throbber
J. Med. Chem. 1991,34,992-998
`992
`genated as for 22, affording a mixture of components: yield 76.5
`Plates were incubated for 72 h at 37 OC in a humidified atmo-
`sphere containing 5% COP After 72 h, 20 pL of 5 mg/mL MTl’
`mg, from which 5 was isolated via chromatotron with u& of ethyl
`acetate/hexane (41); total yield of 5 from 20b, 43.5 mg, 42%; mp
`(3-(4,5-dimethylthiazol-2-yl)-2,5-diphenyltstolium bromide)
`was added, and cells were incubated for 90 min to allow reduction
`219-220 “C; ‘H NMR (CDC13) 6 6.75 (1 H, s, H7), 6.5 (1 H, s, H4),
`6.31 (2 H, S, H2’, 6’), 6.00 (2 H, AB 9, OCHZO), 5.48 (1 H, 9, M H ) ,
`of the formasan by the surviving cells. Following washing and
`4.74 (1 H, d, H3), 4.37 (2 H, AB q, CH,OCO), 3.87 (2 H, br d,
`solubilization by DMSO, absorbance of each well was measured
`spectrophotometrically at 570 nm. The ICw is determined as the
`CHZOH), 3.82 (6 H, S, OCH3 X 2), 3.57 (1 H, d, H2), 1.81 (1 H,
`br t, OH); HRMS (FAB/HRP), calcd for CZ1HmO8 400.1156, found
`concentration of compound tested required to reduce the ab-
`sorbance to 50% of non-drug-treated control values.
`400.1162. Anal. (CzlHzoOs) C, H; C: calcd, 62.98; found, 61.04.
`Biological Assay. Cells were grown in RPMI 1640 supple-
`mented with 10% fetal calf serum. Test compounds were dis-
`solved in dimethyl sulfoxide (DMSO) and diluted first with &le’s
`Balanced Salt Solution, followed by culture medium, to twice the
`highest concentration of compound to be tested. From this
`concentrated stock, 2-fold serial dilutions were prepared in S w e l l
`microtiter trays. Each concentration was tested in triplicate and
`compared to triplicate drug-free controls. A 100-pL aliquot of
`cells (2.5 x 103 cells) was added to the wells of the microtiter plate
`containing 100 pL of growth medium with or without test drugs.
`
`Acknowledgment. We are grateful to Dr. R. Stephens
`in the Analytical division for helpful NOE analysis on
`several of our compounds and also thank Dr. M. Bures of
`CAMD division for molecular modeling analyses.
`
`Supplementary Material Available: Microanalysis and masa
`spectra for several compounds mentioned in the text (31 pages).
`Ordering information is given on any current masthead page.
`
`Relationships between the Structure of Taxol Analogues and Their Antimitotic
`Activity8
`
`Francoise GuBritte-Voegelein,**t Daniel GuBnard,? Francois Lavelle,* Marie-ThBrBse Le Goff,t Lydie Mangatal,+ and
`Pierre Potiert
`Znstitut de Chimie des Substances Naturelles, Centre National de la Recherche Scientifique, avenue de la Terrasse,
`91 198 Gif-sur- Yvette Ceden, France, and Centre de Recherches d e Vitry, RhGne-Poulenc SantB, 94403 Vitry-sur-Seine,
`France. Received April 2, 1990
`
`A variety of synthetic analogues of taxol, a naturally occurring antitumor diterpene, were examined for their potency
`to inhibit microtubule disassembly. For some of the compounds, the in vitro cytotoxic properties showed a good
`correlation with the tubulin assay. This structure-activity relationship study shows that inhibition of microtubule
`disassembly is quite sensitive to the configuration at (2-2’ and C-3’. A correlation between the conformation of the
`side chain at C-13 and the activity is suggested. Of all the compounds examined, one of the most potent in inhibiting
`microtubule disassembly and in inhibiting murine P388 leukemic cells, N-debenzoyl-N-tert-(butoxycarbony1)- 10-
`deacetyltaxol, named taxotere, was selected for evaluation as a potential anticancer agent.
`
`Several antitumor drugs prevent the formation of the
`mitotic spindle during cell division by interfering with the
`tubulin-microtubules system. Among the different classes
`of natural “mitotic spindle poisons”, the anticancer di-
`terpene taxol’ promotes the assembly of microtubules and
`inhibits the disassembly process of microtubules to tubu-
`lin2s3 in contrast to the vinblastine and colchicine type
`compounds which prevent microtubule assembly. Among
`natural substances, relationships between structure and
`microtubule assembly in vitro have been reported mostly
`for vinblastine! colchicine$ maytansine: podophyllotoxin,’
`and steganacine.8 A good correlation between the inhib-
`ition of tubulin assembly and cytotoxicity has been shown
`for some of these compounds. In the vinblastine series,
`a new hemisynthetic “Vinca alkaloid”, 5’-noranhydrovin-
`blastine or Navelbines was selected by using this in vitro
`assay as a possible useful chemotherapeutic agent and this
`compound is now used in clinics.1°
`In the taxol series, investigation of the structureactivity
`relationships has been limited because of the poor avail-
`ability of taxol (1) from natural sources (only 50-150
`mg/kg of dried trunk bark can be isolated from several
`
`8 Dedicated to Professor G. B. Marini-Bettolo on the occasion
`of his 75th anniversary.
