itacitinib (incb039110), a jak1 inhibitor, reduces cytokines … · 2020. 9. 30. · 1 itacitinib...

26
Itacitinib (INCB039110), a JAK1 inhibitor, Reduces Cytokines Associated with Cytokine 1 Release Syndrome Induced by CAR T-Cell Therapy 2 3 Eduardo Huarte 1 , Roddy S. O’Connor 2,3 , Michael T. Peel 1 , Selene Nunez-Cruz 2,3 , John 4 Leferovich 2,3 , Ashish Juvekar 1 , Yan-ou Yang 1 , Lisa Truong 1 , Taisheng Huang 1 , Ahmad Naim 4 , 5 Michael C. Milone 2,3 , and Paul A. Smith 1 6 7 1 Incyte Research Institute, Wilmington, Delaware. 2 Department of Pathology and Laboratory 8 Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, 9 Pennsylvania. 3 Center for Cellular Immunotherapies, Perelman School of Medicine, University 10 of Pennsylvania, Philadelphia, Pennsylvania. 4 Incyte Corporation, Wilmington, Delaware. 11 12 13 Running title: Itacitinib (JAK1 Inhibitor) Prevents CAR T-CellInduced CRS 14 15 16 Corresponding author: Eduardo Huarte, PhD, Incyte Research Institute, 1801 Augustine Cut- 17 off. Wilmington, DE 19810, USA; Phone: 302 304 6332; E-mail: [email protected] 18 19 Disclosure of Potential Conflict of Interests: Eduardo Huarte, Michael T. Peel, Ashish 20 Juvekar, Yan-ou Yang, Lisa Truong, Taisheng Huang, Ahmad Naim and Paul S. Smith are 21 employees and stockholders of Incyte. Michael C. Milone is cofounder and co-chair of Cabaletta 22 Bio and consultant for Novartis. 23 24 on July 6, 2021. © 2020 American Association for Cancer Research. clincancerres.aacrjournals.org Downloaded from Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 30, 2020; DOI: 10.1158/1078-0432.CCR-20-1739

Upload: others

Post on 16-Feb-2021

7 views

Category:

Documents


0 download

TRANSCRIPT

  • Itacitinib (INCB039110), a JAK1 inhibitor, Reduces Cytokines Associated with Cytokine 1

    Release Syndrome Induced by CAR T-Cell Therapy 2

    3

    Eduardo Huarte1, Roddy S. O’Connor

    2,3, Michael T. Peel

    1, Selene Nunez-Cruz

    2,3, John 4

    Leferovich2,3

    , Ashish Juvekar1, Yan-ou Yang

    1, Lisa Truong

    1, Taisheng Huang

    1, Ahmad Naim

    4, 5

    Michael C. Milone2,3

    , and Paul A. Smith1 6

    7 1Incyte Research Institute, Wilmington, Delaware.

    2Department of Pathology and Laboratory 8

    Medicine, Perelman School of Medicine, University of Pennsylvania, Philadelphia, 9

    Pennsylvania. 3Center for Cellular Immunotherapies, Perelman School of Medicine, University 10

    of Pennsylvania, Philadelphia, Pennsylvania. 4Incyte Corporation, Wilmington, Delaware. 11

    12

    13

    Running title: Itacitinib (JAK1 Inhibitor) Prevents CAR T-Cell–Induced CRS 14

    15

    16

    Corresponding author: Eduardo Huarte, PhD, Incyte Research Institute, 1801 Augustine Cut-17

    off. Wilmington, DE 19810, USA; Phone: 302 304 6332; E-mail: [email protected] 18

    19

    Disclosure of Potential Conflict of Interests: Eduardo Huarte, Michael T. Peel, Ashish 20

    Juvekar, Yan-ou Yang, Lisa Truong, Taisheng Huang, Ahmad Naim and Paul S. Smith are 21

    employees and stockholders of Incyte. Michael C. Milone is cofounder and co-chair of Cabaletta 22

    Bio and consultant for Novartis. 23

    24

    on July 6, 2021. © 2020 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 30, 2020; DOI: 10.1158/1078-0432.CCR-20-1739

    http://clincancerres.aacrjournals.org/

  • Translational relevance 25 The development of chimeric antigen receptor (CAR) technology has dramatically altered the 26

    global landscape of cancer cell therapy with unprecedented responses of CAR T-cells targeting 27

    CD19 and other B-cell antigens. However, cytokine release syndrome (CRS) and neurotoxicity 28

    represent serious, potentially life-threatening side effects often associated with CAR T-cell 29

    therapy. In the present study, we demonstrated that JAK1 inhibition with itacitinib reduces levels 30

    of cytokines implicated in CRS without affecting CAR T-cell proliferation or cytolytic activity in 31

    vitro. Our data show that prophylactic JAK1 inhibition reduces hyperinflammation, and does not 32

    affect CAR T-cell antitumor response in preclinical models. This study provides the rationale for 33

    a phase II clinical trial of itacitinib for the prevention of CRS induced by CAR T-cell therapy 34

    (NCT04071366). 35

    on July 6, 2021. © 2020 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 30, 2020; DOI: 10.1158/1078-0432.CCR-20-1739

    http://clincancerres.aacrjournals.org/

  • Abstract 36 Purpose: T-cells engineered to express a chimeric antigen receptor (CAR T-cells) are a 37

    promising cancer immunotherapy. Such targeted therapies have shown long-term relapse-free 38

    survival in patients with B-cell leukemia and lymphoma. However, cytokine release syndrome 39

    (CRS) represents a serious, potentially life-threatening side effect often associated with CAR T-40

    cell therapy. CRS manifests as a rapid (hyper)immune reaction driven by excessive 41

    inflammatory cytokine release, including interferon- and interleukin-6. 42

    Experimental Design: Many cytokines implicated in CRS are known to signal through the Janus 43

    kinase–signal transducers and activators of transcription (JAK-STAT) pathway. Here we study 44

    the effect of blocking JAK pathway signaling on CAR T-cell proliferation, anti-tumor activity 45

    and cytokine levels in in vitro and in vivo models. 46

    Results: We report that itacitinib, a potent, selective JAK1 inhibitor, was able to significantly and 47

    dose-dependently reduce levels of multiple cytokines implicated in CRS in several in vitro and in 48

    vivo models. Importantly, we also report that at clinically relevant doses that mimic human JAK1 49

    pharmacologic inhibition, itacitinib did not significantly inhibit proliferation or antitumor killing 50

    capacity of three different human CAR T-cell constructs (GD2, EGFR, and CD19). Finally, in an 51

    in vivo model, antitumor activity of CD19-CAR T-cells adoptively transferred into CD19+

    tumor 52

    bearing immuno-deficient animals was unabated by oral itacitinib treatment. 53

    Conclusion: Together, these data suggest that itacitinib has potential as a prophylactic agent for 54

    the prevention of CAR T-cell–induced CRS, and a phase II clinical trial of itacitinib for 55

    prevention of CRS induced by CAR T-cell therapy has been initiated (NCT04071366). 56

    on July 6, 2021. © 2020 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 30, 2020; DOI: 10.1158/1078-0432.CCR-20-1739

    http://clincancerres.aacrjournals.org/

  • Introduction 57 T-cells engineered to express a chimeric antigen receptor (CAR T-cells) have recently 58

    been approved by the US Food and Drug Administration, and commercially available CAR T-59

    cell therapies (axicabtagene ciloleucel and tisagenlecleucel) have resulted in an overall response 60

    rate of almost 80% (1-4), including long-term relapse-free survival in patients with B-cell 61

    leukemia and lymphoma (5). However, an exaggerated immune response by the adoptively 62

    transferred CAR T-cells, as well as the host immune system, can lead to a severe cytokine 63

    release syndrome (CRS) in up to 40% of treated patients (6, 7). CRS manifests as a rapid 64

    immune reaction driven by a massive release of inflammatory cytokines. While individual 65

    cytokine levels differs between patients, severe CRS is generally very similar to that seen in 66

    hemophagocytic lymphohistiocytosis (HLH) or macrophages activation syndrome (MAS)(8), 67

    with elevation of both canonical T-cell cytokines (i.e. IFN-) as well as cytokines compatible 68

    with HLH/MAS (i.e. IL-6 and IL-10)(9). Typified by chills and fevers, CRS can also progress to 69

    a more severe, even life-threatening reaction (10, 11). 70

    To date, low-grade CRS is treated symptomatically with antipyretics and fluids (7). 71

    Grade > 2 CRS can be treated with tocilizumab (an anti–IL-6 receptor), recently approved by the 72

    US Food and Drug Administration and the European Medicines Agency for treatment of severe 73

    and life-threatening CRS (12, 13). Importantly, therapeutic administration of tocilizumab does 74

    not influence peak CAR T-cell numbers nor clinical outcomes (14). However, treatment with 75

    corticosteroids is also needed in the case of tocilizumab-refractory patients, and concerns remain 76

    over the effect of high dose corticosteroids on CAR-T cell expansion and anti-tumor activity 77

    (14). To date there are no preventative strategies, and we rely on therapeutic treatments. Thus, 78

    the ability to prophylactically inhibit hyper-inflammatory states and the incidence of CRS would 79

    represent a new clinical management strategy for patients receiving CAR T therapy. 80

