throbber

`
`
`www.bloodjournal.orgFrom
`
`by guest
`
`
`
`For personal use only.on September 13, 2018.
`
`NEOPLASIA
`
`Expression of tumor necrosis factor–related apoptosis-inducing ligand, Apo2L,
`and its receptors in myelodysplastic syndrome: effects on in vitro hemopoiesis
`Dae Young Zang, Ray G. Goodwin, Michael R. Loken, Eileen Bryant, and H. Joachim Deeg
`
`Tumor necrosis factor–related apoptosis-
`inducing ligand (TRAIL), a member of the
`tumor necrosis factor (TNF) family, binds
`to several cell-surface receptors with dis-
`tinct functions (agonistic receptors 1 and
`2 [TRAIL-R1, TRAIL-R2]; decoy receptors
`3 and 4 [TRAIL-R3, TRAIL-R4]). Expres-
`sion and function was characterized in
`patients with myelodysplastic syndromes
`(MDSs). While normal marrow showed
`negligible expression of TRAIL and recep-
`tors (except TRAIL-R3), TRAIL and all
`receptors were constitutively expressed
`Introduction
`
`in MDS marrow. Following TRAIL expo-
`sure, MDS marrow showed significant
`increases in apoptosis, whereas normal
`marrow, except for a subset of CD341
`precursors, did not (P5 .012). Marrow
`from 21 patients with MDS was then
`propagated in long-term cultures in the
`presence or absence of TRAIL. While in
`advanced MDS (refractory anemia with
`excess blasts in transformation [RAEB-T]
`and tAML [MDS transformed into AML]),
`colony numbers decreased in the presence
`of TRAIL (63.0% 6 10.4% of untreated group
`
`[100%]), numbers increased in patients with
`RA or RAEB (160.2% 6 90.5% of untreated
`group). TRAIL eliminated preferentially
`clonally abnormal cells as identified by
`chromosomal markers. Thus, TRAIL and
`receptor expression differed significantly
`between normal and MDS marrow, and
`TRAIL modulated in vitro hemopoiesis in
`MDS dependent upon disease stage but
`not, to a detectable extent,
`in normal
`marrow. (Blood. 2001;98:3058-3065)
`
`© 2001 by The American Society of Hematology
`
`The myelodysplastic syndromes (MDSs) comprise a heteroge-
`neous group of hemopoietic stem cell disorders usually character-
`ized by a cellular marrow with dysplastic features, peripheral blood
`cytopenias, and a variable incidence of transformation into acute
`myeloid leukemia (AML).1 MDS generally occurs in elderly
`the median age at diagnosis being 65 to 70 years.2
`patients,
`Allogeneic hemopoietic stem cell
`transplantation is currently
`considered the only curative treatment modality for MDS; how-
`ever, age and performance status of patients and lack of histocom-
`patible donors limit
`the applicability of this treatment. Also,
`regimen-related toxicity is a major problem in patients with MDS
`undergoing transplantation.3,4 It would be desirable, therefore, to
`develop new therapeutic strategies to improve or restore normal
`hemopoietic function.
`One mechanism invoked to explain the apparent discrepancy
`between cellular marrow and peripheral blood cytopenias in
`patients with MDS is programmed cell death (apoptosis), which
`occurs with increased frequency in MDS marrow.5,6 Several
`cytokines or ligands known to have proapoptotic properties, such
`as interleukin-1b (IL-1b), tumor necrosis factor a (TNFa), and
`Fas-ligand are up-regulated in many patients with MDS.5,7-9 We
`and others have shown that blockade of TNFa or Fas-ligand
`enhances hemopoietic colony transformation from MDS marrow in
`vitro and improves blood cell counts in vivo.5,7,10 However, it is
`clear from those studies that the regulation of hemopoiesis in MDS
`is complex and multiple factors are involved. The TNF-related
`apoptosis-inducing ligand (TRAIL), also known as Apo2 ligand
`
`(Apo2L), is a member of the TNF family and induces apoptosis
`preferentially in transformed and tumor cells but generally not in
`normal cells.11-13 However, these observations must be extrapolated
`with caution because TRAIL effects on normal cells, for example,
`in the brain, have been documented.14 In MDS marrow, normal
`precursors and clonally abnormal cells typically coexist. If expo-
`sure to TRAIL resulted in selective elimination of the abnormal
`clone, TRAIL might be a useful
`therapeutic agent for MDS,
`certainly for marrow purging (and autologous stem cell transplanta-
`tion) if not for in vivo treatment. The exact mechanism by which
`TRAIL eliminates tumor cells preferentially or selectively is not
`known. One possibility is differential expression of agonistic
`receptors 1 and 2 (TRAIL-R1 and -R2, also known as death
`receptors 4 [DR4] and DR5) and antagonistic or modulatory
`receptors, TRAIL-R3 and -R4, also known as decoy receptors 1
`(DcR1) and DcR2.15,16 An alternative mechanism would be differ-
`ential expression of, or functional defects in, cytoplasmatic inhibi-
`tors of apoptosis such as the Fas-associated death domain protein
`(FADD)17 or FADD-like IL-1b–converting enzyme (ICE)–
`inhibitory protein (FLIP).18
`The objective was to exploit the potential selective killing of
`clonal and, presumably, malignant cells by human TRAIL.
