PRT062070

The Novel Kinase Inhibitor PRT062070 (Cerdulatinib) Demonstrates Efficacy in Models of Autoimmunity and B-Cell Cancer s

Greg Coffey, Andreas Betz, Francis DeGuzman, Yvonne Pak, Mayuko Inagaki, Dale C. Baker, Stanley J. Hollenbach, Anjali Pandey, and Uma Sinha
Portola Pharmaceuticals, Inc., San Francisco, California
Received July 8, 2014; accepted September 23, 2014

ABSTRACT

The heterogeneity and severity of certain autoimmune diseases and B-cell malignancies warrant simultaneous targeting of mul- tiple disease-relevant signaling pathways. Dual inhibition of spleen tyrosine kinase (SYK) and Janus kinase (JAK) represents such a strategy and may elicit several benefits relative to selective kinase inhibition, such as gaining control over a broader array of disease etiologies, reducing probability of selection for bypass disease mechanisms, and the potential that an overall lower level sup- pression of individual targets may be sufficient to modulate dis- ease activity. To this end, we provide data on the discovery and preclinical development of PRT062070 [4-(cyclopropylamino)-2-({4-[4- (ethylsulfonyl)piperazin-1-yl]phenyl}amino)pyrimidine-5-carboxamide hydrochloride], an orally active kinase inhibitor that demonstrates activity against SYK and JAK. Cellular assays demonstrated specific
inhibitory activity against signaling pathways that use SYK and JAK1/
3. Limited inhibition of JAK2 was observed, and PRT062070 did not inhibit phorbol 12-myristate 13-acetate–mediated signaling or activation in B and T cells nor T-cell antigen receptor–mediated signaling in T cells, providing evidence for selectivity of action. Potent antitumor activity was observed in a subset of B-cell lymphoma cell lines. After oral dosing, PRT062070 suppressed inflammation and autoantibody generation in a rat collagen- induced arthritis model and blocked B-cell activation and spleno- megaly in a mouse model of chronic B-cell antigen receptor stimulation. PRT062070 is currently under evaluation in a phase I dose escalation study in patients with B-cell leukemia and lymphoma (NCT01994382), with proof-of-concept studies in humans planned to assess therapeutic potential in autoimmune and malignant diseases.

Introduction
Inhibition of spleen tyrosine kinase (SYK) and Janus kinase (JAK) demonstrated therapeutic potential in a variety of im- munologic disorders. SYK is required for signaling and immune cell activation via the B-cell antigen receptor (BCR), activating fragment crystallizable receptors, and integrins (Mocsai et al., 2010). Genetic studies demonstrate SYK is required for certain inflammatory and autoimmune mechanisms in mice (Colonna et al., 2010; Jakus et al., 2010; Elliott et al., 2011; Wex et al., 2011; Ozaki et al., 2012), a concept that has been reproduced pharmacologically in cellular and animal models (Braselmann et al., 2006; Pine et al., 2007; Coffey et al., 2012). Phase II clinical trials of SYK inhibition by R788 [(6-(5-fluoro-2-(3,4,5- trimethoxyphenylamino)pyrimidin-4-ylamino)-2,2-dimethyl-3-

oxo-2,3-dihydropyrido[3,2-b][1,4]oxazin-4-yl)methyl dihydrogen phosphate] demonstrated efficacy in B-cell non-Hodgkin lym- phoma (NHL) and chronic lymphocytic leukemia (CLL) (Friedberg et al., 2010), rheumatoid arthritis (RA) (Weinblatt et al., 2008, 2010), and immune thrombocytopenia (Podolanczuk et al., 2009). SYK inhibition by GS-9973 [6-(1H-indazol-6-yl)-N-(4- morpholinophenyl)imidazo[1,2-a]pyrazin-8-amine] was recently reported to suppress tumor progression in NHL/CLL (Sharman et al., 2013). These data are consistent with the clinical activity observed by inhibition of Bruton’s tyrosine kinase (BTK) (Byrd et al., 2013; Wang et al., 2013) and phosphatidylinositol-4,5- bisphosphate 3-kinase d (PI3Kd) (Brown et al., 2013), both downstream of SYK in the BCR signaling pathway. JAK family kinases control immune responses after ligation of interleukin (IL) receptors and a subset of chemokine receptors (Yamaoka et al., 2004). The JAK inhibitor CP690,550 [tofacitinib; (3R,4R)-

This work was supported by Portola Pharmaceuticals, Inc. dx.doi.org/10.1124/jpet.114.218164.
s This article has supplemental material available at jpet.aspetjournals.org.
4-methyl-3-(methyl-7H-pyrrolo[2,3-d]pyrimidin-4-ylamino)- b-oxo-1-piperidinepropanenitrile] is approved for the treatment of RA and has demonstrated clinical activity in psoriasis (Papp

ABBREVIATIONS: ABC, activated B cell; BCR, B-cell antigen receptor; BTK, Bruton’s tyrosine kinase; CLL, chronic lymphocytic leukemia; CIA, collagen-induced arthritis; CP690,550, (3R,4R)-4-methyl-3-(methyl-7H-pyrrolo[2,3-d]pyrimidin-4-ylamino)-b-oxo-1-piperidinepropanenitrile; DLBCL, diffuse large B-cell lymphoma; ERK, extracellular signal-related kinase; GCB, germinal center B cell; GS-9973, 6-(1H-indazol-6-yl)-N-(4- morpholinophenyl)imidazo[1,2-a]pyrazin-8-amine; IL, interleukin; JAK, Janus kinase; NHL, non-Hodgkin lymphoma; PBS, phosphate-buffered saline; PMA, phorbol 12-myristate 13-acetate; PI3K, phosphatidylinositol-4,5-bisphosphate 3-kinase; PRT060318, 2-((1R,2S)-2-aminocyclohexylamino)-4-(m-tolylamino) pyrimidine-5-carboxamide; PRT062070, 4-(cyclopropylamino)-2-({4-[4-(ethylsulfonyl)piperazin-1-yl]phenyl}amino)pyrimidine-5-carboxamide hydrochloride; R788, (6-(5-fluoro-2-(3,4,5-trimethoxyphenylamino)pyrimidin-4-ylamino)-2,2-dimethyl-3-oxo-2,3-dihydropyrido[3,2-b][1,4]oxazin-4-yl)methyl dihydrogen phosphate; RA, rheumatoid arthritis; STAT, signal transducer and activator of transcription; SYK, spleen tyrosine kinase.

538

et al., 2012) and the prevention of organ transplant rejection (Vincenti et al., 2012). Moreover, early clinical studies suggest that targeting JAK pathways using small molecule JAK in- hibition (Younes et al., 2012), signal transducer and activator of transcription (STAT) 3 knockdown (Hong et al., 2013), or a neutralizing antibody specific for IL-6 (Kurzrock et al., 2013) may be beneficial for the treatment of B-cell malignancies. These data suggest that SYK and JAK independently contrib- ute to the pathogenesis of certain autoimmune and malignant disorders.
Our research has focused on understanding the complexity of cell signaling networks involved in regulating B-cell ac- tivation and survival. It was previously reported that IL-2, IL-4, and tumor necrosis factor a act as costimulatory molecules to the BCR (Waldmann et al., 1984; Zubler et al., 1984; Clark et al., 1989; Braun et al., 2002), promoting proliferation and survival of healthy normal B cells. We previously reported that costimulation of the BCR with IL-2 or IL-4 in healthy normal B cells significantly enhanced cellular activation rel- ative to BCR or cytokine stimulation alone and that combining SYK-selective and JAK-selective inhibitors more potently sup- pressed this activation response relative to either inhibitor alone (Coffey et al., 2013). Hence, BCR/SYK- and cytokine JAK/
STAT-derived signals appear to act in concert to control normal B-cell function. Of particular interest, elevated serum IL-6 and IL-10 are associated with more aggressive disease progression in CLL and NHL (Fayad et al., 2001; Lai et al., 2002; el-Far et al., 2004). IL-4 promotes the survival of CLL cells in culture and also protects them from death induced by treatment with fludarabine and chlorambucil (Steele et al., 2010). IL-6 and IL-10 autocrine stimulation promote the survival of subsets of B-cell NHL (Lam et al., 2008). Combined SYK/JAK inhibition may therefore represent a powerful strategy to control difficult to treat autoimmune diseases and B-cell malignancies.
We present herein the discovery and biologic characterization of PRT062070 [4-(cyclopropylamino)-2-({4-[4-(ethylsulfonyl) piperazin-1-yl]phenyl}amino)pyrimidine-5-carboxamide hy- drochloride], a novel small molecule inhibitor of SYK and JAK family kinases in preclinical and clinical development (Flinn et al., 2014) for the treatment of autoimmune disease and B-cell malignancies.

