throbber
6382s Vol. 10, 6382s– 6387s, September 15, 2004 (Suppl.)
`
`Clinical Cancer Research
`
`Mammalian Target of Rapamycin Inhibition
`
`Janice P. Dutcher
`Comprehensive Cancer Center, Our Lady of Mercy Medical Center,
`Bronx, New York
`
`threonine kinase, as a key regulator of cell cycle and of many
`intracellular functions has emerged and presented itself as a
`potential target for antitumor therapeutics.
`
`ABSTRACT
`The mammalian target of rapamycin (mTOR) is a ser-
`ine/threonine kinase that has been increasingly recognized
`as key to the regulation of cell growth and proliferation.
`mTOR either directly or indirectly regulates translation
`initiation, actin organization, tRNA synthesis, ribosome bio-
`genesis, and many other key cell maintenance functions,
`including protein degradation and transcription functions.
`Inhibition of mTOR blocks traverse of the cell cycle from
`the G1 to S phase. Preclinical data show inhibition of tumor
`growth in a number of cell lines and xenograft models.
`Clinical trials are ongoing. In metastatic renal cell cancer,
`both tumor regression and prolonged stabilization have
`been noted. mTOR inhibition appears to be a key pathway
`that may be useful in antitumor therapy. Renal cell cancer
`may be particularly susceptible through both the translation
`inhibition pathway and pathways that enhance HIF-1␣ gene
`expression, a factor believed to stimulate growth in meta-
`static renal cell cancer. Additional clinical trials that use
`agents that inhibit mTOR are ongoing.
`
`INTRODUCTION
`The identification of the mammalian target of rapamycin
`(mTOR) pathway as a potential target for anticancer therapy
`emerged from efforts to understand the activity of the immuno-
`suppressive drug rapamycin (sirolimus, Rapamune, Wyeth-Ay-
`erst Laboratories, Collegeville, PA). Rapamycin, a natural prod-
`uct derived from the soil bacteria Streptomyces hygrosopius,
`was approved for use in organ transplantation in 1999 (1– 4).
`During its preclinical evaluation, studies demonstrated potent
`antitumor activity, although initially the mechanism was un-
`known (5–7). Subsequently, Dilling et al. (8) demonstrated
`potent inhibition of growth of rhabdomyosarcoma cells by ra-
`pamycin at nanogram concentrations. Clues to the pathways
`involved were derived from this cell line, which required an
`autocrine loop involving signaling through insulin-like growth
`factor receptors (9, 10). Subsequent work by many investigators
`demonstrated that rapamycin produces cell cycle arrest, prevent-
`ing progression of dividing cells from the G1 to S phase of the
`cell cycle (11, 12). Subsequently, the role of mTOR, a serine/
`
`Presented at the First International Conference on Innovations and
`Challenges in Renal Cancer, March 19 –20, 2004, Cambridge, Massa-
`chusetts.
`Requests for reprints: Janice P. Dutcher, Our Lady of Mercy Cancer
`Center, New York Medical College, 600 East 233rd Street, Bronx, NY
`10466. Phone: 718-920-1100; Fax: 718-920-1123; E-mail: jpd4401@
`aol.com.
`©2004 American Association for Cancer Research.
`
`mTOR REGULATORY PATHWAYS
`Rapamycin does not directly inhibit mTOR but binds to its
`immunophilin, FK binding protein (FKBP12). Rapamycin plus
`FKBP12 then interact with mTOR and inhibit its function (12),
`leading to inhibition of cell growth and proliferation. The down-
`stream effects of this inhibition include inhibition of transla-
`tional pathways, with loss of phosphorylation of the eukaryotic
`translation initiation factor, 4E binding protein-1, and inhibition
`of the 40S ribosomal protein p70 S6 kinase (blocking ribosomal
`biogenesis; refs. 13–15). This effect results in a 15% to 20%
`inhibition of overall protein translation and leads to cell cycle
`arrest (12). Other cellular functions that appear to be regulated
`by mTOR and, thus, affected by its inhibition include actin
`organization, membrane traffic, protein degradation, protein ki-
`nase C signaling, and tRNA synthesis (16). There are also
`regulatory effects on synthesis of essential cell cycle proteins,
`such as cyclin D1 and c-myc (15, 17–19). Recent data suggest
`that mTOR regulates protein synthesis when cellular ATP levels
`fluctuate (20).
