throbber
13...-FEB...:.2-00a cBSDS syts N SPA
`,
`.
`.
`ONCOGENE• - BAS t NGSTOKE -
`
`..
`
`LS
`
`7BQ
`
`.
`
`·'l111lliU1ll11 :·~ .
`6256 .. 782Qll0 • VOL 19 ~tJM~t 56
`· ... :.:75 :
`: AC~ ,
`dCP, f,I(
`"
`.
`1:11
`
`f
`
`,.
`
`.,
`
`New Drug Targets and
`Therapies for Cancer
`
`Guest Editor S. Sebti
`
`Volume 19 • Number 56 • 27 December 2000 • Review Issue 6
`
`.·
`
`Breckenridge Exhibit 1006
`Hidalgo
`Page 001
`
`

`

`Volume 19. Number 56. 27 December 2000. Review Issue 6
`
`ONCOGENE
`Reviews
`
`New Drug Targets and Therapies for Cancer
`
`Guest Editor Said Sebti
`
`6549 Guest Editor
`Sa.id M Sebti
`
`6550 The EGF receptor family as targets for cancer therapy
`John Mendelsohn and Jose Baselga
`
`6566 Design of growth factor antagonists with antiangiogenic
`and antitumor properties
`Said M Sebti and Andrew D Hamilton
`
`6574 From oncogene to drug: development of small molecule
`tyrosine kinase inhibitors as anti-tumor and anti(cid:173)
`angiogenic agents
`Michael J Morin
`
`6584 Farnesyltransferase and geranylgeranyltransferase I
`inhibitors and cancer therapy: Lessons from mechanism
`and bench-to-bedside translational studies
`Said M Sebti and Andrew D Hamilton
`
`6594 Development of anticancer drugs targeting the MAP
`kinase pathway
`Judith S Sebolt-Leopold
`
`6600 Small molecule modulators of cyclin-dependent kinases
`for cancer therapy
`Adrian M Senderowicz
`
`6607 Small molecule inhibitors of dual specificity protein
`phosphatases
`Katharine E Pestell, Alexander P Ducruet, Peter Wipf
`and John S Lazo
`
`'
`6627 Bcl-2 family proteins as targets for anticancer drug
`design
`Ziwei Huang
`
`6632 Telomere maintenance mechanisms as a target for drug
`development
`David J Bearss, Laurence H Hurley and
`Daniel D Von Hoff
`
`6642 Critical appraisal of the use of matrix metalloproteinase
`inhibitors in cancer treatment
`Stanley Zucker, Jian Cao and Wen-Tien Chen
`
`6651 Potential roles of antisense technology in cancer
`chemotherapy
`Stanley T Crooke
`
`6660 Replication-selective oncolytic adenoviruses: virotherapy
`aimed at genetic targets in cancer
`David Kirn
`
`6670 ONYX-015 selectivity and the p14ARF pathway
`Frank McCormick
`
`6673 Dendritic cell vaccination for cancer therapy
`Frank 0 Nestle
`
`6680 The rapamycin-sensitive signal transduction pathway as
`a target for cancer therapy
`Manuel Hidalgo and Eric K Rowinsky
`
`6613 ST AT proteins: novel molecular targets for cancer drug
`discovery
`James Turkson and Richard Jove
`
`/
`
`6687 New agents in cancer clinical trials
`Julian Adams and Peter J Elliott
`
`Copyright © 2000 Nature Publishing Group
`
`Subscribing organisations are encouraged to copy and distribute
`this table of contents for internal, non-commercial purposes
`
`This issu'e is now available at:
`www.nature.com/onc
`
`Breckenridge Exhibit 1006
`Hidalgo
`Page 002
`
`

`

`ONCOGENE
`www .nature.com/ one
`
`Oncogene, including Oncogene Reviews, is published by Nature
`Publishing Group, a division of Macmillan Publishers Ltd.
`
`Scope The 2001 volume wi ll include 50 regular issues and 8 issues
`of Oncogene Reviews. The Editors will consider for publication all
`papers and communications on all aspects of cellular growth
`control and the molecular mechanisms underlying malignant
`change.
`
`This journal is covered by Current Contents, EMBASE Excerpta
`Medica and Index Veterinarius.
`
`Editorial Manuscripts and a ll editorial correspondence should be
`sent to: John Jenkins, The Oncogene Editorial Offices, Eden
`House, Enterprise Way, Edenbridge, Kent, TN8 6HF, UK. Tel:
`+ 44
`1732
`860 111 Fax:
`+ 44
`1732
`860222 E-mail:
`oncogene@globalnet.co.uk; or E Premkumar Reddy, The Fels
`Institute for Cancer Resea rch & Molecular Biology, Medical
`Research Building, 3307 North Broad St., Philadelphia, PA
`19140, USA. Tel: + I 215 707 4307 Fax: + 1 2 15 707 1454.
