throbber
mTOR as a Target for Cancer Therapy
`
`P. J. Houghton- S. Huang
`Department of Molecular Pharmacology, St. Jude Children's Research Hospital,
`332 N. Lauderdale, Memphis, TN 38105-2794, USA
`E-mail: peter.houghton@stjude.org
`
`Introduction
`
`.
`
`2
`
`3
`
`Autocrine Growth of Rhabdomyosarcomas . . . . . . .
`
`Selective Tumor Growth Inhibition by Rapamycin .
`
`4 Mechanism of Action of Rapamycin . . . . . . . . .
`
`5
`
`6
`
`Rapamycin Induces Apoptosis in Rhabdomyosarcoma Cells .
`
`The Tumor Suppressor p53 Protects
`from Rapamycin -Induced Apoptosis . .. ..
`
`7 Mechanism(s) of Resistance to Rapamycin ..
`
`8
`
`9
`
`Preclinical Antitumor Activity for Rapamycins .
`
`Clinical Trials for Antitumor Efficacy of Rapamycin s .
`
`10 Conclusions
`
`.
`
`References.
`
`. . . . .
`
`. . . .
`
`340
`
`340
`
`341
`
`343
`
`343
`
`347
`
`347
`
`351
`
`352
`
`353
`
`354
`
`Abstract The target of rapamycin, mTOR, acts as a sensor for mitogenic
`stimuli, such as insulin-like growth factors and cellular nutritional sta(cid:173)
`tus, regulating cellular growth and division. As many tumors are driven
`by autocrine or paracrine growth through the type -I insulin-like growth
`factor receptor, mTOR is potentially an attractive target for molecular(cid:173)
`targeted treatment. Further, a rationale for anticipating tumor-selective
`activity based on transforming events frequently identified in malignant
`disease is becoming established.
`
`G. Thomas et al.(eds.), TOR Target of Rapamycin
`© Springer-Verlag Berlin Heidelberg, 2004
`
`Roxane Labs., Inc.
`Exhibit 1034
`Page 001
`
`

`
`P. ]. Houghton and S. Huang
`
`340
`
`1 I
`
`ntroduction
`
`Since the early 1960s the introduction of cytotoxic agents, thence their
`dose intensification (starting in the 1970s), has dramatically improved
`survival of children with hematologic and solid tumors. For example,
`data from 1960-1963 shows that overall survival for children under fif(cid:173)
`teen years old with a diagno sis of neuroblastoma, bone or joint sarco(cid:173)
`mas, or CNS tumors was 25%, 20%, and 35%, respectively. In contrast,
`for the period 1985-1994 the survival for these same groups had in(cid:173)
`creased to 63%-65%. However, for soft tissue sarcomas, the focus of this
`laboratory, increases in sur vival have been less impressive, increasing
`from 60% in 1974 to 71% (1985-1994)1. Although these result s demon(cid:173)
`strate clear progress in treating childhood malignancies, they do not re(cid:173)
`flect the morbidity and long-term sequelae often associated with inten(cid:173)
`sive use of cytotoxic agents. Consequently, almost two decade s ago we
`started to study pediatric soft tissue sarcomas with the ultimate goal of
`developing novel therapeutic approaches based on specific biological
`characteristics of these tumors. In this chapter we will review the pro(cid:173)
`cess that allowed us to stumble onto rapamycin as a potential therapeu(cid:173)
`tic agent, and the progress in understanding why this macrolide antibi(cid:173)
`otic may exert tumor-specific cytotoxicity.
`
`utocrine Growth of Rhabdomyosarcomas
`
`2 A
`
`Our laboratory has focused on rhabdomyosarcoma, a tumor of skeletal
`mu scle origin, and in parti cular a particularly aggressive "alveolar" vari(cid:173)
`ant thereof. Cytogenetic analysis of several of the se tumors from inde(cid:173)
`pend ent patients that were established as xenografts in immune-de(cid:173)
`prived mice showed a consistent chromosomal
`translocation t(2:13)
`(q35;qI4; Hazelton et al. 1987). A more systematic sur vey of patient tu(cid:173)
`mor biopsies demonstr ated consistent
`translocations in greater than
`90% of alveolar rh abdomyosarcomas (Douglass et al. 1987). We now
`know that this translocation results in expression of a chimeric tran-
`
`I Years 1974 to presen t based on SEE R data [Ries LAG, Miller BA, Guemey JG,
`Linet M, Tamra T, Young JL, Bunin GR (cds)]. SEE R Progra m, 1975-1 995; Tables
`and Graphs, Nationa l Cancer Institute. NIH Pub. No. 99-4649. Bethesda, MD, 1999
`
`Roxane Labs., Inc.
`Exhibit 1034
`Page 002
`
`

