throbber
Downloaded from
`
`http://annonc.oxfordjournals.org/
`
` at University of Chicago on February 8, 2016
`
`Review
`
`Annals of Oncology 16: 525 – 537, 2005
`
`doi:10.1093/annonc/mdi113
`
`Published online 22 February 2005
`
`mTOR-targeted therapy of cancer with rapamycin derivatives
`
`S. Vignot1†, S. Faivre2†, D. Aguirre2 & E. Raymond1,2*
`
`1Department of Oncology, Hospital Saint Louis, Paris; 2Department of Medical Oncology, Beaujon University Hospital, Clichy, France
`
`Received 6 August 2004; revised 12 November 2004; accepted 15 November 2004
`
`Rapamycin and its derivatives (CCI-779, RAD001 and AP23576) are immunosuppressor macrolides
`that block mTOR (mammalian target of rapamycin) functions and yield antiproliferative activity in a
`variety of malignancies. Molecular characterization of upstream and downstream mTOR signaling
`pathways is thought to allow a better selection of rapamycin-sensitive tumours. For instance, a loss
`of PTEN functions results in Akt phosphorylation, cell growth and proliferation; circumstances that
`can be blocked using rapamycin derivatives. From recent studies, rapamycin derivatives appear to
`display a safe toxicity profile with skin rashes and mucositis being prominent and dose-limiting.
`Sporadic activity with no evidence of dose – effect relationship has been reported. Evidence suggests
`that rapamycin derivatives could induce G1 – S cell cycle delay and eventually apoptosis depending
`on inner cellular characteristics of tumour cells. Surrogate molecular markers that could be used to
`monitor biological effects of rapamycin derivatives and narrow down biologically active doses in
`patients, such as the phosphorylation of P70S6K or expression of cyclin D1 and caspase 3, are cur-
`rently evaluated. Since apoptosis induced by rapamycin is blocked by BCL-2, strategies aimed at
`detecting human tumours that express BCL-2 and other anti-apoptotic proteins might allow identifi-
`cation of rapamycin-resistant tumours. Finally, we discuss current and future placements of rapamy-
`cin derivatives and related translational research into novel therapeutic strategies against cancer.
`Key words: cell signal inhibitors, phase I trial, rapamycin, signal transduction inhibitors, sirolimus
`
`Introduction
`
`Cancer cells need several kinases for cell cycle control, pro-
`liferation, invasion and angiogenesis [1]. Treatments targeted
`against cellular signalling pathways have shown promise in
`the management of solid tumours and hematological malig-
`nancies. mTOR (mammalian target of rapamycin) was shown
`to be a key kinase acting downstream of the activation of the
`phosphatidylinositol 3 kinase (PI3K). Cumulative evidence
`supports the hypothesis that mTOR acts as a ‘master switch’
`of cellular catabolism and anabolism, signalling cells to
`expand, grow and proliferate. Although it is found in virtually
`all mammalian cells, it is particularly important in tumour
`cells that proliferate and invade aggressively. In addition,
`mTOR has recently been found to have profound effects in
`the regulation of apoptotic cell death, mainly dictated by the
`cellular context and downstream targets including P53, BAD,
`BCL-2, P27 and C-MYC.
`Rapamycin (sirolimus) is a macrolide antibiotic produced
`by Streptomyces hygroscopicus, which binds FKBP-12
`
`*Correspondence to: Dr E. Raymond, Department of Medical Oncology,
`Beaujon University Hospital, 100 Boulevard du General Leclerc, 92100
`Clichy cedex, France. Tel: +33-01-4087-5617;
`Fax: +33-01-4087-5487; E-mail: eric.raymond@bjn.ap-hop-paris.fr
`† Ste´phane Vignot and Sandrine Faivre participated equally to this work
`and should be considered as joint first authors.
`
`q 2005 European Society for Medical Oncology
`
`(FK506 binding protein). Thereby, the rapamycin – FKBP12
`complex can inhibit mTOR preventing further phosphorylation
`of P70S6K, 4E-BP1 and, indirectly, other proteins involved in
`transcription and translation and cell cycle control. Rapamycin
`is currently used alone or in combination with cyclosporine as
`an immunosuppressive drug to prevent renal graft rejection.
