throbber
Vol. 4, 1183-1191, May 1998
`
`Clinical Cancer Research 1183
`
`Phase I Trial of Subcutaneous Recombinant Human Interleukin-12
`in Patients with Advanced Renal Cell Carcinoma
`
`Robert J. Motzer,1 Ashok Rakhit,
`Lawrence H. Schwartz, Thomas Olencki,
`Thomas M. Malone, Kate Sandstrom,
`Rosemary Nadeau, Harsukh Parmar, and
`Ronald Bukowski
`Genitourinary Oncology Service, Division of Solid Tumor Oncology,
`Department of Medicine [R. J.M., T. M. M.] and Department of
`Medical Imaging [L. H. S.], Memorial Sloan-Kettering Cancer Center,
`New York, New York 10021; Hoffmann-La Roche, Inc., Nutley, New
`Jersey, and Welwyn, United Kingdom [A. R., R. N., H.P.]; and the
`Cleveland Clinic Foundation, Cleveland, Ohio 44195 [T. 0., K. S.,
`R.B.J
`
`ABSTRACT
`Patients with advanced renal cell carcinoma were
`treated in a Phase I trial with escalating doses of recombi(cid:173)
`nant human interleukin-12 (rHuIL-12) given on days 1, 8,
`and 15 of each 28-day cycle. Treatment in the initial dose
`scheme consisted of a fixed dose with dose levels of 0.1, 0.5,
`and 1.0 p.g/kg given to cohorts composed of three or six
`patients. On the basis of the toxicity profile, a second scheme
`(up-titration) was undertaken wherein rHuIL-12 was esca(cid:173)
`lated for each patient from week 1 to week 2, to a target dose
`given week 3 and thereafter; cohort target dose levels were
`0.5, 0.75, 1.0, 1.25, and 1.5 p.g/kg. Fifty-one patients were
`treated: 32 (63%) had prior cytokine therapy and 19 (37%)
`had received no prior systemic therapy. The maximum tol(cid:173)
`erated dose for the faxed dose scheme was 1.0 p.g/kg. Dose(cid:173)
`limiting toxicities included increase in transaminase concen(cid:173)
`tration, pulmonary toxicity, and leukopenia. The most
`severe toxicities occurred with the first injection and were
`milder upon further treatment. With the up-titration dose
`scheme, the maximum tolerated dose was reached at 1.5
`p.g/kg, and dose-limiting toxicity consisted of an increase in
`serum transaminase levels.
`At the maximum tolerated dose of 1.5 p.g/kg, serum
`IL-12 levels increased to a mean peak level of 706 pg/ml.
`Serum levels of IFN--y increased to a mean peak level of
`about 200 pg/ml at 24 h after the first maintenance dose of
`1.5 p.g/kg. The best responses were as follows: one patient
`had complete response, 34 patients were stable, 14 patients
`showed progression, and 1 patient was inevaluable.
`
`In conclusion, rHuIL-12 was relatively well tolerated
`when administered by s.c. injection. The recommended dose
`according to the up-titration schedule of rHuIL-12 (p.g/kg)
`for Phase II trials was as follows: cycle 1, 0.1 (day 1), 0.5
`(day 8), 1.25 (day 15); cycle 2 onwards, 1.25. Phase II trials
`of rHuIL-12 were initiated in previously untreated patients
`with renal cell carcinoma and in patients with melanoma.
`
`INTRODUCTION
`rHulL-12 2 is a heterodimeric protein composed of two
`disulfide-linked subunits having molecular masses of 40 and 35
`kDa, respectively (I, 2). IL-12 as a cytokine has been shown to
`exert a number of regulatory effects on T lymphocytes and NK
`cells (3, 4). These include (a) enhancing the lytic activity of
`NK/LAK-cells; (b) facilitating specific cytolytic T-lymphocyte
`responses; (c) inducing the secretion of IFN--y by both T and NK
`cells; and (d) promoting the development of TH I-type helper T
`cells, thereby contributing to the development of cell-mediated
`immune responses (5-8). Other cell types, including mono(cid:173)
`cytes, macrophages, activated B-cells, dendritic cells, and kera(cid:173)
`tinocytes, also contribute to the biological activity of IL-12
`(5-8). One activity of IL-12 that may contribute to an antitumor
`effect is the ability to inhibit angiogenesis, an effect mediated
`through the induction of IFN--y (9).
`Interest in biological response modifiers as a treatment for
`metastatic renal cell carcinoma is fostered by the reproducible,
`albeit infrequent, responses with IFN-a and IL-2 against this
`chemotherapy-refractory malignancy ( 10). The antitumor activ(cid:173)
`ities of rHuIL-2 were demonstrated in a large number of murine
`tumor models, including Renea, a spontaneously arising renal
`cell carcinoma (8, 11 ). Treatment of mice bearing murine Renea
`renal cell carcinoma resulted in tumor growth inhibition, tumor
`regression, and prolongation of survival; the antitumor effect
`was superior to IL-2 and IFN-a (8, 11 ). These data provided the
`rationale for a Phase I trial of rHuIL-12 in patients with renal
`cell carcinoma. The schedule of s.c. administration at escalating
`dose levels of rHuIL-12 given once weekly for 3 weeks was
`based on single- and multiple-dose toxicology studies per(cid:173)
`formed in cynomolgus monkeys and chimpanzees. 3 Pharmaco(cid:173)
`kinetic and pharmacodynamic parameters were studied, which
`included assessment of IFN--y and serum neopterin, an uncon(cid:173)
`jugated pteridine released by macrophages after activation by
`IFN--y (12).
`
`Received 11/25/97; accepted 2/20/98.
`The costs of publication of this article were defrayed in part by the
`payment of page charges. This article must therefore be hereby marked
`advenisemenr in accordance with 18 U.S.C. Section 1734 solely to
`indicate this fact.
`1 To whom requests for reprints should be addressed, at Memorial
`Sloan-Kettering Cancer Center, 1275 York Avenue, New York.
`NY 10021.
`
`2 The abbreviations used are: rHuIL-12, recombinant human interleu(cid:173)
`kin-12; IL, interleukin; ASAT, aspartate aminotransferase; ALAT, ala(cid:173)
`nine aminotransferase.
`3 Roche, unpublished data.
`
`Downloaded from
`
`on February 22, 2018. © 1998 American Association for Cancerclincancerres.aacrjournals.org
`
`
`Research.
`
`NOVARTIS EXHIBIT 2039
`Breckenridge v. Novartis, IPR 2017-01592
`Page 1 of 10
`
`