Materials and Methods
Synthesis of PRT062070 and Purified Kinase Assays. Exten- sive structure-activity relationship studies identified the potent and orally bioavailable dual SYK/JAK inhibitor PRT062070. PRT062070 is synthesized via a three-step process using starting materials 4-(cyclopropylamino)-2-(methylthio)pyrimidine-5-carboxylic acid and 4-(4-[ethylsulfonyl)piperazine-1-yl]benzenamine. The structure iden- tity and quality of PRT062070 was confirmed by high-performance liquid chromatography, Fourier transform infrared spectroscopy, high- resolution mass spectroscopy, and proton NMR. Potency against purified SYK was determined by fluorescence resonance energy transfer as previously described (Coffey et al., 2012). A broader panel of 270 purified kinases (EMD Milipore, Billerica, MA) was surveyed in which PRT062070 was tested at a fixed concentration of 300 nM. [33P]-labeled substrate was measured after incubation of purified kinase with peptide substrate and [33P]ATP at the Km concentration for the kinase.
Intracellular Signaling in Human and Rat Whole Blood. Human whole blood was collected into lithium heparin vacutainers (VWR, Radnor, PA) from the antecubital vein of subjects who gave

written informed consent (approved by the Human Subjects Commit- tee of Portola Pharmaceuticals, Inc.). Rat whole blood was collected into lithium heparin via cardiac puncture. Aliquots (100 ml) of whole blood were preincubated for 1 hour at 37°C with various concen- trations of PRT062070 (0.1 to 4 mM) before stimulation. Human whole blood was stimulated for 10 minutes with 2.5 ml goat anti-human IgD (Bethyl Laboratories, Montgomery, TX), 10 minutes with 80 nM phorbol 12-myristate 13-acetate (PMA; Sigma-Aldrich, St. Louis, MO), 10 minutes with 10 ml anti-CD3/CD28 Dynabeads (Life Tech- nologies, Grand Island, NY), or 15 minutes with 15 ng/ml of Escherichia coli–derived recombinant human IL-2, IL-4, IL-6, or granulocyte- macrophage colony-stimulating factor (R&D Systems, Minneapolis, MN). Rat whole blood was stimulated for 10 minutes with E. coli–derived recombinant rat IL-6 (R&D Systems). The signaling reaction was ter- minated by addition of 2 ml prewarmed (37°C) 1ti phosflow Lyse/Fix buffer (BD Biosciences, San Jose, CA) followed by incubation at room temperature for 10 minutes. Fixed cells were washed twice in phosphate-buffered saline (PBS; Sigma-Aldrich) and resuspended in 220°C 50% methanol diluted in PBS and stored at 4°C overnight. Unless otherwise indicated, all antibodies used were obtained from BD Biosciences. After permeablization of the cell membrane, cells were washed in PBS containing 1% bovine serum albumin and re- suspended in the same buffer containing mouse anti-human CD19, rabbit anti-human p-extracellular signal-regulated kinase (ERK) Y204 (Cell Signaling Technologies, Danvers, MA), and mouse anti- human pSYK Y352 specific antibodies (anti-IgD and PMA stimula- tions); mouse anti-human CD3, pERK Y204, and mouse anti-human pZap70 Y319 (anti-CD3/CD28 stimulations); mouse anti-human CD3, CD4, CD8, and STAT5 Y694 specific antibodies (human IL-2 sti- mulations); mouse anti-human CD3, CD4, CD8, CD14, CD19, and STAT6 Y641 specific antibodies (human IL-4 stimulations); mouse anti-human CD14 and STAT3 Y705 specific antibodies (human IL-6 stimulations), mouse anti-human CD14 and STAT5 Y694 specific antibodies (human GM-CSF stimulations), and mouse anti-rat CD3, CD4, CD8; and mouse anti-human and mouse STAT1 Y701 specific antibodies (rat IL-6 stimulations) for 1 hour at room temperature. Cells were then washed and resuspended in PBS/bovine serum albumin and assessed by flow cytometry (LSRII; BD Biosciences) for effect of PRT062070 on SYK- and JAK-dependent signaling.
Cellular Activation in Human Whole Blood and Mouse Splenocytes. B-cell and basophil activation experiments in human whole blood were performed as previously described (Coffey et al., 2012). In drug combination experiments, PRT062070 potency was compared with that of the SYK-selective [PRT062607/BIIB057 (Coffey et al., 2012)] and JAK-selective (CP690,550; tofacitinib) inhibitors, with the SYK- and JAK-selective inhibitors combined in a 1:1 ratio for direct comparison with PRT062070. IL-4–mediated functional responses were performed by stimulating human whole blood overnight with 15 ng/ml IL-4 followed by staining with mouse antibodies specific for human CD3, CD4, CD8, CD14, CD19, CD23, CD25, and CD69. Mouse splenocytes were obtained from Balb/c mice by forcing the spleens through a 0.45 mm single cell strainer (BD Biosciences). Washed cells were stimulated overnight with 1 ml goat anti-mouse IgD serum (Ebiosciences, San Diego, CA) or goat serum isotype control (Sigma- Aldrich) in the presence of various concentrations of PRT062070. B-cell activation was determined the next day by staining with CD45R/B220, CD69, CD80, and CD86 as described elsewhere (Coffey et al., 2012).
Tumor Cell Line Viability Experiments. Cell lines obtained from the American Type Culture Collection (Manassas, VA; Ramos RA-1, Daudi, and Toledo) and DSMZ (Braunschweig, Germany; SU-DHL4 and SU-DHL6) were cultured in RPMI media supple- mented with 10% fetal bovine serum (0.5 ti 106 cells/ml). Cells were treated with the indicated concentrations of PRT062070, the SYK- selective inhibitor PRT060318 [2-((1R,2S)-2-aminocyclohexylamino)-4-(m- tolylamino)pyrimidine-5-carboxamide] (Reilly et al., 2011), the pan-JAK inhibitor 1 (EMD Milipore, Darmstadt, Germany), a 1:1 combination of PRT060318 and JAK inhibitor 1, or vehicle control (0.5% dimethylsulfoxide)

for 72 hours. Cell viability assays were performed using CellTiter Glo (Promega, Madison WI) in 384-well plates. Cells were seeded at a density of 5000 cells per well. Apoptosis was measured using the cleaved caspase 3 detection kit (BD Biosciences) as per the supplied protocol. In some experiments, SU-DHL6 cells were mixed with peripheral blood mononuclear cells isolated from healthy donor whole blood to compare the sensitivity of tumor versus normal B cells to PRT062070 in the same tube. Normal and tumor B cells were distinguished by FACS based on CD19 expression and side scatter.
Rat Collagen-Induced Arthritis Model. All in vivo studies were completed in compliance with the “Guide for the Care and Use of Laboratory Animals” and with prior approval from Portola’s Insti- tutional Animal Care and Use Committee. The rat collagen-induced arthritis (CIA) model and histopathology evaluation was performed as previously described (Coffey et al., 2012). Briefly, female Lewis rats (7–8 weeks old and 159–187 g at study initiation) were immunized and boosted with bovine collagen and then randomized to treatment groups upon development of ankle inflammation scores of 1–2. Treatment was initiated after development of disease symptoms. Rats were then administered vehicle or PRT062070 by oral gavage twice daily for the following 2 weeks. At study termination, whole blood from the abdominal aorta was collected for complete blood counts and PRT062070 plasma concentration. Blood for pharmacokinetic analysis was collected im- mediately before the final dose (Cnadir), and at 2 and 6 hours post-final dose for all dose levels. The Tmax for PRT062070 in rats is 2 hours (data not shown). Serum collagen antibody (IgG) titers were monitored both in a pilot study in naive, inflammation score 0, inflammation score 1–2, and inflammation score 4–8 rats, as well as at study termination in the PRT062070 efficacy model using the Rat Anti–Type I and Type II Collagen IgG Assay Kit (Chondrex, Redmond, WA).
Mouse In Vivo BCR Stimulation. BCR stimulation leading to cellular activation and splenomegaly was performed as previously described (Spurgeon et al., 2013). Briefly Balb/c mice (n 5 5 per group) received a single subcutaneous administration of isotype control goat serum or preimmune mouse IgD goat serum. Oral twice daily ad- ministration of vehicle (0.5% methylcellulose in water) or PRT062070 at various dose levels was initiated 1 hour before inoculation with serum and continued for a total of 5 days. On day 5, mice were anesthetized with subcutaneous ketamine cocktail and exsangui- nated via cardiac puncture. Six mice were euthanized immediately before final dose, and remaining mice (at least 3 per dose group) were euthanized 1 hour postfinal dose, which estimates Tmax in this species (data not shown). At the time of euthanasia, spleens were weighed and single cell suspensions were prepared for evaluation of B-cell CD69 and CD80/86 expression by flow cytometry as described above.

