You are on page 1of 10

Journal of Controlled Release 350 (2022) 298–307

Contents lists available at ScienceDirect

Journal of Controlled Release


journal homepage: www.elsevier.com/locate/jconrel

Lipid nanoparticles produce chimeric antigen receptor T cells with


interleukin-6 knockdown in vivo
Jing-e Zhou 1, Lei Sun 1, Yujie Jia, Zhehao Wang, Tengshuo Luo, Jingwen Tan, Xiaoyan Fang,
Hongjia Zhu, Jing Wang, Lei Yu *, Zhiqiang Yan *
Institute of Biomedical Engineering and Technology, Shanghai Engineering Research Center of Molecular Therapeutics and New Drug Development, School of Chemistry
and Molecular Engineering, East China Normal University, Shanghai 200062, PR China

A R T I C L E I N F O A B S T R A C T

Keywords: Chimeric receptor T cells (CAR-T) can effectively cure leukemia; however, there are two limitations: a compli­
Chimeric antigen receptor T cells cated preparation process ex vivo and cytokine release syndrome (CRS). In this study, we constructed a lipid
CD3 antibody nanoparticle system modified by CD3 antibody on the surface, loading with the plasmid containing the com­
Lipid nanoparticles
bination gene of interleukin 6 short hairpin RNA (IL-6 shRNA) and CD19-CAR (AntiCD3-LNP/CAR19 + shIL6).
IL-6 shRNA
The system targeted T cells by the mediation of CD3 antibody and stably transfected T cells to transform them
Cytokine release syndrome
into CAR-T cells with IL-6 knockdown, thus killing CD19-highly expressed leukemia tumor cells and reducing
CRS caused by IL-6. In vivo experiments showed that AntiCD3-LNP/CAR19 + shIL6 could stably transfect T cells
and produce CAR-T within 90 days to kill the tumor. This significantly prolonged the survival time of leukemia
model mice and demonstrated the prepared LNP exhibited the same anti-tumor effect as the traditional CAR-T
cells prepared ex vivo. In this study, CAR-T cells were directly produced in vivo after intravenous injection of
the lipid nanoparticles, without the need of using the current complex process ex vivo. Additionally, IL-6
expression was silenced, which would be helpful to reduce the CRS and improve the safety of CAR-T therapy.
This method improves the convenience of using CAR-T technology and is helpful in further promoting the clinical
application of CAR-T.

1. Introduction used in clinical practice [12,13]. New technology is required for CAR-T
cell preparation to simplify its preparation process, avoid strict quality
Chimeric antigen receptor T-cell (CAR-T) therapy has achieved control ex vivo, and reduce the technical barriers.
remarkable success in the treatment of hematological malignancies Another challenge for CAR-T therapy is cytokine release syndrome
[1–3], with several products currently approved by the FDA for acute B- (CRS) [9]. Studies have shown that about 60% to 80% of patients treated
cell leukemia (B-ALL) [4,5], large B-cell lymphoma and multiple with CAR-T may suffer from CRS [12]. In CRS, CAR-T cells are activated
myeloma [6,7]. However, the clinical application of CAR-T technology after combining with the antigen of tumor cells, constantly expanding
is still limited by many factors [8], such as the complex preparation and release copious amounts of cytokines, thus leading to a systemic
process and cytokine release syndrome (CRS) [9]. inflammatory response or even the death of the patient [14]. Tocilizu­
Currently, obtaining CAR-T cells requires a complex process and mab, a humanized IL-6 receptor monoclonal antibody, is often used
strict quality control [10]. A series of procedures, including blood clinically to block IL-6 receptors and the associated signal transduction
sample collection, separation, activation, transfection, amplification, [15–17], thus alleviating CRS [18]. However, Tocilizumab can often
detection, preparation, transportation, and drug administration, need cause neutropenia and severe bone marrow suppression, increasing the
up to about three weeks [10]. Additionally, the preparation of CAR-T risk of infection [19]. Thus, finding an effective way to control CRS for
cells for clinical applications requires customization and following CAR-T therapy is urgently required [20].
GMP for each patient, making mass production difficult [11]. As CAR-T The Nano-delivery system can be used to deliver DNA to cells and
therapy has high costs and technical barriers, it is difficult to be widely facilitate the direct production of CAR-T cells in vivo [21–23]. For

* Corresponding authors.
E-mail addresses: yulei@nbic.ecnu.edu.cn (L. Yu), zqyan@sat.ecnu.edu.cn (Z. Yan).
1
Jing-e Zhou and Lei Sun contributed equally to this work.

https://doi.org/10.1016/j.jconrel.2022.08.033
Received 6 April 2022; Received in revised form 8 August 2022; Accepted 16 August 2022
Available online 25 August 2022
0168-3659/© 2022 Elsevier B.V. All rights reserved.
J.-e. Zhou et al. Journal of Controlled Release 350 (2022) 298–307

example, in 2017, Nat Nano [24] reported a type of nanoparticle that The IL-6 shRNA sequence was inserted into the basic vector plasmid
encapsulated CAR DNA with cationic polymer and targeted T cells PUT523 by ApaLI, PvuII, and XhoI endonuclease. The transposons were
through the mediation of CD3 antibodies and transformed them directly also added to the plasmid genes, leading to a high transfection efficiency
into CAR-T cells. This study provided new insights into CAR-T treat­ and durable and stable gene expression. The primer was synthesized by
ment. The investigator then developed nanoparticle-mediated delivery Shanghai General Bioengineering Co. Ltd. After the interference frag­
of mRNA for CAR-T therapy that has entered the clinical phase ment was ligated to the expression vector, the target plasmid was
(NCT01837602 [25], NCT03060356 [26], NCT02277522 [27]). How­ transformed and identified by colony PCR, followed by DNA sequencing.
ever, the delivery of CAR DNA with lipid nanoparticles (LNPs) remains
unreported. We choose LNP as gene delivery vector because it has more 2.2. Cell lines
advantages than cationic polymers. Cationic polymers have high mo­
lecular weight (most of them are more than 10KDa), which leads to their Raji-Luc cells, the K562 cell line, and human T cells were acquired
poor biodegradability and easy accumulation in the body, thus causing from Shanghai Unicar-Therapy Biomed-Pharmaceutical Technology
damage to tissues. Co., Ltd. The K562 cells were transfected with lentivirus to obtain the
The LNP delivery systems had the following advantages: 1) LNP is K562 cells expressing CD19. The Raji-Luc cell line and K562 cells were
mainly composed of phospholipids, which has good drug-forming cultured in complete Roswell Park Memorial Institute (RPMI) 1640
property. It is easily metabolized and will not accumulate in the body medium with 10% heat-inactivated fetal bovine serum (FBS), 100 U/mL
for a long time to cause toxicity. 2) The ionizable lipid used in LNP have penicillin, and 100 μg/mL streptomycin.
pH sensitivity. Its positive charge at low pH enables LNPs to escape from
endosomes, and its neutral charge at around pH 7.0 keeps LNPs stable in
2.3. MTAS-NLS peptide-DSPE-PEG synthesis
the blood, avoids being quickly cleared in the blood. 3) LNP, as a gene
drug delivery system, has been marketed for several drugs, while
The sulfhydrylated microtubule-associated-nuclear localization
cationic polymeric system is still in the early stage of preclinical trials.
peptide Cys-MTAS-NLS (sequence.
In this study, we prepared lipid nanoparticles (LNPs) modified by
CGRYLTQETNKVETYKEQPLKTPGKKKKGKPGKRKEQEKKKRRTR)
CD3 antibody, which encapsulated plasmids containing human
was synthesized by Gil Biochemical Company. The addition of this
interleukin-6 (IL-6) short hairpin RNA (shRNA) and the CAR gene. After
peptide into the LNPs could help the plasmid gene to enter the nucleus
intravenous injection, the LNPs successfully transfected the T cells and
and increase transfection efficiency [28]. Next, 10 mg of the nuclear
produced IL-6-knockdown CAR-T cells in vivo. The CAR-T cells showed
localization peptide Cys-MTAS-NLS was dissolved in 0.5 mL of 1X PBS
antitumor effects comparable to that of conventional CAR-T cells and
solution (pH 7.0). Then, 15.8 mg of Mal-PEG6-DSPE was dissolved in 1
reduced the release of IL-6 and the incidence of CRS (Fig. 1). This
mL of dimethylformamide (DMF), mixed with the peptide solution, and
technology could avoid the complex preparation process ex vivo and
stirred for 4 h. After the reaction was completed, the excess polypeptide
strict quality control of the traditional CAR-T technology, reduce the
and DMF were removed by dialysis in ultra-pure water for two days
technical barrier and production costs, and provide a more convenient
(Mw = 3.5 kDa). After freeze-drying, the target compound (DSPE-PEG-
and practical protocol for CAR-T therapy.
MTAS-NLS) was obtained. The structure was characterized by IR spec­
trums and 1H NMR.
2. Methods

