Review Open Access
Copyright ©2013 Baishideng Publishing Group Co., Limited. All rights reserved.
World J Exp Med. Nov 20, 2013; 3(4): 62-73
Published online Nov 20, 2013. doi: 10.5493/wjem.v3.i4.62
RNA vaccines for anti-tumor therapy
Fabian Benencia, Biomedical Engineering Program, Russ College of Engineering and Technology, Ohio University, Athens, OH 45701, United States
Fabian Benencia, Molecular and Cell Biology Program, Ohio University, Athens, OH 45701, United States
Fabian Benencia, Department of Biomedical Sciences, Heritage College of Osteopathic Medicine Academic Research Center, Ohio University, Athens, OH 45701, United States
Author contributions: Benencia F solely contributed to this paper .
Supported by Ohio University and a Research Scholarly Affairs Committee grant award to Benencia F, No. RP1206
Correspondence to: Fabian Benencia, PhD, Assistant Professor of Immunology, Department of Biomedical Sciences, Heritage College of Osteopathic Medicine Academic Research Center, Ohio University, 61 Oxbow Trail, Athens, OH 45701, United States. benencia@ohio.edu
Telephone: +1-740-5972133 Fax: +1-740-5972778
Received: July 30, 2013
Revised: September 27, 2013
Accepted: November 2, 2013
Published online: November 20, 2013

Abstract

The immune system is able to recognize tumor antigens and this has been the basis for the development of cancer immunotherapies. The immune system can be instructed to recognize and attack tumor cells by means of vaccination strategies. One such strategy involves the delivery of tumor antigen as genetic material. Herewith we describe the use of RNA encoding tumor antigens for vaccination purposes in tumor settings. RNA has features that are interesting for vaccination. Upon transfection, the RNA has no possibility of integration into the genome, and the tumor translated proteins enter the intrinsic antigen processing pathway thus enabling presentation by MHC-I molecules. This can specifically activate cytotoxic CD8 T cells that can attack and kill tumor cells. RNA can be delivered as a naked molecule for vaccination purposes or can be used to transfect dendritic cells. The combination of RNA technology with dendritic cell vaccination provides a powerful tool for cancer immunotherapies.

Key Words: RNA, Vaccine, Dendritic cells, Immunization, Cytotoxic T cells, Tumors

Core tip: In this review we discuss the use of RNA encoding tumor antigens for anti-tumor vaccination. RNA has several features that makes it relevant for vaccination purposes. Importantly, the RNA has no possibility of integration into the genome, and the tumor translated proteins enter the intrinsic antigen processing pathway thus enabling presentation by MHC-I molecules thus specifically activating cytotoxic CD8 T. Further, RNA can be delivered as a naked molecule f or can be used to transfect dendritic cells. This combination of RNA technology with dendritic cell vaccination provides a powerful tool for cancer immunotherapies.



TUMOR IMMUNOLOGY

Cancer is one of the leading causes of mortality in humans and most of the successes obtained battling this disease rely on early prevention even though a gamut of treatments such as chemotherapy, radiotherapy and surgery are available to patients. In view of this situation it becomes necessary to generate innovative approaches for the treatment of this disease. One such strategy entails educating the immune system to recognize and destroy tumor cells. To this end, several immunotherapeutic strategies have been designed and tested in preclinical studies and clinical trials.

Tumors are composed not only by cancer cells, but also by other cellular types such as fibroblasts, endothelial cells and infiltrating leukocytes that together with extracellular matrix components constitute the microenvironment of the tumor[1]. In recent years the relevance of the tumor microenvironment as a key player in cancer progression has been highlighted and the role of its cellular populations and extracellular matrix components examined. In this context, immune cells play a double edge sword role[2].

On one hand, the protective role of the immune system against tumors has been widely described and indeed the presence of tumor-infiltrating lymphocytes (TILs) has been reported in numerous studies involving melanoma[3], colorectal[4-7], breast[8,9], ovarian[10-16], prostate[17], renal[18], and esophageal carcinoma[19]. These TILs are able to recognize tumors as demonstrated by their capability to get activated by tumor antigens and kill cancer cells ex vivo[10,20-22]. Notably, several reports showed that the prevalence of certain T cell populations is associated with a better outcome in different types of cancers. Particularly, studies involving ovarian, non-small cell lung, mesothelioma, colon, and urothelial cancers showed that a high CD8/regulatory T cell ratio among TILs is usually associated with a better prognostic or a better response to antitumor treatment[14,23-28].

On the other hand, the presence of a robust number of regulatory T cells within the TILs, or a CD4/CD8 ratio that favors CD4 T cells, has been associated with a worse outcome or tumor growth in various studies[29-33]. These studies highlight the ability of the immune system to recognize tumors and provide a rationale for pursuing immunotherapeutic approaches, but also underscore the hurdles for its success. Similarly, other tumor-associated leukocytes such as myeloid-derived suppressor cells (MDSCs) can promote tumor growth by modulating the immune response[34]. Indeed, we have previously demonstrated the relevance of the tumor microenvironment in attracting MDSCs by a complement-mediated process[35]. Further, the presence of a subset of splenic dendritic cells (DCs) with the ability to suppress antitumor T cells responses via indoleamine 2,3-dioxigenase expression highlights the immunosuppressive role of antigen presenting cells (APCs) in some tumor settings[36]. Notably, leukocyte infiltration can precede the development of a neoplasm, being chronic inflammation a risk factor for the development of cancer[37-39]. Further, inflammatory conditions such as caused by certain types of infections can be involved in the pathogenesis of many human malignancies. For example, gastric carcinomas can arise in a Helicobacter pylori-induced gastritis environment[38] or hepatitis B virus/hepatitis C virus can induce hepatocellular carcinomas[39]. Also, chronic but non-infective inflammatory conditions as in the case of smoking-related bronchial cancer can induce carcinogenesis[40]. In the same way, chronic pancreatitis is considered a risk factor for the development of pancreatic cancer, and many of the growth factors involved in tissue remodeling and regeneration in chronic pancreatitis are present in pancreatic cancer[41]. In addition, there is strong evidence that tumor-associated leukocytes can also promote tumor angiogenesis. In particular, infiltrating inflammatory cells secrete a diverse repertoire of growth factors and proteases that potentiate tumor growth by stimulating angiogenesis. We and others have described the capability of APCs such as DCs or macrophages, to collaborate with neoangiogenesis in human cancers and in different mouse tumor models[42-47]. Thus, tumors exhibit an arsenal of mechanisms in order to inhibit an effective immune response.

Collectively, these data indicate that in some settings immunoablative procedures must precede immunotherapeutic treatments. To this end, some studies have suggested that depletion of regulatory T cell populations or tumor-associated leukocytes can enhance the effectiveness of a subsequent immunotherapy[3,48].

TUMOR IMMUNOTHERAPY

The ability of the immune system to recognize and attack tumors relies on the presence of tumor-specific antigens (TSAs) and tumor-associated antigens (TAAs). As recently reviewed by Aly[49], TSAs are expressed only by tumor cells due to mutations in normal cellular genes, or to the expression of viral antigens or normally suppressed oncogenes in cancer cells. On the other hand, TAAs are molecules expressed both in normal and cancer cells but expressed at higher levels by tumors, or expressed by normal cells only during the embryonic state differentiation. For the purposes of the present review, tumor antigens will be named generically as TAAs.

Pioneering studies performed by Rosenberg et al[50] in melanoma aimed to activate lymphocytes in vivo by treating cancer patients with IL-2. The rationale being that the patients’ T cells have the ability to recognize and attack tumors. Indeed, this is the basis for immunotherapies using TILs. To carry out these T cell adoptive therapies, upon purification from tumor tissues, TILs are expanded and activated ex vivo using TAAs and are subsequently re-infused into patients[51]. Recent advances in this area involve the generation of TAA-specific T cells by means of genetic recombination. As previously described in detail, chimeric antigen receptor (CAR) T cells are engineered to express the portion of an antibody that recognizes an antigen fused to the T cell receptor signaling region[52]. Thus, they recognize TAA on tumor cells with the specificity of an antibody and they kill them using the cytotoxic machinery of T cells[52]. This circumvents the problem of isolating TILs, which might not be present in all patients or present at very low numbers in tumor samples. Recently, by using CAR T cells, Kalos et al[53] were able to completely eradicate cancer cells in patients with advanced leukemia.

Additional immunotherapeutic strategies have been proposed and investigated based on the ability of the immune system to recognize TAAs. One such strategy involves inducing immune responses against TAA by means of vaccination. To this end, TAAs are used as tumor lysates, proteins purified from these lysates, or peptides (derived from tumor protein digests or synthesized in vitro). Furthermore, as described below, tumor vaccination strategies also involve the use of apoptotic or necrotic tumor cells as way of delivering the TAAs. These molecules will be recognized in vivo by resident APCs, which are key components of the innate immune system. The innate immunity is the first line of defense against pathogens. Cells of the innate immune response include macrophages, granulocytes, DCs, and natural killer cells. Macrophages, B lymphocytes and DCs are generally described as APCs. After ingesting a pathogen, APCs are able to eliminate it through various mechanisms involving enzymatic degradation and the use of reactive oxygen or nitrogen species. APCs detect pathogens through the expression of pattern recognition receptors (PRRs) which are able to recognize conserved pathogen associated molecular patterns (PAMPs). Some of the main PRRs include membrane associated toll-like receptors (TLRs) and cytoplasmic NOD-like receptors[54,55]. DCs are highly effective APCs distributed throughout the body, particularly in immunological organs such as thymus, spleen, lymph nodes and Peyer’s patches[56-58].

