You are on page 1of 9

original article © The American Society of Gene & Cell Therapy

A Short Pulse of IL-4 Delivered by DCs


Electroporated With Modified mRNA
Can Both Prevent and Treat Autoimmune
Diabetes in NOD Mice
Rémi J Creusot1, Pearl Chang1, Don G Healey2, Irina Y Tcherepanova2, Charles A Nicolette2
and C Garrison Fathman1
1
Division of Immunology and Rheumatology, Stanford University School of Medicine, Stanford, California, USA; 2Argos Therapeutics, Durham,
North Carolina, USA

Bone marrow-derived dendritic cells (DCs) are cells of the dendritic cells (DCs) have received wide scrutiny because of their
immune system that have been used as a tool to boost, inherent ability to regulate immune responses. Depending on their
modulate, or dampen immune responses. In the con- type and maturation state, DCs can promote immune tolerance as
text of autoimmunity, DCs can be modified to express well as trigger immune responses. Tolerogenic properties of DCs
immuno­regulatory products encoded by transgenes, and can be ascribed not only to their state of maturation (immature),
used therapeutically in adoptive cellular therapy. DCs but also to certain molecules expressed on their surface (e.g., pro-
that were lentivirally transduced (lt) to express ­interleukin grammed death ligand-1), intracellularly (e.g., indoleamine 2,3-­
4 (IL-4) can significantly delay or prevent the onset of dioxygenase) or secreted [e.g., interleukin 10 (IL-10)].3,4 Some
autoimmune diabetes in nonobese diabetic (NOD) other immunoregulatory products that are known to prevent
mice. However, modifying cells using viral vectors ­carries or ameliorate autoimmune disease are either not expressed by
the dual risk of oncogenicity or immuno­genicity. This DCs, or expressed in insufficient amounts. However, DCs remain
study demonstrates that NOD DCs, electroporated with vehicles of choice to express these immunomodulatory products
“translationally enhanced” IL-4 mRNA (eDC/IL-4), can because of their efficient migration to relevant lymphoid tissues
be equally efficient therapeutically, despite the reduced and interaction with T cells. In mice, the pancreatic lymph nodes
amount and shorter duration of IL-4 secretion. Moreover, (PLNs) are among the few lymphoid tissues preferentially targeted
a single injection of eDC/IL-4 in NOD mice shortly after by adoptively transferred bone ­marrow-derived DCs, which makes
the onset of hyperglycemia was able to maintain stable this strategy particularly relevant for the treatment of T1D.5
glycemia for up to several months in a significant ­fraction IL-4 is a pleiotropic cytokine whose contribution to immune
of treated mice. Treatment with eDC/IL-4 boosted regu- regulation is well established. One of its most important roles is to
latory T (Tregs) cell functions and modulated T helper support the differentiation and function of Th2 and B cells, while
responses to reduce pathogenicity. Thus, treatment with antagonizing the development and function of Th1 cells, impli-
cated in numerous autoimmune diseases. IL-4 is not normally
DCs, electroporated with modified IL-4 mRNA to express
produced by DCs, and viral vectors have so far been the method
IL-4 for up to 24 hours, constitutes a viable cellular ther-
of choice to express it ectopically. DCs modified to express IL-4
apy approach for the regulation of autoimmune diabetes,
(DC/IL-4) have shown therapeutic efficacy in NOD mice,6,7 as well
as a preferred alternative to the use of viral vectors.
as in collagen-induced arthritis, a mouse model of rheumatoid
Received 24 May 2010; accepted 14 June 2010; published online arthritis.8,9 The approach of using viral vectors allows relatively
13 July 2010. doi:10.1038/mt.2010.146
stable expression of the transgene, but carries certain risks and
disadvantages that are inherent to the type of viral vector used
Introduction (most typically adenoviral and lentiviral), ranging from immuno-
Type 1 diabetes (T1D) is a multifactorial autoimmune disease genicity to oncogenicity. Nonviral approaches for ectopic expres-
characterized by the progressive T cell-mediated destruction of β sion of immunoregulatory products are not only safer, but should
cells of the pancreatic islets. The nonobese diabetic (NOD) mouse preserve the integrity of the DC, including its homing and T cell
model of T1D shares many similarities with human T1D and has clustering capacity.
been widely used to study the pathogenesis of disease and to test the Modification of DCs by mRNA electroporation has been
efficacy of therapeutic approaches aimed at maintaining or restor- extensively studied for the treatment of cancer, where newly
ing tolerance to and function of β cells.1,2 Among these approaches, introduced mRNAs typically encode tumor antigens and other

Correspondence: C Garrison Fathman, Division of Immunology and Rheumatology, 269 Campus Drive, CCSR Building Room 2225, Stanford, California
94305-5166, USA. E-mail: cfathman@stanford.edu

2112 www.moleculartherapy.org vol. 18 no. 12, 2112–2120 dec. 2010


© The American Society of Gene & Cell Therapy
NOD T1D Treatment With mRNA Electroporated DCs

proteins that boost the immunogenicity of the DCs.10–12 However, ltDCs were in contact with lentiviral particles for the final 2 days
this approach has not yet been successfully applied to autoimmune of a 7-day culture, eDCs were electroporated after the final harvest
diseases. Here, we report the use of an IL-4 mRNA optimized for on day 7. Using our extensively optimized conditions, electropo-
IL-4 protein expression to electroporate bone marrow-derived ration was carried out without significant loss of cells or change
DCs and then treat NOD mice before or immediately after the in viability (data not shown). Electroporation had the highest and
onset of hyperglycemia. DCs electroporated with translationally most consistent transfection efficiency (80–85%), whereas lenti-
enhanced IL-4 mRNA (eDC/IL-4), despite the short duration of viral transduction efficiency was more variable (40–70%), but in
IL-4 expression, had equivalent efficacy in preventing disease as general the transduced cells expressed higher levels of the trans-
did the previously described lentivirally transduced counterparts gene (Figure 1a and Supplementary Figure S1). Lentiviral trans-
(ltDC/IL-4).7 In addition, eDC/IL-4 cells were able to induce duction did not affect the differentiation of DCs (>90% CD11b+
a transient or sustained reversal of hyperglycemia in a signifi- CD11c+ at the end of the culture, Figure 1b), but increased their
cant fraction of mice treated after the onset of hyperglycemia. maturity, as seen by the expression of costimulatory molecules
Treatment with eDC/IL-4 reinforced tolerance through several and class II major histocompatibility complex (Figure 1c).
mechanisms, including induction of regulatory T cells (Tregs) and
skewing of helper T cell populations. This study represents a proof Function of modified DCs
of principle that electroporation of DCs with enhanced mRNA is The function of electroporated and transduced DCs was assessed by
a viable, safe, and effective substitute for viral vectors when using the amount of IL-4 secreted using enzyme-linked immunosorbent
cellular gene therapy to treat autoimmune diseases such as T1D. assay (ELISA). We observed that eDC/IL-4 consistently secreted
less than half the amount of IL-4 produced by ltDC/IL-4 within a
Results culture period of 24 hours in vitro at 37 °C, whereas unmodified
Characterization of electroporated DCs produced no detectable levels of IL-4 (Figure 2a). IL-4 intra-
and transduced DCs cellular staining showed that eDC/IL-4 no longer produced IL-4
We compared mRNA electroporation (eDCs) and lentiviral trans- by 24 hours whereas ltDC/IL-4 maintained IL-4 production, sug-
duction (ltDCs) for their efficiency of transgene transfer and gesting that, as expected, eDC/IL-4 use up their capacity to make
expression and for their effect on the phenotype of DCs. While IL-4 faster than ltDC/IL-4 (Figure 2b).

