throbber
[CANCER RESEARCH 55, 1982-1988, May 1, 1995]
`
`Rapamycin Enhances Apoptosis and Increases Sensitivity to Cisplatin in Vitro1
`
`Yufang Shi, Andrea Frankel, Laszlo G. Radvanyi, Linda Z. Penn, Richard G. Miller, and Gordon B. Mills2
`
`Oncology Research, The Toronto Hospital, Toronto M5G 2C4, Canada ¡Y.S., A. F., G. B. M.], Ontario Cancer institute. Princess Margaret Hospital, Toronto M4X 1K9, Canada
`¡L.G. R., R. G. M.]. and Department of Microbiology,
`Immunology and Cancer, The Hospital
`for Sick Children, Toronto M5G 1X8, Canada [L Z. P.]
`
`ABSTRACT
`
`systems by the
`can be regulated in a number of different
`Apoptosis
`actions of cytokines. Rapamycin
`has been shown to exert
`its effects on
`growth factor-induced
`cell proliferation,
`at least in part, by blocking the
`activation
`of
`the p70 S6 kinase and thus preventing
`the downstream
`signaling process, such as the activation of the members of the cdk family.
`To determine whether
`this pathway plays a role in the regulation of
`apoptosis, we assessed the effect of rapamycin on apoptosis
`induced by
`interleukin 2 deprivation in murine T-cell
`lines, by T-cell receptor ligation
`in a murine T-cell hybridoma, by enforced i--//m- expression in murine
`fibroblasts,
`and by corticosteroids
`in murine T-lymphoma
`cell
`lines. Al
`though rapamycin did not induce apoptosis on its own, rapamycin aug
`mented apoptosis
`in each of the cell
`lines used as indicated by increased
`genomic DNA fragmentation,
`decreased cell viability, and characteristic
`apoptotic
`changes
`in morphology. These
`results
`suggest
`that a signal
`transduction pathway(s)
`inhibited by rapamycin plays an important
`role
`in the susceptibility
`of cells to apoptosis. Many chemotherapeutic
`agents
`kill cancer cells through the induction of apoptosis. Strikingly,
`rapamycin
`increased the ability of the alkylating agent, cisplatin,
`to induce apoptosis
`in the human promyelocytic
`leukemia
`cell
`line HI,-60 and the human
`ovarian cancer cell
`line SKOV3. These data suggest
`that a signal
`trans
`duction pathway,
`likely related to p70 S6 kinase,
`inhibited by rapamycin
`may be an important component of the pathway which prevents cell death
`in many cell lineages and also indicate that rapamycin has the potential
`to
`augment
`the efficacy of selected anticancer
`therapies.
`
`INTRODUCTION
`The discovery of the immunosuppressive
`
`drugs CsA,3 FK506, and
`
`rapamycin has revolutionized organ transplantation and the treatment
`of autoimmune
`diseases
`(1). These immunosuppressants
`exert
`their
`effects by binding to a class of intracellular proteins called immu-
`nophilins, specifically interfering with the signaling pathways leading
`to cytokine production or proliferation of T lymphocytes upon acti
`vation (2). CsA binds to an immunophilin called cyclophilin A, while
`FK-506 and rapamycin bind to FKBP (2). Both cyclophilin and FKBP
`are peptidyl-prolyl
`cis-trans
`isomerases,
`the enzymatic
`activity of
`which is inhibited by the immunosuppressants. However,
`inhibition of
`the peptidyl-prolyl
`cis-trans
`isomerase does not account for the dem
`onstrated immunosuppressive
`activity (3). The complex of cyclophilin
`A and cyclosporin A, as well as the complex of FK506 and FKBP,
`binds to and inactivates calcineurin, an intracellular calcium/calmod-
`ulin-activated protein phosphatase (2); while the complex of rapamy
`cin and FKBP exerts its effect, at least
`in part, on the p70 S6 kinase
`pathway (4). Studies of the mechanisms by which these immunosup-
`
`Received 11/4/94; accepted 3/1/95.
`The costs of publication of this article were defrayed in part by the payment of page
`charges. This article must
`therefore be hereby marked advertisement
`in accordance with
`18 U.S.C. Section 1734 solely to indicate this fact.
`' This project was supported by grants from the Medical Research Council of Canada
`and the National Cancer Institute of Canada (to G. B. M.). G. B. M. is a Medical Research
`Council of Canada Scientist, Y. S.
`is a recipient of a postdoctoral
`fellowship of the
`National Cancer
`Institute of Canada, and L. G. R. was supported in part by an Ontario
`Graduate Studentship.
`2 To whom requests for reprints should be addressed, at Section of Molecular Thera
`peutics, M. D. Anderson Cancer Center, University of Texas, C5.001, 1515 Holcombe
`Boulevard, Houston, TX 77030.
`binding
`3 The abbreviations used are: CsA, cyclosporin A; FKBP, FK-506/rapamycin
`protein;
`IL-2, interleukin 2; MTT, 3-(4, 5-dimethylthiazol-2-yl)-2,5-diphenyl
`tetrazolium
`bromide; 125IUDR, 125I-labeled deoxyuridine.