2.1. Plasmid construction 2.4. Synthesis and characterization of DSPE-PEG-CD3 antibody-targeted


lipid molecules
All the plasmids used in this study were custom-cloned by Shanghai
Unicar-Therapy Biomed-Pharmaceutical Technology Co., Ltd. The anti-human CD3 antibody (Bioxcell.com; cat. no. BE0001–1)
was dissolved in 1 mL of NaHCO3 solution (pH = 8), and 4 mol. of DSPE-

Fig. 1. The design of LNPs and the production of CAR-T cells in vivo. After intravenous injection, the LNPs transfected the T cells and produced IL-6-knockdown
CAR-T cells in vivo.

299
J.-e. Zhou et al. Journal of Controlled Release 350 (2022) 298–307

PEG2000-NHS was dissolved in dimethyl sulfoxide (DMSO). Then, lymphocytes for 48 h, and the number of cells expressing GFP was
DSPE-PEG2000-NHS was slowly added to the CD3 antibody solution and determined by flow cytometry to evaluate the transduction efficiency in
mixed for 4 h. After the reaction was completed, the solution was dia­ vitro.
lyzed in ultra-pure water (cutoff molecular weight: 3.5 kDa) and freeze-
dried. The structures were characterized by HPLC and 1H NMR.
2.8. Functions of AntiCD3-LNP/CAR19 + shIL6 in vitro
2.5. Preparation of AntiCD3-LNP/CAR19 + shIL6
2.8.1. Cytotoxicity assay
The thin-film hydration and extrusion method [29] was used to We selected LNPs transduced T cells as the effector cells and K562 or
prepare the LNPs with IL-6 knockdown (LNP/CAR19 + shIL6). The Raji cells as the target cells to evaluate the ability of killing tumor cells
membrane materials, including DlinMC3, DOPE [30], Chol, PEG-DMG The T cells were transfected with AntiCD3-LNP/CAR19 + shIL6, LNP/
[31], DOTAP, and DSPE-MTAS-NLS (molar ratio 1–20: 5–10: 1–10: CAR19 + shIL6, and AntiCD3-LNP/CAR19 (CD3 antibody modified
1–10: 1–5: 3), were weighed and dissolved in 2 mL of trichloromethane nanoparticles without knocking down IL-6) were incubated for 48 h with
as the lipid oil phase solution. Then, trichloromethane was gradually CD19 overexpressed K562 target cells and Raji target cells in a ratio of
removed using a rotary evaporator to obtain a film of uniform thickness. 10:1, 8:1, 5:1, and 1:1, respectively. The in vitro cytotoxicity of CAR-T
Sodium acetate solution (pH = 4; 25 mM) was prepared as an aqueous cells was verified by the lactate dehydrogenase (LDH) method.
solvent, and the plasmid was dissolved in the solution, with a 1:10 M
ratio of the plasmid to the lipid materials. The prepared aqueous solu­ 2.8.2. Cytokine secretion assay
tion was added to the lipid film and mixed for 1 h. Then, the LNP/ To test the expression level of IL-6 mRNA, the T cells were trans­
CAR19 + shIL6NP/IL-6 was extruded through the nuclear pore mem­ fected with AntiCD3-LNP/CAR19, LNP/CAR19 + shIL6, and AntiCD3-
branes of 400 and 200 nm using a micro-extruder. The LNP/CAR19 + LNP/CAR19 + shIL6, followed by incubation with the target cells for 48
shIL6NP/IL-6 suspension was collected with a 10 kDa ultrafiltration h; the co-incubated cells were collected to extract total RNA and detect
centrifuge tube and centrifuged at 6000 rpm for 30 min, and placed in IL-6 expression by qPCR.
phosphate-buffered saline (PBS). Finally, DSPE-PEG2000-CD3 (in a
molar ratio of 1:10 to the plasmid) was added to the above solution, and 2.9. QPCR
incubated for 4 h at 37 ◦ C to obtain the CD3 antibody-modified LNPs
with IL-6 knockdown (AntiCD3-LNP/CAR19 + shIL6), which was stored To test the expression level of IL-6 mRNA, T cells were transfected
at 4 ◦ C. with AntiCD3-LNP/CAR19, LNP/CAR19 + shIL6, and AntiCD3-LNP/
CAR19 + shIL6, followed by incubation with the target cells for 48 h.
2.6. Nanoparticle characterization Then the co-incubated cells were collected, centrifuged, and the super­
natant was removed. Total RNA was extracted using TRIZOL Reagent
The nanoparticle solution was diluted 50 times with PBS. The size, (Cat#15596–018, Life technologies, Carlsbad, CA, US) following the
polydispersity index (PDI), and zeta potential (ζ) of the LNPs were manufacturer’s instructions. Then A 5 × Primescipt RT Master mix was
determined by dynamic light scattering (DLS) (Malvern, UK). The used to reverse-transcribe the extracted RNA to obtain cDNA. Total IL-6
morphology of the LNPs was observed by transmission electron micro­ expression was detected by qPCR. The primers were provided by
scopy (TEM). The nanoparticle samples were negatively stained with Shanghai Uni-car Biological Co. Ltd. The primer sequence for IL-6
phosphotungstic acid and observed after the samples were dried. The amplification is:
prepared LNP solutions were evenly divided into two parts. DNase (5 Gene Name Sequences (5′ -3′ )
μg/ug DNA) was added into one part to react at 37 ◦ C for 30 min, and
EDTA was used to stop the reaction. After DNase treatment, the un­ Forward: TAACCACCCCTGACCCAACCA
IL-6
Reverse: GCGCAGAATGAGATGAGTTGTCA
coated plasmid was decomposed, while the LNP-coated plasmid was Forward: GGACAGGACCATATTGAGGGACA
protected from degradation. The solution was centrifuged and the lower GAPDH
Reverse: AGGAGTGAGTGGAAGACAGAATGGA
layer was the LNP encapsulated plasmid. We determined the plasmid as
C (encapsulated). The other part was added with 5% TritonX-100, and
reacted at room temperature for 15 min, so that LNP was completely
broken to release the encapsulated plasmid. At this time, the total 2.10. Mouse models
plasmid content was determined as C (total). The encapsulation effi­
ciency was calculated by the following formula: Encapsulation effi­ NOD-SCID IL-2 receptor gamma null (NSG) mice were injected with
ciency = encapsulated DNA/total DNA. We used BCA kit (Beyotime 5 × 106 Raji-Luc cells to construct acute lymphoblastic leukemia models.
Biotechnology) to detect the antibody loading of LNP. Briefly, free CD3 After luciferase-induced fluorescence was observed (in approximately
antibodies that do not bind to LNPs are removed by a sephadex column. one week), tumor-bearing mice were randomly divided into 8 groups (n
Then, AntiCD3-LNPs solution was mixed with methanol, and add 20 μL = 10). The drug was administered by tail vein injection at a dose of 5 μg/
into the 96 well plate. 200 μL of BCA working solution was added to each g (calculated as the mass of the plasmid). The efficacy of LNPs was
well and stand at 37 ◦ C for 30 min. The absorbance of each well at 562 compared with conventional CAR-T cell therapy by treating a group of
nm was measured by microplate reader, and the CD3 antibody con­ mice with a single dose of 5 × 106 △IL-6 CAR-T cells.
centration was calculated according to the standard curve. The antibody
loading rate is calculated as follows: Column-passing antibody/total
antibody*100%. 2.11. In vivo distribution of nanoparticles

