throbber

`
`American Society of Clinical
`Oncology
`Educational Book
`36th Annual Meeting
`Spring 2000
`
`EDITOR
`Michael C. Perry, MD, Columbia, MO
`
`Editorial Projects Manager
`Rich Harrington, Chestnut Hill, MA
`
`Editorial Assistant
`Lindsay McOmber, Chestnut Hill, MA
`
`© 2000 American Society of Clinical Oncology
`Alexandria, VA
`
`West-Ward Exhibit 1016
`Dancey 2000
`Page 001
`
`

`

`Rapa:m.ycin - S ensi ti ve Signal-Transduction
`Pathways: Protein Translation Control of
`Cell Proliferation
`
`By Janet E. Dancey, MD
`
`Abstract: Rapamycin Is a natural product with antimi(cid:173)
`crobial, immunosuppressant, and antitumor activities
`owing to its modulation of signal-transduction pathways
`linking mitogenic stimuli to the synthesis of specific
`proteins needed for cell-cycle progression from the G1 to
`the 8 phase. Rapamycin and its soluble analog CCI-779
`have in vitro and in vivo anti proliferative activity against
`a broad range of human tumor-cell lines, and CCI-779 is
`just entering clinical trials to determine Its value in
`cancer therapy. The pharmacologic action of rapamycin
`is mediated through its binding to the intracellular pro(cid:173)
`tein FK506 binding protein 12 and subsequent inhibition
`of the protein kinase mammalian target of rapamycin
`(mTOR). mTOR signals to two separate pathways, each of
`which controls the translation of specific mRNAs. One
`rapamycin-sensitive pathway affects the activity of the
`
`C ELL PROLIFERATION requires the synthe(cid:173)
`
`sis of proteins necessary for entry into and
`transit through the cell cycle. 1 Thus, regulatory
`mechanisms to increase translation of these pro(cid:173)
`teins are necessary to promoting cell prolifera(cid:173)
`tion. As an anticancer treatment strategy, target(cid:173)
`ing translational regulation has been a relatively
`un exploited area of therapeutic development.
`However, the identification of the antiprolifera(cid:173)
`tive effects of the agent rapamycin (sirolimus)
`and the identification of its novel mechanism of
`action have renewed interest in targeting the
`translational regulatory apparatus as a therapeu(cid:173)
`tic strategy.
`Rapamycin, a natural product, has antimicro(cid:173)
`bial, immunosuppressant, and antitumor activi(cid:173)
`ties that result from the modulation of signal
`transduction pathways that link mitogenic stim(cid:173)
`uli to the synthesis of specific proteins needed for
`cell-cycle progression from the Gl to the S phase.
`
`Address correspondence to Janf't Dancey, MD. Investiga(cid:173)
`tionezZ Drug Branch, Cancel' Treatment Evaluation Program,
`Division ofCelTlcer Treatment and Diagnosis, National Cancer
`Institute, EPN 715, 6130 Executive Blvd, Rockville, MD 20854;
`email danceyj@ctep.nci_ nih.goll.
`© 2000 by American Society of Clinical Oncology.
`1092-9118/00/68-7.5
`
`68
`
`408 ribosomal protein 86 kinase p70s0k, and the other
`affects the function of eukaryotic initiation factor 4E(cid:173)
`binding protein-1 (4E-BP1), also known as phosphory(cid:173)
`lated heat- and acid-stable protein, The observations by
`several groups that the inhibition of mTOR- mediated
`p70 sek and 4E-BP1 phosphorylation by rapamycin were
`coupled to growth arrest in the G1 phase led to the
`hypothesis that the anti proliferative properties of rapa(cid:173)
`mycin result from its effects on the regulation of protein
`translation affected by these targets. However, the pre(cid:173)
`cise mechanisms of cell-cycle arrest are as yet unknown.
`This review will focus on recent advances in the under(cid:173)
`standing of the mechanisms by which rapamycln inhibits
`cell growth and the issues surrounding the development
`of this type of agent as a potential treatment for cancer.
`
`Rapamycin is in late phase III clinical trials as an
`immunosuppressive drug for organ and bone mar(cid:173)
`row transplant recipients, and its ester analog,
`CCI-779, is being evaluated in early clinical trials
`as a therapeutic agent against cancer. The immu(cid:173)
`nosuppressant effects of rapamycin result from
`its inhibition of the biochemical events required
`for the progression ofinterleukin-2-stimulated T
`cells from the Gl to the S phase of the cell cycle.:.!
`However, the growth-inhibitory actions of rapa(cid:173)
`mycin and its analog are not restricted to lym(cid:173)
`phoid cells, as these agents also have cytostatic or
`cytotoxic activities against solid tumor cell lines.
`In addition to its possible clinical utility, rap amy(cid:173)
`cin is a useful pharmacologic probe for studies of
`the signal-transduction pathways that govern
`translation. This review will focus on recent ad(cid:173)
`vances in the understanding of the mechanisms of
`cell-growth inhibition by rapamycin and the is(cid:173)
`sues surrounding the development of this class of
`agent as a potential mode of therapy in the
`treatment of cancer.
`
`THE DISCOVERY OF RAPAMYCIN AND ITS
`ANTIPROLIFERATIVE ACTIVITY
`
`Rapamycin, a macrolide, was first identified as
`a fungicide produced by the bacteria Streptomyces
`hygroscopicU8, which had been isolated from soil
`samples from Easter Island. 3
`,4 Although it was
`
`West-Ward Exhibit 1016
`Dancey 2000
`Page 002
`
`