`
`pressive drugs act have not only demonstrated how they function in
`cells but also have proved that
`they are useful
`tools to dissect cell
`signaling pathways
`(5).
`Unlike cyclosporin A and FK506, which suppress a calcium-de
`pendent pathway in the early stages of T-cell activation,
`rapamycin
`does not alter
`the early events
`following the activation of T cells
`through the T-cell antigen receptor, although it binds to and competes
`with FK506 for
`the same protein, FKBP (6).
`Instead,
`rapamycin
`inhibits signal
`transduction from the IL-2, epidermal growth factor,
`and other cytokine receptors,
`thus blocking the G¡to S phase transi
`tion required for cell cycle progression (6). In addition,
`rapamycin
`also inhibits the proliferation of 3T3 cells (7) and the hepatoma cell
`line H4 (8), presumably by blocking the effects of the growth factor-
`like activity of serum. The signal
`transduction pathway inhibited by
`the rapamycin-FKBP complex is not completely understood. Regard
`less of the mechanism,
`rapamycin blocks the activation of p70 S6
`kinase by diverse agents.
`It has also been shown that
`rapamycin
`blocks the activation of p34cdc2 kinase in T cells and in the myogenic
`cell
`line BC3H1 (9), presumably because p34cdc2 is a downstream
`target of p70 S6 kinase. This, combined with the evidence
`that
`p34cdc2 deregulation is an obligatory component of the induction of
`apoptosis by natural killers in target cells (10), suggests that rapamy
`cin could potentially alter the induction of apoptosis.
`is an active sui
`Apoptosis,
`a physiologically
`programmed event,
`cide process
`requiring energy-dependent
`participation
`of the dying
`cells (11). Apoptosis can be induced in T cells by activation, or in
`growth factor- and hormone-dependent
`cells by deprivation
`of the
`dependent
`factors, or in malignant cells by chemotherapeutic
`agents.
`Cyclosporin A and FK-506 have been shown to block activation-
`induced apoptosis in T-cell
`lines (12), while rapamycin does not (13).
`Herein, we report
`that rapamycin augments apoptosis in a number of
`different
`systems. This effect can be demonstrated
`in the murine
`T-cell
`line CTLL-2 induced by IL-2 withdrawal,
`in T-cell hybridomas
`induced by activation through the T-cell antigen receptor,
`in murine
`S49 cells treated with steroids, and in wye-transformed RAT-1 fibro
`blasts induced by culturing under
`low serum conditions. Strikingly,
`rapamycin also promotes apoptosis
`in the human promyelocytic
`line
`HL-60 cells and the human ovarian cancer SKOV3 cells induced
`by the chemotherapeutic
`drug, cisplatin,
`indicating potential clinical
`applications.
`
`MATERIALS AND METHODS
`
`Reagents. MTT and dexamethasone were purchased from Sigma Chemical
`Co. (St. Louis, MO). Rapamycin was provided by the National Cancer
`Insti
`tute, NIH (Bethesda, MD). Recombinant
`human IL-2 was obtained from the
`former Cetus Corp. (La Jolla, CA). Cisplatin was from David Bull Laboratories
`Pty Ltd. (Mulgrave, Victoria, Australia). FK520 was a gift of Dr. N. H. Sigal
`(Merck, Sharp and Dohme Research Laboratories, Rahway, NJ). Unless oth
`erwise indicated, all other chemicals were the purest grade available and were
`obtained from Sigma.
`Cell Lines. The T-cell hybridoma ALI was a gift of Dr. B. Singh (Uni
`versity of Western Ontario, London, Ontario, Canada; Ref. 14). They were
`recloned
`and selected
`for
`responsiveness
`to activation
`stimuli
`and T-cell
`receptor expression. A hamster anti-murine CD3 B cell hybridoma, 145-2cll
`(15), was used as sources of antibodies
`to the T-cell
`receptor complex. The
`IL-2-dependent mouse T cell
`line, CTLL-2, human promyelocytic
`leukemia
`
`1982
`
`Breckenridge Exhibit 1033
`Shi
`Page 001
`
`

`

`RAPAMYCIN ENHANCES APOPTOSIS
`
`line SKOV3 were obtained
`line HL-60, and human ovarian cancer cell
`cell
`from American Type Culture Collection (Rockville, MD). The mouse lym-
`phoma, S49, was kindly provided
`by Dr. G. T. Williams
`(University
`of
`Birmingham, Birmingham, United Kingdom). Rat-1 cells constitutively
`ex
`pressing c-myc under
`the control of the muLV retroviral promoter have been
`described
`previously
`(16). All cells were cultured
`at 37°C in humidified
`
`containing 5% CO2 in RPMI 1640 (GIBCO Laboratories, Grand
`atmosphere
`Island, NY) supplemented with 2 HIM L-glutamine,
`10 mM HEPES, 50 p,M
`2-mercaptoethanol,
`5-10% heat-inactivated
`fetal bovine serum (Sigma), and
`10 (AMgentamicin (GIBCO).
