throbber
~9-1353.00/0
`Copyri~t © by The American Society for Pharmacology and Expe~mental The~peuti¢~
`All rights of reproduction in any form reserved.
`~OL/~JLAR PHA~ACOI~6~, "4&459--471 (1996).
`
`Carrier- and Receptor-Mediated Transport of Folate
`Antagonists Targeting Folate-Dependent Enzymes: Correlates
`of Molecular-Structure and Biological Activity
`
`(3. ROBBIN WESTERHOF, JAN H. SCHORNAGEL, IETJE KATHMANN, ANN L JACKMAN, ANDRE ROSOWSKY, RONALD
`FORSCH, JOHN B. HYNES, F. THOMAS BOYLE, GODEFRIDUS J. PETERS, HERBERT M. PINEDO, and GERRIT JANSEN
`
`Department of Oncology, University Hospital Vdje Universiteit, Amsterdam, 1007 MB The Netherlands (G.R.W., LK., G.J.P., H.M.P., G.J.),
`Department of Internal Medicine, Netherlands Cancer Institute, Amsterdam, 1066 CX The Netherlands (J.H.S.), Drug Development Section,
`Institute of Cancer Research, Sutton, Surrey, 5N2 5NG United Kingdom (A.L.J.), Dana-Farber Cancer Institute and Department of Biological
`Chemistry and Molecular Pharmacology, Hamard Medical School, Boston, Massachusetts 02215 (A.R., R.A.F.), Department of Pharmaceutical
`Sciences, Medical Univetsily of South Carolina, Charleston, South Carolina 29425 (J.B.H.), and Z
`"cals, Macclesfleld,
`Cheshire, SKIO 4TG United Kingdom (F.T.B.)
`
`Received January 30, 1995; Accepted June 14, 1995
`
`SUMMARY
`The transport propertias and growthAnhibitocy poten’dal of 37 classic
`
`(mFBP), or both. The ;,,becellular targets of these drugs were dihy-
`
`ge:~h-~fbib~ potential as a function of cellular RFC/mFBP ex-
`pression, and the protective effect of either FA or leucovodn against
`
`d~k~.~’~k: a~d (CB3717), ZD1694, 5,~k= add (U~HQ),
`
`.were we~ tolerated. Gmwlt~Wt~lk~ studies iderfdlied a sedes of
`
`turas) or mFBP (CB3717, 2-NH=-ZD1694, or 5,8-dk:ieazabo~ic
`
`port pathways (e.g., DOATHF, ZD1694, BW1843U89, or LY231514).
`
`substrate affinity for all analogues except CB3717, 2-NI-I~-ZD1694,
`
`For more than 40 years, the folate antagonist MTX has had
`an established role in cancer chemotherapy, both as a single
`
`This study was supported by Dutch Cancer Society Grant IKA 89-34. G. J.
`is a recipient of a fellowship from the Royal Netherlands Academy of Arts and
`Sciences.
`
`agent and in combination regimens (I). Knowledge of the
`factors that contribute to the preclinical and clinical activity
`of MTX, i.e., membrane transport, intracellular retention,
`and inhibition of the target enzyme DHFR, has provided a
`solid basis for the design and synthesis of novel antifolates
`that are either transported more efficiently, have a prolonged
`
`ABBREVIATIONS: RFC, reduced folate/methotrexate carder;, mFBP, membrane-associated folate binding protein; FCS, fetal calf serum; HBSS, HEPES
`balanced salt solution; LV, L-leucovodn (L-5-formyttetrahydrofotate); 5-CH3-THF, 5-metf~elrahy~-ofolate; DHFR, d~drofolate reductase; FPGS,
`folytpol~lutamate synthetmm; GARTF, glycinamide dbonucleotide transforrnytase; TS, thymidylate synthase; FA, folic acid; PteGlu, pteroyl glutamate
`(FA); Ivrrx, methotrexate; 2-dMTX, 2-desamino-MTX; 2-CHa-MTX, 2-desamino-2-meft~M3X; AMT, aminopte~; 2-dAIvrr, 2-desamlno-AMT; 2-CI~-
`AMT, 2-desamino-2-methyI-AM’r; 10-EdAM, 10-etttyl-10-d~_ ==u~minoptedn; PT523, N=-(4-amlno4-d~xypteroy~-(hemlpflltBIo~-omilJ3ine;
`DDATHF, 5,10-dideaza-5,6,7,8-tetrahydrofolic acid; 5-d(i)H4PteGlu, 5-desza-5,6,7,8-tetrahydroisofotic acid; N~-CH3-5-d(i)H4PteGlu, Ne-rnet~-5-
`deaza-5,6,7,8-tetrahydroisofolic acid; 5-dPteHCysA, N=-(5-de~v~.nteroyl)-L-homocystetc acid; 5-dPteAPBA, N=-(5-de~_ ,~.nteroyl)-oL-2-andno-4-phos-
`phonobutanoic acid; 5-dPteOrn, N=-(5-de~v-,nteroy~-omithine; 5-dH,PteHCysA, N=-(5-deaza-5,S,7,8-tetrahydropteroyl)-L~ acid;
`5-dI-~PteAPBA, N=-(5-deaza-5,6,7,8-telmhydropteroyl)-DL-2-arnino-4~p/no~c~:x~Jtanol¢ acid; 5-dH~PteOm, N=-(5-deaza-5,6,7,8-tetrahydropteroyl)-L-
`orni~ine; CB3717, N~ °-propargyl-5,8-dideazafolic acid; IC1-198,583, 2-desamino-2-me~lW~°-propargyl-5,8-dideezafolic acid; 4-H-IC1-198,583, 4-de-