`Thus, we determined the effect of TRAIL on apoptosis and
`hemopoiesis in long-term marrow cultures (LTMC) of normal
`and MDS marrow, and characterized the expression of TRAIL
`and its receptors. We also characterized the expression of FLIP
`in marrow cells from patients with MDS and healthy donors.
`
`From the Clinical Research Division, Fred Hutchinson Cancer Research
`Center and the Department of Medicine, University of Washington; Immunex;
`and Hematologics, Seattle, WA.
`
`Supported by PHS grants HL36444, CA18029 and CA87948. H.J.D. is also
`supported by a grant from the Gabrielle Rich Leukemia Fund.
`
`Submitted March 14, 2001; accepted July 6, 2001.
`
`ligand with Genentech.
`
`Reprints: H. Joachim Deeg, Fred Hutchinson Cancer Research Center, 1100
`Fairview Ave N, D1-100, PO Box 19024, Seattle, WA 98109-1024; e-mail:
`jdeeg@fhcrc.org.
`
`The publication costs of this article were defrayed in part by page charge
`payment. Therefore, and solely to indicate this fact, this article is hereby
`marked ‘‘advertisement’’ in accordance with 18 U.S.C. section 1734.
`
`Ray G. Goodwin owns stock in Immunex, which is codeveloping TRAIL/Apo2
`
`© 2001 by The American Society of Hematology
`
`3058
`
`BLOOD, 15 NOVEMBER 2001 z VOLUME 98, NUMBER 10
`
`Dr. Reddy’s Laboratories, Inc. v. Celgene Corp.
`IPR2018-01504
`Exhibit 2018, Page 1
`
`

`

`
`
`
`www.bloodjournal.orgFrom
`
`by guest
`
`
`
`For personal use only.on September 13, 2018.
`
`BLOOD, 15 NOVEMBER 2001 z VOLUME 98, NUMBER 10
`
`TRAIL AND APOPTOSIS IN MDS
`
`3059
`
`Materials and methods
`
`Marrow cells
`
`Bone marrow (BM) samples were obtained from 44 patients with MDS
`(refractory anemia [RA], n 5 20; RA with ring sideroblasts [RARS], n 5 4;
`RA with excess blasts [RAEB], n 5 9; RAEB in transformation [RAEB-T],
`n 5 9; chronic myelomonocytic leukemia [CMML], n 5 2), 6 patients
`whose disease had transformed to AML, and 20 healthy volunteer donors.
`Patients and healthy donors had given informed consent according to the
`procedures approved by the Institutional Review Board of the Fred
`Hutchinson Cancer Research Center (FHCRC). Samples were processed as
`described19,20 for in vitro hemopoiesis, phenotyping, characterization of
`apoptosis, RNA extraction, and cytogenetic analysis with fluorescence in
`situ hybridization (FISH).
`
`Cell lines
`
`Human acute myelogenous leukemia cell line KG1, acute T-cell leukemia
`cell line Jurkat, and human colon cancer cell line Colo 205 were obtained
`from American Type Culture Collection (Rockville, MD).
`
`Antibodies and reagents
`
`The following monoclonal antibodies (mAbs) were obtained from commer-
`cial sources: fluorescein isothiocyanate (FITC) or phycoerythrin (PE)-
`conjugated mouse anti–human CD3 (IgG2a), CD13 (IgG1), CD14 (IgG2a),
`CD34 (HPLC-2, IgG1), and CD45 (IgG1) (Becton Dickinson, San Jose,
`CA, or Caltag, San Francisco, CA); FITC or PE-conjugated mouse F(ab9)2
`IgG1 or IgG2a or IgG2b (Becton Dickinson) or mouse IgM (Catalag) as
`isotype controls; FITC or PE-conjugated goat F(ab9)2 anti–mouse IgG
`(H1L) or IgM (Caltag) as secondary antibodies; rabbit anti–mouse IgG to
`carboxy terminus human FLIP long form for Western blot (Calbiochem, La
`Jolla, CA). The mouse anti–human IgM STRO-1 mAb, which recognizes a
`stromal cell surface antigen,21 was obtained from Developmental Studies
`Hybridoma Bank (Iowa City, IA). The monoclonal mouse anti–human
`TRAIL (IgG1, M180, blocking; M184, nonblocking), TRAIL-R1 (IgG2a,
`M271), TRAIL-R2 (IgG1, M413), TRAIL-R3 (IgG1, M430), TRAIL-R4
`(IgG1, M444), as well as stem cell factor (SCF), and leucine zipper
`(LZ)–tagged soluble trimeric human TRAIL were supplied by Immunex
`(Seattle, WA). IL-1, IL-3, IL-6, granulocyte colony-stimulating factor
`(G-CSF), and granulocyte-macrophage colony-stimulating factor (GM-
`CSF) were purchased from R & DSystems (Minneapolis, MN).
