throbber
Cancer Therapy: Clinical
`
`A Phase I and Pharmacokinetic Study of Temsirolimus (CCI-779)
`Administered Intravenously Daily for 5 Days Every 2 Weeks
`to Patients with Advanced Cancer
`Manuel Hidalgo,1,2 Jan C. Buckner,3 Charles Erlichman,3 Marilyn S. Pollack,2 Joseph P. Boni,4
`Gary Dukart,4 Bonnie Marshall,4 Lisa Speicher,4 Laurence Moore,5 and Eric K. Rowinsky1,2
`
`Abstract Purpose: Patients with advanced cancer received temsirolimus (Torisel, CCI-779), a novel inhib-
`itor of mammalian target of rapamycin, i.v. once daily for 5 days every 2 weeks to determine the
`maximum tolerated dose, toxicity profile, pharmacokinetics, and preliminary antitumor efficacy.
`Experimental Design: Doses were escalated in successive cohorts of patients using a conven-
`tional phase I clinical trial design. Samples of whole blood and plasma were collected to determine
`the pharmacokinetics of temsirolimus and sirolimus, its principal metabolite.
`Results: Sixty-three patients were treated with temsirolimus (0.75-24 mg/m2/d). The most
`common drug-related toxicities were asthenia, mucositis, nausea, and cutaneous toxicity. The
`maximum tolerated dose was 15 mg/m2/d for patients with extensive prior treatment because,
`in the 19 mg/m2/d cohort, two patients had dose-limiting toxicities (one with grade 3 vomiting,
`diarrhea, and asthenia and one with elevated transaminases) and three patients required dose
`reductions. For minimally pretreated patients, in the 24 mg/m2/d cohort, one patient developed
`a dose-limiting toxicity of grade 3 stomatitis and two patients required dose reductions, establish-
`ing 19 mg/m2/d as the maximum acceptable dose. Immunologic studies did not show any
`consistent trend toward immunosuppression.Temsirolimus exposure increased with dose in a less
`than proportional manner. Terminal half-life was 13 to 25 hours. Sirolimus-to-temsirolimus expo-
`sure ratios were 0.6 to 1.8. A patient with non ^ small cell lung cancer achieved a confirmed partial
`response, which lasted for 12.7 months. Three patients had unconfirmed partial responses; two
`patients had stable disease for z24 weeks.
`Conclusion:Temsirolimus was generally well tolerated on this intermittent schedule. Encouraging
`preliminary antitumor activity was observed.
`
`The mammalian target of rapamycin (mTOR) is a serine/
`threonine kinase and a member of the phosphatidylinositol
`family of kinases, which is involved in the response of
`eukaryotic cells to proliferative and nutritional stimuli (1 – 4).
`mTOR is downstream of Akt
`in the phosphatidylinositol
`3-kinase (PI3K)/Akt signaling pathway and regulates the
`ribosomal protein S6 kinase (p70 S6 kinase) and eukaryotic
`translation initiation factor 4E-binding protein-1. Activation
`of these proteins increases the translation of mRNAs with a
`
`Authors’ Affiliations: 1Institute for Drug Development, Cancer Therapy and
`Research Center, Brook Army Medical Center and 2The University of Texas Health
`Science Center, San Antonio, Texas; 3Mayo Clinic, Rochester, Minnesota; 4Wyeth
`Research, Collegeville, Pennsylvania; and 5Wyeth Research, Cambridge,
`Massachusetts
`Received 1/18/06; revised 5/17/06; accepted 6/15/06.
`Grant support: Wyeth Research (Collegeville, PA).
`The costs of publication of this article were defrayed in part by the payment of page
`charges. This article must therefore be hereby marked advertisement in accordance
`with 18 U.S.C. Section 1734 solely to indicate this fact.
`Requests for reprints: Manuel Hidalgo, The Sidney Kimmel Comprehensive
`Cancer Center at Johns Hopkins, 1650 Orleans Street, Room 1M89, Baltimore, MD
`21231-1000. Phone: 410-502-9746; Fax: 410-614-9006; E-mail: mhidalg1@
`jhmi.edu.
`F 2006 American Association for Cancer Research.
`doi:10.1158/1078-0432.CCR-06-0118
`
`5¶-terminal oligopyrimidine tract or 5¶-cap structure, which
`encode for proteins involved in G1-S cell cycle regulation (5, 6).
`The PI3K/Akt pathway is activated in cancer by growth factor
`and/or hormone receptor activation or by mutations in genes,
`such as PI3K or PTEN, or byAkt amplification (7 – 21).
