Fiedler 2016
Fiedler 2016
Fiedler 2016
Abstract
mRNA cancer vaccines are a relatively new class of vaccines, which combine the
potential of mRNA to encode for almost any protein with an excellent safety profile
and a flexible production process. The most straightforward use of mRNA vaccines
in oncologic settings is the immunization of patients with mRNA vaccines encoding
tumor-associated antigens (TAAs). This is exemplified by the RNActive®
technology, which induces balanced humoral and cellular immune responses in
animal models and is currently evaluated in several clinical trials for oncologic
indications. A second application of mRNA vaccines is the production of
personalized vaccines. This is possible because mRNA vaccines are produced by
a generic process, which can be used to quickly produce mRNA vaccines targeting
patient-specific neoantigens that are identified by analyzing the tumor exome. Apart
from being used directly to vaccinate patients, mRNAs can also be used in cellular
therapies to transfect patient-derived cells in vitro and infuse the manipulated cells
back into the patient. One such application is the transfection of patient-derived
dendritic cells (DCs) with mRNAs encoding TAAs, which leads to the presentation
of TAA-derived peptides on the DCs and an activation of antigen-specific T cells
in vivo. A second application is the transfection of patient-derived T cells with
mRNAs encoding chimeric antigen receptors, which allows the T cells to directly
recognize a specific antigen expressed on the tumor. In this chapter, we will review
preclinical and clinical data for the different approaches.
Keywords
RNActive® vaccines Personalized mRNA vaccines mRNA-pulsed dendritic
cells Chimeric antigen receptor (CAR) T cell immunotherapy
Abbreviations
A Adenine
APC Antigen-presenting cell
CAR Chimeric antigen receptor
CTL Cytotoxic T lymphocyte
CTLA4 Cytotoxic T lymphocyte antigen 4
DC Dendritic cell
DNA Deoxyribonucleic acid
EGFR Endothelial growth factor receptor
ELISA Enzyme-linked immunosorbent assay
GC Guanine and cytosine
GITR Glucocorticoid-induced tumor necrosis factor receptor
HLA Human leukocyte antigen
HMGB1 High-mobility group box 1
IDH1 Isocitrate dehydrogenase 1
IFN Interferon
Ig Immunoglobulin
LLC Lewis lung cancer
MAGEC1 Melanoma antigen family C1
MAGEC2 Melanoma antigen family C2
MHC Major histocompatibility complex
mRNA Messenger ribonucleic acid
MUC1 Mucin 1
NK Natural killer
NSCLC Non-small-cell lung cancer
NY-ESO-1 New York esophageal squamous cell carcinoma-1
ORF Open reading frame
OVA Ovalbumin
PAP Prostatic acid phosphatase
PD1 Programmed cell death 1
PDL1 Programmed cell death 1 ligand 1
PDL2 Programmed cell death 1 ligand 2
PFS Progression-free survival
PSA Prostate-specific antigen
PSCA Prostate stem cell antigen
PSMA Prostate-specific membrane antigen
RNA Ribonucleic acid
RT-PCR Reverse transcriptase polymerase chain reaction
STEAP1 Six-transmembrane epithelial antigen of the prostate 1
TAA Tumor-associated antigen
TERT Telomerase reverse transcriptase
Th T helper
TLR Toll-like receptor
mRNA Cancer Vaccines 63
1.1 Introduction
5T4
68 K. Fiedler et al.
In the past, development of cancer vaccines was mainly dedicated toward the
so-called tumor-associated proteins as antigenic source. Mostly, these
tumor-associated antigens can be divided into two classes: Antigens derived from
fetal genes overexpressed in cancer cells (van der Bruggen et al. 1991; Van den
Eynde et al. 1991) or antigens derived from tissue-specific proteins also expressed
in cancer cells (Boon et al. 1994). To offer broadly applicable immune therapies
against cancer, the latest developmental activity in the field of cancer vaccines was
focused on shared tumor-associated antigens that are expressed not only in abun-
dance in different cancer types but also in a certain amount of cancer patients.
But the use of such shared tumor-associated antigens is accompanied with certain
drawbacks: Firstly, T cells easily recognize foreign antigens but in general are
unable to recognize self-antigens due to the negative selection process in the thymus
preventing induction of autoimmunity. Unfortunately, most tumor-associated anti-
gens fall into the group of self-antigens hampering the initiation of a proper immune
response toward these antigens. Secondly, tumor-associated antigens are not soli-
tarily expressed in tumor tissue leading to possible on-target effects in healthy tissue
through the induced immune response. Finally, the expression of tumor-associated
antigens in different tumor tissues or different cancer patients can be highly variable
due to normal biologic heterogeneity. Recent research has shown that ninety-five
percent of the mutations in a given patient seem to be unique to that tumor (Stratton
2011) and that even on a subclonal level a high variability can be found in a certain
tumor (Gerlinger et al. 2015; Martincorena et al. 2015). Therefore, it can be assumed
that expression of shared tumor-associated antigens is subjected to a similar vari-
ability. Moreover, tumor tissues often exploit different escape mechanisms to evade
the antitumoral immune response like downregulation of tumor-associated antigens
or preferential outgrowth of non-expressing clones (Matsushita et al. 2012).
