throbber
1678 Vol. 6, 1678 –1692, May 2000
`
`Clinical Cancer Research
`
`Phase I Trial of Twice-Weekly Intravenous Interleukin 12 in
`Patients with Metastatic Renal Cell Cancer or Malignant
`Melanoma: Ability to Maintain IFN-g Induction Is
`Associated with Clinical Response1
`
`Jared A. Gollob,2 James W. Mier,
`Korina Veenstra, David F. McDermott,
`Daniel Clancy, Marguerite Clancy, and
`Michael B. Atkins
`Beth Israel Deaconess Medical Center, Division of
`Hematology/Oncology, Boston, Massachusetts 02215
`
`ABSTRACT
`The aim of this study was to examine the tolerability,
`antitumor activity, and biological effects of a new schedule
`of i.v. recombinant human interleukin 12 (rhIL-12). Twenty-
`eight patients were enrolled in a Phase I trial in which
`rhIL-12 was administered twice weekly as an i.v. bolus for 6
`weeks. Stable or responding patients were eligible to receive
`additional 6-week cycles until there was no evidence of
`disease or until tumor progression. Patient cohorts were
`treated with escalating doses of rhIL-12 (30 –700 ng/kg). The
`maximum tolerated dose (MTD) was 500 ng/kg, with dose-
`limiting toxicities consisting of elevated hepatic transami-
`nases and cytopenias. At the MTD (n 5 14), there was one
`partial response occurring after 6 cycles of rhIL-12 in a
`patient with renal cell cancer. Two additional renal cell
`cancer patients treated at the MTD had prolonged disease
`stabilization, with one of these exhibiting tumor regression
`after 8 cycles of rhIL-12. IFN-g, IL-15, and IL-18 were
`induced in patients treated with rhIL-12. Whereas IFN-g
`and IL-15 induction were attenuated midway through the
`first cycle in patients with disease progression, those patients
`with tumor regression or prolonged disease stabilization
`were able to maintain IFN-g, IL-15, and IL-18 induction.
`The down-modulation of IFN-g induction during rhIL-12
`treatment did not relate to IL-10 production or alterations
`in rhIL-12 bioavailability but was associated with an ac-
`quired defect in lymphocyte IFN-g production in response
`to IL-12, IL-2, or IL-15. This defect could be partially
`
`Received 12/20/99; revised 2/18/00; accepted 2/18/00.
`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.
`1 Supported in part by NIH Grants CA78055 and CA74401 as well as by
`a stipend from Genetics Institute, Inc.
`2 To whom requests for reprints should be addressed, at Division of
`Hematology/Oncology, Beth Israel Deaconess Medical Center, 330
`Brookline Avenue, East Campus/Room KS-158, Boston, MA 02215.
`Phone: (617) 667-1930; Fax: (617) 975-8030.
`
`overcome in vitro through combined stimulation with IL-12
`plus IL-2. These findings show that the chronic administra-
`tion of twice-weekly i.v. rhIL-12 is well-tolerated, stimulates
`the production of IL-12 costimulatory cytokines and IFN-g,
`and can induce delayed tumor regression. Strategies aimed
`at maintaining IFN-g induction, such as the addition of IL-2,
`may further augment the response rate to this schedule of
`rhIL-12.
`
`INTRODUCTION
`IL3-12 is a cytokine with considerable promise for the
`treatment of human malignancies because of its pleiotropic
`immunostimulatory effects on lymphocytes (1–5), dendritic
`cells (6), and neutrophils (7– 8), as well as its potent antitumor
`activity in murine tumor models (9 –10). Whereas immune ac-
`tivation by IL-12 in mice has resulted in both tumor necrosis
`factor and NO production, the antitumor effect of IL-12 has
`been more dependent on IFN-g production (10) and the activa-
`tion of either CD81 T cells (9 –11) or NKT cells (12). There
`seem to be a number of mechanisms through which IL-12 can
`induce tumor regression, including the direct killing of tumor
`cells by activated lymphocytes, the antiangiogenic effects of
`IL-12-induced IFN-g (13), and injury both to the tumor micro-
`circulation and to the tumor itself by activated neutrophils (11).
`The immunomodulatory activity of IL-12 is considerably
`dependent on costimulatory cytokines. When the ability of
`IL-12 to activate unmanipulated peripheral blood NK cells and
`CD81 T cells in humans was examined, it was found that these
`lymphocyte subsets responded to IL-12 only when stimulated
`together with IL-2 (14). Both IL-15 and IL-18 are also key
`costimulatory cytokines, which, when combined with IL-12,
`induce strong IFN-g production by T and NK cells (15, 16). In
`mice treated with IL-12, the neutralization of endogenous IL-18
`significantly blunts IFN-g production (17), further emphasizing
`the fact that the biological activity of IL-12 in vivo is likely
`dependent on the presence and/or induction of endogenous
`costimulatory cytokines.
