throbber
British Journal of Haematology, 1998, 102, 783–790
`
`Chimaeric anti-interleukin 6 monoclonal antibodies
`in the treatment of advanced multiple myeloma:
`a phase I dose-escalating study
`
`H. C. T. VAN ZAANEN,1 H. M. LOKHORST,2 L. A. AARDEN,3 H. J. A. M. RENSINK,3 S. O. WARNAAR,4
`1
`1 1Department of Haematology, Academic Medical Centre, Amsterdam,
`J. VAN DER LELIE
`AND M. H. J. VAN OERS
`2Department of Haematology, Academic Hospital Utrecht, 3Central Laboratory of the Netherlands Red Cross
`Blood Transfusion Service, Amsterdam, and 4Centocor, Leiden, The Netherlands
`
`Received 18 February 1998; accepted for publication 11 May 1998
`
`Summary. Interleukin 6 plays a key role in the pathogenesis
`of multiple myeloma (MM). Therefore we conducted a phase I
`dose-escalating study with chimaeric monoclonal anti-IL6
`antibodies (cMab) in MM patients resistant to second-line
`chemotherapy. The cMab (CLB IL6/8; Kd 6·25 · 10¹12
`M)
`was given in two cycles of 14 daily infusions, starting on day
`1 and day 28, repectively, with a daily dose of 5 mg in
`patients 1–3, 10 mg in patients 4–6, 20 mg in patients 7–9
`and 40 mg in patients 10–12 (total dose 140 mg, 280 mg,
`560 mg and 1120 mg of anti-IL6, respectively). 11/12
`patients had elevated pretreatment IL6 levels.
`Except for transient thrombocytopenia in two patients
`there was no toxicity. There were no changes in haemoglobin
`levels, granulocyte count, liver enzymes or renal function.
`
`No human anti-chimaeric antibodies were induced. This was
`also reflected in a long half-life time of the cMab (median
`17·8 d), resulting in accumulation of the anti-IL6 cMab and
`high levels of circulating IL6. However, this was in the form
`of biologically inactive IL6/cMab complexes and did not
`result in acceleration of the disease. Although C-reactive
`protein (CRP) levels were decreased to below detection level
`in 11/12 patients, indicating effective IL6 blocking, none of
`the patients achieved a response according to the standard
`criteria. We conclude that this chimaeric anti-IL6 Mab has a
`low toxicity, low immunogenicity and a long T1/2. A dose of
`40 mg/d for 14 d can safely be used in future phase II studies.
`
`Keywords: chimaeric, anti-interleukin 6, multiple myeloma.
`
`Interleukin-6 (IL6) is a cytokine with multiple biological
`activities. It has been shown to be involved in such diverse
`processes as T-cell activation,
`induction of acute-phase
`proteins, and stimulation of haemopoietic precursor cell
`growth and differentiation (Heinrich et al, 1990; Kishimoto,
`1989). In the last decade in vitro and in vivo observations
`have suggested a major role of IL6 in the pathogenesis of
`multiple myeloma (MM) (Anderson et al, 1989; Hilbert et al,
`1995; Kawano et al, 1988; Klein et al, 1989; Lokhorst et al,
`1994; Nordan et al, 1987; van Oers et al, 1993; Zhang et
`al, 1989). Especially in patients with active and/or terminal
`disease, serum IL6 levels have been found to be elevated
`(Bataille et al, 1989; Klein et al, 1990).
`Murine anti-IL6 Mab have been used (in rather hetero-
`genous schedules) in the treatment of myeloma patients
`(Bataille et al, 1995; Klein et al, 1991). No major side-effects
`
`Correspondence: Dr H. C. T. van Zaanen, Sint Franciscus Gasthuis,
`Kleiweg 500, 3045 PM Rotterdam, The Netherlands.
`
`䉷 1998 Blackwell Science Ltd
`
`have been observed, but antibodies to mouse immuno-
`globulin (HAMA) were frequently detected about 2 weeks
`after starting the treatment. This resulted in rapid Mab
`clearance and diminished efficacy of this treatment. These
`HAMAs are frequently directed against the Fc part of the
`mouse immunoglobulin (Hoffman, 1990), and may also
`induce anaphylactic reactions.
`In order to reduce the risk of induction of HAMAs and thus
`to increase efficacy of
`the treatment, we produced a
`chimaeric anti-IL6 antibody (cMab). With this cMab we
`performed a phase I dose-escalating study in MM patients
`who were resistant to second-line chemotherapy. Here we
`report the results of this phase I study.
`
`PATIENTS AND METHODS
`
`All patients had MM according to the criteria of Durie &
`Salmon (1975), and had relapsed after, or were resistant to,
`second-line chemotherapy (VAD (vincristine, adriamycin
`
`783
`
`IPR2018-00685
`Celgene Ex. 2037, Page 1
`
`

`

`784 H. C. T. van Zaanen et al
`Table I. Patients’ characteristics.
