Vaccination with in vitro transcribed RNA encoding tumor antigens is an emerging approach in cancer immunotherapy. Attempting to further improve RNA vaccine efficacy, we have explored combining RNA with immunomodulators such as rapamycin. Rapamycin, the inhibitor of mTOR, was used originally for immunosuppression. Recent reports in mouse systems, however, suggest that mTOR inhibition may enhance the formation and differentiation of the memory CD8+ T-cell pool. Because memory T-cell formation is critical to the outcome of vaccination aproaches, we studied the impact of rapamycin on the in vivo primed RNA vaccine-induced immune response using the chicken ovalbumin-expressing B16 melanoma model in C57BL/6 mice. Our data show that treatment with rapamycin at the effector-to-memory transition phase skews the vaccine-induced immune response toward the formation of a quantitatively and qualitatively superior memory pool and results in a better recall response. Tumor-infiltrating immune cells from these mice display a favorable ratio of effector versus suppressor cell populations. Survival of mice treated with the combined regimen of RNA vaccination with rapamycin is significantly longer (91.5 days) than that in the control groups receiving only one of these compounds (32 and 46 days, respectively). Our findings indicate that rapamycin enhances therapeutic efficacy of antigen-specific CD8+ T cells induced by RNA vaccination, and we propose further clinical exploration of rapamycin as a component of immunotherapeutic regimens. Cancer Immunol Res; 1(6); 386–92. ©2013 AACR.

Direct application of in vitro transcribed antigen-encoding RNA is emerging as a promising approach for antitumor vaccination (1, 2). RNA delivers the complete antigen, is selectively internalized by professional antigen-presenting cells (3), and has an intrinsic adjuvant activity. RNA does not integrate into the genome and with the transient expression of the encoded antigens allows better control of immune responses. The capability of RNA vaccines to confer antitumor immunity has been shown in model systems (4–6). Recombinant RNA is easy to produce in large amounts and high purity by in vitro transcription. These advantages prompted clinical testing of vaccination with antigen-encoding RNA, which has demonstrated excellent safety and provided promising data on immunogenicity in patients with cancer (7).

As reported previously, our group has developed pharmacologically optimized RNA with improved stability and translational performance; the encoded antigen was efficiently presented on both MHC class I and class II molecules (8, 9). Injection of such optimized RNA into lymph nodes propagates a proinflammatory microenvironment, resulting in de novo priming and efficient expansion of antigen-specific CD8+ and CD4+ T cells (5). When tumor-bearing mice were immunized with this RNA vaccine, potent antitumor immunity was elicited, resulting in significant survival benefit (5). We improved the potency of this vaccine platform several logs by molecularly modifying the RNA backbone; our current strategy is to identify synergistic immunomodulatory compounds for combination therapy (10). As CD8+ T memory cells are critical for efficient and sustained immunity against cancer, compounds capable of tuning vaccine-induced memory response are of particular interest.

The mTOR has a pivotal role in the modulation of T-cell immune responses. mTOR inhibition was used as a treatment against allo-rejection reactions in solid organ transplantation and in a number of autoimmune/inflammatory diseases. The recent observations on adoptive T-cell transfer therapy against viral and bacterial infection in mouse model systems (11, 12) have confirmed mTOR as a potential gatekeeper for the differentiation of memory CD8+ T cells. mTOR inhibiton by rapamycin has been shown to enhance the differentiation and the quantity of memory CD8+ T cells (11). Rao and colleagues proposed that the enhanced memory CD8+ T-cell differentiation was due to decreased expression of the transcription factor T-bet and persistent expression of Eomesodermin (13). This observation has generated considerable interest in the use of rapamycin for shaping antigen-specific CD8+ T-cell responses.

The objective of the current study was to investigate the effect of rapamycin on T-cell responses induced by intranodal RNA vaccination. To this end, mTOR inhibition in combination with RNA vaccination was assessed for the first time in the de novo priming setting in a mouse tumor treatment model. To our knowledge, this is the first study in which rapamycin effects on immune cells in the tumor microenvironment were investigated.

This report presents an approach for enhancing RNA vaccination with the multifaceted effects of rapamycin on T-cell–mediated immune responses.