`The promising preclinical data showing IL-12 to be highly
`effective against murine melanoma, renal cell cancer, and sar-
`coma led to its testing in clinical trials in cancer patients starting
`in 1994. In the first published trial, rhIL-12 was administered
`i.v. daily for 5 days, with a 2-week break between cycles. In
`
`3 The abbreviations used are: IL, interleukin; rh, recombinant human;
`NO, nitric oxide; DLT, dose-limiting toxicity; MTD, maximal tolerated
`dose; PR, partial response; PBMC, peripheral blood mononuclear cell;
`CT, computed tomography; NK, natural killer; ppb, parts per billion.
`
`NOVARTIS EXHIBIT 2038
`Breckenridge v. Novartis, IPR 2017-01592
`Page 1 of 15
`
`

`

`Clinical Cancer Research
`
`1679
`
`addition, a single test dose was given 2 weeks before the first
`cycle. With that dosing schedule, the MTD was 500 ng/kg, with
`DLTs consisting of liver function test abnormalities and stoma-
`titis (18). Although signs of immune activation were observed,
`including dose-dependent IFN-g production and reversible de-
`creases in CD81 T cell and NK cell numbers (19), only two
`responses were seen among 40 patients (one PR in a patient with
`renal cell cancer and one transient complete response (CR) in a
`patient with melanoma). Similarly low response rates were
`observed in two subsequent trials of weekly s.c. rhIL-12 in
`melanoma (20) and renal cell cancer (21), as well as in a trial
`testing a thrice weekly schedule of s.c. rhIL-12 (22).
`In patients treated with either i.v. or s.c. rhIL-12, IFN-g
`production induced in vivo by rhIL-12 has attenuated rapidly
`with consecutive cycles (18, 20 –22), which indicates that the
`biological response to rhIL-12 is down-modulated during ther-
`apy. Even a single test dose administered 2 weeks before the
`first cycle of rhIL-12 seemed to attenuate IL-12-induced IFN-g
`production (23). This down-modulation of IFN-g production has
`been shown to result in diminished IL-12-induced tumor regres-
`sion in mice (24). In addition, multiple doses of IL-12 have also
`been shown in animals to induce a temporary state of immuno-
`suppression (25–26), perhaps analogous to the down-modula-
`tion of IFN-g production in patients receiving multiple doses of
`rhIL-12. This paradoxical immunosuppression after a relatively
`brief period of immune activation by rhIL-12 may explain the
`limited antitumor activity observed to date in rhIL-12 clinical
`trials. Although the mechanism of this IL-12-induced down-
`modulation of subsequent IFN-g induction remains undefined,
`data from animal models have suggested that IL-12-induced NO
`may be operative (26), whereas observations from clinical trials
`have also implicated changes in rhIL-12 pharmacokinetics
`(20, 22).
`In June of 1998, we initiated a Phase I dose escalation trial
`of i.v. rhIL-12 in patients with renal cell cancer and melanoma,
`using a new dosing schedule. To try to prevent or delay the
`dampening of IFN-g induction, we eliminated the test dose. In
`addition, we implemented a twice-weekly dosing schedule to
`determine whether moderate and sustained IFN-g production
`could be stimulated without prohibitive toxicity. Although im-
`portant aims of this trial included determining the safety and
`tolerability as well as the antitumor activity of this regimen, this
`study was also undertaken to further explore the mechanism
`through which rhIL-12 activates the immune system in vivo and
`to examine how IFN-g induction by rhIL-12 is modulated with
`chronic dosing.
`
`PATIENTS AND METHODS
`Patient Selection. All of the patients were adults with
`histologically proven advanced malignancy that was metastatic
`or unresectable and for which standard curative or palliative
`measures did not exist or were no longer effective. All of the
`patients had measurable or evaluable disease that was clearly
`progressive. Patients were required to have an Eastern Cooper-
`ative Oncology Group (ECOG) performance status of 0 or 1 and
`adequate organ function defined by WBC .4000/ml, platelet
`count .100,000/ml, creatinine ,1.5 mg/dl, bilirubin ,1.5 mg/
`dl, aspartate aminotransferase ,2 times the upper limit of nor-
`
`mal, and electrocardiogram and chest X-ray without clinically
`significant nonmalignant abnormalities. Patients with brain me-
`tastases, seizure disorders, organ allografts, concurrent require-
`ment for corticosteroids, more than two prior chemotherapy
`regimens, more than two prior immunotherapy regimens, or
`prior IL-12 therapy were ineligible.
