throbber
nature publishing group
`
`STATE
`
`ART
`
`Rapamycin: Something Old, Something New,
`Sometimes Borrowed and Now Renewed
`CM Hartford1,2 and MJ Ratain1,3,4
`
`The molecular target of rapamycin (mTOR) is central to a complex intracellular signaling pathway and is involved in
`diverse processes including cell growth and proliferation, angiogenesis, autophagy, and metabolism. Although sirolimus
`(rapamycin), the oldest inhibitor of mTOR, was discovered more than 30 years ago, renewed interest in this pathway is
`evident by the numerous rapalogs recently developed. These newer agents borrow from the structure of sirolimus and,
`although there are some pharmacokinetic differences, they appear to differ little in terms of pharmacodynamic effects
`and overall tolerability. Given the multitude of potential applications for this class of agents and the decrease in cost
`that can be expected upon the expiration of sirolimus patents, renewed focus on this agent is warranted.
`
`Rapamycin (or sirolimus, the official generic name) is the
`prototypical inhibitor of the molecular target of rapamycin
`(mTOR). It was discovered more than 30 years ago as an
`antifungal agent and was approved in 1999 as an immuno-
`suppressant for the prevention of renal allograft rejection.
`Over
`the years, with the elucidation of
`the complex
`intracellular signaling network in which mTOR participates
`and the realization of the multitude of potential therapeutic
`applications of
`interfering with this network,
`interest in
`agents that can inhibit mTOR has increased. There are
`currently three mTOR inhibitors in clinical trials, all either
`prodrugs or analogs of sirolimus, including temsirolimus,
`which was
`recently approved by the Food and Drug
`Administration (FDA) for the treatment of renal cancer.
`Although these newer agents (rapalogs) exhibit slightly
`different pharmacokinetic properties, they appear to differ
`little pharmacodynamically from sirolimus. This review will
`provide an overview of the mTOR pathway, discuss the
`pharmacokinetics and tolerability of the different mTOR
`inhibitors, and highlight the many potential clinical applica-
`tions of these agents.
`
`THE mTOR PATHWAY
`The mTOR protein is a serine-threonine kinase that is central
`to a complex intracellular signaling pathway (Figure 1) and is
`involved in a number of important processes such as cell
`growth and proliferation, cellular metabolism, autophagy,
`
`and angiogenesis. It responds to signals from the extracellular
`environment such as nutrient and growth factor supply,
`energy, and stress. Signaling through the pathway is
`promoted when there is an abundance of nutrients or energy
`and is downregulated in states of depletion and stress.1
`mTOR exists within the cell complexed to the proteins
`GbL and either raptor or rictor.2 The mTOR/rictor protein
`complex is not responsive to inhibition by rapalogs and will
`not be discussed in detail except to mention its interaction
`with mTOR/raptor. The mTOR/GbL/raptor complex can be
`activated by various stimuli
`through different upstream
`molecules. Insulin (via the insulin receptor substrate-1) or
`other growth factors affect mTOR via the phosphoinositide-3
`kinase (PI3K)/Akt pathway1 (Figure 1). After stimulation,
`PI3K initiates a cascade that ultimately results
`in the
`phosphorylation and activation of Akt. Akt then acts via
`the tuberous sclerosis complex (TSC), consisting of the
`proteins TSC1 and TSC2, to exert its effect on mTOR.
