Genetic modification of dendritic cells and its application
for cancer immunotherapy

Yasuhiko Nishioka, Wen Hua, Naoki Nishimura, and Saburo Sone

DThird Department of Internal Medicine, The University of Tokushima School of Medicine, Tokushima, Japan

Abstract: Dendritic cells (DCs) are the most potent antigen-presenting cells (APCs). DCs pulsed with peptides of tumor-associated antigens (TAA) and tumor lysate have been used in cancer immunotherapy. An early clinical study demonstrated the safety of the use of DCs, but the clinical response was not sufficient. The gene-modification of DCs with TAA and soluble factor genes such as cytokine and chemokine genes has been examined to enhance the antigen-presenting capacity of DCs. Viral vectors including retroviruses and adenoviruses have been reportred to be useful to obtain a sufficient transduction efficiency into DCs. TAA gene-transduced DCs could have several advantages compared with TAA peptide-pulsed DCs as follows : 1) The use of TAA gene-modified DCs are not restricted by MHC haplotypes. 2) The gene transduction with TAA genes is likely to present the unknown TAA peptides on DCs. 3) The gene-modified DCs show the prolonged presentation of TAA peptides. The transduction of DCs with cytokine genes including IL-12 and GM-CSF have also been reported to augument the antitumor effects of DCs. Although the results in the experimental systems were promising, the clinical application of gene-modified DCs includes several problems such as the standardization of methods of manipulation and gene-transduction of DCs. Approaches to solve them require further studies. J. Med. Invest. 49:7-17, 2002

Keywords:dendritic cells (DCs), tumor-associated antigens (TAA), cytokine, chemokine, gene transduction, viral vector

INTRODUCTION
Dendritic cells (DCs) are the most potent antigen-presenting cells (APCs), which distribute in most tissues, capture antigens in situ and migrate to lymphoid organs to activate naive T cells (1, 2). In 1973, Steinman reported the novel cell type in murine spleen that shows the typical phenotype with long dendrites, and named them dendritic cells (3). Since the number of DCs, however, were very few at 1.0-1.6% in the spleen or less in other major organs including peripheral blood, it has been difficult to study the in vitro and in vivo functions of DCs. Since 1992, when the culture methods to generate DCs from monocytes and CD34+ hematopoietic progenitor cells with cytokines in vitro were established (4-6), both basic and clinical research have rapidly progressed. Based on the analysis of DC functions, its clinical application for several diseases, especially for malignant diseases, has been performed using DCs pulsed with peptides and proteins of tumor-associated antigens (TAAs) or tumor lysate (7-10). Tumor cell-dendritic cell hybrids were also used for the treatment of renal cell carcinoma (11). These early clinical studies demonstrated the safety of DC-based immunotherapy, but the clinical responses were not so sufficient irrespective of some objective responses. To improve the antitumor effects in humans, the novel approaches or the combination with other modalities should be examined. One of the most potent approaches to enhance the APC function of DCs could be the genetic modification of DCs with antigen genes (12). In fact, vaccination with gene-modified DCs was more effective in suppressing tumor growth compared with vaccination with gene-modified tumor cells (13). The other genes including cytokines and chemokines have also been examined to augment immune responses against cancer. Here, we review the recent progresses in the study of gene-modified DCs.

Efficient gene transfer into DCs
To modify DCs with foreign genes, the various methods of gene transfer have been examined (Table 1). The transduction markers such as LacZ and luciferase were used in early studies, whereas recent projects to examine the transduction of DCs employeed the green fluorescent protein (GFP) that spontaneously emitted green light without the substrate in living cells (14) (Figure 1). Since DCs are terminally differentiated and not dividing cells (15), it is difficult to transduce the foreign gene into DCs. First, Alijagic et al. reported the gene modification of human monocyte-derived DCs using liposome-mediated transduction of the tyrosinase gene (16). Although they found the proliferation of tyrosinase-specific T cells, the transduction efficiency determined by LacZ as a marker gene was too low to evaluate. Similarly, approaches using non-viral systems could not produce a high transduction efficiency, being about 10% at most (17-22). On the other hand, viral vectors, particularly retroviral and adenoviral vectors were used for the transduction of DCs in most of the studies reported recently. The retroviral transduction is limited to use with CD34+ cell-derived DCs in humans and bone marrow-derived DCs in mice because of the requirement for cell proliferation. Furthermore, the additional techniques and repeated transduction were needed for the retroviral system to yield high efficiency since the conventional method of retroviral transduction could not produce high transduction efficiency. The co-culture of DCs with producer cells or the combination of centrifugation or liposome reported to be effective to enhance the transduction efficiency (23-35). In summary, the transduction efficiency by the retrovirus system varied was reported to be 11.5-86% (Table 1). On the other hand, the other viral vector that was commonly used for DC transduction was an adenoviral system, which was known to infect non-dividing cells with a high efficiency. To date, the adenovirus could be the most useful vector to transduce DCs with foreign genes since most findings using adenoviral transduction showed an efficiency greater than 80% (Table 1) (16, 19, 36-43). Furthermore, there appears to be several advantages in the adenoviral vectors:1) the adenoviral vector is not integrated into host genomes when compared with a retroviral system. 2) most humans have an anti-adenoviral immunity, which might prevent the adverse effects caused by adenoviral vectors. However, even when used in an adenoviral system, DCs were relatively resistant to gene-modification compared with tumor cells. The combined use including liposomes and centrifugation was recommended to achieve a high transduction efficiency (20, 40). Recently, other viral systems such as Lentivirus (44, 45), adeno-associate virus (AAV) (46), influenza virus (47) and pox virus (48) were also reported to be useful for the gene-modification of DCs.