2.7. In vitro T-cell transfection using AntiCD3-LNP/CAR19 + shIL6 The tumor-bearing mice were randomly divided into three groups,
namely, AntiCD3-LNP/CAR19 + shIL6 as the experimental group, LNP/
The AntiCD3-LNP/CAR19 + shIL6 nanoparticles and LNP/CAR19 + CAR 19 + shIL6 as the control group, and the blank normal saline group.
shIL6 nanoparticles loaded with DiO fluorescent dye were prepared, and The in vivo tissue distribution of AntiCD3-LNP/CAR19 + shIL6 was
the uptake of the two types of nanoparticles by T cells was observed with observed using the Small Animal In Vivo Imaging System (Perkin Elmer,
a laser confocal microscope. The AntiCD3-LNP/CAR19 + shIL6 and MA, USA) after tail vein injection of luciferase-labeled nanoparticles in
LNP/CAR19 + shIL6 loaded with GFP were incubated with human T mice.

300
J.-e. Zhou et al. Journal of Controlled Release 350 (2022) 298–307

2.12. Quantification of CAR-T produced by AntiCD3-LNP/CAR19 + particle size of about 200 nm, PDI < 0.2, and a zeta potential close to 0
shIL6 in peripheral blood mV (Fig. 2 A-C). AntiCD3-LNP/CAR19 + shIL6 was incubated in serum
and its size did not significantly change within 7 days (Fig. S6),
After injecting the nanoparticles into tumor-bearing mice, 0.1 mL of demonstrating its good stability in serum. To determine the ability of the
peripheral blood was taken from the mice on the days 7th, 14th, 21st, nanoparticles to target T cells in vitro, the LNPs were incubated with T
35th, and 90th, respectively. Then, white blood cells were separated cells for 4 h. The T cells took up much more CD3 antibody-modified
using a monocyte separation solution and erythrocyte lysis solution. The LNPs than unmodified nanoparticles (Fig. 2D). We investigated the
isolated leukocytes labeled with anti-human CD3 (PE-Cy7), anti-human endosomal escape of LNPs and found that the AntiCD3-LNP/CAR19 +
CD4 (PE), anti-human CD8 (APC), and anti-human CD19 (FITC) were shIL6 were internalized into the cells by the lysosomal pathway and
detected by flow cytometry to determine the number of CAR-T cells. could successfully escape the lysosomes at 4 h in vitro. (Fig. S7). The
detection of T cell transduction efficiency by the LNPs helped to deter­
mine the expression of CAR on the surface of T cells. We found that the
2.13. Statistical analysis transfection efficiency of LNPs was significantly enhanced after CD3
antibody modification, though the enhancement was not very remark­
All data are presented as the mean ± SD. Two groups of data were able (from 4.57 ± 0.30% to 6.9 ± 0.16%, Fig. 2E-F). We detected CD19
analyzed by performing Student’s t-tests (two-tailed). More than three CAR positive% of CAR-T cells produced by LNPs and those by lentivirus
groups of data were analyzed by performing analysis of variance with FACS. The results showed that the CAR positive rate of ΔIL6 CAR-T
(ANOVA), followed by Tukey’s multiple comparison test. We tested the was higher than that of LNPs (Fig. S8), which might be because the
normal distribution of the data using four methods: Anderson-Darling transfection efficiency of the lentiviral vector was significantly higher
test, D’Agostino&Pearson test, Shapiro-Wilk test and Kolmogorov- than that of LNPs. We selected different effector: target ratios (E: T) to
Smirnov test. Statistical significance was defined as * (p < 0.05), ** evaluate the ability to kill tumor cells after T cell transduction. The △IL-
(p < 0.01), or ***(p < 0.001). All analyses were performed using the 6 CAR-T group showed the highest killing ability at the effector: target
software GraphPad Prism v8.0. Pairwise differences in bioluminescent ratio of 10: 1. The AntiCD3-LNP/CAR19 + shIL6 group showed potent
tumors and T-cell signals were analyzed at selected time points using cytotoxicity and could kill the target cells effectively at E:T = 10:1 and
Wilcoxon rank-sum test, and the survival data were analyzed using the 8:1. The AntiCD3-LNP/CAR19 + shIL6 group showed significantly
Log-rank test. enhanced killing effect compared with LNP/CAR19 + shIL6 group at E:
T = 10:1. The AntiCD3-LNP/CAR19 + shIL6 group and LNP/CAR19 +
3. Results shIL6 group both showed weak killing ability at E:T = 5: 1 and 1:1
(Fig. 2G). We also examined the ability of T cells to express IL-6 after
3.1. LNPs programming T cells in vitro killing the target cells. The IL-6 mRNA levels generated in the AntiCD3-
LNP/CAR19 group were significantly higher than those generated in the
We prepared LNPs encapsulated with CAR plasmid and modified LNP/CAR19 + shIL6 and AntiCD3-LNP/CAR19 + shIL6 groups
with CD3 antibody as the targeting ligand on the surface to target T cells. (Fig. 2H), indicating that IL-6 shRNA in the CAR plasmid effectively
CD3 antibody was successfully conjugated with phospholipid (Fig. S5). silenced IL-6 expression.
The nanoparticles were spheroidal as shown in TEM images, with a

Fig. 2. The LNPs transfected T cells in vitro and produced CAR-T cells. A. The AntiCD3-LNP/CAR19 + shIL6 nanoparticles showed a spheroidal structure when
analyzed by TEM. B and C. The particle size of LNP was about 200 nm, and the zeta potential of LNP was nearly 0 mV. The data are represented as the mean ± SD (n
= 6). D. Confocal microscopy confirmed that the LNPs loaded with DiO dye were internalized by T cells within 4 h (This graph is representative of 3 human donors).
F. The efficiency of T cell transduction with AntiCD3-LNP/CAR19 + shIL6 was significantly higher than that with LNP/CAR19 + shIL6 in vitro. The data are
represented as the mean ± SD of three independent experiments. G. The killing ability of LNPs after T cell transduction following stimulation by K562 cells or Raji
cells. Data are represented as the mean ± SD for 3 technical triplicates, with three random fields of view assessed per replicate. The killing ability was highest at E:T
= 10:1. H. Detection of IL-6 mRNA produced by T-cell programming via qPCR. Data are represented as the mean ± SD of 5 technical triplicates. (The symbol *
represents P < 0.05, ** represents P < 0.01, and *** represents P < 0.001).