DC ACTIVATION PROCESS

Immature (non-activated) DCs present in peripheral tissues can detect PAMP-bearing microorganisms through their high expression of cell surface, vesicular and cytoplasmic PRRs[59]. This process leads to the activation of the DCs, which can degrade pathogenic proteins (both recovered from the extracellular space, or from the cytoplasmic pool) and process them into peptides[58]. Antigenic peptide fragments derived from the processed pathogen molecules are the exposed on the surface of the DCs in the context of MHC I or II molecules. During this process, an immature DC will undergo “maturation” due to presence of inflammatory cytokines generated by the DC itself, or by other surrounding cells in response to the pathogen or tissular damage. This maturation process entails upregulation of MHC class II molecules, costimulatory molecules such as CD40, CD80, CD86; OX40L and the chemokine receptor CCR7. This receptor recognizes the chemokines CCL19 and CCL21 which are constitutively expressed at high levels by lymph nodes[60]. Thus, mature DCs migrate from the sites of antigen capture to the T-cell regions of draining lymph nodes, where they contact naïve or memory T cells. Through interaction with specific cell receptors for antigen on the surface of T lymphocytes, DCs select and activate specific T cell clones with the capability to recognize the presented antigen[58,61,62]. In this way, DCs tie the innate and adaptive immunity, being keystones for the development of antigen specific immune responses.

APCs have different ways of processing and presenting antigens. Typically, antigens that are captured by the phagocytosis or endocytosis are degraded in the lysosomal compartment and peptides are presented by MHC-II molecules on the surface of the cells thus interacting and activating CD4 T cells. On the other hand, antigens generated within the cells for example as a result of a viral infection, can be degraded by the proteosome and the peptides presented on the surface of the cell in the context of MHC I molecules[55,61,63]. This strategy selects and activates antigen specific CD8 T cells[55,61,63]. Notably, DCs have the capability to cross-present antigens[64]. This means that DCs can acquire extracellular antigens, like for example apoptotic or necrotic tumor cells, or tumor lysates and also present them to CD8 T cells in the context of MHC I molecules.

DCS AND ANTI-TUMOR THERAPY

A multitude of preclinical studies and clinical trials have been designed in order to determine the anti-tumor efficacy and safety of DC-based vaccines[65]. The development of a successful DC-based tumor vaccination depends heavily on generating robust and long lasting specific CD4 and CD8 T cell responses[66]. To accomplish this, DCs have been generated from bone marrow precursors in the mouse and mostly from monocytes in humans as we previously reviewed[1]. Different steps in the antigen presentation process have been evaluated such as antigen loading, DC maturation, and delivery route and dose scheme as we have recently reviewed[1]. One strategy for loading DCs with TAAs in the mouse model involves pulsing the cells with peptides derived from tumor antigens[67]. In addition, since TAAs are not well characterized for the majority of tumors, vaccines can be prepared with whole tumor antigens[68,69]. To this end, DCs have been loaded with whole tumor lysates[70], apoptotic or necrotic cells[71] alone or conjugated with TLR ligands[72], antigens coated with antibodies to target them to Fcγ receptors[73] or peptides encapsulated in biodegradable polymers[74]. We have showed that inducing the expression of danger signals in tumor cells by means of replication-deficient or replication-restricted virus appears also to be an efficient method to pulse DCs for vaccination purposes, probably by upregulating danger signals in the tumor cells[71]. Finally, other strategies such as fusing DCs with tumor cells have also been successfully pursued[75]. These fused cells express tumor antigen but had the machinery of the DCs to present these antigens to T cells.

This information regarding DC-based antitumor vaccines pulsing has been translated to the human, where clinical trials have involved, among others, DCs pulsed with peptides[76], whole tumor lysates[77], or fused with tumor cells[78-80]. Other strategies involved pulsing human DCs with apoptotic or necrotic cells[81-90]. As we have previously reviewed[91] controversy exists regarding whether necrotic or apoptotic cells are better for pulsing DCs for tumor vaccination purposes[90,92-94]. Nevertheless, inducing tumor cell death by exposure to ultraviolet-B radiation seems to provide a mixture of apoptotic and necrotic cells suitable for vaccination purposes DCs[95,96].

TAA AS GENETIC MATERIAL

Another vaccination strategy entails delivering TAAs as the genetic material that encodes their synthesis. Thus, either DNA or RNA carrying the information to synthesize TAAs can be administered to laboratory animals in preclinical studies or to patients under clinical trials with the aim to induce local synthesis of TAAs. In contrast to delivery of TAAs as protein/peptide formulations, the recombinant antigens synthesized in the cytosol of the cells may enter the degradation process of intracellular molecules, yielding peptides that can be directly presented by MHC I molecules hence inducing a robust CD8 (cytotoxic) T cell immune response. To this end, numerous studies have been performed in order to determine the effectiveness of DNA vaccination in tumor settings[49,97,98]. The genetic material can be administered in vivo by using different techniques such gene gun, ultrasound, electroporation, cationic liposomes, and nanoparticles[99]. Alternatively, viral vectors can deliver DNA encoding for TAAs directly to the DCs. Viral vectors used to transduce human DCs[100] include recombinant adenoviruses[101-103], poxviruses[104], and retrovirus[100]. Lentiviruses have also been used to induce stable transduction of human hematopoietic stem cells or DCs[105,106]. These vectors have the advantage of infecting non-dividing cells, therefore being excellent tools to express different molecules in terminally differentiated DCs which have lost the capability to duplicate. Moreover, hematopoietic stem cells have been transduced with lentiviruses and then differentiated into antigen-expressing DCs[107]. The full scope of DNA vaccination has been extensively reviewed in the literature and will not be discussed here.

RNA VACCINES

An alternative approach for delivering TAAs as genetic material is the use of RNA for vaccinations. The advantage of RNA vaccination in comparison to DNA vaccination is that there is no danger of genome integration with the latent possibility of oncogene activation, and that there is no need to engineer expression vectors for delivery. On the contrary the expression of the antigens in the context of RNA delivery is transient, and then RNA is very labile as compared to DNA. Both DNA and RNA vaccines in addition to carrying TAAs have the potential to non-specifically stimulate the immune response upon recognition of CPG sequences by TLR9 (DNA) or by activation of TLR3 (RNA). RNA vaccination strategies involve naked RNA delivery or the pulsing of DCs with RNA molecules. Further, both whole tumor RNA or TAA specific RNA have been used as inducers of antitumor immunity.

VACCINES WITH NAKED RNA
Murine studies

Several murine studies describe the use of naked RNA for vaccination purposes. The naked RNA can be administered by injection or delivered intradermally through electroporation[108]. In order to decrease degradation, the RNA has been complexed with histidine-rich cationic polymers and histidylated cationic lipids. In this case, systemic injections of specific synthetic messenger(m) RNA encoding the human melanoma MART-1 TAA complexed with polyethylene glycol ylated histidine-rich polylysine and histidylated liposomes (termed lipopolyplexes) were able to delay the growth of B16F10 melanoma in the mouse model[109]. Notably, intravenous injection of mannosylated liposomes containing mRNA encoding for the EGFP protein proved to be taken up by spleen DCs. Further, when mRNA for MART-1 was complexed into these mannosylated liposomes, a decrease in the growth of B6F10 murine melanoma tumors was observed[110].

Another strategy is to deliver naked RNA that could simultaneously activate the immune response by way of TLR signaling. These kind of vaccines are called “two component” since they deliver TAAs while simultaneously activating the immune response. It has been reported that two component OVA-encoding RNA vaccines containing free and protamine-complexed mRNA induced specific immune responses activating both humoral and cellular immune responses against OVA-expressing tumors[111]. In addition, naked RNA can be injected systemically, or can be administered directly to sites harboring high concentration of immune cells by means of intranodal injection[112,113]. This strategy aims to directly target APCs in the site where they interact with T cells.

An innovative approach to RNA vaccine immunotherapy has been the developing of self-replicating RNA vectors (replicons). These vectors encode for a RNA-dependent RNA polymerase derived from alphaviruses which has the capability to amplify a plasmid-encoded TAA RNA[114]. This increases the availability of TAA RNA and consequently, TAA protein availability. In addition, this counteracts the high degradation that naked RNA is subjected to upon injection. Immunization with RNA replicons encoding for HPV antigens was able to decrease the growth of aggressive TC1 tumors, which carry HPV E6 and E7 antigens[115].

Human studies

Naked RNA vaccinations have been assayed in clinical settings. In particular, naked RNA encoding for several TAAs has been delivered intradermally inducing expression of cytotoxic T cells in cancer patients, together with an improve on the clinical response in some individuals[116,117]. In order to enhance the effectiveness of the transfection process while protecting the RNA from degradation, naked RNA has also been delivered complexed with liposomes in human clinical studies[118]. Further, both in mouse and human studies, adjuvants that target APCs such as FLT3 and GM-CSF have been co-delivered in their protein state or as RNA together with the naked RNA vaccines in order to further activate these cells locally[118]. This strategy aims to induce a robust activation of the transfected DCs in vivo, thus potentiating their migratory potential and their ability to induce the activation of T cells capable of recognizing TAAs of interest.

USE OF RNA-PULSED DCS FOR ANTITUMOR THERAPIES
Mouse studies

Foundational studies evaluating the effectiveness of DC-based RNA vaccination in the mouse model and in humans were performed by Dr Eli Gilboa. In 1996 his group was able to demonstrate that murine DCs pulsed with whole tumor RNA were able to induce a robust antitumor immune response in a mouse model of melanoma[119]. Shortly after, they were able to demonstrate the feasibility of this approach in a preclinical setting, inducing specific T cell responses in vitro by pulsing human monocyte-derived DCs with the carcinoembryonic antigen (CEA) antigen[120]. Since then, a multitude of studies have built on these successes in order to generate efficient DC-based RNA vaccines.

In animal experimental models, the efficacy of RNA-pulsed DC vaccination has been extensively tested. Collectively, vaccinated animals showed a decrease in tumor growth together with the activation of tumor specific cell-mediated immunity. In particular, murine DCs have been pulsed with whole tumor RNA as a source of TAAs[121-125]. Interestingly, we have previously reported that DCs pulsed with whole tumor RNA are more effective in inducing antitumor immune responses than DCs loaded with equivalent amounts of apoptotic tumor cells[126]. In order to enhance antigen presentation by DCs and the consequent efficacy of the vaccination procedure, DCs have also been pulsed with specific TAA mRNA replicons[127]. As described above, these constructs aim to increase the amount of TAA RNA present in the APCs with the consequent increase in the levels of expression of the antigen.

Other strategies designed to increase the effectiveness of DC-based RNA vaccination entailed pulsing DCs with TAA mRNA together with mRNA of cytokines such as GM-CSF and particularly IL-12[128-131], the rationale being that these cytokines will potentiate the degree of activation of the pulsed DCs.