    JAKs play an important role in signal transduction following cytokine and growth factor 81

    binding to their cognate receptors. Once activated, JAKs phosphorylate signal transducer and 82

    activator of transcription proteins (STATs), which results in dimerization and translocation of 83

    STATs to the nucleus in order to activate or suppress gene transcription (15). Aberrant 84

    production of JAK-mediated cytokines, such as IL-6 and growth factors, has been associated 85

    with a number of chronic inflammatory conditions (16, 17). Thus, we reasoned that the blockade 86

    of the JAK signaling pathway would attenuate the excessive cytokine response that occurs 87

    during CRS without negatively impacting CAR T-cell function. Itacitinib (INCB039110) is a 88

    small-molecule inhibitor of the Janus kinase (JAK) family of protein tyrosine kinases (18). As 89

    opposed to other JAK inhibitors developed by Incyte (i.e. ruxolitinib) as well as others (i.e. 90

    tofacitanib), itacitinib is a JAK1 selective inhibitor, and is therefore expected to minimize the 91

    risk of long-term infection or other complications associated with pan-JAK inhibitors (19), while 92

    reducing the levels of cytokines that signal trough JAK1, such as IFN-, IL-6, IL-12 or TNF-. 93

    In this study, we show that in both in vitro and in vivo preclinical models, itacitinib doses 94

    relevant to cellular half maximal inhibitory concentration (IC50; 50–100 nM) were more effective 95

    than tocilizumab in reducing CRS-related cytokines produced by CD19-CAR T-cells. Moreover, 96

    itacitinib did not significantly inhibit in vitro proliferation or antitumor killing capacity of CAR 97

    T-cells, nor, in a lymphoma model, did it affect the antitumor activity of CAR T-cells in a 98

    prophylactic setting. Together, these data suggest that itacitinib has potential as a prophylactic 99

    agent for the prevention of CAR T-cell–induced CRS, and a phase II clinical trial of itacitinib for 100

    the prevention of CRS induced by CAR T-cells is ongoing (NCT04071366). 101

    102

    on July 6, 2021. © 2020 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 30, 2020; DOI: 10.1158/1078-0432.CCR-20-1739

    https://en.wikipedia.org/wiki/STAT_proteinhttps://en.wikipedia.org/wiki/STAT_proteinhttp://clincancerres.aacrjournals.org/

  • Methods 103 Animals 104

    C57BL/6 and BALB/c animals were purchased from Taconic Biosciences (Rensselaer, 105

    New York) and were approximately 8 weeks old at the time experiments were initiated. OT-1 106

    TCR-transgenic mice and severely immunodeficient NOD-scid IL2R gamma null (NSG) animals 107

    were purchased from the Jackson Laboratory (Bar Harbor, Maine). All mice were used in 108

    protocols approved by the Institutional Animal Care and Use Committee. 109

    110

    T-cell proliferation assay 111

    Peripheral blood mononuclear cells (PBMCs) were prepared from human whole blood 112

    samples using a Ficoll-Hypaque separation method, and T-cells were then obtained from the 113

    PBMCs by centrifugal elutriation. T-cells were maintained in RPMI 1640 medium (Thermo 114

    Fisher Scientific, Waltham, Massachusetts) supplemented with 10% fetal bovine serum, 1% 115

    HEPES, 2 mM L-glutamine, 0.05 mM 2-mercaptoethanol and 100 μg/mL streptomycin, and 100 116

    units/mL penicillin (complete RPMI or CM). T-cells were activated with Dynabeads (Thermo 117

    Fisher Scientific; immobilized agonist antibodies against CD3/CD28) at a 3:1 ratio, resuspended 118

    at a density of 0.5 × 106 cells/mL in 24-well plates and treated with itacitinib at various 119

    concentrations (from 50 to 1000 nM). The plates were incubated at 37°C in 5% CO2 atmosphere 120

    for 10 days, and the proliferation was determined every other day by bead-based methods 121

    (CountBright™ Absolute Counting Beads, Molecular Probes, ThermoFisher). Cultures were 122

    replenished every other day with fresh CM. 123

    124

    Cytotoxicity assays 125

    Luciferase expressing SY5Y neuroblastoma cells (GD-2+) were plated in a 96-well plate 126

    at 50,000 cells/well. Twenty-four hours later, 150,000 CAR T-cells were added to corresponding 127

    wells in a final volume of 200 L. Target cells alone were seeded in parallel to quantify the 128

    maximum luciferase expression (relative luminescent units; RLUmax). Seventeen hours later 129

    100 L of luciferin substrate (Bright-Glo™ Luciferase Assay System, Promega, Madison, 130

    Wisconsin) was added to the co-culture. Luminescence was measured after a 10-minute 131

    incubation using an EnVision (PerkinElmer, Waltham, Massachusetts) plate reader. The percent 132

    cell lysis was obtained using the following calculation: [1 − (RLUexperimental)/(RLUmax)] × 133

    100. The experiment was performed in triplicates. 134

    135

    Itacitinib modulation of mouse CRS models 136

    CRS was induced in BALB/c animals by intravenous injection (IV) of Concanavalin-A 137

    (ConA; 20 mg/kg) or 100 g of an anti-CD3ε antibody (clone 145-2C11). Corresponding 138

    animals were orally dosed with 60 or 120 mg/kg of itacitinib 60 minutes prior CRS induction 139

    (prophylactic) or 30 minutes after (therapeutic) CRS induction. Two hours after ConA or anti-140

    CD3 injection, mice were sacrificed and blood was collected into K2EDTA tubes for cytokine 141

    measurement. 142

    143

    Activated macrophage models 144

    Spleens were collected from 6- to 8-week-old C57BL-6 female mice and mechanically 145

    dissociated. Splenocytes were cultured in RPMI medium supplemented with 10% fetal bovine 146

    serum and macrophage colony-stimulating factor. On day 6, cells were treated with itacitinib and 147

    on July 6, 2021. © 2020 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 30, 2020; DOI: 10.1158/1078-0432.CCR-20-1739

    http://clincancerres.aacrjournals.org/

  • then treated with 5 ng/mL lipopolysaccharide (LPS) on day 7. The following day, supernatant 148

    was collected and cytokines were measured as described below. Six- to 8-week-old female 149

    C57BL/6 mice were prophylactically orally dosed with vehicle, or 60 or 120 mg/kg of itacitinib 150

    twice a day (b.i.d.) for 3 days. Mice then received intraperitoneal injections of LPS (5 µg per 151

    animal). Two hours after injection, mice were euthanized and 3 mL of sterile saline was injected 152

    into the peritoneum to perform a peritoneal lavage. Subsequent cytokine measurements were 153

    performed as described below. 154

    155

    Cytokine measurement 156

    Plasma cytokines were measured using multi-spot assay system, pro-inflammatory panel 157

    1 (for IFN-, IL-1β, IL-2, IL-4, IL-5, IL-6, KC/GRO, IL-10, IL-1p70, and TNF; Meso Scale 158

    Discovery, Meso Scale Diagnostics [MSD], Rockville, Maryland), following manufacturer’s 159

    instructions. Briefly, samples, standards, and controls were added at 25 µL per well. The plate 160

    was sealed and incubated for 2 hours at room temperature on an orbital shaker (600 rpm). At the 161

    end of the incubation, the wells were washed three times using 200 µL phosphate-buffered saline 162

    + 0.05% Tween 20, soaking for 30 seconds and then discarding. Detection antibody was added at 163

    25 µL per well, and the plate sealed and incubated for 1 hour at room temperature on an orbital 164

    shaker (600 rpm). At the end of the incubation, the plate was washed three times as done 165

    previously. 150 µL of the MSD Read Buffer was added to each well, and the MSD plates were 166

    measured on a MSD Sector S 600 plate reader. The raw data were measured as an 167

    electrochemiluminescence signal detected by photodetectors and analyzed using the MSD 168

    Discovery Workbench software. A four-parameter logistic fit curve was generated for each 169

    analyte using the standards, and the concentration of each sample was calculated. 170

    171

    OT-1 T-cell expansion and antitumor activity 172

    The OT-1 strain is transgenic for the TCR VV5 specific for the OVA257-264 peptide 173

    (SIINFEKL) and restricted to H2-Kb. Naïve OT-1 CD8 cells were isolated from OT-1 spleens by 174

    negative selection using Naïve CD8a+ T Cell Isolation Kit (Miltenyi Biotec, Auburn, California). 175

    To measure OT-1 CD8+ cell expansion, cells were CFSE-labeled (2 M) and 176

    resuspended at 5 × 106 cells/mL in complete RPMI, 20 IU IL-2, 2 g/mL of SIINFEKL peptide. 177

    and increasing itacitinib concentrations, from 0 to 1000 nM. Three to five days later, OT-1 cells 178

    were collected, stained with an APC-anti-CD8 antibody, and the number of divisions was 179

    calculated by measuring the relative carboxyfluorescein succinimidyl ester (CFSE) fluorescence 180

    intensity of the different conditions by flow cytometry (20). 181

    To study the effect of itacitinib on the antitumor activity of CD8 OT-1 cells, C57BL/6 182

    animals received subcutaneous injections of 0.5 × 106 OVA-expressing EG7 tumor cells into the 183

    shaved right flank. At day 7, corresponding animals were orally dosed with itacitinib b.i.d. at 60 184

    or 120 mg/kg, and at day 10, corresponding animals received an intravenous (tail vein) injection 185

    of 5 × 106 naïve OT-1 CD8 cells. Tumor sizes were measured on two perpendicular axes using a 186

    digital caliper at least three times a week. Tumor volumes were calculated with the formula: 187

