当前位置: 首页 > 期刊 > 《干细胞学杂志》 > 2004年第4期 > 正文
编号:11340504
Cellular Immunotherapy with Dendritic Cells in Cancer: Current Status
http://www.100md.com 《干细胞学杂志》
     a Massachusetts Institute of Technology, Center for Cancer Research, Cambridge, Massachusetts, USA;

    b Department of Internal Medicine, University of Genova, Genova, Italy;

    c Department of Hematology, Oncology and Immunology, University of Tübingen, Tübingen, Germany

    Key Words. Dendritic cells ? Tumor antigens ? Vaccinations

    Correspondence: Peter Brossart, M.D., Department of Hematology, Oncology and Immunology, University of Tübingen, Otfried-Müller Str. 10, D-72076 Tübingen, Germany. Telephone: 49-7071-298-2726; Fax: 49-7071-295709; e-mail: peter.brossart@med.uni-tuebingen.de

    ABSTRACT

    The first demonstration that tumor rejection antigens exist goes back to the late 1980s when tumor-infiltrating lymphocytes from melanoma patients were shown to lyse HLA-matched melanoma cell lines, suggesting the existence of shared melanoma antigens . In the subsequent years, the first genes encoding tumor antigens (such as tyrosinase, gp-100, the MART and MAGE genes) were cloned, and the immunogenic epitopes were identified. These and subsequent studies pointed out that tumors often upregulate the expression of molecules that are normally suppressed or expressed at much lower levels in adult tissues. T lymphocytes capable of recognizing these antigens usually exist in the periphery, possibly due to the lack of presentation of these antigens during thymic selection or lower avidity of the T-cell receptor (TCR) . However, in most cases, the immune system fails to recognize and destroy tumor cells that may give rise to clinically relevant malignancies. The tumor escape mechanisms include the inefficiency of tumor cells as antigen-presenting cells (APCs) and the lack of efficient contact between immune system and tumor cells .

    There is also evidence that antitumor immune responses can extinguish established tumors, especially in patients affected by melanoma or renal cell carcinoma. Infiltration of the primary tumor with lymphocytes has been associated with a better prognosis in different types of malignancies . Similarly, immune-mediated paraneoplastic syndromes characterized by an immune response directed against antigens shared by tumor and normal tissues (such as the central nervous system) have been associated with a better clinical outcome and even with spontaneous tumor regressions . However, such spontaneous immune responses are rare and still remain largely elusive. Thus, the goal of modern tumor immunotherapy is to trigger the immune system in order to mimic such rejection events and improve clinical outcome.

    Particularly, the induction CD8+ cytotoxic T lymphocytes (CTLs) directed against tumor epitopes in vivo is the desirable effect of a specific immunotherapy approach, given that these immune effectors are mainly responsible for tumor rejection . These lymphocytes recognize via the TCR 8–11 amino acids–long peptide epitopes in the context of the HLA class I molecules. Upon encounter with cells that express the target antigen, the CTLs activate their lytic machinery and kill the cells. The induction of CD4+ helper T cells also plays a major role in antitumor immunity, and immunization strategies should probably take into account providing immunogenic epitopes for these lymphocytes.

    The first immunization strategies for cancer patients often involved the administration of tumor lysates or irradiated tumor cells together with immunological adjuvants such as bacillus Calmette-Guérin (BCG) . This vaccination method has recently been reported by Vermorken et al. to be possibly associated with a protection from relapses in patients with stage-II colorectal cancer, but this study needs further confirmation by other groups. Such an approach is limited by the requirement of sufficient amount of tumor material and by potential concerns related to the administration of autologous tumor cells, though irradiated, to patients in clinical remission of disease.

    Recent advances in the knowledge of the immune system have opened new perspectives for the development of antitumor immunization strategies. In particular, the administration of immunogenic APCs such as dendritic cells (DCs) loaded with tumor antigens is now considered one of the most promising approaches to the specific cancer immunotherapy and is being evaluated in many cancer centers for different malignancies and in different clinical settings.