`Phosphorylation of TSC2 by Akt inhibits TSC2, thereby
`releasing the inhibition that TSC2 otherwise exerts on mTOR
`via inhibition of Rheb, an activator of mTOR.1
`The energy status of the cell also affects mTOR signaling
`and acts via serine/threonine protein kinase 1 (LKB1) and
`AMP-activated kinase (AMPK). In states of energy depletion
`(increased AMP relative to ATP), LKB1 activates AMPK,
`which then activates TSC2, resulting in inhibition of the
`pathway.1 Stress signals,
`for example, DNA damage and
`
`1Committee on Clinical Pharmacology and Pharmacogenomics, The University of Chicago, Chicago, Illinois, USA; 2Department of Pediatrics, The University of
`Chicago, Chicago, Illinois, USA; 3Department of Medicine, The University of Chicago, Chicago, Illinois, USA; 4Cancer Research Center, The University of Chicago,
`Chicago, Illinois, USA. Correspondence: MJ Ratain (mratain@medicine.bsd.uchicago.edu)
`
`Published online 29 August 2007. doi:10.1038/sj.clpt.6100317
`
`CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 82 NUMBER 4 | OCTOBER 2007
`
`381
`
`Ex. 1069-0001
`
`

`
`STATE
`
`ART
`
`including those that are needed for cell cycle progression and
`are involved in cell cycle regulation.6 Phosphorylation and
`activation of S6K1 are also involved in cell growth and
`proliferation, possibly via translation of mRNAs that have a
`terminal 50-oligopyrimidine tract such as those that encode
`ribosomal proteins and elongation factors. However, because
`translation of these latter genes was shown to be intact in
`/
`/
`/S6K2
`mice, other mechanisms are probably
`S6K1
`involved.7 Importantly, S6K1 also has an inhibitory feedback
`function on the pathway by phosphorylating and inhibiting
`insulin receptor substrate-1, thus reducing growth factor–-
`stimulated signaling through PI3K/Akt/mTOR.8
`It also
`phosphorylates and inactivates BCL2 antagonist of cell death
`(BAD), a proapoptotic molecule.6 The numerous proteins
`whose translation is regulated through mTOR and its
`downstream targets include cyclin D1, MYC avian myelocy-
`tomatosis viral oncogene homolog (c-MYC), and hypoxia-
`inducible factor-la (HIF-1a).6
`
`INHIBITORS OF THE mTOR PATHWAY
`Mechanisms of action and mechanisms of resistance
`Inhibitors of the mTOR pathway that are actively being
`studied include sirolimus,
`temsirolimus, everolimus, and
`AP23573 (now called deferolimus) (Figure 2). Sirolimus and
`the rapalogs exert their effects by the same mechanism. Each
`drug binds to the intracellular binding protein FK506-
`binding protein (FKBP12) to form a complex, which then
`binds to mTOR at the FKBP12–rapamycin binding domain,
`interfering with its ability to signal adequately to its
`downstream effectors. Exactly how the sirolimus–FKBP12
`complex interrupts mTOR signaling is not known, but it may
`involve a destabilization of the interaction between mTOR
`and raptor.1
`Given the complexity of the mTOR pathway, there are
`many potential sites of resistance. Defects in the binding of
`sirolimus to FKBP12 because of mutations in FKBP12 and
`mutations in the FKBP12–rapamycin binding domain both
`confer resistance by interfering with the binding of the
`sirolimus–FKBP12 complex to mTOR.9 Other potential
`mechanisms of resistance include decreased levels of 4E-
`BP1 and mutations in S6K1.10 The complexity of the pathway
`and feedback loops within it may also contribute to
`resistance. For example,
`it is possible that inhibiting the
`negative feedback of S6K1 on insulin receptor substrate-1
`may contribute to resistance to the antiproliferative effects of
`mTOR inhibition.9
`
`In the early 1970s, sirolimus, the first inhibitor of
`Sirolimus.
`mTOR, was discovered as part of a screening program for
`new antifungal agents and was first named rapamycin
`because it was isolated from a soil sample from Rapa
`Nui.11 Not long after, the inhibitory effect on the immune
`system was recognized in rats,12 but it was not until the late
`1980s13 and early 1990s14 that
`it was developed as an
`immunosuppressant. Both the antifungal and immunosup-
`pressive activities are the result of the drug’s ability to
`
`Figure 1 Schematic of the mTOR pathway. mTOR receives input from a
`number of upstream pathways, including PI3K/Akt, TSC1/TSC2, and AMPK,
`and acts through downstream effectors S6K and E4BP1 to exert its effects.
`
`hypoxia, also act via the TSC to cause an inhibition of mTOR
`signaling. The nutrient status of the cell, for example, amino-
`acid supply, influences the activity of mTOR. Although the
`mechanisms by which this occurs are less well characterized,
`it is known that the pathway is active when nutrients are
`readily available and downregulated in states of starvation.3
`A number of other pathways can interact with the
`signaling described thus far. For example, phosphatase and
`tensin homolog (PTEN) is a tumor suppressor that counter-
`acts the effects of PI3K by dephosphorylating phosphati-
`dylinositol
`(3,4,5)-trisphosphate and therefore prevents
`activation of Akt.4 Neurofibromin 1 (NF1) inhibits Ras,
`which can activate the mTOR pathway via PI3K/Akt.5
`Moreover,
`the mTOR/rictor complex can phosphorylate
`Akt, which therefore results in feedback and further signaling
`through the pathway.2
`Downstream of mTOR are two main effectors, S6K1 and
`4E-BP1, both of which control the translation of specific
`mRNAs and the synthesis of particular proteins. Phosphor-
`ylation of 4E-BP1 by mTOR ultimately results
`in the
`initiation of
`translation of certain mRNAs
`that have
`regulatory subunits in the 50-untranslated terminal regions,
`
`382
`
`VOLUME 82 NUMBER 4 | OCTOBER 2007 | www.nature.com/cpt
`
`Ex. 1069-0002
`
`

`
`STATE
`
`ART
`
`Figure 2 Inhibitors of the mammalian target of rapamycin (mTOR) pathway that are actively being investigated. The chemical structure of sirolimus and the
`newer analogues are similar. The bars indicate sites of structural differences among the agents.