Alternative strategies focused on enhancing the processing of the nascent TAA in the transfected DCs. To this end, studies pulsing DCs with RNA encoding for TAAs fused with molecules that augment the delivery of the synthesized proteins to the endoplasmic reticulum, TAAs RNA linked with ubiquitin RNA to target the ubiquitin-proteosome pathway, MHC I and II pathways by fusion with LAMP1 or DC. LAMP sequences, or with immunogenic helper proteins such as EGFP have been used[97,132-134]. In this way, cytoplasmic TAAs will be more efficiently processed by the ER, increasing the levels of TAAs peptides presented in the context of MHC I molecules on the surface of the DCs.

Finally, others strategies to potentiate the efficacy of DC-based RNA vaccines entail the use of different maturation cocktails or immunostimulatory factors to activate the RNA-pulsed cells. For example, soluble CD40 has been shown to act as an adjuvant for cytokine treatment of RNA-pulsed DCs increasing the generation of cytotoxic T cells in a an experimental model of melanoma[135].

Human preclinical

In order to optimize the likelihood of effective translation into the clinic, human DCs have been prepared from monocytes recovered from apheresis products or by differentiation of CD34+ hematopoietic precursors[136,137]. As above, whole tumor RNA or mRNA can be used to transfect these cells by electroporation or lipofection[137,138]. In addition, RNA recovered from tumor cells lines can be used to pulse human DCs. For example, whole RNA from KL562 leukemia cells was delivered to monocyte-derived DCs by electroporation and lipofection being the transfected RNA degraded within 24 h. Notably, the translated TAA proved to be processed through the MHC-I presentation pathway rather than the endosomal-phagocytic pathway indicating that these DCs could be able to activate CD8 cytotoxic T cells[139]. Interestingly, not only monocyte or hematopoietic CD34+ derived DCs have been tested in RNA vaccination studies. Indeed, DCs directly recovered from hepatocellular carcinoma patients could be efficiently pulsed with whole RNA recovered from hepatic cancer cell lines[140].

It has been determined that better expression of TAAs after transfection with whole tumor RNA is achieved when antisense RNAs are eliminated from the whole tumor RNA preparation[141]. This highlights the need to prepare high quality RNA for transfection studies. Further, although most of DC protocols (both in mouse and human) propose to induce maturation of these cells after RNA transfection, a study suggests that RNA transfection of DCs can also be performed after maturation of these cells[142]. Taking into account studies indicating the viability of cryopreserved mature human DCs[143], this opens the possibility of transfecting DCs right before administration to patients.

Human DCs transfected with tumor RNA have been shown to elicit specific T cell responses in vitro. This was demonstrated by their ability to generate TAA specific T cell lines, or by activating ex vivo TILs recovered from cancer tissues. For example, DCs transfected with survivin or TERT RNA were used to generate CD8 cytotoxic cell lines with the capability to eliminate tumor cell lines and primary tumors in vitro[144,145]. Further, RNA recovered from prostate tumor samples by laser capture microdissection was amplified and used to transfect DCs generated from blood precursors. It was shown that these DCs were able to induce cytotoxic T cells in vitro[146].

As described above, mouse studies determined that RNA encoding for TAAs can be engineered to enhance the capability of the DCs to process the nascent antigens. To translate these results into the human setting, DCs generated from human monocytes were transfected with mRNA encoding for the TERT antigen fused with LAMP in order to augment the processing of the TAA upon translation. This strategy induced a robust activation of CD4 T cells specific for TERT as determined in in vitro studies[147].

Human clinical studies

Several clinical trials have been conducted in order to evaluate the efficacy of DC-based RNA vaccines in cancer patients. In these clinical trials, the vaccines were generated by pulsing monocyte-derived DCs either with whole tumor RNA or specific TAA RNA. Altogether, human clinical studies highlight that the administration of DC-based RNA vaccines is safe and does not induce adverse reactions. For example, in a phase I clinical trial involving acute myeloid patients aiming to generate clinical grade DC vaccines, monocyte-derived DCs were pulsed with in vitro transcribed RNA encoding the Wilm’s tumor. Then, these cells were injected repeatedly into patients by the intramuscular route[148]. The results of this study indicated that the vaccination scheme was well tolerated by the patients[148]. This was also observed in a clinical trial involving stage IV malignant melanoma patient[149]. In this case, DCs were pulsed with whole tumor RNA expanded in vitro but no positive effect of the vaccination was observed. This is no surprising taking into account the advanced stage of the illness, but nevertheless the study highlights the safety of using this procedure for antitumor therapies. Other studies showed that DC-based RNA vaccination is able to induce specific T cell responses in cancer patients. In particular, in a clinical trial involving relapsed metastatic ovarian cancer patients, DCs pulsed with mRNA specific for folate receptor α were able to induce a large population of effector memory CD8 cytolytic T cells reactive to the antigen upon repeated injections[150]. Similarly, specific T cell responses were observed in colorectal cancer patients receiving several injections of DCs harboring CEA mRNA[151]. In addition, it has been shown that patients vaccinated with DCs transfected with mRNA recovered from autologous melanoma tumor cells were capable of initiating T cell responses specific to antigens encoded by the pulsed APCs[152]. Finally, in order to ensure a robust activation of T cells, strategies designed to deliver the transfected DCs directly to the lymph nodes have been tested. In a phaseI/II clinical trial with melanoma patients it has been shown that upon intranodal administration, DCs electroporated with mRNA encoding for gp100 or tyrosinase migrate towards T cells areas of the lymph node [153].

CONCLUSION

In closing, in the last 15 years, a growing body of literature has argued for the use of RNA for vaccination purposes. Importantly, RNA is safer than DNA vaccine approaches taking into account that no possibility of genomic integration exists. Furthermore, the combination of RNA technology with DC-based vaccines has made available a powerful strategy for antitumor therapies. Advances in RNA technology (i.e., strategies to increase stability, use of replicons), together with the development of more effective protocols for generating activated DCs (i.e., use of better inflammatory cocktails) and an increase in our knowledge of tumor immunology (i.e., the use of immunoablative therapies to eliminate suppressor populations) will guide further pursuit of tumor immunotherapies using DC-based RNA vaccines. This offers the potential to advance the outcome of cancer immunotherapies for the benefit of patients.

