throbber
Pharmac. Ther. Vol. 55, pp. 31-51, 1992
`Printed in Great Britain. All rights reserved
`
`Specialist Subject Editor: E. HAMEL
`
`0163-7258/92 $15.00
`© 1992 Pergamon Press Ltd
`
`NATURAL PRODUCTS WHICH INTERACT WITH
`TUBULIN IN THE VINCA DOMAIN:
`MAYTANSINE, RHIZOXIN, PHOMOPSIN A,
`DOLASTATINS 10 AND 15 AND
`HALICHONDRIN B
`
`ERNEST HAMEL
`
`Laboratory of Molecular Pharmacology, Developmental Therapeutics Program, Division of
`Cancer Treatment, National Cancer Institute, National Institutes of Health, Bethesda,
`MD 20892, U.S.A.
`
`Abstract-This paper summarizes published data on the interactions of tubulin with antimi(cid:173)
`totic compounds that inhibit the binding of vinca alkaloids to the protein. These are all
`relatively complex natural products isolated from higher plants, fungi and marine invertebrate
`animals. These agents are maytansine, rhizoxin, phomopsin A, dolastatins lO and 15 and
`halichondrin B and their congeners. Effects on tubulin polymerization, ligand binding
`interactions and structure-activity relationships are emphasized.
`
`CONTENTS
`
`I. Introduction
`2. The Maytansinoids
`3. Rhizoxin
`4. Phomopsin A
`5. Dolastatin 10
`6. Dolastatin 15
`7. Halichondrin B, Homohalichondrin B and Halistatins 1 and 2
`8. One Laboratory's Comparative Data
`9. A Preliminary Model of the 'Vinca Domain' of Tubulin
`Acknowledgements
`References
`
`31
`32
`35
`37
`38
`41
`42
`44
`44
`47
`47
`
`I. INTRODUCTION
`
`Antimitotic agents almost universally alter microtubule assembly reactions. With the exception
`of taxol and its congeners (Schiff et al., 1979), the primary effect of these compounds, the one
`observed at lower drug concentrations, is inhibition of the reaction. With the exception of
`estramustine and its phosphate derivative (Stearns and Tew, 1985; Wallin et al., 1985), these
`inhibitors all interact with tubulin, the predominant protein component of micro tubules. Although
`tubulin molecules are not uniform, since there is minor primary structure variability (Krauhs et
`al., 1981; Ponstingl et al., 1981) and substantial variability resulting from post-translational
`modification (Eipper, 1972; Arce et al., 1975; Maruta et al., 1986; Edde et al., 1990; Alexander et
`al., 1991), each tubulin unit in a microtubule consists of two highly similar polypeptide chains (ex(cid:173)
`and p -tubulin) and two guanosine nucleotides. The two polypeptide chains are nearly identical in
`molecular weight (about 50 kDa) and have approximately 40% absolute sequence homology and
`about 70% homology if conservative amino acid substitutions are considered (Krauhs et al., 1981;
`Ponstingl et al., 1981). One guanosine nucleotide is in the form of GOP in the microtubule while
`in unpolymerized tubulin it can be in the form of either GOP or GTP. This nucleotide is
`
`31
`
`IMMUNOGEN 2001, pg. 1
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`32
`
`E. HAMEL
`
`exchangeable with GOP or GTP in solution in unpolymerized tubulin (Weisenberg eta/., 1968;
`Kobayashi, 1974; Levi et a/., 1974; Caplow and Zeeberg, 1980; Lin and Hamel, 1987), but it
`becomes inaccessible in polymerized tubulin (Weisenberg et a/., 1976; Arai and Kaziro, 1977;
`David-Pfeuty et a/., 1977). The exchangeable nucleotide has been localized to the P-subunit of
`tubulin (Geahlen and Haley, 1977; Nath eta/., 1985). The second guanosine nucleotide is always
`in the form of GTP and is always inaccessible to solution nucleotides. Even in cells its half-life
`appears to be equal to that of tubulin itself (Spiegelman et a/., 1977). The location of the
`nonexchangeable nucleotide has not been determined, but the sequence homology of the tubulin
`subunits suggests it may be on cc-tubulin.
`Inhibitors of microtubule assembly fall into two broad classes, which have been essentially
`defined by their effects on the binding to tubulin of two commercially available radio labeled drugs.