Candidate genes for gene transduction of DCs
The candidate genes that have been tested for the gene-modification of DCs are described in Table 2. The effects of gene-tansduction of DCs with TAA genes were examined first. Recent studies reported findings with other genes including cytokines, chemokines and costimulatory molecules.
(1) Tumor-associated antigen (TAA) genes
In 1991, Boon et al. first reported the successful cloning of the TAA gene which is specifically recognized by cytotoxic T lymphocytes (CTLs), and named it MAGE (49). Futhermore, they identified the antigenic peptides of MAGE-3, which were presented on MHC class I of APC and could induce antigen-specific CD8+ CTLs (50, 51). Kawakami et al. also reported the melanocyte-specific antigens MART-1 and gp100 which were recognized by tumor-infiltrating lymphocytes in melanoma (52, 53). These findings allowed us to start tumor vaccine therapy using TAA peptides. The early clinical studies for patients with metastatic melanoma using TAA peptides mixed with an adjuvant showed the induction of tumor specific immune responses and some objective responses (54, 55).
On the other hand, it was reported that the administration of DCs pulsed with TAA peptides was more effective in regressing established tumors than TAA peptides alone (56, 57). Many investigators have now focused on the use of DCs for cancer immunotherapy to obtain better clinical effects. In addition to the use of TAA peptides or tumor lysate, fusion of DCs with tumor cells (58), pulsing with tumor RNA (59), exosomes (60) and the gene-modification of DCs (12) have been reported to be hopeful strategies. Among them, one of the best use of DCs could be the gene-transduced DCs with the TAA gene due to the following possibilities : 1) The use of TAA gene-modified DCs is not restricted by MHC haplotypes. 2) The gene transduction with TAA genes is likely to present unknown TAA peptides on DCs. 3) The gene-modification prolongs the presentation of TAA peptides on DCs. In the early experiments, the tumor cells modified to express foreign antigens such as β-galactosidase (β-Gal) and ov albumin (OVA) have been used (34, 36, 61, 62). However, since these artificial antigens have shown a strong immunogenicity that is different from that of endogenous TAA, experiments with endogenous TAA are necessary before initiating clinical trials of immunotharapy against human cancers. To answer this, Kaplan et al. demonstrated that therapy with DCs transduced with endogenous TAA antigen TRP (tyrosinase-related protein)-2 effectively induced the tumor-specific immunity and regressed B16 tumors (39). This observation could be important since they first demonstrated the possibility that immunization with endogenous TAA, in which immunogenicity was presumably low, was also effective for inducing tumor-specific immunity and inhibiting tumor growth. In humans, Reeves et al. reported MART-1 gene transduction of human CD34+ cell-derived DCs with retrovirus system and the induction of CTLs specific for MART-1 in vitro (23). Butterfield et al. also demonstrated the MART-1 gene-modification of human monocyte-derived DCs with adenoviral vector and the effective CTL induction using gene-modified DCs (63, 64). They next reported the efficient induction of CTLs specific for α-fetoprotein (AFP) by AFP-transduced DCs as an immunotherapy for patients with hepatocellular carcinoma (65, 66). The DCs modified to express other TAA genes including p53, MAGE-1, 3 and MUC-1 have been tested for their ability to induce the antigen-specific CTLs in vitro (18, 24, 37, 48, 67, 68). Although the gene-modified DCs with these TAA genes have been effective in generating CTLs in vitro, it is still unclear what type of TAA genes are most effective for what types of cancer.
