throbber
An Official Journal
`of the
`American Association
`for
`Cancer Research
`
`·'. ·_:;-.·
`
`···- .. ".,, ~- • .• « •• :.~~;.·:-: ·:' .••
`
`. .
`
`':-: .·· ...
`
`i
`
`.. ,.
`
`•
`
`. .::.------~ ..
`... ,
`
`·.
`
`·' ,• -·~
`
`f ~ ••
`
`--·
`, '
`t
`
`~'
`
`.. :: t~·i~{t=_..~~:~~::F~··
`
`. -... .
`_ ... :- .- ... ·-·-·
`
`_:·...
`
`:..::\.•-
`
`'"'
`. -· ...
`' .. ..,: ..
`
`. ~..
`
`" ~~
`<.:·
`
`•
`
`l~lff~
`
`·-<>-
`
`,
`' • r
`
`I
`
`?
`j
`~ ----'"';<'-~--..,
`:~ Supplement to· ·.
`
`l.l
`
`.:r. Clinical
`:~:·cancer::
`:~ 1 Resetirch
`?~----~---"""
`'·' 0
`
`. _. - ·-' . .
`
`. ............ --:
`
`. . . .
`
`I
`,-}
`\
`
`... . ~
`. .
`
`-:.
`.., .....
`
`· I.11 itci-E~Rt~~c~--: <,
`syrrip:osiuni)on,··_.:.; :/::_,:·· .. ·
`. _new d.riig~.J~-~-... ~~-· ~ ... _~):~-..·:.;/.::.:::~· :(cid:173)
`.. , ::. :~·~nee~ ,therapy·~-·:·": ... ,)~ ...
`
`-
`
`• •
`
`'
`
`•
`
`~
`
`.......... ,::.
`
`•
`
`\
`
`~
`
`I
`
`•
`
`~
`n
`r;,
`ll
`tJ
`I
`!I
`b
`/,
`i
`\!
`h
`tl
`I
`n
`u
`II
`II r
`"
`u
`A
`9
`\1
`y
`6
`~
`t
`~
`Q•
`b t
`?' C) ·r
`
`_.-.·
`
`;.;.
`·>· -·
`
`'.
`
`...
`~-:· ·::~
`. '\'·.· .....
`
`,.
`
`..
`
`1-J
`If
`ll
`,?
`·ti ,
`:.t
`. b
`a
`.b
`:+
`(l
`~
`' Q
`' Q
`do ..
`b
`.. ti ~ovem er 2000 o Volume 6 0 Supplement
`-"? 1P. 4467s-4597s o ISSN 1078-0432
`c,i
`II
`~
`
`West-Ward Exhibit 1082
`Hidalgo NCI 2000 Abs #413
`Page 001
`
`

`

`4548s
`
`Poster Session 17
`
`Clinical Cancer Research o Volume 6 o November 2000 (Supplement)
`
`408 Inhibition of Ras expression in lymphoma by isoform specific anti(cid:173)
`sense oligonucleotides: effects on proliferation, survival and gene expres(cid:173)
`sion profiled by cDNA microarrays. Clarke, Paul, A.1. Sumpter, K.1
`·2 • Te
`• Mania. B. 4
`Poele, R. 1 , Di Stefano, F. 1 , Wooster, R. 3 • Cunningham, D.2
`, and
`
`Workman, P1 • CRC Centre for Cancer Therapeutics 1, Dept. Molecular Carcino(cid:173)
`genesis3, Lymphoma Unit2, Institute of Cancer Research and Royal Marsden
`Hospital, UK, and /SIS Pharmaceuticals, USA4
`•
`Ras proteins play a central role in signal transduction. The specific function of
`individual Ras-isofonns is unclear. but evidence suggests that they may have
`distinct functions. Different isoforms are mutated in specific tumours for example
`Kirsten (K~ - ras mutations in colorectal cancer and N-ras mutations in haemato(cid:173)
`logical malignancies. The use of recombinant knock-out mice has demonstrated
`that Ki-ras expression is essential for embryogenesis, while N-ras and Harvey (Ha)
`- ras knockout mice show few defects. Therapeutic strategies targeting the ex(cid:173)
`pression or post-translational modification of Ras are currently under development.