a GFP IL-4
100 100

80 80
Control DCs
% of Max

% of Max

60 60
Electroporated DCs (GFP mRNA on
40 40
left panel, IL-4 mRNA on right panel)
20 20

0 0 Transduced DCs (IL-4-IRES-GFP)


100 101 102 103 104 100 101 102 103 104
FL1-H: FITC FL4-H: APC

b Staining control Control DCs Electroporated DCs Transduced DCs


104 104 104 104

103 103 103 103


CD11c

102 102 102 102

101 101 101 101

100 100 100 100


100 101 102 103 104 100 101 102 103 104 100 101 102 103 104 100 101 102 103 104
CD11b

c 100 100 100 100


CD40
CD40 CD80 CD86 MHC-II
80 80 80 80

60 60 60 60

40 40 40 40

20 20 20 20

0 0 0 0
100 1
102 103 104 0 1
102 103 104 0 1
102 103 104 0 1
10 10 10 10 10 10 10 102 103 104

Figure 1  Phenotype of electroporated and transduced DCs. (a) GFP and IL-4 expression by unmodified DCs (harvested on day 7), mRNA elec-
troporated DCs (harvested and electroporated on day 7), and lentivirally transduced DCs (infected on day 5 and harvested on day 7). (b) CD11c and
CD11b expression by unmodified, mRNA electroporated, and lentivirally transduced DCs. (c) CD40, CD80, CD86, and class II MHC (I-Ag7) expression
by unmodified, mRNA electroporated and lentivirally transduced DCs. DC, dendritic cell; GFP, green fluorescent protein; IL, interleukin; MHC, major
histocompatibility complex.

Molecular Therapy vol. 18 no. 12 dec. 2010 2113


© The American Society of Gene & Cell Therapy
NOD T1D Treatment With mRNA Electroporated DCs

a 90 b GFP IL-4
100 100
eDC/GFP
80 80 80
eDC/IL-4

eDC/IL-4
60 60
70
ItDC/IL-4
40 40
IL-4 (pg per 100 modified DCs)

60
20 20

50 0 0
100 101 102 103 104 100 101 102 103 104

40 100 100

80 80
30

ItDC/IL-4
60 60
20
40 40

10 20 20

0 0
0
0 10 101 102 103 104 100 101 102 103 104
0 2 4 6 8 10 12 14 16 18 20 22 24
Time in culture (hours) 3 hours 6 hours 24 hours

Figure 2  Function of electroporated and transduced DCs. (a) Time course of IL-4 secretion by unmodified DCs, eDC/IL-4, and ltDC/IL-4 in the
first 24 hours after reculture at 37 °C in vitro. For better comparison between electroporation and lentiviral transduction, the values were normalized
based on the efficiency of transfection/transduction, and shown as mean ± SD from different numbers of cell plated (normalized to 100 cells). Data
from two independent experiments are shown. (b) GFP and intracellular IL-4 expression in eDC/IL-4 and ltDC/IL-4 after 3, 6, and 24 hours in culture.
DC, dendritic cell; eDC, enhanced DC; GFP, green fluorescent protein; ltDC, lentivirally transduced DC; IL, interleukin; MHC, major histocompatibility
complex.

Prevention of onset of hyperglycemia detected by ELISA in the serum of treated mice between 1 and
We previously published that ltDC/IL-4, injected into 12-week- 3 days after administration of eDC/IL-4 (data not shown).
old prediabetic NOD mice, can delay the time of disease onset and
prevent disease in about 50% of treated mice.7 We asked whether Reversal of hyperglycemia
eDC/IL-4 could work as a nonviral, potentially safer alternative to Next, we asked whether eDC/IL-4 had any beneficial effect when
ltDC/IL-4. Cohorts of nondiabetic 12-week-old female NOD mice administered after the onset of hyperglycemia. A cohort of female
were treated with a single intravenous injection of phosphate- NOD mice was monitored weekly for glycemia between the age of
buffered saline (PBS), eDC/green fluorescent protein (GFP), eDC/ 11 and 30 weeks, and individual mice that became hyper­glycemic
IL-4, or ltDC/IL-4 (Figure 3). The control DCs (eDC/­GFP) slightly (250–500 mg/dl) during the period of observation were treated
delayed the onset of disease, but had no significant effect com- with a single injection of PBS, or freshly prepared eDC/GFP
pared to PBS (P = 0.692). However, both eDC/IL-4 and ltDC/IL-4 or eDC/IL-4, and monitored twice weekly thereafter for blood
significantly reduced the incidence of disease (P = 0.013 and 0.026, ­glucose. Mice with a blood glucose >500 mg/dl over two consecu-
respectively). As previously shown with ltDC/IL-4,7 IL-4 expres- tive measurements were sacrificed. Data in Figure 4a,b show the
sion by the DCs is required for the full therapeutic effect (eDC/ survival curve of control groups (PBS and eDC/GFP) and the
GFP versus eDC/IL-4, P = 0.023). Finally, despite the reduced eDC/IL-4-treated group. Mice treated with either PBS or eDC/
amount of IL-4 expressed by eDC/IL-4 compared to ltDC/IL-4, GFP had to be sacrificed within 40 days, and there was no sig-
these two treatments were equivalent therapeutically (P = 0.967). nificant difference between the two controls (P = 0.216). However,
It is noteworthy that some mice treated with eDC/IL-4 had mod- one-third of mice treated with eDC-IL-4 survived well beyond 40
est increases in glycemia (150–200 mg/dl) with transient episodes days, with a sustained period of reversal of hyperglycemia, com-
>250 mg/dl. However, because 250 mg/dl was our cutoff thresh- pared to all controls (P = 0.0014). Blood glucose measurements on
old for hyperglycemia in a prevention setting, some treated mice, individual mice are reported in Figure 4c,d and demonstrate that
although initially considered diabetic, returned to <250 mg/dl and the responding mice did not return to a state of normoglycemia,
did not develop full blown hyperglycemia (>500 mg/dl) during the but instead to a state of controlled moderate hyperglycemia fluc-
time they were monitored (Supplementary Figure S2). Thus, the tuating between 150 and 300 mg/dl. Two ­factors can contribute
therapeutic efficacy as shown in Figure 3 was slightly underesti- to how well individual mice respond to the treatment: the sever-
mated. Moreover, based on the amount of IL-4 produced in vitro, ity of hyperglycemia and the age at the time of treatment; these
we estimated that every million of the eDC/IL-4 and ltDC/IL4 parameters are plotted for both responding and nonresponding
injected produce respectively 260 ± 30 and 440 ± 100 ng of IL-4 mice in Supplementary Figure S4. The average glycemia at time
within 24 hours in vivo (the variability is dependent on the effi- of treatment with eDC/IL-4 was 307 ± 37 and 354 ± 53 mg/dl for
ciency of transfection/transduction, which is more consistent responding mice (n = 5) and nonresponding (n  = 10), respec-
with electroporation) (Supplementary Figure S3). No IL-4 was tively. Although the data suggests better effectiveness in mice