mRNA Cancer Vaccines 73
interesting are the results of Snyder et al. and Rizvi et al. demonstrating a close
correlation between the abundance of clonal neoantigens and sensitivity to immune
checkpoint blockade by anti-PD1 or anti-CTLA-4 treatment in patients suffering
from non-small-cell lung cancer or melanoma. Moreover, the predicted neoantigens
elicited specific CD8+ T cell responses in both studies (Snyder et al. 2014; Rizvi
et al. 2015). Also for therapeutic approaches with adoptive tumor-infiltrating
lymphocyte transfer in melanoma patients, a similar correlation between neoantigen
load and therapy efficacy was revealed (Lu et al. 2014; Linnemann et al. 2015).
Additionally, Tran and colleagues could provide evidence that immunotherapy
based on mutation-specific CD4+ T cells in a patient with epithelial cancer is
possible (Tran et al. 2014).
The above-mentioned studies and several further publications demonstrated the
feasibility of tumor exome mining plus immunogenicity prediction based on HLA
allotypes and peptide-binding probability to identify patient-specific highly
immunogenic neoantigens in different types of cancer with a variety of approaches
(Rajasagi et al. 2014; Duan et al. 2014; Yadav et al. 2014; Tran et al. 2015).
Moreover, the potential value of such neoantigens for personalized immunotherapy
approaches has been frequently implied (Gubin et al. 2014; Tran et al. 2014). One
possibility to target specifically neoantigens in cancer patients is to engineer
neoantigen-specific T cell receptors and adoptively transfer these T cells (Leisegang
et al. 2016) using methods also described here. A further option would be the
immunization with vaccines incorporating the mutated sequences of neoantigens.
The efficacy of such an immunization approach using peptides is currently under
evaluation in a phase I clinical trial in IDH1R123H-mutated grade III–IV gliomas
(NCT02454634). This clinical study is based on the results published by Schu-
macher and colleagues showing the antitumor potential of mutant IDH1 peptide
vaccination in tumor-bearing MHC-humanized mice (Schumacher et al. 2014).
However, mutant IDH1 seems to be a unique exception, since the mutation can be
found frequently in diffuse grade II and grade III gliomas.
This high penetrance of a neoantigen appears to be in contrast to other tumor
entities. In most human tumors, a large fraction of the mutations is not shared
between patients at a meaningful frequency. Moreover, only a small portion of
mutations within expressed genes have antigenic potential leading to T cell reac-
tivity (Lu et al. 2014; Linnemann et al. 2015). Due to these limitations, broadly
applicable neoantigens that can be used in huge patient cohorts for vaccinations are
highly unlikely. Hence, mRNA vaccines represent a superior approach to satisfy the
specific demands of a personalized immunotherapy based on neoantigens. The
obvious advantages of mRNA vaccines in comparison with peptide vaccines were
already mentioned above. Particularly, the production under well-defined and
controlled conditions by in vitro transcription and the possibility to produce vac-
cines against different neoantigens by a common process and in a relatively short
period of time fulfill the requirements for a personalized immunotherapy approach.
In a recent study of Kreiter and colleagues, the tumor-rejecting properties of
neoantigen-based RNA vaccines were demonstrated in murine tumor models. In
principle, mutanome analysis and MHC-binding prediction lead to selection of
mRNA Cancer Vaccines 75
on HLA restriction of the patients, meaning that the induced immune response by
the vaccine is limited to the peptides used and that only patients with specific HLA
allotypes can be treated (Van Nuffel et al. 2012). Instead, mRNA molecules can
encode the entire tumor antigen. Therefore, multiple immunogenic epitopes within
the same protein can be presented. In addition, since the protein is endogenously
expressed and presented after mRNA transfection of DCs, multiple peptide–MHC
complexes (pMHC) are generated; thereby, vaccine development is independent on
the patient’s genetic background.