`However it is not entirely clear which particular isoform. if any, iS critical to tumour
`cell proliferation or survival. In this study our aims were to characterise the contri(cid:173)
`bution of each specific isofonn to lymphoma oell proliferation, survival and gene
`expression by using an antisense strategy to specifically inhibit the expression of
`eacn isofonn in non-Hodgkin's lymphoma (NHL) cell lines. Six different lymphoid
`cell lines were treated with antisense oligonucieotides specific for each individual
`Ras isofonn. Western blotting confinned that these antisense oligonucleotides
`specifically inhibited the expression of their target genes. Growth in media supple(cid:173)
`mented with 10% serum was not effected by any of the Ras antisense oligonu(cid:173)
`cleotides. However, under restrictive growth conditions (0.1 % serum) proliferation
`was significantly reduced following treatment with N-ras antisense. FACS analysis
`demonstrated there was no significant increase in cell death following treatment
`with any of the antisense molecules, nor was there a marked change in cell cycle
`distribution. Gene expression profiling using an in-house microarray with 5800
`cDNAs detected changes specific for the inhibition of each Ras-isofonn. In con(cid:173)
`clusion, N-ras appears to be required for lymphoma cell proliferation and would be
`a suitable therapeutic target in NHL
`
`409 Cerivastatin triggers apoptosis of AML cells with higher potency
`than lovastatin. Tan. M., Xia. Z .• Wong, W. W.-L, Dimitroulakos. J .. Minden,
`M., and Penn. LZ. Ontario Cancer lnstitvte, University Health Network, Toronto,
`Canada.
`The vastatin family of drugs inhibits the 3-hydroxy-3-methylglutaryl coen·
`zyme A (HMG-CoA) reductase enzyme that catalyzes the rate-limiting step in
`the mevalonate pathway. We have previously reported that lovastatin, a mem·
`ber of the vastatin family, induces apoptosis in human acute myeloid leukemic
`(AML) cells. We have recently shown that inhibiting protein geranylgeranylation
`downstream of mevalonate is critical for lovastatin-induced apoptosis. In the
`present study we evaluate the potency and mechanism of cerivastatin. a third
`generation member of the vastatin family. Unlike lovastatin, which requires
`activation from pro-drug to active form. cerivastatin is administered in active
`configuration. Our results show that cerivastatin is at least ten fold more potent
`than lovastatin at inducing apoptosis in AML cell-lines as well as in primary AML
`patient samples in vitro. Importantly, like lovastatin, cerivastatin is not cytotoxic
`to normal human bone marrow cells. Moreover, we show that inhibition of
`protein geranylgeranylation is essential for cerivastatin to trigger apoptosis.
`These results strongly suggest that the active form of the vastatin family
`induces apoptosis in AML cells by inhibiting HMG-CoA reductase. In addition,
`the apoptotic activity of the vastatin family occurs through a common mecha(cid:173)
`nism that requires the abrogation of protein geranylgeranylation. Taken to·
`gether, cerivastatin may be the drug of choice for inhibiting HMG-CoA reduc(cid:173)
`tase and triggering tumor cell apoptosis in the clinical management of AML.
`
`410 Blocking protein geranylgeranylation is essential for apoptosis of
`AML cells triggered by inhibitors of HMG-CoA reductase. Zhenlei Xia 1 •
`Melissa M. Tan 1, Jim Dimitroulakos', Mark D. Minden 1·2 , W. Wei-Lynn Wong 1·2 ,
`Linda z. Penn"2 • Department of Cellular and Molecular Biology, the Ontario
`Cancer Institute, University Health Network, Toronto 1 Department of Medical
`Biophysics, University of Toronto, Toronto2•
`We have previously reported that lovastatin, a 3-hydroxy-3-methylglutaryl
`coenzyme A (HMG-CoA) reductase inhibitor, induces apoptosis in human acute
`myeloid leukemia (AML) cells. To understand the critical biochemical mecha(cid:173)
`nism linking inhibition of the mevalonate pathway and apoptosis induced by
`lovastatin in AML cells, add-back experiments were employed in the present
`study. Apoptosis induced by lovastatin was completely prevented by meval·
`onate and geranylgeranyl pyrophosphate (GGPP), while only partially prevented
`by farnesy1 pyrophosphate (FPP). Other products of the mevalonate pathway
`Including cholesterol, squalene, lanosterol, desmosterol, dolichol. dolichol
`phosphate, ubiquinone, and isopentenyladenine had no effect. On the induction
`of apoptosis by lovastatin, our results suggest that the inhibition of geranylgera(cid:173)
`nylation of target proteins is the predominant mechanism of lovastatin-induced
`apoptosis in AML cells. Supporting evidence showed that the geranylgeranyl
`transferase inhibitor (GGTI-298) mimicked the effect of lovastatin, whereas the
`famesyl transferase inhibitor (FTl-277) was much less effective at inducing
`apoptosis in AML cells. In addition, inhibition of Rap1A and Rab5 prenylation
`was observed with lovastatin and GGTl-298 treatment, but not with FTl-277.