`
`Pt
`
`M/F
`
`Age (yr)
`
`M-protein
`
`Stage
`
`CRP
`
`1
`2
`3
`4
`5
`6
`7
`8
`9
`10
`11
`12
`Median
`
`M
`F
`F
`F
`F
`F
`F
`F
`F
`F
`M
`F
`
`53
`70
`58
`71
`74
`63
`60
`58
`54
`53
`69
`64
`61·5
`
`IgG(cid:107)
`(cid:108)
`IgG(cid:107)
`IgG(cid:107)
`IgA(cid:107)
`IgG(cid:107)
`IgA(cid:108)
`IgG(cid:107)
`IgG(cid:107)
`IgG(cid:108)
`IgA(cid:107)
`*
`
`IIIa
`IIIa
`IIIa
`IIIa
`IIIa
`IIIa
`IIIa
`IIa
`IIIa
`IIIa
`IIIa
`IIIa
`
`8
`<3
`3
`12
`6
`4
`6
`<3
`4
`<3
`6
`76
`5
`
`IL6
`
`50
`3
`7
`22
`17
`13
`10
`41
`7
`5
`33
`9
`11·5
`
`(cid:98)2M
`
`1·9
`4·2
`4·4
`7·7
`5·4
`1·5
`3·3
`2·4
`4·0
`2·9
`2·8
`10·2
`3·7
`
`CRP, C-reactive protein (normal value <5 mg/l); IL6, interleukin-6 (normal value <3 pg/ml);
`(cid:98)2M, beta-2 microglobulin (normal value 1·1–2·4 mg/l).
`* Non-secretor.
`
`and dexamethasone) or VAD-like regimens, intermediate to
`high doses of melphalan; 70–140 mg/m2 with or without
`autologous bone marrow or peripheral stem cell support).
`Exclusion criteria were; age <18 or >75 years,
`life
`expectancy <3 months, Karnofsky score <60, diabetes
`mellitus, hypercalcaemia requiring treatment, recent allo-
`geneic bone marrow transplantation, creatinine level
`> 150 (cid:109)mol/l, co-existing malignancies and active infection.
`Pretreatment characteristics of these 12 patients are
`shown in Table I.
`Chimaeric monoclonal anti-IL6 antibody. A murine-human
`chimaeric anti-IL6 monoclonal antibody (chimaeric CLB
`IL6/8) was developed, containing the antigen-binding
`variable region of the murine anti-IL6 antibody (CLB IL6/
`8) (Brakenhoff et al, 1990) and the constant region of a
`human IgG1 kappa immunoglobulin. The neutralizing Mab
`CLB IL6/8 blocks binding of IL6 to the IL6 receptor (CD 126)
`(Ehlers et al, 1994) and has a high affinity for recombinant
`as well as native IL6 (Kd ¼ 6·25 · 10¹12
`M) (Brakenhoff et al,
`1990; van Zaanen et al, 1996). The chimaeric Mab was
`manufactured by Centocor, Leiden, The Netherlands.
`Treatment schedule. After obtaining written informed
`consent according to the guidelines of the participating
`institutes, each patient received two cycles of treatment with
`cMab. Both cycles (starting at day 1 and 28 respectively)
`consisted of 14 daily 2 h i.v. infusions of the cMab (see Fig 1).
`This schedule was chosen to study the possible occurrence
`and response to re-treatment of a rebound phenomenon (i.e.
`acceleration of disease activity following cessation of the
`antibody administration) as has been described in patients
`treated with murine anti-IL6 Mab at the moment therapy
`was stopped (Klein et al, 1991). Before each cycle a test dose
`(10 (cid:109)g) was given by slow i.v. push over 5 min. As none of the
`12 patients developed an immediate hypersensitivity reac-
`tion, in all cases treatment was started 15 min later. The first
`three patients received a daily dose of 5 mg of the cMab (total
`
`dose 140 mg), the next three patients received 10 mg/d (total
`dose 280 mg), patients 7–9 received 20 mg/d (total dose
`560 mg) and the last three patients received 40 mg/d (total
`dose 1120 mg).
`Levels of IL6 and anti-IL6 antibodies. During treatment with
`the chimaeric CLB IL6/8 almost all the IL6 in the plasma
`circulated as a complex with the antibody (van Zaanen et al,
`1996). IL6 levels were determined with the B9 bioassay as
`described before (Aarden et al, 1987; van Zaanen et al,
`1996). One unit of B9-stimulating activity was defined as the
`amount
`inducing half-maximal proliferation and corre-
`sponded to 1 pg of IL6. To determine the total IL6 level (i.e.
