throbber
13
`
`
`
`. Tyrosine Kinase Inhibitors Against
`EGF Receptor-Positive Malignancies
`
`Elise A. Sudbeck, Sutapa Ghosh, Xing-Ping Liu,
`Yaguo Zheng, Dorothea E. Myers, and Fatih M. Uckun
`
`1. Introduction
`The role of protein tyrosine kinases (PTKs)in the survival of cancer cells
`and their potential use in anticancer therapy has led to their selection as anti-
`cancer drugtargets. Tyrosine kinases which are being studied for this purpose
`include epidermal growth factor receptor (EGFR) (—-6), Janus kinases (JAKs)
`(7-13), Bruton’s tyrosine kinase (BTK) (14-16), platelet-derived growth fac-
`tor (PDGF) (17), protein kinase C (PKC) (18-24), Lek (25,26), Trk (27-30),
`and others. The strategies used to attenuate or disable kinases implicated in
`cancer includethe use of antibodies, immunoconjugates, ligand-binding cyto-
`toxic agents, and small-molecule inhibitors. Each of these strategies has shown -
`some promise for the treatment of cancer. Herceptin (31-35), for example,is
`an immunotherapeutic agent that binds to the extracellular domain of HER2
`(also referred to as ErbB-2, a tyrosine kinase belonging to the same family as
`EGFR) at nanomolar levels. EGF-genistein (BGF-gen)is an EGFR-binding
`cytotoxic agentthat also showspotency in the nanomolar range (2,36) and will
`be discussed in this chapter. The search for new small molecules that inhibit
`kinases has involvedtraditional approaches, including the testing of natural
`products, random screening of chemical libraries, the use of classical struc-
`ture—activity relationship studies, and the incorporation of structure-based drug
`design approaches and combinatorial chemistry techniques. As a result, sev-
`eral promising small-molecule inhibitors have also been identified in recent
`years that may prove useful as potent new anticancer drugs.
`Small-molecule inhibitors of kinases that show. promise as anticancer agents
`include inhibitors of EGFR. EGF exerts pleiotropic biologic effects by binding
`
`From: Methods in Molecular Biology, vol. 166: Immunotoxin Methods and Protocols
`Edited by: W. A. Hall © Humana Press Inc., Totowa, NJ
`
`193
`
`j
`
`|
`
`j |
`
`
`
`OSI EXHIBIT 2003
`APOTEX V. OSI
`IPR2016-01284
`
`OSI EXHIBIT 2003
`APOTEX V. OSI
`IPR2016-01284
`
`

`

`194
`
`Sudbecketal.
`
`to ErbB-L (37-40). In breast cancer, the expression of EGFR is a significant
`and independent indicator for recurrence and poorsurvival (41-43). Recent
`studies provided evidence that EGFR serves as an endogenousnegative regu-
`lator of apoptosis in breast cancer cells (2). Many classes of small molecules
`have been reported in recent years that inbibit EGFR kinase (1,3-6,44~-47).
`Genistein (Fig. 1), a naturally occurring isoflavone found in soybeans,is an
`inhibitor of EGFR (6). When genistein is linked to EGF, the potency against
`EGFR increases from an ICsy value of >10 1 to a value in the nanomolar
`range (2).
`Pyrazolopyrimidines(see Fig. 1) were foundto inhibit EGFR with ICsp val-
`ues ranging around 1-8 nM (3). Two pyrazolopyrimidines with reported ICs
`values below 10 nM (3) also showedhigh selectivity towards some nonreceptor
`tyrosine kinases (c-Src, v-Abl, and serine/threonine kinases such as PKC-a
`and CDK1). The quinazoline derivative CP-358,774 (45) inhibits EGFR with
`an ICsg of 2 nM and reduces EGFR autophosphorylation in intact tumorcells
`with an ICsq of 20 nM. This inhibition is selective for EGFR relative to other
`tyrosine kinases examined as determined by assays of isolated kinases and
`whole cells. Despite the reported profound in vitro potency (K; = 5 pM) and
`selectivity of the ATP-competitive brominated quinazoline derivative PD153035
`(ig. 1; 4,5), the compoundfailed to show significant in vitro-orin vivoeffi-
`cacy against cancer cells. Other quinazolines reported include PD168393 and
`PD160678, which selectively target and irreversibly inactivate EGFR through
`covalent modification of a cysteine (Cys?”*) residue presentin the ATP-bind-
`ing pocket (44). These compoundsalsointeract in an analogous fashion with
`ErbB2 (which has a conserved Cys residue at the same position) but have no
`activity against IR, PDGFreceptor, FGFR, and PKC. The compoundshave not
`been tested against BTK and JAK3, which also contain conserved cysteine
`residues at the correspondingposition.
`A series of new quinazoline compoundstargeting EGFR have been designed
`more recently using structure-based methods.In this study, a three-dimensional
`modelof the kinase domain of EGFR wasconstructed (I) using known coordi-
`nates of homologous kinase domains as reference coordinates (Hematopoietic
`cell putative protein tyrosine kinase [HCK; 48], fibroblast growth factor recep-
`tor [FGFR; 49,50], and insulin receptor kinase [IRK; 57]). The EGFR model
`was used along with an inhibitor docking procedureforthe rational design of
`compoundspredicted to bind favorably to EGFR. The EGFR modelindicated
`that inhibition may be significantly improved by increasing the sizeof the func-
`tional groups attached to the 4-anilinoqninazoline molecular scaffold. Chemi-
`cally relevant substitutions at the 3', 4’ and 5' positions on the anilino ring lead
`to the successful design of a dibromo quinazoline derivative, WHI-P97, with
`an ICs9 value of 2.5 1M in EGFR kinase inhibition assays. WHI-P97 effec-
`
`

