throbber
Downloaded from http://ashpublications.org/blood/article-pdf/122/14/2412/1370092/2412.pdf by guest on 18 January 2023
`
`Regular Article
`
`LYMPHOID NEOPLASIA
`
`Egress of CD191CD51 cells into peripheral blood following treatment
`with the Bruton tyrosine kinase inhibitor ibrutinib in mantle cell
`lymphoma patients
`Betty Y. Chang,1 Michelle Francesco,1 Martin F. M. De Rooij,2 Padmaja Magadala,1 Susanne M. Steggerda,1
`Min Mei Huang,1 Annemieke Kuil,2 Sarah E. M. Herman,3 Stella Chang,1 Steven T. Pals,2 Wyndham Wilson,3
`Adrian Wiestner,3 Marcel Spaargaren,2 Joseph J. Buggy,1 and Laurence Elias1
`
`1Research Department, Pharmacyclics, Inc., Sunnyvale, CA; 2Pathology Department, Academic Medical Center, Amsterdam, The Netherlands;
`and 3Hematology Branch, National Heart, Lung, and Blood Institute, National Institutes of Health, Bethesda, MD
`
`Key Points
`
`(cid:129) MCL cells are mobilized
`into the peripheral blood of
`patients treated with the BTK
`inhibitor ibrutinib.
`Ibrutinib dose-dependently
`inhibits BCR- and chemokine-
`mediated adhesion and
`migration of MCL cells.
`
`(cid:129)
`
`Ibrutinib (PCI-32765) is a highly potent oral Bruton tyrosine kinase (BTK) inhibitor in
`clinical development for treating B-cell lymphoproliferative diseases. Patients with chronic
`lymphocytic leukemia (CLL) often show marked, transient increases of circulating CLL cells
`following ibrutinib treatments, as seen with other inhibitors of the B-cell receptor (BCR)
`pathway. In a phase 1 study of ibrutinib, we noted similar effects in patients with mantle
`cell lymphoma (MCL). Here, we characterize the patterns and phenotypes of cells mobilized
`among patients with MCL and further investigate the mechanism of this effect. Peripheral
`blood CD191CD51 cells from MCL patients were found to have significant reduction in
`the expression of CXCR4, CD38, and Ki67 after 7 days of treatment. In addition, plasma
`chemokines such as CCL22, CCL4, and CXCL13 were reduced 40% to 60% after treatment.
`Mechanistically, ibrutinib inhibited BCR- and chemokine-mediated adhesion and chemo-
`taxis of MCL cell lines and dose-dependently inhibited BCR, stromal cell, and CXCL12/
`CXCL13 stimulations of pBTK, pPLCg2, pERK, or pAKT. Importantly, ibrutinib inhibited migration of MCL cells beneath stromal cells in
`coculture. We propose that BTK is essential for the homing of MCL cells into lymphoid tissues, and its inhibition results in an egress of
`malignant cells into peripheral blood. This trial was registered at www.clinicaltrials.gov as #NCT00114738. (Blood. 2013;122(14):2412-2424)
`
`Introduction
`
`Mantle cell lymphoma (MCL) is an aggressive type of B-cell
`malignancy, constituting 8% of non-Hodgkin lymphomas.1-3 MCL
`is typically characterized by the t(11;14)(q13;q32) translocation,
`which drives cyclin D1 overexpression. Constitutive activation of
`the phosphatidylinositol 3-kinase/protein kinase B (PI3K/AKT) and
`nuclear factor kB pathways contribute to the pathogenesis of MCL.1
`The majority of MCL patients present with advanced disease at
`diagnosis, and more than 90% of patients have extranodal man-
`ifestations with circulating MCL cells, bone marrow, and gastroin-
`testinal involvement. In general, MCL patients have a poor prognosis,
`with a median overall survival time of 30 to 43 months and fewer
`than 15% of them are long-term survivors.2,3 This demonstrates a
`clear need for new therapeutics for the treatment of this disease.
