throbber
JOURNAL OF INTERFERON AND CYTOKINE RESEARCH 21:257–263 (2001)
`Mary Ann Liebert, Inc.
`
`Randomized Multicenter Phase II Trial of Subcutaneous
`Recombinant Human Interleukin-12 Versus Interferon-a2a for
`Patients with Advanced Renal Cell Carcinoma
`
`ROBERT J. MOTZER,1 ASHOK RAKHIT,3 JOHN A. THOMPSON,4 JOHN NEMUNAITIS,5
`BARBARA A. MURPHY,6 JULIE ELLERHORST,7 LAWRENCE H. SCHWARTZ,2
`WILLIAM J. BERG,1 and RONALD M. BUKOWSKI8
`
`ABSTRACT
`
`Recombinant human interleukin-12 (rHuIL-12) is a pleiotropic cytokine with anticancer activity against re-
`nal cell carcinoma (RCC) in preclinical models and in a phase I trial. A randomized phase II study of rHuIL-
`12 compared with interferon-a (IFN-a) evaluated clinical response for patients with previously untreated, ad-
`vanced RCC. Patients were randomly assigned 2:1 to receive either rHuIL-12 or IFN-a2a. rHuIL-12 was
`administered by subcutaneous (s.c.) injection on days 1, 8, and 15 of each 28-day cycle. The dose of IL-12 was
`escalated during cycle 1 to a maintenance dose of 1.25 l g/kg. IFN was administered at 9 million units by s.c.
`injection three times per week. Serum concentrations of IL-12, IFN-c , IL-10, and neopterin were obtained in
`10 patients treated with rHuIL-12 after the first full dose of 1.25 l g/kg given on day 15 (dose 3) of cycle 1
`and again after multiple doses on day 15 (dose 6) of cycle 2. Thirty patients were treated with rHuIL-12, and
`16 patients were treated with IFN-a. Two (7%) of 30 patients treated with rHuIL-12 achieved a partial re-
`sponse, and the trial was closed to accrual based on the low response proportion. IL-12 was absorbed rapidly
`after s.c. drug administration, with the peak serum concentration appearing at approximately 12 h in both
`cycles. Serum IL-12 concentrations remained stable on multiple dosing. Levels of IFN-c , IL-10, and neopterin
`increased with rHuIL-12 and were maintained in cycle 2. rHuIL-12 is a novel cytokine with unique pharma-
`cologic and pharmacodynamic features under study for the treatment of malignancy and other medical con-
`ditions. The low response proportion associated with rHuIL-12 single-agent therapy against metastatic RCC
`was disappointing, given the preclinical data. Further study of rHuIL-12 for other medical conditions is un-
`derway. For RCC, the study of new cytokines is of the highest priority.
`
`INTRODUCTION
`
`RECOMBINANT HUMAN INTERLEUKIN-12 (rHuIL-12) is a cy-
`
`tokine with a variety of immunomodulatory effects on T
`lymphocytes and natural killer (NK) cells including (1) en-
`hancing the lytic activity of NK lymphokine-activated killer
`(LAK) cells, (2) facilitating specific cytolytic T lymphocyte re-
`sponses, (3) inducing the secretion of interferon-g (IFN-g) by
`both T and NK cells, and (4) promoting the development of
`
`Th1 helper T cells, thereby contributing to the development of
`cell-mediated immune responses.(1–3) rHuIL-12 has also been
`demonstrated to inhibit angiogenesis, which may contribute to
`its antitumor activity.(4) rHuIL-12 has been shown to have strik-
`ing therapeutic effects in a number of mouse tumor models, in-
`cluding the Renca renal cell model,(5) and mouse models of in-
`fectious disease and airway inflammation. On the basis of these
`studies, clinical trials investigating the potential therapeutic ef-
`fects of rHuIL-12 in the treatment of cancer, human immuno-
`
`1Genitourinary Oncology Service, Division of Solid Tumor Oncology, Department of Medicine, and the 2Department of Medical Imaging,
`Memorial Sloan-Kettering Cancer Center, New York, NY.