Boczkowski and collaborators were the first ones to describe in the late 1990s
that DCs pulsed with mRNA encoding for tumor antigens are potent antigen-
presenting cells (Boczkowski et al. 1996). Only few years later, an early clinical
trial using DCs loaded with RNA encoding the PSA proved the feasibility and
safety of this approach. CTL responses against metastatic prostate tumors
expressing PSA were indeed induced in some patients (Heiser et al. 2002). This
approach was based on passive pulsing of DCs, which relies on the ability of these
cells to take up mRNA through micropinocytosis (Diken et al. 2011). But this
mechanism, involving transport into the endosomes, entails the risk that only a
fraction of the mRNA can reach the cytosol and can then be translated into the
protein. Therefore, several approaches have been established in order to deliver
mRNA directly to the cytoplasm such as electroporation, nucleofection, lipofection,
and more recently sonoporation (Benteyn et al. 2015). Above all, electroporation
has been shown to be a powerful technique to introduce tumor antigens into DCs
(Van Tendeloo et al. 2001).
mRNA encoding specific TAAs or total tumor RNA can be used to transfect
DCs. Vaccine strategies employing DCs transfected with defined TAA mRNA
avoid the need for growth of patient-specific tumor cells and reduce the risk of
autoimmunity, which can be induced in patients by the presence of normally
expressed self-proteins (Nair et al. 1999; Ponsaerts et al. 2003). However, there are
some limitations due to the fact that for many cancers, the TAAs are unknown. An
attractive alternative is to utilize DCs transfected with patient-derived total tumor
RNA. Through this approach, the entire spectrum of tumor-specific antigens is
displayed, thereby eliminating the need for identification of TAAs and allowing the
immune system to use the most effective antigens while reducing the risk of escape
mutants. Another advantage of using tumor-derived RNA as a source of
whole-tumor antigen is that it can be quickly and easily amplified by RT-PCR from
even a small amount of tumor (Heiser et al. 2002).
To date, several clinical trials have been reported using mRNA-transfected DCs.
The majority of these studies have shown that tumor-specific T cell responses can
be induced by mRNA-transfected DCs in several tumor entities such as brain
cancer, melanoma, lung adenocarcinoma, renal cell carcinoma, and ovarian cancer
(Bonehill et al. 2009; Caruso et al. 2004, 2005; Kyte et al. 2007; Nair et al. 2002;
Su et al. 2003; Morse et al. 2003; Dannull et al. 2005; Van Nuffel et al. 2012). In
particular, in the renal cell carcinoma study by Su et al., patients displayed no
evidence of dose-limiting toxicity or induction of autoimmunity (Su et al. 2003).
mRNA Cancer Vaccines 77
Similarly, brain tumor and neuroblastoma studies conducted in nine and seven
patients, respectively, showed a clinical response in a total of three of the patients.
Despite many progresses in mRNA-DC immunotherapies, clinical responses
remain modest and new strategies on how to enhance the efficacy of mRNA-DC
vaccines are being explored. Besides the delivery of tumor antigens, mRNA can be
used to deliver also proteins that can modulate the function of DCs. Cotransfection
of DCs with mRNA encoding TAAs and costimulatory molecules such as CD83
(Aerts-Toegaerr et al. 2007), OX40 (Dannull et al. 2005), and 4-1BBL (Grünebach
et al. 2005) has shown to improve mRNA-DC vaccine efficacy in preclinical
models. Moreover, DCs activated through electroporation with mRNAs encoding
four tumor antigens as well as mRNAs encoding CD40 ligand and constitutively
active TLR4 and CD70 (TriMix-DCs) resulted in a broad T cell response and
durable tumor response in chemorefractory advanced melanoma patients (Van
Nuffel et al. 2012). DCs have been also modified with mRNAs encoding
immunomodulating cytokines. For example, DCs transfected with mRNAs
encoding IL-12 and TAAs were shown to induce high-avidity cytotoxic T cells and
enhance their effector function. In addition, the migratory capacity of DCs has been
modulated using mRNA encoding a chimeric E/L-selectin, a strategy that led to
increased migration of the DCs to lymph nodes upon intravenous administration
(Dörrie et al. 2008).
Finally, further preclinical strategies have focused on increased DC function by
coadministration of DCs transfected with TAA-encoding mRNAs or mRNAs
encoding for antagonistic anti-CTLA-4 or agonistic anti-GITR antibodies (Pruitt
et al. 2011). Thus, the engineered DCs have the ability not only to present the tumor
antigen of interest but also to locally modulate immune checkpoints and the tumor
microenvironment. Because of promising results in preclinical studies, this
approach is currently under investigation in a phase I clinical trial for the treatment
of melanoma patients (NCT01216436).
Taken together, these strategies have the potential to improve cancer
immunotherapy.
2 Conclusion
Acknowledgments We would like to thank Henoch Hong, Ulrike Gnad-Vogt, Ulrich Kruse, and
Mariola Fotin-Mleczek for discussions and suggestions.
