throbber
[Cancer Biology 8& Therapy 2:3, 222-232, May/June 2003]; ©2003 Landes Bioscience
`
`Review
`
`Rapamycins
`Mechanism of Action and Cellular Resistance
`
`Shile Huang
`Mary-AnnBjornsti
`Peter J. Houghton*
`
`Department of Molecular Pharmacology; St Jude Children's Research Hospital; 332
`N. Lauderdale; Memphis; Tennessee USA
`
`*Correspondence to: Peter J. Houghton; Department of Molecular Pharmacalogy; St
`Jude Children’s Research Hospital; 332 N. Lauderdale; Memphis; Tennessee
`38105-2794 USA; Email: peterhoughton@stjude.org
`
`Received 04/24/03; Accepted 05/01/03
`
`Previously published online os 0 CB&T Paperin Press at:
`http://wwwlandesbioscience.com/journals/cbt/toc.php?volume=28issue=3
`
`KEY WORDS
`
`Rapamycins, Translation initiation, Cancer,
`Resistance, Therapy, Ribosomal biogenesis, Yeast
`
`Work reported from these laboratories was supported by PHS awards (A23099,
`(A58755, CA77776, CA96996 and CA21765 (Cancer Center Support Grant) and by
`American, Lebanese, Syrian Associated Charities (ALSAC).
`
`222
`
` ABSTRACT
`
`Rapamycins are macrocyclic lactones that possess immunosuppressive, antifungal and
`antitumor properties. The parent compound, rapamycin, is approved as an immunosup-
`pressive agent for preventing rejection in patients receiving organ transplantation. Two
`analogues, CCI-779 and RADOO1 are currently being investigated as anticancer agents.
`Rapamycinsfirst bind a cyclophilin FKBP12, and this complex binds and inhibits the
`function of mTOR (mammalian target of rapamycin) a serine/threonine (Ser/Thr) kinase
`with homology to phosphatidylinositol 3’ kinase. Currently, as mTORis the only identified
`target,
`this places rapamycins in a unique position of being the most selective kinase
`inhibitor known. Consequently these agents have been powerful fools in elucidating the
`role of mTORin cellular growth, proliferation, survival and tumorigenesis. Increasing
`evidence suggests that mTOR acts as a central controller sensing cellular environment
`(nutritional status or mitogenic stimulation) and regulating translationinitiation through the
`eukaryotic initiation factor 4E, and ribosomal p70 Sé6 kinase pathways. Here we review
`the conserved TOR signaling pathways, conceptual basis for tumor selectivity, and the
`mechanismsofresistance to this class of antitumor agent.
`
`INTRODUCTION
`
`Rapamycin, a macrocyclic lactone product ofthe soil bacteria Streptomyces hygroscopicus,
`was isolated and identified as an antifungal agent in the mid-1970’s.'3 Rapamycin
`(sirolimus),
`is a structural analogue of the macrolide antibiotic FK506 (tacrolimus,
`Prograf®) (Fig. 1), and like FK506 was foundto potently suppress the immunesystem.*7
`The potential for rapamycin to act as an antitumor agent was recognizedearly in its
`development when the drug demonstrated potent inhibitory activity against numerous
`solid tumors in the NCI screening program.®-!9 However, the drug was not developed
`further dueto stability and solubility problems that prevented developmentof a parenteral
`formulation 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 manylaboratories have demonstrated that
`rapamycin, in contrast to FK506,is not only a potent immunosuppressant, but also an
`active antitumor agent. Rapamycin can actas a cytostatic, slowing or arresting cells in G,
`phase. Under specific conditions, or in some tumorcell lines rapamycins may induce