`

`1184 IL-12 in Renal Cell Carcinoma
`
`Table 1 Dosing schemes of rHuIL-12 (µ.g/kg)
`
`Cycle I
`
`Day 8
`Day I
`Cohort
`Dose escalation scheme A (fixed dose)
`I
`0.1
`0.1
`II
`0.5
`0.5
`III
`1.0
`1.0
`Dose escalation scheme B (2-step up-titration)
`I
`0.1
`0.25
`II
`0.1
`0.5
`III
`0.1
`0.5
`IV
`0.1
`0.5
`V
`0.1
`0.5
`
`Day 15
`
`0.1
`0.5
`1.0
`
`0.5
`0.75
`1.0
`1.25
`1.5
`
`Cycle 2 and
`others, days
`I, 8, and 15
`
`No. of
`patients
`
`Median no. of
`cycles (range)
`
`0.1
`0.5
`1.0
`
`0.5
`0.75
`1.0
`1.25
`1.5
`
`3
`15
`6
`
`3
`3
`3
`6
`12
`
`4 (1-9)
`4 (1-17)
`4 (.3-7)
`
`6 (2-16)
`2 (2-3)
`3 (2-4)
`4.5 (2-8)
`3 (2-11)
`
`PATIENTS AND METHODS
`Patients. Between April 1995 and November 1996, 51
`patients with advanced renal cell carcinoma were entered in this
`institutional review board-approved Phase I trial. All patients
`were between 18 and 75 years of age; gave informed consent;
`had measurable disease; had a Kamofsky performance status
`2:80%; had an estimated life expectancy of >4 months; had a
`WBC count of 2:3000 cells/mm3 , a granulocyte count of 2:2000
`cells/mm3, a platelet count of 2:75,000 cells/mm 3, and a hemo(cid:173)
`globin level of 2: 10 g/dl; had normal serum bilirubin and
`transaminase levels and alkaline phosphatase :s 2.5 times nor(cid:173)
`mal; had a normal serum creatinine concentration in patients
`without a prior nephrectomy and :s 1.5 times normal in patients
`with prior nephrectomy; and had a serum calcium level of
`:sl2.5 mg/di (:S3.12 mmol/liter). Exclusion criteria included
`active brain metastases, history of psychiatric disabilities or
`seizures, clinically significant cardiac abnormalities, chronic
`obstructive pulmonary disease, prior history of systemic liver
`disease, active systemic infection, and THI-mediated autoim(cid:173)
`mune diseases. Prior systemic therapy was allowed, but patients
`could not have received more than one previous chemotherapy
`plus one previous immunotherapy.
`rHuIL-12.
`rHuIL-12 was supplied by Hoffmann-La
`Roche, Inc. (Nutley, NJ) as ready-to-use HSA-free solution in
`single-dose glass vials containing 10, 100, 500, or 1000 µg of
`purified rHuIL-12 in 1 ml of sterile solution containing poly(cid:173)
`sorbate 80 (0.2 mg/ml) and 67 mM PBS adjusted to pH 7.0. The
`vials were stored at 2-8°C and protected from light. rHuIL-12
`was administered by s.c. injection using a 25 gauge needle.
`Dose Schedule. All patients were treated with two cycles
`of therapy lasting 28 days (Table 1 ). Each cycle consisted of s.c.
`injections on days 1, 8, and 15. Treatment was given on an
`outpatient basis, except that patients were hospitalized for phar(cid:173)
`macokinetic studies. On the basis of results of the tumor assess(cid:173)
`ment following cycle 2, patients with a response of stable
`disease or better received additional cycles of therapy until
`evidence of progression or unacceptable toxicity.
`In the initial dose scheme (scheme A), patients were treated
`with a fixed dose of rHuIL-12 at planned dose levels ofO. l, 0.5,
`and 1.0 µg/kg. Cohorts included three patients per dose level
`until a grade 2 or higher toxicity occurred, with the exception of
`grade 2 fever or Ieukopenia and grade 3 or 4 lymphopenia or
`fever; for these levels and subsequent dose-escalated levels, an
`
`additional three patients were entered. The maximum tolerated
`dose was defined as the level at which two of six patients
`experienced dose-limiting grade 3 or 4 toxicity.
`As a result of observations made during the initial phase of
`the trial regarding treatment tolerability, a second scheme
`(scheme B) was initiated in August 1995, after patients were
`treated on the trial. In this scheme, the dose of rHuIL-12 was
`escalated for each patient on days 8 and 15 of cycle 1 (Table 1).
`Subsequent cycles were administered at the day 15 dose level.
`Target (day 15) dose levels were 0.5, 0.75, 1.0, 1.25, and 1.5
`µg/kg. Patients weighing more than 80 kg received a maximum
`total dose corresponding to a body mass of 80 kg. The number
`of patients treated per dose level and the maximum tolerated
`dose was defined as described for dose scheme A.
`Patients were monitored by physical examination, com(cid:173)
`plete blood count, and serum chemistry on each day of treat(cid:173)
`ment. Each patient had a reassessment of measurable disease
`after the second cycle of treatment and then every 4 weeks
`thereafter. Patients with stable disease had tumor assessments
`performed every 2 months thereafter. All patients kept a daily
`log documenting symptoms and medications taken. Response
`was assessed according to WHO criteria and toxicity according
`to Common Toxicity criteria.
`Pharmacodynamic and Pharmacokinetic Parameters.
`IFN--y, tumor necrosis factor-a, and neopterin concentrations
`were measured in the serum of patients at preselected times.
`Serum samples were obtained at baseline and 10, 24, 48, 72, 96,
`and 168 h following treatment on day 1 of cycle 1 of treatment
`scheme A and day 15 of cycle 1 of treatment scheme B.
`IFN--y concentrations in serum were determined by a com(cid:173)
`mercial assay (R&D Systems, Minneapolis, MN). In brief, 50 µl
`of the assay diluent were added to each well of the microtiter
`plate. Two hundred µ1 of standard or sample was added to each
`well and incubated for 2.5 h at room temperature. Each well was
`then washed three times with buffer, and any remaining wash
`removed by blotting it against a clean paper towel or by aspi(cid:173)
`ration. Two hundred µl of IFN--y conjugate were added and
`incubated for 2 hat room temperature. Each well was repeatedly
`washed/aspirated, 200 µl of substrate solution were added, and
`the samples were incubated for 20 min. Fifty µl of stop solution
`were added to each well and tapped to ensure thorough mixing.
`The absorbance of each well was determined within 30 min
`
`Downloaded from
`
`clincancerres.aacrjournals.org
`
`on February 22, 2018. © 1998 American Association for Cancer
`Research.
`
`NOVARTIS EXHIBIT 2039
`Breckenridge v. Novartis, IPR 2017-01592
`Page 2 of 10
`
`