`
`interrupt the complex intracellular signaling cascade of the
`mTOR pathway, which is relatively conserved from yeasts to
`humans.15 As described, this ultimately causes a decrease in
`protein synthesis. In yeast and molds, the interruption of
`synthesis of proteins involved in cell cycle progression
`the microorganisms.16
`In
`interferes with growth of
`T lymphocytes, this interferes with the ability of the cell to
`respond to cytokines and therefore blocks their proliferation
`and differentiation,17 leading to immunosuppression. As
`sirolimus had been used for the prevention of allograft
`rejection, most of the pharmacology data come from solid
`organ transplant patients.
`Sirolimus is a macrocyclic lactone produced by Strepto-
`myces hygroscopicus.18 It
`is poorly soluble in water and
`therefore can only be given orally. It is available in both liquid
`and tablet formulations. A study of stable renal allograft
`recipients in which patients were converted from liquid
`sirolimus to the tablet form demonstrated relative bioequi-
`valent pharmacokinetics for the two formulations. Although
`the tablet
`formulation resulted in a lower maximum
`concentration (Cmax), the area under the concentration–time
`the two formulations were similar.19
`curves (AUCs) of
`
`Absorption of sirolimus is rapid with peak concentrations
`attained in about 2 h, but bioavailability is low (B15%)20
`and exhibits wide interpatient variability. This variability has
`been largely attributed to the effects of intestinal cytochrome
`P450 3A enzymes (CYP3A) and P-glycoprotein activity on
`sirolimus absorption.21 Studies on renal transplant patients
`have shown that the coadministration of cyclosporine affects
`the bioavailability of sirolimus and that when the drugs are
`administered concomitantly, both the Cmax and the AUC of
`sirolimus are increased, possibly because of inhibition of
`CYP3A4 and P-glycoprotein by cyclosporine.22 The adminis-
`tration of a high-fat meal will also affect absorption. In a
`study of healthy volunteers, coadministration of sirolimus
`with a high-fat meal resulted in a slower absorption but a
`35% increase in AUC.23
`The volume of distribution of sirolimus is large (B12 l/
`kg), indicating wide distribution into tissues and necessitat-
`ing, in many instances, a loading dose. Most of the drug
`partitions into red blood cells (B95%), with small amounts
`in lymphocytes and granulocytes (B1% each).24 Of the 3%
`in plasma, only 2.5% is free and the remainder is protein-
`bound.24
`
`CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 82 NUMBER 4 | OCTOBER 2007
`
`383
`
`Ex. 1069-0003
`
`

`
`STATE
`
`ART
`
`The metabolism of sirolimus is mainly via hepatic CYP3A
`enzymes. In a study of 18 adults with mild to moderate
`hepatic impairment, AUC and half-life of sirolimus were
`significantly increased and weight-normalized apparent oral
`clearance was significantly decreased, as compared with
`healthy controls,25 suggesting that dose adjustments may be
`needed for patients with hepatic impairment. Sirolimus has
`multiple metabolites, all with low immunosuppressive
`activity (o10% relative to sirolimus).26 The excretion is
`primarily
`fecal.
`The
`clearance
`is widely
`variable
`(1.45–6.93 ml/min/kg) and the half-life is long (B62 h),20
`allowing for once-daily dosing.
`and
`The
`interaction of
`sirolimus with CYPA34
`P-glycoprotein not only leads to wide interpatient variability
`in absorption and metabolism but also to the potential for a
`number of drug interactions. Commonly encountered
`inhibitors of CYP3A4 and P-glycoprotein include ketocona-
`zole, cyclosporine, erythromycin, and ritonavir, along with
`grapefruit juice; inducers of CYP3A4 include dexamethasone,
`phenytoin, carbamazepine, rifampin, and phenobarbital, to
`name a few. These medications should be used cautiously and
`avoided if possible in patients treated with sirolimus.
