throbber
Review
`
`Inhibitors of the mammalian
`target of rapamycin
`
`Janet E Dancey
`Investigational Drug Branch/CTEP/DCTD, National Cancer Institute, 6130 Executive Boulevard,
`Room 7131 Rockville, MD 20852, USA
`
`The mammalian target of rapamycin (mTOR) is a downstream protein
`kinase of the phosphatidylinositol 3′-kinase–Akt signalling pathway. As a
`result of its position within this pathway and its central role in controlling
`cellular growth, mTOR is viewed as an important target for anticancer ther-
`apeutics development. Currently, the mTOR inhibitor rapamycin (sirolimus,
`Wyeth) and its derivatives temsirolimus (CCI-779, Wyeth), everolimus
`(RAD-001, Novartis Pharma AG) and AP-23573 (Ariad Pharmaceuticals) are
`being evaluated in cancer clinical trials. Preclinical studies suggest that sen-
`sitivity to mTOR inhibition may correlate with aberrant activation of the
`phosphatidylinositol 3′-kinase pathway and/or with aberrant expression of
`cell-cycle regulatory or antiapoptotic proteins. Clinical trial results show
`that mTOR inhibitors are generally well tolerated and may induce pro-
`longed stable disease and even tumour regressions in a subset of patients.
`Questions remain regarding optimal dose, schedule, patient selection and
`combination strategies for this novel class of agents.
`
`Keywords: AP-34573, everolimus, mammalian target of rapamycin, sirolimus, temsirolimus
`
`Expert Opin. Investig. Drugs (2005) 14(3):313-328
`
`1. Introduction
`
`The mammalian target of rapamycin (mTOR) is a downstream protein kinase of
`the phosphatidylinositol 3′-kinase (PI3K)–Akt signalling pathway. As a result of its
`position within this pathway and its central role in controlling cellular growth,
`mTOR is considered to be an important target for anticancer therapeutics develop-
`ment. By targeting mTOR, the immunosuppressant and antiproliferative agent
`rapamycin inhibits the signals required for cell-cycle progression, cell growth and
`proliferation in both normal and malignant cells. Currently, mTOR inhibitors
`rapamycin (sirolimus, Rapamune™, Wyeth) and its derivatives temsirolimus
`(CCI-779, Wyeth), everolimus (RAD-001, Novartis Pharma AG) and AP-23573
`(Ariad Pharmaceuticals) are being evaluated in cancer clinical trials. An additional
`agent, TAFA-93 (isotechnika), has recently entered human trials in the prevention
`of organ rejection after transplantation.
`
`2. Biochemistry of protein kinase of the phosphatidylinositol
`3′-kinase–Akt–mammalian target of rapamycin pathway
`
`mTOR is an evolutionarily conserved 290-kDa serine-threonine kinase that reg-
`ulates both cell growth and cell-cycle progression through its ability to integrate
`signals from nutrient and growth factor stimuli [1,2]. mTOR, a member of the
`PI3K-kinase-related kinase (PIKK) superfamily, is composed of 2549 amino
`acids that are grouped into highly conserved, yet functionally poorly understood,
`domains. Modelling of the tertiary structure of mTOR suggests that most of the
`protein consists of helical repeat units that may form an extended superhelical
`structure to create multiple interfaces for protein–protein interactions [3]. mTOR
`
`1. Introduction
`
`2. Biochemistry of protein kinase
`of the PI3K–Akt–mTOR pathway
`
`3. mTOR in human cancer
`
`4. Rapamycin and derivatives
`
`5. Expert opinion
`
`Ashley Publications
`www.ashley-pub.com
`
`10.1517/13543784.14.3.313 2005 Ashley Publications Ltd ISSN 1354-3784
`
`313
`
`NOVARTIS EXHIBIT 2027
`Par v. Novartis, IPR 2016-01479
`Page 1 of 16
`
`

`
`Inhibitors of the mammalian target of rapamycin
`
`Nutrients
`
`Raptor
`
`mLST8
`
`mTOR
`
`Rheb
`-GTP
`
`GF Stimulation
`
`TSC1
`TSC2
`AMPK
`LKB1
`
`PIP3
`
`Akt
`
`PDK-1
`
`PIP2
`
`PI3-K
`
`PTEN
`
`4EBP
`
`PP2A
`
`S6K
`
`eIF-4E
`
`Translation
`
`S6
`
`Transcription
`
`↑ Protein synthesis
`G1 progression
`Cell survival
`
`Figure 1. mTOR signalling. GF receptor stimulation leads to activation of PI3K and 3'-OH phosphorylation of PIP2 to generate PIP3. PIP3
`then recruits PDK-1 and Akt to the plasma membrane to be activated. The tumour suppressor phosphatase PTEN dephosphorylates PIP3
`at the D3 position of the inositol ring. Activated Akt phosphorylates and inhibits TSC, removing its inhibitory effect on Ras-related small
`GTPase Rheb, which acts as a positive upstream regulator of TOR. TSC2 is also inhibited by the presence of amino acids, thus allowing
`Rheb to activate mTOR through an unknown mechanism. Activation of mTOR in complex with other proteins such as raptor and possibly
`other proteins such as mLST8 leads to phosphorylation of eIF-4E-binding protein (4E-BP) and ribosomal protein S6 kinase 1. This
`interaction results in an increase in translation rates of a subset of mRNAs including those encoding proteins required for cell-cycle
`progression. See Section 2 for additional details.