301
J.-e. Zhou et al. Journal of Controlled Release 350 (2022) 298–307

3.2. LNPs targeting T-cells and producing CAR-T cells In vivo gamma null (NSG) mice were injected with Raji-Luc cells through the
tail vein. After seven days, the fluorescence intensity of luciferase was
We used the LNPs to target T lymphocytes in vivo and to convert it determined by the small animal in vivo imaging to confirm the leukemia
into CAR-T cells. The targeting ability and transduction efficiency of model. Then, the tumor-bearing mice were randomly placed into one of
nanoparticles to T cells in vivo are crucial. First, NOD-SCID IL-2 receptor four groups, administered with T cells + CD3 antibody-modified LNPs

Fig. 3. CAR DNA-loaded LNPs programed the T cells and produced CAR-T cells in vivo. A. Representative Flow cytometry of the LNPs that bound to and transfected
the T cells. The LNPs showed the ability to program T cells in vivo. This graph is representative of n = 5 tumor-bearing mice. B. Biodistribution of fluorescent CD3
antibody targeting or non-targeted LNPs 24 h after tail-vein injection. Data are from five mice per treatment condition. The mice on the left were administered LNP/
CAR19 + shIL6, and the mice on the right were administered AntiCD3-LNP/CAR19 + shIL6. The CD3 antibody modified the LNPs targeting the lymphoid tissue in
which the T cells were located. C. The number of CAR-T cells produced by LNPs in vivo. The number of CAR-T cells in T+ AntiCD3-LNP/CAR19 + shIL6 group was
significantly higher than that in other groups on the 21st day. Data are represented as mean ± SD of 5 mice in each group. D. Phenotypic analysis of the LNPs after
transfection of T cells (n = 5, Data are represented as mean ± SD). E. The ratio of CD8+/CD4+ CAR-T cells in the peripheral blood. Large quantities of CD8 + CAR-T
were produced and reached highest on Day21 after LNPs administration. The ratio of CD8+/CD4+ CAR-T cells in T + AntiCD3-LNP/CAR19 + shIL6 group was
significantly more than that in other groups on Day 21.

302
J.-e. Zhou et al. Journal of Controlled Release 350 (2022) 298–307

(AntiCD3-LNP/CAR19 + shIL6), T cells+ LNPs (LNP/CAR19 + shIL6), T System. Twenty-four hours after injection, while most of LNP/CAR19 +
cells +CD3 antibody-modified LNPs without knocking down IL-6 shIL6 remained in the liver, AntiCD3-LNP/CAR19 + shIL6 was more
(AntiCD3-LNP/CAR19), and conventional CAR-T cells with knocked accumulated in the lymph nodes of the mice, (Fig. 3B; LNP/CAR19 +
down IL-6 function (△IL-6 CAR-T), respectively. The LNPs were injec­ shIL6 on the left and AntiCD3-LNP/CAR19 + shIL6 on the right), which
ted 2 h after the T-cell injection in each group. The dose of T cells was attributed to the CD3 antibody modification of LNPs.
injected was 5 × 106 per mouse, and the dose of LNPs was 5 μg/g (based To examine whether the LNPs could program the T cells and produce
on the mass of the CAR plasmid). After the tumor-bearing mice were CAR-T cells in vivo, we determined the number of CAR-T cells produced
injected with human T cells and DiR fluorescent dye-labeled AntiCD3- in vivo by flow cytometry on 7, 14, 21, 35, and 90 days after injecting T
LNP/CAR19 + shIL6 and LNP/CAR19 + shIL6, their fluorescence dis­ cells and nanoparticles. The number of CAR-T cells produced in each
tribution in vivo was observed by the Small Animal In Vivo Imaging group was plotted as a function of time after the administration. T cells

Fig. 4. AntiCD3-LNPs induced an antitumor effect by programming T cells in vivo. Almost all tumor cells were killed after 41 days of injection of T + AntiD3-LNP/
CAR19 + shIL6, which is similar with the positive control group of conventional△IL-6 CAR-T. The antitumor effect of T + LNP/CAR19 + shIL6 was lower than that
of T+ AntiCD3-LNP/CAR19 + shIL6. The injection of T cells, AntiCD3-LNP/CAR19 + shIL6 or LNP/CAR19 + shIL6 alone could not effectively kill tumor cells.

303
J.-e. Zhou et al. Journal of Controlled Release 350 (2022) 298–307

were stably transfected into CAR-T cells in vivo after injecting T+ shIL6 group (Fig. 3A). By contrast, only a few CAR-T cells were produced
AntiCD3-LNP/CAR19 + shIL6 or T+ LNP/CAR19 + shIL6. In the T+ in the T+ AntiCD3-LNP/CAR19 group. The number of CAR-T cells was
AntiCD3-LNP/CAR19 + shIL6 group, a large number of CAR-T cells the highest on the 21st day (Fig. 3C). Additionally, the number of CAR-T
were produced by the nanoparticles within 14–21 days after the treat­ cells in T+ AntiCD3-LNP/CAR19 + shIL6 group was significantly higher
ment (mean 74.6% CAR+ among CD3+ T cells on day 21), but the than that in T + LNP/CAR 19 + shIL6 group on the 21st day. This
number sharply decreased within 35–90 days after killing tumor cells indicated that modifying the CD3 antibody promoted the targeting of
((mean 62.5% CAR+ among CD3+ T cells on day 14, mean 12.1% CAR+ nanoparticles to T cells. The T cell subtypes that internalized the injected
among CD3 + T cells on day 90). In the T+ LNP/CAR19 + shIL6 group, LNPs were measured by flow cytometry, and the proportion of CD8+/
some T cells were programmed into CAR-T cells in vivo, but the number CD4+CAR-T cells was compared (Fig. S9). Most CAR-T cells were CD8+
of CAR-T cells was lesser than that in the T+ AntiCD3-LNP/CAR19 + CAR-T on days 14 and 21, whereas the proportion of CD8+ CAR-T cells

Fig. 5. LNPs programmed T cells caused antitumor effects and knocked down IL-6 in vivo. A. Quantification of bioluminescence intensity of each tumor-bearing
mouse. Each line represents a change in the bioluminescence intensity over time in each mouse. B. Survival of tumor-bearing mice following therapy, depicted
as Kaplan–Meier curves (n = 10). Statistical significance was calculated using the log-rank Mantel-Cox test; * represents P < 0.05, ** represents P < 0.01, and ***
represents P < 0.001. C. The concentration of IL-6 in peripheral blood after nanoparticle injection. AntiCD3-LNP/CAR19 + shIL6 significantly reduced the con­
centration of IL-6 protein, which helped to reduce cytokine syndrome. Data are represented as mean ± SD of 5 mice in each group, and statistical significance was
calculated with ANOVA with Turkey multiple comparisons. D. The concentration of IL-4 in peripheral blood after administration. E. The IL-2 secretion of △IL-6 CAR-
T group in peripheral blood after administration was significantly higher than that of other groups on Day10. F. The secretion of IFN-γ in T + AntiCD3-LNP/CAR19
group, was significantly lower than that of △IL-6 CAR-T, LNP/CAR19 + shIL6, and AntiCD3-LNP/CAR19 + shIL6 groups. The concentration of IFN-γ in T +
AntiCD3-LNP/CAR19 + shIL6 group was significantly higher than that of in T + AntiCD3-LNP/CAR19 group on Day10. G. The concentration of IL-10 in peripheral
blood after administration. T + AntiCD3-LNP/CAR19 group secreted the highest level of IL-10 on Day10. H. The concentration of IL-17α in peripheral blood after
administration. I. The concentration of TNF-α in peripheral blood after administration. T + AntiCD3-LNP/CAR19 group secreted the highest level of TNF-α on Day10.