`
`mTOR as a Target for Cancer Therapy
`
`341
`
`scription factor (PAX3/FKHR) that appears to block myogenic differen(cid:173)
`tiation (Galili et al. 1993; Shapiro et al. 1993; Epstein et al. 1995) leading
`to tumor formation. These studies lead to further characterization of al(cid:173)
`veolar rhabdomyosarcoma cell lines, and revealed overexpression of
`transcripts for type II insulin-like growth factor (IGF-II) specifically
`from the fetal P3 promoter. In collaboration with Lee Helman at the Pe(cid:173)
`diatric Branch, National Cancer Institute (NCI), Bethesda, we were able
`to show that growth of rhabdomyosarcoma cells was driven by an auto (cid:173)
`crine loop. Specifically, cells secreted IGF-II, and signaled through the
`IGF-I receptor (El-Badry et al. 1990). Inhibition of the IGF-I receptor by
`using a neutralizing antibody inhibited tumor cell growth. This suggest(cid:173)
`ed to us that interference with IGF-I-receptor-mediated signaling may
`be a therapeutic strategy for these tumors. To test this concept, the IGF(cid:173)
`1 receptor was downregulated using a stable expression of antisense con(cid:173)
`structs (Shapiro et al. 1994). These studies showed a high correlation be(cid:173)
`tween downregulation of the receptor, decreased growth in soft agar
`(such growth being a characteristic of malignant cells), and decreased
`formation of tumors when cells were inoculated into immune-deprived
`mice. Further, clones with the lowest expression of IGF-I receptor ex(cid:173)
`pressed the highest levels of the myogenic marker, MyoD, and formed
`multinucleate syncytia, thus recapitulating some characteristics of myo(cid:173)
`genic differentiation. While our studies focus on rhabdomyosarcomas,
`there is increasing evidence that many tumors are "driven" by either au(cid:173)
`tocrine or paracrine signaling through the IGF-I receptor. Clearly dereg(cid:173)
`ulated IGF-I signaling is frequent in many pediatric solid tumors (neu(cid:173)
`roblastoma, Ewing's sarcoma, Wilms' tumor, medulloblastoma, glioblas(cid:173)
`toma), as well as many adult carcinomas (Macauly, 1992; Toretsky and
`Helman 1996). Direct inhibition of the IGF-I receptor with antibody,
`while effective in mouse models (Kalebic et al. 1994), was not at that
`time plausible in humans due to antigenicity, although this is now less of
`a problem and such reagents are being developed for clinical application.
`Alternatively, we sought a small molecule inhibitor ofIGF-I signaling.
`
`3 S
`
`elective Tumor Growth Inhibition by Rapamycin
`
`Our studies with rapamycin started in 1992, and were stimulated by a
`chance conversation with Randall Johnson at Smith Kline Beecham. At
`that time it was known that the immunosuppressive agents FK506 and
`
`Roxane Labs., Inc.
`Exhibit 1034
`Page 003
`
`

`
`342
`
`P. J. Houghton and S. Huang
`
`Table 1 Sensitivity of childhood rhabdomyosarcoma cell lines and human colon car(cid:173)
`cinoma celIlines to rapamycin and geldan amycin
`
`Rapamycin ICso (nglml)
`
`Geldanamycin Cso (nM)
`
`Rhabdomyosarcoma cell lines
`Rhl
`Rh18
`Rh28
`Rh30
`Colon carcinoma cell line s
`GC3/cl
`VRCs/cl
`CaCo
`HCT8
`HCT29
`HCn 16
`National Cancer Institute
`screen (60 celIlines)
`
`4,680
`0.1
`8.0
`0.37
`
`9,800
`1,280
`1,570
`8,400
`> 10,000
`> 10,000
`3,160
`
`5.9
`14.3
`17.9
`1.9
`
`3.6
`1.4
`3.4
`NDa
`2.6
`ND
`
`a ND, not determined (from Dilling et aI. 1994).
`
`cyclosporin A blocked T cell activation prior to expression of interleu(cid:173)
`kin-2, whereas rapamycin acted downstream of interleukin-2 expression
`(Flanagan et al. 1991; Schreiber and Crabtree 1992; McCaffrey et al.
`1993). There was also some suggestion that for T cells to progress to S
`phase,
`the IGF-I receptor had to be expressed (Reiss et al. 1992). We
`speculated that perhaps rapamycin acted downstream of the IGF-I re(cid:173)
`ceptor to block cell cycle progression, and if so, may act to inhibit the
`growth of rhabdomyosarcoma cells. The results, shown in Table 1, were
`both surprising and exciting. Three of four rhabdomyosarcoma cell lines
`were exquisitely sensitive to rapamycin whereas one line, (Rh l ), which
`is less dependent on IGF-I mitogenic signaling , was highly resistant. The
`other interesting aspect of these results was the marked selectivity for
`rhabdomyosarcoma cells relative to colon carcinoma cells, or cells used
`in the NCI in vitro screen (Dilling et al. 1994). Intriguing, but not com(cid:173)
`prehended (at that time) was the observation that under serum-free con(cid:173)
`ditions Rh l cells became very sensitive to rapamycin, with the IC50 de(cid:173)
`creasing from 5,800 ng/ml
`to 3.6 ng/ml. Consistent with results from
`other laboratories,
`the effect of rapamycin could be competed using
`FK506, indicating that initial formation of the FKBP-rapamycin complex
`was important.
`
`Roxane Labs., Inc.
`Exhibit 1034
`Page 004
`
`