`its
`The discovery of mTOR and the understanding of
`biological functions have been greatly facilitated by studies
`with sirolimus (rapamycin), a naturally occurring macrolide
`that inhibits mTOR (2, 22). Sirolimus binds to the intracellular
`immunophilin FKBP12 and this complex inhibits mTOR,
`which results in inhibition of p70 S6 kinase and 4E-binding
`protein-1 functions, followed by a decrease in cyclin D1 levels,
`increase in p27 levels, and cell cycle arrest (23). In certain
`preclinical models, sirolimus induces apoptosis (24). Siro-
`limus also has antiangiogenesis effects by decreasing hypoxia-
`inducible factor-1a – induced secretion of vascular endothelial
`growth factor (25). Recently, sirolimus has been shown to
`inhibit the transforming capabilities of PI3K mutants (26),
`which supports the notion that mTOR inhibitors may be useful
`for the treatment of tumors with these mutations.
`Temsirolimus (Torisel, CCI-779) is an ester of sirolimus
`(Fig. 1) selected for clinical development based on a favorable
`pharmacologic and toxicity profile. Temsirolimus inhibited the
`growth of a variety of tumor cells and was particularly effective
`in tumors with a defective PTEN gene (27 – 33). Temsirolimus
`
`www.aacrjournals.org
`Downloaded from
`
`clincancerres.aacrjournals.org
`
`5755
`Clin Cancer Res 2006;12(19) October 1, 2006
`on August 24, 2015. © 2006 American Association for Cancer
`Research.
`
`NOVARTIS EXHIBIT 2056
`Par v. Novartis, IPR 2016-01479
`Page 1 of 9
`
`

`

`Cancer Therapy: Clinical
`
`Fig. 1. Structure of temsirolimus and sirolimus, its principal metabolite.
`
`also was effective in reversing resistance to conventional
`chemotherapy and hormone therapy conferred by PTEN defects
`(30, 34).
`Because of the immunosuppressive effects of sirolimus, an
`expected metabolite of temsirolimus, temsirolimus was evalu-
`ated for inhibition of T lymphocyte function in euthymic mice
`(CCI-779 Investigator’s Brochure). Although i.v. temsirolimus
`inhibited T lymphocyte activity, its effects were reversible and
`T lymphocyte activity returned to normal within 24 hours
`after drug treatment was stopped. Multiple cycles of temsir-
`olimus treatment did not result in cumulative deterioration of
`T lymphocyte function. Further studies in mice indicated that
`antitumor activity could be achieved with different intermittent
`dosing regimens, including a daily 5-day regimen given every
`2 weeks. Accordingly, this intermittent schedule was used in a
`phase I study to minimize the immunosuppressive effects of
`temsirolimus while maintaining antitumor activity.
`Based on the data summarized above, temsirolimus was
`selected for clinical development. Three phase I single-agent
`studies have been conducted with this drug based on different
`administration regimens, including i.v. weekly (35), i.v. once
`daily for 5 days every 2 weeks (this study), and oral once daily
`for 5 days every 2 weeks (36). In this study, patients with
`advanced cancer were treated with temsirolimus to evaluate
`safety, determine the maximum tolerated dose (MTD), charac-
`terize pharmacokinetics, and seek preliminary evidence of
`antitumor activity.
`
`Materials and Methods
`
`In this phase I, dose escalation study, temsirolimus
`Trial design.
`was administered as a 30-minute i.v. infusion once daily on days 1 to
`5 of each treatment cycle of f2 weeks. Patients were observed at least
`9 days after their day 5 dose of temsirolimus before receiving the next
`cycle of drug. Patients could remain on study as long as temsirolimus
`was well tolerated and there was no evidence of disease progression.
`The primary objectives of the study were to determine the safety and
`tolerability and to identify the MTD of temsirolimus given i.v. once
`daily for 5 days every 2 weeks in patients with advanced solid tumors.
`The secondary objectives were to determine the pharmacokinetics of
`temsirolimus on this schedule and to obtain preliminary information
`on antitumor activity.