`
`Publisher All business correspondence and enquiries abou t
`supplement publication and sponsorship opportunities shou ld be
`addressed to Oncogene, Nature Publishing Group, Houndmills,
`Basingstoke, Hampshire RG21 6XS, UK. Tel: + 44 1256 329242
`Fax: + 44 1256 810526.
`Publishing Manager: Sue Deeley
`Production Controller: D ebbie Cole
`
`Oncogene, including Oncogene Reviews, is online at
`www.nature.com/onc
`
`Visit the journal's home pages for details of the aims and scope,
`readership, instructions to autho rs and how to contact the Editors
`a nd publishing staff. Use the website to order a subscription,
`reprints, a sample copy or individual articles.
`
`Free to all readers: tables of contents and abstracts for a ll articles
`published since 1997 and the complete text of the January 2001
`issue. Register to receive the table of contents by e-mail as each
`iss ue is published.
`
`Subscriptions - Outside the USA
`Orders must be accompanied by remittance. Cheques should
`be made payable to Nature Publishing Group and sent to:
`The Subscription Department, Nature Publishing Group,
`Houndmills, Basingstoke, Hampshire RG2 l 6XS, UK. E-mail:
`subscriptions@nature.com. Where appropriate, subscribers may
`make payments into UK Post Office Giro Account No: 519 2455.
`Full details must accompany the payment.
`
`Subscriptions - USA
`USA subscribers can call toll free: 1 800 747 3187. Please send
`check/money order/credit card details
`to: The Subscription
`Department, Nature Publishing Group, Houndmills, Basing(cid:173)
`stoke, Hampshire RG2 l 6XS, UK.
`E-mail: subscriptions@nature.com
`
`Prices are set in UK Sterling. Dollar prices are converted from
`UK Sterling at the current exchange rate. Accordingly, your
`credit card charge may vary slightly from the Dollar rate shown.
`To obtain the exact Dollar rate shown, please remit by check. All
`prices, specifications and details are subject to change without
`prior notification. Single
`issues of Oncogene R eviews are
`available. For information, pleasll contact Marketing Dept,
`Nature
`Publishing
`Group,
`Houndmills,
`Basingstoke,
`Hampshire, RG21 6XS, UK. Tel: + 44 1256 351898 Fax: + 44
`1256 328339.
`'
`
`is
`including Oncogene Reviews (ISSN 0950-9232)
`Oncogene,
`published 58 times a year by Nature Publishing Group, c/o
`Mercury Airfreight International Ltd, 365 Blair Road, Avenel,
`New Jersey, NJ 07001, USA. Subscription price for institutions is
`$3520 per annu m. Periodicals postage is paid a t Rahway NJ.
`Postmaster: send address corrections
`to Oncogene, Nature
`Publishing Group, c/o Mercury Airfreight International Ltd, 365
`Blair Road, Avenel, NJ 07001.
`
`Advertisements Enquiries concerning advertisements should be
`addressed to: Robert Sloan, Advertisement Manager, 84 Amos
`Grove, Southgate, London, Nl4 7AR, UK. Tel: + 44 20 8882
`7199 Fax: + 44 20 8882 7299. E-mail: rsloan @ rsa2.demon.co.uk
`
`Reprints and permissions For reprints of any article in this
`journal or reproduction rights, please contact Trace Noel
`(t.noel @nature.com) at the publisher's address (above).
`
`Subscribers to the 200 I online version of the journal have access to
`PDF and HTML files with the full text of all articles published
`since 1997 .
`
`Copyright © 2000 Nature Publishing Group
`ISSN 0950-9232
`
`Subscriptions - 2001 subscription rates
`INSTITUTIONAL SUBSCRIPTIONS
`Combined (electronic online & print)
`EU
`Rest of World
`Electronic onlinc only
`Print (hard copy) only
`EU
`Rest of World
`
`£23 10
`£2420/US$3872
`£2 l00/US$3360
`
`£2 100
`£2200/ US$3520
`
`for information on
`Site licences and institutional online access -
`multi-user or multi-site access to Nature Publishing Group
`products please contact s.archer@macmillan.co.uk or telephone
`+ 44 20 7843 6426 . For o ther enquir ies please contact
`sjsuppo rt@nature.com or telephone + 44 20 7843 4759.