`oxy-IC1-198,583; 4-OCH3-1C1-198,583, 4-rnethoxy-ICl-198,583; Glu--*Va14CI-198,583, valine-ICl-198,583; Glu-,Sub-ICl-198,583, 2-amino-suberate-
`IC1-198,583; 7-CH3-1C1-1 98,583, 7-methyl-ICl-198,583; ZD1694, N-[5(N-(3~4-dihydr~~2-methy~-4-~x~quinaz~~in-6-y~-me~~n~~)amin~)2-thany~)]-L-g~utamic
`acid; 2-NH~-ZD1694, 2-amino-ZD1694; BW1843U89, (S)-2-{5-(((1~3~methy~1-~x~-~?/qt~‘~m-9-y~)me~j~)an~)-1-~x~-24s~ind~n~
`gluladc acid; LY231514, N-(4-(2-(2-amin~-4~7-dihydr~4-~x~-~H-pyrr~k)~2~3-~]pydmidin-5-y~)e~.~)banz~y~]-L-g~u~amic acid; IAHQ, 5,8-dideezeisofollc
`acid; 2-d-IAHQ, 2-desamino-lAHQ; 2-CH~-dlAHQ, 2-desamino-2-methyl-lAHQ; 5-d(i)PteGlu, 5-deazaisofolic acid; hP-CHa-5-d(i)PteGlu, Ne-methyl-5-
`deazaisofolic acid; hP-CHO-5-d(i)PteGlu, Ne-formyl-5-deazaisofolic acid; AG337, 3,4-dihydro-2-amirm-6-methyl-4-oxo-5-(4-pyrtdytlNo)qulnazol~;
`AG377, 2,4-diamino-6-[N-(4-(pher~jlsu ~ino]q uinazoline.
`
`Sandoz Inc.
`Exhibit 1022-0001
`
`JOINT 1022-0001
`
`

`
`!
`
`intracellular retention, or are more potent inhibitere of the
`target enzyme (2-13). Other than DHFR, key enzymes in
`folate metabolism, such as TS, GARTF, and FPGS, have been
`recognized as potential targets for folate-based chemothera-
`peutic agents (14-19).
`Traditionally, the biological activity of novel antifolates is
`evaluated with in vitro model systems (usn~lly leukemia cell
`lines) that express the RFC as the major transport route for
`natural reduced folate cofactors (e.g., LV and 5-CHs-TI-IF)
`and classical antifolate compounds (6, 20-22). Although the
`role of the RFC in antifolate transport is undisputed, an
`increasing number of reports suggest that at least one other
`folate trsn~port protein, an mFBP, may have an additional
`role in antifolate uptake (23-28X A functional role of mFBP
`in folate uptake has been demonstrated in a number of in
`vitro (28-32) and in vivo (33) studies in which mFBP-medi-
`ated transport of natural reduced folate cofactors supported
`cellular growth at nanomolar extracollular concentrations.
`Other reports have demonstrated mFBP-mediated transport
`of antifolates, particularly ff the mFBP exhibits a high bind-
`ing affinity for these drugs (23, 34-36). Because the mFBPs
`appear to be expressed in a range of normal and neoplastic
`cells and tissues (37-42), further analysis of the potential
`role of this protein in antifolate drug uptake is warranted.
`The RFC and mFBP are’ structurally and functionally un-
`related proteins. The RFC is an integral membrane protein
`with a molecular weight ranging from 43-48 kDa in rodent
`cells (43, 44) to 80-120 kDa in human cells (42, 45, 46). The
`mFBP is a 38-40-kDa protein that is linked to the plasma
`membrane via a glycosylphosphatidylinositol anchor (47, 48).
`The mechanism of RFC-mediated folate and antifolate up-
`take has been the subject of extensive studies, which have
`demonstrated the uptake process is both temperature and
`energy dependent and able to be inhibited by structurally
`unrelated anions (6, 49-52). These observations support the
`hypothesis that an anion exchange mechanism may be in-
`volved in carrier-mediated antifolate uptake (6). At least two
`mechanisms have been described for mFBP-mediated folate
`uptake: one by the classic receptor-mediated endocytosis
`pathway (53) and the other, most extensively studied in
`monkey kidney MA-104 cells, via a novel mechanism called
`potocytosis. Potocytosis (27) comprises a series of events that
`involve clustering of folate-bound mFBP molecules in mem-
`brane invaginations (caveolae), followed by transient closure
`of caveolae from the extracollular medium and acidification
`of their lumen, after which the folate molecule is dissociated
`from the mFBP and translocated across the plasma mem-
`brane via a specific carrier protein (27, 54). Whether the
`latter protein is the RFC is not clear.