`When used as an immunosuppressant for the prevention
`of solid organ graft rejection, therapeutic drug monitoring
`has been an important issue. A study of 150 renal transplant
`patients showed a correlation between trough drug levels
`(which correlate well with sirolimus AUC) and both
`incidence of adverse side effects (for levels 415 ng/ml) and
`acute graft rejection (for levels o5 ng/ml).27 One approach
`that has been recommended is to monitor all patients during
`the initial phase of treatment (for B2 months) and thereafter
`based on individual patient characteristics.28
`
`deferolimus
`and
`everolimus,
`Rapalogs. Temsirolimus,
`(Figure 2) are all structurally similar to sirolimus, differing
`mainly at a single position of
`the lactone ring (C-40)
`unrelated to both the mTOR- and FKBP12-binding sites.
`Temsirolimus is a water-soluble dihydroxymethyl propionic
`acid ester prodrug of sirolimus. Both intravenous and oral
`formulations are available, although recent development has
`focused on the intravenous formulation. To date, temsiro-
`limus has been mainly developed as an anticancer agent, and
`it was approved by the FDA on 30 May 2007 for the
`treatment of advanced renal cell carcinoma. Everolimus is an
`oral 40-O-(2-hydroxyethyl) derivative (not prodrug) of
`sirolimus approved in Europe as an immunosuppressant
`for the prevention of cardiac and renal allograft rejection in
`adults.29 More recently, it has been studied as an anticancer
`agent. Deferolimus is the newest addition to the rapalogs.
`It contains a phosphine oxide substitute on the lactone ring
`and is available in both oral and intravenous formulations. It
`is currently being investigated for the treatment of a number
`of different malignancies.
`As mentioned, sirolimus and the rapalogs inhibit mTOR
`by forming a complex with FKBP12, which then binds to
`mTOR. Few data are available regarding differences in the
`
`ability of the drugs to inhibit mTOR. One study showed that
`the binding of everolimus to FKBP12 was approximately
`threefold weaker than that of sirolimus in vitro.30 However,
`further in vivo studies on rats showed similar efficacy of the
`two agents in terms of immunosuppressive activity. The
`mTOR inhibitors are very specific in their action, and there is
`no evidence to date of any effects other than those on mTOR.
`All are known to result in a decrease in the phosphorylation
`of the downstream effectors 4E-BP1 and S6K131–34 and the
`degree of this effect is being studied as a potential biomarker
`of mTOR inhibition.
`An often stated reason for the development of new mTOR
`inhibitors is to improve the pharmacokinetic properties of
`sirolimus, mainly the poor bioavailability and insolubility in
`water. The chemical modifications of
`temsirolimus and
`deferolimus have resulted in water-soluble formulations that
`are currently being studied as intravenous agents. Few data
`are available regarding the oral formulations of these rapalogs
`and,
`to date,
`the main oral alternative to sirolimus is
`everolimus. Although everolimus exhibits greater polarity
`than sirolimus, the bioavailability is only slightly improved
`and is still relatively low (B16%).35 Similar to sirolimus,
`CYP3A4 and P-glycoprotein affect
`its absorption and
`contribute to wide interpatient variability.36
`The rapalogs share many characteristics with sirolimus,
`including extensive partitioning into red blood cells,36,37
`metabolism by hepatic CYP3A enzymes,38,39 and primarily
`fecal excretion.35 In a study of patients with liver cirrhosis,
`the clearance of everolimus was reduced by 53% and the half-
`life prolonged 84%, showing the need for dose reductions in
`patients with liver impairment.40 Although similar studies of
`temsirolimus have not been published, given the extensive
`hepatic metabolism, it is possible that this agent may also
`require dose adjustment in patients with liver dysfunction. As
`the rapalogs are substrates of CYP3A4 and P-glycoprotein,
`potential drug interactions are also a concern, as with
`sirolimus. There are differences in the half-lives, potentially
`affecting the optimal dosing schedules. The half-life of
`everolimus in stable renal transplant patients was 24–35 h41
`and 13–2542,43 and 45–74 h,37 respectively, for the intravenous
`formulations of temsirolimus and deferolimus.