`Study Design. The study was an open-label, nonrandom-
`ized, single-center Phase I dose escalation trial. The treatment
`protocol was approved by the Cancer Therapy Evaluation Pro-
`gram (CTEP) of the National Cancer Institute (protocol T97-
`0053) and by the Human Institutional Review Board at the Beth
`Israel Deaconess Medical Center (protocol 97-1083), and writ-
`ten informed consent was obtained from each patient. rhIL-12,
`produced by Genetics Institute, Inc. (Cambridge, MA), was
`supplied by the National Cancer Institute (IND 6798). The
`rhIL-12 was administered by i.v. bolus injection.
`The treatment schedule is shown in Fig. 1. Patients were
`treated in the General Clinical Research Center at the Beth Israel
`Deaconess Medical Center, and received i.v. bolus injections of
`rhIL-12 twice weekly, with doses given 3– 4 days apart. A cycle
`of therapy lasted 6 weeks, with patients receiving a total of 12
`doses during that period. During the first cycle only, patients
`were admitted overnight after the first, second, and seventh
`doses of rhIL-12 for observation and serial blood draws. All of
`the remaining doses were administered on an outpatient basis,
`with patients observed for 1 h after each dose. Patients were
`evaluated for tumor response at the end of each 6-week cycle,
`and patients with stable or regressing disease could continue
`receiving additional cycles until there was no evidence of dis-
`ease or until there was disease progression. Patients were al-
`lowed up to a 2-week break between cycles for the resolution of
`any significant rhIL-12-induced toxicity.
`The rhIL-12 dose was increased from 30 to 700 ng/kg in
`successive cohorts of patients. No intrapatient dose escalation
`was permitted. A minimum of three patients were enrolled at
`each dose level, and all of the patients had to have completed the
`first 3 weeks of cycle 1 before initiating enrollment to the next
`dose level. Toxicity was assessed using the National Cancer
`Institute Common Toxicity Criteria. In general, grade 3 or
`greater toxicities were considered dose-limiting. However, liver
`function test abnormalities were not classified as dose-limiting
`until the total bilirubin was .3 times normal or the hepatic
`transaminases or alkaline phosphatase were .10 times normal.
`In addition, the WBC count and neutrophil count were not
`considered dose-limiting until criteria for grade 4 toxicity were
`met, and no degree of lymphopenia was dose-limiting. Grade 2
`cardiovascular toxicity (except for hypotension) and neurolog-
`ical toxicity were considered dose-limiting. The IL-12 dose was
`escalated when 0 of 3 patients at a dose level had a DLT. If 1 of
`3 experienced a DLT, three more patients were enrolled at that
`dose level, and the dose was escalated if no more than 1 of 6
`patients had a DLT. Patients experiencing a DLT could resume
`the IL-12 at the next lowest dose level if the toxicity resolved
`within 2 weeks. When two or more DLTs were experienced at
`a dose level, the MTD was determined to be the next previous
`dose level.
`All of the patients received ranitidine for the duration of
`their IL-12 treatment. Acetaminophen was administered prophy-
`lactically for 24 h after each IL-12 dose and could be taken as
`
`NOVARTIS EXHIBIT 2038
`Breckenridge v. Novartis, IPR 2017-01592
`Page 2 of 15
`
`

`

`1680 Phase I Trial of Twice-Weekly i.v. rhIL-12
`
`X
`
`X
`
`l l l l
`
`Week
`
`1
`
`2
`
`X
`
`l l
`
`3
`
`I
`
`4
`
`l l
`
`5
`
`6
`
`= IL-12 dose administered by IV bolus
`twice weekly on days 1 and 4
`
`trial of
`Fig. 1 Schema for clinical
`twice-weekly i.v. rhIL-12 without a
`test dose.
`
`X = Overnight hospitalizations for serial q4hr blood
`draws to collect serum for measurement of cytokines
`induced following IL-12 injection
`
`•IL-12 dose levels: 30,100,300, 500, 700 ng/kg
`•3-6 patients per dose cohort
`•Tumor response assessed after each 6-week cycle
`•Patients can continue to recehe 6-week cycles until
`no evidence of di,ease or until disease progression
`
`needed thereafter. Indomethacin was used to control fever that
`was not responsive to acetaminophen, and demerol was used to
`treat rigors.
`Assessment of Tumor Response. Tumor measurements
`were obtained by CT scan at the end of each 6-week cycle
`of IL-12.
`Measurement of IL-12- and rhIL-12-Induced Cyto-
`kines. Serial blood specimens were collected in heparinized
`tubes immediately before and 4, 8, 12, 16, 20, and 24 h after the
`first, second, and seventh rhIL-12 doses during the first cycle.
