eradication of hepatocellular carcinoma by nkg2d-specific ...€¦ · 04.09.2019  · tumors,...

35
1 Eradication of hepatocellular carcinoma by NKG2D-specific CAR T- 1 cells 2 3 Bin Sun 1,2,3, # , Dong Yang 1,2, 3, # , Hongjiu Dai 3, #, * , Xiuyun Liu 2 , Ru Jia 4 , Xiaoyue Cui 2 , 4 Wenxuan Li 5 , Changchun Cai 7 , Jianming Xu 4 , Xudong Zhao 1,2,6, * 5 1 Laboratory of tumor animal models and anti-aging, State Key Laboratory of 6 Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 7 610041, Sichuan. China. 8 2 Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese 9 Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, 10 Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 11 650223, China 12 3 Nanjing Kaedi Biotech Co. Ltd., Nanjing, Jiangsu 211100, China 13 4 Department of GI Oncology, the 307 Hospital of Academy of Military Medical 14 Science, Beijing 100071, China 15 5 College of Life Sciences, Sichuan University, Chengdu, Sichuan 610064, China 16 6 Center for Excellence in Animal Evolution and Genetics, Chinese Academy of 17 Sciences, Kunming 650223, China 18 7 Department of Gastroenterology, The Affiliated Hospital of Jiujiang University, 19 Jiujiang, Jiangxi 332000, China 20 21 # These authors contributed equally to the study. 22 23 Running title: NKG2D-BBz CAR-T eliminates hepatocellular cancer 24 25 on March 29, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.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 4, 2019; DOI: 10.1158/2326-6066.CIR-19-0026

Upload: others

Post on 19-Oct-2020

3 views

Category:

Documents


0 download

TRANSCRIPT

  • 1

    Eradication of hepatocellular carcinoma by NKG2D-specific CAR T-1

    cells 2

    3

    Bin Sun1,2,3, #

    , Dong Yang1,2, 3, #

    , Hongjiu Dai3, #, *

    , Xiuyun Liu2, Ru Jia

    4, Xiaoyue Cui

    2, 4

    Wenxuan Li5, Changchun Cai

    7, Jianming Xu

    4, Xudong Zhao

    1,2,6, * 5

    1 Laboratory of tumor animal models and anti-aging, State Key Laboratory of 6

    Biotherapy and Cancer Center, West China Hospital, Sichuan University, Chengdu 7

    610041, Sichuan. China. 8

    2 Key Laboratory of Animal Models and Human Disease Mechanisms of Chinese 9

    Academy of Sciences/Key Laboratory of Bioactive Peptides of Yunnan Province, 10

    Kunming Institute of Zoology, Chinese Academy of Sciences, Kunming, Yunnan 11

    650223, China 12

    3 Nanjing Kaedi Biotech Co. Ltd., Nanjing, Jiangsu 211100, China 13

    4 Department of GI Oncology, the 307 Hospital of Academy of Military Medical 14

    Science, Beijing 100071, China 15

    5 College of Life Sciences, Sichuan University, Chengdu, Sichuan 610064, China 16

    6 Center for Excellence in Animal Evolution and Genetics, Chinese Academy of 17

    Sciences, Kunming 650223, China 18

    7 Department of Gastroenterology, The Affiliated Hospital of Jiujiang University, 19

    Jiujiang, Jiangxi 332000, China 20

    21

    # These authors contributed equally to the study. 22

    23

    Running title: NKG2D-BBz CAR-T eliminates hepatocellular cancer 24

    25

    on March 29, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.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 4, 2019; DOI: 10.1158/2326-6066.CIR-19-0026

    http://cancerimmunolres.aacrjournals.org/

  • 2

    Key words: NKG2D, NKG2DL, HCC, CAR-T cells 26

    27

    Financial Support 28

    This work was financially supported by the National Natural Science Foundation of China 29

    (U1702289 to X.Z., 81802976 to D.Y.) 30

    31

    *Co-corresponding authors 32

    Hongjiu Dai, Ph.D. 33

    NANJING KAEDI BIOTECH INC. 34

    No. 18 Zhilan Road, Building #5 Room 402, Science Park, Jiangning District, Nanjing, 35

    Jiangsu 211100, China 36

    E-mail: [email protected] 37

    Tel: +86-25-52187062 38

    39

    Xudong Zhao, Ph.D. 40

    Kunming Institute of Zoology, the Chinese Academy of Sciences 41

    No. 32 Jiaochang Donglu, Kunming, Yunnan 650223, P.R. China 42

    E-mail: [email protected] 43

    Tel: +86-871-68125430 44

    45

    46

    Disclosure of Potential Conflicts of Interest 47

    No potential conflicts of interest were disclosed. 48

    49

    Total number of figures: 6 50

    Total number of tables: 0 51

    Word count: 3460 52

    53

    on March 29, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.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 4, 2019; DOI: 10.1158/2326-6066.CIR-19-0026

    http://cancerimmunolres.aacrjournals.org/

  • 3

    54

    Abstract 55

    Despite the great success of chimeric antigen receptor T (CAR-T) cell therapy in the 56

    treatment of hematological malignancies, CAR-T cell therapy is limited in solid 57

    tumors, including hepatocellular carcinoma (HCC). NKG2D ligands (NKG2DLs) are 58

    generally absent on the surface of normal cells but are overexpressed on malignant 59

    cells, offering good targets for CAR-T therapy. Indeed, analysis of the Cancer 60

    Genome Atlas and HCC tumor samples showed that the expression of most 61

    NKG2DLs was elevated in tumors compared with normal tissues. Thus, we designed 62

    a novel NKG2D-CAR comprising the extracellular domain of human NKG2D, 4-1BB 63

    and CD3ζ signaling domains (BBz). NKG2D-BBz CAR-T cells efficiently killed the 64

    HCC cell lines SMMC-7721 and MHCC97H in vitro, which express high levels of 65

    NKG2DLs, whereas they less efficiently killed NKG2DL- silenced SMMC-7721 cells 66

    or NKG2DL-negative Hep3B cells. Overexpression of MICA or ULBP2 in Hep3B 67

    improved the killing capacity of NKG2D-BBz CAR-T cells. T cells expressing the 68

    NKG2D-BBz CAR effectively eradicated SMMC-7721 HCC xenografts. 69

    Collectively, these results suggested that NKG2D-BBz CAR-T cells could potently 70

    eliminate NKG2DL-high HCC cells both in vitro and in vivo, thereby providing a 71

    promising therapeutic intervention for NKG2DL-positive HCC patients. 72

    73

    74

    Introduction 75

    Liver cancer is the sixth most common cancer type and the fourth most common 76

    cause of cancer-related deaths worldwide (1) Hepatocellular carcinoma (HCC) 77

    accounts for more than 75–85% of all liver cancer cases (1). In developed countries, 78

    approximately 40% of HCC patients are diagnosed at an early stage due to health 79

    on March 29, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.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 4, 2019; DOI: 10.1158/2326-6066.CIR-19-0026

    http://cancerimmunolres.aacrjournals.org/

  • 4

    surveillance programs (2,3). According to the Barcelona Clinic Liver Cancer (BCLC) 80

    staging system, many therapies, such as partial liver resection, transplantation, and 81

    local ablation, have excellent effects on early-stage HCC patients (stages 0 and A), 82

    providing median survival rates of 60 months and beyond (2,4). Unfortunately, most 83

    HCC patients, especially in developing countries, are diagnosed at a later disease 84

    stage because the symptoms of liver cancer are not obvious until it is in its later stages 85

    (3,5). For more developed stages of HCC, only a few treatments have shown survival 86

    benefits. Patients at intermediate stages (stage B) benefit from chemoembolization 87

    and have an estimated median survival of 26 months (2,6). For patients at advanced 88

    stages (stage C), only two therapies have been approved for clinical use by the Food 89

    and Drug Administration (FDA), sorafenib as a frontline treatment and regorafenib as 90

    a second line treatment (7,8); these drugs extend the overall survival of patients by 2-91

    3 months but are often accompanied by treatment-induced adverse events,such as 92

    hypophosphatemia, weight loss, hand–foot skin reaction, hypertension, etc. (7,8). 93

    Therefore, novel strategies for the treatment of advanced HCC, such as 94

    immunotherapy with PD-1 antibodies or Chimeric antigen receptor T (CAR-T) cells, 95

    are currently being tested in clinical trials (2,3,9). 96

    Immunotherapy is an effective treatment strategy for several cancers, including 97

    HCC. For example, administration of a cytotoxic T-lymphocyte-associated protein 4 98

    (CTLA-4) inhibitor enhances antitumor immunity in a murine HCC model, with a 99

    tumor re-challenge rejection rate of 90% and an elimination rate of 50% for 100

    metastatic tumor(10). In addition, nivolumab (a fully human programmed death-1 101

    inhibitor) shows potential as a treatment for HCC with a manageable safety profile 102