304
J.-e. Zhou et al. Journal of Controlled Release 350 (2022) 298–307

gradually decreased after the tumor was killed (Fig. 3D). We compared LNP/CAR19 + shIL6 groups. This might be the main reason for the low
the CD8+/CD4+ CAR-T cells ratio in different groups on Day 21. The survival rate of mice in T + AntiCD3-LNP/CAR19 group. Tumor
results showed that the number of CD8 + CAR-T cells in T + AntiCD3- microenvironment usually has high levels of IL-4 [18], which promotes
LNP/CAR19 + shIL6 group was significantly more than that in other immunosuppression and the production of pro-inflammatory cytokines.
groups on Day 21 (Fig. 3E), indicating that AntiCD3-LNP/CAR19 + IL-17A can promote the activation of T cells and enhance the recruit­
shIL6 could produce more cytotoxic T cells than other types of LNPs. ment of autoimmune cells in patients. There was no significant differ­
ence of the two cytokines among different groups (Fig. 5D, Fig. 5H). IL-
3.3. LNPs induced an antitumor effect and reduced CRS by programming 2, IL-10, and TNF-α are pro-inflammatory cytokines that can activate a
T cells series of inflammatory-related cascades, which are closely related to the
occurrence of CRS. T + AntiCD3-LNP/CAR19 + shIL6 group produced
To investigate whether LNPs exerted antitumor effects after T cell lower levels of pro-inflammatory factors than T + AntiCD3-LNP/CAR19
programming in vivo, we systemically injected luciferase-expressing group on Day10 (Fig. 5E, G, and I), which is helpful to reduce the
Raji leukemia cells (Raji-Luc) into NSG mice and used bioluminescent incidence of CRS.
imaging to assess the differences of tumor burden among treatment
groups (Fig. 4). To compare the therapeutic efficacy of LNPs with con­ 4. Discussion
ventional CAR T-cell therapy, we set a positive control group of mice
treated with a single dose of 5 × 106 CAR-T cells, which was prepared by The traditional CAR-T cells need to be prepared ex vivo, which has
transduction ex vivo with lentiviral vectors encoding CD19 CAR+shIL-6 several problems, such as a complex production process, long cycle,
(△IL-6 CAR-T). We found that almost all tumor cells were killed after strict quality control requirements, personal customization, and high
41 days of injection of human T cells + AntiD3-LNP/CAR19 + shIL6, cost. Moreover, the problem of cytokine release syndrome (CRS) is
which is similar to the positive control group of conventional CAR T-cell common in the clinical application of CAR-T. To solve these problems, in
therapy. This could be because T cells are programmed by AntiCD3- this study, we used a mixture of ionizable phospholipids DLin-MC3-DMA
LNP/CAR19 + shIL6, and thus, transformed into CAR-T cells in vivo [32,33] and cationic lipids [34–36] as the material to prepare lipid
(Fig. 3C) that show strong antitumor effects comparable to conventional nanoparticles and modified the CD3 antibody on the surface so that it
CAR-T therapy. T cells + LNP/CAR19 + shIL6 also produced antitumor could be actively targeted to and internalized by T cells. The combined
effect in mice, which, however, was lower than that of T+ AntiCD3- plasmids containing IL-6 shRNA and CD19-CAR loaded by LNPs stably
LNP/CAR19 + shIL6. This is probably because LNP/CAR19 + shIL6 transfected T cells under the influence of the nuclear localization peptide
could not efficiently program T cells in vivo, leading to producing an MTAS-NLS to produce IL-6 knockdown CAR-T cells. Therefore, the
inadequate number of CAR-T cells. Moreover, the injection of human T technology could avoid complex preparation process ex vivo and in­
cells alone could not kill tumor cells in mice. Similarly, the injection of crease the convenience of CAR-T therapy; At the same time, it could
AntiCD3-LNP/CAR19 + shIL6 or LNP/CAR19 + shIL6 alone could not effectively reduce the CRS problem associated with CAR-T therapy and
kill tumor cells, which might be due to the lack of T cells in NSG mice improve its clinical safety.
and the failure of LNPs to program T cells to express CAR. We also found The rational design of gene delivery system is the key factor influ­
that T cells + AntiCD3-LNP/CAR19 showed a slight antitumor effect in encing the effective delivery and transfection of genes in vivo. The safety
vivo, which was lower than that of T cells + AntiCD3-LNP/CAR19 + of LNPs editing T cells in vivo determines whether they can be used
shIL6. This was mainly due to the low encapsulation efficiency of clinically. The component of phospholipid used for LNPs is referenced
AntiCD3-LNP/CAR19, which was significantly lower than AntiCD3- from ONPATTRO [37] and the Selective organ targeting (SORT) nano­
LNP/CAR19 + shIL6 (Table S2), thus causing poor transduction of T particles [38]. We used cationic lipid DOTAP and ionizable cationic lipid
cells and fewer production of CAR-T cells in vivo (Fig. 3C). DLinDMA as the main carrier materials of LNPs [39]. DLin-MC3-DMA
Fluorescence intensity analysis was performed on five representative has a neutral charge at around pH 7.0, which keeps LNPs stable in the
mice from each group (Fig. 5A). We found that T cells +AntiCD3-LNP/ blood, and its positive charge at low pH enables LNPs to escape from
CAR19 + shIL6 could reduce tumor burden from day 12, indicating that endosomes. Therefore, ionizable cationic lipids have shown advantages
nanoparticles exhibit an antitumor effect by programming T cells in in nucleic acid delivery in vivo [40]. Currently, EndoTAG-1 [41,42], a
vivo. We evaluated the survival time of tumor-bearing mice in each paclitaxel cationic liposome prepared from DOTAP, has entered clinical
group for 120 days after administration (Fig. 5B). We found that mice phase III research. However, a high positive charge of LNPs might cause
treated with T + AntiCD3-LNP/CAR19 + shIL6 survived significantly serious immune reactions and toxic side effects on normal tissues
longer than those treated with T+ LNP/CAR19 + shIL6. The therapeutic [43–45]. Therefore, maintaining a low or neutral charge is very
effect of T + AntiCD3-LNP/CAR19 + shIL6 was not significantly important for LNPs. We optimized the formulation based on previous
different from that of △IL-6 CAR-T cells. The median survival times of siRNA-loaded LNPs [32,40,46], so that the zeta potential of LNP was
mice injected with PBS, human T cells alone, LNP/CAR19 + shIL6, maintained at 0–2 mV by adjusting the amounts of pDNA, DOTAP, and
AntiCD3-LNP/CAR19 + shIL6, and T + AntiCD3-LNP/GFP were 20, 19, Dlin-MC3-DMA (Table S1).
12, 13, and 31 days, respectively. These results indicated that the in­ The transfection efficiency of LNP depends on the size of the pDNA
jection of human T cells or LNP/CAR19 + shIL6 or AntiCD3-LNP/ and makes it easier for smaller pDNA copies [47,48]. Smaller DNA
CAR19 + shIL6 alone cannot significantly prolong the survival of tumor- fragments are considered to be more easily encapsulated or more easily
bearing mice. To determine whether CRS is produced in tumor-bearing translocated to the nucleus. To ensure the high transfection efficiency of
mice after administration, we collected the serum from the mice on days AntiCD3-LNP/CAR19 + shIL6 in vivo, we added IL-6 shRNA based on
7, 10, and 14 after administration and detected the concentration of the original CAR19 pDNA and deleted many unnecessary components,
seven types of cytokines. After injection of human T cells+ AntiCD3- thus reducing the copies of the plasmid (5314 bp; lesser than CAR19
LNP/CAR19, tumor-bearing mice produced high levels of IL-6 on the (8504 bp) in AntiCD3-LNP/CAR 19 group) to facilitate the lipid
10th and 14th day (Fig. 5C), which was significantly higher than that in encapsulation (Fig. S1 and S2); In addition, we synthesized microtubule-
T + AntiCD3-LNP/CAR19 + shIL6 group. The results indicated that associated nuclear localization peptide (MTAS-NLS)-DSPE (Fig. S3,
AntiCD3-LNP/CAR19 + shIL6 transfected T cells successfully and Fig. S4) and added it into LNPs, promoting LNPs to enter the nucleus
knocked down IL-6 in vivo, which is helpful to reduce the incidence and while simultaneously adsorbing and compressing the plasmid. The main
degree of CRS. We found that the secretion of IFN-γ which related to reason is that delivering large DNA sequences need more positive
tumor killing in T + AntiCD3-LNP/CAR19 group, was significantly charges (high N/P ratio) to increase DNA condensation within the
lower than that of △IL-6 CAR-T, LNP/CAR19 + shIL6, and AntiCD3- nanoparticles. We encapsulated AntiCD3-LNPs/CAR19 with the same

