throbber
Inhibitors of mammalian target of rapamycin as novel antitumor agents:
`From bench to clinic
`
`Shile Huang & Peter J Houghton*
`Address
`Department of Molecular Phamraoology
`St Jude Children's Research Hospital
`332 N Laudardala
`Memphis
`TN 3B105»2?94
`USA
`Email: peter.houghton@stjude.org
`
`‘To whom correspondence should be addressed
`
`Current Opinion In lnvesligatlonal Drugs 2002 3(2):295-304
`O Phan'naPress Ltd ISSN 1472-44‘i'2
`
`Rapamycin and its derivatives, CCI-779 and RAD-D01, inhibit tlie
`mannnaliau target of
`rapamycin
`(n1'1"OR,l, doronregulating
`translation of specific mRNAs required for cell cycle progression
`from C1 to S phase. Preclinically, mTOR inhibitors potently
`suppress growth and proliferation of numerous tumor cell lines in
`culture or when grown in mice as xenografts. CC!-779 and RAD-
`OOI are being developed as antitumor drugs and are undergoing
`clinical
`trials. Clinically, CCl-779 has
`shown evidence of
`antitumor activity but
`imluce.-:l relatively mild side eflfects
`in
`patients. Here we discuss potential antitumor mechanisms and
`resistance meclmnisnts of mTOR inhibitors, and summarize the
`current status of these compounds as novel antitumor agents.
`
`Keywords Antitumor, cell cycle, mammalian target of
`rapamycin (mTOR), rapamycin
`
`Introduction
`Malignant disease is characterized by genetic mutations or
`compensatory changes in cells that result in unregulated
`population growth due to increased proliferation or decreased
`cell death. Since the early 1950s, extensive chemical synthesis
`and screening programs have resulted in clinical
`trials of
`many potential anticancer agents. In some cases, cytotoxic
`agents have significantly increased survival rates in adult
`diseases and notably for children with hematologic as well as
`solid tumors. However, relatively few cytotoxic agents have
`proven to be useful against a wide spectrum of cancers and
`' most cause significant toxicity. The reasons for the relatively
`poor activity of cytotoxic agents are numerous. Most do not
`target the transforming event, rather they induce forms of
`damage that lead to necrosis or activation of cell-suicide,
`apoptosis. As a consequence, agents are cytotoxic to both
`malignant tumor cells and normal healthy cells, often causing
`severe side effects. Understanding pathways known to be
`critical to the growth and survival of tumors is essential to
`developing potentially selective treatments. Validation of this
`concept has been met with compounds such as imatinib
`(Gleevec, STI—571; Novartis AG). This compound inhibits
`tyrosine kinases such as BCR/ABL in chronic myeloid
`leukemia and c-KIT in gastrointestinal stromal tumors. Less
`success has been met by targeting activated Ras with
`inhibitors of farnesyltransferase.
`
`Wyeth-Ayerst
`Rapamune;
`(sirolimus,
`Rapamycin
`Laboratories; Figure 1), an immunosuppressant, has emerged
`as a potent inhibitor of a signaling pathway that may be
`deregulated in some forms of cancer,
`leading to both
`
`increased growth and malignant characteristics of cells. It is a
`lipophilic
`macrolide,
`that
`selectively
`inhibits
`a
`serine/theronine kinase, specifically, the lammalian target gt
`gapamycin
`(mTOR).
`mTOR
`lies
`downstream of
`phosphatidylinositol 3-kinase (PI3I<)
`in the PI3l( signaling
`pathway. Rapamycin was originally isolated as a fungicide
`from the soil bacteria Streptomyces liygroscopicus, collected
`fron1 Easter Island (known as Rapa Nui to the natives) in the
`South Pacific in 1975 [1,2]. Structurally similar
`to the
`imrnunosuppresive reagent FK-506 ttacrolirnus; Fujisawa
`Pharmaceutical Co Ltd), rapamycin was initially developed
`for transplant rejection [3,4] and was approved by the US
`Food and Drug Administration in September 1999 and the
`European Commission in March 2000. While rapamycin was
`being developed as an immunosuppressant, it was also found
`to exert potent antitumor activity in intro and in viva [5-7].
`However, perhaps because its mechanism of action was
`unknown at that time, rapamycin was not developed as a
`cancer therapeutic.
`
`The potential for rapamycin as a cancer therapeutic was
`refocused, in part, by studies of Dilling et al [8]. These were
`the first studies to demonstrate potent and selective
`inhibition of growth by rapamycin. Rapamycin potently
`inhibited
`the growth of
`rhabdomyosarcoma
`cells
`at
`concentrations of approximately 1 ng/ml, whereas human
`colon cancer cells were inhibited only at micromolar
`concentrations in culture (Table 1). Proliferation of many
`rhabdomyosarcoma cells is regulated by an autocrine loop
`involving secretion of type II insulin-like growth factor {lGF-
`II) and signaling through the type I IGF receptor [9,10].
`Indeed,
`the cell
`lines most sensitive to rapamycin were
`dependent on this autocrine pathway.
`
`Additional findings from various research groups around
`the world support rapamycin as a good candidate for a
`cancer therapeutic agent. In many malignant cells in culture,
`rapamycin can act as a cytostatic agent by arresting cells in
`G1 phase. Another potential mode of its antitumor action is
`via the induction of apoptosis. Rapamycin potently inhibits
`proliferation
`or
`growth
`of
`cells
`derived
`from
`rhabdomyosarcoma,
`neuroblastoma,
`glioblastoma,
`medulloblastoma.
`small
`cell
`lung
`cancer
`[8,11-16],
`osteoscarcoma [17], pancreatic carcinoma [18,19], breast and
`prostate carcinoma [20-2], murine melanoma and leukemia,
`and B-cell lymphoma [6,23-25] (Table 1).
`
`Despite the antiproliferative effects of rapamycin, it has poor
`water-solubility and stability in solution, precluding its
`formulation for parenteral use as an anticancer agent. Two
`rapamycin ester analogs, CCI-7'79 (Wyeth-Ayerst Research;
`Figure 1) and RAD-091 (everolimus; Novartis AG; Figure 1),
`with improved pharmaceutical properties, but
`similar
`cellular effects to rapamycin [16,20—22,26,27,28-,29o], are
`currently undergoing antitumor phase III and phase I
`clinical trials, respectively. This review will discuss potential
`antitumor and resistance mechanisms of rapamycin and its
`derivatives, and summarize the preliminary data about
`these compounds, from the bench to the clinic.
`
`est-Ward Pharm.
`Exhibit 1017
`Page 001
`
`West-Ward Pharm.
`Exhibit 1017
`Page 001
`
`

