throbber
Published OnlineFirst January 14, 2015; DOI: 10.1158/1078-0432.CCR-14-2291
`
`Review
`
`Interleukin-6 as a Therapeutic Target
`Jean-Fran¸cois Rossi1,2,3, Zhao-Yang Lu4, Michel Jourdan2, and Bernard Klein2,3
`
`Clinical
`Cancer
`Research
`
`Abstract
`
`Human IL6 is a cytokine produced by many cell types that
`has pleiotropic effects. In agreement, anti-IL6 therapy reduces
`inflammation, hepatic acute phase proteins, and anemia and
`has antiangiogenic effects. Blocking IL6 has demonstrated
`therapeutic efficacy with drug registration in Castleman disease
`and inflammatory diseases (rheumatoid arthritis) without
`major toxicity. Interestingly, the inhibition of C-reactive pro-
`tein (CRP) production is a trustworthy surrogate marker of anti-
`IL6 therapy efficacy. Clinically registered IL6 inhibitors include
`siltuximab, an anti-IL6 mAb, and tocilizumab, an anti-IL6R
`mAb. In various cancers, in particular plasma cell cancers, large
`randomized trials showed no efficacy of IL6 inhibitors, despite
`
`a full inhibition of CRP production in treated patients in vivo,
`the numerous data showing an involvement of IL6 in these
`diseases, and initial short-term treatments demonstrating a
`dramatic inhibition of cancer cell proliferation in vivo. A likely
`explanation is the plasticity of cancer cells, with the presence of
`various subclones, making the outgrowth of cancer subclones
`possible using growth factors other than IL6. In addition,
`current therapeutic strategies used in these cancers already
`target IL6 activity. Thus, anti-IL6 therapeutics are able to neu-
`tralize IL6 production in vivo and are safe and useful in inflam-
`matory diseases and Castleman disease. Clin Cancer Res; 21(6);
`1248–57. Ó2015 AACR.
`
`Introduction
`
`IL6 was first described as a cytokine inducing B lymphocytes
`to produce immunoglobulin or stimulating hepatocytes and it
`was named B-stimulating factor-2 or hepatocyte growth factor
`(HGF; ref. 1; Figs. 1 and 2). IL6 is an inflammatory cytokine
`involved in various biologic processes, including dysimmune
`diseases and cancers (2, 3). It
`is produced by many cell
`lineages, including stromal cells, hematopoietic cells, epithe-
`lial cells, or muscle cells. IL6 binds to a membrane receptor
`(IL6R or CD126) or to its soluble form (sIL6R) with a 109 Kd
`(4). Then, the complexes IL6/IL6R or IL6/sIL6R bind to the
`gp130 IL6 transducer (CD130) with a low Kd (1011 M). These
`bindings result in gp130 dimerization, phosphorylation, and
`activation of
`receptor-associated kinases
`(JAK1,
`JAK2,
`and Tyk2; ref. 4). Gp130 is a common transducing chain used
`by the seven members of the gp130 cytokine family and by
`IL27 (5).
`Anti-IL6 therapies have been developed for the treatment of
`dysimmune diseases and in cancers. Current IL6 inhibitors
`include mAbs to IL6 (siltuximab) or IL6 receptor (tocilizu-
`mab), and other inhibitors are being investigated in clinical
`trials. This review updates the data on the clinical efficacy of
`IL6 inhibitors.
`
`1Department of Hematology, CHU de Montpellier, Montpellier, France.
`2INSERM U1040, Montpellier, France. 3Universite Montpellier
`I,
`Montpellier, France. 4Unite de Therapie Cellulaire, CHU de Montpellier,
`Montpellier, France.
`
`Note: J.-F. Rossi and B. Klein share senior authorship.
`
`Corresponding Author: Jean-Fran¸cois Rossi, Department of Hematology, CHU
`Saint-Eloi, 80 Avenue Augustin Fliche 34295 Montpellier Cedex, France. Phone:
`33-4-67-33-80-79; Fax: 33-4-67-33-83-73; E-mail: jeanfrancoisrossi@me.com
`
`doi: 10.1158/1078-0432.CCR-14-2291
`
`Ó2015 American Association for Cancer Research.
`
`Lessons from Anti-IL6 Murine Monoclonal
`Antibodies
`
`In the early 1990s, our group initiated the first clinical trials
`with anti-IL6 murine mAbs in patients with multiple myeloma or
`metastatic renal cell carcinoma (MRCC).
`
`Clinical observations
`The first treated patients had extramedullary multiple myeloma
`disease, in particular plasma cell leukemia, and the inhibition of
`malignant plasma cell proliferation within a few days of treatment
`confirmed that IL6 was a growth factor for malignant plasma cells
`in vivo (6, 7), as shown using in vitro models (8, 9). These first anti-
`IL6 treatments used anti-IL6 murine mAbs, elsilimomab (BE-8),
`and/or BE-4, or mAb8, and have shown that the C-reactive protein
`(CRP) production by human hepatocytes was completely under
`the control of IL6 in vivo, as evidenced by the loss of circulating
`CRP throughout anti-IL6 treatment and a quick loss reversal at
`treatment discontinuation (7). Anti-IL6 therapy also inhibited
`fever with body temperature normalization (7) in line with IL6
`pyrogenic activity in rats (10). No major toxicity was evidenced,
`except a transient and mild (25%) decrease in platelet count
`(7, 11–13), which is expected given the role of IL6 to drive
`megakaryocyte maturation (14). Objective response with a
`decrease of the monoclonal component was observed in some
`patients, including refractory patients (11). We also treated 18
`patients with MRCC with BE-8 (elsilimomab) and 4 patients
`received both BE-8 and IFNa-2a, resulting in 2 partial responses,
`2 minor responses, one stable disease (12). Patients' conditions
`improved, in particular with an increase in hemoglobin level,
`analgesia, and a lack of flu-like syndrome despite IFN coadmin-
`istration in some patients (12). Serum CRP was no more detect-
`able within 2 days after start of anti-IL6 treatment, making it
`an easy marker of treatment efficacy (7, 11, 12). Other acute
`phase proteins also decreased rapidly (serum amyloid A protein,
`a1 anti-trypsin) and serum albumin increased 20 days after the
`anti-IL6 therapy initiation in agreement with the major role of
`IL6 to control acute phase protein and albumin production by
`
`1248
`
`Clin Cancer Res; 21(6) March 15, 2015
`
`Downloaded from
`
`clincancerres.aacrjournals.org
`
`on July 19, 2019. © 2015 American Association for Cancer Research.
`
`Lassen - Exhibit 1024, p. 1
`
`