`The klnet~c properties of the RFC and mFBP for transport
`also differ significantly. The RFC exhibits a high affinity (Kin,
`1-10/m) for natural reduced folate cofactors and antifolates
`such as MTX (6), which can be internalized at a maximal rate
`of 10-20 molecules per minute per transport molecule (20). A
`characteristic feature of the RFC is its poor affinity for FA
`(Kin, 200-400 ~), which is in contrast to that of mFBP.
`mFBP has a high affinity (Kd, 1-10 n~) for FA and reduced
`folate cofactors, which may favor uptake at a physiological
`folate concentration of 5-50 nM (26). On the other hand, the
`recycling rate of mFBP is much slower than that for the RFC:
`from -30 rnin in MA104 cells (27, 55) to 5 hr in L1210 cells
`(56, 57).
`
`We report on a comprehensive study of the functional as-
`pects of both the RFC and mFBP in antifolate drug transport,
`which allowed us to directly compare the efficiency of these
`transport proteins. As models, three CCRF-CEM human leu-
`kemia cell lines were used, which are characterized by nor-
`real, upregulated, or defective RFC transport (22, 58). For
`comparison, three (variant) L1210 murine leukemia cells
`were included that express the RFC (50), coexpress the
`mFBP and the RFC (59), or lack functional RFC activity
`while retaining mFBP (60). In the present study, we used a
`series of folate-based inhibitors of DHFR (MTX and AMT)
`(61), GARTF (DDATHF and 5-d(i)H4PteGlu) (15, 62), FPGS
`(glutamate side chain-modified 5-deazafolic, 5-deazatetrahy-
`drofolic acid structures) (63), and TS (CB3717, ZD1694, and
`IAHQ) (64-66). These compounds and their analogues with
`structural alterations in the pteridine/quinazoline ring (sub-
`stitutions at C-2, C-7, N-3, and C-4), p-smlnobenzoate ring
`(thiophene for benzoyl), the C~-N1° bridge, and the glutamate
`side chain (replacement by 2-aminosuberate, valine, and or-
`nithine) were used in the study. All of these compounds were
`evaluated for changes in the substrate specificity for the RFC
`and mFBP; the growth-inhibitory effects against cell lines
`with different expression levels of the RFC, mFBP, or both;
`and the protective effect of LV, FA, or both.
`The structure-activity relationships described in the
`present study for antifolate transport via the RFC, mFBP, or
`both may be of predictive value in assessing the preclinical
`and clinical activity of antifolate drugs and can also be used
`for the rational future design of new antifolates.
`
`Experimental Procedures
`
`Materials. RPMI 1640, with and without FA, and FCS, dialyasd
`and nondialysed, were obtained from GIBCO (Grand Island, NY). FA
`and DL-5-CHs-THF were purchased from Sigma Chemical Co. (St.
`Louis, MO). LV (L-stereoisomer) was a gift from Lederle (Pearl River,
`NY). [3’,5’,7,9-SH]FA (35 Ci/mmo]), [3’,5’,7-SH]MTX (20 Ci/mmol),
`and [3,4-SH]glutamic acid (56.6 Ci/mmol) were obtained from
`Moravek Riochemicals (Brea, CA). [SH]FA and [SH]MTX were puri-
`fied as described previously (20, 59, 60). All other chemicals were of
`the highest purity available.
`Drugs. The chemical structures of the antifoiste compounds de-
`scribed in the study are depicted in Fig. 1. The inhibitory potential of
`these compounds against their target enzyme, as well as their sub-
`strate ~fl~nity for FPGS, are described in Tables 1 (DHFR, GARTF,
`and FPGS inhibitors) and 2 (TS inhibitors). It should be noted that
`these data were taken from the indicated references and that the
`experimental conditions were not identical. Nevertheless, these data
`were included to explain possible growth-inhibitory effects on the
`basis of poor enzyme inhibition or substrate affinity for FPGS. Fur-
`ther details regarding chemical synthesis of the drugs are given in
`references as indicated in Tables 1 and 2.
`DHFR inhlbitors. MTX was a gi~ from Pharmachemie (Haar-
`lem, The Netherlands). AMT was purchased from Sigma Chemical
`Co. 2-dMTX, 2-CHs-MTX, 2-dAMT, and 2-CHs-AMT were synthe-
`sized as previously described (61). 10-EdAM was a ~ from CIBA-
`GEIGY (Basel, Switzerland). PT523 (67, 68) was a gift from Dr. W. T.
`McCulloch (Sparta Pharmaceuticals, Research T~gle Park, NC).
`GARTF Inhlbitors. The synthesis of 5-d(i)I-I~PteGlu and Ng-CHs-
`5-d(i)H~PteGlu has been described previously (62). DDATHF was a
`gift from the late Dr. G. B. Grindey (Lilly Research Labs., Indianap-
`otis, IN).
`FPGS i~tbitors. The 5-deazafolate analogues 5-dPteHCysA,
`5-dPteAPBA, 5-dPteOrn, 5-dH~PteHCysA, 5-dH~PteAPBA, and
`5-dH~PteOrn were synthesized by Rosowsky et al. (63).
`
`Sandoz Inc.
`Exhibit 1022-0002
`
`JOINT 1022-0002
`
`

`
`Structure-Activity Relationshilm of Antifolate Transport 461
`
`!