Plasma was prepared from the blood and analyzed for PRT062070 concentration to estimate drug levels achieved.
Pharmacokinetic Analysis. Plasma samples were analyzed for PRT062070 concentration using a liquid chromatography–tandem mass spectrometry. In brief, 100-ml plasma samples were processed in a 96-well Captiva filter plate (0.2 mm; Varian, Inc., Palo Alto, CA). The samples were precipitated with acetonitrile containing 200 ng/ml of an internal standard. The mixture was vortexed and refrigerated at 4°C for at least 30 minutes to allow for complete protein precipitation. The mixture was filtered into a 96-well collection plate. The filtrate was injected onto a Sciex API3000 liquid chromatography–tandem mass spectrometry equipped with a turbo-ion spray source. PRT062070 was separated on a Thermo Hypersil-Keystone C18 column (4.6 ti 50 mm, 5 mm; Fisher Scientific, Houston, TX). A mobile phase gradient mixture of 80% mobile phase A (10 mM ammonium acetate in water) and 20% mobile phase B (10 mM ammonium acetate in 10% water 90% acetonitrile) to 75% mobile phase B was programmed over 2.3 minutes. The peak area of the m/z 446 → 336 product ion (PRT062070) was measured against that of the m/z 357 → 295 product ion (PRT061196) in positive ion mode. The analytical range was 1 to 10,000 ng/ml for PRT062070.
Statistical Analysis. Statistical calculations were performed using the R software. The data of the dose-response studies were analyzed using the three-parameter log-logistic model in the drc package with the upper limit fixed to 100% inhibition. The confidence intervals of the IC50 values were calculated by Taylor expansion. t tests were performed using Excel software, with statistical significance set to P , 0.05.

Results
Characterization of PRT062070 Selectivity and Po- tency against SYK and JAK in Purified Kinase Assays. PRT062070 is a reversible ATP-competitive small molecule kinase inhibitor, the chemical structure of which is shown in Fig. 1A. Potency and specificity of PRT062070 was initially tested at 300 nM, 10ti above the SYK inhibitory IC50, in the Millipore panel of 270 independent purified kinase assays. Kinases that were inhibited by .80% were then reevaluated to determine IC50 using a 10-point concentration-response curve. Twenty-four kinases fit this criterion, and IC50 values for each are shown in Fig. 1B, arranged in order of potency of inhibitory activity.

Fig. 1. PRT062070 chemical structure and potency in purified kinase assays. (A) The chemical structure of PRT062070 is shown.
(B)PRT062070 was screened against the Millipore panel of 270 purified kinases in duplicate at 10-fold above its SYK IC50. Kinases inhibited by greater than 80% were then tested against a 10-point PRT062070 concentration-response curve to determine IC50. The IC50 values for each of these kinases are shown in order of potency.

PRT062070 Inhibits SYK-Dependent Signaling and Functional Responses in B Cells. The potency and spec- ificity of SYK inhibition by PRT062070 was tested in human whole blood from normal donors, in which various leukocyte subsets were stimulated via SYK-dependent or SYK-independent pathways. BCR signaling is initiated by activation of the SRC family kinase LYN, which phosphorylates SYK at Y352. SYK then associates with the Iga/b subunits and propagates the signal to downstream effectors, such as ERK1/2 (Jiang et al., 1998). Hence, we stimulated B cells in human whole blood (n 5 7) with antibody specific for the BCR and measured LYN (SYK Y352) and SYK (ERK Y204) kinase activity. The IC50 against ERK Y204 phosphorylation was 0.5 mM, with no ap- preciable inhibition of SYK Y352 phosphorylation at 4 mM (Fig. 2A). Consistent with the inhibition of SYK, PRT062070 reduced the ability of stimulated B cells to upregulate cell- surface expression of the early activation marker CD69 (IC50 5 0.11 mM). Inhibition of Fc«RI-mediated basophil degranula- tion (IC50 5 0.12 mM) further confirmed the potency of SYK inhibition in whole blood (Fig. 2B). PRT062070 did not inhibit

PMA-induced protein kinase C–mediated signaling to ERK Y204 nor PMA induced CD69 upregulation in B cells at 4 mM (Fig. 2C). Furthermore, T-cell signaling after cross-linking of the T-cell antigen receptor or in response to PMA was un- affected by 4 mM PRT062070 (Fig. 2D), demonstrating spec- ificity of action and lack of broad cross-reactivity to other pathways.
Inhibition of Cytokine Signaling and IL-4 Functional Responses in Human Whole Blood. Cytokine stimulations were performed in human whole blood to assess the potency of PRT062070 against JAK-STAT signaling pathways. IL-2 (JAK1/3) stimulation resulted in STAT5 Y694 phosphorylation in T cells. The PRT062070 IC50 values were 0.3 and 0.16 mM in CD41 and CD81 T cells, respectively (Fig. 3A). IL-4 (JAK1/3) stimulation resulted in phosphorylation of STAT6 Y641 in CD41 T cells, CD81 T cells, CD141 monocytes, and CD191 B cells; PRT062070 demonstrated IC50 values of 0.58, 0.33, 1, and 0.92 mM, respectively (Fig. 3B). IL-6 (JAK1) stimula- tion leading to STAT3 Y705 phosphorylation in monocytes was inhibited with an IC50 of 0.35 mM (Fig. 3C), whereas

Fig. 2. Inhibition of SYK-mediated signaling and functional responses in human whole blood. For each graph, the mean percent inhibition 6 S.E.M. of the mean fluorescent intensity (MFI) normalized to vehicle control is reported on the y-axis and PRT062070 concentration (micromolar) is reported on the I-axis. (A) Human whole blood was stimulated with anti-IgD to measure LYN kinase activity (pSYK Y352; ,, n = 7), SYK kinase activity (pERK Y204; s, n = 8), and B-cell functional response (CD69 upregulation; m, n = 17). (B) Fc«RI-mediated basophil degranulation in human whole blood (n = 4).
(C)Human whole blood stimulated with PMA (n = 6) to induce protein kinase C (PKC)–dependent pERK Y204 (d) and CD69 upregulation (s) on B cells.
(D)Human whole blood stimulated with anti-CD3/28 dynabeads or PMA (n = 6) to induce Zap70 or PKC-dependent, respectively, pERK Y204 (d) and pZap70 Y319 (s) in T cells. Horizontal bars in (C) and (D) depict the median.