`
`PERSONAL SUBSCRIPTIONS
`Combined (electronic online & print)
`EU
`Rest of World
`Electronic online only
`Print (hard copy) only
`£576
`EU
`Rest of World
`£576/US$922
`Prices for airmail delivery on application
`
`£633
`£633/US$1013
`£576/US$922
`
`All rights of reproduction are reserved in respect of a ll papers,
`articles, illustrations, etc., published in this journal in all countries
`of the world.
`
`All ma terial published in this journal is protected by copyright,
`which covers c1<clusivc rights to reproduce and di~tribule the
`material. No material published in this journal may be reproduced
`or stored on microfilm or in electronic, optical or magnetic form
`without the written au thorisation of the Publisher.
`
`Authorisation to photocopy items for internal or personal use of
`specific clients, is granted by Nature Publishing Group, for
`libraries and other users registered with the Copyright Clearance
`Centre (CCC) Transaction Reporting Service, provided that the
`base fee of $ 15.00 per copy is paid directly to CCC, 222 Rosewood
`Drive, Danvers, USA 0950-9232/00 $15.00+$0.00
`
`Apart from any fair dealing for the purposes of research or private
`study, or criticism or review, as permitted under the Copyright,
`Designs and Patent Act 1988, this publication may be reproduced,
`stored or tra nsm itted, in any form or by any means, only with the
`prior permission in writing of the publishers, or in the case of
`reprograp hic reproduction, in accordance with the terms of
`licences issued by the Copyright Licensing Agency.
`
`T ypese t by Elite T ypese tting Tech niques, Eastleigh, Hants a nd printed in Great Brita in by The Friary Press, Dorchester
`Printed on acid-free paper, effecti ve with Vo lume 9, Iss ue I. 1994
`
`Breckenridge Exhibit 1006
`Hidalgo
`Page 003
`
`

`

`Oncogene (2000) 19, 6680 – 6686
`ª 2000 Macmillan Publishers Ltd All rights reserved 0950 – 9232/00 $15.00
`
`www.nature.com/onc
`
`The rapamycin-sensitive signal transduction pathway as a
`target for cancer therapy
`
`Manuel Hidalgo*,1 and Eric K Rowinsky1
`
`1The University of Texas Health Science Center at San Antonio, The Institute for Drug Development, Cancer Therapy and
`Research Center, San Antonio, Texas, USA
`
`The high frequency of mutations in cancer cells which
`result in altered cell cycle regulation and growth signal
`transduction, conferring a proliferative advantage,
`in-
`dicates that many of these aberrant mechanisms may be
`strategic targets for cancer therapy. The macrolide
`fungicide rapamycin, a natural product with potent
`antimicrobial, immunosuppressant, and anti-tumor prop-
`erties, inhibits the translation of key mRNAs of proteins
`required for cell cycle progression from G1 to S phase.
`Rapamycin binds intracellularly to the immunophilin
`FK506 binding protein 12 (FKBP12), and the resultant
`complex inhibits the protein kinase activity of a protein
`kinase termed mammalian target of rapamycin (mTOR).
`The inhibition of mTOR, in turn, blocks signals to two
`separate
`downstream pathways which
`control
`the
`translation of specific mRNAs required for cell cycle
`traverse from G1 to S phase. Blocking mTOR a€ects the
`activity of the 40S ribosomal protein S6 kinase (p70s6k)
`and the function of the eukaryotic initiation factor 4E-
`binding protein-1 (4E-BP1), leading to growth arrest in
`the the G1 phase of the cell cycle. In addition to its
`actions on p70s6k and 4E-BP1,
`rapamycin prevents
`cyclin-dependent kinase activation, inhibits retinoblasto-
`ma protein (pRb) phosphorylation, and accelerates the
`turnover of cyclin D1 that leads to a deficiency of active
`cdk4/cyclin D1 complexes, all of which can inhibit cell
`cycle traverse at
`the G1/S phase transition. Both
`rapamycin and CCI-779, an ester analog of rapamycin
`with improved pharmaceutical properties and aqueous
`solubility, have demonstrated impressive activity against
`a broad range of human cancers growing in tissue culture
`and in human tumor xenograft models, which has
`supported the development of compounds
`targeting
`rapamycin-sensitive signal-transduction pathways. CCI-
`779 has completed several phase I clinical evaluations
`and is
`currently undergoing broad disease-directed
`e(cid:129)cacy studies. The agent appears to be well tolerated
`at doses that have resulted in impressive anti-tumor
`activity in several
`types of
`refractory neoplasms.