`

`Clinical Cancer Research 1185
`
`using a spectrophotometer set at 450 nm. The interassay varia(cid:173)
`bility was < 10%, and the limit of detection was 3.0 pg/ml.
`Neopterin concentration in serum was measured using a
`radio-immunoassay that used 1251-labeled neopterin as a tracer
`(lncstar, Stillwater, MN). Antineopterin rabbit antibody was
`incubated with samples, standards, and 1251-labeled neopterin.
`After a 1-h incubation at 37°C, antirabbit antiserum from sheep
`in polyethylene glycol buffer was added and incubated at room
`temperature for 15 min. Samples were centrifuged, supernatant
`was decanted, and pellets were counted for radioactivity. The
`amount of neopterin in sample was inversely proportional to the
`amount of radioactivity in the pellet. Unknown concentrations
`were calculated from a standard curve. The assay showed neg(cid:173)
`ligible cross-reactivity with other neopterin-like compounds
`(biopterin, monapterin, and tetrahydroneopterin) and a lower
`limit of quantitation of 0.2 ng/ml.
`Tumor necrosis factor-a concentrations were determined
`by a quantitative sandwich enzyme-immunoassay technique
`(R&D Systems). Tumor necrosis factor-a standards and study
`samples were incubated with antibody bound to a microtiter
`plate. After washing away unbound substances, an enzyme(cid:173)
`linked polyclonal antibody specific for tumor necrosis factor-a
`was added to the wells. The plates were washed to remove
`unbound antibody-enzyme reagent, and a substrate solution was
`added. Color development was proportional to the amount of
`tumor necrosis factor-a bound in the initial step. Sample con(cid:173)
`centrations were determined on a standard curve by plotting the
`absorbance versus tumor necrosis factor-a concentration. The
`lower limit of quantitation of the assay was 15.6 pg/ml.
`Serum concentrations of rHuIL-12 were measured at base(cid:173)
`line and at 2, 4, 8, IO, 16, 24, 30, 34, 48, and 72 hon days 1-3
`of cycle I of scheme A (fixed dose) and on day 15 of cycle I
`when first maintenance (target) dose was administered in
`scheme B (up-titration schedule). rHuIL-12 concentration was
`measured by a two-step method of antibody capture to ensure
`specificity followed by a cell proliferation assay. rHuIL-12 was
`isolated from serum by an affinity technique that involved
`incubation of samples in sterile tissue culture plates precoated
`with mouse antihuman IL-12 monoclonal antibody (13).
`Samples and rHuIL-12 standards were incubated with the
`bound monoclonal antibody for 3 h on an orbital shaker at room
`temperature. After a sterile wash with PBS to remove all non(cid:173)
`specific material, KIT 5/K6 cells were added to each well of the
`tissue culture plates. After incubation for 66 h, cells were pulsed
`with [methyl-3H]thymidine for six h, and cell proliferation was
`measured by [methyl-3H]thymidine incorporation. Sample val(cid:173)
`ues were determined on a standard curve obtained from plotting
`radioactive counts against IL-12 concentration. The assay had a
`lower limit of detection of 50 pg/ml of serum using 100-µl
`aliquots. The interassay precision was 8.0%. rHulL-12 is stable
