throbber
295
`
`BANDBIUI-HuY
`
`*To whom correspondence should be addressed
`
`Current Opinion in Investigational Drugs 2002 3(2):295-304
`© PharmaPressLtd ISSN 1472-4472
`
`Inhibitors of mammalian target of rapamycin as novel antitumor agents:
`From bench toclinic
`Shile Huang & Peter J Houghton”
`Address
`increased growth and malignant characteristics of cells. It is a
`Department of Molecular Pharmacology
`
`lipophilic—macrolide, that selectively inhibits =a
`
`
`
`St Jude Children's Research Hospital
`serine/theronine kinase, specifically, the mammalian targetof
`332 N Lauderdale
`rapamycin)
`(mTOR).
`mTOR
`lies
`downstream of
`Memphis
`TN 38105-2794
`phosphatidylinositol 3-kinase (PI3K) in the PI3K signaling
`USA
`pathway. Rapamycin was originally isolated as a fungicide
`Email: peter.houghton @stjude.org
`from the soil bacteria Streptomyces hygroscopicus, collected
`from Easter Island (known as Rapa Nui tothe natives) in the
`South Pacific in 1975 [1,2]. Structurally similar
`to the
`immunosuppresive reagent FK-506 (tacrolimus; 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 antitumoractivity in vitro and in vivo [5-7].
`However, perhaps because its mechanism of action was
`unknown at that time, rapamycin was not developed as a
`cancer therapeutic.
`
`Rapamycin and its derivatives, CCI-779 and RAD-001, inhibit the
`mammalian target
`of
`rapamycin
`(mTOR), downregulating
`translation of specific mRNAsrequired forcell cycle progression
`from G1 to S phase. Preclinically, mTOR inhibitors potently
`suppress growthandproliferation of numerous tumorcell lines in
`culture or when grownin mice as xenografts. CCI-779 and RAD-
`001 are being developed as antitumor drugs and are undergoing
`clinical
`trials. Clinically, CCI-779 has
`shown evidence of
`antitumor activity but
`induced relatively mild side effects
`in
`The potential for rapamycin as a cancer therapeutic was
`patients. Here we discuss potential antitumor mechanisms and
`refocused, in part, by studies of Dilling et al [8]. These were
`resistance mechanisms of mTOR inhibitors, and summarize the
`
`the to demonstrate potent and_selectivefirst studies
`
`current status of these compounds as novel antitumor agents.
`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
`rhabdomyosarcomacells is regulated by an autocrine loop
`involving secretion oftype II insulin-like growth factor (IGF-
`Il) and signaling through the type I IGF receptor [9,10].
`Indeed,
`the cell
`lines most sensitive to rapamycin were
`dependent on this autocrine pathway.
`
`i E
`
`FASAPIA
`
`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 duetoincreased 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 reasonsfor the relatively
`pooractivity 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 tumorcells and normalhealthy 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
`
`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 modeof its antitumor action is
`via the induction of apoptosis. Rapamycin potently inhibits
`proliferation
`or
`growth
`of
`cells
`derived
`from
`rhabdomyosarcoma,
`neuroblastoma,
`glioblastoma,
`small
`medulloblastoma,
`cell
`lung
`cancer
` [8,11-16],
`osteoscarcoma [17], pancreatic carcinoma[18,19], breast and
`prostate carcinoma[20-22], 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-779 (Wyeth-Ayerst Research;
`Figure 1) and RAD-001 (everolimus; Novartis AG; Figure 1),
`with improved pharmaceutical properties, but
`similar
`cellular effects to rapamycin [16,20-22,26,27,28e,29e], are
`currently undergoing antitumor phase III and phase I
`clinicaltrials, respectively. This review will discuss potential
`antitumor and resistance mechanisms of rapamycin andits
`derivatives, and summarize the preliminary data about
`these compounds, from the bench totheclinic.
`
`West-Ward Pharm.
`Exhibit 1017
`Page 001
`
`West-Ward Pharm.
`Exhibit 1017
`Page 001
`
`