`Important challenges during clinical development include
`the definition of a recommended dose range associated
`with optimal biological activity and maximal therapeutic
`indices, as well as the ability to predict which tumors will
`be sensitive or resistant to CCI-779. Oncogene (2000)
`19, 6680 – 6686.
`
`Keywords: rapamycin; CCI-779; signal
`clinical development
`
`transduction;
`
`*Correspondence: M Hidalgo, Department of Medicine, Division of
`Medical Oncology, The University of Texas Health Science Center at
`San Antonio, 7703 Floyd Curl Dr. Mail code 7884. San Antonio,
`Texas, TX, 78229, USA
`
`Cell proliferation is a complex multifaceted process
`that
`requires
`the synthesis of essential
`regulatory
`proteins involved in the transduction of extracellular
`and autocrine proliferative stimuli. Since several of
`these highly regulated processes are aberrant in many
`types of cancers, conferring a proliferative advantage,
`they are potential strategic targets for therapeutic
`development against cancer
`(Sherr, 2000).
`Indeed,
`several novel classes of therapeutics that interfere with
`discrete essential elements of aberrant signal transduc-
`tion and cell cycle regulation, such as inhibitors of
`various receptor tyrosine kinases, oncogenes, critical
`proteins involved in signal transduction (e.g. Ras, Raf),
`and cyclin-dependent kinases, are being developed as
`anti-cancer agents (Rowinsky et al., 1999, Senderowicz
`and Sausville 2000). One such agent,
`rapamycin
`(sirolimus; Rapamune1; Wyeth-Ayerst, PA, USA), a
`macrolide fungicide isolated from the bacteria Strepto-
`myces hygroscopicus, possesses potent antimicrobial,
`immunosuppressant, and antitumor properties (Baker
`et al., 1978; Sehgal et al., 1975; Vezina et al., 1975).
`Because of its profound immunosuppressive actions,
`rapamycin was initially developed and received reg-
`ulatory approval for the indication of prevention of
`allograft
`rejection following organ transplantation
`(Sehgal, 1995). The antiproliferative actions of rapa-
`mycin have been demonstrated to be due to its ability
`to modulate critical signal transduction pathways that
`link mitogenic stimuli
`to the synthesis of proteins
`required for
`cell
`cycle
`traverse
`from G1
`to S
`(Wiederrecht et al., 1995). Impressive antiproliferative
`activity has been demonstrated following treatment of
`a diverse types of experimental tumors with rapamycin
`(Eng et al., 1984, Muthukkumar
`et al., 1995;
`Seu€erlein and Rozengurt, 1996). However, the poor
`aqueous solubility and chemical stability of rapamycin
`precluded its clinical development as an anti-cancer
`agent. Recently, a series of rapamycin analogs with
`improved aqueous solubility and stability have been
`synthesized and evaluated. CCI-779 (Wyeth Ayerst,
`PA, USA), a soluble ester analog of rapamycin, was
`selected for development as an anti-cancer agent based
`on its prominent anti-tumor profile and favorable
`pharmaceutical and toxicological characteristics
`in
`preclinical studies (Gibbons et al., 2000). Several phase
`I studies of CCI-779 have been completed and disease-
`directed e(cid:129)cacy evaluations in a number of tumor
`types are being performed (Raymond et al., 2000;
`Hidalgo et al., 2000). This review will summarize the
`principal mechanisms of anti-tumor action of rapamy-
`cin, specifically its e€ect on rapamycin-sensitive signal
`transduction pathways, and will discuss the preliminary
`results of experimental and clinical studies with this
`novel class of anti-cancer agents.
`
`Breckenridge Exhibit 1006
`Hidalgo
`Page 004
`
`

`

`Mechanism of action of rapamcyin and
`rapamycin analogs
`
`Rapamycin, and its ester analog, CCI-779, uniquely
`interfere with cell cycle progression from G1 to S phase
`in response to proliferative stimuli by blocking the
`translation of mRNAs of essential cell cycle proteins
`(Wiederrecht et al., 1995). The principal mechanisms
`responsible
`for
`these
`actions, which have been
`elucidated only over
`the
`last
`several years, are
`graphically depicted in Figures 1 and 2.