`The tubes were centrifuged immediately after collection, and the
`plasma was then removed and stored at 220°C. Plasma IL-12
`levels were measured using an ELISA that detects only the p70
`IL-12 heterodimer (Endogen, Cambridge, MA, sensitivity ,3
`pg/ml). ELISA kits were also used to measure plasma IFN-g
`(Endogen, sensitivity ,2 pg/ml), IL-10 (Endogen, sensitivity
`,3 pg/ml), IL-15 (R&D, Minneapolis, MN, sensitivity ,1
`pg/ml), and IL-18 (R&D, sensitivity ,15 pg/ml).
`In Vitro Assays of Lymphocyte Cytokine Responsive-
`ness. Blood specimens were collected in heparinized tubes
`immediately before the first and seventh doses of rhIL-12
`during cycle 1. PBMCs were isolated from blood samples
`through density gradient centrifugation using Histopaque-
`1077 (Sigma, St. Louis, MO). PBMCs were incubated in
`96-well U-bottomed plates at 5 3 104 cells/well with medium
`alone (RPMI 1640 plus 15% FCS, 2% L-glutamine, 1%
`sodium pyruvate, 1% gentamicin, and 1% penicillin-strepto-
`mycin) or with medium plus one of the following: (a)
`50 ng/ml IL-2 (Chiron Corporation, Emeryville, CA, specific
`activity 18 3 106 units/mg); (b) 1 nM IL-12 (Genetics Insti-
`tute, Cambridge, MA, specific activity 1.7 3 107 units/mg);
`(c) 10 ng/ml IL-15 (Endogen, specific activity $2 3 106
`units/mg); (d) IL-2 1 IL-12; or (e) IL-15 1 IL-12. Condi-
`tions were plated in triplicate, and after a 72-h incubation at
`37°C, aliquots of supernatants from each well were harvested
`
`immediately before pulsing each well with 1 mCi [3H]thymi-
`dine (DuPont-New England Nuclear, Boston, MA). The
`IFN-g concentration in the harvested supernatants was as-
`sayed using an IFN-g ELISA (Endogen). Cell proliferation
`was determined by measuring [3H]thymidine incorporation
`8 h after pulsing, as described previously (27).
`Measurement of NO in Expired Air. The concentration
`of NO in expired air was measured in five patients receiving
`IL-12 at either the 500-ng/kg or the 700-ng/kg dose levels.
`Measurements were made immediately before and 24 h after the
`first and second IL-12 doses. Expired air was collected in
`self-sealing balloons after first clearing the upper airway of
`ambient air NO by having patients take four deep inspirations
`through a tube fitted with a charcoal filter (Omega Engineering
`Co.). The NO concentration in the air expired after the fourth
`breath was measured using a high-sensitivity NO detector based
`on a gas-phase chemiluminescent reaction between NO and
`ozone (Model 280 Nitric Oxide Analyzer, Sievers Instruments,
`Inc., Boulder, CO). Patients receiving high-dose IL-2 (600,000
`IU/kg i.v. every 8 h) were used as positive controls. IL-2
`patients had NO samples obtained before the start of the 1st
`week of IL-2 and then daily for the 1st 3 days of IL-2 treatment.
`
`RESULTS
`Patient Characteristics
`Between June 1998 and June 1999, 28 patients were en-
`rolled in this study. Patient characteristics are shown in Table 1.
`The majority of patients had metastases to two or more sites
`(including 15 of 28 with liver, adrenal, and/or kidney involve-
`ment and 10 of 28 with bone metastases), and 23 of 28 had
`received one or more prior immunotherapy regimens (primarily
`IL-2-based regimens). Only 3 of 23 patients had responded to
`their prior immunotherapy.
`
`NOVARTIS EXHIBIT 2038
`Breckenridge v. Novartis, IPR 2017-01592
`Page 3 of 15
`
`

`

`Table 1 Patient characteristics
`
`Total patients
`Median age (range)
`Gender (male/female)
`Performance status
`ECOGa 0
`ECOG 1
`Tumor types
`Renal cell cancer
`Melanoma
`Prior therapy
`Chemo/Immunotherapy
`High-dose IL-2
`Low-dose IL-2
`IFN a-2b
`Biochemotherapy
`Chemotherapy
`Surgery
`Radiotherapy
`None
`Prior systemic treatment regimens
`0
`1
`2
`3
`Prior response to chemo/immunotherapy
`Disease sites
`Lung
`Lymph nodes
`Skin or soft tissue
`Liver
`Bone
`Adrenal/kidney
`Number of disease sites
`1
`2
`$3
`a ECOG, Eastern Cooperative Oncology Group.