`‘Centre National de la Recherche Scientifique.
`Centre de Recherches de Vitry.
`
`species of yew (genus Taxus, family Taxaceae)’J1).
`However, some closely related taxol congeners, mostly
`
`(1) Wani, M. C.; Taylor, H. L.; Wall, M. E.; Coggon, P.; McPhail,
`A. T. J. Am. Chem. SOC. 1971,93, 2325-2327.
`(2) Suffness, M.; Cordell, G. A. The Alkaloids, Chemistry and
`Pharmacology; Brossi, A., Ed.; Academic Press. Inc.: New
`York, 1985; Vol XXV, Chapter 1.
`(3) (a) Schiff, P. B.; Fant, J.; Horwitz, S. B. Nature 1979, 277,
`665-667. (b) Manfredi, J. J.; Fant, J.; Horwitz, S. B. Eur. J.
`Cell Biol. 1986, 42, 126.
`(4) Zavala, F.; Guhard, D.; Potier, P. Experientia 1978,34,1497.
`(5) Rosner, M.; Capraro, H. G.; Jacobson, A. E.; Atwell, L.; Brossi,
`A,; Ioro, M. A,; Williams, T. H.; Sik, R. M.; Chignell, C. F. J.
`Med. Chem. 1981,24, 257, and references cited therein.
`(6) Higashide, E.; Asai, M.; Ootsu, K.; Tanida, S.; Kozai, Y.; Ha-
`segawa, T.; Kishi, T.; Sugino, y.; Yoneda, M. Nature, Lond.
`1977, 270, 721.
`(7) Gender, W. J.; Murthy, C. D.; Trammell, M. H. J. Med. Chem.
`1977, 20, 635.
`(8) Zavala. F.: GuCnard. D.: Robin. J-P.: Brown. E. J. Med. Chen.
`.
`,
`
`.
`,
`
`-
`1980, 23, 546.
`(9) Maneenev. P.: Andriamialisoa. 2.: Lallemand. Y.: Lannlois. Y.:
`.
`.
`. .
`Lanaois,”N.; Potier, P. Tetrahekron 1979, 35, 2175.
`(10) For an overview of the clinical studies, see: Seminars in On-
`cology 1989, 16, No. 2, Suppl4.
`(11) (a) Miller, R. W.; Powell, R. G.; Smith, C. R., Jr.; Arnold, E.;
`Clardy, J. J. Org. Chem. 1981, 46, 1469-1474. (b) SCnilh, V.;
`Blechert, S.; Colin, M.; GuCnard, D.; Picot, F.; Potier, P.;
`Varenne, P. J. Nat. Prod. 1984,47, 131-137. (c) Magri, N. F.;
`Kingston, D. G. I. J . Org. Chem. 1986,51, 797-802.
`
`
`
`0022-2623/91/1834-0992$02.50/0 0 1991 American Chemical Society
`
`CabRef0001252
`
`AVENTIS EXHIBIT 2026
`Mylan v. Aventis
`IPR2016-00627
`
`

`
`Structure and Antimitotic Activity of Taxol Analogues
`
`Journal of Medicinal Chemistry, 1991, Vol. 34, No. 3 993
`
`Table I. Structures of Taxol Analogues and Their Inhibition of Pig Brain Microtubule Diassembly”
`
`kw6b
`RZ
`
`R1
`
`IDM/IDm
`R3
`ref
`compd
`23
`CH=CHC6H6
`H
`H
`20b, 21
`4
`100
`CH=CHCH3
`COCH3
`H
`20b
`5
`CH(OH)CH(OH)CeH,
`H
`H
`20b
`7‘
`3
`60
`CH(OH)CH(OH)CH3
`COCH3
`H
`20b
`8‘
`loo0
`CH=CHC6H6
`COZCHzCC13
`COZCHzCC13
`20,21
`1 la
`CH=CHCeHe
`COCH3
`COZCH2CCls
`20,21
`l l b
`-
`The isolation of tubulin from pig brain and inhibition studies were carried out as previously described.13 *ID, is the concentration of
`drugs leading to a 50% inhibition of the rate of microtubule disassembly. The ratio IDSo/ID, (taxol) gives the activity with regard to taxol
`itself (taxol: IDSO (pM) = 0.4). CThreo compounds 2’R,3’R + 2’S,3’S.
`
`acylated at C-2‘ and/or C-7,12-16 have been prepared as
`have water-soluble taxol derivatives, thereby showing that
`esters at C-2’ can serve as prodrugs of taxol.16
`Although previous studies have mostly highlighted the
`importance of the side chain in the 13-position for the in
`vitro disassembly13-assembly14 process and for the cyto-
`toxic activity,’J4 we were interested in studying in more
`details the effects of structural and/or configurational
`modifications at carbons 2’ and 3’ of the side chain and
`carbons 7 and 10 of the taxane skeleton, on the biological
`activity.
`To overcome the serious problems posed by the poor
`availability of taxol (1) and its derivatives, we have used
`simpler natural taxane-type compounds as “chemical
`precursors” to synthesize more complex taxol-like products.
`In this way, 10-deacetylbaccatin I11 (2a), easily extracted
`from the yew 1eave~’’J~~ and baccatin I11 (3a), isolated from
`the heartwood’* or prepared from 2a,19 can be used as
`
`(a) Kingston, D. G. I.; Magri, N. F.; Jitrangsri, C. New Trends
`Nat. Prod. Chem. 1986, 26, 219. (b) Mellado, W.; Magri, N.