`

`EGFRInhibitors for Treatment of Cancer
`
`195
`
`.
`
`,
`
`N
`
`om
`:
`ahhBr
`a
`7
`Oo.
`HO
`Cy
`YN NH
`. C }
`W?.
`a He
`CO on
`OHO
`Pyrazolopyrimidine
`PD153035
`Genistein
`.
`derivative
`(,7,4'-trihydroxyisoflavone)
`HOURx =:52pM@
`
`
`
`4-6 nM (3), 101M (46)
`
`EGFR IC,)> 102M so =25pMEGFR IC,, = 1 nM (5),
`
`
`
`OQ. oeoseoO
`
`WHI-P97
`EGFR IC, = 2.5 pM
`
`LFM (A77 1726)
`EGER IC,= 5.4 1M
`
`PD168393
`EGER IC,, = 0.45 nM
`
`WHI-P154
`EGER IC, = 5.6 1M
`
`H3
`
`a
`
`ay
`
`LFM-A12
`EGER IC,, = 1.71M
`
`PD160678
`EGFR IC,, = 0.70 nM
`oO
`
`WHI-P131
`JAK IC, = 9.1 1M
`EGFR IC,, = 4.2 pM
`Br
`
`Hs!
`
`ng
`
`N Ni
`
`it
`
`LEM-A13
`BTK IC,, = 2.5 1M
`
`Fig. 1. Examples of tyrosine kinase inhibitors.
`
`tively inhibited the in vitro invasiveness of EGFR-positive human cancer cells
`in a concentration-dependent manner. The quinazoline derivatives WHI-P97,
`WHI-P131, WHI-P154, WHI-P180, and WHI-P197 with 3’ or 4'OH substitu-
`tion on the anilino moiety were predicted to form an additional hydrogen bond
`with Asp®3! in the ATP-binding region of EGFR that may enhancebinding.
`The EGFR inhibition values for WHI-P97, WHI-P131, WHI-P154, WHI-P180,
`and WHI-P197 ranged from 2.5 to 5.6 [LM in kinase assays. However, the
`
`

`

`196
`
`Sudbecket al.
`
`quinazolines tested were not specific for EGFR. For example, the EGFR inhib-
`itor WHI-P154 (EGFR ICsp = 5.6 LM) also inhibited other tyrosine kinases
`such as HCK (ICsp = 12 WM), JAK3 (Cs = 130 11.) andspleen tyrosine kinase
`(SYK) (C59 = 150 1) (46).
`‘

`In terms of selectivity for EGFR, leflunomide metabolites may show the
`most promise. Earlier studies reported that the immunosuppressive activity of
`leflunomide is due to its metabolite A77 1726 (a-cyano-B-hydroxy-B-methyl-
`N-[4-(trifluromethyl)phenyl]-propenamide, or LFM), which is rapidly formed
`in vivo, functions as a pyrimidine synthesis inhibitor (52) and also inhibits the
`tyrosine kinase activity of EGFR (53). The leflunomide metabolite analog
`LEM-A12 (Fig. 1) showed inhibition of EGFR with an ICs9 value of 1.7 1M
`and killed >99% of human breast cancercells in vitro bytriggering apoptosis
`(1). Both LFM-A12 and WHI-P97 inhibited the in vitro invasiveness ofEGFR-
`positive human breast cancer cells at micromolar concentrations and induced
`apoptotic cell death. In addition, LFM-A12 inhibited the proliferation (ICs) =
`26.3 uM) andin vitro invasiveness (ICs5y = 28.4 1.4) of EGFR-positive human
`breast-cancercells in a concentration-dependentfashion.
`Like the quinazolines WHI-P97 and WHI-P154, the design of LFM-A12
`wasaided by a model of the EGFR kinase domain.In kinase assays, LFM-A12
`was found to be specific for EGFR and did not inhibit other-PTKs such-as
`BTK, HCK, JAKI, JAK3, IRK, and SYK at concentrations ranging from 175
`to 350 jtM. The observedselectivity of LFM-A12 for EGFR likely results from
`its molecular shape and from favorable interactions with unique EGFR resi-
`dues that are not present in the kinase domains of other PTKs. Likewise, unfa-
`vorable interactions with unique residues of other PTKsthat are not found in
`the EGFR kinase domain may also contribute to this selectivity. This observa-
`tion is in contrast to the observed inhibition of several kinases (EGFR, HCK,
`JAK3 and SYK) by WHI-P154. Thefirst contributing factor for the nonselec-
`tivity of WHI-P154 maybe the inhibitor’s complementary shape with the hinge
`region of the binding cavity of all seven kinases, which in turn leadsto favor-
`able hydrophobic contact between the compoundand the residuesin this cav-
`ity. Additionally, predicted hydrogen bonding interactions with all seven
`kinases may enhanceits binding with each of them.
`The structure-based method used to design leflunomide inhibitors of EGFR
`was also successfulfor the identification of small-molecule inhibitors of JAK3
`(7). JAK3 is expressed abundantly in primary leukemic cells from children with
`acute lymphoblastic leukemia (ALL). The constniction of a three-dimensional
`model ofJAK3 (7) was used to design a quinazoline inhibitor, WHI-P131 (ig. 1,
`shownto have specificity for JAK3. WHI-P131 inhibited JAK3 (Cs) = 9.1 pM)
`but not JAK1 or JAK2 anddid notinhibit the ZAP/SYK-family tyrosine kinase
`SYK, TEC-family tyrosine kinase BTK, Sre-family tyrosine kidase LYN, or
`
`

