throbber

`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`Clin Exp Immunol 2002; 130:75–84
`
`
`
`Blackwell Science, Ltd
`CEIClinical and Experimental Immunology
`Oxford, UK
`0009-9104Blackwell Publishing Ltd, 2002
`130
`
`Original Article
`
`J. B. Marriott
`
`et al.
`
`Diverse effects of Thd and its analogues on TNF-

`
`/TNFR2 during T cell co-stimulation
`
`αααα
`Thalidomide and its analogues have distinct and opposing effects on TNF-
` and
`TNFR2 during co-stimulation of both CD4
` and CD8
` T cells
`+
`+
`
`J. B. MARRIOTT, I. A. CLARKE, K. DREDGE, G. MULLER*, D. STIRLING* & A. G. DALGLEISH
`*
`Division of Oncology, Department of OGEM, St George’s Hospital Medical School, London, UK, and
`Celgene Corporation,
`Warren, New Jersey, USA
`
`(Accepted for publication 28 June 2002)
`
`SUMMARY
`
`Thalidomide (Thd) is clinically useful in a number of conditions where its efficacy is probably related to

` activity. More recently, Thd has also been shown to co-stimulate T cells and second gen-
`its anti-TNF
`-
`eration co-stimulatory (IMiD™) analogues are currently being assessed in the treatment of cancer
`patients. However, in contrast to their known suppressive effects during inflammatory stimuli, the

` and TNF receptors (TNFRs) during T cell co-stimulation are not
`effects of Thd/IMiDs on TNF-
`known. We sought to determine the effect of Thd, two clinically relevant IMiDs (CC-4047, ACTIMID™

` production
`and CC-5013, REVIMID™) and a non-stimulatory SelCID analogue (CC-3052) on TNF-
`and on the expression and shedding of TNFRs during co-stimulation. We found that co-stimulation of

`CD3-induced T cell surface expression of TNFR2
`PBMC with Thd/IMiDs, but not CC-3052, prevented
`and thereby reduced soluble TNFR2 (sTNFR2) levels. However, there was no effect on total (surface/
`intracellular) TNFR2 protein expression, suggesting inhibition of trafficking to the cell membrane. The

` produc-
`extent of co-stimulation by Thd/IMiDs (assessed by CD69/CD25 expression and IL-2/sIL-2R
`+
`+
` and CD8
` T lymphocytes and correlated with TNFR2 inhibition. Co-
`tion) was similar for CD4
`stimulation, but not the early inhibitory effect on TNFR2, was IL-2-dependent and led to increased

`+
`+
` production by both CD4
` and CD8
` T lymphocytes. The clinical relevance of this observation
`TNF-

` during REVIMID™ treatment of patients with
`was confirmed by the elevation of serum TNF-
`advanced cancer. Together, these results suggest a possible role for TNF-mediated events during co-
`stimulation and contrast with the TNF inhibitory effects of Thd and its analogues during inflammatory
`stimuli.
`
`Keywords
`
`co-stimulation immunomodulation thalidomide T lymphocyte TNF
`
`INTRODUCTION
`
`Thalidomide (Thd) is emerging as an important immunothera-
`peutic drug in a number of clinical situations [1–4]. It is well estab-

` agent in the context of monocyte/
`lished as an anti-TNF-

` [5,6] and clinically during TNF-
`-
`macrophage activation
`in vitro
`mediated inflammatory disease [6]. However, other immunomod-
`ulatory properties may explain the extensive range of its clinical
`activity. More recently, it has been shown to inhibit monocyte IL-
`12 production [7] and also to be able to induce the activation and
`+
` T cells stimulated via the T cell receptor in
`proliferation of CD8
`the absence of co-stimulation [8]. Thus, in certain clinical settings,
`such as in the treatment of cancer patients, Thd may act as an
`
`Correspondence: Dr J. B. Marriott, Division of Oncology, St George’s
`Hospital Medical School, Cranmer Terrace, Tooting, London, SW17 ORE,
`UK.
`E-mail: jmarriot@sghms.ac.uk
`
`© 2002 Blackwell Publishing Ltd
`
`adjuvant to promote T-cell responses. However, Thd treatment

` [9–
`has also been known to increase circulating levels of TNF-
`12] and IL-12 [13] and it appears likely that the effects of Thd vary,
`depending on the relative predominance of monocyte/macroph-
`age and T cell activation. Furthermore, the effects of Thd may also
`vary depending on the activation of particular signalling pathways
`[14].
`Significant side effects such as somnolence and peripheral
`neuropathy are associated with clinically effective Thd treatment.
`However, the design and synthesis of Thd analogues has yielded
`compounds with enhanced activity/toxicity profiles [15,16]. These
`analogues are currently being characterized and have been shown
`to segregate into at least two distinct classes. The SelCIDs™
`(selective cytokine inhibitory drugs) consist of phosphodiesterase
`type 4 (PDE4) inhibitors [17]. The IMiDs™ (immunomodulatory
`drugs) are thought to be mechanistically similar to Thd, although
`with enhanced potency and therapeutic indices, and act via as yet
`75
`
`IPR2018-01714
`Celgene Ex. 2009, Page 1
`
`