`free IL6 plus IL6 complexed to cMab), an excess (10 (cid:109)g/ml) of
`CLB IL6/14 was added to each well in order to displace IL6
`from its binding to the in vivo administered neutralizing
`chimaeric CLB IL6/8. CLB IL6/14 and CLB IL6/8 Mab
`recognize partly overlapping sites of IL6. However, CLB
`IL6/14 is not capable of inhibiting IL6 activity in the B9
`bioassay (Aarden, 1991). During treatment with the cMab
`actual
`free IL6 levels cannot be measured, because the
`dilution of the samples necessary for testing in the B9
`bioassay or ELISA immediately influences the equilibrium
`between IL6–cMab complex,
`free IL6 and free cMab.
`Therefore free IL6 levels were calculated using the
`Henderson-Hasselbalch equation, with the Kd, the daily
`serum cMab levels and the total IL6 levels as known
`parameters.
`Levels of the chimaeric CLB IL6/8 monoclonal antibody
`were determined using a radioimmunoassay as described in
`detail before (van Zaanen et al, 1996). The threshold of this
`assay is 0·5 ng/ml of antibody.
`Human anti-chimaeric antibody (HACA) levels were deter-
`mined using an ELISA. Briefly, the cMab (CLB IL6/8, IgG1-
`kappa) was coated overnight at room temperature (2 (cid:109)g/ml
`in 100 (cid:109)l well) on flat-bottomed microtitre plates. The plates
`were washed four times with PBS/Tween solution. Patients
`
`䉷 1998 Blackwell Science Ltd, British Journal of Haematology 102: 783–790
`
`IPR2018-00685
`Celgene Ex. 2037, Page 2
`
`

`

`Chimaeric Anti-IL6 in Advanced Multiple Myeloma
`
`785
`
`Fig 1. Mean serum levels of chimaeric anti-IL6
`Mab in the different dosage groups. Patients
`1–3 (a) ¼ 5 mg/d; patients 4–6 (b) ¼ 10 mg/d;
`patients 7–9 (c) ¼ 20 mg/d; patients 10–12
`(d) ¼ 40 mg/d. Patient 12 received treatment
`on days 1–4 and 16–29. Day 65: n ¼ 9,
`day 100: n ¼ 5.
`
`Patient 5 had fever and an urinary tract infection when
`admitted for the second treatment cycle on day 28. In patient
`9 anti-IL6 was stopped at day 34 because of pneumonia and
`septicaemia. In patient 12 (receiving anti-IL6 until day 5)
`treatment was interrupted because of fever. With antibiotic
`treatment her clinical condition improved and anti-IL6 was
`given from day 16 to day 29 (one treatment cycle of 14 d).
`A second cycle was not given at the patient’s request.
`Pretreatment values of IL6, CRP and B2M are shown in
`Table I. In all but one patient pretreatment IL6 levels were
`elevated. 6/12 patients had elevated CRP levels. There was
`no correlation between CRP and IL6 levels (r ¼ ¹0·13) in
`these patients. Serum (cid:98)2M was increased in nine patients.
`Patient 12 had a non-secreting myeloma, with large
`plasmacytomas on the chest wall and the right upper arm.
`The plasmacytoma on the chest wall was measured
`bidimensionally, before and daily during anti-IL6 treatment.
`Before treatment it had a diameter of 14 · 16 cm and a
`growth rate of 1·5 cm/week in two perpendicular directions.
`
`Levels of cMab and IL6
`Data regarding the pharmacokinetics have been published
`previously (van Zaanen et al, 1996). The median half-life
`time of this cMab was 17·8 d (range 7·8–39·7), with a
`median distribution volume of 6·0 litres (range 3·0–9·7).
`Peak serum levels of the cMab ranged between 6·7 (cid:109)g/ml
`(patient 1) and 288 (cid:109)g/ml (patient 11). No HACAs were
`found in any of the patients during the study period of 100 d.
`During treatment, accumulation of the cMab occurred due
`to its long half-life time (Fig 1). This resulted in high total
`serum IL6 levels, complexed with the cMab (van Zaanen et
`al, 1996). Calculated free IL6 levels decreased to <0·5 pg/ml
`during treatment in all patients (Fig 2).
`
`Toxicity
`During and after administration of the cMab, no changes in
`
`sera were added in different dilutions (1/50 up to 1/800) in
`HPE-buffer (CLB, Biotechnology, Amsterdam, The Nether-
`lands) and incubated for 2 h at room temperature. After
`washing, the plates were incubated with HRP-conjugated
`monoclonal mouse anti-human lambda light chain (KH29,
`CLB, Amsterdam, The Netherlands) in 100 (cid:109)l HPE buffer for
`1 h at room temperature. Subsequently, after washing, the
`bound peroxidase was detected at 450 nm in a Titertek
`Multiskan.
`C-reactive protein (CRP) was determined by nephelometry
`(Behring, Germany); detection level 3 mg/l; normal value
`<5 mg/l.
