Download as pdf or txt
Download as pdf or txt
You are on page 1of 23

Accepted Manuscript

CRISPR knock out CTLA-4 enhances the anti-tumor activity of


cytotoxic T lymphocytes

Long Shi, Tongyu Meng, Zhilong Zhao, Jinsheng Han, Wei


Zhang, Fei Gao, Jianhui Cai

PII: S0378-1119(17)30719-9
DOI: doi: 10.1016/j.gene.2017.09.010
Reference: GENE 42159
To appear in: Gene
Received date: 17 May 2017
Revised date: 12 August 2017
Accepted date: 5 September 2017

Please cite this article as: Long Shi, Tongyu Meng, Zhilong Zhao, Jinsheng Han, Wei
Zhang, Fei Gao, Jianhui Cai , CRISPR knock out CTLA-4 enhances the anti-tumor activity
of cytotoxic T lymphocytes, Gene (2017), doi: 10.1016/j.gene.2017.09.010

This is a PDF file of an unedited manuscript that has been accepted for publication. As
a service to our customers we are providing this early version of the manuscript. The
manuscript will undergo copyediting, typesetting, and review of the resulting proof before
it is published in its final form. Please note that during the production process errors may
be discovered which could affect the content, and all legal disclaimers that apply to the
journal pertain.
ACCEPTED MANUSCRIPT

CRISPR knock out CTLA-4 enhances the anti-tumor activity of cytotoxic T lymphocytes

Long Shi1, Tongyu Meng4, Zhilong Zhao5, Jinsheng Han6, Wei Zhang7, Fei Gao3, Jianhui Cai 2,3

1. Department of Oncology, The Second Hospital of Hebei Medical University, Shijiazhuang, Hebei,

China 050000

T
IP
2. Department of Surgery, Hebei Medical University, Shijiazhuang, Hebei, China 050000

CR
3. Department of Surgery&Oncology, Hebei General Hospital, Shijiazhuang, Hebei, China 050000

US
4.Department of Gynecologic Oncology, The first hospital of SJZ, Shijiazhuang, Hebei, China 050000

5. Department of Surgery, the third affiliated hospital of Jinzhou medical university, Jinzhou, Liaoning,
AN
China 121000
M

6. Department of Surgery, Cangzhou hospital of Traditional Chinese Medicine and Western Medicine
ED

integrated Hebei, Cangzhou, Hebei, China 601000

7. Department of Surgery, HanDan Central Hospital, Handan, Hebei, China 056001


PT

Correspondence to Jianhui Cai; Tel: 86-311-85988977; Fax: 86-311-85988902.


CE

Email: jianhuicai2002@163.com
AC

Abstract

T cell-mediated anti-tumor immunity plays a pivotal role in cancer immune surveillance. Cytotoxic T-

lymphocyte-associated protein 4 (CTLA-4) is a protein receptor mainly expressed in activated T cells and

regulatory T cells. CTLA-4 competes with CD28 for ligand binding and generates inhibitory signals to

attenuate T cell activation. The blockade of CTLA-4 mediated immune inhibitory checkpoint has been

associated with enhanced anti-tumor immunity. In this study, we use CRISPR-Cas9 system to knock out
ACCEPTED MANUSCRIPT

(KO) CTLA-4 from cytotoxic T lymphocytes (CTLs) and evaluate its effect on the anti-tumor activity of

the CTLs. CTLA-4 KO CTLs robustly enhanced tumor cell death by 40% compared to the control and

facilitated apoptosis and caspase activities in tumor cells. The knockout of CTLA-4 also increased TNF-α

and IFN-γ secretion of the CTLs by approximately 2-fold. The effectiveness of CTLA-4 KO in enhancing

anti-tumor activity of the CTLs was verified in vivo using mouse xenograft model. The xenografted mice

T
treated with CTLA-4 KO CTLs demonstrated repressed tumor growth and prolonged survival compared

IP
to the control group. Our data suggest that CRISPR targeting CTLA-4 immune checkpoint could

CR
significantly improve the anti-tumor activity of CTLs.