`BP: Binding protein; elF: Eukaryotic initiation factor; GF: Growth factor; mLST8: Mammalian orthologue of LST8; mTOR: Mammalian target of rapamycin;
`PDK: Phosphatidylinositol-dependent kinase; PDK-1: Phosphatidylinositol-dependent kinase-1; PI3K: Phosphatidylinositol 3′-kinase; PIP2: Phosphatidylinositol-4,5-
`bisphosphate; PIP3: Phosphatidylinositol-3,4,5-triphosphate; PP2A: Protein phosphatase 2A; PTEN: Phosphatase and tensin homologue; Raptor: Regulatory-associated
`protein of mTOR; Rheb: Ras homologue enriched in brain; SC: Tuberous sclerosis complex.
`
`functions in a protein complex that integrates signals from
`a variety of sources, including growth factors, energy stores
`and hypoxia, with the protein translation apparatus [4].
`There remains a number of unresolved questions regarding
`the function and the mechanism of action of mTOR. For
`example, it is not clear whether the intrinsic kinase activity
`of mTOR is sufficient for its full activity in vivo or if some
`mTOR functions may be mediated through protein interac-
`tions independent of its kinase function [5]. Furthermore, it
`is not clear whether mTOR may also serve as a scaffold for
`other proteins with catalytic activity, such as kinases and
`phosphatases that may regulate its overall activity in vivo [6].
`However, it does seem clear that not all the cellular activi-
`ties of mTOR are sensitive to inhibition by rapamycin [5].
`Thus, mTOR function in normal and malignant cells
`requires further elucidation, and a further understanding of
`these functions is likely to lead to optimal strategies for
`therapeutic interventions.
`The pathway from growth factor receptor stimulation to
`mTOR activation proceeds through and in parallel to PI3-K
`and Akt (Figure 1). In response to extracellular stimuli, PI3K
`phosphorylates the 3′-hydroxyl of phosphatidylinositol-4,5-
`bisphosphate (PIP2) to generate phosphatidylinositol-3,4,5-
`triphosphate (PIP3). The formation of PIP3 leads to the bind-
`ing through their pleckstrin homology (PH) regions and
`
`kinase-1
`phosphatidylinositol-dependent
`of
`activation
`(PDK-1) and Akt at the plasma membrane [1]. The tumour
`suppressor phosphatase PTEN (phosphatase and tensin
`homologue deleted on chromosome 10) dephosphorylates
`PIP3 at the 3′ position of the inositol ring, reversing the action
`of PI3K. Activated Akt phosphorylates and inhibits the tuber-
`ous sclerosis complex (TSC). In mammals, TSC1 (hamartin)
`and TSC2 (tuberin) associate to form a heterodimer that
`inhibits cell-cycle progression and cell proliferation [7,8] that, at
`least in part, is mediated through mTOR inhibition. A current
`model suggests that PI3K-dependent activation of Akt results
`in the phosphorylation and inactivation of TSC2 in mamma-
`lian cells [9]. Following its phosphorylation, TSC2 destabilises,
`disrupting the formation of functional TSC1/2 complexes and
`removing their inhibitory effect on mTOR [10-12]. Conversely,
`energy deprivation activates the tumour suppressor gene prod-
`uct LKB1, which in turn phosphorylates and activates TSC2
`[13,14]. TSC2 acts as a GTPase-activating protein (GAP)
`toward the Ras-related small GTPase Rheb (Ras homologue
`enriched in brain) [15], a positive upstream regulator of
`mTOR. Therefore, activation of TSC2 by LKB1, as may
`occur in a nutrient-deprived state, inhibits Rheb and results in
`the downregulation of mTOR. In contrast, inhibition of