`GGPP completely reverted the effect of lovastatin on protein prenylation, while
`FPP had no significant effect on these processes. We conclude that blockage
`of the mevalonate pathway, especially inhibition of protein geranylgeranylation
`is a critical mechanism in lovastatin·induced apoptosis in AML cells.
`
`411 The use of expression profiling to Identify genes regulated by the
`Pl3K inhibitor L Y294002. Robert te Peele, Karine Maillard, Richard Wooster
`and Paul Workman. The Institute of Cancer Research, 15 Cotswold Road,
`Sutton, SM2 5NG, UK.
`The Pl3K pathway plays an important role in regulating cellular proliferation
`and survival, and mutations in components of this pathway have been impli(cid:173)
`cated in cancer. To determine which genes are regulated by the P13K pathway,
`we have used DNA microarrays to establish expression profiles in tumour cells
`treated with the P13K inhibitor L Y294002 versus non-treated colon adenocar(cid:173)
`cinoma cells.
`Our DNA expression arrays are based on cDNAs from the Unigene set of ESTs.
`PCR products from an initial set of 5808 cDNAs were gridded on a single double
`poly-L-Lysine coated glass microsoope slide. Hybridisations were performed using
`Cy3- and CyS-labelled first strand cDNA from treated and untreated cell line
`mRNA. After washing. the slides were scanned using an Axon Genepix 4000
`scanner. Data were analysed with the Genepix software. Ratios from this process
`were normalised and used for interpretation and cluster analysis.
`Concentrations of 30 and 100 "'M of LY294002 were used in time course
`experiments, both concentrations inhibited phosphorylation of the Akt/PKB
`protein. The lower concentration mainly caused cell cycle arrest whereas at the
`higher concentration cell death was induced. Expression changes in genes
`involved in cell cycle regulation and apoptosis were observed. Cluster analysis
`grouped the cells treated with 30 "'M and those treated with 100 "'M on
`separate branches. A subset of differentially expressed genes in the cells
`treated with 30 or 1 00 µ.M of L Y294002 could be identified. One of the genes
`was an apoptotic cysteine protease.
`Expression profiling using DNA microarrays can be used to identify P13K
`inhibitor regulated genes and may help understand signal transduction path·
`ways for proliferation and survival in tumour cells. This work was supported by
`the /CR and the CRC.
`
`412 p53 cooperates in enforcing rapamycin-induced G1 arrest and pro(cid:173)
`tects from apoptosis. S. Huang, LN.Liu, H. Hosoi, M.B.Dilling, T. Shikata, P.J.
`Houghton. Dept. Molecular Pharmacology, St. Jvde Children's Research Hos(cid:173)
`pital, Memphis, TN 38105, U.S.A
`The macrolide antibiotic rapamycin specifically inhibits the Ser!Thr kinase
`activity of mTOR/FRAP, a signaling molecule that links extracellular growth
`factor signals to protein translation. Rapamycin is a potent inhibitor of tumor cell
`growth. and an analog, CCl-779 is currently under Phase I investigation for
`treatment of cancer. In this work, the relationship between G1 checkpoint
`function and rapamycin-induced apoptosis was examined using human rhab·
`domyosarcoma cells, and mouse embryo fibroblasts (MEFs). Serum-starved
`tumor cells expressing mutated p53 alleles became apoptotic when exposed to
`rapamycin, and were protected by expression of a rapamycin resistant mutant
`mTOR. Replication defective adenovirus expressing either p53, or p21"'P'. but
`not control virus. induced Gl accumulation, up-regulation of p21c1P1 , and
`complete protection of cells from raparnycin-induced apoptosis, but not against
`apoptosis induced by cycloheximide. Under serum-free conditions rapamycin
`induced apoptosis of MEFs with p53·1• or p21c•P•-1- genotype, but not wild type
`or p19ARF-t- cells. Cycloheximide induced apoptosis independent of the geno(cid:173)
`type. Ad-p21 and Ad-p53 rescued cells from rapamycin-induced death,
`whereas expression of dominant negative p53 in wild type MEFs restored the
`ability of rapamycin to induce apoptosis. Under serum-containing conditions
`
`rapamycin suppressed levels of cyclin A, only in wild type, and p1gARF·1• MEFs.