`

`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`76
`
`J. B. Marriott
`
` et al.
`
`unknown mechanism(s) [18–24]. Both groups of compounds

`,
`are potent inhibitors of monocyte/macrophage-derived TNF-
`although T cell co-stimulatory activity is limited to the IMiD
`group [17].
`Thd and its co-stimulatory IMiD analogues, in particular CC-
`4047 and CC-5013, are currently being assessed in the treatment
`of patients with advanced multiple myeloma and patients with
`advanced solid tumours [25–32]. Furthermore, we have shown
`recently that CC-4047, when co-administered with autologous or
`allogeneic tumour cell vaccination, is able to enhance protective
`and long-lasting immunity significantly in a murine model of col-
`orectal cancer [33].

` during
`Thd and its derivatives are known to suppress TNF-
`the activation of inflammatory pathways by LPS, but it is not
`known whether this is the case during the direct stimulation of T
`cell activation pathways. Therefore, in this study we planned to
`determine the effect of these compounds, plus the non-co-

` and its
`stimulatory analogue CC-3052, on the expression of TNF-
`. Furthermore, we wanted
`receptors during co-stimulation
`in vitro
`+
`+
` and CD8
` populations
`to assess the co-stimulatory effect on CD4
`+
` T cells were acti-
`because previous results suggested that CD8
`vated preferentially. Because signals generated via the TNF sys-
`tem are important in T cell homeostatic control we hoped to
`provide insights into the T cell co-stimulatory mechanism of these
`novel compounds. Finally, we hoped to show that any effects on
`the TNF system were relevant for clinical non-inflammatory dis-
`ease and to this end we obtained sera from a small phase I study
`of CC-5013 in the treatment of advanced cancer.
`
`MATERIALS AND METHODS
`
`PBMC and whole blood culture
`Thd, two IMiDs (CC-4047 and CC-5013) and a SelCID (CC-3052)
`(provided by Celgene Corporation, NJ, USA) were dissolved
`freshly in DMSO to make a 20 mg/ml stock solution. The com-
`pounds were then diluted directly into the tissue culture wells at
`the required concentrations.
`Venous blood was collected into sodium heparin vacutainers
`(Becton Dickinson, Oxford, UK) and PBMC were prepared sub-
`sequently by density centrifugation over Ficoll Hypaque (Sigma,
`g
` for 20 min cells were
`Poole, UK). After centrifugation at 700
`collected from the interface, washed three times in HBSS

`6
` 10
`/ml in complete
`(Sigma), counted and resuspended at 1–2
`RPMI-1640 medium supplemented with 10% fetal calf serum
` L-glutamine (all
`(FCS), 1% penicillin/streptomycin and 2 m
`M

`−

` Thd/analogues (1 ng
`10
`g/ml;
`Sigma). PBMC were cultured
`0·05%v/v final DMSO concentration) at 1 ml/well in 24-well

`g/ml; R&D systems,
`plates, precoated with anti-CD3 mAb (2·5
`Minneapolis, USA) in bicarbonate coating buffer pH 9·5. PBMC
`−

`7 days at 37
`C in 5% CO
`. For some
`were incubated for 12 h
`2
`experiments PBMC were co-incubated with neutralizing anti-IL-

`g/ml; R&D systems). Cell-free supernatants
`2 mAb (0·005–0·1
`−

`70
`C until assayed by
`were collected and stored in aliquots at
`ELISA. Cells were harvested and resuspended in PBS/BSA prior
`to processing as described below.
` serotype
`For lipopolysaccharide (LPS; Sigma,
`Escherichia coli
`0127:B8)-stimulated cultures heparinized venous blood was
`diluted 1 : 4 in complete RPMI-1640 medium (without FCS) and

`g/ml). All
`stimulated in 24-well plates at 1 ml/well with LPS (1


`C in 5% CO
`
` Thd/analogues
`cultures were incubated at 37
`2

`g/ml). After 24–48 h cell-free supernatants were collected by
`(10
`
`microcentrifugation and stored in aliquots at
`by ELISA.
`
`−
`

