throbber
[Cancer Biology BC Therapy 2:3, 222-252, May/June 2005] ; ©2003 Landes Bioscience
`
`Review
`
`Rapamycins
`
`Mechanism of Action and Cellular Resistance
`
`ABSTRACT
`
`galand
`' munosup-
`ntation. Two
`ticancer agents.
`5 and inhibits the
`
`
`
`Rapamycins are macrocyclic lactones that possess immunosuppressive,
`antitumor properties. The parent compound, rapamycin, is approved
`pressive agent for preventing rejection in patients receiving orga
`analogues, CCI-779 and RAD001 are currently being investigate
`Rapamycins first bind a cyclophilin FKBPI 2, and this comple
`function of mTOR (mammalian target of rapamycin) a serbt
`with homology to phosphatidylinositol 3’ kinase. Currentl
`target,
`this places rapamycins in a unique position o
`inhibitor known. Consequently these agents haveb
`role of mTOR in cellular growth, proliferation,
`evidence suggests that mTOR acts as a centra
`
`onine (Ser/Thr) kinase
`OR is the only identified
`g the most selective kinase
`owerful tools in elucidating the
`and tumorigenesis. Increasing
`troller sensing cellular environment
`
`(nutritional status or mitogenic stimulation) 0; regulating translation initiation through the
`eukaryotic initiation factor 4E, and rib
`70 S6 kinase pathways. Here we review
`the conserved TOR signaling pathwa
`ceptual basis for tumor selectivity, and the
`mechanisms of resistance to this clfif antitumor agent.
`
`INTRODUCTION
`
`9
`
`Rapamycin, a m
`was isolated an
`
`
`
`ic lactone product of the soil bacteria Strrptomyce: /Jygraxcapiau,
`tified as an antifungal agent in the mid-1970’s.1'3 Rapamycin
`ructural analogue of the macrolide antibiotic FK506 (tacrolimus,
`, and like FK506 was found to potently suppress the immune system.4'7
`rail for rapamycin to act as an antitumor agent was recognized early in its
`nt when the drug demonstrated potent inhibitory activity against numerous
`mars in the NCI screening program.8‘1° However, the drug was not developed
`er due to stability and solubility problems that prevented development of a parenteral
`rmulation for use in clinical trials. Also at that time in the early 1980’s, the mechanism
`of action of rapamycin in blocking signal transduction was not understood.
`Rapamycin (Rapamune®), as an immunosuppressive drug, was finally approved by the
`Food and Drug Administration (FDA)
`in the USA in 1999, and the European
`Commission in 2000, respectively. Results from many laboratories have demonstrated that
`rapamycin, in contrast to FK506, is not only a potent immunosuppressant, but also an
`active antitumor agent. Rapamycin can act as a cytostatic, slowing or arresting cells in G1
`phase. Under specific conditions, or in some tumor cell lines rapamycins may induce
`apoptosis in culture. To date, studies have revealed that rapamycin potently arrests growth
`of cells derived from rhabdomyosarcoma, neuroblastoma and glioblastoma, small cell lung
`cancer,“'17 osteoscarcoma,18 pancreatic cancer,19'2° breast and prostate cancer,21'23
`murine melanoma and leukemia, and B-cell lymphoma.9’24‘26 In addition to broad
`spectrum activity in vitro, rapamycin and its derivatives (designated here as rapamycins)
`suppress growth of some human and murine tumor models in vivo.“'3° When combined
`with other chemotherapeutic agents, rapamycins generally show at least additive antitumor
`activity.1°=12’17’31 Preliminary data from clinical trials have indicated that rapamycins are
`well tolerated and successfully suppress growth of various human tu.mors.52'54
`The use of rapamycin as an anticancer drug is clinically impractical, because of its poor
`water-solubility and stability in solution. Recently, rapamycin ester analogues (Fig. 1),
`CCI-779 [rapamycin-42, 2, 2-bis(hydroxymethyl)-propionic acid]
`(Wyeth-Ayerst, PA,
`USA) and RAD001 [everolimus, 40-O-(2—hydroxyethyl)-rapamycin]
`(Novartis, Basel,
`Switzerland), have entered clinical trials. These analogues have improved pharmaceutical
`properties. CCl—779 is being developed for both intravenous and oral administration,
`whereas RAD001 is only for oral administration. The antitumor activity of these analogues
`
`
`
`(sirolimus)
`Prograf®)
`The
`devel
`50
`
`
`
`Shile Huang
`
`Mary-Ann Biornsti
`
`Peter J. Houghton*
`
`Department of Molecular Phormacologm St Jude Children's Research Hospital; 332
`II. Lauderdale; Memphis; Tennessee USA
`
`*Carrespnndence to: Peter I. Houghton; Department of Molecular Pharmacology; St
`Jude Children's Research Hospital; 332 N. Lauderdale; Memphis; Tennessee
`30105-2794 USA; Email: peter.houghton@stiude.org
`
`Received 04/24/03; Accepted 05/0l/03
`
`Previously published online as a [It&T Paper in Press at:
`http://VIww.Iundesbioscience.com/iournals/cbt/toc.php?voIume=2&issue=3
`
`KEY WORDS
`
`Rapamycins, Translation initiation, Cancer,
`Resistance, Therapy, Ribosomal biogenesis, Yeast
`
`Work reported from these laboratories was supported by PHS awards (A23099,
`(AS0755, CA77776, (M6996 and (A2l765 (Cancer Center Support Grant) and by
`American, Lebanese, Syrian Associated Charities (AlSA().