Animals

Six- to 8-week-old female C57BL/6 mice were purchased from Charles River. Animals were kept in accordance with federal and state policies on animal research at the University of Mainz (Mainz, Germany).

Cells

The B16-F10 melanoma cell line expressing the chicken ovalbumin gene (B16-OVA) was a kind gift from H.J. Schild (Institute for Immunology, Mainz, Germany). Cell banks were created immediately upon receipt of the cell line and early passages (third and fourth) of cells were used for tumor experiments. Cells were tested for Mycoplasma every 3 months. Reauthentification of cells was not performed since receipt.

RNA vaccine and synthetic peptides

The RNA vaccine coding for H2-Kb-restricted dominant OVA257-264 epitope (SIINFEKL) from chicken Ovalbumin was generated by in vitro transcription as described previously from the plasmid vector pST1-sec-SIINFEKL-MITD-2hBgUTR-A120 (7, 8). This vector construct features a signal sequence for routing to the endoplasmic reticulum and the MHC class I transmembrane and cytoplasmic domains to improve MHC class I and MHC class II presentation as well as optimized 3′UTR and polyA tail for improved stability and translational efficiency (8, 9). The synthetic SIINFEKL peptide was purchased from Jerini Peptide Technologies.

Immunizations and rapamycin treatment

For intranodal RNA immunization, mice were anesthetized and the inguinal lymph node was surgically exposed. Twenty micrograms of RNA formulated in 10 μL RNase-free water (Ambion) was injected slowly and the wound was closed. In one experiment, mice were boosted on day 35 with s.c. administered SIINFEKL peptide (200 μg) and CpG 1826 (25 μg; MWG Biotech). Rapamycin (Wyeth-Ayerst) was administered intraperitoneally. Mice were treated either during the effector-to-memory transition phase (day 10–31 post RNA vaccination or d11-33 post tumor implantation) with a daily high dose of 600 μg/kg body weight (11) or during the T-cell expansion phase (day 2–8 in relation to RNA vaccinations) with a low dose of 75 μg/kg body weight (11). Control mice received Phosphal 50 PG (PG50; Phospholipid GmbH).

Tumor experiments

A total of 2 × 105 B16-OVA tumor cells were inoculated s.c. into the flanks of C57BL/6 mice (day 0). Mice received three intranodal RNA immunizations at 3-day intervals. Early tumor models (immunizations on days 3, 6, and 9) as well as advanced tumor models (immunization on days 7, 10, and 13) were explored. Rapamycin was administered daily at the effector-to-memory transition phase starting from day 11 in the early and day 15 in the advanced tumor models. Tumor sizes were measured every 3 days. Mice were sacrificed when the tumor diameter reached 15 mm.

Flow cytometric analysis

All monoclonal antibodies for flow cytometric analysis were purchased from BD Bioscience, except for anti-KLRG1, which was obtained from eBioscience San Diego. Phenotyping of peripheral blood cells and MHC/tetramer-staining were performed as described previously (8). Regulatory T cells (Treg) were detected by using the FoxP3 staining kit (eBioscience) according to the manufacturer's instructions.

Tumor-infiltrating leukocytes (TIL) were obtained by mechanical disruption of the tumor tissue. Phenotyping by flow cytometry was performed with the following markers after gating CD45+ leukocytes: myeloid-derived suppressor cells (MDSC; CD11b+, Gr1low/high), Treg (CD4+, CD25+, FoxP3+), NK cells (CD3, NK1.1+), CD8+ T cells (CD3+, CD8+), and SIINFEKL+ CD8+ T cells (CD3+, CD8+, tetramer+).

Flow cytometric data were acquired on a FACSCalibur and FACS Canto analytic flow cytometers (BD Bioscience) and analyzed by FlowJo software (Tree Star).

Statistical analysis

Statistical analysis was performed by GraphPad Prism 5 software employing unpaired two-tailed Student t test, one-way ANOVA test with Bonferroni comparison post-test, and Log rank (Mantel–Cox) test. Values of P < 0.05 were considered statistically significant. Data are presented as mean ± SEM.