`apoptosis in culture. To date, studies have revealed that rapamycin potently arrests growth
`ofcells derived from rhabdomyosarcoma, neuroblastomaandglioblastoma, small cell lung
`cancer,!!-!7 osteoscarcoma,!® pancreatic cancer,!?2° breast and prostate cancer,2!-74
`murine melanoma and leukemia, and B-cell lymphoma.?2*?6 In addition to broad
`spectrum activity in vitro, rapamycin andits derivatives (designated here as rapamycins)
`suppress growth of some human and murine tumor models in vivo.!!-39 When combined
`with other chemotherapeutic agents, rapamycins generally showatleast additive antitumor
`activity.!0:12:17:31 Preliminary data from clinical trials have indicated that rapamycins are
`well tolerated and successfully suppress growth of various human tumors.3234
`The use of rapamycin as an anticancer drugis clinically impractical, because ofits poor
`water-solubility and stability in solution. Recently, rapamycin ester analogues (Fig. 1),
`CCI-779 [rapamycin-42, 2, 2-bis(hydroxymethy])-propionic acid]
`(Wyeth-Ayerst, PA,
`USA) and RADOO1 [everolimus, 40-O-(2-hydroxyethyl)-rapamycin]
`(Novartis, Basel,
`Switzerland), have entered clinical trials. These analogues have improved pharmaceutical
`properties. CCI-779 is being developed for both intravenous and oral administration,
`whereas RADO01 is only for oral administration. The antitumoractivity of these analogues
`
`Cancer Biology & Therapy
`
`2003; Vol. 2 Issue 3
`
`West-Ward Pharm.
`Exhibit 1018
`Page 001
`
`West-Ward Pharm.
`Exhibit 1018
`Page 001
`
`

`

`RAPAMYCINS: MECHANISM OF ACTION AND CELLULAR RESISTANCE
`
`is similar to that of rapamycin.!77!-2327-30 RADO01 is
`in Phase I trials whereas the development of CCI-779
`is more advancedwith several PhaseII 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 tumorcells may affect
`sensitivity to rapamycins. At least
`in some systems
`mutations that occur frequently in malignant trans-
`formations such as GLJ amplification, or mutations
`that inactivate p53, and the dual specificity phos-
`phatase PTEN (phosphatase and tensin homolog
`deleted on chromosometen, also known as MMAC1
`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 mTORincellular regulation, the mechanism
`of action of rapamycins, and currently understood
`resistance mechanisms.
`
`THE RAPAMYCIN TARGET (MTOR)
`
`LLLLLSLLLELLPLLLLL
`
` Rapamycin
`
`CCI-779
`
`Figure 1. The structure of rapamycin and its analogues FK506, CCI-779 and RADOO1. The
`FKBP12 binding face is shown by thefilled bars, whereas the mTOR binding face of
`rapamycin is shownby the hatched bar.
`
`The mammaliantarget of rapamycin, [also named
`FKBP12 and rapamycin-associated protein (FRAP),
`rapamycin and FKBP12 target 1 (RAFT 1), rapamycin
`target
`1
`(RAPT1), or sirolimus effector protein
`(SEP)],
`is a 289 kDa Ser/Thr kinase orthologue of
`TORI and TOR2 in Saccharomyces cerevisiae.>>*6
`TORis 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.3°38 Since the C-terminus ofTORis highly homologousto the
`catalytic domain of phosphatidylinositol 3’ kinase (PI3K), mTORis
`considered to belong to the PI3K-related protein kinase (designated
`PIKK) family, which also includes Mecl, Tell, RAD3, MEI-41,
`DNA-PK, ATM, ATR, and TRRAP?°37 Recently, single TOR
`homologshave also been identified in fungi (TOR1 in Cryptococcus
`neoformans), plants
`(AtTOR in Arabidopsis thaliana), worms
`(CeTORin Caenorhabditis elegans), and flies (ATOR in Drosophila
`menalogaster).”