305
J.-e. Zhou et al. Journal of Controlled Release 350 (2022) 298–307

amount of positive charge as AntiCD3-LNPs/CAR19 + shIL6, whereas [3] C. Sheridan, First approval in sight for Novartis’ CAR-T therapy after panel vote,
Nat. Biotechnol. 35 (2017) 691–693.
the size of CAR19 was much larger than that of CAR19 + shIL6, thus
[4] J.N. Brudno, R.P. Somerville, V. Shi, J.J. Rose, D.C. Halverson, D.H. Fowler, J.
resulting in insufficient condensation of CAR19 and low encapsulation C. Gea-Banacloche, S.Z. Pavletic, D.D. Hickstein, T.L. Lu, S.A. Feldman, A.
efficiency (Table S2). In addition, AntiCD3-LNP/CAR19 has more T. Iwamoto, R. Kurlander, I. Maric, A. Goy, B.G. Hansen, J.S. Wilder, B. Blacklock-
negative charges (− 3.4 ± 0.1). These could be the reasons for poor T cell Schuver, F.T. Hakim, S.A. Rosenberg, R.E. Gress, J.N. Kochenderfer, Allogeneic T
cells that express an anti-CD19 chimeric antigen receptor induce remissions of B-
transduction, CAR-T cell production, and antitumor effect in the cell malignancies that Progress after allogeneic hematopoietic stem-cell
AntiCD3-LNP/CAR19 group. The original transposon iPB7 was inserted transplantation without causing graft-versus-host disease, J. Clin. Oncol. 34 (2016)
in the plasmid for stable transfection of T cells by LNPs, thus T cells were 1112–1121.
[5] J.A. Fraietta, K.A. Beckwith, P.R. Patel, M. Ruella, Z. Zheng, D.M. Barrett, S.
able to stable express CAR gene in vivo. The results showed that it could F. Lacey, J.J. Melenhorst, S.E. McGettigan, D.R. Cook, C. Zhang, J. Xu, P. Do,
last for >90 days after T cell transduction. J. Hulitt, S.B. Kudchodkar, A.P. Cogdill, S. Gill, D.L. Porter, J.A. Woyach, M. Long,
We successfully programmed T cells in vivo, but the off-target effect A.J. Johnson, K. Maddocks, N. Muthusamy, B.L. Levine, C.H. June, J.C. Byrd, M.
V. Maus, Ibrutinib enhances chimeric antigen receptor T-cell engraftment and
during treatment may reduce the safety. We found that some LNPs efficacy in leukemia, Blood 127 (2016) 1117–1127.
transfected macrophages in vivo, and we also examined the number of [6] P. Kebriaei, H. Singh, M.H. Huls, M.J. Figliola, R. Bassett, S. Olivares, B. Jena, M.
macrophages that expressed CAR (Fig. S10). A small portion of macro­ J. Dawson, P.R. Kumaresan, S. Su, S. Maiti, J. Dai, B. Moriarity, M.A. Forget,
V. Senyukov, A. Orozco, T. Liu, J. McCarty, R.N. Jackson, J.S. Moyes, G. Rondon,
phages was transfected and expressed CAR, but these CAR-macrophages M. Qazilbash, S. Ciurea, A. Alousi, Y. Nieto, K. Rezvani, D. Marin, U. Popat,
were difficult to proliferate in vivo. By contrast, the CAR-T cells pro­ C. Hosing, E.J. Shpall, H. Kantarjian, M. Keating, W. Wierda, K.A. Do, D.
duced in vivo could continuously proliferate under the stimulation of A. Largaespada, D.A. Lee, P.B. Hackett, R.E. Champlin, L.J. Cooper, Phase I trials
using sleeping beauty to generate CD19-specific CAR T cells, J. Clin. Invest. 126
tumor cells. No significant toxicity was observed through hematoxylin
(2016) 3363–3376.
and eosin (H&E) staining of sections of major organs (Fig. S11). This [7] C.J. Turtle, L.A. Hanafi, C. Berger, T.A. Gooley, S. Cherian, M. Hudecek,
indicated that the expression of CAR in non-T cells did not cause sig­ D. Sommermeyer, K. Melville, B. Pender, T.M. Budiarto, E. Robinson, N.
nificant damage to other organs. N. Steevens, C. Chaney, L. Soma, X. Chen, C. Yeung, B. Wood, D. Li, J. Cao,
S. Heimfeld, M.C. Jensen, S.R. Riddell, D.G. Maloney, CD19 CAR-T cells of defined
In conclusion, we found that the in vivo programming of T cells into CD4+:CD8+ composition in adult B cell ALL patients, J. Clin. Invest. 126 (2016)
CAR-T cells via LNPs achieved antitumor effect comparable to that of 2123–2138.
conventionally CAR-T therapy and reduced CRS risk by knocking down [8] R.C. Sterner, R.M. Sterner, CAR-T cell therapy: current limitations and potential
strategies, Blood Cancer J. 11 (2021) 69.
IL-6. This technology improves the convenience of using CAR-T tech­ [9] I.R. Marco, L. Davila, Xiuyan Wang, Shirley Bartido, Jae Park, Kevin Curran,
nology, and is helpful to further promote the clinical application. Stephen S. Chung, Jolanta Stefanski, Oriana Borquez-Ojeda,
Malgorzata Olszewska, Jinrong Qu, Teresa Wasielewska, Qing He, Mitsu Fink,
Himaly Shinglot, Maher Youssif, Mark Satter, Yongzeng Wang, James Hosey,
Funding Hilda Quintanilla, Elizabeth Halton, Yvette Bernal, Diana C.G. Bouhassira, Maria
E. Arcila, Mithat Gonen, Gail J. Roboz, Peter Maslak, Dan Douer, Mark G. Frattini,
This work was supported by the National Natural Science Foundation Sergio Giralt Michel Sadelain, Renier Brentjens, Efficacy and toxicity management