`Temsirolimus is the only rapalog that is a prodrug. It
`quickly undergoes hydrolysis to sirolimus after intravenous
`administration. Sirolimus can be seen as early as 15 min after
`the start of temsirolimus infusion, reaches peak concentra-
`tion at 0.5–2.0 h, and then decreases monoexponentially.43
`After a dose of temsirolimus, the exposure to sirolimus
`exceeds that of parent drug because of the differences in half-
`lives, with the mean sirolimus/temsirolimus ratio being
`2.5–3.5.43 Although temsirolimus exhibits inhibitory activity
`against mTOR, most of its clinical effects are probably due to
`the sirolimus metabolite.44,45
`
`Tolerability of the mTOR inhibitors
`The four mTOR inhibitors exhibit remarkably similar side
`effect profiles and are in general well tolerated by patients,
`
`384
`
`VOLUME 82 NUMBER 4 | OCTOBER 2007 | www.nature.com/cpt
`
`Ex. 1069-0004
`
`

`
`STATE
`
`ART
`
`Table 1 Multiple applications of mTOR inhibitors that have been studied, both clinically and preclinically, along with the rationale
`for possible efficacy of mTOR inhibition
`
`Clinical
`data
`
`Preclinical data
`
`Refs. 51–61
`
`Refs. 17, 62–71
`
`Refs. 72–75
`
`Refs. 5, 67, 76–89
`
`Application
`
`Rationale
`
`Comments
`
`Immune
`
`Graft rejection
`GVHD
`Autoimmune diseases
`ALPS
`Asthma
`
`Inhibition of proliferation of T cells in response to
`growth-promoting cytokines (IL-2)
`Inhibition of proliferation of B cells and mast
`cells
`Suppression of B-cell immunoglobulin
`production in response to certain stimuli
`
`Cancer
`
`Malignant tumors
`Benign hamartomas
`Radiation and
`chemosensitization
`
`Antiproliferative effect by inhibiting cell cycle
`progression and causing G1 arrest
`Proapoptotic effect by interfering with
`phosphorylation of BAD
`Antiangiogenic effect by interfering with HIF-1a
`Many proteins involved in hamartoma
`syndromes are linked to the mTOR pathway
`Radiation can induce signaling of Akt/mTOR,
`which can be attenuated by mTOR inhibition
`
`Benign diseases characterized by abnormal proliferation
`
`Effects of mTOR inhibitors on T-cell subsets and
`function:
`Increase in the suppressor function of CD4+ T cells
`Thymic atrophy and a reduction in CD4+CD8+
`thymocytes secondary to increased apoptosis
`without affecting total numbers of T and B cells in
`the periphery
`An increase in the percent of CD4+CD25+
`regulatory T cells
`An increase in the numbers of alloreactive
`CD103+CD8+ regulatory T cells
`
`Many different tumor types have shown
`responsiveness to mTOR inhibition, including both
`solid tumors and hematologic malignancies
`At doses used for anticancer treatment, mTOR
`inhibitors do not appear to be immunosuppressive
`TSC1/TSC2 (tuberous sclerosis), PTEN (cowden
`disease), LKB1 (Peutz–Jeghers syndrome), NF1
`(neurofibromatosis) are hamartomas syndromes
`linked to the mTOR pathway
`Other potential applications of the antiangiogenic
`effect of mTOR inhibitors include diseases
`characterized by neovascularization, such as
`endometriosis and keratitis
`
`Cardiac stents
`Hypertrophic
`myocarditis
`Pulmonary fibrosis
`Hepatic fibrosis
`ADPKD
`
`Neurodegenerative disorders
`
`Huntington disease
`
`Infectious diseases
`
`Fungal infections
`HIV
`
`Metabolic disorders
`
`Type II diabetes
`Obesity
`
`By blocking progression through the cell cycle
`and causing G1 arrest, mTOR inhibitors have an
`antiproliferative effect that may be beneficial for
`a number of different benign diseases
`
`Sirolimus-eluting stents were approved in 2003 for
`angioplasty to open clogged coronary arteries
`
`Refs. 90–92
`
`Refs. 93–98
`
`Huntington disease is characterized by
`accumulation of intraneuronal proteins that
`interfere with cellular processes. Increased
`autophagy, through inhibition of mTOR, may
`increase the degradation of the intracellular
`proteins that characterize Huntington disease
`
`The mTOR pathway is relatively conserved from
`yeasts to humans. Blocking the pathway in
`yeasts and fungi interferes with their growth and
`proliferation
`Infection with HIV-1 requires expression of the
`viral coreceptor CCR5 on the cell surface. In T
`cells, this expression depends on signaling
`through IL-2, which is blocked by mTOR
`inhibition
`
`Via intracellular connection with the insulin
`receptor/IRS-1/PI3K/Akt pathway and its role in
`cellular response to nutrients, mTOR may be
`important in the development of insulin
`resistance and obesity
`
`Early studies demonstrated potent activity against
`Candida albicans and Aspergillus, among other
`species. However, because of the
`immunosuppressive effects, these are not typically
`used for treatment of fungal infections
`
`Refs. 99, 100
`
`Refs. 16, 18, 101,
`102
`
`Refs. 103, 104
`
`ADPKD, autosomal dominant polycystic kidney disease; ALPS, autoimmune lymphoproliferative syndrome; CCR5, CC chemokine receptor 5; GVHD, graft-versus-host disease;
`HIV, human immunodeficiency virus; IL-2, interleukin-2; IRS-1, insulin receptor substrate-1; mTOR, molecular target of rapamycin; PI3K, phosphoinositide-3 kinase; TSC,
`tuberous sclerosis complex.