`

`296 Current Opinion in Investigational Drugs 2002 Vol 3 No 2
`
`Table 1. Sensitivity of different tumorcell lines to rapamycin.
`Cell Line
`Rhabdomyosarcomacells
`Rh1 (10% serum)
`Rhi (serum free)
`Rh18
`Rh28
`Rh3o
`Colon carcinomacells
`GC/c1
`VRC,/c1
`CaCo
`HCT8
`HCT29
`HCT116
`Smail cell lung cancer cells
`H69
`H345
`H510
`Neuroblastoma cells
`NB-SD
`NS-1643
`NB-EB
`NB-1691
`NB-1382.2
`Glioblastomacells
`si-62
`SJ-G3
`Medullablastomacells
`DAOY
`Osteoblasts-like osteosarcoma cells
`ROS 17/2.8
`Pancreatic cancercells
`Panc-1
`MiaPaCa-2
`Leukemia cells
`RBL-2H3
`B-cell lymphoma cells
`BKS-2
`ge
`‘NFS1.1
`WEHI-279
`Thymomacells
`eid
`
`Rapamycin (IC
`(ng/ml)
`4680
`3.6
`0.1
`8.0
`0.37
`(ng/ml)
`9800
`1280
`1570
`8400
`> 10,000
`> 10,000
`(nM)
`~1
`~1
`~1
`(ng/ml)
`4
`~1
`2.9
`18
`639
`ng/ml
`UB
`> 10,000
`(ng/ml)
`~1
`(nM)
`< 100
`(ng/ml)
`1
`3
`(nM)
`~10
`(ng/ml)
`0.31
`pet
`0.60
`inom)
`
`Reference
`[8]
`
`[8]
`
`(11,12]
`
`[13]
`
`13
`im
`
`[16]
`
`[17]
`
`[18,19]
`
`[23]
`
`[25]
`
`[25]
`
`
`
`
`Antitumor mechanism ofinhibitors 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
`antitumoraction,ie, inhibiting mTOR, which, links mitogen
`stimulation to protein synthesis andcell cycle progression.
`mTOR
`antitumor mechanism of
`To better understand the
`rapamycin and its derivatives, we will briefly review the
`emerging cellular role of mTOR. mTORisreferred to by
`various other names, some of which are derived from its
`binding partner FK-506-binding protein, FKBP12 (discussed
`below). These names are FRAP (FKBP12 and rapamycin-
`associated protein), RAFT1 (rapamycin and FKBP12 target
`1), RAPT1 (rapamycin target 1) and SEP (sirolimus 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 kinases in
`eukaryotes.
`In the yeasts, Saccharomyces
`cerevisiae and
`Schizosaccharomyces 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 cerevisiae, and 56% identity with dTOR in
`overall sequence [39,40]. Human, mouse and rat mTOR
`proteins share 95% identity at the aminoacid level [40,41].
`mTORcontains a catalytic kinase domain and a FKBP12-
`rapamycin binding (FRB) domain near the C-terminus, and
`up to 20 tandemly repeated HEAT (Huntingtin, EF3, A
`subunit of PP2A 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 PlIK-related kinase family,
`which also includes MEC1, TEL1, RAD3, MEI-41, DNA-PK,
`ATM, ATR and TRRAP [42¢¢]. Both PI3K and potentially
`protein kinase B (PKB; Akt) lie upstream of mTOR, whereas
`ribosomal p70S6 kinase (p70S6K) and eukaryotic initiation
`factor-4E (eIF-4E) binding protein isoforms (4E-BP1-3) are the
`
`West-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 & Houghton 297
`
`
`
`rapamycin
`(Wyeth-Ayerst)
`
`ccl-779
`
`(Wyeth-Ayerst)
`
`
`RAD-001
`(Novartis)
`
`‘cH,
`
`H,C
`