`
`Phenotyping of marrow cells
`
`Phenotypic analysis of marrow cells was carried out by flow cytometry as
`described.5 For determination of the expression of TRAIL and its receptors, 105
`cells were incubated with 10 mg/mL mAb against human TRAIL or TRAIL-R1
`to -R4 for 30 minutes on ice. After washes in phosphate buffered saline (PBS),
`cells were incubated for 30 minutes on ice with PE-conjugated (secondary) goat
`F(ab9)2 anti–mouse IgG. For 2-color immunofluorescence, FITC-conjugated
`mAbs for CD3, CD19 (for the lymphocyte gate), CD13 (for the granulocyte
`gate), CD14 (for the monocyte gate), and CD34 (for the hemopoietic progenitor
`cell gate) were incubated after staining with mAbs to human TRAIL and
`TRAIL-R1 to -R4, PE-conjugated (secondary) goat F(ab9)2 anti–mouse IgG.
`Blast cells were isolated on the basis of orthogonal light scatter and CD45
`labeling as described.22,23 Cells labeled with irrelevant isotype-matched mAb and
`incubated with the secondary detection antibody served as negative controls.
`Data on at least 10 000 events were collected using a FACScan (Becton
`Dickinson) and analyzed using CellQuest software (Becton Dickinson).
`
`Reverse transcription–polymerase chain reaction amplification
`
`RNA preparation and reverse transcription (RT) were carried out as
`described.5,18 Total RNA was prepared from cell pellets using the RNeasy
`Total RNA kit (Qiagen, Chatsworth, CA). Cells were lysed in buffer RLT (4
`M guanidinium isothiocyanate, 0.5% sarcosyl, and 0.1 M 2b-mercaptoetha-
`nol) and homogenized using a QIAshredder spin column. After centrifuga-
`
`tion, an equal volume of 70% ethanol was added to the lysate, and the
`mixture was applied onto RNeasy spin columns. Total RNA, which binds to
`the silica gel membrane under high salt-buffer conditions, was washed with
`buffers RW1 and RPE (supplied with the kit), and then eluted in
`DEPC-treated water. Finally, any remaining DNA was removed by
`digestion with RNase-free DNase. The concentration and purity of the RNA
`was determined by measuring the absorbance at A260/A280 and by gel
`electropheresis.
`To synthesize cDNA, 1 mg the total RNA (generally corresponding to
`’ 0.5 3 106 cells) was combined with 1 mL Oligo (dT)12-18 (500 mg/mL;
`Gibco/BRL, Gaithersburg, MD) and heated for 5 minutes to 70°C; the
`resulting heteroduplex was resuspended to 20 mL in RT buffer and
`incubated for 30 minutes at 37°C with 200 U Moloney murine leukemia
`virus (M-MLV) reverse transcriptase (Gibco/BRL). After adding a second
`100 U aliquot M-MLV, the reaction was continued for an additional 15
`minutes prior to heat-inactivating the enzyme for 2 minutes at 80°C. cDNA
`was stored at 270°C. Samples were then thawed in batches for polymerase
`chain reaction (PCR) amplification.
`The cDNA was amplified by PCR in a reaction mixture consisting of 50
`mM KCl, 20 mM Tris-HCl (pH 8.4), 0.1% Triton X-100, 1 mM DTT, 2.0
`mM MgCl2, all 4 dNTPs (each at 0.2 mM), 10 pmol of each oligonucleotide
`primer (see below), and 2.5 units Taq DNA Polymerase (Boehringer
`Mannheim). Amplification was performed in 0.5-mL Gene Amp tubes in a
`final volume of 50 mL. The PCR mixes were overlaid with mineral oil and
`amplified for 30 to 40 cycles (see below) denaturation (at 94°C for 1
`minute), annealing (at 55°C to 65°C [see below] for 1 minute, and extension
`at 72°C for 1 minute). PCR products were size-fractionated and analyzed by
`1.5% agarose gel electropheresis and normalized according to the amount
`of b-actin in the same cDNA sample. PCR product sizes were determined
`from comparison to the Ready-Load 100 base pair (bp) DNA standard
`ladder (Gibco/BRL).