`3Hoffmann-La Roche, Inc., Nutley, NJ
`4University of Washington, Seattle, WA
`5US Oncology, Dallas, TX
`6Vanderbilt University, Nashville, TN
`7University of Texas M.D. Anderson Cancer Center, Houston, TX
`8The Cleveland Clinic Foundation, Cleveland, OH
`
`257
`
`Downloaded by Guangxi University for Nationalities from online.liebertpub.com at 02/22/18. For personal use only.
`
`NOVARTIS EXHIBIT 2075
`Breckenridge v. Novartis, IPR 2017-01592
`Page 1 of 7
`
`

`

`258
`
`MOTZER ET AL.
`
`deficiency virus (HIV) infection, chronic viral hepatitis, and
`asthma were initiated.
`IFN-a and IL-2 have a low level of antitumor effector against
`renal cell carcinoma (RCC).(6,7) As this malignancy is associ-
`ated with a poor prognosis and is chemotherapy resistant,(8) the
`study of new cytokines is a priority. A phase I trial of rHuIL-
`12 was conducted in patients with advanced RCC using two
`schedules, as a fixed dose an an uptitration schedule, whereby
`the dose of rHuIL-12 was increased for each patient to a target
`dose.(9) The study showed antitumor activity against metasta-
`tic RCC and improved tolerability for the uptitration schedule
`and suggested a dose suitable for phase II study.(9) The phar-
`macokinetic and pharmacodynamic studies showed that serum
`levels of IL-12, IFN-g, and IL-10 produced in response to
`IL-12 were all highest in the week after the first dose of rHuIL-
`12, were dose related, and decreased after long-term adminis-
`tration when rHuIL-12 was administered as a fixed dose.(10)
`Clinical responses associated with rHuIL-12 treatment
`against RCC were reported in phase I trials conducted by oth-
`ers, as well.(11–14) To further define efficacy, toxicity, and phar-
`macology, we conducted a randomized phase II trial of rHuIL-
`12 in patients with RCC. Patients were randomized to receive
`rHuIL-12 by an uptitration schedule based on our phase I trial(9)
`or IFN-a2a (Roferon®-A) (Hoffman-La Roche, Inc., Nutley,
`NJ). A proportion of patients treated with rHuIL-12 had phar-
`macologic and pharmacokinetic studies performed. The results
`of this trial and these studies follow.
`
`rHuIL-12 therapy
`
`rHuIL-12 was supplied by Hoffman-La Roche, Inc., as
`ready-to-use human serum albumin (HAS)-free solution in
`single-use glass vials in three concentrations: 10, 50, and 100
`mg/ml purified rHuIL-12 in 1 ml sterile solution containing
`polysorbate 80 (0.2 mg/ml) and 67 mM phosphate-buffered
`saline (PBS) adjusted to pH 7.0. The vials were stored at 2–8°C
`and protected from light. rHuIL-12 was administered by sub-
`cutaneous (s.c.) injection using a 25G needle.
`Patients were treated by s.c. injection on days 1, 8, and 15
`of each 28-day cycle. The dose was escalated during the first
`cycle: day 1 dose 5 0.1 m/kg, day 8 dose 5 0.5 mg/kg, and day
`15 dose 5 1.25 mg/kg, with a maximum dose of 100 mg. Dur-
`ing subsequent cycles, all treatment was administered at a main-
`tenance dose of 1.25 mg/kg. Treatment was delivered on an out-
`patient basis except when pharmacokinetic studies were being
`performed. Patients were treated until disease progression or
`unacceptable toxicity. The treatment dose was modified for tox-
`icity according to a nomogram.
`
`IFN-a therapy
`
`IFN-a2a (Roferon®-A) was supplied by Hoffmann-La
`Roche, Inc., as ready-to-use, HSA-free solution in single-use
`glass vials. IFN-a2a was administered by s.c. injection at a dose
`of 9 million units three times each week. Dose modifications
`were based on toxicity grade.
`
`PATIENTS AND METHODS
`
`Patient follow-up
`
`Patients
`
`Forty-six patients with advanced RCC were accrued from six
`centers to this Institutional Review Board-approved clinical trial
`between February and May 1997. All patients entered in the
`trial gave informed consent. They were required to have mea-
`surable disease, Karnofsky performance status $80%, white
`blood cell (WBC) count $3000 cells/mm3, granulocytes $2000
`cells/mm3, platelet count $75,000/mm3, hemoglobin $9 g/dl,
`serum bilirubin #1.5 times normal, transaminase levels and al-
`kaline phosphatase $2.5 times normal, serum creatinine con-
`centration #1.5 times normal, and PT/PTT within normal lim-
`its. Exclusion criteria included prior systemic treatment for RCC,
`active brain metastases, history of psychiatric disabilities or
`seizures, clinically significant comorbid conditions, active in-
`fection, and a history of any Th1-mediated autoimmune disease.