`Most inhibitors of polymerization, including virtually all synthetic compounds, inhibit the binding
`of radiolabeled colchicine to tubulin and do not affect the binding of radiolabeled vinblastine to
`tubulin. Generally, despite rather diverse but relatively uncomplicated molecular structures, when
`carefully examined such compounds show a competitive pattern of inhibition against colchicine.
`Inhibitors of radiolabeled vinblastine binding are much less common. Thus far, all such
`inhibitors are natural products (or closely related synthetic analogs) and their structures are
`complex. These agents are the subject of this paper. None of them inhibit the binding of
`radiolabeled colchicine to tubulin. Their effects on vinblastine and vincristine binding fall into both
`competitive and noncompetitive patterns. In addition, all interfere with interactions of guanosine
`nucleotides at the exchangeable site. Finally, while no colchicine-site drug has a role in the
`treatment of neoplastic diseases, the efficacy of the vinca alkaloids in cancer chemotherapy (see
`Rowinsky and Donehower, 1992) lends particular interest to the diverse group of natural products
`that interfere with the tubulin-vinblastine interaction.
`
`0
`"-c-R
`I
`
`0
`
`0
`
`II
`
`R=CH-N-C-~
`
`CH
`
`I 3
`
`I
`CH3
`
`MaytanSine:
`
`Ansamitocin P-3:
`
`Ansamitocin P-4:
`
`FIG. I. Structural formulae of maytansine, ansamitocin P-3 and ansamitocin P-4.
`
`IMMUNOGEN 2001, pg. 2
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Interactions of tubulin in the vinca domain
`
`33
`
`2. THE MAYTANSINOIDS
`
`Maytansine (NSC 153858; structure presented in Fig. I) is the most thoroughly studied member
`of a series of ansa macrolide compounds isolated and characterized by Kupchan and his
`collaborators (Kupchan eta/., 1972, 1974, 1975, 1977, 1978). These agents were obtained from the
`plants Maytenus ovatus, M. buchananii, M. serrata and the related Putterlickia verrucosa.
`Chemically related compounds have also been isolated from Colubrina texensis, a member of a
`different plant family (Wani eta/., 1973) and from a Nocardia species (Higashide eta/., 1977; Asai
`et a/., 1979). The best studied compounds from the microorganism are ansamitocin P-3 and
`ansamitocin P-4 (structures in Fig. 1). Maytansine (Remillard eta/., 1975; Wolpert-DeFilippes et
`a/., 1975a,b; Kupchan et al., 1978; Bai eta/., 1990b) and the ansamitocins (Higashide eta/., 1977;
`Ootsu et a/., 1980) are highly cytotoxic compounds (see below) which inhibit mitosis, with cells
`accumulating in apparent metaphase arrest (condensed chromosomes; absent nuclear membrane;
`no spindle or poorly defined spindle with few if any microtubules). Except for taxol (Rowinsky
`and Donehower, 1992), maytansine is the only antimitotic agent to have gone through extensive
`clinical trials for the treatment of neoplastic diseases (e.g. Cabanillas eta/., 1979; Rosenthal eta/.,
`1980), but thus far no useful role for the drug has been established in clinical practice. Recently
`maytansine derivatives have been coupled to tumor-specific antibodies in a new approach to clinical
`use of this agent (Chari et a/., 1992).
`Radiolabeled maytansine binds to tubulin in a reversible reaction which is relatively fast, occurs
`at ooc and is inhibited by vincristine (Mandelbaum-Shavit eta/., 1976) and vinblastine (Batra et
`a/., 1986). The apparent Kd value for the reaction at 37°C is 0.7 JI.M (Mandelbaum-Shavit eta/.,
`1976). Vincristine was found to act as a competitive inhibitor of maytansine with an apparent K;
`value of 10 JI.M (Mandelbaum-Shavit et al., 1976). Neither vincristine (Mandelbaum-Shavit eta/.,
`1976) nor vinblastine (Batra eta/., 1986) displaces all radiolabeled maytansine bound to tubulin.
`This phenomenon has not been studied in detail, but may be related to observations of Takahashi
`et a/. (1987b) on apparently contradictory effects of vinblastine on the binding of radiolabeled
`rhizoxin to tubulin (competitive) as compared with the effects of rhizoxin on the binding of
`radiolabeled vinblastine (not purely competitive) (see below).