Footnotes

P- Reviewer: Midoux P S- Editor: Zhai HH L- Editor: A E- Editor: Wang CH

References
1.  Benencia F, Sprague L, McGinty J, Pate M, Muccioli M. Dendritic cells the tumor microenvironment and the challenges for an effective antitumor vaccination. J Biomed Biotechnol. 2012;2012:425476.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 58]  [Article Influence: 4.8]  [Reference Citation Analysis (0)]
2.  Talmadge JE, Donkor M, Scholar E. Inflammatory cell infiltration of tumors: Jekyll or Hyde. Cancer Metastasis Rev. 2007;26:373-400.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 214]  [Cited by in F6Publishing: 233]  [Article Influence: 14.6]  [Reference Citation Analysis (0)]
3.  Jacobs JF, Nierkens S, Figdor CG, de Vries IJ, Adema GJ. Regulatory T cells in melanoma: the final hurdle towards effective immunotherapy? Lancet Oncol. 2012;13:e32-e42.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 176]  [Cited by in F6Publishing: 176]  [Article Influence: 14.7]  [Reference Citation Analysis (0)]
4.  Waldner M, Schimanski CC, Neurath MF. Colon cancer and the immune system: the role of tumor invading T cells. World J Gastroenterol. 2006;12:7233-7238.  [PubMed]  [DOI]  [Cited in This Article: ]
5.  Pagès F, Berger A, Camus M, Sanchez-Cabo F, Costes A, Molidor R, Mlecnik B, Kirilovsky A, Nilsson M, Damotte D. Effector memory T cells, early metastasis, and survival in colorectal cancer. N Engl J Med. 2005;353:2654-2666.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1553]  [Cited by in F6Publishing: 1556]  [Article Influence: 81.9]  [Reference Citation Analysis (0)]
6.  Galon J, Costes A, Sanchez-Cabo F, Kirilovsky A, Mlecnik B, Lagorce-Pagès C, Tosolini M, Camus M, Berger A, Wind P. Type, density, and location of immune cells within human colorectal tumors predict clinical outcome. Science. 2006;313:1960-1964.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4318]  [Cited by in F6Publishing: 4595]  [Article Influence: 255.3]  [Reference Citation Analysis (0)]
7.  Naito Y, Saito K, Shiiba K, Ohuchi A, Saigenji K, Nagura H, Ohtani H. CD8+ T cells infiltrated within cancer cell nests as a prognostic factor in human colorectal cancer. Cancer Res. 1998;58:3491-3494.  [PubMed]  [DOI]  [Cited in This Article: ]
8.  DeNardo DG, Coussens LM. Inflammation and breast cancer. Balancing immune response: crosstalk between adaptive and innate immune cells during breast cancer progression. Breast Cancer Res. 2007;9:212.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 442]  [Cited by in F6Publishing: 485]  [Article Influence: 30.3]  [Reference Citation Analysis (0)]
9.  Marrogi AJ, Munshi A, Merogi AJ, Ohadike Y, El-Habashi A, Marrogi OL, Freeman SM. Study of tumor infiltrating lymphocytes and transforming growth factor-beta as prognostic factors in breast carcinoma. Int J Cancer. 1997;74:492-501.  [PubMed]  [DOI]  [Cited in This Article: ]
10.  Conejo-Garcia JR, Benencia F, Courreges MC, Khang E, Zhang L, Mohamed-Hadley A, Vinocur JM, Buckanovich RJ, Thompson CB, Levine B. Letal, A tumor-associated NKG2D immunoreceptor ligand, induces activation and expansion of effector immune cells. Cancer Biol Ther. 2003;2:446-451.  [PubMed]  [DOI]  [Cited in This Article: ]
11.  Adams SF, Levine DA, Cadungog MG, Hammond R, Facciabene A, Olvera N, Rubin SC, Boyd J, Gimotty PA, Coukos G. Intraepithelial T cells and tumor proliferation: impact on the benefit from surgical cytoreduction in advanced serous ovarian cancer. Cancer. 2009;115:2891-2902.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 89]  [Cited by in F6Publishing: 106]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
12.  Clarke B, Tinker AV, Lee CH, Subramanian S, van de Rijn M, Turbin D, Kalloger S, Han G, Ceballos K, Cadungog MG. Intraepithelial T cells and prognosis in ovarian carcinoma: novel associations with stage, tumor type, and BRCA1 loss. Mod Pathol. 2009;22:393-402.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 204]  [Cited by in F6Publishing: 235]  [Article Influence: 15.7]  [Reference Citation Analysis (0)]
13.  Hamanishi J, Mandai M, Iwasaki M, Okazaki T, Tanaka Y, Yamaguchi K, Higuchi T, Yagi H, Takakura K, Minato N. Programmed cell death 1 ligand 1 and tumor-infiltrating CD8+ T lymphocytes are prognostic factors of human ovarian cancer. Proc Natl Acad Sci USA. 2007;104:3360-3365.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1041]  [Cited by in F6Publishing: 1143]  [Article Influence: 67.2]  [Reference Citation Analysis (0)]
14.  Sato E, Olson SH, Ahn J, Bundy B, Nishikawa H, Qian F, Jungbluth AA, Frosina D, Gnjatic S, Ambrosone C. Intraepithelial CD8+ tumor-infiltrating lymphocytes and a high CD8+/regulatory T cell ratio are associated with favorable prognosis in ovarian cancer. Proc Natl Acad Sci USA. 2005;102:18538-18543.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1705]  [Cited by in F6Publishing: 1835]  [Article Influence: 96.6]  [Reference Citation Analysis (0)]
15.  Shah CA, Allison KH, Garcia RL, Gray HJ, Goff BA, Swisher EM. Intratumoral T cells, tumor-associated macrophages, and regulatory T cells: association with p53 mutations, circulating tumor DNA and survival in women with ovarian cancer. Gynecol Oncol. 2008;109:215-219.  [PubMed]  [DOI]  [Cited in This Article: ]
16.  Tomsová M, Melichar B, Sedláková I, Steiner I. Prognostic significance of CD3+ tumor-infiltrating lymphocytes in ovarian carcinoma. Gynecol Oncol. 2008;108:415-420.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 144]  [Cited by in F6Publishing: 155]  [Article Influence: 9.1]  [Reference Citation Analysis (0)]
17.  Vesalainen S, Lipponen P, Talja M, Syrjänen K. Histological grade, perineural infiltration, tumour-infiltrating lymphocytes and apoptosis as determinants of long-term prognosis in prostatic adenocarcinoma. Eur J Cancer. 1994;30A:1797-1803.  [PubMed]  [DOI]  [Cited in This Article: ]
18.  Nakano O, Sato M, Naito Y, Suzuki K, Orikasa S, Aizawa M, Suzuki Y, Shintaku I, Nagura H, Ohtani H. Proliferative activity of intratumoral CD8(+) T-lymphocytes as a prognostic factor in human renal cell carcinoma: clinicopathologic demonstration of antitumor immunity. Cancer Res. 2001;61:5132-5136.  [PubMed]  [DOI]  [Cited in This Article: ]
19.  Schumacher K, Haensch W, Röefzaad C, Schlag PM. Prognostic significance of activated CD8(+) T cell infiltrations within esophageal carcinomas. Cancer Res. 2001;61:3932-3936.  [PubMed]  [DOI]  [Cited in This Article: ]
20.  Freedman RS, Tomasovic B, Templin S, Atkinson EN, Kudelka A, Edwards CL, Platsoucas CD. Large-scale expansion in interleukin-2 of tumor-infiltrating lymphocytes from patients with ovarian carcinoma for adoptive immunotherapy. J Immunol Methods. 1994;167:145-160.  [PubMed]  [DOI]  [Cited in This Article: ]
21.  Ioannides CG, Freedman RS, Platsoucas CD, Rashed S, Kim YP. Cytotoxic T cell clones isolated from ovarian tumor-infiltrating lymphocytes recognize multiple antigenic epitopes on autologous tumor cells. J Immunol. 1991;146:1700-1707.  [PubMed]  [DOI]  [Cited in This Article: ]
22.  Ioannides CG, Platsoucas CD, Rashed S, Wharton JT, Edwards CL, Freedman RS. Tumor cytolysis by lymphocytes infiltrating ovarian malignant ascites. Cancer Res. 1991;51:4257-4265.  [PubMed]  [DOI]  [Cited in This Article: ]
23.  Liu H, Zhang T, Ye J, Li H, Huang J, Li X, Wu B, Huang X, Hou J. Tumor-infiltrating lymphocytes predict response to chemotherapy in patients with advance non-small cell lung cancer. Cancer Immunol Immunother. 2012;61:1849-1856.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 99]  [Article Influence: 8.3]  [Reference Citation Analysis (0)]
24.  Hwang WT, Adams SF, Tahirovic E, Hagemann IS, Coukos G. Prognostic significance of tumor-infiltrating T cells in ovarian cancer: a meta-analysis. Gynecol Oncol. 2012;124:192-198.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 403]  [Cited by in F6Publishing: 490]  [Article Influence: 37.7]  [Reference Citation Analysis (0)]
25.  Yamada N, Oizumi S, Kikuchi E, Shinagawa N, Konishi-Sakakibara J, Ishimine A, Aoe K, Gemba K, Kishimoto T, Torigoe T. CD8+ tumor-infiltrating lymphocytes predict favorable prognosis in malignant pleural mesothelioma after resection. Cancer Immunol Immunother. 2010;59:1543-1549.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 128]  [Cited by in F6Publishing: 131]  [Article Influence: 9.4]  [Reference Citation Analysis (0)]
26.  Stumpf M, Hasenburg A, Riener MO, Jütting U, Wang C, Shen Y, Orlowska-Volk M, Fisch P, Wang Z, Gitsch G. Intraepithelial CD8-positive T lymphocytes predict survival for patients with serous stage III ovarian carcinomas: relevance of clonal selection of T lymphocytes. Br J Cancer. 2009;101:1513-1521.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 77]  [Cited by in F6Publishing: 87]  [Article Influence: 5.8]  [Reference Citation Analysis (0)]
27.  Morris M, Platell C, Iacopetta B. Tumor-infiltrating lymphocytes and perforation in colon cancer predict positive response to 5-fluorouracil chemotherapy. Clin Cancer Res. 2008;14:1413-1417.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 88]  [Cited by in F6Publishing: 96]  [Article Influence: 6.0]  [Reference Citation Analysis (0)]
28.  Fukunaga A, Miyamoto M, Cho Y, Murakami S, Kawarada Y, Oshikiri T, Kato K, Kurokawa T, Suzuoki M, Nakakubo Y. CD8+ tumor-infiltrating lymphocytes together with CD4+ tumor-infiltrating lymphocytes and dendritic cells improve the prognosis of patients with pancreatic adenocarcinoma. Pancreas. 2004;28:e26-e31.  [PubMed]  [DOI]  [Cited in This Article: ]
29.  Kim HI, Kim H, Cho HW, Kim SY, Song KJ, Hyung WJ, Park CG, Kim CB. The ratio of intra-tumoral regulatory T cells (Foxp3+)/helper T cells (CD4+) is a prognostic factor and associated with recurrence pattern in gastric cardia cancer. J Surg Oncol. 2011;104:728-733.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 38]  [Cited by in F6Publishing: 41]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