`CH, H.C...
`
`y
`
`cH,
`
`
`
`Figure 1. Molecular structures of rapamycin, CCI-779 and RAD-001. °o
`FRB
`
`z:: J
`
`Po
`|
`
`ey
`=
`i]
`°P|
`
`BINDSBAMIPIED
`
`best characterized downstream mTOR effector molecules.
`Increasing evidence has implicated mTOR as a central
`controller of cell growth and proliferation. mTOR may
`directly or indirectly regulate translation initiation, actin
`organization, membranetraffic and protein degradation,
`protein kinase C signaling, ribosome biogenesis and tRNA
`synthesis, as well as transcription [42ee]. 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
`tumorcells, which is clearly a consequence of binding
`mTOR. Whetherthis action is a consequence ofinhibiting
`mTORkinase activity per seis less clear. Rapamycin 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 vitro and
`mTOR autophosphorylation. However,
`the specificity of
`rapamycin action can be demonstrated in vivo as certain
`mutations in the FRB domain of mTOR affect FKBP12-
`rapamycin binding, and significantly reduce the cellular
`sensitivity of
`rapamycin. The first
`rapamycin-resistant
`alleles, TORI-1 and TOR2-1, identified in a Saccharomyces
`cerevisiae genetic screen were shown to confer dominant
`resistance. These mutant TORproteins lost the ability for
`FKBP-rapamycin
`complex
`binding
`[44].
`Similarly,
`mammalian
`cells
`also
`became
`highly
`resistant
`to
`rapamycin when a mutation (Ser“”lle"”) occurred in the
`FRB domain of mTOR, whichresulted in decreased affinity
`for binding of FKBP12-rapamycin complex [14,45,46]. In
`the yeast, Saccharomyces 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].
`
`
`Figure 2. Schematic representation of mTOR domains.
`
`[eat|]vem||a
`
`HEAT repeats
`
`ON
`
`HEAT Huntingtin, EF3, A subunit of PP2A and TOR, FAT FRAP-
`ATM-TRRAP, FRB FKBP12-rapamycin binding, CD catalytic
`domain, FATC FAT C-terminus.
`
`Potential models for rapamycin 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 alonedirectly inhibits the kinase activity of mTOR
`is still controversial. In vitro, rapamycin inhibited the modest
`increase in kinase activity of
`immunoprecipitated mTOR
`inducedbyinsulin [48]. The FKBP12-rapamycin complex also
`inhibited the autokinase activity of mTOR, although a much
`higher concentration of rapamycin was needed invitro than in
`vivo to inhibit
`the activity of mTOR [49]. Conversely,
`treatment of cells with rapamycin did not alter
`the
`autophosphorylationlevel of Ser, and hadlittle or noeffect
`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 [50ee,51e]. Consistent with this model, we [52,
`Houghton & Huang, unpublished data] have suggested that
`mTORregulates the catalytic subunit of PP2A 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
`
`