2114 www.moleculartherapy.org vol. 18 no. 12 dec. 2010


© The American Society of Gene & Cell Therapy
NOD T1D Treatment With mRNA Electroporated DCs

100 with less severe hyperglycemia, statistical significance was not


PBS iv (n = 27)
90
reached (P = 0.071). The median age of treatment of controls and
eDC/GFP iv (n = 24) eDC/IL-4 treated mice (in weeks) was similar (18 and 19, respec-
80
eDC/IL-4 iv (n = 25) tively), but within eDC/IL-4 treated mice, the median age at the
70 ItDC/IL-4 iv (n = 22) time of treatment was greater for responding mice (24) than for
­nonresponding mice (19).
Percentage diabetic

60

50 Changes in immune cell populations and cytokine


40 profiles in targeted tissues
Targeted tissues (PLNs and spleen) and nontargeted tissue (­cervical
30
lymph nodes) were collected 1 and 3 days after intravenous injec-
20 tion of PBS, eDC/GFP, or eDC/IL-4. No major change in the per-
10
centage of the overall population of CD25+ Foxp3+ cells among
CD4+ T cells was observed, except for a modest but significant
0
10 15 20 25 30
increase in the spleen with eDC/IL-4 treatment (Figure  5a and
Age of mice (weeks) Supplementary S5a). However, a small population (up to 1% of
CD4+ T cells) emerged with higher levels of CD25 and CD4 than
Figure 3  Incidence of diabetes after a single intravenous injection the general CD4+ CD25+ population (Supplementary Figure
of PBS, eDC/GFP, eDC/IL-4, or ltDC/IL-4 in 12-week-old prediabetic
S5b). This small population significantly increased between day 1
NOD mice. The arrow indicates the time of treatment. Data are pooled
from two independent experiments. Log rank test was used for statisti- and 3 following treatment with both eDC/GFP and eDC/IL-4, but
cal analysis. eDC, enhanced dendritic cell; GFP, green fluorescent pro- was further increased with eDC/IL-4 (Figure 5b). Moreover, these
tein; ltDC, lentivirally transduced DC; IL, interleukin; NOD, nonobese CD25hi cells were made up of two populations of Foxp3+ cells, one
diabetic; PBS, phosphate-buffered saline.
expressing higher levels of Foxp3 than the general CD25+ Foxp3+

a 100 b 100

90 90

80 Controls (n = 20)
80 PBS (n = 15)
eDC/IL-4 (n = 15)
eDC/GFP (n = 5)
70
70
60
60
Survival rate

50
50
40
40
P = 0.216 30
30
20 P = 0.0014
20
10
10
0
0 0 20 40 60 80 100 120 140 160 180 200 220 240 260 280 300 320
0 20 40 60
c 600 d 600

500 500
Blood glucose (mg/dl)

400 400

300 300

200 200

100 100
Normoglycemia

0 0
0 20 40 60 0 20 40 60 80 100 120 140 160 180 200 220 240 260 280 300 320
Days after treatment

Figure 4 Reversal of hyperglycemia after treatment with eDC/IL-4. (a,b) Survival curve of mice treated with (a) PBS or eDC/GFP, and (b) of all
control mice (PBS and eDC/GFP) or eDC/IL-4-treated mice. Mice were sacrificed when their blood glucose reached over 500 mg/dl after two consecu-
tive measurements. Log rank test was applied for statistical analysis. (c,d) Blood glucose measurements in mice treated with (c) PBS (dashed line,
n = 15) or eDC/GFP (solid lines, n = 5), and in mice treated with (d) eDC/IL-4 (n = 15). In d, responders were eDC/IL-4-treated mice that survived
longer than control mice (n = 5, solid lines); nonresponders (n = 10) are represented in dashed lines. Initial blood glucose value (day 0) is the mean
of two consecutive measurements (between 250 and 500 mg/dl) before treatment. eDC, enhanced dendritic cell; GFP, green fluorescent protein;
IL, interleukin; NOD, nonobese diabetic; PBS, phosphate-buffered saline.

Molecular Therapy vol. 18 no. 12 dec. 2010 2115


© The American Society of Gene & Cell Therapy
NOD T1D Treatment With mRNA Electroporated DCs

a 12 Day 1 Day 3 P = 0.037 d 700 Splenocytes from


12
PBS-treated
600
10 10
500
% CD25 + Foxp3+

8 8 400
6 300
6
200
4 4
100

Blood glucose (mg/dl)


2
2 0
10 12 14 16 18 20 22 24 26
0 0
PBS eDS/GFP eDC/IL-4 eDC/IL-4 PBS eDS/GFP eDC/IL-4
700 Splenocytes from
PLNs CLNs Spleen eDC/IL-4-treated
600
b 1.2 P = 0.009 P = 0.004
1 500
1 400
0.8
P = 0.004
0.8 300
hi

0.6
% CD25

200
0.6
0.4 100
0.4 P = 0.0004
0
0.2 0.2 10 12 14 16 18 20 22 24 26

0 0
PBS eDC/GFP eDC/IL-4 eDC/IL-4 PBS eDC/GFP eDC/IL-4 100 Splenocytes from
PBS-treated ( ---- )
PLNs CLNs Spleen 80 eDC/IL-4-treated ( ___ )

