throbber
PCT
`
`WORLD INTELLECTUAL PROPERTY ORGANIZATION
`International Bureau
`
`INTERNATIONAL APPLICATION PUBLISHED UNDER THE PATENT COOPERATION TREATY (PC1)
`
`(51) International Patent Classification 5 :
`A61K 31/33, C12N 9/12
`C12Q 1/42, G0lN 33/48, 33/573
`C07D267/00
`
`(11) International Publication Number:
`
`WO 93/19752
`
`Al
`
`(43) International Publication Date:
`
`14 October 1993 (14.10.93)
`
`\.
`
`J' ,._
`
`(21) International Application Number:
`
`PCT/US93/02459
`
`(22) International Filing Date:
`
`19 March 1993 (19.03.93)
`
`(81) Designated States: AU, CA, JP, European patent (AT, BE,
`CH, DE, DK, ES, FR, GB, GR, IE, IT, LU, MC, NL,
`PT, SE).
`
`(30) Priority data:
`07/864,806
`
`7 April 1992 (07.04.92)
`
`US
`
`Published
`With international search report.
`
`(7l)Applicant: DANA-FARBER CANCER
`INSTITUTE,
`INC. [US/US]; 44 Binney Street, Boston, MA 02115
`(US).
`
`(72)1nventors: BIERER, Barbara, E. ; 399 Hammond Street,
`Newton, MA 02167-1225 (US). AVRUCH, Joseph; 277
`St. Paul Street, Brookline, MA 02146 (US).
`
`(74) Agent: FRASER, Janis, K.; Fish & Richardson, 225 Fran(cid:173)
`klin Street, Boston, MA 02110-2804 (US).
`
`(54) Title: INHIBITION OF P70 S6 KINASE
`
`(57) Abstract
`
`A method of screening for an antiproliferative or immunosuppressive agent, which method includes the steps of (1) con(cid:173)
`tacting a eukaryotic cell with a candidate antiproliferative or immunosuppressive composition; and (2) determining the level of
`activity of a serine/threonine kinase or a serine/threonine phosphatase in the p70 S6 kinase cascade of said cell in the presence
`of the candidate composition, wherein a level of said activity that results in a lower p70 S6 kinase activity in the presence of the
`composition than in the absence of the composition is an indication that the composition is antiproliferative or immunosuppres(cid:173)
`sive agent; and methods of treatment using such compositions.
`
`1·
`/ ..
`
`

`

`FOR THE PURPOSES OF INFORMATION ONLY
`
`Codes used to ;ctentify States party to the PCT on the front pages of pamphlets publishing international
`applications under the PCT.
`
`AT
`AU
`BB
`BE
`BF
`BG
`BJ
`BR
`CA
`CF
`CG
`CH
`Cl
`CM
`cs
`CZ
`DE
`DK
`ES
`Fl
`
`Au,tria
`Australia
`Barbados
`Belgium
`Burkina Faso
`Bulgaria
`Benin
`Brazil
`Canada
`Ccntml African Republic
`Congo
`Swiu.crland
`Cote d'Ivoire
`Cameroon
`c~.cchoslovakia
`(:...cch Republic·
`Germany
`Denmark
`Spain
`Finland
`
`FR
`GA
`GB
`GN
`GR
`HU
`IE
`IT
`JP
`KP
`
`KR
`KZ
`LI
`LK
`I.U
`MC
`MG
`ML
`MN
`
`France
`Gabon
`United Kingdom
`Guinea
`Greece
`Hungary
`Ireland
`Italy
`Japan
`Democratic People', Republic
`of Korea
`Republic of Korea
`K.!7.akhstan
`Liechtenstein
`Sri l.anl.a
`Luxembourg
`Monaco
`Mad~gascar
`Mali
`Mongolia
`
`MR
`MW
`NL
`NO
`NZ
`PL
`PT
`RO
`RU
`SD
`SE
`SK
`SN
`SU
`TD
`TG
`UA
`us
`VN
`
`Mauritania
`Malawi
`Netherlands
`Norway
`New Zealand
`Poland
`Portugal
`Romania
`Russian Federation
`Sudan
`Sweden
`Slovak Republic
`Senegal
`Soviet Union
`Chad
`Togo
`Ukraine
`United States of America
`Viet Nam
`
`-!
`
`r ,._~ ._" ..
`
`