RNA vaccination combined with mTOR inhibition enhances differentiation of antigen-specific memory CD8+ T cells

Once a CD8+ T-cell is activated by antigen recognition, it proliferates and differentiates into an effector cell. Peaking of the CD8+ T-cell response is followed by the contraction phase, when most of the effector cells die and the remaining become long-lived memory cells (14). High-dose rapamycin administered during the contraction period had been shown in an LCMV infection model to facilitate transition of adoptively transferred effector CD8+ T cells to memory T cells, whereas its administration in the priming phase was observed to deplete CD8+ T cells (11).

On the basis of those findings, we studied immune responses induced by intranodal RNA vaccination in conjunction with mTOR inhibition applied during the contraction phase. Naïve mice received repetitive intranodal immunizations (days 0, 3, and 6) with RNA coding for the SIINFEKL epitope of chicken ovalbumin (SIINFEKL-RNA) followed by rapamycin treatment (days 10–31; Fig. 1A, left). We monitored the phenotype of antigen-specific CD8+ T cells in the peripheral blood using markers of memory CD8+ T cells such as CD127 (also known as interleukin-7 receptor; ref. 15) and the inhibitory killer cell lectin–like receptor G1 (KLRG1; ref. 15).

Figure 1.

Rapamycin treatment during the effector-to-memory transition phase promotes generation of memory CD8+ T cells. A–D, C57BL/6 mice (n = 10) were immunized intranodally with SIINFEKL-RNA (20 μg) on days 0, 3, and 6. Rapamycin (Rapa; 600 μg/kg body weight) or PG50 (control) were injected intraperitoneally daily from day 10 to day 31 (effector-to-memory transition phase). A, Treatment plan and frequencies of SIINFEKL-specific CD8+ T cells (gated in total CD8+ T cells) assessed via tetramer staining in the peripheral blood over time. B, Frequencies of SIINFEKL-specific short-lived effector CD8+ T cells (KLRG1+) and memory CD8+ T cell precursors (KLRG1CD127+; gated in SIINFEKL-specific CD8+ T cells) monitored in the peripheral blood. Representative histograms of KLRG1 expression on these cells are also shown for day 28. C, Frequencies of SIINFEKL-specific memory CD8+ T precursors (CD44+KLRG1CD127+; gated in SIINFEKL-specific CD8+ T cells) in the peripheral blood on day 28. D, Mice were boosted with s.c. administration of SIINFEKL peptide (200 μg) and CpG (25 μg) on day 35, and SIINFEKL-specific CD8+ T cells in the peripheral blood were quantified by tetramer staining 6 days later. *, P < 0.05; **, P < 0.01; ***, P < 0.001 [Student t test (B, C, and D)].

Figure 1.

Rapamycin treatment during the effector-to-memory transition phase promotes generation of memory CD8+ T cells. A–D, C57BL/6 mice (n = 10) were immunized intranodally with SIINFEKL-RNA (20 μg) on days 0, 3, and 6. Rapamycin (Rapa; 600 μg/kg body weight) or PG50 (control) were injected intraperitoneally daily from day 10 to day 31 (effector-to-memory transition phase). A, Treatment plan and frequencies of SIINFEKL-specific CD8+ T cells (gated in total CD8+ T cells) assessed via tetramer staining in the peripheral blood over time. B, Frequencies of SIINFEKL-specific short-lived effector CD8+ T cells (KLRG1+) and memory CD8+ T cell precursors (KLRG1CD127+; gated in SIINFEKL-specific CD8+ T cells) monitored in the peripheral blood. Representative histograms of KLRG1 expression on these cells are also shown for day 28. C, Frequencies of SIINFEKL-specific memory CD8+ T precursors (CD44+KLRG1CD127+; gated in SIINFEKL-specific CD8+ T cells) in the peripheral blood on day 28. D, Mice were boosted with s.c. administration of SIINFEKL peptide (200 μg) and CpG (25 μg) on day 35, and SIINFEKL-specific CD8+ T cells in the peripheral blood were quantified by tetramer staining 6 days later. *, P < 0.05; **, P < 0.01; ***, P < 0.001 [Student t test (B, C, and D)].