`In contrast, nonradiolabeled maytansine almost totally displaces radiolabeled vinblastine or
`vincristine from tubulin (Mandelbaum-Shavit e.t al., 1976; Batra et a/., 1986; Safa et al., 1987).
`Maytansine has been found to competitively inhibit the binding of both vinca agents to tubulin.
`Apparent K; values vs vincristine have been reported as 0.4 JI.M (Mandelbaum-Shavit eta/., 1976;
`York eta/., 1981) and 3.1 JI.M (Bai eta/., 1990a) and versus vinblastine as 0.5 JI.M (Bhattacharyya
`and Wolff, 1977) and 0.9 JI.M (Bai eta/., 1991). Lacey eta!. (1987) found that the IC50 value of
`maytansine versus radiolabeled vinblastine was 1.6 Ji.M. All workers have concluded that may(cid:173)
`tansine binds to tubulin with greater affinity than these two vinca agents. In addition, ansamitocin
`P-3 inhibits the binding of radiolabeled vinblastine to tubulin, but the data indicated that the
`inhibition was not competitive (Takahashi et al., 1987b).
`Maytansine, like vinblastine, has only minor effects on the binding of radiolabeled colchicine to
`tubulin (Mandelbaum-Shavit eta/., 1976; Batra et al., 1986; Lacey eta/., 1987). Unlike vinblastine,
`however, maytansine does not stabilize the colchicine binding activity of tubulin (Bhattacharyya
`and Wolff, 1977; Bai eta/., 1990a). Tubulin stabilization by vinblastine, but not maytansine, has
`also been demonstrated by the ability of the former, but not the latter, to reduce the rate at which
`bis(8-anilinonaphthalene-l-sulfonate) interacts nonspecifically with denatured hydrophobic regions
`of tubulin (Prasad et a/., 1986).
`Maytansine has specific effects on the alkylation of tubulin sulfhydryl groups, which have been
`studied by Luduefia and his colleagues (Luduefia and Roach, 198la,b; Luduefia eta/., 1982; Roach
`and Ludueiia, 1984). This work has been reviewed in detail in this series (Luduefia and
`Roach, 1991). In brief, these workers have evaluated the effects of a large variety of tubulin
`ligands, including many antimitotic agents, on the pattern of alkylation observed with
`iodoacetamide and on the formation of intrapolypeptide chain crosslinks in p -tubulin following
`reaction with the bifunctional agent N,N'-ethylenebis(iodoacetamide). There are two potential
`major crosslinks. The first, between cys 239 and cys 354 (Little and Ludueiia, 1985) (referred
`to throughout this paper as the 'first crosslink'), occurs in all tubulin preparations. The second,
`
`IMMUNOGEN 2001, pg. 3
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`34
`
`E. HAMEL
`
`between cys 12 and either cys 201 or cys 211 (Little and Ludueiia, 1987) (referred to as the
`'second crosslink'), only occurs in nucleotide-depleted tubulin with its formation strongly inhibited
`by GTP. Maytansine had minimal effects on alkylation by iodoacetamide, in contrast to strong
`inhibition by vinblastine. Formation of the first crosslink was enhanced in the presence of both
`maytansine and vinblastine, while formation of the second crosslink was almost completely
`inhibited by maytansine and weakly inhibited by vinblastine. Since the exchangeable nucleotide site
`has been localized to the f3 -subunit, this finding suggests that the maytansine binding site may also
`be on f3 -tubulin.
`Our own studies also led to this conclusion. Maytansine potently inhibits tubulin-dependent GTP
`hydrolysis (Lin and Hamel, 1981; Bai et al., 1990b) and nucleotide binding to tubulin, particularly
`at lower reaction temperatures (Huang et al., 1985; Bai et al., l990a, 1991). Maytansine does not
`displace nucleotide bound in the exchangeable site (Huang et al., 1985; Bai et al., 1990a), and,
`moreover, the drug prevents nucleotide loss from tubulin during gel filtration chromatography (Lin
`and Hamel, 1987). The most reasonable explanation for these observations is that maytansine binds
`to f3 -tubulin in a manner that prevents entry and exit of guanosine nucleotides at the exchangeable
`site. We also have evaluated vinblastine as an inhibitor of nucleotide exchange on tubulin (Huang
`et al., 1985; Bai et al., 1990a, 1991). Although the vinca alkaloid has some effect on the reaction,
`significant inhibition is only observed at high concentrations (at least 100 /lM).