`In addition to the downstream activities of mTOR, which
`are affected by its inhibition, important upstream regulators of
`its activity may be altered in malignant cells. This may make
`this pathway particularly important in antitumor therapeutics.
`Both phosphatidylinositol 3⬘-kinase and Akt are upstream to
`mTOR. Akt activity is regulated by PTEN, a tumor suppressor
`gene,
`thus regulating mTOR activity. This counteracts Akt
`activation through phosphatidylinositol 3⬘-kinase. Aberrations
`in these upstream regulators, therefore, may lead to alterations
`in mTOR regulatory activity. The most striking examples of this
`are the dysregulation of phosphatidylinositol 3⬘-kinase activity
`when PTEN is mutated, deleted, or methylated (21–23). In these
`situations, this could lead to uncontrolled activity of mTOR,
`leading to uncontrolled cell proliferation.
`An additional pathway influenced by mTOR that appears
`to be particularly important in renal cell carcinoma involves the
`hypoxia-inducible factor (HIF). With loss of VHL function
`commonly seen in clear cell renal cell cancer, there is accumu-
`lation of the oxygen-sensitive transcription factors HIF-1␣ and
`HIF-2␣ (24). An increase in accumulation of these factors yields
`increased stimulation of vascular endothelial growth factor
`(VEGF), platelet-derived growth factor, and transforming
`growth factor ␣ (25). This effect is augmented by the activation
`of mTOR, which stimulates both a protein stabilization function
`and a protein translational function and, thus, increases HIF-1␣
`activity (26, 27).
`In addition, it has been determined that mutations of tuber-
`ous sclerosis complex TSC1 and -2 gene products function
`together to inhibit mTOR-mediated downstream signaling (28).
`Mutations of these genes occur in tuberous sclerosis, and their
`loss of function yields yet another pathway, which leads to
`
`Downloaded from
`
`on May 21, 2015. © 2004 American Association for Cancer
`clincancerres.aacrjournals.org
`Research.
`
`West-Ward Pharm.
`Exhibit 1012
`Page 001
`
`

`

`Clinical Cancer Research
`
`6383s
`
`increased activity of mTOR and disrupts phosphatidylinositol
`3⬘-kinase-Akt signaling through down-regulation of platelet-
`derived growth factor receptor (28 –30). Loss of TSC1 or TSC2
`gene activity induces VEGF production through mTOR (30).
`Signaling through this pathway with activation of Akt and
`mTOR results in increased HIF activity and increased VEGF.
`However, in TSC2-negative cells, platelet-derived growth factor
`receptor is markedly reduced (29) TSC2 regulates VEGF
`through both mTOR-dependent and -independent pathways
`(31). TSC2 also regulates HIF. Thus, studies evaluating the
`impact of TSC1 and TSC2 mutations demonstrate the connec-
`tion of increased VEGF and activated mTOR pathways to
`angiogenesis. Thus, inhibition of mTOR, leading to an antian-
`giogenic effect, can be explained by its impact on several
`proangiogenic pathways.
`
`PRECLINICAL EVALUATION OF mTOR
`INHIBITION
`As stated previously, early work evaluating the immuno-
`suppressive activity of rapamycin also demonstrated antitumor
`activity (5, 6), but
`this was not
`initially further evaluated.
`Subsequently, studies in rhabdomyosarcoma cells suggested an
`antitumor effect, particularly in cells that required stimulation
`by insulin-like growth factor (8). Subsequent studies in multiple
`other cell lines have shown both cytostatic and cytotoxic effects
`of rapamycin (8, 15, 22, 32– 41). Of interest, during these
`evaluations, it became apparent that rapamycin not only pro-
`duced cell cycle arrest in G1 but also produced effects that led
`to cell death. Experimental findings have demonstrated induc-
`tion of programmed cell death (apoptosis) in B-cells (42, 43)
`and rhabdomyosarcoma cells (35, 36).
`In tests performed by the National Cancer Institute (NIH,
`Bethesda, MD) human tumor cell line panel, rapamycin and its
`derivatives showed significant growth inhibition in breast can-
`cer, prostate cancer, leukemia, melanoma, renal cell cancer,
`glioblastoma, and pancreatic cancer (22). It is known that half of
`glioblastomas have PTEN mutations, which could make them
`increasingly sensitive to mTOR inhibition, and this is being
`investigated clinically (21, 22, 44).