Close modal

We found that the frequencies (Fig. 1A, right) and absolute numbers (Supplementary Fig. S1A) of RNA vaccine-induced antigen-specific CD8+ T cells were not altered by rapamycin as comparable with those of the PG50-treated control group (Fig. 1A). When we assessed CD8+ T-cell subpopulations, however, we found that a lower frequency of short-lived antigen-specific effector cells (KLRG1+) resulted from RNA vaccination in combination with rapamycin (Fig. 1B, left graph). In contrast to this skewing of the effector pool, the frequency (Fig. 1B, right graph) and absolute number (Supplementary Fig. S1B) of the antigen-specific memory CD8+ T-cell precursors (KLRG1CD127+) were higher in these mice. In the advanced tumor B16-OVA melanoma model, similar results were obtained in the blood and spleen of immunized mice treated with rapamycin (Supplementary Fig. S1C). Moreover, antigen-specific effector T cells of rapamycin-treated mice expressed CD44 at a higher frequency (Fig. 1C), which is known to contribute to the overall cellular fitness and survival of CD8+ T cells (15). In line with these findings, CD8+ T-cell memory precursors of rapamycin-treated mice were also superior with regard to their proliferation rate in response to boosting with SIINFEKL peptide on day 35 (Fig. 1D and Supplementary Fig. S1D). This was not the case, when a low dose of rapamycin was administered during the priming and the expansion phase (days 2–8) of vaccine-induced antigen-specific immune response (Supplementary Fig. S2A). The frequency (Supplementary Fig. S2B) and the absolute number (Supplementary Fig. S2C) of antigen-specific CD8+ T cells were markedly lower on day 10 in the peripheral blood of rapamycin-treated mice. We ruled out a general suppression of the hematopoietic system by rapamycin, as hemogram parameters are not altered with rapamycin treatment (Supplementary Fig. S3). Previous data, which demonstrated that mTOR inhibition may have negative effects on priming by suppressing the in vivo differentiation and antigen presentation capacity of dendritic cells (16), may provide an explanation for this observation. Moreover, it was shown that factors like antigen format and antigen strength might influence the outcome of rapamycin in combination with vaccines, suggesting that rapamycin treatment protocols require specific tuning for each vaccine setting (17).

In summary, our data show that targeted modulation with rapamycin treatment during the effector-to-memory transition phase shapes immune responses induced by RNA vaccines toward the formation of a quantitatively and qualitatively superior memory pool of antigen-specific T cells.

mTOR inhibition augments the therapeutic effect of antitumor RNA vaccination

Next, we wanted to know whether immunomodulation by rapamycin treatment might enhance the efficacy of RNA vaccination in vivo.

It had been reported that the modest effects of recombinant protein-based tumor antigen vaccines in RENCA renal cell cancer and B16 melanoma models can be augmented by the mTOR inhibitor temsirolimus (18). However, the study used adoptively transferred antigen-specific T cells and therefore only indirect evidence for the modulation of memory T cells was available.

We have reported that intranodal vaccination with SIINFEKL-RNA could elicit strong protective and therapeutic antitumor immune responses in C57BL/6 mice against B16 melanoma cells expressing chicken ovalbumin (B16-OVA; ref. 5). We used this tumor model to assess whether immunomodulation by the combined regimen of RNA vaccine with rapamycin affects the growth kinetics of tumors and the survival of the animals.

Mice were implanted with B16-OVA cells and 3 days later were immunized with SIINFEKL-RNA for three cycles at 3-day intervals (i.e., on days 3, 6, and 9). On day 10, all mice with similar antigen-specific CD8+ T-cell frequency (Supplementary Fig. S4) were divided into two groups for treatment with or without rapamycin during the T-cell effector-to-memory transition phase from days 11 to 33.

Consistent with our previous reports, immunization with the RNA vaccine alone had a significant effect on tumor growth compared with the nonimmunized control groups (Fig. 2A; ref. 5). When rapamycin was added, the growth reduction was even more profound (Fig. 2A). Tumor latency, the period of time required to reach a defined tumor volume (300 mm3; ref. 19), was significantly prolonged from 34 and 32 days in the group that received either RNA vaccination or rapamycin treatment alone, respectively, to 61 days in the group that received both the RNA vaccine and the rapamycin treatment (Fig. 2B and Table 1). The inhibition of tumor growth translated to a significantly longer survival for the mice (Fig. 2C). The combination of RNA vaccination with rapamycin treatment extended the median survival from 32 and 46 days in the rapamycin- or RNA-treated groups, respectively, to 91.5 days in the group treated with the combination (Table 1). In an advanced B16-OVA model in which the RNA immunization was initiated 7 days after tumor inoculation, the superiority of RNA vaccination was confirmed (Fig. 2D).