`
`Trial design
`
`Patients were prospectively randomized to receive treatment
`with either rHuIL-12 or IFN-a. The ratio of assignment for
`treatment with rHuIL-12 to IFN-a was 2:1. The target accrual
`was 80 patients treated with rHuIL-12 and evaluated for re-
`sponse. If an objective tumor response rate of 25% occurred in
`80 patients, the 95% confidence interval (CI) would range from
`16.0% to 35.9%. Thus, the lower limit of normal would be sim-
`ilar to the response rate induced by other therapies.(8) No for-
`mal interim analysis was planned, but an early stopping rule
`was implemented by the sponsor based on a low response pro-
`portion observed in the rHuIL-12 arm.
`
`Patients were seen and examined at periodic intervals, with
`laboratory evaluation that included hematologic, coagulation,
`and biochemistry panels. Tumor assessments were made after
`
`TABLE 1. RESULTS OF CLINICAL TRIAL
`
`Characteristic
`
`Patients
`Males (%)
`Females (%)
`Median age (range)
`Nephrectomy (%)
`Total (%)
`Partial (%)
`Disease sites (%)
`One (%)
`Two (%)
`Three or more (%)
`Response
`Evaluable
`Favorable (partial)
`response (%)
`Toxicity: grade $3
`Flu-like symptoms (%)
`Liver function test
`elevation (%)
`Neutropenia (%)
`Ascites (%)
`Stomatitis (%)
`
`rHuIL-12
`No.
`
`30
`18 (60)
`12 (40)
`55 (40–77)
`16 (53)
`15 (50)
`1 (3)
`
`8 (27)
`6 (20)
`16 (53)
`
`29
`
`2 (7)
`
`3 (10)
`
`2 (7)
`1 (3)
`1 (3)
`1 (3)
`
`IFN-a
`No.
`
`16
`9 (56)
`7 (44)
`60 (41–70)
`9 (56)
`8 (50)
`1 (6)
`
`1 (6)
`4 (25)
`11 (69)
`
`14
`
`0 (0)
`
`1 (7)
`
`1 (7)
`0 (0)
`0 (0)
`0 (0)
`
`Downloaded by Guangxi University for Nationalities from online.liebertpub.com at 02/22/18. For personal use only.
`
`NOVARTIS EXHIBIT 2075
`Breckenridge v. Novartis, IPR 2017-01592
`Page 2 of 7
`
`

`

`PHASE II TRIAL OF IL-12
`
`259
`
`every two cycles. Responses were graded according to the
`World Health Organization (WHO) Criteria, and toxicity was
`graded by the National Cancer Institute (NCI) Common Tox-
`icity Criteria.
`
`Pharmacodynamic and pharmacokinetic methods
`
`Serum concentrations of rHuIL-12 were measured at base-
`line, and 4, 6, 8, 10, 12, 16, 20, 24, 30, 36, and 48 h after dos-
`ing on day 15 of the first two cycles in 10 patients treated at
`two of the centers. rHuIL-12 concentration was measured by a
`two-step method of antibody capture to insure specificity, fol-
`lowed by a cell proliferation assay. rHuIL-12 was isolated from
`serum by an affinity technique that involved incubation of sam-
`ples in sterile tissue culture plates precoated with mouse anti-
`human IL-12 monoclonal antibody (mAb).(15) The method has
`been described previously.(9,10)
`IFN-g, neopterin, and IL-10 concentrations were measured in
`the serum of 10 patients at preselected times. Serum samples
`were obtained at baseline and 10, 24, 48, 72, 96, and 168 h
`after treatment on day 15 of the first two cycles of treatment.
`IFN-g concentrations in serum were determined by a commer-
`cial assay (R&D Systems, Minneapolis, MN). Neopterin con-
`centration in serum was measured using a radioimmunoassay
`that used 125I-neopterin as a tracer (Incstar, Stillwater, MN).