    (11). Chimeric antigen receptor T (CAR-T) cell therapy is another emerging 103

    immunotherapeutic option for cancer treatment (2,3). CAR-T cells are engineered to 104

    on March 29, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.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 4, 2019; DOI: 10.1158/2326-6066.CIR-19-0026

    http://cancerimmunolres.aacrjournals.org/

  • 5

    express a specific CAR, which recognizes tumor-associated antigens on the surface of 105

    tumor cells and then kills these cells in a targeted manner (12). However, clinical 106

    application of this approach requires the identification of the tumor-associated 107

    antigens and the design of highly specific CARs. 108

    CAR-T immunotherapy has shown promise in the fight against cancer, especially 109

    for hematologic malignancies (12,13). Currently, the FDA has approved two CAR-T 110

    therapies for blood cancer: Yescarta (axicabtagene cioleucel) developed by Kite 111

    Pharma Inc. and Kymriah (tisangenlecleucel) developed by Novartis International AG 112

    (14). However, insufficient persistence of tumor-specific antigens (15), heterogeneity 113

    among tumor cells (16,17), presence of an immunosuppressive microenvironment 114

    (16), and toxicity due to off-target effects currently (17) compromise the therapeutic 115

    efficiency of CAR-T immunotherapy in solid cancers. There are a few of ongoing 116

    preclinical studies of CAR-T therapies for HCC, which target the HCC associated 117

    antigens GPC-3, MUC-1, and CEA. However, the clinical results of CAR-T cells in 118

    HCC have been disappointing and associate with severe side effects (18). Therefore, 119

    there is a need to further develop CAR-T therapies for the treatment of HCC. 120

    NK group 2 member D (NKG2D) is a type II transmembrane-anchored C-type 121

    lectin-like protein receptor expressed on natural killer (NK) cells, CD8+ T cells, 122

    subsets of γδ T cells, and some autoreactive CD4+ T cells (19,20). NKG2D 123

    recognizes its ligands (NKG2DLs), such as MHC I chain-related molecules A and B 124

    (MICA and MICB) (21) and six cytomegalovirus UL16-binding proteins (ULBP1–6) 125

    (22). NKG2DLs are generally absent on the surface of normal cells but are 126

    overexpressed on tumor cells, and their expression can be further increased by 127

    chemotherapy or radiation (23). Accordingly, NKG2DL is a potential target for CAR-128

    T therapy. Previous studies have indicated that ligation of NKG2D with its ligands 129

    on March 29, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.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 4, 2019; DOI: 10.1158/2326-6066.CIR-19-0026

    http://cancerimmunolres.aacrjournals.org/

  • 6

    can activate NK cells and stimulate T cells in vitro (24), and ectopic expression of 130

    NKG2DLs on tumor cells was sufficient to cause tumor rejection mediated by NK 131

    cells (25). NKG2D-expressing CARs exhibited robust anti-tumor efficacy in several 132

    different xenograft models, including models of multiple myeloma (26), ovarian 133

    carcinoma (27), osteosarcoma (28), glioblastoma (29), pancreatic cancer (30), and 134

    relapsed/refractory acute myeloid leukemia (31). NKG2DLs are also overexpressed in 135

    human HCC, and the expression is significantly and negatively associated with poor 136

    prognosis or early recurrence (32,33). However, no NKG2D-CAR has been reported 137

    for HCC treatment. Therefore, we designed a NKG2D-BBz CAR with high 138

    specificity and selectivity for HCC, and evaluated its antitumor activities using HCC 139

    cell lines in vitro and in a xenograft mouse model in vivo. 140

    141

    Materials and methods 142

    143

    Plasmid construction and lentiviral package 144

    The lentiviral vectors pTomo-pCMV-MICA-IRES-EGFP and pTomo-pCMV-ULBP2-145

    IRES-puro, which were used for overexpression of MICA and ULBP2 in Hep3B cells 146

    as described below, were constructed in a pTomo vector backbone (Addgene, USA). 147

    The full-length human MICA (accession_NM_000247, primer: F, 148

    ATGGGGCTGGGCCCGGTCTT; R, CTAGGCGCCCTCAGTGGAGCCAG ) and 149

    ULBP2 (accession_NM_025217, primer: F, ATGGCAGCAGCCGCCGCTACCAAG; 150

    R, TCAGATGCCAGGGAGGATGAAG) sequences were PCR-amplified using 151

    PrimeSTAR HS DNA Polymerase (Takara, China) and inserted into the plasmids 152

    between the XbaI and BamHI restriction sites. For the MICA andULBP2 shRNA 153

    plasmids, the target sequences were cloned into a plko.1 puro vector obtained from 154

    on March 29, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.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 4, 2019; DOI: 10.1158/2326-6066.CIR-19-0026

    http://cancerimmunolres.aacrjournals.org/

  • 7

    Addgene according to the manufacturer’s protocol (http://www.addgene.org/8453/). 155

    The target sequences were as follow: shMICA-1, GCAGAAGATGTCCTGGGAAAT; 156

    shMICA-2, ATTCAATTCCCTGCCTGGAT; shULBP2-1, 157

    CCTCCTCTTTGACTCAGAGAA; shULBP2-2, TGAGCACGGTCTTGATCAAAC. 158

    A codon-optimized targeting domain comprising the extracellular domain of human 159

    NKG2D or CD19 scFv (as shown in the Supplementary Table) was synthesized 160

    (Idobio, China) and fused to a CAR backbone comprising a human CD8 hinge spacer 161

    and transmembrane domain, 4-1BB co-stimulatory domain and CD3ζ (BBz) (as 162

    shown in the Supplementary Table 1). The entire encoding sequence of the CAR 163

    expression molecule (as shown in Supplementary Table 1) was cloned into the 164

    lentiviral vector LentiGuide-Puro (Addgene, USA) between the SmaI and MluI 165

    restriction sites to replace PuroR and an EF1a promoter was inserted in front of the 166

    CAR sequences by SmaI single digestion. For lentiviral package, the lentiviral 167

    plasmids were co-transfected into HEK293T cells with the packaging plasmids 168

    psPAX2 and pCMV-VSVG (Addgene, USA) at a ratio of 10:8:5. Lentivirus were 169

    harvested as described before (34). 170

    171

    Cell lines and culture 172

    The human liver cancer cell lines SMMC-7721, Hep3B, and MHCC97H were 173

    purchased from Guangzhou Jennio Biotech Co., Ltd. in March 2018 and validated 174

    using short tandem repeat (STR) profiling in October 2018.Hep3B cells were infected 175

    with pTomo-CMV-ULBP2-IRES-puro and pTomo-CMV-MICA-IRES-EGFP 176

    lentivirus [multiplicity of infection (MOI) = 10] to generate Hep3B-ULBP2 and 177

    Hep3B-MICA cell lines, respectively. SMMC-7721 cells were infected with pTomo-178

    CMV-luciferase-IRES-puro, plko-shMICA, and plko-shULBP2 lentivirus (MOI = 10) 179

    on March 29, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.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 4, 2019; DOI: 10.1158/2326-6066.CIR-19-0026

    http://cancerimmunolres.aacrjournals.org/

  • 8

    and subsequently selected by puromycin (1 μg/ml) for 2 weeks to generate SMMC-180

    7721-luciferase, SMMC7721-shMICA, SMMC7721-shULPB2, and SMMC7721-181

    shMICA-shULBP2 cell lines, respectively. Hep3B, Hep3B-MICA, and Hep3B-182

    ULBP2 cells were cultured in Dulbecco’s modified Eagle’s medium (Life 183

    Technologies, USA) supplemented with 10% FBS (Life Technologies, USA), 100 184

    U/ml penicillin, and 100 mg/ml streptomycin sulfate (Life Technologies, USA). 185

    SMMC-7721 and MHCC97H cells were cultured in RPMI1640 (Life Technologies, 186

    USA) supplemented with 10% FBS, 100 U/ml penicillin, and 100 mg/ml 187

    streptomycin sulfate. HEK293T cells used for lentiviral package were obtained from 188

    A. Lasorella (The Institute for Cancer Genetics, Columbia University Medical Center) 189

    in December 2011 and cultured in DMEM (Life Technologies, USA) supplemented 190

    with 10% FBS, 100 U/ml penicillin, and 100 mg/ml streptomycin sulfate. HEK293T 191

    cells were not validated within the past year. All the cells were cultured at 37°C in a 192

    humidified incubator with 5% CO2 and routinely confirmed to be Mycoplasma-free 193

    by PCR. 194

    195

    Generation of CAR-T cells 196

    Primary T cells were isolated from peripheral blood of three healthy donors and two 197