    V = (W2 × L)/2 188

    Where V is tumor volume, W is tumor width, and L is tumor length. 189

    190

    Cell Viability Assay 191

    Cell proliferation was measured using CellTiter-Glo® Luminescent Cell Viability Assay 192

    (Promega, Madison, WI), following manufacturer’s instructions. Briefly, 1000 EG7 cells were 193

    on July 6, 2021. © 2020 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 30, 2020; DOI: 10.1158/1078-0432.CCR-20-1739

    http://clincancerres.aacrjournals.org/

  • seeded in a total volume of 200 l of RPMI + 10% FBS per well in a 96-well plate. The next 194

    day, compound plates were reconstituted in complete media with a 10 point dose curve. Cell 195

    media of the plate was aspirated and compound with a total volume of 100 l was incubated with 196

    the cells for 72 hours. 100 l of CellTiter-Glow reagent was added to each well and protected 197

    from the light with a plate seal. Plates were shaken at room temperature for 2 minutes to induce 198

    cell lysis and then allowed to sit for approximately 10 minutes. The raw data was measured on a 199

    TopCount Luminescence Counter (Perkin Elmer, Waltham, MA) and analyzed in Prism using an 200

    [inhibitor] vs. response – variable slope (four parameters) equation. 201

    202

    Generation of CAR constructs and CAR T-cell preparation 203

    The CD19-BBζ CAR consisting of a CD8 hinge, 4-1BB costimulatory domain, and CD3ζ 204

    signaling domain was generated as previously described (21). PBMCs were stimulated with 205

    Dynabeads (3:1 ratio) and then plated in 24-well plates in X-Vivo medium (Lonza, Morristown, 206

    New Jersey) containing IL-2 (200 IU/mL), IL-7 (10 ng/mL), IL-15 (5 ng/mL), and IL-21 (5 207

    ng/mL) at 0.5 × 106 cells/mL. Three days later, T-cells were transduced using a lentiviral vector 208

    expressing the CAR construct, as described elsewhere (22). 209

    Generation of the GD2-E101K CAR lentiviral plasmid was previously described (23). 210

    The CAR contains the 14G2a scFv, as well as an EF1α promoter, CD8 hinge, 4-1BB 211

    costimulatory domain, and CD3ζ signaling domain. The anti-EGFR CAR, with 3C10 scFv, was 212

    previously described (24). The CAR contains the 3C10 scFV, as well as an EF1α promoter, CD8 213

    hinge, 4-1BB costimulatory domain, and CD3ζ signaling domain. 214

    215

    Antitumor activity (Nalm6 model) 216

    To study whether JAK1 inhibition affects the antitumor effect of CAR T-cells, a 217

    xenograft model was used as previously reported (25). Briefly, 6- to 10-week-old 218

    immunodeficient NOD-SCID γc−/−

    (NSG) mice were injected intravenously via tail vein with 2.5 219

    × 106 luciferase expressing Nalm6 acute lymphoblastic leukemia cells. Corresponding animals 220

    received 120 mg/kg of itacitinib per 10 days (orally, b.i.d.), starting 1 day after the tumor 221

    challenge. Mice were then randomized into groups for adoptive transfer of 3 × 106 CAR T-cells, 222

    non-transduced cells, or vehicle intravenously at day 4. Anesthetized mice were imaged using a 223

    Xenogen IVIS Spectrum system (Caliper Life Sciences, Hopkinton, Massachusetts) once a week. 224

    Mice were given an intraperitoneal injection of D-luciferin (150 mg/kg; Caliper Life Sciences). 225

    Total flux was quantified using Living Image 4.4 (PerkinElmer) by drawing rectangles of 226

    identical area around mice, reaching from head to 50% of the tail length. Background 227

    bioluminescence was subtracted for each image individually. Animals were monitored daily, and 228

    survival rates were compared between groups. 229

    230

    Antitumor activity (NAMALWA model) 231

    Six- to 8-week-old male NSG mice were subcutaneously inoculated with luciferase-expressing 232

    CD19+ NAMALWA lymphoma cells (5 × 10

    6). Mice were weighed and monitored two times a 233

    week, and bioluminescence signals were measured every 5 days. At day 5 after tumor challenge, 234

    animals started a b.i.d. oral treatment of vehicle or INCB039110 (120 mg/kg) for 8 days. At day 235

    8 after tumor challenge, animals received an adoptive transfer of 5 × 106 CD19-CAR T-cells. 236

    Bioluminiscence was measured as done previously. Animals were monitored daily, and survival 237

    rates were compared between groups. 238

    239

    on July 6, 2021. © 2020 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 30, 2020; DOI: 10.1158/1078-0432.CCR-20-1739

    http://clincancerres.aacrjournals.org/

  • Data and statistical analysis 240

    Data are reported as mean ± SD or mean ± SEM in the relevant figures. Differences between two 241

    groups were analyzed by nonparametric Mann-Whitney test. Statistical analysis for multiple 242

    groups was performed by Kruskal-Wallis with Dunn’s post hoc test for nonparametric data sets, 243

    or ANOVA with Holm-Sidak’s test for parametric results. All tests were performed using 244

    GraphPad Prism (GraphPad Software Inc, San Diego, California). 245

    246

    Results 247 Itacitinib reduces cytokine levels in murine models of acute hyperinflammation 248

    As CRS is the most common side effect associated with CAR T-cell treatment, we 249

    investigated whether itacitinib is able to reduce the levels of cytokines associated with acute 250

    hyperactivation leading to CRS. Therefore, we conducted experiments in which naïve animals 251

    were challenged with ConA, a potent T-cell mitogen capable of inducing broad inflammatory 252

    cytokine releases and proliferation (26). Similar to individuals experiencing CRS, animals 253

    receiving ConA have elevated serum levels of multiple inflammatory cytokines as well as 254

    behavioral changes such as fever, malaise, hypotension, hypoxia, capillary leak, multi-organ 255

    toxicity, and potentially death. To study the effect of itacitinib in this model, corresponding 256

    animals were prophylactically dosed with 60 or 120 mg/kg of itacitinib to achieve JAK1 257

    inhibition coverage equivalent to that observed in clinical trials (27-29). When compared with 258

    vehicle-dosed animals, itacitinib was able to significantly reduce serum levels of many of the 259

    cytokines implicated in CRS (i.e., IL-6, IL-12, and IFN-) in a dose-dependent manner (Fig. 1A). 260

    As expected, itacitinib did not have a significant effect on cytokines independent of the JAK1 261

    pathway (i.e., IL-5, Fig. 1A). However, not all JAK-mediated cytokines were significantly 262

    decreased (i.e. IL-4, Fig 1A). Additionally, itacitinib was also able to dose-dependently reduce 263

    CRS-implicated cytokines in a therapeutic mode, where animals were dosed with itacitinib 30 264

    minutes after ConA challenge (Fig. 1B). 265

    To confirm the capacity of itacitinib to reduce hyperinflammation, naïve animals were 266

    challenged with anti-CD3 to induce nonspecific T-cell activation and cytokine response. Once 267

    again, corresponding animals were either prophylactically or therapeutically dosed with 120 268

    mg/kg of itacitinib. Compared with vehicle-treated mice, itacitinib was able to significantly 269

    reduce serum levels of many of the cytokines implicated in CRS, but have no effect on cytokines 270

    independent of the JAK1 pathway (Supplementary Fig. S1A). 271

    272

    Itacitinib reduces IL-6 production by macrophages 273

    Recent evidence shows that host macrophages are the major producers of IL-6 after CAR 274

    T-cell treatment (30). Therefore, we queried whether itacitinib would prevent IL-6 production by 275

    host macrophages. First, murine bone marrow–derived macrophages were expanded in vitro with 276

    granulocyte-colony stimulating factor, and itacitinib was added to the cultures at day 6. LPS was 277

    added to the culture at day 7 to activate the macrophages (31). Prophylactic treatment with 278

    itacitinib reduced IL-6 production in a dose-dependent manner, indicating that the activity of 279

    itacitinib in reducing production of inflammatory cytokines is not exclusive to T-cells (Fig. 2A). 280

    We also observed a non-significant trend towards reduction on several other cytokines (i.e. IL-281