`Rapamycin also inhibited DNA replication or S-phase progression significantly
`more in wild type, p19ARF-1-. and p21c•p1-1- MEFs compared to p53-1· cells.
`Suppression of G1 progression by p53 was therefore partly independent of
`p21c•P1
`• However, p21ciP1-mediated G1 block ultimately protected cells from
`apoptosis. The data suggest p53 cooperates in enforcing Gt cell cycle arrest
`leading to a cytostatic response to rapamycin. In contrast, in tumor cells having
`deficient p53 function the response to this agent may be apoptosis. These data
`suggest a mechanistic basis for selective tumor cytotoxicity for rapamycin and
`CCl-779. Supported by USPHS grant CA77776 and by ALSAC.
`
`413 Phase I and pharmacological study of CCI-n9, a cell cycle Inhibi(cid:173)
`tor. M Hidalgo, E Rowinsky, C Erlichman, R Drengler, B Marshall, A Adjei, L
`Hammond, E Galanis, T Edwards, J Burton, J Boni, A Teicher, G Dukart, and J
`Buckner. CTRC, San Antonio, TX; Mayo Clinic, Rochester, MN; Wyeth-Ayerst
`Research, Radnor, PA.
`CCl-779, an ester of rapamycin, binds to FKBP-12 intracellularly, forming a
`complex that inhibits the kinase activity of mammalian target of rapamycin
`(mTOR). This interferes with key signal transduction pathways, including those
`regulated by p70s6 kinase and PHAS-1 protein, resulting in inefficient translation
`of proteins involved in cell cycle progression. CCI· 779 inhibited tumor growth in
`preclinical models and is being developed for treatment of cancer. This study is
`evaluating the safety and pharmacokinetics (PK) of escalating doses of CCl-779
`administered as a 30·minute IV infusion daily x 5 every 2 weeks in patients (pts)
`with solid neoplasms. As of May 2000, 63 patients had been entered. This
`abstract is based on 45 patients in the data base as of April 2000, treated with
`<;loses ranging from 0.75-19.1 mg/m2/d. A total of 214 courses were adminis(cid:173)
`tered. Two episodes of DLT in the first cycle were observed: asymptomatic,
`grade 3 hypocalcemia (2.16 mg/m2/d) and grade 3 elevation in transaminases
`(19.1 mg/m2/d). In addition. grade 3 thrombocytopenia requiring dose reduction
`was observed in 5 pts, including 3 heavily pretreated (HP) pts at 19.1 mg/m2/d.
`Idiosyncratic reactions to the infusion (including flushing, SOB, chest pain) were
`
`Proceedings of the 2000 NCloEORTCoAACR Symposium
`
`West-Ward Exhibit 1082
`Hidalgo NCI 2000 Abs #413
`Page 002
`
`

`

`Clinical Cancer Research o Volume 6 o November 2000 (Supplement)
`
`Poster Session 17
`
`4549s
`
`observed also. Other adverse events, generally mild-moderate in severity, oc(cid:173)
`curred over a broad range of doses. Toxicities include asthenia, cutaneous
`toxicity, mucositis, and hypertriglyceridemia. In 17 pis receiving 0.75 to 3.12
`mg/m2/d, CCI-779 exhibited little accumulation from cycle to cycle, preferenlial
`binding to RBCs. dose-related increase in AUC, and mean to,., of 32.6 h.
`Preliminary evidence of antitumor activity has been· observad, with 1 PR (non(cid:173)
`small cell lung cancer) and minor responses in other tumor types. The safety
`profile and antitumor activity observed to date, associated with plasma con(cid:173)
`centrations at which biological activity was observed in vitro, are encouraging.
`
`414 CCl-779, an ester analogue of rapamycin that interacts with PTEN/
`Pl3 kinase pathways: A phase I study utilizing a weekly intravenous sched(cid:173)
`ule. E. Raymond, J. Alexandre, H. Depenbrock, N. Ady Vago, S. Faivre, A.