`
`70
`
`C until assayed
`
`Clinical CC-5013 study: patient selection, treatment and sample
`collection
`We collected serum from a small phase I study designed to assess
`the safety, tolerability and efficacy of REVIMID™ (CC-5013) in
`the treatment of patients with advanced and heavily pretreated
`cancer. Of the 20 patients enrolled we obtained samples from 11
`subjects, aged between 18 and 75 years, with histologically proven
`=
`
` 6), histological or cytological
`stage IV metastatic melanoma (
`n
`=
`
` 2) or
`proof of adenocarcinoma of the exocrine pancreas (
`n
`=
`
`other malignancy (one each of breast, renal and lung cancer;
`n
`3). Before initiating therapy, patients were subject to a complete
`medical history, physical examination and baseline evaluation of
`signs and symptoms, including full blood counts and neurological
`examination. These were repeated at weekly intervals during the
`study period and also at the end of the study.
`All patients gave written informed consent prior to participa-
`tion in the study. The protocol was approved by the Wandsworth
`Local Research Ethics Committee (LREC). All patients were
`treated with 5 mg oral REVIMID™ daily (tablets were taken
`every evening) for 1 week with the dose escalating to 10 mg daily
`at week 2, 25 mg daily at week 3 and 50 mg daily at week at
`which the treatment was maintained indefinitely depending on
`tolerance.
`Blood was taken at the same time at each visit (and therefore
`at the same time relative to drug administration), collected into
`serum separator (SST; Vacutainer, BD) tubes and left to clot for
`∼
`g
`30 min. Tubes were spun at 950
` for 10 min and serum collected.
`−

`70
`C until being assayed for TNF-
`Sera were frozen in aliquots at


` and sIL-2 receptor
` (sIL-2R) by ELISA. Serum was collected
`at baseline, during the first 1–3 weeks (10–25 mg daily) of treat-
`ment and again at weeks 4–5 (50 mg daily).
`REVIMID™ tablets were manufactured by Penn pharmaceu-
`ticals (Tredegar, Wales, UK) and supplied by Celgene Corpora-
`tion (Warren, NJ, USA).
`
`Enzyme-linked immunosorbent assay (ELISA)
`Culture supernatants were assayed for sTNFR1, sTNFR2, sIL-2R
`using kits provided by R&D Systems (Minneapolis, MN, USA)

` using an assay procedure and reagents
`and for IL-2 and TNF-
`provided by Pharmingen (BD). Briefly, 96-well flat-bottomed

`C with anti-
`ELISA plates (Nunc) were precoated overnight at 4
`cytokine capture mAb in bicarbonate coating buffer, pH 9·5 and
`) prior to incubation of standards and
`washed (with PBS/Tween
`20

`C. After washing, plates
`test (supernatant) samples overnight at 4
`were incubated with biotinylated anticytokine mAb for 1 h. After
`further washing, plates were developed using chromogen (tetram-
`ethylbenzidine)/hydrogen peroxide for 20–30 min at RT. Stan-
`dard absorbances (405 nm) of duplicate wells (minus control zero
`standard) were used to calculate concentration of cytokine/recep-
`tor levels which were corrected for any dilution factor. Data are
`expressed as percentage compared to control (DMSO alone),
`which is represented as 100%.

`, sTNFR2, IL-2
`Sera from patients were assayed for TNF-
`and sIL-2R using kits provided by R&D Systems. In each case,
`the manufacturer’s instructions were followed exactly. Data are
`expressed as pg/ml at baseline (pretreatment) compared to fol-
`low-up at 1–3 and 4–5 weeks.
`
`© 2002 Blackwell Publishing Ltd,
`
`
`
`Clinical and Experimental Immunology,
`
`
`
`130:75–84
`
`IPR2018-01714
`Celgene Ex. 2009, Page 2
`
`

`

`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`
`a
`Diverse effects of Thd and its analogues on TNF-
`/TNFR2 during T cell co-stimulation
`
`77
`
`Flow cytometric analysis of surface CD25, CD69, TNFR1 and
`TNFR2
`PBMC were harvested from 24-well plates by gentle aspiration,
`washed and suspended in wash buffer (PBS/0·5% BSA/0·1%

`CD3-
`sodium azide). Three-colour phenotypical analysis of
`stimulated PBMC was performed by surface staining cells (by

`C) with the following fluoro-
`incubating cells for 30 min at 4
`chrome conjugated monoclonal antibodies: anti-CD25 FITC
`(Becton Dickinson Immunocytometry Systems, BDIS, Oxford,
`UK), anti-CD69 FITC (L78; BDIS) or anti-TNFR1 FITC
`(16803·161; R&D Systems) and anti-TNFR2 PE (22235·311;
`R&D Systems) with either anti-CD4 PerCP (SK3; BDIS) or anti-
`CD8 PerCP (SK1; BDIS) plus appropriate isotype-matched and
`compensation controls. Cells were then washed once in 2 ml wash

`l Cellfix™ (BDIS) prior to
`buffer and fixed for analysis in 200
`flow cytometric analysis. Upon analysis lymphocytes were gated
` side scatter (SSC) properties.
`on forward scatter (FSC)
`versus
`PerCP-positive T cell subsets were then selected by gating on SSC
` FL-3 dot-plots and displayed as two-colour dot-plots, with
`versus
`quadrants set according to isotype-matched controls. For each
`sample 10 000 lymphocytes were acquired on a Becton Dickinson
`FACScan using CellQuest™ software.
`