Fig. 3. PRT062070 exhibits differential potency against cytokine JAK/STAT signaling pathways. Eight-point concentration response curves (n = 6–8) were performed in human whole blood stimulated with IL-2 (A), IL-4 (B), IL-6 (C), or granulocyte-macrophage colony-stimulating factor (GM-CSF) (D). Percent inhibition of STAT phosphorylation is depicted on the y-axis (mean percent inhibition 6 S.E.M. of MFI). Cell types affected include CD4- and CD8-positive T cells, CD14-positive monocytes, and CD19-positive B cells. The x-axis shows the concentrations of PRT062070 (micromolar) tested.

granulocyte-macrophage colony-stimulating factor (JAK2)– induced STAT5 Y694 phosphorylation in monocytes was not potently inhibited by this compound (Fig. 3D), indicating an en- hanced inhibitory potency of PRT062070 against signaling pathways that use JAK1/JAK3 relative to JAK2 in cellular systems. IL-4 stimulation results in the upregulation of various cell surface activation markers in leukocytes. We therefore incubated human whole blood with IL-4 overnight in the pres- ence of various concentrations of PRT062070 and measured the impact on functional responses in cells. In the presence of IL-4, B cells upregulate cell surface CD23, CD25, and CD69; the PRT062070 IC50 values for each were 1.5, 0.26, and 0.13 mM, respectively (Supplemental Fig. 1, left panel). Monocytes upregu- late cell surface CD23 and CD25 expression, which were inhibited by PRT062070 with IC50 values of 0.23 and 0.36 mM, respectively (Supplemental Fig. 1, right panel).
BCR-induced cellular activation is potentiated by cytokine costimulation (Waldmann et al., 1984; Zubler et al., 1984; Clark et al., 1989; Braun et al., 2002; Coffey et al., 2013). Consistently, BCR-induced activation was more sensitive to the effects of PRT062070 (CD69 IC50 5 0.11 mM; Fig. 2A) than would have been predicted from the potency of cellular SYK kinase inhibition after the same stimulation (ERK Y204 IC50 5 0.5 mM; Fig. 2A). This difference in potency between inhibition of BCR-mediated SYK signaling and B cell activation was not observed with SYK-selective inhibition, where overlapping IC50 values were reported in these two assays (Coffey et al., 2012). To corroborate this observation, we tested the ability of selective
SYK versus JAK inhibition alone and in combination, relative to PRT062070, to suppress BCR-mediated CD69 upregulation in human whole blood. SYK inhibition alone (PRT062607/BIIB057) resulted in full suppression of CD69 upregulation with an IC50 of approximately 0.3 mM. Interest- ingly, whereas JAK inhibition had only a partial effect in suppressing B-cell function, the combination of SYK- and JAK-selective inhibitors exerted greater suppression than did either inhibitor alone, which mimicked the potency of PRT062070 in this assay (Supplemental Fig. 2). These data indicate that SYK and JAK contribute to the overall response of B cells to BCR ligation and provide evidence that PRT062070 affects BCR-mediated B-cell activation by dual inhibition of SYK and JAK.
SYK and JAK Cooperate to Reduce Viability in a Sub- set of NHL Cell Lines. Cell lines with differential sensitiv- ities to SYK- and JAK-selective inhibition were tested for survival in the presence of combined SYK plus JAK-selective inhibitors and the dual SYK/JAK inhibitor PRT062070. As shown in Fig. 4A, the germinal center B-cell subtype (GCB) diffuse large B-cell lymphoma (DLBCL) cell lines SU-DHL4 and SU-DHL6 were sensitive to SYK-selective (PRT060318; Fig. 4A, left) but not to pan-JAK (JAK inhibitor 1; Fig. 4A, right) inhibition. This contrasts with two Burkitt lymphoma cell lines, Ramos and Daudi, which demonstrated comparable sensitivity to both SYK and JAK kinase inhibition. Combining the SYK and JAK selective inhibitors (Fig. 4A, bottom left) resulted in greater inhibition of cell viability relative to either

Fig. 4. SYK and JAK contribute to the survival of a subset of NHL cell lines. (A) Concentration-response curves are shown in the presence of the selective SYK inhibitor PRT060318 (top left), the pan-JAK inhibitor 1 (top right), combined SYK plus JAK-selective inhibitors (bottom left panel), and the dual SYK/JAK inhibitor PRT062070 (bottom right). Concentration in micromoles is displayed on the x-axis and percent inhibition of cell viability is shown on the y-axis. Data represent the mean 6 S.E.M. of three independent experiments. (B) The table depicts the IC50 and 95% confidence intervals for each treatment condition.

inhibitor alone. This conclusion was supported by the IC50 values with nonoverlapping 95% confidence intervals (Fig. 4B). The 95% confidence interval for inhibition of Ramos cell viability was 0.49–0.72 mM with combined SYK plus JAK- selective inhibition versus 0.77–1.77 mM and 0.98–2 mM for SYK- and JAK-selective inhibition, respectively. Similarly, the 95% confidence interval for inhibition of Daudi cell viability was 0.85–1.15 mM with combined SYK plus JAK-selective in- hibition versus 1.77–3.54 and 1.21–2.85 mM for SYK- and JAK- selective inhibition, respectively. PRT062070 (Fig. 4A, bottom right) resulted in viability effects similar to that of the com- bined SYK plus JAK-selective inhibition, although the confi- dence intervals did overlap with that of SYK- and JAK-selective inhibitors (Fig. 4B).
Additional studies were conducted to evaluate the potential for broad cytotoxic effects on B cells. For these experiments, SU-DHL4, SU-DHL6, and Ramos cells were used, each of which express functional cell-surface BCRs. By comparison, the Toledo cell line of DLBCL origin does not express surface BCR and has thus adapted BCR-independent survival mech- anisms (Gabay et al., 1999). As shown in Fig. 5A, SU-DHL4, SU-DHL6, and Ramos cells each underwent apoptosis in response to treatment with PRT062070, whereas there was no detectable activity in the BCR-negative Toledo cell line. SU- DHL6 tumor cells were added to healthy normal human periph- eral blood mononuclear cells and treated with PRT062070 to determine the relative sensitivity of tumor versus nontumor B cells. As shown in Fig. 5B, nearly 80% of the tumor B cells

Fig. 5. PRT062070 induces apoptosis in BCR-signaling competent NHL cell lines. (A) The percent of cells expressing cleaved caspase 3 after 48-hour treatment with vehicle (Vh), 1 or 3 mM PRT062070 is plotted on the y-axis (mean + S.D.; n = 3). (B) The scatter plots depict side scatter (SSC; y-axis) and CD19 expression (x-axis) for a mixture of SU-DHL6 cells and healthy human volunteer peripheral blood mononuclear cells. In this manner, SU-DHL6 and primary B cells were resolved from each other and separately gated, as indicated by the arrows. Histograms depict extent of caspase 3 cleavage in primary B cells (top row) and SU-DHL6 (bottom row) after 48-hour treatment with vehicle or 3 mM PRT062070.

underwent apoptosis in the presence of PRT062070, whereas under the same conditions there was no effect on the non- tumor primary B cells.
PRT062070 Elicits Dose-Dependent Efficacy in the Rat CIA Model. We tested the potential for PRT062070 to modulate inflammation in the rat CIA treatment model after oral dosing. Animals treated with vehicle control exhibited a rapid onset of hind paw inflammation within 2–3 days of boosting with adjuvant, with maximal inflammation occur- ring by day 7. Treatment with 0.5 mg/kg PRT062070 (attaining average Cmax plasma concentration at 2 hours of 0.18 mM) resulted in a nonstatistically significant trend toward reduced ankle inflammation, whereas significant reductions in in- flammation were achieved with the 1.5, 3, and 5 mg/kg doses, with average Cmax plasma concentrations at 2 hours of 0.52, 0.58, and 1.49 mM, respectively. Inflammation was abolished at the 3 mg/kg dose and reversed relative to pretreatment levels at 5 mg/kg (Fig. 6A). Blinded histologic observations were consistent with the inflammation scores. Significant improve- ments in inflammatory infiltrate within the synovium and the integrity of the articular cartilage were observed in a dose- dependent manner (Fig. 6B). Representative histologic evalua- tions are shown in Supplemental Fig. 3.
As expected, PRT062070 treatment also affected anti- collagen antibody formation. Relative to the time of treatment initiation (ankle inflammation score of 1–2), circulating levels
of anticollagen antibody approximately doubled in control animals with inflammation scores of 4–8 over the experimen- tal time course of 16 days (Fig. 6C). In animals treated with PRT062070, we observed a 50% reduction in circulating antibody titers at the 1.5, 3, and 5 mg/kg doses (Fig. 6D). These data suggest that a threshold plasma concentration of PRT062070 was reached at which continued terminal differ- entiation of mature B cells to antibody-secreting plasma cells was disrupted. Importantly, the anti-inflammatory effects of PRT062070 occurred within the pharmacologically active range of this compound (i.e., 0.52–1.49 mM). To estimate the level of target inhibition achieved by these concentrations in rat blood, we performed IL-6 stimulations in the presence of a range of PRT062070 concentrations. The IC50 for IL-6 sig- naling in CD31 T cells was 1.4 mM (Supplemental Fig. 4). Rela- ting this to average Cmax plasma concentrations observed in the rat CIA model, 0.52 to 1.49 mM achieved approximately IC25 to IC50 against this pathway. These data suggest that a low level of combined SYK and JAK inhibition is likely sufficient for meaningful anti-inflammatory effects in this stringent model.
Inhibition of Chronic BCR Stimulation and Spleno- megaly in Mice. Chronic BCR stimulation in secondary lymphoid organs may contribute to the proliferation and survival of certain B-cell malignancies. Chronic BCR signal- ing in vivo can be modeled using subcutaneous administra- tion of an activating anti-mouse BCR antibody, which was