(2) The cytokine and chemokine genes
Various cytokines and chemokines were involved in the process of antigen presentation and CTL induction by DCs (1). Interleukin (IL)-12 enhances NK cell and CTL activities, plays a key role in the induction of Th1 immune responses including IFN-? production (69), and promotes the growth of T and NK cells (70, 71). The administration of IL-12 protein and IL-12 gene-transduction into tumor cells has shown profound antitumor effects (72, 73). Granulocyte-macrophage colony stimulating factor (GM-CSF) is known to be an essential cytokine to generate DCs from both bone marrow cells and monocytes and stimulate the survival of DCs (4-6). For these reasons, studies regarding the transduction of DCs with cytokine genes were initially examined using IL-12 and GM-CSF genes. Melero et al. and we demonstrated that the intratumoral injection of IL-12 gene-transduced DCs induced the tumor specific immune responses and regressed the established tumors in mice (32, 74). The antitumor effects of intratumoral injection of IL-12 gene-modified DCs were found to be better than that of IL-12 gene-modified fibroblasts that have been used in clinical trial as a phase I and II study (32). GM-CSF gene-modified DCs pulsed with TAA peptides were also demonstrated to be more effective in inducing antitumor immunity than nontransduced DCs (21). It was suggested that these effects were mediated by the increased survival and migration to draining lymph nodes (21). The approaches of intratumoral injection were applied for IL-7 gene-modified DCs (75). They compared IL-7 gene-modified DCs with TAA-loading DCs, and found DC-IL-7 to be as effective as TAA-loaded DCs and superior to tumor lysate-pulsed DCs (75). Even when human DCs were transduced with IL-12 and IL-7 genes, these cytokine gene-transduced DCs showed the enhancement of the allogeneic MLR, indicating that these approaches could be applicable for humans (30, 76)
Chemokines would also be better candidates to enhance immune responses in vivo. The transduction of the lymphotactin gene was examined using combinations with TAA peptides (77). The vaccination of DC-lymphotactin pulsed with TAA peptide was more effective in inducing specific antitumor immunity and reducing lung metastases of 3LL tumors when compared with nontransduced DCs (77).
(3) The cell surface molecules
CD40L is a costimulatory molecule that is expressed on activated CD4+ T cells and stimulates APCs through the CD40-CD40L interaction (1). To activate DCs directly, Kikuchi et al. transduced the CD40L gene to murine DCs and evaluated the antitumor effects of CD40L gene-modified DCs (78). Infection of DCs by AdCD40L induced IL-12 and MIP-1α productions, and the intratumoral administration of CD40L-transduced DCs induced the regression of pre-existing tumors (78).
DCs express CD80 molecules, but the level of CD80 expression is not high. Tsang et al. examined CD80 gene transduction into human DCs and found that the CD80 gene-modification of DCs enhanced IFN-? production and cytotoxicity of antigen-specific CTLs using CEA-specific T cells (79). They extended this study and reported the imunostimulatory effect of transduction of three costimulatory molecules (CD80, CD54 and CD58) using avipox virus (48). The human DCs transduced with a triad of costimulatory molecules significantly generated peptide-specific CTLs in vitro (48). It might be a great advantage that the avipox viral vector could express three transgenes on human DCs at the same time.

Future perspectives and problems with the application of gene-modified DCs
The schema of DC-based cancer immunotherapy in humans is shown in figure 2. There have been three types of human DCs used in the clinical trials such as monocyte-derived, CD34+ cell-derived and blood DCs. Among them, the monocyte-derived DCs are convenient to use in clinics due to their ease in preparation. However, the standard method of DC-based immunotherapy has not yet been established. There are some problems that needed to be clarified to optimize DC therapy as follows : 1) the kind of DCs best for tumor immunotherapy 2) the optimal protocol of DC administration 3) the most effective TAA. To date, some studies have answered these questions. For example, the findings reported by Eggert et al. showed that the s.c. injection of DCs was better than the i.v. injection for inducing the antitumor immunity in mice (80). Mores et al. demonstrated that the migration of DCs into lymph nodes was much better after s.c. injection when compared with i.v. route in humans (81). Based on these observations, the s.c. injection is suggested to be the most useful route for DC administration. Furthermore, the comparative studies on the function between monocyte-derived and CD34+ cell-derived DCs have been reported. Mortarini et al. and Felazzo et al. showed that CD34+ cell-derived DCs were more potent to induce CTLs than monocyte-derived DCs (82, 83). However, further studies are required to clarify whether CD34+ cell-derived DCs are better than monocyte-derived DCs for cancer immunotherapy. The subset of DCs (84) and the novel findings of DC function such as the interaction with innate immunity (85, 86) and the trafficking capacity (87) should be also considered to establish the standard protocol of DC-based immunotherapy. Since the gene-modified DCs have shown strong antitumor effects against various types of tumors in animal models, future studies would be expected to lead to clinical trials.

REFERENCES
1.Banchereau J, Steinman RM : Dendritic cells and the control of immunity. Nature 392:245-252, 1998.
2.Steinman RM, Dhodapkar M : Active immunization against cancer with dendritic cells : the near future. Int J Cancer 94:459-473, 2001.
3.Steinman RM, Cohn ZA : Identification of a novel cell type in peripheral lymphoid organs of mice. I. Morphology, Quantitation, Tissu distribution. J Exp Med 137:1142-1162, 1973.
4.Inaba K, Inaba M, Romani N, Aya H, Deguchi M, Ikehara S, Muramatsu S, Steinman RM : Generation of large numbers of dendritic cells from mouse bone marrow cultures supplemented with granulocyte/macrophage colony-stimulating factor. J Exp Med 176:1693-702, 1992.