`Rapamycin analogues currently selected for clinical devel-
`opment are CCI-779 (intravenous formulation currently in
`phase III from Wyeth Ayest), RAD001 (oral formulation cur-
`rently in phase I-II from Novartis Pharma) and AP23573
`(intravenous formulation currently in phase I from Ariad
`Pharma). In clinical settings, using intermittent administration
`of CCI-779, RAD001 and AP23576, no evidence of immuno-
`suppressive effects has been observed. Dose-limiting toxicities
`consist of skin reactions, mucositis and minimal myelosup-
`pression. Evidence of antitumour activity has been reported in
`several patients with renal clear cell carcinoma and breast can-
`cer. Interestingly, rapamycin and its analogues antagonise
`tumour growth induced by loss of the PI3K antagonist, PTEN.
`Selection of patients based on the detection of activated
`P70S6K/AKT and/or loss of PTEN expression might help to
`predict the sensitivity of tumour cells to rapamycin analogues.
`Pharmacodynamic monitoring of the biological activity of
`rapamycin in clinical trials using molecular endpoints such as
`the phosphorylation of AKT, P70S6K and/or 4E-BP1 might
`also help to determine biological relevant dose(s) and plasma
`ARGENTUM
`Exhibit 1039
`
`000001
`
`

`

`Downloaded from
`
`http://annonc.oxfordjournals.org/
`
` at University of Chicago on February 8, 2016
`
`Figure 2. Several molecules involved in cell survival, including mTOR,
`are regulated by the PI3K/AKT pathway.
`
`IL-6, insulin-like growth factor (IGF), epidermal growth factor
`(EGF), platelet derived growth factor (PDGF), insulin growth
`factors (IGF-1 and IGF-2) and colony stimulating factor
`(CSF). Activated PI3K phosphorylates inositol lipids at the 30
`position of the ring inositol, generating the lipid products PI3-
`phosphate
`[PI(3)P], PI3,4-biphosphonate
`[PI(3,4)P2]
`and
`PI3,4,5-triphosphate [PI(3,4,5)P3]. These lipid products are
`involved in a number of cellular processes including cell
`proliferation, survival, cytoskeletal reorganisation, membrane
`trafficking, cell adhesion, motility, angiogenesis and insulin
`action [8, 9]. Downstream to PI3K, protein kinase B (PKB),
`also named AKT, impacts on cell survival at multiple levels
`[10]. Substrates of AKT include glycogen synthase kinase
`(GSK3), 6-phosphofructo-2-kinase, the protein BAD, forkhead
`family of
`transcription factors, endothelial nitric oxide
`synthase (eNOS), mTOR, BRCA1 and others (Figure 2).
`GSK3 appears negatively regulated by AKT-dependent phos-
`phorylation. Reduced GSK-3 activity leads to increased levels
`of the growth stimulator beta-catenin [11]. The pro-apoptotic
`protein BAD is also inactivated by AKT-dependent phos-
`phorylation, thus enhancing cell survival. On the contrary,
`AKT indirectly activates mTOR via TSC, which in turn phos-
`phorylates and activates several targets involved in translation
`of specific mRNAs, apoptosis and/or cell cycle, as we will
`discuss later [12].
`Kinase activities are regulated by phosphatases that act in
`opposition to kinases by removing phosphates from the target
`proteins. The phosphatase and tensin homologue gene (PTEN,
`also named MMAC1 or TEP1) is a tumour suppressor gene,
`located on human chromosome 10q23 [13]. PTEN was found
`to be mutated in several human sporadic cancers such as
`breast, endometrial, ovarian (type endometroid), brain, renal
`carcinoma, melanoma and prostate tumour cell
`lines and
`primary tumours. Patients with germline mutations of PTEN
`develop inherited Bannayan Zoanna syndrome characterised
`by multiple hamartomas and Cowden disease, and sub-
`sequently are susceptible to developing breast, thyroid and
`several others cancers [14, 15].
`The PTEN product has a protein tyrosine phosphatase
`domain and extensive homology to tensin (related protein with
`focal adhesions), suggesting that PTEN suppresses tumour cell
`growth by antagonising protein tyrosine kinases, and regulates
`tumour cell
`invasion and metastasis through interactions
`
`526
`
`rapamycin
`treated with
`individuals
`in
`concentration(s)
`analogues. In addition, rapamycin and its analogues may sen-
`sitise cancer cells to apoptosis induction by cisplatin and gem-
`citabine.