`Patient selection. Patients with histologically confirmed advanced
`cancer (solid tumors or lymphomas) who failed to respond to standard
`therapy or for whom standard therapy was not available were eligible
`for this study. Eligibility criteria also included age z18 years; an Eastern
`Cooperative Oncology Group performance status V2 (ambulatory and
`capable of self-care); life expectancy z12 weeks; no prior chemother-
`apy, radiation therapy, or immunosuppressive therapy (except cortico-
`
`steroids for management of emesis or peritumoral edema) within
`3 weeks of starting study treatment; no treatment with investigational
`agents within 30 days before commencing study treatment; adequate
`hematopoietic (hemoglobin level z9 g/dL, absolute neutrophil
`count z1,500/AL, platelet count z100,000/AL), hepatic [bilirubin
`<1.5 mg/dL, aspartate and alanine aminotransaminases <3 times
`institutional normal upper limit (<5 times institutional normal
`upper limit for patients with liver metastases)], and renal (creatinine
`<2 mg/dL) functions; measurable or evaluable disease; and no active
`infections or history of hypersensitivity to macrolide antibiotics,
`unstable angina, or myocardial infarction within 6 months or coexist-
`ing medical problems of sufficient severity to limit compliance in the
`study. Due to the known toxicities of sirolimus, patients who entered
`the trial were also required to have serum levels of cholesterol
`and triglycerides V350 and V300 mg/dL, respectively. Patients with
`clinically and radiologically stable brain tumors were eligible. Patients
`receiving hepatic enzyme-inducing anticonvulsants or antiarrhythmic
`agents were ineligible. Before treatment, patients were required to give
`written informed consent according to federal and institutional guide-
`lines.
`Because patients who have received extensive anticancer therapy tend
`to have greater drug-related toxicity than those who have received less
`extensive therapy, patients treated with higher dose levels of temsir-
`olimus were classified as being minimally pretreated or heavily
`pretreated. Heavily pretreated patients were defined as having received
`radiotherapy to z25% of bone marrow – producing areas, more than six
`cycles of an alkylating agent (except low-dose cisplatin), more than four
`courses of a carboplatin-containing regimen, or more than two courses
`of carmustine or mitomycin C (37).
`Drug dosage and administration. The starting dose of temsirolimus
`was 0.75 mg/m2 based on animal toxicology studies and prior clinical
`experience with sirolimus. A modified version of the Continual Reas-
`sessment Method (38, 39) was to be used to guide dose escalation.
`
`Table 1. Patient characteristics
`
`Characteristics
`
`n
`
`63
`60
`39/24
`
`Patients
`Fully assessable patients
`Sex (men/women)
`Age (y)
`56
`Median
`19-79
`Range
`Eastern Cooperative Oncology Group performance status, patients
`0
`20
`1
`30
`2
`13
`Prior therapy, patients
`Chemotherapy alone
`Radiotherapy alone
`Chemotherapy and radiotherapy
`Tumor type, patients
`Renal
`Colorectal
`Non – small cell lung cancer
`Soft-tissue sarcoma
`Endometrial
`Ovarian
`Sarcoma
`Other*
`
`58
`2
`28
`
`16
`10
`9
`7
`3
`2
`2
`14
`
`*One each of anaplastic astrocytoma, cervical, esophageal,
`gastric, head and neck-adenoid cystic carcinoma, hepatocellular,
`non-Hodgkin’s lymphoma, nasopharyngeal, osteosarcoma, pan-
`creatic, prostate, squamous cell carcinoma of the skin, thyroid,
`and unknown.
`
`Clin Cancer Res 2006;12(19) October 1, 2006
`clincancerres.aacrjournals.org Downloaded from
`
`
`5756
`www.aacrjournals.org
`on August 24, 2015. © 2006 American Association for Cancer
`Research.
`
`NOVARTIS EXHIBIT 2056
`Par v. Novartis, IPR 2016-01479
`Page 2 of 9
`
`

`

`Phase I Study of Temsirolimus
`
`Table 2. Dose escalation and toxicity experience
`
`Temsirolimus
`dose (mg/m2/d  5)
`
`No. patients
`entered (inevaluable*)
`
`DLT (cycle 1)
`
`No. patients
`
`Toxicity and grade
`
`No. patients
`reduced to dosec
`
`Total at dose
`
`No. evaluable
`patients
`
`No.
`cycles
`
`0.75
`1.25
`1.5
`1.8
`2.16
`2.6
`3.12
`3.74
`4.5
`5.4
`6.5
`7.8
`9.4
`11.3
`Minimally pretreated
`15
`19
`24
`Heavily pretreated
`15
`19
`
`3
`4 (1)
`1
`1
`6
`1
`2
`2
`4
`2
`2
`3
`1
`4 (1)
`
`3
`6
`6
`
`6 (1)
`6
`
`Total
`
`63 (3)
`
`0
`0
`0
`0
`1
`0
`0
`0
`0
`0
`0
`0
`0
`0
`
`1
`0
`1
`
`0
`2
`
`4
`
`Grade 3 hypocalcemia
`
`Grade 3 hyperglycemia
`
`Grade 3 stomatitis
`
`Grade 3 aspartate and alanine
`aminotransaminase elevations
`Grade 3 vomiting, diarrhea,
`and asthenia
`
`0
`0
`0
`1
`0
`0
`0
`0
`0
`0
`0
`0
`0
`1
`
`2
`3
`0
`
`5b
`0
`
`3
`3
`1
`2
`6
`1
`2
`2
`4
`2
`2
`3
`1
`4
`
`5
`9
`6
`
`10
`6
`
`10
`7
`2
`15
`32
`4
`12
`24
`50
`5
`10
`29
`3
`17
`
`33
`45
`24
`
`31
`7
`
`361
`
`*Reasons inevaluable for determining dose escalation: two disease progression (1.25 and 15 mg/m2/d) during cycle 1 and one hypersensitivity
`reaction (11.3 mg/m2/d) during the first 24 hours after the first temsirolimus dose.