`TSC2 as occurs in the presence of amino acids, as well as
`binding to 14-3-3 proteins [16-18], with Akt phosphorylation
`
`314
`
`Expert Opin. Investig. Drugs (2005) 14(3)
`
`NOVARTIS EXHIBIT 2027
`Par v. Novartis, IPR 2016-01479
`Page 2 of 16
`
`

`
`or through loss of TSC2 function through mutation, as occurs
`in TS patients, allows Rheb to activate mTOR and leads to the
`phosphorylation of the downstream mTOR targets [8]. The
`molecular mechanism by which Rheb activates mTOR is cur-
`rently unknown. The net result of these signalling interactions
`suggests a model in which growth factor signalling through
`PI3K–Akt is coordinated with nutrient availability signalling
`through LKB1–TSC1/2 to Rheb and mTOR.
`mTOR functions in a complex with at least two other pro-
`teins: regulatory associated protein of mTOR (raptor) [19,20]
`and mammalian orthologue of LST8 (mLST8, also known as
`G-protein β-subunit-like protein [GβL]) [20,21]. The function
`of raptor is not completely understood; however, it appears
`to be essential for mTOR signalling in vivo and is critical for
`mTOR substrate phosphorylation in vitro [6]. One model
`that has been proposed to explain the role of mTOR–raptor–
`mLST8 suggests that a change in the configuration of the
`mTOR–raptor complex, which is mediated by nutrient con-
`ditions such as amino acid availability, affects the ability of
`mTOR to interact with and phosphorylate its substrates [6].
`In the absence of amino acids, the mTOR–raptor–mLST8
`complex prevents mTOR from binding to its substrates and/
`or prevents the access of mTOR (or mTOR-associated
`kinases) to the substrates [6]. In the presence of amino acids, a
`conformational change promotes the interaction between
`raptor and mTOR substrates and/or increases access of the
`substrates to mTOR and its associated kinases [6]. Further
`studies are required to address how amino acids elicit these
`proposed changes in mTOR–raptor complex and verify this
`model [6]. However, current evidence suggests that activated
`mTOR, in complex with raptor, and possibly other proteins,
`leads to phosphorylation of two key proteins; eukaryotic ini-
`tiation factor-4E (eIF-4E) binding protein 1 (4E-BP1) and
`protein S6 kinase 1 (S6K1) [3,22]. S6K1 or 4E-BP1 phosphor-
`ylation is often used as in vitro and in vivo readouts of
`mTOR activity (described in this section) in laboratory and
`clinical research studies.
`Unphosphorylated 4E-BP1 binds to RNA cap-binding
`protein eIF-4E, inhibiting its coupling to mRNA methyl-7
`GpppN cap and the multi-protein translational–initiation
`complex, required for initiating translation of cap-dependent
`mRNAs, a subset of mRNAs with regulatory elements located
`in the 5′-untranslated regions (UTRs) [23]. Stimulation of cells
`by hormones, mitogens, growth factors, cytokines and G-pro-
`tein-coupled agonists results in the activation of mTOR, lead-
`ing to multi-site phosphorylation of 4E-BP1, release of eIF4E
`to bind to cap mRNA transcripts and other initiation com-
`plex proteins and the initiation of cap-dependent translation.
`This interaction results in an increase in translation rates of
`cap-dependent mRNAs, which include those encoding a
`number of proteins required for cell-cycle progression, such as
`mRNAs that encode cell-cycle regulator proteins cyclin D and
`ornithine decarboxylase [24]. This effect on translation of cer-
`tain regulatory mRNAs may be one means by which mTOR
`regulates cell growth.