CRISPR-Cas9 - Clustered regularly interspaced short palindromic repeats and CRISPR-associated protein
9

US
CTLA-4 - Cytotoxic T-lymphocyte-associated protein 4
KO – Knock out
CTL - cytotoxic T lymphocytes
AN
TNF-α - Tumor necrosis factor-α
IFN-γ - interferon gamma
DC - dendritic cells
CIK - Cytokine-induced killer cells
M

APC - antigen-presenting cell


CAR - Chimeric antigen receptor
TRAC - T-cell receptor α constant locus
ED

PBMC - peripheral blood mononuclear cells


GM-CSF - Granulocyte-macrophage colony-stimulating factor
IL-4 - interleukin 4
PT

IL-1β - Interleukin 1 beta


SDS-PAGE - sodium dodecyl sulfate polyacrylamide gel electrophoresis
ELISA - enzyme-linked immunosorbent assay
CE

SCID - Severe Combined Immunodeficiency


PARP - Poly (ADP-ribose) polymerase
AC
ACCEPTED MANUSCRIPT

Keywords: CRISPR, CTLA-4, cytotoxic T lymphocytes, T cell-mediated antitumor immunity,


cancer immunotherapy

T
IP
CR
US
AN
M
ED
PT
CE
AC
ACCEPTED MANUSCRIPT

1. Introduction

Human adaptive immunity is finely tuned by the balance between stimulatory and inhibitory signals[1].

Two stimulatory signals are required for naive T cells activation [2-5]. The interaction of T-cell receptor

(TCR) with the antigenic peptide presented by the antigen-presenting cell (APC) provides the antigen-

specific signal[6]. The interaction of T cell surface molecule CD28 with the B7 molecules (CD80 and

T
CD86) on APC generates co-stimulatory signal[7-16]. Activation of the TCR in the presence of co-

IP
stimulatory signals leads to T cell clonal expansion and initiation of effector functions. Cytotoxic T-

CR
lymphocyte-associated protein 4 (CTLA-4), also known as cluster of differentiation 152 (CD152), is a

protein receptor primarily expressed in activated T cells and regulatory T cells [17]. CTLA-4 is

US
homologous to the T cell co-stimulatory protein CD28 and competes with CD28 in binding the B7

ligands[18]. Unlike CD28 that delivers a co-stimulatory signal for T cells activation, CTLA-4 transmits
AN
an inhibitory signal to T cells through interacting with B7 ligands[17, 19]. CTLA-4 down-regulates
M

immune response through inhibiting IL-2 production and cell cycle progression[19, 20].
ED

The co-stimulatory and co-inhibitory signals mediated by CD28 and CTLA-4 deliberately modulate the T

cell-mediated immunity. The blockade of CTLA-4 with monoclonal antibodies led to enhanced the anti-
PT

tumor immune response in a range of preclinical investigations[21-31]. These preclinical findings suggest

the activation of cytotoxic T-lymphocytes (CTLs) trough targeting CTLA-4 is a feasible strategy in
CE

cancer immunotherapy.
AC

The emerging CRISPR gene editing technology has taken cancer immunotherapy to the next level.

CRISPR has been recently used to precisely deliver Chimeric antigen receptor (CAR) to the T-cell

receptor α constant (TRAC) locus to improve the endurance and potency of CAR T cells. In this study,

we edited CTLA-4 out of CTLs with the CRISPR-Cas9 system to investigate its effect on T cell

activation and cytotoxicity. This study will provide the foundation for the future application of CRISPR

technology in CTLA-4 targeting.


ACCEPTED MANUSCRIPT

2. Materials and methods

2.1 Cell culture and reagents

Peripheral blood from healthy donors was obtained were with the written consent and approved from

Hebei Blood Center (Hebei, China). Human PBMCs (peripheral blood mononuclear cells) isolated from

peripheral blood were cultured in RPMI-1640 complete medium (Gibco BRL, USA) supplemented with

T
10% fetal bovine serum (FBS, Gibco), 100 U/ml penicillin, and 100 mg/ml streptomycin (Life

IP
Technologies, Rockville, MD, USA). Human colonic adenocarcinoma cell line HCT116 cell line [32] was

CR
purchased from American Type Culture Collection (ATCC; Manassas, VA, USA) and cultured in RPMI-

1640 medium containing 10% FBS and antibiotics in a humidified 37 °C incubator with 5% CO2.

US
2.2 DCs induction and maturation
AN
PBMCs were isolated from whole blood samples by Ficoll-Paque density gradient centrifugation (GE

Healthcare, Chicago, IL, USA). Mononuclear cells from the interphase were collected, washed with
M

complete medium three times, and then cultured in the complete medium at 37 oC with 5% CO2. After 2h
ED

incubation, unattached cells and medium were removed. Attached cells were washed and then cultured in

RPMI complete medium supplemented with 100ng/ml human recombinant GM-CSF (PeproTech, Rocky
PT

Hill, NJ, USA) and 100ng/ml human recombinant IL-4 (PeproTech) for 6 days. On day 6, irradiated

HCT116 cell lysates were added into cultured DCs. After 24h co-culture, 25 ng/ml IL-1β (PeproTech)
CE

and 100 ng/ml TNF-α (Millipore Sigma, St. Louis, MO, USA) were added into the culture. Mature DCs
AC

were tested and collected on day 8.