`

`298 Current Opinion in Investigational Drugs 2002 Vol 3 No 2
`
`p44/42
`inhibits
`rapamycin
`cells,
`these
`In
`cells.
`phosphorylation on Thr™ following IGF-I
`stimulation.
`However, more studies
`are necessary to confirm the
`generality of this phosphatase model. An alternative modelis
`for mTORto act as a scaffold and for the FKBP12-rapamycin
`complex to disrupt higher order mTOR-protein complexes.
`
`Rapamycin inhibition of mTOR-controlled signaling
`pathways
`Althoughspecific details of how rapamycin inhibits function of
`mTORremain tobe resolved,it has been widely accepted that
`inhibition of mTOR by rapamycin blocks growth factor
`stimulationof 40S ribosomal p70S6 kinase and phosphorylation
`of 4E-BP1 (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 suppressorof eIF-4E, has been reported to
`be a direct substrate for mTORin cells [56,57]. In vitro, mTOR
`selectively phosphorylates 4E-BP1 at least at two and possibly
`four Ser/Thr residues (Thr”, Thr“, Thr” and Ser") in the N-
`terminal region [58ee,59]. Phosphorylation of 4E-BP1 appears to
`be an ordered process [58¢e,59,60]. Phosphorylation of Ser
`depends
`on
`phosphorylation
`of
`all
`three Ser/Thr
`phosphorylation sites
`[59,60], whereas mutations of Thr”
`and/or Thr” to Ala(s) prevents phosphorylation of Ser” and
`Thr”, suggesting that phosphorylation of Thr” and Thr”serves
`as a requisite ‘priming’ event[51e]. It appears that mTORalso
`plays a critical role in regulating the phosphorylation of Ser”
`and Thr”.
`In the presence of rapamycin, 4E-BP] becomes
`hypophosphorylated and associates with elF-4E. This prevents
`formation of the elF-4F initiation complex and cap-dependent
`translation of mRNA.
`
`
`Figure 3. Rapamycin-inhibited signaling pathways controlled by mTOR.
`
`
`
`
`IGF-IR
`
`FKB12-rapamycin
`
`
` [ere|[exert_|NR |
`Phosphatase '—Cimtor
`
`A
`(PHAS-1)
`
`4E-BP'1
`
`elF-4E
`
`
`
`insulin-like growth factor, IRS1 insulin receptor substrate 1,
`Arrows representactivation, whereas bars representinhibition. IGF-IR type |
`PI3K phosphatidylinositol 3-kinase, PTEN phosphatase and tensin homolog deleted on chromosometen, PKB/Akt protein kinase B, FKB12-
`rapamycin FK-506-binding protein 12-rapamycin complex, mTOR mammalian target of rapamycin, elF-4E eukaryotic initiation factor-4E,
`4E-BP1 (PHAS-1) elF-4E-binding protein 1, S6 40S ribosomal protein, p70S6K p70S6kinase.
`
`West-Ward Pharm.
`Exhibit 1017
`Page 004
`
`West-Ward Pharm.
`Exhibit 1017
`Page 004
`
`