`In contrast to results from the alkylation and GTP exchange experiments with maytansine,
`however, are genetic results. The only maytansine-resistant mutant reported thus far has been a
`Chinese hamster ovary cell line with an altered oc-tubulin (Schibler and Cabral, 1985). Takahashi
`eta/. (1990) have, however, described an interesting manipulation by site-directed mutagenesis in
`a /3-tubulin gene of Schizosaccharomyces pombe. Ileu 100 was converted to asn 100 (analogous to
`the situation in mammalian f3 -tubulin) and an organism with increased sensitivity to ansamitocin
`P-3, as well as rhizoxin (see below), was produced.
`Maytansine and the ansamitocins inhibit microtubule assembly requiring microtubule-associated
`proteins (Remillard eta/., 1975; Bhattacharyya and Wolff, 1977; Kupchan eta/., 1978; Ootsu eta/.,
`1980; York eta/., 1981; Fellous eta/., 1985; Huang eta/., 1985; Lacey eta/., 1987; Takahashi et
`a/., 1987a) and the glutamate-induced polymerization of purified tubulin (Bai eta/., 1990b). With
`microtubule-associated proteins, a higher concentration of maytansine is required with micro(cid:173)
`tubule-associated protein 2 as compared with tau factor to obtain equivalent inhibition (Fellous
`et a/., 1985). With the exception of one report where an IC50 value for polymerization of 0.4 /lM
`was obtained (Bhattacharyya and Wolff, 1977), most workers have obtained IC50 values for
`maytansine and the ansamitocins in the 1-5 /lM range (Remillard eta/., 1975; Ootsu eta/., 1980;
`Huang et al., 1985; Lacey eta/., 1987; Takahashi eta/., 1987a; Bai et al., 1990b). Despite binding
`more avidly to tubulin than vinblastine (see above), in direct comparisons maytansine (and the
`ansamitocins) has always been found to be less effective than vinblastine as an inhibitor of
`polymerization (Bhattacharyya and Wolff, 1977; Ootsu et al., 1980; Huang et al., 1985; Lacey et al.,
`1987; Takahashi eta/., 1987a; Bai et a/., 1990b).
`/lM cause extensive
`Concentrations of both maytansine and the ansamitocins below 20
`disassembly of preformed microtubules (Remillard et al., 1975; Ootsu et a/., 1980) and the
`ansamitocins cause intracellular microtubules to disappear (Ootsu et a/., 1980).
`Unlike the vinca alkaloids (reviewed by Himes, 1991), neither maytansine nor the ansamitocins
`induce formation of spiral aggregates oftubulin (Bhattacharyya and Wolff, 1977; Ootsu eta/., 1980;
`Fellous et al., 1985; Takahashi et al., 1987a). Instead maytansine potently inhibits this reaction,
`for maytansine at concentrations substoichiometric to those of both tubulin and vinblastine (e.g.
`2 /lM maytansine, 10 /lM tubulin, 140 /lM vinblastine) totally prevents tubulin spiralization (Fellous
`et al., 1985). In addition, maytansine causes dissolution of preformed vinblastine-induced
`aggregates (Fellous et al., 1985).
`There is minimal information about tubulin interactions with any maytansinoids other than the
`ansamitocins, which differ solely in the ester substituent at position C(3) (see Fig. 1). York et al.
`(1981) examined four analogs as inhibitors of vincristine binding to tubulin. A compound with an
`altered ester substituent at position C(3) was essentially equivalent to maytansine, but three analogs
`with no substituent at this position and a 2-3 double bond were significantly less potent as
`inhibitors. One of these (which also lacked the N-methyl group and the 4-5 epoxide) had an
`
`IMMUNOGEN 2001, pg. 4
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Interactions of tubulin in the vinca domain
`
`35
`
`apparent noncompetitive pattern of inhibition, but its K; value was 5 J.lM as opposed to the 0.4 J.lM
`value obtained for maytansine. Kupchan et al. (1978) obtained initial results with a large series of
`analogs. They compared inhibitory effects on microtubule assembly, mitosis of fertilized sea urchin
`eggs and growth of KB carcinoma cells in culture. There was little correlation between effects
`observed in the three assays and they only described compounds with significant inhibitory effects
`on the assembly reaction. The following modifications (see Fig. I) did not seem to reduce
`maytansine's effectiveness as an inhibitor of assembly: altered ester at position C(3), hydroxyl group
`at position C(3), etherification of the hydroxyl at position C(9), thioether instead of hydroxy group
`at position C(9), no substituent at position C(3) together with introduction of a 2-3 double bond
`(in contrast to the finding of York eta/., 1981), noN-methyl group and acetyl group at position
`C(l5).