% diabetic
eDC/GFP eDC/IL-4 eDC/GFP eDC/IL-4 60 P = 0.041
100 100 100 100
40
80 80 80 80
Spleen

60 60 60 60 20
PLNs

40 40 40 40
20 0
20 20 20
10 12 14 16 18 20 22 24 26
0 0 0 0
100 101 102 103 104 100 101 102 103 104 100 101 102 103 104 100 101 102 103 104 Age (weeks)

Figure 5  Induction of functional Tregs. (a) Frequency of CD25+ Foxp3+ cells gated on CD4+ T cells in select lymphoid tissues 1 and 3 days after
treatment. (b) Frequency of induced CD25hi cells among CD4+ T cells (gating shown in Supplementary Figure S5b). Mean ± SD is from n = 5 mice
per group per time point. t-Test was performed for statistical analysis. P values shown on graph are between day 1 and day 3. Difference between
eDC/GFP and eDC/IL-4 in spleen on day 3 was also significant (P = 0.035 on a; P = 0.01 on b). (c) Expression level of Foxp3 on CD4+ CD25−
cells (shaded histogram, no line), CD4+ CD25+ cells (shaded histogram with line) and induced CD4+ CD25hi cells (thick line). Gating is shown in
Supplementary Figure S5b. (d) Development of hyperglycemia in NOD.SCID mice injected at 10 weeks of age with 3 × 106 splenocytes from PBS-
treated (upper panel) or eDC/IL-4-treated (middle panel) NOD mice (n = 6 mice per group). The incidence of diabetes is reported in the lower panel
(mice were considered diabetic after two consecutive measurement of blood glucose over 250 mg/dl). Log rank test was used in statistical analysis.
eDC, enhanced dendritic cell; GFP, green fluorescent protein; IL, interleukin; NOD, nonobese diabetic; PBS, phosphate-buffered saline.

population, and one expressing lower levels (Figure 5c). Adoptive the target tissue. In the PLNs, eDC/IL-4 reduced the expression
transfer of splenocytes from eDC/IL-4-treated mice into NOD. of IFN-γ significantly, resulting in a more than sixfold increase of
SCID mice did not efficiently transfer disease when compared to the IL-4/IFN-γ ratio to 7.8. In the spleen, eDC/IL-4 increased IL-4
splenocytes from PBS-treated mice (Figure 5d). Treatment with and IL-10 more than it increased IFN-γ, resulting in a >14-fold
modified DCs did not significantly alter the population of acti- increase of the IL-4/IFN-γ ratio to 0.1. The expression profile of
vated (CD44+ CD69+) T cells (Supplementary Figure S6), nor IL-10 mimicked that of IL-4.
did it lead to dramatic changes in different populations of splenic
DCs (Supplementary Figure S7). Cells isolated from selected tis- Discussion
sues were also cultured in vitro without stimulation to assess the DCs play a crucial role in regulating immune responses, and
relative levels of interferon γ (IFN-γ), IL-4, and IL-10 induced have attracted much attention as tools to manipulate the immune
(Figure 6). No expression of these cytokines was seen from any response toward a desired outcome (stronger immune responses
tissue obtained at 1 day after treatment, from the tissues from PBS- against infections and tumors, versus dampened autoimmune
treated mice or from control cervical lymph nodes from eDC/ or allergic responses). The use of DCs generated ex vivo to treat
IL-4-treated mice. The cytokine profile on day 3 following treat- T1D has been proposed and tested by several groups.13–17 These
ment with control DCs (eDC/GFP) differed substantially between approaches consist, for example, of rendering the DCs more
PLNs and spleen. eDC/GFP induced similar amounts of IFN-γ tolerogenic by the use of particular cytokines in the culture, or
and IL-4 in the PLNs (IL-4/IFN-γ ratio = 1.26) but mostly IFN-γ introducing antisense oligonucleotides to downregulate costimu-
in the spleen (IL-4/IFN-γ ratio = 0.007). The additional effects of latory molecules. DCs can also be used as vehicles to express and
IL-4 delivered by eDC/IL-4 led to different changes depending on deliver immunoregulatory products to sites of relevance in vivo.

2116 www.moleculartherapy.org vol. 18 no. 12 dec. 2010


© The American Society of Gene & Cell Therapy
NOD T1D Treatment With mRNA Electroporated DCs

a 1,800
1,600
1,400
1,200 P = 0.77 P = 0.036

IL-4 (pg/ml)
1,000
800
600
P = 0.014
400 P = 0.39
200
0
−200 PBS eDC/GFP eDC/IL-4 eDC/IL-4 PBS eDC/GFP eDC/IL-4
b 8,000
7,000 P = 0.00068
36 hours 72 hours
6,000
5,000
IFN-γ (pg/ml)

4,000
P = 0.024
3,000
2,000 P = 0.036

1,000
0

−1000 PBS eDC/GFP eDC/IL-4 eDC/IL-4 PBS eDC/GFP eDC/IL-4


c 5,000
P = 0.81 P = 0.0039
4,000
IL-10 (pg/ml)

3,000

2,000
P = 0.027
P = 0.56
1,000

0
PBS eDC/GFP eDC/IL-4 eDC/IL-4 PBS eDC/GFP eDC/IL-4

PLN CLN Spleen

Figure 6 Local cytokine profile in targeted tissues. Cells from PLNs, CLNs, and spleen obtained 3 days after different treatments were cultured
in vitro. (a) IL-4, (b) IFN-γ, and (c) IL-10 were measured by ELISA on supernatant collected 36 hours (white bars) or 72 hours (gray bars) after replat-
ing. Mean ± SD is from n = 5 mice per group. t-Test was performed for statistical analysis. P values shown on graph are between eDC/GFP and eDC/
IL-4 for each time point. CLN, cervical lymph node; eDC, enhanced dendritic cell; GFP, green fluorescent protein; IFN-γ, interferon-γ; IL, interleukin;
NOD, nonobese diabetic; PBS, phosphate-buffered saline.