`

`WO93/19752
`
`PCT /US93/02459
`
`- 1 -
`
`INHIBITION OF P70 S6 KINASE
`The invention was made in the course of work
`funded in part by a grant from the U.S. government, which
`5 has certain rights in the invention. The field of the
`invention is antiproliferative or immunosuppressive
`agents.
`
`Background of the Invention
`A conserved response of many eukaryotic cell types
`10 to mitogenic signals is the phosphorylation of multiple
`serine residues on the 40S ribosomal subunit protein S6
`(Erikson, J. Biol. Chem. 266:6007-6010, 1991). This
`phosphorylation increases the efficiency of protein
`synthesis, which appears to be required in several steps
`15 of cell cycle progression (Erikson, supra). Recently,
`two families of S6 kinases have been characterized at the
`enzymatic and molecular levels:
`the 85-90 kDa (rsk) S6
`kinase family (Jones et al., Proc. Natl. Acad. Sci. USA
`85:3377-3381,, 1988), referred to herein as p90 S6
`20 kinase, and the 70 kDa S6 kinase family (Banerjee et al.,
`Proc. Natl. Acad. Sci. USA 87:8550-8554, 1990; Kozma et
`al., Proc. Natl. Acad. Sci. USA 87:7365-7369, 1990),
`referred to herein as p70 S6 kinase. The activity of
`both types of kinases is regulated by serine/threonine
`25 phosphorylation (Erikson, supra), and both are
`serine/threonine kinases themselves.
`Eukaryotic cells contain many different kinases as
`part of various cascades that transmute signals from
`cell-surface receptors to effector molecules within the
`30 cell.
`In some cases the cell-surface receptor is itself
`a kinase that is activated upon binding its ligand; in
`other cases, the receptor is associated with a separate
`protein that acquires kinase activity when the receptor
`
`,.
`
`,,.
`
`

`

`WO93/19752
`
`PCT /US93/02459
`
`-
`
`2 -
`
`binds its ligand. The newly-activated kinase then
`phosphorylates the next member of the relevant cascade,
`thereby activating (or in some cases, deactivating) it.
`Phosphatases also form an integral part of the cascade,
`5 acting to remove the phosphate groups added by the
`kinases, and thereby deactivating (or in some cases,
`activating) the substrate polypeptide. In general, each
`member of such a cascade transmutes signals by
`sequentially phosphorylating (if it is a kinase) or
`10 dephosphorylating (if it is a phosphatase) certain
`critical residues in the next member of the cascade,
`thereby activating or deactivating such next member, as
`the case may be.
`In T cells expressing the interleukin-2 (IL-2)
`15 receptor, binding of IL-2 to this receptor triggers a
`response culminating in proliferation of the T cell
`(Smith, Ann. Rev. Immunol. 2:319-333, 1984). Evans et
`al. observed that one aspect of the IL-2 generated
`response is an increase in S6 phosphorylation and a
`20 concomitant increase in the rate of protein synthesis (J.
`Biol. Chem. 262:4624-4630, 1987). Similarly, the multi(cid:173)
`faceted response of insulin-dependent H4 hepatoma cells
`to stimulation by insulin includes an increase in the
`level of S6 phosphorylation, apparently attributable at
`25 least in part to increased activity of the S6 kinases.
`Rapamycin and FK506 are macrolide antibiotics
`which are potent immunosuppressive agents. Although the
`two compounds share certain structural features (see Fig.
`9) and are capable of binding to the same family of
`30 cellular proteins (termed FK506-binding proteins, or
`FKBPs) to form a biologically active complex, their
`mechanisms of immunosuppression differ significantly.
`The FK506/FKBP complex inhibits a very early step in
`antigen-induced T cell activation, preventing
`35 proliferation of activated T cells by blocking the
`
`

`

`WO93/19752
`
`PCT /US93/02459
`
`-
`
`3
`
`-
`
`induction of cytokine gene transcription. Liu et al.
`(Cell 66:807-815, 1991) have recently shown that this
`occurs through the binding of the FK506/FKBP complex to
`calcineurin, a calcium-dependent phosphatase thought to
`5 play a role in T cell signal transduction, thereby
`blocking the phosphatase activity of this enzyme and
`short-circuiting signal transduction along this pathway.
`In contrast, the rapamycin/FKBP complex apparently does
`not bind to or affect the activity of calcineurin, and
`10 acts by blocking a later step in antigen-induced T cell
`activation, one that occurs subsequent to binding of IL-2
`to its receptor (i.e., at a point after the initial
`induction of cytokine gene transcription). The entity
`directly targeted by the rapamycin/FKBP complex has not
`15 been identified, although it has been shown not to be the
`IL-2 receptor itself.
`
`Summary of the Invention
`As disclosed herein, it has now been shown that
`the addition of rapamycin to an IL-2-stimulated T cell
`20 line results in the inhibition of p70 S6 kinase, which
`kinase may constitute the direct target of the rapamycin
`effector complex, or may be downstream of the actual
`target. Furthermore, experiments are herein disclosed
`which demonstrate that rapamycin will inhibit both basal
`25 and insulin-stimulated proliferation and p70 S6 kinase
`activity in an insulin-dependent hepatoma cell line, an
`observation that has broad implications with respect to
`novel medical applications for rapamycin. The
`information linking p70 S6 kinase inhibition to the
`immunosuppressive and newly discovered general
`antiproliferative effects of rapamycin may be utilized to
`design an assay for screening for immunosuppressive
`and/or antipr9liferative agents which act by the same or
`a related mechanism to that of rapamycin:
`i.e., which
`
`30
`
`~
`
`•
`
`