`The crystal structure of maytansine 3-bromopropyl ether has been summarized (Kupchan et a/.,
`1972, 1974; Bryan et al., 1973). Substituents are oriented in a manner that minimizes intramolecular
`repulsions. The two longer sides of the 19-member ring are approximately parallel and about 5.4
`A apart. The ring appears to be open in its center. Thr face of the ring with the ester group is
`relatively hydrophilic, the opposite face hydrophobic.
`
`3. RHIZOXIN
`
`The fungus Rhizopus chinensis is the etiologic agent for a disease known as rice seedling blight
`in which there is an abnormal swelling of plant roots secondary to the failure of cell division
`(Ibaragi, 1973). A number of compounds which reproduced the disease process were isolated and
`characterized by Iwasaki and coworkers (Iwasaki eta/., 1984, 1986a,b; Kobayashi eta/., 1986). The
`most important of these agents was called rhizoxin (NSC 332598; structure in Fig. 2). The most
`prominent structural feature of rhizoxin is its 16-member macrolide ring. Besides its toxicity for
`plant tissues, rhizoxin also has antitumor (Tsuruo eta/., 1986) and antifungal (Iwasaki eta/., 1984)
`activity and it causes the accumulation of cells arrested in mitosis (Tsuruo eta/., 1986; Bai eta/.,
`1990b). Rhizoxin is in the early stages of clinical evaluation in human cancer patients (Bisset eta/.,
`1992).
`Radiolabeled rhizoxin binds rapidly to tubulin at 37°C and the reaction is reversible. Scatchard
`analysis of binding data indicated one high affinity binding site with a Kd value of0.2 J.lM (Takahashi
`et a/., 1987b). Binding of the drug to tubulin was inhibited by both vinblastine and ansamitocin
`P-3 (Takahashi eta/., 1987b). Lineweaver-Burk analysis indicated that both inhibitors were acting
`in a competitive manner, with apparent K; values of 0.1 J.lM for ansamitocin P-3 and 3 J.lM for
`vinblastine (Takahashi et al., 1987b). The binding of radiolabeled rhizoxin to tubulin is also
`strongly inhibited by phomopsin A (Li eta/., 1992). The IC50 value obtained with 3 J.lM rhizoxin
`was just over 0.1 J.lM for phomopsin A, but the type of inhibition (competitive or noncompetitive)
`was not defined in this study.
`
`0
`
`FIG. 2. Structural formula of rhizoxin.
`
`IMMUNOGEN 2001, pg. 5
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`36
`
`E. HAMEL
`
`Takahashi et a/. (1987b) reported that inhibition by rhizoxin of the binding of radiolabeled
`vinblastine to tubulin did not appear to be purely competitive, although the apparent Ki value for
`rhizoxin was quite low, about 0.01 JlM. In my laboratory we have found that rhizoxin was a classic
`competitive inhibitor of the binding ofradiolabeled vincristine to tubulin, with an apparent~ value
`of 12 JlM (Bai et a/., 1990a).
`Rhizoxin does not inhibit the binding of colchicine to tubulin (Takahashi eta/., 1987b) nor does
`it stabilize the colchicine site (Bai et a/., 1990a). Further, it does not alter the rate of interaction
`of bis(S-anilinonaphthalene-1-sulfonate) with tubulin (Sullivan et a/., 1990).
`The effect of rhizoxin on the alkylation of tubulin sulfhydryls in terms of crosslink formation
`was examined and qualitatively it was found to behave identically to maytansine, with increased
`formation of the first crosslink and markedly reduced formation of the second crosslink (Sullivan
`eta/., 1990).
`Rhizoxin, like maytansine, inhibits tubulin-dependent GTP hydrolysis (Bai et a/., 1990b) and
`guanosine nucleotide exchange at the exchangeable site (Bai eta/., 1991). It is, however, less potent
`than maytansine as an inhibitor in both reactions (see below).