`In this review, we will describe the molecular pathways
`involved in rapamycin activity and we will present recent pre-
`clinical and clinical data on rapamycin and its analogues. We
`will then discuss the current and future placement of those
`molecules into current therapeutic strategies against cancer.
`
`PI3K signaling pathway and mTOR
`
`Overview of the PI3K-related kinases (PIKKs)
`
`Following activation of membrane receptors by a variety of
`growth factors, secondary molecular signals are generated to
`transmit the stimulus toward the nucleus and activate a num-
`ber of events. Many of these signals involve the phosphoryl-
`ation of proteins known as kinases (Figure 1). Among those
`kinases, PI3K and PI3K-related kinases (PIKK) belong to a
`family of high molecular mass kinases whose catalytic
`domains show a strong resemblance. This family and the ribo-
`somal protein P70S6K, mTOR, the DNA-dependent protein
`kinase,
`the ataxia telangiectasia mutated gene (ATM),
`the
`ataxia-telangiectasia related (ATR) protein and key com-
`ponents of the histone acetylase complex are involved in
`checkpoint regulation of cell cycle, DNA repair,
`telomere
`length and cell death [2].
`The PI3K pathway is very often activated in cancer and
`contributes to cell cycle progression, to decrease apoptosis
`and to increase metastatic capabilities of cancer cells [3, 4].
`The uncontrolled activation of the PI3K pathway has been
`implicated in cell transformation and tumour progression in
`several tumour types including brain tumours, breast, ovarian
`and renal carcinomas [5 – 7]. Activation of the PI3K pathway
`is mediated by activated RAS or directly by some tyrosine-
`kinase receptors, under the control of several growth factors
`and cytokines including interleukin 1 (IL-1), IL-2, IL-3, IL-4,
`
`Figure 1. Multiple signaling pathways involved in signal transduction
`from tyrosine kinase receptors (TKR).
`
`000002
`
`

`

`527
`
`Downloaded from
`
`http://annonc.oxfordjournals.org/
`
` at University of Chicago on February 8, 2016
`
`Figure 3. Overview on mTOR main activities in normal cells.
`
`mTOR is a serine/threonine kinase of 289 kDa, highly
`related to yeast TORs that belong to the PIKK family with a
`dual regulation by amino acid availability and by mitogen
`activated PI3K/AKT. TOR proteins in Sacharomyces cerevi-
`sae and the mammalian related proteins (mTOR) are required
`for signalling translational initiation and therefore cell cycle
`progression from the G0/G1 to S phase [22]. Yeast TOR 2
`protein also controls the actin cytoskeleton during cell cycle
`progression but it is not clear whether this function is con-
`served by mTOR [23].
`In humans, mTOR primarily appears to be a nutrient-
`sensing protein: mTOR is constitutively activated in the pre-
`sence of growth factor and nutrients and acts as a master
`switch of cellular catabolism and anabolism [12, 24]. mTOR
`is also regulated by hypoxia and by AMP levels. mTOR is
`inhibited through deacetylated tRNA species accumulating as
`a result of amino acid shortage (but the exact pathway remains
`to be elucidated) and via C-ABL protein tyrosine kinase that
`phosphorylates mTOR and inhibits its action. mTOR is also
`activated by TSC2 mutations or loss of LKB1.
`As discussed above, upregulation of mTOR can be related
`to loss of the tumour suppressor gene PTEN and activation of
`AKT.
`
`Translational control by mTOR
`
`mTOR modulates translation of specific mRNAs via the regu-
`lation of the phosphorylation state of several different trans-
`lation proteins, mainly 4E-BP1, P70S6K and eEF2.
`
`4E-BP1. Protein synthesis is regulated in many instances at
`the initiation phase, when a ribosome is recruited to the 50 end
`of an mRNA. Eukaryotic ribosomes do not have the ability to
`locate and bind to the 50 end of mRNA and need translation
`initiation factors to guide them. The cap structure at the 50 end
`of an mRNA is recognised by the eukaryotic translation
`initiation factor 4E(eIF4E). eIF4E, in association with eIF4G,
`directs the translation machinery to the 50 end of the mRNA.
`The 4E-binding proteins
`(4E-BP)
`are essential
`in the
`regulation of
`the interaction between eIF4E and eIF4G.
