throbber
295
`
`Inhibitors of mammalian target of rapamycin as novel antitumor agents:
`From bench to clinic
`Shile Huang & Peter J Houghton*
`
`Address
`Department of Molecular Pharmacology
`St Jude Children's Research Hospital
`332 N Lauderdale
`Memphis
`TN 38105-2794
`USA
`Email: peter.houghton@stjude.org
`
`*To whom correspondence should be addressed
`
`Current Opinion in lnvestigational Drugs 2002 3(2):295-304
`© PharmaPress Ltd ISSN 1472-4472
`
`Rapamycin and its derivatives, CCl-779 and RAD-001, inhibit the
`mammalian
`target of rapamycin
`(mTOR), downregulating
`translation of specific mRNAs required for cell cycle progression
`from GI 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. CCl-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
`patients. Here we discuss potential antitumor mechanisms and
`resistance mechanisms 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, STl-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.
`
`Rapamune; Wyeth-Ayerst
`(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 mammalian .target Qf
`rapamycin
`(mTOR). mTOR
`lies
`downstream
`of
`phosphatidylinositol 3-kinase (P13K) in the Pl3K signaling
`pathway. Rapamycin was originally isolated as a fungicide
`from the soil bacteria Streptomyces hygroscopicus, collected
`from Easter Island (known as Rapa Nui to the 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 antitumor activity 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.
`
`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 (IGF(cid:173)
`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.
`
`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
`Gl 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 [20-22], 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 [16,20-22,26,27,28•,29•], are
`currently undergoing antitumor phase l1I 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.
`
`ARGENTUM
`Exhibit 1017
`
`000001
`
`

`

`296 Current Opinion in lnvestigational Drugs 2002 Vol 3 No 2
`
`Table 1. Sensitivity of different tumor cell lines to rapamycin.
`Cell Line
`Rhabdomyosarcoma cells
`Rh1 (10% serum)
`Rh1 (serum free)
`Rh18
`Rh28
`Rh30
`Colon carcinoma cells
`GCjc1
`VRC/c1
`Ca Co
`HCTS
`HCT29
`HCT11 6
`Small cell lung cancer cells
`H69
`H345
`H510
`Neuroblastoma cells
`NB-SD
`NS-1643
`NB-EB
`NB-1691
`NB-1382.2
`Glioblastoma cells
`SJ-G2
`SJ-G3
`Medul/ablastoma cells
`DAOY
`Osteoblasts-like osteosarcoma cells
`ROS 17/2.8
`Pancreatic cancer cells
`Panc-1
`MiaPaCa-2
`Leukemia cells
`RBL·2H3
`B-cell lymphoma cells
`BKS-2
`L1 .2
`' NFS1 .1
`WEHl-279
`Thymoma cells
`EL4
`
`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)
`- 1
`- 1
`2.9
`18
`639
`(ng/ml)
`0.5
`> 10,000
`(ng/ml)
`- 1
`(nM)
`< 100
`(ng/ml)
`1
`3
`(nM)
`- 10
`(ng/ml)
`0.31
`0.27
`0.31
`0.60
`(ng/ml)
`0.17
`
`Reference
`[8]
`
`[8]
`
`[11 , 12]
`
`(13]
`
`(13]
`
`[16)
`
`(17]
`
`[18,19)
`
`[23]
`
`(25)
`
`[25)
`
`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
`anti tumor action, ie, inhibiting mTOR, which, links mitogen
`stimulation to protein synthesis and cell cycle progression.
`
`mTOR
`the antitumor mechanism of
`To better understand
`rapamycin and its derivatives, we will briefly review the
`emerging cellular role of mTOR. mTOR is referred to by
`various other names, some of w hich are derived from its
`binding partner FK-506-bind ing protein, FKBP12 (discussed
`below). These names are FRAP (FKBP12 and rapamycin(cid:173)
`associated protein), RAFTl (rapamycin and FKBP12 target
`1), RAPTl (rapamycin target 1) and SEP (sirolimus effector
`protein). In the mid-1990s, mTOR was identified as a
`mammalian serine/ threonine kinase of approximately 289
`in humans, mice and ra ts [30-33]. TOR proteins
`kDa
`represent a class of evolutionarily conserved kinases in
`the yeasts, Saccharomyces cerevisiae and
`eukaryotes. In
`Schizosaccharomyces pombe, two TOR genes, TORl 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 cha racterized, sharing 38% identity with
`TOR2 from Saccharomyces cerevisiae [37,38]. mTOR shares
`approximately 45% identity with TORl 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 amino acid level [40,41].
`mTOR contains a catalytic kinase domain and a FKBP12-
`rapa mycin 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 ca talytic domain of Pl3K, mTOR is
`considered a member of the PIK-related kinase fa mily,
`which also includes MECl, TELl, RAD3, MEl-41, DNA-PK,
`ATM, ATR and TRRAP [42••]. Both PI3K and potentially
`protein kinase B (PKB; Akt) lie upstream of mTOR, w hereas
`ribosomal p70S6 kinase (p70S6K) and eukaryotic initiation
`factor-4E (eIF-4E) binding protein isoforms (4E-BP1-3) are the
`
`000002
`
`