`In an elegant series of experiments Takahashi and coworkers (Takahashi et a/., 1989,
`1990) isolated rhizoxin-resistant mutants of Aspergillus nidulans and found an altered {3 -tubulin.
`This was demonstrated to result from replacement of asn 100 with an isoleucine moiety.
`Further, they found that both Schizosaccharomyces pombe and Sacchoromyces cerevisiae were
`naturally resistant to rhizoxin and lacked asparagine at position 100. When site-directed muta(cid:173)
`genesis was employed to place asparagine at position 100 in both yeasts, rhizoxin-sensitive strains
`were obtained.
`Microtubule assembly was inhibited by rhizoxin with an IC50 value of 5 J,LM; the agent was less
`potent than vinblastine and equivalent to ansamitocin P-3 (Takahashi et a/., 1987a). It was
`somewhat less potent as an inhibitor of the glutamate-induced polymerization of purified tubulin.
`Its IC 50 value was about 7 JlM, approximately 6 times that of vinblastine and twice that of
`maytansine (Bai eta/., 1990b, 1991). Rhizoxin also causes extensive depolymerization of preformed
`microtubules (Takahashi et a/., 1987a).
`There is no evidence that rhizoxin induces formation of spiral aggregates (Takahashi et al.,
`1987a,b). In fact, like maytansine, rhizoxin potently inhibits the vinblastine-induced reaction, for
`the drug at 2 JlM eliminated spiral formation with 10 JlM tubulin and 100 JlM vinblastine (Takahashi
`eta/., 1987b).
`Several analogs of rhizoxin have been evaluated for their effects on microtubule assembly
`(Takahashi et al., 1987a). Equivalent activity was observed in compounds in which the meth(cid:173)
`oxy substituent at position C(l7) was replaced with an hydroxyl group, in which the 2-3 epoxide
`was replaced with a double bond, or in which both epoxide groups were replaced with double
`bonds. A four-fold loss of activity was observed when the lactone group was disrupted. Major loss
`of activity occurred when either epoxide was hydrolyzed and replaced with an hydroxyl group at
`position C(3) or C(12), or if the position C(13) hydroxyl group was esterified.
`The side chain of rhizoxin has been studied for its role in the compound's antitubulin
`properties (Kato et a/., 1991). Significant loss of activity in inhibiting microtubule assembly
`occurred with all compounds with less than six carbon atoms in the side chain. That is, C(16)
`through C(21) are required for good antitubulin activity. Replacement of the distal side chain with
`a carbonyl oxygen at C(20) led to complete loss of activity, while its replacement with a methylene
`group (that is, restoring C(21) alone) yielded a compound with almost full activity. Reduction
`of the inactive C(20) ketone to the alcohol did not restore activity, but several esters of this
`alcohol had partial activity. All changes made in the distal portion of the side chain, beyond
`C(21), had relatively minor effects on inhibitory properties of the derivatives relative to that of
`rhizoxin. These changes included conversion of configuration at C(22}-C(23) from trans to cis,
`reduction of the C(20}-C(21) double bond, introduction of an ester linkage at C(22) and
`several derivatives in both cis and trans configuration at C(21). This work led Sawada et a/.
`(1991) to prepare fluorescent and photoaffinity analogs of rhizoxin derivatized at position
`C(22). Both analogs bound to tubulin and their binding was substantially reduced in the
`presence of rhizoxin. They may prove useful as probes for the mechanism of binding of the drug
`to tubulin.
`
`IMMUNOGEN 2001, pg. 6
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`Interactions of tubulin in the vinca domain

`:,;c,
`CO-NH-C
`CHz
`H...,.
`7 'co
`ru_NJ.l_._._c/ 9
`110
`8
`61
`s I
`,.en
`'
`NH
`HO'
`
`/CH3
`
`.... '"]'.&&
`
`37
`
`Cl
`
`OH
`
`2S
`24
`23
`22
`21
`3 /C..........17 019 20
`2
`O-C
`CO-N-c-CO-NH-C-CO-NH-C-~H
`18
`I "CH3CHz
`II
`II
`CH3
`C
`C26
`/\
`/\
`H ~H
`CH3 C~~
`27
`
`H
`
`\
`
`FIG. 3. ~ 1ctural formula of phomopsin A.
`
`The crystal structure of one relatively inactive analog has been evaluated (Iwasaki eta/., 1986b).
`In the compound studied the side chain projects out from the 16-member ring.