`

`CANCER THERAPIES TARGETING TRANSlATION
`
`originally identified as an antifungal agent, sub(cid:173)
`sequent studies demonstrated impressive antitu(cid:173)
`mor and immunosuppressant activities. Rapamy(cid:173)
`cin was originally evaluated by the National
`Cancer Institute (NCD in the late 1970s. At that
`time, it was found to have antiproliferative activ(cid:173)
`ity in a variety of murine tumor systems, includ(cid:173)
`ing B16 melanoma and P388 leukemia models.5.6
`Rapamycin has since been shown to inhibit the
`growth of B-celllymphoma celllines,7 and small(cid:173)
`cell lung cancer cell lines.8 In addition to its
`growth-inhibiting effects, rapamycin inducedp53
`independent apoptosis of childhood rhabdomyo(cid:173)
`sarcoma cell lines. 9 Treatment with rapamycin
`inhibited cyclin Dl expression and proliferation of
`MiaPaCa-2 and Panc-l human pancreatic cancer
`cell lines. 10 Rapamycin also augmented cisplatin(cid:173)
`induced apoptosis in murine T-cell lines, the hu(cid:173)
`man promyelocytic cell line HL-60, and the hu(cid:173)
`man ovarian cancer cell line SKOV3,u These
`data suggest that the anti proliferative action of
`rapamycin may be an important component of the
`pathway that prevents cell death and that rapa(cid:173)
`mycin may enhance the efficacy of selected cyto(cid:173)
`toxic agents.
`
`THE TARGET OF RAPAMYCIN
`Our understanding of molecular mechanisms
`underlying the biologic effects of rapamycin has
`advanced considerably in recent years (Fig 1).
`Similar to other natural immunosuppressants
`such as cycIosporine and FK506, rapamycin binds
`to members of the ubiquitous immunophilin fam(cid:173)
`ily of FK506 binding proteins (FKBPs), inhibiting
`their enzymatic activity as prolyl isomerases. 12,13
`Although this enzymatic function is important for
`altering protein conformation, it is, surprisingly,
`not relevant to the action of rapamycin.14 Al(cid:173)
`though there are many members of the FKBP
`family, biochemical and genetic studies suggest
`that FKBP12 is the most relevant binding protein
`for the pharmacologic activity of rapamycin in
`eukaryotic cells. Yeast mutants lacking FKBP12
`are viable and resistant to rapamycin toxicity,
`indicating that both the protein and the drug are
`required for rapamycin action. 14 Overexpression
`of FKBP12 in mammalian cells increases their
`sensitivity to rapamycin, and cell lines with re(cid:173)
`duced levels of FKBP12 are rapamycin-resistant,
`providing further evidence for a model in which
`the cellular effects of rapamycin result from its
`
`69
`
`BAD
`1
`Apoptosis
`
`mTOR
`
`I- [ Rapamycin~D
`
`PI-3 kinase I- PTEN
`~
`... - - PKBfAkt
`1
`/~
`I
`\
`
`4E-BP-IIPI-IAS-1
`
`1'70 S6 kinase
`
`Translation
`
`GI
`Fig 1. Rapamycin-sensitive signaling pathways. The target
`of rapamycin kinase (TOR) functions to regulate the activities of
`the translational regulators 4E-BP 1 /PHAS-. and p70 S6 kinase.
`Rapamycin binds to FKBP12 and the complex inhibits mTOR.
`
`binding to FKBP12. 15 Thus, rapamycin may ac(cid:173)
`tually be considered a "pro drug" for the active
`agent at the cellular level, the FKBP12-rapamy(cid:173)
`cin complex.
`The target of the rapamycin-immunophilin
`complex was initially identified in yeast and con(cid:173)
`firmed in mammalian cells. In mammalian cells,
`the complex interacts with a large polypeptide
`kinase of 290 kd, termed mammalian target of
`rapamycin (mTOR)16 (also known as FRAP,17
`RAFTl,18 and RAPT1 19
`), blocking its activity.
`The yeast TOR proteins exhibit a high degree of
`overall sequence identity (> 40%) to mTOR, with
`even greater identity (> 65%) observed in their
`carboxy-terminal catalytic domains (reviewed
`in2o). The high level of sequence conservation,
`together with the strikingly similar effects of
`rapamycin on yeast and mammalian cell growth,
`suggests that TOR proteins are important to cell
`function because they have been highly conserved
`during eukaryotic evolution.
`Yeast and mammalian TOR proteins are mem(cid:173)
`bers of a recently described family of protein
`
`West-Ward Exhibit 1016
`Dancey 2000
`Page 003
`
`