`
`Dancey
`
`The second target of mTOR is the phosphorylation and
`activation of S6K1. Previously, activation of S6K1 had been
`correlated with increased translation of 5′-terminal oligopyri-
`midine tract (TOP) mRNAs, which encode components of
`the translational apparatus [25]. However, the requirement for
`S6K1 activity in translation of TOP-containing mRNAs has
`been disputed. Recent studies indicate that the translation of
`TOP mRNAs may occur independently of S6K1 function, as
`S6K1 activation is insufficient to relieve translational repres-
`sion of TOP RNAs and complete inhibition of mTOR by
`rapamycin had only a slight repressive effect on translation of
`TOP mRNAs [24,26]. These recent studies led to the conclu-
`sion that the regulation of TOP translation by growth factors
`and mitogens is primarily through the PI3K pathway with lit-
`tle role for mTOR [24]. Instead, S6K1 has been implicated in
`glucose homeostasis and regulation of eukaryotic elongation
`factor 2 kinase [24]. The exact mechanism(s) by which mTOR
`regulates translation and cell growth are complex and require
`further study.
`Consistent with its role as a central controller of cellular
`growth, mTOR activation leads to the phosphorylation of
`several downstream signalling effectors and transcription fac-
`tors in addition to its effects on 4E-BP1 and S6K1, which in
`turn influence cell proliferation, survival and angiogenesis. Of
`particular note for therapeutics development is that many,
`although not all, of the protean functions of mTOR appear to
`be sensitive to inhibition by rapamycin. The additional cellu-
`lar effects of mTOR include its direct phosphorylation of sig-
`nal transducer and activator of transcription-3 (STAT3) [27].
`Similarly, S6K1 phosphorylation of the transcription factor
`cAMP response element modulator (CREM) [27,28] is reported
`to be rapamycin sensitive. mTOR has been reported to regu-
`late autophagy [29,30]. mTOR signalling may also provide an
`antiapoptotic function; Akt-mediated protection from apop-
`tosis is mediated, at least in part, by mTOR-dependent stabi-
`lisation of cell surface amino acid transporters [31], and the
`proapoptotic protein BAD has been reported to be a substrate
`of S6K1 [32]. In addition, microarray analysis of RNA isolated
`from cells deprived of nutrients or treated with rapamycin has
`demonstrated an important role for mTOR in controlling the
`expression of genes involved in many metabolic and biosyn-
`thetic pathways [33]. Clearly, the biochemical effects of mTOR
`signalling are multiple, incompletely catalogued, poorly
`understood and potentially context specific [24]. However, the
`multitude of cellular signalling processes in which mTOR
`participates in normal and malignant cells and the inhibition
`of some of these processes by pharmacological inhibition has
`contributed to the keen interest in mTOR inhibition as a
`strategy for development of therapeutics.
`
`3. Mammalian target of rapamycin in human
`cancer
`
`Although mutations of mTOR have not been reported in
`human cancers, both aberrant PI3K-dependent signalling and
`
`Expert Opin. Investig. Drugs (2005) 14(3)
`
`315
`
`NOVARTIS EXHIBIT 2027
`Par v. Novartis, IPR 2016-01479
`Page 3 of 16
`
`

`
`Inhibitors of the mammalian target of rapamycin
`
`aberrant protein translation have been identified in a wide
`variety of malignancies and may contribute to the oncogenesis
`and malignant progression. For example, components of the
`PI3K pathway that are mutated in different human tumours
`include activation mutations of growth factor receptors,
`amplification and/or overexpression of PI3K and Akt. The
`resultant aberrant pathway signalling not only leads to a
`growth advantage during carcinogenesis and stimulates cancer
`cell proliferation but also contributes to treatment resistance
`due to a high PI3K–Akt-mediated survival threshold [1]. If
`such cancer cells are ‘addicted’ to the growth and survival-sig-
`nalling effects of the PI3K–Akt pathway, it is possible that this
`dependency will
`result
`in cancer-cell
`sensitivity
`to
`mTOR inhibition [1].
`In addition to cancer-cell dependency on aberrant PI3K
`signalling for proliferation and survival, endothelial cell pro-
`liferation may also be dependent on mTOR signalling.