`F.; Kingston, D. G. I.; Garcia-Arenas, R.; Orr, G. A.; Horwitz,
`S. B. Riochem. Riophys. Res. Commun. 1984, 224, 2; 329.
`(a) Lataste, H.; SBnilh, V.; Wright, M.; GuBnard, D.; Potier, P.
`Proc. Natl. Acad. Sci. U.S.A. 1984, 81, 4090. (b) SBnilh, V.
`PhD Thesis, March 2, 1984, UniversitC de Paris-Sud, Orsay.
`Parness, J.; Kingston, D. G. I.; Powell, R. G.; Harracksingh, C.;
`Horwitz, S. B. Biochena. Biophys. Res. Commun. 1982, 105,
`1082.
`Magri, N. F.; Kingston, D. G. I. J . Nat. Prod. 1988, 298-306.
`Deutsch, H. M.; Glinski, J . A.; Hernandez, M.; Haugwitz, R.
`D.; Narayanan, V. L.; Suffness, M.; Zalkow, L. H. J. Med.
`Chem. 1989,32, 788-792.
`Chauvisre, G.; GuCnard, D.; Picot, F.; SBnilh, V.; Potier, P. C.R.
`Acad. Sci. Paris 1981, 293, 501-503.
`(a) Della Casa de Marcano, D. P.; Halsall, T. G. J. Chem. SOC.
`Chem. Comm. 1975, 365. (b) Miller, R. W.; Powell, R. G.;
`Smith, C. R., Jr.; Arnold, E.; Clardy, J. J. Ore. Chem. 1981,46,
`1469-1474. (c) SCnilh, V.; Blechert, S.; Colin, M.; GuBnard, D.;
`Picot, F.; Potier, P.; Varenne, P. J. Not. Prod. 1984, 47,
`131-137.
`
`starting materials for the preparation of taxol and deriv-
`atives. Recently, we reported two partial syntheses of taxol
`(1) and structural analogues from 2a and 3a. One ap-
`proach relies on the double bond functionalization of
`cinnamoyl derivatives of taxane,20p21 the other makes use
`of the direct esterification of a taxol-like side chain on
`10-deacetylbaccatin III.’9921c
`Using these two synthetic approaches, we have been able
`to prepare a number of new taxol-like substances and
`consequently to further study the structure-activity rela-
`tionships in this series. The potential antimitotic activities
`of these new compounds have been investigated by using
`the in vitro tubulin assay. For some of the compounds,
`the in vitro cytotoxic properties were also determined.
`Chemistry
`Structures of compounds, literature references con-
`cerning their preparation together with their activity on
`tubulin are given in Tables I and 11.
`Esterification of cinnamic, crotonic, and 3-phenyl-
`propionic acids with compounds 2b or 3b, followed by
`
`(19) Denis, J-N.; Greene, A.; GuBnard, D.; Gugritte-Voegelein, F.;
`Mangatal, L.; Potier, P. J . Am. Chem. SOC. 1988, 120,
`5917-5919.
`(20) (a) SBnilh, V.; GuBritte, F.; GuBnard, D.; Colin, M.; Potier, P.
`(b)
`C.R. Acad. Sci. Paris, 1984, 299, SBrie 11, 1039-1049.
`GuBritte-Voegelein, F.; SBnilh, V.; David, B.; GuBnard, D.;
`Potier, P. Tetrahedron 1986, 42, 4451-4460.
`(21) (a) Colin, M.; GuBnard, D.; GuBritte-Voegelein, F.; Potier, P.
`(Rh6ne-Poulenc SantC) Eur. Pat. Appl. EP 253,738 (Cl.
`C07D305/14), January 20, 1988, Fr Appl86/10,400, 17 July
`1986; Chem. Abstr. 1988,209,22762~. (b) Mangatal, L.; Ad-
`eline, M-T.; Gudnard, D.; GuBritte-Voegelein, F.; Potier, P.
`Tetrahedron 1989, 42, 4451. (c) Mangatal, L., PhD Thesis,
`April 19, 1989, UniversitB de Paris XI Orsay. (d) Colin, M.;
`GuCnard, D.; GuBritte-Voegelein, F.; Potier, P. (Rhhe-Poulenc
`SantB) Eur. Pat. Appl. EP 253,739 (CI.C07D305/14), Jan 20,
`1988, Fr Appl86/10,401, Jul17,1986; Chem. Abstr. 1988,109,
`22763~.
`
`CabRef0001253
`
`

`
`994 Journal of Medicinal Chemistry, 1991, Vol. 34, No. 3
`
`GuBritte- Voegelein et al.