`

`WO 93/19752
`
`PCT /US93/02459
`
`-
`
`4 -
`
`ultimately result in a decrease in p70 S6 kinase activity
`such an assay will pick up not
`in the treated cell.
`only those compounds or complexes which act on the same
`enzyme as that targeted by the rapamycin/FKBP complex,
`5 but also those which act upstream or downstream of that
`particular enzyme in the p70 S6 cascade. The screened
`compounds or complexes (generally referred to as
`compositions} which result in decreased p70 S6 kinase
`activity may act to inhibit a kinase (e.g., p70 S6 kinase
`10 itself, or a kinase upstream of p70 S6 kinase), or may
`activate a phosphatase (e.g., one which specifically
`dephosphorylates and thus inactivates p70 S6 kinase, or
`an enzyme upstream of p70 S6 kinase). The screen will
`also identify those compositions which activate a kinase
`15 or inhibit a phosphatase upstream of p70 S6 kinase,
`provided that this activation or inhibition ultimately
`results in a decrease in p70 S6 kinase activity.
`The screening method of the invention includes the
`steps of (1) contacting a eukaryotic cell (e.g., a
`20 mammalian cell such as a human cell} with a candidate
`antiproliferative or immunosuppressive composition; and
`(2) determining the level of activity of a
`serine/threonine kinase or a serine/threonine phosphatase
`in the p70 S6 kinase cascade of said cell in the presence
`25 of the candidate composition, wherein a level of said
`activity that results in a lower p70 S6 kinase activity
`in the presence of the composition than in the absence of
`the composition is an indication that the composition is
`an antiproliferative or immunosuppressive agent. The
`30 serine/threonine kinase may be p70 S6-kinase itself, or a
`kinase which phophorylates and thereby activates p70 S6
`kinase in vivo, or a kinase which acts upstream of that
`point. The serine/threonine phosphatase may be a
`phosphatase which dephosphorylates and thereby
`35 inactivates p70 S6 kinase in vivo, or a phosphatase which
`
`

`

`WO93/t9752
`
`PCT /US93/02459
`
`-
`
`5 -
`
`In preferred embodiments,
`acts upstream of that point.
`step 2 may be readily accomplished by simply measuring
`the level of p70 S6 kinase activity, which is directly or
`inversely related to the activities of each of the
`5 kinases and phosphatases upstream of p70 S6 kinase in the
`p70 S6 kinase cascade.
`In order to maximize the window
`of response in this assay, the cell may optionally be
`exposed to an appropriate mitogen (preferably a growth
`factor) before or during step 1. For example, where the
`10 cell is a hematopoietic cell, a cytokine such as
`interleukin 1 (IL-1), IL-2, IL-3, IL-4, IL-5, IL-6, IL-7,
`IL-8, IL-9, IL-10, IL-11, erythropoietin (EPO), Steel
`factor (stem cell factor); granulocyte colony stimulating
`factor (G-CSF), macrophage colony stimulating factor (M-
`15 CSF), or granulocyte/macrophage colony stimulating factor
`(GM-CSF) may be used to trigger proliferation signals;
`preferably, the cell is a lymphocyte such as a T cell or
`B cell, and the mitogen of choice is a lymphokine such as
`one of the interleukins.
`Alternatively, candidate antiproliferative or
`immunosuppressive agents (including, for example, analogs
`of rapamycin and analogs of the rapamycin/FKBP complex)
`may be screened in vitro in a method including the
`following steps:
`(1) combining (a) a sample containing p70 S6
`kinase, (b) a substrate for the kinase, and (c) a
`candidate antiproliferative or immunosuppressive
`composition; and
`(2) determining whether the candidate composition
`30 inhibits the biological activity of the kinase, such
`inhibition being an indication that the candidate
`composition is an antiproliferative or immunosuppressive
`agent. Alternatively, the in vitro assay may employ p70
`S6 kinase as the substrate, and an enzyme just upstream
`
`25
`
`20
`
`...
`
`.•
`
`