`
`Upstream actions and the target of rapamycin
`
`Rapamycin binds intracellularly to members of the
`immunophilin family of FK506 binding proteins
`(FKBPs), inhibiting their enzymatic activity as prolyl
`isomerases (Heitman et al., 1991; Koltin et al., 1991;
`Fruman et al., 1995). Although there are many
`members of
`the FKBP family, a large body of
`biochemical and genetic studies suggest that FKBP12
`is the most important binding protein with respect to
`the rapamycin-sensitive signal
`transduction pathway
`(Heitman et al., 1991; Koltin et al., 1991; Fruman et
`al., 1995). The resultant rapamycin-FKBP12 complex
`interacts with and inhibits the activity of a 290 kd
`kinase,
`termed mammalian target of
`rapamycin
`(mTOR) (also known as FRAP, RAFT1, and RAP1)
`(Figure 1) (Sabatini et al., 1994; Sabers et al., 1995;
`Brown et al., 1994; Chiu et al., 1994). mTOR is a
`member of a recently identified family of protein
`kinases
`termed phosphoinositide
`3-kinase
`related
`kinases (PIKKs), which are involved in many critical
`regulatory cellular functions pertaining to cell cycle
`progression, cell cycle checkpoints that govern cellular
`responses to DNA damage, DNA repair, and DNA
`recombination (Sarkaria et al., 1998).
`cells
`In response
`to growth stimuli, quiescent
`increase the translation of a subset of mRNAs whose
`protein products are required for traverse through the
`G1 phase of the cell cycle. mTOR regulates essential
`signal transduction pathways and is involved in the
`
`transduction pathway.
`signal
`Figure 1 Rapamycin-sensitive
`Rapamycin and CCI-779 bind to the immunophilin FK506
`binding protein-12 (FKBP-12). The rapamycin-FKBP12 complex
`blocks the kinase activity of the mammalian target of rapamycin
`(mTOR). The inhibition of mTOR kinase activity inhibits the
`downstream translational regulators 4E-BP1/PHAS and p70s6k.
`inhibition of 4E-BP1/PHAS and p70s6k decrease
`the
`The
`translation of mRNA of specific proteins essential for cell cycle
`progression from G1 to S phase
`
`Targeting rapamycin-sensitive pathway
`M Hidalgo and EK Rowinsky
`
`GROWTH
`FACTOR
`
`mTOR
`
`6681
`
`Figure 2 Rapamycin and CCI-779 inhibits the phosphorylation
`of 4E-BP1/PHAS, preventing the release of the eIF-4E and the
`activation of the eIF4F complex
`
`coupling of growth stimuli with cell cycle progression.
`Phosphatidylinositol 3-kinase (PI3K)/protein kinase B
`(Akt) (PI3K/Akt) appears to be the key modulatory
`factor in the upstream pathway by which growth
`factor-growth factor receptor interactions a€ect
`the
`phosphorylation state of mTOR (Figure 1) (Downward
`1998; Scott et al., 1998; Nave et al., 1999). PI3K plays
`a central
`role
`in cellular proliferation, motility,
`neovascularization, viability, and senescence and is
`upregulated in cancer cells (Shayestech et al., 1999;
`Cantley et al., 1991). Its main physiological function is
`the phosphorylation of the D3 portion of membrane
`phosphoinositols (Cantley et al., 1991; Carpenter et al.,
`1990). Although the role of PI3K and its lipid products
`in signal transduction processes is not clear, the activity
`of this enzyme on tyrosine kinases induces mitogenesis,
`cellular growth, and cellular transformation (Carpenter
`et al., 1990; Varticovski et al., 1994; Hu et al., 1995).
`Recently, several studies have investigated the role of
`small molecule-inhibitors of PI3K as potential tumor
`suppressor agents. For example, the flavonoid deriva-
`tive, LY294002 (Eli Lilly, Indianapolis, IN, USA), a
`potent PI3K inhibitor,
`is a competitive, reversible
`inhibitor of
`the ATP binding site of
`the enzyme
`(Vlahos et al., 1994; Hu et al., 2000). The agent
`induces G1 arrest
`in proliferating cells,
`leading to
`almost complete inhibition of melanoma cell prolifera-
`tion, partial
`inhibition of MG-63 osteosarcoma cell
`growth, and inhibitor of OVCAR-3 ovarian carcinoma
`inducing prominent apoptotic e€ects (Hu et al., 2000;
`Casagrande et al., 1998; Thomas et al., 1997). The
`inhibitor also completely inhibits the retinoblastoma
`protein (pRb) hyperphosphorylation that normally
`occurs during G1 progression and induces up-regula-
`tion of
`the cyclin-dependent kinase inhibitor p27
`(Casagrande et al., 1998).
`There are ample experimental data indicating that
`mTOR functions downstream of the PI3K/Akt path-
`way and is phosphorylated in responses to stimuli that
`activate the PI3K/Akt pathway (Scott et al., 1998;
`Nave et al., 1999; Hu et al., 1995; Sekulic et al., 2000).