`PCR reactions were performed using the following primers: b-actin (for-
`ward: 59-TCCTGTGGCATCCACGAAACT-39, reverse: 59-ATCGTCCACCG-
`CAAATGCTTC-39); TRAIL (forward: 59-GGAACCCAAGGTGGGTAGAT-
`39, reverse: 59-TCTCACCACACTGCAACCTC-39); TRAIL-R1 (forward: 59-
`CTGAGCAACGCAGACTCGCTGTCCAC-39, reverse: 59-TCCAAGGAC-
`ACGGCAGAGCCTGTGCCAT-39); TRAIL-R2 (forward: 59-GCCTCATGGA-
`CAATGAGATAAAGGTGGCT-39, reverse: CCAAATCTCAAAGTACGCA-
`CAAACGG-39); TRAIL-R3 (forward: 59-GAAGAATTTGGTGCCAATGCCA-
`CTG-39, reverse: 59-CTCTTGGACTTGGCTGGGAGATGTG-39); TRAIL-R4
`(forward: 59-CTTTTCCGGCGGCGTTCATGTCCTTC-39, reverse: 59-GTT-
`TCTTCCAGGCTGCTTCCCTTTGTAG-39); and long form of FLIP (forward:
`59-AATTCAAGGCTCAGAAGCGA-39, reverse: 59-GGCAGAAACTCTGCT-
`GTTCC-39). The sizes of the expected products were 304, 192, 506, 502, 612,
`453, and 226 bp, respectively. Human b-actin PCR-cycle conditions were 95°C
`for 45 seconds, 55°C for 1 minute, and 72°C for 45 seconds for 30 cycles. Human
`TRAIL-R1, -R2, -R3, and FLIPL conditions were 94°C for 1 minute, 55°C for 1
`minute, 72°C for 1 minute for 30 cycles. Human TRAIL-R4 conditions were
`95°C for 4 minutes 15 seconds, followed by 30 cycles of 95°C for 45 seconds,
`60°C for 45 seconds, and 72°C for 45 seconds for 30 cycles.18 Samples were
`resolved on 2% agarose gels and visualized with ethidium bromide.
`
`Western blot analysis
`
`Western blots were performed as previously described.18 Briefly, marrow
`mononuclear cells (MMNCs) from patients with MDS or from healthy
`donors were lysed in PBS containing 1% Nonidet P-40, 0.35 mg/mL PMSF,
`9.5 mg/mL leupeptin, and 13.7 mg/mL pepstatin A. The lysed cells were
`washed, and protein concentrations of the extracts were determined by
`colorimetric assay (Bio-Rad, Richmond, CA). Equal amounts of protein
`were separated by sodium dodecyl sulfate–polyacrylamide gel electrophore-
`sis (SDS-PAGE), transferred to polyvinylidine difluoride (PVDF; Bio-
`Rad), and blocked with 5% nonfat dry milk in 1 3 Tris buffered saline for 1
`hour. The membranes were then incubated with the FLIP antiserum (diluted
`1:1000) overnight, washed and incubated with an anti–rabbit horseradish
`peroxidase antibody (diluted 1:10 000; Amersham, Arlington Heights, IL)
`for 1 hour. Following several washes,
`the blots were developed by
`chemiluminescence reagent (Pierce, Rockford, IL).
`
`Dr. Reddy’s Laboratories, Inc. v. Celgene Corp.
`IPR2018-01504
`Exhibit 2018, Page 2
`
`

`

`
`
`
`www.bloodjournal.orgFrom
`
`by guest
`
`
`
`For personal use only.on September 13, 2018.
`
`3060
`
`ZANG et al
`
`BLOOD, 15 NOVEMBER 2001 z VOLUME 98, NUMBER 10
`
`replicates of 3. LZ-TRAIL (300 ng/mL) and M180 as a blocking Ab to
`TRAIL (300 ng/mL) were added to culture wells as indicated. Cultures
`were maintained at 33°C in a humidified air of 5% CO2 atmosphere. After 1
`week, nonadherent cells were collected, plates were washed twice with
`Ca11- and Mg21-free Hanks balanced salt solution, and 100 mL 0.05%
`trypsin was added to each well. Adherent cells were removed and combined
`with nonadherent cells and washed once. The cell mixture was then
`resuspended in 3.5 mL 2 3 Dulbecco medium containing 40% fetal calf
`serum, mixed with an equal volume of 0.06% agar (Difco, Detroit, MI) and
`aliquoted into 35 3 10 mm culture dishes (2 mL/dish). Ten mL of a
`cytokine mixture (including IL-1, IL-3, IL-6, G-CSF, GM-CSF, and SCF at
`concentrations of 10 mg/mL each) was added to each dish.27 Untreated cells
`served as controls. After 2 weeks, the plates were scored for colony
`numbers. Groups of at least 40 cells were counted as colonies.
`
`Statistical analysis
`
`The Mann-Whitney U test was used to compare the levels of expression of
`TRAIL and TRAIL receptors between normal and MDS marrow. A
`Wilcoxon signed-rank test was used to compare apoptosis percentages and
`colony numbers between untreated and LZ-TRAIL–treated normal and
`MDS marrow. Significant differences between data from specific groups
`were defined as those with P # .05.