`The domain structure of mTORis 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 PP2A 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.°*® The remarkable conservation of mTORatthe aminoacid
`level suggests that multiple domainsof 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.3”39*? The Tor signaling pathway in yeast
`is depicted in Figure 3, and controls translation initiation, protein
`turnover, transcription, andactin cytoskeleton organization. In yeast
`these pathwayshave been rigorously established,*” andareat least in
`part maintained in mammalian cells. The Torl/2 complex (desig-
`nated TORC1) comprising Kogl
`(the yeast homologue of the
`mammalian protein raptor) and Lst8 controls translation, protein
`stability and transcription,#3“> whereas the TORC2 complex
`controls actin organization. As TORC2is not a rapamycin targetit
`will not be considered further. The evolutionarily conserved TORC1
`complex controls translation initiation probably through activation
`of eIF4E, and transcription of ribosomal genes, stress response
`genes, ribosomal biogenesis and tRNAsynthesis.
`Tor, through the TORC1 complex,controls protein turnover and
`some aspects of transcription through regulation of protein phos-
`
`Figure 2. Schematic representation of the domains of 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 PlK-related kinases located N-terminal to
`the FKBP12-rapamycin binding domain (FRB);
`the role of
`FAT sequencesis less clear, but they are associated with
`C+erminal
`FAT (FATC)
`sequences
`in mTOR.
`Interaction
`between FAT and FATC domains mayfacilitate protein bind-
`ing or act asastructural scaffold; CD: Catalytic domain;
`RD: regulatory domain.
`
`
`|RO]}FATC
`
`2549 aa
`
`1| C
`
`www.landesbioscience.com
`
`ancer Biology & Therapy
`
`DIA
`
`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
`?
`
`
`Sale
`
`Translation
`
`rr
`
`= GD
`
`Le
`
`Actin
`organization
`
`protein turnover
`
`Transcription
`
`Figure 3. Nutrient signaling in yeast. (Adapted from Jacinto and Hall.37)
`
`phatases. Tor positively regulates Tap42, which binds to and inacti-
`vates the Sit4 and Pphprotein phosphatases.464” Pph21 and Pph22
`are calaytic subunits of PP2A,andSit4 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 someaspects of tran-
`scription appearto be regulated through Tap42 binding andinactivat-
`ing Sit4, whereas pathwaysregulated byrelease ofPph phosphatases are
`less well defined. In the presence ofnutrients Tor signaling represses
`autophagy andleads to stabilization of proteins by suppressing
`ubiquitin-dependent degradation.4® For example, Tor signaling
`prevents ubiquitylation, vacuolar targeting and degradation of the
`tryptophan transporter Tat2 by maintaining phosphorylation and
`inactivation ofNprl a putative Tat2 kinase.4?>° Under conditions of
`starvation Sit4 becomesactivated leading to dephosphorylation of
`Npr1 and degradation ofTat2. The Tap42-Sit4 pathwayalso 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.*94 The pathway(s) downstream of Tap42 involving Pph
`are less clear. Similarly, Tor negatively regulates the heterodimeric
`transcription factor Rtgl-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 ofstress-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 Bmh1 and Bmh2.°°
`Proximal Signaling in Mammalian Cells. In mammaliancells,
`mTORis regulated not only by nutrients but also by growth fac-
`tors.3”39-42 Tt appears that growth factors regulate mTORsignaling
`through both PI3K and Akt pathways, whereas proximal activators
`regulated by nutrients and ATPare less well characterized. In mam-
`malian cells mTORis activated as a consequenceofligand binding
`to various growth factorreceptors that result in activation of PI3K,
`Figure 4. Activated PI3K catalyzes the conversion ofphostidylinosi-
`tol (4,5)-bisphosphate (PIP2) to phosphatidylinositol-3,4,5-trispho-
`sphate (PIP3). This pathwayis negatively regulated by a dual-speci-
`ficity protein andlipid phosphatase PTEN. Full activation of Akt,
`downstream of PI3K requires binding of PIP3 to the pleckstrin
`homology domain of Akt, and phosphorylation by phosphoinositide-
`dependent kinases 1/2 (PDK1/2) and other unidentified kinases.