References
Aerts-Toegaert C, Heirman C, Tuyaerts S, Corthals J, Aerts JL, Bonehill A, Thielemans K,
Breckpot K (2007) CD83 expression on dendritic cells and T cells: correlation with effective
immune responses. Eur J Immunol 37(3):686–695
Anderson BR, Muramatsu H, Nallagatla SR, Bevilacqua PC, Sansing LH, Weissman D, Karikó K
(2010) Incorporation of pseudouridine into mRNA enhances translation by diminishing PKR
activation. Nucleic Acids Res 38(17):5884–5892
Bai Y, Kan S, Zhou S, Wang Y, Xu J, Cooke JP, Wen J, Deng H (2015) Enhancement of the
in vivo persistence and antitumor efficacy of CD19 chimeric antigen receptor T cells through
the delivery of modified TERT mRNA. Cell Discov 1:Article number: 15040
Barrett DM, Zhao Y, Liu X, Jiang S, Carpenito C, Kalos M, Carroll RG, June CH, Grupp SA
(2011) Treatment of advanced leukemia in mice with mRNA engineered T cells. Hum Gene
Ther 22(12):1575–1586
Barrett DM, Liu X, Jiang S, June CH, Grupp SA, Zhao Y (2013) Regimen-specific effects of
RNA-modified chimeric antigen receptor T cells in mice with advanced leukemia. Hum Gene
Ther 24(8):717–727
Beatty GL, Haas AR, Maus MV, Torigian DA, Soulen MC, Plesa G, Chew A, Zhao Y, Levine BL,
Albelda SM, Kalos M, June CH (2014) Mesothelin-specific chimeric antigen receptor
mRNA-engineered T cells induce anti-tumor activity in solid malignancies. Cancer Immunol
Res 2(2):112–120
Benteyn D, Heirman C, Bonehill A, Thielemans K, Breckpot K (2015) mRNA-based dendritic cell
vaccines. Expert Rev Vaccines 14(2):161–176
Boczkowski D, Nair SK, Snyder D, Gilboa E (1996) Dendritic cells pulsed with RNA are potent
antigen-presenting cells in vitro and in vivo. J Exp Med 184(2):465–472
Bonehill A, Van Nuffel AM, Corthals J, Tuyaerts S, Heirman C, François V, Colau D, van der
Bruggen P, Neyns B, Thielemans K (2009) Single-step antigen loading and activation of
dendritic cells by mRNA electroporation for the purpose of therapeutic vaccination in
melanoma patients. Clin Cancer Res 15(10):3366–3375
Boon T, Kellermann O (1977) Rejection by syngeneic mice of cell variants obtained by
mutagenesis of a malignant teratocarcinoma cell line. Proc Natl Acad Sci USA 74(1):272–275
Boon T, Cerottini JC, Van den Eynde B, van der Bruggen P, Van Pel A (1994) Tumor antigens
recognized by T lymphocytes. Annu Rev Immunol 12:337–365
80 K. Fiedler et al.
Brentjens RJ, Davila ML, Riviere I, Park J, Wang X, Cowell LG, Bartido S, Stefanski J, Taylor C,
Olszewska M, Borquez-Ojeda O, Qu J, Wasielewska T, He Q, Bernal Y, Rijo IV, Hedvat C,
Kobos R, Curran K, Steinherz P, Jurcic J, Rosenblat T, Maslak P, Frattini M, Sadelain M
(2013) CD19-targeted T cells rapidly induce molecular remissions in adults with
chemotherapy-refractory acute lymphoblastic leukemia. Sci Transl Med 5(177):177ra38
Caruso DA, Orme LM, Neale AM, Radcliff FJ, Amor GM, Maixner W, Downie P, Hassall TE,
Tang ML, Ashley DM (2004) Results of a phase 1 study utilizing monocyte-derived dendritic
cells pulsed with tumor RNA in children and young adults with brain cancer. Neuro Oncol. 6
(3):236–246
Caruso DA, Orme LM, Amor GM, Neale AM, Radcliff FJ, Downie P, Tang ML, Ashley DM
(2005) Results of a Phase I study utilizing monocyte-derived dendritic cells pulsed with tumor
RNA in children with Stage 4 neuroblastoma. Cancer 103(6):1280–1291
Castle JC, Kreiter S, Diekmann J, Löwer M, van de Roemer N, de Graaf J, Selmi A, Diken M,
Boegel S, Paret C, Koslowski M, Kuhn AN, Britten CM, Huber C, Türeci O, Sahin U (2012)