`The mTOR signalling
`pathway modulates
`4E-BP1
`phosphorylation and mediates its dissociation from eIF-4E
`[25, 26]. This dissociation is a crucial step toward activating
`translation of mRNAs with specific regulatory elements in
`the 50-untranslated terminal region (50UTR), especially c-myc,
`cyclin D1 and ornithine decarboxylase. In contrast, when
`
`focal adhesions. Davies et al.
`[16] and others, have
`at
`demonstrated that PTEN plays an important role in anchorage-
`dependant cell survival. Additionally, loss of PTEN protects
`cells from apoptosis triggered by matrix detachment (anoikis),
`and the re-expression of PTEN in PTEN-mutated cells causes
`apoptosis in cells in suspension.
`PTEN is involved in the regulation of the PI3K pathway
`[3]. There is evidence that PTEN dephosphorylates phospha-
`tydilinositol 3,4,5-triphosphate while mutated PTEN cannot
`dephosphorylate
`phosphoinositides
`at
`the D3
`position
`(D3-PPI). PTEN ± mice spontaneously develop neoplasia,
`the normal PTEN allele and an
`associated with loss of
`increased activation of AKT, mTOR and P70S6K. In vitro and
`in vivo, the growth of PTEN-deleted human cancer cells and
`PTEN/ mouse cells can be preferentially inhibited by phar-
`macologic mTOR inhibition [17]. This growth inhibition then
`involves both a decrease in proliferation and an increase in
`apoptosis.
`However, although PTEN inactivation might be required, it
`might not be sufficient to explain the sensitivity to rapamycin
`since there is also evidence to show that cancer cells with
`PTEN inactivation might remain resistant to rapamycin. Con-
`versely, a dose dependent tumour growth delay is observed in
`mice bearing PTEN proficient cancer cells. In that case,
`reports have suggested that the effects of rapamycin might be
`related to the inhibitory effects against endothelial cells block-
`ing tumour angiogenesis.
`Several studies have suggested that genomic integrity, tran-
`script and protein levels, phosphorylation and activity of all
`the multiples components of the PI3K pathway, should be
`evaluated to determine whether they predict prognosis or
`response to therapy in several cancers. The members of the
`PIKK family are key components of signals that coordinate
`the activity of the cell cycle and their functional characteris-
`ation gives important insights into cell growth and cell cycle
`checkpoint function. Further, development of molecular thera-
`peutics targeting the PI3K pathway is clearly warranted in
`different types of cancer. Wortmannin inhibits the multiple
`effects of PI3Ks and yield anti-inflammatory, immunosuppres-
`sive, cytotoxic and radio-sensitising properties with potential
`as an anti-neoplastic drug [18, 19]. The multiple molecular
`targets inhibited by this agent (PI3, PI4 and PIKK) raise cau-
`tion about
`its clinical use. Besides, wortmannin presents
`chemical instability and hepatotoxicity, limiting its develop-
`ment. Thus, we need to evaluate more specific molecules in
`this pathway as individual targets.
`
`Focus on mTOR (Figure 3)
`
`mTOR was identified in 1994 by several groups of investi-
`gators as the kinase targeted by rapamycin linked to the cel-
`lular protein FKBP12 (FK506-binding protein).
`It was
`therefore also named FKBP-RAP associated protein (FRAP),
`RAP FKBP12 target
`(RAFT1) and RAP target
`(RAPT1)
`[20, 21].
`
`000003
`
`

`

`528
`
`growth factor or nutrients are lacking, or in the presence of
`mTOR inhibitors, 4E-BP1 becomes hypophosphorylated,
`which increases
`its binding with EIF-4E and prevents
`initiation of translation.
`
`involved in amino acid biosynthesis, regulates the activity of
`amino acid permeases and represses autophagy. In the absence
`of the TOR signal, ribosomal biosynthesis is inhibited and
`autophagy is activated.
`
`Downloaded from
`
`http://annonc.oxfordjournals.org/
`
` at University of Chicago on February 8, 2016
`
`Neuronal function and role in brain development
`
`Recent evidence also suggests that mTOR may be involved in
`neuronal protein synthesis. mTOR could play a role in
`embryonic brain development and in the learning and memory
`process [34]. mTOR could inhibit eEF2 phosphorylation in
`active synapses to locally unrepress translation, whereas some
`studies have reported an increase of eEF2 phosphorylation in
`response to various neurotransmitters.