2.3 Generation of Cytokine-induced killer cells (CIK) and CTLs

After 2h incubation in the complete medium at 37 °C with 5% CO2, unattached PBMCs were used to

generate CIKs according to the protocol by Schmidt-Wolf et al [33]. Briefly, unattached cells were

cultured in RPMI-1640 complete medium supplemented with 1000U/ml IFN-γ (Millipore Sigma),

1000U/ml IL-2 (Millipore Sigma), 5µg/ml CD3 monoclonal antibody (Millipore Sigma). The medium
ACCEPTED MANUSCRIPT

was half changed every 2-3 days with new medium contain 1000U/ml IL-2. CIK cells were mature and

ready to use after two weeks. CIKs were co-cultured with the mature DCs at the ratio of DC: T = 1:20 for

4 days to induce antigen-specific CTLs. CTLs were expanded in RPMI-1640 complete medium

supplemented with 50U/ml recombinant human IL-2.

2.4 Detection of immunophenotyping by flow cytometry

T
The FC500 flow cytometer (Beckman Coulter, USA) was used to characterize the immune phenotypes of

IP
DCs, CIKs, and CTLs. DCs were evaluated by CD80/CD86/CD83/CD11c; CIKs were evaluated by

CR
CD3+/CD56+; CTLs were evaluated by CD3+/CD8+. All the antibodies were purchased from BioLegend

(San Diego, CA, USA). The flow cytometry data were analyzed with FlowJo v.10.2 (TreeStar, Ashland,

US
OR, USA).
AN
2.5 CRISPR/Cas9 Lentivirus production

The non-targeting single guide RNA control and three single guide RNA sequences targeting human
M

CTLA-4 gene were designed. Control: ACGGAGGCTAAGCGTCGCAA; CTLA-4 sg1:


ED

CCTTGGATTTCAGCGGCACA; CTLA-4 sg2: GCACAAGGCTCAGCTGAACC; and CTLA-4 sg3:

CTCAGCTGAACCTGGCTACC. Single guide RNAs were cloned into a LentiCrispr v2 vector at BsmBI
PT

site following the protocol described previously[34]. Human embryonic kidney 293T cells were co-

transfected with single guide RNA containing LentiCrispr V2, packaging vector ps-PAX2 vector, and
CE

envelop vector pCL-VSVG. The culture medium was removed and changed to RPMI-1640 complete
AC

medium 24h later. The supernatant containing lentivirus was harvested 48h later and filtered through

0.45µm syringe filter.

2.6 Generation of CTLA-4 knockout cell lines

CTLs were transduced with lentivirus expressing control or CTLA-4 sgRNAs. After 48h incubation,

growth medium was changed to selection medium containing 1µg/ml puromycin. Once growing normally
ACCEPTED MANUSCRIPT

in selection medium, CTLs were then expanded in RPMI-1640 complete medium supplemented with

50U/ml recombinant human IL-2. The knockdown of CTLA-4 was confirmed using western blot analysis.

2.7 Western blot analysis

Cells were pelleted and lysed in lysis buffer (150 mM NaCl, 1% Triton X-100, 0.5% Na deoxycholate,

0.1% SDS, 50mM Tris-HCL, pH 7.4 with protease and phosphor-protease inhibitors). After incubation

T
for 15min on ice, cell lysates were centrifuged at 15,000 rpm for 15 min and then the supernatant was

IP
collected. Protein concentration was determined by Bradford assay (Bio-Rad Laboratories, Hercules, CA,

CR
USA). Equal amounts of total cell lysates were resolved by SDS-PAGE, transferred to a PVDF membrane

and then immunoblotted with antibodies against CTLA-4 or GAPDH followed by horseradish

US
peroxidase-conjugated secondary antibodies. Blots were then visualized by enhanced chemiluminescence

(Bio-Rad Laboratories).
AN
2.8 Cell viability assay
M

HCT116 cell viability was measured using WST-1 assay. Briefly, HCT116 cells were seeded in triplicate
ED

wells in 96-well-plates at the density of 5 x 103/well. After attachment, the control or CTLA-4 KO CTLs

were added to the plates at the density of 5 x 104/well. CTLs were co-cultured with HCT116 cells for
PT

another 18h at 37°C. HCT116 cell viability was assessed by WST-1 assay as described previously [35].