`
`CLINICAL PHARMACOLOGY & THERAPEUTICS | VOLUME 82 NUMBER 4 | OCTOBER 2007
`
`385
`
`Ex. 1069-0005
`
`

`
`STATE
`
`ART
`
`adding to the attractiveness of this class of drugs. Classically
`described adverse effects include fatigue, rash, mucositis,
`anorexia, and gastrointestinal effects such as diarrhea and
`nausea; hematologic
`effects
`include
`thrombocytopenia,
`leukopenia, and anemia; and metabolic effects
`include
`hyperlipidemia,
`hyperglycemia,
`and
`hypercholesterole-
`mia.20,33,43,46 In most cases,
`the adverse effects can be
`managed with dose reductions or, in the case of metabolic
`disturbances, with the addition of insulin or a cholesterol-
`lowering medication.
`A rarer and potentially serious side effect that has been
`noted with these agents is interstitial pneumonitis.47 In a
`series of 24 renal
`transplant patients who developed
`pneumonitis associated with sirolimus, symptoms included
`cough, fatigue, and fever in the majority of patients, and eight
`patients also developed dyspnea.48 One patient was asympto-
`matic. Imaging by computed tomography revealed patchy
`bilateral
`infiltrates to be the most common finding (19
`patients). Sirolimus was discontinued in all patients and all
`recovered completely within 6 months. Similar reports after
`treatment with temsirolimus exist49 and pneumonitis is listed
`as an uncommon side effect of everolimus (Certican,
`prescribing information).
`The first approved application of mTOR inhibitors was as
`an immunosuppressant. The effects on the immune system
`may be of some concern when these agents are used for other
`purposes and especially when administered to patients with
`cancer. However, despite the ability to block proliferation of
`certain cells of the immune system, there has not been a
`significant number of infectious complications with the doses
`and schedules used in clinical oncology trials. Most published
`trials have not reported specific analyses of T-cell subsets or
`function, but a phase I study of temsirolimus mentioned that
`lymphocyte cell-surface phenotype analysis and mitogen
`proliferation assays did not
`show any trend toward
`immunosuppression.42
`
`Applications of mTOR inhibitors
`Because of mTOR’s complex effects on protein synthesis and
`cell cycle progression, interfering with its signaling can lead
`to a number of potentially therapeutic outcomes. For
`example, by interfering with the translation of proteins
`involved in cell cycle progression, inhibition of mTOR results
`in arrest in G1 of the cell cycle and an antiproliferative effect
`that may be beneficial for the treatment of cancer and a
`number of benign diseases associated with abnormal
`proliferation. Immunosuppression can occur by interfering
`with protein synthesis within certain cells of the immune
`system, such as T cells. Interfering with the translation of
`HIF-1a has an antiangiogenic effect, and inhibiting the
`phosphorylation of BAD by S6K1 may promote apoptosis.
`Another effect of mTOR inhibition is to promote autophagy,
`although the exact mechanism of this remains unknown.50
`These multiple and diverse effects may potentially result in a
`number of
`important
`therapeutic applications
`for
`the
`treatment of a wide range of diseases. The various diseases
`
`for which inhibition of the mTOR pathway has been studied,
`both clinically and preclinically, along with the rationale for
`why inhibiting the pathway may be efficacious, are listed in
`Table 1.