`
`Rapamycin and analogues
`
`Rapamycin development
`
`Rapamycin, also named sirolimus, is a natural antibiotic pro-
`duced by S. hygroscopicus. This molecule was found 30 years
`ago in the Easter Island Rapa Nui soil from which rapamycin
`was named. Rapamycin was subsequently isolated in Montreal
`by Ayerst Research laboratories in 1972. Rapamycin is a
`macrocyclic lactone developed initially as an anti-fungal drug
`directed against Candida albicans, Cryptococcus neoformans,
`and Aspergillus fumigatus [35 – 38]. It is a white crystalline
`solid insoluble in aqueous solutions, but soluble in organic
`solvents. The chemical structure is shown in Figure 4.
`Recently, rapamycin has been tested by the Developmental
`Therapeutic Branch, National Cancer
`Institute (NCI) and
`identified as a noncytotoxic agent that delays tumour prolifer-
`ation, finding evidence of cytostatic activity against several
`human cancers in vitro and in vivo. However, the development
`program of rapamycin as an anticancer agent was halted in
`1982 and only resumed in 1988 after demonstration of a safe
`toxicological profile in animals.
`In the meantime, rapamycin was developed as an immuno-
`supressive agent and those studies have enabled us to under-
`stand the mechanism of action of this agent. Rapamycin, via
`
`Figure 4. Rapamycin’s chemical structure including FKBP12 and mTOR
`binding domains.
`
`P70S6K. mTOR also phosphorylates and activates P70S6K to
`favour the recruitment of the 40S ribosomal subunit
`into
`actively translating polysomes and enhance the translation of
`mRNAs with 50 terminal oligopyrimidine tracts. These tran-
`scripts can encode up to 20% of the mRNAs [27].
`
`eEF2. Finally, mTOR also acts at the level of the elongation
`phase. The eukaryotic elongation factor 2 (eEF2) promotes
`translocation of the mRNA and mTOR regulates the activity
`of eEF2 kinase, apparently via regulation of a phosphatase
`activity (PP2A) [28].
`
`Anti-apoptotic and pro-apoptotic effects
`
`the downstream target of mTOR,
`There is evidence that
`P70S6K, binds to mitochondrial membranes and phosphory-
`lates the pro-apoptotic molecule BAD [29]. The binding of
`P70S6K to BAD inactivates BAD and increases cell survival.
`In contrast, mTOR might translocate from the cytoplasm to
`the nucleus shortly after the formation of syncitium between
`cells expressing the HIV envelope and CD4 cells. Once in the
`nucleus, it causes phosphorylation of P53, transcriptional acti-
`vation and induction of pro-apoptotic proteins such as BAX,
`and activation of the intrinsic cell death pathway [30].
`
`Cell cycle regulation
`
`mTOR inhibition results in an increase in the turnover of
`cyclin D1, at both mRNA and protein levels [31], and a
`decrease in the elimination of the cyclin dependant kinase
`inhibitor P27. Additionally, mTOR downregulates cyclin-A-
`dependent kinase activity in exponentially growing cells. The
`pharmacological inhibition of mTOR decreases G1 transit in
`the cell cycle [32].
`
`Metabolic modulation
`
`Cells have the ability to adapt to the dynamic pool of nutrients
`in their immediate environment. Mammalian cells respond
`continually to changes in available blood glucose and amino
`acids. mTOR plays an important role in the modulation of
`metabolic pathways, including those related to insulin [12,
`33]. The initiation of translation appears to be the limiting
`phase in protein synthesis. The central role played by mTOR
`in protein translation leads to the control of skeletal muscle
`protein synthesis. Because mTOR inhibition causes cellular
`responses indicating the physiological state of starvation, these
`proteins are thought to be mediators of nutrient-sensing path-
`ways. mTOR can detect nutrients such as carbon and nitrogen,
`signaling cells to grow and proliferate, a fact particularly
`important
`in tumour cells that proliferate aggressively. In
`yeast, TOR functions are well established genetically, with
`somewhat less compelling data in mammalian cells. In yeast,
`TOR signaling modulates the transcription of genes that are
`
`000004
`
`

`

`Downloaded from
`
`http://annonc.oxfordjournals.org/
`
` at University of Chicago on February 8, 2016
`
`its methoxy group, crosslinks the immunophilin FK506 bind-
`ing protein (FKBP12). The rapamycin – FKBP12 complex
`specifically interacts with mTOR to inhibit mTOR signalling
`to downstream targets [38]. Rapamycin inhibits T-cell pro-
`liferation induced by antigen, mitogenic lectins, alloantigen
`and crosslinking of T-cell surface markers with monoclonal
`antibodies. Rapamycin can inhibit proliferative responses
`induced by cytokines, including IL-1, IL-2, IL-3, IL-4 and IL-
`6, IGF, PDGF and CSFs.