`Pharmacological studies with albeit relatively non-specific inhibitors
`of PI3K (wortmannin and LY294002) indicate that mTORis down-
`stream of PI3K. How mTORisregulated by PI3K or Akt, however,
`is still not well understood. Akt can phosphorylate mTOR
`(Ser2448) directly, although the significance remains to be deter-
`mined.3738 Recent studies have placed the tuberous sclerosis
`(TSC1/2) complex as a modulator between PI3K/Akt and mTOR.>?>4
`The TSC1/2 complex comprises harmartin (TSC1) and tuberin
`(TSC2). These proteins form a physical and functional complex in
`
`224
`
`Cancer Biology & 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.
`Conversely co-expression of TSC1 and TSC2 inhibits amino
`acid-induced activation of SGK1 in nutrient-deprived cells.>4
`Mitogenic stimuli, such as insulin or serum, activate Akt which can
`directly phosphorylate TSC2 on multiple sites in vitro and in
`vivo.>25556 Phosphorylation of TSC2 at Ser939 and Thr1462is
`PI3K-dependent.°2>° Akt-mediated phosphorylation of TSC2
`destabilizes TSC2 and thereby inhibits the formation of TSC1/2
`complex,
`leading to de-repression of mTOR, and consequently
`increasing the kinase activity of mTOR. By contrast, treatment with
`rapamycin does notinfluence the phosphorylation of TSC2,°? and
`together with other data,>>4 suggests that mTORlies downstream
`of TSC2. Other results?”°* 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 mTORand TSC1/2.
`mTORAssociated Proteins. mTOR forms a scaffold complex
`with other proteins, such as raptor (regulatory associated protein of
`mTOR) and mLST8,°™© which are the mammalian counterparts of
`yeast Kog] (kontroller of growth 1) and Lst8,°! respectively. The
`exactrole ofraptor remainsunclear.>8°?-6! Raptor mayactas a scaffold
`protein linking mTORto SGK1and 4E-BP1.°Alternatively, it may
`have a dual function, inhibiting mTOR under nutrient-deficient
`conditions andstimulating mTORina nutrient-replete environment.>?
`
`Thus TSC1/2, raptor and possibly mLST$8actas potential modulators
`of mTORfunction in responseto availability of nutrients.
`Signaling Distal to mTOR. mTORcontrols translation initiation
`through two pathways, S6K1 and eukaryotic initiation factor 4E
`(eIF4E) binding proteins (4E-BPs). mTOReitherdirectly phospho-
`rylates Thr389 of S6K1, or suppresses a phosphatase responsible for
`maintaining the hypophosphoryaltedstate ofthis residue. Activation
`of S6K1 enhances translation of mRNAs bearing a 5’
`terminal
`oligopyrimidine tract (5’ TOP).Inactivation of S6K1 decreases
`synthesis of ribosomal proteins and translation factors.°%
`Activation of S6K1 is complex. The process involves interplay
`between fourdifferent domainsandat least seven specific sites medi-
`ated by multiple upstream kinases.°4 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.% Atypical
`PKC isoforms and the Rho family of small G proteins (cdc42 and
`Racl) maycontribute to phosphorylation of S6K1,©although the
`specific sites regulated by these kinases remain to be determined. In
`vitro, mTOR phosphorylates only Thr389 in the regulatory
`domain.®”-However, whether this phosphorylation is directly or
`indirectly regulated by mTORis in question, since recent data suggest
`that mTOR may regulate S6K1 activation by inhibiting phos-
`phatases rather than directly phosphorylating S6K1.°7°
`
`www.landesbioscience.com
`
`Cancer Biology & 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) of4E-BP1 can be phosphorylated.©
`So far, only mTORand ATM havebeen identified to be involved in
`phosphorylation of 4E-BP1.7!-74 Other kinases that phosphorylate
`4E-BP1 remainto be characterized. ATM phosphorylates 4E-BP1 at
`Serl12,”4 however the physiological significance of this remains to
`be elucidated. In vitro mTOR phosphorylates 4E-BP1 at twosites
`(Thr37, Thr46) and possibly at two additional Ser/Thr residues
`(Thr70 and Ser65) in the N-terminal region.”*7° Phosphorylation
`is a hierarchical process.’3”>’7 Phosphorylation of Thr37/Thr46is
`followed by Thr70 phosphorylation. Ser65 is phosphorylated last”7
`and is dependent on phosphorylation ofall three Thr/Prosites.”>»”°
`Mutation ofThr37 and/or Thr46 toalanine(s) prevents phosphory-
`lation of Ser65 and Thr70,