5.Caux C, Dezutter-Dambuyant C, Schmitt D, Banchereau J : GM-CSF and TNF-α cooperate in the generation of dendritic langerhans cells. Nature 360 :258-261, 1992.
6.Sallusto F, Lanzavecchia A : Efficient presentation of soluble antigen by cultured human dendritic cells is maintained by granulocyte/macrophage colony-stimulating factor plus interleukin 4 and downregulated by tumor necrosis factor α. J Exp Med 179:1109-1118, 1994.
7.Hsu F.J, Benike C, Fagnoni F, Marie Liles T, Czerwinski D, Taidi B, Engelman EG, Levy R :Vaccination of patients with B-cell lymphoma using autologous antigen-pulsed dendritic cells. Nature Med. 2:52-58, 1996.
8.Nestle FO, Alijagic S, Gilliet M, Sun Y, Grabbe S, Dummer R, Burg G, Schadendorf D : Vaccination of melanoma patients with peptide- or tumor lysate-pulsed dendritic cells. Nature Med 4:328-332, 1998.
9.Geiger J, Hutchinson R, Hohenkirk L, McKenna E, Chang A, Mule J : Treatment of solid tumors in children with tumor-lysate-pulsed dendritic cells. Lancet 356:1163-1165, 2000.
10.Banchereau J, Palucka AK, Dhodapkar M, Burkeholder S, Taquet N, Rolland A, Taquet S, Coquery S, Wittkowski KM, Bhardwaj N, Pineiro L, Steinman R and Fay J : Immune and clinical responses in patients with metastatic melanoma to CD34+ progenitor-derived dendritic cell vaccine. Cancer Res 61:6451-6458, 2001.
11. Kugler A, Stuhler G, Walden P, Zoller G, Zobywalski A, Brossart P, Trefzer U, Ullrich S, Muller CA, Becker V, Gross AJ, Hemmerlein B, Kanz L, Muller GA, Ringert R-H : Regression of human metastatic renal cell carcinoma after vaccination with tumor cell-dendritic cell hybrids. Nature Med 6:332-336, 2000.
12.Kirk CJ, Mule JJ : Gene-modified dendritic cells for use in tumor vaccine. Hum Gene Ther 11:797-806, 2000.
13.Klein C, Bueler H, Mulligan RC : Comparative analysis of genetically modified dendritic cells and tumor cells as therapeutic cancer vaccines.:J Exp Med 191:1699-708, 2000.
14.Chalfie M, Tu Y, Euskirchen G, Ward WW, Prasher DC : Green fluorescent protein as a markere for gene expression. Nature 263:802-805, 1994.
15.Palucka KA, Taquet N, Sanchez-Chapuis F, Gluckman JC:Dendritic cells as the terminal stage of monocyte differentiation. J Immunol 160:4587-4595, 1998.
16.Alijagic S, Moller P, Artuc M, Jurgovsky K, Czarnetzki, Schadendorf D : Dendritic cells generated from peripheral blood transfected with human tyrosinase induce specific T cell activation. Eur J Immunol 25:3100-3107, 1995.
17.Arthur JF, Butterfield LH, Roth MD, Bui LA, Kiertscher SM, Lau R, Dubinett S, Glaspy J, McBride WH, Economou JS : A comparison of gene transfer methods in human dendritic cells. Cancer Gene Ther 4:17-25, 1997.
18.Tuting T, DeLeo AB, Lotze MT, Storkus WJ : Genetically modified bone marrow-derived dendritic cells expressing tumor-associated viral or "self" antigens induce antitumor immunity in vivo. Eur J Immunol 27:2702-2707, 1997.
19.VanTedelooV, Snoeck H-W, Lardon F, Vanham G, Nijs G, Lenjou M, Hendriks L, Broeckhoven AV, Moulijn A, Rodrigus I, Verdonk P, Van Bockstaele Drand Berneman ZN : Nonviral transfection of distinct types of human dendritic cells : High-efficiency gene transfer by electroporation into hematopoietic progenitor- but not monocyte-derived dendritic cells. Gene Ther 5:700-707, 1998.
20.Dietz AB and Vuk-Pavlovic:High efficiency adenovirus-mediated gene transfer to human dendritic cells. Blood 91:392-398, 1998.
21.Curiel-Lewandrowski C, Mahnke K, labeur M, Roters B, Schmidt W, Granstein RD, Luger TA, Schwarz T, Grabbe S : Transfection of immature murine bone marrow-derived dendritic cells with the granulocyte-macrophage colony-stimulating factor gene potently enhances their in vivo antigen-presenting capacity. J Immunol 163:174-183, 1999.
22.Diebold SS, Lehemann H, Kursa M, Wagner E, Cotton M, Zenke M : Efficient gene delivery into human dendritic cells by adenovirus polyethylenimine and mannose polyethylenimine transfection. Hum Gene Ther 10:775-786, 1999.