`

`eagogjul-juy
`
`
`
`__geinoseAcipreg
`
`<Tt
`
`=o
`
`Inhibitors of mammalian target of rapamycin as novel antitumor agents: From bench to clinic Huang & Houghton 299
`
`Ribosomal p70S6K represents the other well characterized
`downstream target of mTOR. Twop70S6 kinases have been
`characterized, namely, p70S6K1 and p70S6K2. The activation
`of both these kinases can be inhibited by rapamycin [61,62].
`mTOR maydirectly or indirectly phosphorylate p70S6K1 at
`Thr” or Thr” [50¢*,63,64,65¢e,66¢,67]. Phosphorylation of
`these two residues is blocked by rapamycin. Furthermore,
`mutation ofeither of these residues can abrogatethe ability of
`rapamycin to inhibit p70S6K activation. p70S6K functions to
`increase translation of 5'
`terminal oligopyrimidine (5'TOP)
`tract mRNAs, primarily coding for
`elements of
`the
`translational machinery,
`such
`as_
`ribosomal
`proteins,
`elongation factors, the poly(A) binding protein [61] and IGF-II
`[68]. Inhibition of mTOR by rapamycin thusselectively causes
`decreased translation of 5'TOP-containing mRNAs.
`
`In addition to pathways controlling translation initiation,
`mTORhas been implicated in regulating the retinoblastoma
`protein (pRb), RNA polymerase (Pol) 1/II/III-transcription
`andtranslation of rRNA and tRNA, and phosphatases (PP2A,
`PP4, PP6) [69]. It seems that these pathwaysare cell type-
`dependent. For example,
`in vascular smooth musclecells,
`rapamycin may act upstream of pRb to slow or arrest cell
`cycle transit [70]. In this model, rapamycininhibits activation
`of cyclin-dependent kinases
`(CDKs), which results
`in
`hypophosphorylation of pRb protein, and inhibits cells
`progressing from G1 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 E-
`p33“ complex [71,72]. G1 arrest by rapamycin may also be
`due to prevention of the degradation of CDK inhibitory
`protein p27""' that occurs whencells 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 NIH3T3cells, rapamycin inhibits the G1 to S transition in
`part through decrease of cyclin D1 mRNAlevel and protein
`stability [76], or delay of the expression of cyclin A [77].
`
`Antitumoractivity of rapamycin
`As previously mentioned, rapamycin has been approved as an
`immunosuppressive 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 caninhibit T-
`cell activation and proliferation. Increasing evidence indicates
`that rapamycin is not only a potent immunosuppressant, but
`also a promising antitumor agent. As reviewed above(Table 1),
`rapamycin potently inhibits the growth of many tumorcell
`lines in vitro, and has demonstrated antitumoractivity in both
`xenograft 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 programmed 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 rhabdomyosarcoma cells [14,15], and monocyte- and
`CD34-derived dendritic cells [80]. When combined with other
`chemotherapeutic
`agents
`in
`vitro,
`rapamycin enhanced
`cisplatin-induced apoptosis in human small cell lung cancercell
`lines
`[11],
`potentiated
`apoptosis
`of
`the murine T-
`lymphoblastoid cell line S49, induced by dexamethasone [81]
`
`and augmented cisplatin- or camptothecin-induced cytotoxicity
`in DAOY human medulloblastoma cell lines [16]. Rapamycin
`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 tumorcells. However, Huang
`et 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 Gl
`phase and maintain viability. In contrast, when grown under
`autocrine conditions
`(ie,
`serum-free)
`in the presence of
`rapamycin, p53 mutant cells accumulate in Gl phase, but
`progress to S-phase and undergo apoptosis. More than 90% of
`apoptotic Rh30 cells (mutant p53 alleles, Arg”Cys”) 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 toreinforce
`a Gl arrest. This model has been further tested using Rh30
`infected with adenovirus expressing wild-type p53 (Ad-p53)
`and p53 +/+ or p53 -/- murine embryofibroblasts (MEFs) [15].
`Restoring the p53-mediated G1 checkpoint by Ad-p53 infection
`causes Rh30 rhabdomyosarcoma cells to arrest
`in Gl and
`prevents rapamycin-induced apoptosis (Figure 4). Similarly,
`when exposed to rapamycin, p53 -/- MEF cells continuecell
`cycle progression, and become apoptotic, whereas p53 +/+
`MEFcells arrest in G1 and remainviable. Exactly whycells that
`fail to arrest in G1 in the presence of rapamycin dieis currently
`unknown.
`
`Mechanismsof resistance to rapamycin
`As shown by Dilling ef al [8], under similar conditions of
`growth, variouscell lines demonstrated several thousand-
`fold differences in sensitivity to rapamycin. The mechanism
`for this intrinsic resistance is under investigation. Cells may
`alsoacquire resistance either with or without mutagenesis.
`
`Mutations in FKBP12, mTOR and p70S6K
`Budding
`yeast
`Saccharomyces
`cerevisiae
`treated with
`rapamycin irreversibly arrested in the G1 phase. However,
`when yeast TORI 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 FKBP12-rapamycin 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 mTOR (Ser™’->lle’), having reduced
`affinity for binding the FKBP12-rapamycin complex,
`is
`expressed [14,46]. mTOR is essential
`for activation of
`ribosomal
`p70S6K1
`through
`phosphorylation
`of
`the
`rapamycin-sensitive
`sites
`at Thr” 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
`rapamycinis less clear, and maybe cell context-specific.
`
`West-Ward Pharm.
`Exhibit 1017
`Page 005
`
`West-Ward Pharm.
`Exhibit 1017
`Page 005
`
`