`Studies in human tumor xenografts in mice have also
`demonstrated prolonged time to tumor growth (37). In studies in
`a pediatric brain tumor model that used CCI-779, an ester of
`rapamycin, there was significant growth inhibition and an ad-
`ditive effect when CCI-779 was administered with cisplatin
`(37). This interesting evaluation demonstrated new clinical char-
`acteristics: (1) there was not a linear-dose response effect but
`more of a threshold level effect, (2) intermittent dosing was
`effective, and (3) 2 weeks of daily dosing was superior to 1
`week, one large single dose, and dosing for ⬎2 weeks.
`Additionally, renal transplantation investigators have com-
`pared the tumor-promoting effect of the immunosuppressive
`agents used in renal transplantation in a murine model of met-
`astatic human renal cell cancer (45). Of interest, and consistent
`with these preclinical data, the number of pulmonary metastases
`was reduced when the animals were exposed to rapamycin but
`increased when exposed to cyclosporine, another immunosup-
`pressive agent used in transplantation (45). In these studies,
`
`rapamycin also reduced circulating levels of VEGF-A and trans-
`forming growth factor ␤1 (45).
`Because rapamycin has poor water solubility and stability
`in solution, it is a poor candidate for parenteral administration.
`Therefore, two ester analogues of rapamycin have been devel-
`oped with improved pharmaceutical properties and cellular ef-
`fects similar to rapamycin in the cell line screening evaluations:
`CCI-779 (Wyeth-Ayerst Research, Cambridge, MA) and RAD-
`001 (everolimus, Novartis AG, Basel, Switzerland). Both of
`these agents have entered clinical trials (46 – 48). A third mTOR
`inhibitor, ap23573 (ARIAD Pharmaceuticals, Cambridge, MA;
`ref. 49), is completing preclinical trials and is scheduled for
`clinical trials in late 2004.
`
`PHASE I CLINICAL INVESTIGATION IN
`CANCER PATIENTS
`Most of the current clinical data with agents that inhibit
`mTOR come from clinical trials of CCI-779 (Wyeth-Ayerst).
`This agent has been evaluated extensively in two Phase II trials,
`one using a weekly schedule and one using a daily for 5 days
`schedule. Raymond et al. (46) studied the weekly dosing as a
`30-minute infusion at doses ranging from 7.5 to 220 mg/m2 per
`week. There were no dose-limiting toxic effects, although grade
`3 mucositis was observed. There was a partial response in one
`patient each with renal cell cancer, breast cancer, and a neu-
`roendocrine tumor. The second Phase I trial, by Hidalgo et al.
`(47) evaluated daily 30-minute infusions given for 5 days every
`2 weeks. Dose-limiting toxic effects were grade 3 thrombocy-
`topenia, grade 3 elevations of liver function tests, and grade 3
`hypocalcemia. The maximally tolerated dose was 19 mg/m2
`daily for minimally pretreated patients and 15 mg/m2 daily for
`heavily pretreated patients. A partial response was noted in one
`patient with non–small cell lung cancer.
`Other relatively frequent toxic effects that occurred with
`some frequency included dermatologic effects such as eczema-
`toid reactions, maculopapular rash, nail bed changes, and fol-
`liculitis. The hematologic effect was thrombocytopenia. Eleva-
`tions in liver
`function tests and hypertriglyceridemia and
`hypercholesterolemia were noted. There were reversible decre-
`ments in testosterone. There was occasional mucositis. All of
`these effects were described as being mild.
`
`PHASE II CLINICAL TRIAL OF CCI-779 IN
`PATIENTS WITH METASTATIC RENAL
`CELL CANCER
`A Phase II trial has been completed and reported involving
`patients treated previously with metastatic renal cell cancer (50,
`51). This study used weekly administration of one of three dose
`levels of CCI-779: 25 mg/m2, 75 mg/m2, and 250 mg/m2. The
`dose used was blinded to the treating physician and patient.
`Dose reductions were prescribed for grade 3 toxic effects. Treat-
`ment was continued until evidence of progression or unaccept-
`able toxic effects. Patients were premedicated with diphenhy-
`dramine to preclude allergic reactions that were observed early
`in the trial. There were 111 patients enrolled and 110 received
`treatment, 36 at 25 mg/m2, 38 at 75 mg/m2, and 36 at 250
`mg/m2. Ninety percent of patients had received prior therapy for
`metastatic disease (usually a cytokine), and more than half had
`
`Downloaded from
`
`
`on May 21, 2015. © 2004 American Association for Cancerclincancerres.aacrjournals.org
`Research.