`

`Inhibitors of mammalian target of rapamycin as novel antitumor agents: From bench to clinic Huang & Houghton 297
`
`Figure 1. Molecular structures of rapamycin, CCl-779 and RAD-001 .
`
`(J ,, H OH
`
`' y"'y
`
`0
`
`OH
`
`0
`
`~0.--Y-OH
`y ' CH3
`
`0
`
`O~OH
`
`Qyo
`
`0
`
`0
`
`0
`
`CH3
`
`rapamycin
`(Wyeth·Ayerst)
`
`CCl-779
`(Wyeth·Ayerst)
`
`RAD·001
`(Novartis)
`
`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, membrane traffic and protein degradation,
`protein kinase C signaling, ribosome biogenesis and tRNA
`synthesis, as well as transcription [42•• ]. 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. 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
`inhibits
`function of mTOR. High
`(Figure 2), and
`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, TOR1 -1 and TOR2 -1, identified in a Saccharomyces
`cerevisiae 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
`.
`l
`.
`(S
`II 2005)
`d .
`th
`rapamycm w 1en a mutat10n
`er
`-? e
`occurre m
`e
`2035
`FRB domain of mTOR, which resulted in decreased affinity
`for binding of FKBP12-rapamycin complex [14,45,46]. In
`the yeast, Saccharomyces cerevisiae, decreased RBPl, a
`
`homolog of mammalian FKBP12, or mutation at Tyr8' , led
`to decreased binding of rapamycin and conferred a
`recessive resistance phenotype [47].
`
`Figure 2. Schematic representation of mTOR domains.
`
`2549 aa
`
`11 HEAT 11 HEAT
`
`HEAT repeals
`
`FRB
`
`FATC
`
`HEAT Huntingtin, EF3, A subunit of PP2A and TOR, FAT FRAP(cid:173)
`ATM-TRRAP, FRB FKBP12-rapamycin binding, CD catalytic
`domain, FATC FAT C-terminus.
`
`Potential models fQr 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 alone directly inhibits the kinase activity of mTOR
`is still controversial. In vitro, rapamycin inhibited the modest
`increase in kinase activity of immunoprecipitated mTOR
`induced by insulin [48]. The FKBP12-rapamycin complex also
`inhibited the autokinase activity of mTOR, aliliough a much
`higher concentration of rapamycin was needed in vitro ilian in
`vivo
`to
`inhibit the activity of mTOR [49]. Conversely,
`treatment of cells with
`rapamycin did not alter
`the
`
`autophosphorylation level of Ser2'"1, 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 [50 .. ,51•]. Consistent with
`this model, we [52,
`Houghton & Huang, unpublished data] have suggested that
`mTOR regulates the catalytic subunit of PP2A associated with
`p44/42 mitogen-activated protein (MAP) kinases in some
`
`000003
`
`