`Endothelial cell proliferation
`is stimulated by vascular
`endothelial cell growth factor (VEGF) activation of the
`PI3K–Akt–mTOR pathway and, furthermore, VEGF pro-
`duction may be partly controlled by mTOR signalling
`through mTOR effects on the expression of hypoxia-induci-
`ble factor-1α (HIF-1α) [34-36]. HIF-1α is a heterodimeric
`transcription
`factor containing an
`inducibly expressed
`HIF-1α subunit and a constititutively expressed HIF-1β sub-
`unit. Under hypoxic conditions, the HIF-1α subunit accu-
`mulates due to a decrease in the rate of proteolytic
`degradation through the ubiquitin–proteasome pathway. The
`resulting HIF-1α–HIF-1β heterodimers lead to the transcrip-
`tion of certain gene products, including VEGF. Recent studies
`suggest that amplified signalling through PI3K and its down-
`stream target, mTOR, enhances HIF-1-dependent gene
`expression in vitro and that this expression is partially sensitive
`to mTOR inhibition [37]. Therefore, tumour angiogenesis
`may depend on mTOR signalling in two ways: through
`hypoxia-induced production of VEGF by tumour and stro-
`mal cells, and through VEGF stimulation of endothelial cell
`proliferation and survival through PI3K–Akt–mTOR path-
`way. Furthermore, the antitumour effects noted by inhibiting
`mTOR may be related to antiproliferative effects within
`tumour cells as well as endothelial cells.
`More directly related to mTOR effects on protein transla-
`tion in cancer cells, aberrantly high rates of protein biosynthe-
`[38], and certain tumour
`sis are observed in tumours
`suppressors and proto-oncogenes may regulate malignant pro-
`gression by altering the protein synthesis machinery [39]. For
`example, signalling through Ras and Akt acts rapidly to
`increase the association of mRNA transcripts with polyribo-
`somes, and, therefore, may
`immediately and broadly
`influence protein translation [40]. In addition, dysregulation of
`cap-dependent translation through overexpression of eIF-4E
`confers malignant characteristics and induces cancer by sup-
`pressing apoptosis in a breast-cancer model, underscoring the
`potential of therapeutics that selectively target the Akt–
`mTOR–eIF-4E pathway [41,42]. Further investigation will be
`
`needed to clarify to what extent deregulation of translation of
`total or specific mRNAs contributes to tumorigenesis. How-
`ever, deregulated components of the translational machinery,
`or specific oncoproteins that are overexpressed in cancer cells
`and that are under mTOR translational control, could be sen-
`sitive to mTOR targeted therapy. Thus, mTOR inhibition
`may lead to the inhibition of malignant progression by alter-
`ing the translation of multiple proteins including those that
`control cell size, cell-cycle progression and cell survival as well
`as angiogenesis.
`
`4. Rapamycin and derivatives
`
`Rapamycin is a macrocyclic lactone produced by Streptomy-
`ces hygroscopicus, a soil bacterium native to Easter Island
`(Rapa Nui). Rapamycin possesses fungicidal, immunosup-
`pressive and antiproliferative properties [43]. Because of its
`ability to suppress lymphocyte activation, rapamycin was
`developed and received regulatory approval as an immuno-
`suppressant for the prophylaxis of renal allograft rejection.
`Rapamycin’s immunosuppressant effects are due to its inhi-
`bition of the biochemical events required for the progression
`of IL-2-stimulated T cells from G1 to S phase of the cell
`cycle. However, rapamycin and derivatives temsirolimus,
`everolimus and AP-23573 also inhibit cellular proliferation
`in a variety of tumour models, and are currently under clini-
`cal evaluation as potential cancer therapeutics. These clinical
`studies will help to validate mTOR as an anticancer drug tar-
`get. A review of the mechanisms of action and resistance and
`a summary of clinical trial results for these agents is
`described in Section 4.1 – 4.8.
`
`4.1 Mechanism of action
`For many years, rapamycin has been used as a pharmacological
`probe; thus, much is known about its mechanism of action and
`by inference, if not actual experimentation, about the other
`agents of this pharmaceutical class. Rapamycin targets the ubiq-
`uitously expressed peptidyl-prolyl cis-trans isomerase FK506-
`binding protein of 12 kDa (FKBP12) and the rapamycin–
`FKBP12 complex binds to the FKBP12–rapamycin-binding
`(FRB) domain adjacent to the kinase domain of mTOR. The
`mechanism by which rapamycin inhibits mTOR is unclear.