Exploiting the mutanome for tumor vaccination. Cancer Res 72(5):1081–1091
Cerundolo V, Hermans IF, Salio M (2004) Dendritic cells: a journey from laboratory to clinic. Nat
Immunol 5(1):7–10
Cheadle EJ, Sheard V, Hombach AA, Chmielewski M, Riet T, Berrevoets C, Schooten E,
Lamers C, Abken H, Debets R, Gilham DE (2012) Chimeric antigen receptors for T-cell based
therapy. Methods Mol Biol 907:645–666
Coulie PG, Lehmann F, Lethé B, Herman J, Lurquin C, Andrawiss M, Boon T (1995) A mutated
intron sequence codes for an antigenic peptide recognized by cytolytic T lymphocytes on a
human melanoma. Proc Natl Acad Sci U S A. 92(17):7976–7980
Dannull J, Nair S, Su Z, Boczkowski D, DeBeck C, Yang B, Gilboa E, Vieweg J (2005)
Enhancing the immunostimulatory function of dendritic cells by transfection with mRNA
encoding OX40 ligand. Blood 105(8):3206–3213
Demaria S, Formenti SC (2012) Role of T lymphocytes in tumor response to radiotherapy. Front
Oncol 2:95
Diken M, Kreiter S, Selmi A, Britten CM, Huber C, Türeci Ö, Sahin U (2011) Selective uptake of
naked vaccine RNA by dendritic cells is driven by macropinocytosis and abrogated upon DC
maturation. Gene Ther 18(7):702–708
Dörrie J, Schaft N, Müller I, Wellner V, Schunder T, Hänig J, Oostingh GJ, Schön MP, Robert C,
Kämpgen E, Schuler G (2008) Introduction of functional chimeric E/L-selectin by RNA
electroporation to target dendritic cells from blood to lymph nodes. Cancer Immunol
Immunother 57(4):467–477
Duan F, Duitama J, Al Seesi S, Ayres CM, Corcelli SA, Pawashe AP, Blanchard T, McMahon D,
Sidney J, Sette A, Baker BM, Mandoiu II, Srivastava PK (2014) Genomic and bioinformatic
profiling of mutational neoepitopes reveals new rules to predict anticancer immunogenicity.
J Exp Med 211(11):2231–2248
Echchakir H, Mami-Chouaib F, Vergnon I, Baurain JF, Karanikas V, Chouaib S, Coulie PG (2001)
A point mutation in the alpha-actinin-4 gene generates an antigenic peptide recognized by
autologous cytolytic T lymphocytes on a human lung carcinoma. Cancer Res 61(10):4078–4083
Formenti SC, Demaria S (2013) Combining radiotherapy and cancer immunotherapy: a paradigm
shift. J Natl Cancer Inst 105(4):256–265
Fotin-Mleczek M, Duchardt KM, Lorenz C, Pfeiffer R, Ojkić-Zrna S, Probst J, Kallen KJ (2011)
Messenger RNA-based vaccines with dual activity induce balanced TLR-7 dependent adaptive
immune responses and provide antitumor activity. J Immunother 34(1):1–15
Fotin-Mleczek M, Zanzinger K, Heidenreich R, Lorenz C, Thess A, Duchardt KM, Kallen KJ
(2012) Highly potent mRNA based cancer vaccines represent an attractive platform for
combination therapies supporting an improved therapeutic effect. J Gene Med. 14(6):428–439
Fotin-Mleczek M, Zanzinger K, Heidenreich R, Lorenz C, Kowalczyk A, Kallen KJ, Huber SM
(2014) mRNA-based vaccines synergize with radiation therapy to eradicate established tumors.
Radiat Oncol 9:180
mRNA Cancer Vaccines 81
Garnett CT, Schlom J, Hodge JW (2008) Combination of docetaxel and recombinant vaccine
enhances T-cell responses and antitumor activity: effects of docetaxel on immune enhance-
ment. Clin Cancer Res 14(11):3536–3544
Gerlinger M, Catto JW, Orntoft TF, Real FX, Zwarthoff EC, Swanton C (2015) Intratumour
heterogeneity in urologic cancers: from molecular evidence to clinical implications. Eur Urol
67(4):729–737
Golden EB, Frances D, Pellicciotta I, Demaria S, Helen Barcellos-Hoff M, Formenti SC (2014)
Radiation fosters dose-dependent and chemotherapy-induced immunogenic cell death.