`Serum IL-10 was measured by an ELISA with a commercial
`assay (Boehringer Mannheim, Mannheim, Germany). The
`methodologies have been described previously.(9,10)
`Serum concentration compared with time data was analyzed
`for rHuIL-12 and other immunologic markers by a noncom-
`partmental model.
`
`RESULTS
`
`Clinical trial
`
`Forty-six patients were enrolled (Table 1). Thirty patients
`were randomized to treatment with rHuIL-12, and 16 were ran-
`domized to treatment with IFN-a2a. The median age was 55
`years on the rHuIL-12 arm and 60 years on the IFN-a2a arm.
`Approximately one half of all patients had a prior nephrectomy.
`Two of 29 (7%, 95% C.I. 0-16%) evaluable patients treated
`with rHuIL-12 achieved a partial response. One patient who re-
`sponded had prior nephrectomy with lung-only metastases, and
`the second patient had prior nephrectomy with three metastatic
`sites, lung, bone, and skin. No patient treated with IFN-a2a
`achieved a partial or complete response. Based on the low re-
`sponse to rHuIL-12, accrual to the trial was stopped.
`The most common toxicity associated with rHuIL-12 was
`flu-like symptoms. Grade 3 or 4 elevation of hepatic transam-
`inases occurred in 2 (7%) patients receiving rHuIL-12 and 1
`(7%) receiving IFN-a2a. Other severe toxicities occurring on
`the rHuIL-12 arm were neutropenia, ascites, and stomatitis.
`
`Pharmacokinetic studies
`
`Serum concentrations of IL-12 were obtained in 10 patients
`treated with rHuIL-12 at two centers participating in this study.
`Because of gradual dose escalation in the first cycle of treat-
`ment, serum concentrations were measured after the first full
`dose of 1.25 mg/kg given on day 15 (dose 3) of cycle 1 and
`again after multiple doses on day 15 (dose 6) of cycle 2. IL-12
`was absorbed rapidly after s.c. administration, with peak serum
`concentration noted at about 12 h in both cycles. We reported
`
`500
`
`450
`
`400
`
`350
`
`Cll!,le 2 D!!l£ 15
`
`E --O> a. 300
`-N
`::, ... Q)
`
`T"""
`I
`...J
`E
`
`(f)
`
`250
`
`200
`
`150
`
`100
`
`50
`
`0
`0.00
`
`10.00
`
`20.00
`
`30.00
`Time (hr)
`
`40.00
`
`50.00
`
`60.00
`
`FIG. 1. Mean (6SE) serum concentrations of IL-12 in RCC patients after 1.25 mg/kg/week s.c. administration of rHuIL-12 us-
`ing a slow dose escalation scheme.
`
`Downloaded by Guangxi University for Nationalities from online.liebertpub.com at 02/22/18. For personal use only.
`
`NOVARTIS EXHIBIT 2075
`Breckenridge v. Novartis, IPR 2017-01592
`Page 3 of 7
`
`

`

`260
`
`MOTZER ET AL.
`
`TABLE 2. COMPARISON OF PHARMACOKINETIC AND IMMUNOLOG IC SURROGATE PARAMETERS IN
`CYCLE 1 VS. CYCLE 2 AFTER ADMINISTRATION OF rHUIL-12 IN RCC PATIENTS
`
`Analyte
`
`IL-12
`Mean
`SE
`IFN-g (pg/ml)
`Mean
`SE
`IL-10 (pg/ml)
`Mean
`SE
`Neopterin (ng/ml)
`Mean
`SE
`
`Cmax
`Dose 3
`Dose 6
`
`Tmax (h)
`Dose 3
`Dose 6
`
`AUC
`
`Dose 3
`
`Dose 6
`
`384
`88
`
`170
`39
`
`65
`8
`
`432
`112
`
`146
`33
`
`96
`13
`
`8.65
`1.37
`
`10.34
`1.67
`
`15
`4
`
`30
`5
`
`36
`5
`
`80
`14
`
`10
`1
`
`20
`5
`
`25
`3
`
`69
`6
`
`10,646 pg ? h/ml
`2,583
`
`11,031
`2,886
`
`7,982 pg ? h/ml
`2,185
`
`6,483 pg ? h/ml
`1,373
`
`793 ng ? h/ml
`154
`
`6,011
`1,525
`
`9,345
`1,403
`
`1,306
`336
`
`that serum concentrations of IL-12 decreased gradually after
`multiple doses in a fixed-dose scheme.(10) In contrast, the cur-
`rent study showed serum IL-12 concentrations were stable on
`multiple dosing when drug was administered in the slow dose-
`escalation pattern (Fig. 1). The maximum serum concentrations
`of IL-12 were maintained at comparable levels in cycles 1 and
`2 (dose 3, 384 pg/ml, vs. dose 6, 432 pg/ml). Serum area un-
`der the curve (AUC) during the dosing interval was also main-
`tained on multiple dosing (dose 3, 10,646 pg/ml, vs. dose 6,
`11031 pg/ml). Individual pharmacokinetic parameters were
`compared for dose 3 (cycle 1) vs. dose 6 (cycle 2) (Table 2).