`Lahr-Aandak, E. Matennan, J. Boni, S. Abbas, E. Angevin, B. Escudier, J.P.
`Armand. /nstitut Gustave Roussy, Villejuif, France; Onkologische Tagesklinik &
`Wyeth Ayerst Research I Genetics Institute, Munich, Germany.
`Background. CCl-779 inhibits mTOR. thus the phosphorylation of elF4E(cid:173)
`BP1 and p7056 kinases, prevents e1F4E to initiate protein synthesis and the
`phosphorylation of the ribosomal protein S6 required for the translation of
`mRNAs. Patients and Methods. CCl-779 was given as a weekly 30-min
`infusion in patients (pts} with advanced tumors using the modified CAM. Re(cid:173)
`sults. 1 Bpts (MiF: 12/6) received: 7.5 (1 pt). 15 (2pts}, 22.5 (1 pt}, 34 (3pts}, 45
`(4pts), 60 (1pt), 80 (1pt}, 110 (1pt}, 165 (1pt} and 220 mg/m2/week (3~ts}. OLT
`was observed in only 1 pt; MTO has not been reached. No prolonged 1mmuno(cid:173)
`suppression has been induced. Grade (Gr) 1-2 skin toxicity was observed:
`dryness with mild itching (6pts), eczema-like lesions (2pts}, ?~b-acute .u.rticaria
`(2pts}, and aseptic folliculitis (11 pis}. Gr1-2 and Gr-3 m~cos1t1s/stomatit1s were
`observed in 10pts and 1pt. respectively. All pts rece1v1ng ;;,, 8 doses experi(cid:173)
`enced Gr-1 nail changes. Thrombocytopenia was observed in 9pts; 2pts with
`G·3 at 34 and 45 mg/m2/week. Leukopenia was reported in 4pts and anemia in
`7pts. Asymptomatic increases of triglyceride and cholesterol levels were ob(cid:173)
`served in 9pts and 5pts, respectively. A reversible decrease in testosterone
`concentrations with increased levels of LH/FSH were observed in 5/9 men
`receiving :;,4 doses at dosages 2:15mg/m2/week. Pharmacokinetic analysis
`from 12pts (doses: 7.5-60 mg/m2/week) indicates that CCl-779 Cmax increased
`linearly but AUC increased sub-proportionately. Clearance and volume of dis(cid:173)
`tribution at steady state increased with increasing dose. Mean half-life was
`about 20hrs. Of the 16pts evaluable for anti-tumor activity, 3pts had a partial
`response (renal cell carcinoma with lung metastases; neuro-endocrine tumor
`with hepatic metastases and breast cancer with liver, lymph node and peri(cid:173)
`orbital metastases). Conclusion. Current data show that CCl-779 has promis(cid:173)
`ing activity and mild-moderate toxicity over a broad range of doses.
`
`415 Effect of the proteasome inhibitor PS-341 on cell cycle progression
`and bcl-2: A potentially unique mechanism of action. A. Perez-Solert. YH
`L!ngt. B Ngt. J Adams', P Elliott', L Liebest. Kaplan Cancer Center, tNew York
`University School of Medicine, New York, NY, and "Millennium Pharmaceuticals,
`Cambridge, MA.
`PS-341 is a proteasome inhibitor currently in Phase I clinical evaluation. We
`studied the effects of PS-341 on cell cycle progression and related events in
`human NSCLC H460 (p53 wild type} and H358 (p53 null} cells. Exposure to 0.1
`tJ.M PS-341 for 6 h resulted in a marked accumulation of cells at G2/M. This
`blockade was associated with a 6-10 fold time-dependent accumulation of
`cyclins A and Band a 10-fold elevation of cyclins A and B kinase activities as
`assessed by 32 P-,-ATP incorporation into histone-H1. In addition, bcl-2 phos(cid:173)
`phorylation, a marker of mitotic arrest, was detected as early as 3 h after
`exposure to PS341. More importantly, a 25 kDa bcl-2 degradation product was
`detected as early as 12 h after exposure to PS341. This degradation product
`appeared specific for proteasome inhibition since it was observed with the
`proteasome inhibitors MG132 and PSI but not with the chemotherapeutic
`agents paclitaxel. vinblastine, camptothecin, etoposide, and cisplatin, ?r the
`PKC inhibitor staurosporine. In addition, it was not caspase-dependent since 1t
`was observed in the presence of caspase inhibitors and appeared to localize in
`the triton X-100 insoluble cellular fraction. In view of the ability of PS341 to
`induce arrest at G2/M we then studied in vitro cytotoxicity of the combination of
`PS341 and the antitubulin agent docetaxel against H460 and H358 cells. Cells
`were treated concomitantly with PS341 (0.1 tJ.M or 0.5tJ.M) and docetaxel (0.1 to
`4tJ.M) for 48 h. An additive cytotoxic effect was observed with the combination
`0.5tJ.M PS341 and 0.5 and 1 tJ.M docetaxel. In conclusion, our results indicate
`that PS341 induces unique changes in bcl-2 that appear to be specific for
`proteasome inhibition. The functional consequences of these bcl-2 changes
`and their potential relationship with the demonstrated ability of this agent to
`retain its cytotoxicity against bc!-2 transfected cells is being investigated.