30.  Chen KJ, Zhou L, Xie HY, Ahmed TE, Feng XW, Zheng SS. Intratumoral regulatory T cells alone or in combination with cytotoxic T cells predict prognosis of hepatocellular carcinoma after resection. Med Oncol. 2012;29:1817-1826.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 67]  [Cited by in F6Publishing: 72]  [Article Influence: 5.5]  [Reference Citation Analysis (0)]
31.  Shah W, Yan X, Jing L, Zhou Y, Chen H, Wang Y. A reversed CD4/CD8 ratio of tumor-infiltrating lymphocytes and a high percentage of CD4(+)FOXP3(+) regulatory T cells are significantly associated with clinical outcome in squamous cell carcinoma of the cervix. Cell Mol Immunol. 2011;8:59-66.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 152]  [Cited by in F6Publishing: 188]  [Article Influence: 13.4]  [Reference Citation Analysis (0)]
32.  Liotta F, Gacci M, Frosali F, Querci V, Vittori G, Lapini A, Santarlasci V, Serni S, Cosmi L, Maggi L. Frequency of regulatory T cells in peripheral blood and in tumour-infiltrating lymphocytes correlates with poor prognosis in renal cell carcinoma. BJU Int. 2011;107:1500-1506.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 81]  [Cited by in F6Publishing: 88]  [Article Influence: 6.3]  [Reference Citation Analysis (0)]
33.  Shen Z, Zhou S, Wang Y, Li RL, Zhong C, Liang C, Sun Y. Higher intratumoral infiltrated Foxp3+ Treg numbers and Foxp3+/CD8+ ratio are associated with adverse prognosis in resectable gastric cancer. J Cancer Res Clin Oncol. 2010;136:1585-1595.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 143]  [Cited by in F6Publishing: 158]  [Article Influence: 11.3]  [Reference Citation Analysis (0)]
34.  Condamine T, Gabrilovich DI. Molecular mechanisms regulating myeloid-derived suppressor cell differentiation and function. Trends Immunol. 2011;32:19-25.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 567]  [Cited by in F6Publishing: 634]  [Article Influence: 45.3]  [Reference Citation Analysis (0)]
35.  Markiewski MM, DeAngelis RA, Benencia F, Ricklin-Lichtsteiner SK, Koutoulaki A, Gerard C, Coukos G, Lambris JD. Modulation of the antitumor immune response by complement. Nat Immunol. 2008;9:1225-1235.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 492]  [Cited by in F6Publishing: 532]  [Article Influence: 33.3]  [Reference Citation Analysis (0)]
36.  Baban B, Hansen AM, Chandler PR, Manlapat A, Bingaman A, Kahler DJ, Munn DH, Mellor AL. A minor population of splenic dendritic cells expressing CD19 mediates IDO-dependent T cell suppression via type I IFN signaling following B7 ligation. Int Immunol. 2005;17:909-919.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 148]  [Cited by in F6Publishing: 153]  [Article Influence: 8.1]  [Reference Citation Analysis (0)]
37.  Rüegg C. Leukocytes, inflammation, and angiogenesis in cancer: fatal attractions. J Leukoc Biol. 2006;80:682-684.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 56]  [Article Influence: 3.1]  [Reference Citation Analysis (0)]
38.  Peek RM, Crabtree JE. Helicobacter infection and gastric neoplasia. J Pathol. 2006;208:233-248.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 368]  [Cited by in F6Publishing: 417]  [Article Influence: 21.9]  [Reference Citation Analysis (0)]
39.  Szabó E, Páska C, Kaposi Novák P, Schaff Z, Kiss A. Similarities and differences in hepatitis B and C virus induced hepatocarcinogenesis. Pathol Oncol Res. 2004;10:5-11.  [PubMed]  [DOI]  [Cited in This Article: ]
40.  Williams MD, Sandler AB. The epidemiology of lung cancer. Cancer Treat Res. 2001;105:31-52.  [PubMed]  [DOI]  [Cited in This Article: ]
41.  Jura N, Archer H, Bar-Sagi D. Chronic pancreatitis, pancreatic adenocarcinoma and the black box in-between. Cell Res. 2005;15:72-77.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 64]  [Article Influence: 3.4]  [Reference Citation Analysis (0)]
42.  Conejo-Garcia JR, Benencia F, Courreges MC, Kang E, Mohamed-Hadley A, Buckanovich RJ, Holtz DO, Jenkins A, Na H, Zhang L. Tumor-infiltrating dendritic cell precursors recruited by a beta-defensin contribute to vasculogenesis under the influence of Vegf-A. Nat Med. 2004;10:950-958.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 340]  [Cited by in F6Publishing: 342]  [Article Influence: 17.1]  [Reference Citation Analysis (0)]
43.  Curiel TJ, Cheng P, Mottram P, Alvarez X, Moons L, Evdemon-Hogan M, Wei S, Zou L, Kryczek I, Hoyle G. Dendritic cell subsets differentially regulate angiogenesis in human ovarian cancer. Cancer Res. 2004;64:5535-5538.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
44.  Ribatti D. The paracrine role of Tie-2-expressing monocytes in tumor angiogenesis. Stem Cells Dev. 2009;18:703-706.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 16]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
45.  Riboldi E, Musso T, Moroni E, Urbinati C, Bernasconi S, Rusnati M, Adorini L, Presta M, Sozzani S. Cutting edge: proangiogenic properties of alternatively activated dendritic cells. J Immunol. 2005;175:2788-2792.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
46.  Conejo-Garcia JR, Buckanovich RJ, Benencia F, Courreges MC, Rubin SC, Carroll RG, Coukos G. Vascular leukocytes contribute to tumor vascularization. Blood. 2005;105:679-681.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 144]  [Cited by in F6Publishing: 151]  [Article Influence: 7.6]  [Reference Citation Analysis (0)]
47.  Gough PJ, Gomez IG, Wille PT, Raines EW. Macrophage expression of active MMP-9 induces acute plaque disruption in apoE-deficient mice. J Clin Invest. 2006;116:59-69.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 307]  [Cited by in F6Publishing: 321]  [Article Influence: 16.9]  [Reference Citation Analysis (0)]
48.  Allavena P, Mantovani A. Immunology in the clinic review series; focus on cancer: tumour-associated macrophages: undisputed stars of the inflammatory tumour microenvironment. Clin Exp Immunol. 2012;167:195-205.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 283]  [Cited by in F6Publishing: 282]  [Article Influence: 23.5]  [Reference Citation Analysis (0)]
49.  Aly HA. Cancer therapy and vaccination. J Immunol Methods. 2012;382:1-23.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 43]  [Cited by in F6Publishing: 45]  [Article Influence: 3.8]  [Reference Citation Analysis (0)]
50.  Rosenberg SA, Mulé JJ, Spiess PJ, Reichert CM, Schwarz SL. Regression of established pulmonary metastases and subcutaneous tumor mediated by the systemic administration of high-dose recombinant interleukin 2. J Exp Med. 1985;161:1169-1188.  [PubMed]  [DOI]  [Cited in This Article: ]
51.  Freedman RS, Platsoucas CD. Immunotherapy for peritoneal ovarian carcinoma metastasis using ex vivo expanded tumor infiltrating lymphocytes. Cancer Treat Res. 1996;82:115-146.  [PubMed]  [DOI]  [Cited in This Article: ]
52.  Park TS, Rosenberg SA, Morgan RA. Treating cancer with genetically engineered T cells. Trends Biotechnol. 2011;29:550-557.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 155]  [Cited by in F6Publishing: 174]  [Article Influence: 13.4]  [Reference Citation Analysis (0)]
53.  Kalos M, Levine BL, Porter DL, Katz S, Grupp SA, Bagg A, June CH. T cells with chimeric antigen receptors have potent antitumor effects and can establish memory in patients with advanced leukemia. Sci Transl Med. 2011;3:95ra73.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
54.  Feldmann M, Maini RN. Discovery of TNF-alpha as a therapeutic target in rheumatoid arthritis: preclinical and clinical studies. Joint Bone Spine. 2002;69:12-18.  [PubMed]  [DOI]  [Cited in This Article: ]
55.  Kapsenberg ML. Dendritic-cell control of pathogen-driven T-cell polarization. Nat Rev Immunol. 2003;3:984-993.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1000]  [Cited by in F6Publishing: 984]  [Article Influence: 49.2]  [Reference Citation Analysis (0)]
56.  Bonasio R, von Andrian UH. Generation, migration and function of circulating dendritic cells. Curr Opin Immunol. 2006;18:503-511.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 91]  [Cited by in F6Publishing: 85]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
57.  Lanzavecchia A, Sallusto F. The instructive role of dendritic cells on T cell responses: lineages, plasticity and kinetics. Curr Opin Immunol. 2001;13:291-298.  [PubMed]  [DOI]  [Cited in This Article: ]
58.  Banchereau J, Briere F, Caux C, Davoust J, Lebecque S, Liu YJ, Pulendran B, Palucka K. Immunobiology of dendritic cells. Annu Rev Immunol. 2000;18:767-811.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4895]  [Cited by in F6Publishing: 4737]  [Article Influence: 197.4]  [Reference Citation Analysis (0)]
59.  Doherty TM, Fisher EA, Arditi M. TLR signaling and trapped vascular dendritic cells in the development of atherosclerosis. Trends Immunol. 2006;27:222-227.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 34]  [Cited by in F6Publishing: 37]  [Article Influence: 2.1]  [Reference Citation Analysis (0)]
60.  Sánchez-Sánchez N, Riol-Blanco L, de la Rosa G, Puig-Kröger A, García-Bordas J, Martín D, Longo N, Cuadrado A, Cabañas C, Corbí AL. Chemokine receptor CCR7 induces intracellular signaling that inhibits apoptosis of mature dendritic cells. Blood. 2004;104:619-625.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 130]  [Cited by in F6Publishing: 136]  [Article Influence: 6.8]  [Reference Citation Analysis (0)]
61.  Kadowaki N. Dendritic cells: a conductor of T cell differentiation. Allergol Int. 2007;56:193-199.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 70]  [Cited by in F6Publishing: 70]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
62.  Timmerman JM, Levy R. Dendritic cell vaccines for cancer immunotherapy. Annu Rev Med. 1999;50:507-529.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 356]  [Cited by in F6Publishing: 344]  [Article Influence: 13.8]  [Reference Citation Analysis (0)]
63.  Cahalan MD, Parker I. Close encounters of the first and second kind: T-DC and T-B interactions in the lymph node. Semin Immunol. 2005;17:442-451.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 40]  [Cited by in F6Publishing: 44]  [Article Influence: 2.3]  [Reference Citation Analysis (0)]