`
`West-Ward Pharm.
`Exhibit 1012
`Page 002
`
`

`

`6384s mTOR Inhibition
`
`received more than one prior regimen. Sixty-five percent of
`patients were Eastern Cooperative Oncology Group perform-
`ance status 1, and 35% were Eastern Cooperative Oncology
`Group performance status 0.
`As in the Phase I trials, the most common toxic effects
`observed were maculopapular rash (76%) and mucositis (70%).
`Additionally, with prolonged therapy, patients developed asthe-
`nia (50%) and nausea (43%). Grade 3 or 4 laboratory adverse
`events were hyperglycemia (17%), hypophosphatemia (13%),
`anemia (9%), and hypertriglyceridemia (6%). There did not
`appear to be a dose-toxicity relationship, although there were
`more dose reductions at the higher dose levels. Responses were
`observed with all of the dose levels.
`The objective response rate was 7%, with one complete and
`seven partial responses. Median time to tumor progression was
`5.8 months, and median survival was 15 months. The cumula-
`tive response rate, including complete responses, partial re-
`sponses, minor response, and stable disease for ⬎24 weeks, was
`51% (51).
`In this study, patients were evaluated for prognosis, using
`the prognostic factor criteria developed by Motzer et al., (52) in
`an analysis of ⬎300 patients treated with interferon at Memorial
`Hospital, New York, NY. The five factors that were determined
`to have significant prognostic impact were performance status,
`lactate dehydrogenase, serum calcium, hemoglobin, and time
`from initial diagnosis to treatment (52). Although initially de-
`veloped for untreated patients, the factors appear to segregate
`patients in the second-line setting as well (51). In this study of
`CCI-779, survival was decidedly different based on prognostic
`group among all of the dose-level patient groups (51).
`into the
`Eighty-seven percent of CCI-779 patients fell
`intermediate or poor prognosis categories for metastatic renal
`cell cancer, with ⬍10% in the good prognosis group. Median
`survival for these CCI-779 –treated patients in the intermediate
`or poor groups appeared to be 1.6- to 1.7-fold longer than those
`in the original study by Motzer et al. (52) of interferon-treated
`patients when compared with prognostic group by prognostic
`group (of first-line patients). Thus, mTOR inhibition may be of
`particular value in the poorer prognosis patients. This will be
`additionally tested in a randomized Phase III trial.
`Preclinical data have suggested synergy of CCI-779 admin-
`istered in combination with interferon ␣, which has led to the
`conduct of a Phase I/II clinical trial of this combination in renal
`cell cancer (53, 54). The study design began with an initial dose
`of interferon at 6 million units three times per week and of
`CCI-779 at 5 mg/m2 weekly. In the initial Phase I component of
`the study, the dose of CCI-779 was escalated, and interferon
`␣was kept at 6 million units three times per week. Doses of
`CCI-779 ranged from 5 to 25 mg/m2. A small cohort was also
`treated at 15 mg/m2 and 9 million units of interferon ␣. The
`Phase II component of the trial has extended the number of
`patients treated at 15 mg/m2 of CCI-779 and 6 million units of
`interferon ␣. As of December 2003, 71 patients have been
`entered, and several continue with treatment. The median time
`undergoing treatment for all of the cohorts is 7 months, and
`more than half have continued treatment for ⬎6 months. Re-
`sponses have been confirmed, and evaluation is continuing. The
`combination appears to show favorable activity and safety. A
`Phase III study has been initiated in which the combination of
`
`CCI-779 and interferon ␣ is compared with either agent alone as
`first-line therapy in patients with a poor prognosis for metastatic
`renal cell cancer. An additional randomized study is planned for
`patients who have had prior therapy.
`
`OTHER AGENTS
`The other agents that inhibit mTOR are undergoing early
`phases of evaluation. RAD001 is an oral formulation and is
`currently in Phase I clinical trials (48). The agent ap23573 is
`currently in preclinical evaluation, with human clinical trials
`planned (49).
`
`DISCUSSION
`Preclinical data demonstrate the importance of mTOR as a
`regulator of cell growth and proliferation. Additional evaluation
`of malignant cells demonstrates that the constitutive activity of
`mTOR leads to unregulated growth. Human malignancies have
`been demonstrated to have a constitutive activation of mTOR or
`its upstream regulators, leading to enhanced mTOR activity.