`
`$0
`V
`"3
`
`©"'§
`
`222
`
`Cancer Biology 86 Therapy
`
`2003; Vol. 2 Issue 3
`
`We
`
`West-Ward Pharm.
`Exhibit 1018
`Page 001
`
`

`

`RAPAMYCTNS: MECHANISM OF ACTION AND CELLULAR RESISTANCE
`
`is similar to that of rapamycin.17’21‘23’27’30 RADOOI is
`in Phase I trials whereas the development of CCI—779
`is more advanced with several Phase II trials completed.
`Why should it be anticipated that
`rapamycins
`could exhibit tumor—selectivity, in a manner analogous
`to the activity of another kinase inhibitor, Gleevec, in
`Bcr—Abl expressing chronic lymphocytic leukemia?
`Accumulating evidence suggests that genetic mutations
`or compensatory changes in tumor cells may affect
`sensitivity to rapamycins. At least
`in some systems
`mutations that occur frequently in malignant trans-
`formations such as GL1 amplification, or mutations
`that inactivate p53, and the dual specificity phos—
`phatase PTEN (phosphatase and tensin homolog
`deleted on chromosome ten, also known as MMACI
`for mutated in multiple advanced cancers) or lead to
`activation of Akt appear to determine rapamycin
`sensitivity. On the other hand there may be multiple
`loci that confer intrinsic or acquired resistance. This
`review will summarize the current knowledge of the
`role of mTOR in cellular regulation, the mechanism
`of action of raparnycins, and currently understood
`resistance mechanisms.
`
`THE RAPAMYCIN TARGET (MTOR)
`
`W;fl’;W fi
`
`Rapamycin
`
`
`
`CCI-T79
`
`Figure l. The structure of rapamycin and its analogues FK506, CCI-779 and RADOOI . The
`FKBPI2 binding Face is shown by the filled bars, whereas the mTOR binding Face of
`rapamycin is shown by the hatched bar.
`
`The mammalian target of rapamycin, [also named
`FKBPI2 and rapamycin—associated protein (FRAP),
`rapamycin and FKBP12 target 1 (RAFT1), rapamycin
`target
`I
`(RAPTI), or sirolimus effector protein
`(SEP)],
`is a 289 kDa Ser/Thr kinase orthologue of
`TORI and TOR2 in Sac:/mromyces cerevz'5z'ae.35’36
`TOR is an atypical serine/threonine kinase highly
`conserved from yeast to mammals. Human, mouse
`and rat mTOR proteins share 95% identity at the amino acid
`level.36‘38 Since the C—terminus ofTOR is highly homologous to the
`catalytic domain of phosphatidylinositol 3' kinase (PI3K), mTOR is
`considered to belong to the PI5K—related protein kinase (designated
`PIKK) family, which also includes Mecl, Tell, RAD3, MEI—4I,
`DNA—PK, ATM, ATR, and TRRAP3637 Recently, single TOR
`homologs have also been identified in fungi (TORI in Cryptococcus
`neofiirmzzm), plants
`(AtTOR in Aralzidopsix t/mliana), worms
`(CeTOR in Caenor/aabziitis elegant), and flies (dTOR in Drosophila
`memz[oga:ter).37
`The domain structure of mTOR is depicted in Figure 2. The
`protein consists of a catalytic kinase domain, an FKBP12—rapamycin
`binding (FRB) domain and a putative auto—inhibitory domain
`(“repressor domain”) near the C—terminus, and up to 20 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. HEAT motifs may serve as
`protein—protein interaction parts, whereas FAT and FATC domains
`
`may participate in modulation of catalytic kinase activity of
`mTOR.36 The remarkable conservation of mTOR at the amino acid
`
`level suggests that multiple domains of this protein are essential for
`its cellular functions.
