal dezvelo} "nem. CTmv:~'/zr {,)1*)i1/::' 7; in Zt1r)Z:‘=wl:1rI7)empeuiic5. 1:3 72385.
`21. ()]iET, A. 1‘)
`/. Fazrne‘ ‘y]Lr;1nsfer;zseinhibizorsz tazrgazing the mc-]ecL11;zr
`
`
`
`'
`h‘. Biofibfys. Arm. 1423‘ 19
`‘
`'9. lrzhibiwrs (1J‘pt'c:-Lain prenyizxti-;m.
`11 ofa HOV-';‘] inhibiuz-r ofrniLc:-gen—
` 2:1
`
`
`Z'1—l38-0.
`ase. J. Biol. Chem. 273:1-3623—l86;”w2.
`jp2x€€11E:3.
`£5 ‘
`aczivamd prozein kinase
`l‘am;m-Ji, F.
`1
`drugs?
`34. L ‘scher, A. 1998. Analcz-gs ofstzzumspcmnez p0Lentia1;mI.ic;m
`
`
`
`inhibizors induce C}’[OL me c release and caspase 3 aczivailon prefab
`Sew. P/)4}/77:’/lrlrfloll. 31:72l—728.
`
`7: 01‘ the P'3'K—AKT
`czivgn.
`
`
`entiafly in I.t'ansfot'rned cm Prat. ,\/1112. Aczui
`'\.Lz. USA. 95:1S356— 15
`D‘-.1, W
`
`
`
`Z4.Eerrante,K.,\X/ ogr;Ld,B,,.md (Lmetta, R.
`the p.n popmtic effects of .
`.nesy'1tr;n1sferase
`.9 Promising new deve1—
`p;zt'm'my masks
`
`(Lmcew Cbwnotlyer. P0117“/natal.
`inhibiL<1rs;.Qmcerfles.59:42O8—4Z12.
`opmsms in ca :+:r chemothergzpy.
`
`20.
`
`
`
`
`’I'b,e_}'<>urna10fC1inica] Tzwestigatti-on
`
`I
`
`]2m,uzary20O()
`
`] Valmne 105
`
`] Number 1
`
`1 3
`
`APOTEX EX. 1010-005

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