    DCs are leukocytes that are highly specialized in the capture and presentation of antigens to T cells . They are presently believed to control the induction (and, possibly, the suppression) of antigen-specific immune responses in vivo . DCs for clinical use can be generated in sufficient numbers from circulating precursors, including peripheral blood CD14+ monocytes and CD34+ stem cells . Injection of DCs loaded with tumor-associated antigens (TAAs) into patients was shown to break tolerance and to induce antitumor cytotoxic immune responses in vivo . The DC-based clinical trials performed so far have demonstrated that this form of immunotherapy is feasible and safe . Moreover, some studies reported cases of tumor regression or growth arrest following DC administration.

    DEFINITION OF DCS AND METHODS FOR DC GENERATION

    Anticancer vaccinations attempt to elicit tumor-directed CD8+ CTLs that lyse tumor cells presenting MHC class I–associated peptides derived from tumor-associated proteins. Several different strategies are currently available to deliver antigens into DCs during the ex vivo manipulation for further presentation to T cells in the recipient. DCs can be pulsed with synthetic peptide epitopes derived from known TAAs such as MUC1, Her-2/neu, survivin, tyrosinase, telomerase, CEA, p53, MAGE, or Melan-A/MART . Although most of these peptides are designed to bind HLA-A2, the most common HLA class I molecule among Caucasians, several peptide epitopes have been identified that bind to other HLA class I alleles. Moreover, HLA class II binding peptides have also been reported that either unspecifically trigger CD4+ lymphocyte activation or are derived from tumor antigens and induce antigen-specific CD4+ helper T cells . Many of these peptides are now commercially available and ready to use under GMP conditions. Hence, peptide-based vaccinations are potentially applicable to most patients. In some immunization studies, peptides are injected directly into the patients without previous incubation with DCs ex vivo. For this kind of approach, the peptides are usually coinjected with immune cytokines such as GM-CSF, which favors DC migration and activation in situ, or with incomplete Freund adjuvant . Major drawbacks related to the use of peptides are (a) the restriction to some HLA class I alleles, (b) the need to determine the expression of the target antigen by a tumor, and (c) the likelihood that targeting single or few tumor epitopes may impede the detection of tumor cells that downregulate those antigens. However, to some extent, tumor escape may be prevented by the expansion of lymphocytes directed against epitopes other than those used for immunization, a phenomenon named "epitope spreading," which has already been observed in some clinical studies .

    Another approach is to use recombinant proteins as antigens. These are captured by DCs, then processed and presented in the form of immunogenic peptides in the context of HLA molecules. This approach bypasses the HLA restriction of the peptides and was successfully applied to the treatment of patients with follicular lymphoma . In this case, DCs were pulsed ex vivo with tumor-specific idiotype protein to induce antitumor immunity, with encouraging clinical results. The effectiveness of the same approach for the treatment of myeloma is still under investigation . Similarly, a recombinant prostatic acid phosphatase (PAP) has been used to load autologous DCs by Fong et al. . An alternative strategy is gene-based delivery of TAAs into DCs. DCs can be transduced with recombinant viruses (retroviral or adenoviral vectors, vaccinia virus) or transfected with RNA encoding for a specific tumor antigen .

    Several other approaches also exist that, instead of using single or few antigens, make use of whole tumor material as an antigenic source. These approaches use tumor lysates, dead tumor cells (apoptotic bodies, necrotic cells), DCs fused with tumor cells, or total tumor RNA . All of these methods were shown to induce immunity against the parental tumor and are being evaluated in the clinical setting. Importantly, whole tumor–derived materials represent the entire antigenic repertoire of a tumor; thus the resulting immune response simultaneously targets many tumor antigens. In fact, it is likely that preferential expansion of CTLs directed against immunodominant epitopes will happen in some cases; this may be related to the higher frequency of some epitope-specific effectors or to the strong immunogenicity of some tumor-derived peptides (or both) . One potential advantage of the use of RNA compared with the other tumor-derived materials cited above is that methods exist for the unspecific amplification of messenger RNA . This translates into the applicability of this method also in those cases when small tumor specimens such as needle biopsies would not be sufficient to obtain lysates or apoptotic tumor cells for DC pulsing. Moreover, the antigens encoded by the transfected RNA may be processed and presented on both HLA class I and class II molecules, thus inducing CD8+ as well as CD4+ antitumor lymphocytes . The elicited immune response, at least according to the in vitro experiments, seems to be restricted to immunodominant tumor epitopes while saving nonmalignant autologous cells . Thus, RNA transfection of DCs appears as a very attractive approach for the induction of antitumor CTLs in a variety of malignancies.