`
`4. PHOMOPSIN A
`
`The fungus Phomopsis leptostomiformis grows on lupins and produces toxins which cause
`lupinosis, a severe liver disease of grazing animals. One of the pathologic changes observed in
`lupinosis is hepatocytes arrested in mitosis. Culvenor and coworkers (Culvenor eta/., 1977, 1983;
`Mackay et a/., 1986; Lacey et a/., 1987) isolated and characterized active components in fungal
`extracts. The predominant agent was named phomopsin A (NSC 381839; structure presented in
`Fig. 3). It is a hexapeptide formed from modified amino acids. Its structure includes a 13-member
`ring containing an ether linkage. Phomopsin A was used to produce experimental lupinosis,
`including the characteristic mitotic hepatocytes (Culvenor et a/., 1977). In addition, it caused
`mitotic arrest of cultured cells (Petterson eta/., 1979; Tonsing et al., 1984; Bai eta/., 1990b ), leading
`tothe prediction that it would interfere with microtubule assembly (Petterson et al., 1979).1t should
`be noted that phomopsin A in tissue culture studies has generally been used at micromolar
`concentrations (Petterson eta/., 1979; Tonsing eta/., 1984; Bai eta/., 1990b) and its IC50 value for
`the growth (at 24 hr) of Ll210 murine leukemia cells is 7 J.lM (Bai et a/., 1990b). This IC50 value
`is about 100-10,000-fold higher than that obtained with other natural product antimitotic agents
`(see below).
`Li eta/. (1992) recently reported the preparation of radiolabeled phomopsin A by fermentation
`with C4C)isoleucine. The peptide bound rapidly and stably to tubulin at 37°C and Scatchard
`analysis indicated two classes of binding site with Kd values of 10 and 300 nM. Rhizoxin and
`ansamitocin P-3 weakly inhibited the binding of the radiolabeled phomopsin A to tubulin, with
`IC50 values of about I 00 J.lM, while the inhibitory effect of vinblastine was still more feeble. Whether
`these inhibitory effects were competitive or noncompetitive was not determined. Although Li
`et a/. (1992) reported that they found no evidence for phomopsin A-induced aggregation of
`tubulin, other workers have described such aggregates (see below). This is of importance in
`evaluation of binding data. Ligand-induced aggregation of a protein can result in biphasic
`Scatchard plots even though there is actually only a single binding site for the ligand (Timasheff
`et al., 1991).
`Phomopsin A was initially reported to inhibit the binding of radio labeled vinblastine to tubulin
`more effectively than either maytansine or nonradiolabeled vinblastine, with an IC50 value of 0.8
`J.lM. The type of inhibition was not determined in this study (Lacey et a/., 1987). Subsequently,
`phomopsin A was demonstrated to be a classic noncompetitive inhibitor of the binding of
`radiolabeled vincristine to tubulin, with an apparent Ki value of 2.8 J.lM (Bai et a/., 1990a).
`Phomopsin A moderately enhances the binding ofradiolabeled colchicine to tubulin (Lacey eta/.,
`1987; Luduefia et a/., 1989; Bai et a/., 1990a). Moreover, it seems to completely stabilize tubulin.
`
`JPT SS/1-D
`
`IMMUNOGEN 2001, pg. 7
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`38
`
`E. HAMEL
`
`There is little or no decay of colchicine binding in its presence (Luduefia et al., 1989, 1990; Bai
`eta/., 1990a), nor any increase in the amount of binding of bis(8-ani1inonaphthalene-l-sulfonate)
`(Ludueiia et a/., 1989).
`Alkylation of tubulin sulfhydryl groups is altered by phomopsin A in a manner that resembles
`effects of both vinblastine and maytansine (Luduefia et al., 1990). Like vinblastine, phomopsin A
`strongly inhibits the reaction of tubulin with iodoacetamide and enhances formation of the first
`crosslink. Like maytansine, phomopsin A strongly inhibits formation of the second crosslink.