`in serum for 24 h at room temperature. Serum samples were also
`found to be stable after three freeze/thaw cycles.
`Anti-rHuIL-12 antibodies were measured in serum at base(cid:173)
`line and periodically thereafter. Anti-rHuIL-12 antibodies were
`determined by a sandwich enzyme immunological assay. The
`assay was based on the ability of the multivalent anti-IL-12
`antibodies to simultaneously bind rHuIL-12 coated on the wells
`of microtiter plate and soluble peroxidase-conjugated rHuIL-12.
`The intra- and interassay variabilities were 11 and 20%, respec-
`
`Table 2 Patient characteristics
`
`Characteristic
`
`Patients
`Male/female
`Median age (range)
`Median Kamofsky performance status
`(range)
`Prior nephrectomy
`Yes
`No
`Prior therapy
`IFN
`IL-2
`IFN plus IL-2 with/without 5-
`fluorouracil or floxuridine
`Granulocyte-macrophage colony-
`stimulating factor plus IL-6
`Chemotherapy (Tallamustine)
`Hormonal therapy (Tamoxifen)
`Radiotherapy
`No systemic therapy
`Evaluable sites
`Lung
`Mediastinum
`Retroperitoneal lymph node
`Kidney
`Adrenal gland
`Bone
`Liver
`Spleen
`Peripheral node
`Muscle
`Skin
`
`No.(%)
`
`51
`38 (74)/13 (26)
`56 (38-73)
`90 (80-90)
`
`43 (84)
`8 (16)
`
`14 (27)
`6 (12)
`11 (26)
`
`1 (2)
`
`2 (4)
`1 (2)
`6 (12)
`19(37)°
`
`31 (61)
`9 (18)
`9 (18)
`5 (10)
`5(10)
`5 (10)
`5 (10)
`2(4)
`2(4)
`I (2)
`I (2)
`
`"Includes two patients treated with radiation therapy.
`
`tively. The level of detection for this assay was 29 ng/ml. The
`specificity of the assay for anti-IL-12 antibodies was confirmed
`by the absence of any measured response in serum samples
`containing antibodies against various irrelevant proteins.
`
`RESULTS
`Patient Characteristics. Fifty-one patients were treated
`with rHuIL-12 (Table 2). The median age was 56 years, and 43
`(75%) had a prior nephrectomy. Thirty-two (63%) had prior
`cytokine therapy (IFN-a, IL-2, or IL-6), and 19 (37%) had
`received no prior systemic therapy.
`Treatment Administered and Toxicity. Twenty-four
`patients were treated with a fixed-dose schedule (scheme A) and
`27 were treated with the up-titration schedule (scheme B; Table
`I). Fever, fatigue, and a rapid, transient decrease in WBC counts
`after the first injection were common to all dose levels. The
`decrease in WBC count included a decrease in lymphocytes and
`neutrophils; the lymphocyte count decreased to a greater extent.
`Other frequent adverse events at doses equal to or greater than
`0.5 µg/kg were mild to moderate chills, diaphoresis, anorexia,
`headaches, transient cough, and nausea and vomiting. There
`were no treatment-related deaths, and none of the patients re(cid:173)
`quired intensive care unit support.
`With the fixed-dose regimen, the 0.1 µg/kg dose was well
`tolerated, and there were no grade 3 toxicities (except fever,
`which was not dose-limiting). One of three patients treated with
`0.1 µg/kg rHuIL-12 had a grade 2 increase in serum transami-
`
`Downloaded from
`
`on February 22, 2018. © 1998 American Association for Cancerclincancerres.aacrjournals.org
`
`
`Research.
`
`NOVARTIS EXHIBIT 2039
`Breckenridge v. Novartis, IPR 2017-01592
`Page 3 of 10
`
`