`Now clinical data are beginning to demonstrate that this path-
`way is a viable target for antitumor therapeutics, and the out-
`come demonstrated in the Phase II trial of CCI-779 in metastatic
`renal cell cancer is promising. The ability to evaluate and
`interpret prolonged stable disease in metastatic renal cell cancer
`trials continued to be problematic but appears to be a real
`outcome and will need to be carefully quantitated in future
`randomized trials with many of the agents currently undergoing
`evaluation, including the inhibitors of mTOR.
`As noted in the early in vitro work by Dilling et al. (8), the
`rhabdomyosarcoma cell line that they studied is regulated by an
`autocrine loop involving secretion of type 2 insulin-like growth
`factor, and signaling is through the insulin-like growth factor
`receptor. In previous studies (8, 12), the cells most sensitive to
`rapamycin were dependent on this autocrine pathway. Others
`have reported the interaction of rapamycin with control of
`glucose and lipids and the role of mTOR in the insulin-signaling
`pathway (55, 56). In view of the incidence of hyperglycemia and
`hypertriglyceridemia observed during treatment with CCI-779,
`it is possible that these biochemical parameters may correlate
`with the degree of mTOR inhibition (51). This will be evaluated
`by additionally investigating the clinical outcome in the current
`trials with CCI-779 and the levels of glucose and lipids. This
`will need additional confirmation by observations during trials
`of the other mTOR inhibitors, as to whether these clinical effects
`indeed do reflect a pharmacodynamic surrogate for mTOR
`inhibition.
`The inhibition of mTOR appears to be a promising targeted
`approach to antitumor therapy. Additional evaluation of these
`agents for schedule, route of administration, and combination
`therapy approaches is clearly warranted. Renal cell cancer ap-
`pears to be sensitive to this therapeutic approach, and it will be
`of great interest to determine whether combinations of mTOR
`inhibitors with other agents will enhance the therapeutic effi-
`cacy in this difficult disease. It will also be important to evaluate
`the biochemical parameters that might predict favorable outcome.
`
`clincancerres.aacrjournals.org Downloaded from
`
`on May 21, 2015. © 2004 American Association for Cancer
`
`Research.
`
`West-Ward Pharm.
`Exhibit 1012
`Page 003
`
`

`

`Clinical Cancer Research
`
`6385s
`
`I think these are the right
`
`OPEN DISCUSSION
`Dr. Robert J. Motzer: Can you comment on the lung
`toxicity or the pulmonary infiltrates that are associated with the
`drug?
`Dr. Janice P. Dutcher: There have been some asymp-
`tomatic patients, but we did not see them at all in our group.
`Dr. Michael B. Atkins:
`In the paper that was published
`in JCO, there were 6 patients out of approximately 100 who had
`pulmonary infiltrates (J Clin Oncol 2004;22:909 –18). Some of
`these individuals had associated symptoms. All had their treat-
`ment held, 2 were not restarted probably because their disease
`progressed, 4 were restarted when their symptoms got better.
`Two did not have a recurrence of the problem, and 2 had a
`recurrence of the pulmonary infiltrates. Five of 6 happened at
`the 75-mg dose level, 1 at the 250-mg level, and none at the
`25-mg dose level. It is hard to say whether any of these side
`effects were dose dependent.
`Dr. Robert A. Figlin: My understanding of signal trans-
`duction inhibition with CCI-779 is that administered once a
`week it will not inhibit the target for an extended period. Do we
`have any readout from laboratory biology that can help us
`understand whether once a week, which is very convenient for
`the patient but might not be good at impacting the tumor, is a
`better strategy? Is that born out by laboratory models? How do
`we need to be targeting these pathways via pharmacological
`interventions?
`Dr. William G. Kaelin, Jr.:
`questions.
`Dr. Atkins: There were a lot of patients in this study who
`had minor responses. I think it was 26% of the patients who had
`more than 25% tumor regression without satisfying the criteria
`for partial response. A lot of the patients who were benefiting
`were patients who we thought would never have benefited from
`immunotherapy. There were people with hypercalcemia, fa-
`tigue, a performance status of 2, and multiple prior treatments.