`Apoptosis
`Induction. The T-cell hybridoma Al.l was activated by anti-
`CD3 coated on tissue culture plastic by incubating with 0.05 M Tris-HCl
`(pH
`9.0) overnight
`at 4°Cor for
`l h at 37°C.Plates were washed with PBS to
`
`cells were added. Cells were harvested
`remove unbound antibody before Al.l
`for DNA fragmentation
`analysis after a 12-h incubation at 37°Cin humidified
`5% CO2. Apoptosis
`in CTLL-2 cells was induced by IL-2 starvation,
`in S49
`cells by dexamethasone
`treatment,
`in Rat-1-m^c cells by low serum, and in
`HL-60 cells and SKOV3 cells by cisplatin treatment as indicated.
`Genomic DNA Fragmentation Assay. Cells (4-6 X IO5) were harvested
`
`tube in 30 p.1 PBS and lysed with 30 fil of
`in an Eppendorf
`and resuspended
`lysis buffer
`[80 mM EDTA, 200 mM Tris (pH 8.0), 1.6% (w/v) sodium lauryl
`sarcosinate,
`and 5 mg proteinase K/ml]. The lysate was mixed and then
`incubated in a 50°Cwater bath for 1.5 h. After adding 0.2 mg/ml RNase A, the
`mixture was incubated in a 37°Cwater bath for an additional 30 min. The
`
`resulting DNA solution was analyzed on 1% agarose gels
`(10 mM Tris and 1 mM EDTA).
`by labeling
`Alternatively,
`genomic DNA fragmentation was quantitated
`actively dividing cells with 125IUDR at the concentration
`of IO6 cells/ml with
`1 fiCi/ml of 125IUDR at 37°Cfor 8-10 h. Labeled cells were harvested and
`
`in TAE buffer
`
`300000-1
`
`200000-
`
`100000-
`
`O ^^
`Q. ÃœJ
`
`5»
`
`O -H
`
`2 _-il>0
`
`[Rapamycln]
`
`ng ml
`
`cell proliferation. CTLL-2 cells were
`Fig. 1. Effect of rapamycin on IL-2-induced
`plated in 96-well plates at 1 X 10" cells/well with different concentrations
`of rapamycin
`and recombinant human IL-2. After 20 h, l ^tCi of [3H]thymidine was added to each well
`for 3 h. Cell proliferation was determined by measuring [3H]thymidine
`incorporation by
`scintillation counting. Results represent
`the mean of six wells; bars, SE.
`
`washed with cold media at least three times. Treatments were then carried out
`in 200 fj.1media in 96-well
`tissue culture plates; genomic DNA fragmentation
`was then assayed as follows. Cells were harvested in 1.5-ml Eppendorf
`tubes
`and lysed by adding 900 ju.1lysis buffer [5 mM Tris (pH 7.4), 2 mM EDTA, and
`0.5% Triton X-100 (nonionic detergent);
`total volume, 1.1 ml]. The Eppendorf
`tubes were then vortexed vigorously to ensure complete lysis of cells. After
`incubating on ice for 20 min, the tubes were centrifuged at 14,000 cpm for 20
`min in a microfuge. One ml of supernatant
`(containing fragmented DNA) was
`transferred to a new Eppendorf
`tube, leaving 100 ß\supernatant with the pellet
`to ensure that
`the pellets were not cotransferred.
`The radioactivity
`of
`the
`supernatant
`and the pellet were measured with a gamma counter. The percent
`age of fragmentation was calculated by:
`
`% DNA fragmentation =
`
`Supernatant cpm X 1.1
`Supernatant cpm + pellet cpm
`
`x 100
`
`MTT Staining. Cell viability was assessed essentially as described by
`Mosmann (17). Briefly, cells were incubated in 100 fil media in 96-well plates
`with additions
`as indicated. Following
`incubation,
`10 /nl of MTT solution
`(5 mg MTT/ml
`in H2O) were added and incubated at 37°Cfor 4 h. One
`hundred (¿1of acid-isopropanol
`(0.04 N HC1 in isopropanol) were added to each
`culture and mixed by pipetting or shaking on a plate shaker
`to dissolve the
`reduced MTT crystals;
`the relative cell viability was obtained by scanning with
`an ELISA reader with a 570-nm filter.
`Cell Proliferation Assay. Cells were incubated in 96-well plates with
`appropriate treatments. The proliferative
`response was determined by [3H]thy-
`midine incorporation in which 1 ^iCi of [3H]thymidine was added to each well
`and incubated for 3 h. Cells were then harvested onto glass-fiber
`filter paper,
`and the rate of [3H]thymidine
`uptake was quantitated
`by liquid scintillation
`
`counting.