`

`Published OnlineFirst January 14, 2015; DOI: 10.1158/1078-0432.CCR-14-2291
`
`Anti-IL6 Therapy
`
`Inflammation Immunity Reproduction Metabolism Hematopoiesis
`
`Neural development Bone remodeling Angiogenesis
`
`Figure 1.
`IL6 is a pleiotropic cytokine. gp130,
`glycoprotein 130; Treg, regulatory
`T cell.
`
`IL6 (4-helix bundle, 212 AA)
`
`B lymphocytes: plasma cell differentiation,
`survival/proliferation factor for multiple
`myeloma
`
`T lymphocytes: in association with TGFβ
`differentiation of TH17 and IL27 inhibition
`of Treg
`
`Megakaryocytes: differentiation
`
`Cardiac stem cells: cell survival
`
`Adipocytes: lipolysis
`Osteoclasts: Cox-2, Wnt, NF-κB, RANK
`
`Neural stem cells: astrocyte differentiation
`
`© 2015 American Association for Cancer Research
`
`hepatocytes (15). Hemoglobin level slightly increased by 1 to 1.5
`g/dL due to inflammation control and no major change was seen
`in circulating T cells, natural killer cells, or B lymphocytes and
`in complement molecules. Overall, these first clinical trials with
`
`murine anti-IL6 mAb have shown the lack of toxicity of anti-IL6
`therapy, identified CRP as an easy and quick surrogate marker of
`IL6 bioactivity, and shown antitumor effects in some patients with
`multiple myeloma or MRCC. In addition, they made it possible to
`
`sIL6R
`
`IL6/sIL6R
`
`1
`
`IL11
`
`CLC
`
`CNTF
`
`LIF
`OSM
`
`CT-1
`
`OSM
`
`2
`
`IL27
`
`Free
`IL6
`
`gp130
`
`WSX-1
`
`PI3K
`
`AKT
`
`IL6R
`
`IL11R
`
`P P
`
`JAK
`
`STAT3
`
`STAT3
`
`STAT3
`
`P
`
`P
`
`MAPK
`
`Survival and cell cycle
`Increased RANKL expression
`
`Cell cycle
`Cell growth
`
`Survival
`
`© 2015 American Association for Cancer Research
`
`Figure 2.
`The IL6 receptor and glycoprotein
`(gp) 130 transducer cytokine family.
`Part 1 illustrates usual signaling. Part 2
`shows trans-signaling. AKT, protein
`kinase B; CLC, cardiotrophin-like
`cytokine; CNTF, ciliary neurotrophic
`factor; CT-1, cardiotrophin-1; gp,
`glycoprotein; LIF, leukemia inhibitory
`factor; OSM, oncostatin M; WSX-1, IL27
`receptor subunit a.
`
`www.aacrjournals.org
`
`Downloaded from
`
`clincancerres.aacrjournals.org
`
`Clin Cancer Res; 21(6) March 15, 2015
`
`on July 19, 2019. © 2015 American Association for Cancer Research.
`
`1249
`
`Lassen - Exhibit 1024, p. 2
`
`