`
`o
`
`DHF~RGARTF F~PGS
`
`TS
`
`Rg. 1. Molecular structure of folate-based inhibltors of DHFR, GARTF, FPGS, and TS.
`
`TS ;-hibitors. CB3717 (64, 69) and ICI-198,583 (70) served as
`basic structures for the following ~nalogues: 3-deaza-ICI-198,583,
`4-H-ICI-198,583, 4-OCHa-ICI-198,583, Glu--~Val-ICI-198,583 (71,
`72), 7-CHa-ICI-198,583 (73, 74), Glu-~Sub-ICI-198,583 (72), ZD1694
`(65), and 2-NH2-ZD1694 (75, 76). The isofolic analogues IAHQ, 2-des-
`Amino-IAHQ, and 2-des*mino-2-methyl-IAHQ and the 5-deazaisofo-
`lates 5-d(i)PteGlu, N~-CHa-5-d(i)PteGin, and l~-CHO-5d(i) eGlu
`were synthesized as described previously (62, 66, 77, 78). LY231514
`(79) was a gift from the late Dr. G. B. Grindey. BW1843U89 (80) was
`
`provided by R. Ferone (Burroughs Wellcome Co., Research Triangle
`Park, NC). AG337 (81, 82) and AG377 (83) were gitts from Agouron
`Pharmaceuticals, Inc. (San Diego, CA).
`Cell culture. The cell lines used in the present study include
`RFC-expreseing CCRF-CEM human leukemic lymphoblaste (20-22);
`a variant of this line, CEM/MTX, lacking functional RFC (58); and
`another variant (CEM-7A), characterized by a 30-fold overexpression
`of the RFC compared with CCRF-CEM coils (22). In addition, the
`following murine cell lines were used: wild-type RFC expressing
`
`Sandoz Inc.
`Exhibit 1022-0003
`
`JOINT 1022-0003
`
`

`
`Charactedstics of folate-based inhibitors of DHFR, GARTF, and FPGS with respect to enzyme inhibition and sublimate activity for
`
`Compound Main target FPGS substrete activity~ Ref.
`DHFR TS GARTF
`
`! TABLE I
`
`MTX
`2-dMTX
`2-CH3-MTX
`AMT
`2-dAMT
`2-CH3-AMT
`10-EdAM
`PT523
`DDATHF
`5-d(i)H4PteGlu
`N~-CH3-5-d(i)H4PteGlu
`
`5-dPteHCysA
`5-dPteAPBA
`5-dPteOm
`5-dH4PteHCysA
`5-dH4PteAPBA
`5odH4PteOm
`
`DHFR
`
`GARTF
`
`FPGS
`
`0.024
`6
`>20
`0.025
`19
`>50
`2.3 pM
`0.052
`>100
`>1000
`>1000
`
`0.4
`4.7
`8.3
`12
`220
`
`29
`
`>10
`
`>100
`> 100
`>100
`
`55
`300
`
`2100
`4200
`
`0.12
`5.3
`26
`
`0.19
`0.047
`3.7
`
`+
`NR
`NR
`+ + +
`NR
`NR
`+++
`NS
`+++
`+ +
`+ + +
`
`570
`9.0
`5.7
`22
`3.2
`0.027
`
`61,110
`61
`61
`61, 87
`61
`12, 61
`111
`68
`112,113
`62
`62
`
`63
`63
`63
`63
`63
`63
`
`"The data in this table are obtained from the indicated references. Please note that Kin determinations of enzyme inhibition and substrate activity determinetion
`for FPGS may not have been carded out under Identical (x)ndlttons with respect to substr~e, Inhibitor, and cofactor concentration. For experimental details, see the
`indicsted references.
`~ FPGS subetrate activity (Kin) is defined as follows: Km 5-25 M,M: (+++); Km 25-100/ZM: (++); Km > 100 ~: (+). NR, not reported; NS, nonsubstrate.
`
`TABLE 2
`Characted~cs of folate-based inhibitom of TS with respect to enzyme Inhibition and substrate activity for FPG~’
`
`Compound Main target FPGS substrate activity~ Ref.
`DHFR TS GARTF
`
`MTX
`CB3717
`IC1-198,583
`3-deaza-ICl-198,583
`4-H-IC1-198,583
`4-OCH3-1C1-198,583
`Glu--,VaI-ICI-198,583
`Glu--*Sub-ICl-198,583
`7-CH3-1C1-198,583
`ZD1694
`2-NH2-ZD1694
`BW1843U89
`LY231514
`IAHQ
`2-dlAHQ
`2-CHa-IAHQ
`5-d(i)PteGlu
`N~-CH3-5-d(i)PteGIu
`NS-CHO-5-d(i)PteGlu
`AG337
`AG377
`
`DHFR
`TS
`
`0.024
`0.91
`5.5
`
`12
`
`0.11
`0.32
`3.1
`0.21
`0.25
`1.95
`
`0.02
`0.046
`6.3
`7.9
`6.0
`0.042
`0.018
`0.028
`0.65
`0.44
`0.00009
`0.44
`1.3
`25
`17
`7.1
`0.48
`28.8
`0.011
`0.033
`
`> 17
`
`85.9
`>100
`> 100
`
`+
`+ +
`+ +
`+
`
`NS
`+ + + +
`
`+ + + +
`+ + + +
`+++
`+ + +
`+ + +
`+
`+ +
`+
`NS
`NS
`
`110
`7O
`
`110
`70,
`69,
`72
`114
`114
`71
`72
`73
`65
`76
`80
`79
`78
`78
`78
`62
`62
`62
`81
`63
`
`¯ The data in this table are depicted from the indicated references. Please note that KI,.~ determinations of enzyme inhibition and substrat.e activity determination
`for FPGS may not have been ~ out under identical conditions with respect to subetr~e, inhibitor, and cofactor concentration. For expenmental details, see the
`indicated references. Moreover, it should be noted that the KI for the compounds listed may not reflect the increased potency of inhibiting TS when polyglutamete
`
`~ FPGS substrate activity (Kin) is defined as follows: Km < 5/~M: (++++); Km 5-25 p.u: (+++); Km 25-100 p~: (++); K,, > 100 p~: (+). NS, nonsubstrate.