`
`Table 11. Inhibition of Pig Brain Microtubule Disassembly by Taxol Analogues at 4 O c a
`
`compd
`
`ref
`
`R1
`
`20a
`20a
`20a
`20a
`21
`21
`21
`21
`
`H
`6
`9a (2’R)
`H
`9b (2’s)
`H
`10a (3’s or 3’R)
`H
`10b (3’R or 3’s)
`H
`12a (2’R,3’S)
`H
`12b (2’S,3’R)
`H
`12c (2’R,3’S)
`H
`12d (2’S,3’R)
`H
`13a (2’R,3’S)
`H
`13b (2’S,3’R)
`H
`13c (2’R,3’S)
`H
`13d (2’S,3’R)
`H
`H
`1 3ea
`H
`13F
`14a (2’R,3’S)
`H
`14b (2’S,3’R)
`H
`14c (2’R,3’S)
`H
`14d (2’S,3’R)
`H
`H
`1 4ea
`H
`14F
`15 (2’R,3’S)
`H
`16a (2’R,3’S)
`H
`16b (2’S,3’R)
`H
`16c (2’R,3’S)
`H
`16d (2’S,3’R)
`H
`l a (2’R,3’S)
`H
`17b (2’S,3’R)
`H
`17c (2’R,3’S)
`H
`17d (2’S,3’R)
`H
`18 (2’R,3’S)
`H
`H
`19aa
`H
`19ba
`20 (2’R,3’S)
`H
`H
`21 (2’R,3’S)
`22 (2’R,3’S)
`CO(CH2)&02H
`23 (2’RJ’S)
`CO(CH2)&02H
`COCH2NH2
`24 (2’R,3’S)
`25 (2’R.3’S)
`COCH(NH9)CHGHc.
`-.
`, . ” ”
`a Erythro compounds 2’R,3’R or 2’S,3’S.
`
`21
`21
`21
`21
`
`21c
`21
`21
`21
`21
`21
`21
`21
`21
`
`R2
`
`H
`H
`H
`H
`H
`H
`H
`H
`H
`H
`H
`H
`H
`H
`H
`H
`H
`H
`H
`H
`H
`H
`COCHS
`COCH,
`COCH3
`COCH3
`COCH3
`COCH3
`COCH3
`COCH3
`COCH3
`H
`H
`H
`H
`COCH,
`CO(CH2)3C02H
`COCHzNHz
`H
`
`R3
`
`H
`OH
`OH
`H
`H
`OH
`OH
`NHTs
`NHTs
`OH
`OH
`NHCOztBu
`NHCOztBu
`OH
`OH
`OH
`OH
`NHCOPh
`NHCOPh
`OH
`OH
`OH
`OH
`OH
`NHCOztBu
`NHCOZtBu
`OH
`OH
`NHCOPh
`NHCOPh
`OH
`OH
`OH
`OH
`OH
`OH
`OH
`OH
`OH
`
`R4
`
`H
`H
`H
`NHC02tBu
`NHCOztBu
`NHTs
`NHTs
`OH
`OH
`NHC0,tBu
`NHC02tBu
`OH
`OH
`NHC02tBu
`NHC0,tBu
`NHCOPh
`NHCOPh
`OH
`OH
`NHCOPh
`NHCOPh
`NH2
`NHC0,tBu
`NHCOztBu
`OH
`OH
`NHCOPh
`NHCOPh
`OH
`OH
`NH,
`NH,
`NH,
`NHkO(CH2)3C02H
`NHCO(CGH&S03H
`NHCOPh
`NHC02tBu
`NHC0,tBu
`NHCOdBu
`
`ID,/ID,(taxol)
`17
`4.5
`3.5
`2.3
`4.1
`5
`-
`15
`-
`0.5
`30
`10
`160
`1.8
`4.3
`1.3
`4
`10
`170
`1.3
`1.3
`-
`0.5
`30
`10
`108
`1
`4.5
`10
`110
`44
`30
`30
`1
`5.5
`1
`2
`1.2
`1
`
`deprotection, led, respectively, to esters 4, 5, and 6.
`2’,3’-Dihydroxy derivatives 7 and 8 were easily obtained
`from esters 4 and 5. Coupling of racemic O-(l-ethoxy-
`ethyl)-3-phenyllactic acid with 2b led, after deprotection,
`to a mixture of esters 9a and 9b in a 82/18 ratio showing
`that an asymmetric induction in favor of the 2’R isomer
`took place during esterification.21c Under the same con-
`ditions, coupling of L-O-( l-ethoxyethyl)-3-phenyllactic acid
`yielded compounds 9a and 9b in a 60140 ratio, showing
`that epimerization at C-2’ also occurred during esterifi-
`cation. Racemic N-tert-(butoxycarbonyl)-3-amino-
`propionic acid yielded the (2-3’ functionalized derivatives
`10a and 10b after removal of the protecting groups.
`Application of the Sharpless vicinal oxyamination re-
`action2* to cinnamate derivatives such as lla using chlo-
`ramine T or tert-butyl-N-chloro-N-argentocarbamate
`followed by deprotection of the C-7 and C-10 hydroxyl
`groups afforded, respectively, a mixture of threo hydroxy
`p-toluenesulfonamide isomers 12a-d and threo hydroxy-
`carbamates 13a-d. Compound 13a was correlated with
`
`(22) Sharpless, K. B.; Patrick, D. W.; Truesdale, L. K.; Biller, S. A.
`J. Am. Chem. Soc. 1975,97, 2305-2307.
`
`10-deacetyltaxol(14a) while 2‘-epi,3‘-epi, 10-deacetyltaxol
`14b was obtained from 13b. In the same way regioisomers
`14c and 14d were prepared from the oxyaminated com-
`pounds 13c and 13d. Amino alcohol 15 was prepared from
`hydroxycarbamate 13a after deprotection of the amino
`group. The same reactions were applied to the cinnamate
`derivative of baccatin I11 1 l b to provide oxycarbamates
`16a-d, N-benzoyl-3-phenylisoserine isomers 1 (taxol),
`17b-d, and amino alcohol 18. Taxol (11, 10-deacetyltaxol
`(14a), and hydroxycarbamates were also obtained by direct
`esterification.