`Beta-2 microglobulin ((cid:98)2M) was determined by a micro-
`particle enzyme immunoassay (Abbott Laboratories, U.S.A.);
`normal value 1·1–2·4 mg/l.
`levels were determined once
`Paraprotein (M-protein)
`weekly by immunonephalometry (Behring Nephalometer
`100 Analyzer; Behring Diagnostics, Amsterdam, The
`Netherlands).
`Levels of IL6 and the cMab were determined daily from day
`0 until day 14 and day 28 until day 41 (samples were drawn
`before starting infusion of anti-IL6), and on days 17, 21, 44,
`48, 56 and 100.
`Response to treatment was defined as a decrease in M-
`protein level of >50% or a decrease in plasmacytoma size in
`patient 12.
`
`RESULTS
`
`Dosage of chimaeric anti-IL6
`Twelve patients (three in each dosage group) completed at
`least one treatment cycle of 14 d. Eight of
`them also
`completed a second 14 d treatment cycle. In the other four
`patients anti-IL6 treatment was stopped because they
`fulfilled a predefined stopping criterion: patient 1 required
`radiotherapy for neurological complications at day 30.
`
`䉷 1998 Blackwell Science Ltd, British Journal of Haematology 102: 783–790
`
`IPR2018-00685
`Celgene Ex. 2037, Page 3
`
`

`

`786 H. C. T. van Zaanen et al
`
`Fig 2. Free IL6 levels. (A) The eight patients who received two
`complete treatment cycles. Free IL6 levels <0·3 pg/ml during
`treatment in all patients. (B) The three patients who completed
`one treatment cycle of 14 d (see Results). Patient 1: anti-IL6 until
`day 30; patient 5: anti-IL6 until day 14; patient 9: anti-IL6 until day
`34. (C) Patient 12 received anti-IL6 on days 1–4 and 16–29.
`Because of
`fever of unknown origin, anti-IL6 treatment was
`interrupted at day 5.
`
`blood pressure, pulse rate or temperature were observed.
`These parameters were measured every 15 min for 2 h
`immediately after the administration of anti-IL6. No changes
`in liver function were found. Creatinine levels fluctuated in
`
`patients 2, 5, 10 and 12 but remained <150 (cid:109)mol/l; in the
`other eight patients no changes in renal
`function were
`found. Haemoglobin levels remained stable, while minor
`changes occurred in platelet and leucocyte counts.
`In
`patients 2 and 5 we observed a transient thrombocytopenia
`with a nadir of 24 and 58 · 109/l respectively (on days 56
`and 14,
`respectively). Before anti-IL6 treatment both
`patients had hypocellular bone marrow aspirate and
`biopsy (with a decreased number of megakaryocytes). In 6/
`12 patients (patients 2, 4, 7, 8, 9 and 11) a mild transient
`granulocytopenia was documented after one dose of anti-
`IL6. The median decrease in the number of granulocytes was
`32·5% (range 20–46%), with a median time to nadir of 1 d.
`The granulocytes returned to base-line levels within 2 d. No
`alterations in lymphocyte subsets were found.
`Three patients developed infectious complications; patient
`5 (with known recurrent urinary tract infections) had fever
`with low blood pressure due to a urinary tract infection with
`Escherichia coli on the day she was admitted for the second
`treatment cycle. Patient 9 died because of pneumonia and
`septicaemia with Staphylococcus aureus. Patient 12 developed
`fever of unknown origin on day 5. She was treated with
`antibiotics; all cultures remained negative.
`
`Clinical effects
`In all but one patient the M-protein level did not change
`during both cycles of anti-IL6 treatment. In eight patients
`(patients 1, 3, 4, 7–11) this indicated an unaffected disease
`course because they had similar M-protein levels at 2
`months before anti-IL6 treatment (Figs 3A and 3B). In
`patient 2 disease activity remained progressive during the
`first treatment cycle; this was reflected by a rise of M-protein
`and (cid:98)2M levels (77% and 145% increase in relation to day 0,
`respectively). However, during the second treatment cycle,
`M-protein and (cid:98)2M levels remained stable (Fig 3B). After day
`60 she received 70 mg/m2 melphalan intravenously, result-
`ing in a temporary decrease of M-protein level. In two
`patients (patients 5 and 6) a marked stabilization of the M-
`protein occurred during therapy (Fig 3B). This was also true
`for the growth rate of the plasmacytoma of patient 12. In
`two patients (patients 1 and 7) we observed a possible
`acceleration in the increase of the M-protein levels after
`stopping anti-IL6 (Fig 3B). Likewise, after the anti-IL6
`treatment was stopped in patient 12, the growth rate of
`the plasmacytoma was increased when compared to the
`growth rate before therapy (1·5 cm/week versus 2 cm/
`week).