`
`No. of patients
`28
`56 yr (32–72 yr)
`20/8
`
`15
`13
`
`20
`8
`
`24
`14
`6
`12
`3
`1
`20
`13
`1
`
`4
`12
`10
`2
`3
`
`21
`13
`5
`9
`10
`6
`
`6
`9
`13
`
`Treatment Administered and Toxicity
`Dose Escalation Phase. Three patients were treated at
`each of the first three rhIL-12 dose levels (30, 100, and 300
`ng/kg), with no DLTs. Common side effects included self-
`limited fever and chills, occurring 6 –10 h after the dose, and
`mild malaise. These side effects were observed even at the
`30-ng/kg dose level, and were greatly attenuated by the start of
`the 3rd week of therapy. One patient, treated at 100 ng/kg,
`developed a supraventricular tachycardia (Table 2) in the setting
`of fever after the second rhIL-12 dose, which resolved sponta-
`neously. Small, minimally symptomatic, transient oral aphthous
`ulcers developed during the first cycle of therapy in one patient
`at the 300-ng/kg dose level. No significant (Grade 2 or greater)
`cytopenias or liver-function test abnormalities were noted at the
`first three dose levels.
`At the 500-ng/kg dose level, the fever and chills were more
`severe with previously untreated patients or with patients for
`whom more than 1 year had passed since prior therapy. Fevers
`were highest after the second dose and were minimal-to-absent
`by the 3rd week of therapy in the majority of patients. Indo-
`methacin was added to acetaminophen for the control of fevers
`and chills in only 3 of 14 patients and was never required
`beyond the first week of therapy. Minimal nausea and anorexia
`
`Clinical Cancer Research
`
`1681
`
`were observed, but no diarrhea or gastrointestinal bleeding.
`Stomatitis was uncommon and was never greater than grade 2.
`Grade 1–2 elevations of serum transaminases were common,
`usually peaking after the second dose and normalizing by the
`start of week 3 (Table 2). Orthostatic hypotension 24 h after
`the second rhIL-12 dose occurred in one patient and constituted
`the one DLT among the six patients treated at the 500-ng/kg
`dose level during the escalation phase. No fluid retention or
`evidence of capillary leak syndrome was observed at either the
`500-ng/kg dose level or any other rhIL-12 dose level, nor was
`there any renal or pulmonary toxicity.
`A total of five patients were treated at the 700-ng/kg dose
`level. Two patients (one with melanoma and one with renal cell
`cancer) who had received high-dose IL-2 ,6 months before the
`rhIL-12 had either no fever or low-grade fevers and minimal-
`to-no liver function test abnormalities during rhIL-12 treatment.
`In contrast, the other three patients who received either high-
`dose IL-2 therapy .1 year previously or low-dose IL-2 .6
`months previously experienced higher and more sustained fe-
`vers (requiring both acetaminophen and indomethacin during
`the first 2 weeks of therapy) as well as more protracted consti-
`tutional symptoms (including malaise and anorexia). Two DLTs
`were observed among these three patients, including grade-3
`hemolytic anemia (occurring during week 5 of cycle 1) in one
`patient and a grade-3 elevation of serum hepatic transaminases
`(occurring after the second dose of rhIL-12) in another (Table
`2). The hemolytic anemia was Coombs negative and required
`both the discontinuation of the rhIL-12 and a 1-week course of
`prednisone to resolve. IL-12-induced hypersplenism leading to
`extravascular hemolysis was suspected because CT scans
`showed the development of splenomegaly after the first cycle of
`rhIL-12 (not shown). The grade 3 transaminase elevation re-
`solved within 1 week of stopping the rhIL-12.
`Safety Phase. On the basis of the two DLTs observed at
`the 700-ng/kg dose level, the MTD for the twice-weekly sched-
`ule of i.v. rhIL-12 administered without a test dose was deter-
`mined to be 500 ng/kg. To better assess the safety of the MTD,
`an additional eight patients were treated at 500 ng/kg. As shown
`in Table 3, 7 of 8 patients tolerated the rhIL-12 well without any
`DLTs. One patient tolerated cycle 1 without difficulty but then
`developed grade-4 neutropenia after the first 2 weeks of cycle 2.
`Bone marrow biopsy revealed agranulocytosis, which resolved
`after discontinuation of the IL-12 and treatment with prednisone
`plus low-dose oral cyclophosphamide.
`With the exception of the case of agranulocytosis, no
`unusual or severe toxicities occurred among patients receiving
`two or more uninterrupted 6-week cycles of rhIL-12, including
`two patients who had been on rhIL-12 for 36 and 48 weeks,
`respectively (Table 3). Several patients, including the one on
`rhIL-12 for 36 weeks, experienced grade 1–2 arthralgias (Table
`2), involving primarily the shoulders and fingers, beginning
`with the second cycle of therapy. The arthralgias were episodic,
`unaccompanied by joint swelling or tenderness, and responsive
`to therapy with nonsteroidal anti-inflammatory drugs.