`
`CONCLUSION
`The mTOR pathway plays a central role in a number of
`important intracellular processes. Blocking the pathway with
`sirolimus or a rapalog has implications for the treatment of
`many diverse diseases. Although many studies are currently
`underway and more data will be collected, the newer mTOR
`inhibitors appear to be potentially interchangeable with
`sirolimus
`(if administered at a comparable dose and
`schedule). Wide interpatient variability in pharmacokinetics,
`metabolism by CYP3A4, and therefore the potential
`for
`numerous drug interactions, poor bioavailability of the oral
`agents, and generally good tolerability are characteristics of
`this class of drugs. There is no convincing evidence thus far
`to suggest that the newer agents offer a large advantage to
`sirolimus. Given the limited patent exclusivity and the
`decrease in cost that can be expected upon expiration of
`the sirolimus patents, perhaps more focus should be given to
`the oldest agent so that it can be effectively applied for the
`multitude of new potential uses being discovered, rather than
`continuing to borrow from it for the production of more new
`inhibitors of mTOR.
`
`ACKNOWLEDGMENTS
`Dr Hartford was supported by T32 GM007019 from the NIH.
`
`CONFLICT OF INTEREST
`The authors declared no conflict of interest.
`
`& 2007 American Society for Clinical Pharmacology and Therapeutics
`
`1.
`
`2.
`
`3.
`
`4.
`
`5.
`
`6.
`
`7.
`
`Wullschleger, S., Loewith, R. & Hall, M.N. TOR signaling in growth and
`metabolism. Cell 124, 471–484 (2006).
`Sarbassov, D.D., Ali, S.M. & Sabatini, D.M. Growing roles for the mTOR
`pathway. Curr. Opin. Cell. Biol. 17, 596–603 (2005).
`Avruch, J., Lin, Y., Long, X., Murthy, S. & Ortiz-Vega, S. Recent
`advances in the regulation of the TOR pathway by insulin and
`nutrients. Curr. Opin. Clin. Nutr. Metab. Care 8, 67–72 (2005).
`Rowinsky, E.K. Targeting the molecular target of rapamycin (mTOR).
`Curr. Opin. Oncol. 16, 564–575 (2004).
`Johannessen, C.M. et al. The NF1 tumor suppressor critically regulates
`TSC2 and mTOR. Proc. Natl. Acad. Sci. USA 102, 8573–8578 (2005).
`Faivre, S., Kroemer, G. & Raymond, E. Current development of mTOR
`inhibitors as anticancer agents. Nat. Rev. Drug Discov. 5, 671–688
`(2006).
`Pende, M. et al. S6K1(/)/S6K2(/) mice exhibit perinatal lethality
`and rapamycin-sensitive 50-terminal oligopyrimidine mRNA
`translation and reveal a mitogen-activated protein kinase-dependent
`S6 kinase pathway. Mol. Cell. Biol. 24, 3112–3124 (2004).
`Petroulakis, E., Mamane, Y., Le Bacquer, O., Shahbazian, D. &
`Sonenberg, N. mTOR signaling: implications for cancer and anticancer
`therapy. Br. J. Cancer 94, 195–199 (2006).
`Kurmasheva, R.T., Huang, S. & Houghton, P.J. Predicted mechanisms
`of resistance to mTOR inhibitors. Br. J. Cancer 95, 955–960 (2006).
`Huang, S., Bjornsti, M.A. & Houghton, P.J. Rapamycins: mechanism of
`action and cellular resistance. Cancer Biol. Ther. 2, 222–232 (2003).
`Sehgal, S.N., Baker, H. & Vezina, C. Rapamycin (AY-22,989), a new
`antifungal antibiotic. II. Fermentation, isolation and characterization.
`J. Antibiot. (Tokyo) 28, 727–732 (1975).
`12. Martel, R.R., Klicius, J. & Galet, S. Inhibition of the immune response by
`rapamycin, a new antifungal antibiotic. Can. J. Physiol. Pharmacol. 55,
`48–51 (1977).
`
`8.
`
`9.
`
`10.
`
`11.
`
`386
`
`VOLUME 82 NUMBER 4 | OCTOBER 2007 | www.nature.com/cpt
`
`Ex. 1069-0006
`
`

`
`13.
`
`14.
`
`15.
`
`16.
`
`17.
`
`18.
`
`19.
`
`20.
`
`22.
`
`23.
`
`24.
`
`25.
`
`26.
`
`27.
`
`28.
`
`29.
`
`30.
`
`31.
`
`32.
`
`33.
`
`Calne, R.Y. et al. Rapamycin for immunosuppression in organ
`allografting. Lancet 2, 227 (1989).
`Kahan, B.D., Chang, J.Y. & Sehgal, S.N. Preclinical evaluation of a new
`potent immunosuppressive agent, rapamycin. Transplantation 52,
`185–191 (1991).
`Cardenas, M.E. et al. Antifungal activities of antineoplastic agents:
`Saccharomyces cerevisiae as a model system to study drug action.