`Analysis of intracellular TNFR2, IL-2 and TNF-


`PBMC were stimulated with
`CD3
` CC-4047 for 24–96 h with

`g/ml; Sigma)
`the protein transport inhibitor Brefeldin A (10
`present for the last 6 h of culture to allow intracellular accumu-
`lation of protein. Cells were harvested, washed and suspended in
`wash buffer, then fixed and permeabilized using the specially for-
`mulated Becton Dickinson FACS lysis and permeabilization solu-
`tions as per the manufacturer’s instructions. Cells were then
`stained with anti-TNFR2 FITC, anti-IL-2 FITC (5344·111, BDIS)

` FITC (6401·1111, BDIS) and anti-CD4 PE or anti-
`or anti-TNF-
`CD8 PE plus appropriate isotype-matched and compensation
`controls for 30 min before washing once in 2 ml wash buffer fol-

`l Cellfix™. Stained samples
`lowed by a final resuspension in 200

`C prior to two-colour analysis on the
`were kept in the dark at 4
`permeabilized cells. Upon flow cytometric analysis lymphocytes
` side scatter (SSC)
`were gated on forward scatter (FSC)
`versus
`properties and displayed as two-colour dot-plots, with quadrants
`set according to isotype-matched controls. For each sample 10 000
`lymphocytes were acquired on a Becton Dickinson FACScan
`using CellQuest™ software.
`
`Proliferation assay

`5
`PBMC (1–2
` 10
`) were resuspended in complete RPMI-1640


` Thd/analogues) onto
`CD3 precoated 96-well
`round-
`(

`l per well.
`bottomed plates (Falcon, BD) to a final volume of 200

`3
`C in 5% CO
` for 5 days with [
`H]-
`Cells were cultured at 37
`2
`thymidine (NEN Life Science Products, Hounslow, UK) added
`for the last 24 h (2 mCi/well). Cells were harvested onto glass fibre
`filter paper and counted in a matrix 96 direct beta counter (Can-
`3
`H]-thymidine was determined as
`berra Packard). Uptake of [
`mean cpm of sextuplet culture wells and expressed as percentage
`change compared to control (DMSO alone).
`
`Statistical analysis
`For analysis of serum data comparisons between groups were
`examined by the Kruskal–Wallis test with inclusion of the Dunn’s
`
`multiple comparison test. The statistics were performed using
`Primer of Biostatistics (3·02) statistical software.
`
`RESULTS
`
`Co-stimulatory IMiD CC-4047 strongly inhibits the secretion

`of sTNFR2 by
`CD3-stimulated PBMC cultures whereas
`non-co-stimulatory SelCID CC-3052 augments sTNFR2
`A single treatment of CC-4047 leads to potent inhibition of

`CD3-stimulated PBMC supernatants at 24 h
`sTNFR2 levels in
`(Fig. 1a). This effect was consistent in four of four experiments


`=
`CD3 alone, 2676 pg/ml; + CC-4047
` 32·3%
`(24 h: mean
`23·5% of control). Inhibition was decreased at 48 h (mean


`=
`CD3 alone, 4436 pg/ml; + CC-4047
` 52·7%
` 13% of control),
`although a small inhibitory effect was still apparent after 72 h.
`Soluble TNFR2 was not consistently detectable in 12 h cultures
`(data not shown).
`We found that the non-co-stimulatory analogue, CC-3052,
`had the opposite effect on sTNFR2 when compared to CC-4047 in

`=
` 222%
` 141% of control). The
`all experiments (24 h: + CC-3052

`=
` 159%
`effect of CC-3052 also decreased at 48 h (+ CC-3052
`50·4%) and a smaller augmentary effect was apparent at 72 h.
`Thd had very little overall effect on sTNFR2 (for example, at

`=
` 102·7%
` 9·3% of control).
`48 h: + Thd
`In contrast to the effects on sTNFR2 none of the compounds
`had a consistent or significant effect on the levels of sTNFR1 at


` 10·9%; CC-3052, mean 86·8%
`24 h (+ CC-4047, mean 110%

`=
`13·6%; Thd
` 104%
` 11·2%; Fig. 1b). However, CC-3052 did
`inhibit sTNFR1 at 48 and 72 h.
`We also found that CC-4047 and Thd strongly induced the


` at 72 h (mean
`CD3 alone control, 1443 pg/
`production of TNF-


`=
`=
` 365%
` 233%; + Thd
` 247%
` 122%; Fig. 1c),
`ml; + CC-4047

`CD3, 2753 pg/ml;
`although there was very little effect at 24 h (


`=
`=
` 114%
` 37·8%; Thd
` 129%
` 39·8%).
`CC-4047
`
`sTNFR2 production by LPS-stimulated whole blood cultures
`was not affected by CC-4047, Thd or CC-3052
`We next determined whether the divergent effects of CC-4047
`and CC-3052 on sTNFR2 production seen during T cell co-

`CD3 were also apparent during LPS activation
`stimulation with
`of monocyte/macrophage cell populations. In contrast to the
`effect during T cell activation we found that there was no effect of
`any compound on sTNFR1 or sTNFR2 production by LPS-
`stimulated whole blood cultures at any time-point (Fig. 1d,e).
`As expected, CC-3052 and CC-4047 both strongly inhibited LPS-