`
`Results
`
`TRAIL-induced apoptosis in normal and MDS marrow
`
`In ancillary dose/response experiments, apoptosis was determined
`in the TRAIL-sensitive cell lines Colo 205, Jurkat, and KG1. The
`extent of apoptosis was assessed by Annexin V–staining and DNA
`histograms (data not shown). Based on those results, marrow from
`healthy donors and patients with MDS was exposed to TRAIL at
`concentrations as high as 1000 ng/mL for up to 24 hours. Results
`are summarized in Figures 1 and 2. In unfractionated mononuclear
`cells from normal marrow, even at the highest TRAIL concentra-
`tions and exposure for 24 hours, no increase in the proportion of
`apoptotic cells was detectable. In MDS marrow,
`in contrast,
`apoptosis increased above baseline already at TRAIL concentra-
`tions of 10 ng/mL and reached a maximum/plateau with a 100%
`increase at TRAIL concentrations of 300 ng/mL and exposure
`durations of 6 to 12 hours. An increase in apoptosis was observed in
`all subpopulations of cells studied (Figure 2):
`total marrow,
`24.8% 6 14.0% increased to 36.3% 6 18.0%, P 5 .011; CD31,
`34.3% 6 20.0% to 44.4% 6 24.9%, P 5 .002; CD141, 19.2% 6 17.2%
`29.7% 6 24.2%, P 5 .006; CD131,
`27.9% 6 24.5% to
`to
`37.1% 6 27.1%, P 5 .004; CD341, 33.3% 6 35.1% to 43.2% 6 30.7%,
`
`Figure 2. Apoptosis in subpopulations of normal and MDS marrow cells. Shown
`is the extent of apoptosis before (left column) and after TRAIL exposure (right
`column) obtained with 10 normal and 14 MDS marrow and exposure for 6-12 hours at
`300 ng TRAIL/mL. Shown are mean values; standard deviations ranged from 6 4%
`to 6 7% in normal, and 6 6% to 6 15% for MDS marrow.
`
`Figure 1. TRAIL-induced apoptosis in normal and MDS marrow cells. The extent
`of apoptosis in normal and MDS marrow cells exposed to TRAIL at 0 to 1000 ng/mL
`for 12 hours was assessed by Annexin V staining and by way of DNA histograms (see
`“Materials and methods”). Shown are the mean values for normal marrow (n 5 10)
`and for MDS marrow (n 5 22); standard deviations ranged from 6 6% to 6 8% for
`normal, and 6 8% to 6 14% for MDS marrow.
`
`Apoptosis assay
`
`Marrow cells from healthy donors and from patients with MDS were
`incubated in 48-well plates (5 3 105 cells/well) with progressively increas-
`ing concentrations of purified LZ–human TRAIL (0, 10, 30, 100, 300, 1000,
`and 3000 ng/mL) for 3, 6, 12, and 24 hours.12,13,18 Apoptotic changes were
`identified by staining with FITC-conjugated Annexin V (Immunotech,
`Westbrook, ME)24,25 and by means of DNA histograms to identify
`hypodiploid nuclei (sub–G1 peak) using flow cytometric analysis as
`described.26 To determine a possible effect of LZ-TRAIL on the marrow
`“microenvironment” and on stroma cells, adherent cell cultures were
`established.27 Adherent cell layers from marrow cells and STRO-1–positive/
`glycophorin-negative sorted cells were established in T25 tissue culture
`flasks (Corning, NY) using Iscoves modified Dulbecco medium (Gibco,
`Grand Island, NY) supplemented with 12.5% each of heat-inactivated horse
`and fetal calf serum (FCS), 0.4 mg/mL L-glutamine, 1 mM sodium
`pyruvate, 10-6 M hydrocortisone, 10-4 M 2-mercaptoethanol, 100 U/mL
`penicillin, and 100 mg/mL streptomycin.28 Flasks were demidepleted of
`cells at weekly intervals. After 4 weeks, nonadherent cells and monocytes
`were removed by 3 trypsinization passes, and negative selection by
`PE-conjugated anti-CD14 mAb. Apoptosis was determined as described.
`
`Cytogenetic and fluorescence in situ hybridization analysis
`
`To determine whether TRAIL-mediated apoptosis occurred preferentially
`in clonal cells (identifiable by a chromosomal marker), we performed
`cytogenetic and fluorescence in situ hybridization (FISH) analysis on
`marrow cells from 9 patients with MDS before and after exposure to
`LZ-TRAIL (300 ng/mL).29 We also determined the proportion of cytogeneti-
`cally abnormal cells among apoptotic (Annexin V–stained) and nonapop-
`totic (Annexin V–negative) marrow cells from 3 patients with MDS
`following sorting by flow cytometry after exposure to LZ-TRAIL
`(300 ng/mL).