    10, IL-12p70, KC/GRO, Supplementary Fig. S2) 282 Having determined that itacitinib is able to reduce IL-6 production in vitro, we next 283

    expanded our studies to an in vivo context to assess the effect of itacitinib on activated 284

    on July 6, 2021. © 2020 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 30, 2020; DOI: 10.1158/1078-0432.CCR-20-1739

    http://clincancerres.aacrjournals.org/

  • macrophages in mice. Mice were prophylactically treated with itacitinib or vehicle for 3 days to 285

    achieve steady state before receiving an intraperitoneal injection of LPS. Two hours after 286

    injection, cytokines were measured from intraperitoneal lavages. As seen in Fig. 2B, doses of 287

    itacitinib that mimic the JAK1 inhibition coverage achieved in clinical trials significantly 288

    reduced IL-6 production by activated macrophages in vivo. Thus, data from both in vitro and in 289

    vivo systems indicate that itacitinib is capable of downregulating the major cellular source of 290

    inflammatory IL-6 during an experimentally induced model of CRS. 291

    292

    Itacitinib reduces CAR T-cell cytokine production 293

    Whereas cytokine production by the host immune system is a primary cause of CRS, 294

    production of inflammatory cytokines by CAR T-cells is also a major contributor (32). Having 295

    shown that itacitinib is capable of reducing inflammatory cytokine production by the host 296

    immune system, we next wanted to examine whether itacitinib reduces inflammatory cytokine 297

    production by CAR T-cells. Human CD19-CAR T-cells were expanded under concentrations of 298

    tocilizumab or itacitinib equivalent to human doses achieved in clinical trials (33). After a 3-day 299

    expansion, CAR T-cells were co-cultured with CD19 expressing NAMALWA target cells, and 6 300

    hours later supernatants were collected to quantify cytokines. Itacitinib, but not tocilizumab, was 301

    able to significantly reduce the levels of many inflammatory cytokines (i.e., IL-2, IFN-, IL-6, 302

    and IL-8) (Fig. 3). As an internal control (background), we also measured cytokines produced by 303

    non-transduced T-cells (Fig. 3). 304

    305

    Itacitinib at clinically relevant concentrations does not affect PBMC proliferation 306 As T-cells play an important antitumor role, the effect of itacitinib on T-cells was studied 307

    to determine if treatment may result in broad immunosuppression potentially interfering with 308

    normal T-cell proliferation and function. T-cells were obtained from freshly isolated human 309

    PBMCs of healthy adults. Following activation with anti-CD3/anti-CD28 coated beads (23), T-310

    cells were expanded in the presence of increasing concentrations of itacitinib, and proliferation 311

    was measured by flow cytometry. When compared with the dimethyl sulfoxide (DMSO) control, 312

    concentrations of itacitinib relevant to the IC50 (50–100 nM) (34) did not have a significant effect 313

    on anti-CD3/anti-CD28–induced expansion of human T-cells (Fig. 4A), demonstrating that 314

    itacitinib treatment has no negative impact on the ability of T-cells to proliferate. As an internal 315

    control, and consistent with the disruption of the JAK/STAT signaling activity, higher 316

    concentrations of itacitinib (1000 nM) blocked T-cell proliferation (Fig. 4A). 317

    318

    Itacitinib does not affect CAR T-cell proliferation 319

    To study the effect of itacitinib specifically on CAR T-cell expansion and cytolytic 320

    activity, GD2-CAR T-cells (23) were expanded with anti-CD3/anti-CD28 coated beads in the 321

    presence of increasing itacitinib concentrations, ranging from 100 to 500 nM. Once again, when 322

    compared with the DMSO control, itacitinib at 100–250 nM did not have a significant effect on 323

    CD3/CD28-induced expansion of GD2-CAR T-cells, whereas 500 nM was enough to block 324

    CAR T-cell proliferation (Fig. 4B). Importantly, GD2-CAR T-cells expanded in the presence of 325

    low (100 and 250 nM) itacitinib concentrations were able to efficiently lyse GD2-expressing 326

    tumor cells (Fig. 4C). As a negative (background) control, we also measured target cell lysis by 327

    non-transduced (nonspecific) T-cells. As expected, GD2-CAR T-cells expanded in the presence 328

    of a high dose of itacitinib (500 nM) and were unable to induce the lysis of target cells beyond 329

    on July 6, 2021. © 2020 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 30, 2020; DOI: 10.1158/1078-0432.CCR-20-1739

    http://clincancerres.aacrjournals.org/

  • background levels (Fig. 4C). Taken together, these data indicate that IC50 relevant doses of 330

    itacitinib do not interfere with CAR T-cell proliferation or antitumor activity in vitro. 331

    Next, we queried whether itacitinib has an effect on CAR T-cell antigen-specific 332

    proliferation. EGFR-CAR T-cells (35) were expanded with anti-CD3/CD28 beads for 2 weeks 333

    and then restimulated with EGFR beads in the presence of itacitinib or DMSO controls. Once 334

    again, doses of itacitinib relevant to the IC50 (100 or 250 nM) did not affect CAR T-cell antigen-335

    specific proliferation (Fig. 4D). 336

    337

    Itacitinib does not impair T-cell antitumor activity in vivo 338

    To further test the effect of itacitinib on antigen-specific T-cell proliferation and in vivo 339

    antitumor activity, we isolated splenocytes from OT-1 mice, transgenic for the TCR V2V5 340

    specific for the peptide OVA257-264 (SIINFEKL) (36). OT-1 T-cells were expanded with the 341

    SIINFEKL peptide in the presence of increasing concentrations of itacitinib, and their expansion 342

    was measured by flow cytometry. As expected, itacitinib concentrations relevant to the IC50 have 343

    a minimal effect on the expansion rate (Supplementary Fig. S3A). Itacitinib concentrations 344

    relevant to maximum free concentrations (189 and 244 nM, in the absence or presence of potent 345

    CYP3A4 inhibitors, respectively) induced a modest reduction on T-cell proliferation 346

    (Supplementary Fig. S3A). To evaluate the effect of itacitinib on the in vivo antitumor efficacy 347

    of OT-1 CD8, we conducted experiments involving adoptive transfer of OVA-specific CD8 cells 348

    into C57BL/6 mice previously transplanted with an OVA-expressing tumor cell line (EG7). Five 349

    days after tumor challenge, corresponding animals were orally dosed b.i.d. with vehicle or 60 or 350

    120 mg/kg of itacitinib for 2 weeks. Eight days after tumor inoculation, animals received an 351

    adoptive transfer of OVA-specific OT-1 naïve CD8 cells. When compared with the control 352

    group, adoptive transfer of OT-1 CD8 was able to significantly reduce tumor growth (Fig. 4). 353

    Importantly, itacitinib doses equivalent to the JAK1 coverage target doses do not affect the 354

    antitumor efficacy of OT-1 cells (Fig. 5). 355

    To rule out a direct effect of itacitinib on EG7 tumor cell growth, we expanded EG7 cells 356

    in the presence of increasing itacitinib concentration. Even very high concentrations of itacitinib 357

    (25 M) were unable to inhibit tumor proliferation (Supplementary Fig. S3B). 358

    359

    Itacitinib does not affect CD19-CAR T-cell cytolytic activity in vitro 360

    To study the effect of itacitinib on human CAR T-cells targeting the CD19 antigen, we 361

    treated CD19-CAR T-cells with either itacitinib or anti–IL-6 receptor (tocilizumab) for 3 days 362

    and then measured their cytolytic activity against CD19-expressing target cells. Whereas 363

    concentrations of tocilizumab that mimic human pharmacologic activity significantly reduced the 364

    cytolytic activity of CAR T-cells (37), 100 nM of itacitinib showed no significant effect when 365

    compared with control CAR T-cells (Fig. 6A). CAR T-cells expanded with a higher dose of 366

    itacitinib (300 nM, approximately five-fold higher that the cellular IC50) showed a modest but 367

    statistically significant inhibition on the antitumor activity (Fig. 6A). As internal controls, the 368

    antitumor activity of non-transfected PBMCs was also tested. Non-transduced T-cells were 369

    unable to specifically lyse the target cells and were unaffected by neither itacitinib nor 370

    tocilizumab treatment beyond background levels (Fig. 6B). 371

    372

    Itacitinib does not affect CD19-CAR T-cell antitumor activity in vivo 373

    To test the effect of itacitinib in vivo, we studied the effect of oral itacitinib on the 374

    antitumor efficacy of CD19-CAR T-cells. Immunodeficient mice (NSG) were challenged with 375

    on July 6, 2021. © 2020 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 30, 2020; DOI: 10.1158/1078-0432.CCR-20-1739

    http://clincancerres.aacrjournals.org/

  • CD19+ nalm6-luciferase expressing human lymphoma cells. Once the tumor was engrafted (day 376

    4), the mice received an adoptive transfer of 3 × 106 human CD19-CAR T-cells. In this model, 377

    CAR T-cells were able to control tumor growth, measured by luciferin expression (Fig. 7). 378

    Importantly, daily oral itacitinib dosing (120 mg/kg) of the animals did not affect the antitumor 379

    activity of adoptively transferred CAR T-cells, thus indicating that, at targeted doses, itacitinib 380

    does not have a significant effect on the antitumor activity of CAR T-cells. 381

    Finally, to confirm that itacitinib would not affect CAR T-cell activity on a more 382

    aggressive model, immunodeficient mice (NSG) were challenged with 5 × 106 CD19

    + 383

    NAMALWA-luciferase expressing human lymphoma cells. Once the tumor was engrafted (day 384

    7), the mice received an adoptive transfer of human CD19-CAR T-cells. When compared with 385

    animals receiving control cells, animals receiving an adoptive transfer of CAR T-cells had a 386

    significant tumor growth delay, measured both by luciferin expression (Supplementary Fig. 387