    In vivo DC loading has also been evaluated in preclinical models. In particular, immunization with DNA vaccines by gene gun represents an attractive approach; here gold particles coated with expression plasmid DNA encoding target genes are "bombarded" into the skin . This procedure transfects plasmid DNA directly into the DCs present in the skin. Transfected DCs express the encoded antigen and present the processed peptides to the antigen-specific T cells to initiate an immune response in the afferent lymph nodes. In light of the results reported by Sudowe et al. and Garg et al. in the animal model, this approach may reveal as an effective method for antitumor immunity induction.

    ROUTES OF DC DELIVERY

    The detection of the immune response to tumor antigens following vaccination represents one of the major endpoints of the clinical vaccination studies. The DTH assay represents a possible approach to this goal. It is usually performed by intradermal injection of tumor-derived material or DCs loaded with tumor antigen(s) before and after the vaccination course . In the case of tumor regression after vaccine administration, the detection of either infiltrating lymphocytes or inflammatory cells (or both) in tumor specimens, whenever these are easily reachable, should be performed in order to correlate the clinical outcome with the elicited immune response . In some cases, the tumor-infiltrating lymphocytes can be isolated and further characterized . However, in most studies the lymphocytes reacting to the tumor antigens have been detected in the peripheral blood mononuclear cells. The T cells specific for a defined tumor-derived epitope can be tracked via different approaches, which typically include ELISPOT, cytokine secretion assay from Miltenyi, intracellular staining for IFN-, tetramers, proliferation assays, ELISAs, cytotoxicity assays, and real-time polymerase chain reaction for IFN- . The results obtained with different methods are often, though not necessarily, consistent, and further refining of these techniques is still required .

    When whole tumor–derived material (tumor lysates, total tumor RNA, fusions, tumor-derived peptides) is used as an antigen source for vaccine preparation, the autologous tumor cells or tumor material, when available, can be used to determine immunoreactivity before and after vaccine administration . The same DCs loaded with tumor antigens may work as a suitable target in immunological monitoring . In this kind of approach, the antigens involved in the immune response are often not known. However, in selected patients who express defined HLA alleles and tumor antigens, the immunization against known tumor epitopes could also be evaluated .

    Some studies found no correlation between the immune response to the antigen used for immunization and the clinical outcome, since some tumor regressions were observed in patients who showed little response to vaccination . Besides, different studies have already reported the expansion of lymphocytes specific for different tumor epitopes following vaccination , and in the study by Butterfield et al. the only complete clinical remission was induced in a patient showing epitope spreading. These data indicate that immunity versus an array of different tumor antigens, including molecules not present in the vaccine preparation, should possibly be monitored.

    An improved characterization (phenotypic and functional) of the antitumor T lymphocytes will also be necessary for a better understanding of the lymphocyte subsets involved in tumor rejections. This goal can be pursued by combining tetramer staining with antibodies for surface markers such as CD45RA, CD45RO, CD27, CCR7, CD28, and CD25 or with intracellular staining for cytokines such as IFN-, IL-4, IL-10 . The antigen-specific lymphocytes can be isolated by fluorescence-activated cell sorter (FACS) or magnetic cell sorting (MACS) technology, expanded, and further characterized.

    Finally, pulsing DCs with immunogenic epitopes (such as influenza peptides or CD4 epitopes) or antigens (such as keyhole limpet hemocyanin or HBsAg) has been performed with the double intent to exploit them as an immune adjuvant and to use them as an immunological tracer to evaluate DC priming efficacy in vivo and responsiveness of the immune system to vaccine administration . The use of these immunogens may be particularly useful in order to compare different vaccine administration routes and schedules or DCs generated according to different protocols.