`

`102 ananse
`
`300 Current Opinion in Investigational Drugs 2002 Vol 3 No 2
`
`Figure 4. Protective effect of the tumor suppressor p53 on rapamycin-induced apoptosis.
`
`@Oo
`
`c 8a2o=—o2G2E>n
`
`Control
`
`Rap 100
`
`Ad-Bgal
`
`
`
`Ad-Bgal + Rap 100
`
`Ad-p63
`
`Ad-p63 + Rap 100
`
`0°
`10° 101 102 103 104100
`
`101
`
`103
`
`104
`
`°a
`2oO
`
`Annexin V-FITC fluorescence
`
`Rh30 rhabdomyosarcoma cells were infected with either adenovirus expressing wild-type p53 (Ad-p53) or Ad-f-galactosidase ([}-gal) at a
`multiplicity of infection of 1. After 24 h, medium wasreplaced with serum-free N2E, andcells were grownfor a further 6 daysin the absence
`or presence of 100 ng/ml rapamycin (Rap 100). Cells were harvested and apoptosis determined by quantitative FACs analysis (ApoAlert).
`Left panels show dualstaining for propidium iodide uptake and annexin V-FITC. Right panels show corresponding distribution of annexin V-
`FITC staining in populationsofcells. (Adapted from Huangetal [15].)
`
`
`Decrease of 4E-BP1 protein expression
`Mechanisms of acquired resistance (withoutuse of mutagens)
`or intrinsic resistance have received less attention. Murine
`BC3H1 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,
`Rh30/Rapal0K and C2, have also been obtained by growing
`
`rhabdomyosarcoma cells in the continuous
`Rh30 parental
`presence of increasing concentrations of rapamycin, without
`prior mutagenesis [82, Huang & Houghton, unpublished
`data]. Whenresistant clones were grown without rapamycin
`for 6 or 10 weeks, they reverted to rapamycin sensitivity (in
`terms of IC,, 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
`
`West-Ward Pharm.
`Exhibit 1017
`Page 006
`
`West-Ward Pharm.
`Exhibit 1017
`Page 006
`
`