`

`Published OnlineFirst January 14, 2015; DOI: 10.1158/1078-0432.CCR-14-2291
`
`Rossi et al.
`
`understand the main biologic mechanisms involved in anti-IL6
`therapy.
`
`Basis for Pharmacologic Effects of
`Antagonists
`
`Initial investigations of the biologic mechanisms involved in
`anti-IL6 treatment
`At the stop of the anti-IL6 mAb treatment, a quick recovery of
`CRP production, fever, and cancer disease was observed (7). The
`reason is that the anti-IL6 mAb binds IL6, prevents its consump-
`tion by cells and its renal clearance, and accumulates large levels of
`circulating IL6 in the form of stable monomeric IL6/anti-IL6
`complexes (16). The half-life of circulating IL6 increased from
`a few minutes in untreated individuals to several days, actually the
`same half-life of the free mAb, in anti-IL6–treated patients (16).
`Using this observation, we calculated the in vivo daily production
`of IL6 by integrating pharmacologic and affinities parameters and
`showed a high variation in daily IL6 production in vivo, from
`several mg/day to mg/day, and in patients with the lower IL6
`production, a complete blockade of CRP production and objec-
`tive response (17). In a patient developing acute Escherichia coli
`septicemia during anti-IL6 treatment, a production of IL6 over
`7 mg/day was calculated (18). Using CRP serum levels as a
`surrogate marker for IL6 bioactivity in vivo, we have shown that
`the anti-IL6 mAb was able to fully block IL6 activity in patients
`producing less than 18 mg IL6 per day, in association with an
`antitumor effect (11, 17). This finding indicated that the dose of
`anti-IL6 mAb injected could be several 100-fold too low to
`neutralize a huge IL6 production in vivo and to control CRP
`production and tumor growth in some patients.
`In addition, at the end of anti-IL6 mAb treatment, we have
`shown that the ratio of the concentrations of free anti-IL6 mAbs to
`IL6/anti-IL6 mAb complexes decreased, making it possible for
`soluble or cell membrane IL6 receptors to disrupt IL6/anti-IL6
`mAb complexes and use this large amount of circulating IL6 to
`trigger cell activation. This mechanism explains the quick recovery
`of CRP production, fever, and cancer cell progression occurring at
`the end of anti-IL6 treatment in some patients (11). A possibility
`to avoid this rebound effect is to use a combination of two or three
`anti-IL6 mAbs recognizing different epitopes. This would result in
`the formation of polymeric IL6/IL6 mAb complexes, which are
`captured mainly by hepatocytes leading to a rapid clearance, as we
`demonstrated in a murine model (19).
`
`The Anti-IL6 Drugs
`
`Several anti-IL6 mAbs have been developed. The CNTO328
`chimeric anti-IL6 mAb (siltuximab) was used in clinical trials for
`multiple myeloma (20, 21), MRCC (22), and prostate cancer (23)
`and was recently registered for treating patients with Castleman
`disease (24). Sirukumab, a humanized anti-IL6 mAb, has been
`assayed in healthy subjects to determine PK/PD and safety. This
`drug has a half-life ranging from 18.5 to 29.6 days with no serious
`adverse events (25). Anti-IL6R mAb, particularly atlizumab (also
`called tocilizumab), has been assayed in dysimmune diseases
`(26). Recently, CytomX Therapeutics Inc. developed targeted
`"masked" antibodies, which are activated by disease-associated
`proteases (27). Sant7, a potent antagonist of the IL6 receptor,
`was engineered through targeted amino acid substitutions in
`key residues of the human IL6 molecule (28). Sant7 shows
`higher affinity than IL6 for the gp80 receptor subunit, but
`completely lacks binding capacity to the gp130 receptor sig-