`cIncludes all patients reduced from the next higher dose level at any subsequent cycle.
`bOne patient required a second dose reduction to 11.3 mg/m2/d.
`
`However, because of adverse events observed at the first two dose
`levels and after discussions with the U.S. Food and Drug Administration,
`the protocol was amended and a fixed 20% dose escalation was used.
`A later amendment allowed fixed dose escalation increments of up
`to 30%.
`The National Cancer Institute Common Toxicity Criteria version 2.0
`was used to grade toxicity. Unacceptable toxicities included temsir-
`
`olimus-related (a) grade 3/4 nonhematologic toxicity (excluding
`nausea or vomiting in patients on suboptimal antiemetic prophylaxis
`or serum triglycerides <1,500 mg/dL if recovery occurred by the next
`cycle), (b) grade 4 thrombocytopenia, or (c) grade 4 neutropenia
`lasting >5 days. If a patient had an unacceptable toxicity, dose reduction
`by one to two levels and/or a delay in treatment could occur. If a grade
`3 toxicity was observed in a patient at a given dose level, the cohort was
`
`Fig. 2. Frequently occurring toxicities of
`temsirolimus included, for all 63 patients,
`asthenia (35 patients, 56%), mucositis
`(34 patients, 54%), nausea (26 patients,
`41%), cutaneous toxicity (26 patients, 41%),
`hypertriglyceridemia (23 patients, 37%),
`thrombocytopenia (21patients, 33%),
`hypercholesterolemia (14 patients, 22%),
`elevated transaminases (12 patients, 19%),
`and hyperglycemia (11patients, 17%).
`Temsirolimus doses: 0.75 to 11.3, 15, 19, and
`24 mg/m2/d. MP, minimally pretreated; HP,
`heavily pretreated.
`
`www.aacrjournals.org
`Downloaded from
`
`5757
`Clin Cancer Res 2006;12(19) October 1, 2006
`
`Research.
`
`on August 24, 2015. © 2006 American Association for Cancerclincancerres.aacrjournals.org
`
`NOVARTIS EXHIBIT 2056
`Par v. Novartis, IPR 2016-01479
`Page 3 of 9
`
`

`

`Cancer Therapy: Clinical
`
`Table 3. Pharmacokinetic variables of temsirolimus on day 5, mean F SD (no. patients)
`
`Dose group (mg/m2)
`
`0.75
`1.25
`2.16
`4.5
`15
`19
`
`C max (ng/mL)
`72 F 16 (3)
`133 F 64 (3)
`186 F 51 (5)
`331 F 72 (4)
`503 F 293 (5)
`796 F 226 (12)
`
`t max (h)
`0.67 F 0.29 (3)
`0.46 F 0.28 (30)
`0.59 F 0.24 (5)
`0.46 F 0.11 (4)
`0.26 F 0.18 (5)
`0.41 F 0.13 (12)
`
`t 1/2 (h)
`24.8 F 7.5 (2)
`12.6 F 5.1 (3)
`16.4 F 6.9 (5)
`13.9 F 2.6 (4)
`20.0 F 22.0 (5)
`15.4 F 15.6 (12)
`
`AUC (cycle 1; ng h/mL)
`
`1,355 F 732 (2)
`2,502 F 1,531 (3)
`3,896 F 986 (5)
`5,350 F 792 (4)
`8,619 F 2,188 (5)
`9,838 F 3,504 (12)
`
`Abbreviations: Cmax, peak observed concentration; t max, time to C max; t1/2, terminal half-life; AUC, area under the concentration versus time
`curve; CLc, central clearance; Vdss, steady-state volume of distribution; AR, accumulation ratio of day 5 to day 1; B/Pratio, blood-to-plasma
`concentration ratio.
`
`expanded to three patients. If an unacceptable toxicity was observed
`in a patient at a given dose level in cycle 1, a dose-limiting toxicity
`(DLT) occurred and that cohort was expanded to six patients. The MTD
`was defined as the highest dose for which two or fewer patients had a
`DLT. However, the combination of DLTs and dose reductions that
`occurred at a given dose level were taken into account in identifying
`the MTD.