`

`Inhibitors of mammalian target of rapamycin as novel antitumor agents: From bench to clinic Huang & Houghton 301
`
`agar. Consistent with increased c-Myc,the levels of the 4E-
`BP suppressor proteins bound to eIF-4E were significantly
`lower (approximately 10-fold), as were total cellular levels of
`4E-BP proteins. Steady state levels of 4E-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-BP:eIF4E levels. In colon
`carcinoma cells, very low levels of 4E-BP were detected,
`whereas eIF4E levels were similar to those in sensitive
`
`lines. In contrast, no significant changes were
`tumor cell
`determined for p7056 kinase levels or activity between
`parental andresistant clones.
`
`demonstrate elevated levels of phosphorylated Akt and
`activated p70S6K. While CCI-779 had noeffect on Akt
`activation, as anticipated, it normalized p7OS6K activity.
`Whether,
`loss of PTEN function consistently sensitizes
`tumor
`cells
`to
`rapamycin
`analogs
`remains
`to be
`demonstrated.
`
`Thesepreclinical results have revealed that CCI-779 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.
`
`
`
`BANOOIUI-QUY
`
`Early data from phase I trials have shown promise of CCI-
`779 in treatment of some cancers [88]. In European phase|
`clinical trials [89], CCI-779 was administered as a weekly 30-
`min intravenousinfusion in 18 patients with different types
`of advanced solid tumors. Doses ranged from 7.5 to 220
`mg/m’/week. After > 8 weekly doses, significant
`tumor
`regressions were observed in twopatients (receiving 15
`mg/m’ /week) with lung metastasis of renal cell carcinomas
`and in one patient (receiving 22.5 mg/m’/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
`necessarytoelicit antitumoractivity. Considering the high
`plasma levels of CCI-779 measured in patients (1000-fold
`greater than required for rapamycin activity in vitro), it is
`conceivable that CCI-779 is acting through a secondary
`mechanism independent of mTOR.
`
`Development of CCI-779 and RAD-001 as
`antitumor agents
`CCI-779
`2,2-
`rapamycin-42,
`inhibitor-779;
`cycle
`(cell
`CCI-779
`of
`is
`an
`ester
`bis(hydroxymethyl)-propanoic
`acid)
`rapamycin (Figure 1), which was developed by Wyeth-
`Ayerst as an antitumor agent. Like rapamycin, CCI-779
`acts
`by
`inhibition
`of mTOR,
`preventing
`the
`phosphorylation
`of
`4E-BPs
`and
`p70S6K [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-
`22,26,28¢,29e]. CCI-779
`potently
`inhibits growth
`of
`numerous cultured human tumor cell
`lines including
`trials were also conducted to
`In the US, phase I clinical
`human breast, prostate, pancreatic and small cell
`lung
`determine the safety and tolerability of CCI-779 [90]. CCI-
`carcinomas, glioblastoma, medulloblastoma, melanoma,
`779 was administeredasa daily intravenous 30-min infusion
`rhabdomyosarcoma and T-cell leukemia, with IC,, values
`for 5 days every 2 weeks. Of45 patients with various types
`in the nanomolar range [16,20-22]. These observations have
`of cancer, nine achieved some evidence of tumor response.
`been further supported by the significant inhibitory effect
`Importantly, results from the above twotrials indicate that
`of CCI-779 on growth of human tumor xenografts in
`CCI-779 is well
`tolerated in patients with only mild side
`athymic nude mice [16,20,21,24]. When combined with
`effects,
`such as acneform rash, mild mucositis,
`some
`cisplatin or camptothecin, CCI-779 showed additive
`thrombocytopenia, and elevated triglyceride and cholesterol
`cytotoxicity in subcutaneous implants of human brain
`levels. Rapamycin and its analogs potently inhibit T-cell
`tumors [16]. Interestingly,
`in vivo CCI-779 also inhibited
`the growth of human U251 malignant glioma cells that
`activation,
`thus the potential
`for
`immunosuppression in
`patients treated with CCI-779 wasanticipated. Interestingly
`were
`resistant
`to rapamycin in
`vitro, although the
`
`CCI-779~—induced evidence_ofonly modest
`
`
`mechanism is unknown [16].
`In addition, more recent
`results
`have
`revealed
`that
`the
`tumor
`suppressor
`immunosuppression. Rapamycins
`also
`inhibit
`insulin
`phosphatase and tensin homolog deleted on chromosome
`signaling, but in the clinical
`trials reported there was no
`ten (PTEN)-mutated or -deficient cancer cells are more
`toxicity consistent with inhibition of insulin signaling. CCI-
`779 is currently in phaseIII clinical trials. In athymic nude
`sensitive to CCI-779 (21 280,29]. PTEN acts as a major
`negative regulator of the PI3K/Akt signaling pathway [83-
`mouse xenografts, apart from breast and prostate cancers,
`85]. Loss of PTEN by deletion or mutation occurs in as
`human glioblastoma
`(U87MG)
`tumors
`and_ pancreatic
`many as 50%of all solid human tumors [86], resulting in
`carcinoma were also very sensitive to CCI-779. Thus, with
`activation of Akt. Conceptually, this could activate mTOR-
`an expanded focus,
`it is probable that clinical
`trials will
`dependent pathways, hence forming the basis for CCI-779
`identify other tumortypesthatare sensitive to 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 p70S6K
`activation [87]. Of considerable interest, however,
`is the
`report by Podsypaninaet al [28e] 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
`
`RAD-001
`another
`is
`(40-O-(2-hydroxyethyl)-rapamycin)
`RAD-001
`rapamycin analog that is being developed as an antitumor
`agent. Like rapamycin and CCI-779, 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 vitro, RAD-001 potently
`inhibits growth of numerous human tumorcell lines, with
`50% inhibition of growth in the femtomolar range [27].
`
`
`
`West-Ward Pharm.
`Exhibit 1017
`Page 007
`
`West-Ward Pharm.
`Exhibit 1017
`Page 007
`
`

`

`302 Current Opinion in Investigational Drugs 2002 Vol 3 No 2
`
`Unlike CCI-779, RAD-001 is a hydroxyethyl ether derivative
`of
`rapamycin, designed for oral administration.
`In vivo
`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, R

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