23.Reeves ME, Royal RE, Lam JS, Rosenberg SA, Hwu P : Retroviral transduction of human dendritic cells with tumor-associated antigen gene. Cancer Res 56:5672-5677, 1996.
24.Henderson RA, Nimgaonkar MT, Watkins SC, Robbins PD, Ball ED, Finn OJ : Human dendritic cells genetically engineered to express high levels of the human epithelial tumor antigen mucin (MUC-1). Cancer Res 56:3763-3770, 1996.
25.Specht JM, Wang G, Do MT, Lam JS, Royal RE, Reeves ME, Rosenberg SA, Hwu P:Dendritic cells retrovirally transduced with a model antigen gene are therapeutically effective against established pulmonary metastases. J Exp Med 186 :1213-1221, 1997.
26.Aicher A, Westermann J, Cayeux S, Willimsky G, Daemen K, Blankenstein T, Uckert W, Dorken B, Pezzutto A:Successful retroviral mediated transduction of a reporter gene in human dendritic cells : Feasibility of therapy with gene-modified antigen presenting cells. Exp Hematol 25:39-44, 1997.
27.Szabolcs P, Gallardo HF, Ciocon DH, Sadelain M, Young JW : Retrovirally transduced human dendritic cells express a normal phenotype and potent T-cell stimulatory capacity. Blood 90 : 2160-2167, 1997.
28.Verhasselt B, De Smedt M, Verhelst R, Naessens E, Plum J : Retrovirally transduced CD34+ human cord blood cells generate T cells expressing high levels of the retroviral encoded green fluorescent protein marker in vitro. Blood 91:431-440, 1998.
29.SchilzAJ, BrounsG, Knob H, Ottmann OG, Hoelzer D, Fauser AA, Thrasher AJ, Grez M:High efficiency gene transfer to human hematopoietic SCID-repopulating cells under serum-free conditions. Blood 92:3136-3171, 1998.
30.Westermann J, Aicher A, Qin Z, Cayeux S, Daemen K, Blankenstein Th, Dorken B, Pezzutto A : Retroviral interleukin-7 gene transfer into human dendritic cells enhances T cell activation. Gene Ther 5:264-271, 1998.
31.TakayamaT, NishiokaY, Lu L, Lotze MT, Tahara H, Thomson AW : Retrovirally delivery of viral interleukin-10 into myeloid dendritic cells markedly inhibits their allostimulatory activity and promotes the induction of T-cell hyporesponsiveness. Transplantation 66:1567-1574, 1998.
32.Nishioka Y, Hirao M, Robbins PD, Lotze MT, Tahara H : Induction of systemic and therapeutic antitumor immunity using intratumoral injection of dendritic cells genetically modified to express interleukin-12. Cancer Res 59:4035-4041, 1999.
33.Gasperi C, Rescigno M, Granucci F, Citterio S, Matyszak MK, Sciurpi MT, Lanfrancone L, Ricciardi-Castagnoli P : Retroviral gene transfer, rapid selection, and maintenance of the immature phenotype in mouse dendritic cells. J Leukocyte Biol 66:263-267, 1999.
34.De Veerman M, Heirman C, Van Meirvenne S, Devos S, Corthals J, Moser M, Thielemans K :Retrovirally transduced bone marrow-derived dendritic cells require CD4+ T cell help to elicit protective and therapeutic antitumor immunity. J Immunol 162:144-151, 1999.
35.Movassagh M, Baillou C, Cosset FL, Klatzmann D, Guigon M, Lemoine FM:High level of retrovirus-mediated gene transfer into dendritic cells derived from cord blood and mobilized peripheral blood CD34+ cells. Hum Gene Ther 10 :175-187, 1999.
36.Brossart P, Goldrath AW, Butz EA, Martin S, Bevan MJ : Virus-mediated delivery of antigenic epitopes into dendritic cells as a means to induce CTL. J Immunol 158:3270-3276, 1997.
37.Gong J, Chen L, Chen D, Kashiwaba M, Manome Y, Tanaka T, Kufe D : Induction of antigen-specific antitumor immunity with adenovirus-transduced dendritic cells. Gene Ther 4:1023-1028, 1997.
38.Song W, Kong H-L, Carpenter H, Torii H, Granstein R, Rafli S, Moore MAS, Crystal RG : Dendritic cells genetically modified with an adenovirus vector encoding the cDNA for a model antigen induce protective and therapeutic antitumor immunity. J Exp Med 186:1247-1256, 1997.
39.Kaplan JM, Yu Q, Piraino ST, Pennington SE, Shankara S, Woodwrth LA, Roberts BL : Induction of antitumor immunity with dendritic cells transduced with adenovirus vector-encoding endogenous tumor-associated antigens. J Immunol 163:699-707, 1999.
40.Nishimura N, Nishioka Y, Shinohara T, Ogawa H, Yamamoto S, Tani K, Sone S : Novel centrifugal method for simple and highly efficient adenovirus-mediated green fluorescence protein gene transduction into human monocyte-derived dendritic cells. J Immunol Methods 253:113-124, 2001.