`Tubulin-dependent GTP hydrolysis is potently inhibited by phomopsin A and it is slightly more
`potent than maytansine (Bai et al., 1990b). The peptide is also a powerful inhibitor of nucleotide
`exchange on tubulin, although, like maytansine, phomopsin A does not displace GDP bound in
`the exchangeable site (Bai et al., 1990a). In fact, phomopsin A is the most potent inhibitor of
`nucleotide exchange yet examined (Bai et al., 1991). It is particularly notable in that exchange is
`inhibited almost as extensively at 37°C as at ooc. We have interpreted the loss of inhibition of
`nucleotide exchange by maytansine at 37°C as resulting from the reversibility of maytansine's
`binding to tubulin (Huang et al., 1985). If correct, this explanation implies that phomopsin A will
`dissociate very slowly, if at all, once bound to the protein. Initial studies with radiolabeled
`phomopsin A support this hypothesis (Li et al., 1992), as does the peptide's potent stabilization
`of tubulin (see above).
`In direct comparisons to other drugs phomopsin A was equivalent to vinblastine and more
`effective than maytansine as an inhibitor of microtubule assembly from microtubule protein (Lacey
`et al., 1987) and of the glutamate-induced polymerization of purified tubulin (Bai et al., 1990b).
`IC50 values of 0.6 J1M for inhibition of microtubule assembly (Tonsing et al., 1984; Lacey et al.,
`1987), of 0.4 J1M for inhibition of glycerol-induced polymerization of purified tubulin (Tonsing
`et al., 1984) and of 1.4 J1M for inhibition of glutamate-induced polymerization (Bai et al., 1990b)
`have been reported. Phomopsin A causes the disappearance of microtubules in cultured cells and
`the complete in vitro depolymerization of preformed microtubules (Tonsing et al., 1984).
`The disappearance of preformed microtubules was not associated with a complete loss of
`turbidity (Tonsing et al., 1984). Instead, the polymer was replaced by aggregates consisting of rings
`and spirals. These appeared to be much smaller and morphologically distinct (see below) from those
`induced by the vinca alkaloids (see Himes, 1991 and Fig. 4A). Rings and spirals were formed from
`microtubules derived from both microtubule protein and purified tubulin in glycerol (Tonsing et al.,
`1984). In our laboratory we have observed formation of morphologically similar oligomeric
`structures from unpolymerized tubulin in the presence of superstoichiometric concentrations of
`phomopsin A (Fig. 4B). These structures have been observed in reaction mixtures containing 0.1 M
`4-morpholine ethanesulfonate and heat-treated microtubule-associated proteins (Fig. 4B). In
`reaction mixtures containing only purified tubulin our initial studies indicate that rings, both single
`and in apparently aggregated clusters, are the predominant structure, with relatively few spirals.
`Substoichiometric concentrations of phomopsin A inhibit the formation of the vinblastine(cid:173)
`induced aggregate (4 J1M phomopsin A, 10 J1M tubulin, 40-150 J1M vinblastine), but the peptide is
`less potent than maytansine in this regard (Ludueiia et al., 1989).
`Lacey eta/. (1987) examined three analogs of phomopsin A for effects on microtubule assembly
`and vinblastine binding and they found little difference between the four compounds. The equally
`active agents were phomopsin B, which lacks the chloride atom at position C(l4), octahydropho(cid:173)
`mopsin A, which has the four carbon-carbon double bonds reduced, and phomopsinamine A,
`which lacks the substituent at position N(24).
`The crystal structure of phomopsin A (Mackay et al., 1986) has two molecules per unit, with
`the phenyl ring having somewhat different orientations. The side chain projects out from the
`13-member ring in both molecules.
`
`Pettit and his collaborators have been isolating cytotoxic agents produced by the marine animal
`Dolabella auricularia, a shell-less mollusc known as a sea hare (Pettit et al., 1987a,b, l989a,b,c,
`
`5. DOLASTATIN 10
`
`IMMUNOGEN 2001, pg. 8
`Phigenix v. Immunogen
`IPR2014-00676
`
`

`

`-::l c> .,
`
`0
`::l
`
`~ :::.
`"' 0 ....,
`2
`0"
`E.. :;·
`:;·
`:;.
`0
`
`FIG. 4. Spiral and ring oligomers formed in the presence of superstoichiometric concentrations of vinblastine (panel A), phomopsin A (panel B) and dolastatin
`I 0 (panel C). The reaction mixture used for the micrograph presented in panel A contained 1.0 mg/ml tubulin, 0.5 mg/mL heat-treated microtubule-associated
`proteins, 0.1 M 4-morpholineethanesulfonate (pH 6.9 with NaOH), 0.5 mM MgC12, 0.1 M GTP and 50 f1M vinblastine and was incubated

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