`We were seeing the disease course change in those patients, so
`it suggested to us that maybe there was a different patient
`population that was responding to CCI-779 than would typically
`respond to immunotherapy. What would be the appropriate
`targets to measure in an mTOR inhibition trial and when would
`be the appropriate time to measure them?
`Dr. Figlin: That’s in fact what our trial will be looking at
`in patients who are undergoing cytoreductive nephrectomy with
`metastatic disease. We will be looking at inhibition of all of the
`targets in the mTOR pathway.
`Dr. Walter M. Stadler: There is a lot of publicity about
`these drugs being more active in the PTEN-deficient tumors.
`Has any of that been born out clinically?
`Dr. Figlin: The mTOR pathway appears to have impor-
`tance not just inside the tumor cell but also in terms of the
`angiogenesis. There may be some synergy in terms of that
`pathway being important in multiple places.
`Dr. Atkins: What about the connection between mTOR
`inhibition and HIF? Is that a direct effect or is that a more
`general effect on protein synthesis or on molecules that are
`turning over quicker?
`Dr. Kaelin: That is still being worked out, and I certainly
`don’t want to oversell the HIF connection. There clearly is a HIF
`
`connection, but mTOR is a fairly important enzyme that does
`other things. An emerging theme that is coming out of at least
`preclinical studies with various targets and cancer is this notion
`that cancer cells, after mutating a particular pathway, become
`addicted to that pathway and hypersensitive to inhibitors of that
`pathway. This happens to be one of those scenarios that have
`been described for some other targets as well.
`Dr. Michael S. Gordon: Those have been the settings
`where the greatest results, positive results, have been shown.
`Dr. Kaelin: Preclinically, in retrospect, there was evi-
`dence that leukemic cells become addicted to BCR-ABL sig-
`naling and become hypersensitive to an inhibitor. In some of
`these models, for example, after you introduce BCR-ABL into
`cells, they become hypersensitive and they will undergo apo-
`ptosis after acute interruption of that signal, which might favor
`intermittent dosing to allow normal cells to recover. If I am not
`mistaken, rapamycin is primarily antiproliferative, meaning cy-
`tostatic. There is an influence of PTEN status on responsiveness,
`but still what you see is a cytostatic effect. In that scenario, you
`could make the case for a continuous exposure.
`Dr. Daniel J. George:
`It sounds like there is still a huge
`dose range that people are struggling with. Obviously, there is
`toxicity, but does that toxicity vary? Should we be using a dose
`lower than 25 mg?
`Dr. Dutcher: The sense was that the toxicity was clearly
`manageable at all dose levels, and the dose that is in the Phase
`III trial is the 25-mg dose. There are some that would argue that
`you should stay at the 250 mg, because it would allow dose
`reductions and there was a slight difference in survival.
`Dr. Atkins: When we looked at all of the efficacy pa-
`rameters of response, time to progression, and survival, there
`was no clearcut difference between doses, indicating that even
`the 25-mg dose was probably high enough to hit the target.
`Although the survival was longer in the 250-mg patients, the
`time to progression was less, so there may have been patient
`selection that accounted for their prolonged survival. It is inter-
`esting that in the more recent studies with CCI-779 and inter-
`feron, we have seen several patients who have had significant
`hypertension as well. It was not reported in the single-agent
`Phase II study, because it probably did not get to exceed the
`10% to 15% range. Now we are seeing patients who have had
`hypertension because they have been on treatment for a while,
`which may be an indication that CCI-779 is hitting VEGF or
`HIF.
`
`Dr. Gordon:
`Is the rash an EGFR-type rash? Is it the
`same as we would see with gefitinib?
`Dr. Dutcher:
`It is not as severe, but it is similar. It
`usually did not last through the whole treatment.
`Dr. Atkins: A lot of patients got steroids to control the
`rash, which could be a potential problem. It is sort of strange to
`think about combining this with interferon even though there
`was synergy, but it is clearly an immunosuppressive agent if you
`give it frequently enough.
`Dr. Dutcher: Some of the people got antibiotics for the
`nail bed problems.
`Dr. Gordon: But that, again, is something that we have
`been seeing with the targeted therapies.
`
`Downloaded from
`
`
`on May 21, 2015. © 2004 American Association for Cancerclincancerres.aacrjournals.org
`Research.
`
`West-Ward Pharm.