`Immediately after starting the anti-IL6 treatment the CRP
`levels decreased to below detection level in all patients except
`for patient 12 in which CRP decreased from 76 to 10 mg/l.
`Other acute-phase proteins tested, pre-albumin, alpha-1
`antitrypsin and alpha-1 acid glycoprotein, remained stable
`and within normal ranges.
`
`Survival
`Six patients were still alive 9 months after treatment with
`anti-IL6. Three patients died within the study period of 100 d
`(patients 5, 9 and 12). Patient 5 went off study because of
`an urinary tract infection on day 28. After appropriate
`
`䉷 1998 Blackwell Science Ltd, British Journal of Haematology 102: 783–790
`
`IPR2018-00685
`Celgene Ex. 2037, Page 4
`
`

`

`Chimaeric Anti-IL6 in Advanced Multiple Myeloma
`
`787
`
`Fig 3. Percentage of M-protein levels before,
`during and after anti-IL6 treatment in relation
`to day 0 (day 0 ¼ 100%). Anti-IL6 cMab was
`given in two cycles of 14 d (two boxes below
`abscissa). (A) Patients 3, 4, 8, 9, 10 and 11; (B)
`patients 1, 2, 5, 6 and 7. At day 60, patient 2
`received melphalan i.v. (70 mg/m2).
`
`treatment she was discharged and died several weeks later at
`home from progressive disease. Patient 9 died on day 35 due
`to an irreversible septic shock. Although radiotherapy was a
`treatment option for the large plasmacytomas in patient 12,
`no further treatment was given according to the wish of the
`patient. She died shortly afterwards because of progressive
`disease with pleural effusion. Three patients died after day
`100 because of refractory multiple myeloma.
`
`DISCUSSION
`
`From this phase I dose-escalating study with chimaeric anti-
`IL6 Mab in patients with advanced MM, two important
`conclusions can be drawn. First, the use of this cMab is
`safe. Despite high levels of circulating cMab, no toxicity
`was observed and no HACAs were induced. Second,
`
`accumulation of the high-affinity cMab resulted in high
`levels of circulating IL6. However, this was in the form of
`biologically inactive complexes and did not
`result
`in
`acceleration of the disease during anti-IL6 treatment.
`
`Toxicity
`Although IL6 is a multi-functional cytokine, blocking its
`biological activity by chimaeric anti-IL6 Mabs did not result
`in serious side-effects. Only a transient thrombocytopenia
`occurred in two patients who had hypocellular bone marrow
`before starting anti-IL6 treatment. The mechanism of this
`thrombocytopenia during anti-IL6 treatment is not com-
`pletely understood. Although from studies in primates and
`humans it has become evident that IL6 is able to induce
`megakaryocyte maturation and thrombocytopoiesis (Asano
`et al, 1990; Stahl et al, 1991; Wickenhauser et al, 1995;
`
`䉷 1998 Blackwell Science Ltd, British Journal of Haematology 102: 783–790
`
`IPR2018-00685
`Celgene Ex. 2037, Page 5
`
`

`

`788 H. C. T. van Zaanen et al
`Stouthard et al, 1996), its role in steady-state thrombo-
`cytopoiesis
`is not clear because IL6 knock-out mice
`have normal platelet counts. Thrombopoietin (TPO) levels
`did not change during anti-IL6 treatment (unpublished
`observations).
`Three patients developed infectious complications during
`therapy. Although a relationship with anti-IL6 cannot be
`totally excluded, it is not likely because infectious complica-
`tions are common in end-stage multiple myeloma and are a
`major cause of death (Foerster, 1993). Moreover, in IL6
`knock-out mice the outcome of TNF-induced shock was not
`different from IL6 wild-type mice (Libert et al, 1994). The
`three patients involved (patients 5, 9 and 12) were still able
`to respond to the infection with an increase of
`their
`endogenous IL6 production, resulting in increased free IL6
`serum levels, fever and increased CRP levels.
`
`High levels of circulating IL6
`In agreement with other studies (Heremans et al, 1992;
`Martens et al, 1993; May et al, 1993, 1994; Mihara et al,
`1991), we found that the use of anti-IL6 Mab led to
`accumulation of circulating IL6 in the form of
`immune
`complexes. An important question is whether this complexed
`IL6 has biological activity. If so, treatment with anti-IL6
`might result in disease progression and/or clinical deteriora-
`tion. Several observations make this possibility unlikely.