`
`Tor Signaling in Yeast. In yeast, Tor kinase activity is regulated
`by availability of nutrients (amino acids and glucose), whereas in
`mammalian cells, mTOR is regulated also by phosphatidic acid,
`ATP, and growth factors.37’39'42 The Tar signaling pathway in yeast
`is depicted in Figure 3, and controls translation initiation, protein
`turnover, transcription, and actin cytoskeleton organization. In yeast
`these pathways have been rigorously established,37 and are at least in
`part maintained in mammalian cells. The TorI/2 complex (desig-
`nated TORC1) comprising Kogl
`(the yeast homologue of the
`mammalian protein raptor) and Lst8 controls translation, protein
`stability and transcription,43'45 whereas the TORC2 complex
`controls actin organization. As TORC2 is not a rapamycin target it
`will not be considered further. The evolutionarily conserved TORCI
`complex controls translation initiation probably through activation
`of eIF4E, and transcription of ribosomal genes, stress response
`genes, ribosomal biogenesis and tRNA synthesis.
`Tor, through the TORCI complex, controls protein turnover and
`some aspects of transcription through regulation of protein phos—
`
`2549 aa
`
`Figure 2. Schematic representation at the domains at mTOR. Structural
`domains of mTOR. HEAT :[huntingtin elongation Factor A subunit of PP2A
`and TOR)
`repeats [positions 71-522 and 628-1147); FAT:
`(FRAP-ATM-
`TRAPP) domain, which is unique to PIK-related kinases located N-terminal to
`the FKBPl2-rapamycin binding domain [FRB),'
`the role of
`FAT sequences is less clear, but they are associated with
`C-terminal
`FAT (FATC)
`sequences
`in mTOR.
`Interaction
`between FAT and FATC domains may Facilitate protein bind-
`ing or act as a structural scatlold; CD: Catalytic domain;
`RD: regulatory domain.
`
`1 I
`
`
`II > II
`ll
`
`www.landesbioscience.com
`
`Cancer Biology 8: Therapy
`
`77%
`
`West-Ward Pharm.
`Exhibit 1018
`Page 002
`
`West-Ward Pharm.
`Exhibit 1018
`Page 002
`
`

`

`RAPAMYCINS: MECHANISM OF ACTION AND CELLULAR RESISTANCE
`
`Amino acids/glucose
`
`Cell membrane
`
`
`Translation
`
`Actin
`organization
`
`protein turnover
`
`Figure 3. Nutrient signaling in yeast. (Adapted from Jacinio and Hc1||.37)
`
`Transcription
`
`phatases. Tor positively regulates Tap42, which binds to and inacti-
`vates the Sit4 and Pph protein phosphatases.46’47 Pph21 and Pph22
`are calaytic subunits of PPZA, and Sit4 is the catalytic subunit of a
`PP2A—related phosphatase in yeast. Nutrient deficiency or rapamycin
`leads to dissociation of these phosphatases from Tap42, resulting in
`increased phosphatase activity. This leads to dephosphorylation of
`Nprl and Gln3 involved in protein turnover and transcription,
`respectively. Control of protein turnover and some aspects of tran-
`scription appear to be regulated through Tap42 binding and inactivat-
`ing Sit4, whereas pathways regulated by release of Pph phosphatases are
`less well defined. In the presence of nutrients Tor signaling represses
`autophagy and leads to stabilization of proteins by suppressing
`ubiquitin—dependent degradation.“ For example, Tor signaling
`prevents ubiquitylation, vacuolar targeting and degradation of the
`tryptophan transporter Tat2 by maintaining phosphorylation and
`inactivation ofNpr1 a putative Tat2 kinase.49’50 Under conditions of
`starvation Sit4 becomes activated leading to dephosphorylation of
`Nprl and degradation ofTat2. The Tap42—Sit4 pathway also controls
`the Gln3 transcription factor. Under nutrient replete conditions
`Gln3 is phosphorylated and is bound to the Ure2 protein in the
`cytoplasm. Inhibition of Tor by rapamycin or nitrogen starvation
`leads to dephosphorylation of Gln3, its nuclear translocation and
`transcription of genes required for the use of secondary nitrogen
`sources.43’44 The pathway(s) downstream of Tap42 involving Pph
`are less clear. Similarly, Tor negatively regulates the heterodimeric
`transcription factor Rtg1—Rtg3 through an unknown mechanism.“
`The Tor pathway also is important in control of stress—responses
`through modulation of transcription. The TORC1 complex nega-
`
`tively controls transcription of stress-responsive genes through the cyto-
`plasmic sequestration of the general stress transcription factors Msn2
`and Msn4. Although the mechanism is not fully understood this
`may occur through Tor signaling promoting the binding of these
`transcription factors to the 14-3-3 homologues Bmhl and Bmh2.50
`Proximal Signaling in Mammalian Cells. In mammalian cells,
`mTOR is regulated not only by nutrients but also by growth fac-
`tors.37’39‘42 It appears that growth factors regulate mTOR signaling
`through both I’I3K and Akt pathways, whereas proximal activators
`regulated by nutrients and ATP are less well characterized. In mam-
`malian cells mTOR is activated as a consequence of ligand binding
`to various growth factor receptors that result in activation of PI3K,
`Figure 4. Activated PI3K catalyzes the conversion of phostidylinosi—
`tol (4,5)—bisphosphate (PIP2) to phosphatidylinositol—3,4,5—trispho-
`sphate (PIP3). This pathway is negatively regulated by a dual—speci—
`flcity protein and lipid phosphatase PTEN. Full activation of Akt,
`downstream of PI3K requires binding of PIP3 to the pleckstrin
`homology domain ofAkt, and phosphorylation by phosphoinositide—
`dependent kinases 1/2 (PDK1/2) and other unidentified kinases.