`

`WO93/19752
`
`PCT/US93/02459
`
`-
`
`6
`
`-
`
`of p70 S6 kinase as the effector. This method would
`include the following steps:
`(l) providing a sample containing p70 S6 kinase
`and either (i) a serine/threonine kinase capable of
`5 phosphorylating and activating p70 S6 kinase, or (ii) a
`serine/threonine phosphatase capable of dephosphorylating
`and deactivating p70 S6 kinase;
`(2) contacting the sample with a candidate
`antiproliferative or immunosuppressant composition; and
`(3) determining whether the amount of
`phosphorylation of the p70 S6 kinase in the presence of
`the candidate composition is lower than the amount of
`phosphorylation of p70 S6 kinase in the absence of the
`candidate composition, such a lower amount of
`l5 phosphorylation being an indication that the candidate
`composition is an antiproliferative or immunosuppressive
`agent.
`
`io
`
`The invention also includes methods of inhibiting
`the proliferation or immune response of a cell (e.g., a
`20 hematopoietic cell such as a T cell, stimulated by a
`cytokine such as IL-2) of an animal (preferably a mammal
`such as a human)r by introducing into the animal a
`composition that interrupts the p70 S6 kinase cascade via
`a mechanism such as (i) directly inhibiting the activity
`25 of the p70 S6 kinase of the cell, or (ii) directly
`inhibiting the activity of a kinase which phosphorylates
`in vivo a serine or threonine residue on p70 S6 kinase,
`or (iii) directly increasing the activity of a
`phosphatase which dephosphorylates in vivo a serine or
`30 threonine residue on p70 S6 kinase. By "directly" is
`meant that the altered activity of the given kinase or
`phosphatase results from the interaction between the
`composition (or a complex containing the composition) and
`the given kinase or phosphatase, and not between the
`35 composition and an enzyme upstream to the given kinase or
`
`

`

`WO93/19752
`
`PCT /US93/02459
`
`- 7 -
`
`phosphatase. Although it is not known which, if any, of
`these possible mechanisms is the mechanism of
`rapamycin/FKBP, the experimental results obtained with
`rapamycin indicate that a composition which functioned by
`5 any one of these mechanisms would produce the desired
`result.
`
`Also within the invention is a method of
`inhibiting cellular proliferation in response to a
`mitogen other than IL-2, which method includes the steps
`10 of (1) providing a cell which proliferates in response to
`the mitogen; and (2) treating the cell with a composition
`(which may include rapamycin or an analog of rapamycin)
`that modulates the activity of a serine/threonine kinase
`(such a p70 S6 kinase or a kinase which activates p70 S6
`15 kinase) or a serine/threonine phosphatase (such one which
`dephosphorylates, and thereby deactivates, p70 S6 kinase)
`in the p70 S6 kinase cascade of the cell, thereby
`resulting in a decrease in the activity of p70 S6 kinase
`in the cell. This method may be used to treat an animal
`20 having a condition characterized by proliferation of a
`cell in response to the given mitogen. The term mitogen
`is meant to include any entity which, when contacted with
`a cell, stimulates the cell to proliferate at a rate
`higher than the rate in its absence. Examples of such
`25 mitogens include lectins and growth factors (defined as
`naturally-occurring proteins or glycoproteins which acts
`as endogenous mitogens in vivo) such as IL-1, IL~3, IL-4,
`IL-5, IL-6, IL-7, IL-8, IL-9, IL-10, IL-11, Steel factor,
`G-CSF, M-CSF, GM-CSF, EPO, epidermal growth factor (EGF),
`30 fibroblast growth factor (FGF), platelet-derived growth
`factor (PDGF), and insulin.
`Also within the invention is an analog of
`rapamycin or of the rapamycin/FKBP complex, which analog,
`when introduced into a cell, modulates the activity of a
`35 serine/threonine kinase or serine/threonine phosphatase
`
`