`Temsirolimus (25 mg/mL in 100% ethanol, Wyeth Research,
`Collegeville, PA) is a light-sensitive drug and was protected from
`sunlight and unshielded fluorescent
`light during preparation and
`administration. The drug-ethanolic concentrate was diluted 10-fold in a
`polyethylene glycol/polysorbate diluent and then further diluted with
`0.9% saline solution to a total volume of 50 to 100 mL, which was
`administered for f30 minutes using glass or polyolefin infusion kits
`and an automatic dispensing pump.
`Evaluation of patients. Physical examination and routine laboratory
`evaluations were done before treatment and weekly.
`For assessment of
`immunologic activity, whole blood samples
`were collected before treatment, on days 1 and 5 of cycles 1 to 3, and
`on day 8 of cycle 1. Three assays were done. (a) WBC counts and
`differentials were monitored to check for changes in lymphocyte
`numbers. (b) Proliferative responses (uptake of tritiated thymidine) of
`patient’s lymphocytes to pokeweed mitogen, phytohemagglutinin, and
`concanavalin A and to pooled allogeneic cells were monitored as
`standard indicators of altered lymphocyte function (40). (c) Lympho-
`cyte subsets (cell surface phenotypes CD4/CD3, CD8/CD3, CD14, and
`CD45 and the CD4/CD3:CD8/CD3 ratio) were monitored using
`standard methods (41). Measured variables were graphically depicted
`and visually analyzed.
`Radiologic studies for disease assessment were repeated after
`alternate cycles or as needed. A complete response was reported if
`there was disappearance of all active disease. A partial response was
`reported if there was at least a 50% reduction in total tumor size (the
`sum of
`the product of
`the bidimensional measurements of all
`lesions). A confirmed response was reported if two measurements
`separated by a minimum of 4 weeks indicated a response and an
`unconfirmed response was reported if a response occurred but did not
`meet the criteria required for a confirmed response. Stable disease was
`scored if there was <50% reduction in total tumor size or <25%
`increase in the size of one or more measurable lesions. An increase in
`the size of one or more measurable lesions by at least 25% or the
`appearance of any new lesion was considered disease progression
`(42). Clinical benefit included the number of patients with confirmed
`and unconfirmed complete and partial responses and the number of
`patients with stable disease for at least 24 weeks. Time to tumor
`progression was measured from day 1 of temsirolimus treatment until
`documented disease progression.
`Pharmacokinetic analyses. Whole blood samples for the determina-
`tion of temsirolimus and sirolimus concentrations were collected in
`sodium EDTA tubes (3 mL each) in cycles 1 and 3: on days 1 and 5 at
`0 (before treatment), 0.25, 0.50, 1, 2, 4, and 6 hours; on days 2 to 4 at
`
`0 hours; and on days 8, 10, and 12. The samples were frozen at 70jC
`until assayed. To determine the blood to plasma partitioning of
`temsirolimus, 6 mL blood samples were collected in cycle 1 on days
`1 and 5 at 0.5 hour after drug administration and in cycle 2 on day 1
`before drug administration. These samples were centrifuged immedi-
`ately and the plasma was stored at 70jC until assayed.
`Temsirolimus and sirolimus concentrations in whole blood were
`measured using a liquid chromatography-tandem mass spectrometry
`procedure (Taylor Technology,
`Inc., Princeton, NJ) as described
`(35). Both temsirolimus and sirolimus concentration data were
`analyzed by noncompartmental methods. A compartmental model
`was also used to fit temsirolimus concentration data. Pharmacoki-
`netic analyses were based on concentrations derived in whole blood
`due to the limited stability of
`temsirolimus in plasma. A two-
`compartment open model was fit to the concentration data with
`dose administration and elimination from the central compartment.
`Variable estimation for each patient and treatment period was
`individually derived using the maximum likelihood estimation
`algorithm in the ADAPTII software, release 4, March 1997 (Bio-
`medical Simulations Resource, University of Southern California,
`Los Angeles, CA).
`Dose-dependent variables were normalized and all pharmacokinetic
`variables were log transformed before performing ANOVA. The ANOVA
`assessed variability factors for course (j) and patient (k) using the
`model: yjk = l + coursej + patientk + ejk, in which l is the overall mean
`and e is the within-patient random error in variable y. Statistical
`
`Fig. 3. Representative pustular skin rash in a patient treated with temsirolimus.
`
`Clin Cancer Res 2006;12(19) October 1, 2006
`Downloaded from
`clincancerres.aacrjournals.org
`
`5758
`www.aacrjournals.org
`on August 24, 2015. © 2006 American Association for Cancer
`Research.