`
`416 A phase I pharmacodynamic study of the proteasome inhibitor
`PS-341. J.P. Thomas. A. Adjei, C. Ehrlichman, P. Geiger. A. Haas, R. Arzooma(cid:173)
`nian. D. Alberti, R. Mamocha. K. Binger. J. Volkman, C. Feierabend, K. Tutsch,
`J. Adams, P. Eliot and G. Wilding. University of Wisconsin Comprehensive
`Cancer Center, Madison, WI, Mayo Clinic, Rochester, MN and Millenium Phar(cid:173)
`maceuticals, Cambridge, MA.
`The ubiquitin-proteasome pathway is the principal enzymatic degradation path(cid:173)
`way for most intracellular proteins incli.;ding those involved in cell cycle regulatio~.
`apoptosis and angiogenesis. PS-341 is a dipeptide boronic acid compound that JS
`a potent inhibitor of the 20S/26S proteasome. Proteasome inhibition affects such
`
`diverse metabolic processes including stabilization of cell cycle regulatory proteins
`and inhibition of NF-KB activation. PS·341 has broad activity including MOR and
`Bcl-2 overcxpresslng cancer cell lines. In vivo PS-341 inhibits the growth of a
`number of tumors including the HT-29, NCl-H23 and PC-3 models. Toxicity was
`seen in preclinical models when the proteasome was inhibited by greater than
`60%. We are conducting a phase I trial of PS-341 in patients with advanced
`refractory cancers. PS-341 is administered intravenously twice weekly for 4 weeks
`followed by a two week break. Dose levels of 0.5, 0.9, 1.25 and 1.50 mg/m2 have
`been explored. A total of 9 patients have been treated at the UW. Toxicities seen
`have included rash, fatigue and thrombocytopenia. A MTO has not yet been
`reached. Proteasome inhibition by PS-341 has been monitored by measuring 20$
`proteasome activity in whole blood samples using a ftourogenic peptide substrate.
`20S proteasome inhibition in this study measured 1 hour after PS-341 administra(cid:173)
`tion correlates highly with PS-341 dose. We are achieving levels of proteasome
`inhibition (> 60%) associated with anti-tumor activity in the preclinical models. We
`have also examined patient peripheral blood mononuclear cells to determine
`whether levels of proteasome inhibition achieved in this study may be associated
`with accumulation of ubiquitinated proteins. Cell lysates were analyzed by Western
`blot for ubiquitin protein conjugates. Up to a 3 fold increase in ubiquitinated
`proteins were seen in some patients, peaking at 5 hours after PS-341 admini(cid:173)
`stration.
`
`417 Proteasome inhibition by PS-341: A phase I study. A Hamilton'. JP
`Eder, A Pavlick 1
`, JW Clark", A Chachoua 1
`, DP Ryan3, K Farrell', H Wasserstrom 1,
`L Liebes 1
`• J Wright'. P Elliott5. J Adams5 and F Muggia'. 1 NYU Sch. of Med .. 2Dana
`Farber Can. Inst., 3Mass. General Hosp., 4CTEP NCl,5Millennium Pharm.