64.  Dresch C, Leverrier Y, Marvel J, Shortman K. Development of antigen cross-presentation capacity in dendritic cells. Trends Immunol. 2012;33:381-388.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 51]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
65.  Ridgway D. The first 1000 dendritic cell vaccinees. Cancer Invest. 2003;21:873-886.  [PubMed]  [DOI]  [Cited in This Article: ]
66.  Gilboa E. DC-based cancer vaccines. J Clin Invest. 2007;117:1195-1203.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 442]  [Cited by in F6Publishing: 437]  [Article Influence: 25.7]  [Reference Citation Analysis (0)]
67.  Yamaguchi S, Tatsumi T, Takehara T, Sasakawa A, Hikita H, Kohga K, Uemura A, Sakamori R, Ohkawa K, Hayashi N. Dendritic cell-based vaccines suppress metastatic liver tumor via activation of local innate and acquired immunity. Cancer Immunol Immunother. 2008;57:1861-1869.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 17]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
68.  Hsueh EC. Tumour cell-based vaccines for the treatment of melanoma. BioDrugs. 2001;15:713-720.  [PubMed]  [DOI]  [Cited in This Article: ]
69.  Scanlan MJ, Jäger D. Challenges to the development of antigen-specific breast cancer vaccines. Breast Cancer Res. 2001;3:95-98.  [PubMed]  [DOI]  [Cited in This Article: ]
70.  Hatfield P, Merrick AE, West E, O‘Donnell D, Selby P, Vile R, Melcher AA. Optimization of dendritic cell loading with tumor cell lysates for cancer immunotherapy. J Immunother. 2008;31:620-632.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
71.  Courrèges MC, Benencia F, Conejo-García JR, Zhang L, Coukos G. Preparation of apoptotic tumor cells with replication-incompetent HSV augments the efficacy of dendritic cell vaccines. Cancer Gene Ther. 2006;13:182-193.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 17]  [Article Influence: 0.9]  [Reference Citation Analysis (0)]
72.  Shirota H, Klinman DM. CpG-conjugated apoptotic tumor cells elicit potent tumor-specific immunity. Cancer Immunol Immunother. 2011;60:659-669.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 27]  [Cited by in F6Publishing: 31]  [Article Influence: 2.4]  [Reference Citation Analysis (0)]
73.  Pilon-Thomas S, Verhaegen M, Kuhn L, Riker A, Mulé JJ. Induction of anti-tumor immunity by vaccination with dendritic cells pulsed with anti-CD44 IgG opsonized tumor cells. Cancer Immunol Immunother. 2006;55:1238-1246.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 14]  [Article Influence: 0.7]  [Reference Citation Analysis (0)]
74.  Ma W, Smith T, Bogin V, Zhang Y, Ozkan C, Ozkan M, Hayden M, Schroter S, Carrier E, Messmer D. Enhanced presentation of MHC class Ia, Ib and class II-restricted peptides encapsulated in biodegradable nanoparticles: a promising strategy for tumor immunotherapy. J Transl Med. 2011;9:34.  [PubMed]  [DOI]  [Cited in This Article: ]
75.  Xu F, Ye YJ, Liu W, Kong M, He Y, Wang S. Dendritic cell/tumor hybrids enhances therapeutic efficacy against colorectal cancer liver metastasis in SCID mice. Scand J Gastroenterol. 2010;45:707-713.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 13]  [Cited by in F6Publishing: 15]  [Article Influence: 1.1]  [Reference Citation Analysis (0)]
76.  Wierecky J, Müller MR, Wirths S, Halder-Oehler E, Dörfel D, Schmidt SM, Häntschel M, Brugger W, Schröder S, Horger MS. Immunologic and clinical responses after vaccinations with peptide-pulsed dendritic cells in metastatic renal cancer patients. Cancer Res. 2006;66:5910-5918.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 175]  [Cited by in F6Publishing: 185]  [Article Influence: 10.3]  [Reference Citation Analysis (0)]
77.  Ovali E, Dikmen T, Sonmez M, Yilmaz M, Unal A, Dalbasti T, Kuzeyli K, Erturk M, Omay SB. Active immunotherapy for cancer patients using tumor lysate pulsed dendritic cell vaccine: a safety study. J Exp Clin Cancer Res. 2007;26:209-214.  [PubMed]  [DOI]  [Cited in This Article: ]
78.  Akasaki Y, Kikuchi T, Irie M, Yamamoto Y, Arai T, Tanaka T, Joki T, Abe T. Cotransfection of Poly(I: C) and siRNA of IL-10 into fusions of dendritic and glioma cells enhances antitumor T helper type 1 induction in patients with glioma. J Immunother. 2011;34:121-128.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 13]  [Article Influence: 1.0]  [Reference Citation Analysis (0)]
79.  Rosenblatt J, Vasir B, Uhl L, Blotta S, Macnamara C, Somaiya P, Wu Z, Joyce R, Levine JD, Dombagoda D. Vaccination with dendritic cell/tumor fusion cells results in cellular and humoral antitumor immune responses in patients with multiple myeloma. Blood. 2011;117:393-402.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 164]  [Cited by in F6Publishing: 156]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
80.  Zhang Y, Ma B, Zhou Y, Zhang M, Qiu X, Sui Y, Zhang X, Ma B, Fan Q. Dendritic cells fused with allogeneic breast cancer cell line induce tumor antigen-specific CTL responses against autologous breast cancer cells. Breast Cancer Res Treat. 2007;105:277-286.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 24]  [Cited by in F6Publishing: 26]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
81.  Kacani L, Wurm M, Schwentner I, Andrle J, Schennach H, Sprinzl GM. Maturation of dendritic cells in the presence of living, apoptotic and necrotic tumour cells derived from squamous cell carcinoma of head and neck. Oral Oncol. 2005;41:17-24.  [PubMed]  [DOI]  [Cited in This Article: ]
82.  Brusa D, Garetto S, Chiorino G, Scatolini M, Migliore E, Camussi G, Matera L. Post-apoptotic tumors are more palatable to dendritic cells and enhance their antigen cross-presentation activity. Vaccine. 2008;26:6422-6432.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 41]  [Cited by in F6Publishing: 43]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
83.  Chen W, Wang J, Shao C, Liu S, Yu Y, Wang Q, Cao X. Efficient induction of antitumor T cell immunity by exosomes derived from heat-shocked lymphoma cells. Eur J Immunol. 2006;36:1598-1607.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 129]  [Cited by in F6Publishing: 143]  [Article Influence: 7.9]  [Reference Citation Analysis (0)]
84.  Chen Z, Moyana T, Saxena A, Warrington R, Jia Z, Xiang J. Efficient antitumor immunity derived from maturation of dendritic cells that had phagocytosed apoptotic/necrotic tumor cells. Int J Cancer. 2001;93:539-548.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 115]  [Cited by in F6Publishing: 119]  [Article Influence: 5.2]  [Reference Citation Analysis (0)]
85.  Henry F, Boisteau O, Bretaudeau L, Lieubeau B, Meflah K, Grégoire M. Antigen-presenting cells that phagocytose apoptotic tumor-derived cells are potent tumor vaccines. Cancer Res. 1999;59:3329-3332.  [PubMed]  [DOI]  [Cited in This Article: ]
86.  Hoffmann TK, Meidenbauer N, Dworacki G, Kanaya H, Whiteside TL. Generation of tumor-specific T-lymphocytes by cross-priming with human dendritic cells ingesting apoptotic tumor cells. Cancer Res. 2000;60:3542-3549.  [PubMed]  [DOI]  [Cited in This Article: ]
87.  Russo V, Tanzarella S, Dalerba P, Rigatti D, Rovere P, Villa A, Bordignon C, Traversari C. Dendritic cells acquire the MAGE-3 human tumor antigen from apoptotic cells and induce a class I-restricted T cell response. Proc Natl Acad Sci USA. 2000;97:2185-2190.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 109]  [Cited by in F6Publishing: 112]  [Article Influence: 4.7]  [Reference Citation Analysis (0)]
88.  Shaif-Muthana M, McIntyre C, Sisley K, Rennie I, Murray A. Dead or alive: immunogenicity of human melanoma cells when presented by dendritic cells. Cancer Res. 2000;60:6441-6447.  [PubMed]  [DOI]  [Cited in This Article: ]
89.  Schnurr M, Scholz C, Rothenfusser S, Galambos P, Dauer M, Röbe J, Endres S, Eigler A. Apoptotic pancreatic tumor cells are superior to cell lysates in promoting cross-priming of cytotoxic T cells and activate NK and gammadelta T cells. Cancer Res. 2002;62:2347-2352.  [PubMed]  [DOI]  [Cited in This Article: ]
90.  Scheffer SR, Nave H, Korangy F, Schlote K, Pabst R, Jaffee EM, Manns MP, Greten TF. Apoptotic, but not necrotic, tumor cell vaccines induce a potent immune response in vivo. Int J Cancer. 2003;103:205-211.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 164]  [Cited by in F6Publishing: 150]  [Article Influence: 7.1]  [Reference Citation Analysis (0)]
91.  Chiang CL, Benencia F, Coukos G. Whole tumor antigen vaccines. Semin Immunol. 2010;22:132-143.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 145]  [Cited by in F6Publishing: 155]  [Article Influence: 11.1]  [Reference Citation Analysis (0)]
92.  Gallucci S, Lolkema M, Matzinger P. Natural adjuvants: endogenous activators of dendritic cells. Nat Med. 1999;5:1249-1255.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 1227]  [Cited by in F6Publishing: 1168]  [Article Influence: 46.7]  [Reference Citation Analysis (0)]
93.  Basu S, Binder RJ, Suto R, Anderson KM, Srivastava PK. Necrotic but not apoptotic cell death releases heat shock proteins, which deliver a partial maturation signal to dendritic cells and activate the NF-kappa B pathway. Int Immunol. 2000;12:1539-1546.  [PubMed]  [DOI]  [Cited in This Article: ]
94.  Sauter B, Albert ML, Francisco L, Larsson M, Somersan S, Bhardwaj N. Consequences of cell death: exposure to necrotic tumor cells, but not primary tissue cells or apoptotic cells, induces the maturation of immunostimulatory dendritic cells. J Exp Med. 2000;191:423-434.  [PubMed]  [DOI]  [Cited in This Article: ]
95.  Kotera Y, Shimizu K, Mulé JJ. Comparative analysis of necrotic and apoptotic tumor cells as a source of antigen(s) in dendritic cell-based immunization. Cancer Res. 2001;61:8105-8109.  [PubMed]  [DOI]  [Cited in This Article: ]
96.  Schlienger K, Chu CS, Woo EY, Rivers PM, Toll AJ, Hudson B, Maus MV, Riley JL, Choi Y, Coukos G. TRANCE- and CD40 ligand-matured dendritic cells reveal MHC class I-restricted T cells specific for autologous tumor in late-stage ovarian cancer patients. Clin Cancer Res. 2003;9:1517-1527.  [PubMed]  [DOI]  [Cited in This Article: ]