`The interaction of neoplastic B cells with stromal cells in the
`lymph node (LN) or bone marrow microenvironment plays a critical
`role in the survival, progression, and drug resistance of various
`B-cell malignancies,4-7 including MCL.6,8,9 Importantly, the homing
`and trafficking of B cells into the microenvironment is tightly
`controlled and regulated by the interaction of chemokine receptors
`and adhesion molecules.10-15 The contact between MCL cells and
`
`mesenchymal stromal cells (MSCs) is established and maintained
`by chemokine receptors and adhesion molecules. Stromal cells in
`lymphoid tissues constitutively express chemokines such as CXCL12
`and CXCL13, forming gradients that allow the homing of
`B lymphocytes from the periphery into tissue compartments. MCL
`cells express G protein–coupled chemokine receptors such as
`CXCR4 and CXCR5 that bind CXCL12 and CXCL13, respec-
`tively.6 Adhesion is facilitated by binding of integrins such as
`VLA-4 on B cells to VCAM-1 on stromal cells and fibronectin in the
`extracellular matrix.16 In addition to the chemokine receptor and
`integrin engagement, it has been shown that B-cell receptor (BCR)
`activation is involved in integrin (such as VLA-4) -mediated
`adhesion7,14,16-18 and is thought to contribute to the growth and
`survival of most types of B-cell malignancies.17,19-21 BCR signaling
`pathway phosphoproteins are represented abundantly in MCL cell
`lines,1,22,23 and genomic lesions or constitutive activation of sig-
`naling proteins downstream of the BCR pathways such as SYK and
`PI3KA have been reported in MCL.23-25
`Since BCR signaling is important for integrin-mediated adhesion,
`growth, and survival of B lymphocytes, Bruton tyrosine kinase
`
`Submitted February 1, 2013; accepted August 2, 2013. Prepublished online as
`Blood First Edition paper, August 12, 2013; DOI 10.1182/blood-2013-02-
`482125.
`
`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 USC section 1734.
`
`M.F. and M.F.M.D.R. contributed equally to the manuscript.
`
`The online version of this article contains a data supplement.
`
`© 2013 by The American Society of Hematology
`
`2412
`
`BLOOD, 3 OCTOBER 2013 x VOLUME 122, NUMBER 14
`
`SANDOZ INC.
`
`IPR2023-00478
`
`Ex. 1042, p. 1 of 13
`
`

`

`Downloaded from http://ashpublications.org/blood/article-pdf/122/14/2412/1370092/2412.pdf by guest on 18 January 2023
`
`BLOOD, 3 OCTOBER 2013 x VOLUME 122, NUMBER 14
`
`EGRESS OF MCL CELLS AFTER IBRUTINIB TREATMENT
`
`2413
`
`(BTK), a key component of the BCR pathway, is thus significant in
`B lymphocyte adhesion and survival.7,14,16,18 More recently, it has
`been shown that BTK plays a role in chemokine (such as CXCL12)
`-controlled B-cell chemotaxis and homing.14 The BTK inhibitor
`ibrutinib (PCI-32765) is an irreversible covalent inhibitor with a
`50% inhibitory concentration of 0.5 nM against BTK in biochemical
`assays, has been found to have broad antitumor activity in B-cell
`malignancies including MCL,26,27 and is currently being evaluated
`in phase 3 clinical studies. In the initial phase 1 study, we noted
`that many MCL patients had rapid increases of CD51 B lymphocytes
`in the peripheral blood (PB) following ibrutinib treatment. This
`often occurred concomitantly with rapid reductions of lymphade-
`nopathy, suggesting an egress of malignant cells from tissues into
`PB. The schedule of drug administration for most patients in the
`phase 1 study was in cycles of daily administration for 28 days,
`followed by 7 days without drug administration. We observed that
`the lymphoid flux was rapidly reversed during the 7-days-off portion
`of the treatment, with prompt reappearance at the beginning of the
`next cycle, resulting in a sawtooth pattern in PB. These patterns,
`along with immunophenotypic characterization of the cells involved,
`are presented here. We further investigated the mechanism of this
`effect by using MCL cell lines and primary cells and also in an
`MCL–stromal coculture system. We demonstrated that ibrutinib
`suppressed BCR- and CXCL12-/CXCL13-mediated adhesion and
`chemotaxis, suppressed migration of cells beneath the stromal cells
`(pseudo-emperipoiesis), and inhibited the phosphorylation of
`BTK, PLCg, and ERK in MCL cells. These observations effec-
`tively highlight the importance of BTK catalytic activity in the
`homing of MCL cells into lymphoid tissues and provide insight
`into the relevant mechanisms.
`
`Materials and methods
`
`Refer to supplemental Methods for details (available on the Blood Web site).
`
`Primary human MCL specimens from drug-treated patients
`
`Blood was drawn from MCL patients enrolled in PCYC-0475326 or PCYC-
`110427 (before April 1, 2012) in accordance with International Conference
`on Harmonisation Good Clinical Practice guidelines and principles of the
`Declaration of Helsinki, with informed consent from each patient and in
`compliance with the protocols approved by the relevant institutional review
`board. The blood samples were drawn into sodium heparin cell preparation
`tubes (BD) and shipped overnight to Pharmacyclics, Inc., within 24 hours.