` production (Fig. 1f) and were more effective than
`induced TNF-
`Thd in this respect.
`
`CC-4047 and CC-5013 strongly reduced the surface expression

`+
`+
`of TNFR2 on
`CD3-stimulated CD4
` and CD8
` T cells during
`PBMC co-stimulation
`We then determined whether the inhibitory effect of CC-4047 on
`supernatant levels of sTNFR2 during co-stimulation was due to
`reduced surface TNFR2 expression or to reduced receptor shed-
`ding. We found that CC-4047 and CC-5013 (and to a lesser extent
`Thd) reduced the surface expression of TNFR2 and that this
`correlated with the induction of surface CD25 (IL-2 receptor)
`+
` (Fig. 2a)
`(Fig. 2). Furthermore, this was seen equally in both CD4
`+
` populations (Fig. 2b). Results from two dose–response
`and CD8
`experiments indicated that co-stimulation (gauged by CD69
`
`© 2002 Blackwell Publishing Ltd,
`
`
`
`Clinical and Experimental Immunology,
`
`
`
`130:75–84
`
`IPR2018-01714
`Celgene Ex. 2009, Page 3
`
`

`

`
`
`
`
`24
`
`48
`h
`
`72
`
`96
`
`24
`
`48
`
`72
`
`h
`
`J. B. Marriott et al.
`
`(c)
`
`500
`
`400
`
`300
`
`200
`
`100
`
`0
`
`0
`
`(f)
`
`% TNF-alpha relative to control
`
`500
`
`400
`
`300
`
`200
`
`100
`
`0
`
`0
`
`% TNF-alpha relative to control
`
`24
`
`48
`h
`
`72
`
`96
`
`24
`
`48
`
`72
`
`h
`
`(b)
`
`500
`
`400
`
`300
`
`200
`
`100
`
`% TNFR1 relative to control
`
`0
`
`0
`
`(e)
`
`500
`
`400
`
`300
`
`200
`
`100
`
`% TNFR1 relative to control
`
`0
`
`0
`
`24
`
`48
`h
`
`72
`
`96
`
`24
`
`48
`
`72
`
`h
`
`(a)
`
`500
`
`400
`
`300
`
`200
`
`100
`
`0
`
`0
`
`(d)
`
`500
`
`400
`
`300
`
`200
`
`100
`
`0
`
`0
`
`78
`
`% TNFR2 relative to control
`
`% TNFR2 relative to control
`
`Fig. 1. Co-stimulatory CC-4047 strongly inhibits the secretion of sTNFR2 by αCD3-stimulated PBMC cultures (but not LPS-stimulated
`cultures) whereas non-co-stimulatory SelCID CC-3052 augments sTNFR2. Divergent effects of CC-4047 (Ο), CC-3052 (䊐), and Thd (∆)
`on the production of sTNFR2, sTNFR1 & TNF-α by PBMC-stimulated with αCD3 (a–c) and whole blood cultures stimulated with LPS
`(d–f). Cultures were incubated ± Thd/analogues (10 µg/ml) and supernatants collected at the indicated times as per Materials and methods.
`Results are expressed as percentage change compared to control cultures with DMSO alone. Data presented are from four normal donors
`used in two separate experiments.
`
`expression) and inhibition of surface TNFR2 could both be
`detected at CC-4047 concentrations as low as 10 ng (data not
`shown).
`We were unable to detect the presence of surface TNFR1
`either with αCD3 alone or in the presence of the co-stimulatory
`compounds using cells from a total of four donors in two separate
`experiments (data not shown).
`
`CC-4047 did not alter the level of total cellular TNFR2
`(intracellular plus surface) in T cells during PBMC co-
`stimulation
`We investigated whether the inhibitory effect of CC-4047 on sur-
`face TNFR2 was due to the inhibition of TNFR2 protein produc-
`tion. We utilized the protein transport inhibitor Brefeldin A and
`two-colour flow cytometry in order to measure the total amount
`of cellular TNFR2 within CD4+ and CD8+ cells during PBMC co-
`stimulation. The data presented in Fig. 3 show that when total
`protein was analysed CC-4047 did not appear to exert an inhibi-
`tory effect on TNFR2. The data presented suggest strongly that
`TNFR2 protein production itself is not being affected. Therefore,
`it is likely that trafficking to the cell surface that is being inhibited.
`Furthermore, our data also rule out an inhibitory effect on
`TNFR2 transcription.
`
`The relative extent of TNFR2 inhibition by CC-4047, CC-5013
`and Thd corresponds to co-stimulatory activity as gauged by
`the production of IL-2, sIL-2R and proliferation in αCD3
`stimulated PBMC cultures
`We then investigated whether there was a possible connection
`between the inhibitory effect of these compounds on TNFR2 sur-
`face expression and co-stimulatory activity. The co-stimulatory
`activity of CC-4047, CC-5013 and Thd was assessed by IL-2 and
`sIL-2R production and also by the incorporation of tritiated thy-
`midine (to measure proliferation).
`We found that IL-2 production was not detected in control
`(αCD3 alone) cultures. However, after 24 h CC-4047, CC-5013
`and, to a lesser extent Thd, strongly induced IL-2 production
`(Fig. 4a). For CC-4047 this effect was still apparent after 7 days of
`culture. The increased expression of sIL-2R became apparent
`after 72 h and followed a very similar pattern (Fig. 4b). Prolifer-
`ation was assessed at days 5–6 with a consistent pattern of results
`(Fig. 4c). Therefore, using these three markers of co-stimulation
`the same relative pattern of effectiveness emerged: CC-
`4047 > CC-5013 >> Thd.
`In contrast to these effects the non-co-stimulatory SelCID,
`CC-3052, strongly inhibited IL-2 and sIL-2R although it had no
`effect on proliferation, as we have shown previously [19].
`
`© 2002 Blackwell Publishing Ltd, Clinical and Experimental Immunology, 130:75–84
`
`IPR2018-01714
`Celgene Ex. 2009, Page 4
`
`