97.  Bonehill A, Heirman C, Tuyaerts S, Michiels A, Breckpot K, Brasseur F, Zhang Y, Van Der Bruggen P, Thielemans K. Messenger RNA-electroporated dendritic cells presenting MAGE-A3 simultaneously in HLA class I and class II molecules. J Immunol. 2004;172:6649-6657.  [PubMed]  [DOI]  [Cited in This Article: ]
98.  Sun L, Kong B, Sheng X, Sheu JJ, Shih IeM. Dendritic cells transduced with Rsf-1/HBXAP gene generate specific cytotoxic T lymphocytes against ovarian cancer in vitro. Biochem Biophys Res Commun. 2010;394:633-638.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 5]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
99.  Bolhassani A, Safaiyan S, Rafati S. Improvement of different vaccine delivery systems for cancer therapy. Mol Cancer. 2011;10:3.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 171]  [Cited by in F6Publishing: 151]  [Article Influence: 11.6]  [Reference Citation Analysis (0)]
100.  Lotem M, Zhao Y, Riley J, Hwu P, Morgan RA, Rosenberg SA, Parkhurst MR. Presentation of tumor antigens by dendritic cells genetically modified with viral and nonviral vectors. J Immunother. 2006;29:616-627.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
101.  Linette GP, Shankara S, Longerich S, Yang S, Doll R, Nicolette C, Preffer FI, Roberts BL, Haluska FG. In vitro priming with adenovirus/gp100 antigen-transduced dendritic cells reveals the epitope specificity of HLA-A*0201-restricted CD8+ T cells in patients with melanoma. J Immunol. 2000;164:3402-3412.  [PubMed]  [DOI]  [Cited in This Article: ]
102.  Dietz AB, Vuk-Pavlović S. High efficiency adenovirus-mediated gene transfer to human dendritic cells. Blood. 1998;91:392-398.  [PubMed]  [DOI]  [Cited in This Article: ]
103.  Miyazawa M, Iwahashi M, Ojima T, Katsuda M, Nakamura M, Nakamori M, Ueda K, Naka T, Hayata K, Iida T. Dendritic cells adenovirally-transduced with full-length mesothelin cDNA elicit mesothelin-specific cytotoxicity against pancreatic cancer cell lines in vitro. Cancer Lett. 2011;305:32-39.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 23]  [Article Influence: 1.8]  [Reference Citation Analysis (0)]
104.  Bonini C, Lee SP, Riddell SR, Greenberg PD. Targeting antigen in mature dendritic cells for simultaneous stimulation of CD4+ and CD8+ T cells. J Immunol. 2001;166:5250-5257.  [PubMed]  [DOI]  [Cited in This Article: ]
105.  Cui Y, Golob J, Kelleher E, Ye Z, Pardoll D, Cheng L. Targeting transgene expression to antigen-presenting cells derived from lentivirus-transduced engrafting human hematopoietic stem/progenitor cells. Blood. 2002;99:399-408.  [PubMed]  [DOI]  [Cited in This Article: ]
106.  Lizée G, Gonzales MI, Topalian SL. Lentivirus vector-mediated expression of tumor-associated epitopes by human antigen presenting cells. Hum Gene Ther. 2004;15:393-404.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 51]  [Cited by in F6Publishing: 56]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
107.  Cui Y, Kelleher E, Straley E, Fuchs E, Gorski K, Levitsky H, Borrello I, Civin CI, Schoenberger SP, Cheng L. Immunotherapy of established tumors using bone marrow transplantation with antigen gene--modified hematopoietic stem cells. Nat Med. 2003;9:952-958.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
108.  Johansson DX, Ljungberg K, Kakoulidou M, Liljeström P. Intradermal electroporation of naked replicon RNA elicits strong immune responses. PLoS One. 2012;7:e29732.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
109.  Mockey M, Bourseau E, Chandrashekhar V, Chaudhuri A, Lafosse S, Le Cam E, Quesniaux VF, Ryffel B, Pichon C, Midoux P. mRNA-based cancer vaccine: prevention of B16 melanoma progression and metastasis by systemic injection of MART1 mRNA histidylated lipopolyplexes. Cancer Gene Ther. 2007;14:802-814.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 106]  [Cited by in F6Publishing: 113]  [Article Influence: 6.6]  [Reference Citation Analysis (0)]
110.  Perche F, Benvegnu T, Berchel M, Lebegue L, Pichon C, Jaffrès PA, Midoux P. Enhancement of dendritic cells transfection in vivo and of vaccination against B16F10 melanoma with mannosylated histidylated lipopolyplexes loaded with tumor antigen messenger RNA. Nanomedicine. 2011;7:445-453.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 158]  [Cited by in F6Publishing: 153]  [Article Influence: 11.8]  [Reference Citation Analysis (0)]
111.  Fotin-Mleczek M, Duchardt KM, Lorenz C, Pfeiffer R, Ojkić-Zrna S, Probst J, Kallen KJ. Messenger RNA-based vaccines with dual activity induce balanced TLR-7 dependent adaptive immune responses and provide antitumor activity. J Immunother. 2011;34:1-15.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
112.  Kreiter S, Diken M, Selmi A, Diekmann J, Attig S, Hüsemann Y, Koslowski M, Huber C, Türeci Ö, Sahin U. FLT3 ligand enhances the cancer therapeutic potency of naked RNA vaccines. Cancer Res. 2011;71:6132-6142.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 52]  [Cited by in F6Publishing: 53]  [Article Influence: 4.1]  [Reference Citation Analysis (0)]
113.  Kreiter S, Selmi A, Diken M, Koslowski M, Britten CM, Huber C, Türeci O, Sahin U. Intranodal vaccination with naked antigen-encoding RNA elicits potent prophylactic and therapeutic antitumoral immunity. Cancer Res. 2010;70:9031-9040.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 201]  [Cited by in F6Publishing: 205]  [Article Influence: 14.6]  [Reference Citation Analysis (0)]
114.  Diebold SS, Schulz O, Alexopoulou L, Leitner WW, Flavell RA, Reis e Sousa C. Role of TLR3 in the immunogenicity of replicon plasmid-based vaccines. Gene Ther. 2009;16:359-366.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 22]  [Cited by in F6Publishing: 24]  [Article Influence: 1.5]  [Reference Citation Analysis (0)]
115.  Cheng WF, Hung CF, Lee CN, Su YN, Chang MC, He L, Wu TC, Chen CA, Hsieh CY. Naked RNA vaccine controls tumors with down-regulated MHC class I expression through NK cells and perforin-dependent pathways. Eur J Immunol. 2004;34:1892-1900.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 11]  [Article Influence: 0.6]  [Reference Citation Analysis (0)]
116.  Rittig SM, Haentschel M, Weimer KJ, Heine A, Muller MR, Brugger W, Horger MS, Maksimovic O, Stenzl A, Hoerr I. Intradermal vaccinations with RNA coding for TAA generate CD8+ and CD4+ immune responses and induce clinical benefit in vaccinated patients. Mol Ther. 2011;19:990-999.  [PubMed]  [DOI]  [Cited in This Article: ]
117.  Weide B, Pascolo S, Scheel B, Derhovanessian E, Pflugfelder A, Eigentler TK, Pawelec G, Hoerr I, Rammensee HG, Garbe C. Direct injection of protamine-protected mRNA: results of a phase 1/2 vaccination trial in metastatic melanoma patients. J Immunother. 2009;32:498-507.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
118.  Hess PR, Boczkowski D, Nair SK, Snyder D, Gilboa E. Vaccination with mRNAs encoding tumor-associated antigens and granulocyte-macrophage colony-stimulating factor efficiently primes CTL responses, but is insufficient to overcome tolerance to a model tumor/self antigen. Cancer Immunol Immunother. 2006;55:672-683.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 75]  [Cited by in F6Publishing: 82]  [Article Influence: 4.3]  [Reference Citation Analysis (0)]
119.  Boczkowski D, Nair SK, Snyder D, Gilboa E. Dendritic cells pulsed with RNA are potent antigen-presenting cells in vitro and in vivo. J Exp Med. 1996;184:465-472.  [PubMed]  [DOI]  [Cited in This Article: ]
120.  Nair SK, Boczkowski D, Morse M, Cumming RI, Lyerly HK, Gilboa E. Induction of primary carcinoembryonic antigen (CEA)-specific cytotoxic T lymphocytes in vitro using human dendritic cells transfected with RNA. Nat Biotechnol. 1998;16:364-369.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 315]  [Cited by in F6Publishing: 323]  [Article Influence: 12.4]  [Reference Citation Analysis (0)]
121.  Pan K, Zhao JJ, Wang H, Li JJ, Liang XT, Sun JC, Chen YB, Ma HQ, Liu Q, Xia JC. Comparative analysis of cytotoxic T lymphocyte response induced by dendritic cells loaded with hepatocellular carcinoma -derived RNA or cell lysate. Int J Biol Sci. 2010;6:639-648.  [PubMed]  [DOI]  [Cited in This Article: ]
122.  Jarnjak-Jankovic S, Saebøe-Larssen S, Kvalheim G, Gaudernack G. mRNA transfection of DC in the immature or mature state: comparable in vitro priming of Th and cytotoxic T lymphocytes against DC electroporated with tumor cell line-derived mRNA. Cytotherapy. 2007;9:587-592.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 5]  [Cited by in F6Publishing: 4]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
123.  Ashley DM, Faiola B, Nair S, Hale LP, Bigner DD, Gilboa E. Bone marrow-generated dendritic cells pulsed with tumor extracts or tumor RNA induce antitumor immunity against central nervous system tumors. J Exp Med. 1997;186:1177-1182.  [PubMed]  [DOI]  [Cited in This Article: ]
124.  Liao X, Li Y, Bonini C, Nair S, Gilboa E, Greenberg PD, Yee C. Transfection of RNA encoding tumor antigens following maturation of dendritic cells leads to prolonged presentation of antigen and the generation of high-affinity tumor-reactive cytotoxic T lymphocytes. Mol Ther. 2004;9:757-764.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 47]  [Cited by in F6Publishing: 44]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
125.  Nair SK, Heiser A, Boczkowski D, Majumdar A, Naoe M, Lebkowski JS, Vieweg J, Gilboa E. Induction of cytotoxic T cell responses and tumor immunity against unrelated tumors using telomerase reverse transcriptase RNA transfected dendritic cells. Nat Med. 2000;6:1011-1017.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 278]  [Cited by in F6Publishing: 265]  [Article Influence: 11.0]  [Reference Citation Analysis (0)]
126.  Benencia F, Courrèges MC, Coukos G. Whole tumor antigen vaccination using dendritic cells: comparison of RNA electroporation and pulsing with UV-irradiated tumor cells. J Transl Med. 2008;6:21.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 53]  [Cited by in F6Publishing: 58]  [Article Influence: 3.6]  [Reference Citation Analysis (0)]