`The preclinical development of rapamycin as an immuno-
`suppressor has been extensively reviewed [39, 40]. It has
`demonstrated a high degree of synergy with cyclosporin [41]
`both in vitro and in vivo, lowering the dose of cyclosporin
`necessary for
`immunosuppression, enhancing the rejection
`prevention in renal
`transplantation and minimising cyclo-
`sporin-induced toxicity [42, 43]. There are observations that
`high doses of rapamycin block the proliferative responses to
`cytokines by vascular and smooth muscle cells after mechan-
`ical injury, such as balloon angioplasty or allo-rejection [44,
`45]. In a non-human primate model, supra-therapeutic concen-
`tration of rapamycin stabilised and possibly reversed the inti-
`mal vascular lesion caused by the progression of immune
`injury in aortic allograft [46]. Rapamycin treatment concomi-
`tant with monoclonal antibody blockade of the co-stimulatory
`signal by anti-CD154 in mice induces tolerance, and the com-
`bination of rapamycin with anti-B7 in non-human primates
`seems to facilitate tolerance induction [47]. IC50 values of
`rapamycin as an immunosuppressor are in the range of
`0.1 – 300 nM.
`A relevant point of rapamycin as an immunosuppressor is
`the absence of the vasomotor renal side effects exhibited by
`CsA and tacrolimus. Treatment with rapamycin preserves glo-
`merular filtration and renal blood flow in normal, salt-depleted
`and spontaneously hypertensive rats [48]. The renal
`tissue
`seems to be protected during the rapamycin treatment by an
`inhibition of the intrarenal angiotensin II cascade. However,
`rapamycin does produce a dose-dependent tubular toxicity in
`rats, which is related to the delayed recovery of tubular epi-
`thelial function after injury [49].
`Over the last 8 years, rapamycin has undergone clinical
`trials as an immunosuppressive agent, progressing from phase
`I safety,
`tolerability and pharmacokinetic investigation to
`phase II dose-finding studies and limited sized evaluations of
`drug combination regimens. The completion of phase III trials
`led to approval of rapamycin by the Food and Drug Adminis-
`tration (FDA) of the USA in 1999 to prevent acute rejection
`in combination with cyclosporin and steroids. One year later,
`the drug was approved by the European Agency as an alterna-
`tive to calcineurin antagonists for
`long-term maintenance
`therapy to avoid graft
`rejection.
`Interestingly,
`rapamycin,
`unlike cyclosporin, does not seem to increase the risk of
`malignancy but rather to decrease the risk of post-transplant
`lymphoproliferative disorders.
`Apart from its immunosupressive capacity, rapamycin was
`also recently shown to be capable of preventing coronary
`artery re-stenosis [50, 51]. Growth, migration and differen-
`
`529
`
`tiation of vascular smooth-muscle cells are two major features
`of neointimal proliferation after vascular injury. The proposed
`mechanism of inhibition of proliferation of vascular smooth-
`muscle cells by sirolimus includes binding of the immuno-
`philin FKBP12, blockage of P70S6K, impairment of retino-
`blastoma protein phosphorylation, and prevention of p27
`downregulation. Additionally, rapamycin has been shown to
`be effective in inhibiting PDGF-induced migration of human
`vascular smooth cells in vitro, without affecting the ability of
`these cells to bind collagen and without disrupting their cyto-
`skeletal components [52, 53]. To avoid the systemic effects of
`rapamycin, it has been used locally in an impregnated stent to
`prevent coronary restenosis [51].