41.Tillman BW, de Gruijl TD, Luykx-de Bakker SA, Scheper RJ, Pinedo HM, Curiel TJ, Gerritsen WR, Curiel DT:Maturation of dendritic cells accompanies high-efficiency gene transfer by a CD40-targeted adenoviral vector. J Immunol 162:6378-6383, 1999.
42.Zhong L, Granelli-Piperno A, Choi Y, Steinman RM : Recombinant adenovirus is an efficient and non-perturbing genetic vector for human dendritic cells. Eur J Immunol 29:964-972, 1999.
43.Diao J, Smythe JA, Smyth C, Rowe PB, Alexander IE : Human PBMC-derived dendritic cells transduced with an adenovirus vector induce cytotoxic T-lymphocyte responses against a vector-encoded anatigen in vitro. Gene Ther 6:845-853, 1999.
44.Chinnasamy N, Chinnasamy D, Toso JF, Lapointe R, Candotti F, Morgan RA Hwu P : Efficient gene transfer to human peropheral blood monocyte-derived dendritic cells using human immunodeficiency virus type1-based lentivirus vectors. Hum Gene Ther 11:1901-1909, 2000.
45.Evans JT, Cravens P, Lipsky PE, Garcia JV : Differentiation and expansion of lentivirus vector-marked dendritic cells derived from human CD34+ cells. Hum Gene Ther 11:2483-2492, 2000.
46.Ponnazhagan S, Mahendra G, Curiel DT, Shaw DR :Adeno-associated virus type 2-mediated transduction of human monocyte-derived dendritic cells : implication for ex vivo immunotherapy. J Virol 75:9493-9501, 2001.
47.Strobel I, Krumbholz M, Menke A, Hoffmann E, Dunbar PR, Bender A, Hobom G, Steinkasserer A, Schuler G, Grassmann R : Efficient expression of the tumor-associated antigen MAGE-3 in human dendritic cells, using an avian influenza virus vector. Hum Gene Ther 11:2207-2218, 2000.
48.Zhu M, Terasawa H, Gulley J, Panicali D, Arlen P, Schlom J, Tsang KY : Enhanced activation of human T cells via avipox vector-mediated hyperexpression of a triad of costimulatory molecules in human dendritic cells. Cancer Res 61:3725-3734, 2001.
49.Van der Bruggen P, Traversari C, Chomez P, Lurquin C, De Plaen E, Van den Eynde B, Knuth A, Boon T : A gene encoding an antigen recognized by cytolytic T lymphocytes on a human melanoma. Science 254:1643-1647, 1991.
50.Gaugler B, Van den Eynde B, Van der Bruggen P, Romero P, Gaforio JJ, De Plaen E, Lethe B, Brasseur F, Boon T : Human gene MAGE-3 codes for an antigen recognized on a melanoma by autologous cytolytic T lymphocytes. J Exp Med 179:921-930, 1994.
51.Van der Bruggen P, Bastin J, Gajewski T, Coulie PG, Boel P, De Smet C, Traversari C, Townsend A, Boon T : A peptide encoded by human gene MAGE-3 and presented by HLA-A2 induces cytolytic T lymphocytes that recognize tumor cells expressing MAGE-3. Eur J Immunol 24:3038-3043, 1994.
52.Kawakami Y, Eliyahu S, Delgado CH, Robbins PF, Rivoltini L, Topalian SL, Miki T, Rosenberg SA : Cloning of the gene coding for a shared human melanoma antigen recognized by autologous T cells infiltrating into tumor. Proc Natl Acad Sci U S A 91:3515-3519, 1994.
53.Kawakami Y, Eliyahu S, Delgado CH, Robbins PF, Sakaguchi K, Appella E, Yannelli JR, Adema GJ, Miki T, Rosenberg SA : Identification of a human melanoma antigen recognized by tumor-infiltrating lymphocytes associated with in vivo tumor rejection. Proc Natl Acad Sci USA 91:6458-6462, 1994.
54.Marchand M, van Baren N, Weynants P, Brichard V, Dreno B, Tessier MH, Rankin E, Parmiani G, Arienti F, Humblet Y, Bourlond A, Vanwijck R, Lienard D, Beauduin M, Dietrich PY, Russo V, Kerger J, Masucci G, Jager E, De Greve J, Atzpodien J, Brasseur F, Coulie PG, van der Bruggen P, Boon T : Tumor regressions observed in patients with metastatic melanoma treated with an antigenic peptide encoded by gene MAGE-3 and presented by HLA-A1. Int J Cancer 80:219-230, 1999.
55.Rosenberg SA, Yang JC, Schwartzentruber DJ, Hwu P, Marincola FM, Topalian SL, Restifo NP, Dudley ME, Schwarz SL, Spiess PJ, Wunderlich JR, Parkhurst MR, Kawakami Y, Seipp CA, Einhorn JH, White DE : Immunologic and therapeutic evaluation of a synthetic peptide vaccine for the treatment of patients with metastatic melanoma. Nat Med 4:321-327, 1998.