Briefly, 10 µl of WST-1 reagent was added into each well of the cell culture and incubated at 37 °C for 1
CE

h. Absorbance at wavelength 450 nm was read by standard microplate reader, and data were recorded and
AC

normalized by T cell culture alone.

2.9 Caspase enzymatic activity assay

Caspase assay buffer and caspase substrates were purchased from Enzo Life Science, Inc. (Farmingdale,

NY, USA). Caspase-3 substrate (Ac-DMQD-AMC), caspase-8 substrate(Ac-LETD-AFC) and caspase-9

substrate (Ac-LEHD-AFC) were used to test the activity of caspase-3, caspase-8 and caspase-9

respectively following the manufacture’s instruction.

2.10 cytokine induction assay


ACCEPTED MANUSCRIPT

1.5 x 104 HCT116 cells (target cells) were seeded in U-bottom 96-well-plate overnight at 37°C. On the

second day, 1.5 x 105 control CTLs or CTLA-4 KO CTLs (effector cells) were added to the culture. After

another 24 h, cell-free supernatants was collected by centrifuging at 1,000 g for 10 min and assayed for

cytokine release. Human IFN-gamma ELISA kit (Thermo Fisher Scientific Inc., Waltham, MA, USA)

and TNF-alpha ELISA Kit (Abcam Inc., Cambridge, MA, USA) were used to test IFN gamma, TNF

alpha, and IL-2 release following manufacture’s protocol.

T
IP
2.11 Xenograft mice model

CR
All the animal studies were approved by Hebei Medical University Animal Care and Use Committee.

HCT116 cells were washed and suspended in phosphate-buffered saline at the concentration of 5 × 107/ml.

US
100 µl of HCT116 cells were subcutaneously injected into the right flank of CB-17 SCID mice (6 weeks

old) that received irradiation (200 rad) on day 0. Mice were randomly separated into two groups (10 mice
AN
each group). On day 7 and day 14, 1 x 107 control CTLs or CTLA-4 KO CTLs were injected
M

intravenously into the mice. Tumors were measured twice a week by a digital caliper. Tumor volume was

calculated using the formula v = ab2 x 0.5, where a is the length and b is the width of the tumor. Mice
ED

were sacrificed when mice died, or tumor size reached 1000 mm3. Overall survival was examined by
PT

Kaplan-Meier method.

2.12 Statistical analysis


CE

All experiments were independently repeated three times. The results were presented as the mean 
AC

standard deviation. Two-tailed Student’s t test was used to determine statistical significance. The

GraphPad prism 5 software was used to evaluate the significance of the difference between groups. P

value less than 0.05 was considered as statistically significant.

3. Result

3.1 Immunophenotypes of DCs, CIKs and CTLs


ACCEPTED MANUSCRIPT

Flow cytometry was used to monitor the immunophenotypes of the ex vivo cultivated cells. The high-

level expression of CD11c (97.53 ± 1.08%; Figure 1A), co-stimulatory molecules CD80 (83.12 ± 1.15%;

Figure 1B), CD86 (93.49 ± 3.52%; Figure 1C) and the maturation marker CD83(83.24 ± 0.74%, Figure

1D) at day 8 demonstrated the maturation of DCs. The proportion of CD3+/CD56+ CIK cells reached

21.73 ± 2.04% (Figure 1E) on Day 10. The proportion of CD3+/CD8+ CTLs reached 87.86 ± 1.73%

T
(Figure 1F) at day 13, indicating the successful activation and expansion of CTLs by mature DCs.

IP
3.2 CRISPR knockout of CTLA-4 increased the anti-tumor activity of the CTLs

CR
The CTLs were transduced with lentivirus expressing guide RNA against CTLA-4 sequence to generate

CTLA-4 KO CTLs. The lentivirus expressing non-targeting guide RNA was used to generate parallel

US
control CTLs with intact CTLA-4. The efficiency of CTLA-4 knockout in the CTLs was tested 10 days

after lentivirus transduction. An equal amount of cell lysates was subjected to SDS-PAGE and then
AN
transferred to a PVDF membrane for western blot analysis. Compared to the control, the CTLA-4
M

expression was significantly reduced in the CTLs transduced with lentivirus expressing CTLA-4 guide

RNA, indicating the successful knockout of CTLA-4 from most of the CTLs by the CRISPR-Cas9 system
ED

(Figure 2A).
PT

We next examined if the knockout of CTLA-4 could improve the anti-tumor activity of the CTLs. We

first assayed the cytotoxicity of CTLA-4 KO CTLs on HCT116 colon cancer cell line. After being co-
CE

cultured with CTLs (control or CTLA-4 KO) for 24 h, HCT116 cells was assessed for cell viability using
AC

WST-1 assay. Results showed that the HCT116 cell viability in CTLA-4 KO co-culture decreased to

58.26% ± 4.55% of that in the control treatment (p < 0.005, Figure 2B), clearly demonstrating that

CTLA-4 KO enhanced the cytotoxicity of CTLs.