`In a laminar flow hood, the peripheral blood mononuclear cells (PBMCs) were
`collected, washed with phosphate-buffered saline (PBS), and frozen in 90%
`fetal bovine serum and 10% dimethylsulfoxide in liquid nitrogen until use.
`
`Cell lines and primary material for ex vivo studies
`
`Adhesion studies were performed with PBMCs from MCL patients of the
`Academic Medical Center (Amsterdam, The Netherlands). For ex vivo
`CXCR4 and CD38 staining studies, PB and LN biopsies were collected
`from treatment-naive MCL patients enrolled in National Cancer Institute Study
`05-C-0170 (www.clinicaltrials.gov #NCT00114738) with approval from
`the National Institutes of Health Institutional Review Board and informed
`consent. Matched PB and LN samples were obtained on the same day and
`were processed and analyzed in parallel.
`
`and fixed in PBS and 1.6% paraformaldehyde. Cells to be analyzed for
`proliferation with Ki67 were permeabilized with 70% ethanol at 220°C
`overnight, rehydrated with PBS, and stained with Ki67 antibody.
`
`Flow cytometry
`
`BD FACS (fluorescence-activated cell sorter) Canto II was used for all flow
`cytometry collections. Phosphoflow assays were stained and performed as
`described.28 At least 10 000 CD191 cells were collected from each staining
`sample. The data were analyzed and quantified by using FlowJo v7.6, and
`the geometric mean was derived and presented for the mean fluorescence
`intensities (MFIs) of histograms for the specifically stained cell populations.
`
`Coculture assays
`
`Cocultures of M2-10B4 stromal cells and the MCL cell line Mino were
`established according to Burger et al.10 Mino cells were treated with
`vehicle, pertussis toxin, or ibrutinib for 1 hour at 37°C and then added to
`confluent monolayers of stromal cells. The cocultures were incubated at
`37°C for 5 hours to overnight to allow migration of Mino cells beneath the
`stromal cell layer, after which they were washed extensively to remove
`unmigrated cells. For cocultures using live-cell tracer dyes, M2-10B4 and
`Mino cells were first loaded with CellTracker Green 5-chloromethylfluorescein
`diacetate (CMFDA) and CellTracker Orange 5-(and-6)-(((4-chloromethyl)
`benzoyl)amino)tetramethylrhodamine (CMTMR), respectively. For micros-
`copy, cells were fixed with paraformaldehyde and mounted on slides with
`4,6 diamidino-2-phenylindole mounting medium (Vectashield). For quantifica-
`tion of migration of Mino cells in cocultures by flow cytometry, cells were
`trypsinized and stained with APC-Cy7–labeled anti-CD19 antibody. Cells were
`counted by using CountBright absolute counting beads (Life Technologies).
`
`Actin polymerization in Mino cells
`
`Mino cells were adhered to coverslips in serum-free media for 30 minutes at
`37°C and treated with dimethylsulfoxide, pertussis toxin, or ibrutinib for 1 hour.
`Cells were fixed with paraformaldehyde, permeabilized with Triton X-100,
`and stained with Alexa Fluor 594–labeled phalloidin (Molecular Probes).
`Coverslips were mounted on glass slides by using Vectashield mounting
`medium containing 4,6 diamidino-2-phenylindole. Microscopy was performed
`on a Zeiss Axioplan 2 microscope using a 633/1.40 oil immersion Plan-
`Apochromat objective, and images were acquired with a Zeiss AxioCam
`MRm charge coupled device camera and AxioVision v.4.8 software. For
`densitometry, at least 30 cells were imaged for each condition.
`
`Adhesion and migration assays
`
`Cell adhesion7,14 and migration assays were performed as described.14,29
`
`Immunoblotting
`
`Western blots were performed as previously described.7,29
`
`Statistical analysis
`
`Analyses were performed by using GraphPad Prism 4.0. Statistically
`significant differences were determined by using either analysis of variance
`with Bonferroni’s post hoc comparison, or unpaired two-tailed Student t test
`was used to determine the significance of differences between two means.
`The one-sample t test was used to determine the significance of differences
`between means and normalized values (100%).