`

`1186 IL-12 in Renal Cell Carcinoma
`
`Table 3 Number of patients experiencing selected adverse events in cycle I, day 1, versus cycle 2, day 15 (grades 2, 3, or 4), from fixed dosing
`scheme A
`
`0.1 µ.g/kg (n = 3)
`
`0.5 µ.g/kg (n = 15)
`
`1.0 µ.g/kg (n = 6)
`
`Adverse event
`Fever
`WBC
`ASAT
`ALAT
`
`Cycle I,
`day I
`(2,0)
`(0, 0)
`(1. 0)
`(1,0)
`
`Cycle 2,
`day 15
`(0, 0)
`(0, 0)
`(0, 0)
`(0, 0)
`
`Cycle I,
`day 1
`(8. 2)
`(3, 1)
`(0, 0)
`(1,0)
`
`Cycle 2,
`day 15
`(1,0)
`(1. 0)
`(0,0)
`(0, 0)
`
`Cycle I,
`day 1
`(4,0)
`(2, 1)
`(1. 1)
`(0, 1)
`
`Cycle 2.
`day 15
`(1,0)
`(0,0)
`(1. 0)
`(0, 0)
`
`4IO r-------------------------------------
`
`-3IO
`
`180
`
`100
`
`· · · · · · ·
`
`· · · -&rade~ · · · · · · · · · · · · · · · · · · · · ·
`
`IO
`
`..... ·Grac1e1· .......................................•.................................
`
`Grade
`a
`u u
`
`C
`
`Q
`u
`
`0
`
`ID
`
`I
`
`"' I N I .,. !
`
`Q
`u
`
`Q
`~
`
`u
`Q
`u
`Fig. I Changes in ALAT levels following fixed doses of weekly s.c. administration of rHuIL-12 in a patient (patient 103) at 1.0 µ.g/kg dose.
`Increases in transaminase levels were highest after the first dose and decreased on subsequent dosing.
`
`nase concentration in cycle I, but the patient met grade I
`toxicity pretreatment.
`Fifteen patients were treated with a fixed-dose schedule of
`0.5 µg/kg. All reported a mild to moderate fever within 36 h of
`the first injection, and all had mild to moderate increases in
`serum transaminase concentrations after the first injection. One
`patient had grade 4 gastrointestinal toxicity. This patient had a
`distant history of recurrent colitis and a family history of ulcer(cid:173)
`ative colitis undisclosed at study entry. He developed bloody
`diarrhea during the second cycle, and a colonoscopy showed
`pancolitis. The patient was taken off the study and improved
`with medical management that included steroids.
`Six patients were treated with a fixed-dose schedule of 1.0
`µg/kg; two experienced dose-limiting toxicity composed of a
`grade 3 increase in transaminase concentration plus grade 4
`pulmonary toxicity in one patient and a grade 3 leukopenia in
`
`one patient. The pulmonary toxicity was characterized by acute
`onset of shortness of breath in the setting of fever approximately
`40 h following day I of cycle I treatment during hospital stay.
`With supplemental oxygen, the patient recovered immediately
`without sequelae. Evaluation included a chest radiograph, elec(cid:173)
`trocardiogram, and ventilation/perfusion scan, which failed to
`reveal an etiology. The patient weighed 112 kg, and the rela(cid:173)
`tively high dose of 112 µg of rHuIL-12 may have been a factor;
`subsequent patients treated on the study were dosed according to
`a maximum 80 kg weight. At the 1.0 µg/kg dose treatment,
`fever and leukopenia were more persistent with slower recovery.
`Nearly all patients treated with 1.0 µg/kg had an increase in
`transaminase levels 5-10 days after the first injection, but only
`one patient had grade 3 toxicity.
`A comparison of the severity of fever, leukopenia, and
`elevated serum transaminase concentration according to dose in
`
`Downloaded from
`
`on February 22, 2018. © 1998 American Association for Cancerclincancerres.aacrjournals.org
`
`
`Research.
`
`NOVARTIS EXHIBIT 2039
`Breckenridge v. Novartis, IPR 2017-01592
`Page 4 of 10
`
`