`Exhibit 1012
`Page 004
`
`

`

`6386s mTOR Inhibition
`
`REFERENCES
`1. Vezina C, Kudelski A, Sehgal SN. Rapamycin (AY-22,989), a new
`antifungal antibiotic, I: taxonomy of the producing stretomycete and
`isolation of the active principle. J Antibiot 1975;28:721– 6.
`2. Sehgal SN, Baker H, Vezina C. Rapamycin (AY-22,989), a new
`antifungal antibiotic, II: fermentation, isolation and characterization. J
`Antibiot 1975;28:727–32.
`3. Caine RY, Collier DS, Lim S, Pollard SG, Samaan A, White DJ,
`Thiru S. Rapamycin for immunosuppression in organ allografting. Lan-
`cet 1989;2:227.
`4. Schreiber SL. Chemistry and biology of the immunophilins and their
`immunosuppressive ligands. Science 1991;251:283–7.
`5. Douros J, Suffness M. New antitumor substances of natural origin.
`Cancer Treat Rev 1981;8:63– 87.
`6. Houchens DP, Ovejera AA, Riblet SM, Slagel DE. Human brain
`tumor xenografts in nude mice as a chemotherapy model. Eur J Cancer
`Clin Oncol 1983;19:799 – 805.
`7. Eng CP, Sehgal SN, Vezina C. Activity of rapamycin (AY-22,989)
`against transplanted tumors. J Antibiot 1984;37:1231–7.
`8. Dilling MB, Dias P, Shapiro DN, Germain GS, Johnson RK, Hough-
`ton PJ. Rapamycin selectively inhibits the growth of childhood rhab-
`domyosarcoma cells through inhibition of signaling via the type I
`insulin-like growth factor receptor. Cancer Res 1994;54:903–7.
`9. El-Badry OM, Minniti C, Kohn EC, Houghton PJ, Daughaday WH,
`Helman LJ. Insulin-like growth factor II acts as an autocrine growth and
`motility factor in human rhabdomyosarcoma tumors. Cell Growth Differ
`1990;1:325–31.
`10. Shapiro DN, Jones BG, Shapiro LH, Dias P, Houghton PJ. Anti-
`sense-mediated reduction in insulin-like growth factor-I receptor expres-
`sion suppresses the malignant phenotype of a human alveolar rhab-
`domyosarcoma. J Clin Investig 1994;94:1235– 42.
`11. Wiederrecht GJ, Sabers CJ, Brunn GJ, Martin MM, Dumont FJ,
`Abraham RT. Mechanism of action of rapamycin: new insights into the
`regulation of G1-phase progression in eukaryotic cells. Prog Cell Cycle
`Res 1995;1:53–71.
`12. Huang S, Houghton PJ. Inhibitors of mammalian target of rapamy-
`cin as novel antitumor agents: from bench to clinic. Curr Opinion Invest
`Drugs 2002;3:295–304.
`13. Hidalgo M, Rowinsky EK. The rapamycin-sensitive signal trans-
`duction pathway as a target for cancer therapy. Oncogene 2000;19:
`6680 – 6.
`14. Dudkin L, Dilling MB, Cheshire PJ, Harwood FC, Hollingshead M,
`Arbuck SG, Travis R, Sausville EA, Houghton PJ. Biochemical corre-
`lates of mTOR inhibition by the rapamycin ester CCI-779 and tumor
`growth inhibition. Clin Cancer Res 2001;7:1758 – 64.
`15. Yu K, Toral-Barza L, Discafani C, Zhang, W.-G., Skotnicki J, Frost
`P, Gibbons JJ. mTOR, a novel target in breast cancer: the effect of
`CCI-779, an mTOR inhibitor, in preclinical models of breast cancer.
`Endocr Relat Cancer 2001;8:249 –58.
`16. Schmelzle T, Hall MN. TOR, a central controller of cell growth.
`Cell 2000;103:253– 62.
`17. Rosenwald IB, Kaspar R, Rousseau D, et al. Eukaryotic translation
`initiation factor 4E regulates expression of cyclin D1 at transcriptional
`and post-transcriptional levels. J Biol Chem 1995;270:21176 – 80.
`18. Shantz LM, Pegg AE. Overproduction of ornithine decarboxylase
`caused by relief of translational repression is associated with neoplastic
`transformation. Cancer Res 1994;54:2313– 6.