`
`mTOR as a Target for Cancer Therapy
`
`343
`
`echanism of Action of Rapamycin
`
`4 M
`
`The action of rapamycin will be covered in detail elsewhere. Briefly, ra(cid:173)
`pamycin first binds the immunophilin FKBP-12 (the 12-kDa FK506
`binding protein) and this complex is now known to be a specific inhibi(cid:173)
`tor of a serine/threonine kinase mTOR (the mammalian target of ra(cid:173)
`pamycin, also called FRAP/RAPT/RAFT; Brown et al. 1994; Sabatini et
`al. 1994; Chiu et al. 1994; Sabers et al. 1995). Kinase mTOR is a member
`of the PIKK superfamily that includes ATR, ATM, Mecl, and Tell pro (cid:173)
`teins having homology to phosphatidylinositol lipid kinases. Evidence
`increasingly implicates mTOR as a central controller of cell growth and
`proliferation, and it controls initiation of translation of ribosomal pro(cid:173)
`teins and several proteins that regulate cell cycle. Activation of ribosom(cid:173)
`al S6Kl after mitogen stimulation is dependent on mTOR (Chung et al.
`1992; Kuo et al. 1992; Terada et al. 1992). Cap-dependent translation is
`facilitated by mTOR's phosphorylation and inactivation of 4E-BPs, sup (cid:173)
`pressors of eukaryotic initiation factor 4E (eIF4E; Lin et al. 1994, Pause
`et al. 1994, Beretta et al. 1996). More recent findings have shown that
`mTOR may directly or indirectly control transcription, ribosomal bio(cid:173)
`genesis, actin cytoskeleton organization, and protein kinase C (reviewed
`in Schmelzle and Hall, 2000). Recently, our studies with rhabdomyosar(cid:173)
`coma cells showed that activation of MAP kinases (p44/42) by growth
`factors was mTOR-dependent (Houghton et al. 2001; Harwood et al.,
`manuscript submitted),
`implicating mTOR in cross-talk between the
`PI3K and MAPK pathways in some cell lines. Thus, the emerging picture
`places mTOR in a central role in which it senses mitogenic stimuli and
`amino acid (Iiboshi et al. 1999), ATP (Dennis et al. 2001), or nutrient
`(Rohde et al. 2001) conditions; mTOR coordinates many cellular pro(cid:173)
`cesses related to growth and proliferation. The signaling pathways from
`IGF-IR to mTOR and downstream targets are depicted in Fig. 1.
`
`apamycin Induces Apoptosis in Rhabdomyosarcoma Cells
`
`5 R
`
`The basis for the differential sensitivity of Rhl cells under serum-con(cid:173)
`taining or serum-free conditions was of interest. The results suggested
`that some component of serum was able to rescue Rh l cells but not
`Rh30 cells. Consequently, we attempted to identify components of serum
`
`Roxane Labs., Inc.
`Exhibit 1034
`Page 005
`
`

`
`344
`
`P. J. Houghton and S. Huang
`
`Ribosomal
`proteins
`IGF-II
`
`Cyelin DI
`ODe.
`c- M YC
`, I IIIF I
`
`,=-
`
`-
`
`Fig. 1 Simplified schema of signaling pathways from the IGF-I receptor to mTOR
`and Ras in mammalian cells
`
`that protected from rapamycin (Hosoi et al. 1999). As shown in Fig. 2,
`under serum-free conditions IGF-I completely protected Rh l cells, but
`had no apparent effect on the action of rapamycin against Rh30 cells.
`Further, IGF-I rescue was not a consequence of reversing rapamycin in(cid:173)
`hibition of ribo somal S6Kl activation, as S6Kl activity remained sup(cid:173)
`pressed. So far the only growth factors shown to protect Rh1 cells are
`IGF-I, IGF-II, and to a lesser extent insulin (Thimmaiah et al. 2003). As
`IGF-I has been reported to prevent apoptosis in cells undergoing differ(cid:173)
`ent forms of stress (Butt et al. 1999; Fujio et al. 2000; Kulik et al. 1997;
`Sell et al. 1995), we examined the fate of Rh l and Rh30 cells treated un(cid:173)
`der serum-free conditions with or without IGF-I supplementation. As
`shown in Fig. 3, under serum-free conditions rapamycin induces quite
`dramatic apoptosis of both Rhl and Rh30 cells, and IGF-I essentially
`completely protects against the effect of rapamycin. Thus , under serum(cid:173)
`free conditions the response to rapamycin is apoptosis, whereas under
`seru m-containing conditions Rh30 cells are growth-arrested, but Rhl
`cells continue to proliferate. Interestingly, expression of a rapamycin re(cid:173)
`sistant mTOR mutant (Ser2035-> Ile) conferred resist ance to both the
`growth-inhibiting and apoptosis-inducing activities of rap amycin. Thus,
`
`Roxane Labs., Inc.
`Exhibit 1034
`Page 006
`
`