`Clin. Microbiol. Rev. 12, 583–611 (1999).
`Singh, N. & Heitman, J. Antifungal attributes of immunosuppressive
`agents: new paradigms in management and elucidating the
`pathophysiologic basis of opportunistic mycoses in organ transplant
`recipients. Transplantation 77, 795–800 (2004).
`Dumont, F.J. & Su, Q. Mechanism of action of the
`immunosuppressant rapamycin. Life Sci. 58, 373–395 (1996).
`Singh, K., Sun, S. & Vezina, C. Rapamycin (AY-22,989), a new antifungal
`antibiotic. IV. Mechanism of action. J. Antibiot. (Tokyo) 32, 630–645
`(1979).
`Kelly, P.A., Napoli, K. & Kahan, B.D. Conversion from liquid to solid
`rapamycin formulations in stable renal allograft transplant recipients.
`Biopharm. Drug Dispos. 20, 249–253 (1999).
`Napoli, K.L. & Taylor, P.J. From beach to bedside: history of the
`development of sirolimus. Ther. Drug Monit. 23, 559–586 (2001).
`21. Mahalati, K. & Kahan, B.D. Clinical pharmacokinetics of sirolimus.
`Clin. Pharmacokinet. 40, 573–585 (2001).
`Kaplan, B., Meier-Kriesche, H.U., Napoli, K.L. & Kahan, B.D. The effects
`of relative timing of sirolimus and cyclosporine microemulsion
`formulation coadministration on the pharmacokinetics of each agent.
`Clin. Pharmacol. Ther. 63, 48–53 (1998).
`Zimmerman, J.J., Ferron, G.M., Lim, H.K. & Parker, V. The effect of a
`high-fat meal on the oral bioavailability of the immunosuppressant
`sirolimus (rapamycin). J. Clin. Pharmacol. 39, 1155–1161 (1999).
`Yatscoff, R., LeGatt, D., Keenan, R. & Chackowsky, P. Blood distribution
`of rapamycin. Transplantation 56, 1202–1206 (1993).
`Zimmerman, J.J. et al. Pharmacokinetics of sirolimus (rapamycin) in
`subjects with mild to moderate hepatic impairment. J. Clin.
`Pharmacol. 45, 1368–1372 (2005).
`Trepanier, D.J., Gallant, H., Legatt, D.F. & Yatscoff, R.W. Rapamycin:
`distribution, pharmacokinetics and therapeutic range investigations:
`an update. Clin. Biochem. 31, 345–351 (1998).
`Kahan, B.D. et al. Therapeutic drug monitoring of sirolimus:
`correlations with efficacy and toxicity. Clin. Transplant. 14, 97–109
`(2000).
`Stenton, S.B., Partovi, N. & Ensom, M.H. Sirolimus: the evidence for
`clinical pharmacokinetic monitoring. Clin. Pharmacokinet. 44, 769–786
`(2005).
`Chapman, T.M. & Perry, C.M. Everolimus. Drugs 64, 861–872 (2004);
`discussion 864–873.
`Schuler, W. et al. SDZ RAD, a new rapamycin derivative:
`pharmacological properties in vitro and in vivo. Transplantation 64,
`36–42 (1997).
`Dudkin, L. et al. Biochemical correlates of mTOR inhibition by the
`rapamycin ester CCI-779 and tumor growth inhibition. Clin. Cancer
`Res. 7, 1758–1764 (2001).
`Galanis, E. et al. Phase II trial of temsirolimus (CCI-779) in recurrent
`glioblastoma multiforme: a North Central Cancer Treatment Group
`Study. J. Clin. Oncol. 23, 5294–5304 (2005).
`Yee, K.W. et al. Phase I/II study of the mammalian target of
`rapamycin inhibitor everolimus (RAD001) in patients with relapsed or
`refractory hematologic malignancies. Clin. Cancer Res. 12, 5165–5173
`(2006).
`34. Mita, M.M. et al. Phase I, pharmacokinetic (PK), and pharmacodynamic
`(PD) study of AP23573, an mTOR inhibitor, administered IV daily X 5
`every other week in patients (pts) with refractory or advanced
`malignancies. J. Clin. Oncol. (Meeting Abstracts) 22, 3076 (2004).
`Kirchner, G.I., Meier-Wiedenbach, I. & Manns, M.P. Clinical
`pharmacokinetics of everolimus. Clin. Pharmacokinet. 43, 83–95
`(2004).
`Kovarik, J.M. et al. Longitudinal assessment of e

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