`
`Results
`
`MCL phenotyping
`
`PBMCs were washed, pelleted, and resuspended in PBS and 2% fetal bovine
`serum containing phenotyping surface antibodies. All staining cocktails were
`run in duplicate tubes. Cells were stained for 30 minutes, washed, pelleted,
`
`Transient increase in absolute lymphocyte count following
`ibrutinib administration to MCL patients
`
`In a phase 1 study that enrolled patients with various non-Hodgkin
`lymphomas, MCL patients were treated with ibrutinib in 35-day
`
`SANDOZ INC.
`
`IPR2023-00478
`
`Ex. 1042, p. 2 of 13
`
`

`

`Downloaded from http://ashpublications.org/blood/article-pdf/122/14/2412/1370092/2412.pdf by guest on 18 January 2023
`
`2414
`
`CHANG et al
`
`BLOOD, 3 OCTOBER 2013 x VOLUME 122, NUMBER 14
`
`cycles during which the drug was administered once a day for
`28 days with a 7-day drug holiday between cycles.26 Under these
`conditions, a cyclical pattern of increasing and decreasing absolute-
`lymphocyte count (ALC) was observed. This was demonstrated
`by an increase in ALC following the first few weeks of treatment
`followed by a return to baseline after the 7-day drug holiday
`(Figure 1A). This cyclic ALC pattern continued for the duration of
`the treatment (data not shown). During the course of ibrutinib treat-
`ments, tumor volumes (quantified by sum of perpendicular diameters
`measurements from scans) decreased by 75% on average (Figure 1A)
`during 2 to 6 treatment cycles. Thus, during the first 6 cycles of
`treatment, the cyclic changes of PB ALC occurred concomitantly
`with nodal responses in these patients. The increase in ALC was
`observed in a subsequent (phase 2) study in which MCL patients
`were treated with a fixed continuous daily dose of 560 mg without
`interruption. In this trial, the ALC increased by 100% to 200%
`following the first 2 to 4 weeks of treatment, followed by notable
`gradual reductions in ALC commencing by the eighth week of
`treatment and continuing over the following months (Figure 1B).
`
`Elevated ALC is due to an increase of light chain–restricted
`CD191CD51 cells
`
`In order to define the population of lymphocytes increased by
`ibrutinib, the PBMCs of patients isolated before (day 1 [D1]) and
`after 1 week of treatment (day 8 [D8]) were stained with CD3, CD19,
`and CD5 and analyzed by flow cytometry. The increased lymphocytes
`were characterized as CD3–CD191CD51; both the absolute count
`and the percentage of CD191CD51 cells in the lymphocyte popu-
`lation were significantly increased after 1 week of ibrutinib treat-
`ment (P , .05), whereas the absolute count and the percentage of
`CD191CD5– cells was not (Figure 1C). An illustrative patient is
`shown in Figure 1D, in whom the CD191CD3– and CD191CD51
`populations before drug treatments were 9.29% and 84.4%, respec-
`tively, and they increased to 63% and 98.8% after 1 week of treat-
`ment. The CD191CD3–CD51 cells were light chain–restricted (data
`not shown), likely reflecting increased circulating MCL cells in the
`periphery following 1 week of drug treatment. In some cases, the
`mobilized cells made up a distinct subset of CD45dim small cells,
`which is also consistent with MCL (data not shown).
`To confirm that full inhibition of the target BTK was achieved in
`these patients, occupancy of the BTK active site by ibrutinib was as-
`sessed in PBMCs from MCL patients by using a competitive binding
`fluorescent probe assay.30 On average, more than 90% target occupancy
`was observed in patients following 1 week of treatment (Figure 1E-F).
`
`The peripheral CD191CD51 population has decreased Ki67,
`pERK, and surface CD38 and CXCR4 expression following
`drug treatment
`Peripheral CD191CD51 cells were analyzed for markers commonly
`associated with cell proliferation/activation states such as CD38,31,32
`Ki67,33 and pERK. CD381 surface expression was higher in the
`CD191CD51 cells than in the normal CD191CD5– cells prior to
`drug treatment and became even higher in some patients immediately
`after treatment (Figure 2A), likely because of the higher CD38 ex-
`pression in tissues (Figure 2D), but it decreased significantly
`following 1 week of treatment (P , .05) (Figure 2B) and decreased
`even further with longer treatment, while the CD38 expression in
`the normal B cells did not change (Figure 2A). In vitro ibrutinib
`treatment did not affect CD38 expression in MCL cells (supplemental
`Figure 1). Furthermore, Ki67 expression, commonly used as a marker
`of proliferation, was significantly reduced after treatment (P , .01)
`
`(Figure 2B; supplemental Figure 7). pERK expression was generally
`higher in the CD201CD51 cells of MCL patients compared with
`healthy volunteers and was significantly reduced by ibrutinib treatment
`(Figure 2B, lower panel; P , .05).