`

`296 Current Opinion in Investigations! Drugs 2002 Vol 3 No 2
`
`Table 1. sensitivity at different tumor cell lines to rapamycln.
`cell Line
`flhabdomyosaraoma ceiis
`Fth1 (10% serum)
`H111 {serum free)
`H1118
`H1128
`Ft!-I30
`Colon carcinoma cells
`GCJG1
`VFiCJc1
`Ceca
`HCT8
`HOT29
`HCT116
`small cell lung manner cells
`H69
`H345
`H510
`Neurobiastama cells
`NB-SD
`NS-1643
`NB-EB
`NB-1691
`NB-1382.2
`Giiobiastama cells
`SJ-G2
`SJ-G3
`Medullabiastoma ceiis
`DAOY
`Ostoobiaete-like ostaosarcoma coils
`ROS 17.~‘2.B
`Pancreatic cancer cells
`Pane-1
`MiaPaca-2
`Leukemia cells
`FIBL-2H3
`B-col.‘ lymphoma cells
`BKS-2
`L1 .2
`' NFS1.1
`WEHI-279
`Thyrnama coils
`EL4
`
`Fla
`
`In cln C
`tnsfmli
`4630
`3.6
`
`0:80
`iflfiimil
`0.17
`
`
`
`Antitumor mechanism of inhibitors of mTOR
`Rapamycin and its analogs, CCI-779 and RAD-001, are the
`most potent and selective inhibitors of mTOR reported so
`far. The three agents share a common mechanism of
`antitumor action, ie, inhibiting mTOR, which, links mirage,-.
`stimulation to protein synthesis and cell cycle progresgiol-L
`mTOR
`
`antitumor mechanism of
`To better understand the
`rapamycin and its derivatives, we will briefly review the
`emerging cellular role of mTOR. mTOR is referred to by
`various other names, some of which are derived from its
`binding partner Fl(~506-binding protein, FKBP12 (discugsed
`below). These names are FRAP (FKBP12 and rapamycjn-
`associated protein}, RAI-T1 (rapamycin and FKBP12 target
`1), RAPT1 {rapamycin target 1) and SE13 {sirofilnus effector
`protein}.
`In the mid-1990s, mTOR was identified as a
`mammalian serine/threonine kinase of approximately 289
`kDa in humans, mice and rats [30-33]. TOR proteins
`represent a class of evolutionarily conserved kirtases in
`eukaryotes.
`In the yeasts, Saccharomyces
`cerevisiae and
`Scltfzosacclmromyces pombe, two TOR genes, TOR] and TOR2,
`
`have been cloned, which share 67% identity and encode
`proteins of approximately 280 kDa [34-36]. In the fruit fly,
`Drosophila melanogaster, a single TOR ortholog,
`termed
`dTOR, has been characterized, sharing 38% identity with
`TOR2 from Saccharomyces cerevisiae [37,38]. mTOR shares
`approximately 45% identity with TORI and TOR2 from the
`yeast Saccharomyces ccrwisiac, and 56% identity with dTOR in
`overall sequence [39,40]. Human, mouse and rat mTOR
`proteins share 95% identity at the amino acid level [40,41].
`mTOR contains a catalytic kinase domain and a Fl(BP12-
`rapamycin binding (FRB) domain near the C«terminus, and
`up to 2.0 tandemly repeated HEAT {Huntingtin, EF3, A
`subunit of PPZA and TOR) motifs at the N-terminus, as well
`as FAT (FRAP-ATM-TRRAP) and FATC {FAT C-terminus)
`domains (Figure 2}. Since the C-terminus of mTOR shares
`strong homology to the catalytic domain of PI3K, mTOR is
`considered a member of the Pll<-related kinase family,
`which also includes MEC1, TELL RAD3, MEI-41, DNA-PK,
`ATM, ATR and TRRAP [42»]. Both PI3l< and potentially
`protein kinase B (PKB; Akt) lie upstream of mTOR, whereas
`ribosomal p70S6 kinase (p70S6l() and eukaryotic initiation
`factor-4E (eIF—4E) binding protein isoforms (4E-BP1-3) are the
`
`st-Ward Pharm.
`Exhibit 1017
`Page 002
`
`West-Ward Pharm.
`Exhibit 1017
`Page 002
`
`