    S4B) and expansion of survival (P = 0.0014) (Supplementary Fig. S4C). Even in this 388

    aggressive tumor model, daily oral itacitinib dosing of the animals did not affect the antitumor 389

    activity of adoptively transferred CAR T-cells (P = 0.1860), thus confirming the safety of 390

    prophylactic itacitinib dosing to prevent CRS. 391

    392

    Discussion 393 CAR T-cell therapy has shown unprecedented remission rates in patients with relapsed or 394

    refractory B-cell acute lymphoblastic leukemia. Unfortunately, the high frequency of side effects 395

    (mainly CRS and neurotoxicity) that often require extensive critical care support remains a 396

    barrier to the further development of CAR T-cell therapy. Current therapeutic options are limited 397

    to treating severe cases of CRS with tocilizumab or corticosteroids, but unfortunately severe 398

    toxicities and death still occasionally occur (38). Therefore, there is a clear need to develop 399

    prophylactic treatments to prevent (rather than treat) CRS without impairing antitumor efficacy. 400

    Despite being a very active research field, only recently clear predictors of CAR T-cell 401

    associated toxicities have been identified, and several factors, such as ferritin, C-reactive protein 402

    or tumor burden can be reliable predictors of severe CRS (11). A recent clinical trial on pediatric 403

    leukemia patients suggests that early therapeutic intervention with tocilizumab is effective at 404

    reducing CRS rates without affecting CAR T-cell proliferation or antitumor efficacy (39). 405

    However, this data should not be generalized to other conditions such as lymphoma as clinical 406

    management strategies are different than leukemia and concerns remain over the potential impact 407

    of corticosteroids on CAR T-cell activation and proliferation (14). 408

    Recent advances in our understanding of the JAK/STAT pathway have highlighted its 409

    role in T-cell priming, activation, and expansion. Indeed, several small-molecules targeting the 410

    JAK/STAT pathway have shown clinical potential in a number of indications (reviewed in (40)). 411

    Biochemical and cellular profiling of itacitinib confirmed it to be a potent JAK1 inhibitor with 412

    large fold-selectivity margins over the other family members (JAK2, JAK3, and TYK2) or 413

    unrelated kinases (41). Importantly, JAK1 either independently or in cooperation with other 414

    members of the JAK family, mediate the signaling of a number of inflammatory cytokines 415

    implicated in CRS, including GM-CSF, IFN-, IL-6 and IL-12 (42, 43). Crucially, specific 416 inhibition of JAK1 may prevent some of the side effects associated with JAK1/JAK2 inhibitors 417

    such as lymphopenia (42). 418

    Consistent with the potent disruption of the JAK1-specific STAT signaling activity, here 419

    we show that itacitinib inhibited multiple cytokines involved in CRS (including IL-6, IL-12, and 420

    on July 6, 2021. © 2020 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 30, 2020; DOI: 10.1158/1078-0432.CCR-20-1739

    http://clincancerres.aacrjournals.org/

  • IFN-) in two different models of systemic inflammation in a dose-dependent manner. 421

    Importantly, only cytokines dependent upon JAK1 were affected, as levels of IL-5 and other 422

    JAK1-independent cytokines were unaffected by itacitinib treatment. It is also important to note 423

    that while the effect on inflammatory cytokine levels was profound and statistically significant, 424

    plasma levels of such cytokines were still elevated when compared with naïve animals, thus 425

    potentially dampening the hyper-immune activation state, but avoiding broad 426

    immunosuppression. 427

    In addition to the effect of itacitinib on the production of cytokines by T-cells and CAR 428

    T-cells, our findings show that itacitinib is also able to reduce the secretion of inflammatory 429

    cytokines by macrophages, both in vitro and in vivo. Recent studies have shown that IL-1 and IL-430

    6 produced by host monocytes/macrophages play a key role in the pathogenesis of CRS and 431

    neurotoxicity (30, 44). Our data demonstrate that itacitinib is able to inhibit IL-6 production by 432

    murine macrophages in vivo, demonstrating its potential utility in treating the most prominent 433

    source of inflammatory cytokines during CRS. The ability of itacitinib to inhibit the production 434

    of CRS-associated cytokines produced by a range of cell types (both of murine and human 435

    origin, an important distinction considering the differences between mouse and human biology 436

    (45)) is particularly noteworthy, as such a strategy may be necessary for the effective treatment 437

    of CRS. 438

    To achieve clinical efficacy while preventing CRS, it is imperative that clinical doses of 439

    itacitinib do not interfere with CAR T-cell proliferation and antitumor activity. Our findings 440

    confirm that itacitinib concentrations relevant for cellular IC50 (in vitro) and drug 441

    pharmacokinetic exposures that mimic JAK1 target coverage in vivo have no significant effect 442

    on the proliferation rate of PBMCs or four different CAR T-cell constructs. More importantly, 443

    our data show that in preclinical models, where animals receive an oral dose of itacitinib that 444

    mimic human JAK1 pharmacologic inhibition, prophylactic treatment with itacitinib does not 445

    affect the in vivo antitumor efficacy of CAR T-cells. 446

    In summary, we have demonstrated that itacitinib at clinically relevant doses can prevent 447

    CAR T-cell associated CRS in preclinical models without negatively affecting antitumor 448

    efficacy. Reducing the incidence and severity of CRS has the potential to benefit clinical 449

    practice, including the use of CAR T-cell therapy in patients in previously high-risk populations. 450

    Based on these results, a phase II clinical trial of itacitinib for the prevention of CRS induced by 451

    CD19-CAR T-cell therapy was initiated (NCT04071366). Results from our preclinical study may 452

    have implications in other settings; future studies are required to further investigate the role of 453

    JAK inhibition beyond CAR T-cells induced CRS (i.e., bispecific antibodies (46)) or conditions 454

    (including HLH (9, 47) and respiratory infections such as SARS (48), MERS (49), or Covid-19 455

    (50-52)) where CSR may play a role. 456

    457

    Acknowledgments 458 This study was funded by Incyte Corporation, and supported in part by an NIH grant 459

    RO1CA226983-03 awarded to R.O’C. 460

    We are thankful to April Barbour, Michael Howell, and Lea Burke for useful discussions about 461

    pharmacokinetics and translational relevance of the manuscript. Editorial assistance was 462

    on July 6, 2021. © 2020 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 30, 2020; DOI: 10.1158/1078-0432.CCR-20-1739

    http://clincancerres.aacrjournals.org/

  • provided by Envision Pharma Group, Inc. (Philadelphia, PA, USA), and funded by Incyte 463

    Corporation. 464

    465

    References 466 1. Jain MD, Bachmeier CA, Phuoc VH, Chavez JC. Axicabtagene ciloleucel (KTE-C19), an 467

    anti-CD19 CAR T therapy for the treatment of relapsed/refractory aggressive B-cell non-468

    Hodgkin's lymphoma. Ther Clin Risk Manag 2018;14:1007–17. 469

    2. Lee DW, Kochenderfer JN, Stetler-Stevenson M, Cui YK, Delbrook C, Feldman SA, et 470

    al. T cells expressing CD19 chimeric antigen receptors for acute lymphoblastic 471

    leukaemia in children and young adults: a phase 1 dose-escalation trial. Lancet 472

    2015;385:517–28. 473

    3. Schuster SJ, Bishop MR, Tam CS, Waller EK, Borchmann P, McGuirk JP, et al. 474

    Tisagenlecleucel in adult relapsed or refractory diffuse large B-cell lymphoma. N Engl J 475

    Med 2019;380:45–56. 476

    4. Gill S, Maus MV, Porter DL. Chimeric antigen receptor T cell therapy: 25 years in the 477

    making. Blood Rev 2016;30:157–67. 478

    5. Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, Bunin NJ, et al. Chimeric antigen 479

    receptor T cells for sustained remissions in leukemia. N Engl J Med 2014;371:1507–17. 480

    6. Santomasso B, Bachier C, Westin J, Rezvani K, Shpall EJ. The other side of CAR T-cell 481

    therapy: cytokine release syndrome, neurologic toxicity, and financial burden. Am Soc 482

    Clin Oncol Educ Book 2019;39:433–44. 483

    7. Neelapu SS. Managing the toxicities of CAR T-cell therapy. Hematol Oncol 2019;37 484

    Suppl 1:48–52. 485

    8. Maude SL, Barrett D, Teachey DT, Grupp SA. Managing cytokine release syndrome 486

    associated with novel T cell-engaging therapies. Cancer J 2014;20:119-22. 487

    9. Das R, Guan P, Sprague L, Verbist K, Tedrick P, An QA, et al. Janus kinase inhibition 488

    lessens inflammation and ameliorates disease in murine models of hemophagocytic 489

    lymphohistiocytosis. Blood 2016;127:1666-75. 490

    10. Bugelski PJ, Achuthanandam R, Capocasale RJ, Treacy G, Bouman-Thio E. Monoclonal 491

    antibody-induced cytokine-release syndrome. Expert Rev Clin Immunol 2009;5:499–521. 492