`

`Clinical Cancer Research 1187
`
`Table4 Number of patients experiencing selected adverse events in cycle 1 versus cycle 2 (grades 2, 3, or 4) from up-titration dosing scheme B
`0.5 µ.g/kg (n = 3)
`0.75 µ.g/kg (n = 3)
`1.0 µ.g/kg (n = 3)
`1.25 µ.g/kg (n = 6)
`1.5 µ.g/kg (n = 12)
`
`Adverse event
`Fever
`WBC
`ASAT
`ALAT
`
`Cycle 1,
`day 15
`(0,0)
`(0, 0)
`(0, 0)
`(I, 0)
`
`Cycle 2,
`day 15
`(0, 0)
`(1,0)
`(0, 0)
`(0, 0)
`
`Cycle I,
`day 15
`(I, 0)
`(I, 0)
`(0,0)
`(I, 0)
`
`Cycle 2,
`day 15
`(0, 0)
`(1,0)
`(0, 0)
`(0, 0)
`
`Cycle I,
`day 15
`(2, 0)
`(2, 0)
`(0, 0)
`(0, 0)
`
`Cycle 2.
`day 15
`(I. 0)
`(1,0)
`(I, 0)
`(0, 0)
`
`Cycle 1,
`day 15
`(2,0)
`(2, 0)
`(0,0)
`(0,0)
`
`Cycle 2.
`day 15
`(I. 0)
`(3, 0)
`(0,0)
`(0,0)
`
`Cycle I.
`day 15
`(3,0)
`(4,0)
`(0, 1)
`(0, I)
`
`Cycle 2,
`day 15
`(0,0)
`(4, 0)
`(0, I)
`(I, 1)
`
`100 0 . - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - - -
`
`900
`
`800
`
`700
`
`300
`
`200
`
`100
`
`..............
`..
`..
`...
`0 .,
`.,
`.,
`.,
`.,
`.,
`.,
`.,
`....
`.,
`.,
`.,
`"'
`N
`"'
`.5
`C
`0
`Ei
`Ei
`~ C
`Ei
`Ei
`ai u u C u
`0
`u e ~ ~ e
`0
`c5
`u u ..... u u u u
`.....
`C
`~
`"'
`ca
`a,
`"'
`Ei
`Ei
`..,
`Ill
`C
`Ei
`C
`C
`Ei
`u
`c5
`c5
`l3
`u
`u
`l3
`Fig. 2 Changes in ALA T levels following slow intrapatient dose escalation of rHuIL-12 in a patient (patient 120) at a 1.5 µ.g/kg maintenance dose.
`Grade 3 increases occurred after 4 weeks of treatment and decreased on subsequent dosing at 50% level.
`
`0,
`C
`N
`
`.,
`"'
`0
`C
`.....
`a,
`C
`N
`u
`
`0
`
`0
`
`0
`
`0
`
`N
`C
`
`0,
`C
`
`0
`
`N
`
`0,
`
`N
`N
`C
`
`N
`C
`N
`u
`
`.,
`.,
`0
`C
`.....
`Ei
`N
`u
`
`cycle 1 versus cycle 2 suggested a dose-effect relationship
`(Table 3). At all dose levels, the most severe toxicities occurred
`mainly after the first injection and were milder upon further
`treatment with rHuIL-12 (Fig. 1). Most patients showed only
`mild (grade 1) fever and other common adverse events (leuko(cid:173)
`penia and elevated ALA T and/or ASA T) in cycle 2 at all doses.
`Therefore, the maximum tolerated dose was reached at 1.0
`µg/kg with the first dose. Because tolerability improved with
`subsequent therapy, a slow escalation dosing scheme was fol(cid:173)
`lowed for a second cohort of patients.
`In the second dose scheme, the dose of rHuIL-12 was
`up-titrated in two steps for each patient following day 1 and day
`8 treatments, and patients were treated with the maintenance
`dose ranging from 0.5 to 1.5 µg/kg from day 15 of cycle 1 until
`they were taken off study or required a dose reduction. No
`dose-limiting toxicity was observed in patients treated at target
`maintenance dose levels of 1.25 µg/kg or less. The maximum
`
`tolerated dose was 1.5 µg/kg. At this dose level, two of the first
`six patients had dose-limiting toxicity. The one patient had
`grade 4 serum transaminase (ALAT) concentrations during
`week 2 of cycle 2 at the maintenance dose of 1.5 µg/kg. A
`second patient had grade 3 serum transaminase elevations fol(cid:173)
`lowing the first injection of the 1.5 µg/kg dose during cycle 1.
`An additional six patients were treated at this dose level without
`any dose-limiting toxicity.
`The severity of changes in serum transaminase and leuko(cid:173)
`cyte concentrations, as well as fever, was compared between
`cycle 1 and cycle 2 following escalation doses (Table 4 ). In
`contrast to patients treated with the fixed dosing scheme, the
`slow escalation of rHuIL-12 was better tolerated, although sim(cid:173)
`ilar adverse events occurred at later times ( cycle 2) and at higher
`doses (Fig. 2).
`An additional toxicity observed, albeit not dose-limiting,
`was stomatitis. Grade I mucositis was first noted in two patients
`
`Downloaded from
`
`on February 22, 2018. © 1998 American Association for Cancerclincancerres.aacrjournals.org
`
`
`Research.
`
`NOVARTIS EXHIBIT 2039
`Breckenridge v. Novartis, IPR 2017-01592
`Page 5 of 10
`
`