`Since the chemokine receptor CXCR4 is important for B-cell
`homing to lymphoid tissues,34,35 we determined the surface CXCR4
`expression in the MCL cells and found that CXCR4 was signif-
`icantly reduced (P , .05) in the CD191CD51 population following
`1 week of treatment (Figure 2C). We then analyzed CXCR4 ex-
`pression on patient-matched LN- and PB-resident MCL cells from
`3 untreated patients and found that CXCR4 expression was lower
`in MCL cells isolated from LNs compared with PB in all 3 patients
`examined (Figure 2D), similar in trend to those noted in chronic
`lymphocytic leukemia (CLL) patients in whom CXCR4 surface
`expression is lower in LN compared with PB36,37 because of receptor
`endocytosis from high tissue concentrations of CXCL12.4,10,36,38
`Therefore, the newly circulating CXCR4lo MCL cell population
`is consistent with the mobilized cells originating from tissues such
`as LNs. This interpretation is further supported by the notable
`reduction in lymphadenopathy observed during the same period
`(Figure 1A). Importantly, ibrutinib treatment in vitro did not have a
`direct effect on CXCR4 surface expression in primary MCL cells at
`concentrations of 1 to 1000 nM (supplemental Figure 1).
`We also examined changes to plasma chemokines in the treated
`patients and found that chemokines important in B-cell trafficking/
`homing (CXCL13) and T-cell/accessory-cell attractions (CCL22,
`CCL4, CCL17)6,39,40 were reduced on average by more than 50%
`following 1 week of treatment. By the end of the first cycle of
`treatment, in addition to the decrease of CCL4 and CCL22, cytokines
`important for MCL proliferation—interleukin 10 (IL-10) and
`tumor necrosis factor a (TNF-a)41,42—were also reduced by 50%
`(Figure 2E).
`
`Ibrutinib inhibits pseudo-emperipoiesis in
`MCL–stromal coculture
`
`The transient increase of ALC in MCL patients treated with ibrutinib
`may be due to a disruption in cellular adhesion and migration within
`the LN or tissue compartment. To investigate this, we established
`the in vitro effect of ibrutinib on the pseudo-emperipoiesis of MCL
`in stromal cell cocultures. Primary MCL cells or the Mino cell line
`were grown in coculture with M2-10B4 murine bone marrow
`stromal cells. We found that primary MCL cells or Mino cells both
`adhered to and migrated beneath the M2-10B4 cells. Significant
`inhibition of pseudo-emperipoiesis by ibrutinib was observed, as
`demonstrated by light microscopy (Figure 3A-B), and the number
`of Mino cells or primary MCL cells remaining in the coculture were
`quantified by flow cytometry of hCD191 cells harvested by gentle
`washing following 4 hours of coculture (Figure 3D-E, left panels).
`Ibrutinib dose-dependently inhibited migration of Mino cells beneath
`the stromal cells, and the inhibition was significant at 100 nM
`(P , .01) and 1000 nM (P , .001). Pertussin toxin, a well-studied
`G protein coupled receptor inhibitor used as a positive control for
`inhibition of Mino cell migration significantly inhibited migra-
`tion at 200 ng/mL (P , .001). In addition, CXCL12, an important
`chemokine for B-cell homing produced by stromal cells, increased
`cortical actin of Mino cells, as assessed by phalloidin fluorescence
`microscopy, and this response was also dose-dependently and
`significantly inhibited by ibrutinib treatments at 10 and 100 nM
`(P , .001) (Figure 3C-D, right panel). Ibrutinib also suppressed
`actin polymerization of primary MCL in coculture at 100 nM
`(P , .001) (Figure 3E, right panel).
`
`SANDOZ INC.