`The rapamycin–FKBP12 complex may act by altering the com-
`position and/or conformation of the multi-protein mTOR
`complexes. By disrupting these protein complexes, rapamycin
`may impair either upstream signalling leading to mTOR activa-
`tion or kinase access to downstream substrates [44,45]. Rapamy-
`cin and its derivatives share the following features: inhibition of
`cellular proliferation by inducing G1 phase arrest, induction of
`apoptosis in selected models and limited normal tissue toxicity.
`The antiproliferative effects of rapamycins have been evalu-
`ated in numerous in vitro and in vivo tumour models. Results
`from these experiments indicate that these agents may inhibit
`tumour and endothelial cell proliferation in picomolar to
`nanomolar concentrations, and may add to the cytotoxicity of
`
`316
`
`Expert Opin. Investig. Drugs (2005) 14(3)
`
`NOVARTIS EXHIBIT 2027
`Par v. Novartis, IPR 2016-01479
`Page 4 of 16
`
`

`
`other chemotherapeutic agents and radiation [46-51]. The
`means by which rapamycin induces antiproliferative effects are
`not completely understood but are associated with the inhibi-
`tion of transition from G1 to S phase of the cell cycle. Among
`the reported cellular alterations in the presence of rapamycin
`include reduction of cyclins, particularly cyclin D [52,53], which
`would lead to inhibition of cyclin-dependent kinase activity
`and phosphorylation of retinoblastoma protein (pRb) as well
`as increases in cyclin-dependent kinase inhibitors p21cip1 and
`p27kip1 [54,55]. These observed effects would be consistent with
`the ability of rapamycin to impede G1 progression.
`In most instances, inhibition of mTOR by rapamycin leads
`to an antiproliferative response. However, there are examples
`in which rapamycin induces apoptosis of certain cell lines
`[56,57]. The molecular determinants of the antiproliferative ver-
`sus apoptotic response of cells after exposure to rapamycins are
`poorly understood. One proposed trigger for rapamycin-
`induced apoptosis may depend on the functions of p53,
`p27cip1 and p27kip1. In a series of studies, Huang and colleagues
`reported that rapamycin induced a cellular stress response
`characterised by rapid and sustained activation of the apoptosis
`signal-regulating kinase 1 (ASK1) signalling pathway. Selective
`apoptosis was seen in cells lacking functional p53 or p27kip1,
`and the apoptotic response correlated with and was dependent
`on 4E-BP1 expression. In contrast, wild-type p53 or p27cip1
`suppressed the apoptotic response to rapamycin [56-58]. Thus,
`the antitumour activity of rapamycin may depend on the func-
`tion of cell-cycle regulatory proteins as well as upstream cellu-
`lar signalling through mTOR. Based on these laboratory
`models, the expected clinical activity of rapamycin would be
`delayed tumour progression rather than tumour regression in
`most patients with sensitive disease. However, tumour regres-
`sion through rapamycin-induced apoptosis could occur.
`In addition to the effects on tumour cell proliferation, inhi-
`bition of mTOR by rapamycin also potently inhibits angio-
`genesis and endothelial cell proliferation in vitro and in vivo.
`Humar and colleagues showed that hypoxia directly enhances
`dose-dependent induction of DNA synthesis and cellular pro-
`liferation by platelet-derived growth factor (PDGF) and basic
`fibroblast growth factor (bFGF) and rapamycin specifically
`blocked the increase in proliferation observed under hypoxia
`in mouse and rat vascular smooth muscle and endothelial cell
`angiogenesis models [36]. The antiangiogenic properties of
`rapamycin are also associated with a decrease in VEGF pro-
`duction and a reduction in the response of vascular endo-
`thelial cells to stimulation by VEGF [36,59]. In vitro studies
`revealed that rapamycin is capable of blocking the activation
`of HIF-1 through enhanced degradation of HIF-1α [37]. In
`summary, there is considerable evidence that rapamycin is
`antiangiogeneic, and these antiangiogenic effects may be
`multifold in that the agent inhibits endothelial cell prolifera-
`tion in the presence of hypoxia, inhibits endothelial cell pro-
`liferation to VEGF stimulation through inhibition of mTOR,
`and decreases VEGF synthesis through enhanced HIF-1α
`degradation [59-61].