3.3 CTLA-4 KO CTLs enhance apoptosis and caspase activities in tumor cells

The apoptosis of HCT116 cells were analyzed by Annexin V/PI staining followed by flow cytometry

analysis after being co-cultured with CTLs for 12 h. 40.3% of HCT116 cells entered late apoptotic stage
ACCEPTED MANUSCRIPT

in the CTLA-4 KO co-culture, whereas only 20.3% of HCT116 cells entered this stage in the control co-

culture (Fig 3A). Accordingly, the percentage of early apoptotic HCT116 cells in the CTLA-4 KO and

control co-culture are 22.7% and 18.3% respectively. These data clearly show that CTLA-4 KO CTLs

induced a higher level of apoptosis in the co-cultured tumor cells than did the control CTLs.

Caspases play essential roles in programmed cell death. We found the HCT116 cells in the CTLA-4 KO

T
co-culture have more cleaved form of caspase-3, caspase-7, caspase-8, caspase-9 and PARP than the

IP
control (Fig 3B), showing that CTLA-4 KO CTLs lead to an elevated caspase activation in tumor cells.

CR
We then incubated HCT116 cell lysates with different caspase substrates to test their caspase activities.

Results confirmed that the caspase-3, caspase-8, and caspase-9 activities of the HCT116 cells in CTLA-4

US
KO co-culture are respectively 10.35 ± 1.53, 9.18 ± 0.67 and 7.92 ± 0.25 times higher than that of the

control.
AN
3.4 CTLA-4 KO CTLs produce increased cytokines
M

CTLs release cytokines such as Tumor necrosis factor-α (TNF-α) and interferon-γ (IFN-γ), which is

associated with the activation of host immune response to kill viral infected cells and tumor cells. Since
ED

we found that CTLA-4 KO enhanced the anti-tumor activity of the CTLs, we then examined if CTLA-4
PT

KO could enhance the cytokines secretion of CTLs. ELISA assays showed that CTLA-4 KO CTLs

secreted 2.38 ± 0.45 and 2.17 ± 0.26 times more TNF-α and IFN-γ respectively than did the control CTLs
CE

(Figure 4A and 4B). This result clearly shows that CTLA-4 KO could increase the cytokines production

of CTLs.
AC

3.5 CTLA-4 KO CTLs decrease the growth of HCT116 cells xenografted in mice

The efficacy of CTLA-4 KO CTLs in repressing tumor growth was validated in vivo using human tumor

xenograft model. Two groups of CB-17 SCID mice were subcutaneously inoculated with 5 x 10 6

HCT116 cells following 200 rad irradiation. The mice were then treated with either control CTLs

(control group) or CTLA-4 KO CTLs for four different times to evaluate the repression of CTLs on tumor

growth. The tumor became detectable after 2 weeks and the tumor growth in the CTLA-4 KO CTLs
ACCEPTED MANUSCRIPT

treated mice was dramatically repressed compared to that in the control group (Figure 5A, p < 0.01). All

the mice in the control group died from progressive tumors by 62 days. In contrast, only 10% of CTLA-4

KO CTLs treated mice died at the same time (Figure 5B, p < 0.0001). Thus, CTLA-4 KO in CTLs

effectively inhibited human MM cell growth in vivo and significantly improved overall survival of the

xenografted mice.

T
4. Discussion

IP
T-cell mediated anti-tumor responses are the major component of cancer immune surveillance that

CR
controls tumor development and progression [36]. Tumor cells can escape immune surveillance in the

presence of tumor-associated antigen-specific T cells [37]. The mechanisms utilized by cancer cells to

US
repress T-cell function including the production of soluble immunosuppressive factors (e.g. TGF-β, IL-10)

and the recruitment of suppressor immune cells. The cellular molecules involved in the inhibition of T
AN
cell mediated anti-tumor response have been intensively investigated and a group of inhibitory molecules
M

have been identified. Among these molecules, PD-1 and CTLA-4 are two most studied targets for the

development of cancer therapy[17, 38, 39].