`

`1188 IL-12 in Renal Cell Carcinoma
`
`1000
`
`900
`
`800
`
`700
`
`--+-1.0 ug/kg; Fixed C1D1
`--0.5 ug/kg; Esc C1D15
`--1.0 ug/kg; EscC1D15
`--M--1.25 ug/kg; EscC1D15
`---1.50 ug/kg; Esc C1D15
`
`C, 600
`E
`~
`8 500
`_,
`:i t
`
`N
`
`400
`
`Fig. 3 Mean serum concentration pro(cid:173)
`files of rHuIL-12 after the first dose of
`1.0 µ.g/kg in the fixed dose scheme com(cid:173)
`pared to the first maintenance dose of
`0.5-1.5 µ.g/kg in the intrapatient escala(cid:173)
`tion dose scheme.
`
`300
`
`200
`
`100
`
`0
`
`0
`
`2
`
`4
`
`8
`
`10
`
`16
`Time (hr)
`
`24
`
`30
`
`34
`
`48
`
`Table 5 Mean (:!:SE) pharmacokinetic parameters of rHuIL-12 at various doses (cycle I, day 1, for fixed dose and cycle I, day 15, for
`escalation doses)
`
`Fixed Dose
`
`Escalation Dose
`
`0.5 µ.g/kg
`(n = 15)
`320 (70)
`13
`7031 (2031)
`13 (2)
`
`Cmax (pg/ml)
`T max (h)
`AUC" (pg-h/ml)
`l112 (h)
`"AUC, area under the curve.
`h Adequate sample in one patient.
`
`1.0 µ.g/kg
`(n = 6)
`1092 (275)
`16
`26589 (6633)
`10 (2)
`
`0.5 µ.g/kg
`(n = 3)
`70 (35)
`16
`454 (237)
`9b
`
`1.0 µ.g/kg
`(n = 3)
`353 (79)
`14
`7631 (2745)
`15 (7)
`
`1.25 µ.g/kg
`(n = 6)
`383 (53)
`15
`9109 (1705)
`16 (3)
`
`1.5 µ.g/kg
`(n = 12)
`706 (159)
`15
`14269 (3145)
`12 (I)
`
`treated at the 1.0 µg/kg dose level in the fixed-dose scheme and
`one patient treated at the 1.0 µg/kg dose level in the up-titration
`dose scheme. When higher doses of rHuIL-12 were given in the
`up-titration dose scheme, the severity increased and reached
`grade 2 toxicity in two patients treated at 1.25 µg/kg and two
`patients treated at 1.5 µg/kg.
`Phannacokinetics/Phannacodynamics. Serum concen(cid:173)
`trations of rHuIL-12 are shown in Fig. 3 at various doses after
`the first dose (day l) in fixed dose scheme A or after the first
`maintenance dose (day 15) in escalation dose scheme B. Serum
`concentration of IL-12 increased slowly after s.c. administra(cid:173)
`tion, with peak serum concentration observed between 8 and
`24 h. Serum concentration then decreased gradually with a
`half-life ranging from 7 to 21 h. Mean pharmacokinetic param(cid:173)
`eters of IL-12 are described in Table 5 for both dose schemes.
`The serum concentrations of IL-12 for equivalent target doses
`were lower after escalation dose scheme B compared to fixed
`
`dose scheme A, although the peak time and half-life were
`unchanged.
`After treatment for as long as 8 months, no serum antibod(cid:173)
`ies were identified against rHuIL-12. Serum levels of anti(cid:173)
`rHulL-12 antibody were below the detection limit (29 ng/ml) in
`all patients.
`Three different immunological markers (IFN--y, neopterin,
`and tumor necrosis factor-a) were followed to investigate THI
`immune stimulation by rHuIL-12. Serum levels of IFN--y were
`nonmeasurable (<3.0 pg/ml) at baseline. The mean peak level
`achieved in patients treated with 1.0 µg/kg by the fixed dose
`schedule was 250 pg/ml (Fig. 4). Similar to the levels of serum
`IL-12, IFN--y increased but to a lesser extent when dosing
`scheme was changed from fixed to the up-titration schedule. The
`peak level of IFN--y was 126 pg/ml at 1.0 µg/kg dose in the
`escalation dose scheme. The levels, however, increased as the
`dose was increased within the escalation scheme to 205 pg/ml at
`
`Downloaded from
`
`on February 22, 2018. © 1998 American Association for Cancerclincancerres.aacrjournals.org
`
`
`Research.
`
`NOVARTIS EXHIBIT 2039
`Breckenridge v. Novartis, IPR 2017-01592
`Page 6 of 10
`
`