`

`298 Current Opinion in lnvestigational Drugs 2002 Vol 3 No 2
`
`p44/ 42
`inhibits
`rapamycin
`cells,
`these
`In
`cells.
`phosphorylation on Thr202
`IGF-1 stimulation.
`following
`to confirm
`the
`However, more studies are necessary
`general ity of this phosphatase model. An alterna tive model is
`for mTOR to act as a scaffold and for the FKBP12-rapamycin
`complex to disrupt higher order mTOR-protein complexes.
`
`Rapamycin inhibition of mTOR-control/ed signaling
`pathways
`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 p70S6 kinase and phosphorylation
`of 4E-BP1 (also designated PHAS-l). This results in a 15 to 20%
`inhibition of overall protein translation and arrests cell cycle
`progression in Gl. Consistent with this observation, mTOR
`controls the synthesis of essential proteins involved in cell cycle
`progression (cyclin Dl and ornithinine decarboxylase) [53,54]
`
`and survival (c-Myc) [55] . A scheme of mTOR-controlled
`signaling pathways based on rapamycin effects is shown in
`Figure 3. 4E-BP1, the suppressor of e!F-4E, has been reported to
`be a direct substrate for mTOR in cells [56,57]. /11 vil.ro, mTOR
`selectively phosphorylates 4E-BP1 at least at two and possibly
`four Ser /Thr residues (Thr37
`, Thr'w', Thr"' and Ser"') in the N(cid:173)
`termina] region [58••,59]. Phosphorylation of 4E-BP1 appears to
`be an ordered process [58••,59,60]. Phosphorylation of Ser"'
`depends on phosphorylation
`of
`all
`three Ser /Thr
`phosphorylation sites [59,60], whereas mutations of Thr37
`and/or Thr46 to Ala(s) prevents phosphorylation of Ser65 and
`Thr"', suggesting that phosphorylation of Thr37 and Thr46 serves
`as a requisite 'priming' event [51•]. It appears that mTOR also
`plays a critical role in regulating the phosphorylation of Ser'"
`and Thr"'. In the presence of rapamycin, 4E-BP1 becomes
`hypophosphorylated and associates with e!F-4E. This prevents
`formation of the eIF-4F initiation complex and cap-dependent
`translation of mRNA.
`
`Figure 3. Rapamycin-inhibited signaling pathways controlled by mTOR.
`
`===Hi=====
`
`IGF-IR
`
`Pl3K
`
`FKB 12-rapamycin
`
`§J--l _P_KB_IA_kt---1~ l
`I Phosphatase I t--8
`/ "Yr~-
`l
`
`1 p70S6
`86 I
`
`4E-BP1
`(PHAS-1)
`
`elF-4E
`
`I
`
`Arrows represent acti~ation , .whereas bars represent inhibition. IGF-IR type I insulin-like growth factor, IRS1 insulin receptor substrate 1,
`P13K phosphat1dyllnos1tol 3-kmase, PTEN phosphatase and tensin homolog deleted on chromosome ten, 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, SG 40S ribosomal protein, p70S6K p70S6 kinase.
`
`000004
`
`

`

`Inhibitors of mammalian target of rapamycin as novel antitumor agents: From bench to clinic Huang & Houghton 299
`
`Ribosomal p7056K represents the other well characterized
`downstream target of mTOR. Two p7056 kinases have been
`characterized, namely, p7056Kl and p7056K.2. The activation
`of both these kinases can be inhibited by rapamycin [61,62].
`mTOR may directly or indirectly phosphorylate p7056Kl at
`Thr2~J or Thr:ll«' [S0 .. ,63,64,6S .. ,66•,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 p7056K activation. p7056K functions to
`increase translation of S' terminal oligopyrimidine (STOP)
`for elements of
`the
`tract mRNAs, primarily coding
`translational machinery,
`such as
`ribosomal proteins,
`elongation factors, the poly(A) binding protein [61] and JGF-II
`[68]. Inhibition of mTOR by rapamycin thus selectively causes
`decreased translation of STOP-containing mRNAs.
`
`In addition to pathways controlling translation initiation,
`mTOR has been implicated in regulating the retinoblastoma
`protein (pRb), RNA polymerase (Pol) l/II/llI-transcription
`and translation of rRNA and tRNA, and phosphatases (PP2A,
`PP4, PP6) [69]. It seems that these pathways are cell type(cid:173)
`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
`(CDKs), which results
`in
`hypophosphorylation of pRb protein, and
`inhibits cells
`progressing from Gl to S-phase [70]. ln T-lymphocytes,
`rapamycin induces Gl arrest, in part through inhibition of
`
`activation of CDKl (p34''k2) and the formation of the cyclin E(cid:173)
`p33"'k2 complex [71,72] . Gl arrest by rapamycin may also be
`due to prevention of the degradation of CDK inhibitory
`protein p27K'P' that occurs when cells are stimulated by growth
`factors [73,74]. This is further supported by the observation
`that p27K'P' deficient fibroblasts are somewhat resistant to
`rapamycin as determined by assaying for DNA synthesis [7S] .
`In NIH3T3 cells, rapamycin inhibits the Gl to S transition in
`part through decrease of cyclin Dl mRNA level and protein
`stability [76], or delay of the expression of cyclin A [77].
`
`Antitumor activity 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 can inhibit T(cid:173)
`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 tumor cell
`lines in vitro, and has demonstrated antitumor activity 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(cid:173)
`cells [24,2S]. Consistent with these findings, recent studies
`indicate that rapamycin alone can also induce apoptosis of
`certain rhabdomyosarcoma cells [14,lS], and monocyte- and
`CD34-derived dendritic cells [80]. When combined with other
`in vitro,
`rapamycin enhanced
`chemotherapeutic agents
`cisplatin-induced apoptosis in human small cell lung cancer cell
`lines
`[11], potentiated
`apoptosis of
`the murine T(cid:173)
`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 S-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
`et al [lS] have observed that the responses of malignant and
`normal cells to rapamycin are qualitatively different. When
`treated with rapamycin, cells with wild-type pS3 arrest in Gl
`phase and maintain viability. In contrast, when grown under
`autocrine conditions (ie, serum-free)
`in
`the presence of
`rapamycin, pS3 mutant cells accumulate in Gl phase, but
`progress to S-phase and undergo apoptosis. More than 90% of
`apoptotic Rh30 cells (mutant pS3 alleles, Argm ~Cys273) were
`BrdU-labeled, suggesting that the cells died after initiating
`replication. Thus, rapamycin-induced death appea1·s to be a
`consequence of continued cell cycle progression, suggesting
`that pS3 senses inhibition of mTOR and co-operates to reinforce
`a Gl arrest. This model has been further tested using Rh30
`infected with adenovirus expressing wild-type pS3 (Ad-pS3)
`and pS3 +/+or pS3-/- murine embryo fibroblasts (MEFs) [lS].
`Restoring the pS3-mediated Gl checkpoint by Ad-pS3 infection
`causes Rh30 rhabdomyosarcoma cells to arrest in Gl and
`prevents rapamycin-induced apoptosis (Figure 4). Similarly,
`when exposed to rapamycin, pS3 -/- MEF cells continue cell
`cycle progression, and become apoptotic, whereas pS3 +I+
`MEF cells arrest in Gl and remain viable. Exactly why cells that
`fail to arrest in Gl in the presence of rapamycin die is currently
`unknown.
`
`Mechanisms of resistance to rapamycin
`As shown by Dilling et al [8], under similar conditions of
`growth, various cell lines demonstrated several thousand(cid:173)
`fold differences in sensitivity to rapamycin. The mechanism
`for this intrinsic resistance is under investigation. Cells may
`also acquire resistance either with or without mutagenesis.
`
`Mutations in FKBP12, mTOR and p7056K
`Budding yeast Saccharomyces
`cerev1siae
`treated with
`rapamycin irreversibly arrested in the Gl phase. However,
`when yeast TORl and TOR2 were genetically mutated to
`TORl-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 RBPl, a homolog of mammalian FKBP12, or a
`mutation altering Tyr89
`, 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 [4S]. 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 (Ser2035 ~Ile2 035), having reduced
`affinity for binding the FKBP12-rapamycin complex, is
`expressed [14,46]. mTOR is essential for activation of
`ribosomal p70S6Kl
`through phosphorylation of
`the
`rapamycin-sensitive sites at Thr229 or Thr389
`[63,64].
`Substitution of either of these residues can also abroga te 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.
`
`000005
`
`