`
`RESULTS
`
`reciprocal effect; the higher the concentration of IL-2 in the culture
`medium, the higher the concentration of rapamycin required to sup
`press the proliferation of CTLL-2 cells to background levels. (The
`effect of rapamycin on IL-2-induced proliferation was analyzed by
`two-way ANOVA, P < 0.0001.) This suggests that the concentration
`of IL-2 determines the quantity or quality of the transmembrane
`signals and that rapamycin is able to completely block IL-2-induced
`proliferation only in the presence of relatively low concentrations of
`IL-2.
`Since IL-2 induces proliferation in responsive cells, IL-2 must
`provide both mitogenic and cell survival signals upon the interaction
`with its receptor. It is conceivable that the effect of rapamycin on
`IL-2-induced proliferation of CTLL-2 cells might be due to the
`inhibition of cell survival signals rather than on the mitogenic signal
`induced by IL-2. We tested this hypothesis by using a model in which
`IL-2 deprivation induces apoptosis in CTLL-2 cells. As shown pre
`viously (19), 24 to 48 h after IL-2 withdrawal, the majority of CTLL-2
`cells undergo apoptosis. When rapamycin was added, there was a
`significant increase of apoptosis in CTLL-2 cells as indicated by
`genomic DNA fragmentation assessed on agarose gels (Fig. 2) and by
`release of label from 125IUDR-labeled cells (data not presented).
`However, this effect was only apparent when limiting concentrations
`of IL-2 were present. Indeed, in the presence of IL-2 (10 units/ml),
`rapamycin did not induce apoptosis at any concentration tested (Fig.
`2 and data not shown). This is apparently discordant to Fig. 1, in
`which cell proliferation induced by as much as 20 units/ml of IL-2 can
`still be decreased by high concentrations of rapamycin. The most
`likely explanation for this discrepancy is that apoptosis only occurs
`when IL-2 signals are decreased below a critical
`threshold. The
`immunosuppressant FK-520, an analogue of FK506 which competes
`with rapamycin for FKBP, did not promote apoptosis, but rather
`reversed the effect of rapamycin when FK520 was present at a 20-fold
`excess (Fig. 3),
`indicating that the apoptosis-promoting effect of
`rapamycin depends on the binding of rapamycin to FKBP.
`The distinction between cell survival and cell proliferation signals
`has been well established by genetic complementation of bcl-2 and
`The immunosuppressive effect of rapamycin is not through its
`myc (20, 21). The proto-oncogene c-myc, which plays an important
`effect on the signals from the T-cell antigen receptor but rather
`through the effects on the signals induced by IL-2 produced after
`role in cell proliferation and transformation, is also required for the
`T-cell activation (18). We tested the effect of rapamycin on IL-2-
`induction of apoptosis in some cell lineages (22, 23). When cells
`induced proliferation of a murine T-cell line, CTLL-2, by varying the
`constitutively expressing myc are cultured under reduced serum con
`concentrations of both IL-2 and rapamycin. As indicated by [3H]thy-
`ditions, they undergo apoptosis (23). In this case, apoptosis can be
`midine incorporation shown in Fig. 1, IL-2 and rapamycin showed a
`suppressed by cell survival signals provided by the constitutive
`1983
`
`Breckenridge Exhibit 1033
`Shi
`Page 002
`
`

`

`[rapamycin] ng/ml
`= 8 _
`08
`_
`
`RAPAMYCIN ENHANCES APOPTOSIS
`
`cally promoted anti-CD3-induced apoptosis in Al.l cells as indicated
`by genomic DNA fragmentation detected by agarose gel electrophore
`sis (Fig. 5, a and b) and by the release of 125IUDRfrom labeled cells
`(Fig. 5c). Rapamycin alone did not induce apoptosis in the T-cell
`hybridoma cells (Fig. 5).
`
`D Cl M 9
`a am a
`o «us rf
`u oÃ(cid:173)».ai
`
`O
`
`0.5
`[IL-2] lU/ml
`
`10
`
`I
`
`0.30
`
`[Rapa]
`
`ng/ml
`
`U ml
`
`genomic DNA fragmentation.
`IL-2 deprivation-induced
`Fig. 2. Rapamycin increases
`CTLL-2 cells (1 X Id6) were cultured without or with IL-2 at 0.5 or 10 units/ml, and
`rapamycin at (I, 1, 10, or 100 ng/ml. After 20 h, cells were harvested and lysed, and
`genomic DNA was extracted and analyzed by agarose gel electrophoresis
`(a). Densito-
`metric analysis of the fragmented DNA of the second hands in the agarose gel
`is also
`presented (h).
`
`expression of bcl-2 or the presence of high concentrations of serum
`(20, 23). In accordance with previous studies, when Rat-1 cells
`constitutively expressing c-mvc were cultured in media containing
`0.5% serum, there was a dramatic decrease in cell viability as indi
`cated by MTT reduction. This decrease in cell viability under low
`serum conditions was augmented by the addition of 10 ng/ml rapa
`mycin (Fig. 4). Thus, rapamycin increases programmed cell death
`induced by the constitutive expression of c-myc, likely by interfering
`with cell survival signals mediated by the low concentration of serum
`present in the assays (one-way ANOVA, P < 0.001). As with CTLL-2
`cells incubated with high concentrations of IL-2 (Figs. 1 and 2),
`rapamycin exerted little effect on apoptosis in RAT-1 cells in the
`presence of high concentrations of serum (P < 0.05).