`First, during anti-IL6 treatment the CRP level (which is
`completely IL6 dependent; Heinrich et al, 1990) decreased to
`below detection level in 11/12 patients. In the remaining
`patient CRP decreased by 87% compared to the pretreatment
`value. Second, we did not observe an accelerated disease
`course during anti-IL6 treatment. However, after anti-IL6
`was stopped, in three patients a possible rebound phenom-
`enon was seen, resulting in increased M-protein levels in two
`patients and an accelerated plasmacytoma growth in
`one patient. A possible explanation might be that when free
`cMab levels decrease some time after stopping administration
`of cMab,
`IL6 gradually dissociates from the cMab/IL6
`complex. The free IL6 level will increase according to the
`Henderson-Hasselbalch equation: Kd ¼ [IL6free] [cMabfree] /
`[complex].
`It has been suggested that the rebound phenomenon
`might be prevented by the formation of multivalent IL6/Mab
`complexes which are rapidly cleared from the circulation.
`Indeed in mice it has been found that simultaneous
`treatment with three anti-IL6 Mab (binding to three distinct
`epitopes of IL6) induced a rapid uptake of these complexes by
`the liver, leading to enhanced elimination of IL6 from the
`circulation (Montero-Julian et al, 1995).
`
`whereas mature myeloma cells display a low proliferative
`activity with higher secretion of M-protein (Kawano et al,
`1993). Joshua et al (1996) found that the in vivo labelling
`index was almost entirely determined by a subpopulation of
`‘primitive’ (¼immature) plasma cells. It is possible that
`myeloma cell proliferation and CRP production are blocked
`by anti-IL6 cMab, whereas M-protein production by mature
`myeloma cells is less affected. This assumption is supported
`by the observation by Sonneveld et al (1991) that in vitro Ig-
`synthesis and proliferation of MM cells are stimulated by
`different cytokines and that anti-IL6 can block plasma cell
`proliferation, but not Ig-synthesis. Indeed,
`for the three
`patients in whom we were able to analyse the plasma cell
`labelling index before and after anti-IL6 treatment we found
`a strong decrease in LI (data not shown).
`The existence of IL6-independent myeloma cells in these
`end-stage patients might be another explanation for the
`minor
`clinical
`responses during anti-IL6 treatment.
`Decreased IL6 dependence of myeloma cells in the course
`of the disease has been described (Asaoku et al, 1988). On
`the other hand, Zhang et al (1992) have shown that in the
`terminal phase of MM and plasma cell leukaemia, in vitro
`tumour growth was totally dependent on IL6. Moreover,
`from the pleural effusion of patient 12 we were able to obtain
`an IL6-producing myeloma cell line. IL6 production and
`growth of this cell
`line can totally be blocked by anti-
`IL6 cMab (unpublished observation). Likewise, in the two
`patients (patients 2 and 3) in whom we were able to test it,
`the myeloma cells were still IL6-dependent in vitro.
`In vitro studies showed that IL6 (which is overproduced in
`MM) reversed the growth arrest of dexamethasone on all
`myeloma cell lines tested (Juge-Morinea et al, 1995). Thus,
`neutralization of IL6 by anti-IL6 Mab might improve the
`response of MM patients to dexamethasone. Therefore in
`future studies it would be relevant to combine this chimaeric
`anti-IL6 Mab with, for example, dexamethasone or VAD. An
`additional theoretical advantage of the use of anti-IL6
`antibodies could be the beneficial effects on myeloma-
`associated bone disease. Several data suggest that IL6,
`probably in synergy with IL-1 beta and TNF, also plays an
`important role in the pathogenesis of this aspect of the
`disease (Kurihara et al, 1990; Bataille et al, 1992; Roodman,
`1995; Barille et al, 1995).
`In conclusion, this phase I dose-escalating study indicated
`that treatment of end-stage MM patients with chimaeric
`anti-IL6 Mabs had a very low toxicity. A dose of 40 mg/d for
`14 d can safely be used in future phase II studies.
`
`REFERENCES
`
`Efficacy
`Although this phase I dose-escalating study does not allow
`formal conclusions as to efficacy, it was rather striking to see
`that during anti-IL6 treatment we observed only minor
`effects on M-protein levels, whereas a profound decrease of
`CRP levels was found in all patients. A possible explanation
`might be that myeloma cells in the bone marrow of MM
`patients consist of both immature and mature cells.
`Immature myeloma cells are considered to be proliferating,
`
`Aarden, L. (1991) Plasmacytomas. Mechanisms of B-cell Neoplasm
`(ed. by F. Melchens and M. Potter), pp. 419–433. Editione Roche,
`Basel.
`(1987)
`Aarden, L., De Groot, E., Schaap, O. & Lansdorp, P.
`Production of hybridoma growth factor by human monocytes.
`European Journal of Immunology, 17, 1411–1416.
`Anderson, K., Jones, R., Morimoto, C., Leavitt, P. & Barut, B. (1989)
`Response patterns of purified myeloma cells to hematopoietic
`growth factors. Blood, 73, 1915–1924.