`
`NOVARTIS EXHIBIT 2056
`Par v. Novartis, IPR 2016-01479
`Page 4 of 9
`
`

`

`Phase I Study of Temsirolimus
`
`Table 3. Pharmacokinetic variables of temsirolimus on day 5, mean F SD (no. patients) (Cont’d)
`
`AUC (cycle 3; ng h/mL)
`
`—
`2,812 (1)
`4,705 F 1,781 (3)
`5,439 F 2,223 (3)
`7,756 (1)
`9,353 F 1,053 (4)
`
`CLc (L/h)
`6.2 F 3.3 (2)
`5.2 F 2.7 (3)
`5.5 F 1.4 (5)
`7.6 F 1.5 (4)
`16.5 F 5.3 (5)
`19.9 F 7.5 (12)
`
`Vdss (L)
`132.1 F 14.2 (2)
`57.1 F 21.7 (3)
`81.5 F 23.7 (5)
`111.1 F 8.7 (4)
`232.5 F 110.9 (5)
`239.2 F 116.0 (12)
`
`AR
`
`2.2 F 1.1 (3)
`1.0 F 0.6 (3)
`1.3 F 0.4 (5)
`1.1 F 0.2 (4)
`0.7 F 0.4 (4)
`0.8 F 0.1 (11)
`
`B/Pratio (day 1)
`10.9 F 13.9 (2)
`10.4 F 3.2 (4)
`9.1 F 6.3 (6)
`3.7 F 1.7 (4)
`1.5 F 0.9 (6)
`1.5 F 0.6 (12)
`
`differences with P < 0.05 were considered significant. Before statistical
`analysis, C max was normalized to the daily temsirolimus dose, and
`AUC and AUCsum were normalized to the cumulative dose adminis-
`tered over the respective 2-week cycle. All available data were included
`in the statistical analysis. To assess the proportionality of exposure
`with dose, C max, AUC, and AUCsum were analyzed using the power
`b
`model Y = aDOSE
`, in which Y is the pharmacokinetic variable of
`interest, b is the variable estimate for slope, and a is the intercept. For
`this analysis, the null hypothesis, Ho: b = 1 was tested. Rejection of
`the relationship between Y and DOSE is not
`Ho indicates that
`proportional.
`
`Results
`
`General. A total of 63 patients, whose relevant character-
`istics are shown in Table 1, were enrolled on this study from
`August 1998 to May 2000. The last patient completed the study
`in February 2002. Patients received a total of 361 2-week cycles
`of temsirolimus. The median number of cycles administered
`per patient was 4 (range, 1-21). Fifty-eight patients had received
`prior treatment with chemotherapy alone and 30 had received
`prior treatment with radiation therapy either alone (2) or
`combined with chemotherapy (28).
`the temsirolimus dose
`Dose escalation. The results of
`escalation are shown in Table 2. The first patient
`in the
`0.75 mg/m2/d cohort experienced grade 3 neutropenia.
`Because of this grade 3 toxicity, the cohort was expanded to
`three patients as dictated by the protocol. The two additional
`patients who were treated at this dose developed no adverse
`events. The first patient in the next cohort (1.25 mg/m2/d)
`also experienced grade 3 neutropenia and three additional
`patients were treated at this dose and developed no adverse
`the 2.16 mg/m2/d
`events. No DLTs were observed until
`cohort. In this cohort, one patient had a DLT of grade 3
`hypocalcemia; five additional patients were treated and had
`no DLTs. Dose escalation continued without additional DLTs
`until the 15 mg/m2/d cohort. In this cohort, one patient had a
`DLT of grade 3 hyperglycemia; two additional patients were
`treated and had no DLTs. In the 19 mg/m2/d cohort, one
`patient had DLTs of grade 3 elevations in transaminases; five
`additional patients were treated and one of these had grade 3
`thrombocytopenia. To further evaluate this dose level, six
`additional patients were treated and one patient had DLTs of
`grade 3 vomiting, diarrhea, and asthenia and two had grade 3
`thrombocytopenia, which required dose reductions. The two
`patients with the DLTs and the three with the dose reductions
`in the 19 mg/m2/d cohort were heavily pretreated. Thus, the
`decision was made to classify patients based on whether they
`had been heavily pretreated or minimally pretreated for the
`remainder of the dose escalation.
`
`Five additional heavily pretreated patients were treated
`with 15 mg/m2/d temsirolimus for a total of six in the
`heavily pretreated cohort and no DLTs were observed. Of
`the six heavily pretreated patients who had been treated with
`19 mg/m2/d temsirolimus, two had DLTs and three required
`dose reductions. Based only on DLTs, the MTD would have
`been 19 mg/m2/d but, because of the dose reductions, the dose
`of 15 mg/m2/d was considered the MTD for heavily pretreated
`patients.