`indicating that phosphorylation of
`Thr37 and Thr46serves as a requisite “priming” event.>° Single
`phosphorylation of these residues is not sufficient
`to dissociate
`4E-BP1 from elF4E,
`indicating the requirement of combined
`phosphorylation of at least Thr37, Thr46, Ser65, and Thr70 in
`4E-BP1 to suppress association with eIF4E.9%78 Inhibition of
`mTORleads to rapid hypophosphorylation of 4E-BP1 which then
`tightly binds to eIF4E. This prevents formation of elF4F complex
`that contains eIF4E, eIF4G, eIF4A and elIF3, and inhibits
`cap-dependenttranslation initiation.>° Once 4E-BP1 is hyperphos-
`phorylated, it releases eIF4E,facilitating e[F4F complex formation
`and promoting cap-dependentprotein synthesis.>° Overall inhibition
`of mTORby rapamycin leads to a decrease in protein synthesis of
`15 to 20 percent. However, as the elF4E pathway is required for
`translation of mRNA’s encoding cyclin D1,7%°° and ornithine
`decarboxylase®! inhibition of mTORleads to slowing or arrest of
`cells in G, phase ofthe cell cycle. However, the exact mechanism(s)
`by which mTORregulatescell 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 pathwayregulates trans-
`lation initiation of survival
`factors
`such as
`c-MYC®* and
`hypoxia-inducible factor 1a, and consequently vascular endothelial
`growth factor.8384 In addition, mTORis involved in the regulation
`of cyclin A, cyclin dependent kinases (cdk1/2), cdk inhibitors
`(p21C'P! and p27KiP!), retinoblastoma protein, RNA polymerase
`I/II/I1-transcription and translation of rRNA and tRNA,protein
`phosphatases (PP2A, PP4 and PP6), and CLIP-170.337.85-91
`
`TUMORSELECTIVITY OF RAPAMYCINS
`
`To date there are noreports suggesting that activating mutations
`of mTOR,or overexpression occur as primary events in malignant
`transformation. However, activation of signaling pathways both
`proximal anddistal to mTOR appearto occur frequently in human
`cancer. Loss of the phosphatase PTEN by deletion, silencing or
`mutation leads to constitutive activation of Akt,2%3%9? and upregu-
`lation of mTOR-dependent pathways. In PTEN-deficient tumor
`cells or mouse embryofibroblasts (MEFs), activated Akt is associat-
`ed with enhanced activity of S6K1 and hyperphosphorylation of
`4E-BP1,?9° or increased levels of c-MYC.”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 cancercells,
`multiple myelomacells and in MEFs.?39 The association of PTEN
`deficiency andsensitivity to rapamycinis further supported by the
`
`activity of CCI-779 against the growth of human tumors implanted
`in athymic nude mice.2339 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 independentvariable predicting
`for rapamycinsensitivity 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.?> These studies suggest that tumors
`developing in patients with tuberous sclerosis may besensitive to
`rapamycins. Oncogeneexpression mayalso regulate the response to
`rapamycin. For example, in RK3Ecells transformed with c-MYC or
`Ras rapamycin treatmentincreased global protein synthesis. In contrast
`rapamycin inhibited global protein synthesis and turnover in GLI
`transformed isogeniclines leading to inhibition ofproliferation.™4
`Although generally considered to be cytostatic agents, rapamycins
`can induce apoptosis in some cell systems. Rapamycins induce
`apoptosisof B-cells, rhabdomyosarcomacells, renal tubular cells and
`dendritic cells.1%9>°” Rapamycin enhances transforming growth
`factor-B induced cell cycle arrest,98 and through blocking survival
`factor signaling”? rapamycins enhancecell death. Ourresults 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
`p21! 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 inducesa cellular stress response characterized by rapid
`andsustained activation ofASK1 (apoptosis signal-regulating kinase
`1) signaling in p53-mutantcells.!°° This leads to sustained phos-
`phorylation of c-JUN (Ser63) that appears to be responsible for
`inducing apoptosis. Rapamycin-inducedstress appears distinct from
`that inducedbyultra violet radiation in that MEKK1is notactivated,
`and from cytotoxic agents such as DNA damaging agents where
`other stress pathways (p38 or ERK1/2) are also activated.!0!:!0? In
`contrast, cells expressing wild type p53,
`(or constitutive p21@P!