`
`mTOR as a Targe t for Cancer Therapy
`
`345
`
`A
`
`1 20
`
`Rh1
`
`___ t
`
`12 0
`
`100
`
`Rh30
`
`100
`
`.._-.t- .-~ 80
`
`e~
`
`8 0
`
`o"
`
`60
`
`40
`
`20
`
`O L..J...---'-
`
`-'--
`
`---'--
`
`--'---
`
`-'----'
`
`100 1000
`1 0
`1
`0 .1
`0.01
`Rapamycin (ng/ml)
`
`0
`
`2 , '
`
`L'
`
`.,.
`
`Q'!.
`:;;6 0
`
`~ C
`
`)40
`
`OJo
`
`20
`
`o L..-!._'--_ -'-_ -'-_ -'-_ -'-_ .....
`o
`
`10
`1
`0 .1
`0 .0 1
`Aapamycin (ng/mll
`
`1 0 0 10 0 0
`
`2 ,'
`
`B
`
`m
`:;
`
`cE
`
`~1
`.2:'t:l
`u~
`c( ~ 1 .5
`m(cid:173)
`
`.. "'"'cc'"~::
`
`1
`
`., .!
`..
`IIIc.c
`,..0.
`o.~-a.,.
`
`a:
`
`:E
`0.
`
`~
`
`.
`..
`
`l:
`.t
`..
`E
`•
`;;
`
`e
`:i
`...
`!2
`
`e
`:i
`...
`~
`!2
`
`..
`c
`:i
`...
`...
`!2
`
`s:
`:i
`
`..
`0.
`...
`a:
`s
`
`e
`:i
`~
`0.
`
`..
`...
`0:
`S!
`
`e
`:i
`...
`..
`0
`...
`...
`S!
`
`0:
`
`.
`..
`
`E
`;;
`
`e
`:i
`
`. ...
`...
`~
`..
`E
`;;
`
`!2
`
`e
`2
`:e
`...
`!2
`
`e
`2
`II
`!2
`
`..
`
`e
`:i
`...
`..
`0.
`...
`0:
`!2
`
`e
`:i
`=
`..
`
`0.
`~
`
`!2
`
`e
`:i
`..
`~
`...
`..
`
`0:
`!2
`
`Fig.2A, B Inhibitory effect of rapamycin and effect of IGF-I on Rh i (left panels) and
`Rh30 (right panels) rhabdomyosarcoma cells. A Rapamycin sensitivity. Cells were seeded
`and allowed to attach overnight, washed, and cultured in serum-free N2E in medium con(cid:173)
`taining serial concentr ations of rapamycin with (closed square) or without (open square)
`addition of IGF-I (10 ng/ml) for 7 days, Growth was assessed by lysing cells and counting
`nuclei. Results are presen ted as percent control. Cell nu mber for control Rhl cells was
`I.74xl05, and for Rh30 was s.zxio', respectively. B Activation of p70S6 K by IGF-I is
`blocked in Rhl (left panel) and Rh30 (right panel) by rap amycin, Cells were serum(cid:173)
`starved overn ight, then stimulated with IGF-I (IO nglml) without or with prein cubation
`for 15 min with rapamycin (IOOng/ml). Ribosomal p70S6 K assays were performed on im(cid:173)
`munoprecipitates derived from 3x106 cells. Each value repre sents the mean±SD for 4 de(cid:173)
`terminations, and shows a representative experi ment. (From Hosoi et al. 1999)
`
`Roxane Labs., Inc.
`Exhibit 1034
`Page 007
`
`

`
`346
`
`P. J. Houghton and S. Huang
`
`Rh30
`
`~tit=j B. Serum -free
`
`C. Rapamycin 100ng/ml
`
`D. Rapamy cin 100n g/m l
`IGF- 1 10ng /ml
`
`,a,
`
`'0'
`
`10'
`
`Annexin V-FITC Fluorescence
`
`Fig.3 Rapamycin induces apoptosis that is blocked by IGF-I. Rhl (left panels), and
`Rh30 cells (right panels) were grown und er seru m-containing (FBS) or serum-free
`(N2E) culture conditions supplemented with rapamycin (lOO ng/m!) or rapamycin
`plus IGF-I (lO nglml). Cells were har vested after 6 (RhI) or 4 days (Rh30), and ap(cid:173)
`optosis measured by the ApoAlert method. Cells were analyzed by FACS for annex(cid:173)
`in-V-FITC and propidium iodide fluorescence. (From Hosoi et al. 1999)
`
`apoptosis is a consequence of inhibiting mTOR, and not through a sec(cid:173)
`ond mechanism of action. Of note is that both RhI and Rh30 cells are
`mutant for the p53 tumor suppressor gene. This raised the possibility
`that p53 may be involved in sensing mTOR inhibition and cooperating
`in enforcing a G, arrest.
`
`Roxane Labs., Inc.
`Exhibit 1034
`Page 008
`
`