Fig. 6. Dose responsive effect of PRT062070 in rat CIA treatment model. (A) Hind paw inflammation scores (mean 6 S.E.M.; y-axis; n = 8 per group) are plotted over time in days (x-axis) during the course of treatment with vehicle (s), 0.5 mg/kg (d), 1.5 mg/kg (m), 3 mg/kg (n), and 5 mg/kg PRT062070 (,). The average Cmax plasma concentrations (2-hour post final dose) achieved in each dosing group is shown at the right of the graph in micromoles. (B) Corresponding mean modified Mankin histopathology scores (6 S.E.M.; y-axis) determined in a blinded fashion are presented, with the treatment conditions shown on the x-axis. (C) and (D) Rat serum anticollagen antibody titers (micrograms per milliliter; y-axis) in which titers from naive immunized but noninflamed (score 0) and inflamed rats at scores of 1–2 and 4–8 are shown from a pilot study (C), and titers from naive and immunized rats treated with vehicle or various PRT062070 dose levels (milligrams per kilogram; x-axis) from a treatment study are shown (D). Horizontal bars depict the median. Statistically significant differences relative to vehicle control treated rats: *P , 0.05.

previously shown to induce immune cell activation in vivo (Mountz et al., 1987), BCR signaling in mouse whole blood (Coffey et al., 2012), and a rapid onset of splenomegaly in mice (Spurgeon et al., 2013). PRT062070 inhibited mouse splenic B-cell activation in response to this antibody in vitro (Fig. 7A) and in vivo (Fig. 7B). Figure 7B depicts spleens from 2 mice repre- sentative of each treatment group stimulated with isotype control goat serum and treated with vehicle (first set of spleens), stimulated with anti-BCR and treated with vehicle (second set of spleens), and stimulated with anti-BCR but treated with 15 mg/kg PRT062070 (third set of spleens). As shown in the bar graph of Fig. 7B, PRT062070 also suppressed upregulation of splenic B-cell surface CD80/86 and CD69 by .60%. The inhibi- tion of splenomegaly was dose- and concentration-dependent (Fig. 7, C and D). Average Cmax plasma concentrations were 15, 213, 1210, 1395, and 1913 ng/ml for the 1, 5, 15, 20, and 30 mg/kg dose groups, respectively. Statistically significant effects on splenomegaly relative to control were achieved at Cmax concentrations of 1210 ng/ml and greater. These data demonstrate that PRT062070 can inhibit BCR signaling and activation in the spleen after oral dosing in mice.

Discussion
We describe herein the preclinical characterization of the kinase inhibitor PRT062070. In the Millipore Kinase Profiler
screen of 270 purified kinases, PRT062070 inhibited 24 kinases by more than 80% when tested at a concentration 10-fold above SYK IC50. The purified kinase screen was used to help initially define the kinase-inhibitory profile of PRT062070, enabling a more focused evaluation in cellular assays. Potency in the purified kinase assays largely predicted potency in cellular as- says. The major exception was lack of cellular potency against JAK2, providing evidence for potential false-positives in the purified kinase assays. Moreover, the rank-order of potency for SYK and JAK family members in the purified kinase assays did not translate directly into rank-order in cellular assays, likely reflecting the greater complexity of signaling pathways within cells at physiologic ATP concentration. We observed very comparable potency in cellular assays measuring JAK/
STAT, BCR/SYK, and Fc«RI/SYK, with limited to no activity against signaling pathways utilizing protein kinase C, lymphocyte- specific protein tyrosine kinase (LCK), and z-chain–associated protein kinase 70 (ZAP70). Consistent with this kinase- inhibitory profile, PRT062070 in a dose-dependent manner suppressed inflammation and auto-antibody generation in the rat CIA model, and prevented BCR-mediated splenomegaly and B cell activation in mice.
Small molecule inhibitors of SYK and JAK have inde- pendently demonstrated activity in rodent inflammation models (Fridman et al., 2010; Coffey et al., 2012). B-cell activation is required for disease development in the rat CIA

Fig. 7. PRT062070 blocks BCR-induced B-cell activation and splenomegaly in mice. (A) FACS plots represent mouse splenic B cells stimulated with isotype control (control) or anti-BCR antibody in the presence of vehicle or PRT062070 at 0.25 and 0.05 mM. B cell activation (mean + S.D., n = 4) was measured by upregulation of CD69 (bottom row) and CD80/86 (top row). (B) Mice (n = 5 per group) were injected subcutaneously with control goat serum or serum preimmunized with mouse IgD (anti-BCR). The photograph depicts two representative spleens from each group of mice treated with control goat serum and vehicle control (first set, no stimulation), anti-BCR with vehicle (second set, induced B-cell activation), and anti-BCR with 15 mg/kg PRT062070 (third set, B-cell activation inhibited by PRT062070). The bar graph beneath the picture (mean + S.D., n = 5) depicts CD69 and CD80/86 surface expression levels on splenic B cells isolated from the anti-BCR administered animals treated with vehicle or 15 mg/kg PRT062070. (C) Percent inhibition of splenomegaly (determined by normalization to mice treated with anti-BCR and vehicle) is plotted as a bar graph (n = 4–5 mice per group; mean + S.D.), with the PRT062070 dose levels depicted on the x-axis. (D) Cmax plasma concentrations of PRT062070 (nanograms per milliliter; x-axis) is plotted against the percent inhibition of splenomegaly observed in each individual animal on study (y-axis). Statistically significant differences relative to vehicle control-treated mice: *P , 0.05.

and mouse BCR-induced splenomegaly models. We therefore studied the relative contributions of SYK and JAK to B-cell activation in human whole blood, enabling more control over the pharmacological mechanisms. We found that by combin- ing SYK and JAK inhibitors we could not only more potently inhibit B cell functional responses in human whole blood but that this particular combination mimicked the potency ob- served with PRT062070, supporting a dual SYK/JAK mecha- nism for B cell inhibition by this molecule. These data are consistent with the observation that combined SYK and JAK inhibitors mimicked the potency of PRT062070 in Burkitt lymphoma cell lines. It is therefore likely that dual SYK/JAK inhibition is at least partly responsible for the activity seen with PRT062070 in the various ex vivo and in vivo models presented here. However, we cannot rule out the possibility that inhibition of other kinases contribute to the overall activity of PRT062070.
The same cell signaling networks that promote inflamma- tion and autoimmunity also appear to promote the survival
and proliferation of certain B-cell malignancies. In particu- lar, SYK is an intriguing target for CLL, which appears to be heavily dependent upon signaling via the BCR (Chiorazzi et al., 2005; Duhren-von Minden et al., 2012). This is evi- denced by the impressive clinical responses observed with SYK (fostamatinib; Friedberg et al., 2010), BTK (ibrutinib; Byrd et al., 2013) and PI3Kd [idelalisib (GS1101; CAL-101); Brown et al., 2013] inhibition. SYK is upstream of BTK and PI3Kd on the BCR pathway (Jiang et al., 1998; Beitz et al., 1999; Hatton et al., 2012; Sharman et al., 2013), which allows us to postulate that targeting this kinase may more broadly affect BCR signaling. SYK is also required for in- tegrin signaling and integrin-mediated adhesion of CLL to stromal cells, facilitating maintenance and survival in the tumor microenvironment (Mocsai et al., 2002; Frommhold et al., 2007; Buchner et al., 2010). Hence, inhibition of SYK alone is likely to affect multiple disease-relevant survival mechanisms in vivo. In addition, serum levels of several