`
`Biological Effects of Twice-Weekly i.v. rhIL-12
`IFN-g levels were obtained in
`In Vivo IFN-g Induction.
`eight patients treated at the 500-ng/kg dose level as well as
`in two patients enrolled at the 700-ng/kg dose level. As shown
`
`NOVARTIS EXHIBIT 2038
`Breckenridge v. Novartis, IPR 2017-01592
`Page 4 of 15
`
`

`

`1682 Phase I Trial of Twice-Weekly i.v. rhIL-12
`
`Table 2 Number of patients experiencing select toxicities during treatment with IL-12 (grades 2, 3, 4)
`
`Dose level, ng/kg (no. of patients)
`
`Toxicity
`
`30 (3)
`
`100 (3)
`
`300 (3)
`
`500 (14)
`
`Hepatic
`ASTa
`Bilirubin
`Alk phosphatase
`Hematologic
`Neutropenia
`Anemia
`Hemolytic anemia
`Thrombocytopenia
`Oral mucositis
`Cardiovascular (hypotension)
`Cardiovascular (arrhythmia)
`Fever
`Arthralgia
`a AST, aspartate aminotransferase; Alk, alkaline.
`
`(1, 0, 0)
`
`(1, 0, 0)
`(1, 0, 0)
`
`(4, 0, 0)
`(1, 0, 0)
`(1, 0, 0)
`
`(2, 1, 1)
`(2, 0, 0)
`
`(1, 0, 0)
`(2, 0, 0)
`(1, 0, 0)
`
`(2, 1, 0)
`(2, 0, 0)
`
`700 (5)
`
`(0, 1, 0)
`
`(1, 0, 0)
`
`(2, 0, 0)
`
`(0, 1, 0)
`(1, 0, 0)
`(1, 0, 0)
`
`(2, 0, 0)
`
`Table 3 Summary of tolerability of IL-12 among patients treated at the MTD of 500 ng/kg
`
`Dose escalation phase
`
`Safety phase
`
`Patient
`no.
`10
`11
`12
`
`13
`14
`15
`21
`22
`
`23
`24
`25
`26
`27
`28
`
`No. of
`cycles
`completed
`8
`3
`1
`
`6
`1
`1
`1
`2a
`
`1
`2
`1b
`1
`3
`2
`
`Results
`
`No DLT
`No DLT
`Grade 2 orthostatic hypotension; IL-12
`dose reduced to 300 ng/kg
`No DLT
`No DLT
`No DLT
`No DLT
`Grade 4 neutropenia (agranulocytosis);
`IL-12 discontinued
`No DLT
`No DLT
`No DLT
`No DLT
`No DLT
`No DLT
`
`a Received four doses in cycle 2.
`b Received only two doses in cycle 1 because of rapid disease progression.
`
`in Fig. 2 and Table 4, we were able to discern three patterns of
`IFN-g induction among these 10 patients. In all of the patterns,
`the first significant rise in plasma IFN-g occurred between 4 and
`8 h after the rhIL-12 dose, corresponding to the onset of fevers/
`chills. In the type-I pattern (Table 4 and Fig. 2A, top), the IFN-g
`level peaked at a modest 450-1600 pg/ml (with peaks occurring
`between 8 and 24 h for individual patients) after the first rhIL-12
`dose (week 1/day 1). After the second dose (week 1/day 4), peak
`levels were 2–3-fold higher than those induced by the first dose.
`However, after the seventh dose (week 4/day 1), peak IFN-g
`levels were comparable with those after the first dose. Patients
`with this type-I pattern tended to have modest fever/chills after
`each rhIL-12 dose during cycle 1, with the most prominent
`symptoms occurring after the second dose. However, whereas
`IFN-g could be detected in the plasma 24 h after an IL-12 dose,
`it always dropped to undetectable levels by the time of the next
`dose 2–3 days later (Fig. 2, A-C, top). Patients exhibiting the
`
`type-I pattern of IFN-g induction had all been treated previously
`with an IL-2-based regimen and were either .6 months past a
`low-dose IL-2 regimen or .1 year past a high-dose IL-2
`regimen.
`In the type-II pattern, peak IFN-g levels after the first dose
`were, on the average, 2-fold higher than those measured in
`patients with the type-I pattern (Table 4 and Fig. 2B, top). The
`augmentation in peak IFN-g levels after the second dose was
`also higher in the type-II pattern compared with the type-I
`pattern, increasing 2- to 4-fold over the peak levels after dose 1.
`This difference in the magnitude of IFN-g production was
`associated with higher fevers and more pronounced chills/rigors
`in these patients after the first two doses of rhIL-12 compared
`with patients exhibiting the type-I pattern of IFN-g induction.