`Activation-induced apoptosis in T-cell hybridoma cells has been
`used as a model system to explore the mechanism regulating negative
`selection during T-cell development in the thymus (24). Cyclosporin
`A and FK-506 can completely block activation-induced apoptosis in
`T-cell hybridoma A1.1 cells (12, 13) and other cells (data not shown).
`However, when the effect of rapamycin was tested, it was found that,
`unlike CsA or FK506, rapamycin does not block activation-induced
`apoptosis (13). We sought to determine whether rapamycin would
`promote apoptosis in this system following activation of ALI cells
`with varying concentrations of anti-CD3. At an optimal dose of
`anti-CD3 (2 ju.g/ml),rapamycin did not alter activation-induced ap
`optosis in A 1.1 cells, likely because the majority of the cells were
`committed to undergo apoptosis by anti-CD3 alone (25). However, at
`lower doses of anti-CD3 (0.5 /xg/ml or lower), rapamycin dramati
`
`1984
`
`«api . FK520
`
`on genomic DNA fragmentation
`rapamycin
`the effect of
`Fig. 3. FK520 reverses
`induced by IL-2 deprivation. CTLL-2 cells (1 x IO6) were cultured with or without
`rapamycin al 1 ng/ml
`in the presence or absence of FK520 at 20 ng/ml. Cells were
`harvested and assessed for genomic DNA fragmentation
`on an agarose gel following a
`20-h incubation (a). Densitometric analysis of the second bands in the gel is presented in b.
`
`rapamycin
`
`trapamycin
`
`0.5 i
`
`0.4-
`
`0.3-
`
`0.2-
`
`°U7
`«w
`
`f¡
`o S
`
`"*
`= 9
`
`z oit"
`s E
`at
`te
`
`0.5
`
`Serum Concentration
`
`(%)
`
`fibroblasts
`cell death in myr-transformed
`Fig. 4. Rapamycin enhances programmed
`induced by serum deprivation, mvc-transformed Rat-1 cells were cultured in RPMI
`supplemented with 5 or 0.5% fetal bovine serum for 50 h with or without rapamycin at 10
`ng/ml. The relative number of viable cells was determined by the reduction of MTT.
`
`Breckenridge Exhibit 1033
`Shi
`Page 003
`
`

`

`a
`
`.- et
`
`«
`+
`
`+
`
`+
`SÃ(cid:173)
`«
`,,¿..£333333
`S ä.3. Elv; in c; r»—-.
`
`RAPAMYCIN ENHANCES APOPTOSIS
`
`cells were stimulated with different
`cells. T-cell hybridoma Al.l
`in mouse T-cell hybridoma
`genomic DNA fragmentation
`activation-induced
`Fig. 5. Rapamycin augments
`concentrations
`of anti-CD3 (145-2cl 1) coated on tissue culture plastic with or without
`rapamycin at 10 ng/ml. Cells were harvested 12 h after culture and assessed for genomic DNA
`fragmentation by agarose gel electrophoresis
`(a). Densitometric analysis of the fragmented genomic DNA is presented in b. Al.l
`cells were also labeled with l25IUDRat 1 /xCi/1 X IO6
`cells/ml. After 10 h, cells were washed and cultured with 0.5 (J.g/ml anti-CD3 for 12 h. The percentage of genomic DNA fragmentation was assessed by the release of 12iIUDR-labeled
`fragmented DNA (c); bars, SE.
`
`*
`
`Rapa
`
`[Anli-CD3]
`
`Rapa
`
`Anti-COS
`
`AHII-CD3
`
`t Rapa
`
`is the
`in T cells and thymocytes
`apoptosis
`Corticosteroid-induced
`best characterized model system for studying programmed cell death
`(26). The corticosteroid-sensitive
`mouse lymphoma
`cell
`line S49
`undergoes
`characteristic
`apoptosis upon treatment with dexametha-
`sone. When 125IUDR-labeled S49 cells were incubated with a subop
`timal concentration
`of dexamethasone
`(10~7 M), rapamycin signifi
`
`(Fig. 6; one-way
`plus
`rapamycin;
`
`genomic DNA fragmentation
`increased
`cantly
`ANOVA;
`dexamethasone
`versus dexamethasone
`P < 0.001).
`reagents can induce
`It has been demonstrated that chemotherapeutic
`apoptosis
`in target cells (27). Rapamycin has already been tested
`clinically as an immunosuppressant
`in patients and is relatively non-
`toxic in short-term administration
`(28).
`If rapamycin can augment
`apoptosis
`induced by chemotherapeutic
`reagents,
`then the addition of
`rapamycin to chemotherapy
`protocols could potentially increase the
`efficacy of chemotherapy. Cisplatin, an effective chemotherapy agent,
`has been shown to induce apoptosis in a number of cell lineages (29).