cytokines are increased in CLL (Mahadevan et al., 2009) and have predictive value in disease outcome (Fayad et al., 2001; Lai et al., 2002; Yan et al., 2011). IL-2, IL-4, and alpha in- terferon promote CLL survival in vitro, reportedly by in- ducing the upregulation of B-cell lymphoma 2 family proteins (Dancescu et al., 1992; Panayiotidis et al., 1993; Jewell et al., 1994; Castejon et al., 1999). Stimulation of CLL cells with IL-4 induces upregulation of B-cell lymphoma-XL and myeloid leukemia cell differentiation protein and protects CLL from apoptosis via cytotoxic agents (Steele et al., 2010). Thus, JAK inhibition may provide therapeutic benefit in CLL patients, particularly in combination with SYK inhibition.
Subsets of NHL also demonstrate a reliance on SYK and JAK pathways for survival. DLBCL has been discriminated genetically into several subtypes, most notably the GCB and activated B-cell type (ABC) (Alizadeh et al., 2000). The ABC/
GCB designation appears to predict survival (Rosenwald et al., 2002; Wright et al., 2003) as well as response of tumor cell lines to small-molecule kinase inhibition of the BCR signaling pathway (Davis et al., 2010). Based on work using cell lines and primary tumors, the clinical response to BTK inhibition (ibrutinib) in DLBCL was genetically defined and appears to be determined by the lymphoma subtype (GCB or ABC) and by the occurrence of mutations that drive nuclear factor k–light chain-enhancer of activated B cell–mediated JAK/pSTAT3 activation (Davis et al., 2010). BTK inhibition did not yield clinical responses in patients with GCB subtype DLBCL (overall response rate was 5%) (Wilson et al., 2012); it should be noted that this subtype represents approxi- mately 50% of all DLBCL cases. No responses were seen in patients with myeloid differentiation primary response gene 88 (MYD88) mutation alone (Wilson et al., 2012), and ex- perimental evidence suggests that no responses may be seen in patients with caspase recruitment domain-containing protein 11 (CARD11) mutations (Davis et al., 2010). In a separate study, 43% of DLBCL could be characterized by amplification of the BCR pathway (termed BCR/proliferation), and interest- ingly, 25% were characterized by the infiltration of nontumor leukocytes to the tumor environment (termed host response) (Monti et al., 2005). In the host response cluster, there was evi- dence for gamma interferon signaling and increased expression of receptors for IL-2, IL-6, and IL-15. Targeting SYK and JAK may therefore be relevant not only to tumor-intrinsic prosur- vival signaling but also to suppressing prosurvival signals pro- vided by the tumor microenvironment.
Of interest, SYK inhibition appears to affect a survival mechanism in GCB subtype lymphoma cell lines that BTK inhibition does not affect (Figs. 4 and 5; Cheng et al., 2011), and therefore SYK inhibition may have distinct clinical ac- tivity in this population. Moreover, subsets of ABC subtype NHL cell lines secrete IL-6 and/or IL-10, engaging an au- tocrine JAK/pSTAT3 signaling pathway that contributes to survival (Ding et al., 2008; Lam et al., 2008). Dual targeting of these survival signals via IKKB and JAK inhibition more effectively killed ABC DLBCL cell lines relative to either inhibitor alone (Lam et al., 2008), suggesting that dual in- hibition of BCR and JAK/STAT signaling pathways may have an advantage over selective inhibition of the BCR pathway alone. This hypothesis was recently tested genetically; com- bined short hairpin RNA knockdown of SYK and JAK was found to have greater antitumor activity in a DLBCL cell line relative to knockdown of each kinase alone (Ma et al., 2013).

Consistently, broad antitumor activity in DLBCL cell lines was observed with PRT062070, including cell lines bearing MYD88 and CARD11 mutation (Ma et al., 2013). Our data are consistent with previously published reports where DLBCL cell lines of GCB origin demonstrated no sensitivity to JAK inhibition (Ding et al., 2008; Lam et al., 2008). By comparison two Burkitt lymphoma cell lines demonstrated sensitivity to SYK and JAK inhibition separately and enhanced sensitivity when combined (Fig. 4). We are currently conducting studies to evaluate the dependency of a much broader panel of ge- netically diverse DLBCL cell lines on SYK and JAK survival pathways. This will include a pharmacogenomic evaluation to identify mutations that influence the cells dependencies on SYK and JAK pathways for survival. Ongoing pharmacody- namic evaluation in B-cell lymphoma and leukemia patients treated with PRT062070 (Flinn et al., 2014) should further elucidate the specific contribution of SYK and JAK pathways to disease pathology.

Acknowledgments
The authors thank the PRT062070 project team for helping to advance this compound into clinical development.

Authorship Contributions
Participated in research design: Coffey, Betz, Pak, Baker, Hollenbach, Pandey, Sinha.
Conducted experiments: Coffey, DeGuzman, Inagaki, Baker. Contributed new reagents or analytic tools: Pandey. Performed data analysis: Coffey, Betz, DeGuzman.
Wrote or contributed to the writing of the manuscript: Coffey, Betz, Baker, Hollenbach, Pandey, Sinha.
References
Alizadeh AA, Eisen MB, Davis RE, Ma C, Lossos IS, Rosenwald A, Boldrick JC, Sabet H, Tran T, and Yu X, et al. (2000) Distinct types of diffuse large B-cell lymphoma identified by gene expression profiling. Nature 403:503–511.
Beitz LO, Fruman DA, Kurosaki T, Cantley LC, and Scharenberg AM (1999) SYK is upstream of phosphoinositide 3-kinase in B cell receptor signaling. J Biol Chem 274:32662–32666.
Braselmann S, Taylor V, Zhao H, Wang S, Sylvain C, Baluom M, Qu K, Herlaar E, Lau A, and Young C, et al. (2006) R406, an orally available spleen tyrosine kinase inhibitor blocks fc receptor signaling and reduces immune complex-mediated in- flammation. J Pharmacol Exp Ther 319:998–1008.
Braun D, Caramalho I, and Demengeot J (2002) IFN-alpha/beta enhances BCR- dependent B cell responses. Int Immunol 14:411–419.
Brown JR, Furman RR, Flinn I, Coutre SE, Wagner-Johnston ND, Kahl BS, Spur- geon SEF, Benson DM, Peterman S, and Johnson DM, et al. (2013) Final results of a phase I study of idelalisib (GS-1101) a selective inhibitor of PI3Kd, in patients with relapsed or refractory CLL. J Clin Oncol 31 (Suppl) 7003.
Buchner M, Baer C, Prinz G, Dierks C, Burger M, Zenz T, Stilgenbauer S, Jumaa H, Veelken H, and Zirlik K (2010) Spleen tyrosine kinase inhibition prevents che- mokine- and integrin-mediated stromal protective effects in chronic lymphocytic leukemia. Blood 115:4497–4506.
Byrd JC, Furman RR, Coutre SE, Flinn IW, Burger JA, Blum KA, Grant B, Sharman JP, Coleman M, and Wierda WG, et al. (2013) Targeting BTK with ibrutinib in relapsed chronic lymphocytic leukemia. N Engl J Med 369:32–42.
Castejón R, Vargas JA, Romero Y, Briz M, Muñoz RM, and Durántez A (1999) Modulation of apoptosis by cytokines in B-cell chronic lymphocytic leukemia. Cytometry 38:224–230.
Cheng S, Coffey G, Zhang XH, Shaknovich R, Song Z, Lu P, Pandey A, Melnick AM, Sinha U, and Wang YL (2011) SYK inhibition and response prediction in diffuse large B-cell lymphoma. Blood 118:6342–6352.
Chiorazzi N, Hatzi K, and Albesiano E (2005) B-cell chronic lymphocytic leukemia, a clonal disease of B lymphocytes with receptors that vary in specificity for (auto) antigens. Ann N Y Acad Sci 1062:1–12.
Clark EA, Shu GL, Lüscher B, Draves KE, Banchereau J, Ledbetter JA, and Valentine MA (1989) Activation of human B cells. Comparison of the signal transduced by IL-4 to four different competence signals. J Immunol 143: 3873–3880.
Coffey G, Betz A, Graf J, Stephens G, Lin PH, Imboden J, and Sinha U (2013) Methotrexate and a spleen tyrosine kinase inhibitor cooperate to inhibit responses to peripheral blood B cells in rheumatoid arthritis. Pharmacology Research and Perspectives 1:1–11.
Coffey G, DeGuzman F, Inagaki M, Pak Y, Delaney SM, Ives D, Betz A, Jia ZJ, Pandey A, and Baker D, et al. (2012) Specific inhibition of spleen tyrosine kinase suppresses leukocyte immune function and inflammation in animal models of rheumatoid arthritis. J Pharmacol Exp Ther 340:350–359.