`

`Diverse effects of Thd and its analogues on TNF-a/TNFR2 during T cell co-stimulation
`
`79
`
`(a)
`
`a CD3 alone
`
`a CD3 + CC-4047
`
`a CD3 + CC-5013
`
`a CD3 + Thd
`
`100101102103104
`
`100 101 102 103 104
`CD25 FITC
`
`TNF-R2 PE
`
`100 101 102 103 104
`CD25 FITC
`
`100101102103104
`
`TNF-R2 PE
`
`100 101 102 103 104
`CD25 FITC
`
`100101102103104
`
`TNF-R2 PE
`
`100 101 102 103 104
`CD25 FITC
`
`100101102103104
`
`48·81
`
`17·38
`
`22·28
`
`46·33
`
`100101102 103 104
`TNF-R2 PE
`
`100101102 103 104
`TNF-R2 PE
`
`100101102 103 104
`TNF-R2 PE
`
`100101102 103 104
`TNF-R2 PE
`
`114·03
`
`250·22
`
`210·49
`
`165·96
`
`Events
`
`Events
`
`100101102 103 104
`CD25 FITC
`
`100101102 103 104
`CD25 FITC
`
`100101102 103 104
`CD25 FITC
`
`100101102 103 104
`CD25 FITC
`
`(b)
`
`a CD3 alone
`
`a CD3 + CC-4047
`
`a CD3 + CC-5013
`
`a CD3 + Thd
`
`100101102103104
`
`100101102103104
`
`100 101 102 103 104
`CD25 FITC
`
`TNF-R2 PE
`
`100 101 102 103 104
`CD25 FITC
`
`TNF-R2 PE
`
`100 101 102 103 104
`CD25 FITC
`
`100101102103104
`
`TNF-R2 PE
`
`100 101 102 103 104
`CD25 FITC
`
`100101102103104
`
`TNF-R2 PE
`
`TNF-R2 PE
`
`41·18
`
`10·73
`
`14·97
`
`35·84
`
`100101102 103 104
`TNF-R2 PE
`
`100101102 103 104
`TNF-R2 PE
`
`100101102 103 104
`TNF-R2 PE
`
`100101102 103 104
`TNF-R2 PE
`
`99·20
`
`149·64
`
`149·19
`
`145·49
`
`Events
`
`Events
`
`100101102 103 104
`CD25 FITC
`
`100101102 103 104
`CD25 FITC
`
`100101102 103 104
`CD25 FITC
`
`100101102 103 104
`CD25 FITC
`
`Fig. 2. CC-4047 and CC-5013 strongly reduced the surface expression of TNFR2 on αCD3 stimulated CD4+ and CD8+ T cells during PBMC
`co-stimulation. PBMC were incubated ± Thd/analogues (10 µg/ml) for 48 h. Three-colour flow cytometric analysis was performed by surface
`staining stimulated PBMC with anti-CD25 FITC, anti-TNFR2 PE and anti-CD4/CD8 PerCP plus appropriate isotype matched and
`compensation controls as described in Materials and methods. (a) Dot plots represent TNFR2 versus CD25 (IL-2 receptor) gated on (a)
`CD4 and (b) CD8 T cell subsets. Histograms show mean fluorescent intensity of TNFR2 expression on CD25+ T cells. This effect was highly
`consistent when performed in 10 separate experiments each using PBMC from two normal donors although data from a single represen-
`tative experiment are shown.
`© 2002 Blackwell Publishing Ltd, Clinical and Experimental Immunology, 130:75–84
`
`IPR2018-01714
`Celgene Ex. 2009, Page 5
`
`