    HCC patients using the RosetteSep™ Human T Cell Enrichment Cocktail 198

    (STEMCELL, Canada) according to the manufacturer’s protocol. The purity of the 199

    isolated cells was detected by flow cytometry using phycoerythrin-conjugated anti-200

    human CD3 (Biolegend, 300408).T cells were cultured in RPMI1640 (Gibco, USA) 201

    supplemented with 10% FBS, 100 U/ml penicillin, 100 mg/ml streptomycin sulfate, 202

    and 200 U/ml IL-2 (PeproTech, USA). To generate CAR-T cells, T cells were 203

    stimulated with CD3/CD28 beads (Life Technologies, USA) at a ratio of 1:1 for 48 h 204

    on March 29, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.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 4, 2019; DOI: 10.1158/2326-6066.CIR-19-0026

    http://cancerimmunolres.aacrjournals.org/

  • 9

    and then infected with lentiviral particles at a MOI of 10. All the patient studies were 205

    approved by the Institutional Review Board at Kunming Institute of Zoology, Chinese 206

    Academy of Sciences (approved ID: SMKX-2019022) with written informed consent 207

    obtained from participants and conducted in accordance with the international ethical 208

    guidelines for biomedical research involving human subjects. 209

    210

    In vitro cytotoxicity assays 211

    The specific cytotoxicity of the CAR-modified T cells was tested against the various 212

    HCC cell lines at variable effector-to-target (E/T) ratios of 0.5:1, 1:1, 2:1, 4:1, and 213

    8:1. After 16 h of culture in RPMI1640 (Gibco, USA) supplemented with 10% FBS, 214

    100 U/ml penicillin, 100 mg/ml streptomycin sulfate, cytotoxicity was measured 215

    using Cell-Mediated Cytotoxicity Fluorometric Assay Kit (BioVison, K315-100, 216

    USA) according to the manufacturer’s protocol. 217

    218

    Flow cytometry 219

    Cells were harvested, washed twice with 1× PBS, and resuspended in cold PBS 220

    containing 2% FCS, 1% sodium azide (at a density of 1 × 106 cells/ml). Subsequently, 221

    labelled primary antibodies were added into the cell suspension according to the 222

    manufacturers’ instructions and incubated for 1h at 4℃ in the dark. To evaluate CAR 223

    expression, CD19-BBz CAR-T cells and NKG2D-BBz CAR-T cells were incubated 224

    with Alexa Fluor 647-conjugated goat anti-mouse F(ab)2 (Jackson 225

    ImmunoResearch,115-606-006) and allophycocyanin (APC)-conjugated anti-226

    NKG2D (Biolegend, 320808), respectively. APC-conjugated MICA (R&D Systems, 227

    FAB1300A), MICB (R&D Systems, FAB1599A), ULBP1 (R&D Systems, 228

    FAB1380A), ULBP2/5/6 (R&D Systems, FAB1298A), and ULBP3 (R&D Systems, 229

    on March 29, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.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 4, 2019; DOI: 10.1158/2326-6066.CIR-19-0026

    http://cancerimmunolres.aacrjournals.org/

  • 10

    FAB1517A) were used to determine the expression of NKG2DLs on different HCC 230

    cells. APC-anti-CD4 (Biolegend, 357408) and FITC-anti-CD8 (Biolegend, 344704) 231

    were used to detect the ratio of CD4-positive to CD8-positive T cells. All flow 232

    cytometry readings were performed on the BD LSR Fortessa system and analyzed 233

    using FlowJo software. 234

    Cell growth analysis 235

    To detect the proliferation capacity, a 5-ethynyl-uridine (EdU)-labeling assay was 236

    performed using Click-iT EdU imaging Kits (Invitrogen, C10337). Briefly, EdU was 237

    added to the cell culture medium at a final concentration 10 μM and incubated for 1 h 238

    at 37°C in a humidified incubator with 5% CO2. Then the cells were harvested and 239

    fixed with 4% paraformaldehyde. Permeabilization was performed with 0.3% Triton 240

    X-100 followed by incubation withClick-iT reaction cocktail containing Alexa 241

    Fluor® azide for 30 min at room temperature. The EdU incorporation rate was 242

    analyzed by flow cytometry. To measure the proportion of apoptotic cells, cells were 243

    harvested, washed with 1× PBS, and resuspended at a density of 1 × 106 cells/ml. 244

    Thereafter, 7-aminoactinomycin D (Sangon Biotech, E607304-0200, China) was 245

    added according to the manufacturer’s protocol and the ratio of stained cell was 246

    analyzed by flow cytometry. 247

    Cytokine release assay 248

    CD19-BBz CAR-T and NKG2D-BBz CAR-T cells were co-cultured with SMMC-249

    7721 cells in RPMI1640 (Gibco, USA) supplemented with 10% FBS, 100 U/ml 250

    penicillin, 100 mg/ml streptomycin sulfate at an E/T ratio of 0:1 and 5:1, respectively, 251

    for 16 h. 10 μL of the supernatant was collected and the concentrations of IFN-γ (BD 252

    Biosciences,550612), interleukin (IL)-10 (BD Biosciences,550613), tumor 253

    on March 29, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.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 4, 2019; DOI: 10.1158/2326-6066.CIR-19-0026

    http://cancerimmunolres.aacrjournals.org/

  • 11

    necrosis factor (TNF)-α (BD Biosciences,550610), and IL-2 (BD Biosciences,254

    550611) were determined using respective enzyme-linked immunosorbent assay 255

    (ELISA) kits as described above according to the manufacturer instructions. The 256

    quantification was performed on the Synergy H1 (BioTek, USA) by measuring 257

    absorbance at 450 nm. 258

    In vivo HCC xenograft model 259

    All protocols were approved by the animal ethics committee of the Kunming Institute 260

    of Zoology, Chinese Academy of Sciences. Five to six-week-old NOD-Prkdcscid

    261

    Il2rgtm1

    /Bcgen mice (B-NDG) were purchased from Jiangsu Biocytogen Co., Ltd. 262

    (Jiangsu, China). A total of 1 × 106 SMMC-7721-luciferase cells were suspended in 263

    PBS containing 30% Matrigel (BD Bioscience) and subcutaneously injected into the 264

    B-NDG mice. When the mean tumor bioluminescence reached ~5 × 106 265

    photons/second at one-week after tumor cell injectiona, the mice were anesthetized 266

    with 2.5% avertin by intraperitoneal injection (15 ml/kg) followed by the 267

    intraperitoneal injection of 150 mg/kg D-luciferin (BioVison, USA). Ten minutes 268

    later, bioluminescent signals were recorded using an in vivo imaging software (IVIS) 269

    system (Lumina Xr, USA), and the mice were randomly divided into the following 270

    four groups: (i) tail intravenous injection of 100 l of sterile saline only without T 271

    cells; (ii) 1 × 107 non-transduced T cells (NTD) in sterile saline; (iii) 1 × 10

    7 272

    genetically modified CD19-BBz CAR-T cells in sterile saline (CD19-BBz CAR); and 273

    (iv) 1 × 107 genetically modified NKG2D-BBz CAR-T cells in sterile saline 274

    (NKG2D-BBz CAR). After that, the bioluminescent signals were measured 275

    approximately every 10 days. The data were quantified using Living Image software 276

    (Caliper Life Science, USA). The mice were sacrificed when the tumor volume 277

    reached approximately 2000 mm3. 278

    on March 29, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.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 4, 2019; DOI: 10.1158/2326-6066.CIR-19-0026

    http://cancerimmunolres.aacrjournals.org/

  • 12

    Tissue microarray and immunohistochemistry (IHC) Analysis 279

    Immunohistochemistry analysis was performed as described previously (34). Briefly, 280

    sections cut from paraffin-embedded samples were deparaffinized, rehydrated, and 281

    processed for antigen retrieval with sodium citrate buffer (Beyotime, china). The 282

    sections were blocked with 10% serum from the same species as the source of the 283

    secondary antibody for 1 hour at room temperature and incubated with primary 284

    antibodies at 4°C overnight. Then, the sections were washed thrice in TBS 0.025% 285

    Triton with gentle agitation and incubated with secondary antibodies for 1 hour at 286

    room temperature. Chromogenic staining was performed on the sections using 3,3′-287

    Diaminobenzidine (Beyotime, P0203, china) according to the manufacturer’s 288

    protocol. The OD-CT-DgLive02-002 microarray (Outdo Biotech, Shanghai, China) 289

    containing 32 human liver cancer samples, HOrgN090PT02 microarray (Outdo 290

    Biotech, Shanghai, China) containing 90 normal tissues from major human organs 291

    (including the thyroid, tongue, esophagus, stomach, duodenum, colon, liver, pancreas, 292

    trachea, lung, heart, artery, skeletal muscle, skin, seminal vesicle, prostate, testis, 293

    bladder, brain, and spleen) and HLivH060CD03 microarray (Outdo Biotech, 294

    Shanghai, China) containing 7 non-tumor cirrhotic liver tissues were immunostained 295

    using anti-MICA (Abcam, ab93170, USA) at a dilution ratio of 1:200 or anti-ULBP2 296