`Asano, S., Okano, A., Ozawa, K., Nakahota, T., Ishibashi, T., Koike,
`
`䉷 1998 Blackwell Science Ltd, British Journal of Haematology 102: 783–790
`
`IPR2018-00685
`Celgene Ex. 2037, Page 6
`
`

`

`789
`Chimaeric Anti-IL6 in Advanced Multiple Myeloma
`K., Kimura, H., Tanioko, Y., Hirano, T., Kishimoto, T., Takahu, F. &
`immature and mature myeloma cells in the bone marrow of
`Akiyoma, Y.
`(1990)
`In vivo effects of recombinant human
`human myelomas. Blood, 82, 564–570.
`interleukin-6 in primates: stimulated production of platelets.
`Kishimoto, T. (1989) The biology of interleukin-6. Blood, 74, 1–10.
`Blood, 75, 1602–1605.
`Klein, B., Wijdenes,
`J., Zhang, X.G.,
`Jourdan, M., Boiron,
`J.,
`Iwato, K., Tanabe, O., Tanaka, T.,
`Asaoku, H., Kawano, M.,
`Brochier, J., Liautard, J., Merlin, M., Clement, C., Morel-Fournier, B.,
`Hirano, T., Kishimoto, T. & Kuramoto, A. (1988) Decrease in
`Lu, Z., Mannoni, P., Sany, J. & Bataille, R. (1991) Murine anti-
`BSF-2/IL-6 response in advanced cases of multiple myeloma.
`interleukin-6 monoclonal antibody therapy for a patient with
`Blood, 72, 429–432.
`plasma cell leukemia. Blood, 78, 1198–1204.
`Barille, S., Colette, M., Bataille, R. & Amiot, M. (1995) Myeloma cells
`Klein, B., Zhang, X.G., Jourdan, M., Boiron, J., Portier, M., Lu, Z.,
`upregulate IL6 secretion in osteoblastic cells through cell-to-cell
`Wijdenes, J., Brochier, J. & Bataille, R. (1990) IL6 is the central
`contact but downregulate osteocalcin. Blood, 86, 3151–3159.
`tumor growth factor in vitro and in vivo in multiple myeloma.
`Bataille, R., Barlogie, B., Lu, Z., Rossi, J., Lavabre-Bertrand, T.,
`European Cytokine Network, 1, 193–201.
`Beck, T., Wijdenes, J., Brochier, J., & Klein, B. (1995) Biologic
`Klein, B., Zhang, X.G., Jourdan, M., Content, J., Houssiau, J.,
`effects of anti-interleukin 6 murine monoclonal antibody in
`Aarden, L., Piechaczyk, M. & Bataille, R. (1989) Paracrine rather
`advanced multiple myeloma. Blood, 86, 685–691.
`than autocrine regulation of myeloma cell growth and differ-
`Bataille, R., Chappard, D. & Klein, B. (1992) Mechanism of bone
`entiation by IL6. Blood, 73, 517–526.
`lesions. In: ‘Multiple myeloma’. Hematology/Oncology Clinics of
`Kurihara, N., Bertolini, D., Suda, T., Akiyama, Y. & Roodman, G.
`North America, 6, 285–295.
`(1990) IL6 stimulates osteoclast-like multinucleated cell forma-
`Bataille, R., Jourdan, M., Zhang, X.G. & Klein, B. (1989) Serum levels
`tion in long-term human marrow cultures by inducing IL-1
`of IL6, a potent myeloma cell growth factor, as a reflect of disease
`release. Journal of Immunology, 144, 4226–4230.
`severity in plasma cell dyscrasias. Journal of Clinical Investigation,
`Libert, C., Takahashi, N., Cauwels, A., Brouckaert, P., Bluethmann, H. &
`84, 2008–2011.
`Fiers, W. (1994) Response of IL-6 deficient mice to TNF-induced
`Brakenhoff, J., Hart, M., De Groot, E., Di Padova, F. & Aarden, L.
`metabolic changes and lethality. European Journal of Immunology,
`(1990) Structure–function analysis of human IL6: epitope
`24, 2237–2242.
`mapping of neutralizing monoclonal antibodies with amino- and
`Lokhorst, H., Lamme, M., de Smet, M., Klein, M., de Weger, R.,
`carboxyl-terminal deletion mutants. Journal of Immunology, 145,
`van Oers, R. & Bloem, A. (1994) Primary tumor cells of myeloma
`561–568.
`patients induce IL6 secretion in long-term bone marrow cultures.
`Durie, B. & Salmon, S. (1975) A clinical staging system for multiple
`Blood, 84, 2269–2277.
`myeloma. Cancer, 36, 842–854.