Figure 2.

Rapamycin treatment improves the therapeutic efficacy of antitumor RNA vaccination in early and advanced tumor models. A–C, syngeneic tumor cells (2 × 105 B16-OVA/mouse) were injected s.c. into C57BL/6 mice (n = 10) on day 0. Mice were immunized intranodally three times using SIINFEKL-RNA (20 μg) (days 3, 6, and 9). From day 11 to day 33, mice received daily intraperitoneal injections of rapamycin (RNA + Rapa) or PG50 (RNA). Additional control groups (n = 10 mice/group) were treated with rapamycin (Rapa) or PG50 (control) alone without RNA immunization. A, tumor growth curves (dotted line shows 300 mm3 tumor volume and numbers in parentheses represent number of tumor-bearing mice/number of all mice in that group). B, tumor latency defined as time (in days) to reach 300 mm3 tumor volume. C, survival curves (dotted line shows median survival) of all groups described above. D, syngeneic tumor cells (2 × 105 B16-OVA/mouse) were injected s.c. into C57BL/6 mice (n = 15) on d0. Mice were immunized intranodally three times using SIINFEKL-RNA (20 μg, days 7, 10, and 13). From day 15 to day 37, mice received daily intraperitoneal injections of rapamycin (RNA + Rapa),PG50 (RNA) or controls as described above. Survival curves of advanced melanoma model were presented. **, P < 0.01; ***, P < 0.001 [one-way ANOVA test with Bonferroni comparison post-test (C) and log-rank (Mantel–Cox) test (D)].

Figure 2.

Rapamycin treatment improves the therapeutic efficacy of antitumor RNA vaccination in early and advanced tumor models. A–C, syngeneic tumor cells (2 × 105 B16-OVA/mouse) were injected s.c. into C57BL/6 mice (n = 10) on day 0. Mice were immunized intranodally three times using SIINFEKL-RNA (20 μg) (days 3, 6, and 9). From day 11 to day 33, mice received daily intraperitoneal injections of rapamycin (RNA + Rapa) or PG50 (RNA). Additional control groups (n = 10 mice/group) were treated with rapamycin (Rapa) or PG50 (control) alone without RNA immunization. A, tumor growth curves (dotted line shows 300 mm3 tumor volume and numbers in parentheses represent number of tumor-bearing mice/number of all mice in that group). B, tumor latency defined as time (in days) to reach 300 mm3 tumor volume. C, survival curves (dotted line shows median survival) of all groups described above. D, syngeneic tumor cells (2 × 105 B16-OVA/mouse) were injected s.c. into C57BL/6 mice (n = 15) on d0. Mice were immunized intranodally three times using SIINFEKL-RNA (20 μg, days 7, 10, and 13). From day 15 to day 37, mice received daily intraperitoneal injections of rapamycin (RNA + Rapa),PG50 (RNA) or controls as described above. Survival curves of advanced melanoma model were presented. **, P < 0.01; ***, P < 0.001 [one-way ANOVA test with Bonferroni comparison post-test (C) and log-rank (Mantel–Cox) test (D)].

Close modal
Table 1.

Mean tumor latency and median survival of groups shown in Fig. 2B and Fig. 2C 

TreatmentMean tumor latency, dMedian survival, d
Control 24.2 23.5 
Rapamycin 32.4 32 
RNA 34.1 46 
RNA + rapamycin 61.5 91.5 
TreatmentMean tumor latency, dMedian survival, d
Control 24.2 23.5 
Rapamycin 32.4 32 
RNA 34.1 46 
RNA + rapamycin 61.5 91.5 

Our findings show that the modulation of RNA vaccination-induced immune responses by rapamycin translates into superior antitumor effects.

Increased tumor-infiltrating leukocytes and decreased MDSC frequencies are observed after RNA-rapamycin combination therapy

We investigated the composition of immune effector cell within tumors of treated mice. As described in Fig. 2A, tumors were harvested on day 25 for flow cytometric analysis of TIL.