`
`In vitro hemopoiesis
`
`Marrow cultures were carried out using the Dexter method.30,31 Stromal
`layers were established in T25 tissue culture flasks (Corning) using Iscoves
`modified Dulbecco medium. Flasks were demidepleted of cells at weekly
`intervals. At 2 to 3 weeks, the stromal cells were irradiated with 1800 cGy,
`removed from the flask by a brief exposure to 0.05% trypsin, and
`1.25 3 105 viable cells/mL were plated as a supportive layer in flat-
`bottomed 48-well plates (Costar, Cambridge, MA). After 1 to 5 days,
`MMNCs (0.5 3 106 cells/well) were layered onto the stromal layers in
`
`Dr. Reddy’s Laboratories, Inc. v. Celgene Corp.
`IPR2018-01504
`Exhibit 2018, Page 3
`
`

`

`
`
`
`www.bloodjournal.orgFrom
`
`by guest
`
`
`
`For personal use only.on September 13, 2018.
`
`BLOOD, 15 NOVEMBER 2001 z VOLUME 98, NUMBER 10
`
`TRAIL AND APOPTOSIS IN MDS
`
`3061
`
`Figure 3. TRAIL-induced apoptosis in MDS marrow.
`TRAIL-induced apoptosis in aberrant MDS blasts (A) and
`in CD341 precursors from MDS marrow (B). Panels in the
`left columns represent data before, and in the right
`columns, after TRAIL exposure. Shown are the gates
`used (top), scattergrams (middle), and DNA histograms
`(bottom) of gated cells. FSC indicates forward scatter;
`SSC, orthogonal scatter; FLI-FITC, fluorescence intensity
`after labeling with FITC-conjugated Annexin V; FL2-PI,
`fluorescence intensity after staining with propidium iodine;
`counts, relative number of cells. Percentage figures in the
`bottom panels indicate proportions of apoptotic cells.
`
`P 5 .043).
`between CD341
`difference
`no
`There was
`/CD331 and CD341/CD332 precursors. Two examples, MDS blasts
`and CD341 precursors, are shown in Figure 3. The most prominent
`increase in apoptosis was seen in MDS blasts (22.9% 6 12.4% to
`39.6% 6 16.1%). The increase in TRAIL-mediated apoptosis was
`prevented in the presence of the blocking anti-TRAIL mAb, M180 (data
`not shown). In normal marrow, no change in the proportion of apoptotic
`cells following LZ-TRAIL exposure was detected in the CD31, CD141,
`or CD131 gates. However, in the gate of CD341 precursors, the
`proportion of apoptotic cells was 4.3% 6 2.8% before and 7.3% 6
`4.3% after LZ-TRAIL exposure (n 5 7; P 5 .06). This finding suggests
`a physiologic role for TRAIL and its receptors in the regulation of
`normal hemopoiesis at a CD341 precursor stage as recently suggested
`by others.32
`Differences in regard to susceptibility to apoptosis were also
`observed among adherent cell layers, whereas TRAIL exposure
`failed to have a significant effect on adherent layers derived from
`normal marrow (10.2% apoptosis before TRAIL exposure, 14.4%
`after TRAIL exposure),
`the proportion of apoptotic cells in
`adherent layers derived from MDS marrow increased significantly
`after TRAIL exposure from 23.6% 6 7.1% to 47.4% 6 9.4%
`(P 5 .04). Apoptosis was limited to CD141 and other mononuclear
`cells; no apoptosis was observed in STRO2/1 glycophorin2/
`CD142 stromal cells.
`
`Surface expression of TRAIL and its receptors on normal and
`MDS marrow cells
`
`The higher TRAIL susceptibility of malignant cells may be due to
`differential expression of the agonistic (TRAIL-R1 and -R2) and
`decoy (TRAIL-R3 and -R4) receptors.15,16 We determined surface
`expression of TRAIL and its receptors on marrow cells from 21
`patients with MDS and from 10 healthy donors. Results are
`summarized in Figure 4A. In normal marrow, the expression of
`TRAIL and its receptors was negligible except for TRAIL-R3. On
`MDS marrow cells, in contrast, TRAIL and receptors R1, R2, R3,
`
`and R4 were constitutively expressed: TRAIL, 1.4% 6 0.6%
`(mean 6 SD) of cells in normal vs 7.9% 6 10.0% in MDS marrow
`(P 5 .011); TRAIL-R1, 1.4% 6 0.4% vs 16.3% 6 23.1% (P 5 .017);
`TRAIL-R2, 1.4% 6 0.6% vs 14.9% 6 21.2% (P 5 .001); TRAIL-R3,
`28.3% 6 20.7% vs 23.6% 6 17.2% (P 5 .495); TRAIL-R4,
`1.6% 6 0.6% vs 7.0% 6 11.3% (P 5 .043). TRAIL-R3 expression
`on normal cells was restricted to the granulocyte gate (CD131)
`(65.0% 6 25.8%). TRAIL-R2 expression in MDS marrow was
`significantly higher than in normal marrow in the lymphocyte gates
`(CD31 or CD191), in the monocyte gate (CD141), and in the
`granulocyte gate. In purified CD341 cells or blasts, a distinct
`pattern of expression was noted especially for TRAIL-R2. Among
`CD341 cells from MDS marrow, 17.9% 6 34.3% expressed
`TRAIL-R2 vs 0.2% 6 0.1% in normal marrow. Among MDS
`blasts, 50.5% 6 48.1% were TRAIL-R2–positive vs 1.4% 6 1.2%
`in normal marrow.