`

`Inhibitors of mammalian target of rapamycin as novel antitumor agents: From bench to clinic Huang 8. Houghton 297
`
`Figure 1. Molecular structures or raparnycin, col-779 and RAD-001.
`
`ocr-in
`(Wyeth-Ayersii
`
`best characterized downstream mTOR effector molecules.
`
`Figure 2. Schematic representation of mTOR domains.
`
`Increasing evidence has implicated mTOR as a central
`controller of cell growth and proliferation. mTOR may
`directly or indirectly regulate translation initiation, actin
`organization, membrane traffic and protein degradation,
`protein kinase C signaling, ribosome biogenesis and tRNA
`synthesis, as well as transcription [42u]. Recent results also
`suggest
`that mTOR may sense
`cellular ATP levels,
`suppressing protein synthesis when ATP levels decrease
`[43].
`
`Specificity of rapamycin action
`Rapamycin inhibits proliferation and growth of many
`tumor cells, which is clearly a consequence of binding
`mTOR. Whether this action is a consequence of inhibiting
`mTOR kinase activity per se is less clear. Raparnycin cannot
`directly bind to mTOR. It first has to bind to the 12 kDa
`cytosolic immunophilin, FKBP12, found in mammalian
`cells,
`to form the FKBP12-rapamycin complex. The
`complex then interacts with the FRB domain in mTOR
`(Figure 2),
`and inhibits
`function of mTOR. High
`concentrations of rapamycin together with FKBP12 are
`required to inhibit mTOR kinase activity in oiiro and
`mTOR autophosphorylation. However,
`the specificity of
`rapamycin action can be demonstrated in vivo as certain
`mutations in the FRB domain of mTOR affect FI(BP12-
`rapamycin binding, and significantly reduce the cellular
`sensitivity of rapamycin. The first
`rapamycin-resistant
`alleles, TORI-I and TOR2—1, identified in a Saccimromyces
`cereoisiae genetic screen were shown to confer dominant
`resistance. These mutant TOR proteins lost the ability for
`FKBP-rapamycin
`complex
`binding
`[44].
`Similarly,
`mammalian
`cells
`also
`became
`highly
`resistant
`to
`rapamycin when a mutation (Ser’°"—>Ile’“”} occurred in the
`FRB domain of mTOR, which resulted in decreased affinity
`for binding of FKBP12-rapamycin complex [14,45,46]. in
`the yeast. Smtcharonryces cerevisiae, decreased RBP1,
`a
`homolog of mammalian FKBP12, or mutation at Tyr", led
`to decreased binding of
`rapamycin and conferred a
`recessive resistance phenotype [47].
`
`HEAT Huntingtin, EF3, A subunit of PP2A and TOR, FAT FFlAP-
`ATM-TFIFIAP. FFIB FKBP12-rapamycin binding, CD catalytic
`domain. FATC FAT C-tenninus.
`
`Potential models for raparnyc-in inhibition of mTOR
`Small molecule kinase inhibitors act directly,
`regulating
`kinase activity generally by competition for ATP binding.
`However, whether
`the FKBP12-rapamycin complex or
`rapamycin alone directly inhibits the kinase activity of mTOR
`is still controversial. in vitro, rapamycin inhibited the modest
`increase in kinase activity of imrnunopreczipitated mTOR
`induced by insulin [48]. The FKBP12-rapamycin complex also
`inhibited the autokinase activity of mTOR, although a much
`higher concentration of rapamycin was needed in vitro than in
`trim to inhibit
`the activity of mTOR [49]. Conversely,
`treatment of cells with raparnycin did not alter
`the
`autophosphorylation level of Serm, and had little or no effect
`on the kinase activity of immunoprecipitated mTOR [37,49].
`More recently. an alternative model for mTOR function has
`been proposed. Specifically, mTOR may repress phosphatase
`activity associated with downstream targets. The inhibition of
`mTOR induced by bound FKBP12-rapamycin complex, may
`result
`in activation of
`this phosphatase, which then
`dephosphorylates downstream effector molecules such as
`p70S6K [500-,5!-]. Consistent with this model, we [52,
`Houghton 8: Huang, unpublished data] have suggested that
`mTOR regulates the catalytic subunit of PPZA associated with
`p44/42 mitogen-activated protein (MAP) kinases in some
`
`West-Ward Pharm.
`Exhibit 1017
`Page 003
`
`West-Ward Pharm.
`Exhibit 1017
`Page 003
`
`