`
`IPR2023-00478
`
`Ex. 1042, p. 3 of 13
`
`

`

`BLOOD, 3 OCTOBER 2013 x VOLUME 122, NUMBER 14
`
`EGRESS OF MCL CELLS AFTER IBRUTINIB TREATMENT
`
`2415
`
`Downloaded from http://ashpublications.org/blood/article-pdf/122/14/2412/1370092/2412.pdf by guest on 18 January 2023
`
`Figure 1. Transient mobilization of lymphocytes in MCL patients treated with ibrutinib. The mean percentage change of ALC (over baseline) is graphed against
`treatment time. (A) Ibrutinib treatment was in 35-day cycles of 28 days on and 7 days off. Mean percentage ALC change and percentage of the sum of perpendicular
`diameters (SPD) change are plotted to treatment cycles (n 5 9). (B) Ibrutinib treatment was continuous with no gap in treatment. Mean percentage ALC change compared
`with baseline (plotted as mean 1 standard error [SE]; n 5 17) is plotted against treatment time. Time points plotted are D0 (day zero), D1, D8, D15, D22, and end of cycles
`1
`1
`1
`CD5– cells following 1 week of ibrutinib treatment. Note the statistically significant
`1, 2, 3, 4, and 5. (C) The absolute count (ABS) and percentage of CD19
`CD5
`vs CD19
`1
`1
`cells of MCL patients following treatment. *P , .05 (paired t test; n 5 16). (D) Flow plot of gated lymphocytes of PBMC samples
`change in the absolute count of CD19
`CD5
`1
`CD3–
`from a representative MCL patient before and after ibrutinib treatment (560 mg per day) for 7 days. PBMCs were stained with CD3, CD19, and CD5. Note increase of CD19
`1
`1
`and CD19
`CD5
`population after 7 days of drug treatment. (E) Fluorescent probe occupancy assay of BTK in PBMCs isolated from a representative MCL patient before treatment
`(predose), and after 4 hours, 24 hours after first dose, before treatment on the eighth day (Day 8, predose), and 4 hours after the eighth day dose (Day 8, 4HR). The arrows point to
`the 75 kDa BTK band on a scanned fluorescent gel (top) and a western blot (bottom). (F) An average of .90% occupancy of BTK by drug is achieved in MCL patients who were
`administered ibrutinib in the first week, determined by fluorescent probe assays (n 5 6).
`
`SANDOZ INC.
`
`IPR2023-00478
`
`Ex. 1042, p. 4 of 13
`
`

`

`2416
`
`CHANG et al
`
`BLOOD, 3 OCTOBER 2013 x VOLUME 122, NUMBER 14
`
`Downloaded from http://ashpublications.org/blood/article-pdf/122/14/2412/1370092/2412.pdf by guest on 18 January 2023
`
`Figure 2. CD191CD51 cells have decreased CXCR4, CD38, and Ki67 expression following ibrutinib treatment. (A) Reduction of CD38 expression (mean fluorescence
`1
`1
`1
`CD5– cells during 4 weeks of treatment in 4 patients treated with ibrutinib. (B) Surface CD38 expression (left
`intensity ratio [MFIR]) in CD19
`CD5
`cells but not CD19
`panel; *P , .05) and intracellular Ki67 (right panel; **P , .01) is significantly reduced following 1 week of treatment. MFIR of intracellular phospho-ERK (pT202/Y204/
`1
`1
`ERK1/2) of CD20
`CD5
`cells from healthy participants or MCL patients treated with ibrutinib before treatment (day 1 [D1]) and after 1 week of treatment (D8) (lower
`1
`1
`panel; *P , .05). (C) Significant reduction of surface CXCR4 expression (MFIR) in CD19
`cells following 1 week of ibrutinib treatment (n 5 14; *P , .05). The line
`CD5
`between the dot plots shows the mean. (D) CXCR4 and CD38 expression from LN biopsies and PBMCs (PB) of three MCL patients (patients A, B, and C) not treated with
`drug. (E) Plasma chemokine and cytokine concentrations on day 8 (D8; left) or day 29 (D29; right) of ibrutinib-treated MCL patients compared with pretreatment times
`100% (n 5 9).
`
`SANDOZ INC.