`To estimate TRAIL sensitivity on the basis of expression of
`TRAIL receptors, we calculated the ratio of agonistic (TRAIL-R1
`and -R2) to antagonistic (TRAIL-R3 and -R4) receptors. As shown
`in Figure 4B, in MDS marrow this ratio was significantly higher
`than in normal marrow (18.4% 6 20.3 vs 5.1% 6 6.3, P 5 .015).
`These data are compatible with the higher susceptibility to
`TRAIL-induced death observed in MDS marrow.
`
`Expression of mRNA for TRAIL and its receptors in
`marrow cells
`
`Estimation of mRNA expression was based on band intensities of
`electropheresed RT-PCR products. Although mRNA for TRAIL
`and all TRAIL receptors was detected in cells from both normal
`and MDS marrow, in agreement with the data on cell surface
`expression, the ratio of agonistic TRAIL-R1 and -R2 to decoy
`TRAIL-R3 and -R4 receptors was higher in MDS than in normal
`marrow (Figure 5). When only CD341 cells were considered, the
`ratio was again higher in CD341 cells from MDS marrow. Further,
`within MDS marrow, there was a higher ratio in blast cells than in
`
`Dr. Reddy’s Laboratories, Inc. v. Celgene Corp.
`IPR2018-01504
`Exhibit 2018, Page 4
`
`

`

`
`
`
`www.bloodjournal.orgFrom
`
`by guest
`
`
`
`For personal use only.on September 13, 2018.
`
`3062
`
`ZANG et al
`
`BLOOD, 15 NOVEMBER 2001 z VOLUME 98, NUMBER 10
`
`Figure 4. Surface expression of TRAIL and TRAIL receptors on marrow cells
`and ratio of surface expression of TRAIL-R1 and R2 to TRAIL-R3 and R4.
`(A) Surface expression of TRAIL and receptors R1 to R4 on marrow cells from 10
`healthy donors (Healthy) and 20 patients with MDS (MDS). Shown are results for total
`marrow, CD31, CD141, CD131, and CD341 cells (mean 6 1 SD); b-actin served as
`internal standard. (B) Ratio of surface expression (as shown in part A) of TRAIL
`receptors R1 and R2 (agonistic) over R3 and R4 (decoy) in marrow from 10 healthy
`donors and from 20 patients with MDS. Shown are the results (mean 6 1 SD) for total
`marrow and for CD31, CD141, CD131, and CD341 cells, respectively.
`
`Figure 5. Expression of TRAIL and TRAIL receptors R1-R4 mRNA in normal and
`in MDS marrow. Shown are examples for total mononuclear cells (MNC) and CD341
`precursors, and separately, MDS blasts sorted on the basis of CD45 expression and
`orthogonal light scatter as described previously; b-actin served as internal standard.5,22
`
`consistent with the notion that apoptosis is facilitated in blasts,
`thereby controlling the size of the clonal (blast) pool.
`
`Cytogenetics and FISH analysis
`
`the non–blast cell population. This suggests that blast cells might
`be particularly vulnerable to TRAIL-induced apoptosis, a fact that
`might explain the phenomenon that a clonal (blast) population can
`exist
`in MDS marrow for extended periods of time without
`expanding and developing into frank leukemia.
`
`To determine whether TRAIL-mediated apoptosis eliminated selec-
`tively or preferentially clonal (malignant) cells, we performed
`FISH analysis on the marrow from patients with MDS with a
`cytogenetic marker before and after TRAIL exposure. Based on
`results of ancillary time course studies, cells were exposed to
`
`Expression of apoptosis inhibitory protein FLIP
`
`In addition to the expression of agonistic and decoy receptors,
`differences in intracellular modification of TRAIL signals could
`explain differential effects in normal and malignant cells. FLIP, a
`cytoplasmatic inhibitor of apoptosis, can interfere with TRAIL-
`induced cell death.33 Griffith et al described a correlation between
`the level of expression of FLIP and sensitivity to TRAIL in a
`line.18 Thus, we compared the level of FLIP
`melanoma cell
`expression in normal and MDS marrow at the mRNA and protein
`levels. FLIP mRNA was present in both normal and MDS marrow,
`though quantitative differences were observed in MDS marrow.