`Erythro isomers 13e,f and 14e,f were prepared from
`cinnamate ester 1 la. Epoxidation of 1 la yielded a mixture
`of two diastereoisomers which were treated with sodium
`azide. After reduction and deprotection with zinc dust in
`acetic acid, two amino alcohols 19a and 19b were obtained.
`Treatment of compounds 19a and 19b with benzoyl
`chloride or di-tert-butyl dicarbonate yielded, respectively,
`the erythro isomers of 10-deacetyltaxol 14e,f and the er-
`ythro hydroxycarbamates 13e,f. Compounds 20 and 21
`were obtained by condensing the appropriate anhydride
`with the amino alcohol 15. Esterification of taxol (1) and
`compound 13a with glutaric anhydride led, respectively,
`
`CabRef0001254
`
`

`
`Structure and Antimitotic Activity of Taxol Analogues
`to products 22 and 23. Finally, acylation of 13a with
`suitably protected amino acids such as glycine and phe-
`nylalanine gave compounds 24 and 25. All new compounds
`were principally characterized by NMR and MS.
`Results and Discussion
`Different in vitro assays have been used to determine
`the activity of taxol congeners on tubulin (promotion of
`microtubule assembly in the absence of GTP,14 inhibition
`of binding of tritiated taxol to microtubule^,'^ and inhib-
`ition of microtubule disassembly at 4 OCU$lc). Concerning
`the drug-tubulin interaction, the two procedures involving
`inhibition of microtubule assembly or inhibition of mi-
`crotubule disassembly gave the same results. However,
`because of the rapidity of the latter method (5 min per
`sample), all the compounds described in Tables I and I1
`were assayed for their ability to inhibit the disassembly
`process of microtubules at 4 “C.
`Previous structure-activity studies have shown that (a)
`the side chain at C-13 is necessary for a good drug-receptor
`interaction,lJ3J4 and (b) modification of substituents at
`C-10 and/or C-7 such as replacement of a hydroxyl group
`by an acyl or xylosyl group has little effect on the activi-
`ty.12b,13,14
`With respect to structural modifications on the side
`chain, structural modifications have mainly been made at
`C-2’ and have shown that acylation of this carbon results
`in a loss of activity in the tubulin assay12bJ3 but not in the
`cytotoxicity assay. These observations recently led to the
`preparation of water soluble derivatives of taxol.16
`The ID,, values obtained in this study (Tables I and 11)
`are in good agreement with those previously described and
`also provide much more information concerning the spe-
`cific influence of configuration and structural modifications
`of the side chain on the activity of taxol-type molecules:
`(a) Replacement of the C-10 acetoxy group with a hydroxyl
`group did not lead to loss of potency in different series of
`taxane derivatives (Compare series 13a-d with 16a-b, and
`14a-d and 1,17b-d). (b) Replacement of a 3’-phenyl group
`with a methyl group resulted in a major loss of activity.
`Thus the 2’,3’-dihydroxycrotonyl ester 8 is 20-fold less
`potent than its cinnamoyl analogue 7. (c) Branching of
`large groups such as [ (trichloroethyl)oxy]carbonyl at C-7
`and/or (2-10 (lla,b) resulted in a loss of activity whereas
`compounds with polar substituents such as xylo~yl,’~ glu-
`taryl (22,23), or aminoacyl (24,25) are as potent as taxol
`in the tubulin assay. (d) No loss of potency was observed
`for compounds having different kinds of hydrophobic
`substituents on the amido group at C-3’. Thus, replace-
`ment of the phenyl group in taxol with tiglyl (cephalo-
`manine),I4 tosyl (12a), or hexanoyl groups13b gave com-
`pounds as active as taxol. Moreover, the tert-butyloxy-
`carbonyl compounds (13a,16a) were shown to be the most
`potent inhibitors of microtubule disassembly so far pre-
`pared by us. In contrast, compounds having a free amino
`group at C-3’ are less potent than their N-amido analogues
`(compare 18 and taxol (1)).
`More interesting are the results obtained with com-
`pounds having different configurations at (3-2’ and/or C-3’.
`Both threo ((2’R,3’S) 13a,14a,16a and 1; (2’S,3’R)
`13b,14b,16b, and 17b) and erythro ((2’R,3’R; 2’S,3’S) 13e,
`13f, 14e, and 14f) diastereoisomers were assayed, showing
`that inhibition of microtubule disassembly is quite sen-
`sitive to these two configurations. Thus in all cases, the
`2’R,3‘S diastereoisomer (natural configuration), was found
`to be the most potent. If this is true, then one question
`remains unanswered with regard to our observations: do
`the differences in activity seen among the various ana-
`logues synthesized arise from the presence or absence of
`
`Journal of Medicinal Chemistry, 1991, Vol. 34, No. 3 995
`t
`
`IQH
`
`Figure 1. Three-dimensional view of compound 13a.
`particular functionalities per se or are these the result of
`differences in the side chain conformations imposed by
`these modifications, resulting in a favorable or unfavorable
`fit to‘the activity site?