`However, despite this larger surge of IFN-g production after the
`second dose, IFN-g induction after the seventh dose of rhIL-12
`was markedly curtailed compared with IFN-g levels after the
`
`NOVARTIS EXHIBIT 2038
`Breckenridge v. Novartis, IPR 2017-01592
`Page 5 of 15
`
`

`

`A
`
`3000
`
`2500
`
`2000
`
`150-0
`
`1000
`
`500
`
`12
`
`10
`
`B
`
`10000---·
`
`8000 -
`
`I
`
`60001
`I
`4000 ~
`
`2000
`
`__ l
`
`Wk104
`
`10
`
`15
`
`20
`
`25
`
`30
`
`(cid:157) -t \\kJlJ4
`'r j
`i. ,</ l WklDI
`/ /~ __ ------r
`Jl
`I Wk4Dl
`;,/· 1
`•------..., ------
`,,
`
`10
`
`20
`
`25
`
`30
`
`I
`
`I
`
`I
`
`12
`
`10 •
`
`' \ I !_iWk!Dl i
`-~~-·- ~!~:1
`
`:
`.
`
`.
`.
`
`I
`'
`
`j
`
`15
`
`20
`
`25
`
`/ .+:1:+1::::
`
`·•····•····•wk4DI
`
`·•··
`
`1
`
`C
`
`3000
`
`2500
`
`2000
`
`1500
`
`1000
`
`500
`
`12
`
`10
`
`Clinical Cancer Research
`
`1683
`
`10
`
`15
`
`20
`
`25
`
`30
`
`WklDI
`WklD4
`
`Wk4D1
`
`10
`
`30
`
`10
`
`15
`
`20
`
`25
`
`30
`
`10
`
`15
`
`20
`
`25
`
`30
`
`600 ~ - - - - - - - - - - - - - - -
`
`200 - - - - - - - - - - - - - - - -
`
`200 ~ - - - - - - - - - - - - - - - ,
`
`500
`
`400
`
`300
`
`200
`
`100
`
`'i Wk!D4
`.....
`- _.,-- .---!-= . · ·: · · .. · . i Wk4D1
`
`.._
`
`I
`
`WklDl
`
`175 .;
`
`150'
`
`125 ~
`
`100·
`
`75
`
`50·
`
`25·'
`
`/
`.• _./
`/~·.........-;-_
`
`,,,,,.•Wk1D4
`
`....
`
`Wk4D1
`
`WklDl
`
`150
`
`100
`
`50
`
`.,.~'--
`~
`
`_.. WklD4
`' . .,- WklDI
`.
`
`Wk4D1
`
`.
`
`10
`
`15
`
`20
`
`25
`
`30
`
`10
`
`15
`
`20
`
`25
`
`30
`
`10
`
`15
`
`20
`
`25
`
`30
`
`Time (hours post IL-12 injection)
`Fig. 2 Patterns of in vivo cytokine induction in patients during rhIL-12 therapy. Plasma concentrations of IFN-g, IL-15, and IL-18 were measured
`in 10 patients treated with rhIL-12 at the 500-ng/kg and 700-ng/kg dose levels. Cytokine concentrations were determined before and after (every 4 h
`over a 24-h period) the first (Wk1D1), second (Wk1D4), and seventh (Wk4D1) rhIL-12 injections during cycle 1. The values for IFN-g, IL-15, and
`IL-18 at each time point in A represent the mean and SD derived from four patients with the type-I pattern of IFN-g induction. In B and C, representing
`the type-II and type-III patterns of IFN-g induction, respectively, the values at each time point for IFN-g and IL-15 represent the mean and SD derived
`from three patients for each pattern, whereas the data for IL-18 were from one patient (similar results seen with two patients examined) for each
`pattern.
`
`first dose. This was associated with a greatly diminished-to-
`absent febrile response to rhIL-12 by the third week of cycle 1.
`All of the three patients with this pattern of IFN-g induction had
`not received any prior immunotherapy for their metastatic dis-
`ease.
`
`The type-III pattern was characterized by modest peak
`IFN-g levels in response to the first dose of rhIL-12, followed
`by the rapid attenuation of IFN-g production (Table 4 and Fig.
`2C, top). Whereas peak IFN-g levels reached their maximum
`after the second rhIL-12 dose in the type-I and type-II patterns,
`in the type-III pattern, peak IFN-g levels were lower after dose
`2 compared with those after dose 1 (Table 4), and they contin-
`ued to decline when measured again after the seventh dose. This
`type-III pattern was associated with a weak-to-absent febrile
`response to IL-12, and, of the three patients exhibiting this
`pattern, all had received one course or multiple courses of
`high-dose IL-2 ,6 months before starting the rhIL-12.