`

`70
`
`kinases called phosphoinositide 3 kinase (PI3K)(cid:173)
`related kinases (PIKKs). Members of this family
`are involved in a range of essential cellular func(cid:173)
`tions, including cell-cycle progression, cell-cycle
`checkpoints, DNA repair, and DNA recombina(cid:173)
`tion.21 ,22 Among these PIICK family members are
`the cell-cycle regulatory protein kinases ataxia
`telangiectasia mediated, ataxia telangiectasia re(cid:173)
`lated, and DNA-dependent protein kinase cata(cid:173)
`lytic subunit. In contrast to the TOR proteins,
`ataxia telangiectasia mediated, ataxia telangiec(cid:173)
`tasia related, and DNA-dependent protein ki(cid:173)
`nases participate in cell-cycle checkpoints that
`govern cellular responses to DNA damage.21 The
`PIKK family members share a carboxy-terminal
`catalytic domain that bears significant sequence
`homology to the lipid kinase domains of PI3Ks,
`although no intrinsic lipid kinase activity has
`been described for mTOR.
`
`REGULATION OF PROTEIN SYNTHESIS
`BY mTOR
`Stimulation of quiescent cells with growth fac(cid:173)
`tors leads to a dramatic increase in the transla(cid:173)
`tion of a subset of mRNAs whose protein products
`are required for progression through the G I
`phase of the cell cycle. 23 mTOR regulates key
`pathways affecting the efficiency of protein trans(cid:173)
`lation, The upstream signaling pathway that cou(cid:173)
`ples growth-factor-receptor occupancy to mTOR
`protein activation is only partially understood.
`mTOR is a phosphoprotein, and its phosphoryla(cid:173)
`tion state, as well as its catalytic activity, have
`been reported to be modulated by the mitogen(cid:173)
`activated phosphatidylinosol (PI) 3 kinase-Pro(cid:173)
`tein Kinase B (PKB)/Akt. 24
`,25 PI3 kinase and Akt
`are considered to be proto-oncogenes, and the
`pathway is inhibited by the tumor suppressor
`genePTEN. 26 Although other signaling pathways
`are activated downstream of PI3K, the Akt path(cid:173)
`way is of particular interest because of its role in
`inhibiting apoptotic pathways and promoting cell
`proliferation (reviewed in26,27). In mammalian
`cells, activated mTOR signals to two separate
`pathways that control translation of specific sub(cid:173)
`sets of mRNAs. These are the 40S ribosomal
`protein S6 kinase, p70s6 k,8 and the eukaryotic
`initiation factor (eIF)-4E-binding protein-I C4E(cid:173)
`BPI), also known as PHAS-I (phosphorylated
`heat- and acid-stable protein).28,29 Among subset
`mRNAs regulated by these pathways are those
`
`JANET E. DANCEY
`