`Pharmacological studies with albeit relatively non—specif1c inhibitors
`of PI3K (wortmannin and LY294002) indicate that mTOR is down-
`stream of PI3K. How mTOR is regulated by I’I3K or Akt, however,
`is still not well understood. Akt can phosphorylate mTOR
`(Ser2448) directly, although the significance remains to be deter-
`mined.37’38 Recent studies have placed the tuberous sclerosis
`(TSC1/2) complex as a modulator between PI3K/Akt and mTOR.52‘54
`The TSC1/2 complex comprises harmartin (TSC1) and tuberin
`(TSC2). These proteins form a physical and functional complex in
`
`224
`
`Cancer Biology 8: Therapy
`
`2003; Vol. 2 Issue 3
`
`West-Ward Pharm.
`Exhibit 1018
`Page 003
`
`West-Ward Pharm.
`Exhibit 1018
`Page 003
`
`

`

`RAPAMYCINS: MECHANISM OF ACTION AND CELLULAR RESISTANCE
`
`Figure 4. Growth Factor signaling in mammalian cells.
`
`Vivo, which binds and inhibits mTOR.52‘54 Loss of TSC1/2 results
`in mTOR—dependent increase of ribosomal p70 S6 kinase (S6K1)
`activity, and confers resistance of cells to amino acid starvation.53
`Conversely co—expression of TSC1 and TSC2 inhibits amino
`acid—induced activation of S6K1 in nutrient—deprived cells.“
`Mitogenic stimuli, such as insulin or serum, activate Akt which can
`directly phosphorylate TSC2 on multiple sites in vitro and in
`vivo.52’55’56 Phosphorylation of TSC2 at Ser939 and Thr1462 is
`PI3K-dependent.52’56 Akt—mediated phosphorylation of TSC2
`destabilizes TSC2 and thereby inhibits the formation of TSCI/2
`complex,
`leading to de—repression of mTOR, and consequently
`increasing the kinase activity of mTOR. By contrast, treatment with
`rapamycin does not influence the phosphorylation of TSC2,52 and
`together with other data,5354 suggests that mTOR lies downstream
`of TSC2. Other results5758 imply that the TSC1/2 complex may
`mediate S6K1 activation through a pathway parallel
`to mTOR.
`However, further studies will be required to address the relationship
`of mTOR and TSC1/2.
`
`mTOR Associated Proteins. mTOR forms a scaffold complex
`with other proteins, such as raptor (regulatory associated protein of
`mTOR) and mLST8,59'60 which are the mammalian counterparts of
`yeast Kogl (kontroller of growth 1) and Lst8,61 respectively. The
`exact role of raptor remains unclear.38’59‘61 Raptor may act as a scaffold
`protein linking mTOR to S6K1 and 4E—BP1.60 Alternatively, it may
`have a dual function, inhibiting mTOR under nutrient—def1cient
`conditions and stimulating mTOR in a nutrient—replete environment.”
`
`Thus TSC1/2, raptor and possibly mLST8 act as potential modulators
`of mTOR function in response to availability of nutrients.