`
`Dancey
`
`4.2 Determinants of sensitivity and resistance to
`mammalian target of rapamycin inhibition
`Considerable research is underway to identify markers associ-
`ated with tumour cell sensitivity and resistance to rapamycin.
`Laboratory studies suggest that genetic mutations and/or
`compensatory changes leading to aberrant signal transduc-
`tion, both upstream and downstream of mTOR, influence
`tumour cell sensitivity to rapamycins [62,63]. Among the
`described abnormalities in cellular molecules that correlated
`with rapamycin resistance are mutations of mTOR or
`FKBP12 that prevent rapamycin from binding to mTOR
`[62,63]. Although such mutations have been induced in labora-
`tory models, these mutations have not been described in
`human cancers. Mutations or defects of mTOR-regulated
`proteins, including S6K1 and 4E-BP1, which impair their
`interaction with mTOR, also render cells insensitive to
`rapamycin [62]. Although inhibition of phosphorylation of
`S6K1, its target ribosomal S6 protein, and 4E-BP1 correlates
`with rapamycin sensitivity in laboratory models, inhibition
`of S6K1 and 4E-BP1 is not sufficient to guarantee sensitivity
`to rapamycin as hypophosphorylation of these mTOR targets
`has been seen in rapamycin-resistant as well as -sensitive cells
`[62,64]. Thus, additional factors must mediate cellular depend-
`ency on mTOR signalling. Expression and function of
`ataxia-telangiectasia gene product ATM, as well as 14-3-3,
`p53, PI3K–Akt and PTEN have been reported to correlate
`with rapamycin sensitivity [62,63]. Data supporting these last
`two potential mechanisms of sensitivity will be discussed in
`greater detail.
`There is considerable evidence that aberrant stimulation of
`the PI3K–Akt signalling pathway in cancer cells may increase
`the dependency of such tumours on mTOR signalling func-
`tions and their sensitivity to signal modulation by inhibiting
`mTOR [1]. Studies have reported that rapamycin and related
`compounds exert selective cytostatic/cytotoxic effects on
`PTEN-/- tumours in vivo [65,66]. In vitro and in vivo studies of
`isogenic PTEN+/+ and PTEN-/- mouse cells, as well as human
`cancer cells with defined PTEN status. showed that the
`growth of PTEN null cells was blocked preferentially by
`mTOR inhibition [65]. However, the loss of PTEN function
`does not correlate with rapamycin sensitivity in all models. In
`a series of breast cancer cell lines, overexpression of S6K1 and
`phosphorylated Akt, independent of PTEN status, was associ-
`ated with rapamycin sensitivity [64]. As noted in other studies,
`the differential sensitivity to mTOR inhibition was not
`explained by differences in biochemical blockade of the
`mTOR pathway because S6 phosphorylation was inhibited in
`sensitive and resistant cell lines. Thus, aberrant stimulation of
`the PI3K–Akt pathway, which may occur through hyper-
`action growth factor receptor, PI3K, Akt or loss of PTEN may
`be markers of tumour cell responsiveness to rapamycins.
`Loss of functional TSC, as occurs in patients with tuber-
`ous sclerosis syndrome (TS), may also confer sensitivity to
`rapamycins. Although TS-associated tumours are usually
`benign, they can affect almost every major organ system and
`
`Expert Opin. Investig. Drugs (2005) 14(3)
`
`317
`
`NOVARTIS EXHIBIT 2027
`Par v. Novartis, IPR 2016-01479
`Page 5 of 16
`
`

`
`Inhibitors of the mammalian target of rapamycin
`
`are thus responsible for considerable morbidity of affected
`individuals [7]. TSC is rarely associated with malignant
`tumours, although patients with TSC are at increased risk
`for clear-cell renal carcinoma [7,67]. Loss or reduction of the
`function of the TSC1–TSC2 protein complex in individuals
`with TSC leads to hyperactive mTOR signalling in hamar-
`tomatous tissues, implying that the loss of TSC1–TSC2 and
`dysregulation of mTOR signalling is a causative factor in
`TSC disease [67,68]. In addition, TSC2-/- p53-/- cells, as well
`as tumours from TSC2+/- mice, display an mTOR-activation
`signature with constitutive activation of S6K. Both the acti-
`vation of S6K and the growth advantage of TSC2-/- p53-/-
`cells are reverted by treatment with rapamycin [69]. Activa-
`tion of mTOR was associated with downregulation of
`PDGF receptor–PI3K–Akt signalling in cells lacking TSC1
`or TSC2. The low rate of malignant tumour development in
`TS patients may result from the negative feedback of the
`PDGF receptor–PI3K–Akt signalling pathway to balance
`activation of mTOR [69]. The observation that mTOR sig-
`nalling is upregulated in TS-associated hamartomas suggest
`that rapamycin and its derivatives may prove to be effective
`in the treatment of these patients.