`
`IPR2023-00478
`
`Ex. 1042, p. 5 of 13
`
`

`

`BLOOD, 3 OCTOBER 2013 x VOLUME 122, NUMBER 14
`
`EGRESS OF MCL CELLS AFTER IBRUTINIB TREATMENT
`
`2417
`
`Downloaded from http://ashpublications.org/blood/article-pdf/122/14/2412/1370092/2412.pdf by guest on 18 January 2023
`
`Figure 3. Ibrutinib inhibits migration of MCL cells beneath stromal cells (pseudo-emperipoiesis) and the formation of CXCL12-stimulated cortical actin. (A) Phase
`contrast (left panel) and 4,6 diamidino-2-phenylindole (DAPI) staining (right panel) of Mino and BM stromal cell M2-10B4 coculture 24 hours after Mino cells were pretreated
`with vehicle (dimethylsulfoxide [DMSO]) (panels a and b) or ibrutinib (1000 nM) (panels c and d). Highlighted yellow outlines show typical cobblestone appearance of
`migrated Mino cells beneath stromal cells. Black arrow points to a cell that is adhered on top of stromal cells but not migrated underneath. Yellow arrow points between two
`migrated Mino cells. (B) Mino cell and stromal cell coculture of Mino cells loaded with 5-(and-6)-(((4-chloromethyl)benzoyl)amino)tetramethylrhodamine (red), and M2 cells
`loaded with 5-chloromethylfluorescein diacetate (green). (A) Stromal cells alone and (B) in coculture with Mino cells. (C) Mino cells pretreated with G protein coupled
`receptor inhibitor pertussis toxin at 200 ng/mL or (D) ibrutinib at 1000 nM. (C) Mino cells were stimulated with CXCL12 at 100 ng/mL and stained with rhodamine-phalloidin to
`determine actin polymerization and were counterstained with DAPI to identify nuclei of cells. Phalloidin staining of Mino cells (A) before and (B) after CXCL12 stimulation and
`after treatment with (C) pertussis toxin at 200 ng/mL or (D) ibrutinib at 100 nM. Magnification, 3200. (D) Mino cells were pretreated with ibrutinib, pertussis toxin, or vehicle
`for 30 minutes and then placed on a stromal cell–populated plate. After 4 hours, coculture was washed several times and migrated, and adhered Mino cells were counted in
`a flow cytometer with calibrated beads after staining with hCD19. Both pertussis toxin and ibrutinib dose-dependently inhibited migration and adhesion of Mino cells (left
`panel). Mino cells stimulated with CXCL12 and treated with vehicle or drug were stained with phalloidin, and its intensity was determined by using flow cytometry (right
`1
`panel). (E) Ibrutinib (100 nM) inhibited pseudo-emperipoiesis of primary MCL (hCD19
`cells) in coculture with M2-10B4 stromal cells (left panel). Actin polymerization as
`assessed by phalloidin staining was significantly reduced by ibrutinib treatment in primary MCL cells. **P , .01; ***P , .001. One-way analysis of variance (ANOVA)
`compared with vehicle control.
`
`SANDOZ INC.
`
`IPR2023-00478
`
`Ex. 1042, p. 6 of 13
`
`

`

`Downloaded from http://ashpublications.org/blood/article-pdf/122/14/2412/1370092/2412.pdf by guest on 18 January 2023
`
`2418
`
`CHANG et al
`
`BLOOD, 3 OCTOBER 2013 x VOLUME 122, NUMBER 14
`
`Ibrutinib inhibits BTK activity in MCL/stromal cell coculture
`and suppresses stromal cell–induced chemokine and
`cytokine secretion
`
`To further understand the drug effect on MCL cells in coculture
`with stromal cells, Mino cells were treated with drug and cocultured
`with M2-10B4 or stimulated with anti-immunoglobulin M (IgM).
`Ibrutinib dose-dependently inhibited pBTK, pPLCg2, pAKT, and
`pERK in Mino cells in coculture with M2-10B4 cells and in Mino
`cells with BCR stimulation (Figure 4A). Chemokine and cytokine
`concentrations of conditioned media were determined from ibrutinib-
`treated Mino cells alone or stimulated with anti-IgM or in coculture
`(Figure 4B). Mino cells increased chemokine and cytokine secretions
`following BCR stimulation or coculture with M2 cells. Similar
`results were observed with the Jeko cell line (supplemental Figure 2).
`Ibrutinib dose-dependently and potently suppressed production of
`human IL-10, CCL22, CCL3, CCL4, TNF-a, and CCL17 following
`BCR activation or in coculture, whereas the murine stromal cells
`alone did not produce human chemokines or cytokines (Figure 4B).
`Similarly, ibrutinib suppressed the production of IL-10, CCL22,
`CCL3, CCL4, and TNF-a of Jeko1 cells in coculture with M2-10B4
`(supplemental Figure 2) or human stromal cell line HS-5 (data not
`shown). Interestingly, these chemokines/cytokines were also reduced
`in ibrutinib-treated patients, but the degree of reduction in plasma
`CCL3 was far less than that in vitro (Figure 2E).