    (thermo fisher, PA5-47118, USA) at a dilution ratio of 1:50. To investigate the 297

    persistence of the administrated human T cells in the mice, the sections of formalin-298

    fixed, paraffin-embedded lung, liver, bone marrow, hippocampus, spleen, kidney, 299

    pancreas, and tumor tissues from the CD19-BBz CAR-T group and NKG2D-BBz 300

    CAR-T groups were immuno-stained using an CD3-ξ antibody (Santa Cruz 301

    Biotechnology, sc-1239, USA) at a dilution ratio of 1:100. 302

    Karyotype assays 303

    on March 29, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.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 4, 2019; DOI: 10.1158/2326-6066.CIR-19-0026

    http://cancerimmunolres.aacrjournals.org/

  • 13

    NKG2D-BBz CAR-T cells were collected 10 days after infection with NKG2D-BBz 304

    CAR and normal T cells were used as controls. Karyotype assays were conducted as 305

    previously described (35). Briefly, cells were treated with colcemid (Sangon Biotech, 306

    A600322, China) at 37℃ for 2 hours, then harvested, washed in 0.075mol/L KCl at 307

    37℃ for 10 min. Then the cells were fixed in freshly prepared fixative 308

    (methanol/glacial acetic acid = 3/1), dropped onto slides, and dried at room 309

    temperature. The chromosome images were captured on the Olympus DP71 310

    microscope. 311

    Statistical analysis 312

    All statistical analyses were performed using GraphPad Prism 7.0 statistical software. 313

    The data were all presented as mean ± standard deviation (SD). Statistical differences 314

    between two groups were analyzed using Student’s t-tests with Welch correction. 315

    Statistical differences among three or more groups were analyzed by one-way 316

    ANOVA with Sidak correction. Statistical significance was defined as *P ≤ 0.05,

    **P ≤ 317

    0.01, ***

    P ≤ 0.001. 318

    319

    Results 320

    NKG2DLs were overexpressed in human liver cancer 321

    To clarify the expression of NKG2DLs in clinical samples, we analyzed the RNA-Seq 322

    data of paired liver cancer and normal tissues from the Cancer Genome Atlas (TCGA; 323

    N = 50). The results showed that MICA, MICB, ULBP1, ULBP2, ULBP4, and ULBP5 324

    were upregulated compared with those in paired normal tissues (Supplemental Fig. 325

    S1). Similarly, immunohistochemistry in human liver cancer specimens on the OD-326

    CT-DgLive02-002 microarray chip showed that expression levels of MICA and 327

    ULBP2 were elevated in 72% (23/32) and 97% (31/32) of the liver cancer tissues, 328

    on March 29, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.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 4, 2019; DOI: 10.1158/2326-6066.CIR-19-0026

    http://cancerimmunolres.aacrjournals.org/

  • 14

    respectively (Fig. 1A, B). Although the leading cause of HCC is cirrhosis and most 329

    HCC patients have liver cirrhosis, the expression of MICA and ULBP2 was absent in 330

    non-tumor cirrhotic liver tissue (Supplemental Fig. S2). In addition, flow cytometry 331

    showed that the expression levels of all NKG2DLs were low in the Hep3B cell line, 332

    while MICA and ULBP2 were highly expressed in the SMMC-7721 and MHCC97H 333

    cell lines (Fig. 1C). These results clearly supported the possibility of using NKG2DL 334

    as a target in HCC therapy. 335

    336

    NKG2D-BBz CAR-T cells lysed HCC cells in a NKG2DL-dependent manner 337

    We designed the lentiviral expression vectors encoding CD19-BBz and NKG2D-BBz 338

    as shown in Fig. 2A. T cells were isolated from PBMCs of healthy donors, and 99.2% 339

    of the cells were confirmed to be CD3-positive by flow cytometry (Supplemental Fig. 340

    S3). After activation with CD3/28 beads for 48 h, T cells were infected with CD19-341

    BBz and NKG2D-BBz lentivirus. After 72 h, CD19 and NKG2D-BBz CAR were 342

    detected in 42.3% and 46.1% of the T cells, respectively (Fig. 2B). The results of the 343

    cytotoxicity assay showed that at the E:T ratio of 8:1, NKG2D-BBz CAR-T cells 344

    could efficiently lyse SMMC-7721 and MHHC97H cells (approximately 100%), but 345

    not the NKG2DLs-negative Hep3B cell line (less than 30%). By contrast, CD19-BBz 346

    CAR-T cells failed to initiate the specific lysis of these HCC cell lines (Fig. 2C). 347

    Cytotoxic T cells secrete several cytokines upon killing target cells. To confirm the 348

    specific cytotoxicity of NKG2D-BBz CAR-T cells, cytokine were assessed in the cell 349

    culture medium, demonstrating that TNF-α, IFN-γ, IL-10, and IL-2 were significantly 350

    increased in SMMC-7721 cells cultured with NKG2D-BBz CAR-T cells, but not in 351

    the culture medium of CD19-BBz CAR-T or NTD T cells (Fig. 2D). 352

    To investigate if the cytotoxicity of NKG2D-BBz CAR-T cell was positively 353

    on March 29, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.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 4, 2019; DOI: 10.1158/2326-6066.CIR-19-0026

    http://cancerimmunolres.aacrjournals.org/

  • 15

    correlated with the cell surface expression of NKG2D ligands, lentiviral particles that 354

    induced MICA or ULBP2 silencing were constructed and used to infect SMMC-7721 355

    cells (Supplemental Fig. S4). Silencing MICA and ULBP2 in SMMC7721-shMICA-356

    shULBP2 cells was confirmed by flow cytometry (MICA, 72.5%; ULBP2, 80.1%; 357

    Fig 3A). Correspondingly, NKG2D-BBz CAR-T cells showed less potent cytotoxicity 358

    against SMMC7721-shMICA-shULBP2 cells compared to other cell lines (Fig 3B). 359

    Ectopic NKG2DL-expressed cell lines Hep3B-MICA and Hep3B-ULBP2 were also 360

    generated (Fig 3C). The cytotoxicity of NKG2D-BBz CAR-T cells towards the 361

    Hep3B-MICA or Hep3B-ULBP2 cells was greater than 60%, even at the lowest E:T 362

    ratio of 0.5:1 (Fig 3D), whereas the cytotoxicity towards the wild-type Hep3B cells 363

    was only about 4% at the same E:T ratio (Fig 3D). These data indicate that the 364

    cytotoxicity of NKG2D-BBz CAR-T on HCC cells is NKG2DL-dependent. 365

    366

    NKG2D-BBz CAR-T cells suppressed the growth of SMMC-7721 xenografts 367

    To explore the therapeutic efficacy of NKG2D-BBZ CAR-T cells in vivo, 368

    subcutaneous xenografts were established by injecting transfected SMMC-7721-369

    luciferase cells into B-NDG mice, followed by of the CAR T-cell therapy 7 days later. 370

    In all mice receiving saline, NTD, or CD19-BBz CAR-T, SMMC-7721 xenografts 371

    progressively grew. However, the xenografts in mice receiving NKG2D-BBz CAR-T 372

    cells had delayed tumor growth than the three control groups. Overall, 50% (3/6) of 373

    the mice showed no tumors 19 days after infusion with NKG2D-BBz CAR-T cells, 374

    while the other three mice only had minimal residual tumor remaining (Fig. 4A, B). 375

    At the end of the study, four out of the six mice in the group receiving the NKG2D-376

    BBz CAR-T cells were tumor-free, and the other two mice had very small tumors 377

    remaining with bioluminescence of approximately 5 × 106 photons/second (Fig. 4A, 378

    on March 29, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.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 4, 2019; DOI: 10.1158/2326-6066.CIR-19-0026

    http://cancerimmunolres.aacrjournals.org/

  • 16

    B). Collectively, these data demonstrate that NKG2D-BBz CAR-T cells can 379

    successfully inhibit the tumorigenesis of subcutaneous SMMC-7721 xenografts. 380

    Since the direct interaction between CAR-T cells and cancer cells is necessary 381

    for a therapeutic effect, human T cells in the xenograft tumor and normal tissues were 382

    detected via immunohistochemistry using anti-CD3-ξ. The immunohistochemistry 383

    results showed that NKG2D-BBz CAR-T cells accumulated in SMMC-7721 384

    xenografts, while only small quantities of CD19-BBz CAR-T cells were found in 385

    tumors. Human T cells were also detected in the bone marrow of both the NKG2D-386

    BBz CAR-T and CD19-BBz CAR-T groups but were absent in the lung, liver, kidney, 387

    and brain (Fig. 4C). These results indicate that NKG2D-BBz CAR-T cells 388

    preferentially infiltrated and resided in the HCC tumors, correlating to the efficient 389

    suppression of tumor growth. 390

    391

    NKG2D-BBz CAR-T cells derived from HCC patients showed antitumor activity 392

    HCC patients have reported abnormalities in their lymphocyte function (36), thus 393

    the antitumor activity of NKG2D CAR-T cells derived from HCC patient T cells 394

    needed to be verified. Peripheral blood collected from two HCC patients without 395

    radiotherapy and chemotherapy was used to isolate T cells. Compared with the T cells 396

    of a healthy donor, the patients’ T cells proliferated more slowly (Supplemental Fig. 397