`Studies evaluating the addition of rapamycin to standard
`anticancer therapy suggest that rapamycin may add to the
`cytotoxic effects of standard cancer drugs; however, tumour
`cell responsiveness to these combinations will be determined
`by the molecular phenotype of cancer cells. For example,
`Wendel and colleagues [70] used a mouse model of B-cell
`lymphoma to explore the consequences of inhibiting Akt
`signalling. They showed that Akt promotes tumorigenesis
`and cancer drug resistance by disrupting apoptosis. Lym-
`phomas expressing Akt but not those expressing Bcl-2 were
`sensitised by rapamycin to chemotherapy-induced apop-
`tosis. In another example, apoptosis induced by the c-myc
`gene is suppressed by mTOR downstream target eIF-4E [71].
`Although eIF-4E recapitulates Akt’s action in tumorigenesis
`and drug resistance, it renders cells insensitive to concomi-
`tant administration of rapamycin and chemotherapy. These
`results suggest that Akt signalling through mTOR and
`eIF-4E may be an important mechanism of oncogenesis and
`in vivo cancer drug resistance, and that targeting the apop-
`totic programme may restore drug sensitivity [70,72]. How-
`ever, cells overexpressing Akt that are exposed to rapamycin
`may be rendered sensitive to standard cancer drugs and be
`triggered to undergo apoptosis when exposed to the combi-
`nation. However, those tumour cells overexpressing eIF-4E
`or Bcl-2 may be insensitive to rapamycin and remain
`resistant to standard cancer therapies.
`Collectively, these data suggest that tumour cells sensitive to
`mTOR inhibition are
`likely to be those with aberrant
`signalling through the PI3K–Akt pathway, such as those with
`activating mutations and/or amplification of growth factor
`receptors, PI3K, Akt, or through the loss of PTEN or TSC1/2.
`Sensitive cells may be triggered to undergo G1 arrest or apopto-
`sis. The apoptotic effect may be determined, in part, by p53,
`
`p21cip1, p27kip and Bcl-2 function. Conversely, cells with amplifi-
`cation and/or overexpression of downstream target eIF-4E may
`be insensitive to rapamycins. In addition to the presence and
`function of these pathway proteins, microarray and proteomic
`analyses have identified many proteins whose levels were
`changed in response to rapamycin [33]. Interpreting the data and
`investigating the leads generated by these screens may lead to a
`clearer understanding of the control of gene expression by
`mTOR and patterns that are predictive of the sensitivity or resist-
`ance to its inhibition. Thus, predicting sensitivity or resistance of
`tumours to rapamycins will probably require the assessment of
`multiple, rather than individual, molecular markers.
`
`4.3 Preclinical anticancer activity of mammalian target
`of rapamycin inhibitors
`All the rapamycins under clinical development have anti-
`proliferative activity in a variety of haematological and solid
`tumour systems as single agents and in combination with stand-
`ard cancer therapeutic agents and radiation [43,47-49,51,73-80].
`Because of their high lipophilic indices, rapamycins easily cross
`the blood–brain barrier [47] and, thus, may be of value for the
`treatment of primary and secondary malignancies of the central
`nervous system. These studies suggest that rapamycins may have
`a role as single agents and in combination with standard
`therapies in a variety of malignancies.
`Relatively few studies have assessed various rapamycins
`under identical experimental conditions. Two studies sug-
`gest that antitumour effects of rapamycin and temsirolimus
`or everolimus are similar. Exposure of IGROV1 ovarian car-
`cinoma cells to rapamycin and everolimus in vitro was asso-
`ciated with substantial cellular apoptosis with both agents
`[81]. Co-treatment of rapamycin in vitro or temsirolimus
`in vivo inhibited mTOR activity and restored sensitivity of
`the MCF7–Akt transfected cell line to t

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