If  such immunoregulatory products are not normally expressed et al.22 have conducted a similar comparison and also noted that
by DCs, their respective genes must be introduced and adequately ­electroporation was more efficient, but that transduction led
expressed. To date, only viral means of DC modification have to higher levels of transgene expression, especially at later time
been successfully applied to achieve prevention or treatment of points (beyond 24 hours). In agreement with this study, we found
autoimmune diseases in mouse models, but safety concerns have that certain proteins, like GFP, are still present in high amount in
impeded the use of this approach in attempts at human therapy. cells up to 3 days after electroporation. Expression of such pro-
In the field of tumor immunology, researchers have achieved teins does not necessarily reflect the duration of mRNA expres-
expression of certain tumor antigens using electroporation of auto­ sion if the expressed protein is very stable (slowly degraded).
logous DCs with mRNA or translationally enhanced mRNA for In the case of a secreted protein like IL-4, however, we did not
vaccination strategies.10–12 This has led to several trials in humans, detect expression beyond 24 hours, whereas ltDCs were still
demonstrating the safety of mRNA electroporated DCs.18–21 In making IL-4 at this time. Unlike transduced cells, electroporated
this present study, we explore and report for the first time the use DCs bear a finite quantity of mRNA, most of which will in turn
of translationally enhanced mRNA electroporation as an alterna- be translated into protein as long as the mRNA persists. In the
tive to viral means of modifying DCs to express immunomodula- case of cytokines like IL-4, most of the mature protein presum-
tory cytokines for adoptive cellular gene therapy of autoimmune ably gets secreted within 12 hours. Although eDC/IL-4 produced
diseases. Electroporation with mRNA was more efficient than less IL-4 in a 24-hour period than ltDC/IL-4, they expressed
lentiviral transduction in modifying a greater number of DCs, higher levels on per cell basis within the first 6 hours after elec-
and did so with no apparent effect on their phenotype. Dullaers troporation. This elevated initial level of expression, at the time

Molecular Therapy vol. 18 no. 12 dec. 2010 2117


© The American Society of Gene & Cell Therapy
NOD T1D Treatment With mRNA Electroporated DCs

of administration, may account for the biological effect of delayed limited and specific lymphoid tissues.5 Overall, it appears that the
disease ­progression  which was ­comparable to that of ltDCs. effect of eDC/IL-4 was more pronounced in the spleen than in the
Interestingly, electroporated DCs, in contrast to unmodified PLNs, possibly because DCs migrate faster to the spleen than to
and transduced DCs, may become more refractory to producing the PLNs5 and can therefore secrete more IL-4 before the mRNA
IL-12 under certain maturation conditions.12,22 This phenomenon get depleted and the expression shut down. In other published
is worth further exploration, as impaired induction of IL-12 can studies,7 we have determined that the efficacy of ltDC/IL-4 was
enhance the effect of IL-4 expressed by therapeutic DCs. more consistent using the intravenous route (which allows spleen
Although various studies have reported an effect of DCs alone homing) compared to the intraperitoneal route (which improves
in preventing diabetes when injected into young NOD mice, DCs PLN homing, but prevents spleen homing), suggesting that hom-
transduced by adenovirus or lentivirus to express IL-4, but not ing to the spleen may be important in addition to homing to the
untransduced DCs, were able to significantly delay and/or prevent PLNs. We have previously reported that certain changes in gene
the onset of disease when injected in older prediabetic mice.6,7 One expression affect the PLNs very early in young NOD mice, but
major concern with the electroporation method was the transient appear in the spleen only around 12 weeks of age.26 Because, we
nature of IL-4 expression (limited to 24 hours). However, we have treated mice at 12 weeks of age, it is possible that homing to both
also reported that DCs are detected in the spleen within 4 hours the spleen and the PLNs is desired.
and in the PLNs between 7 and 10 hours after injection,5 thus Although the therapeutic effect of eDC/IL-4 may not appear
sufficiently early to expect IL-4 to still be produced in the ­tissue as effective as other therapies being investigated in NOD mice,
following transfer. We showed that eDC/IL-4 had a significant including treatment with anti-CD3 antibodies or ex vivo amplified
effect in preventing onset of disease, whereas control eDC/GFP and stimulated Tregs, mRNA electroporation of DCs offers poten-
did not, suggesting that IL-4, even transiently expressed, played tial flexibility and room for improvement. First, different types of
an important role. Thanks to recent developments, T1D can be mRNA may be combined. For cancer immunotherapy, a combina-
diagnosed earlier, allowing prevention strategies to be considered tion of mRNA encoding both tumor antigens and factors enhanc-
for patients with a high risk of disease (family history, permissive ing DC maturity and stimulatory function has been used.13,27,28 In
major histocompatibility complex, and the presence of relevant the case of autoimmune diseases, retaining DC immaturity and/or
autoantibodies in the serum),23 and treatments that are still effec- inducing tolerogenic properties is often preferable. In that respect,
tive at a late stage before the onset of overt hyperglycemia deserve electroporation is a safer choice over transduction with viral vec-
more attention. Currently, most efforts are still focused on treating tors, which tends to increase DC maturity. Although IL-4 is an
recently diagnosed patients, and most successful therapies in the effective cytokine in the treatment of autoimmune diabetes, other
NOD mouse are only seriously considered if they are also able to molecules may be coexpressed to improve the effects of the DCs
reverse overt hyperglycemia. We showed that a single injection by, for example, preventing the maturation of the DCs, express-
of eDC/IL-4 after onset of hyperglycemia had a significant effect, ing inhibitory molecules (e.g., programmed death ligand-1,
decreasing the severity of disease in a third of the mice for pro- indoleamine 2,3-dioxygenase), enhancing trafficking to lymphoid
longed periods of time. The treatment was most effective in mice or inflamed tissues (via certain chemokine receptors and adhesion
that had less severe hyperglycemia. In addition, older mice were molecules) or inducing more antigen-specific responses (expres-
more likely to respond to the treatment, possibly a reflection of sion of peptides recognized by self-reactive T cells). Secondly,
late onset being the result of a more slowly progressing and less the effect of both prophylactic and therapeutic treatments was
aggressive disease. achieved with a single injection of eDC/IL-4, but it is possible
We have previously shown that the homing of DCs to target that several treatments with eDC/IL-4 at different intervals may
tissues results in increased expression of CD25 and CD69 on increase the duration of the effect. To this end, cryopreservation
CD4+ T cells.5 In this study, with a fivefold lower number of DCs of DCs will be required as adoptive cellular therapy with DCs
used for therapy, we continued to observe induction of CD25hi ­represents a personalized medicine in which the cells cannot be
cells that express Foxp3. This, combined with the lack of CD69 continuously prepared. We have started to test cryopreserved
upregulation, suggests that induction of Tregs may contribute eDC/IL-4 in therapy settings, and found that a similar proportion
to the therapeutic effect of the DCs. The fact that splenocytes of mice responded to treatment, but with a more transient rever-
from eDC/IL-4-treated mice lose their ability to transfer disease sal of hyperglycemia. Although optimization will be required to
in NOD.SCID mice is further suggestive that Tregs have been improve recovery, viability, and function of cryopreserved murine
induced, but an enhanced deletion of autoreactive T cell within bone marrow-derived eDC/IL-4, there is evidence that human
3  days of treatment cannot be ruled out. Although CD25hi cells (monocyte-derived) DCs retain excellent viability and function
were induced by all electroporated DCs, the presence of IL-4 following recovery from cryopreservation.11,29
boosted their frequency, particularly in the spleen. Such beneficial In conclusion, we have demonstrated that electroporation
effect of IL-4 on Tregs has been reported before.24,25 Another aspect with translationally enhanced mRNA can be used to modify DCs
of the therapeutic effect of eDC/IL-4 is a significant shift from to express immunoregulatory products for adoptive cellular gene
Th1 to Th2 compared to eDC/GFP, characterized by a reduced therapy of autoimmune diseases. In the case of IL-4, which is
Th1 response and/or an increased Th2 response, depending on secreted and not retained in the cells, the timeline of expression is
the tissue ­analyzed, as well as significantly greater levels of IL-10 short and lasts no longer than 24 hours. Nevertheless, this short
produced. The lack of response in the cervical lymph nodes of “pulse” of IL-4 delivered in vivo was sufficient to induce thera-
eDC/IL-4-treated mice confirms that therapeutic DCs only target peutic effects in NOD mice, both in prevention and treatment of