`

`293 Current Opinion in lnvestigational Drugs 2002 Vol 3 No 2
`
`p44/42
`inhibits
`rapamyciri
`cells,
`these
`In
`cells.
`phosphorylation on Thrm following IGF-I
`stimulation.
`However, more studies are necessary to confirm the
`generality of this phosphatase model. An alternative model is
`for n1TOR to act as a scaffold and for the FKBP12-rapamycin
`complex to disrupt higher order mTOR-protein complexes.
`
`Rapamycin inhibition of mTOFt-controlled signaling
`nathways
`Although specific details of how rapamycin inhibits function of
`mTOR remain to be resolved, it has been widely accepted that
`inhibition of mTOR by rapamycin blocks growth factor
`stimulation of 40S ribosomal p7t}S6 kinase and phcsphorylation
`of 4E-Bl"'l (also designated PHAS-I). This results in a 15 to 20%
`inhibition of overall protein translation and arrests cell cycle
`progression in_ G1. Consistent with this observation, mTOR
`controls the synthesis of essential proteins involved in cell cycle
`progression (cyclin D1 and ornithinine decarboxylase) {53,54]
`
`[55]. A scheme of mTOR-controlled
`(c-Myc)
`and survival
`signaling pathways based on rapamycin effects is shown in
`Figure 3. 4E-BP1, the suppressor of elF-4E, has been reported to
`be a direct substrate for mTOR in cells [56,57]. In oilm, mTOR
`selectively phosphorylates 4E-BP1 at least at two and possibly
`four Ser/Thr residues (Thr", Thr“, 'I‘hr"’ and Set”) in the N-
`terminal region [58--,59]. Phosphorylation of 4E-BP1 appears to
`be an ordered process [53..,59,50]. Phosphorylation of Ser“
`depends
`on
`phosphorylation
`of
`all
`three Ser/Thr
`phosphorylation sites 159,60], whereas mutations of Thr”
`and/or Thr"‘ to Ala(s) prevents phosphorylation of Ser“ and
`Thrw, suggesting that ph0spl'|orylal'it)rt of Th!” and 'I‘hr"’ serves
`as a requisite ‘pr-in-ring‘ event [51I]. lt appears that mTOR also
`plays a critical role in regulating the phosphorylation of Serif
`and Thr”.
`In the presence of raparnycin. 4-IE-BP1 becomes
`hypophosphorylated and associates with elF-4E. This prevents
`formation of the elF-4F initiation complex and cap-dependent
`translation of mRNA.
`
`Figure 3. Rapamyclfl-lflhlbllbd 8l9f|8ll|'|9 pathways controlled by n1'l'OFt.
`
`FKB12-rapamycin
`
`4E—BP1
`
`(PHAS-I)
`
`Arrows represent activation. whereas bars represent inhibition. IGF-IFI type I Insulin-like growth factor, IRS1 insulin receptor substrate 1,
`Plait phosphatldvlinositoi 3-klnase. PTEN phosphatase and tensin tlomolog deleted on chromosome ten. PKBJAICI protein kinase B, FKB12-
`fapamyclrl FK-506-binding protein 12-rapamycin complex, rn'l'OR mammalian target oi rapamycln. alF-4E eukaryotic initiation taotor-4E,
`4E-BP1 (PHA3-I} elF~4E—bincIing protein 1. S6 40S ribosomal protein, prosaic p70S6 kinase.
`
`
`Ward Pharm.
`Exhibit 1017
`Page 004
`
`West-Ward Pharm.
`Exhibit 1017
`Page 004
`
`