127.  Edlich B, Hogdal LJ, Rehermann B, Behrens SE. Dendritic cells transfected with Her2 antigen-encoding RNA replicons cross-prime CD8 T cells and protect mice against tumor challenge. Vaccine. 2010;28:7764-7773.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 4]  [Cited by in F6Publishing: 4]  [Article Influence: 0.3]  [Reference Citation Analysis (0)]
128.  Bontkes HJ, Kramer D, Ruizendaal JJ, Meijer CJ, Hooijberg E. Tumor associated antigen and interleukin-12 mRNA transfected dendritic cells enhance effector function of natural killer cells and antigen specific T-cells. Clin Immunol. 2008;127:375-384.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 42]  [Cited by in F6Publishing: 44]  [Article Influence: 2.8]  [Reference Citation Analysis (0)]
129.  Naka T, Iwahashi M, Nakamura M, Ojima T, Nakamori M, Ueda K, Katsuda M, Miyazawa M, Ishida K, Yamaue H. Tumor vaccine therapy against recrudescent tumor using dendritic cells simultaneously transfected with tumor RNA and granulocyte macrophage colony-stimulating factor RNA. Cancer Sci. 2008;99:407-413.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 10]  [Cited by in F6Publishing: 12]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
130.  Bontkes HJ, Kramer D, Ruizendaal JJ, Kueter EW, van Tendeloo VF, Meijer CJ, Hooijberg E. Dendritic cells transfected with interleukin-12 and tumor-associated antigen messenger RNA induce high avidity cytotoxic T cells. Gene Ther. 2007;14:366-375.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 56]  [Cited by in F6Publishing: 57]  [Article Influence: 3.2]  [Reference Citation Analysis (0)]
131.  Koido S, Kashiwaba M, Chen D, Gendler S, Kufe D, Gong J. Induction of antitumor immunity by vaccination of dendritic cells transfected with MUC1 RNA. J Immunol. 2000;165:5713-5719.  [PubMed]  [DOI]  [Cited in This Article: ]
132.  Kim SG, Park MY, Kim CH, Sohn HJ, Kim HS, Park JS, Kim HJ, Oh ST, Kim TG. Modification of CEA with both CRT and TAT PTD induces potent anti-tumor immune responses in RNA-pulsed DC vaccination. Vaccine. 2008;26:6433-6440.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 18]  [Cited by in F6Publishing: 20]  [Article Influence: 1.3]  [Reference Citation Analysis (0)]
133.  Hosoi A, Takeda Y, Sakuta K, Ueha S, Kurachi M, Kimura K, Maekawa R, Kakimi K. Dendritic cell vaccine with mRNA targeted to the proteasome by polyubiquitination. Biochem Biophys Res Commun. 2008;371:242-246.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 9]  [Cited by in F6Publishing: 7]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
134.  Steitz J, Britten CM, Wölfel T, Tüting T. Effective induction of anti-melanoma immunity following genetic vaccination with synthetic mRNA coding for the fusion protein EGFP.TRP2. Cancer Immunol Immunother. 2006;55:246-253.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 42]  [Article Influence: 2.2]  [Reference Citation Analysis (0)]
135.  Onaitis MW, Kalady MF, Emani S, Abdel-Wahab Z, Tyler DS, Pruitt SK. CD40 ligand is essential for generation of specific cytotoxic T cell responses in RNA-pulsed dendritic cell immunotherapy. Surgery. 2003;134:300-305.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in F6Publishing: 1]  [Reference Citation Analysis (0)]
136.  Mu LJ, Gaudernack G, Saebøe-Larssen S, Hammerstad H, Tierens A, Kvalheim G. A protocol for generation of clinical grade mRNA-transfected monocyte-derived dendritic cells for cancer vaccines. Scand J Immunol. 2003;58:578-586.  [PubMed]  [DOI]  [Cited in This Article: ]
137.  Van Tendeloo VF, Ponsaerts P, Lardon F, Nijs G, Lenjou M, Van Broeckhoven C, Van Bockstaele DR, Berneman ZN. Highly efficient gene delivery by mRNA electroporation in human hematopoietic cells: superiority to lipofection and passive pulsing of mRNA and to electroporation of plasmid cDNA for tumor antigen loading of dendritic cells. Blood. 2001;98:49-56.  [PubMed]  [DOI]  [Cited in This Article: ]
138.  Ponsaerts P, Van den Bosch G, Cools N, Van Driessche A, Nijs G, Lenjou M, Lardon F, Van Broeckhoven C, Van Bockstaele DR, Berneman ZN. Messenger RNA electroporation of human monocytes, followed by rapid in vitro differentiation, leads to highly stimulatory antigen-loaded mature dendritic cells. J Immunol. 2002;169:1669-1675.  [PubMed]  [DOI]  [Cited in This Article: ]
139.  Gao L, Fan HH, Lu HZ, Nie XX, Liu Y, Yang YM, Qian KC, Gao F. Impact of transfection with total RNA of K562 cells upon antigen presenting, maturation, and function of human dendritic cells from peripheral blood mononuclear cells. Transfusion. 2007;47:256-265.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 2]  [Cited by in F6Publishing: 3]  [Article Influence: 0.2]  [Reference Citation Analysis (0)]
140.  Zhang HM, Zhang LW, Liu WC, Cheng J, Si XM, Ren J. Comparative analysis of DC fused with tumor cells or transfected with tumor total RNA as potential cancer vaccines against hepatocellular carcinoma. Cytotherapy. 2006;8:580-588.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 15]  [Cited by in F6Publishing: 13]  [Article Influence: 0.8]  [Reference Citation Analysis (0)]
141.  Harris J, Monesmith T, Ubben A, Norris M, Freedman JH, Tcherepanova I. An improved RNA amplification procedure results in increased yield of autologous RNA transfected dendritic cell-based vaccine. Biochim Biophys Acta. 2005;1724:127-136.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 7]  [Cited by in F6Publishing: 8]  [Article Influence: 0.4]  [Reference Citation Analysis (0)]
142.  Schaft N, Dörrie J, Thumann P, Beck VE, Müller I, Schultz ES, Kämpgen E, Dieckmann D, Schuler G. Generation of an optimized polyvalent monocyte-derived dendritic cell vaccine by transfecting defined RNAs after rather than before maturation. J Immunol. 2005;174:3087-3097.  [PubMed]  [DOI]  [Cited in This Article: ]
143.  John J, Dalgleish A, Melcher A, Pandha H. Cryopreserved dendritic cells for intratumoral immunotherapy do not require re-culture prior to human vaccination. J Immunol Methods. 2005;299:37-46.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 8]  [Cited by in F6Publishing: 10]  [Article Influence: 0.5]  [Reference Citation Analysis (0)]
144.  Zeis M, Siegel S, Wagner A, Schmitz M, Marget M, Kühl-Burmeister R, Adamzik I, Kabelitz D, Dreger P, Schmitz N. Generation of cytotoxic responses in mice and human individuals against hematological malignancies using survivin-RNA-transfected dendritic cells. J Immunol. 2003;170:5391-5397.  [PubMed]  [DOI]  [Cited in This Article: ]
145.  Heiser A, Maurice MA, Yancey DR, Coleman DM, Dahm P, Vieweg J. Human dendritic cells transfected with renal tumor RNA stimulate polyclonal T-cell responses against antigens expressed by primary and metastatic tumors. Cancer Res. 2001;61:3388-3393.  [PubMed]  [DOI]  [Cited in This Article: ]
146.  Heiser A, Maurice MA, Yancey DR, Wu NZ, Dahm P, Pruitt SK, Boczkowski D, Nair SK, Ballo MS, Gilboa E. Induction of polyclonal prostate cancer-specific CTL using dendritic cells transfected with amplified tumor RNA. J Immunol. 2001;166:2953-2960.  [PubMed]  [DOI]  [Cited in This Article: ]
147.  Su Z, Vieweg J, Weizer AZ, Dahm P, Yancey D, Turaga V, Higgins J, Boczkowski D, Gilboa E, Dannull J. Enhanced induction of telomerase-specific CD4(+) T cells using dendritic cells transfected with RNA encoding a chimeric gene product. Cancer Res. 2002;62:5041-5048.  [PubMed]  [DOI]  [Cited in This Article: ]
148.  Van Driessche A, Van de Velde AL, Nijs G, Braeckman T, Stein B, De Vries JM, Berneman ZN, Van Tendeloo VF. Clinical-grade manufacturing of autologous mature mRNA-electroporated dendritic cells and safety testing in acute myeloid leukemia patients in a phase I dose-escalation clinical trial. Cytotherapy. 2009;11:653-668.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 79]  [Cited by in F6Publishing: 85]  [Article Influence: 5.7]  [Reference Citation Analysis (0)]
149.  Markovic SN, Dietz AB, Greiner CW, Maas ML, Butler GW, Padley DJ, Bulur PA, Allred JB, Creagan ET, Ingle JN. Preparing clinical-grade myeloid dendritic cells by electroporation-mediated transfection of in vitro amplified tumor-derived mRNA and safety testing in stage IV malignant melanoma. J Transl Med. 2006;4:35.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 25]  [Cited by in F6Publishing: 26]  [Article Influence: 1.4]  [Reference Citation Analysis (0)]
150.  Hernando JJ, Park TW, Fischer HP, Zivanovic O, Braun M, Pölcher M, Grünn U, Leutner C, Pötzsch B, Kuhn W. Vaccination with dendritic cells transfected with mRNA-encoded folate-receptor-alpha for relapsed metastatic ovarian cancer. Lancet Oncol. 2007;8:451-454.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 46]  [Cited by in F6Publishing: 49]  [Article Influence: 2.9]  [Reference Citation Analysis (0)]
151.  Lesterhuis WJ, De Vries IJ, Schreibelt G, Schuurhuis DH, Aarntzen EH, De Boer A, Scharenborg NM, Van De Rakt M, Hesselink EJ, Figdor CG. Immunogenicity of dendritic cells pulsed with CEA peptide or transfected with CEA mRNA for vaccination of colorectal cancer patients. Anticancer Res. 2010;30:5091-5097.  [PubMed]  [DOI]  [Cited in This Article: ]
152.  Kyte JA, Kvalheim G, Aamdal S, Saebøe-Larssen S, Gaudernack G. Preclinical full-scale evaluation of dendritic cells transfected with autologous tumor-mRNA for melanoma vaccination. Cancer Gene Ther. 2005;12:579-591.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 50]  [Cited by in F6Publishing: 51]  [Article Influence: 2.7]  [Reference Citation Analysis (0)]
153.  Schuurhuis DH, Verdijk P, Schreibelt G, Aarntzen EH, Scharenborg N, de Boer A, van de Rakt MW, Kerkhoff M, Gerritsen MJ, Eijckeler F. In situ expression of tumor antigens by messenger RNA-electroporated dendritic cells in lymph nodes of melanoma patients. Cancer Res. 2009;69:2927-2934.  [PubMed]  [DOI]  [Cited in This Article: ]  [Cited by in Crossref: 48]  [Cited by in F6Publishing: 50]  [Article Influence: 3.3]  [Reference Citation Analysis (0)]