`naling subunit. Other inhibitors have been developed (Tables 1
`and 2).
`
`In the initial clinical trials with the BE-8 anti-IL6 murine mAb, a
`full inhibition of CRP production was achieved only in patients
`producing less than 18 mg/day of IL6 (11). Considering the
`molecular weights of IL6 and BE-8 anti-IL6 mAb, the Kd of
`binding of BE-8 to IL6 (1011 M), and the concentration of
`circulating BE-8 in patients (mean concentration of 10 mg/mL),
`a production of IL6 less than 18 mg/day in vivo meant a 100-fold
`molar excess of BE-8 mAb to IL6 (Fig. 3). In patients with multiple
`myeloma treated with the siltuximab humanized anti-IL6 mAb, a
`siltuximab concentration of 5 mg/mL was proposed to achieve a
`300-fold molar excess of siltuximab to IL6, which is considered
`necessary to block IL6 activity and tumor growth with that
`mAb (29). In a phase I study in patients with MRCC, the admin-
`istration of 6 mg/kg of siltuximab every 2 weeks efficiently
`suppressed serum CRP in patients who had a baseline CRP level
` 30 mg/L (30). These data fit well with the pharmacokinetics
`parameters of siltuximab (30). Indeed, clinically relevant sche-
`dules of siltuximab were simulated predicting a dosage of 6 mg/kg
`every 2 weeks or 9 mg/kg every 3 weeks would decrease CRP to
`below the lower limit of quantification (30).
`Regarding tocilizumab, an administration of 8 mg/kg once
`every 2 weeks resulted in a marked increase in serum sIL6R in
`the form of sIL6R/tocilizumab complexes and reached a steady
`state at day 42 of treatment (31). A decrease of CRP level was
`found as long as the concentration of free tocilizumab not
`engaged in sIL6R/tocilizumab complexes and able to inhibit
`biding of IL6 to membrane or soluble IL6R remained above
`1 mg/mL in serum (31).
`
`Diseases Improved by Anti-IL6 Therapies
`
`Dysimmune diseases
`In the early phases of inflammation, IL6 is produced by
`monocytes and macrophages, in particular though the stimu-
`lation of Toll-like receptors. A deregulated and persistent IL6
`production has been observed in various chronic inflammatory
`and/or autoimmune diseases, including in animal models. IL6
`blockade by means of gene-knockout or administration of anti-
`IL6 or anti-IL6R antibody can suppress such disease develop-
`ment either preventively or therapeutically. The anti-IL6R mAb
`tocilizumab has demonstrated efficacy either as a monotherapy
`or in combination with disease-modifying antirheumatic drugs
`for adult patients with moderate to severe rheumatoid arthritis
`(for review refs. 26, 31). A Cochrane database systematic review
`concluded that tocilizumab-treated patients were four times
`more likely to achieve American College of Rheumatology 50%
`improvement (38.8% vs. 9.6%) and 11 times more likely to
`achieve Disease Activity Score remission as compared with
`control patients (30.5% vs. 2.7%; ref. 32). Thus, the anti-IL6R
`mAb tocilizumab is now approved for the treatment of rheu-
`matoid arthritis in more than 90 countries. The outstanding
`results obtained with tocilizumab in rheumatoid arthritis led to
`a change in the treatment objective from protection against
`joint destruction to prolongation of life expectancy with nor-
`mal activities in daily life. Safety has been reported from six
`studies performed in Japan. The incidence of adverse events
`(AE), including abnormal laboratory test results, was 465.1 per
`100 patient-years, with infection being the most common AE
`
`1250
`
`Clin Cancer Res; 21(6) March 15, 2015
`
`Downloaded from
`
`clincancerres.aacrjournals.org
`
`on July 19, 2019. © 2015 American Association for Cancer Research.
`
`Clinical Cancer Research
`
`Lassen - Exhibit 1024, p. 3
`
`