`
`Pharmacodynamic studies
`
`Three immunologic surrogate markers were monitored in the
`serum of the 10 patients treated with rHuIL-12 (Table 2). Serum
`IFN-g increased after IL-12 dosing, with peak concentrations
`
`appearing at about 24 h into both cycles 1 and 2. Serum IFN-g
`was not measurable at predose baseline in any of these patients.
`Similar to IL-12, the average peak serum concentration of
`IFN-g was relatively unchanged (170 vs. 146 pg/ml) between
`the two cycles (Fig. 2). Five patients showed a small decrease,
`and 4 patients’ peak concentration increased slightly. Serum
`samples could not be obtained in 1 patient in cycle 2.
`The concentration of neopterin increased significantly from
`baseline as the dose was increased from 0.5 to 1.25 mg/kg/week
`in cycle 1, but induction was maintained at comparable levels
`as the 1.25 m/kg dose was maintained on subsequent cycles
`(dose 3 vs. dose 6: Cmax 8.65 vs. 10.34 ng/ml, and AUC 793
`vs. 1306 ng ? h/ml), demonstrating maintenance of the immuno-
`logic activity of rHuIL-12 in these patients at the end of 2
`months treatment (Fig. 3).
`Serum concentrations of IL-10 increased after administration
`of rHuIL-12. Peak serum IL-10 concentrations appeared be-
`
`200
`
`180
`
`160
`
`- 140
`E -C) 120
`
`.e,
`~ z 100
`!±
`E
`::::, ... Q)
`
`80
`
`60
`
`Cl)
`
`40
`
`20
`
`0
`
`0
`
`20
`
`40
`
`60
`
`100
`80
`Time (hr)
`
`120
`
`140
`
`160
`
`180
`
`FIG. 2. Maintenance of serum concentrations of IFN-g after s.c. administration of 1.25 mg/kg/week of rHuIL-12 in RCC pa-
`tients given in a slow dose escalation scheme.
`
`Downloaded by Guangxi University for Nationalities from online.liebertpub.com at 02/22/18. For personal use only.
`
`NOVARTIS EXHIBIT 2075
`Breckenridge v. Novartis, IPR 2017-01592
`Page 4 of 7
`
`

`

`PHASE II TRIAL OF IL-12
`
`261
`
`25 ~ - - - - - - - - - - - - - - - - - - - - - - - - - -~
`
`20
`
`C:
`
`:::::-
`E ---0)
`~ 15
`·.:::
`Cl>
`C.
`0
`Cl> z 10
`E
`....
`:::J
`Cl>
`(/)
`
`Cycle 1 Dav 15
`
`5
`
`0-+----~---r------r------,-------r------,---,----~------j
`60
`120
`20
`40
`80
`100
`140
`160
`180
`0
`Time (hr)
`
`FIG. 3. Mean (6SE) serum concentrations of neopterin after s.c. administration of 1.25 mg/kg/week of rHuIL-12 in RCC pa-
`tients given a slow dose escalation.
`
`tween 24 and 48 h of IL-12 administration in both cycles. Sim-
`ilar to IL-12 and IFN-g, IL-10 concentrations did not decrease
`on multiple dosing in this study. The mean Cmax and AUC in-
`creased to a small but significant extent (p , 0.05 by paired t-
`test) (Fig. 4).