`

`EGFRInhibitors for Treatment of Cancer
`
`197
`
`the receptor-family tyrosine kinase IRK, even at concentrationsas high as 350 1M
`(7). WHI-P131 induced apoptosis in JAK3-expressing human leukemia cell
`_lines but not in JAK3-negative melanoma or squamouscarcinomacells.
`Anotherstudy to identify kinase inhibitors focused on inhibitors of BTK as
`antileukemic agents with apoptosis-promoting properties (14). A three-dimen-
`sional homology model of the BTK kinase domain was constructed(14) and
`- inhibitor docking proceduresled to the identification of an LFM analog, LFM-
`.A13 (Fig. 1), which was found to be a potent and specific inhibitor ofBTK. LFM-
`AJ3 inhibited recombinant BTK with an IC.value of 2.5 [LM,butit did notaffect
`the enzymaticactivity of other protein tyrosine kinases including JAK1 and JAK2,
`Sre-family kinase HCK, and receptor-family tyrosine kinases EGFR and IRK,at
`concentrations as high as 278 iM. LFM-A13 also enhaiiced the chemosensitivity
`of BTK-positive B-lineage leukemiacells to vincristine atidCeramide.
`Although several agents have been identified in recentyears that inhibit tyro-
`sine kinases such as EGFR, JAK3, and BTK,a future challengeis to ensure the
`specificity of inhibitors for one targeted tyrosine kinase. A successful strategy
`to accomplish this involves conjugating small-molecule inhibitors to ligand-
`binding entities; this enables the inhibitor to be delivered to a specific tyrosine
`kinase. An example of this strategy is to link a kinase inhibitor (soybean-
`derived genistein, 5,7,4'-trihydroxyisoflavone) to a protein (recombinantliuman
`EGF)that binds to a receptor kinase (EGFR). The resulting protein-inhibitor
`conjugate is an EGFR-directed cytotoxic agent (EGF-gen) with PTK inhibi-
`tory activity (2,36), which will be describedin further detail in this chapter. (A
`similar method was successfully appliedto the targeted delivery of genistein to
`CD19-receptor—associated vital PTK and shows considerable promise for more
`effective treatment of human leukemias and lymphomas/25,54))
`
`2. Materials and Methods
`
`2.1. Structure-Based Design of Small-Molecule Inhibitors of EGFR
`
`The three-dimensional coordinates ofthe EGFR kinase domain used in protein—
`inhibitor modeling studies (Fig. 2) were constructed based on a structural align-
`mentof the sequence of EGFR with the sequences of knowncrystal structures of
`other protein kinases (kinase domains ofHCK[48], FGFR [50], IR[55], and cAPK
`[56)) as described previously (1). The procedure was also usedto construct homol-
`ogy models for JAK1, JAK3 (7), BTK (14), and SYK (Mao, C., unpublished data).
`Molecular docking and scoring procedures were used to estimate binding of
`inhibitors in the catalytic site of EGFR (Tables 1 and 2, Figs. 3 and 4)(1,47).
`Leflunomide metabolite analogs such as LFM-A12 (Scheme1) and quinazoline
`compounds such as WHI-P97 (Schemes 2&3) were synthesized, and their ability
`to inhibit EGFR in breast cancercells was tested as previously described (1,57).
`
`