ED

CTLA-4 plays an essential role in modulating the T-cell mediated anti-tumor response [40]. The blockade
PT

of CTLA-4 mediated co-inhibitory signaling with antibody has been shown to enhance the immune

response against cancer cells [41]. The monoclonal CTLA-4 antibody ipilimumab has been approved for
CE

melanoma treatment and others are in development [42-44]. In addition to therapeutic antibody

development, the application of CRISPR gene-editing technology in T cell genome engineering also
AC

holds great promise in cancer immunotherapy. The advantage of CRISPR is the sequence specific

interventions in cells. Recent studies have demonstrated the potential of CRISPR technology in

improving the efficacy of CAR T cells.

In this study, we used CRISPR technology to disrupt the CTLA-4 mediated co-inhibitory signaling and

tested its effect on CTLs’ anti-tumor activity. Compared to the control, the CTLA-4 KO CTLs could

further reduce tumor cell viability by about 40%. The mechanism study showed the enhanced anti-tumor
ACCEPTED MANUSCRIPT

effect is associated with the increased apoptosis and augmented caspase activities in tumor cells. Since

secretion of cytokines (primarily TNF-α and IFN-γ) is one of the mechanisms for CTLs to kill tumor cells,

we next measured if knockout of CTLA-4 in CTLs increases TNF-α and IFN-γ secretion. The production

of TNF-α and IFN-γ in CTLA-4 KO CTLs increased about 2-fold compared to that in the control CTLs.

In vivo animal study shows that CTLA-4 KO CTLs are active in the xenograft mouse model. The CTLA-

T
4 KO CTLs treatment inhibited tumor growth more efficiently than did the control and prolonged the

IP
overall survival of the host.

CR
In conclusion, our data showed that CRISPR knockout CTLA-4 enhanced the T cell mediated anti-tumor

immunity, suggesting the potential application of CRISPR technology in immune checkpoints targeting.

Conflicts of interest
US
AN
The authors declare that there is no conflict of interest regarding the publication of this paper.
M

Reference
ED

1. Salama, A.K. and F.S. Hodi, Cytotoxic T-lymphocyte-associated antigen-4. Clin Cancer Res,

2011. 17(14): p. 4622-8.


PT

2. Bretscher, P. and M. Cohn, A theory of self-nonself discrimination. Science, 1970. 169(3950): p.


CE

1042-9.

3. Lafferty, K.J. and A.J. Cunningham, A new analysis of allogeneic interactions. Aust J Exp Biol
AC

Med Sci, 1975. 53(1): p. 27-42.

4. Schwartz, R.H., A cell culture model for T lymphocyte clonal anergy. Science, 1990. 248(4961):

p. 1349-56.

5. Bretscher, P., The two-signal model of lymphocyte activation twenty-one years later. Immunol

Today, 1992. 13(2): p. 74-6.

6. Kalergis, A.M., et al., Efficient T cell activation requires an optimal dwell-time of interaction

between the TCR and the pMHC complex. Nat Immunol, 2001. 2(3): p. 229-34.
ACCEPTED MANUSCRIPT

7. Lenschow, D.J., T.L. Walunas, and J.A. Bluestone, CD28/B7 system of T cell costimulation.

Annu Rev Immunol, 1996. 14: p. 233-58.

8. Hara, T., S.M. Fu, and J.A. Hansen, Human T cell activation. II. A new activation pathway used

by a major T cell population via a disulfide-bonded dimer of a 44 kilodalton polypeptide (9.3

antigen). J Exp Med, 1985. 161(6): p. 1513-24.

T
9. Martin, P.J., et al., A 44 kilodalton cell surface homodimer regulates interleukin 2 production by

IP
activated human T lymphocytes. J Immunol, 1986. 136(9): p. 3282-7.

CR
10. Damle, N.K., et al., Differential regulatory signals delivered by antibody binding to the CD28

(Tp44) molecule during the activation of human T lymphocytes. J Immunol, 1988. 140(6): p.

US
1753-61.

11. Lindstein, T., et al., Regulation of lymphokine messenger RNA stability by a surface-mediated T
AN
cell activation pathway. Science, 1989. 244(4902): p. 339-43.

12. Ledbetter, J.A., et al., CD28 ligation in T-cell activation: evidence for two signal transduction
M

pathways. Blood, 1990. 75(7): p. 1531-9.


ED

13. Van Lier, R.A., et al., T cell receptor/CD3 and CD28 use distinct intracellular signaling

pathways. Eur J Immunol, 1991. 21(7): p. 1775-8.


PT

14. June, C.H., et al., Role of the CD28 receptor in T-cell activation. Immunol Today, 1990. 11(6): p.
CE

211-6.

15. Jenkins, M.K., et al., CD28 delivers a costimulatory signal involved in antigen-specific IL-2
AC

production by human T cells. J Immunol, 1991. 147(8): p. 2461-6.