`The conformation of taxol-like compounds is imposed
`by the three-ring system having the highly strained
`eight-membered ring B cis-fused to ring A and trans-fused
`to ring C and, in addition, a bridgehead double bond, a
`hindering geminal dimethyl group, and a planar oxetane
`ring. This particular structure gives rise to a “caged-type
`conformation”. A recent X ray analysis of the 2’R,3’S
`compound 13a,24 giving interatomic bond lengths, showed
`that the side chain adopts a particular conformation due
`to intramolecular hydrogen bonds and repulsive interaction
`between the substituents at carbons 2’ and 3’ and those
`of the taxane skeleton (Figure 1). Moreover, recent
`ROESY experiment^^^ with threo isomers showed that
`compounds having a 2’R,3’S configuration exhibit a num-
`ber of NOE’s, indicating interactions between C-3’H and
`the C-4 acetyl group and between C-2’H and C-18H3. In
`contrast, the 2’S,3’R diastereoisomers are characterized by
`NOE involving C-2’H/C-4 acetyl group and C-3’H/C-18H3.
`Though these observations can also be predicted by mo-
`lecular modeling, they need to be confirmed by other NMR
`experiments under different conditions of temperature and
`solvent. Configurations at C-2’ and/or C-3’ of compounds
`10a,b and erythro isomers 13e,f and 14e,f are still unknown
`
`(23) Beloeil, J-C.; Dubois, J.; Gillet, B.; Guenard, D.; Gueritte-
`Voegelein, F. Unpublished results.
`(24) Gugritte-Voegelein, F.; Gugnard, D.; Mangatal, L.; Potier, P.;
`Guilhem, J.; Cesario, M.; Pascard, C. Acta Crist. 1990, C46,
`781-784.
`(25) Lavelle, F.; Fizames, C.; Gueritte-Voegelein, F.; Gubnard, D.;
`Potier, P. Proc. Am. Assoc. Cancer Res. 1989,30, 2254.
`
`CabRef0001255
`
`

`
`996 Journal of Medicinal Chemistry, 1991, Vol. 34, No. 3
`
`Table 111. Cvtotoxicitv of Taxol Analogues
`P388O
`IDW/IDm(13a)'
`IC,/ICW(13a)*
`IC,
`compd
`0.27
`taxol (1)
`2
`2
`taxotere (13a)
`0.13
`1
`1
`> 10
`>77
`60
`13b
`20
`4
`31
`13c
`1
`0.17
`1.3
`16a
`7
`54
`17b
`9
`20
`6
`46
`17c
`>4
`>31
`21
`11
`1.4
`1.5
`0.19
`24
`0.12
`2
`1
`25
`"Murine P388 leukemic cells were obtained from the tumor
`bank of the National Cancer Institute. P388 cells were grown at 37
`"C as suspensions in RPMI 1640 medium containing 10 pM 2-
`mercaptoethanol, 2 mM L-glutamine, 200 U/mL penicillin, 200
`pg/mL streptomycin, and supplemented with 10% (v/v) fetal calf
`serum. Exponentially growing P388 cells suspended in complete
`medium were seeded in tubes at IO6 cells/mL.
`the compounds
`were added at different concentrations on day 0 (3 tubes/concen-
`tration). Cells were allowed to grow for 3 or 4 days at 37 "C under
`5 % COP Final cell numbers were measured by using a Coulter
`counter. The results were expressed as the concentration (pg/mL)
`which inhibits 50% of the cell proliferation (ICw). The ICM were
`estimated by regression analysis concentration-response data.
`*Cytotoxicity of drugs in comparison to compound 13a. cIDM for
`disassembly of microtubule: see Table 11. The ratio IDw/IDM-
`(13a) gives the activity with regard to 13a itself(l3a: IDM (pM) =
`0.2).
`but some information can also be provided by ROESY
` experiment^.^^ These studies could show the direct in-
`fluence of the side-chain conformation on the inhibition
`of microtubule disassembly. It is already interesting to
`note that the gain in activity, in going from compound 6,
`having no substituent at (2-2' and C-3', to compound 13a,
`bearing hydroxyl and hydroxycarbamate groups at C-2'
`and C-3', may be the product of the separate contributions
`brought by the substituents at carbons 2' (compound 9)
`and 3' (compound 10).
`Other structural features such as the benzoate group at
`C-2 or the oxetane ring are probably essential for exhibiting
`a good activity. Indeed, products lacking these groups,
`though identified by others in yew extracts (i.e. taxanes
`with C4-C20 double bond),26 were not isolated by using
`the tubulin bioassay guided fra~tionation."~J~~
`Previous results have shown that taxanes lacking the
`side chain at C-13 have the same affinity as taxol for
`Physarum t ~ b u 1 i n . l ~ ~ This may be the result of a point
`mutation on the peptide chain in the neighborhood of the
`binding site, allowing a specific noncovalent bond between
`the side chain and mammalian tubulin; moreover, the side
`chain by itself is inactive and has no effect on the binding
`of taxol or its analogues. These two observations allow us
`to imagine the binding process to occur as follows: (a) the
`taxane skeleton is recognized and binds to its site on tu-
`bulin, and (b) the drug-tubulin bond is stabilized by the
`specific interaction of the side chain, a situation which is
`probably not restricted to the field of taxane derivatives.