of 19-28z CAR T cell therapy in B cell acute lymphoblastic leukemia, Cancer 6
of China (81872812, 82073800). (2014).
[10] B.L. Levine, J. Miskin, K. Wonnacott, C. Keir, Global manufacturing of CAR T cell
Credit author statement therapy, Mol. Ther. Methods Clin. Dev. 4 (2017) 92–101.
[11] X. Wang, I. Riviere, Clinical manufacturing of CAR T cells: foundation of a
promising therapy, Mol. Ther. Oncolytics 3 (2016) 16015.
Jing-e Zhou: Perform experiments, analyze data, and writing the [12] S.S. Neelapu, S. Tummala, P. Kebriaei, W. Wierda, C. Gutierrez, F.L. Locke, K.
manuscript. V. Komanduri, Y. Lin, N. Jain, N. Daver, J. Westin, A.M. Gulbis, M.E. Loghin, J.
F. de Groot, S. Adkins, S.E. Davis, K. Rezvani, P. Hwu, E.J. Shpall, Chimeric antigen
Lei Sun: Perform experiments, analyze data, and writing the receptor T-cell therapy - assessment and management of toxicities, Nat. Rev. Clin.
manuscript. Oncol. 15 (2018) 47–62.
Yujie Jia: Perform experiment. [13] S.S. Neelapu, S. Tummala, P. Kebriaei, W. Wierda, F.L. Locke, Y. Lin, N. Jain,
N. Daver, A.M. Gulbis, S. Adkins, K. Rezvani, P. Hwu, E.J. Shpall, Toxicity
Zhehao Wang:Perform experiment. management after chimeric antigen receptor T cell therapy: one size does not fit
Tengshuo Luo: Perform experiments. ‘ALL’, Nat. Rev. Clin. Oncol. 15 (2018) 218.
Jingwen Tan: Perform experiments. [14] S.L. Maude, N. Frey, P.A. Shaw, R. Aplenc, D.M. Barrett, N.J. Bunin, A. Chew, V.
E. Gonzalez, Z. Zheng, S.F. Lacey, Y.D. Mahnke, J.J. Melenhorst, S.R. Rheingold,
Xiaoyan Fang:Perform experiments.
A. Shen, D.T. Teachey, B.L. Levine, C.H. June, D.L. Porter, S.A. Grupp, Chimeric
Hongjia Zhu:Perform experiments. antigen receptor T cells for sustained remissions in leukemia, N. Engl. J. Med. 371
Jing Wang:Support the program and review the manuscript. (2014) 1507–1517.
[15] F.L. Locke, S.S. Neelapu, N.L. Bartlett, T. Siddiqi, J.C. Chavez, C.M. Hosing,
Lei Yu: Design and support the program and review the manuscript.
A. Ghobadi, L.E. Budde, A. Bot, J.M. Rossi, Y. Jiang, A.X. Xue, M. Elias, J. Aycock,
Zhiqiang Yan: Design and support the program, writing and review J. Wiezorek, W.Y. Go, Phase 1 results of ZUMA-1: a multicenter study of KTE-C19
the manuscript. anti-CD19 CAR T cell therapy in refractory aggressive lymphoma, Mol. Ther. 25
(2017) 285–295.
[16] R.Q. Le, L. Li, W. Yuan, S.S. Shord, L. Nie, B.A. Habtemariam, D. Przepiorka, A.
Declaration of Competing Interest T. Farrell, R. Pazdur, FDA approval summary: tocilizumab for treatment of
chimeric antigen receptor T cell-induced severe or life-threatening cytokine release
syndrome, Oncologist 23 (2018) 943–947.
The authors have no competing interests. [17] L. Kang, X. Tang, J. Zhang, M. Li, N. Xu, W. Qi, J. Tan, X. Lou, Z. Yu, J. Sun,
Z. Wang, H. Dai, J. Chen, G. Lin, D. Wu, L. Yu, Interleukin-6-knockdown of
chimeric antigen receptor-modified T cells significantly reduces IL-6 release from
Appendix A. Supplementary data monocytes, Exp. Hematol. Oncol. 9 (2020) 11.
[18] T. Jain, M. Bar, A.J. Kansagra, E.A. Chong, S.K. Hashmi, S.S. Neelapu, M. Byrne,
Supplementary data to this article can be found online at https://doi. E. Jacoby, A. Lazaryan, C.A. Jacobson, S.M. Ansell, F.T. Awan, L. Burns,
V. Bachanova, C.M. Bollard, P.A. Carpenter, J.F. DiPersio, M. Hamadani, H.
org/10.1016/j.jconrel.2022.08.033.
E. Heslop, J.A. Hill, K.V. Komanduri, C.A. Kovitz, H.M. Lazarus, J.M. Serrette,
M. Mohty, D. Miklos, A. Nagler, S.Z. Pavletic, B.N. Savani, S.J. Schuster, M.
References A. Kharfan-Dabaja, M.A. Perales, Y. Lin, Use of chimeric antigen receptor T cell
therapy in clinical practice for relapsed/refractory aggressive B cell non-Hodgkin
lymphoma: an expert panel opinion from the American Society for Transplantation
[1] FDA approves second CAR T-cell therapy, Cancer Discov. 8 (2018) 5–6.
and Cellular Therapy, Biol. Blood Marrow Transpl. 25 (2019) 2305–2321.
[2] T.J. Fry, N.N. Shah, R.J. Orentas, M. Stetler-Stevenson, C.M. Yuan, S. Ramakrishna,
[19] F. Bonacina, D. Coe, G. Wang, M.P. Longhi, A. Baragetti, A. Moregola,
P. Wolters, S. Martin, C. Delbrook, B. Yates, H. Shalabi, T.J. Fountaine, J.F. Shern,
K. Garlaschelli, P. Uboldi, F. Pellegatta, L. Grigore, L. Da Dalt, A. Annoni,
R.G. Majzner, D.F. Stroncek, M. Sabatino, Y. Feng, D.S. Dimitrov, L. Zhang,
S. Gregori, Q. Xiao, D. Caruso, N. Mitro, A.L. Catapano, F.M. Marelli-Berg, G.
S. Nguyen, H. Qin, B. Dropulic, D.W. Lee, C.L. Mackall, CD22-targeted CAR T cells
D. Norata, Myeloid apolipoprotein E controls dendritic cell antigen presentation
induce remission in B-ALL that is naive or resistant to CD19-targeted CAR
and T cell activation, Nat. Commun. 9 (2018) 3083.
immunotherapy, Nat. Med. 24 (2018) 20–28.