`Six minimally pretreated patients had been treated with
`19 mg/m2/d temsirolimus and none had DLTs. Thus, six
`minimally pretreated patients were treated with 24 mg/m2/d
`temsirolimus. One had a DLT of grade 3 stomatitis and two
`required dose reductions, one because of grade 2 thrombo-
`cytopenia and the other because of grade 2 erythema
`nodosum. Based on the DLT and the two dose reductions, a
`MTD was not formally identified but the dose of 19 mg/m2/d
`was considered the maximum acceptable dose in minimally
`pretreated patients.
`Toxicity. Selected temsirolimus-related toxicities as a func-
`tion of dose that occurred in at least 10% of patients in any
`treatment cycle are summarized in Fig. 2. The most common
`drug-related adverse events observed across all dose levels were
`asthenia (56%), mucositis (54%), nausea (41%), and cutane-
`ous toxicity (41%). The two most frequent drug-related grades
`3 to 4 adverse events were hypophosphatemia and hypergly-
`cemia in 11% and 8% of patients, respectively. Overall, 10
`patients required dose reductions; 7 of these and 20 additional
`patients required dose delays.
`Hematologic toxicity consisted mainly of thrombocytopenia
`(33%) and leukopenia (27%). Grade 3 thrombocytopenia
`occurred in five patients, including three heavily pretreated
`patients treated at the 19 mg/m2/d dose (Fig. 2). Thus, this
`incidence seemed to be dose related. Thrombocytopenia
`was the most common cause for dose reductions and delays
`(four patients with both and seven with only delays). Five
`patients developed grade 3 neutropenia; three were treated
`with <15 mg/m2/d temsirolimus, suggesting that severe
`neutropenia was not dose related. Neutropenia contributed
`to dose reduction and delay in one patient. Seventeen (27%)
`patients developed temsirolimus-related grades 1 to 2 epistaxis,
`which resolved rapidly; 10 were treated with doses of at least
`15 mg/m2/d.
`Treatment with temsirolimus resulted in few severe non-
`hematologic toxicities. Although 54% of patients developed
`mucositis, only one patient who was treated with 24 mg/m2/d
`temsirolimus developed grade 3 mucositis, a DLT (Table 2).
`Drug-related cutaneous toxicity was commonly observed in
`patients treated with temsirolimus over a wide range of doses
`
`www.aacrjournals.org
`Downloaded from on August 24, 2015. © 2006 American Association for Cancerclincancerres.aacrjournals.org
`
`5759
`Clin Cancer Res 2006;12(19) October 1, 2006
`
`Research.
`
`
`
`NOVARTIS EXHIBIT 2056
`Par v. Novartis, IPR 2016-01479
`Page 5 of 9
`
`

`

`Cancer Therapy: Clinical
`
`Table 4. Pharmacokinetic variables of sirolimus on day 5, mean F SD (no. patients)
`
`Dose group (mg/m2)
`
`0.75
`1.25
`2.16
`4.5
`15.0
`19.1
`
`C max (ng/mL)
`9.3 F 4.1 (3)
`27.3 F 7.6 (3)
`34.3 F 21.2 (5)
`48.7 F 16.7 (4)
`57.8 F 24.3 (5)
`133.9 F 70.3 (12)
`
`t max (h)
`25.9* F 37.5 (3)
`3.6 F 2.2 (3)
`3.6 F 2.1 (5)
`2.8 F 2.3 (4)
`1.8 F 1.5 (5)
`2.0 F 1.5 (12)
`
`t 1/2 (h)
`102.0 F 46.3 (3)
`80.3 F 15.5 (3)
`49.4 F 27.2 (6)
`50.3 F 6.8 (4)
`39.9 F 1.3 (2)
`52.3 F 14.5 (11)
`
`AUC (cycle 1; ng h/mL)
`1,866 F 1,324 (3)
`4,186 F 1,293 (3)
`3,240 F 2,463 (6)
`5,583 F 2,728 (4)
`4,729 F 545 (2)
`15,503 F 8,573 (11)
`
`Dose gr
`
`Abbreviations: AUCratio, uncorrected ratio of sirolimus to temsirolimus AUC; AUCsum, arithmetic sum of temsirolimus and sirolimus AUC.
`*One of three patients exhibited an abnormally prolonged t max.
`
`(26 total patients, 41%; Fig. 2). These included maculopapular
`rashes, acne, pustular rashes (Fig. 3), and pruritus. All skin
`reactions were grades 1 to 2 in severity and reversible. One
`patient treated with 24 mg/m2/d temsirolimus presented on
`day 94 with a clinical picture consistent with erythema
`nodosum (grade 2), which was considered to be possibly
`related to study medication, resulted in a dose reduction, and
`resolved with prednisone treatment. Three patients developed
`symptoms consistent with an allergic drug reaction, which
`began shortly after the start of the first i.v.