`expression)
`there is only transient activation of ASK1,!°
`Suppression of ASK1 is associated with binding of p21@P!
`in
`rapamycin treated cells, and protection from apoptosis. Since the
`rapamycin-induced apoptosis is specifically prevented by insulin-like
`growthfactors (IGF-I/II) and insulin,®© combination of IGF receptor
`inhibitors with rapamycins maybeselectively cytotoxic and induce
`regression of tumors with p53 mutations. Whether such anticipation
`is justified requires vigoroustesting.
`
`MECHANISMS OF RESISTANCE TO RAPAMYCINS
`
`Intrinsic sensitivity to rapamycins betweencell lines may vary by
`several orders of magnitude. For example, rhabdomyosarcomacells
`in vitro are very sensitive (concentration for 50% inhibition (IC50)
`of proliferation ~ 1 nM) comparedto colon carcinomacells (IC50 >
`5000 nM).!! Mechanismsofintrinsic and acquired resistance may
`have either a genetic or epigenetic basis.
`Mutations in FKBP12 and mTOR.Rapamycinsfirst 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 & 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
`
`Synthetic lethality
`a genetic defect alters cellular response to drug action
`MEFs
`
`
`
`o 1a es
`
`2cc
`
`Se=
`
`1cw
`s ue
`Be
`Ey
`
`p53"+
`
`Rapamycin|
`
`Alive
`
`p53"
`
`Rapamycin|
`
`
`Dead
`
`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 mTORthatblock binding
`of FKBP-rapamycin complex to mTOR. Such
`mutations werefirst identified in budding yeast
`S. cerevisiae in which treatment with rapamycin
`irreversibly arrests cells in the G, phase. In the
`yeast S. cerevisiae, 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.!°4
`This observation has been further confirmed by
`RBP1 disruption experiments using the patho-
`genic
`yeast Candida
`albicans. Wild-type
`RBP1/RBP1 parental strain and the rbp1/RBP1
`heterozygous mutantweresensitive to rapamycin
`inhibition, whereas
`rbp1/rbp1 homozygous
`mutantwas rapamycin resistant.!In addition, in
`S. cerevisiae mutation ofa specific residue (Tyr89)
`which is conserved in RBPJ or FKBPs, also
`resulted in decreased binding of rapamycin and
`conferred a recessive resistance phenotype.!° In
`murine mastcells, two distinct point mutations
`in FKBP12 confer
`resistance. By altering a
`hydrophobic residue within the drug-binding
`pocket (Trp59Leu) or changing a charged. sur-
`face residue (Arg49>Gln), the bindingaffinity for
`rapamycin decreases substantially.!6
`A genetic screen identified rapamycin-resist-
`ant alleles with mutations in genes designated
`TORI and TOR2. Strains with mutatedto tor]-1
`(Ser1972>Arg) and tor2-1 (Ser1975>Arg), were
`completely resistant
`to the growth- inhibitory
`effect ofrapamycin. Theseresistantalleles encode mutantTorproteins
`that do not bind the FKBP-rapamycin complex.!03:107-111 This
`result suggests that a conserved serine residue (Ser1972 in Torl;
`Ser1975 in Tor2) in Tor proteinsis critical for FKBP-rapamycin
`binding. In mammalian cells mutations in the FRB domain confer
`a dominantresistant phenotype consistent with decreased affinity
`for binding of the FKBP- rapamycin complex.'!! Expression of a
`mutant mTOR(Ser2035 lle), having greatly reduced bindingaffin-
`ity for the FKBP-rapamycin complex, confers high level resist-
`ance, 14112,113
`Deregulation of eIF4E. mTORphosphorylates and regulates the
`function of 4E-BP1, the suppressor of e[F4E.2° Recently, our group
`has found that acquired resistance to rapamycin wasassociated with