`
`Ibrutinib inhibits BCR- and chemokine-mediated adhesion and
`migration in vitro
`
`We measured the direct effect of ibrutinib on adhesion and migration
`of the MCL cell lines Jeko1, HBL2, Mino, and JVM-1. First, the
`effect of ibrutinib on BTK signaling was determined. As expected,
`ibrutinib inhibited phosphorylation of BTK and downstream signal-
`ing proteins PLCg2, MAP kinases ERK, JNK, and AKT following
`stimulation by anti-IgM and chemokines CXCL12 and CXCL13
`(Figures 5A-B and 6A; supplemental Figures 5 and 6). Cell surface
`expression of CXCR4, CXCR5, CCR7, surface IgM, and a4b1
`integrin was confirmed by flow cytometry (supplemental Figure 3),
`and subsequent in vitro adhesion and chemotaxis assays were per-
`formed with the drug. Ibrutinib significantly inhibited anti-IgM–
`stimulated adhesion of Jeko1 and HBL1 cells onto fibronectin or
`VCAM-1 at 100 nM (a clinically relevant concentration of ibrutinib)
`with more than 50% to 70% inhibition. The inhibition of adhesion
`by ibrutinib was also dose-dependent (supplemental Figure 4).
`Similarly, the adhesion of both Mino and Jeko1 cells to VCAM-1
`or fibronectin was inhibited by ibrutinib at 100 nM following
`CXCL12 or CXCL13 activation. The extent of inhibition was
`greater in Mino cells (50% to 70%) than in Jeko1 cells (20% to 30%)
`(Figure 6B-C). We found that, in addition to changes in adhesion,
`ibrutinib dose-dependently inhibited CXCL12-induced migration
`of Mino, Jeko1, and JVM-1 cells, with Mino and Jeko1 cells being
`more sensitive to drug than JVM-1 cells (Figure 6D). Ibrutinib also
`significantly inhibited CXCL13-stimulated migration of Mino cells
`dose-dependently from 1 nM to 1 mM (Figure 6D).
`Next, we examined the effect of ibrutinib on signaling and
`adhesion in primary MCL cells. In primary MCL, pY223BTK was
`increased compared with that in normal B lymphocytes, consistent
`with elevated BCR signaling in malignant B cells. Ibrutinib inhibited
`pBTK in both primary MCL and normal B cells on Y223, the auto-
`phosphorylation site of BTK, and Y551 (phosphorylated by Src
`family kinases) and reduced pPLCg2 on Y759 and Y1217 (Figure 7A)
`at concentrations of 10 nM and above. These results demonstrate
`
`that ibrutinib directly inhibits BTK activity in MCL primary cells.
`Importantly, ibrutinib also inhibited CXCL12- or CXCL13-activated
`adhesion to VCAM-1 as well as BCR-stimulated adhesion to fibro-
`nectin at 100 nM in primary MCL cells. The degree of inhibition in
`these primary cells was about 10% to 20%, and the magnitude was
`less impressive compared with that in the MCL cell lines but the
`inhibition was statistically significant (Figure 7B).
`These studies collectively demonstrate that ibrutinib inhibits
`BCR-, CXCL12-, and CXCL13-activated adhesion and migration in
`MCL cell lines as well as in primary MCL cells, which is associated
`with the BTK inhibition in these cells.
`
`Discussion
`
`Dramatic early increases of PB lymphocytes occur consistently
`among CLL patients treated with small-molecule antagonists of the
`BCR signaling pathway (Syk, PI3Kd, and BTK inhibitors).26,43,44
`Here we report and characterize similar early transient elevations
`of lymphocytes among MCL patients treated with the irreversible
`BTK inhibitor ibrutinib. The increase in ALC commenced as early
`as the first week, was maximal after 2 to 3 weeks of treatment, and
`then gradually subsided over several cycles of treatment. As with
`CLL patients,26 MCL patients dosed on a 28-days-on and 7-days-
`off schedule exhibited decreases of ALC following the 7-day-off
`period at the end of each cycle, and subsequent increases of ALC
`were observed periodically at the beginning of each subsequent
`cycle. Despite these increases in circulating cells, lymphatic masses
`were noted to decrease concomitantly, consistent with cellular
`mobilization from tissues as opposed to disease progression.
`We found that the newly appearing cells were light chain–
`restricted CD191CD51 cells, consistent with their identification as
`MCL cells. The identity of these cells as lymphomatous is not
`surprising since circulating PB tumor cells are often detectable
`among patients with MCL. Increase of circulating MCL cells fol-
`lowing treatment has not been previously reported and, moreover,
`it was highly specific, not involving CD191CD3–CD5– 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