`
`DISCUSSION
`
`This randomized phase II trial stopped accrual after an in-
`terim analysis showed a low response proportion associated
`with rHuIL-12 treatment in previously untreated patients with
`
`120
`
`100
`
`E 80
`---0)
`-9:
`0 ..... 60
`I
`...J
`E
`:::,
`.... 40
`Q)
`Cl)
`
`20
`
`0
`
`0
`
`20
`
`40
`
`60
`
`100
`80
`Time (hr)
`
`120
`
`140
`
`160
`
`180
`
`FIG. 4. Mean (6SE) serum concentrations of IL-10 after s.c. administration of 1.25 mg/kg/week of rHuIL-12 in RCC patients
`given a slow dose escalation scheme.
`
`Downloaded by Guangxi University for Nationalities from online.liebertpub.com at 02/22/18. For personal use only.
`
`NOVARTIS EXHIBIT 2075
`Breckenridge v. Novartis, IPR 2017-01592
`Page 5 of 7
`
`

`

`262
`
`MOTZER ET AL.
`
`advanced RCC. The low response proportion was consistent
`with the results of the phase I trial, whereby 1 (2%) of 50 pa-
`tients achieved a partial response.(9) However, that trial included
`patients treated with lower doses according to a phase I trial
`design, and approximately 60% had progressed through prior
`cytokine therapy. The current study was undertaken to define
`activity in cytokine-naive patients treated at an optimal dose
`and schedule derived from the phase I trial. Nevertheless, the
`antitumor activity of rHuIL-12 was disappointing, given the
`high degree of response observed with rHuIL-12 in the mouse
`RCC model.(5) Ongoing studies are addressing the efficacy of
`combination treatment using rHuIL-12 with IL-2, as synergy
`for the two cytokines was shown in animal studies.(16)
`In the first part of the phase I trial, rHuIL-12 was adminis-
`tered in fixed weekly doses, and serum concentrations de-
`creased rapidly on multiple dosing.(10) The extent of reduction
`in serum AUC was observed to be dependent on the initial rate
`of exposure. Similar to serum profiles of IL-12, the immuno-
`logic markers IFN-g, IL-10, and neopterin showed a gradual
`decrease on chronic dosing, suggesting a potential downregu-
`lation of the cytokine cascade. This was consistent with the clin-
`ical findings, as adverse events (e.g., fever, fatigue) also de-
`creased in intensity with chronic administration of the drug.
`These observations in the phase I trial led us to study a grad-
`ual dose-escalation scheme to potentially reduce acute adverse
`events (first-dose toxicity) and allow better adaptation of this
`cytokine, thereby reducing the extent of regulatory feedback re-
`sponse.(9) In the current phase II study, patients were treated
`with a slow dose-escalation scheme of 0.1 mg/kg in week 1,
`then 0.5 mg/kg in week 2, followed by a maintenance dose of
`1.25 mg/kg/week during the rest of the treatment period. In con-
`trast to observations from a fixed dosing scheme,(9,10) the serum
`concentration of IL-12 appeared to be maintained in cycle 2
`compared with that observed after the first full dose of 1.25
`mg/kg given in week 3. When compared with first-dose Cmax
`given at 1 mg/kg in an earlier fixed-dose study,(10) concentra-
`tions in the current study were lower, at 1.25 mg/kg dose, sug-
`gesting that the influence of regulatory feedback was still pres-
`ent in this dosing scheme. The Cmax for the fixed first dose of
`1 mg/kg in the phase I trial was 1092 6 SE 275 pg/ml(10) com-
`pared with the first escalation dose of 1.25 mg/kg 384 6 88
`pg/ml in the current study. Therefore, IL-12 exposure may not
`necessarily have been gained by this dosing scheme. However,
`tolerability was improved substantially, as toxicity was unac-
`ceptable in the first dose of the fixed schedule at doses .0.5
`mg/kg.(9)
`Three different immunologic surrogate markers were moni-
`tored in the serum of these 10 patients treated with rHuIL-12.