`

`300 Current Opinion in lnvestigational Drugs 2002 Vol 3 No 2
`
`Figure 4. Protective effect of the tumor suppressor p53 on rapamycin-induced apoptosis.
`
`QJ
`
`QJ
`
`() c
`() en
`~
`0
`::::I
`:;::
`QJ
`32
`"C
`.Q
`E
`::::I
`'6 ·o.
`e a..
`
`Control
`
`Rap 100
`
`Ad-~gal + Rap 100
`
`Ad-p63
`
`Ad-p63 + Rap 100
`
`101 102 103 104 100
`
`101
`
`102
`
`103
`
`104
`
`Annexin V-FITC fluorescence
`
`Rh30 rhabdomyosarcoma cells were infected with either adenovirus expressing wild··type p53 (Ad-p53) or Ad-~-galactosidase (~-gal) at a
`multiplicity of infection of 1. After 24 h, medium was replaced with serum-free N2E, and cells were grown for a further 6 days in 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 dual staining for propidium iodide uptake and annexin V-FITC. Right panels show corresponding distribution of annexin V(cid:173)
`FITC staining in populations of cells. (Adapted from Huang et al [15].)
`
`Decrease of 4E-BP1 protein expression
`Mechanisms of acquired resistance (without use of mutagens)
`or intrinsic resistance have received less attention. Murine
`BC3Hl cells selected for acquired resistance demonstra ted
`reduced levels of p27K'"', and consistent w ith this, embryo
`fibroblasts with disrupted p27K;pi are relatively resistant to
`rapa mycin, as determined by inhibition of DNA precursors
`[75]. These data are consistent with Gl arrest, being in part
`due to rapamycin stabilizing this cyclin-dependent kinase
`inhibitor protein. Recently, rapa mycin-resistant cell
`lines,
`Rh30/ Rapa10K and C2, have also been obtained by growing
`
`Rh30 parental rhabdomyosa rcoma cells in the continuous
`presence of increasing concentra tions of rapamycin, without
`prior mutagenesis (82, Huang & Houghton, unpublished
`data]. When resistan t clones were grown w ithout rapamycin
`for 6 or 10 weeks, they reverted to rapa mycin sensiti vity (in
`terms of IC50 by growth in hibition assay). Thus, acquired
`rapamycin resistance was unstable.
`
`Analysis of several clones revea led increased levels of the c(cid:173)
`Myc protein. Of interest, these rapa mycin-resistan t clones
`exhibited increased anchorage-independent grow th in soft
`
`000006
`
`