`encoding components of the protein synthesis
`machinery itself.
`Several recent reports indicated that activation
`of either PI3K or Akt is sufficient to induce the
`phosphorylation of both p70s6k and 4E-BPI/
`PHAS-I through mTOR.30,31 Treatment of acti(cid:173)
`vated PI3K or Akt expressing cells with rap amy(cid:173)
`the p70s6k and 4E-BPl/PHAS-I
`cin blocks
`phosphorylation, indicating that mTOR is re(cid:173)
`quired for these responses.29,32 In addition, there
`is evidence that Akt phosphorylates the carboxyl
`terminus of mTOR, contributing to its activa(cid:173)
`tion. 24,25 These results clearly link the PI3K-Akt
`pathway to p70s6k and 4E-BPl/PHAS-l transla(cid:173)
`tional control pathways through mTOR.
`Compared with its upstream effectors, the
`downstream actions of mTOR on translation are
`more fully characterized. For the subset of mR(cid:173)
`NAs that contain regulatory elements located in
`the 5'-untranslated regions, the binding of the
`mRNA to the ribosomal subunit and the efficient
`initiation of translation is mediated by the mul(cid:173)
`tisubunit eukaryotic initiation factor-4 (eIF-4l
`complex.33 4E-BPl/PHAS-I is a low-molecular(cid:173)
`weight protein that inhibits the initiation of
`translation through its association with eIF-4E,
`the mRNA cap-binding subunit of the elF -4F
`complex.23 Binding of 4E-BPs to eIF-4E is depen(cid:173)
`dent on the phosphorylation status of 4E-BP. In
`quiescent cells, 4E-BPI/PHAS-I is relatively un(cid:173)
`derphosphorylated and binds tightly to elF -4E
`(reviewed in34
`). Stimulation of cells by hormones,
`mitogens, growth factors, cytokines, and G-pro(cid:173)
`tein-coupled agonists results in 4E-BPl/PHAS-I
`phosphorylation through the action ofmTOR and,
`possibly, other kinases, which promotes the dis(cid:173)
`sociation of the 4E-BPI/PHA8-I-eIF-4E complex
`(Fig 2). eIF-4E can then bind to the eIF-4F com(cid:173)
`plex, and this interaction will then lead to an
`increase in translation rates. Conversely, growth(cid:173)
`factor deprivation or treatment with rapamycin
`results in 4E-BPlIPHAS-l dephosphorylation, an
`increase in eIF-4E binding, and a concomitant
`decrease in translation.34
`The second downstream target of mTOR is
`p70s6k, the kinase that phosphorylates the 40S
`ribosomal protein S6. In response to mitogenic
`stimuli, p70s6k phosphorylates 86 on multiple
`sites, and these modifications favor the recruit(cid:173)
`ment of the 40S subunit into actively translating
`polysomes35 and enhance the translation of mR-
`
`West-Ward Exhibit 1016
`Dancey 2000
`Page 004
`
`