    S5). The T cells were infected with CD19-BBz and NKG2D-BBz lentiviral particles 398

    and the expression of CARs was detected by flow cytometry. The patients’ CAR-T 399

    cells had similar CAR expression efficiency and CD4/CD8 ratios to that of T cells 400

    derived from healthy donor (Supplemental Fig. S6&S7). To determine the 401

    cytotoxicity of NKG2D-BBz CAR-T cells from different sources against HCC cells, 402

    we incubated T cells with SMMC7721 cells at different E:T ratios. The results 403

    on March 29, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.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 4, 2019; DOI: 10.1158/2326-6066.CIR-19-0026

    http://cancerimmunolres.aacrjournals.org/

  • 17

    showed that patients’ NKG2D CAR-T cell have a comparable antitumor activity with 404

    NKG2D CAR-T cell derived from a healthy donor (Fig. 5A). Correspondingly, both 405

    NKG2D-BBz CAR-T cells showed effective antitumor activity against xenografts 406

    formed by SMMC-7721 cells in B-NDG mice (Fig. 5B&C). As expected, xenografts 407

    treated with normal saline (NS), or NTD, CD19-BBz CAR-T cells derived from HCC 408

    patients grew rapidly (Fig. 5B&C). 409

    Safety evaluation of CAR-T cells 410

    The expression of tumor-associated antigens on normal cells often leads to severe off-411

    target effects from CAR-T therapy, limiting their clinical application (37). Analysis of 412

    the HOrgN090PT02 microarray chip demonstrated the lack of MICA expression in 413

    most human tissues (thyroid, tongue, esophageal epithelium, gastric mucosa, jejunal 414

    mucous membrane, ileal mucous membrane, appendix, mucous membrane of the 415

    rectum, liver, pancreas, trachea, lung, myocardium, artery, skeletal muscle, seminal 416

    vesicle, prostate, bladder, testis, medulla oblongata, telencephalon, epencephal, 417

    brainstem, and spleen) except for the skin (3/3) (Fig. 6A). Considering gene transfer 418

    mediated by lentivirus can possibly induce genome instability and cell malignant 419

    transformation, karyotype and proliferation assays were performed to evaluate the 420

    safety of CAR expression by lentivral infection. NKG2D-BBz CAR-T cells 421

    maintained a normal karyotype compared with that of untransduced T cells for up to 422

    14 days post-transduction (Fig. 6B). At 72 h after infection with the lentivirus 423

    expressing CD19-BBz CAR and NKG2D-BBz CAR, there were no significant 424

    alterations detected in the proliferation and apoptotic status of T cells (Supplemental 425

    Fig. S8). 426

    427

    Discussion 428

    on March 29, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.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 4, 2019; DOI: 10.1158/2326-6066.CIR-19-0026

    http://cancerimmunolres.aacrjournals.org/

  • 18

    The potential therapeutic efficacy of NKG2D-based CAR-T cells has been tested in 429

    several malignant tumors, but their efficacy in liver cancer has not been assessed until 430

    now. In this study, we designed a novel NKG2D-BBz CAR using the extracellular 431

    domain of NKG2D instead of the full-length sequence, followed by 4-1BB and CD3ζ, 432

    and successfully produced NKG2D-BBz CAR-T cells that showed strong cytotoxicity 433

    against HCC cell lines in vitro as well as a therapeutic effect against HCC cell 434

    xenografts in vivo These results demonstrated that NKG2D-BBz CAR-T can 435

    specifically eradicate HCC cells in an NKG2DL-dependent manner, providing a 436

    scientific basis for proceeding to a clinical trial for NKG2DL-positive patients. 437

    CAR-T immunotherapy has excellent effects in hematologic malignancies (38). 438

    Accordingly, many studies have been performed to extrapolate the success to solid 439

    tumors, yet few have shown success (39). To date, several tumor-associated antigens 440

    (TAA) expressed in liver tumor cells have been exploited for CAR-T therapy, and 441

    were demonstrated to lyse HCC cells in vitro and eliminate mouse xenografts in vivo, 442

    including GPC-3 (40), MUC1 (41), AFP (42), and CEA (43). However, the clinical 443

    results of CAR-T cells in the treatment of HCC have yielded poor therapeutic 444

    responses (18,44). The immunosuppressive tumor microenvironment is one of the 445

    main contributors to the limited antitumor effects of CAR-T in liver cancers (18,36). 446

    NKG2D-based CAR-T cells eliminate the inhibitive tumor microenvironment through 447

    the following mechanisms: 1) killing the tumor neovasculature expressing NKG2DLs 448

    to ameliorate the microenvironment and enhance the immunotherapy effect (45); 2) 449

    killing immunosuppressive myeloid-derived suppressor cells and regulatory T cells; 450

    and 3) recruiting myeloid cells and activated macrophages to modulate the immune 451

    tumor microenvironment (46,47). Tumor heterogeneity is a major cause of tumor 452

    immune escape and often leads to recurrence and metastasis. NKG2D-based CAR-T 453

    on March 29, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.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 4, 2019; DOI: 10.1158/2326-6066.CIR-19-0026

    http://cancerimmunolres.aacrjournals.org/

  • 19

    cells can target multiple ligands expressed in tumor cells and thus may be beneficial 454

    for the radical treatment of tumors (48). T lymphocytes, both CD4+ T helper cells and 455

    CD8+ cytotoxic T cells usually play an important role in tumor suppression, and a 456

    significant decrease in T cell activity has been observed in HCC patients (36). 457

    However, in the present study, the CD4+/CD8+ ratio in NKG2D CAR-T cells in HCC 458

    patients was still within the normal range (from 0.39 to 7.43) (49). In addition, 459

    NKG2D CAR-T derived from patient T cells showed strong killing activity 460

    specifically toward liver cancer cells. These findings suggest NKG2D CAR-T as a 461

    potential multi-functional CAR-T therapy. 462

    A major concern of CAR-T cells in clinical practice is their potential off-tumor 463

    effects. For example, treatment with the receptor tyrosine-protein kinase ERBB2-464

    CART resulted in acute respiratory distress syndrome in a patient with colorectal liver 465

    metastasis due to the expression of Erbb2 in the lungs (50). In addition, treatment of 466

    CEA antigen-directed CAR-T cells in colon cancer patients resulted in severe colitis 467

    due to antigen recognition of the normal colonic tissue (51). Therefore, the specificity 468

    of TAAs is of great importance in the application of CAR-T therapy. Using a tissue 469

    chip including 96 samples from 27 tissue types, we confirmed that MICA, one of the 470

    most important NKG2DLs, is not expressed in most normal tissues, which is in line 471

    with previous reports (52,53). MICA is localized in the cytoplasm of most MICA-472

    positive epithelial cells (54), suggesting that these MICA-positive epithelial cells 473

    would not be targeted by NKG2D-BBz CAR-T cells. CAR NKR-2 (developed by 474

    Celyad) was the first-generation NKG2D CAR with infusion of full-length NKG2D, 475

    DAP10, and the CD3ζ cytoplasmic domain. Although MICA was found to be slightly 476

    expressed in the skin, a phase I clinical trial of NKR-2 (NCT02203825) revealed that 477

    seven patients with acute myeloid leukemia and five patients with multiple myeloma 478

    on March 29, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.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 4, 2019; DOI: 10.1158/2326-6066.CIR-19-0026

    http://cancerimmunolres.aacrjournals.org/

  • 20

    were successfully treated with NKR-2 cells without obvious toxicities or other 479

    adverse events (55). These data demonstrated the safety of NKG2D-based CAR-T 480

    therapy. 481

    Taken together, this study clarified the therapeutic roles of NKG2D-BBz CAR-T 482

    cells in preclinical HCC models. Considering the proven safety of NKG2D CAR-T 483

    cells in clinical trials with other cancers, their use can be expected to be applied in 484

    clinical trials for NKG2DL-positive patients. 485

    486

    Acknowledgements 487

    We would like to thank Guolan Ma from the Public Technology Service Center, 488

    Kunming Institute of Zoology, Chinese Academy of Sciences for her technical 489

    support in the flow cytometric analysis. 490

    491

    References 492

    1. Bray F, Ferlay J, Soerjomataram I, Siegel RL, Torre LA, Jemal A. Global cancer 493

    statistics 2018: GLOBOCAN estimates of incidence and mortality worldwide for 36 494

    cancers in 185 countries. CA Cancer J Clin 2018;68(6):394-424 doi 495

    10.3322/caac.21492. 496

    2. Llovet JM, Zucman-Rossi J, Pikarsky E, Sangro B, Schwartz M, Sherman M, et al. 497

    Hepatocellular carcinoma. Nat Rev Dis Primers 2016;2:16018 doi 498

    10.1038/nrdp.2016.18. 499

    3. Llovet JM, Montal R, Sia D, Finn RS. Molecular therapies and precision medicine for 500

    hepatocellular carcinoma. Nat Rev Clin Oncol 2018;15(10):599-616 doi 501

    10.1038/s41571-018-0073-4. 502

    4. Yang JD, Roberts LR. Hepatocellular carcinoma: A global view. Nat Rev 503

    Gastroenterol Hepatol 2010;7(8):448-58 doi 10.1038/nrgastro.2010.100. 504

    5. Dimitroulis D, Damaskos C, Valsami S, Davakis S, Garmpis N, Spartalis E, et al. 505

    From diagnosis to treatment of hepatocellular carcinoma: An epidemic problem for 506