Colonna L, Catalano G, Chew C, D’Agati V, Thomas JW, Wong FS, Schmitz J, Masuda ES, Reizis B, and Tarakhovsky A, et al. (2010) Therapeutic targeting of Syk in autoimmune diabetes. J Immunol 185:1532–1543.
Dancescu M, Rubio-Trujillo M, Biron G, Bron D, Delespesse G, and Sarfati M (1992) Interleukin 4 protects chronic lymphocytic leukemic B cells from death by apo- ptosis and upregulates Bcl-2 expression. J Exp Med 176:1319–1326.
Davis RE, Ngo VN, Lenz G, Tolar P, Young RM, Romesser PB, Kohlhammer H, Lamy L, Zhao H, and Yang Y, et al. (2010) Chronic active B-cell-receptor signalling in diffuse large B-cell lymphoma. Nature 463:88–92.
Ding BB, Yu JJ, Yu RY, Mendez LM, Shaknovich R, Zhang Y, Cattoretti G, and Ye BH (2008) Constitutively activated STAT3 promotes cell proliferation and survival in the activated B-cell subtype of diffuse large B-cell lymphomas. Blood 111:1515–1523.
Dühren-von Minden M, Übelhart R, Schneider D, Wossning T, Bach MP, Buchner M, Hofmann D, Surova E, Follo M, and Köhler F,, et al. (2012) Chronic lymphocytic leukaemia is driven by antigen-independent cell-autonomous signalling. Nature 489:309–312.
el-Far M, Fouda M, Yahya R, and el-Baz H (2004) Serum IL-10 and IL-6 levels at diagnosis as independent predictors of outcome in non-Hodgkin’s lymphoma. J Physiol Biochem 60:253–258.
Elliott ER, Van Ziffle JA, Scapini P, Sullivan BM, Locksley RM, and Lowell CA (2011) Deletion of Syk in neutrophils prevents immune complex arthritis. J Immunol 187: 4319–4330.
Fayad L, Keating MJ, Reuben JM, O’Brien S, Lee BN, Lerner S, and Kurzrock R
(2001)Interleukin-6 and interleukin-10 levels in chronic lymphocytic leukemia: correlation with phenotypic characteristics and outcome. Blood 97:256–263.
Flinn IW, Patel M, Wagner-Johnston N, Pandey A, Coffey G, Leeds J, Hollenbach SJ, Levy G, and Curnutte J(2014) Pharmacokinetics and pharmacodynamics of the dual Syk/Jak inhibitor PRT062070 (Cerdulatinib) in patients with advanced B cell malignancies. J Clin Oncol 32 (Suppl):5s.
Fridman JS, Scherle PA, Collins R, Burn TC, Li Y, Li J, Covington MB, Thomas B, Collier P, and Favata MF, et al. (2010) Selective inhibition of JAK1 and JAK2 is efficacious in rodent models of arthritis: preclinical characterization of INCB028050. J Immunol 184:5298–5307.
Friedberg JW, Sharman J, Sweetenham J, Johnston PB, Vose JM, Lacasce A, Schaefer-Cutillo J, De Vos S, Sinha R, and Leonard JP, et al. (2010) Inhibition of Syk with fostamatinib disodium has significant clinical activity in non-Hodgkin lymphoma and chronic lymphocytic leukemia. Blood 115:2578–2585.
Frommhold D, Mannigel I, Schymeinsky J, Mocsai A, Poeschl J, Walzog B, and Sperandio M (2007) Spleen tyrosine kinase Syk is critical for sustained leu- kocyte adhesion during inflammation in vivo. BMC Immunol 8:31.
Gabay C, Ben-Bassat H, Schlesinger M, and Laskov R (1999) Somatic mutations and intraclonal variations in the rearranged Vkappa genes of B-non-Hodgkin’s lym- phoma cell lines. Eur J Haematol 63:180–191.
Hatton O, Lambert SL, Krams SM, and Martinez OM (2012) Src kinase and Syk activation initiate PI3K signaling by a chimeric latent membrane protein 1 in Epstein-Barr virus (EBV)1 B cell lymphomas. PLoS ONE 7:e42610.
Hong DS, Younes A, Fayad L, Fowler NH, Hagemeister FB, Mistry R, Nemunaitis JJ, Borad MJ, Bryce AH, and Yamashita M, et al. (2013) A phase I study of ISIS 481464 (AZD9150), a first-in-human, first-in-class, antisense oligonucleotide in- hibitor of STAT3, in patients with advanced cancers. J Clin Oncol 31 (Suppl): 8523.
Jakus Z, Simon E, Balázs B, and Mócsai A (2010) Genetic deficiency of Syk protects mice from autoantibody-induced arthritis. Arthritis Rheum 62:1899–1910.
Jewell AP, Worman CP, Lydyard PM, Yong KL, Giles FJ, and Goldstone AH (1994) Interferon-alpha up-regulates bcl-2 expression and protects B-CLL cells from ap- optosis in vitro and in vivo. Br J Haematol 88:268–274.
Jiang A, Craxton A, Kurosaki T, and Clark EA (1998) Different protein tyrosine kinases are required for B cell antigen receptor-mediated activation of extracel- lular signal-regulated kinase, c-Jun NH2-terminal kinase 1, and p38 mitogen- activated protein kinase. J Exp Med 188:1297–1306.
Kurzrock R, Voorhees PM, Casper C, Furman RR, Fayad L, Lonial S, Borghaei H, Jagannath S, Sokol L, and Usmani SZ, et al. (2013) A phase I, open-label study of siltuximab, an anti-IL-6 monoclonal antibody, in patients with B-cell non-Hodgkin lymphoma, multiple myeloma, or Castleman disease. Clin Cancer Res 19:3659–3670.
Lai R, O’Brien S, Maushouri T, Rogers A, Kantarjian H, Keating M, and Albitar M
(2002)Prognostic value of plasma interleukin-6 levels in patients with chronic lymphocytic leukemia. Cancer 95:1071–1075.
Lam LT, Wright G, Davis RE, Lenz G, Farinha P, Dang L, Chan JW, Rosenwald A, Gascoyne RD, and Staudt LM (2008) Cooperative signaling through the signal transducer and activator of transcription 3 and nuclear factor-kappaB pathways in subtypes of diffuse large B-cell lymphoma. Blood 111:3701–3713.
Ma J, Coffey G, Lu P, Cheng S, Guo A, Pandey A, and Wang YL(2013) Dual SYK/JAK inhibition has a broader anti-tumor activity in both ABC and GCB types of diffuse large B cell lymphoma. Blood 122: 1833.
Mahadevan D, Choi J, Cooke L, Simons B, Riley C, Klinkhammer T, Sud R, Maddipoti S, Hehn S, and Garewal H, et al. (2009) Gene expression and serum cytokine profiling of low stage CLL identify WNT/PCP, Flt-3L/Flt-3 and CXCL9/
CXCR3 as regulators of cell proliferation, Survival and Migration. Hum Genomics Proteomics 2009:453634.
Mócsai A, Ruland J, and Tybulewicz VL (2010) The SYK tyrosine kinase: a crucial player in diverse biological functions. Nat Rev Immunol 10:387–402.
Mócsai A, Zhou M, Meng F, Tybulewicz VL, and Lowell CA (2002) Syk is required for integrin signaling in neutrophils. Immunity 16:547–558.
Monti S, Savage KJ, Kutok JL, Feuerhake F, Kurtin P, Mihm M, Wu B, Pasqualucci L, Neuberg D, and Aguiar RC, et al. (2005) Molecular profiling of diffuse large B-cell lymphoma identifies robust subtypes including one characterized by host inflammatory response. Blood 105:1851–1861.
Mountz JD, Smith J, Snapper CM, Mushinski JF, and Finkelman FD (1987) Poly- clonal activation of the murine immune system by an antibody to IgD. VIII. Stimulation of IgD secretion. J Immunol 139:2172–2178.