Tumors of RNA-vaccinated mice contained a significantly higher frequency of total CD8+ T cells as well as of NK cells as compared with those from the untreated or rapamycin only-treated control mice (Fig. 3A), confirming the capability of RNA vaccine to efficiently induce antitumor immunity (20). Combining RNA vaccination with rapamycin had only a modest added value on the total CD8+ T-cell and NK cell frequencies as compared with RNA vaccination alone. However, the frequency of antigen-specific CD8+ T cells was enhanced in the tumors of mice treated with rapamycin after RNA vaccination (Fig. 3C, left).

Figure 3.

Rapamycin treatment with RNA vaccination alters the infiltration of leukocytes into the tumor. A–D, representative tumor experiment with the B16-OVA system in C57BL/6 mice as described in Fig. 2. Tumors were harvested for the analysis of TIL by flow cytometry between day 20 and day 26 [control (n = 7), Rapa (n = 7), RNA (n = 6), RNA+Rapa (n = 7)]. Frequency of total CD8+ T cells and NK cells (A), MDSC (B), SIINFEKL-specific CD8+ T cells, and Treg (C). D, ratio of intratumoral SIINFEKL-specific CD8+ T cells in relation to Treg.

Figure 3.

Rapamycin treatment with RNA vaccination alters the infiltration of leukocytes into the tumor. A–D, representative tumor experiment with the B16-OVA system in C57BL/6 mice as described in Fig. 2. Tumors were harvested for the analysis of TIL by flow cytometry between day 20 and day 26 [control (n = 7), Rapa (n = 7), RNA (n = 6), RNA+Rapa (n = 7)]. Frequency of total CD8+ T cells and NK cells (A), MDSC (B), SIINFEKL-specific CD8+ T cells, and Treg (C). D, ratio of intratumoral SIINFEKL-specific CD8+ T cells in relation to Treg.

Close modal

Analysis of tumors for infiltration by the two major myeloid-derived suppressor cell (MDSC) populations (Gr1hiCD11b+ and Gr1lowCD11b+ phenotypes) revealed that rapamycin significantly lowers the frequency of these immunosuppressive cell populations in situ in both vaccinated and unvaccinated mice (Fig. 3B). This effect of mTOR inhibition has not been reported so far. The mechanism of this rapamycin effect has not been determined; one possibility is that it may function via rapamycin-mediated inhibition of VEGF production, which is known to have a role in the generation of MDSCs (21, 22).

Rapamycin has been described to promote the expansion of human CD4+FoxP3+ (23, 24). However, we did not observe any significant increase in intratumoral Treg in rapamycin-treated animals as compared with that in the control groups (Fig. 3C, right). Results from another study in the B16 melanoma model indicated that not the absolute number of Treg but the ratio of intratumoral CD8+ T cell to Treg (Teff/Treg) is correlated with therapeutic efficacy (25). We found that tumors from RNA-vaccinated mice displayed a high intratumoral Teff/Treg ratio, which was further improved by mTOR inhibition (Fig. 3D). The high Teff/Treg ratio together with the low numbers of MDSC in the tumor microenvironment may explain the superior control of tumor growth resulting from the combined treatment of RNA vaccination and mTOR inhibition.

In conclusion, this report shows for the first time synergy between rapamycin and intranodal RNA vaccination in de novo priming of the endogenous CD8+ T-cell repertoire resulting in significantly improved outcome in a therapeutic tumor model. We provide direct evidence that mTOR inhibition supports memory T-cell formation and promotes immune-beneficial alterations of the tumor microenvironment.

Given the ease of translation of such a concept, these findings may open up new paths for clinical development of RNA vaccines.

S. Kreiter is a consultant/advisor and has ownership interest (including patents) in Ribological GmbH. C. Huber is a cofounder and member of the supervisory board, has an ownership interest (including patents), and is a consultant/advisory board member of BioNTech, and has commercial research support in Cluster individualized immune intervention CI3. U. Sahin is employed on a consulting basis as founder and CEO of BioNTech. No potential conflicts of interest were disclosed by the other authors.