`

`Published OnlineFirst January 14, 2015; DOI: 10.1158/1078-0432.CCR-14-2291
`
`Anti-IL6 Therapy
`
`Table 1. Anti-IL6 antagonists in clinical trials
`
`Drug (company)
`
`Elsilimomab or BE-8
`
`Siltuximab, chimeric (CNTO328)
`
`Sirukumab, CNTO 136
`Humanized anti-IL6
`ALD518 or BMS-945429
`
`Anti-IL6, humanized
`
`Alder Biopharm. Inc./BMS
`
`Clinical trial (disease/type)
`
`ClinicalTrials.gov
`
`Lymphoma, MM, MRCC
`Phase I/II
`Mab 1339, fully human from BE-8 OPi EUSA/Vaccinex/GSK
`MM
`Phase I
`Phase II multicentric CNTO 328 þ Dex
`Randomized phase II, multicentric CNTO328 þ Bor
`First-line phase Ib/II Len/Bor/Dex/CNTO328
`MGUS, SMM, indolent MM
`Phase I (effect on heart)
`SMM high-risk
`Phase II randomized
`Myelodysplastic syndrome
`Randomized phase II
`CNTO328þBSC vs. placeboþBSC
`Solid cancers
`Phase I/II, ovarian, pancreatic, colon, head and neck, lung cancers
`MRCC
`Phase I/II
`Prostate cancer metastatic, hormone-refractory
`Mitoxantrone þ prednisone þ CNTO328 phase II
`Prostate cancer not responding to hormone therapy
`Multicentric Castleman disease
`Randomized phase II (long-term evaluation)
`Active RA
`Under methotrexate therapy
`Cancer
`Phase I
`NSCLC
`Phase I/II (fatigue and cachexia) multicentric
`RA
`Phase II
`Crohn disease
`Phase II (discontinued)
`
`NCTO1309412
`NCT00402181
`
`NCTO1531998
`
`NCT01219010
`
`NCT01484275
`
`NCT015133317
`
`NCT00841191
`
`NCT00385827
`
`NCT00433446
`NCT0400503
`
`NCT00718718
`
`NCT00867516
`
`NCT01545050
`
`Olokizumab (CDP6038)
`Humanized anti-IL6
`UCB Group/Russia's R-Pharm
`
`RA
`Anti-TNF failure phase II
`
`NCT01533714 and NCT01463059
`
`Abbreviations: Bor, bortezomib; BSC, best supportive care; Carbo, carboplatin; Dex, dexamethasone; Doxo, doxorubicin; Len, lenalidomide; MGUS, monoclonal
`gammopathy of undetermined significance; MM, multiple myeloma; NSCLC, non–small cell lung carcinoma; RA, rheumatoid arthritis; SMM, smouldering multiple
`myeloma.
`
`with 6.22 per 100 patient-years (32). As expected, increases in
`liver function and lipid parameters were observed. Tocilizumab
`is also a promising drug for systemic lupus erythematosus,
`systemic sclerosis, polymyositis, Takayasu and giant cell arter-
`itis, Crohn disease, relapsing polychondritis, multiple sclerosis,
`Still disease, and Beh¸cet disease (26, 31).
`
`Castleman disease
`Castleman disease is a lymphoproliferative disease character-
`ized by benign hyperplastic lymph nodes, follicular hyperplasia
`with polyclonal plasmablastic proliferation and capillary prolif-
`eration associated with vascular hyperplasia and high IL6 activity,
`mainly due to viral IL6 (33, 34). Viral IL6 is produced by cells
`infected by Kaposi sarcoma-associated herpes virus (KSHV) and
`directly binds and stimulates gp130 IL6 transducer in the absence
`of IL6R (33, 34). All of the HIV-positive and half of the HIV-
`negative patients with multicentric Castleman disease were
`infected with KSHV (33). The rationale for developing anti-IL6
`therapy was first that IL6 is a main growth factor for plasmablasts
`(35) and that it has pleiotropic activities, which may explain
`vascular hyperplasia in particular. In particular, transgenic mice
`bearing an IL6 transgene driven by the immunoglobulin Em
`enhancer develop massive polyclonal plasmacytoses and rapidly
`die (36).
`
`The BE-8 anti-IL6 murine mAb was first used for a patient with
`Castleman disease, who showed obvious disease improvement
`after treatment (37). A clinical trial with the anti-IL6R mAb
`tocilizumab confirmed this benefit and led to its approval in
`Japan as an orphan drug for this disease in 2005. Recently, the
`anti-IL6 mAb siltuximab was also proven to provide major clinical
`benefit in a randomized phase II study enrolling 79 patients, and it
`has been approved for treatment of this disease (24).
`
`Diseases with No Demonstrated Benefit of
`Anti-IL6 Therapies
`
`Multiple myeloma
`Before reviewing the efficacy of randomized anti-IL6 trials in
`patients with multiple myeloma, it is worthwhile to update the
`current knowledge about the place of IL6 as a growth factor for
`multiple myeloma cells (MMC). IL6 was recognized in the late
`1980s as an important growth factor for MMCs, being produced
`mainly by the tumor environment (6, 7) and also by MMCs (8).
`Although most studies confirmed these findings (38), current
`data indicate that insulin-like growth factor-1 (IGF-1) is the
`major growth factor for MMCs, and that IL6 is active mainly in
`CD45þ MMCs (39, 40). The phosphatase CD45 dephosphor-
`ylates IGF-1R in MMCs and weakens IGF-1R signaling, making
`
`www.aacrjournals.org
`
`Downloaded from
`
`clincancerres.aacrjournals.org
`
`Clin Cancer Res; 21(6) March 15, 2015
`
`on July 19, 2019. © 2015 American Association for Cancer Research.
`
`1251
`
`Lassen - Exhibit 1024, p. 4
`
`