`PI3K and Akt are considered proto-oncogenes, and the
`pathway is inhibited by the tumor suppressor gene
`PTEN (Wu et al., 1998). There are other signaling
`pathways that are activated downstream of PI3K, but
`the Akt pathway is of particular interest because of its
`role
`in inhibiting apoptosis and promoting cell
`proliferation by a€ecting the phosphorylation status
`
`Oncogene
`
`Breckenridge Exhibit 1006
`Hidalgo
`Page 005
`
`

`

`Targeting rapamycin-sensitive pathway
`M Hidalgo and EK Rowinsky
`
`6682
`
`of cell-survival and apopotosis-inducing proteins like
`BAD (Downward, 1998).
`
`Downstream effects
`
`Following activation via phosphorylation, mTOR
`modulates two separate downstream pathways that
`control
`translation of
`specific subsets of mRNAs
`including the eukaryotic initiation factor 4E binding
`protein-1 (4E-BP1), also known as PHAS-1 (phos-
`phorylated heat- and acid-stable protein), and the 40S
`ribosomal protein S6 kinase (p70s6k) (Figure 1) (Brunn
`et al., 1997; Gingras et al., 1998; Hara et al., 1997).
`There is ample evidence indicating that the activation
`of either PI3K or Akt
`is su(cid:129)cient
`to induce the
`phosphorylation of both 4E-BP1/PHAS-1 and p70s6k
`through mTOR (Chung et al., 1994; Petrisch et al.,
`1995). Furthermore, treatment of activated PI3K- or
`Akt-expressing cells with rapamycin blocks the phos-
`phorylation of p70s6k and 4E-BP1/PHAS-1, suggesting
`that mTROR is required for these activities (Gingras et
`al., 1998; Burgering and Co€er, 1995).
`The first downstream regulator modulated by the
`phosphorylation status of mTOR, 4E-BP1/PHAS-1, is
`a low molecular-weight protein that
`inhibits
`the
`initiation of translation through its association with
`eIF-4E,
`the mRNA cap-binding subunit of
`the
`eukaryotic initiation factor-4 (eIF-4F) complex (Figure
`2). The binding of 4E-BPs to eIF-4E is dependent on
`the phosphorylation status of 4E-BP. In the unpho-
`sphorylated state that predominates in quiescent cells,
`4E-BP1/PHAS-1 binds to eIF-4E which inhibits its
`activity (Sonenberg et al., 1998).
`In response to
`proliferative stimuli, 4E-BP1/PHAS-1 becomes phos-
`phorylated through the action of mTOR and other
`kinases which decrease its binding a(cid:129)nity for eIF-4E.
`These actions promote the dissociation of the 4E-BP1/
`PHAS-1 complex, increasing the availability of eIF-4E,
`which can then bind to eIF-4G, -4B, and -4A, forming
`the multisubunit eIF-4F complex. These interactions
`lead to an increase in the translation of mRNAs with
`regulatory elements in the 5’-untranslated region such
`as cyclin D1 and ornithine decarboxylase (Sonenberg
`and Gingras, 1998; Rosenwald et al., 1995; Shantz and
`Pegg 1995). In contrast, growth factor deprivation or
`treatment with rapamycin results in the dephosphor-
`ylation of 4E-BP1/PHAS-1, an increase in eIF-4E
`binding, and a concomitant decrease in the translation
`of mRNAs for cell cycle progression from G1 to S
`phase, as shown in Figure 2 (Brunn et al., 1997;
`Gingras et al., 1998).
`The second downstream target modulated by mTOR
`is the kinase p70s6k. Upon activation by proliferative
`stimuli mediated by the PI3K/Akt signal transduction
`pathway, mTOR phosphorylates/activates
`p70s6k,
`which,
`in turn, phosphorylates the 40S ribosomal
`protein S6 (Hu et al., 2000). The phosphorylation of
`S6 leads to the recruitment of
`the 40S ribosomal
`subunit into actively translating polysomes,
`thereby
`enhancing the translation of mRNAs with a 5’ terminal
`oligopolypyrimidine including those that encode for
`ribosomal proteins, elongation factors, and insulin
`growth factor – II. Rapamycin treatment results in a
`rapid and profound dephosphorylation of p70s6k,
`suppressing its activity (Seu€erlein and Rozengurt
`1996; Grewe et al., 1999).