    both developed and developing world. World J Gastroenterol 2017;23(29):5282-94 507

    doi 10.3748/wjg.v23.i29.5282. 508

    6. Llovet JM, Real MI, Montana X, Planas R, Coll S, Aponte J, et al. Arterial 509

    embolisation or chemoembolisation versus symptomatic treatment in patients with 510

    on March 29, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.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 4, 2019; DOI: 10.1158/2326-6066.CIR-19-0026

    http://cancerimmunolres.aacrjournals.org/

  • 21

    unresectable hepatocellular carcinoma: a randomised controlled trial. Lancet 511

    2002;359(9319):1734-9 doi 10.1016/S0140-6736(02)08649-X. 512

    7. Llovet JM, Ricci S, Mazzaferro V, Hilgard P, Gane E, Blanc JF, et al. Sorafenib in 513

    advanced hepatocellular carcinoma. N Engl J Med 2008;359(4):378-90 doi 514

    10.1056/NEJMoa0708857. 515

    8. Bruix J, Qin S, Merle P, Granito A, Huang YH, Bodoky G, et al. Regorafenib for 516

    patients with hepatocellular carcinoma who progressed on sorafenib treatment 517

    (RESORCE): a randomised, double-blind, placebo-controlled, phase 3 trial. Lancet 518

    2017;389(10064):56-66 doi 10.1016/S0140-6736(16)32453-9. 519

    9. Lee S, Loecher M, Iyer R. Immunomodulation in hepatocellular cancer. J Gastrointest 520

    Oncol 2018;9(1):208-19 doi 10.21037/jgo.2017.06.08. 521

    10. Chen ZB, Shen SQ, Peng BG, Tao JP. Intratumoural GM-CSF microspheres and 522

    CTLA-4 blockade enhance the antitumour immunity induced by thermal ablation in a 523

    subcutaneous murine hepatoma model. Int J Hyperther 2009;25(5):374-82 doi 524

    10.1080/02656730902976807. 525

    11. El-Khoueiry AB, Sangro B, Yau T, Crocenzi TS, Kudo M, Hsu C, et al. Nivolumab in 526

    patients with advanced hepatocellular carcinoma (CheckMate 040): an open-label, 527

    non-comparative, phase 1/2 dose escalation and expansion trial. Lancet 528

    2017;389(10088):2492-502 doi 10.1016/S0140-6736(17)31046-2. 529

    12. Maude SL, Frey N, Shaw PA, Aplenc R, Barrett DM, Bunin NJ, et al. Chimeric 530

    antigen receptor T cells for sustained remissions in leukemia. N Engl J Med 531

    2014;371(16):1507-17 doi 10.1056/NEJMoa1407222. 532

    13. Curran KJ, Brentjens RJ. Chimeric antigen receptor T cells for cancer 533

    immunotherapy. J Clin Oncol 2015;33(15):1703-6 doi 10.1200/JCO.2014.60.3449. 534

    14. Yip A, Webster RM. The market for chimeric antigen receptor T cell therapies. Nat 535

    Rev Drug Discov 2018;17(3):161-2 doi 10.1038/nrd.2017.266. 536

    15. Kershaw MH, Westwood JA, Parker LL, Wang G, Eshhar Z, Mavroukakis SA, et al. A 537

    phase I study on adoptive immunotherapy using gene-modified T cells for ovarian 538

    cancer. Clinical Cancer Research 2006;12(20):6106-15 doi 10.1158/1078-0432.Ccr-539

    06-1183. 540

    16. Gilham DE, Debets R, Pule M, Hawkins RE, Abken H. CAR-T cells and solid tumors: 541

    tuning T cells to challenge an inveterate foe. Trends Mol Med 2012;18(7):377-84 doi 542

    10.1016/j.molmed.2012.04.009. 543

    17. Kakarla S, Gottschalk S. CAR T cells for solid tumors: armed and ready to go? 544

    Cancer J 2014;20(2):151-5 doi 10.1097/PPO.0000000000000032. 545

    18. Chen Y, E CY, Gong ZW, Liu S, Wang ZX, Yang YS, et al. Chimeric antigen 546

    receptor-engineered T-cell therapy for liver cancer. Hepatobiliary Pancreat Dis Int 547

    2018;17(4):301-9 doi 10.1016/j.hbpd.2018.05.005. 548

    19. Raulet DH. Roles of the NKG2D immunoreceptor and its ligands. Nat Rev Immunol 549

    2003;3(10):781-90 doi 10.1038/nri1199. 550

    on March 29, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.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 4, 2019; DOI: 10.1158/2326-6066.CIR-19-0026

    http://cancerimmunolres.aacrjournals.org/

  • 22

    20. Zhang J, Basher F, Wu JD. NKG2D Ligands in Tumor Immunity: Two Sides of a 551

    Coin. Front Immunol 2015;6:97 doi 10.3389/fimmu.2015.00097. 552

    21. Champsaur M, Lanier LL. Effect of NKG2D ligand expression on host immune 553

    responses. Immunol Rev 2010;235(1):267-85 doi 10.1111/j.0105-2896.2010.00893.x. 554

    22. El-Gazzar A, Groh V, Spies T. Immunobiology and conflicting roles of the human 555

    NKG2D lymphocyte receptor and its ligands in cancer. J Immunol 2013;191(4):1509-556

    15 doi 10.4049/jimmunol.1301071. 557

    23. Dhar P, Wu JD. NKG2D and its ligands in cancer. Curr Opin Immunol 2018;51:55-61 558

    doi 10.1016/j.coi.2018.02.004. 559

    24. Bauer S, Groh V, Wu J, Steinle A, Phillips JH, Lanier LL, et al. Activation of NK cells 560

    and T cells by NKG2D, a receptor for stress-inducible MICA. Science 561

    1999;285(5428):727-9. 562

    25. Cerwenka A, Baron JL, Lanier LL. Ectopic expression of retinoic acid early inducible-563

    1 gene (RAE-1) permits natural killer cell-mediated rejection of a MHC class I-bearing 564

    tumor in vivo. Proc Natl Acad Sci U S A 2001;98(20):11521-6 doi 565

    10.1073/pnas.201238598. 566

    26. Barber A, Zhang T, Megli CJ, Wu JL, Meehan KR, Sentman CL. Chimeric NKG2D 567

    receptor-expressing T cells as an immunotherapy for multiple myeloma. Exp Hematol 568

    2008;36(10):1318-28 doi 10.1016/j.exphem.2008.04.010. 569

    27. Barber A, Zhang T, DeMars LR, Conejo-Garcia J, Roby KF, Sentman CL. Chimeric 570

    NKG2D receptor-bearing T cells as immunotherapy for ovarian cancer. Cancer 571

    Research 2007;67(10):5003-8 doi 10.1158/0008-5472.Can-06-4047. 572

    28. Fernandez L, Metais JY, Escudero A, Vela M, Valentin J, Vallcorba I, et al. Memory T 573

    Cells Expressing an NKG2D-CAR Efficiently Target Osteosarcoma Cells. Clinical 574

    Cancer Research 2017;23(19):5824-35 doi 10.1158/1078-0432.Ccr-17-0075. 575

    29. Weiss T, Schneider H, Silginer M, Steinle A, Pruschy M, Polic B, et al. NKG2D-576

    Dependent Antitumor Effects of Chemotherapy and Radiotherapy against 577

    Glioblastoma. Clin Cancer Res 2018;24(4):882-95 doi 10.1158/1078-0432.CCR-17-578

    1766. 579

    30. Demoulin B, Cook WJ, Murad J, Graber DJ, Sentman ML, Lonez C, et al. Exploiting 580

    natural killer group 2D receptors for CAR T-cell therapy. Future Oncol 581

    2017;13(18):1593-605 doi 10.2217/fon-2017-0102. 582

    31. Sallman DA, Brayer J, Sagatys EM, Lonez C, Breman E, Agaugue S, et al. NKG2D-583

    based chimeric antigen receptor therapy induced remission in a relapsed/refractory 584

    acute myeloid leukemia patient. Haematologica 2018 doi 585

    10.3324/haematol.2017.186742. 586

    32. Kamimura H, Yamagiwa S, Tsuchiya A, Takamura M, Matsuda Y, Ohkoshi S, et al. 587

    Reduced NKG2D ligand expression in hepatocellular carcinoma correlates with early 588