Oncoimmunology 3:e28518
Grünebach F, Kayser K, Weck MM, Müller MR, Appel S, Brossart P (2005) Cotransfection of
dendritic cells with RNA coding for HER-2/neu and 4-1BBL increases the induction of tumor
antigen specific cytotoxic T lymphocytes. Cancer Gene Ther 12(9):749–756
Grupp SA, Kalos M, Barrett D, Aplenc R, Porter DL, Rheingold SR, Teachey DT, Chew A,
Hauck B, Wright JF, Milone MC, Levine BL, June CH (2013) Chimeric antigen
receptor-modified T cells for acute lymphoid leukemia. N Engl J Med 368(16):1509–1518
Gubin MM, Zhang X, Schuster H, Caron E, Ward JP, Noguchi T, Ivanova Y, Hundal J,
Arthur CD, Krebber WJ, Mulder GE, Toebes M, Vesely MD, Lam SS, Korman AJ, Allison JP,
Freeman GJ, Sharpe AH, Pearce EL, Schumacher TN, Aebersold R, Rammensee HG,
Melief CJ, Mardis ER, Gillanders WE, Artyomov MN, Schreiber RD (2014) Checkpoint
blockade cancer immunotherapy targets tumour-specific mutant antigens. Nature 515
(7528):577–581
Heiser A, Coleman D, Dannull J, Yancey D, Maurice MA, Lallas CD, Dahm P, Niedzwiecki D,
Gilboa E, Vieweg J (2002) Autologous dendritic cells transfected with prostate-specific antigen
RNA stimulate CTL responses against metastatic prostate tumors. J Clin Invest. 109
(3):409–417
Hoerr I, Obst R, Rammensee HG, Jung G (2000) In vivo application of RNA leads to induction of
specific cytotoxic T lymphocytes and antibodies. Eur J Immunol 30(1):1–7
Jäger E, Jäger D, Knuth A (2002) Clinical cancer vaccine trials. Curr Opin Immunol 14
(2):178–182
Jensen MC, Riddell SR (2015) Designing chimeric antigen receptors to effectively and safely
target tumors. Curr Opin Immunol 33:9–15
Kallen KJ, Theß A (2014) A development that may evolve into a revolution in medicine: mRNA
as the basis for novel, nucleotide-based vaccines and drugs. Ther Adv Vaccines. 2(1):10–31
Kallen KJ, Heidenreich R, Schnee M, Petsch B, Schlake T, Thess A, Baumhof P, Scheel B,
Koch SD, Fotin-Mleczek M (2013) A novel, disruptive vaccination technology:
self-adjuvanted RNActive(®) vaccines. Hum Vaccin Immunother. 9(10):2263–2276
Kantoff PW, Higano CS, Shore ND, Berger ER, Small EJ, Penson DF, Redfern CH, Ferrari AC,
Dreicer R, Sims RB, Xu Y, Frohlich MW, Schellhammer PF; IMPACT Study Investigators
(2010) Sipuleucel-T immunotherapy for castration-resistant prostate cancer. N Engl J Med. 363
(5):411–22
Karikó K, Muramatsu H, Welsh FA, Ludwig J, Kato H, Akira S, Weissman D (2008)
Incorporation of pseudouridine into mRNA yields superior nonimmunogenic vector with
increased translational capacity and biological stability. Mol Ther 16(11):1833–1840
Kershaw MH, Westwood JA, Darcy PK (2013) Gene-engineered T cells for cancer therapy. Nat
Rev Cancer 13(8):525–541
Knapp DC, Mata JE, Reddy MT, Devi GR, Iversen PL (2003) Resistance to chemotherapeutic
drugs overcome by c-Myc inhibition in a Lewis lung carcinoma murine model. Anticancer
Drugs 14:39–47
Kochenderfer JN, Dudley ME, Feldman SA, Wilson WH, Spaner DE, Maric I, Stetler-Stevenson
M, Phan GQ, Hughes MS, Sherry RM, Yang JC, Kammula US, Devillier L, Carpenter R,
Nathan DA, Morgan RA, Laurencot C, Rosenberg SA (2012) B-cell depletion and remissions
of malignancy along with cytokine-associated toxicity in a clinical trial of anti-CD19