    CLINICAL STUDIES WITH DCS

    The phase I and II clinical studies with DCs are hardly comparable, given that different methods for DC culture, antigen loading, and administration have been used. Altogether, the data reported so far indicate that these ex vivo–generated APCs are immunogenic in vivo and that DC injection was associated with a clinical response in some patients. Phase III studies are necessary to evaluate the potential clinical advantages of DC vaccination and are already ongoing for some diseases, such as melanoma and prostate cancer . It is a general conviction that, if any, the clinical benefits associated with this immunotherapeutic approach are more likely to be recorded among patients in remission of disease or with small tumor burden. Meanwhile, it seems probable that the efficacy of DC vaccinations will be improved by the novel methods of antigen loading and by the concomitant administration of cytokines or immunogenic factors such as IL-2, IL-12, or CpG dinucleotides, which should amplify the immune response in vivo.

    Particularly appealing is the application of DCs to allogeneic bone marrow and PBPC transplantations. In this context, the recently developed protocols for reduced-intensity conditioning (the so-called mini-allo) have increased the safety of this kind of treatment and extended its applicability in leukemia (also in older patients), Hodgkin and non-Hodgkin lymphoma, myeloma, and nonhematological malignancies such as renal cell carcinoma and breast cancer . In this context, DCs could be used for the expansion and adoptive transfer of lymphocytes against TAAs or minor histocompatibility antigens . This may help to selectively target the graft-versus-tumor reaction, while possibly minimizing the graft-versus-host effect.

    ACKNOWLEDGMENTS

    Rosenberg SA. Progress in human tumour immunology and immunotherapy. Nature 2001;411:380–384.

    Pardoll DM. Cancer vaccines. Nat Med 1998;4:525– 531.

    Dunn GP, Bruce AT, Ikeda H et al. Cancer immunoediting: from immunosurveillance to tumor escape. Nat Immunol 2002;3:991–998.

    Ochsenbein AF, Sierro S, Odermatt B et al. Roles of tumour localization, second signals and cross priming in cytotoxic T-cell induction. Nature 2001;411: 1058–1064.

    Drake CG, Pardoll DM. Tumor immunology: towards a paradigm of reciprocal research. Semin Cancer Biol 2002;12:73–80.

    Darnell RB, Posner JB. Paraneoplastic syndromes involving the nervous system. N Engl J Med 2003;349: 1543–1554.

    Vermorken JB, Claessen AM, van Tinteren H et al. Active specific immunotherapy for stage II and stage III human colon cancer: a randomised trial. Lancet 1999; 353:345–350.

    Banchereau J, Briere F, Caux C et al. Immunobiology of dendritic cells. Annu Rev Immunol 2000;18:767–811.

    Lanzavecchia A, Sallusto F. Regulation of T cell immunity by dendritic cells. Cell 2001;106:263–266.

    Steinman RM, Hawiger D, Nussenzweig MC. Tolerogenic dendritic cells. Annu Rev Immunol 2003;21: 685– 711.

    Banchereau J, Schuler-Thurner B, Palucka AK et al. Dendritic cells as vectors for therapy. Cell 2001;106: 271–274.

    Brossart P, Wirths S, Brugger W et al. Dendritic cells in cancer vaccines. Exp Hematol 2001;29:1247–1255.

    Schuler G, Schuler-Thurner B, Steinman RM. The use of dendritic cells in cancer immunotherapy. Curr Opin Immunol 2003;15:138–147.

    Fong L, Engleman EG. Dendritic cells in cancer immunotherapy. Annu Rev Immunol 2000;18:245–273.

    Ribas A, Butterfield LH, Glaspy JA et al. Current developments in cancer vaccines and cellular immunotherapy. J Clin Oncol 2003;21:2415–2432.

    Kurts C, Heath WR, Carbone FR et al. Constitutive class I-restricted exogenous presentation of self antigens in vivo. J Exp Med 1996;184:923–930.

    Mahnke K, Schmitt E, Bonifaz L et al. Immature, but not inactive: the tolerogenic function of immature dendritic cells. Immunol Cell Biol 2002;80:477–483.