`

`CANCER THERAPIES TARGETING TRANSlATION
`
`71
`
`I ~;~t~~ / Rapamycin ---£rJ
`~,:;:\
`[4i~~
`~ /
`13~~!J)@ ~@ I ActiveeIF4EI
`1
`(~'j1E£AUG - AAAAA
`
`[I~11Ctiv~~
`
`-,_±~1-,/
`[eIF4F Compi;] --~ ~~~ation ofTransla~
`Fig 2. Activation of mTOR leads to the phosphorylation of
`4E-BP l/PHAS·l. eIF-4E is released and then binds to the eIF-4G
`scaffolding protein of the eIF-4F complex at the 5' cap site of the
`mRNA template. The eIF·4F complex contains a 7·methyl(cid:173)
`guanosine cab-binding subunit called eIF-4E, an RNA helicase
`called eIF-4A, and a multifunctional scaffolding protein eIF-4G
`that bridges the 405 ribosome and the mRNA, while also
`binding eIF-4E, eIF-4A, e1F-3, and the poly(A)-binding protein.
`
`NAs bearing 5' terminal oligopolypyrimidine
`tracts. Although these transcripts represent only
`100 to 200 genes, they can encode up to 20% of the
`cell's mRNA. 86
`Rapamycin treatment triggers the rapid de(cid:173)
`phosphorylation and inactivation ofp70s6k in mi(cid:173)
`togen-stimulated cells. Although p70s6k activa(cid:173)
`tion involves a complex series of phosphorylation
`events catalyzed by multiple protein kinases, the
`prompt inhibitory effect of rapamycin suggests
`that persistent stimulatory input from mTOR
`leads to the activated state of p70s6k.36 The exact
`nature of the input supplied by mTOR is unclear;
`however, a recent study suggested that the mTOR
`phosphorylates and suppresses the activity of a
`type 2A protein phosphatase bound directly to
`p70s6k.87
`The observations by several groups that the
`inhibition of mTOR-mediated p70s6k and 4E-BPl
`phosphorylation by rapamycin were coupled to
`growth arrest led to the hypothesis that the
`anti proliferative properties of rapamycin are a
`-40
`result of its effects on translational contro1.38
`Inhibition of these key signaling pathways results
`in the inefficient translation of the mRNAs of
`proteins such as cyclin D141 and ornithine decal'(cid:173)
`boxylase,42 which are important for cell-cycle pro(cid:173)
`gression through the G1 phase. However, in ad(cid:173)
`dition to its actions on p70s6k and 4E-BPll
`PHAS-1, rapamycin prevents cyclin-dependent
`
`kinase activation and retinoblastoma protein
`(pRb) phosphorylation.43-46 Rapamycin
`also
`seems to accelerate the turnover of cyclin Dl,
`both at the mRNA and protein levels, resulting in
`a deficiency of active cdk4lcyclin Dl complexes
`required for pRB phosphorylation and the release
`of E2F transcription factor and increased associ(cid:173)
`ation of p27kip1 with cyclin E/cdk2. These two
`events, along with the inhibition of translation of
`other mRNAs, can certainly explain the observed
`inhibition at the GllS phase transition.40,47 How(cid:173)
`ever, cells derived from mice in which the p27
`gene has been disrupted by homologous recombi(cid:173)
`nation are only partially rapamycin-resistant, in(cid:173)
`dicating that rapamycin can inhibit cell-cycle pro(cid:173)
`gression by p27-independent mechanisms.48
`Whether the effects of rapamycin on cyclin cdks
`and cdk inhibitor p27 are mediated through its
`inhibition of translation remains to be defined
`with precision in different cell types. Thus, al(cid:173)
`though the direct target of rapamycin has been
`identified, the downstream pathway from the
`target to the inhibition of cell-cycle progression
`requires further study.
`
`CLINICAL DEVELOPMENT
`Unfortunately, poor aqueous solubility and in(cid:173)
`stability compromised the development of rapa(cid:173)
`mycin as an anticancer agent. However, the NCI,
`in collaboration with Wyeth-Ayerst, examined
`several derivatives of rapamycin and selected one
`agent, CCI-779, for further development, on the
`basis on its mechanism of action and favorable in
`vitro and in vivo efficacy and toxicity data.
`CCI-779 is a soluble ester analog ofrapamycin
`with impressive in vitro and in vivo cytostatic
`'activity. Results from the NCI human tumor cell
`line screen showed that CCI-779 and its parent
`compound, rapamycin, share a mechanism of ac(cid:173)
`tion that is distinct from those of other cancer
`therapeutic agents. The two agents are similar:
`the Pearson correlation coefficient of the in vitro
`anti proliferative activities and potencies of the
`two agents across the 60-cellline screen is .86. In
`vitro, human prostate and breast cancer lines as
`well as CNS, melanoma, small-cell lung carci(cid:173)
`noma, and T-cell leukemia human tumor lines
`were among the most sensitive to CCI-779 with a
`50% inhibitory concentration of less than 10- 8
`M.49 Platelet-derived growth factor stimulation of
`the human glioblastoma line T98G was markedly
`
`West-Ward Exhibit 1016
`Dancey 2000
`Page 005
`
`