`
`mTOR as a Target for Cancer Therapy
`
`347
`
`6 T
`
`he Tumor Suppressor p53 Protects
`from Rapamycin-Induced Apoptosis
`
`Cellular response to mTOR inhibition by rapamycin is generally charac(cid:173)
`terized by p53-independent (Metcalfe et al. 1997) cytostasis without cell
`death. In contrast, our studies have shown that
`the response to ra(cid:173)
`pamycin in rhabdomyosarcoma cells lacking functional p53 is apoptosis
`(Hosoi et al. 1999; Huang et al. 2001). Apoptotic cells appeared to pro(cid:173)
`gress from Gl to S phase, as >90% had incorporated BrdUrd in the pres(cid:173)
`ence of rapamycin. To determine whether cellular response to rapamycin
`was determined by p53 function, Rh30 cells were infected with aden(cid:173)
`oviruses expressing wild-type p53 or the cyclin-dependent kinase inhib(cid:173)
`itor p21Cipl (Huang et al. 2001). Expression of p53 or p21cipl protected
`cells. We speculated that the mechanism was probably an enforcement
`of Gl arrest and prevention of cells from initiating DNA replication.
`More recent data (Huang et al. 2003) suggest that cell cycle arrest is not
`the mechanism, as expression of a truncated p21Cipl allele lacking the
`nuclear localization signal does not cause G1 accumulation, but protects
`against rapamycin-induced apoptosis.
`To further investigate the role of p53 and p21Cipl
`in a non-trans(cid:173)
`formed background we used murine embryo fibroblasts (MEFs) with
`disrupted p53 or p21c ipl genes. Specifically, MEFs with disruption of ei(cid:173)
`ther p53 or p21cipl undergo apoptosis whereas wild-type cells arrest in
`Gl without loss of viability when treated with rapamycin (Fig. 4). Re-ex(cid:173)
`pression of wild-type p53 confers resistance to rapamycin-induced ap(cid:173)
`optosis (Huang et al. 2001). Exactly how mTOR signaling feeds into p53
`remains to be determined. However, of importance for potential thera(cid:173)
`peutic application to cancer treatment is that rapamycin is selectively
`toxic to cells with attenuated p53-mediated Gl checkpoint responses, at
`least in the absence ofIGF-I.
`
`echanism(s) of Resistance to Rapamycin
`
`7 M
`
`Rapamycin resistance was first found in yeast (Heitman et al. 1991).
`Mechanisms of rapamycin resistance are multiple and complicated;
`some of them have been identified and some remain to be elucidated.
`Cells may acquire resistance with or without mutagenesis. For instance,
`
`Roxane Labs., Inc.
`Exhibit 1034
`Page 009
`
`

`
`348
`A.
`.,.r-- .,....-- ...,
`
`P. J. Houghton and S. Huang
`
`B.
`,,' y---
`
`.,--
`
`...,
`
`g
`8
`"j
`...
`'"
`'2
`~
`E
`.2
`X
`0
`0.:.
`
`\.,.lo'_..
`
`p53 ·/·
`C<>ntrol
`
`p21 ·' ·
`C ontrof
`
`8
`c:
`~
`'"~
`ii:
`~
`:B

`:2a.

`
`,,'
`
`..I
`
`\
`
`I
`
`.
`.".
`:' V
`
`j
`
`!\.. '\
`I
`\
`
`p53 . '+
`Rap 100
`
`p53 -I.
`Control
`
`p53 .f·
`Rap 100
`.h.
`\
`
`"-
`,
`
`p53 ./.
`Ad-p53
`
`\.A .~
`
`p53 .f·
`Ad-p53
`• Rapl00
`
`\,
`
`w ,IS'
`,OJ' wI
`10' 1iT .0'
`trI'
`Anne llin Y·FITC Fluonlscence
`
`to'
`
`td 10' ttl
`,,'
`to'
`1ft'
`10'
`Anne xin Y· FITC Fluorescence
`
`\G'
`
`Fig. 4 The cellular response to rapamycin is dependent on functional p53 or p21cip l
`in murine embryo fibrobl asts. Left: Wild-type, p53-/', and p21-/- MEFs were grown
`und er serum-free condition s with or without rapamycin (IOO ng/ml). After 5 days
`cells were harvested and apopt osis determined by ApoAlert assay. Result s show a
`representative experiment. Percent distribution of cells in each qu adrant is present(cid:173)
`ed. Right: Wild-type, and p53-/- MEFs, and p53-1- MEFs infected with Ad-p53 (MOl of
`100) were grown without or with rapamycin (100 ng/ml). Cells were harvested after
`5 days and apoptosis determined by quantitative FACS analy sis (ApoAlert) , as de(cid:173)
`scribed in Fig. 3. The percent distribution of cells in each quad rant is presented. Re(cid:173)
`sults show a representative experiment. (From Huang et al. 2001)
`
`Roxane Labs., Inc.
`Exhibit 1034
`Page 010
`
`