`Serum IFN-g, a well-recognized Th1 marker of IL-12 activity,
`increased after IL-12 dosing, with peak concentration appear-
`ing at about 24 h; the average peak serum concentration was
`similar between the two cycles. Neopterin, a marker for mac-
`rophage activation, was induced by rHuIL-12 in all patients
`monitored. Similar to IL-12, IFN-g and neopterin did not de-
`crease on multiple dosing in this study, suggesting that immune
`activity was maintained. IL-10 levels increased following
`rHuIL-12 administration. IL-10 can inhibit the synthesis and
`action of IL-12.(17,18) Therefore, the ability of rHuIL-12 to in-
`duce IL-10 production could represent a negative feedback
`mechanism by which IL-12 limits its own effects.
`
`In summary, rHuIL-12 has unique pharmacologic and phar-
`macokinetic properties that warrant further study for patients
`with malignancy and other medical conditions. The lack of an-
`titumor activity for single-agent rHuIL-12 against RCC in this
`study was disappointing, given promising preclinical data. For
`RCC, the study of new cytokines is of the highest priority.
`
`ACKNOWLEDGMENTS
`
`We thank Lucy Dantis, Patricia Fischer, and the immunol-
`ogy nursing staff for nursing support and Carol Pearce for her
`review of the manuscript.
`
`REFERENCES
`
`1. BANKS, R.E., PATEL, P.S., and SELBY, P.J. (1995). Interleukin-
`12: a new clinical player in cytokine therapy. Br. J. Cancer 71,
`655–659.
`2. BRUNDA, M.J. (1997). Interleukin-12. J. Leukocyte Biol. 55,
`280–288.
`3. GALETTI, T.P. (1993). Interleukin-12: a recently discovered cy-
`tokine with potential for enhancing cell-mediated immune re-
`sponses to tumors. Cancer Invest. 11, 500–506.
`4. VOEST, E.E., KENYON, B.M., O’REILLY, M., TRUITT, G.,
`AMATO, R.J., and FOLKMAN, J. (1995). Inhibition of angio-
`genesis in vivo by interleukin-12. J. Natl. Cancer Inst. 87, 581–586.
`5. BRUNDA, M.J., LUISTRO, L., WARRIER, R.R., WRIGHT, R.B.,
`HUBBARD, B.R., MURPHY, M., WOLF, S.F., and GATELY,
`M.K. (1993). Antitumor and anti-metastatic activity of interleukin-
`12 against murine tumors. J. Exp. Med. 178, 1223–1230.
`6. MOTZER, R.J., BANDER, N.H., and NANUS, D.M. (1996). Re-
`nal-cell carcinoma. N. Engl. J. Med. 335, 865–875.
`7. MOTZER, R.J., MAZUMDAR, M., BACIK, J., RUSSO, P.,
`BERG, W.J., and METZ, E.M. (2000). Effect of cytokine therapy
`on survival for patients with advanced renal cell carcinoma. J. Clin.
`Oncol. 18, 1928–1935.
`8. MOTZER, R.J., and RUSSO, P. (2000). Systemic therapy for re-
`nal cell carcinoma. J. Urol. 163, 408–417.
`9. MOTZER, R.J., RAKHIT, A., SCHWARTZ, L.H., OLENCKI, T.,
`MALONE, T., SANDSTROM, K., NADEAU, R., PARMAR, H.,
`and BUKOWSKI, R. (1998). Phase I trial of subcutaneous recom-
`binant human interleukin-12 in patients with advanced renal cell
`carcinoma. Clin. Cancer Res. 4, 1183–1191.
`10. RAKHIT, A., YEON, M.M., FERRANTE, J., FETTNER, J., FET-
`TNER, S., NADEAU, R., MOTZER, R., BUKOWSKI, R., CAR-
`VAJAL, D., WIILKINSON, V., PRESKY, D., MAGRAM, J., and
`GATELY, M. (1999). Down-regulation of the pharmacokinetic-
`pharmacodynamic response to interleukin-12 during long-term ad-
`ministration to patients with renal cell carcinoma and evaluation
`of the mechanism of this “adaptive response” in mice. Clin. Phar-
`macol. Ther. 65, 615–629.
`11. ATKINS, M.B., ROBERTSON, M.J., GORDON, M., LOTZ, M.,
`DECOSTE, M., DUBOIS, J., RITZ, J., SANDLER, A., EDING-
`TON, H., GARZONE, P., MIER, J., CANNING, C., BATTIAO,
`L., TAHARA, H., and SHERMAN, M.I. (1997). Phase I evalua-
`tion of intravenous recombinant human interleukin 12 in patients
`with advanced malignancies. Clin. Cancer Res. 3, 409–417.