Conception and design: M. Diken, S. Kreiter, C. Huber, U. Sahin

Development of methodology: M. Diken, S. Kreiter, A. Selmi, S. Attig

Acquisition of data (provided animals, acquired and managed patients, provided facilities, etc.): M. Diken, A. Selmi, S. Attig, J. Diekmann

Analysis and interpretation of data (e.g., statistical analysis, biostatistics, computational analysis): M. Diken, S. Kreiter, F. Vascotto, A. Selmi, S. Attig, J. Diekmann

Writing, review, and/or revision of the manuscript: M. Diken, S. Kreiter, F. Vascotto, S. Attig, C. Huber, Ö. Türeci, U. Sahin

Administrative, technical, or material support (i.e., reporting or organizing data, constructing databases): A. Selmi, S. Attig

Study supervision: S. Kreiter, Ö. Türeci, U. Sahin

The authors thank M. Holzmann and R. Roth for excellent technical assistance and Dr. John Castle for critical proofreading of the article.

This work was supported by the GO-Bio and CI3 excellence cluster programs of the Federal Ministry of Education and Research (BMBF).

1.
Kreiter
S
,
Diken
M
,
Selmi
A
,
Türeci
O
,
Sahin
U
. 
Tumor vaccination using messenger RNA: prospects of a future therapy
.
Curr Opin Immunol
2011
;
23
:
399
406
.
2.
Van
LS
,
Heirman
C
,
Thielemans
K
,
Breckpot
K
. 
mRNA: From a chemical blueprint for protein production to an off-the-shelf therapeutic
.
Hum Vaccin Immunother.
2013 Jan 4
.
[Epub ahead of print]
.
3.
Diken
M
,
Kreiter
S
,
Selmi
A
,
Britten
CM
,
Huber
C
,
Türeci
O
, et al
Selective uptake of naked vaccine RNA by dendritic cells is driven by macropinocytosis and abrogated upon DC maturation
.
Gene Ther
2011
;
18
:
702
8
.
4.
Scheel
B
,
Aulwurm
S
,
Probst
J
,
Stitz
L
,
Hoerr
I
,
Rammensee
HG
, et al
Therapeutic anti-tumor immunity triggered by injections of immunostimulating single-stranded RNA
.
Eur J Immunol
2006
;
36
:
2807
16
.
5.
Kreiter
S
,
Selmi
A
,
Diken
M
,
Koslowski
M
,
Britten
CM
,
Huber
C
, et al
Intranodal vaccination with naked antigen-encoding RNA elicits potent prophylactic and therapeutic antitumoral immunity
.
Cancer Res
2010
;
70
:
9031
40
.
6.
Van
LS
,
Goyvaerts
C
,
Maenhout
S
,
Goethals
L
,
Disy
A
,
Benteyn
D
, et al
Preclinical evaluation of TriMix and antigen mRNA-based antitumor therapy
.
Cancer Res
2012
;
72
:
1661
71
.
7.
Weide
B
,
Carralot
JP
,
Reese
A
,
Scheel
B
,
Eigentler
TK
,
Hoerr
I
, et al
Results of the first phase I/II clinical vaccination trial with direct injection of mRNA
.
J Immunother
2008
;
31
:
180
8
.
8.
Holtkamp
S
,
Kreiter
S
,
Selmi
A
,
Simon
P
,
Koslowski
M
,
Huber
C
, et al
Modification of antigen-encoding RNA increases stability, translational efficacy, and T-cell stimulatory capacity of dendritic cells
.
Blood
2006
;
108
:
4009
17
.
9.
Kreiter
S
,
Selmi
A
,
Diken
M
,
Sebastian
M
,
Osterloh
P
,
Schild
H
, et al
Increased antigen presentation efficiency by coupling antigens to MHC class I trafficking signals
.
J Immunol
2008
;
180
:
309
18
.
10.
Kreiter
S
,
Diken
M
,
Selmi
A
,
Diekmann
J
,
Attig
S
,
Husemann
Y
, et al
FLT3 ligand enhances the cancer therapeutic potency of naked RNA vaccines
.
Cancer Res
2011
;