`infusion and
`ended after stopping the infusion. One patient treated with
`11.3 mg/m2/d temsirolimus developed a grade 4 allergic
`reaction and discontinued treatment. The other two patients
`were treated with 24 mg/m2/d temsirolimus, developed grades
`1 and 3 allergic reactions, and had dose delays.
`Some laboratory abnormalities were frequently reported as
`adverse events. Temsirolimus-related hyperglycemia was
`reported in 11 (17%) patients and was grades 3 to 4 in 5
`(8%) patients (Fig. 2). Hyperglycemia was a DLT for one
`patient treated with the 15 mg/m2/d dose (Table 2). Temsir-
`olimus-related elevations in plasma triglyceride and choles-
`terol levels occurred in 23 (37%) and 14 (22%) patients,
`respectively, and reached grades 3 to 4 levels in 3 (5%) and
`2 (3%) patients, respectively (Fig. 2). One minimally pretreated
`patient treated with the 15 mg/m2/d dose developed grade 3
`hypertriglyceridemia and discontinued treatment. Other tem-
`sirolimus-related grades 3 to 4 laboratory abnormalities
`included hypophosphatemia (7 patients, 11%), hypokalemia
`(3 patients, 5%), and hypocalcemia (2 patients, 3%).
`Hypocalcemia was a DLT for one patient treated with the
`2.16 mg/m2/d dose. Elevations of aspartate and alanine
`aminotransaminases occurred in 9 (14%) and 8 (13%)
`patients, respectively. Elevations in aspartate and alanine
`aminotransaminases were DLTs in one heavily pretreated
`patient treated with the 19 mg/m2/d dose.
`Immunologic studies. There were no episodes of infections
`or any other clinical manifestation that indicated an opportu-
`nistic infection or immunosuppressed state. Lymphocyte cell
`surface phenotype analysis and mitogen proliferation assays
`did not show any consistent trend toward immunosuppression
`(data not shown). Although intersubject variability was
`considerable, results for individual patients were consistent.
`Proliferative responses to mitogens and pooled allogeneic cells
`were within the control ranges, with two exceptions that
`recovered to the reference range within the study period.
`Pharmacokinetic analysis. Whole blood and plasma samples
`were available from 62 patients receiving doses of 0.75 to
`
`24 mg/m2 (1.42-55.2 mg). Following drug administration,
`temsirolimus concentrations decreased with time in a poly-
`exponential manner. A summary of relevant pharmacokinetic
`variables measured for temsirolimus and sirolimus is provided
`in Tables 3 and 4, respectively. Figures 4 and 5 show the
`relationship between temsirolimus exposure (C max and AUC,
`respectively) and dose. Over the wide range of doses evaluated,
`temsirolimus exposure increased with dose in a less than pro-
`portional manner. Steady-state volume of distribution (Vdss)
`was extensive,
`increased with dose, and exhibited values
`typically exceeding total body weight. At <15 mg/m2, temsir-
`olimus exhibited preferential partitioning into RBC, with mean
`blood-to-plasma ratios of 3.7 to 10.9 (coefficient of variation,
`31-128%), whereas at z15 mg/m2 the ratios approached unity.
`Mean clearance from whole blood increased with increasing
`dose from 5.2 to 19.9 L/h and was associated with modest
`to moderate intersubject variability (coefficient of variation,
`20-54%). Mean terminal half-life was 13 to 25 hours.
`Sirolimus was a major metabolite that was observed early
`(after 15 minutes of infusion) and decreased with time in an
`apparent monoexponential or biexponential fashion. Sirolimus
`exposure was generally comparable with temsirolimus expo-
`sure, with sirolimus-to-temsirolimus AUC ratios of 0.6 to 1.8.
`No statistically significant differences in pharmacokinetic
`variables were apparent when cycle was a factor in ANOVA
`analysis, a finding consistent with limited degrees of drug
`accumulation observed with multiple cycles of treatment. No
`age-related (Figs. 4 and 5) or sex-related effects were apparent.
`Analysis of toxicity events as a function of exposure revealed
`a positive correlation between temsirolimus C max and the
`severity of thrombocytopenia (P = 0.014). Temsirolimus AUC
`showed a positive correlation with the severity grades of
`aspartate aminotransaminase elevation and hypocalcemia
`(P = 0.012 and 0.030, respectively). Sirolimus AUC showed a
`positive correlation with the severity grades of hypophospha-
`temia and thrombocytopenia (P = 0.003 and 0.011, respec-
`tively). AUCsum were positively r

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