`

`Clinical Cancer Research 1189
`
`250 ...... - - - - - - - - - - - - - - - - - - - - - - - - - - - .
`
`--1.0 ugkg; Fixed C101
`...... o.5 ug.'llg; Eec: C1015
`-+-1.0 ug.'llg; Eec: C1D15
`--125 ug.'llg; Eec: C1015
`-+-1.50 ug.'llg; Eec: C1D15
`
`Fig. 4 Mean serum concentration profiles
`of IFN--y after the first dose of rHuIL-12 at
`1.0 µ.g/kg in the fixed dose scheme com(cid:173)
`pared to the first maintenance dose of
`rHuIL-12 at 0.5-1.5 µ.g/kg in the intrapa(cid:173)
`tient escalation dose scheme.
`
`200
`
`50
`
`0F-----.,:::.----+------+------+--------1
`72
`0
`24
`48
`10
`98
`
`Time lhr)
`
`--1.0 ugkg; Fixed C101
`...... o.5 ug.'llg; Elc C1D15
`-+-1.0 ug/kg; Elc C1015
`--1.25 ug/kg; Elc C1015
`-+-1.50 ug/kg; Elc C1D15
`
`Fig. 5 Mean serum concentration profiles of
`neopterin after the first dose of rHuIL-12 at
`1.0 µ.g/kg in the fixed dose scheme compared
`to first maintenance dose of rHuIL-12 at 0.5-
`1.5 µ.g/kg in the intrapatient escalation dose
`scheme.
`
`IO
`
`50
`
`20
`
`o----------+---------+----------1
`0
`10
`24
`48
`111
`72
`98
`Tlmelhr)
`
`1.5 µg/kg dose, demonstrating a dose-related increase in T8 1
`stimulation. The onset of such stimulation was relatively rapid,
`with peak time for IFN--y occurring at about 24 h after IL-12
`administration. The levels then decreased during subsequent
`days of the cycle to nonmeasurable levels by day 8 of next dose.
`
`Neopterin serum concentration increased more slowly than
`IFN--y, with peak concentration reached between 72 and 96 h
`following treatment with rHuIL-12. The predose baseline neop(cid:173)
`terin concentration ranged from 1.2 to 8 ng/ml. The mean ± SE
`peak neopterin concentration after the first dose of 1.0 µg/kg
`
`Downloaded from
`
`on February 22, 2018. © 1998 American Association for Cancerclincancerres.aacrjournals.org
`
`
`Research.
`
`NOVARTIS EXHIBIT 2039
`Breckenridge v. Novartis, IPR 2017-01592
`Page 7 of 10
`
`

`

`1190 IL-12 in Renal Cell Carcinoma
`
`was 70 ± 25 ng/ml. For patients treated with the up-titration
`schedule (scheme B), the mean ± SE peak neopterin levels after
`the first maintenance (week 3) dose were 15.4 ± 2.7, 19.5 ±
`7.9, 35.0 ± 11.2, and 31.0 ± 5.0 ng/ml at 0.5, 1.0, 1.25, and 1.5
`µg/kg doses, respectively (Fig. 5). The peak concentration oc(cid:173)
`curred at about 96 h and maintained at higher than predose
`levels prior to administration of the next dose (168 h). Few
`sporadic levels of tumor necrosis factor-a were detected during
`the treatment, but no consistent increase in levels was observed
`in relation to severity of the measured adverse events.
`Response and Survival. Fifty patients were evaluable
`for response. One patient, who was considered inevaluable,
`received day I of treatment and was removed from study be(cid:173)
`cause of pulmonary toxicity. Two patients discontinued treat(cid:173)
`ment because of progression following one month of therapy. At
`the 2-month tumor assessment following two cycles of therapy,
`the response was as follows: stable, 34 patients; progressive, 14
`patients. One patient with a response of stable disease following
`the first two cycles had tumor regression and subsequently
`achieved a complete response. The extent of disease in this
`patient was confined to lung parenchyma, and he was treated
`with 1.5 µg/kg of rHuIL-12 according to the up-titration sched(cid:173)
`ule. This patient continues on treatment following 12 cycles of
`therapy and remains progression-free at > 12 months. For the
`remaining 34 patients with a best response of stable, the median
`time to progression was 4 months (range, 2-15 months).
`
`DISCUSSION
`Common adverse events observed in this study were fever,
`chills, fatigue, transient leukopenia, and increase in serum con(cid:173)
`centrations of hepatic transaminase. Other toxicities included
`stomatitis, respiratory distress in the setting of fever, and colitis.
`The colitis may have represented reactivation of an autoimmune
`disorder; rHuIL-12 facilitated the development of autoimmune
`disease in several murine models (

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