`

`198
`
`.
`
`Sudbecketal.
`
`2.2. EGF-Genistein Immunoconjugates
`2.2.1. Preparation ofEGF-Genistein Immunoconjugates
`Recombinant human EGF (chEGF) was produced in £. coli harboring a
`_ genetically engineered plasmid that.contains a synthetic gene for haman EGF
`fused at the N-terminus to a hexapeptide leader sequence for optimal protein
`expression and folding. The rhEGF fusion protein precipitated in the form of
`inclusion bodies, and the mature protein was recovered by trypsin cleavage
`followed by purification using ion-exchange chromatography and HPLC (2,36).
`The recently published photochemical conjugation method using the hetero
`bifunctional, photoreactive crosslinking agent sulfosuccinimidyl 6-(4’azido-2'-
`nitrophenylamino)hexanoate (Sulfo-SANPAH) (25)-wasused to prepare the
`EGF-genistein (EGF-gen) conjugate. Photolytic generationOf areactive sing-
`let nitrene on the other terminus of EGF-SANPAH in the presenceof a 10-fold
`molar excess ofdifferentially hydroxyl-protected genistein (gen) resulted in
`the attachmentofgenviaits available C7-hydroxyl group to the Lys?® or Lys*®
`residues of EGF. The resulting sample was purified using size-exclusion
`HPLC, and reverse-phase HPLC (2). Electrospray-ionization mass spectrom-
`etry (62,63) was used to determine the stoichiometry of gen and EGF in EGF-
`gen. }*J-gen was also used to confirm the stoichiometry of gen and EGFin
`EGF-gen and to verify the removal of free gen and gen-labeled EGF-EGF
`homoconjugates by the described purification procedure. The purity of EGF—
`125]_sen was assessed by SDS-PAGEandautoradiography.
`2.2.2. Binding of EGF-'*5|-Gen to Breast Cancer Cells
`Ligand-binding assays using EGF—!5I-gen (2.0 x 108 cpm/pimol), !*I-gen
`(3.8 x 108 cpm/umol) and !4I-EGF (2.2 x 10'* cpm/imol; Amersham) were
`performed using standard procedutes as previously described (25,64). Thecell
`lines in ligand-binding assays included the EGFR-positive breast cancer cell lines
`MDA-MB-231 and BT-20, as well as the EGFR-negative human leukemiacells
`lines NALM-6(pre-B leukemia) and HL-60 (promyelocytic leukemia).
`
`Fig. 2. (opposite page) Themolecular-surface representation of the homology
`model of the EGFR kinase domain. A small-molecule inhibitor (multicolor) is shown
`docked into the ATP-bindingsite (active site, yellow) ofEGFR. Taken fromn Ghosh et
`al., (1998) Clin. Cancer Res. 4, 2657-2668 (1).
`
`Fig. 3. (opposite page) Docked position ofLFM-A12 (yellow)in the catalytic site of
`the EGFR kinase domain model. The EGFR catalytic site residues are shown as space-
`filling atoms. Taken from Ghoshetal. (1999) Clin. Cancer Res. 5, 4264-4272 (46).
`
`
`emeeeee
`
`Nemaueearned:Petetenenets
`
`vere
`
`

`

`
`ee —T
`
`Fig.2.
`
`

`

`
`
`200
`
`.
`
`Sudbecketal.
`
`eat
`
`Raatg
`
`
`Fig. 4. The superimposed docked positions of several 4-anilinoquinazolines-in the
`catalytic site of the EGFR kinase domain model. Taken from Ghoshet al. (1999)
`Anticancer Drug Des., 14, 403-410 (47).
`
`Fig. 5. The effects of EGF-gen on tumor progression in SCID mice xenografted
`with MDA-MB-231 human breast cancer cells. White arrows indicate the approxi-
`mate size of the tumors in (A) a PBS-treated mouse (control) and (B) an EGF-gen—
`treated mouse after 60 d. Ruler units shown are in centimeters.
`
`—
`
`asmeeAeeteiaee
`
`