    recurrence. J Hepatol 2012;56(2):381-8 doi 10.1016/j.jhep.2011.06.017. 589

    33. Fang L, Gong JY, Wang Y, Liu RR, Li ZS, Wang Z, et al. MICA/B expression is 590

    on March 29, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.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 4, 2019; DOI: 10.1158/2326-6066.CIR-19-0026

    http://cancerimmunolres.aacrjournals.org/

  • 23

    inhibited by unfolded protein response and associated with poor prognosis in human 591

    hepatocellular carcinoma. J Exp Clin Canc Res 2014;33 doi ARTN 76 592

    10.1186/s13046-014-0076-7. 593

    34. Yang D, Sun B, Zhang XH, Cheng DM, Yu XP, Yan LZ, et al. Huwe1 Sustains 594

    Normal Ovarian Epithelial Cell Transformation and Tumor Growth through the 595

    Histone H1.3-H19 Cascade. Cancer Res 2017;77(18):4773-84 doi 10.1158/0008-596

    5472.Can-16-2597. 597

    35. Chou CH, Yang NK, Liu TY, Tai SK, Hsu DS, Chen YW, et al. Chromosome 598

    instability modulated by BMI1-AURKA signaling drives progression in head and neck 599

    cancer. Cancer Res 2013;73(2):953-66 doi 10.1158/0008-5472.CAN-12-2397. 600

    36. Sachdeva M, Chawla YK, Arora SK. Immunology of hepatocellular carcinoma. World 601

    J Hepatol 2015;7(17):2080-90 doi 10.4254/wjh.v7.i17.2080. 602

    37. Bonifant CL, Jackson HJ, Brentjens RJ, Curran KJ. Toxicity and management in CAR 603

    T-cell therapy. Mol Ther-Oncolytics 2016;3 doi UNSP 16011 604

    10.1038/mto.2016.11. 605

    38. Gauthier J, Yakoub-Agha I. Chimeric antigen-receptor T-cell therapy for 606

    hematological malignancies and solid tumors: Clinical data to date, current limitations 607

    and perspectives. Curr Res Transl Med 2017;65(3):93-102 doi 608

    10.1016/j.retram.2017.08.003. 609

    39. Harper K. CAR T-cell Therapy for Solid Tumors? Cancer Discov 2018;8(11):1341- doi 610

    Doi 10.1158/2159-8290.Cd-Nd2018-008. 611

    40. Gao H, Li K, Tu H, Pan X, Jiang H, Shi B, et al. Development of T cells redirected to 612

    glypican-3 for the treatment of hepatocellular carcinoma. Clin Cancer Res 613

    2014;20(24):6418-28 doi 10.1158/1078-0432.CCR-14-1170. 614

    41. Ma YD, Wang Z, Gong RZ, Li LF, Wu HP, Jin HJ, et al. Specific cytotoxicity of MUC1 615

    chimeric antigen receptor-engineered Jurkat T cells against hepatocellular 616

    carcinoma. 2014. 1177-82 p. 617

    42. Liu H, Xu Y, Xiang J, Long L, Green S, Yang Z, et al. Targeting Alpha-Fetoprotein 618

    (AFP)-MHC Complex with CAR T-Cell Therapy for Liver Cancer. Clin Cancer Res 619

    2017;23(2):478-88 doi 10.1158/1078-0432.CCR-16-1203. 620

    43. Burga RA, Thorn M, Point GR, Guha P, Nguyen CT, Licata LA, et al. Liver myeloid-621

    derived suppressor cells expand in response to liver metastases in mice and inhibit 622

    the anti-tumor efficacy of anti-CEA CAR-T. Cancer Immunol Immunother 623

    2015;64(7):817-29 doi 10.1007/s00262-015-1692-6. 624

    44. Li K, Lan Y, Wang J, Liu L. Chimeric antigen receptor-engineered T cells for liver 625

    cancers, progress and obstacles. Tumour Biol 2017;39(3):1010428317692229 doi 626

    10.1177/1010428317692229. 627

    45. Zhang T, Sentman CL. Mouse tumor vasculature expresses NKG2D ligands and can 628

    be targeted by chimeric NKG2D-modified T cells. J Immunol 2013;190(5):2455-63 doi 629

    10.4049/jimmunol.1201314. 630

    on March 29, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.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 4, 2019; DOI: 10.1158/2326-6066.CIR-19-0026

    http://cancerimmunolres.aacrjournals.org/

  • 24

    46. Barber A, Rynda A, Sentman CL. Chimeric NKG2D Expressing T Cells Eliminate 631

    Immunosuppression and Activate Immunity within the Ovarian Tumor 632

    Microenvironment. Journal of Immunology 2009;183(11):6939-47 doi 633

    10.4049/jimmunol.0902000. 634

    47. Spear P, Barber A, Rynda-Apple A, Sentman CL. Chimeric Antigen Receptor T Cells 635

    Shape Myeloid Cell Function within the Tumor Microenvironment through IFN-gamma 636

    and GM-CSF. Journal of Immunology 2012;188(12):6389-98 doi 637

    10.4049/jimmunol.1103019. 638

    48. Soriani A, Fionda C, Ricci B, Iannitto ML, Cippitelli M, Santoni A. Chemotherapy-639

    elicited upregulation of NKG2D and DNAM-1 ligands as a therapeutic target in 640

    multiple myeloma. Oncoimmunology 2013;2(12) doi ARTN e26663 641

    10.4161/onci.26663. 642

    49. Amadori A, Zamarchi R, Desilvestro G, Forza G, Cavatton G, Danieli GA, et al. 643

    Genetic-Control of the Cd4/Cd8 T-Cell Ratio in Humans. Nat Med 1995;1(12):1279-644

    83 doi DOI 10.1038/nm1295-1279. 645

    50. Morgan RA, Yang JC, Kitano M, Dudley ME, Laurencot CM, Rosenberg SA. Case 646

    report of a serious adverse event following the administration of T cells transduced 647

    with a chimeric antigen receptor recognizing ERBB2. Mol Ther 2010;18(4):843-51 doi 648

    10.1038/mt.2010.24. 649

    51. Parkhurst MR, Yang JC, Langan RC, Dudley ME, Nathan DAN, Feldman SA, et al. T 650

    Cells Targeting Carcinoembryonic Antigen Can Mediate Regression of Metastatic 651

    Colorectal Cancer but Induce Severe Transient Colitis. Molecular Therapy 652

    2011;19(3):620-6 doi 10.1038/mt.2010.272. 653

    52. Groh V, Bahram S, Bauer S, Herman A, Beauchamp M, Spies T. Cell stress-654

    regulated human major histocompatibility complex class I gene expressed in 655

    gastrointestinal epithelium. Proc Natl Acad Sci U S A 1996;93(22):12445-50. 656

    53. Groh V, Rhinehart R, Secrist H, Bauer S, Grabstein KH, Spies T. Broad tumor-657

    associated expression and recognition by tumor-derived gamma delta T cells of 658

    MICA and MICB. Proc Natl Acad Sci U S A 1999;96(12):6879-84. 659

    54. Hue S, Mention JJ, Monteiro RC, Zhang S, Cellier C, Schmitz J, et al. A direct role for 660

    NKG2D/MICA interaction in villous atrophy during celiac disease. Immunity 661

    2004;21(3):367-77 doi 10.1016/j.immuni.2004.06.018. 662

    55. Nikiforow S, Werner L, Murad J, Jacobs M, Johnston L, Patches S, et al. Safety Data 663

    from a First-in-Human Phase 1 Trial of NKG2D Chimeric Antigen Receptor-T Cells in 664

    AML/MDS and Multiple Myeloma. Blood 2016;128(22). 665

    666

    Figures 667

    668

    on March 29, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.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 4, 2019; DOI: 10.1158/2326-6066.CIR-19-0026

    http://cancerimmunolres.aacrjournals.org/

  • 25

    Figure 1. NKG2D ligands were overexpressed in liver cancer. The 32 liver cancer 669

    specimens in the OD-CT-DgLive02-002 microarray were stained with MICA 670

    antibody (A) and ULBP2 antibody (B), respectively. The samples with overexpression 671

    of MICA or ULBP2 are marked with a triangle, scale bar = 500 μm. (C) NKG2D 672

    ligand expression in Hep3B, SMMC-7721, and MHCC97H cell lines was detected by 673

    flow cytometry. Data are representative or two independent experiments. Percentage 674

    of positive cells is detailed in the histograms. 675

    676

    Figure 2. NKG2D-BBz CAR-T cells efficiently lysed HCC cells. (A) Schematic 677

    representation of CD19-BBz CAR and NKG2D-BBz CAR. (B) CD19 and NKG2D 678

    CAR expression on human T cells transduced with a lentivirus and analyzed using 679

    flow cytometry. Data are representative of three independent experiments. Percentage 680

    of positive cells is detailed in the histograms. (C). Primary human T cells transduced 681

    with the indicated lentivirus were co-incubated with the three HCC cell lines at 682

    varying effector: target (E: T) ratios for 16 h, respectively. Cell lysis was determined 683

    by a standard nonradioactive cytotoxic assay. Two independent experiments were 684

    performed. Data are presented as the mean ± SD of triplicates, statistical significance 685

    between the NKG2D-BBz group and the other two groups was calculated using one-686

    way ANOVA with Sidak correction, * P < 0.05, *** P < 0.001. (D) Cytokines released 687

    in the co-culture supernatant by NTD, CD19-BBz, and NKG2D-BBz CAR-T cells 688

    when co-cultured with SMMC-7721 cells at E:T ratios of 0:1 and 5:1; TNF-α, IL-10, 689