2118 www.moleculartherapy.org vol. 18 no. 12 dec. 2010


© The American Society of Gene & Cell Therapy
NOD T1D Treatment With mRNA Electroporated DCs

disease, which were in part ascribed to Treg induction and helper l-glutamine, 100 U/ml penicillin, 100 μg/ml ­streptomycin, 0.1 mmol/l
T cell skewing. Modification by mRNA electroporation is a flex- nonessential amino acids, 1 mmol/l sodium pyruvate, 14.3 μmol/l
ible method that allows for coexpression of multiple products that β-mercaptoethanol) in presence of recombinant mouse granulocyte-
could be used to expand current therapeutic options. macrophage colony-stimulating factor and IL-4 (Peprotech, Rocky Hill,
NJ) at 10 ng/ml each. On day 5, cells were infected with lentiviral ­particles
(multiplicity of infection = 15–20) in presence of 10 μg/ml protamine sul-
Materials and Methods fate (Sigma, St Louis, MO). After 16–24 hours incubation with virus, the
Mice. Female NOD and NOD.SCID mice were purchased from the Jackson medium was changed. Infected and noninfected DCs were collected on
Laboratories (Bar Harbor, ME), and kept in our animal facility under spe- day 7. Noninfected DCs were washed three times in OptiMEM and elec-
cific pathogen free conditions. Bone marrow donors were 8–10 weeks old, troporated in a 4 mm cuvette (5 × 106 DCs in 200 μl OptiMEM with 30 μg
while recipient mice were used as nondiabetic at 12 weeks of age (pre- mRNA) on a Gene Pulser Xcell II electroporator (Bio-rad, Hercules, CA)
vention experiments), or as recent onset hyperglycemic between 11 and using optimized parameters (300 V, 150 μF, 100 Ω).
30 weeks of age. NOD.SCID mice were adoptively transferred at 10 weeks
of age. All manipulations were approved by the Stanford Administrative Treatments and monitoring. 1 × 106 DCs (in 200 μl PBS) were injected
Panel on Laboratory Animal Care. intravenously into 12-week-old nondiabetic NOD mice (prevention) or
11–30-week-old NOD mice with recent onset hyperglycemia. In preven-
Subcloning of mIL4Rot6 for RNA generation. mIL4 coding sequence was
tion studies, blood glucose was measured weekly and animals with levels
subcloned from pHR-IL4IG plasmid7 into pGem 4Z 64T ­plasmid  using >250 mg/dl over two consecutive bleedings were considered diabetic. In
SmaI restriction sites. Translation enhancer element from the 3′ untrans- therapy studies, blood glucose was monitored weekly. Mice with high-
lated region of rotavirus gene 6 mRNA (rot 6 element)30 was PCR ampli- blood glucose (250–500 mg/dl) were bled again 1 or 2 days later to confirm
fied from synthetically generated construct (Blue Heron Biotechnology, hyperglycemia and immediately treated. Treated mice were monitored
Bothell, WA) encoding the rot6 sequence using primers forward 5′-ACT twice weekly and animals with levels >500 mg/dl over two consecutive
GCTCGAGGACCAAGCTAACAACTTGG-3′ and reverse 5′-ACTGTTA bleedings were sacrificed. In transfer experiments, splenocytes obtained
ATTAAGGTCACATCCTCTCACTATACC-3′ containing XhoI and PacI 3 days after treatment were injected intravenously into 10-week-old NOD.
recognition sites indicated in italics, respectively. The amplified PCR frag- SCID mice (3 × 106 cells per recipient). Recipient mice were followed
ment containing rot 6 element was subcloned downstream of stop codon every other week until the first onset of disease, and weekly thereafter.
of mIL4 in pGem4Z64T using XhoI and PacI restriction sites. The final Mice were considered diabetic using the same criteria as in prevention
construct was verified by sequencing through the coding region of mIL4 studies.
and junction to pGem4Z64T plasmid.
Flow cytometric analysis. DCs were analyzed for GFP and surface mark-
Generation of the GFP and mIL4Rot6 type I cap RNAs. Plasmid
ers expression (anti-CD11b, CD11c and I-Ak (also stains I-Ag7) were from
pGem4Z64TmIL4Rot6 was purified using QiaFilter maxiprep kit (Qiagen,
BD Biosciences (San Jose, CA); anti-CD40, CD80, CD86, CD8a and SIRPα
Valencia, CA) and linearized with SpeI. Twenty-five microgram of puri-
were from eBioscience). For intracellular staining of IL-4, transduced and
fied linearized template was used to establish 1-ml transcription reaction
electroporated DCs were recultured for 3–24 hours in complete medium
using T7 Flash kit according to manufacturing instruction (Epicentre
without cytokines and with GolgiStop (BD Biosciences) added in the last
Biotechnologies, Madison, WI). The in vitro transcription was conducted
3–4 hours, processed with BD Fix/Perm kit (BD Biosciences) and stained
for 2 hours at 37 °C. After digestion with added 50 μl of DNAseI for addi-
using allophycocyanin-conjugated anti-IL-4 (eBioscience). T cells were
tional 30 minutes at 37 °C, the uncapped RNA was purified using RNeasy
stained for CD4, CD8a, CD25, CD44, CD69, and Foxp3 using antibodies
maxi columns (Qiagen). The purified uncapped RNA was capped in the
from BD Biosciences and eBioscience. Fix/Perm kit from eBioscience was
post-transcriptional capping reaction for 1 hour at 37 °C using Script
used for Foxp3 intracellular staining. All cells were analyzed on LSR1 (BD
Cap Type I Capping kit scaled up to 3,340 μl reaction volume (Epicentre
Biosciences) and data were computed using the FlowJo software (Tree Star,
Biotechnologies). The polyadenylation reaction was carried out using
Ashland, OR).
A-plus PolyA Tailing kit (Epicentre Biotechnologies). The unpurified cap-
ping reaction was adjusted to 6,664 μl with water, 10× A-plus reaction buffer ELISA. DCs were washed and replated at different numbers and several
and ATP substrate according to manufacturer’s instructions and incubated replicates in complete RPMI without granulocyte-macrophage colony-
for 45 minutes at 37 °C swirling the reaction every 15 minutes. The final stimulating factor or IL-4. Supernatant was collected at various time points
polyadenylated RNA modified with type I cap and ~200 polyA tail was ranging from 30 minutes to 24 hours. ELISA was performed using stan-
purified using RNeasy maxi columns (Qiagen). The length of polyA tail dard protocol and reagents from BD Biosciences (anti-IL-4 antibody pair),
was measured by comparing relative migration of unpolyadenylated and Peprotech (IL-4 standard) and Sigma (ExtrAvidin peroxidase and TMB
polyadenylated mIL4 RNAs on denaturing agarose gel followed by collect- substrate). Data shown are mean ± SD from different number of live cells
ing image with Alphaimager and size analysis using ImageQuant Software plated (5 × 103, 2.5 × 104, 1 × 105). The amount of IL-4 secreted was lin-
(Alpha Innotech, San Leandro, CA). 2.2 mg of RNA eluted in water was early correlated with the number of DCs (R2 > 0.98). For cytokine profile,
adjusted to final concentration of 1 mg/ml and stored frozen in single- splenocytes (1 × 106/well), PLN, or cervical lymph node cells (0.8 × 106/
size aliquots in pyrogen-free RNAse-free Safe Lock tubes (Eppendorf, well) were replated in complete c-RPMI in four replicates. Supernatant
Hamburg, Germany). GFP RNA was generated from pGem4Z/GFP/A64 was ­collected after 36 and 72 hours (one duplicate per time point). ELISA
plasmid as described previously.31 was performed as described above for IL-4, or using ELISA Max kits
(Biolegend, San Diego, CA) for IFN-γ and IL-10.
Lentivirus generation, DC culture, transduction, and electroporation.
Lentiviral particles expressing IL-4 and GFP were generated using the SUPPLEMENTARY MATERIAL
pHR-IL4IG vector as previously described.7 Bone marrow was harvested Figure S1.  Efficiency of electroporation or transduction of DCs.
from the femurs, tibias, and pelvis of female mice. The bone marrow cells Figure S2.  Evolution of blood glucose on a selection of mice treated
were depleted of CD3+, B220+, and Gr-1+ cells on AutoMACS (Miltenyi with eDC/IL-4, showing occasional transient increase of glycemia over
Biotec, Bergisch Gladbach, Germany) using biotinylated antibodies (eBio- 250 mg/dl.
science, San Diego, CA) and antibiotin beads (Miltenyi Biotec), and then Figure S3.  Consistency in the amount of IL-4 secreted by modified
­cultured at 37 °C in complete RPMI medium (10% fetal calf serum, 2 mmol/l DCs in a 24 hours culture.