`

`“S—
`
`al,
`
`the
`19)
`
`[Japss00e7Zwos
`
`
`
`
`ISSEVCVCIN§«oTet-ag-vaN«(AM)DTTi)Ey,oDg:SsBION(%)(y)xpunoduop
`
`
`
`
`
`
`
`
`
`
`
`0}yuveur)azoydssqozdAueunpimouinjoajoArepunogsyjsepouoq*(69)urexdoidsysAffouuoDBusnpaye]nayedwaresoRpINsJeINI2;0Wy
`S0616861vs880S(145S0O®—s«OSZ*yo-dWaT
`£97vesLtel68osS6Ioe=DOF=ZLV-WT
`ry;Lsige.HeNO
`O0E<o0E<oor<oot<OvE9€POLIOZ=JGIP-S‘TSsELV-INST
`00€<o0€<SbL6991PO€pOLI0%ro|LY-WATvoniqiyuyYADs‘sone,'ypayzinayeg‘saioaguopoeIa}U]
`
`
`
`aoe<O0E<-O00T;<oat<vee16€Sol«EZIo“=O1V-WAT00E<00E<oor<oor<SpESBELSI,Le1ID- OV-WATo0E<o0E<-001<2%Sov26b681orefySY-WATaoe<O0E<oot'<ool<pSLOE8919€Zig-u6V-WHT
`
`
`00E<00€<oot<O01<LOEL6E.8STplzeUuON=PLYgoe<00E<ooli<ool<L8€aorE9181ZauTIV-WaT
`o0E<o0E<ool<001<Z9€L6E9112td€V-WIT
`
`Q0E<o0E<001<oor<SEtrp1816%"19-9PVT
`
`00g<ode<ool<001<E8Eber891OE1g-0SY-WAT
`
`O0E<00E<ool<LEery9p0818@Zlod=@V-WTone<-Q0E<oor<ze6¢P2pLLI9%jg=1V-WAT
`Gant)5]Anorxojorh-yuOnIqIyuT=IPN'TpnydeygsiSW
`
`
`
`
`Sy(1)sBojeuyWAT]0Ayo}xo}O}AQ[199J89UeDeAnISOg-YADTpue‘(°%D))
`
`
`
`
`
`
`
`
`(a2)(WD‘O8sICues‘sHOTENWISIENIS|OWE“TLSIsu])wresTordipuryaryuyuorourysso0syeownduraay)uopaseLytpn],
`
`
`
`
`
`
`
`‘suonisodpayoopuopaseq‘IpmryAqpoyepnoreourajosdtrayoeIMOOUrgoRTINSTe[Nds[OUrJoadeUsoIedety‘soRJINSPILING,
`
`‘gnoajoursipdnoyeuryeuysworeazoydspreyIIAOuINIOAOUBuLTEYsSNIPEwaATsJo(o[Naa[oUT3ayeMvJUasaidar
`
`
`
`
`
`
`
`
`
`
`
`
`(I)8997-LSOT“pSAN420UDD"UND(8661)Te39YsoUDWayuoyEL
`
`
`
`201
`
`
`
`

`

`202
`
`.
`
`Sudbecketal.
`
`Table 2
`EGFRInteraction Scores, Estimated K;, Values,
`
`and Measured!Cz, Data for 4-Anilinoquinazolines
`
`EGFR
`MS? BS? Lipo Ludi?—InhibitionLudi
`
`
`Compound R3 R4
`RS (A2
`(A?) Score HB‘ Score ‘Kj(uM)
`ICs (UM)
`WHI-P79
`Br H
`H 302
`208
`610
`1
`570
`2.0
`10.0
`(PD153035)
`2.5
`0.09
`701
`2
`655
`224
`329
`Br OH Br
`WHI-P97
`4.0
`0.60
`622
`1
`661
`226
`WHI-PI11 Br CH; H 305
`4.2
`0.40
`639
`2
`593
`202
`WHI-P131
`H OH H 290
`5.6
`0.14
`685
`2
`639
`218
`WHI-P154 Br OH H 307
`645 036 __ 40
`2
`599
`204
`WHLPI80 OH H
`H 292
`
`
`
`
`
`
`215 629 2 675 0.18WHLP197 Cl OH H 298 3.5aavaOC
`
`“Molecular surface area calculated using Connolly’s MS program (69). Defined as the bound-
`ary of volume within any probe sphere (meant to represent a water molecule) of given radius
`sharing no volumewith hard sphere atoms that make up the molecule.
`’8uried surface, the molecular surface in contact with protein calculated by Ludi based on
`docked positions.
`°The numberof hydrogen bonds between the protein andthe inhibitor.
`“Ludi K; values were calculated based on the empirical score function in the Ludi program
`(71). Ideal hydrogen bond distances and angles between compoundsandprotein are assumedin
`all cases for Ludi score and KX; calculation.
`Taken from Ghosh et al. (1999) Anticancer Drug Des. 14, 403-410 (47).
`
`2.2.3. Immunocytochemistry of EGF-Gen
`Immunocytochemistry was used to examine the surface expression of EGFR
`on breast cancer cells, to ‘evaluate the uptake of EGF-gen by breast cancer
`cells, and to examine the morphologic features of EGF-gen-treated cancercells.
`To detect the EGFR-EGF-gen complexes, cells were incubated with a mixture
`of a monoclonal antibody directed at the extracellular domain of haman EGFR
`and a polyclonal rabbit anti-gen antibody (2). After rinsing with PBS,cells were
`incubated with a mixtureof a goat antimouse IgG antibody conjugated to FITC,
`and donkeyantirabbit IgG conjugated to Texas Red, Cells were washed in PBS,
`counterstained with toto-3, and viewed using a confocal microscope.
`
`

`

`EGFRInhibitors for Treatment of Cancer
`
`203
`
`noJL, +nn\cits OG J,
`ciisoprepy
`.
`2-
`
`1
`
`HeFLT
`oO”
`iLFM-AI2
`
`CHscoct
`
`Scheme1. Synthesis of LFM-A12 (59,60).
`
`sO R ——r CH30
`
`CH30
`
`CHO
`
`R=3'5-diBr, 4-0H
`or 3°-Br,4-OH
`
`3
`4
`i ‘ 5
`=p
`
`HN
`
`SN
`
`2 7
`WHI-P97: A = 3'5'-diBr, 4'-OH °
`WHI-P154: R = 3'-Br,4'-OH
`
`Scheme 2. Synthesis of quinazoline derivatives WH1-P97 and WH1-P154 (58).
`
`9°