`
`L1210 leukemia cells, one variant (L1210-B73) expressing beth the
`RFC and mFBP (59), and another variant (L1210-FBP) in which
`mFBP is the only functional transport protein (60). (This cell line
`was erroneously reported to be derived from CEM/MTX cells, but on
`karyotype and restriction polymorphism analysis it appeared to be a
`subline of L1210-B73 cells lacking functional RFC activity (60a).
`Parental cells (CCRF-CEM and L1210) and CEM/MTX cells were
`
`grown in RPMI-1640 medium cent~ining 2 /~M FA supplemented
`with 10% FCS, 2 mM L-ghit~mine, and 100 units/m] each of penicillin
`and streptomycin. CEM-7A, L1210-FBP, and L1210-B73 cells were
`grown in folate-free RPMI 1640 supplemented with 10% dialysed
`FCS, antibiotics, and ghit~mine as described. LV was added as folate
`source at final concentrations of 0.2, 1.0, and 1.0 nM, respectively
`(unless otherwise indicated). The cell cultures ofL1210, L1210-FBP,
`
`Sandoz Inc.
`Exhibit 1022-0004
`
`JOINT 1022-0004
`
`

`
`!
`
`and L1210-B73 cells also contained 50 ~M ~-mercaptoethanol. Cells
`were kept at 37° in a humidified atmosphere with 5% CO2.
`DHFR and FPGS levels in cell lines. The DHFR concentration
`of the cell lines used in the present study as assessed by [SH]MTX-
`bindin~ (84) was comparable in CEM, CEM/MTX, and CEM-7A cells
`(1.3 pmol/mg protein). Likewise, DHFR levels in L1210 cells and
`L1210-FBP cells were similar (2.6 pmol/mg protein). The enzyme
`level in L1210-B73 cells was 2.8-fold higher (7 pmol/mg) than in
`L1210 cells. FPGS activity was measured in all cell lines as described
`by Jansen et al. (85) with 250 /~M MTX as a substrate. Enzyme
`activities expressed as picemoles of MTX-[SH]glutamato formed per
`hour per mi|ligram of protein were as follows: 402 (L1210-FBP), 618
`(L1210-B73), 829 (CEM), 992 (CEM-7A), 1168 (L1210), and 1200
`(CEM/MTX) (results not shown).).
`ItFC substrate specificity. CEM-7A cells (3 × l0s) in the loga-
`l~t.hmlc phase of growth were washed with 10 mi HBSS buffer (107
`mM NaC1, 20 mM HEPES, 26.2 mM NaHCOs, 5.3 mM KC1, 1.9 mM
`CaC12, 1.0 mM MgC12, and 7.0 mM D-glucose adjusted to pH 7.4 with
`NaOH), centrifuged, and suspended in 1 ml HBSS-buffer at 37°.
`Influx of [SH]M~X (0.5 Ci/mmol) was measured over a period of 1.5
`rain at 37° at an extracellular concentration of 5 p~M in the absence or
`presence of increasing concentrations of unlabeled folate or antifo-
`late compound. Uptake was terminated by the addition of 9 ml of
`ice-celd HBSS buffer, centrifugation at 800 x g for 5 rain at 4°, and
`a second wash with 10 ml ice-celd buffer. Cell pellets were resus-
`pended in water and analyzed for radioactivity with Ultima-Gold
`scintillation fluid and a scintillation counter (both from Packard,
`Brussels, Belgium) with a counting efficiency for [3H] of approxi-
`mately 55%. The drug concentration required to inhibited [SH]MTX
`influx by 50% of control values (221 pmol/min/107 cells) was taken to
`be a measure of relative affinity for the RFC.
`mFBP substrate specificity. An intact cell binding assay for
`competitive binding of [SH]FA was performed as described previously
`(86). Briefly, L1210-FBP cells were washed twice with ice-cold HBSS
`buffer. One milliliter of L1210-FBP cell suspension (3 × 10s cells)
`was incubated with 100 pmol [OH]FA (specific activity, 0.5 Ci/mmol)
`in the presence or absence of increasing concentrations of unlabeled
`folate or antifolate compound. After 10 rain, the cells were collected
`by centrifugation (for 5 rain at 800 x g at 4°), after which the
`supernatant was removed. Pellets were resuspended in water and
`analyzed for radioactivity as described. Relative aiYmities are de-
`fined as the inverse molar ratio of cempeund required to displace
`50% of [aH]FA from mFBP on L1210-FBP cells. The relative affinity
`of mFBP for FA is set at 1.