`

`80
`
`J. B. Marriott et al.
`
`Isotype matched
`control
`
`Unstimulated
`
`a CD3 alone
`
`a CD3 + CC-4047
`
`CD4 PE
`
`CD4 PE
`
`CD4 PE
`
`IgG1 FITC
`
`TNF-R2 FITC
`
`TNF-R2 FITC
`
`TNF-R2 FITC
`
`CD8 PE
`
`CD8 PE
`
`CD8 PE
`
`CD4 PE
`
`CD4
`
`CD8 PE
`
`CD8
`
`IgG1 FITC
`
`TNF-R2 FITC
`
`TNF-R2 FITC
`
`TNF-R2 FITC
`
`Fig. 3. CC-4047 did not alter the level of total cellular TNFR2 (intracellular plus surface) in T cells during PBMC co-stimulation.
`Unstimulated PBMC and cells stimulated with αCD3 ± CC-4047 (10 µg/ml) for 24 h (TNFR2) (the last 6 h in the presence of the protein
`transport inhibitor, Brefeldin A) were harvested and processed as described in Materials and methods. Lymphocytes were gated on forward
`scatter (FSC) versus side scatter (SSC) properties and displayed as two-colour dot-plots, with quadrants set according to isotype-matched
`controls. No difference was seen in the expression of intracellular TNFR2 during the co-stimulation of CD4+ or CD8+ T cells. The data
`presented are representative of three separate experiments in which very similar results were obtained.
`
`(b)
`
`(c)
`
`CC-
`3052
`
`CC-
`4047
`
`CC-
`5013
`
`Thd
`
`500
`
`400
`
`300
`
`200
`
`100
`
`0
`
`% proliferation relative to control
`
`CC-
`3052
`
`CC-
`4047
`
`CC-
`5013
`
`Thd
`
`400
`
`300
`
`200
`
`100
`
`0
`
`% sIL-2R (at 72 h) relative to control
`
`CC-
`3052
`
`CC-
`4047
`
`CC-
`5013
`
`Thd
`
`(a)
`
`900
`
`800
`
`700
`
`600
`
`500
`
`400
`
`300
`
`200
`
`100
`
`0
`
`% IL-2 (at 24 h) relative to control
`
`Fig. 4. The relative extent of TNFR2 inhibition by CC-4047, CC-5013 and Thd corresponds to the degree of co-stimulation by these
`compounds. PBMC cultures were co-incubated for 24 h (IL-2), 72 h (sIL-2R) and 120 h (proliferation) ± Thd/analogues (10 µg/ml) as
`described in Materials and methods. Cell culture supernatants were assayed for IL-2 and sIL-2R by ELISA. Proliferation was assessed by
`[3H]-thymidine incorporation. Results are expressed as percentage change in (a) IL-2, (b) sIL-2R and (c) proliferation compared to control
`cultures. Data are from four normal donors used in two separate experiments.
`
`Co-stimulation by CC-4047 and subsequent TNF-α production,
`but not early inhibition of TNFR2, is dependent on IL-2-
`mediated signalling
`We next determined the importance of IL-2-mediated signalling
`for the process of CC-4047-mediated co-stimulation. We wanted
`to determine whether elevation of TNF-α was a consequence of T
`cell activation and not a direct effect of CC-4047. We also wanted
`to confirm that TNFR2 inhibition was not due to co-stimulation
`itself and raise the possibility that this early effect may be signif-
`icant in the initiation of the co-stimulatory process.
`Inhibition of IL-2 signalling was achieved with the use of a
`neutralizing anti-IL-2 mAb precoated onto the culture well.
`Under co-stimulatory conditions anti-IL2 mAb decreased super-
`natant IL-2 levels in a dose-dependent manner (Fig. 5a) and also
`
`decreased the surface expression of IL-2 receptor (data not
`shown). In these same cultures inhibition of IL-2 prevented the
`superinduction of TNF-α in a similar dose–response fashion
`(Fig. 5b) Therefore, co-stimulation-induced TNF-α production is
`clearly mediated via IL-2 generated signalling. However, in iden-
`tical studies we found that the inhibition of IL-2-mediated signal-
`ling did not prevent TNFR2 inhibition by CC-4047 (data not
`shown).
`
`Co-stimulation-induced IL-2 and TNF-α is derived from both
`CD4+ and CD8+ T cells
`Having established a requirement for IL-2 production and (sub-
`sequent IL-2-mediated signalling) during CC-4047- mediated
`co-stimulation we sought to determine the source of this
`
`© 2002 Blackwell Publishing Ltd, Clinical and Experimental Immunology, 130:75–84
`
`IPR2018-01714
`Celgene Ex. 2009, Page 6
`
`