`

`
`
`..*.JII_I|'.'F.-'E'l.'Ji:J'a|_iI_.l-!]|.J'>._."
`
`
`
`
`
`Inhibitors at mammalian target at rapamycin as novel antltumor agents: From bench to clinic Huang & Houghton 299
`
`Ribosomal p70S6K represents the other well characterized
`downstream target of mTOR. Two p70S6 kinases have been
`characterized, namely. p70S6K1 and p70S6l(2. The activation
`of both these kinases can be inhibited by rapamycin [61,62].
`rnTOR may directly or indirectly phosphorylate p7{lS6I(1 at
`Thrm or Thr” [50--,63,64,65I--,66o,67]. Phosphorylation of
`these two residues is blocked by rapamycin. Furthermore,
`mutation of either of these residues can abrogate the ability of
`rapamycin to inhibit p'70S6I( activation. p7l}S6l( functions to
`increase translation of 5' terminal oligopyrirnidine (STOP)
`tract
`IHRNAS, primarily Coding
`for
`elements of
`the
`translational machinery,
`such
`as
`ribosomal
`proteins,
`elongation factors, the poIy(A) binding protein [61] and [GF—I[
`[68]. Inhibition of mTOR by rapamycin thus selectively causes
`decreased translation of 5"IlOP-containing mRNAs.
`
`In addition to pathways controlling translation initiation,
`mTOR has been implicated in regulating the retinoblastoma
`protein {pRb), RNA polymerase {Pol} l/ll/III-transcription
`and translation of rRNA and ERNA, and phosphatases (PPZA,
`PP4, PP6) [69]. It seems that these pathways are cell type-
`dependent. For example, in vascular smooth muscle cells,
`rapamycin may act upstream of pRb to slow or arrest cell
`cycle transit ['70]. In this model, rapamycin inhibits activation
`of cyclin—dependent kinases
`{CDl(s), which results
`in
`hypophosphorylation of pRb protein, and inhibits cells
`progressing from C1 to S-phase [70].
`In T—lymphocytes,
`rapamycin induces G1 arrest, in part through inhibition of
`activation of CDK1 (p34'“"’) and the formation of the cyclin B-
`p33"” complex [71,72]. G1 arrest by rapamycin may also be
`due to prevention of the degradation of CDK inhibitory
`protein p27""" that occurs when cells are stimulated by growth
`factors [73,74]. This is further supported by the observation
`that p27""" deficient fibroblasts are somewhat resistant to
`rapamycin as determined by assaying for DNA synthesis [75].
`In NIHST3 cells, rapamycin inhibits the G1 to S transition in
`part through decrease of cyclin D1 mRNA level and protein
`stability ['76], or delay of the expression of cyclin A [77].
`
`Arititumor activity of rapamycin
`As previously mentioned, rapamycin has been approved as an
`irnmunosuppmiive drug for organ transplantation by the
`FDA. So far, rapamycin has been used clinically in organ
`transplantation with great success, particularly in kidney
`transplantation [78,79]. This is because rapamycin can inhibit T-
`cell activation and proliferation. Increasing evidence indicates
`that rapamycin is not only a potent immunosuppressant, but
`also a promising antilurnor agent. As reviewed above (Table 1),
`rapamycin potently inhibits the growth of many tumor cell
`lines in intro, and has demonstrated antiturnor activity in both
`xeno-graft and syngeneic murine tumor models.
`
`However, rather than acting as a cytostatic, rapamycin induces
`cell death under some conditions. Early data show that
`rapamycin induces prograrnrned cell death or apoptosis of B-
`cells [24,25]. Consistent with these findings, recent studies
`indicate that rapamycin alone can also induce apoptosis of
`certain rhabdomyosarcorna cells [14,15] and monocyte- and
`CD34-derived dendritic cells [80]. When combined with other
`chemotherapeutic
`agents
`in wire,
`rapamycin enhanced
`cisplatin-induced apoptosis in human small cell lung cancer cell
`lines
`[11],
`potentiated
`apoptosis
`of
`the murine T-
`lymphoblastoid cell line S49, induced by dexarnethasone [81]
`
`and augmented cisplatin- or camptothecin-induced cytotoxicity
`in DAOY human medulloblastoma cell lines [16]. Raparnycin
`produced
`additive
`cytotoxicity with 5-fluorouracil
`and
`cyclophosphamide in a Colon 38 tumor model [7]. At present,
`little is known about
`the molecular mechanism by which
`rapamycin induces apoptosis of tumor cells. However, Huang
`at al [15] have observed that the responses of malignant and
`normal cells to rapamycin are qualitatively different. When
`treated with rapamycin, cells with wild-type p53 arrest in G1
`phase and maintain viability. In contrast, when grown under
`autocrine conditions
`(ie, serum-free)
`in the presence of
`rapamycin, p53 mutant cells accumulate in G1 phase, but
`progress to S-phase and undergo apoptosis. More than 90% of
`apoptotic Rh:-10 cells {mutant p53 alleles, Argm—>Cysm) were
`BrdU-labeled, suggesting that
`the cells died after initiating
`replication. Thus, rapamycin-induced death appears to be a
`consequence of continued cell cycle progression, suggesting
`that p53 senses inhibition of mTOR and co-operates to reinforce
`a GI arrest. This model has been further tested using Rh30
`infected with adenovirus expressing wild-type p53 (Ad-p53)
`and p53 +/+ or p53 -/- murine embryo fibroblasts (MEFs) [15].
`Restoring the p53-mediated G1 checkpoint by Ad-p53 infection
`causes R1130 rhabdomyosarcorna cells to arrest
`in G1 and
`prevents rapamycin-induced apoptosis (Figure 4). Similarly,
`when exposed to rapamycin, p53 -/- MEF cells continue cell
`cycle progression, and become apoptotic, whereas p53 +/+
`MEF cells arrest in G1 and remain viable. Exactly why cells that
`fail to anest in G1 in the presence of rapamycin die is currently
`unknown.
`
`Mechanisms of resistance to rapamycin
`As shown by Dilling at al [8], under similar conditions of
`growth, various cell lines demonstrated several thousand-
`fold differences in sensitivity to rapamycin. The mechanism
`for this intrinsic resistance is under investigation. Cells may
`also acquire resistance either with or without rnutagenesis.
`
`Mutations in FKBP12, mTOR and p70S6K
`Budding
`yeast
`Saccltaramyces
`cerevisiae
`treated with
`rapamycin irreversibly arrested in the G1 phase. However,
`when yeast TOR1 and TOR2 were genetically mutated to
`TOR1-1 and TOR2-1, these strains were completely resistant
`to the growth-inhibitory effect of rapamycin. These resistant
`alleles encode proteins that have reduced affinity for
`binding the Fl<Bl’12—raparnycin complex [44]. Also in yeast,
`a recessive resistance phenotype was associated with
`decreased RBP1, a homolog of mammalian FKBP12, or a
`mutation altering Tyr”,
`leading to decreased binding of
`rapamycin [47]. In mammalian cells, resistance to rapamycin
`selected after mutagenesis
`is
`related to a dominant
`phenotype consistent with mutation in mTOR [45]. Similar
`to results in yeast, mTOR mutants are associated with
`decreased affinity for binding of the FKBP12-«rapamycin
`complex. High-level resistance to rapamycin is obtained
`when a mutant
`rnTOR (Ser””—)lle’°”), having reduced
`affinity for binding the FKBP12-rapamycin complex,
`is
`expressed [14,46]. mTOR is essential
`for activation of
`ribosornal
`p70S6I<1
`through
`phosphorylation
`of
`the
`rapamycin-sensitive
`sites
`at Thrm or Thr”
`[63,64].
`Substitution of either of these residues can also abrogate the
`ability of rapamycin to inhibit p70S6K activation. Whether
`this results in resistance to the growth inhibitory effect of
`rapamycin is less clear, and may be cell context-specific.
`
`Ward Pharm.
`Exhibit 1017
`Page 005
`
`West-Ward Pharm.
`Exhibit 1017
`Page 005
`
`