`Concerning the in vitro cytotoxic activity, a generally
`good correlation with the tubulin assay may be noted:
`compounds such as 1, 13a, 16a, 24, and 25 have the ability
`to inhibit both cell growth and disassembly of microtubules
`(Table 111). However, the C-2' esters, less active in di-
`sassembling microtubules, are promptly hydrolysed in
`intra- or extracellular media, resulting in good cytotoxic
`activity.I2b,l3,14,16
`
`(26) Ud- Khan, N.; Parveen, N. J. Sci. Ind. Res. 1987,46,512, and
`referenced cited therein.
`
`~~
`
`~
`
`GuBritte-Voegelein et al.
`Some attempts have been made to solubilize taxol de-
`rivatives in water, but the conditions used to obtain sol-
`ubilization are incompatible with the stability of the final
`products (Le. acidic or basic medium). The diglycine de-
`rivative 24, soluble in water as its hydrochloride salt, is a
`potent inhibitor of microtubule disassembly and in vitro
`cellular proliferation but has no effect in vivo on murine
`le~kemia.~' The other amino acid or acid derivatives are
`good inhibitors of disassembly but they are only partially
`soluble in an ethanol-water mixture.
`Conclusion
`About 40 synthetic taxane-type compounds have been
`tested as potential inhibitors of disassembly of mammalian
`tubulin. The tubulin assay has proven to be a convenient
`method for studying structure-activity relationships,
`particularly in regard to the conformations of the "active"
`molecules. Correlation with activity against P388 murine
`leukemia shows that the tubulin assay is also a very effi-
`cient tool for a preliminary evaluation of new active
`products in the taxane family.
`Of all the compounds examined in Tables I and 11, one
`the most potent, N-debenzoyl-N-(tert-butoxy-
`of
`carbonyl)-lO-deacetyltaxol(13a) named taxotere, was se-
`lected for evaluation as a potential anticancer agent and
`so far has shown excellent antitumor activity against
`several models of grafted murine tumors.26 Moreover
`taxotere (13a) showed a better solubility in excipient
`system (polysorbate 80/ethanol, 1:l) than the two others
`most active compounds taxol (1) and N-debenzoyl-N-
`(tert-butoxycarbony1)taxol (16a).
`Experimental Section
`The purity of the samples was checked by chromatographic
`methods (HPLC and TLC) and careful analysis of NMR spectra.
`Melting points were taken on a Kofler hot bench and are un-
`corrected. Optical rotations (c, cg/mL) were determined on a
`Perkin-Elmer 141 MC polarimeter using a 10 cm path length cell.
`Infrared spectra (cm-', CHC1, or Nujol) were recorded on a
`'H and 13C NMR were
`Perkin-Elmer 257 spectrophotometer.
`recorded on a Bruker AM 200 or AM 400 spectrometer. Chemical
`shifts are in ppm relative to TMS (0.00). Multiplicities are in-
`dicated in parentheses with coupling constants expressed in hertz.
`Mass spectra were recorded on an AEI MS9 (CI) or on a Kratos
`MS8O (FAB). The 10-deacetylbaccatin I11 used in this study was
`isolated from the leaves of the yew tree Taxus baccata L.
`General procedures for esterification of 7,10-bis[ [ (2,2,2-tri-
`chloroethyl)oxy]carbonyl]-l0-deacetylbaccatin I11 (2b) with
`different acids and for the deprotection of C-7 and/or C-10 troc
`group are described in refs 20b and 21.
`3-(Phenylpropionyl)-lO-deacetylbaccatin 111 (6). Esteri-
`fication of 2b (500 mg, 0.56 mmol) with 3-phenylpropionic acid
`(330 mg, 2.2 mmol) gave 93% of the corresponding ester which
`was deprotected to yield compound 6 (70%): MS-CI m / z 677
`(MH'); 'H NMR (CDCl,) 6 1.11 (s, C-17H3), 1.21 (s, C-16H3), 1.74
`(s, C-19H3), 1.82 (s, C-18H3), 2.20 (s, OCOCH,), 2.74 (m, C-2'H2),
`3.05 (m, C-3'Hz), 3.91 (d, J = 7, C-3H2), 4.18 and 4.31 (s d, J =
`9, C-20H2), 4.23 (m, C-7H), 4.95 (d, J = 9, C-5H), 5.20 (s, C-IOH),
`5.67 (d, J = 7, C-2H), 6.17 (t, J = 8, C-l3H), 7.25 and 7.33 (C,H,),
`7.50, 7.62, and 8.06 (OCOCGH6).
`Compounds 9a and 9b. O-(l-Ethoxyethyl)-3-phenyllactic acid
`was prepared by classical procedures from 3-phenyllactic acid
`(first, protection of the carboxyl group as a benzyl ester (MS-E1
`m / z 256 (M'+)), second, protection of the hydroxyl group as
`ethoxyethyl ether (MS-E1 m / z 328 (M'+)), third, cleavage of the
`benzyl ester by hydrogenolysis).
`Esterification of 2b (669 mg, 0.75 mmol) with DL-O-(l-eth-
`oxyethyl)-3-phenyllactic acid (714 mg, 3 mmol) gave 98% of a
`mixture of two esters which were deprotected to give 9a (56%)
`and 9b (12%). Diastereoisomers were separated by silica gel
`column chromatography using hexane/ethyl acetate (2:8) as
`
`(27) Lavelle, F. Personal communication.