`
`Oncogene
`
`In addition to its well characterized inhibitory
`actions on p70s6k and 4E-BP1/PHAS-1,
`rapamycin
`interferes with other intracellular processes involved
`in cell cycle progression which undoubtedly contribute
`to its antiproliferative actions. These other actions are
`particularly important in exponentially growing cells
`were inhibition of p70s6k phosphorylation by rapamycin
`does not result in G1 cell cycle arrest (Kawamata et al.,
`1998). Rapamycin increases the turnover of cyclin D1
`at the mRNA and protein level (Hashemolhosseini et
`al., 1998). This e€ect,
`in addition to the decrease
`translation of cyclin D1 mRNA subsequently to 4E-
`BP1/PHAS inhibition, results in a relative deficiency of
`cyclin D1 in the cdk4/cyclin D1 complexes required for
`retinoblastoma protein phosphorylation (Morice et al.,
`1993; Nourse et al., 1994). Rapamycin also blocks the
`elimination of
`the cyclin dependent kinase (cdks)
`inhibitor p27 and facilitates the formation of
`the
`cyclin/cdks-p27 complexes (Nourse et al., 1994; Luo
`et al., 1996). Furthermore,
`in exponentially growing
`cells, rapamycin upregulates p27 at both the mRNA
`and protein level and inhibits cyclin-A-dependent
`kinase activity (Kawamata et al., 1998). It appears
`that, although rapamycin treatment results in cell cycle
`arrest at the G1/S transistion, the precise mechanism
`responsible for this e€ect is both cell cycle- and cell
`type-specific.
`By inhibiting the translation of critical mRNAs
`involved in the G1 to S phase transition in response to
`mitogenic stimuli, and by interfering with the balance
`of cyclin/cyclin-dependent-kinase/cyclin-dependent ki-
`nase inhibitors in the early phases of the cell cycle,
`rapamycin inhibits
`the
`growth of
`cancer
`cells.
`Rapamycin exerts concentration-dependent inhibition
`of cell proliferation and tumor growth in a variety of
`murine and human cancers growing in both cell culture
`and xenograft models including B16 melanoma, P388
`leukemia, MiaPaCa-2 and Panc-1 human pancreatic
`carcinoma and tumors derived from B-cell lymphoma,
`small cell
`lung cancer carcinoma, and childhood
`rhapdomyosarcoma (Muthukkumar et al., 1995; Seuf-
`ferlein and Rozengurt 1996; Hosoi et al., 1999).
`Rapamycin also induces p53-independent apoptosis in
`childhood rhabdomyosarcoma cell lines and enhances
`the apoptotic-inducing e€ects of cisplatin in murine T-
`cell, human HL-60 promyelocytic leukemia, and hu-
`man ovarian SKOV3 cancer cell
`lines (Hosoi et al.,
`1999; Shi et al., 1995).
`
`Clinical development
`
`The unfavorable pharmaceutical properties of rapamy-
`cin, particularly its poor aqueous
`solubility and
`instability, preclude its clinical development as an
`anti-cancer agent, and, therefore, soluble ester analogs
`were synthesized and evaluated as a collaborative e€ort
`between investigators at Wyeth-Ayerst and the Na-
`tional Cancer Institute. CCI-779, a water soluble ester
`analog of rapamcyin, was selected for further develop-
`ment based on favorable pharmaceutical, toxicologic,
`and antitumor profiles in preclinical evaluations. In the
`National Cancer
`Institute human tumor cell
`line
`screen, CCI-779 and rapamycin demonstrated similar
`anti-tumor profiles and potencies (Pearson correlation
`coe(cid:129)cient, 0.86), with IC50 values frequently less than
`
`Breckenridge Exhibit 1006
`Hidalgo
`Page 006
`
`

`

`Targeting rapamycin-sensitive pathway
`M Hidalgo and EK Rowinsky
`
`6683
`
`1078 M (Gibbons et al., 2000). Platelet-derived growth
`factor stimulation of
`the human glioblastoma line
`T98G was markedly inhibited (IC50, 1 pM), consistent
`with its proposed mechanism of action as an inhibitor
`of signal transduction, and growth-inhibited cells were
`arrested in G1 (Gibbons et al., 2000). In studies
`involving cancer cell
`lines growing in tissue culture,
`human prostate, breast, and small cell lung carcinomas,
`glioblastoma, melanoma, and T-cell
`leukemia were
`among the most sensitive cancers to CCI-779, with
`IC50 values in the nanomolar range (Gibbons et al.,
`2000). Significant growth inhibition was also observed
`following treatment of a variety of human tumor
`xenografts with CCI-779, but the preponderance of
`tumor growth inhibition,
`in contrast to overt tumor
`regression, suggests that subsequent disease-directed
`trials
`should be designed to assess
`this potential
`outcome. In addition, several
`intermittent CCI-779
`dosing regimens were
`e€ective
`in human tumor
`xenograft studies, which is important in view of the
`possibility that prolonged immunosuppression can
`result from both rapamycin and CCI-779 administered
`on continuous dose-schedules and since the immuno-
`suppressive e€ects of rapamycin analogs have been
`demonstrated to resolve in approximately 24 h follow-
`ing treatment (Gibbons et al., 2000).