`

`WO 93/19752
`
`PCT /US93/02459
`
`-
`
`8
`
`of the p70 S6 kinase cascade, such that the activity of
`p70 S6 kinase in the cell is lower in the presence of the
`analog than in its absence. Such analogs may be designed
`to mimic the molecular structure of rapamycin by using
`5 the information known about rapamycin, FK506, and the
`FKBP-binding analog known as 506BD (shown in Fig. 9),
`which does not function as an immunosuppressant.
`Such
`analogs would retain the common FKBP-binding site of all
`three molecules, and would have changes to some portion
`10 of the remainder of the molecule. The design and
`preparation of such analogs is well within the abilities
`of one of ordinary skill in the art of synthetic organic
`chemistry. Given the simple and rapid assays provided
`herein for determining whether a given analog functions
`15 in the same manner as rapamycin, the rapid screening of
`large numbers of such analogs is now feasible.
`Other features and advantages of the invention
`will be apparent from the following detailed description,
`and from the claims.
`
`20
`
`Detailed Description
`The drawings are first described.
`Drawings
`Fig. 1 is a set of SDS-PAGE gels illustrating the
`effect of IL-2 and PMA on S6 kinase activity, MAP kinase
`25 activity, and tyrosine phosphorylation in CTLL-20 cells.
`Cells were treated with IL-2 or PMA for the indicated
`times. Lysates were analyzed for (A) total S6 kinase
`activity, (B) S6 kinase activity due to p90 S6 kinase,
`(C) MAP kinase activity, or (D)
`induc~ion of tyrosine
`30 phosphorylation. For the in vitro kinase assays (A-C),
`the positions of the corresponding substrates are
`indicated on the left. For the antiphosphotyrosine
`immunoblot (D), the positions of molecular weight markers
`(in kDa) .are indicated on the left.
`
`

`

`WO93/19752
`
`PCT /US93/02459
`
`-
`
`9
`
`-
`
`10
`
`15
`
`Fig. 2 is a graph and a set of immunoblot
`autoradiograms illustrating the chromatographic
`resolution of p70 and p90 S6 kinases.
`IL-2-deprived
`cells were either left untreated or treated with
`5 rapamycin for 60 min, and either left unstimulated or
`stimulated with IL-2 for an additional period of 60 min.
`Cell lysates were applied to a Mono Q anion-exchange
`column and eluted with a linear gradient of NaCl from o.o
`to 0.5 M, and fractions were collected and analyzed.
`(A) S6 kinase activity. Open circles: untreated,
`unstimulated.
`·closed triangles: untreated, stimulated
`with IL-2. Closed circles:
`treated with rapamycin,
`stimulated with IL-2.
`(B) Immunoblotting with p70 S6 kinase-specific antiserum.
`(C) Immunoblotting with p90 S6 kinase-specific antiserum.
`Treatments and stimulations are indicated on the left,
`fraction numbers are indicated at the bottom, and the
`positions of molecular weight markers (in kDa) are
`indicated on the right. The positions of p70 and p90 S6
`20 kinases are indicated with arrows on the right. The 90
`kDa proteins detected by the anti-p70 S6 kinase antiserum
`(fractions 14-15) correspond to the recently described
`high molecular weight forms of the p70 S6 kinases (Grove
`et al., Mel. Cell. Biol. 11:5541-5550, 1991). Note that
`25 the protein bands migrating at approximately 100-110 kDa
`which are recognized by the p70 S6 kinase-specific
`antiserum (fractions 9-10 and 19-21) elute in fractions
`devoid of detectable S6 kinase activity.
`Fig. 3 is a set of SOS-PAGE gels illustrating the
`30 effects of rapamycin and FK506 on S6 ~inase activity, MAP
`kinase activity, and tyrosine phosphorylation in CTLL-20
`cells. Cells were preincubated with rapamycin or FK506
`as indicated and either left untreated for 5 min (lanes
`1-3), treated with IL-2 for 5 min (lanes 4-6) or 60 min
`(lanes 7-9), or treated with PMA for 5 min (lanes 10-12).
`
`35
`
`

`

`WO93/19752
`
`PCT /US93/02459
`
`- 10 -
`
`Lysates were analyzed for (A) total S6 kinase activity,
`(B) S6 kinase activity due to p90 S6 kinase, (C) MAP
`kinase activity, or (D) induction of tyrosine
`phosphorylation. For the in vitro kinase assays (A-C),
`5 the positions of the corresponding substrates are
`indicated on the left. For the antiphosphotyrosine
`immunoblot (D), the positions of molecular weight markers
`(in kOa) are indicated on the left.
`Fig. 4A is a set of SOS-PAGE gels illustrating the
`10 rapamycin dose-response of S6 kinase activity in H4
`cells. Serum-starved H4 cells were treated with the
`indicated concentrations of rapamycin for 1 hr at 37°C,
`5% co2 . Cells were then treated with insulin (10- 6 M,
`30 min) or were left untreated. Cells were harvested
`15 into homogenization buffer (10 mM KPi, pH 6.5/1 mM
`EDTA/SmM EGTA/l0mM MgC12/2 mM DTT/ 1 mM V04/50 mM ~(cid:173)
`glycerophosphate/0.1% Triton X-100/2 mM leupeptin/2mM
`pepstatin/0.2 mM PMSF); balanced for protein; and assayed
`for S6 kinase activity according to Nemenoff (Arch.
`20 Biochem. Biophys. 245: 196-203, 1986).
`Fig. 4B is an SOS-PAGE gel illustrating the in
`vivo incorporation of 32Pi into ribosomal protein S6. H4
`hepatoma cells (24-hr serum-starved) were washed with
`phosphate-free, serum-free DMEM, then incubated with 0.5
`25 mCi 32Pi/plate in DMEM for 1 hr. Rapamycin was then added
`at the indicated final concentrations (in quadruplicate).
`After 1 hr of rapamycin incubation, one half of the
`plates were incubated for 30 min with 150 mU/ml insulin
`(10-6 M), and the other half left untreated. Cells were
`30 then harvested in 0.5 ml extraction buffer (10 mM KPi, pH
`6.5/1 mM EDTA/5mM EGTA/10 mM MgC12/2 mM DTT/1 mM V04/
`50 mM ~-glycerophosphate/2 mM leupeptin/2 mM pepstatin/
`10 U/ml aprotinin/0.2 mM PMSF). Cells were broken in a
`dounce homogenizer and centrifuged 10 min at 1000 x g.
`35 Supernatants were then balanced for protein (BioRad), and
`
`.
`
`