`Growth-lnhlbition studies. All cell lines were plated at an
`initial density of 7.5 x 104 cells/m] in individual wells of a 24-well
`tissue culture plato. The growth medium for CCRF-CEM, CEM/
`MTX, and L1210-cells was RPM11640 with 10% FCS, supplemented
`as described. CEM-7A cells were grown in folate-fi, ee RPMI 1640
`with 10~ dialyzed FCS and 1.0 nM LV as the sole folate source;
`L1210-FBP and L1210-B73 cells were grown in the same medium
`but with 1 nM LV, 20 nM LV, or 20 nM FA as folate source. Drugs were
`added at the time of plating. After 72 hr of exposure, cell counts and
`viability were determined with a hemocytometer by trypan blue
`exclusion. IC~o values are given as the concentration of drug at which
`the growth is inhibited by 50% compared with controls.
`
`Results
`RFC substrate specificity. The affinity of the RFC for
`the series of antifolate drugs, expressed as the drug concen-
`tration required to inhibit [aH]MTX influx by 50%, is shown
`in Fig. 2. MTX (represented by a black bar and vertical line)
`was used as a reference compound. Compounds on the left
`side of this line were better substrates for the RFC than
`MTX, whereas compounds on the right side were poorer
`substrates. All DHFR inkibitors included in the study ap-
`
`Structure-Activity Relationships of Antlfolate Transport 463
`
`peared to be better substrates by RFC than ~ Replace-
`ment of the 2-Rmino group by 2-methyl in MTX and AMT led
`to an enhanced affinity. For example, 2-CHs-dAMT was a
`10-fold better substrate for RFC than MTY~
`In the series of GARTF inhibitors, only DDATHF is a more
`efficient substrate for RFC than MTX. The group of FPGS
`inhibitors [5-deazatetrahydrofolic acid analogues with the
`glutamate side chain replaced by other charged residues
`(homocysteic acid, 2-~mino-4-phosphonobutanoic acid, and
`ornithine)] are very poor substrates for RFC-mediated trans-
`
`Within the group ofTS inhibitors, the 2-amino-based struc-
`tures CB3717, 2-NH~-ZD1694, and IAHQ are low affinity
`substrates for the RFC. However, their 2-methyl analogues
`(ICI-198,583, ZD1694, and 2-CH3-IAHQ) are excellent sub-
`strates for the RFC. Modification at the 4-oxo position of
`ICI-198,583 did not alter the substrate affinity for the RFC.
`A modification at the N-3 position led to a 2-fold better
`affinity for the RFC. Replacement of the glutamate side chain
`by valine and 2-~mlnosuberate or a 7-CHa substitution re-
`suited in a 4-5-fold and a 3-fold lower affinity for the RFC
`than for ICI-198,583, respectively. The RFC had the highest
`affinity for BW1843U89. LY231514 was a 2-fold better sub-
`strate for the RFC than MT~ The ai~inity of the RFC for
`5-d(i)PteGlu and its N~-substituted analogues was more than
`10-fold lower compared with MTT~ As might be expected, the
`lipophilic antifolates AG337 and AG377 were poor substratos
`for the RFC.
`mFBP substrate specificity. Relative affinities of the
`mFBP for the series of antifolate drugs are presented in Fig.
`3. FA (represented by the black bar and vertical line), for
`which mFBP has a binding ~fl~nlty constant (Kd) of 0.5-1 aM
`(not shown), was used as the reference compound. Compared
`with FA, the mFBP had a lower affinity for all of the DHFR,
`GARTF, and FPGS inhibitors, especially the 4-amlno-based
`compounds, e.g., MTX, AMT, and their 2-des~mlno/2-methyl
`analogues 10-EdAM and PT523. Modifications at sites other
`than the pteridine ring, such as the Ce-N1° bridge and the
`glutamate side chain, were better tolerated with respect to
`efficient binding by mFBP. For eT~mple, mFBP demon-
`strated good binding affinity for DDATHF and 5-dPteHCysA.
`In contrast to 5-dPteHCysA, the tetrahydrofolate forms of
`5-dPteAPBA and 5-dPteOrn were better substrates for
`mFBP than were their nonreduced counterparts.
`From the group of TS inhibitors, four compounds were
`identified for which mFBP has a higher affinity than for FA:
`LY213514, CB3717, IAHQ, and 2-NH2-ZD1694. Unlike ob-
`servations for the RFC, the 2-methyl analogues of the latter
`three compounds are characterized by significantly de-
`creased binding affinity to mFBP. Most dramatic in terms of
`abrogation of binding affinity are modifications at the N-3
`position (3-deaza-ICI-198,583) and structural alterations at
`the 4-oxo position (4-deoxy-ICI-198,583), although the 4-
`methexy analogue of ICI-198,583 retained the same binding
`affinity as the parent compound. Modifications at other sites
`of the molecule (the qulnazoline ring [substitution at the
`7-position], the C~-N~° bridge, the p-aminobenzoyl, and the
`glutamate side chain) did not dramatically influence the
`binding affinity with respect to binding by mFBP. mFBP
`exhibits a moderately high binding ~Winity for BW1843U89.