`

`Inhibitors of mammalian target of rapamycin as novel antitumor agents: From bench to clinic Huang & Houghton 301
`
`aga r. Consisten t with increased c-Myc, the levels of the 4E(cid:173)
`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 ]. ln clones that reverted to
`rapamycin sensitivity, total levels of 4E-BP1 became similar
`to those in parental cells. ln some cases, intrinsic resistance
`also appears to relate to low 4E-BP:eIF4E levels. ln colon
`carcinoma cells, very low levels of 4E-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 CCI-779 and RAD-001 as
`antitumor agents
`CC/-779
`rapamycin-42, 2,2-
`inhjbitor-779;
`(cell cycle
`CCI-779
`is
`an
`ester of
`bis(hydroxymethyl)-propanoic acid)
`rapamycin (Figure 1), which was developed by Wyeth(cid:173)
`Ayerst as an antitumor agent. Like rapamycin, CCl-779
`by
`inhibition
`of mTOR,
`preventing
`the
`acts
`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•,29• ]. CCI-779 potently
`inhibits growth of
`numerous cultured human tumor cell Jines including
`human breast, prostate, pancreatic and small cell lung
`carcinomas, glioblastoma, medulloblastoma, melanoma,
`rhabdomyosarcoma and T-cell leukemia, with IC50 values
`in the nanomolar range [16,20-22] . These observations have
`been further supported by the significant inhibitory effect
`of CCI-779 on growth of human tumor xenografts in
`athymic nude mice [16,20,21,24]. When combined with
`cisplatin or camp tothecin, CCI-779 showed additive
`cytotoxicity in subcutaneous implants of human brain
`tumors [16]. Interestingly, in vivo CCI-779 also inhibited
`the growth of human U251 malignant glioma cells that
`were resistant
`to
`rapamycin
`in vitro, although
`the
`mechanism is unknown [16]. ln 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 [21,28•,29•]. 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(cid:173)
`dependent pathways, hence forming the basis for CCl-779
`hypersensitivity of PTEN deficient cells. However, it is
`unclear if Akt activates mTOR i11 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 Podsypanina et al [28•] showing inhibition of
`in PTEN +/- mice. These
`neoplastic
`transformation
`anima ls 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-779 had no effect on Akt
`activation, as anticipated, it normalized p70S6K activity.
`Whether, loss of PTEN function consistently sensitizes
`tumor cells
`to
`rapamycin analogs
`remains
`to be
`d emonstrated.
`
`These preclinical results have revealed that CCl-779 exhibits
`in some instances, cytotoxic
`impressive cytostatic, and
`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 CCI-
`779 in treatment of some cancers [88•]. In European phase I
`clinical trials [89], CCl-779 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/m2 /week. After ;::.: 8 weekly doses, significant tumor
`regressions were observed in two patients (receiving 15
`mg/m2 /week) with lung metastasis of renal cell carcinomas
`and in one patient (receiving 22.5 mg/m2 /week) with a
`two
`neuroendocrine tumor of the lung. Additionally,
`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-779 measw-ed 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.
`
`In the US, phase I clinical trials were also conducted to
`determine the safety and tolerability of CCl-779 [90]. CCI-
`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.
`Importantly, results from the above two trials indicate that
`CCl-779 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
`modest
`evidence
`of
`CCl-779
`induced
`only
`immunosuppression. Rapamycins also
`inhibit
`insulin
`signaling, but in the clinical trials reported there was no
`toxicity consistent w ith inhibition of insulin signaling. CCI-
`779 is cw-rently in phase Ill clinical trials. In athymic nude
`mouse xenografts, apart from breast and prostate cancers,
`human glioblastoma
`(U87MG)
`tumors and pancreatic
`carcinoma were also very sensitive to CCI-779. Thus, with
`an expanded focus, it is probable that clinical trials will
`identify other tumor types that are sensitive to CCI-779.
`
`RAD-001
`is another
`(40-0-(2-hydroxyethyl)-rapamycin)
`RAD-001
`rapamycin analog that is being developed

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