    11. Teachey DT, Lacey SF, Shaw PA, Melenhorst JJ, Maude SL, Frey N, et al. Identification 493

    of predictive biomarkers for cytokine release syndrome after chimeric antigen receptor T-494

    cell therapy for acute lymphoblastic leukemia. Cancer Discov 2016;6:664–79. 495

    12. Le RQ, Li L, Yuan W, Shord SS, Nie L, Habtemariam BA, et al. FDA approval 496

    summary: tocilizumab for treatment of chimeric antigen receptor T cell-induced severe or 497

    life-threatening cytokine release syndrome. Oncologist 2018;23:943–7. 498

    13. Neelapu SS, Locke FL, Bartlett NL, Lekakis LJ, Miklos DB, Jacobson CA, et al. 499

    Axicabtagene ciloleucel CAR T-cell therapy in refractory large B-cell lymphoma. N Engl 500

    J Med 2017;377:2531–44. 501

    14. Davila ML, Riviere I, Wang X, Bartido S, Park J, Curran K, et al. Efficacy and toxicity 502

    management of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia. Sci 503

    Transl Med 2014;6:224ra25. 504

    15. Howell MD, Kuo FI, Smith PA. Targeting the Janus kinase family in autoimmune skin 505

    diseases. Front Immunol 2019;10:2342. 506

    on July 6, 2021. © 2020 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 30, 2020; DOI: 10.1158/1078-0432.CCR-20-1739

    http://clincancerres.aacrjournals.org/

  • 16. Banerjee S, Biehl A, Gadina M, Hasni S, Schwartz DM. JAK-STAT signaling as a target 507

    for inflammatory and autoimmune diseases: current and future prospects. Drugs 508

    2017;77:521–46. 509

    17. Fragoulis GE, McInnes IB, Siebert S. JAK-inhibitors. New players in the field of 510

    immune-mediated diseases, beyond rheumatoid arthritis. Rheumatology (Oxford) 511

    2019;58:i43–i54. 512

    18. Zhang Y, Warren MS, Zhang X, Diamond S, Williams B, Punwani N, et al. Impact on 513

    creatinine renal clearance by the interplay of multiple renal transporters: a case study 514

    with INCB039110. Drug Metab Dispos 2015;43:485–9. 515

    19. Verstovsek S, Mesa RA, Gotlib J, Gupta V, DiPersio JF, Catalano JV, et al. Long-term 516

    treatment with ruxolitinib for patients with myelofibrosis: 5-year update from the 517

    randomized, double-blind, placebo-controlled, phase 3 COMFORT-I trial. J Hematol 518

    Oncol 2017;10:55. 519

    20. Quah BJC, Warren HS, Parish CR. Monitoring lymphocyte proliferation in vitro and in 520

    vivo with the intracellular fluorescent dye carboxyfluorescein diacetate succinimidyl 521

    ester. Nat Protoc 2007;2:2049–56. 522

    21. Milone M, Batish SD, Daube JR. Myotonic dystrophy type 2 with focal asymmetric 523

    muscle weakness and no electrical myotonia. Muscle Nerve 2009;39:383–5. 524

    22. Burga RA, Thorn M, Point GR, Guha P, Nguyen CT, Licata LA, et al. Liver myeloid-525

    derived suppressor cells expand in response to liver metastases in mice and inhibit the 526

    anti-tumor efficacy of anti-CEA CAR-T. Cancer Immunol Immunother 2015;64:817–29. 527

    23. Richman SA, Nunez-Cruz S, Moghimi B, Li LZ, Gershenson ZT, Mourelatos Z, et al. 528

    High-affinity GD2-specific CAR T cells induce fatal encephalitis in a preclinical 529

    neuroblastoma model. Cancer Immunol Res 2018;6:36–46. 530

    24. Johnson LA, Scholler J, Ohkuri T, Kosaka A, Patel PR, McGettigan SE, et al. Rational 531

    development and characterization of humanized anti-EGFR variant III chimeric antigen 532

    receptor T cells for glioblastoma. Sci Transl Med 2015;7:275ra22. 533

    25. Ghassemi S, Nunez-Cruz S, O'Connor RS, Fraietta JA, Patel PR, Scholler J, et al. 534

    Reducing ex vivo culture improves the antileukemic activity of chimeric antigen receptor 535

    (CAR) T cells. Cancer Immunol Res 2018;6:1100–9. 536

    26. Gantner F, Leist M, Lohse AW, Germann PG, Tiegs G. Concanavalin A-induced T-cell-537

    mediated hepatic injury in mice: the role of tumor necrosis factor. Hepatology 538

    1995;21:190–8. 539

    27. Juvekar A, Ruggeri B, Condon S, Borkowski A, Huber R, Smith P. Itacitinib, a JAK1 540

    Selective Inhibitor Preserves Graft-Versus-Leukemia (GVL), Enhances Survival and Is 541

    Highly Efficacious in a MHC-Mismatched Mouse Model of Acute GvHD. Blood 542

    2018;132:4522. 543

    28. Huarte E, O'Connor RS, Parker M, Huang T, Milone MC, Smith P. Prophylactic 544

    Itacitinib (INCB039110) for the Prevention of Cytokine Release Syndrome Induced By 545

    Chimeric Antigen Receptor T-Cells (CAR-T-cells) Therapy. Blood 2019;134:1934. 546

    29. Schroeder MA, Khoury HJ, Jagasia M, Ali H, Schiller GJ, Staser K, et al. A phase 1 trial 547

    of itacitinib, a selective JAK1 inhibitor, in patients with acute graft-versus-host disease. 548

    Blood Adv 2020;4:1656-69. 549

    30. Norelli M, Camisa B, Barbiera G, Falcone L, Purevdorj A, Genua M, et al. Monocyte-550

    derived IL-1 and IL-6 are differentially required for cytokine-release syndrome and 551

    neurotoxicity due to CAR T cells. Nat Med 2018;24:739–48. 552

    on July 6, 2021. © 2020 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 30, 2020; DOI: 10.1158/1078-0432.CCR-20-1739

    http://clincancerres.aacrjournals.org/

  • 31. Qin H, Holdbrooks AT, Liu Y, Reynolds SL, Yanagisawa LL, Benveniste EN. SOCS3 553

    deficiency promotes M1 macrophage polarization and inflammation. J Immunol 554

    2012;189:3439–48. 555

    32. Brudno JN, Kochenderfer JN. Toxicities of chimeric antigen receptor T cells: recognition 556

    and management. Blood 2016;127:3321–30. 557

    33. Levi M, Grange S, Frey N. Exposure-exposure relationship of tocilizumab, an anti-IL-6 558

    receptor monoclonal antibody, in a large population of patients with rheumatoid arthritis. 559

    J Clin Pharmacol 2013. doi: 10.1177/0091270011437585. 560

    34. Barbour AM, Punwani N, Epstein N, Landman R, Cimino E, Yuska B, et al. Effect of 561

    itraconazole or rifampin on itacitinib pharmacokinetics when administered orally in 562

    healthy subjects. J Clin Pharmacol 2019;59:1641–7. 563

    35. Li H, Huang Y, Jiang DQ, Cui LZ, He Z, Wang C, et al. Antitumor activity of EGFR-564

    specific CAR T cells against non-small-cell lung cancer cells in vitro and in mice. Cell 565

    Death Dis 2018;9:177. 566

    36. Lu T, Ramakrishnan R, Altiok S, Youn JI, Cheng P, Celis E, et al. Tumor-infiltrating 567

    myeloid cells induce tumor cell resistance to cytotoxic T cells in mice. J Clin Invest 568

    2011;121:4015–29. 569

    37. Frey N, Grange S, Woodworth T. Population pharmacokinetic analysis of tocilizumab in 570

    patients with rheumatoid arthritis. J Clin Pharmacol 2010;50:754–66. 571

    38. Rafiq S, Hackett CS, Brentjens RJ. Engineering strategies to overcome the current 572

    roadblocks in CAR T cell therapy. Nat Rev Clin Oncol 2020;17:147–67. 573

    39. Gardner RA, Ceppi F, Rivers J, Annesley C, Summers C, Taraseviciute A, et al. 574

    Preemptive mitigation of CD19 CAR T-cell cytokine release syndrome without 575

    attenuation of antileukemic efficacy. Blood 2019;134:2149–58. 576

    40. Covington M, He X, Scuron M, Li J, Collins R, Juvekar A, et al. Preclinical 577

    characterization of itacitinib (INCB039110), a novel selective inhibitor of JAK1, for the 578

    treatment of inflammatory diseases. Eur J Pharmacol 2020;885:173505. 579

    41. Srinivas N, Barbour AM, Epstein N, Zhou G, Petusky S, Xun Z, et al. The effect of renal 580

    impairment on the pharmacokinetics and safety of itacitinib. J Clin Pharmacol 2020. doi: 581

    10.1002/jcph.1601. 582

    42. Murray PJ. The JAK-STAT signaling pathway: input and output integration. J Immunol 583

    2007;178:2623–9. 584

    43. Spinelli FR, Conti F, Gadina M. HiJAKing SARS-CoV-2? The potential role of JAK 585

    inhibitors in the management of COVID-19. Sci Immunol 2020;5. 586

    44. Giavridis T, van der Stegen SJC, Eyquem J, Hamieh M, Piersigilli A, Sadelain M. CAR 587

    T cell-induced cytokine release syndrome is mediated by macrophages and abated by IL-588