`

`300 Current Opinion in Investigations] Drugs 2002 Vol 3 No 2
`
`Figure 4. Protective effect at the tumor suppressor p53 on repernvclrl-induced epoptosis.
`
`8C
`(D
`
`Ad-Bgal + Flap 100
`
`32o 3t
`
`oE BE
`
`.3
`ED.
`ED.
`
`Ad-p63 + Rap 100
`
`100 101 102 103 104100
`
`to‘
`
`102
`
`103
`
`10‘
`
`Annexin V~FITC fluorescence
`
`Hhao rhabdomyosarooma oells were infected with either adenovirus expressing wild-type p53 (Ad-p53] or Ad-B-galactosidase (|3-gal} at a
`mumpficfly °l i"'°°“°" °' 1' M3’ 2‘? hi "‘°°“'-1m W33 fflnleoecl with serum-tree N2E. and cells were grown for a further 6 days in the absence
`0|’ Pffifieflce 0* 100 n91m|_f§Pam}‘0|n (Fiap 100}. Cells were harvested and apoptosis determined by quantitative FACs analysis [ApoAIert).
`Left panels show dual staining for propidium Iodide uptake and annexin V-FITC. Flight panels show corresponding distribution oi annexin V.
`FITC staining in populations oi cells. (Adapted from Huang 9; a;[-I5”
`
`Decrease of 4E-BP1 protein expression
`Mechanisms of acquired resistance (without use of mutagens)
`or intrinsic resistance have received less attention. Murine
`BC3I-I1 cells selected for acquired resistance demonstrated
`reduced levels of p27""", and consistent with this, embryo
`fibroblasts with disrupted p27'°"" are relatively resistant to
`rapamycin, as determined by inhibition of DNA precursors
`['75]. These data are consistent with G1 arrest, being in part
`due to rapamycin stabilizing this cyclin-dependent kinase
`inhibitor protein. Recently,
`rapamycin-resistant cell
`lines,
`R1130/Rapa1OK and C2, have also been obtained by growing
`
`Rh3C| parental rhabdomyosarcoma cells in the continuous
`presence of increasing concentrations of rapamycin, without
`prior mutagenesis [82, Huang 6: Houghtorb 1-ITIP‘-lblifihed
`data]. When resistant clones were grown without rapamycin
`for 6 or 10 weeks, they reverted to rapamycin sensitivity (in
`terms of ICE by growth inhibition assay). Thus, acquired
`rapamycin resistance was unstable.
`
`Analysis of several clones revealed increased levels of the c-
`Myc protein. Of interest, these rapamycin-resistant clones
`exhibited increased anchorage-independent growth in soft
`
`est-Ward Pharm.
`Exhibit 1017
`Page 006
`
`West-Ward Pharm.
`Exhibit 1017
`Page 006
`
`