    Steinman RM, Nussenzweig MC. Avoiding horror autotoxicus: the importance of dendritic cells in peripheral T cell tolerance. Proc Natl Acad Sci U S A 2002;99:351– 358.

    Nencioni A, Grunebach F, Zobywlaski A et al. Dendritic cell immunogenicity is regulated by peroxisome proliferator-activated receptor gamma. J Immunol 2002;169:1228–1235.

    Von Herrath MG, Harrison LC. Antigen-induced regulatory T cells in autoimmunity. Nat Rev Immunol 2003;3:223–232.

    Lechmann M, Berchtold S, Hauber J et al. CD83 on dendritic cells: more than just a marker for maturation. Trends Immunol 2002;23:273–275.

    Grunebach F, Weck MM, Reichert J et al. Molecular and functional characterization of human Dectin-1. Exp Hematol 2002;30:1309–1315.

    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 alpha. J Exp Med 1994;179: 1109–1118.

    Brossart P, Grunebach F, Stuhler G et al. Generation of functional human dendritic cells from adherent peripheral blood monocytes by CD40 ligation in the absence of granulocyte-macrophage colony-stimulating factor. Blood 1998;92:4238–4247.

    Dhodapkar MV, Krasovsky J, Steinman RM et al. Mature dendritic cells boost functionally superior CD8+ T-cell in humans without foreign helper epitopes. J Clin Invest 2000;105:R9–R14.

    Jego G, Palucka AK, Blanck JP et al. Plasmacytoid dendritic cells induce plasma cell differentiation through type I interferon and interleukin 6. Immunity 2003; 19:225–234.

    Scheding S, Kratz-Albers K, Meister B et al. Ex vivo expansion of hematopoietic progenitor cells for clinical use. Semin Hematol 1998;35:232–240.

    Brugger W, Brossart P, Scheding S et al. Approaches to dendritic cell-based immunotherapy after peripheral blood stem cell transplantation. Ann N Y Acad Sci 1999;872:363–371.

    Klangsinsirikul P, Russell NH. Peripheral blood stem cell harvests from G-CSF-stimulated donors contain a skewed Th2 CD4 phenotype and a predominance of type 2 dendritic cells. Exp Hematol 2002;30:495–501.

    Gazitt Y. Comparison between granulocyte colony-stimulating factor and granulocyte-macrophage colony-stimulating factor in the mobilization of peripheral blood stem cells. Curr Opin Hematol 2002; 9:190–198.

    Brossart P, Zobywalski A, Grunebach F et al. Tumor necrosis factor alpha and CD40 ligand antagonize the inhibitory effects of interleukin 10 on T-cell stimulatory capacity of dendritic cells. Cancer Res 2000;60: 4485–4492.

    Mortarini R, Anichini A, Di Nicola M et al. Autologous dendritic cells derived from CD34+ progenitors and from monocytes are not functionally equivalent antigen-presenting cells in the induction of melan-A/Mart-1(27–35)-specific CTLs from peripheral blood lymphocytes of melanoma patients with low frequency of CTL precursors. Cancer Res 1997;57:5534–5541.

    Banchereau J, Palucka AK, Dhodapkar M et al. Immune and clinical responses in patients with metastatic melanoma to CD34+ progenitor-derived dendritic cell vaccine. Cancer Res 2001;61:6451–6458.

    Mackensen A, Herbst B, Chen JL et al. Phase I study in melanoma patients of a vaccine with peptide-pulsed dendritic cells generated in vitro from CD34+ hematopoietic progenitor cells. Int J Cancer 2000;86: 385– 392.

    Triozzi PL, Kim J, Aldrich W. Infusion of unpulsed dendritic cells derived from granulocyte/macrophage colony-stimulating factor-mobilized peripheral blood CD34+ cells and monocytes in patients with advanced carcinoma. J Hematother Stem Cell Res 2003;12: 279–287.

    Hsu FJ, Benike C, Fagnoni F et al. Vaccination of patients with B-cell lymphoma using autologous antigen-pulsed dendritic cells. Nat Med 1996;2:52–58.