Molecular Therapy vol. 18 no. 12 dec. 2010 2119


© The American Society of Gene & Cell Therapy
NOD T1D Treatment With mRNA Electroporated DCs

Figure S4.  Factors influencing effectiveness of eDC/IL-4 treatment. 14. Lo, J and Clare-Salzler, MJ (2006). Dendritic cell subsets and type I diabetes: focus
Figure S5.  CD25+ Foxp3+ cells in the PLNs and spleen of treated upon DC-based therapy. Autoimmun Rev 5: 419–423.
15. Trucco, M and Giannoukakis, N (2007). Immunoregulatory dendritic cells to
mice. prevent and reverse new-onset Type 1 diabetes mellitus. Expert Opin Biol Ther 7:
Figure S6.  Activated CD4+ and CD8+ T cells in the PLNs and spleen 951–963.
16. Besin, G, Gaudreau, S, Ménard, M, Guindi, C, Dupuis, G and Amrani, A (2008).
of treated mice. Thymic stromal lymphopoietin and thymic stromal lymphopoietin-conditioned
Figure S7.  Frequency of several DC subsets in the spleen of treated dendritic cells induce regulatory T-cell differentiation and protection of NOD mice
mice. against diabetes. Diabetes 57: 2107–2117.
17. Giannoukakis, N, Phillips, B and Trucco, M (2008). Toward a cure for type 1 diabetes
mellitus: diabetes-suppressive dendritic cells and beyond. Pediatr Diabetes 9(3 Pt 2):
ACKNOWLEDGMENTS 4–13.
D.G.H., I.Y.T., and C.A.N. are employed by Argos Therapeutics. The 18. Kyte, JA, Mu, L, Aamdal, S, Kvalheim, G, Dueland, S, Hauser, M et al. (2006).
Phase I/II trial of melanoma therapy with dendritic cells transfected with autologous
authors also thank Melissa Adam and Aijing Starr, also employed by tumor‑mRNA. Cancer Gene Ther 13: 905–918.
Argos Therapeutics, for preparation of optimized IL-4 cDNA template 19. Van Driessche, A, Van de Velde, AL, Nijs, G, Braeckman, T, Stein, B, De Vries, JM et al.
and for mRNA generation, respectively. This work was supported by (2009). Clinical-grade manufacturing of autologous mature mRNA-electroporated
dendritic cells and safety testing in acute myeloid leukemia patients in a phase I
grants from the National Institutes of Health (DK078123, to C.G.F.), dose‑escalation clinical trial. Cytotherapy 11: 653–668.
the Juvenile Diabetes Research Foundation (3-2005-1019, to R.J.C.) 20. Schuurhuis, DH, Verdijk, P, Schreibelt, G, Aarntzen, EH, Scharenborg, N, de Boer, A
and by a gift from Argos Therapeutics (to C.G.F.). et al. (2009). In situ expression of tumor antigens by messenger RNA-electroporated
dendritic cells in lymph nodes of melanoma patients. Cancer Res 69: 2927–2934.
21. Routy, JP, Boulassel, MR, Yassine-Diab, B, Nicolette, C, Healey, D, Jain, R et al. (2010).
REFERENCES Immunologic activity and safety of autologous HIV RNA-electroporated dendritic
1. Anderson, MS and Bluestone, JA (2005). The NOD mouse: a model of immune cells in HIV-1 infected patients receiving antiretroviral therapy. Clin Immunol 134:
dysregulation. Annu Rev Immunol 23: 447–485. 140–147.
2. Shoda, LK, Young, DL, Ramanujan, S, Whiting, CC, Atkinson, MA, Bluestone, JA et al. 22. Dullaers, M, Breckpot, K, Van Meirvenne, S, Bonehill, A, Tuyaerts, S, Michiels, A et al.
(2005). A comprehensive review of interventions in the NOD mouse and implications (2004). Side-by-side comparison of lentivirally transduced and mRNA-electroporated
for translation. Immunity 23: 115–126. dendritic cells: implications for cancer immunotherapy protocols. Mol Ther 10:
3. Smits, HH, de Jong, EC, Wierenga, EA and Kapsenberg, ML (2005). Different faces of 768–779.
regulatory DCs in homeostasis and immunity. Trends Immunol 26: 123–129. 23. Sherr, J, Sosenko, J, Skyler, JS and Herold, KC (2008). Prevention of type 1 diabetes:
4. Morelli, AE and Thomson, AW (2007). Tolerogenic dendritic cells and the quest for the time has come. Nat Clin Pract Endocrinol Metab 4: 334–343.
transplant tolerance. Nat Rev Immunol 7: 610–621. 24. Skapenko, A, Kalden, JR, Lipsky, PE and Schulze-Koops, H (2005). The IL-4 receptor