`Similar to the RFC, the mFBP has a poor affinity for the
`lipophilic drugs AG337 and AG377.
`
`Sandoz Inc.
`Exhibit 1022-0005
`
`JOINT 1022-0005
`
`

`
`RFC
`
`2-d~
`2-CH~-MTX
`
`2-dAMT
`=-CH,-AM’r
`tO-F=dAM
`
`S-d~P~C~A
`S-dH~UmA
`5-dH4Pt~)m
`
`CB3717
`K:1-198.583
`3-d~z~Cl-198,583
`4-H4C1-198,583
`
`Glu-~Vd-ICI-1S8,583
`Glu~Sub-1~-198,583
`7~H=-IC1-1~8,583
`
`2-NH=-ZD1694
`
`LY231514
`IAHQ
`2-dlAHQ
`
`5-d(i)Pt~Glu
`
`AG337
`AG377
`
`-)
`
`0)
`
`Fig. 2. Concentrations for 50%
`inhibition of RFC-mediated influx
`of [~H]MTX. Values represent the
`concentrations of folate or anti-
`folate compound necessary to
`inhibit RFC-mediated [~H]MTX
`influx in CEM-7A cells by 50% at
`5 pJ~ extracelluler concentration.
`Verffcal line and black bar, con-
`
`centration of unlabeled M’rx
`necessary to inhibit [~H]MTX up-
`take by 50%. For further details,
`see Experimental Procedures.
`Ba/s labeled with a through g,
`actual value exceeds the indi-
`cated value. Residual uptake val-
`ues as percentage of control at
`the indicated drug concentration
`were as follows: a, 57% at 100
`p.M; b, >90% at 100 /~M; C,
`>90% at 100 p~; d, 57% at 100
`p.M; e, >90% at 200 p~; f, >90%
`at 50 ~; g, >90% at 50
`
`I I
`
`;)
`
`0.1
`
`1
`
`10
`
`pM
`
`100 600
`
`Growth-;nk|bition studies. The role of the P,~’C in drug
`transport and growth inhibition was analyzed by using three
`cell lines that differ in RFC expression and MTX transport
`(22) (Tables 3 and 4): CEM-7A cells are characterized by a
`30-fold overexpression of RFC compared with parental
`CCRF-CEM cells, whereas CEM/MTX cells exhibit a MTX
`transport defect. In general, compounds that are good sub-
`strates for the RFC (e.g., MTX and AMT with their 2-deso
`nmino and 2-methyl Rnnlogues ICI-198,583, ZD1694,
`LY231514, BW1843U89, and DDATHF) were potent growth
`inhibitors provided that the compounds were efficiently poly-
`glutamated, are potent inhibitors of their target enzymes, or
`both (Tables 1 and 2). For e~Rmple, drugs like 2-dMTX,
`2-CHa-dAMT, 3-deaza-ICI-198,583, and 4-H-ICI-198,583 ap-
`peared to be efficient substrates for the RFC, but because
`
`they are poor inhibitors of both DHFR and TS, their growth-
`inhibitory activity was low. Nevertheless,’the role of the RFC
`in transport of these compounds and of most of the TS inhib-
`itors is illustrated by the fact that RFC-overproducing
`CEM-7A cells are more sensitive than parental coils, whereas
`MTX transport-defective CEM/MTX cells are cross-resistant
`to these drugs. In this respect, it is of interest to note that
`cross-resistant factors for DDATHF and PT523 were signifi-
`cantly lower than for MTX, AMT, and 10-EdAM. Poor growth
`inhibition associated with inefficient RFC transport was ob-
`served for CB3717, 2-NH2-ZD1694, IAHQ, and the series of
`glutamate side chain-modified FPGS inhibitors. In addition,
`for 5-deazaisofolic acid, 5-deazatetrahydroisofolic acid and
`their N~-substituted analogues, the poor growth-inhibitory
`effect correlates with a low transport efficiency and target
`
`Sandoz Inc.
`Exhibit 1022-0006
`
`JOINT 1022-0006
`
`

`
`mFBP
`
`Sb’uctwe-Actlvity Relationships of Antlfo~te Transport
`
`0.008
`0.005
`0.005
`0.008
`0.002
`0.011
`0.009
`0.009
`
`2-d~
`2-CH,-MTX
`
`2-dAMT
`2-C~,-AMT
`10-EdAM
`m23
`
`DDATHF
`
`N~-CH=-5-d(i)H4Pte~u
`
`S-dPt~W:~A
`
`CB3717
`IC1-198,583
`3-deaz~lCl-198,583
`4-H4C1-198,583
`
`Glu--Vd-ICl-198,583
`Glu-,Sd~-198.583
`7-CH=4C!o198o583
`
`2-NH=-ZD1
`
`10.019
`
`LY231614
`LAHO
`2-dlAHO
`2-CH,4AHO
`5-dli)PIsGiu
`
`I
`
`AG337
`AG377
`
`|0.028
`|o.o$$
`
`I
`
`Fig. 3. Relative affinity of
`mFBP for folate and antifolate
`compounds. Values indicate
`
`inverse molar ratio of com-
`pound required to displace
`50% of [~H]FA from mFBP in
`L1210-FBP ceils. Relative af-
`finity of FA Is set at I (b/ack bar
`and vertical line).