56.Mayordomo JI, Zorina T, Storkus WJ, Zitvogel L, Celluzzi C, Falo LD, Melief CJ, Ildstad ST, Martin Kast W, Deleo AB, Lotze MT : Bone marrow-derived dendritic cells pulsed with synthetic tumour peptides elicit protective and therapeutic antitumor immunity. Nature Med 1:1297-1302, 1995.
57.Porgador A, Gilboa E : Bone marrow-generated dendritic cells pulsed with a class I-restricted peptide are potent inducers of cytotoxic T lymphocytes. J Exp Med 182:255-260, 1995.
58.Gong J, Chen D, Kashiwaba M, Kufe D : Induction of antitumor activity by immunization with fusions of dendritic and carcinoma cells. Nat Med 3:558-561, 1997.
59.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 186 : 1177-1182, 1997.
60.Zitvogel L, Regnault A, Lozier A, Wolfers J, Flament C, Tenza D, Ricciardi-Castagnoli P, Raposo G, Amigorena S:Eradication of established murine tumors using a novel cell-free vaccine : dendritic cell-derived exosomes. Nat Med 4:594-600, 1998.
61.Specht JM, Wang G, Do MT, Lam JS, Royal RE, Reeves ME, Rosenberg SA, Hwu P : Dendritic cells retrovirally transduced with a model antigen gene are therapeutically effective against established pulmonary metastases. J Exp Med 186, 1213-1221, 1997.
62.Song W, Kong HL, Carpenter H, Torii H, Granstein R, Rafii S, Moore MA, Crystal RG:Dendritic cells genetically modified with an adenovirus vector encoding the cDNA for a model antigen induce protective and therapeutic antitumor immunity. J Exp Med 186:1247-1256, 1997.
63.Butterfield LH, Jilani SM, Chakraborty NG, Bui LA, Ribas A, Dissette VB, Lau R, Gamradt SC, Glaspy JA, McBride WH, Mukherji B, Economou JS : Generation of melanoma-specific cytotoxic T lymphocytes by dendritic cells transduced with a MART-1 adenovirus. J Immunol 161 : 5607-5613, 1998.
64.Perez-Diez A, Butterfield LH, Li L, Chakraborty NG, Economou JS, Mukherji B : Generation of CD8+ and CD4+ T-cell response to dendritic cells genetically engineered to express the MART-1/Melan-A gene. Cancer Res 58:5305-5309, 1998.
65.Butterfield LH, Koh A, Meng W, Vollmer CM, Ribas A, Dissette V, Lee E, Glaspy JA, McBride WH, Economou JS : Generation of human T-cell responses to an HLA-A2.1-restricted peptide epitope derived from alpha-fetoprotein. Cancer Res 59 :3134-3142, 1999.
66.Vollmer CM Jr, Eilber FC, Butterfield LH, Ribas A, Dissette VB, Koh A, Montejo LD, Lee MC, Andrews KJ, McBride WH, Glaspy JA, Economou JS : Alpha-fetoprotein-specific genetic immunotherapy for hepatocellular carcinoma. Cancer Res 59:3064-3067, 1999.
67.Ishida T, Chada S, Stipanov M, Nadaf S, Ciernik FI, Gabrilovich DI, Carbone DP : Dendritic cells transduced with wild-type p53 gene elicit potent anti-tumour immune responses. Clin Exp Immunol 117:244-251, 1999.
68.Tuting T, Wilson CC, Martin DM, Kasamon YL, Rowles J, Ma DI, Slingluff CL Jr, Wagner SN, van der Bruggen P, Baar J, Lotze MT, Storkus WJ :Autologous human monocyte-derived dendritic cells genetically modified to express melanoma antigens elicit primary cytotoxic T cell responses in vitro : enhancement by cotransfection of genes encoding the Th1-biasing cytokines IL-12 and IFN-alpha. J Immunol160:1139-1147, 1998.
69.Lamont, A.G. and Adorini, L. IL-12 : a key cytokine in immune regulation. Immunol Today 17, 214-217, 1996.
70.Mehrotra, P T, James A, D W, Mostowski, H S Siegel, J P : Effects of IL-12 on the generation of cytotoxic activity in human CD8+ T lymphocytes. J Immunol. 151:2444-2452, 1993.
71.Aste-Amezaga, M, D'Andrea, A, Kubin, M, Trinchieri, G : Cooperation of natural killer cell stimulatory factor/Interleukin-12 with other stimuli in the induction of cytokines and cytotoxic cell-associated molecules in human T and NK cells. Cell Immunol 156:480-492, 1994.