Ozaki N, Suzuki S, Ishida M, Harada Y, Tanaka K, Sato Y, Kono T, Kubo M, Kitamura D, and Encinas J, et al. (2012) Syk-dependent signaling pathways in neutrophils and macrophages are indispensable in the pathogenesis of anti- collagen antibody-induced arthritis. Int Immunol 24:539–550.
Panayiotidis P, Ganeshaguru K, Jabbar SA, and Hoffbrand AV (1993) Interleukin-4 inhibits apoptotic cell death and loss of the bcl-2 protein in B-chronic lymphocytic leukaemia cells in vitro. Br J Haematol 85:439–445.
Papp KA, Menter A, Strober B, Langley RG, Buonanno M, Wolk R, Gupta P, Krishnaswami S, Tan H, and Harness JA (2012) Efficacy and safety of tofacitinib, an oral Janus kinase inhibitor, in the treatment of psoriasis: a Phase 2b ran- domized placebo-controlled dose-ranging study. Br J Dermatol 167:668–677.
Pine PR, Chang B, Schoettler N, Banquerigo ML, Wang S, Lau A, Zhao F, Grossbard EB, Payan DG, and Brahn E (2007) Inflammation and bone erosion are suppressed in models of rheumatoid arthritis following treatment with a novel Syk inhibitor. Clin Immunol 124:244–257.
Podolanczuk A, Lazarus AH, Crow AR, Grossbard E, and Bussel JB (2009) Of mice and men: an open-label pilot study for treatment of immune thrombocytopenic purpura by an inhibitor of Syk. Blood 113:3154–3160.
Reilly MP, Sinha U, André P, Taylor SM, Pak Y, Deguzman FR, Nanda N, Pandey A, Stolla M, and Bergmeier W, et al. (2011) PRT-060318, a novel Syk inhibitor, pre- vents heparin-induced thrombocytopenia and thrombosis in a transgenic mouse model. Blood 117:2241–2246.
Rosenwald A, Wright G, Chan WC, Connors JM, Campo E, Fisher RI, Gascoyne RD, Muller-Hermelink HK, Smeland EB, and Giltnane JM, et al.; Lymphoma/
Leukemia Molecular Profiling Project (2002) The use of molecular profiling to predict survival after chemotherapy for diffuse large-B-cell lymphoma. N Engl J Med 346:1937–1947.
Sharman JP, Klein L, Boxer M, Kolibaba KS, Hawkins MJ, Di Paolo JA, Hu J, Reddy A, Jin F, and Melchor-Khan F, et al. (2013) Phase 2 trial of GS-9973, a selective Syk inhibitor, in chronic lymphocytic leukemia (CLL) and non-hodgkin lymphoma (NHL). Blood 642:1634.
Spurgeon SE, Coffey G, Fletcher LB, Burke R, Tyner JW, Druker BJ, Betz A, DeGuzman F, Pak Y, and Baker D, et al. (2013) The selective SYK inhibitor P505- 15 (PRT062607) inhibits B cell signaling and function in vitro and in vivo and augments the activity of fludarabine in chronic lymphocytic leukemia. J Pharmacol Exp Ther 344:378–387.
Steele AJ, Prentice AG, Cwynarski K, Hoffbrand AV, Hart SM, Lowdell MW, Samuel ER, and Wickremasinghe RG (2010) The JAK3-selective inhibitor PF-956980 reverses the resistance to cytotoxic agents induced by interleukin-4 treatment of chronic lymphocytic leukemia cells: potential for reversal of cytoprotection by the microenvironment. Blood 116:4569–4577.
Vincenti F, Tedesco Silva H, Busque S, O’Connell P, Friedewald J, Cibrik D, Budde K, Yoshida A, Cohney S, and Weimar W, et al. (2012) Randomized phase 2b trial of tofacitinib (CP-690,550) in de novo kidney transplant patients: efficacy, renal function and safety at 1 year. Am J Transplant 12:2446–2456.
Waldmann TA, Goldman CK, Robb RJ, Depper JM, Leonard WJ, Sharrow SO, Bongiovanni KF, Korsmeyer SJ, and Greene WC (1984) Expression of interleukin 2 receptors on activated human B cells. J Exp Med 160:1450–1466.
Wang ML, Rule S, Martin P, Goy A, Auer R, Kahl BS, Jurczak W, Advani RH, Romaguera JE, and Williams ME, et al. (2013) Targeting BTK with ibrutinib in relapsed or refractory mantle-cell lymphoma. N Engl J Med 369:507–516.
Weinblatt ME, Kavanaugh A, Burgos-Vargas R, Dikranian AH, Medrano-Ramirez G, Morales-Torres JL, Murphy FT, Musser TK, Straniero N, and Vicente-Gonzales AV, et al. (2008) Treatment of rheumatoid arthritis with a Syk kinase inhibitor: a twelve-week, randomized, placebo-controlled trial. Arthritis Rheum 58:3309– 3318.
Weinblatt ME, Kavanaugh A, Genovese MC, Musser TK, Grossbard EB, and Magilavy DB (2010) An oral spleen tyrosine kinase (Syk) inhibitor for rheumatoid arthritis. N Engl J Med 363:1303–1312.
Wex E, Bouyssou T, Duechs MJ, Erb KJ, Gantner F, Sanderson MP, Schnapp A, Stierstorfer BE, and Wollin L (2011) Induced Syk deletion leads to suppressed allergic responses but has no effect on neutrophil or monocyte migration in vivo. Eur J Immunol 41:3208–3218.
Wilson WH, Gerecitano JF, Goy A, Vos S, Kenkre VP, Barr PM, Blum KA, Shustov AR, Advani RH, and Lih J, et al. (2012) The Bruton’s tyrosine kinase (BTK) inhibitor, ibrutinib (PCI-32765), has preferential activity in the ABC subtype of relapsed/refractory de novo diffuse large B-cell lymphoma (DLBCL): interim results of a multicenter, open-label, phase 2 study. Blood 120:686.
Wright G, Tan B, Rosenwald A, Hurt EH, Wiestner A, and Staudt LM (2003) A gene expression-based method to diagnose clinically distinct subgroups of diffuse large B cell lymphoma. Proc Natl Acad Sci USA 100:9991–9996.
Yamaoka K, Saharinen P, Pesu M, Holt VE, 3rd, Silvennoinen O, and O’Shea JJ (2004) The Janus kinases (Jaks). Genome Biol 5:253.
Yan XJ, Dozmorov I, Li W, Yancopoulos S, Sison C, Centola M, Jain P, Allen SL, Kolitz JE, and Rai KR, et al. (2011) Identification of outcome-correlated cytokine clusters in chronic lymphocytic leukemia. Blood 118:5201–5210.
Younes A, Romaguera J, Fanale M, McLaughlin P, Hagemeister F, Copeland A, Neelapu S, Kwak L, Shah J, and de Castro Faria S, et al. (2012) Phase I study of a novel oral Janus kinase 2 inhibitor, SB1518, in patients with relapsed lymphoma: evidence of clinical and biologic activity in multiple lymphoma subtypes. J Clin Oncol 30:4161–4167.
Zubler RH, Lowenthal JW, Erard F, Hashimoto N, Devos R, and MacDonald HR (1984) Activated B cells express receptors for, and proliferate in response to, pure interleukin 2. J Exp Med 160:1170–1183.

Address correspondence to: Greg Coffey, Portola Pharmaceuticals, 270 E. Grand Ave., South San Francisco, CA 94080. E-mail: [email protected]