`

`2-.
`
`Inhibitors of mammalian target at rapamycin as novel antltumor agents: From bench to clinic Huang 8. Houghton 301
`
`agar. Consistent with increased c-Myc, the levels of the 4E-
`BP suppressor proteins bound to elF—4E were significantly
`lower (approximately 10-fold), as were total cellular levels of
`4E-BP proteins. Steady state levels of £1-E-BP transcripts
`remained unaltered, however, the rate of synthesis appeared
`to be decreased in rapamycin-resistant clones [Huang &:
`Houghton, unpublished data]. In clones that reverted to
`rapamycin sensitivity, total levels of 4E-BP1 became similar
`to those in parental cells. In some cases, intrinsic resistance
`also appears to relate to low 4E-BI’:eIF4E levels. In colon
`carcinoma cells, very low levels of 4-E-BP were detected,
`whereas eIF4E levels were similar to those in sensitive
`
`tumor cell lines. In contrast, no significant changes were
`determined for p70S6 kinase levels or activity between
`parental and resistant clones.
`
`Development of CCl—779 and RAD-001 as
`antitumor agents
`CC!-779
`2,2—
`rapamycin-42,
`inhibitor-779;
`cycle
`(cell
`CCI-7'79
`of
`is
`an
`ester
`bis(hydroxymethyl)-propanoic
`acid)
`rapamycin (Figure 1}, which was developed by Wyeth-
`Ayerst as an antitumor agent. Like rapamycin, CCI-7'79
`acts
`by
`inhibition
`of mTOR,
`preventing
`the
`phosphorylation
`of 4E-BPs
`and p7(}S6I(
`[20-22,26].
`However, in contrast to rapamycin, CCI-779 is stable in
`aqueous
`solution, hence
`it
`can be
`formulated for
`intravenous administration. In preclinical
`tests, CCI-779
`possesses similar antitumor profiles to rapamycin [16,20-
`22,26,28o,290]. CCI-779
`potently inhibits growth
`of
`numerous cultured human tumor cell
`lines including
`human breast, prostate, pancreatic and small cell
`lung
`carcinomas, glioblastoma, medulloblastoma, melanoma,
`rhabdomyosarcoma and T-cell leukemia, with ICE values
`in the nanomolar range ['l6,20-22]. These observations have
`been further supported by the significant inhibitory effect
`of CCI-779 on growth of human tumor xenografts in
`athymic nude mice [16,20,2‘l,24]. When combined with
`cisplatin or carnptothecin, CCI-7'79
`showed additive
`cytotoxicity in subcutaneous implants of human brain
`tumors [16]. Interestingly,
`in viva CCI-7'79 also inhibited
`the growth of human U251 malignant gliorna cells that
`were resistant
`to rapamycin in oitro, although the
`mechanism is unknown [16].
`In addition, more recent
`results have
`revealed
`that
`the
`tumor
`suppressor
`phosphatase and tensin homolog deleted on chromosome
`ten (PTEN)-mutated or -deficient cancer cells are more
`sensitive to CCI-779 [21,28o,29t]. PTEN acts as a major
`negative regulator of the PI3K/Akt signaling pathway [83-
`85]. Loss of PTEN by deletion or mutation occurs in as
`many as 50% of all solid human tumors [86], resulting in
`activation of Akt. Conceptually, this could activate mTOR-
`dependent pathways, hence forming the basis for CCI-779
`hypersensitivity of PTEN deficient cells. However,
`it
`is
`unclear if Akt activates mTOR in situ, as mutation of
`putative Akt phosphorylation sites in the C-terminus of
`mTOR does not abrogate insulin stimulation of p7DS6K
`activation [87]. Of considerable interest, however, is the
`report by Podsypanina at at [280] showing inhibition of
`neoplastic transformation in PTEN +/- mice. These
`animals develop spontaneous multifocal complex atypical
`hyperplasia in the uterine secretory epithelium that
`progresses
`to neoplastic transformation. Tumor cells
`
`demonstrate elevated levels of phosphorylated Akt and
`activated p70S6K. While CCI-7'79 had no effect on Akt
`activation, as anticipated,
`it normalized p’70S6K activity.
`Whether,
`loss of PTEN function consistently sensitizes
`tumor
`cells
`to
`rapamycin analogs
`remains
`to be