`
`CabRef0001256
`
`

`
`Structure and Antimitotic Activity of Taxol Analogues
`
`Journal of Medicinal Chemistry, 1991, Vol. 34, No. 3 997
`
`eluant. Under the same conditions, esterification of 2b with
`L-O-( 1-ethoxyethyl)-3-phenyllactic acid gave after deprotection
`a 60/40 mixture of 9a and 9b.
`(C = 1.00, MeOH); MS-FAB' m/z 693 (MH');
`9a: [a]=D = -57'
`'H NMR (CDC13) 6 1.11 (8, C-17H3), 1.22 (8, C-16H3), 1.72 and
`1.73 (29, C-19H3 and C-18H3), 1.82 (m, C-6H), 2.12 (m, C-14Hz),
`2.21 (s, OCOCH3), 2.55 (m, C-6H), 3.12 (m, C-3'Hz), 3.85 (d, J
`= 7, C-3H), 4.18 (m, C-7H), 4.15 and 4.27 (2 d, J = 8, C-20Hz),
`4.50 (m, C-2'H), 4.92 (d, J = 9, C-5H), 5.17 (s, C-lOH), 5.62 (d,
`J = 7, C-2H), 6.11 (t, J = 9, C-l3H), 7.18,7.25,7.30 (CeH5), 7.48,
`7.60, and 8.05 (OCOC6H5).
`(c = 1.00, MeOH); MS-FAB' m/z 693 (MH+);
`9 b [a]l"D -37'
`'H NMR (CDCl3 + CD30D) 6 1.11 (8, C-17&), 1.22 (s, C-16H3),
`1.75 (8, C-19H3), 1.84 (s, C-18H3), 1.92 (m, C-6H), 2.15 (dd, J =
`15 and 9, C-l4H), 2.26 (8, OCOCH3), 2.28 (m, C-14H), 2.53 (m,
`C-6H), 3.03 and 3.24 (2 dd, C-3'Hz), 3.84 (d, J = 7, C-3H), 4.15
`(m, C-7H), 4.18 a i d 4.28 (2 d, J = 9, C-20Hz), 4.47 (dd, J = 8 and
`4.5, C-2'H), 4.96 (d, J = 9, C-SH), 5.20 (s, C-lOH), 5.66 (d, J =
`7, C-2H), 6.17 (t, J = 9, C-13H), 7.25 (C&5), 7.45, 7.60, and 8.00
`(OCOCsH,).
`Compounds 10a and lob. DL-3-[ (tert-Butoxycarbony1)-
`amino]-3-phenylpropionic acid was prepared in 86% from DL-3-
`amino-3-phenylpropionic acid.21c Esterification of this acid (999
`mg, 3.77 mmol) with 2b (838 mg, 0.94 mmol) gave a mixture of
`two diastereoisomeric esters in 98% yield. Removal of the pro-
`tecting groups at C-7 and C-10 provided diastereoisomers 10a
`(60%) and 10b (40%) after purification by HPLC (RP-18; mobile
`phase, H,O/MeOH 36:64; flow rate; 2 mL min-').
`loa: mp 160 'c (MeOH); [a]"D = -18'
`(c = 1.10, CHCl,);
`MS-FAB+ m/z 792 (MH+), 774, 692, 527, 509, 266; 'H NMR
`(CDC13) 6 1.08 (8, C-ITHa), 1.15 (9, C-l6H3), 1.42 (9, tBu), 1.72
`(C-18H3) and C-19H3), 1.82 (m, C-6H), 2.13 (m, C-14H2), 2.28 (e,
`OCOCH3), 2.56 (m, C-6H), 2.93-3.10 (m, C-2'Hz), 3.85 (d, J = 7,
`C-3H), 4.18 (m, C-7H), 4.15 and 4.25 (2 d, J = 9, C-20Hz), 4.94
`(d, J = 9, C-5H), 5.15 (s, C-lOH), 5.18 (br d, C-3'H), 5.60 (br d,
`NH), 5.70 (d, J = 7, C-2H), 6.05 (t, J = 8, C13H), 7.30 (C&), 7.50,
`7.60, and 8.05 (OCOC&,); 13C NMR (CDCl3) 69.90 (C19), 14.40
`(C18), 20.40 (C17), 22.60 (CH3-acetate), 26.50 (ClS), 28.40
`(CH,-tBu), 36.00 (C14), 36.80 (C6), 41.60 (C-2'), 43.00 (C15), 46.70
`(C3), 54.7 ((239, 57.80 (C8), 70.30 (C7), 71.90 (C13), 74.60 (ClO),
`75.10 (C2), 76.50 (C20 and C-l), 78.80 (C-tBu), 81.20 (C4), 84.30
`(C5), 126.10, 127.70, 128.70, 128.80, 130.10, 133.70 (0-Bz, m-Bz,
`p-Bz, o-Ph, m-Ph, p-Ph), 129.50 (Cl-Bz), 135.90,138.70 and 138.50
`(C11, C12, and C1-Ph), 155.20 ( C 4 of carbamate), 166.90 (C=O
`of Bz), 169.80 (C=O of Ac), 170.60 (Cl'), 211.13 (C9).
`lob: mp 156 'c (MeOH); [(YlzZD = -12'
`(c = 0.90, CHCI,);
`MS-FAB+ m/z 792 (MH'), 774, 692, 527, 509, 266; 'H NMR
`(CDC13) 6 1.10 (s, C-17H3), 1.18 (s, C-16H3), 1.43 (e,

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