`A 24-h incubation with cisplatin at 30 JAMor more induced DNA
`fragmentation in the human promyelocytic
`leukemia cell line HL-60,
`and as predicted,
`rapamycin augmented cisplatin-induced DNA frag
`mentation (Fig. la). The densitometric analysis of the gel is presented
`in Fig. Ib. However, as before, rapamycin did not augment
`the effect
`of cisplatin at doses which already induced maximal apoptosis
`(i.e.,
`100 fiM cisplatin). When HL-60 cells were incubated with cisplatin
`for 96 h, there was a dose-dependent
`reduction of cell viability, as
`determined
`by MTT reduction, which was detectable
`at 1 /XMand
`reached maximal effects at 5 to 10 /XM(Fig.
`lc; R2 = 0.995 from
`
`0.625 fiM to 5 JAM;note: the different concentrations of cisplatin in the
`experiments
`in Fig. 7, a and b, reflect
`the different periods of incu
`bation). We selected 2.5 /AM cisplatin as a suboptimal
`dose and
`determined
`the effect of
`rapamycin.
`In this
`system,
`rapamycin
`induced
`a dose-dependent
`augmentation
`of cisplatin-induced
`re
`duction of cell viability (Fig. Id;
`two-way ANOVA, P < 0.001 and
`R2 = 0.982 for
`rapamycin
`from 0.01 nM to 10 nM). In contrast,
`rapamycin
`alone did not
`significantly
`alter HL-60 viability
`at
`several concentrations
`tested (Fig. 7). The effect of rapamycin on
`1985
`
`of cisplatin was also
`doses
`by suboptimal
`induced
`cell death
`readily observed
`by changes
`in morphology
`at
`the microscopic
`level (Fig.
`le). Cisplatin at 2.5 LIMinduced morphological
`changes
`consistent with apoptosis
`in only a small proportion
`of cells;
`rapamycin alone had no effect. However, when cells were coincu-
`
`2.0-
`
`111
`
`COI*
`o cs s
`
`O s
`
`fi n
`0) â„¢
`Ü
`«I
`o I'
`
`Rapa
`
`Dex
`
`Dex + Rapa
`
`cells.
`in steroid-sensitive
`Fig. 6. Rapamycin increases genomic DNA fragmentation
`S49 cells were labeled with 125IUDR at 1 nCi/1 x 10" cells/ml. After 10 h, cells were
`washed and cultured with dexamethasone
`at 10~7 M for 36 h. Percentage
`of genomic
`DNA fragmentation
`was assessed
`by the release
`of
`'25IUDR-labeled
`fragmented
`DNA. Bars. SE.
`
`Breckenridge Exhibit 1033
`Shi
`Page 004
`
`

`

`RAPAMYCIN ENHANCES APOPTOSIS
`
`3
`
`S
`S
`(as T man 'ois -ero 'u.«)
`jo jaquiriN eAiiejau
`
`°.
`
`OS '
`
`w 'ois -Q'o 'iiw)
`to jaqiunN aAiieio
`
`I
`° I
`
`.= o
`
`i^Iïa
`
`; -o 5 5! .
`
`Lu -a
`= =
`
`i8|i
`
`t. PS p
`? fi
`il
`
`IIP
`
`o
`
`al[:
`o t
`>HI
`lili"
`
`•>^^_,e >*
`00«^
`
`E *
`
`*•*S
`
`ill'*'
`
`i
`
`tÃ(cid:173)•?'3 •=
`
`•**fclf~|
`
`S^
`B« o •~
`eie N
`ï* ï5 .s
`
`(as T ura«'oÃ(cid:173)s'Q'o 'um)
`suso
`8|qe!A |o jaquinN
`8A|)Bia|
`
`„5
`
`!i
`
`1986
`
`(as + UM«'OÃ(cid:173)s'Q'o 'iiw)
`SII30
`0|qt!IA
`10 JaqiunN
`0'AHE|r>a
`
`«¡DWT!oc
`BdB«+ so wTl OC
`SK>WTlOf
`«dim+ so WT! Ofr
`«OWTl OS
`
`BdBH+ «ov«TlOOI
`
`CO
`
`Breckenridge Exhibit 1033
`Shi
`Page 005
`
`

`

`RAPAMYCIN ENHANCES APOPTOSIS
`
`rapamycin,
`
`nearly all of
`
`bated with 2.5 JU.Mcisplatin and 1 ng/ml
`the cells underwent
`apoptosis.