`Signaling Distal to mTOR. mTOR controls translation initiation
`through two pathways, S6Kl and eukaryotic initiation factor 4E
`(eIF4E) binding proteins (4E-BPS). mTOR either directly phospho-
`rylates Thr389 of S6K1, or suppresses a phosphatase responsible for
`maintaining the hypophosphoryalted state of this residue. Activation
`of S6K1 enhances translation of mRNAs bearing a 5’
`terminal
`oligopyrimidine tract (5’ TOP).62’63 Inactivation of S6K1 decreases
`synthesis of ribosomal proteins and translation factors.62’63
`Activation of S6K1 is complex. The process involves interplay
`between four different domains and at least seven specific sites medi-
`ated by multiple upstream kinases.64 At
`least 12 sites
`(Serl7,
`Thr229, Thr367, Thr371, Thr389, Ser404, Ser411, Ser418, Tr421,
`Ser424, Ser429, and Thr447) can be phosphorylated in response to
`serum stimulation.“ However, the responsible kinases have not been
`fully characterized. Phosphoinositide—dependent protein kinase 1
`(PDK1) phosphorylates Thr229 in vitro and in vivo.66 Atypical
`PKC isoforms and the Rho family of small G proteins (cdc42 and
`Racl) may contribute to phosphorylation of S6K1,65 although the
`specific sites regulated by these kinases remain to be determined. In
`vitro, mTOR phosphorylates only Thr389 in the regulatory
`domain.67'69 However, whether this phosphorylation is directly or
`indirectly regulated by mTOR is in question, since recent data suggest
`that mTOR may regulate S6K1 activation by inhibiting phos-
`phatases rather than directly phosphorylating S6K1.64’70
`
`wwvv.landesbioscience.com
`
`Cancer Biology 8: Therapy
`
`225
`
`West-Ward Pharm.
`Exhibit 1018
`Page 004
`
`West-Ward Pharm.
`Exhibit 1018
`Page 004
`
`

`

`RAPAMYCINS: MECHANISM OF ACTION AND CELLULAR RESISTANCE
`
`mTOR phosphorylates and inactivates the 4E—BP suppressor
`proteins causing their dissociation from the RNA cap—binding
`protein eIF4E. In response to mitogens, six sites (Thr37, Thr46,
`Ser65, Thr70, Ser83, and Ser112) of 4E—BP1 can be phosphorylated.“
`So far, only mTOR and ATM have been identified to be involved in
`phosphorylation of 4E—BP1.71‘74 Other kinases that phosphorylate
`4E—BP1 remain to be characterized. ATM phosphorylates 4E—BP1 at
`Ser112,74 however the physiological significance of this remains to
`be elucidated. In vitro mTOR phosphorylates 4E—BP1 at two sites
`(Thr37, Thr46) and possibly at two additional Ser/Thr residues
`(Thr70 and Ser65) in the N—terminal region.73’75 Phosphorylation
`is a hierarchical process.73’75'77 Phosphorylation of Thr37/Thr46 is
`followed by Thr70 phosphorylation. Ser65 is phosphorylated last77
`and is dependent on phosphorylation of all three Thr/Pro sites.75’76
`Mutation of Thr37 and/or Thr46 to alanine(s) prevents phosphory—
`lation of Ser65 and Thr70,
`indicating that phosphorylation of
`Thr37 and Thr46 serves as a requisite “priming” event.36 Single
`phosphorylation of these residues is not sufficient
`to dissociate
`4E—BP1 from eIF4E,
`indicating the requirement of combined
`phosphorylation of at least Thr37, Thr46, Ser65, and Thr70 in
`4E—BP1
`to suppress association with eIF4E.36’78 Inhibition of
`mTOR leads to rapid hypophosphorylation of 4E—BP1 which then
`tightly binds to eIF4E. This prevents formation of eIF4F complex
`that contains eIF4E, eIF4G, eIF4A and eIF3, and inhibits
`cap—dependent translation initiation.36 Once 4E—BP1 is hyperphos—
`phorylated, it releases eIF4E, facilitating eIF4F complex formation
`and promoting cap—dependent protein synthesis.36 Overall inhibition
`of mTOR by rapamycin leads to a decrease in protein synthesis of
`15 to 20 percent. However, as the eIF4E pathway is required for
`translation of mRNA’s encoding cyclin D1,79’80 and ornithine
`decarboxylase81 inhibition of mTOR leads to slowing or arrest of
`cells in G1 phase of the cell cycle. However, the exact mechanism(s)
`by which mTOR regulates cell cycle progression are complex, poor-
`ly understood,
`and potentially context
`specific. Although
`rapamycins are highly specific inhibitors, the TOR pathway regu-
`lates multiple cellular processes. The mTOR pathway regulates trans-
`lation initiation of survival
`factors
`such as
`c—MYC82 and
`hypoxia—inducible factor 10., and consequently vascular endothelial
`growth factor.83’84 In addition, mTOR is involved in the regulation
`of cyclin A, cyclin dependent kinases (cdkl/2), cdk inhibitors
`(p21CiP1 and p27KiP1), retinoblastoma protein, RNA polymerase