`

`WO 93/19752
`
`PCT /US93/02459
`
`- 11 -
`
`a an aliquot was centrifuged. The pellet was resuspended
`in 200 ml of extraction buffer including 0.1% Triton x-
`100, and then treated with SDS sample buffer and run on a
`15% polyacrylamide gel (sample balanced for BioRad
`5 protein).
`Fig. 5 is a set of graphs depicting the results of
`Mono Q chromatography of H4 hepatoma extracts. H4
`hepatoma cells, serum-starved as in Fig. 4, were treated
`with 10-6 M insulin for 30 min (left panels), or 10-6 M
`10 insulin (30 min) preceded by a 1 hr incubation with 20 nM
`rapamycin (right panels). Cells were extracted in
`homogenization buffer and centrifuged as in Fig. 4.
`Extracts were diluted 3-fold in chromatography buffer (50
`mM ~-glycerophosphate, pH 7.2/lmM DTT/lmM EGTA/0.lmM
`15 vanadate) and applied to a Mono Q HR (5/5) column
`equilibrated with chromatography buffer. The column was
`eluted with a 90 ml gradient (0-0.4 M NaCl) in
`chromatography buffer with collection of 1 ml fractions.
`Individual fractions were assayed for kinase activities
`20 using 40S ribosomes and SKAIPS peptide. Fractions were
`also assayed for total protein and conductivity.
`Fig. 6 is a set of SDS-PAGE gels illustrating the
`time course of insulin-stimulated S6 kinase activity in
`the presence and absence of rapamycin. Serum-starved H4
`25 hepatoma cells were incubated in the presence (right) or
`the absence (left) of 20 nM rapamycin for 1 hr at 37°C,
`5% CO2 , after which cells were harvested and extracts
`prepared. Top panel: Extracts were assayed for total S6
`kinase activity using 40S ribosomes as substrates for
`30 phosphorylation. Middle panel: Extracts were
`imm_µnoprecipitated with an affinity-purified polyclonal
`p70 S6 kinase antibody (1 µg/ tube) in the presence of 10
`µl protein A Sepharose beads. After a 3 hr incubation at
`5°C, immunoprecipitated material was washed 3X in
`35 homogenization buffer and 0.25 M NaCl+ 0.1% Triton x-
`
`'>
`
`

`

`WO93/19752
`
`PCT /US93/02459
`
`- 12 -
`
`100, followed by one wash in 20 mM Tris HCL pH7.4/lmM
`EGTA/2mM EDTA/2mM DTT/lOmM ~-glycerophosphate/0.1% Triton
`X-100/10% glycerol.
`Immunoprecipitates were then assayed
`for S6 kinase activity using exogenous 40S ribosomes as
`5 substrate. Lower panel: Extracts were
`immunoprecipitated with an affinity-purified polyclonal
`p90 S6 kinase antibody (1 µg per tube) as in the middle
`panel, followed by a determination of S6 kinase activity.
`
`Fig. 7A is a pair of graphs comparing the effects
`10 of rapamycin on p70 S6 kinase, p90 S6 kinase, and
`erkl/MBP kinase. cos cells were transfected with
`expression constructs encoding the rat p70 S6 kinase
`(Grove et al., Mal. Cell. Biol. 11:5541-5550, 1991), the
`rat p90 S6 kinase or the rat p44 erkl/MBP kinase, all of
`15 which had been tagged at the amino terminus with a
`nonapeptide epitope from influenza hemagglutinin (Field
`et al., Mol. Cell. Biol. 8:2159-2165, 1988}. Two days
`following transfection, the cells were treated with the
`indicated concentrations of rapamycin for 60 min and with
`20 100 nM phorbol 12-myristate 13-acetate (PMA) for the
`final 15 min. Cells were lysed with homogenization
`buffer [as in Fig. 4a, but containing 0.1% (w/v) Triton
`X-100], and the extract was clarified by centrifugation
`at 100,000 x g. Recombinant protein was
`immunoprecipitated with the anti-epitope antibody 12CA5
`(immobilized on Sepharose beads) for 3 hat 4°C; pellets
`were washed (3 x 1 ml) in assay buffer [homogenization
`buffer without the MgC12 and with 10% (w/v) glycerol].
`S6 kinase activity was determined as above and p44
`30 erkl/MBP kinase was assayed similarly, using 0.5 mg/ml
`myelin basic protein (MBP) as substrate. Results are
`expressed as the mean pmol of substrate labeled during
`the 15 min assay; error bars indicate standard deviation
`of replicate determinations (upper panel, n=5; lower
`
`25
`
`