`Cisplatin is currently the drug of choice for the treatment of ovarian
`cancer. Therefore, we examined the effect of rapamycin on the in
`duction of cell death in a relatively cisplatin-resistant
`ovarian cancer
`cell line, SKOV3. Since ovarian cancer cells frequently do not dem
`onstrate cisplatin-induced DNA ladders despite clearly undergoing
`apoptosis (30), we used cell viability as assessed by MTT reduction as
`a measure of apoptosis
`in these cells. As shown in Fig. If, cisplatin
`induced a dose-dependent
`reduction in cell viability of SKOV3 cells
`as determined by MTT reduction. The effect of cisplatin was detect
`able at 3 /xM and reached maximal effects at 25 to 50 /AM(R2 = 0.967
`
`from 1.56 JIM to 25 JU.M).We selected 5 /J.Mcisplatin as a suboptimal
`dose and studied the effect of rapamycin on cisplatin-induced
`cell
`death. We found that rapamycin induced a concentration-dependent
`enhancement
`of cisplatin-induced
`cell death (Fig. 7g;
`two-way
`ANO VA, P < 0.01 and R2 = 0.954 for rapamycin from 0.1 nM to 100
`
`could also enhance
`rapamycin
`HM). Thus,
`cisplatin in ovarian cancer cells.
`
`cell death induced by
`
`DISCUSSION
`
`and FK506, which inhibit activation-induced
`Unlike cyclosporin
`apoptosis
`in T cells (12, 13), rapamycin enhances
`the induction of
`apoptosis
`in a number of cell
`lineages mediated by a number of
`different mechanisms. Many cancer chemotherapeutic
`agents have
`been shown to exert their effects by inducing apoptosis in cancer cells.
`Our observation
`that
`low doses of
`rapamycin
`increase
`apoptosis
`induced by suboptimal
`doses of cisplatin,
`at
`least
`in vitro,
`in the
`human promyelocytic
`leukemia cell line HL-60 and the human ovar
`ian cancer cell line SKOV3 suggests a potential clinical application, as
`both drugs are currently in clinical use.
`transformation have led to
`Studies of the mechanisms of malignant
`the identification of genes that regulate cell proliferation,
`cell viabil
`ity, or both. Genes with the capacity to regulate cell viability include
`bcl-2, bcl-x, box, p53, retinoblastoma gene, raf, ras, ahi, Fas/APO-l,
`and c-myc (reviewed in Refs. 31 and 32). The hypothesis
`that cells
`require both proliferative
`and survival
`signals
`is best supported by
`genetic complementation
`experiments
`in which myc overexpression-
`induced cell death is suppressed by bcl-2 expression (20, 21). Our
`results demonstrating
`that
`rapamycin
`promotes
`serum deprivation-
`induced cell death in c-myc transfected RAT-1 cells suggest
`that
`rapamycin promotes cell death through the inhibition of cell survival
`signal(s).
`In IL-2-dependent
`cell lines, IL-2 provides both survival and
`proliferative signals. It has been shown that p70 S6 kinase is activated
`in CTLL-2 cells after
`IL-2 stimulation,
`and rapamycin specifically
`inhibits p70 S6 kinase activation (6). Therefore, p70 S6 kinase might
`be important
`in providing survival signals. Recent reports show that
`one of the downstream targets of p70 S6 kinase is p34cdc2 kinase,
`which has been shown to be important
`in the regulation of apoptosis
`in target cells by cytotoxic killing (10).
`It is currently used in the
`Cisplatin is a potent antitumor
`agent.
`treatment of many malignancies,
`including small cell lung, testicular,
`ovarian, head and neck, bladder, and esophageal
`cancers
`(33). Cis
`platin induces the formation of DNA adducts,
`including cross-links
`between DNA and protein or inter- and intrastrand cross-links in DNA
`(29). However,
`there is no correlation between the amount of cisplatin
`required to induce cell death and to inhibit DNA synthesis
`in DNA
`repair-deficient
`and DNA repair-proficient
`cells. Thus,
`the cytotoxic
`effect of cisplatin is not solely due to the inhibition of DNA synthesis
`or DNA damage.
`Instead,
`the ability of cisplatin to decrease cell
`viability was inhibited by cyclohexamide
`and was accompanied
`by
`genomic DNA fragmentation,
`characteristic of cells dying by apop
`
`the
`rapamycin enhances
`that
`results demonstrating
`tosis (33). Our
`cytotoxic effects of cisplatin suggests that intracellular
`signaling mol
`ecules inhibited by rapamycin,
`such as p70 S6 kinase, may play a
`generalized role in regulating apoptosis induced by chemotherapeutic
`agents.
`Although cisplatin has been widely used as an antitumor agent, high
`doses lead to severe multiorgan toxicities
`including kidney and bone
`marrow failure,
`intractable vomiting, peripheral neuropathy, deafness,
`seizures, and blindness, preventing dose intensification
`(34). Poten
`tially,
`rapamycin could reduce the dosage and augment
`antitumor
`activity of cisplatin without
`increasing toxic side effects. Furthermore,
`the amount of rapamycin required for this effect
`in vitro is only 1
`ng/ml, which is readily achieved in patients. For example,
`in renal
`transplant
`recipients,
`serum rapamycin concentrations
`as high as 10
`ng/ml have been achieved 72 h after administration without
`toxicity
`(35). Thus,
`if rapamycin potentiates
`the effects of cisplatin or other
`drugs on tumor cells without
`increasing multiorgan toxicity of the
`chemotherapeutic
`agents,
`the combination of rapamycin with conven
`tional chemotherapeutic
`agents may result
`in functional
`"dose inten
`sity," perhaps increasing survival
`rates.