`1)s
`CH,;0 OH1)80, CH30 NH, _£uSO4
`
`
`CH30
`2) NH,OH
`CH30:
`NaBH:
`wee
`NOz
`NO,
`
`(1)
`
`(@)
`
`1
`9
`
`
`
`
`CHy
`CH30
`CHO
`2
`NH2
`N
`
`CH; NH, _HCO,H=CHs0 POC CH30 “N
`
`(3)
`
`‘
`
`(5)
`
`Scheme 3, Synthesis of a quinazoline derivative precursor (61,62).
`
`2.2.4. In Vitro Treatment of Cells with EGF-Gen
`
`In order to determine the cytotoxic activity of EGF-gen against breast
`cancercells, cells were treated with various concentrations of EGF-gen and
`then used in either apoptosis assays or clonogenic assays (2). Controls
`included cells treated with G-CSF-gen (an irrelevant cytokine—gen conjugate
`that does not react with EGFR), cells treated with unconjugated EGF plus
`unconjugated gen, cells treated with unconjugated gen or unconjugated EGF,
`and cells treated with PBS, pH 7.4. In some experiments, excess G-CSF or EGF
`’ was added to the EGF-gen—containing treatment medium to showthat the cyto-
`toxicity of EGF-gen can beselectively blocked by excess EGF but not G-CSF.
`
`
`
`[men
`
`

`

`204
`
`.
`
`Sudbecket al.
`
`2.2.5. Immune-Complex Kinase Assays and Antiphosphotyrosine
`Immunoblotting
`
`-
`
`After treatment with EGF-Gen, cells were stimulated with EGF and cell
`lysates were immunoprecipitated with an anti-EGFR antibody reactive with
`the Ala35!_Asp*64 sequence of human EGFR. EGFR immune complexes were
`examined for tyrosine phosphorylation by Western blot analysis as previously
`described (66). All antiphosphotyrosine Western blots were subjected to den-
`sitometric scanning and for each time point a percent inhibition value was
`determined.
`
`2.2.6. Apoptosis Assays Using EGF-Gen
`
`MC540 binding and propidium iodide (PI) permeability (indicators of
`apoptosis) were simultaneously measuredin breast cancercells after exposure to
`EGF-gen(either without any cytokine preincubation or following preincubation
`with excess unconjugated EGF or G-CSF), unconjugated gen, unconjugated EGF
`plus unconjugated gen, or G-CSF-gen,as previously described (66). To detect
`the DNA fragmentation in apoptotic cells, cells were harvested after treatment
`with EGF-gen andDNAwas prepared from Triton-X-100detergentlysates for
`analysis of fragmentation as previously described (66).
`-
`van
`
`2.2.7, Clonogenic Assays
`
`After treatment with EGF-gen, G-CSF—gen, unconjugated EGF, unconju-
`gated gen, or PBS, cells were resuspended in clonogenic medium. Cells were
`cultured and cancer-cell colonies were enumerated on a grid using an inverted-
`phase microscope of high optical resolution. Results were expressed as the
`percent inhibition of clonogenic cells at a particular concentration of thetest
`agent. Dose-survival curves were constructed using the percent control sur-
`vival results for each drug concentration as the data points, and the IC.) values
`were calculated.
`
`2.2.8. Crossreactivity of Human EGF and Antihuman EGFR
`Antibodies with MouseEGFR
`Livers and thymus of BALB/c mice were frozen in liquid nitrogen and 5 pm—
`thick tissue sections were prepared using a cryostat. The sections were pro-
`cessed for standard indirect immunofluorescence using a monoclonal antibody
`directed at the extracellular domain of human EGFR as the primary antibody
`and a goat antimouse IgG conjugated to FITC as the secondary antibody (2). In
`parallel, sections were also stained by direct immunofluorescencestaining tech-
`niques with FITC-conjugated EGF.
`
`