`The proteasome is a multimeric protease complex that regulates cellular
`proteins by degrading ubiquinated proteins. Proteasome inhibition results in
`increased levels of a variety of key cellular proteins that may contribute to
`anti-tumor activity; IKB inhibits nuclear factor KB (NF-KB) mediated transcrip·
`tion, p53 inhibits apoptosis, and p21 inhibits cyclin-dependent kinase (CDK)
`activity. PS-341 is a dipeptide boronic acid derivative that inhibits the protea(cid:173)
`some by stabilization of its active site. Animal models predicted dose limiting
`gastrointestinal toxicity at 2:60% proteasome inhibition (Pl). PS-341 was ad(cid:173)
`ministered as an IV bolus on 01 &4 of a 2-week cycle. Five dose levels have
`, 0.6mg/m2 • 1mg/m2
`been studied to date: 0.25mg/m2
`, 1.2mg/m2 and 1.45mg/
`m2 • 19 pts have been treated: 11M /BF. Age: median 57, range 25-76. Primary
`tumors: colorectal (3), renal (3), NSCLC (3), melanoma (2), ST sarcoma (2),
`osteosarcoma (1), lymphoma (1), prostate (1), endometrial (1), esophagus (1),
`hepatoma (1). Prior therapies: chemotherapy (17), radiotherapy (13). To~icities
`have been mild and non-specific. 1/6 pts treated at 1.2mg/m2 expe11enced
`self-limiting G3 diarrhea. No objective respenses have been documented. One
`pt with melanoma treated at 1 mg/m2 maintained a PR in lung with SO in skin for
`6 months. Pl was measured at 1, 4 and 24hrs after dosing. At all dose levels,
`peak Pl was seen at 1 hr, and recovery to approximately 50% of peak Pl was
`seen at 24h. Peak mean Pl were 21 %, 54%, 46% and 59% at dose levels 1, 2,
`3 and 4 respectively. Tumor Pl at 24h in one pt was 67% and averaged 54% in
`2 biopsies at 2-3 h in another pt. Accrual is ongoing at 1.9mg/m2 , and phase 11
`studies are planned. Supported by U01 CA76642, M01 RR00096 and the Lynne
`Cohen Foundation (NY), and U01 62490 (Boston}.
`
`418 Pharmacodynamlc evaluation of the protein kinase C (PKC) Inhib(cid:173)
`itor CGP41251 (PKC412) in patients with metastatic melanoma. M. Mill(cid:173)
`ward'. C. House'. L Webster'. B. Linahan'. I. Olver, G. Toner'. J. Zalcberg'.
`o. Bowtell'. 'Peter MacCal/um Cancer Institute, Melbourne, 2Royal Adelaide
`Hospital, Australia.
`PKC412 selectively inhibits PKC (IC50 <1 µ.M) and has preclinical activity as a
`cytostatic and modulator of MOR. The recommended Phase II dose is 75mg tds
`which produces potentially active trough plasma levels (1 Oµ.molll), and suppresses
`cytokine release and lymphocyte ERK2 levels (Thavasu 1999). Patients (pts) with
`measurable metastatic melanoma and 2 2 superficial lesions received 75mg tds;
`tumor biopsies and plasma were collected prior to and after 28 days treatment.
`lntra-tumoral total PKC activity was measured in cytosotic and particulate fractions
`using protamine sulphate as the substrate. Initial experiments showed addition of
`10tJ.M PKC412 to melanoma biopsies inhibited phosphorylation. Ability of plasma
`to modulate ex vivo intracellular daunorubicin accumulation in MOR cells was
`measured with activity of 20µ.g/ml vaJspodar (PSC833} defining 100% reversal.
`Compared to the pretreatment biopsy, cytosoiic PKC activity was reduced by 7%
`to 91 % in 7i9 pts. Particulate PKC activity was reduced by 11 % to 79% 1n 4/9 pts.
`Only 1 pt had >50% inhibition in both fractions. Tumor PKG iS?fonn profile .in 1 pt
`resistant to PKC412 (unchanged cytosolic activity and >200% increase ~~ulate
`activity) showed an abundance of PKC~. an isoform refractory to 1nh1bit1on by
`PKC412 (IC50 >1000tJ.M}. Addition of 20µ.g/ml PKC412 to pretreatment plasma
`produced 14%-64% (mean 40%) reversal of MOR. Plasma taken following 28
`days PKC412 showed <10% reversal in 818 patients. All patients had .P"?9ressive
`disease. This Phase llA trial did not demonstrate consistent target inh1b1tton or
`phannacodynamic efficacy of PKC412 in melanoma patients.
`
`Proceedings of the 2000 NC!oEORTCoAACR Symposium
`
`West-Ward Exhibit 1082
`Hidalgo NCI 2000 Abs #413
`Page 003
`
`

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