`
`mTOR as a Target for Cancer Therapy
`
`349
`
`specific mutations in FKBP12 that prevent the formation of FKBP-ra(cid:173)
`pamycin complex (Fruman et al. 1995), or certain mutations in FKBP-ra(cid:173)
`pamycin-binding domain of mTOR (Ser2035~Ile) that block binding of
`FKBP-rapamycin complex to mTOR (Chen et al. 1995; Hosoi et al. 1998),
`conferred rapamycin resistance. Mutation of Thr389 ~Glu (Dennis et al.
`1996) or Thr229~Ala or Glu (Sugiyama et al. 1996) of S6K1, a down(cid:173)
`stream effector of mTOR, also rendered S6K1 insensitivity to rapamycin.
`As discussed in other chapters, the signaling pathway directly down(cid:173)
`stream of mTOR bifurcates (von Manteuffel et al. 1997). In mammalian
`cells, mTOR signals to both S6K1 and 4E-BP, and probably to other
`unidentified effectors. We were interested in understanding whether cel(cid:173)
`lular responses to rapamycin (growth inhibition and apoptosis) were
`dictated by inhibition of signaling to either S6K1 or 4E-BP, or both. One
`approach to understanding which of these pathways is critical to cellular
`proliferation and survival was to select for rapamycin resistance and de(cid:173)
`termine which pathway(s) altered to accommodate rapamycin inhibition
`of mTOR. Rapamycin-resistant clones (Rh30/RapalOK) were obtained
`by continuously growing Rh30 cells in the presence of increasing con(cid:173)
`centrations of rapamycin, without prior mutagenesis (Hosoi et al. 1998).
`These initial studies showed that in resistant clones, rapamycin still in(cid:173)
`hibited IGF-I-stimulated mTOR-dependent activation of S6Kl. However,
`of note, resistance was characterized by elevated c-MYC. Further inde(cid:173)
`pendent clones were selected, either with or without mutagenesis (Dil(cid:173)
`ling et al. 2002). Interestingly, resistance was unstable in each of the
`clones characterized. When the selecting pressure (i.e., rapamycin) was
`withdrawn, cells reverted to rapamycin sensitivity within 6-10 weeks. In
`resistant cells, as compared to parental cells, approximately tenfold less
`4E-BP was bound to eIF4E, and total cellular 4E-BP was markedly re(cid:173)
`duced . In contrast, levels of eIF4E were unchanged. Steady-state levels of
`4E-BP transcripts remained unaltered, but the rate of 4E-BP synthesis
`was reduced in resistant cells. Of importance, in cells that reverted to ra(cid:173)
`pamycin sensitivity, levels of total 4E-BP returned to those of parental
`cells. Compared to parental cells, resistant clones had either similar or
`lower levels and activity of ribosomal S6K1, but c-MYC levels were ele(cid:173)
`vated in both resistant and revertant clones. Further, anchorage-inde(cid:173)
`pendent growth was enhanced in c-MYC-overexpressing cells, suggesting
`that the eIF4E pathway controls some aspects of the malignant pheno(cid:173)
`type (Lazaris-Karatzas et al. 1990; DeBenedetti et al. 1994). These results
`indicate that accommodation in the eukaryotic-initiation-factor 4E
`
`Roxane Labs., Inc.
`Exhibit 1034
`Page 011
`
`

`
`350
`
`P. J. Houghton and S. Huang
`
`_
`
`4E·BPI
`
`_
`
`fH ubo lin
`
`_
`
`_
`
`eIF4E
`
`13 ·tubu lon
`
`co
`l(cid:173)o
`J:
`
`HCT8·4E-BPI clon es
`
`2
`
`3
`
`4
`
`5
`
`I
`
`B.
`
`4E-BP1 . .
`
`elF4E . .
`
`c.
`
`l00 '-lli!= _ _
`
`ec
`8 60
`
`40
`
`C2
`
`lCii
`a.
`
`o i~~_......
`,--__"",
`"","--
`lit
`1000
`100
`10
`1
`Rapamyc in (ng/ml)
`
`!
`
`Roxane Labs., Inc.
`Exhibit 1034
`Page 012
`
`