`

`Published OnlineFirst January 14, 2015; DOI: 10.1158/1078-0432.CCR-14-2291
`
`Rossi et al.
`
`Table 2. Anti-IL6 receptor antagonists in clinical trials
`
`Clinical trial (disease/type)
`
`208 studies including
`Ovarian cancer
`Phase I/II
`(Carbo/pegylated liposomal doxorubicin hydrochloride or Carbo/Doxo) þ
`tocilizumab/Peginterferon alfa-2b
`Erdheim–Chester disease
`(histiocytose)
`Phase II
`Hematophagocytic lymphohistiocytosis
`Phase II
`Graft-versus-host disease
`Phase II
`Steroid-refractory
`Castleman disease KSHV
` Antiviral drugs
`Continuing in responding patients
`14 studies: 12 in RA, including a phase III trial,
`1 in noninfectious uveitis, and 1 in ankylosing spondylitis
`
`ClinicalTrials.gov
`
`NCT01637532
`
`NCT01727206
`
`NCT02007239
`
`NCT02174263
`
`NCT01475162
`NCT01441063
`
`NCT01183598
`
`Drug (company)
`
`Tocilizumab
`
`Sarilumab IL6R
`Sanofi/Regeneron
`VX30 (Vaccinex)
`
`ARGX-109 (arGEN-X)
`
`FM101 (Formatech)
`
`Sant7 (receptor superantagonist)
`
`Abbreviations: Bor, bortezomib; BSC, best supportive care; Carbo, carboplatin; Dex, dexamethasone; Doxo, doxorubicin; Len, lenalidomide; MGUS, monoclonal
`gammopathy of undetermined significance; MM, multiple myeloma; NSCLC, non–small cell lung carcinoma; RA, rheumatoid artritis; SMM, smouldering multiple
`myeloma.
`
`MMCs more dependent on IL6 (39). Besides IGF-1 and IL6,
`other MMC growth factors have been described, including
`mainly BAFF/APRIL, produced by osteoclasts (41), and mem-
`bers of the EGF family, such as HGF and VEGF (see ref. 42 for
`review). In addition, CRP, whose production is under IL6
`control, also increases IL6 production in MMCs, enhances their
`
`proliferation under stress conditions, and protects them from
`chemotherapy (43).
`Regarding signaling pathways, IL6 triggered the JAK/STAT3
`pathway, which drives an antiapoptotic response in MMCs,
`mainly through upregulation of the MCL1 antiapoptotic protein
`)44 ). It also triggers the MAP kinase ERK1/2, activating the cell
`
`sIL6R
`
`10–11 M
`
`IL6
`
`10–9 M
`
`10–9 M
`
`10–11 M
`
`Anti-IL6
`mAb
`
`10–11 M
`
`Figure 3.
`Basis for pharmacologic effects of IL6
`antagonists. gp, glycoprotein.
`
`IL6R
`
`gp130
`
`1252
`
`Clin Cancer Res; 21(6) March 15, 2015
`
`Downloaded from
`
`clincancerres.aacrjournals.org
`
`© 2015 American Association for Cancer Research
`
`on July 19, 2019. © 2015 American Association for Cancer Research.
`
`Clinical Cancer Research
`
`Lassen - Exhibit 1024, p. 5
`
`