`demonstrated.
`
`These preclinical results have revealed that CCI-7'79 exhibits
`impressive cytostatic, and in some instances, cytotoxic
`properties, and may be valuable to delay tumor progression
`and to improve survival when used alone, or in combination
`with other chemotherapeutic agents.
`
`Early data from phase I trials have shown promise of CC!-
`779 in treatment of some cancers [S80]. In European phase 1
`clinical trials [89], CCI-7'79 was administered as a weekly 30-
`min intravenous infusion in 18 patients with different types
`of advanced solid tumors. Doses ranged from 7.5 to 220
`mg/m’/week. After 2 8 weekly doses, significant tumor
`regressions were observed in two patients (receiving 15
`mg/tn’/week) with lung metastasis of renal cell carcinomas
`and in one patient (receiving 22.5 mg/ml/week} with a
`neuroendocrine tumor of
`the lung. Additionally,
`two
`patients experienced tumor stabilization.
`It
`is unclear
`whether the very high dose levels used in this study are
`necessary to elicit antitumor activity. Considering the high
`plasma levels of CCI-7'79 measured in patients (1000-fold
`greater than required for rapamycin activity in vitro), it is
`conceivable that CCI-7'79 is acting through a secondary
`mechanism independent of mTOR.
`
`In the US, phase I clinical trials were also conducted to
`determine the safety and tolerability of CCI-779 [90]. CC]-
`779 was administered as a daily intravenous 30-min infusion
`for 5 days every 2 weeks. Of 45 patients with various types
`of cancer, nine achieved some evidence of tumor response.
`Irnportantly, results from the above two trials indicate that
`CCI-7'79 is well tolerated in patients with only mild Side
`effects,
`such as acneform rash, mild mucositis,
`some
`thrombocytopenia, and elevated triglyceride and cholesterol
`levels. Rapamycin and its analogs potently inhibit T-cell
`activation,
`thus the potential
`for
`immunosuppression in
`patients treated with CCI-779 was anticipated. Interestingly
`CCI-779
`induced
`only
`modest
`evidence
`of
`immunosuppression. Rapamycins
`also
`inhibit
`insulin
`signaling, but in the clinical trials reported there was no
`toxicity consistent with inhibition of insulin signaling. CC]-
`779 is currently in phase III clinical trials. In athymic nude
`mouse xenografts, apart from breast and prostate cancers,
`human glioblastorna
`(U8'7MG)
`tumors and pancreatic
`carcinoma were also very sensitive to CCI-7'79. Thus, with
`an expanded focus,
`it is probable that clinical trials will
`identify other tumor types that are sensitive to CCI-779.
`
`RAD-001
`
`another
`is
`(40-O-(2-hydroxyethyl)-rapamycin)
`RAD-001
`rapamycin analog that is being developed as an antitumor
`agent. Like rapamycin and CC]-7'79, RAD-001 works by
`inhibition of mTOR, downregulating p70S6 kinase activity
`and decreasing the phosphorylation level of 4E-BP1 [27].
`Preliminary data indicate that in wire, RAD-001 potently
`inhibits growth of numerous human tumor cell lines, with
`50% inhibition of growth in the femtomolar range [27].
`
`- st-Ward Pharm.
`Exhibit 1017
`Page 007
`
`West-Ward Pharm.
`Exhibit 1017
`Page 007
`
`

`

`302 Current Opinion in lnvostlgational Drugs 2002 Vol 3 No 2
`
`Unlike CCI-779, RAD-D01 is a hydroxyethyl ether derivative
`of rapamycin, designed for oral administration.
`In viva
`experimental results reveal
`that RAD-001 inhibited the
`growth of human tumor xenografts in nude mice at doses
`ranging from 0.5 to 5.0 mg/kg/day. At these doses, RAD-
`001 was well tolerated, suggesting that it is a very promising
`chemotherapeutic agent. Currently, RAD-001 is entering
`phase I clinical trials for solid tumor.
`
`Summary
`in summary, inhibitors of mTOR, including rapamycin,
`CCI-7'79 and RAD-001, are promising novel anti

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