`Martens, E., Dillen, C., Put, W., Heremans, H., Van Damme, J. &
`Ehlers, M., Grotzinger, J., de Hon, F., Mullberg, J., Brakenhoff, J.,
`Billiau, A. (1993) Increased circulating interleukin 6 activity in
`Liu, J., Wollmer, A. & Rose-John, S. (1994) Identification of two
`endotoxin-challenged mice pretreated with anti-IL6 antibody is
`novel regions of human IL-6 responsible for receptor binding and
`due to IL6 accumulated in antigen–antibody complexes. European
`signal transduction. Journal of Immunology, 153, 1744–1751.
`Journal of Immunology, 23, 2026–2029.
`Foerster, J. (1993) Multiple myeloma. Wintrobe’s Clinical Hematology,
`May, L., Neta, R., Moldawer, L., Kenney, J., Patel, K. & Sehgal, P.
`9th edn (ed. by G. Lee, Th. Bithell, J. Foerster, J. Athens and
`(1993) Antibodies chaperone circulating IL-6: paradoxical effects
`J. Lukens), p. 2222. Lea and Febiger, Philadelphia.
`of anti-IL6 ‘neutralizing’ antibodies in vivo. Journal of Immunology,
`Heinrich, P., Castell, J. & Andus, T. (1990) Interleukin-6 and the
`151, 3225–3236.
`acute phase response. Biochemical Journal, 265, 621–636.
`May, L., Patel, K., Garcia, D., Ndubuisi, M., Ferrone, S., Mittelman, A.,
`Heremans, H., Dillen, C., Put, W., Van Damme, J. & Billiau, A. (1992)
`Mackiewicz, A. & Sehgal, P. (1994) Sustained high levels of
`Protective effect of anti-IL6 antibody against endotoxin, associated
`circulating chaperoned interleukin-6 after active specific cancer
`with paradoxically increased IL6 levels. European Journal of
`immunotherapy. Blood, 84, 1887–1895.
`Immunology, 22, 2395–2401.
`Mihara, M., Koishihara, Y., Fukui, H., Yasukawa, K. & Ohsugi, Y.
`Hilbert, D., Kopf, M., Mock, B., Kohler, G. & Rudikoff, S. (1995)
`(1991) Murine anti-human IL6 monoclonal antibody prolongs
`Interleukin 6 is essential for in vivo development of B lineage
`the half-life time in circulating blood and thus prolongs the
`neoplasms. Journal of Experimental Medicine, 182, 243–248.
`bioactivity of human IL6 in mice. Immunology, 74, 55–59.
`Hoffman, T.
`(1990) Anticipating, recognizing and preventing
`Montero-Julian, F., Klein, B., Gautherot, E. & Brailly, H. (1995)
`hazards associated with in vivo use of monoclonal antibodies:
`Pharmacokinetic study of IL6 therapy with monoclonal anti-
`special considerations related to human anti-mouse antibodies.
`bodies: enhancement of IL6 clearance by cocktails of anti-IL6
`Cancer Research, 50, 1049s–1050s.
`antibodies. Blood, 85, 917–924.
`Joshua, D., Petersen, A., Brown, R., Pope, B., Snowden, L. & Gibson, J.
`Nordan, R., Pumphrey, J. & Rudikoff, S. (1987) Purification and
`(1996) The labelling index of primitive plasma cells determines the
`NH2-terminal sequence of a plasmacytoma growth-factor derived
`clinical behaviour of patients with myelomatosis. British Journal of
`from the murine macrophage cell
`line P 388 D1. Journal of
`Haematology, 94, 76–81, 1996.
`Immunology, 139, 813–817.
`Juge-Morinea, N., Francois, S., Puthier, D., Godard, A., Bataille, R. &
`Roodman, G. (1995) Osteoclast function in Paget’s disease and
`Amiot, M. (1995) The gp130 family cytokines IL-6, LIF and OSM
`multiple myeloma. Bone, 17, 57–61S.
`but not
`IL-11 can reverse the anti-proliferative effect of
`Sonneveld, P., Schoester, M. & de Leeuw, K. (1991) In vitro Ig-
`dexamethasone on human myeloma cells. British Journal of
`synthesis and proliferative activity in multiple myeloma are
`Haematology, 90, 707–710.
`stimulated by different growth factors. British Journal of Haema-
`Kawano, M., Hirano, T., Matsuda, T., Taga, T., Horii, Y., Iwato, K.,
`tology, 79, 589–594.
`Asaoku, H., Tang, B., Tanaka, H., Kuramoto, A. & Kishimoto, T.
`Stahl, C., Zucker-Franklin, D., Evatt, B. & Winton, E. (1991) Effects of
`(1988) Autocrine generation and requirement of BSF 2/IL6 for
`human Interleukin-6 on megakaryocyte development and
`human multiple myelomas. Nature, 332, 83–85.
`thrombocytopoiesis in primates. Blood, 78, 1467–1475.
`Kawano, M., Huang, N., Harada, H., Harada, Y., Sakai, A.,
`Stouthard, J., Goey, H., de Vries, E., de Mulder, P., Groenewege

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