`
`In Vivo Induction of IL-15 and IL-18. To determine
`whether IL-15 and IL-18 were induced by rhIL-12 in cancer
`patients and to examine whether there was an association be-
`tween IL-15/IL-18 induction and IFN-g induction by rhIL-12,
`we measured the plasma levels of IL-15 and IL-18 at the same
`time points used to measure IFN-g levels. As shown in Fig. 2,
`A-C (middle), IL-15 was not detectable in the plasma before
`starting rhIL-12 but was detectable at low levels 4 h after the
`first injection. With each pattern of IFN-g induction, plasma
`IL-15 levels reached their maximum either before, or at the
`same time as, peak IFN-g levels. However, the magnitude of
`IL-15 induction did not correlate with the magnitude of IFN-g
`induction, because peak IFN-g levels after the second rhIL-12
`dose in the type-I and type-II patterns were not associated with
`similar increases in peak IL-15 induction. Nonetheless, there
`was an association between the ability to sustain comparable
`levels of IL-15 induction during a cycle of rhIL-12 and the
`
`NOVARTIS EXHIBIT 2038
`Breckenridge v. Novartis, IPR 2017-01592
`Page 6 of 15
`
`

`

`1684 Phase I Trial of Twice-Weekly i.v. rhIL-12
`
`Table 4 Relation between pattern of IFN-g induction by IL-12 during cycle 1, clinical response, and cytopenias
`
`IFN-g
`induction
`pattern
`Type I
`Patient 10
`13
`18
`22
`
`Type II
`Patient 24
`26
`28
`
`Cycle 1 Peak IFN-g level (pg/ml)
`
`Disease
`
`Wk1D1a
`
`Wk1D4
`
`Wk4D1
`
`IL-12 dose
`level
`(ng/kg)
`
`RCC
`RCC
`RCC
`RCC
`
`RCC
`RCC
`Mel
`
`1600
`1100
`450
`1000
`
`2365
`2040
`1560
`
`2560
`2235
`1290
`1840
`
`4960
`9060
`5918
`
`1200
`1340
`400
`850
`
`220
`248
`77
`
`500
`500
`700
`500
`
`500
`500
`500
`
`Response
`
`Cytopenia
`
`SD @48 wk
`PR @36 wk
`SD @20 wk
`SD @24 wk
`
`PD @12 wk
`PD @6 wk
`PD @12 wk
`
`None
`None
`Hemolytic anemia
`Agranulocytosis
`
`None
`None
`None
`
`Type III
`None
`PD @6 wk
`500
`159
`1476
`2229
`Mel
`Patient 21
`None
`PD @6 wk
`500
`90
`951
`1256
`Mel
`23
`None
`PD @6 wk
`700
`125
`890
`1800
`RCC
`19
`a Wk1D1, week 1 day 1; Wk1D4, week 1 day 4; Wk4D1, week 4 day 1; RCC, renal cell cancer; Mel, melanoma; SD, stable disease; PD,
`progressive disease.
`
`ability to sustain IFN-g induction. As shown in Fig. 2A (middle),
`only small differences in the peak and plateau levels of IL-15
`after the first, second, and seventh rhIL-12 doses were evident in
`patients with the type-I IFN-g pattern. In contrast, there was a
`50 –70% drop in the peak and plateau IL-15 levels by week 4 in
`patients with the type-II and type-III IFN-g patterns (Fig. 2,
`B-C, middle), and no IL-15 could be detected in the plasma
`before the seventh rhIL-12 dose in these patients.
`In all of the patients tested, small amounts of IL-18 were
`detected in the plasma before starting rhIL-12 (Fig. 2, A-C,
`bottom), with higher levels present before the second and sev-
`enth rhIL-12 doses. However, plasma IL-18 levels after an
`rhIL-12 injection usually peaked later than IFN-g. In addition,
`the loss of IFN-g production after the seventh rhIL-12 dose
`among patients with the type-II and type-III IFN-g patterns
`occurred despite continued IL-18 production at levels compara-
`ble with those measured immediately before and after the first
`rhIL-12 dose.
`IL-10 Induction and rhIL-12 Pharmacokinetics. As
`shown in Fig. 3A, IL-10 was induced after the first dose of
`rhIL-12 in a similar manner in all of the patients, returning to
`undetectable levels before the next rhIL-12 dose. A stronger
`increase in IL-10 production was observed after the second dose
`in all of the patients, with the highest peak levels detected in
`patients with the type-II IFN-g pattern (Fig. 3A, middle). How-
`ever, IL-10 induction continued after the seventh dose in all of
`the patients, including those with the type-I IFN-g pattern (Fig.
`3A, top), as well as those with the type-III pattern exhibiting a
`significant suppression of IFN-g (Fig

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