chimeric-antigen-receptor-transduced T cells. Blood 119(12):2709–2720
82 K. Fiedler et al.
Melero I, Berman DM, Aznar MA, Korman AJ, Pérez Gracia JL, Haanen J (2015) Evolving
synergistic combinations of targeted immunotherapies to combat cancer. Nat Rev Cancer 15
(8):457–472
Moore MW, Carbone FR, Bevan MJ (1988) Introduction of soluble protein into the class I
pathway of antigen processing and presentation. Cell 54(6):777–785
Morse MA, Nair SK, Mosca PJ, Hobeika AC, Clay TM, Deng Y, Boczkowski D, Proia A,
Neidzwiecki D, Clavien PA, Hurwitz HI, Schlom J, Gilboa E, Lyerly HK (2003)
Immunotherapy with autologous, human dendritic cells transfected with carcinoembryonic
antigen mRNA. Cancer Invest 21(3):341–349
Nair SK, Hull S, Coleman D, Gilboa E, Lyerly HK, Morse MA (1999) Induction of
carcinoembryonic antigen (CEA)-specific cytotoxic T-lymphocyte responses in vitro using
autologous dendritic cells loaded with CEA peptide or CEA RNA in patients with metastatic
malignancies expressing CEA. Int J Cancer 82(1):121–124
Nair SK, Morse M, Boczkowski D, Cumming RI, Vasovic L, Gilboa E, Lyerly HK (2002)
Induction of tumor-specific cytotoxic T lymphocytes in cancer patients by autologous tumor
RNA-transfected dendritic cells. Ann Surg 235(4):540–549
Palmer M, Parker J, Modi S, Butts C, Smylie M, Meikle A, Kehoe M, MacLean G,
Longenecker M (2001) Phase I study of the BLP25 (MUC1 peptide) liposomal vaccine for
active specific immunotherapy in stage IIIB/IV non-small-cell lung cancer. Clin Lung Cancer 3
(1):49–57 discussion 58
Petsch B, Schnee M, Vogel AB, Lange E, Hoffmann B, Voss D, Schlake T, Thess A, Kallen KJ,
Stitz L, Kramps T (2012) Protective efficacy of in vitro synthesized, specific mRNA vaccines
against influenza A virus infection. Nat Biotechnol 30(12):1210–1216
Ponsaerts P, Van Tendeloo VF, Berneman ZN (2003) Cancer immunotherapy using RNA-loaded
dendritic cells. Clin Exp Immunol 134(3):378–384
Porter DL, Levine BL, Kalos M, Bagg A, June CH (2011) Chimeric antigen receptor-modified T
cells in chronic lymphoid leukemia. N Engl J Med 365(8):725–733
Pruitt SK, Boczkowski D, de Rosa N, Haley NR, Morse MA, Tyler DS, Dannull J, Nair S (2011)
Enhancement of anti-tumor immunity through local modulation of CTLA-4 and GITR by
dendritic cells. Eur J Immunol 41(12):3553–3563
Rajasagi M, Shukla SA, Fritsch EF, Keskin DB, DeLuca D, Carmona E, Zhang W, Sougnez C,
Cibulskis K, Sidney J, Stevenson K, Ritz J, Neuberg D, Brusic V, Gabriel S, Lander ES,
Getz G, Hacohen N, Wu CJ (2014) Systematic identification of personal tumor-specific
neoantigens in chronic lymphocytic leukemia. Blood 124(3):453–462
Rammensee HG (2006) Some considerations on the use of peptides and mRNA for therapeutic
vaccination against cancer. Immunol Cell Biol 84(3):290–294
Ramos CA, Dotti G (2011) Chimeric antigen receptor (CAR)-engineered lymphocytes for cancer
therapy. Expert Opin Biol Ther 11(7):855–873
Reits EA, Hodge JW, Herberts CA, Groothuis TA, Chakraborty M, Wansley EK, Camphausen K,
Luiten RM, de Ru AH, Neijssen J, Griekspoor A, Mesman E, Verreck FA, Spits H, Schlom J, van
Veelen P, Neefjes JJ (2006) Radiation modulates the peptide repertoire, enhances MHC class I
expression, and induces successful antitumor immunotherapy. J Exp Med 203(5):1259–1271
Restifo NP, Dudley ME, Rosenberg SA (2012) Adoptive immunotherapy for cancer: harnessing
the T cell response. Nat Rev Immunol 12(4):269–281
Rizvi NA, Hellmann MD, Snyder A, Kvistborg P, Makarov V, Havel JJ, Lee W, Yuan J, Wong P,
Ho TS, Miller ML, Rekhtman N, Moreira AL, Ibrahim F, Bruggeman C, Gasmi B,
Zappasodi R, Maeda Y, Sander C, Garon EB, Merghoub T, Wolchok JD, Schumacher TN,
Chan TA (2015) Cancer immunology. Mutational landscape determines sensitivity to PD-1
blockade in non-small cell lung cancer. Science 348(6230):124–128
Robbins PF, Lu YC, El-Gamil M, Li YF, Gross C, Gartner J, Lin JC, Teer JK, Cliften P,
Tycksen E, Samuels Y, Rosenberg SA (2013) Mining exomic sequencing data to identify
mutated antigens recognized by adoptively transferred tumor-reactive T cells. Nat Med 19