`I/II/III—transcription and translation of rRNA and tRNA, protein
`phosphatases (PPZA, PP4 and PP6), and CLIP—170.36’37v85‘91
`
`TUMOR SELECTIVITY OF RAPAMYCINS
`
`To date there are no reports suggesting that activating mutations
`of mTOR, or overexpression occur as primary events in malignant
`transformation. However, activation of signaling pathways both
`proximal and distal to mTOR appear to occur frequently in human
`cancer. Loss of the phosphatase PTEN by deletion, silencing or
`mutation leads to constitutive activation of Akt,29’30’92 and upregu—
`lation of mTOR—dependent pathways. In PTEN—deficient tumor
`cells or mouse embryo fibroblasts (MEFS), activated Akt is associat-
`ed with enhanced activity of S6K1 and hyperphosphorylation of
`4E—BP1,29’30 or increased levels of c—MYC.92 It is speculated that
`Akt—activated cells become dependent on upregulated mTOR
`signaling for proliferation, hence become more susceptible to
`rapamycin or CCI—779. Increased sensitivity to rapamycins has been
`demonstrated in a panel of brain, prostate, and breast cancer cells,
`multiple myeloma cells and in MEFs.25’29 The association of PTEN
`deficiency and sensitivity to rapamycin is further supported by the
`
`activity of CCI—779 against the growth of human tumors implanted
`in athymic nude mice.23’3° There are, however, some exceptions;
`cells with functional PTEN and low constitutive activation of Akt
`
`are equally sensitive to inhibition of proliferation by rapamycins.
`Consequently the role of PTEN as an independent variable predicting
`for rapamycin sensitivity remains to be demonstrated. Disruption of
`the TSC1 or TSC2 gene leads to the development of tumors in
`multiple organs, notably kidney brain heart and lung. Even low
`doses of rapamycin causes apoptosis and necrosis of spontaneous
`renal tumors in Eker rats with germline mutation in TSC2 and
`results in tumor regression.93 These studies suggest that rumors
`developing in patients with tuberous sclerosis may be sensitive to
`rapamycins. Oncogene expression may also regulate the response to
`rapamycin. For example, in RKSE cells transformed with c—MYC or
`Ras rapamycin treatment increased global protein synthesis. In contrast
`rapamycin inhibited global protein synthesis and turnover in GLI
`transformed isogenic lines leading to inhibition of proliferation.”
`Although generally considered to be cytostatic agents, rapamycins
`can induce apoptosis in some cell systems. Rapamycins induce
`apoptosis of B—cells, rhabdomyosarcoma cells, renal tubular cells and
`dendritic cells.16’95'97 Rapamycin enhances transforming growth
`factor—[3 induced cell cycle arrest,98 and through blocking survival
`factor signaling” rapamycins enhance cell death. Our results suggest
`that the functional status of the p53 tumor suppressor may dictate
`the cellular fate of rapamycin treated cells, as depicted in Figure 5.
`For example, under serum free conditions,
`the response to
`rapamycin in cells lacking functional p53 is apoptosis, suggesting
`that only in the absence of p53/p21 inhibition of mTOR becomes
`lethal
`(so—called synthetic lethality). Ectopic expression of p53 or
`p21CiP1 protects cells from rapamycin-induced apoptosis. The
`implication is that rapamycins may have potential tumor—selective
`therapeutic effects.“ Recent results show that inhibition of mTOR
`by rapamycin induces a cellular stress response characterized by rapid
`and sustained activation of ASK1 (apoptosis signal—regulating kinase
`1) signaling in p53—mutant cells.100 This leads to sustained phos-
`phorylation of c—]UN (Ser63) that appears to be responsible for
`inducing apoptosis. Rapamycin—induced stress appears distinct from
`that induced by ultra violet radiation in that MEKK1 is not activated,
`and from cytotoxic agents such as DNA damaging agents where
`other stress pathways (p38 or ERK1/2) are also activated.1°1*1°2 In
`contrast, cells expressing wild type p53,
`(or constitutive p21CiP1
`expression)
`there is only transient activation of ASK1.100
`Suppression of ASK1 is associated with binding of p21CiP1 in
`rapamycin treated cells, and protection from apoptosis. Since the
`rapamycin-induced apoptosis is specifically prevented by insulin—like
`growth factors (IGF—I/II) and insulin,96 combination of IGF receptor
`inhibitors with rapamycins may be selectively cytotoxic and induce
`regression of tumors with p53 mutations. VVhether such anticipation
`is justified requires vigorous testing.