`Thus far, CCI-779 has been evaluated in two phase I
`studies, in which the agent has been administered as a
`30-min IV infusion weekly and as a 30-min IV infusion
`daily for 5 days every 2 weeks (Raymond et al., 2000;
`Hidalgo et al., 2000). These schedules were selected for
`initial clinical evaluation to avoid prolonged drug-
`induced immunosuppression which resolves
`shortly
`after drug treatment. However, similar to the tradi-
`tional paradigm used to develop nonspecific cytotoxic
`agents,
`these phase
`I
`studies were designed to
`determine the maximum tolerated dose based on
`dose-limiting toxicities as classically defined. To date,
`16 patients have been treated with CI-779 at doses
`ranging from 7.5 to 220 mg/m2/week on the weekly
`schedule and 35 patients have received doses ranging
`from 0.75 to 24 mg/m2/day on the daily for-5-days
`every-2-week schedule. The principal toxicities of CCI-
`779 have included dermatologic toxicity, myelosuppres-
`sion, reversible elevations in liver function tests, and
`asymptomatic hypocalcemia. The cutaneous e€ects,
`which have been relatively more common with CCI-
`779 on the weekly schedule, have been multifaceted.
`Dermatologic manifestations have included aseptic
`folliculitis, erythematous maculopapular rashes, ezce-
`matoid reactions, dry skin, vesicular lesions, and nail
`disorders. The principal hematologic toxicity has been
`thrombocytopenia, whereas anemia,
`leukopenia, and
`neutropenia have generally been less common and
`severe. Other toxicities and biochemical abnormalities,
`which have generally been mild to moderate in severity,
`reversible, and noted over wide dosing ranges include
`mucositis, hypertriglyceridemia, hypercholesterolemia,
`and reversible decrements in serum testosterone. The
`maximum tolerated doses of CCI-779 on the daily-for-
`5-day-every-2-week schedule are projected to be 15 and
`24 mg/m2/day in patients with minimal or extensive
`prior myelotoxic therapy, respectively, whereas the
`maximum tolerated dose has not been determined for
`CCI-779 administered on a weekly schedule. The
`preliminary results of pharmacokinetic studies indicate
`
`dose-dependent pharmacokinetics, elimination half-life
`values of approximately 15 – 17 h, and preferential
`partitioning of CCI-779 in red blood cells (Raymond
`et al., 2000; Hidalgo et al., 2000). Major tumor
`responses (partial responses; 450% reduction in the
`sum of the bidimensional product of all measurable
`lesions) have been noted in previously-treated patients
`with renal cell carcinoma and non-small cell
`lung
`(550%
`carcinoma,
`and minor
`tumor
`responses
`reduction in the sum of the bidimensional product of
`all measurable lesions) have been observed in pre-
`viously-treated patients with soft tissue sarcoma, serous
`papillary carcinoma of
`the
`endometrium, breast
`carcinoma, squamous cell carcinoma of the skin, and
`non-Hodgkins
`lymphoma. The fact
`that CCI-779
`consistently induced tumor regressions at relatively
`nontoxic doses in the phase I studies is particularly
`noteworthy, since this observation suggests that the
`optimal therapeutic dose of CCI-779 may be lower
`than the maximum tolerated dose.
`Disease-directed e(cid:129)cacy studies of CCI-779 in a
`broad range of tumor types will be initiated following
`the completion of phase I studies. Based on the results
`of experimental studies involving malignant gliomas
`that rely on paracrine or autocrine stimulation of
`receptors that trigger the PI3/Akt pathway, studies
`directed at assessing the anti-tumor activity of CCI-779
`in patients with glioma are planned. However, since
`CCI-779 is principally metabolized by cytochrome
`P450 mixed function oxidases that are induced by
`many types of anticonvulsant agents commonly co-
`administered to patients with malignant gliomas, the
`toxicities, pharmacokinetics, and optimal dose-schedule
`of CCI-779 are being evaluated in patients concurrently
`receiving treatment with P450 mixed function oxidase-
`inducing anticonvulsant agents. In addition, phase I
`studies evaluating the feasibility of administering CCI-
`779 in combination with various cytotoxic chemother-
`apeutics such as 5-fluorouracil

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