`
`Pharmacokinetic and metabolic information
`
`These data were initially obtained from studies that evaluated
`rapamycin as an immunosuppressor [54, 55]. The systemic
`bio-availability of rapamycin is approximately 15%, it has a
`maximal concentration at about 1 h and is widely distributed
`in tissues compared with plasma. The ratio blood cells/plasma
`ranges between 36 in renal transplant cases to 79 in healthy
`volunteers. In vitro experiments using human liver micro-
`somes suggest
`that cytochrome Cyp450 3A4 is the major
`biotransformation system, generating the inactive metabolites,
`hydroxy, dihydroxy, hydroxy-demethyl, didemethyl, 7-0
`demethyl and 41-0 demethyl. More than 90% of the drug is
`recovered in the faeces. Urine represents only 2% of the drug
`elimination. The average elimination half life is variable,
`ranging from 10 h in children to 110 h in patients with hepatic
`impairment.
`Rapamycin exposure is increased by diltiazem and ketoco-
`nazole and decreased by rifamycin and anticonvulsants [56].
`Regarding the interaction between rapamycin and cyclosporin
`(CsA), rapamycin concentrations are increased by concomitant
`administration of Neoral, the microemulsion formulation of
`CsA, and rapamycin increases CsA exposure approximately 2-
`fold, presumably because of competition for metabolism by
`Cyp450 3A4 and, possibly, drug extrusion by P-glycoprotein
`[57].
`Initial clinical studies show that a dose-dependent reversible
`reduction in mean platelet number and, to a far lesser extent,
`leukocyte count, was accompanied by increased serum choles-
`terol and triglyceride values. There were no changes in blood
`pressure, kidney or liver function test results.
`Corroborating preclinical studies, rapamycin does not affect
`glomerular filtration, but hypokalemia and hypophosphatemia
`has been reported as evidence of renal tubular abnormalities
`[58, 59].
`Additionally, rapamycin augments reactions to CsA: hyper-
`tension, acne and hirsutism. It has been associated with minor
`adverse effects such as diarrhea, tachycardia and arthralgia, as
`well as with non-infectious pneumonitis [60].
`
`Rapamycin as an anticancer drug
`
`Rapamycin was shown to inhibit the growth of several murine
`and human cancer cell
`lines in a concentration-dependent
`
`000005
`
`

`

`Downloaded from
`
`http://annonc.oxfordjournals.org/
`
` at University of Chicago on February 8, 2016
`
`530
`
`manner, both in tissue culture and xenograft models: B16
`melanoma, P388 leukemia, MiaPaCa-2 and Panc-1 human
`pancreatic carcinomas and others [61 – 63]. In the 60 tumour
`cell lines screened at the National Cancer Institute in the USA
`(COMPARE program), the average GI50 obtained for rapamy-
`cin over all cell lines was 8.2 nM when the highest concen-
`tration tested is 1000 nM and 1800 nM when the highest
`concentration tested is 106 nM. In practical terms, they found
`general sensitivity to the drug at doses under 2000 ng/ml,
`more evident
`in leukemia, ovarian, breast, central nervous
`system and small cell lung cancer cell lines.
`Rapamycin induces P53-independent apoptosis in childhood
`rhabdomyosarcoma [64] and enhances the apoptosis induced
`in vitro by cisplatin in murine T-cell and human HL-60 pro-
`myelocytic leukemias and human ovarian SKOV3 carcinoma
`[65]. On the contrary, it inhibits taxol-induced apoptosis in
`human B-cell lines, probably through preventing BCL-2 inac-
`tivation and can inhibit hybridoma cell death in bioreactors,
`thereby increasing the production of monoclonal antibody
`[66].
`In addition, rapamycin inhibits the oncogenic transform-
`ation of human cells induced by either PI3K or AKT and has
`shown metastatic tumour growth inhibition and anti-angio-
`genic effect in in vivo mouse models [67]. Considering this
`effect, there is evidence that the PI3K – P70S6K intracellular
`signaling pathway is required for HIF1 and VEGF expression
`and also for VEGF stimulation of endothelial cells. The
`anti-angiogenic effect of rapamycin seems to be related to
`the inhibition of these effects [68].
`Based on these pre-clinical results, studies with rapamycin
`as an anticancer drug were begun and rapamycin analogues
`were
`developed with more
`favourable
`pharmaceutical
`properties.