`
`MECHANISMS OF RESISTANCE T0 RAPAMYCINS
`
`Intrinsic sensitivity to rapamycins between cell lines may vary by
`several orders of magnitude. For example, rhabdomyosarcoma cells
`in vitro are very sensitive (concentration for 50% inhibition (ICSO)
`of proliferation ~ 1 nM) compared to colon carcinoma cells (IC50 >
`5000 nM).“ Mechanisms of intrinsic and acquired resistance may
`have either a genetic or epigenetic basis.
`Mutations in FKBPIZ and mTOR. Rapamycins first bind to the
`cyclophilin FKBP12 in mammalian cells,
`forming the FKBP—
`rapamycin complex. This complex then interacts with the FRB
`domain in mTOR (Fig. 2), and inhibits the function of mTOR.
`
`226
`
`Cancer Biology 8: Therapy
`
`2003; Vol. 2 Issue 3
`
`West-Ward Pharm.
`Exhibit 1018
`Page 005
`
`West-Ward Pharm.
`Exhibit 1018
`Page 005
`
`

`

`RAPAMYCINS: MECHANISM OF ACTION AND CELLULAR RESISTANCE
`
`Rapamycin resistance may be conferred by
`mutations in FKBP12 that prevent the forma-
`tion of FKBP—rapamycin complex, or mutations
`in the FRB domain of mTOR that block binding
`of FKBP—rapamycin complex to mTOR. Such
`mutations were first identified in budding yeast
`5. cerevixiae in which treatment with rapamycin
`irreversibly arrests cells in the G1 phase. In the
`yeast 3. caret/iriae, deletion of the RBPI gene, a
`homologue of mammalian FKBP—12, results in a
`recessive rapamycin resistance, whereas expres-
`sion of RBPI restores rapamycin sensitivity.103
`This observation has been further confirmed by
`RBPI disruption experiments using the patho-
`genic
`yeast Czzndida
`at/Means. Wild—type
`RBP1/RBPI parental strain and the rbpl/RBPI
`heterozygous mutant were sensitive to rapamycin
`inhibition, whereas
`rbpl/rbpl homozygous
`mutant was rapamycin resistant.104 In addition, in
`S. cerevisiae mutation of a specific residue (Tyr89)
`which is conserved in RBPI or FKBPs, also
`resulted in decreased binding of rapamycin and
`conferred a recessive resistance pl1eI10type.105 In
`murine mast cells, two distinct point mutations
`in FKBP12 confer
`resistance. By altering a
`hydrophobic residue within the drug—binding
`pocket (Trp599Leu) or changing a charged sur-
`face residue (Arg49-)Gln), the binding aflinity for
`rapamycin decreases substantially_106
`A genetic screen identified rapamycin—resist-
`ant alleles with mutations in genes designated
`TORI and TORZ. Strains with mutated to torl-1
`
`Synthetic lethality
`a genetic defect alters ceilular response to drug action
`
`p53+.l+
`
`p53~‘-
`
`Rapatnytiril
`
`Atiue
`
`Rapamycinl
`
`
`
`PrupidiumIodideFluorescence
` =‘:.5ti..'ii.
`
`+ Rap 11]!)
`
`Ad-p53
`
`W”
`
`in‘
`
`.-_._
`In‘
`
`In’
`10’
`lo‘
`10‘ In“
`11?
`in’
`Annexin V—F|TC Fluorescence
`
`in‘
`
`Figure 5. Loss of p53 function alters cellular response to rapamycin from cytostasis to apoptosis
`in murine embryo fibroblasts (MEFs). Left: schematic representation of synthetic lethality. Right:
`Wild type, and p53’/’MEFs, and p53’/‘ MEFs infected withAd-p53 (MOI of 100) were grown
`without or with rapamycin (100 ng/ml). Cells were harvested after 5 days and apoptosis deter-
`mined by quantitative FACs analysis (ApoAlert) assay. The per cent distribution of cells in each
`quadrant is presented (from Huang et al. 200i '6).
`
`(Ser19729Arg) and t0r2—I (Ser19759Arg), were
`completely resistant
`to the growth— inhibitory
`effect of rapamycin. These resistant alleles encode mutant Tor proteins
`that do not bind the FKBP—rapamycin complex.103’107'1“ This
`result suggests that a conserved serine residue (Ser1972 in Torl;
`Ser1975 in Tor2) in Tor proteins is critical for FKBP—rapamycin
`binding. In mammalian cells mutations in the FRB domain confer
`a dominant resistant phenotype consistent with decreased affinity
`for binding of the FKBP— rapamycin complex.1” Expression of a
`mutant mTOR (Ser2035-)Ile), having greatly reduced binding affin-
`ity for the FKBP—rapamycin complex, confers high level resist-
`anCe_14,112,113
`
`Deregulation of eIF4E. mTOR phosphorylates and regulates the
`function of 4E—BP1, the suppressor of eIF4E.36 Recently, our group
`has found that acquired resistance to rapamycin was associated with
`decreased levels of 4E—BP1.121 In the absence of selective pressure
`(rapamycin), resistance was unstable and cells reverted to being
`sensitive to growth inhibition of rapamycin within ten weeks. In
`resistant cells
`the levels of 4E—BP1 were reduced significantly
`(~10—fold), whereas in rapamycin—sensitive revertants the 4E—BPI
`levels increased to those in wild type cells. Levels of 4E—BP1 transcripts
`were unaltered in rapamycin resistant clones suggesting post—tran—
`scriptional regulation. Further studies indicate that the synthesis of
`4E—BPI
`significantly decreased in rapamycin—resistant clones.