`
`mTOR as a Target for Cancer Therapy
`
`351
`
`(eIF4E) pathway occurs in rapamycin resistance through suppression of
`translation of 4E-BP. This suggests that
`inhibition of cap-dependent
`translation mediated by eIF4E is critical for rapamycin-induced growth
`arrest and apoptosis. Further, intrinsic or acquired resistance may be a
`consequence of low levels of 4E-BP suppressor proteins. To examine
`whether intrinsic resistance was associated with low 4E-BP, we examined
`levels in colon carcinoma cell lines previously characterized as ra(cid:173)
`pamycin resistant (Dilling et al. 1994). Several of these cell lines with in(cid:173)
`trinsic rapamycin resistance were found to have low 4E-BP relative to
`eIF4E (Fig. 5A). To explore the role of 4E-BP in rapamycin resistance,
`stable clones of HCT8 colon carcinoma were engineered to overexpress
`4E-BP. As shown in Fig. 5, rapamycin sensitivity increased markedly
`(> l,OOO-fold) as 4E-BP expression increased. These results suggest that
`the 4E-BP:eIF4E ratio is an important determinant of rapamycin resis(cid:173)
`tance in some cell lines. However,
`this mechanism clearly does not ac(cid:173)
`count for resistance in other cell lines.
`
`reclinical Antitumor Activity for Rapamycins
`
`8 P
`
`Initial in vivo testing of rapamycin as a potential antitumor agent was
`undertaken through the Division of Cancer Treatment at the National
`Cancer Institute (NCI; Douros and Suffness 1981; Houchens et al. 1983;
`
`Fig.5A-C Overexpression of 4E-BP abrogates resistance to rapamycin. A Western
`blot analysis of 4E-BP,elF4E, and tubulin (loading control) in cell lines that have dif(cid:173)
`ferent intrinsic sensitivities to rapamycin. Colon carcinoma cell lines CaCo2, GC3/cl,
`HCTS, HCT29, Hcn I6, and VRC5/cI are intrinsically resistant to rapamycin with
`IC50 concentrations >1,200 ng/ml. Pediatric solid tumor lines SJ-G2 (glioblastoma)
`and RhlS and Rh30 (rhabdomyosarcoma) are sensit ive to rapamycin (lC50 < I ng/
`ml). B Expression of 4E-BP and elF4E in HCTS clones stably transfected with a 4E(cid:173)
`BP expression plasmid (pcDNA3-PHAS-I). Expression of 4E-BP was greater in clones
`C2, C4, and C5 than in parental HCT8 cells, but expression of was similar in parental
`and CI and C3 transfected clones. C Sensitivity to rapamycin. Cells were plated at
`low density in increasing concentrations of raparnycin, and colonies were counted
`after 7 days of exposure to rapamycin. Symbols: Parental HCT8 (closed circle) and
`clones Cl (open circle), C2 (closed square), C3 (open square), C4 (closed triangle),
`and C5 (open triangle) . Each point is the mean±SD of three determinations. (From
`Dilling et al. 2002)
`
`Roxane Labs., Inc.
`Exhibit 1034
`Page 013
`
`

`
`352
`
`P. J. Houghton and S. Huang
`
`Eng et al. 1984). These preliminary results revealed that rapamycin sig(cid:173)
`nificantly inhibited the growth of syngeneic B16 melanoma, colon carci(cid:173)
`noma models 26 and 38 tumor, CD8Fl mammary tumor, and EM epen(cid:173)
`dymoblastoma. Rapamycin, in the active dose range, was less toxic than
`conventional antitumor drugs, such as 5-fluorouracil, cyclophospha(cid:173)
`mide, and adriamycin (Eng et al. 1984). However, at that time Ayerst Re(cid:173)
`search Laboratories (Montreal, Canada), where rapamycin was first iso(cid:173)
`lated and characterized, abandoned rapamycin as an antitumor agent
`because they failed to develop a satisfactory intravenous formulation for
`clinical
`trials. Recently, two rapamycin ester analogues, CCI-779 [ra(cid:173)
`pamycin-42, 2, 2-bis(hydroxymethy1)-propionic acid; Wyeth-Ayerst] and
`RADOOI [everolimus, 40-0-(2-hydroxyethyl)-rapamycin; Novartis, Basel,
`Switzerland], with improved pharmaceutical properties have been syn(cid:173)
`thesized and evaluated. CCI-779 is designed for intravenous injection,
`whereas RADOOI for oral administration. Both have antitumor effects
`similar to rapamycin, are currently being developed as antitumor agents,
`and are undergoing phase I-III clinical trials. In culture, rapamycin and
`CCI-779 potently inhibit growth of numerous malignant cell lines, in(cid:173)
`cluding those derived from rhabdomyosarcoma, neuroblastoma, glio(cid:173)
`blastoma, medulloblastoma, small cell lung cancer (Dilling et al. 1994;
`Shi et al. 1995; Seufferlein and Rozengurt, 1996; Hosoi et al. 1998; Geo(cid:173)
`erger et al. 2001; Dudkin et al. 2001), osteoscarcoma (Ogawa et al. 1998),
`pancreatic carcinoma (Grewe et al. 1999; Shah et al. 2001), breast and
`prostate carcinoma (Gibbons et al. 2000; Yu et al. 2001), murine melano(cid:173)
`ma, T-cell
`leukemia, and B-cell lymphoma (Houchens et al. 1983;
`Hultsch et al. 1992; Gottschalk et al. 1994; Muthukkumar et al. 1995).
`CCI-779 in vivo also inhibited the growth of human U251 malignant gli(cid:173)
`oma cells that were resistant
`to rapamycin in vitro (Geoerger et al.
`2001). CCI-779 induced tumor growth inhibition correlated with de(cid:173)
`creased phosphorylation of 4E-BP (Dudkin et al. 2001).
`
`9 C
`
`linical Trials for Antitumor Efficacy of Rapamycins
`
`As alluded to, RADOOI and CCI-779 are undergoing phase I and phases
`II/III clinical trials, respectively. However, only preliminary results from
`phase I trials of CCI-779 are currently available. Two groups conducted
`phase I trials for CCI-779 using different schedules (Hidalgo et al. 2000;
`Raymond et al. 2000). In the USA, CCI-779 was administered as a 30-
`
`Roxane Labs., Inc.
`Exhibit 1034
`Page 014
`
`