`

`Published OnlineFirst January 14, 2015; DOI: 10.1158/1078-0432.CCR-14-2291
`
`Anti-IL6 Therapy
`
`cycle (41). The PI3K and AKT signaling pathway is also important
`to promote MMC growth. The AKT kinase is activated in MMCs
`(45). But, in our opinion, IL6 does not activate this pathway,
`unlike IGF-1 or other MMC growth factors (44). This may explain
`the additive effects between IL6 and other growth factors to
`support MMC growth.
`Two randomized trials were recently published showing a
`lack of effect of anti-IL6 therapy. In one trial, the anti-IL6 mAb
`silxutimab was given to transplant-ineligible patients in asso-
`ciation with a regimen of bortezomib–melphalan–prednisone
`compared with bortezomib–melphalan–prednisone and then
`as a maintenance therapy for 18 months or until relapse (20).
`In another trial, siltuximab was given in relapsed/refractory
`multiple myeloma in association with bortezomib compared
`with bortezomib alone and then as maintenance therapy until
`progression (21). These two trials showed no benefit for event-
`free or overall survivals. In the first study, an improvement
`of response was found. The initial anti-IL6 treatments per-
`formed by our group did not investigate event or overall
`survival, but showed a quick blockade of tumor proliferation
`and reduction in tumor mass in patients with fulminant dis-
`ease, mainly with extramedullary proliferation (7, 11). In a
`series of 24 newly diagnosed patients, the BE-8 anti-IL6 mAb
`was given together with 140 mg/m2 of melphalan and autol-
`ogous stem cell transplantation for 21 days (13). This combi-
`nation was shown to be as active as 200 mg/m2 of melphalan to
`decrease the tumor mass, based on historical comparison, and
`without impairment on the hematopoietic recovery (13).
`How can we explain this lack of benefit in randomized trials
`with anti-IL6 mAb despite the major role of IL6 to control healthy
`plasmablast proliferation (35, 36) and despite the blockage of
`myeloma cell proliferation by anti-IL6 mAb in patients with
`fulminant disease (7, 11)? In these randomized trials, siltuximab
`was able to block CRP production, indicating its neutralization of
`IL6 bioactivity in the liver. Thus, a lack of antimyeloma effect of
`siltuximab was not due to an inability to block a too large IL6
`production. In line with this, median CRP levels are not higher in
`patients with multiple myeloma compared with those with rheu-
`matoid arthritis for whom anti-IL6 therapy had benefit (Table 3).
`Several mechanisms could explain this lack of effect of anti-IL6
`therapy. First of all, the efficacy of anti-IL6 therapy was compared
`with drug combination (bortezomib or bortezomib–melphalan–
`prednisone), which already reduces IL6 production and inhibits
`CRP production (20). Second, as summed up above, other factors
`could stimulate MMC growth in the absence of IL6, in particular
`IGF1 in CD45/low MMCs, and favor the emergence of MMC
`subclones independent of IL6 (39, 40). This emergence of IL6-
`independent subclones could not be investigated in our initial
`short-term treatments in patients with fulminant disease. Third,
`other cytokines of the IL6 family can trigger the gp130 IL6
`transducer and also the growth of MMCs and could substitute
`for IL6 to promote MMC survival and growth in vivo (46).
`
`Other hematologic malignancies
`IL6 and/or CRP are prognostic factors for several B-cell malig-
`nancies, particularly malignant lymphoma (47). IL6 is a biologic
`marker in other lymphoid malignancies, including Hodgkin
`disease, mantle-cell lymphoma, cutaneous CD30-positive lym-
`phomas, and KSHV-associated malignancies (48–52). Poly-
`morphisms of different cytokine genes, and particularly of IL6
`gene (IL6-174GG genotype) are associated with treatment failure
`
`in Hodgkin disease (53). The majority of B-cell tumors expressed
`the IL6 receptors, in particular mantle cell lymphoma (54). BE-8
`was used in 11 patients with HIV-associated lymphoma and
`showed a clinical benefit, particularly on B symptoms (55), and
`in one patient with acute monoblastic leukemia (56). Therefore,
`IL6 and more generally gp130 IL6 transducer signaling could play
`an important role in the pathogenesis of certain B-cell neoplasias,
`and new clinical programs could be developed in such diseases.
`
`Metastatic renal cell carcinoma
`IL6 was shown to be an autocrine proliferation factor for tumor
`cell lines obtained from patients with MRCC (57). Mutations in
`the TP53 gene could contribute to the overexpression of IL6 in
`renal cell carcinoma (58). We and others have demonstrated that
`CRP/IL6 is a prognostic factor in MRCC (12, 59, 60). Furthermore,
`we demonstrated a significant association between the presence of
`the IL6R in tumors and tumor stage, nuclear grade, proliferation
`index, and serum IL6 (61). Treatment of renal cell carcinoma cells
`with cisplatinum (CDDP) in combination with anti-IL6 or anti-
`IL6R mAbs can overcome their CDDP resistance by downregula-
`tion of glutathione S-transferase p expression (62). Furthermore,
`in a phase I study, the use of IL6 and GM-CSF in patients with renal
`cell carcinoma was associated with inverse clinical effects (63). We
`treated 18 MRCC patients with BE-8 mAb and demonstrated a
`clinical benefit, including abrogation of fever, hypercalcemia, and
`inflammatory syndrome, reduction of anemia and morphine
`intake, and weight increase, with objective responses observed
`(2/18 patients with partial responses, one with a minor response,
`and one with stable disease; ref. 12). In a phase I/II study,
`siltuximab was shown to stabilize the disease in more than
`50% of progressive MRCC patients, with one partial response
`and a favorable safety profile, a situation that could authorize
`further evaluation of dose-escalation strategies and/or combina-
`tion therapy (22). Nevertheless, to our knowledge, no further
`studies have been performed.
`
`Prostate cancer
`IL6 acts as a paracrine and autocrine growth factor for both
`benign and cancer prostate cells. The levels of IL6 and IL6R
`are increased during prostate carcinogenesis and tumor pro-
`gression (64). In addition, a correlation between increased
`serum IL6 and soluble IL6R levels with aggressiveness of the
`disease was reported (65). During the treatment of prostate
`cancer, castration resistance usually leads to the death of the
`patient. The signaling pathway mediated by IL6 represents an
`alternative pathway in the cancer-resistance phenotype acqui-
`sition and cancer progression (64, 65). During androgen dep-
`rivation therapy, a regulation loop may emerge between sex
`steroids and IL6, with a strong positive correlation with total-
`testosterone, androstenedione, and estradiol levels (66). Other
`gp130 cytokines have shown involvement in prostate tumor
`progression by promoting VEGF and urokinase plasminogen
`activator (67). A meta-analysis was recently performed to
`evaluate the association between the IL6 174GC genotype and
`susceptibility to prostate cancer with an increased risk observed
`in two cohort studies. Additional well-designed studies are
`needed to validate the role of IL6 genetic polymorphism in
`prostatic cancer risk (68).
`All the above-mentioned data show the importance of the
`IL6/IL6R pathway in the regulation of growth and drug resistance
`of prostate cancer cells. In a phase I study, 20 patients scheduled
`
`www.aacrjournals.org
`
`Downloaded from
`
`clincancerres.aacrjournals.org
`
`Clin Cancer Res; 21(6) March 15, 2015
`
`on July 19, 2019. © 2015 American Association for Cancer Research.
`
`1253
`
`Lassen - Exhibit 1024, p. 6
`
`