`19. DeBenedetti A, Joshi B, Graff JR, Zimmer SG. CHO cells trans-
`formed by the translation factor elF4E display increased c-Myc expres-
`sion but require overexpression of Max for tumorigenicity. Mol Cell
`Differ 1994;2:347–71.
`20. Dennis PB, Jaeschke A, Saito M, Fowler B, Kozma SC, Thomas G.
`Mammalian TOR: a homeostatic ATP sensor. Science 2001;294:
`1102–5.
`21. Shi Y, Gera J, Hu L, Hsu J-H, Bookstein R, Li W, Lichtenstein A.
`Enhanced sensitivity of multiple myeloma cells containing PTEN mu-
`tations to CCI-779. Cancer Res 2002;62:5027–34.
`
`22. Gibbons JJ, Discafani C, Peterson R, Hernandez R, Skotnicki J,
`Frost P. The effect of CCI-779, a novel macrolide antitumor agent, on
`the growth of human tumor cells in vitro and in nude mouse xenograft
`in vivo. Proc Am Assoc Cancer Res 2000;40:301.
`23. Xu Q, Simpson S-E, Scialla TJ, Bagg A, Carroll M. Survival of
`acute myeloid leukemia cells requires PI3 kinase activation. Blood
`2003;102:972– 80.
`24. Ohh M, Park CW, Ivan M, et al. Ubiquitination of hypoxia-induc-
`ible factor requires direct binding to the beta-domain of the von Hippel-
`Lindau protein. Nat Cell Biol 2000;2:423–7.
`25. Iliopoulos O, Levy AP, Jiang C, Kaelin WG, Goldberg MA. Neg-
`ative regulation of hypoxia-inducible genes by the von Hippel-Lindau
`protein. Proc Natl Acad Sci USA 1996;93:10595–9.
`26. Hudson CC, Liu M, Chiang GG, et al. Regulation of hypoxia-
`inducible factor 1-alpha expression and function by the mammalian
`target of rapamycin. Mol Cell Biol 2002;22:7004 –14.
`27. Hopfl G, Wenger RH, Ziegler U, et al. Rescue of hypoxia-inducible
`factor-1␣-deficient tumor growth by wild-type cells is independent of
`vascular endothelial growth factor. Cancer Res 2002;62:2962–70.
`28. Tee AR, Fingar DC, Manning BD, Kwiatkowski DJ, Cantley LC,
`Blenis J. Tuberous sclerosis complex-1 and –2 gene products function
`together to inhibit mammalian target of rapamycin (mTOR)-mediated
`downstream signaling. Proc Natl Acad Sci USA 2002;99:13571– 6.
`29. Zhang H, Cicchetti G, Onda H, et al. Loss of Tsc1/Tsc2 activates
`mTOR and disrupts PI3K-Akt signaling through downregulation of
`PDGFR. J Clin Invest 2003;112:1223–33.
`30. El-Hashemite N, Walker V, Zhang H, Kwiatkowski DJ. Loss of
`Tsc1 or Tsc2 induces vascular endothelial growth factor production
`through mammalian target of rapamycin Cancer Res 2003;63:5173–7.
`31. Brugarolas JB, Vazquez F, Reddy A, Sellers WR, Kaelin WG. Jr.
`TSC2 regulates VEGF through mTOR-dependent and –independent
`pathways. Cancer Cell 2003;4:147–58.
`32. Shi Y, Frankel A, Radvanyi LG, Penn LZ, Miller RG, Mills GB.
`Rapamycin enhances apoptosis and increases sensitivity to cisplatin in
`vitro. Cancer Res 1995;55:1982– 8.
`33. Seufferlein TI, Rozengurt E. Rapamycin inhibits constitutive p70s6k
`phosphorylation, cell proliferation and colony formatin in small cell
`lung cancer cells. Cancer Res 1996;56:3895–7.
`34. Hosoi H, Dilling MB, Liu LN, et al. Studies on the mechanism of
`resistance to rapamycin in human cancer cells Mol Pharmacol 1998;54:
`815–24.
`35. Hosoi H, Dilling MB, Shikata T, et al. Rapamycin causes poorly
`reversible inhibition of mTOR and induces p53-independent apoptosis
`in human rhabdomyosarcoma cells. Cancer Res 1999;59:886 –94.
`36. Huang S, Liu LN, Hosoi H, Dilling MB, Shikata T, Houghton PJ.
`p53/p21CIP1 cooperate in enforcing rapamycin-induced G1 arrest

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