`
`mTOR as a Target for Cancer Therapy
`
`353
`
`min intravenous infusion daily for 5 days every 2 weeks at doses of
`15-24 rng/rrr'per day. Of 45 patients with various types of cancer, nine of
`them showed some evidence of tumor response (Hidalgo et al. 2000). In
`Europe, CCI-779 was administered as a 30-min intravenous infusion
`weekly at doses of 7.5-220 mg/rrr' per week (Raymond et al. 2000). After
`2:8 weekly doses, of 18 patients, significant tumor regressions were ob(cid:173)
`served in two patients with lung metastasis of renal cell carcinomas
`(both treated with 15 mg/rrr' per week) and in one patient with a neuro(cid:173)
`endocrine tumor of the lung treated with 22.5 mg/rn! per week. Two pa(cid:173)
`tients experienced tumor stabilization. Consistently, these two groups
`showed that CCI-779 was well tolerated in patients with only mild side
`effects, such as acneform rash, mild mucositis, some thrombocytopenia,
`and elevated triglyceride and cholesterol levels (Hidalgo and Rowinsky
`2000). It is anticipated that these agents will be evaluated against addi(cid:173)
`tional histiotypes in phase II trials.
`
`10
`Conclusions
`
`Rapamycin and its derivatives represent novel agents for therapy of hu(cid:173)
`man cancer. In this review we have detailed some of the studies from this
`and other laboratories that have assisted in development of this class of
`drug specifically as cancer chemotherapeutic agents. Other studies,
`many reported in this book, have elucidated the mechanism of action of
`rapamycin, and have identified pathways proximal and distal to mTOR
`that have lead to a greater understanding of how this class of drug exerts
`cytostatic activity. Our studies have suggested that cellular response to
`rapamycin is converted from G1 cytostasis to apoptosis when p53 is mu(cid:173)
`tated in tumor cells, or disrupted in embryo fibroblasts. These results
`suggest that combining rapamycin, or its derivatives, with an inhibitor
`of the IGF-I receptor may induce apoptosis selectively in tumor cells
`with mutated p53. As both humanized antibodies against this receptor
`and small molecule inhibitors are in development, the validity of this ap(cid:173)
`proach will be amenable to testing relatively quickly. Recently, Neshat et
`al. (2001) have shown that PTEN (phosphatase and tensin homolog
`deleted on chromosome ten) -mutated or PTEN-deficient
`tumor cells
`were more sensitive to CCI-779 (Podsypanina et al. 2001; Neshat et al.
`2001). The PTEN-mutated tumor cells demonstrated elevated levels of
`phosphorylated Akt and activated S6Kl (Podsypanina et al. 2001). Loss
`
`Roxane Labs., Inc.
`Exhibit 1034
`Page 015
`
`

`
`354
`
`P. J. Houghton and S. Huang
`
`of PTEN by deletion or mutation has been reported in approximately
`50% of all solid human tumors (Simpson and Parsons 2001). Thus, there
`are at least two mechanisms by which to anticipate tumor-selective ac(cid:173)
`tivity of rapamycin analogues as cancer chemotherapeutic agents.
`
`Acknowledgment. We wish to dedicate this review to the memory of Charles B. Pratt,
`M.D., who dedicated his life to helping children with cancer at St. Jude and through(cid:173)
`out
`the world . Studies from this laboratory were supported by USPHS awards
`CA23099, CA77776, CA96696 and CA21765 .
`
`References
`
`Beretta L, Gingras AC, Svitkin YV, Hall MN, Sonenberg N (1996) Rapamycin blocks
`the phosphorylation of 4E-BPI and inhibits cap-dependent initiation of tra nsla(cid:173)
`tion . EMBO J 15:658-664
`Brown EJ, Albers MW, Shin TB, Ichikawa K, Keith CT, Lane WS, Schreiber SL (1994)
`A mammalian protein targeted by Gl-arresting rapamycin-receptor complex. Na(cid:173)
`ture 369:756-758
`Butt A.J., Firth S.M. and Baxter R.C. (1999) The IGFaxis and programmed cell death .
`Immunol and Cell BioI. 77:256-262
`Chen J, Zheng XF, Brown EJ, Schreiber SL (1995) Identification of an ll-kDa
`FKBPI2-rapamycin-binding domain within the 289-kDa FKBPI2-rapamycin-as(cid:173)
`sociated protein and characterization of a critical serine residue . Proc Nat! Acad
`Sci USA 92:4947-4951
`Chiu MI, Katz H, Berlin V (1994) RAPTl, a mammalian homolog of yeast Tor, inter(cid:173)
`acts with the FKBPl2/rapamycin complex . Proc Nat! Acad Sci USA 91:12574(cid:173)
`12578
`Chung J, Kuo CJ, Crabtree GR. Blenis J.(1992) Rapamycin -FKBP specifically blocks
`growth-dependent activation of and signaling by the 70 kd S6 protein kinases.
`Cell 69:1227-1236
`Damiens E (2000) Molecular events that regulate cell proliferation : an

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