16. Linsley, P.S. and J.A. Ledbetter, The role of the CD28 receptor during T cell responses to

antigen. Annu Rev Immunol, 1993. 11: p. 191-212.

17. Brunet, J.F., et al., A new member of the immunoglobulin superfamily--CTLA-4. Nature, 1987.

328(6127): p. 267-70.

18. Collins, A.V., et al., The interaction properties of costimulatory molecules revisited. Immunity,

2002. 17(2): p. 201-10.


ACCEPTED MANUSCRIPT

19. Walunas, T.L., et al., CTLA-4 can function as a negative regulator of T cell activation. Immunity,

1994. 1(5): p. 405-13.

20. Krummel, M.F. and J.P. Allison, CD28 and CTLA-4 have opposing effects on the response of T

cells to stimulation. J Exp Med, 1995. 182(2): p. 459-65.

21. Yang, Y.F., et al., Enhanced induction of antitumor T-cell responses by cytotoxic T lymphocyte-

T
associated molecule-4 blockade: the effect is manifested only at the restricted tumor-bearing

IP
stages. Cancer Res, 1997. 57(18): p. 4036-41.

CR
22. Mangsbo, S.M., et al., Enhanced tumor eradication by combining CTLA-4 or PD-1 blockade with

CpG therapy. J Immunother, 2010. 33(3): p. 225-35.

US
23. Espenschied, J., et al., CTLA-4 blockade enhances the therapeutic effect of an attenuated poxvirus

vaccine targeting p53 in an established murine tumor model. J Immunol, 2003. 170(6): p. 3401-7.
AN
24. Dewan, M.Z., et al., Fractionated but not single-dose radiotherapy induces an immune-mediated

abscopal effect when combined with anti-CTLA-4 antibody. Clin Cancer Res, 2009. 15(17): p.
M

5379-88.
ED

25. Pilones, K.A., et al., Invariant natural killer T cells regulate breast cancer response to radiation

and CTLA-4 blockade. Clin Cancer Res, 2009. 15(2): p. 597-606.


PT

26. Demaria, S., et al., Immune-mediated inhibition of metastases after treatment with local radiation
CE

and CTLA-4 blockade in a mouse model of breast cancer. Clin Cancer Res, 2005. 11(2 Pt 1): p.

728-34.
AC

27. Saha, A. and S.K. Chatterjee, Combination of CTL-associated antigen-4 blockade and depletion

of CD25 regulatory T cells enhance tumour immunity of dendritic cell-based vaccine in a mouse

model of colon cancer. Scand J Immunol, 2010. 71(2): p. 70-82.

28. Lute, K.D., et al., Human CTLA4 knock-in mice unravel the quantitative link between tumor

immunity and autoimmunity induced by anti-CTLA-4 antibodies. Blood, 2005. 106(9): p. 3127-33.

29. Fevery, S., et al., CTLA-4 blockade in murine bone marrow chimeras induces a host-derived

antileukemic effect without graft-versus-host disease. Leukemia, 2007. 21(7): p. 1451-9.


ACCEPTED MANUSCRIPT

30. Kwon, E.D., et al., Elimination of residual metastatic prostate cancer after surgery and

adjunctive cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) blockade immunotherapy. Proc

Natl Acad Sci U S A, 1999. 96(26): p. 15074-9.

31. Waitz, R., et al., Potent induction of tumor immunity by combining tumor cryoablation with anti-

CTLA-4 therapy. Cancer Res, 2012. 72(2): p. 430-9.

T
32. Gayet, J., et al., Extensive characterization of genetic alterations in a series of human colorectal

IP
cancer cell lines. Oncogene, 2001. 20(36): p. 5025-32.

CR
33. Schmidt-Wolf, I.G., et al., Use of a SCID mouse/human lymphoma model to evaluate cytokine-

induced killer cells with potent antitumor cell activity. J Exp Med, 1991. 174(1): p. 139-49.

US
34. Sanjana, N.E., O. Shalem, and F. Zhang, Improved vectors and genome-wide libraries for

CRISPR screening. Nat Methods, 2014. 11(8): p. 783-4.


AN
35. Yin, L.M., et al., Simultaneous application of BrdU and WST-1 measurements for detection of the

proliferation and viability of airway smooth muscle cells. Biol Res, 2014. 47: p. 75.
M

36. Dunn, G.P., et al., Cancer immunoediting: from immunosurveillance to tumor escape. Nat
ED

Immunol, 2002. 3(11): p. 991-8.