`

`72
`
`inhibited (50% inhibitory concentration of ap(cid:173)
`proximately 1 pM), consistent with its proposed
`mechanism of action as an inhibitor of signal(cid:173)
`transduction pathways. Growth-inhibited cells
`were arrested in the G 1 phase, and growth-inhib(cid:173)
`itory effects were blocked by the FKBP inhibitory
`molecule ascomycin, suggesting that the mecha(cid:173)
`nism of action of CCI-779 is similar to that of
`rapamycin. 49 In vivo human tumor xenograft
`studies showed significant tumor growth inhibi(cid:173)
`tion, suggesting that CCI-779 should be devel(cid:173)
`oped as a cytostatic rather than a cytotoxic agent.
`Several intermittent dosing regimens of CCI-779
`were effective in these animal models. These
`findings are important because preclinical studies
`have shown that when given intermittently, the
`immunosuppressive effects of CCI-779 resolve
`within 24 hours after the last dose.
`Given its proposed properties as a cytostatic
`agent, CCI-779 may be of value in delaying time
`to tumor progression and in increasing survival in
`patients when used alone or in combination with
`other anticancer agents. Two schedules are cur(cid:173)
`rently being evaluated in phase I trials: a weekly
`schedule and a daily for 5 days every 2 weeks
`schedule. Both phase I trials were designed with
`the traditional phase I objectives of determining
`the maximum-tolerated dose and dose-limiting
`toxicities of the agent. Preliminary results from
`the phase I study of the weekly 30-minute infu(cid:173)
`sion have recently been reported. 50 Twelve pa(cid:173)
`tients treated at dose levels of 7.5 mg/m2 to 60
`mg/m2/wk did not experience dose-limiting toxic(cid:173)
`ities. Interestingly, one partial response and one
`minor response were observed in two patients
`with renal cell carcinoma and lung metastases
`treated at 15 mg/m2
`. Only mild grade 1-2 skin
`reactions and mucositis were seen. The range of
`skin reactions has been described as dryness,
`urticaria, eczema-like lesions, and erythematous
`papules that did not seem to worsen with re(cid:173)
`peated dosing. Because CCI-779 is structurally
`similar, some of the adverse effects reported in
`clinical trials of rapamycin may also be seen
`during the clinical evaluation ofCCI-779. Among
`the most common adverse events that occur as a
`result of oral dosing of rapamycin are hyperlipid(cid:173)
`emia; elevated lactate dehydrogenase levels; hy(cid:173)
`pophosphatemia; hypokalemia; reduced WBC,
`RBC, and platelet counts; stomatitis; and arthral-
`
`JANET E. DANCEY
`
`gias. Phase II studies of a broad range of tumor
`types, sponsored by the NCI and Wyeth-Ayerst,
`will be initiated once phase I studies have been
`completed.
`The observation that CCI-779 induced tumor
`regression in patients treated at relatively non(cid:173)
`toxic doses on the phase I study is particularly
`noteworthy. On the basis of the preclinical re(cid:173)
`sults, it was thought that CCI-779 would act as a
`cytostatic agent, and phase I studies were de(cid:173)
`signed with the traditional objectives of determin(cid:173)
`ing maximum-tolerated dose and dose-limiting
`toxicities. IfCCI-779 is biologically active at lower
`doses, it may not be necessary to treat patients
`with higher doses of this agent that place them at
`risk for toxicity. Ideally, once the biologically
`effective dose to affect the rapamycin target(s) is
`achieved, further dose escalation is unnecessary.
`Defining and limiting drug concentrations to the
`biologically effective range would prevent cross(cid:173)
`reactions with other molecules that cause toxicity
`at higher drug concentrations. In addition, mea(cid:173)
`suring the drug's effect on its target(s) could
`potentially be used as a surrogate efficacy end
`point, assuming there is a strong correlation
`between target modulation and cell survival 01'
`proliferation.
`Unfortunately, determining the optimal biolog(cid:173)
`ically active dose in patients is problematic. Pre(cid:173)
`clinical studies using animal models could define
`plasma and tumor drug concentrations that per(cid:173)
`turb the important cellular target(s) of rapam(cid:173)
`cyin. In turn, the drug's effect on its targets(s)
`should be correlated with any antiproliferative or
`apoptotic effects. Although preclinical studies can
`be helpful in developing assays of target effects
`and defining active drug concentrations, the opti(cid:173)
`mal dose, in principle, should be defined in hu(cid:173)
`man tumor tissue. In practice, this requires a
`valid and reliable assay of the drug's effect on the
`intended target and that patients with accessible
`lesions agree to undergo repeated biopsies. Al(cid:173)
`though these requirements increase the complex(cid:173)
`ity and cost of drug development, efforts to ac(cid:173)
`quire these data can, nonetheless, be amply
`rewarded because information resulting from
`such studies may facilitate subsequent clinical
`development. For CCI-779, assays to determine
`the 4E-BPI/PHAS-1 and/or p70s6k phosphoryla(cid:173)
`tion status may be helpful in defining a pharma-
`
`West-Ward Exhibit 1016
`Dancey 2000
`Page 006
`
`