`
`Table 1. Doses and schedules of rapamycin derivatives in phase I trials
`
`Rapamycin derivatives (Table 1)
`
`CCI-779, a more water-soluble ester derivative of sirolimus,
`was identified by investigators at Wyeth Ayerst as a noncyto-
`toxic agent that delayed tumour proliferation. CCI-779 was
`designed to increase the solubility of rapamycin making this
`compound readily available for intravenous formulation. At
`several non-toxic doses, CCI-779 demonstrated antitumour
`activity alone or in combination with cytotoxic agents in a
`variety of human cancer models such as gliomas, rhabdomyo-
`sarcoma, primitive neuroectodermal tumour such as medullo-
`blastoma, head and neck, prostate, pancreatic and breast
`cancer cells [69 – 73]. Treatment of mice with CCI-779
`inhibits P70S6K activity and reduces neoplastic proliferation.
`As with sirolimus, PTEN-deficient human tumours are more
`sensitive to CCI-779-mediated growth inhibition than PTEN-
`expressing cells. Specifically, studies in vitro in a panel of
`eight human breast cancer cell lines showed that six of eight
`cancer lines studied were inhibited by CCI-779 with IC50 in
`the low nanomolar range. Two lines, however, were found to
`be resistant with IC50>1 mM. The sensitive cell lines were
`estrogen receptor positive, or overexpressed HER-2/Neu, or
`had lost the tumour suppressor gene product PTEN [74]. Inter-
`estingly, preclinical studies indicate that intermittent adminis-
`tration of CCI-779 reduces its immunosuppressive properties
`while retaining its antitumour activity.
`CCI-779 has recently completed phase I evaluation in cancer
`patients and phase II results are starting to be reported. In phase I
`studies, the drug has been administrated as a single agent on a
`weekly schedule and daily for 5 days every other week. The
`main toxicities of CCI-779 included dermatological toxicities
`[aseptic folliculitis, erythematous macular rashes (Figure 5),
`eczematous reactions, dry skin, herpes-type lesions and nail
`disorders], mild myelosuppression (mainly thrombocytemia at
`
`Drugs
`
`CCI779
`
`Schedules
`
`Range of doses
`
`0.75 – 19.1 mg/m2/day q2 weeks
`
`Limiting toxicities
`
`Thrombocytopenia
`
`MTD
`
`Reached
`
`7.5 – 220 mg/m2/week
`
`Mucositis, asthenia, bipolar disorders
`
`Not reached
`
`25 – 100 mg/m2/day q2 weeks
`
`Mucositis
`
`Reached
`
`RAD001
`
`5 – 30 mg/week
`
`Mucositis, asthenia
`
`Not reached
`
`5 – 10 mg/day
`
`Ongoing
`
`AP23573
`
`3 – 28 mg/day q2 weeks
`
`Mucositis
`
`Ongoing
`
`Ongoing
`
`Ongoing
`
`Reached
`
`Ongoing
`
`000006
`
`

`

`Downloaded from
`
`http://annonc.oxfordjournals.org/
`
` at University of Chicago on February 8, 2016
`
`531
`
`pharmacokinetic analysis of CCI-779 administrated intra-
`venously, through limited dose ranges has revealed an increase
`in drug exposure with dose and elimination life of 18 – 30 h.
`Based on these results, weekly doses of 25, 75, and 250 mg
`CCI-779, which are not based on classical definitions of maxi-
`mum tolerated doses, were chosen for phase II trials in
`patients with breast and renal cancer [79].
`A recent phase II trial evaluated the safety and efficacy of
`three dose levels of CCI-779 in previously treated patients
`with advanced renal cell carcinoma. The drug was well toler-
`ated, occurrences of partial responses, minor responses and
`prolonged stabilisation of disease in these heavily pretreated
`patients was found, suggesting the evaluation as a single agent
`in phase III trials in renal cell carcinoma [81].
`Another phase II study is currently ongoing in patients with
`advanced breast cancer [82]. The majority of patients in this trial
`received more than two lines of prior chemotherapy containing
`anthracyclines and taxanes. The study was designed to explore
`two doses of weekly intravenous administration of CCI-779 (75
`and 250 mg/m2). Toxicity was significantly higher in patients
`receiving higher doses (requiring dose reduction in 45.1% of
`those patients). At
`the higher dose, psychic disorders were
`reported, including lethargy and depression. Activity was similar
`in the two treatment groups with an overall response rate of
`10%. Immunohistochemistry was pe

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