`

`Diverse effects of Thd and its analogues on TNF-a/TNFR2 during T cell co-stimulation
`
`81
`
`+
`–
`
`+
`+
`+
`+
`0·1 0·05 0·01 0·005
`
`(b)
`
`100
`
`80
`
`60
`
`40
`
`20
`
`co-stimulated cultures
`% TNF-alpha relative to
`
`0
`CC-4047
`+ anti-IL-2 mAb
`
`(a)
`
`100
`
`80
`
`60
`
`40
`
`20
`
`co-stimulated cultures
`
`% IL-2 relative to
`
`0
`CC-4047
`+ anti-IL-2 mAb
`
`+
`–
`
`+
`+
`+
`+
`0·1 0·05 0·01 0·005
`
`Fig. 5. Co-stimulation by CC-4047 and subsequent TNF-α production, but not early inhibition of TNFR2, is dependent on IL-2-mediated
`signalling. PBMC cultures were stimulated with αCD3 in the presence of CC-4047 (10 µg/ml) and anti-IL-2 mAb (0·1–0·005 µg/ml) as
`described in Materials and methods. Results are expressed as percentage values relative to cultures co-stimulated with αCD3 + CC-4047
`(0% represents αCD3 alone). Cultures with increasing concentrations of anti-IL-2 mAb show a reduction in (a) supernatant IL-2 levels
`and (b) supernatant TNF-α levels. Data shown are from four normal donors in two separate experiments.
`
`Unstimulated
`
`0·61%
`
`a CD3 alone
`2·74%
`
`a CD3 + CC-4047
`8·72%
`
`CD4 PE
`
`8·04%
`
`IL-2 FITC
`
`IL-2 FITC
`
`CD4 PE
`
`1·43%
`
`IL-2 FITC
`
`IL-2 FITC
`
`CD4 PE
`
`0·81%
`
`CD8 PE
`
`CD8 PE
`
`CD8 PE
`
`Unstimulated
`
`0·71%
`
`a CD3 alone
`1·57%
`
`a CD3 + CC-4047
`14·86%
`
`CD4 PE
`36·34%
`
`TNF-a FITC
`
`TNF-a FITC
`
`CD4 PE
`6·22%
`
`TNF-a FITC
`
`TNF-a FITC
`
`CD4 PE
`1·32%
`
`(a)
`
`IL-2 FITC
`
`CD4
`
`IL-2 FITC
`
`CD8
`
`(b)
`
`TNF-a FITC
`
`TNF-a FITC
`
`CD4
`
`CD8
`
`CD8 PE
`
`CD8 PE
`
`CD8 PE
`
`Fig. 6. Co-stimulation-induced IL-2 and TNF-α are derived from both CD4+ and CD8+ T cells. Unstimulated PBMC and cells stimulated
`with αCD3 ± CC-4047 (10 µg/ml) for 96 h (IL-2/TNF-α) (the last 6 h in the presence of the protein transport inhibitor, Brefeldin A) were
`harvested and processed as described in Materials and methods. Lymphocytes were gated on forward scatter (FSC) versus side scatter
`(SSC) properties and displayed as two-colour dot-plots, with quadrants set according to isotype-matched controls. The induction of
`intracellular IL-2 and TNF-α was detected in both CD4+ and CD8+ T cells. Data shown are from one normal donor and are representative
`of very similar data obtained from three donors.
`
`© 2002 Blackwell Publishing Ltd, Clinical and Experimental Immunology, 130:75–84
`
`IPR2018-01714
`Celgene Ex. 2009, Page 7
`
`

`

`82
`
`J. B. Marriott et al.
`
`(b)
`
`6000
`
`4000
`
`2000
`
`TNF-alpha (ng/mL)
`
`(a)
`
`2400
`
`2000
`
`1600
`
`1200
`
`800
`
`400
`
`sIL-2R (pg/mL)
`
`0
`Baseline
`
`Weeks 1–3 Weeks 4–5
`
`0
`Baseline Weeks 1–3 Weeks 4–5
`
`Fig. 7. Treatment of advanced cancer patients with CC-5013/REVIMID™ results in increased TNF-α and sIL-2 receptor levels. Patient
`serum samples were obtained from a small phase I study as detailed in Materials and methods. Serum was collected at baseline and twice
`after commencing CC-5013 treatment (weeks 1–3 and weeks 4–5) and assayed for (a) sIL-2R and (b) TNF-α by ELISA. Each symbol
`represents a single patient.
`
`cytokine. In particular we wished to determine conclusively
`whether co-stimulation affects both CD4+ and CD8+ T cells. This
`was achieved by intracellular cytokine specific staining of CD4+
`and CD8+ T cells within the PBMC population. Data are shown
`from one of two separate experiments in which almost identical
`data were obtained. Figure 6a confirms the lack of IL-2 produc-
`tion observed by ELISA in control (αCD3 alone) cultures
`because very few cells from either subset produce IL-2. However,
`upon CC-4047-mediated co-stimulation there are increased num-
`bers of IL-2-producing cells from both T cell subsets. Further-
`more, the elevated production of TNF-α, which is secondary to
`co-stimulation, is also due to both T cell types (Fig. 6b). Thus,
`co-stimulation by CC-4047 affects CD4+ and CD8+ T cell subsets
`rather than inducing preferential activation of CD8+ cells, as has
`been reported previously [8].
`
`Treatment of cancer patients with CC-5013/REVIMID™ leads
`to increased serum levels of sIL-2R and TNF-α
`We wanted to extend our in vitro observations and determine
`whether the clinical use of a co-stimulatory IMiD (CC-5013/
`REVIMID™) could lead to elevation of serum TNF-α in the c

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