`12. OHNO, R., YAMAGUCHI, Y., TOGUE, T., KINOUCHI, T., KO-
`TAKE, T., SHIBATA, M., KIYOHARA, Y., IDEDA, S., FUKUI,
`I., GOHCHI, A., SUGIYAMA, Y., SAJI, S., HAZAMA, S., OKA,
`M., OHNISHI, K., OHHASHI, Y., TSUKAGOSHI, S., and
`TAGUCHI, T.I. (2000). A dose-escalation and pharmacokinetic
`
`Downloaded by Guangxi University for Nationalities from online.liebertpub.com at 02/22/18. For personal use only.
`
`NOVARTIS EXHIBIT 2075
`Breckenridge v. Novartis, IPR 2017-01592
`Page 6 of 7
`
`

`

`PHASE II TRIAL OF IL-12
`
`263
`
`study of subcutaneously administered recombinant human inter-
`leukin 12 and its biological effects in Japanese patients with ad-
`vanced malignancies. Clin. Cancer Res. 6, 2661–2669.
`13. GOLLUB, J.A., MIER, J.W., VEENSTRA, K., McDERMOTT, D.,
`CLANCY, D., CLANCY, M., and ATKINS, M. (2000). Phase I
`trial of twice-weekly intravenous interleukin 12 in patients with
`metastatic renal cell cancer or melanoma: ability to maintain IFN-
`gamma induction is associated with clinical response. Clin. Can-
`cer Res. 6, 1678–1692.
`14. PORTIELJE, J.E., KRUIT, W.H., SCHULER, M., BECK, J.,
`LAMARS, C.H., STOTER, G., HUBER, C., DE BOER-DEN-
`NERT, M., RAHKIT, A., BOLHUIS, R.L., and AULITZKY, W.E.
`(2000). Phase I study of subcutaneously administered recombinant
`human interleukin 12 in patients with advanced renal cell cancer.
`Clin. Cancer Res. 5, 3983–3989.
`15. NADEAU, R., OSTROWSKI, C., NI-WU, G., and LIBERATO,
`D.J. (1995). Pharmacokinetics and pharmacodynamics of recom-
`binant human interleukin-12 (rHuIL-12) in male rhesus monkeys.
`J. Pharmacol. Exp. Ther. 274, 78–83.
`16. WIGGINTON, J.M., KOMSCHLIES, K.L., BACK, T.C.,
`FRANCO, J.L., BRUNDA, M.J., and WILTROUT, R.H. (1996).
`Administration of interleukin 12 with pulse interleukin 2 and the
`rapid and complete eradication of murine renal carcinoma. J. Natl.
`Cancer Inst. 88, 38–43.
`
`17. TRIPP, C.S., WOLF, S.F., and UNANUE, E.R. (1993). Interleukin
`12 and tumor necrosis factor alpha are costimulators of interferon
`gamma production by natural killer cells in severe combined im-
`munodeficiency mice with listeriosis, and interleukin 10 is a phys-
`iologic antagonist. Proc. Natl. Acad. Sci. USA 90, 3725–3729.
`18. D’ANDREA, A., ASTE-AMEZAGA, M., VALIANTE, N.M.,
`MA, X., KUBIN, M., and TRINCHIERI, G. (1993). Interleukin-
`10 inhibits human lymphocyte interferon gamma production by
`suppressing natural killer cell stimulatory factor/IL-12 synthesis in
`accessory cells. J. Exp. Med. 178, 1041–1048.
`
`Address reprint requests to:
`Dr. Robert J. Motzer
`Memorial Sloan-Kettering Cancer Center
`1275 York Avenue, Howard 906
`New York, NY 10021
`
`Tel: (212) 639-6667
`Fax: (212) 717-3133
`E-mail: motzerr@mskcc.org
`
`Received 27 October 2000/Accepted 9 January 2001
`
`Downloaded by Guangxi University for Nationalities from online.liebertpub.com at 02/22/18. For personal use only.
`
`NOVARTIS EXHIBIT 2075
`Breckenridge v. Novartis, IPR 2017-01592
`Page 7 of 7
`
`

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