    Timmerman JM, Czerwinski DK, Davis TA et al. Idiotype-pulsed dendritic cell vaccination for B-cell lymphoma: clinical and immune responses in 35 patients. Blood 2002;99:1517–1526.

    Fong L, Brockstedt D, Benike C et al. Dendritic cell-based xenoantigen vaccination for prostate cancer immunotherapy. J Immunol 2001;167:7150–7156.

    Fong L, Hou Y, Rivas A et al. Altered peptide ligand vaccination with Flt3 ligand expanded dendritic cells for tumor immunotherapy. Proc Natl Acad Sci U S A 2001;98:8809–8814.

    Small EJ, Fratesi P, Reese DM et al. Immunotherapy of hormone-refractory prostate cancer with antigen-loaded dendritic cells. J Clin Oncol 2000;18:3894–3903.

    Jefford M, Schnurr M, Toy T et al. Functional comparison of DCs generated in vivo with Flt3 ligand or in vitro from blood monocytes: differential regulation of function by specific classes of physiologic stimuli. Blood 2003;102:1753–1763.

    Scandella E, Men Y, Gillessen S et al. Prostaglandin E2 is a key factor for CCR7 surface expression and migration of monocyte-derived dendritic cells. Blood 2002; 100:1354–1361.

    Dhodapkar MV, Steinman RM, Krasovsky J et al. Antigen-specific inhibition of effector T cell function in humans after injection of immature dendritic cells. J Exp Med 2001;193:233–238.

    Jonuleit H, Giesecke-Tuettenberg A, Tuting T et al. A comparison of two types of dendritic cell as adjuvants for the induction of melanoma-specific T-cell responses in humans following intranodal injection. Int J Cancer 2001;93:243–251.

    Thomas R, Chambers M, Boytar R et al. Immature human monocyte-derived dendritic cells migrate rapidly to draining lymph nodes after intradermal injection for melanoma immunotherapy. Melanoma Res 1999;9:474–481.

    Nestle FO, Alijagic S, Gilliet M et al. Vaccination of melanoma patients with peptide- or tumor lysate-pulsed dendritic cells. Nat Med 1998;4:328–332.

    Heiser A, Coleman D, Dannull J et al. Autologous dendritic cells transfected with prostate-specific antigen RNA stimulate CTL responses against metastatic prostate tumors. J Clin Invest 2002;109:409–417.

    Su Z, Dannull J, Heiser A et al. Immunological and clinical responses in metastatic renal cancer patients vaccinated with tumor RNA-transfected dendritic cells. Cancer Res 2003;63:2127–2133.

    Brossart P, Wirths S, Stuhler G et al. Induction of cytotoxic T-lymphocyte responses in vivo after vaccinations with peptide-pulsed dendritic cells. Blood 2000;96:3102–3108.

    Berger TG, Feuerstein B, Strasser E et al. Large-scale generation of mature monocyte-derived dendritic cells for clinical application in cell factories. J Immunol Methods 2002;268:131–140.

    Motta MR, Castellani S, Rizzi S et al. Generation of dendritic cells from CD14+ monocytes positively selected by immunomagnetic adsorption for multiple myeloma patients enrolled in a clinical trial of anti-idiotype vaccination. Br J Haematol 2003;121:240–250.

    Brossart P, Stuhler G, Flad T et al. Her-2/neu-derived peptides are tumor-associated antigens expressed by human renal cell and colon carcinoma lines and are recognized by in vitro induced specific cytotoxic T lymphocytes. Cancer Res 1998;58:732–736.

    Brossart P, Heinrich KS, Stuhler G et al. Identification of HLA-A2-restricted T-cell epitopes derived from the MUC1 tumor antigen for broadly applicable vaccine therapies. Blood 1999;93:4309–4317.

    Schmidt SM, Schag K, Muller MR et al. Survivin is a shared tumor-associated antigen expressed in a broad variety of malignancies and recognized by specific cytotoxic T cells. Blood 2003;102:571–576.

    Vonderheide RH, Hahn WC, Schultze JL et al. The telomerase catalytic subunit is a widely expressed tumor-associated antigen recognized by cytotoxic T lymphocytes. Immunity 1999;10:673–679.