`
`/
`
`0.00 0.25 0.50 0.75 1.00 1.25 1.50
`
`Relative affinity
`
`enzyme inhibition. The lack of correlation between RFC ex-
`pression and growth-inhibitory effect of the lipophilic drugs
`AG337 and AG377 is indicative of an RFC-independent pro-
`cess of drug uptake.
`The drug sensitivity profiles of human CCRF-CEM coils
`(Tables 3 and 4) and routine L1210 colls (Tables 5 and 6)
`were in large part similar. Nevertheless, absolute IC~o values
`for the individual drugs were most often lower for L1210 colls
`than for CCRF-CEM coils, which may be consistent with a
`3-fold higher level of RFC expression in L1210 cells versus
`CCRF-CEM cells, whereas other transport kinetics proper-
`ties are similar (20). The one excoption is the uptake of
`BW1843U89 (80), which is a 17-fold poorer substrate for the
`routine RFC (K~, 14 p~) compared with human RFC (Ki, 0.8
`
`/~; see Fig. 2). This difference in transport characteristics is
`reflected in a 10-fold lower growth-lnhibitory activity of
`BW1843U89 ag~inRt L1210 versus CCRF-CEM colls (80).
`In addition to binding of antifolates, the results given in
`Tables 7 and 8 support a functional role for mFBP in the
`uptake process of antifolates. In the absence of competing
`folate cofactore in the growth medium, most of the com-
`pounds tested demonstrated a growth-inhibitory effect
`against (RFC-/mFBP+ + +) L1210-FBP cells, provided they
`were efficiently polyglutamated or were potent inhlbitere of
`their target enzyme (Tables 1 and 2). Compounds for which
`the RFC has poor Aff;nlty (e.g., CB3717, IAHQ, and 2-NH=-
`ZD1694) appeared to be good growth inhibitore, consistent
`with the high bindin£ affixdty of mFBP for these compounds.
`
`Sandoz Inc.
`Exhibit 1022-0007
`
`JOINT 1022-0007
`
`

`
`!
`
`466 Westedmf et al.
`
`TABLE 3
`Growth-inhibitory effects (IC.o values in nM’) Of antifolate
`=)mpounds against human CF.M cells with upregulated and
`downregulated RFC-medlated transport (RFC expression in
`parentheses)
`
`TABLE 5
`Growth-inhibitow effects (IC~ values in ~ of anUfolate
`compounds against routine L1210- (RFC+)b and L1210-B73
`(RFC+/mFBP+++)~ cells grown in different folate
`concentrations in the medium
`
`Cell line (RFC expression)a
`
`L1210
`
`L1210-B73
`
`Compound
`
`MTX
`2-dMTX
`2-CH3-MTX
`AMT
`2-dAMT
`2-CH3-AMT
`10-EdAM
`PT523
`DDATHF
`5-d(i)H4PteGlu
`NS-CH~-5-d(i)H4PteGlu
`5-dPteHCysA
`5-dPteAPBA
`5-dPteOm
`5-dH4Ptel.-ICysA
`5-dH4PteAPBA
`5-dH4PteOm
`
`CCRF.-CEM
`(+)
`
`8.1
`1,870
`837
`2.0
`500
`1,157
`1.3
`1.1
`11
`37,020
`42,670
`>25,000
`>25,000
`>50,000
`>25,000
`6,100
`>50,000
`
`CEM/MTX
`(-)
`1,950
`40,000
`41,300
`1,010
`12,600
`10,000
`533
`33
`80
`36,025
`>100,000
`>50,000
`>50,000
`>50,000
`>50,000
`>50,000
`>50,000
`
`CEM-7A
`(++++)
`1.2
`49
`31
`0.70
`14.6
`42
`0.3
`0.7
`2.2
`164
`1,740
`4,500
`>25,000
`31,500
`13,000
`1,400
`3,200
`
`¯ IC~o values are given as the concentration of drug at which growth is inhibited
`by 50% compared wflh controls. VaJuas ~e the mas~ of at lasst four separate
`experiments (SD < 15%).
`a RFC expression levels: CCRF-CEM, CEM-MTX, and CEM-TA-ceiis, 0.23,
`<0.02, and 8.1 pmoVmg protein, respectiv~y (22).
`
`TABLE 4
`Growth-lnhibitow effect~ (IC~o values in nM=) of antifolate
`compounds against human CEM cells with upregulated and
`downregulated RFC-medtated transport (Rr-C expression in
`pa~)
`
`Cell line (RFC expression)a
`
`Compound
`
`MTX
`CB3717
`IC1-198,583
`3-deaza-ICl-198,583
`4-H-IC1-198,583
`4-OCHa-IC1-198,583
`Glu-*Val-ICl-198,583
`Glu--*Sub-lCI-198,583
`7-CH~-IC1-198,583
`ZD1694
`2-NH=-ZD1694
`BW1843U89
`LY231514
`IAHQ
`2-dlAHQ
`2-CH~-IAHQ
`5-d(i)PteGlu
`Ng-CHa-5-d(i)PteGlu
`Ng-CHO-5-d(i)PteGlu
`AG3

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