    1 blockade. Nat Med 2018;24:731–8. 589

    45. Mestas J, Hughes CC. Of mice and not men: differences between mouse and human 590

    immunology. J Immunol 2004;172:2731-8. 591

    46. Duell J, Lammers PE, Djuretic I, Chunyk AG, Alekar S, Jacobs I, et al. Bispecific 592

    antibodies in the treatment of hematologic malignancies. Clin Pharmacol Ther 593

    2019;106:781–91. 594

    47. Castillo L, Carcillo J. Secondary hemophagocytic lymphohistiocytosis and severe 595

    sepsis/systemic inflammatory response syndrome/multiorgan dysfunction 596

    syndrome/macrophage activation syndrome share common intermediate phenotypes on a 597

    spectrum of inflammation. Pediatr Crit Care Med 2009;10:387–92. 598

    on July 6, 2021. © 2020 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 30, 2020; DOI: 10.1158/1078-0432.CCR-20-1739

    http://clincancerres.aacrjournals.org/

  • 48. Castilletti C, Bordi L, Lalle E, Rozera G, Poccia F, Agrati C, et al. Coordinate induction 599

    of IFN-alpha and -gamma by SARS-CoV also in the absence of virus replication. 600

    Virology 2005;341:163–9. 601

    49. Zheng Y, Wang QY. Bioinformatics analysis on molecular mechanism of ribavirin and 602

    interferon-alpha in treating MERS-CoV. Zhonghua Liu Xing Bing Xue Za Zhi 603

    2016;37:291–3. 604

    50. Huang C, Wang Y, Li X, Ren L, Zhao J, Hu Y, et al. Clinical features of patients infected 605

    with 2019 novel coronavirus in Wuhan, China. Lancet 2020;395:497–506. 606

    51. Mehta P, McAuley DF, Brown M, Sanchez E, Tattersall RS, Manson JJ, et al. COVID-607

    19: consider cytokine storm syndromes and immunosuppression. Lancet 2020;395:1033–608

    4. 609

    52. Cao Y, Wei J, Zou L, Jiang T, Wang G, Chen L, et al. Ruxolitinib in treatment of severe 610

    coronavirus disease 2019 (COVID-19): A multicenter, single-blind, randomized 611

    controlled trial. J Allergy Clin Immunol 2020. doi: 10.1016/j.jaci.2020.05.019. 612

    613

    on July 6, 2021. © 2020 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 30, 2020; DOI: 10.1158/1078-0432.CCR-20-1739

    http://clincancerres.aacrjournals.org/

  • Figures Legends 614

    615

    Figure 1. 616

    Itacitinib reduces cytokine levels in murine models of acute inflammation. (A) BALB/c mice 617

    were orally dosed with vehicle control, or 60 or 120 mg/kg of itacitinib. One hour later, animals 618

    were challenged with Concanavalin-A (ConA), and 120 minutes later sacrificed and plasma 619

    collected (prophylactic). (B) BALB/c mice were challenged with ConA, 30 minutes later were 620

    orally dosed with vehicle control, or 60 or 120 mg/kg of itacitinib and 2 hours after ConA 621

    challenge sacrificed (therapeutic). N = 5 animals per group. MSD analysis was performed to 622

    detect the levels of pro-inflammatory cytokines. Data represent mean ± SEM, and P values were 623

    calculated by two-way ANOVA. *P < 0.05, ** P < 0.01, ***P < 0.001, **** P < 0.0001. Data 624

    are representative of four independent experiments. 625

    626

    Figure 2. 627

    Itacitinib reduces IL-6 production by macrophages. (A) Macrophages harvested from 628

    C57BL-6 mice were treated with increasing doses of itacitinib 1 day in advance of being 629

    activated with 5 ng/mL LPS. Twenty-four hours after LPS treatment, supernatant was harvested 630

    and cytokine levels measured. (B) C57BL/6 mice were prophylactically orally dosed with 631

    vehicle, or 60 or 120 mg/kg of itacitinib b.i.d. for 3 days. Mice then received intraperitoneal 632

    injections of LPS (5 µg per animal). Two hours after injection, mice were euthanized and IL-6 633

    levels were measured in the peritoneal lavage. Data represent mean ± SEM, and P values were 634

    calculated by two-way ANOVA. *P < 0.05, ***P < 0.001. Data are representative of two 635

    independent experiments. 636

    637

    Figure 3. 638

    Itacitinib reduces CAR T-cell cytokine production. CD19-CAR T-cells were expanded with 639

    either itacitinib or the anti–IL-6 receptor tocilizumab. Three days later, they were co-cultured 640

    with CD19+ lymphoma cells (E:T ratio = 2.5:1) for 6 hours and cytokines were measured in the 641

    supernatant by MSD. (A) Experiment scheme. (B) Data represent mean ± SEM, and P values 642

    were calculated by two-way ANOVA. *P < 0.05, **P < 0.01, ***P < 0.001. Data are 643

    representative of two independent experiments. 644

    645

    Figure 4. 646

    Itacitinib does not affect PBMC proliferation nor cytolytic activity. (A) Flow cytometry 647

    quantification of T-cell expansion of human PBMCs expanded with DMSO (positive control) or 648

    50, 150, or 1000 nM of itacitinib. T-cells were expanded using anti-CD3/CD28-coated beads, 649

    and proliferation was measured every other day by flow cytometry. (B) GD2-CAR T-cells were 650

    expanded in the presence of different itacitinib concentrations and proliferation was measured 651

    every other day by flow cytometry. (C) Lytic activity of GD2-CAR T-cells against SY5Y 652

    neuroblastoma cells (GD-2+) was determined by measuring luciferase activity. As an internal 653

    control, lytic activity of non-transduced T cells was also measured (background dotted line). (D) 654

    on July 6, 2021. © 2020 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 30, 2020; DOI: 10.1158/1078-0432.CCR-20-1739

    http://clincancerres.aacrjournals.org/

  • EGFR-CAR T-cells were expanded with anti-CD3/CD28 beads for 2 weeks and then 655

    restimulated with EGFR beads in the presence of itacitinib (100 or 250 nM). Proliferation was 656

    measured every other day by flow cytometry. 657

    658

    Figure 5. 659

    Itacitinib does not impair T-cell antitumor activity in vivo. C57BL/6 animals were 660

    subcutaneously inoculated with an OVA-expressing EG-7 tumor cell line. Starting at day 5, 661

    corresponding groups received oral doses of itacitinib or vehicle for 2 weeks, and at day 8 they 662

    received an adoptive transfer (A.T.) of naïve OT-1 CD8 T-cells. (A) Experiment scheme. (B) 663

    Tumor growth was monitored and compared between groups. N = 10 animals per group. Data 664

    represent mean ± SEM, and P values were calculated by two-way ANOVA. * P < 0.05, n.s., not 665

    significant. Data are representative of two independent experiments. 666

    667

    Figure 6. 668

    Itacitinib does not affect CD19-CAR T-cell cytolytic activity in vitro. (A) CD19-CAR T-cells 669

    or (B) nontransduced PBMC were expanded with either itacitinib or anti–IL-6 receptor 670

    (tocilizumab) at the indicated concentrations. Three days later, they were co-cultured with 671

    luciferin expressing CD19+ lymphoma cells (E:T ratio = 2.5:1) for 6 hours, luciferase substrate 672

    was added and luminescence was recorded by a luminometer. Data represent mean ± SEM, and 673

    P values were calculated by two-way ANOVA. *P < 0.05, **P < 0.01, ***P < 0.001, n.s., not 674

    significant. 675

    676

    Figure 7. 677

    Itacitinib does not affect CD19-CAR T-cell antitumor activity in vivo. Immunodeficient NSG 678

    mice were inoculated with 2.5 × 106 CD19

    + human lymphoma Nalm6 luciferase-expressing 679

    Nalm6 cell line. Starting 1 day later, animals received b.i.d. doses of itacitinib or vehicle for 10 680

    days. At day 4, post-tumor injection, corresponding animals received an adoptive transfer of 3 × 681