5. Creusot, RJ, Yaghoubi, SS, Chang, P, Chia, J, Contag, CH, Gambhir, SS et al. (2009). alpha-chain-binding cytokines, IL-4 and IL-13, induce forkhead box P3-expressing
Lymphoid-tissue-specific homing of bone-marrow-derived dendritic cells. Blood 113: CD25+CD4+ regulatory T cells from CD25-CD4+ precursors. J Immunol 175:
6638–6647. 6107–6116.
6. Feili-Hariri, M, Falkner, DH, Gambotto, A, Papworth, GD, Watkins, SC, Robbins, PD 25. Pillai, AB, George, TI, Dutt, S and Strober, S (2009). Host natural killer T cells induce
et al. (2003). Dendritic cells transduced to express interleukin-4 prevent diabetes in an interleukin-4-dependent expansion of donor CD4+CD25+Foxp3+ T regulatory cells
nonobese diabetic mice with advanced insulitis. Hum Gene Ther 14: 13–23. that protects against graft-versus-host disease. Blood 113: 4458–4467.
7. Creusot, RJ, Yaghoubi, SS, Kodama, K, Dang, DN, Dang, VH, Breckpot, K et al. (2008). 26. Kodama, K, Butte, AJ, Creusot, RJ, Su, L, Sheng, D, Hartnett, M et al. (2008). Tissue-
Tissue-targeted therapy of autoimmune diabetes using dendritic cells transduced to and age-specific changes in gene expression during disease induction and progression
express IL-4 in NOD mice. Clin Immunol 127: 176–187. in NOD mice. Clin Immunol 129: 195–201.
8. Kim, SH, Kim, S, Evans, CH, Ghivizzani, SC, Oligino, T and Robbins, PD (2001). 27. Michiels, A, Breckpot, K, Corthals, J, Tuyaerts, S, Bonehill, A, Heirman, C et al.
Effective treatment of established murine collagen-induced arthritis by systemic (2006). Induction of antigen-specific CD8+ cytotoxic T cells by dendritic cells
administration of dendritic cells genetically modified to express IL-4. J Immunol 166: co‑electroporated with a dsRNA analogue and tumor antigen mRNA. Gene Ther 13:
3499–3505. 1027–1036.
9. Morita, Y, Yang, J, Gupta, R, Shimizu, K, Shelden, EA, Endres, J et al. (2001). Dendritic 28. Bonehill, A, Tuyaerts, S, Van Nuffel, AM, Heirman, C, Bos, TJ, Fostier, K et al.
cells genetically engineered to express IL-4 inhibit murine collagen-induced arthritis. J (2008). Enhancing the T-cell stimulatory capacity of human dendritic cells by co-
Clin Invest 107: 1275–1284. electroporation with CD40L, CD70 and constitutively active TLR4 encoding mRNA.
10. Michiels, A, Tuyaerts, S, Bonehill, A, Corthals, J, Breckpot, K, Heirman, C et al. (2005). Mol Ther 16: 1170–1180.
Electroporation of immature and mature dendritic cells: implications for dendritic cell- 29. Ponsaerts, P, Van Tendeloo, VF, Cools, N, Van Driessche, A, Lardon, F, Nijs, G et al.
based vaccines. Gene Ther 12: 772–782. (2002). mRNA-electroporated mature dendritic cells retain transgene expression,
11. Van Tendeloo, VF, Ponsaerts, P and Berneman, ZN (2007). mRNA-based gene transfer phenotypical properties and stimulatory capacity after cryopreservation. Leukemia 16:
as a tool for gene and cell therapy. Curr Opin Mol Ther 9: 423–431. 1324–1330.
12. Calderhead, DM, DeBenedette, MA, Ketteringham, H, Gamble, AH, Horvatinovich, JM, 30. Yang, AD, Barro, M, Gorziglia, MI and Patton, JT (2004). Translation enhancer in the
Tcherepanova, IY et al. (2008). Cytokine maturation followed by CD40L mRNA 3’-untranslated region of rotavirus gene 6 mRNA promotes expression of the major
electroporation results in a clinically relevant dendritic cell product capable of inducing capsid protein VP6. Arch Virol 149: 303–321.
a potent proinflammatory CTL response. J Immunother 31: 731–741. 31. Boczkowski, D, Nair, SK, Nam, JH, Lyerly, HK and Gilboa, E (2000). Induction of tumor
13. Feili-Hariri, M, Flores, RR, Vasquez, AC and Morel, PA (2006). Dendritic cell immunity and cytotoxic T lymphocyte responses using dendritic cells transfected with
immunotherapy for autoimmune diabetes. Immunol Res 36: 167–173. messenger RNA amplified from tumor cells. Cancer Res 60: 1028–1034.

2120 www.moleculartherapy.org vol. 18 no. 12 dec. 2010

You might also like