(6):747–752
84 K. Fiedler et al.
Savai R, Schermuly RT, Pullamsetti SS, Schneider M, Greschus S, Ghofrani HA, Traupe H,
Grimminger F, Banat GA (2007) A combination hybrid-based vaccination/adoptive cellular
therapy to prevent tumor growth by involvement of T cells. Cancer Res 67(11):5443–5453
Scheel B, Teufel R, Probst J, Carralot JP, Geginat J, Radsak M, Jarrossay D, Wagner H, Jung G,
Rammensee HG, Hoerr I, Pascolo S (2005) Toll-like receptor-dependent activation of several
human blood cell types by protamine-condensed mRNA. Eur J Immunol 35(5):1557–1566
Schnee M, Vogel AB, Voss D, Petsch B, Baumhof P, Kramps T, Stitz L (2016) An mRNA vaccine
encoding rabies virus glycoprotein induces protection against lethal infection in mice and
correlates of protection in adult and newborn Pigs. PLoS Negl Trop Dis 10(6)
Schuler G, Schuler-Thurner B, Steinman RM (2003) The use of dendritic cells in cancer
immunotherapy. Curr Opin Immunol 15(2):138–147
Schumacher TN, Schreiber RD (2015) Neoantigens in cancer immunotherapy. Science 348
(6230):69–74
Schumacher T, Bunse L, Pusch S, Sahm F, Wiestler B, Quandt J, Menn O, Osswald M, Oezen I,
Ott M, Keil M, Balß J, Rauschenbach K, Grabowska AK, Vogler I, Diekmann J, Trautwein N,
Eichmüller SB, Okun J, Stevanović S, Riemer AB, Sahin U, Friese MA, Beckhove P, von
Deimling A, Wick W, Platten M (2014) A vaccine targeting mutant IDH1 induces antitumour
immunity. Nature 512(7514):324–327
Sebastian M, Papachristofilou A, Weiss C, Früh M, Cathomas R, Hilbe W, Wehler T, Rippin G,
Koch SD, Scheel B, Fotin-Mleczek M, Heidenreich R, Kallen KJ, Gnad-Vogt U, Zippelius A
(2014) Phase Ib study evaluating a self-adjuvanted mRNA cancer vaccine (RNActive®)
combined with local radiation as consolidation and maintenance treatment for patients with
stage IV non-small cell lung cancer. BMC Cancer 14:748
Shojaei F, Wu X, Malik AK, Zhong C, Baldwin ME, Schanz S, Fuh G, Gerber H-P, Ferrara N
(2007) Tumor refractoriness to anti-VEGF treatment is mediated by CD11b+ Gr1+ myeloid
cells. Nat Biotechnol 25:911–920
Snyder A, Makarov V, Merghoub T, Yuan J, Zaretsky JM, Desrichard A, Walsh LA, Postow MA,
Wong P, Ho TS, Hollmann TJ, Bruggeman C, Kannan K, Li Y, Elipenahli C, Liu C,
Harbison CT, Wang L, Ribas A, Wolchok JD, Chan TA (2014) Genetic basis for clinical
response to CTLA-4 blockade in melanoma. N Engl J Med 371(23):2189–2199
Stratton MR (2011) Exploring the genomes of cancer cells: progress and promise. Science 331
(6024):1553–1558
Su Z, Dannull J, Heiser A, Yancey D, Pruitt S, Madden J, Coleman D, Niedzwiecki D, Gilboa E,
Vieweg J (2003) Immunological and clinical responses in metastatic renal cancer patients
vaccinated with tumor RNA-transfected dendritic cells. Cancer Res 63(9):2127–2133
Tran E, Turcotte S, Gros A, Robbins PF, Lu YC, Dudley ME, Wunderlich JR, Somerville RP,
Hogan K, Hinrichs CS, Parkhurst MR, Yang JC, Rosenberg SA (2014) Cancer immunotherapy
based on mutation-specific CD4+ T cells in a patient with epithelial cancer. Science 344
(6184):641–645
Tran E, Ahmadzadeh M, Lu YC, Gros A, Turcotte S, Robbins PF, Gartner JJ, Zheng Z, Li YF,
Ray S, Wunderlich JR, Somerville RP, Rosenberg SA (2015) Immunogenicity of somatic
mutations in human gastrointestinal cancers. Science 350(6266):1387–1390
Van den Eynde B, Lethé B, Van Pel A, De Plaen E, Boon T (1991) The gene coding for a major
tumor rejection antigen of tumor P815 is identical to the normal gene of syngeneic DBA/2
mice. J Exp Med 173(6):1373–1384
van der Bruggen P, Traversari C, Chomez P, Lurquin C, De Plaen E, Van den Eynde B, Knuth A,
Boon T (1991) A gene encoding an antigen recognized by cytolytic T lymphocytes on a human
melanoma. Science 254(5038):1643–1647
Van Nuffel AM, Wilgenhof S, Thielemans K, Bonehill A (2012) Overcoming HLA restriction in
clinical trials: Immune monitoring of mRNA-loaded DC therapy. Oncoimmunology 1
(8):1392–1394
van Rooij N, van Buuren MM, Philips D, Velds A, Toebes M, Heemskerk B, van Dijk LJ,
Behjati S, Hilkmann H, El Atmioui D, Nieuwland M, Stratton MR, Kerkhoven RM, Kesmir C,
mRNA Cancer Vaccines 85