`Whether the steady state level of 4E—BP1 is also regulated by
`increased degradation remains to be determined. Thus, the changes
`in 4E—BP1 levels are reminiscent of those reported for p27KiP1 in
`rapamycin resistant BC5H cells.“5 In contrast, no consistent
`changes were detected in the level or activity of S6K1 between parental
`
`and resistant clones. Rapamycin also inhibited growth factor activa-
`tion of SGKI equally in parental and rapamycin—resistant clones.
`Intrinsic resistance to rapamycin has been shown in glioblastoma
`cells and colon adenocarcinoma that have very low 4E—BP1.“4 For
`example, 4E—BP1 is barely detected in HCT8 colon carcinoma cells
`that are highly resistant to rapamycin (IC50 > 10,000 ng/ml). \X/hen
`4E—BP1 is overexpressed, these cells become sensitive (IC50 < 10
`ng/ml) to rapamycin, Figure 6.114 These data suggest that low levels
`of 4E—BPI results in de—regulation of eIF4E, conferring rapamycin
`resistance.
`
`These results suggest that rapamycin—regulation of the eIF4E
`pathway is crucial in inducing growth arrest. Further de-regulation
`of eIF4E may facilitate a malignant phenotype. Of interest is that
`both rapamycin—resistant and —revertant cells exhibited elevated
`c—MYC levels, and increased anchorage—independent growth. That
`deregulation of the eIF4E pathway is associated with increased
`malignancy is supported by certain clinical observations that dereg-
`ulation of the eIF4E pathway does promote tumor progression.“6
`In addition to decreased 4E—BP1 expression, as described above,
`increased eIF4E levels may also cause de—regulation of eIF4E. In
`advanced head and neck carcinoma,“7 breast carcinoma“8 gas-
`trointestinal carcinoma,119 and peripheral carcinoma of the lung120
`eIF4E levels are elevated. However, levels of 4E—BPI suppressor
`proteins have not been reported in a consistent manner. Potentially,
`the ratio of 4E—BP:eIF4E may determine whether inhibition of
`mTOR elicits a biologically significant tumor response. Further
`studies will be necessary to determine if this ratio has predictive
`value for drug sensitivity of tumors.
`
`www.landesbioscience.com
`
`Cancer Biology 8: Therapy
`
`227
`
`West-Ward Pharm.
`Exhibit 1018
`Page 006
`
`West-Ward Pharm.
`Exhibit 1018
`Page 006
`
`

`

`RAPAMYCINS: MECHANISM OF ACTION AND CELLULAR RESISTANCE
`
`Fiapamycin
`Sensitive
`
`3 7 4- 4E—BP‘t I
`
`-1- (3-tubulin
`
`B
`
`'03 HCT8-4E-BP1 clones
`[MI
`E
`1
`2
`3
`
`4 51""
`
`""""C*- Du 4-4E-BP1
`
`~ --e|F4E
`
`100 -7
`
`Mutations in S6K1. Ribosomal S6K1 is the other principal
`downstream effector of mTOR. Inhibition of mTOR by rapamycin
`primarily inhibits phosphorylation of Thr389 in the regulatory
`domain.“ However, since phosphorylation of S6K1 is hierarchical
`with phosphorylation of several other sites dependent on phospho-
`rylation of Thr389,64 rapamycin in vivo influences phosphorylation
`of other sites, including Thr229 in the catalytic domain, and S404
`in the regulatory domain.67 Mutation ofThr3899Glu abrogates the
`ability of rapamycin to inhibit S6K1 activation.67’121 Similarly,
`substitution of Thr229 by either a neutral amino acid Alanine
`(Thr2299Ala) or by an acidic amino acid Glu (Thr2299Glu),
`renders S6K1 insensitive to rapamycin.122 In addition, deletion of
`the 77 N—terminal codons (AN77) confers rapamycin resistance.123
`Of note truncation of

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