    106 CD19-CAR T-cells. (A) Experiment scheme. (B) Anesthetized mice were imaged using a 682

    Xenogen IVIS Spectrum system to measure bioluminescence once a week. N = 5–10 animals per 683

    group. 684

    on July 6, 2021. © 2020 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 30, 2020; DOI: 10.1158/1078-0432.CCR-20-1739

    http://clincancerres.aacrjournals.org/

  • 15,000

    12,000

    9000

    6000

    3000

    0

    40

    30

    20

    10

    0

    400

    300

    200

    100

    0

    2500

    2000

    1500

    1000

    500

    0

    A

    Co

    nc

    en

    tra

    tio

    n, p

    g/m

    L

    **

    **** ****

    IL-6 IL-12 IFNγ IL-5

    4000

    3000

    2000

    1000

    0

    400

    300

    200

    100

    0

    300

    200

    100

    0

    300

    200

    100

    0

    B

    Co

    nc

    en

    tra

    tio

    n, p

    g/m

    L

    Na

    ïve

    Ve

    hic

    le

    60

    mg

    /kg

    12

    0 m

    g/k

    g

    Na

    ïve

    Ve

    hic

    le

    60

    mg

    /kg

    12

    0 m

    g/k

    g

    Na

    ïve

    Ve

    hic

    le

    60

    mg

    /kg

    12

    0 m

    g/k

    g

    Na

    ïve

    Ve

    hic

    le

    60

    mg

    /kg

    12

    0 m

    g/k

    g

    Itacitinib Itacitinib Itacitinib Itacitinib

    **** **** ********

    ****

    6000

    4000

    2000

    0

    IL-4

    1500

    1000

    500

    0

    Na

    ïve

    Ve

    hic

    le

    60

    mg

    /kg

    12

    0 m

    g/k

    g

    Itacitinib

    Itacitinib

    Concanavalin-A

    0 60 180 mins

    Plasma collection

    and cytokines

    Concanavalin-A

    Itacitinib

    0 30 120 mins

    Plasma collection

    and cytokines

    Figure 1.on July 6, 2021. © 2020 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 30, 2020; DOI: 10.1158/1078-0432.CCR-20-1739

    http://clincancerres.aacrjournals.org/

  • A B

    15,000

    10,000

    5000

    0

    IL-6

    co

    nc

    en

    tra

    tio

    n, p

    g/m

    L

    20,000

    15,000

    10,000

    5000

    0

    IL-6

    co

    nc

    en

    tra

    tio

    n, p

    g/m

    L

    Un

    tre

    ate

    d

    Na

    ïve

    Ve

    hic

    le

    Ita

    cin

    itib

    60

    mg

    /kg

    Ita

    cin

    itib

    12

    0 m

    g/k

    g

    LP

    S

    LP

    S +

    ita

    citin

    ib 1

    00

    nM

    LP

    S +

    ita

    citin

    ib 2

    00

    nM

    LP

    S +

    ita

    citin

    ib 5

    00

    nM

    LP

    S +

    ita

    citin

    ib 1

    00

    0 n

    M

    *

    *** ****

    *

    Day 4 Day 8

    Figure 2.on July 6, 2021. © 2020 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 30, 2020; DOI: 10.1158/1078-0432.CCR-20-1739

    http://clincancerres.aacrjournals.org/

  • 400

    300

    200

    100

    0

    Co

    nc

    en

    tra

    tio

    n, p

    g/m

    LC

    on

    ce

    ntr

    ati

    on

    , p

    g/m

    L

    1000

    800

    600

    400

    200

    0

    100

    80

    60

    40

    20

    0

    2000

    1500

    1000

    500

    0

    IL-2 IFNγ

    IL-6 IL-8

    DM

    SO

    20

    μg

    /mL

    20

    0 μ

    g/m

    L

    10

    0 n

    M

    30

    0 n

    M

    T-c

    ells

    10

    0 μ

    g/m

    L

    Tocilizumab Itacitinib

    DM

    SO

    20

    μg

    /mL

    20

    0 μ

    g/m

    L

    10

    0 n

    M

    30

    0 n

    M

    T-c

    ells

    10

    0 μ

    g/m

    L

    Tocilizumab Itacitinib

    DM

    SO

    20

    μg

    /mL

    20

    0 μ

    g/m

    L

    10

    0 n

    M

    30

    0 n

    M

    T-c

    ells

    10

    0 μ

    g/m

    L

    Tocilizumab Itacitinib

    DM

    SO

    20

    μg

    /mL

    20

    0 μ

    g/m

    L

    10

    0 n

    M

    30

    0 n

    M

    T-c

    ells

    10

    0 μ

    g/m

    L

    Tocilizumab Itacitinib

    **

    ***

    **

    ******

    *

    *

    ***

    *** ***

    A

    B

    CAR T-cell transduction

    Itacitinib or tocilizumab

    0 1 2 3 days

    CD19+ NAWALMA

    Supernatant collection

    and cytokine quantification

    Figure 3.on July 6, 2021. © 2020 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 30, 2020; DOI: 10.1158/1078-0432.CCR-20-1739

    http://clincancerres.aacrjournals.org/

  • A

    8

    6

    4

    2

    0Po

    pu

    lati

    on

    do

    ub

    lin

    gs

    0 2 4 6 8 10

    Days of culture

    B

    4

    3

    2

    1

    0

    Po

    pu

    lati

    on

    do

    ub

    lin

    gs

    0 2 4 6 8 10

    Days of culture

    C

    80

    60

    40

    20

    10

    Tu

    mo

    r c

    ell ly

    sis

    , %

    Non-

    transduced

    0 100 250 500

    D

    8

    6

    4

    2

    0Po

    pu

    lati

    on

    do

    ub

    lin

    gs

    0 2 4 6 8 10 12 14 16 18 20

    Days of cultureItacitinib, nM

    DMSOItacitinib 50 nM

    Itacitinib 100 nM

    Itacitinib 1000 nM

    DMSOItacitinib 100 nM

    Itacitinib 250 nM

    Itacitinib 500 nM

    Non-transducedDMSO

    Itacitinib 100 nM

    Itacitinib 250 nM

    Figure 4.on July 6, 2021. © 2020 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 30, 2020; DOI: 10.1158/1078-0432.CCR-20-1739

    http://clincancerres.aacrjournals.org/

  • 2500

    2000

    1500

    1000

    500

    0Tu

    mo

    r v

    olu

    me

    , m

    ea

    n ±

    SE

    M

    7 9 11 13 16 20 22

    Days

    *n.s.n.s.

    Vehicle

    A.T. + vehicle

    A.T. + itacitinib 60 mg/kg

    A.T. + itacitinib 120 mg/kg

    A

    B

    EG7 (OVA+) cells

    Itacitinib

    OT-I transfer

    0 5 8 2218 days

    Tumor monitoring

    Figure 5.on July 6, 2021. © 2020 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 30, 2020; DOI: 10.1158/1078-0432.CCR-20-1739

    http://clincancerres.aacrjournals.org/

  • 100

    80

    60

    40

    20

    0

    Cy

    toto

    xic

    ity

    , %

    DMSO 20 100 200 100 300

    Tocilizumab, μg/mL Itacitinib, nM

    **** **

    ***

    A B

    n.s.

    100

    80

    60

    40

    20

    0

    Cy

    toto

    xic

    ity

    , %

    DMSO 20 100 200 100 300

    Tocilizumab, μg/mL Itacitinib, nM

    CAR T-cells Nontransduced T-cells

    Figure 6.on July 6, 2021. © 2020 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 30, 2020; DOI: 10.1158/1078-0432.CCR-20-1739

    http://clincancerres.aacrjournals.org/

  • 1000

    100

    10

    1

    0.1

    0.01

    0.001

    To

    tal fl

    ux

    , P

    /S ×

    10

    9

    0 10 20 30 40

    Days elapsed

    Vehicle

    Non-transduced + vehicle

    CAR T-cells + vehicle

    CAR T-cells + itacitinib 120 mg/kg

    A

    B

    CD19+ Nalm6

    Itacitinib

    CD19+ CAR T-cells transfer

    0 1 4 3511 days

    Survival

    Figure 7.on July 6, 2021. © 2020 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 30, 2020; DOI: 10.1158/1078-0432.CCR-20-1739

    http://clincancerres.aacrjournals.org/

  • Published OnlineFirst September 30, 2020.Clin Cancer Res Eduardo Huarte, Roddy S. O'Conner, Michael T. Peel, et al. T-Cell TherapyAssociated with Cytokine Release Syndrome Induced by CAR Itacitinib (INCB039110), a JAK1 inhibitor, Reduces Cytokines

    Updated version

    10.1158/1078-0432.CCR-20-1739doi:

    Access the most recent version of this article at:

    Material

    Supplementary

    http://clincancerres.aacrjournals.org/content/suppl/2020/09/30/1078-0432.CCR-20-1739.DC1

    Access the most recent supplemental material at:

    Manuscript

    Authoredited. Author manuscripts have been peer reviewed and accepted for publication but have not yet been

    E-mail alerts related to this article or journal.Sign up to receive free email-alerts

    Subscriptions

    Reprints and

    [email protected] at

    To order reprints of this article or to subscribe to the journal, contact the AACR Publications

    Permissions

    Rightslink site. Click on "Request Permissions" which will take you to the Copyright Clearance Center's (CCC)

    .http://clincancerres.aacrjournals.org/content/early/2020/09/30/1078-0432.CCR-20-1739To request permission to re-use all or part of this article, use this link

    on July 6, 2021. © 2020 American Association for Cancer Research.clincancerres.aacrjournals.org Downloaded from

    Author manuscripts have been peer reviewed and accepted for publication but have not yet been edited. Author Manuscript Published OnlineFirst on September 30, 2020; DOI: 10.1158/1078-0432.CCR-20-1739

    http://clincancerres.aacrjournals.org/lookup/doi/10.1158/1078-0432.CCR-20-1739http://clincancerres.aacrjournals.org/content/suppl/2020/09/30/1078-0432.CCR-20-1739.DC1http://clincancerres.aacrjournals.org/cgi/alertsmailto:[email protected]://clincancerres.aacrjournals.org/content/early/2020/09/30/1078-0432.CCR-20-1739http://clincancerres.aacrjournals.org/

    Article FileFigure 1Figure 2Figure 3Figure 4Figure 5Figure 6Figure 7