306
J.-e. Zhou et al. Journal of Controlled Release 350 (2022) 298–307

[20] E. Roselli, R. Faramand, M.L. Davila, Insight into next-generation CAR [36] J. Buck, P. Grossen, P.R. Cullis, J. Huwyler, D. Witzigmann, Lipid-based DNA
therapeutics: designing CAR T cells to improve clinical outcomes, J. Clin. Invest. therapeutics: hallmarks of non-viral gene delivery, ACS Nano 13 (2019)
131 (2021). 3754–3782.
[21] N.N. Parayath, M.T. Stephan, In situ programming of CAR T cells, Annu. Rev. [37] A. Akinc, M.A. Maier, M. Manoharan, K. Fitzgerald, M. Jayaraman, S. Barros,
Biomed. Eng. 23 (2021) 385–405. S. Ansell, X. Du, M.J. Hope, T.D. Madden, B.L. Mui, S.C. Semple, Y.K. Tam,
[22] A.R.K. Kumar, Y. Shou, B. Chan, K.L.A. Tay, Materials for improving immune cell M. Ciufolini, D. Witzigmann, J.A. Kulkarni, R. van der Meel, P.R. Cullis, The
transfection, Adv. Mater. 33 (2021), e2007421. Onpattro story and the clinical translation of nanomedicines containing nucleic
[23] M.M. Billingsley, A.G. Hamilton, D. Mai, S.K. Patel, K.L. Swingle, N.C. Sheppard, C. acid-based drugs, Nat. Nanotechnol. 14 (2019) 1084–1087.
H. June, M.J. Mitchell, Orthogonal Design of Experiments for optimization of lipid [38] L.T.J. Qiang Cheng, J. Daniel, Tuo Wei Siegwart, Lukas Farbiak, Sean A. Dilliard,
nanoparticles for mRNA engineering of CAR T cells, Nano Lett. 22 (1) (2021) Selective organ targeting (SORT) nanoparticles for tissue-specific mRNA delivery
533–542. and CRISPR–Cas gene editing, Nat. Nanotechnol. 15 (2020) 313–320.
[24] T.T. Smith, S.B. Stephan, H.F. Moffett, L.E. McKnight, W. Ji, D. Reiman, [39] P. de Antonellis, L. Liguori, A. Falanga, M. Carotenuto, V. Ferrucci, I. Andolfo,
E. Bonagofski, M.E. Wohlfahrt, S.P.S. Pillai, M.T. Stephan, In situ programming of F. Marinaro, I. Scognamiglio, A. Virgilio, G. De Rosa, A. Galeone, S. Galdiero,
leukaemia-specific T cells using synthetic DNA nanocarriers, Nat. Nanotechnol. 12 M. Zollo, MicroRNA 199b-5p delivery through stable nucleic acid lipid particles
(2017) 813–820. (SNALPs) in tumorigenic cell lines, Naunyn Schmiedeberg’s Arch. Pharmacol. 386
[25] X. Han, Y. Wang, W.D. Han, Chimeric antigen receptor modified T-cells for cancer (2013) 287–302.
treatment, Chronic. Dis. Transl. Med. 4 (2018) 225–243. [40] D. Adams, A. Gonzalez-Duarte, W.D. O’Riordan, C.C. Yang, M. Ueda, A.V. Kristen,
[26] R. Ballotti, Y. Cheli, C. Bertolotto, The complex relationship between MITF and the I. Tournev, H.H. Schmidt, T. Coelho, J.L. Berk, K.P. Lin, G. Vita, S. Attarian,
immune system: a melanoma ImmunoTherapy (response) factor? Mol. Cancer 19 V. Plante-Bordeneuve, M.M. Mezei, J.M. Campistol, J. Buades, T.H. Brannagan 3rd,
(2020) 170. B.J. Kim, J. Oh, Y. Parman, Y. Sekijima, P.N. Hawkins, S.D. Solomon,
[27] N.N. Parayath, S.B. Stephan, A.L. Koehne, P.S. Nelson, M.T. Stephan, In vitro- M. Polydefkis, P.J. Dyck, P.J. Gandhi, S. Goyal, J. Chen, A.L. Strahs, S.V. Nochur,
transcribed antigen receptor mRNA nanocarriers for transient expression in M.T. Sweetser, P.P. Garg, A.K. Vaishnaw, J.A. Gollob, O.B. Suhr, Patisiran, an RNAi
circulating T cells in vivo, Nat. Commun. 11 (2020) 6080. therapeutic, for hereditary transthyretin amyloidosis, N. Engl. J. Med. 379 (2018)
[28] K. Narayanan, S.K. Yen, Q. Dou, P. Padmanabhan, T. Sudhaharan, S. Ahmed, J. 11–21.
Y. Ying, S.T. Selvan, Mimicking cellular transport mechanism in stem cells through [41] A. Awada, I.N. Bondarenko, J. Bonneterre, E. Nowara, J.M. Ferrero, A.V. Bakshi,
endosomal escape of new peptide-coated quantum dots, Sci. Rep. 3 (2013) 2184. C. Wilke, M. Piccart, C.T.s. group, a randomized controlled phase II trial of a novel
[29] K. Kogure, R. Moriguchi, K. Sasaki, M. Ueno, S. Futaki, H. Harashima, Development composition of paclitaxel embedded into neutral and cationic lipids targeting
of a non-viral multifunctional envelope-type nano device by a novel lipid film tumor endothelial cells in advanced triple-negative breast cancer (TNBC), Ann.
hydration method, J. Control. Release 98 (2004) 317–323. Oncol. 25 (2014) 824–831.
[30] J.A. Kulkarni, D. Witzigmann, J. Leung, Y.Y.C. Tam, P.R. Cullis, On the role of [42] U. Fasol, A. Frost, M. Buchert, J. Arends, U. Fiedler, D. Scharr, J. Scheuenpflug,
helper lipids in lipid nanoparticle formulations of siRNA, Nanoscale 11 (2019) K. Mross, Vascular and pharmacokinetic effects of EndoTAG-1 in patients with
21733–21739. advanced cancer and liver metastasis, Ann. Oncol. 23 (2012) 1030–1036.
[31] E. Ambegia, S. Ansell, P. Cullis, J. Heyes, L. Palmer, I. MacLachlan, Stabilized [43] H. Lv, S. Zhang, B. Wang, S. Cui, J. Yan, Toxicity of cationic lipids and cationic
plasmid-lipid particles containing PEG-diacylglycerols exhibit extended circulation polymers in gene delivery, J. Control. Release 114 (2006) 100–109.
lifetimes and tumor selective gene expression, Biochim. Biophys. Acta 1669 (2005) [44] S. Daniel, A.C. Friend, J. Robert, C. Debs, Endocytosis and intracellular processing
155–163. accompanying transfection mediated by cationic liposomes, Biochim. Biophys.
[32] J.J. Wheeler, L.P.M. Ossanlou, P.R. Cullis, Stabilized plasmid-lipid particles: Acta 1278 (1996) 41–50.
construction and characterization, Gene Ther. 6 (1999) 271–281. [45] D. Mirska, K. Schirmer, S.S. Funari, A. Langner, B. Dobner, G. Brezesinski,
[33] S.C. Semple, A. Akinc, J. Chen, A.P. Sandhu, B.L. Mui, C.K. Cho, D.W. Sah, Biophysical and biochemical properties of a binary lipid mixture for DNA
D. Stebbing, E.J. Crosley, E. Yaworski, I.M. Hafez, J.R. Dorkin, J. Qin, K. Lam, K. transfection, Colloids Surf. B: Biointerfaces 40 (2005) 51–59.
G. Rajeev, K.F. Wong, L.B. Jeffs, L. Nechev, M.L. Eisenhardt, M. Jayaraman, [46] S.M.A. Muthusamy Jayaraman, Barbara L. Mui, Ying K. Tam, Jianxin Chen,
M. Kazem, M.A. Maier, M. Srinivasulu, M.J. Weinstein, Q. Chen, R. Alvarez, S. Xinyao Du, David Butler, Laxman Eltepu, Shigeo Matsuda, Jayaprakash
A. Barros, S. De, S.K. Klimuk, T. Borland, V. Kosovrasti, W.L. Cantley, Y.K. Tam, K. Narayanannair, Kallanthottathil G. Rajeev, Ismail M. Hafez, Akin Akinc, Martin
M. Manoharan, M.A. Ciufolini, M.A. Tracy, A. de Fougerolles, I. MacLachlan, P. A. Maier, Mark A. Tracy, Pieter R. Cullis, Thomas D. Madden, Muthiah Manoharan,
R. Cullis, T.D. Madden, M.J. Hope, Rational design of cationic lipids for siRNA J. Michael, Hope, maximizing the potency of siRNA lipid nanoparticles for hepatic
delivery, Nat. Biotechnol. 28 (2010) 172–176. gene silencing in vivo, Angew. Chem. Int. Ed. 51 (2012) 8529.
[34] N. Skandrani, A. Barras, D. Legrand, T. Gharbi, H. Boulahdour, R. Boukherroub, [47] K. Remaut, N.N. Sanders, F. Fayazpour, J. Demeester, S.C. De Smedt, Influence of
Lipid nanocapsules functionalized with polyethyleneimine for plasmid DNA and plasmid DNA topology on the transfection properties of DOTAP/DOPE lipoplexes,
drug co-delivery and cell imaging, Nanoscale 6 (2014) 7379–7390. J. Control. Release 115 (2006) 335–343.
[35] L. Zhang, P. Wang, Q. Feng, N. Wang, Z. Chen, Y. Huang, W. Zheng, X. Jiang, Lipid [48] M. Brgles, M. Santak, B. Halassy, D. Forcic, J. Tomasic, Influence of charge ratio of
nanoparticle-mediated efficient delivery of CRISPR/Cas9 for tumor therapy, NPG liposome/DNA complexes on their size after extrusion and transfection efficiency,
Asia Mater. 9 (2017) e441. Int. J. Nanomedicine 7 (2012) 393–401.

307

You might also like