`
`ACKNOWLEDGMENTS
`
`We are indebted to Mary Hill, Feng Wang, and Ajay Sharma for technical
`support.
`
`REFERENCES
`
`protein phosphatase
`
`action in cell signaling
`
`immunosuppressants meet protein phosphalases.
`
`1. Schreiber, S. L. Immunophilin-sensitive
`pathways. Cell, 70: 365-368,
`1992.
`2. McKeon, F. When worlds collide:
`Cell, 66: 823-826,
`1991.
`hit the target. Current Biol., 2: 18-20, 1992.
`3. Cyert, M. S. Immunosuppressants
`4. Kuo, C. J., Chung, J., Fiorentino, D. F., Flanagan, W. M., Blenis, J., and Crabtree,
`G. R. Rapamycin selectively inhibits interleukin-2 activated p70 S6 kinase. Nature
`(Lond.), 358: 70-73,
`1992.
`5. Siga), N. H., and Dumont, F. J. Cyclosporin A, FK-506, and rapamycin: pharmacological
`probes of lymphocyte signal transduction. Annu. Rev. Immunol., 10: 519-560,
`1992.
`6. Chung. J., Kuo, C. J., Crabtree, G. R., and Blenis, J. Rapamycin-FKBP
`specifically
`blocks growth-dependent
`activation of and the signaling by the 70 kd S6 protein
`kinases. Cell, 69: 1227-1236,
`1992.
`7. Jefferies, H. B, Reinhard, C., Kozma, S. C., and Thomas, G. Rapamycin selectively
`represses translation of the polypyrimidine
`tract mRNA family. Proc. Nati. Acad. Sci.
`USA, 91: 4441-4445,
`1994.
`8. Price, D. J., Grove, J. R., Calvo, V., Avruch, J., and Bierer, B. E. Rapamycin-induced
`inhibition of the 70-kilodalton S6 protein kinase. Science (Washington DC), 257:
`973-977,
`1992.
`induces
`blocks proliferation,
`9. Jayaraman, T., and Marks, A. R. Rapamycin-FKBP12
`differentiation,
`and inhibits cdc2 kinase activity in a myogenic
`cell
`line. J. Biol.
`Chem., 268: 25385-25388,
`1993.
`J., Bradbury, E. M., Litchfield, D. W., and Green-
`10. Shi, L., Nishioka, W. K., Th'ng,
`burg, A. H. Premature p34cd32 activation required for apoptosis. Science (Washing
`ton DC), 263: 1143-1145,
`1994.
`11. Wyllie, A. H., Kerr, J. F. R., and Currie, A. R. Cell death:
`apoptosis.
`Int. Rev. Cytol., 68: 251-305,
`1980.
`12. Shi, Y., Sahai, B. M., and Green, D. R.. Cyclosporin A inhibits activation-induced
`cell death in T-cell hybridomas
`and thymocytes. Nature (Lond.), 339: 625-626,
`1989.
`L. A., Burakoff,
`P. S., Slandaert, R. F., Herzenburg,
`13. Birer, B. E., Malilla,
`S. J., Crabtree, G., and Schreiber, S. L. Two distinct
`signal
`transmission
`pathways
`in T lymphocytes
`are inhibited by complexes
`formed between an immunophilin
`and either FK-506 or rapamycin.
`Proc. Nati. Acad. Sci. USA, 87: 9231-9235,
`1990.
`J., Fraga, E., and Singh, B. Fine
`14. Fotedar, A., Boyer, M., Smart, W., Widtman,
`specificity of antigen recognition by a T cell hybridoma clone specific for polylS:
`a
`synthetic polypcptide
`of defined sequence
`and conformation.
`J.
`Immunol.,
`135:
`3028-3033,
`1985.
`15. Leo, O., Foo, M., Sachs, D. H., Samelson, L. E., and Bluestone, J. A.. Identification
`of a monoclonal antibody specific for a murine T3 polypeptide. Proc. Nati. Acad. Sci.
`USA, 84: 1374, 1987.
`J. P.,
`16. Penn, L. J. Z, Brooks, M. W., Laufer, E. M., Littlewood, T. D., Morgenstern,
`Evan, G. I., Lee, W. M. F., and Land, H. Domains of human c-myc protein required
`for autosuppression
`and cooperation with ras oncogenes are overlapping. Mol. Cell.
`Biol., 10: 4961-4966,
`1990.
`assay for cellular growth and survival: application
`17. Mosmann, T. Rapid colorimetrie
`to proliferation and cytotoxicity assays. J. Immunol. Methods, 65: 55-63,
`1983.
`
`the significance
`
`of
`
`1987
`
`Breckenridge Exhibit 1033
`Shi
`Page 006
`
`

`

`RAPAMYCIN ENH

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