`

`CANCER THERAPIES TARGETING TRANSLATION
`
`cologie ally active dose. However, it is possible
`that effects observed at these molecular targets
`may not correlate with antiproliferative effects.
`In fact, there is evidence that cell-cycle progres(cid:173)
`sion and translation can proceed despite the de(cid:173)
`phosphorylation of 4E-BPIIPHAS-I and the inac(cid:173)
`tivation ofp70,,6k by rapamycin. 51 52 These results
`suggest that either these pathways are not the
`only mechanisms by which cell-cycle progression
`is regulated, or-under certain conditions(cid:173)
`mTOR-4E-BPIIPHAS-1 and mTOR_p70s6k path(cid:173)
`ways are redundant. 51,52 Thus, assessing drug
`effects by use of these targets may assist re(cid:173)
`searchers in determining a pharmacologically ac(cid:173)
`tive dose but may not predict drug efficacy either
`because the assays are assessing targets that are
`not related to drug effects on proliferation or
`because downstream factors are rendering the
`cells resistant. In the absence of data, pharmaco(cid:173)
`kinetic studies may be helpful because doses that
`provide concentrations in humans which are ac(cid:173)
`tive in preclinical models are likely to be effective.
`In addition to a definition of the appropriate
`dose, choosing the appropriate efficacy end point
`for phase II studies is also problematic. Preclini(cid:173)
`cal data suggested that CCI-779 would prevent or
`delay the growth of tumors rather than induce
`tumor regressions. Solely on the basis of the
`preclinical results, one could argue that efficacy
`end points other than response should be used in
`phase II trials of CCI -779. Although it is possible
`that by reducing the rate of cell proliferation
`relative to the rate of apoptosis could lead to a
`reduction in tumor mass over time, cytostatic
`agents are more likely to cause prolonged disease
`stabilization rather than objective tumor re(cid:173)
`sponse. Possible surrogate phase II end points
`that have been proposed for the evaluation of
`other cytostatic agents include time to progres(cid:173)
`sion, changes in tumor markers, target inhibition,
`and positron emission tomography scan-assayed
`indices of cell proliferation, as well as the propor(cid:173)
`tion of patients with early disease progression
`and an assessment of clinical benefit (reviewed
`in53
`). Unfortunately, none of these proposed end
`points has been shown to correlate with patient
`benefit.
`Given our current understanding of the mech(cid:173)
`anism of action of rapamycin, a number of hy(cid:173)
`potheses regarding the molecular abnormalities
`that may correlate with the efficacy of CCI-779
`
`73
`
`can be generated. On the basis of preclinical
`results in studies of glioma,49 small-cell carcino(cid:173)
`ma, 8 and rhabdomyosarcoma, 9 tumors that rely
`on paracrine or autocrine stimulation of receptors
`that trigger the PI3K1AktimTOR pathway or tu(cid:173)
`mors with mutations that cause constitutive acti(cid:173)
`vation of the PI3/ Akt pathway may depend on
`rapamycin-sensitive pathways for growth. In fact,
`abnormal activation of this pathway is relatively
`common because mutations of the tumor-suppres(cid:173)
`sor gene PTEN, which encodes for a lipid phos(cid:173)
`phatase that inhibits PI3K-dependent activation
`of PKB/Akt, occur in multiple tumor types with a
`frequency approaching that of p53. 26 Deletion or
`inactivation of PTEN results in unregulated Akt
`activity. Thus, the presence of PTEN mutations
`may also predict for the activity of CCI-779.
`Because CCI-779 seems to induce cytostasis by
`preventing progression through the G 1 phase, it
`is possible that abnormalities of regulators of the
`Gl checkpoint such as pRB, p16, p27, and cyclin
`D may predict for drug efficacy. The parent com(cid:173)
`pound, rapamycin, affects the efficiency with
`which cdks are activated by alteration of the
`expression of the cyclin D subunit.4o Because p16
`inhibits the cyclin n-cdk4/6 phosphorylation of
`pRb required for progression through the G 1
`phase, loss of p16 results in unregulated cyclin
`D/cdk activity. Decreasing cyclin D might reintro(cid:173)
`duce a cdk-inhibitory effect and arrest the cell
`cycle. Defining the molecular characteristics of
`tumors that correlate with the activity or inactiv(cid:173)
`ity of agents may help to identify the groups of
`patients who may benefit from treatment. Such
`information can only be obtained by systemati(cid:173)
`cally collecting and analyzing tumor samples
`from patients. Although these studies add to the
`complexity of the trial, it is possible that molecu(cid:173)
`lar characterization of patients' tumors may be
`more predicative of drug efficacy than histology. 53
`Although our understanding of the regulation
`of translation and its effects on cell proliferation
`are incomplete, targeting translation to control
`cell proliferation is a novel therapeutic strategy
`worthy of further evaluation. Although our un(cid:173)
`derstanding of the mechanisms of action this
`class of agents is imperfect and will undoubtedly
`deepen over time, these agents have significant in
`vitro and in vivo antiproliferative activity against
`a broad range of human tumor cell lines, which
`supports the initiation of clinical trials of CCI-799
`
`West-Ward Exhibit 1016
`Dancey 2000
`Page 007
`
`