`

`peneeeeumnaetnaornRennerCoNPAA
`
`pe
`
`EGFRinhibitors for Treatment of Cancer
`
`205
`
`2.2.9, Mouse Toxicity Studies
`The toxicity profile of EGF-gen in BALB/c mice was examinedas previ-
`ously reported for other biotherapeutic agents (25,67). In single-dosetoxicity
`studies, female BALB/c mice were administered an intraperitonealbolus injec-
`tion of EGF-gen in 0.2 mL PBS, or 0.2 mL PBSalone (control mice). In cumu-
`lative toxicity studies, mice received a total of 2800 lg (140 mg/kg) EGF-gen
`intraperitoneally over 28 consecutive days. Mice were monitored daily for
`mortality to determine the 30-d LDsg values (36).
`
`2.2.10. Treatment of SCID Mouse Xenograft Model of Human
`Breast Cancer
`The left hind legs of CB.17 SCID mice were inoculated.subcutaneously with
`MDA-MB-231 breast cancercells in 0.2 mL PBS. SCID miceinoculated with
`human breast cancer cells were treated with EGF-gen (36). Mice were moni-
`tored daily for health status and tumor growth. Primary endpoints of interest
`were tumor growth and tumor-free survival outcome. Estimationoflife table
`outcome and comparisons of outcome between groups were doneas previ-
`ously reported (6,25,67). The efficacy of EGF-gen against established tumors
`was examinedby treating SCID mice with subcutaneous MDA-MB-231 xeno-
`grafts with EGF-gen on 10 consecutive days and determining the tumor diam-
`eter daily for 20 d from the start of therapy. Control mice weretreated with 0.2
`mL PBSfor 10 consecutive days.
`
`2.2.11. Pharmacokinetic Studies of SCID Mice Treated
`with EGF-Genistein
`Tissue distribution studies in SCID mice were performed using EGF—!*I-
`gen and !?5]-gen as described previously (25). A flow-limited physiologic phar-
`macokinetic model was used to characterize the tissue disposition of EGF-gen
`in non—tumor-bearing as well as tumor bearing SCID mice (25,36,68).
`
`3. Results
`
`3.1. Structure-BasedDesign of Small-Molecule Inhibitors of EGFR
`3.1.1. Predicted Binding of LFM-A12 with the EGFR Kinase Domain
`Based on modeling studies of the kinase domain of EGFR, a binding mode
`was proposed for LFM analogs (Z). The predicted binding mode allows LFM
`analogs to maintain close contact with the hinge region of EGFR.Theinhibitor
`can fit into a space in the EGFR catalytic site defined by Leu®™ and Val’ on
`one side, and Leu®?° and Thr®*? on the other. Thenitrile nitrogen of LFM-A12
`waspredicted to interact with the amide of Met’®? via hydrogen bonding. In
`
`

`

`206
`
`.
`
`Sudbecketal.
`
`addition, the para-substituted OCF; group on LFM-A12 can form close con-
`tacts with residues Thr’®and Asp®#!, Interaction scores, calculated K, values,
`and measured IC.9data for several LFM analogsarelisted in Table 1. Of the
`compoundspredicted to interact most favorably with EGFR, LFM and LFM-
`. Al2 showedthe mostpotent activity against EGFR in kinaseassays, and LFM-
`A12 was the most cytotoxic against breast cancer cells.
`
`3.1.2, Modeling Studies of 4-Anilinoquinazoline Derivatives
`with the EGFR Kinase Domain
`
`Kinaseinhibition properties have also been evaluated for derivatives of 4-ani-
`linoquinazoline(4,5,47). In modeling studies aimed at identifying quinazoline
`derivatives with a high likelihood to bind favorably to theCatalytic site of the
`EGFR, K, values were estimated based on predicted binding interactions
`- between the inhibitor and catalytic-site residues of the EGFR (47). The model
`of the EGER binding pocketwas used in combination with docking procedures
`to predict the favorable placement of chemical groups with defined sizes on a
`molecular template. These studies led to the design of six quinazoline deriva-
`tives, WHI-P97, WHI-P111, WHI-P131, WHI-P154, WHI-P180, and WHI-
`P197. The various docked positions of each quinazoline dérivative were
`qualitatively evaluated in terms of an estimated K; value and consequently com-
`pared with the ICs values of the compounds in EGFR kinaseinhibition assays.
`Table 2 lists the interaction scores and estimated K;, valuesfor the quinazoline
`derivatives. This most favorable docked position allowedthe quinazoline inhib-
`itor to maintain close contacts with the hinge region of EGFR. The quinazoline
`moiety in the molecule can align itself along the hinge region ofEGFR,andthe
`N1nitrogen ofthe quinazoline group can form a hydrogen bondwith the back-
`bone carbonyl atom of the EGFR Met’?residue.
`In the final docking mode, the 6,7-OCH; groups ofthe inhibitor faced the
`solvent accessible region, and the anilino ring was surrounded by residues
`Thr76, Asp®3!, Thr®3°, and Val”, This modelofthe anilinoquinazoline bound
`to the EGFR kinase domain is consistent with that reported by others (71).
`WHI-P97, WHI-P131, WHI-P154, WHI-P180, and WHI-P197 with 3’ or 4'-OH
`substitutions on the anilino moiety can form an additional hydrogen bond with
`Asp®3!, WHI-P79 was the least potent inhibitor in the series evaluated,fol-
`lowed by WHI-P111. The activity of these two compounds maybe affected by
`the absenceof 3' or 4'-OH substitutions that can form a second hydrogen bond
`with EGFR residues and enhance binding.
`The crystal structures of the HCK—quercetin complex (48) and two FGFR—
`inhibitor complexes (50) revealed that PTK inhibitors can bind to the PTK
`catalytic (ATP-binding) site. When the catalytic sites of these PTK crystal
`
`