`

`Published OnlineFirst January 14, 2015; DOI: 10.1158/1078-0432.CCR-14-2291
`
`Rossi et al.
`
`Abbreviations:EU,EuropeanUnion;J,Japan;MCCD,MulticentricCastlemandisease;MM,multiplemyeloma;NR,notreported;RA,rheumatoidarthritis;Paraneop.,paraneoplastic.
`
`Tocilizumab
`

`

`

`
`NR
`NR
`NR
`
`NR
`
`
`

`

`
`No
`NR
`
`NR
`

`
`NR
`NR
`NR
`
`NR
`
`
`

`

`
`5.8(1.7–8.1)
`
`1.55(0.8–130)
`
`Crohndisease
`
`Prostatecancer
`
`No
`Paraneop.syndrome
`
`No
`Paraneop.syndromeeffectHCa
`
`No
`-effectHCa
`
`Tocilizumab(J;situximabEUandU.S.)
`þþ
`
`Tocilizumab
`þþ
`
`Drugregistration
`Diseaseimprovement
`

`

`
`NR
`NR
`

`

`

`

`
`withHIVinfection
`associated
`lymphoma
`
`Effecton
`
`29.2(0–129)
`
`Lymphoma
`

`

`
`Nomajorchange
`
`
`

`

`

`

`

`

`

`
`NR
`
`NR
`

`
`
`

`

`
`4.9(0.3–183)
`
`3.75(0–121)
`
`MRCC
`
`MM
`

`

`
`NR
`

`

`

`

`

`

`

`
`NR
`þþ
`NR
`

`

`

`
`þþ
`
`þþ
`
`17.6(0.1–181)
`
`5.6(2–19.4)
`
`MCCD
`
`RA
`
`1–8weeks
`(1.1–1.5g/dL)within
`Hemoglobinincrease
`Plateletdecrease(20%–30%decrease)
`Lymphocytecounts(peripheralblood)
`Analgesia
`Fever(Bsymptom)
`after3–4weeks)
`(20%–30%increase
`
`Albuminincrease
`
`2–4weeks)
`40%–70%reductionin
`20%decrease;other,
`proteins(ferritin
`Otheracute-phase
`within2–4days
`100%inhibition
`
`CRPinhibition90%–
`
`inhibitors
`
`EffectsofIL6
`
`(mg/L;range)
`
`MedianCRPlevels
`
`Table3.Clinicaleffectsofanti-IL6therapies(monoclonalantibodiestocilizumabandsiltuximab)
`
`1254
`
`Clin Cancer Res; 21(6) March 15, 2015
`
`Downloaded from
`
`clincancerres.aacrjournals.org
`
`on July 19, 2019. © 2015 American Association for Cancer Research.
`
`Clinical Cancer Research
`
`Lassen - Exhibit 1024, p. 7
`
`

`

`Published OnlineFirst January 14, 2015; DOI: 10.1158/1078-0432.CCR-14-2291
`
`Anti-IL6 Therapy
`
`for radical prostatectomy received either no drug or siltuximab
`(6 mg/kg, 5 patients/group with administration once, twice, and
`three times, prior to surgery). A decrease in phosphorylation of
`Stat3 transcription factor and p44/p42 MAPKs, with a down-
`regulation of genes immediately downstream of the IL6 signaling
`pathway and key enzymes of the androgen signaling pathway, was
`observed (69). A phase II study designed by SWOG was com-
`pleted in chemotherapy-pretreated patients with castration-resis-
`tant prostate cancer, showing stable disease in 7 patients (23%).
`An increase of IL6 was observed after treatment, likely due to the
`accumulation of stable IL6/anti-IL6 mAb complexes, as we dem-
`onstrated in other trials (16). This may explain the lack of clinical
`benefit (70). The combination of mitoxantrone/prednisone and
`siltuximab was not associated with a clinical improvement in a
`randomized phase II study, as compared with chemotherapy
`alone (23). Other molecules have been shown efficacious on
`tumorigenesis in prostate cancer,
`including polyphenols, by
`mediating a decrease in IL6 signaling (71). Because of a rise in
`IL6 both in vitro and in vivo in the presence of tyrosine kinase–
`resistant cells, IL6 may represent a biomarker for tyrosine kinase
`resistance (72).
`
`Other Potential Indications
`
`IL6 inhibition may have therapeutic indications in other can-
`cers, including ovarian cancer. Recently, a correlation between
`thrombocytosis,

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