`

`74
`
`JANET E. DANCEY
`
`in cancer patients. As with other molecular-tar(cid:173)
`geted therapies, the challenge to investigators
`will be to efficiently determine what roles these
`translational regulatory pathways and this class
`of agent will play in the treatment of cancer
`patients.
`
`ACKNOWLEDGMENT
`
`I thank Robert Abraham, PhD, and Peter Houghton, PhD,
`for their insights into the mechanisms of action and antipro(cid:173)
`liferative activities of these agents and Edward Sausville, MD,
`PhD, Susan Arbuck, MD, and Howard Streicher, MD, for their
`helpful critique of the manuscript.
`
`REFERENCES
`16. Sabers CJ, Martin MM, Brunn GJ, et al: Isolation of a
`1. Proud CG, Denton RM: Molecular mechanisms for the
`control of translation by insulin. Biochem J 328:329-341, 1997
`protein target of the FKBP12-rapamycin complex in mamma(cid:173)
`2. Rebollo A, Merida I, Gomez J, et al: Differential effect of
`lian cells. J BioI Chern 270:815-822, 1995
`rapamycin and cyclosporin A in proliferation in a murine T
`17. Brown EJ, Albers MW, Shin TB, et al: A mammalian
`cell line expressing either intermediate or high affinity recep(cid:173)
`protein targeted by Gl-arresting rapamycin-receptor complex.
`tor for IL-2. Cytokine 7:277-286, 1995
`Nature 369:756-758, 1994
`3. Sehgal SN, Baker H, Vezina C: Rapamycin (AY-22,989),
`18. Sabatini DM, Erdjument-Bromage H, Lui M, et al:
`a new antifungal antibiotic: II. Fermentation, isolation and
`RAFT!: A mammalian protein that binds to FKBP12 in a
`characterization. J Antibiot (Tokyo) 28:727-732, 1975
`rapamycin-dependent fashion and is homologous to yeast
`4. Vezina C, Kudelski A, Sehgal SN: Rapamycin
`TORs. Cell 78:35-43, 1994
`(AY-22,989), a new antifungal antibiotic: I. Taxonomy of the
`19. Chiu MI, Katz H. Berlin V: RAPTl, a mammalian
`producing streptomycete and isolation of the active principle.
`homolog of yeast Tor, interacts with the FKBPl21rapamycin
`J Antibiot (Tokyo) 28:721-726, 1975
`complex. Proc Nat! Acad Sci USA 91:12574-12578,1994
`5. Dotil·os J, Suffness M: New antitumor substances of
`20. Abraham RT: Mammalian target of rapamycin: Immu(cid:173)
`natural origin. Cancer Treat Rev 8:63-87, 1981
`nosuppressive drugs uncover a novel pathway of cytokine
`6. Eng CP, Sehgal SN, Vezina C: Activity of rapamycin
`receptor signaling. Curr Opin Immunol 10:330-336, 1998
`(AY-22,989) against transplanted tumors. J Antibiot (Tokyo)
`21. Keith CT, Schreiber SL: PIK-related kinases: DNA
`37:1231-1237, 1984
`repair, recombination, and cell cycle checkpoints. Science
`7. Muthukkumar S, Ramesh TM, Bondada S: Rapamycin, a
`270:50-51, 1995
`potent immunosuppressive drug, causes programmed cell
`22. Sarkaria IN, Tibbetts RS, Busby EC, et al: Inhibition of
`death in B lymphoma cells_ Transplantation 60:264-270, 1995
`the radio(cid:173)
`phosphoinositide 3-kinase related kinases by
`8. Seufferlein T, Rozengurt E: Rapamycin inhibits constitu(cid:173)
`sensitizing agent wortmannin. Cancer Res 58:4375-4382,
`tive p70s6k phosphorylation, cell proliferation, and colony
`1998
`formation in small cell lung cancel' cells. Cancer Res 56:3895-
`23. Brown EJ, Schreiber 8L: A signaling pathway to trans(cid:173)
`3897, 1996
`lational controL Ce1186:517-520, 1996
`9. Hosoi H, Dilling MB, Shikata T, et al: Rapamycin causes
`24. Scott PH, Brunn GJ, Kohn AD, et al: Evidence of
`poorly reversible

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