37. Kim, R., M. Emi, and K. Tanabe, Cancer immunoediting from immune surveillance to immune
PT

escape. Immunology, 2007. 121(1): p. 1-14.


CE

38. Ishida, Y., et al., Induced expression of PD-1, a novel member of the immunoglobulin gene

superfamily, upon programmed cell death. EMBO J, 1992. 11(11): p. 3887-95.


AC

39. Freeman, G.J., et al., Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family

member leads to negative regulation of lymphocyte activation. J Exp Med, 2000. 192(7): p. 1027-

34.

40. Mocellin, S. and D. Nitti, CTLA-4 blockade and the renaissance of cancer immunotherapy.

Biochim Biophys Acta, 2013. 1836(2): p. 187-96.

41. Korman, A.J., K.S. Peggs, and J.P. Allison, Checkpoint blockade in cancer immunotherapy. Adv

Immunol, 2006. 90: p. 297-339.


ACCEPTED MANUSCRIPT

42. Grosso, J.F. and M.N. Jure-Kunkel, CTLA-4 blockade in tumor models: an overview of

preclinical and translational research. Cancer Immun, 2013. 13: p. 5.

43. Ribas, A., Overcoming immunologic tolerance to melanoma: targeting CTLA-4 with

tremelimumab (CP-675,206). Oncologist, 2008. 13 Suppl 4: p. 10-5.

44. Weber, J., Overcoming immunologic tolerance to melanoma: targeting CTLA-4 with ipilimumab

T
(MDX-010). Oncologist, 2008. 13 Suppl 4: p. 16-25.

IP
CR
US
AN
M
ED
PT
CE
AC
ACCEPTED MANUSCRIPT

T
IP
CR
US
Figure 1. Flow cytometry analysis of the immunophenotypes of the ex vivo cultivated cells. A-D, the DC
AN
cell marker CD80, CD83, CD11c and CD83 were analyzed. E, The CD3+ cells were further gated for
M

CD56 to show the percentage of CIK cells. F, The CTL cells were analyzed by CD3 and CD8 antibody.
ED
PT
CE
AC
ACCEPTED MANUSCRIPT

T
Figure 2. CRISPR knockout of CTLA-4 in CTLs. A. western blot analysis showing knockout of CTLA-4

IP
in CTLs. GAPDH is used as loading control. B. The cell viability of HCT116 cells co-cultured with the

CR
control or CTLA-4 KO CTLs were compared and normalized to the control. Data is presented as mean 

standard deviation, experiment was independently repeated for 3 times.

US
AN
M
ED
PT
CE
AC
ACCEPTED MANUSCRIPT

T
IP
Figure 3. CTLA-4 disruption in CTLs increased apoptosis and caspase activities in tumor cells. A.

CR
Annexin V/PI staining shows that HCT116 cells co-cultured with CTLA-4 KO CTLs have higher level of

early stage and late stage apoptotic cells. Annexin V+/PI- means early apoptotic, Annexin V+/PI+ means

US
late apoptotic. B. Compared to the control, HCT116 cells co-cultured with CTLA-4 KO CTLs
AN
demonstrated higher level of the cleaved form (CF) of PARP, caspase-3, caspase-7, caspase-8 and

caspase-9. C. Compared to the control, HCT116 cells co-cultured with CTLA-4 KO CTLs demonstrated
M

higher level of caspase enzymatic activity.


ED
PT
CE
AC
ACCEPTED MANUSCRIPT

T
Figure 4. CTLA04 KO increased the cytokine secretion of CTLs. A. The TNF-α secretion in the control

IP
and CTLA-4 KO CTLs were tested and normalized to control CTLs. B. The IFN-γ secretion in the control

CR
and CTLA-4 KO CTLs were tested and normalized to control CTLs.

US
AN
M
ED
PT
CE
AC
ACCEPTED MANUSCRIPT

T
Figure 5. CTLA-4 KO CTLs repress tumor growth more efficiently than the control CTLs in xenografted

IP
mice. A. The tumor growth in human HCT116 bearing mice treated with either control or CTLA-4 KO

CR
CTLs. Data were presented as mean  standard deviation. B. Overall survival was examined by Kaplan-

Meier method (n = 10, p < 0.0001).

US
AN
M
ED
PT
CE
AC
ACCEPTED MANUSCRIPT

Highlights
 CTLA-4 knockout increases the anti-tumor activity of the CTLs
 CTLA-4 knockout CTLs enhance apoptosis and caspase activities in tumor cells
 CTLA-4 knockout increases the secretion of cytokines in CTLs

T
IP
CR
US
AN
M
ED
PT
CE
AC

You might also like