71
:
6132
42
.
11.
Araki
K
,
Turner
AP
,
Shaffer
VO
,
Gangappa
S
,
Keller
SA
,
Bachmann
MF
, et al
mTOR regulates memory CD8 T-cell differentiation
.
Nature
2009
;
460
:
108
12
.
12.
Pearce
EL
,
Walsh
MC
,
Cejas
PJ
,
Harms
GM
,
Shen
H
,
Wang
LS
, et al
Enhancing CD8 T-cell memory by modulating fatty acid metabolism
.
Nature
2009
;
460
:
103
7
.
13.
Rao
RR
,
Li
Q
,
Odunsi
K
,
Shrikant
PA
. 
The mTOR kinase determines effector versus memory CD8+ T cell fate by regulating the expression of transcription factors T-bet and Eomesodermin
.
Immunity
2010
;
32
:
67
78
.
14.
Araki
K
,
Youngblood
B
,
Ahmed
R
. 
The role of mTOR in memory CD8 T-cell differentiation
.
Immunol Rev
2010
;
235
:
234
43
.
15.
Kaech
SM
,
Tan
JT
,
Wherry
EJ
,
Konieczny
BT
,
Surh
CD
,
Ahmed
R
. 
Selective expression of the interleukin 7 receptor identifies effector CD8 T cells that give rise to long-lived memory cells
.
Nat Immunol
2003
;
4
:
1191
8
.
16.
Hackstein
H
,
Taner
T
,
Logar
AJ
,
Thomson
AW
. 
Rapamycin inhibits macropinocytosis and mannose receptor-mediated endocytosis by bone marrow-derived dendritic cells
.
Blood
2002
;
100
:
1084
7
.
17.
Li
Q
,
Rao
R
,
Vazzana
J
,
Goedegebuure
P
,
Odunsi
K
,
Gillanders
W
, et al
Regulating mammalian target of rapamycin to tune vaccination-induced CD8(+) T cell responses for tumor immunity
.
J Immunol
2012
;
188
:
3080
7
.
18.
Wang
Y
,
Wang
XY
,
Subjeck
JR
,
Shrikant
PA
,
Kim
HL
. 
Temsirolimus, an mTOR inhibitor, enhances anti-tumour effects of heat shock protein cancer vaccines
.
Br J Cancer
2011
;
104
:
643
52
.
19.
DuPage
M
,
Mazumdar
C
,
Schmidt
LM
,
Cheung
AF
,
Jacks
T
. 
Expression of tumour-specific antigens underlies cancer immunoediting
.
Nature
2012
;
482
:
405
9
.
20.
Smyth
MJ
,
Thia
KY
,
Street
SE
,
Cretney
E
,
Trapani
JA
,
Taniguchi
M
, et al
Differential tumor surveillance by natural killer (NK) and NKT cells
.
J Exp Med
2000
;
191
:
661
8
.
21.
Zhong
H
,
Chiles
K
,
Feldser
D
,
Laughner
E
,
Hanrahan
C
,
Georgescu
MM
, et al
Modulation of hypoxia-inducible factor 1alpha expression by the epidermal growth factor/phosphatidylinositol 3-kinase/PTEN/AKT/FRAP pathway in human prostate cancer cells: implications for tumor angiogenesis and therapeutics
.
Cancer Res
2000
;
60
:
1541
5
.
22.
Gabrilovich
D
,
Ishida
T
,
Oyama
T
,
Ran
S
,
Kravtsov
V
,
Nadaf
S
, et al
Vascular endothelial growth factor inhibits the development of dendritic cells and dramatically affects the differentiation of multiple hematopoietic lineages in vivo
.
Blood
1998
;
92
:
4150
66
.
23.
Strauss
L
,
Whiteside
TL
,
Knights
A
,
Bergmann
C
,
Knuth
A
,
Zippelius
A
. 
Selective survival of naturally occurring human CD4+CD25+Foxp3+ regulatory T cells cultured with rapamycin
.
J Immunol
2007
;
178
:
320
9
.
24.
Battaglia
M
,
Stabilini
A
,
Roncarolo
MG
. 
Rapamycin selectively expands CD4+CD25+FoxP3 +regulatory T cells
.
Blood
2005
;
105
:
4743
8
.
25.
Quezada
SA
,
Peggs
KS
,
Curran
MA
,
Allison
JP
. 
CTLA4 blockade and GM-CSF combination immunotherapy alters the intratumor balance of effector and regulatory T cells
.
J Clin Invest
2006
;
116
:
1935
45
.