`

`ereaeneaaceetdAoeaNeeeeecereeeeARnAN
`
`structures were superimposed, all atoms of the three PTK inhibitors present
`fell within the plane of a triangle defining the ATP-binding region, and each
`molecule was in close contact with the hinge region and a conserved Aspresi-
`due (Asp*?! in EGFR). Theinhibitors characteristically occupied only half of
`the bindingsite, closest to the hinge region. The fact that the inhibitors reside
`close to the hinge region seemsto correlate with tighter binding and may be an
`important determinant for inhibitor binding. In the case of EGFR,the size and
`relatively planar shapeofthe catalytic site within the constructed EGFR kinase
`domain may contribute to its ability to form favorable interactions with mol-
`ecules such as quinazoline derivatives and LFM analogs. This observation was
`in good agreement with conclusions derived from structure~activity relation- .
`ships for pyrrolo- and pyrazoloquinazoline compounds (71) and wasincorpo-
`rated into the described modeling strategy forEGFR. ~~
`While most of the catalytic site residues of the EGFR kinase domain are
`conserved relative to other tyrosine kinases, a few specific variations are
`observed. In the EGFR modeling studies, the docked inhibitors were located
`between tworegions of mostly hydrophobic residues. EGFR residues on one
`side of the dockedinhibitor included Leu®*, Val’, Lys?2!, and Ala”!9, which
`are conserved in EGFR, HCK, FGFR and IRK. EGFR residues on the other
`side of the docked inhibitor included Leu®2° and Thr®°, whith varyint FGFR
`(Leu, Ala) and IRK (Met, Gly). EGFR residues Asn®!8 and Asp*?! (opposite
`the hinge) are conservedin all four PTKs. Residue Thr’in the EGFR hinge
`region changesto Val in FGFR,and to Metin IRK. Residue Thr®*° in EGFR
`changes to Ala in FGPR,and to Gly in IRK. Oneside of the binding pocket
`contains Cys’73 in EGFR andis therefore considerably more hydrophobic than
`the corresponding residue ofplatelet derived growth factor (PDGF)R (Asp),
`FGFR (Asn), and IRK (Asn). These residue identity differences may provide
`the basis for designing selective inhibitors of the EGFR tyrosine kinase.
`Thecatalytic site of EGFR may have specific features that can be advanta-
`geous for the design of inhibitors. Molecules that can fit into the triangular
`binding region of the EGFR catalytic site that can also form favorable contacts
`with the hinge region are likely to bind more strongly and inhibit EGFR more
`effectively. The docked position of LFM-A12 in the catalytic site of EGFR is
`shownin Fig. 3. It indicates that the molecule can maintain close contact with
`the hinge region of EGFR and can form a hydrogen bond betweenthenitrile
`nitrogen and the amide of Met’®,
`The docked positions of several quinazoline derivatives in the EGFR cata-
`lytic site are shownin Fig. 4. Like LFM-A12, quinazolines can maintain close
`contacts with the hinge region of EGFR,and the N1 nitrogen of the quinazoline
`group can hydrogen bond with the backbone carbonyl atom ofMet”, WHI-P97,
`which can form the most favorable interactions with EGFR (two hydrogen
`
`
` 4
`
`is
`ieni
`in
`eett
`iitie
`it
`ifot
`if
`
`pjae
`
`EGFRInhibitors for Treatment of Cancer
`
`207
`
`ngeaSenettngnnerapnpeteot
`
`
`TesteleottetgSeheSUreSieittWarWoeecerpameneroomet
`
`
`
`fo
`
`

`

`208
`
`:
`
`Sudbecketal.
`
`bonds and hydrophobic contacts), was the most potent of the quinazolines
`tested (EGFR ICs = 2.5 1M). However, in the EGFR model, the binding vol-
`ume of the EGFR-catalytic site is much larger than the volume occupied by
`WHI-P97 and LFM-A12.Increasing thesize of the ligand by using larger ring
`systems might increase the contact area between the receptor and ligand and
`thus enhancebinding. Interactions ofthe inhibitor with nonconserved residues
`such as Cys?>! and Thr®° in the catalytic site of EGFR mayalso beutilized for
`the design of more potentand selective inhibitors of EGFR.
`,
`
`3.2. EGF-Gen Immunoconjugates
`
`3.2.1. Biologic Activity of EGF-Gen Immunoconjugates
`Treatment of EGFR with EGF-genresulted in decreasédtyrosine phospho-
`rylation of the EGFR in a concentration-dependent fashion (2). Whereas EGF-
`gen exhibited marked PTK-inhibitory activity in MDR-MB-231 cells at
`concentrations as low as 0.1 1M in the treatment medium, unconjugated gen
`_ did not significantly affect the EGFR tyrosine phosphorylation even at a 10 uM
`concentration. The inhibitory effect of EGF-gen was blocked by preincubation
`of cells with excess EGF but not by excess G-CSF. Immune-complex kinase
`assays were usedto assessthe effects of EGF-gen on-the enzymatic activities
`of EGFR-associated Src PTK in MBA-MB-231 cells. EGF-gen treatmentinhib-
`ited the Src kinase. Unlike EGF-gen, a mixture of unconjugated gen and EGF
`or G-CSF-—gen did not inhibit the Src kinase activity in MDA-MB-231 cells.
`Thus, EGF-gen is a potent inhibitor of both the EGFR tyrosine

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