`

`WO 93/19752
`
`PCT /US93/02459
`
`- 13 -
`
`panel, n=6). The insert shows a Western blot of the
`recombinant p70 protein detected with the 12CA5
`monoclonal antibody and visualized by chemiluminescence
`(ECL, Amersham), demonstrating that recovery in each of
`5 the immunoprecipitants was equivalent.
`Fig. 7B is a bar graph which presents data
`indicating that rapamycin but not FK506 inhibits
`cos cells were transfected
`recombinant p70 S6 kinase.
`with vector or a p70 S6 kinase expression construct, as
`10 indicated; treated with 100 nM PMA and with 2 nM
`rapamycin or 200 nM FK506 (as indicated); and extracted,
`immunoprecipitated, and assayed as in 7A. The insert
`shows a Western blot of the recombinant p70 protein
`detected with the 12CA5 monoclonal antibody and
`15 visualized by chemiluminescence, demonstrating that
`recovery in each of the immunoprecipitants was
`equivalent.
`Fig. 7C is a graph, an autoradiogram of an SDS(cid:173)
`PAGE gel, and a Western blot illustrating the rapamycin
`20 dose response of p70 S6 kinase with respect to
`autophosphorylation of p70 S6 kinase and phosphorylation
`of 40S ribosomal subunits. COS cells were transfected,
`treated with rapamycin, extracted, and immunoprecipitated
`as in 7A. Upper panel: 40S kinase activity was
`25 determined from replicate sampling of duplicate
`immunoprecipitations (mean and standard deviation, n=6).
`Middle panel: Autophosphorylation was determined from
`kinase assays lacking substrate using aliquots of the
`immunoprecipitations from cells transfected with vector
`30 or recombinant p70 S6 kinase (as indiGated) and treated
`as indicated with no addition or with rapamycin at the
`following concentrations: 0.03 nM, 0.1 nM, 0.3 nM, 1 nM,
`or 3 nM. Lower panel: Western blot of samples in the
`same order as in the middle panel. Recombinant p70
`35 protein w.as detected with the 12CA5 monoclonal antibody
`
`

`

`WO 93/19752
`
`PCT/US93/02459
`
`- 14 -
`
`and visualized by chemiluminescence, demonstrating that
`recovery in each of the immunoprecipitants was
`equivalent.
`Fig. 8 is a pair of graphs illustrating the
`5 rapamycin-mediated inhibition of basal and insulin(cid:173)
`stimulated proliferation of H4 cells. H4 hepatoma cells
`were grown to confluence in Swims S77 medium (Sigma) with
`15% horse serum and 5% fetal calf serum. Confluent cells
`were maintained on serum-free medium 18 hours prior to
`10 assay. Serum-starved (24 hr) rat H4 hepatoma cells, 4 x
`104 cells/well, were cultured at 37°C, 5% CO2 in the
`absence or presence of 10-6 M insulin in the absence or
`presence of rapamycin at the indicated concentrations.
`Proliferation was assessed by the incorporation of [ 3H]-
`15 thymidine in a 16 hr pulse following a 32 hr incubation.
`This experiment is representative of four similar
`experiments.
`Fig. 9 is an illustration of the chemical and 3-
`dimensional structures of FK506, rapamycin, and 506BD
`(Bierer et al., Science 250:556-559).
`
`20
`
`Inhibition of IL2 stimulation of p70 S6
`
`Example 1:
`kinase
`MATERIALS AND METHODS
`Cell culture and stimulation of cells. The IL-2-
`25 dependent murine cell line CTLL-20 (American Type Culture
`Collection, Rockville, MD) was cultured in RPMI-10%FCS
`medium as described (Calvo et al., Eur. J. Immunol.
`22:457-462, 1992, herein incorporated by reference),
`containing human recombinant IL-2 (12~5-25 units/ml,
`30 kindly donated by Hoffmann-LaRoche, Inc.). Cells were
`recovered by centrifugation, washed 3 times with RPMI
`1640, resuspended in RPMI-10%FCS at 1-5 x 106 cells/ml
`and incubated for 3 hat 37° c. Cells were left
`untreated or treated with 100 nM rapamycin, 100 nM FK506
`
`