`Differences in RNA levels for FLIP were reflected in differences in
`the protein levels as determined by Western blots (Figure 6). The
`ratio of mRNA expression of agonistic (R1 and R2) receptors to the
`cytoplasmatic inhibitor FLIP was higher in MDS marrow than in
`normal marrow, and within MDS marrow this ratio was higher in
`blasts than in the remainder of cells. This observation was
`
`Figure 6. Expression of FLIP protein in normal (n 5 3) and in MDS marrow
`(n 5 5). Shown is an original gel (top) and a Western blot for FLIP using marrow cells
`from 3 healthy donors (1,2,3), 4 marrow (JLP [RA], JAC [RA], GRD [tAML], and FWH
`[RA]), and one example of peripheral blood cells (PJC [RAEB]) from patients with
`MDS. MW indicates molecular weight.
`
`Dr. Reddy’s Laboratories, Inc. v. Celgene Corp.
`IPR2018-01504
`Exhibit 2018, Page 5
`
`

`

`
`
`
`www.bloodjournal.orgFrom
`
`by guest
`
`
`
`For personal use only.on September 13, 2018.
`
`BLOOD, 15 NOVEMBER 2001 z VOLUME 98, NUMBER 10
`
`TRAIL AND APOPTOSIS IN MDS
`
`3063
`
`Table 1. Cytogenetically abnormal cells in MDS marrow before and after
`exposure to TRAIL as determined by FISH analysis
`
`Cytogenetically
`abnormal cells (%)
`
`Patient
`ID
`
`Diagnosis
`(FAB)
`
`Cytogenetic
`abnormality
`
`Before
`TRAIL
`
`After
`TRAIL
`
`255
`167
`221
`187
`256
`209
`236
`183
`218
`
`RA
`RA
`RA
`RA
`RA
`RAEB
`RAEB
`RAEB-T
`tAML
`
`5q2
`121
`5q2
`220
`27
`5q2
`27
`18
`18
`
`84
`14
`38
`51
`28
`73
`9
`3
`13
`
`0
`0
`0
`29
`15
`53
`4
`5
`32
`
`Fresh marrow cells from patients with known clonal cytogenetic abnormalities
`were cultured for 6 to 12 hours in the presence of TRAIL (300 ng/mL). After washing,
`FISH analysis with the appropriate probes was carried out on pre-exposure and
`postexposure samples, side by side.
`
`TRAIL for 6 hours. Results in 8 individuals are summarized in
`Table 1. After TRAIL exposure, the proportion of cytogenetically
`abnormal cells decreased in 6 of 9 MDS marrows studied,
`especially in marrow from patients with RA. While the results
`suggest that LZ-TRAIL eliminated predominantly abnormal cells
`in certain MDS marrows, results indicate considerable heterogeneity.
`
`In vitro hemopoiesis
`
`To determine whether the effects of human LZ-TRAIL on marrow
`cells were of functional relevance, we characterized in vitro
`hemopoiesis of marrow mononuclear cells from 21 patients with
`MDS (RA n 5 8, RARS n 5 3, RAEB n 5 3, CMML n 5 1,
`RAEBtn 5 4, and tAML from MDS n 5 2) and 6 healthy donors
`were propagated in LTMC in the presence or absence of human
`TRAIL (Figure 7). As postulated, numbers of colonies from normal
`marrow did not change in the presence of LZ-TRAIL (104% 6 5%
`of untreated group [100%]). In MDS marrow, colony numbers were
`slightly increased in the presence of LZ-TRAIL when results of all
`MDS marrows were combined (114% 6 14% of untreated group).
`However, results were strikingly dependent upon disease stage. In
`advanced MDS (RAEB-T and tAML) colony and cluster numbers
`decreased significantly in the presence of TRAIL (65% 6 7% of
`untreated marrow; P 5 .004 compared to normal marrow), whereas
`in patients with less advanced MDS (RA, RARS, and RAEB),
`colony numbers increased in the presence of TRAIL (157% 6 21%
`of untreated marrow, P 5 .037 compared to normal). These results
`suggest complex regulatory mechanisms and reflect
`the great
`heterogeneity of MDS. Nevertheless, the data are consistent with a
`model that proposes that hemopoiesis in less advanced MDS is
`suppressed by negative signals, presumably generated by clonal
`precursors,5 and elimination of those cells should result in im-
`proved hemopoiesis. Conversely, in advanced MDS, the abnormal
`precursor pool has expanded and is a major contributor to overall
`hemopoiesis.34 As a consequence, elimination of those cells should
`result in a decline in colony formation. An alternative consideration
`might be that cellular responses to TRAIL signaling differed for
`different disease stages.
`
`Discussion
`
`Increased cellular proliferation, along with exaggerated pro-
`grammed cell death (apoptosis), is one paradigm that has been used
`
`to explain the presence of a cellular or even hypercellular marrow
`concurrently with peripheral blood cytopenias in patients with
`MDS.35 Proliferation is likely to be due to dysregulated cytokine
`expression, while apoptosis could be mediated by either a lack of
`positive (survival) signals or an overproduction of negative (death)
`signals.5,8,27,36 Such a pattern could arise with an excess of
`bifunctional cytokines showing a posit

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