`decreased levels of 4E-BP1.!?! In the absenceofselective pressure
`(rapamycin), resistance was unstable andcells 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-BP1
`levels increased to thosein wild type cells. Levels of 4E-BP1 transcripts
`were unaltered in rapamycinresistant clones suggesting post-tran-
`scriptional regulation. Further studies indicate that the synthesis of
`4E-BP1 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 p27*P! in
`rapamycin resistant BC3H cells.!!> In contrast, no consistent
`changes were detectedin thelevel or activity of S6K1 between parental
`
`10 1 1"
`10"
`a0" 40"
`me jo"
`Annexin V-FITC Fluorescence
`
`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 p537 MEFs, and p537 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
`quadrantis presented (from Huanget al. 20014).
`
`andresistant clones. Rapamycin also inhibited growth factor activa-
`tion of S6K1 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 HCT8colon carcinomacells
`that are highly resistant to rapamycin (IC50 > 10,000 ng/ml). When
`4E-BP1 is overexpressed, these cells becomesensitive (IC50 < 10
`ng/ml) to rapamycin, Figure 6.'!4 These data suggest that low levels
`of 4E-BP1 results in de-regulation of eIF4E, conferring rapamycin
`resistance.
`These results suggest that rapamycin-regulation of the elF4E
`pathwayis crucial in inducing growth arrest. Further de-regulation
`of eIF4E mayfacilitate a malignant phenotype. Of interest is that
`both rapamycin-resistant and -revertant cells exhibited elevated
`c-MYClevels, and increased anchorage-independent growth. That
`deregulation of the e[F4E pathway is associated with increased
`malignancy is supported bycertain clinical observations that dereg-
`ulation of the e[F4E pathway does promote tumor progression.!1°
`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,!!? and peripheral carcinomaofthe lung!?°
`elF4E levels are elevated. However, levels of 4E-BP1 suppressor
`proteins have not been reported in a consistent manner. Potentially,
`the ratio of 4E-BP:eIF4E may determine whether inhibition of
`mTORelicits 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 & Therapy
`
`227
`
`West-Ward Pharm.
`Exhibit 1018
`Page 006
`
`West-Ward Pharm.
`Exhibit 1018
`Page 006
`
`

`

`RAPAMYCINS: MECHANISM OF ACTION AND CELLULAR RESISTANCE
`
`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 ofseveral other sites dependent on phospho-
`rylation ofThr389,rapamycin in vivo influences phosphorylation
`ofothersites, including Thr229 in thecatalytic domain, and $404
`in the regulatory domain.” Mutation ofThr389>Glu abrogates the
`ability of rapamycin to inhibit S6K1 activation.o”!2! Similarly,
`substitution of Thr229 by either a neutral amino acid Alanine
`(Thr229>Ala) or by an acidic amino acid Glu (Thr229>Glu),
`renders S6K1 insensitive to rapamycin.!?? In addition, deletion of
`the 77 N-terminal codons (AN77) confers rapamycin resistance.!?4
`Ofnote truncationofthe first 54 residues of N-terminus blocks the
`serum-induced phosphorylation of three rapamycin-sensitive sites,
`Thr229, Thr389 and Ser404, causing rapamycin insensitivity.°”
`Whetherthis results in resistance to the growth inhibitory effect of
`rapamycinis less clear, and maybecell context specific.
`Mutations of PP2A-Related Phosphatases. The regulation of
`protein phosphataseactivity is thoug

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