`

`WO 93/19752
`
`PCT /US93/02459
`
`- 15 -
`
`or equivalent ethanol diluent (final concentration 0.1%)
`for the last hour and during the subsequent stimulation
`period. Aliquots of 4-10 x 106 CTLL-20 cells were either
`left untreated or incubated in the presence of human
`5 recombinant IL-2 (25 units/ml) or phorbol-12-myristate-13
`acetate (PMA, 50 ng/ml, Sigma) for the indicated times at
`In vitro
`37°C, and then divided into two aliquots.
`In vitro kinase assays were performed
`kinase assays.
`essentially as
`described (Calvo et al., supra). 1-2 x
`10 106 cells were
`spun after stimulation and lysed in 0.2 ml
`of 10 mM potassium phosphate, pH 7.05, 1 mM EDTA,
`0.5% Triton x-100, 5 mM EGTA, 10 mM MgCl2, 50 mM ~(cid:173)
`glycerophosphate, 1 mM sodium vanadate, 2 mM DTT, 40
`µg/ml PMSF, 10 µg/ml leupeptin, and 1 µg/ml pepstatin.
`15 After 30 min on ice, nuclei were pelleted by a 5-min
`microfuge centrifugation, and supernatants were used for
`kinase assays.
`For the detection of total S6 kinase activity,
`direct S6 phosphorylation assays were performed with 5 µl
`20 of supernatant in a total volume of 30 µl containing 20
`mM Tris-HCl pH 7.25, 10 mM MgC12 , BSA 10-100 µg/ml, 50 µM
`ATP with 5 µCi [y-32P]ATP, and 40S ribosomal subunits as
`substrate (0.1-0.25 mg/ml) prepared from Xenopus laevis
`as described (Erikson et al., J. Biol. Chem. 261:350-355,
`25 1986). For S6 kinase assays specific for p90 S6 kinase,
`100 µl of supernatant were incubated with 5 µl of the
`specific rabbit antiserum 125 (Sweet et al., Mel. Cell.
`Biol. 10:2787-2792, 1990), and immunocomplexes were
`adsorbed to staphylococcus aureus, washed as previously
`30 described (Vik et al., Proc. Natl. Acad. Sci USA 87: 2685-
`2689, 1990), and subjected to S6 phosphorylation assays
`as described above.
`To assay for MAP kinase activity, 5 µl of
`supernatant were used in a total reaction volume of 30 µl
`35 containing 20 mM Tris-HCl pH 7. 25, 10 mM MgCl2, 100 µM
`
`

`

`WO93/19752
`
`PCT /US93/02459
`
`15
`
`- 16 -
`ATP with 5 ~Ci [y-32PJATP, and -1 ~g of unactivated
`Xenopus p90 S6 kinase (referred to as rsk) obtained as
`described (Vik et al., supra).
`In all cases kinase
`reactions were carried out at 30°C for 15 min and
`5 analyzed as previously described (Calvo et al., supra).
`Antiphosphotyrosine immunoblotting analysis. 3-8
`x 106 cells were washed, lysed, and centrifuged as
`described (Calvo et al., supra). The protein
`concentration in the supernatants was determined by a
`10 colorimetric method (Bradford, Anal. Biochem. 72:248-254,
`1976). Equivalent protein amounts of each supernatant
`were resolved by reducing 8-10% SOS-PAGE and analyzed by
`immunoblotting with the antiphosphotyrosine mAb 4G10 and
`anti-mouse IgG alkaline phosphatase-conjugated antibody
`(Promega) as described (Calvo et al., supra).
`Ion-exchange chromatography and anti-p70/p90 S6
`kinase immunoblotting analysis. 1.4 x 108 cells were
`lysed in homogenization buffer (5 mM EGTA, 1 mM EDTA, 10
`mM MgC12 , 50 mM fi-glycerophosphate, 2 mM DTT, 0.2% Triton
`20 x-100, 10% glycerol, 1 mM PMSF, 10 ~g/ml leupeptin, and
`10 ~g/ml pepstatin) and centrifuged at 100,000 x g for
`30 min at 4°C.
`supernatants, normalized for total
`protein content, were applied to a Mono Q (Pharmacia)
`anion-exchange chromato

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