72.Nastala CL, Edington HD, McKinney TG, Tahara H, Nalesnik MA, Brunda MJ, Gately MK, Wolf SF, Schreiber RD, Storkus WJ, Lotze MT:Recombinant IL-12 administration induces tumor regression in association with IFN-gamma production. J Immunol 153:1697-1706, 1994.
73.Tahara, H, Zitvogel, L, Storkus, W J, Zeh III, H J, McKinney, T G, Schreiber, R D, Gubler, U, Robbins, P D, Lotze, M T : Effective eradication of established murine tumors with IL-12 gene therapy using a polycistronic retroviral vector. J Immunol 154, 6466-6474, 1995.
74.Melero I, Duarte M, Ruiz J, Sangro B, Galofre J, Mazzolini G, Bustos M, Qian C, Prieto J: Intratumoral injection of bone-marrow derived dendritic cells engineered to produce interleukin-12 induces complete regression of established murine transplantable colon adenocarcinomas. Gene Ther 6:1779-1784, 1999.
75.Miller PW, Sharma S, Stolina M, Butterfield LH, Luo J, Lin Y, Dohadwala M, Batra RK, Wu L, Economou JS, Dubinett SM:Intratumoral administration of adenoviral interleukin 7 gene-modified dendritic cells augments specific antitumor immunity and achieves tumor eradication. Hum Gene Ther 11:53-65, 2000.
76.Nishimura, N, Nishioka, Y, Shinohara, T, Sone, S : Enhanced efficiency by centrifugal manipulation of adenovirus-mediated interleukin 12 gene transduction into human monocyte-derived dendritic cells. Hum Gene Ther. 12:333-346, 2001.
77.Cao X, Zhang W, He L, Xie Z, Ma S, Tao Q, Yu Y, Hamada H, Wang J : Lymphotactin gene-modified bone marrow dendritic cells act as more potent adjuvants for peptide delivery to induce specific antitumor immunity. J Immunol 161:6238-6244, 1998.
78.Kikuchi T, Moore MA, Crystal RG : Dendritic cells modified to express CD40 ligand elicit therapeutic immunity against preexisting murine tumors. Blood 96:91-99, 2000.
79.Tsang KY, Zhu M, Even J, Gulley J, Arlen P, Schlom J : The infection of human dendritic cells with recombinant avipox vectors expressing a costimulatory molecule transgene (CD80) to enhance the activation of antigen-specific cytolytic T cells. Cancer Res 61:7568-7576, 2001.
80.Eggert AA, Schreurs MW, Boerman OC, Oyen WJ, de Boer AJ, Punt CJ, Figdor CG, Adema GJ:Biodistribution and vaccine efficiency of murine dendritic cells are dependent on the route of administration. Cancer Res 59:3340-3345, 1999.
81.Morse MA, Coleman RE, Akabani G, Niehaus N, Coleman D, Lyerly HK : Migration of human dendritic cells after injection in patients with metastatic malignancies. Cancer Res 59:56-58, 1999.
82.Mortarini R, Anichini A, Di Nicola M, Siena S, Bregni M, Belli F, Molla A, Gianni AM, Parmiani G:Autologous dendritic cells derived from CD34+ progenitors and from monocytes are not functionally equivalent antigen-presenting cells in the induction of Melan-A/Mart-127-35-specific CTLs from peripheral blood lymphocytes of melanoma patients with low frequency of CTL precursors. Cancer Res 57:5534, 1997.
83.Ferlazzo G, Wesa A, Wei WZ, Galy A : Dendritic cells generated either from CD34+ progenitor cells or from monocytes differ in their ability to activate antigen-specific CD8+ T cells. J Immunol 163:3597-604, 1999.
84.Rissoan MC, Soumelis V, Kadowaki N, Grouard G, Briere F, de Waal Malefyt R, Liu YJ:Reciprocal control of T helper cell and dendritic cell differentiation. Science 283:1183-1186, 1999.
85.Nishioka, Y, Nishimura, N, Suzuki, Y, Sone, S :Human monocyte-derived and CD83+ blood dendritic cells enhance NK cell-mediated cytotoxicity. Eur J Immunol 31:2633-2641, 2001.
86.Cella M, Jarrossay D, Facchetti F, Alebardi O, Nakajima H, Lanzavecchia A, Colonna M : Plasmacytoid monocytes migrate to inflamed lymph nodes and produce large amounts of type I nterferon. Nat Med 185:1101-1111, 1999.
87.Hirao M, Onai N, Hiroishi K, Watkins SC, Matsushima K, Robbins PD, Lotze MT, Tahara H : CC chemokine receptor-7 on dendritic cells is induced after interaction with apoptotic tumor cells:critical role in migration from the tumor site to draining lymph nodes. Cancer Res 60:2209-2217, 2000.

Received for publication December 10, 2001 ; accepted January 21, 2002.

Address correspondence and reprint requests to Yasuhiko Nishioka, M.D., Third Department of Internal Medicine, The University of Tokushima School of Medicine, Kuramoto-cho, Tokushima770-8503, Japan and Fax:+81-88-633-2134.