    IL-2, and IFN-γ were detected in the supernatant collected 16 h after the culture. Two 690

    independent experiments were performed. Data are presented as the mean ± SD of 691

    triplicates and analyzed by two-tailed unpaired Student t test with Welch correction, 692

    ***P < 0.001. 693

    on March 29, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.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 4, 2019; DOI: 10.1158/2326-6066.CIR-19-0026

    http://cancerimmunolres.aacrjournals.org/

  • 26

    694

    Figure 3. Cytotoxic effect of NKG2D-BBz CAR-T was dependent on expression 695

    of NKG2D ligands. (A) MICA and ULBP2 expression in indicated cells was assessed 696

    by flow cytometry. Data are representative of two independent experiments. 697

    Percentage of positive cells is detailed in the histograms. (B) Cytotoxicity of NKG2D-698

    BBz CAR-T cells on SMMC7721-shCOO2, SMMC7721-shMICA, SMMC7721-699

    shULBP2 and SMMC7721-shMICA-shULBP2 cells. Two independent experiments 700

    were performed. Data are presented as the mean ± SD of triplicates, statistical 701

    significance between the SMMC7721-shMICA-shULBP2 group and the other three 702

    groups was calculated using one-way ANOVA with Sidak correction, * P < 0.05, ** P 703

    < 0.01. (C) Flow cytometry analysis of MICA and ULBP2 expression in Hep3B-704

    MICA or Hep3B-ULBP2 cells. Data are representative of three independent 705

    experiments. Percentage of positive cells is detailed in the histograms. (D) Cytotoxic 706

    effect of NKG2D-BBz CAR-T on Hep3B-MICA and Hep3B-ULBP2. Two 707

    independent experiments were performed. Data are presented as the mean ± SD of 708

    triplicates, statistical significance between the Hep3B-vector and the other two groups 709

    was analyzed by one-way ANOVA with Sidak correction, ***P < 0.001. 710

    711

    Figure 4. NKG2D-BBz CAR-T potently suppressed tumorigenesis in an SMMC-712

    7721 xenograft model. (A) B-NDG mice bearing SMMC-7721-luciferase xenografts 713

    were i treated with 100 l of saline (N=5), 1 × 107 non-transduced T cells (N=5), 1 × 714

    107 CD19-BBz CAR-T cells (N=6), and 1 × 10

    7 NKG2D-BBz CAR-T cells (N=6) by 715

    tail vein injection. All the mice were imaged with an IVIS imager at the indicated 716

    times. Tumor growth was assessed by total bioluminescence signals. Data are 717

    representative of two independent experiments. (B) Growth curve of SMMC-7721-718

    on March 29, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.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 4, 2019; DOI: 10.1158/2326-6066.CIR-19-0026

    http://cancerimmunolres.aacrjournals.org/

  • 27

    luciferase xenografts treated as described above. (C) NKG2D-BBz CAR-T cells 719

    accumulated in SMMC-7721-luciferase xenograft tumors. Tumors and normal tissues 720

    were collected from mice bearing SMMC-7721-luciferase subcutaneous xenografts 721

    treated with CD19-BBz CAR-T cells and NKG2D-BBz CAR-T cells. Formalin-fixed, 722

    paraffin-embedded tumor sections were consecutively cut and stained for human CD3 723

    expression (brown), data are representative of two independent experiments, scale bar 724

    = 100 μm. 725

    726

    Figure 5. Patient derived NKG2D-BBz CAR-T potently suppress liver cancer 727

    both in vitro and in vivo. (A) Cytotoxic effect of patient derived NKG2D-BBz CAR-728

    T cells on SMMC-7721 cells. Two independent experiments were performed. Data 729

    are presented as the mean ± SD of triplicates, statistical significance between the 730

    NKG2D-BBz group and the corresponding CD19-BBz group was calculated using 731

    two-tailed unpaired Student t test with Welch correction, ***P < 0.001. (B) B-NDG 732

    mice bearing SMMC-7721-luciferase xenografts were treated with 100 l of saline or 733

    1 × 107 patient derived T cells by tail vein injection. NKG2D-BBz CAR-T cells derived 734

    healthy donor were used as the positive control. Mice were imaged at the indicated 735

    times. Tumor growth was assessed by total bioluminescence signals. This experiment 736

    was done once. (C) Growth curve of SMMC-7721-luciferase xenografts treated with 737

    indicated T cells or saline (n = 4 per group). 738

    739

    Figure 6. Safety assessment of NKG2D-BBz CAR-T. (A) The 90 normal human 740

    tissues in the HOrgN09PT02 microarray were stained with MICA antibody. The 741

    samples containing more than 10% MICA positive cells are marked in red. A1–A4 : 742

    Thyroid; A5–A7: tongue; A8–A11: esophageal epithelium; A12–B5: gastric mucosa; 743

    on March 29, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.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 4, 2019; DOI: 10.1158/2326-6066.CIR-19-0026

    http://cancerimmunolres.aacrjournals.org/

  • 28

    B6–B7: duodenal mucosa; B8–C1: jejunal mucous membrane; C2–C4: ileal mucous 744

    membrane; C5–C9: appendix; C10–D1: mucous membrane of the colon; D2–D3: 745

    mucous membrane of the rectum; D4–D5: liver; D6–D7: pancreas; D8–D10: trachea; 746

    D11–E3: lung; E4–E6: myocardium; E7–E9: artery; E10–F4: skeletal muscle; F5–F7: 747

    skin; F8: seminal vesicle; F9–F11: prostate; F12–G6: testis; G7–G10: bladder; G11: 748

    medulla oblongata; G12–H1: telencephalon; H2–H3: epencephala; H4: brainstem; 749

    H5–H6: spleen, scale bar = 500 μm. (B) Karyotype of non-transduced T cells and 750

    NKG2D-BBz CAR-T cells, data are representative of two independent experiments. 751

    752

    on March 29, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.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 4, 2019; DOI: 10.1158/2326-6066.CIR-19-0026

    http://cancerimmunolres.aacrjournals.org/

  • on March 29, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.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 4, 2019; DOI: 10.1158/2326-6066.CIR-19-0026

    http://cancerimmunolres.aacrjournals.org/

  • on March 29, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.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 4, 2019; DOI: 10.1158/2326-6066.CIR-19-0026

    http://cancerimmunolres.aacrjournals.org/

  • on March 29, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.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 4, 2019; DOI: 10.1158/2326-6066.CIR-19-0026

    http://cancerimmunolres.aacrjournals.org/

  • on March 29, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.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 4, 2019; DOI: 10.1158/2326-6066.CIR-19-0026

    http://cancerimmunolres.aacrjournals.org/

  • on March 29, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.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 4, 2019; DOI: 10.1158/2326-6066.CIR-19-0026

    http://cancerimmunolres.aacrjournals.org/

  • on March 29, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.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 4, 2019; DOI: 10.1158/2326-6066.CIR-19-0026

    http://cancerimmunolres.aacrjournals.org/

  • Published OnlineFirst September 4, 2019.Cancer Immunol Res Bin Sun, Dong Yang, Hongjiu Dai, et al. CAR T-cellsEradication of hepatocellular carcinoma by NKG2D-specific

    Updated version

    10.1158/2326-6066.CIR-19-0026doi:

    Access the most recent version of this article at:

    Material

    Supplementary

    C1

    http://cancerimmunolres.aacrjournals.org/content/suppl/2019/09/04/2326-6066.CIR-19-0026.DAccess the most recent supplemental material at:

    Manuscript

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

    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://cancerimmunolres.aacrjournals.org/content/early/2019/09/04/2326-6066.CIR-19-0026To request permission to re-use all or part of this article, use this link

    on March 29, 2021. © 2019 American Association for Cancer Research. cancerimmunolres.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 4, 2019; DOI: 10.1158/2326-6066.CIR-19-0026

    http://cancerimmunolres.aacrjournals.org/lookup/doi/10.1158/2326-6066.CIR-19-0026http://cancerimmunolres.aacrjournals.org/content/suppl/2019/09/04/2326-6066.CIR-19-0026.DC1http://cancerimmunolres.aacrjournals.org/content/suppl/2019/09/04/2326-6066.CIR-19-0026.DC1http://cancerimmunolres.aacrjournals.org/cgi/alertsmailto:[email protected]://cancerimmunolres.aacrjournals.org/content/early/2019/09/04/2326-6066.CIR-19-0026http://cancerimmunolres.aacrjournals.org/

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