    Alexander J, Sidney J, Southwood S et al. Development of high potency universal DR-restricted helper epitopes by modification of high affinity DR-blocking peptides. Immunity 1994;1:751–761.

    Weber J, Sondak VK, Scotland R et al. Granulocyte-macrophage-colony-stimulating factor added to a multipeptide vaccine for resected stage II melanoma. Cancer 2003;97:186–200.

    Muderspach L, Wilczynski S, Roman L et al. A phase I trial of a human papillomavirus (HPV) peptide vaccine for women with high-grade cervical and vulvar intraepithelial neoplasia who are HPV 16 positive. Clin Cancer Res 2000;6:3406–3416.

    Butterfield LH, Ribas A, Dissette VB et al. Determinant spreading associated with clinical response in dendritic cell-based immunotherapy for malignant melanoma. Clin Cancer Res 2003;9:998–1008.

    Reichardt VL, Okada CY, Liso A et al. Idiotype vaccination using dendritic cells after autologous peripheral blood stem cell transplantation for multiple myeloma: a feasibility study. Blood 1999;93:2411–2419.

    Reichardt VL, Milazzo C, Brugger W et al. Idiotype vaccination of multiple myeloma patients using monocyte-derived dendritic cells. Haematologica 2003;88:1139–1149.

    Van Tendeloo VF, Ponsaerts P, Lardon F et al. 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.

    Palmowski MJ, Choi EM, Hermans IF et al. Competition between CTL narrows the immune response induced by prime-boost vaccination protocols. J Immunol 2002;168:4391–4398.

    Muller MR, Grunebach F, Nencioni A et al. Transfection of dendritic cells with RNA induces CD4- and CD8-mediated T cell immunity against breast carcinomas and reveals the immunodominance of presented T cell epitopes. J Immunol 2003;170:5892–5896.

    Heiser A, Maurice MA, Yancey DR et al. Induction of polyclonal prostate cancer-specific CTL using dendritic cells transfected with amplified tumor RNA. J Immunol 2001;166:2953–2960.

    Grunebach F, Muller MR, Nencioni A et al. Delivery of tumor-derived RNA for the induction of cytotoxic T-lymphocytes. Gene Ther 2003;10:367–374.

    Sullenger BA, Gilboa E. Emerging clinical applications of RNA. Nature 2002;418:252–258.

    Muller MR, Tsakou G, Grunebach F et al. Induction of chronic lymphocytic leukemia (CLL)-specific CD4-and CD8-mediated T-cell responses using RNA-transfected dendritic cells. Blood 2004;103:1763–1769.

    Nencioni A, Muller MR, Grunebach F et al. Dendritic cells transfected with tumor RNA for the induction of antitumor CTL in colorectal cancer. Cancer Gene Ther 2003;10:209–214.

    Milazzo C, Reichardt VL, Muller MR et al. Induction of myeloma-specific cytotoxic T cells using dendritic cells transfected with tumor-derived RNA. Blood 2003;101:977–982.

    Sudowe S, Ludwig-Portugall I, Montermann E et al. Transcriptional targeting of dendritic cells in gene gun-mediated DNA immunization favors the induction of type 1 immune responses. Mol Ther 2003;8:567–575.

    Garg S, Oran A, Wajchman J et al. Genetic tagging shows increased frequency and longevity of antigen-presenting, skin-derived dendritic cells in vivo. Nat Immunol 2003;4:907–912.

    Triozzi PL, Khurram R, Aldrich WA et al. Intratumoral injection of dendritic cells derived in vitro in patients with metastatic cancer. Cancer 2000;89:2646–2654.

    Mullins DW, Sheasley SL, Ream RM et al. Route of immunization with peptide-pulsed dendritic cells controls the distribution of memory and effector T cells in lymphoid tissues and determines the pattern of regional tumor control. J Exp Med 2003;198:1023–1034.

    Mackensen A, Krause T, Blum U et al. Homing of intravenously and intralymphatically injected human dendritic cells generated in vitro from CD34+ hematopoietic progenitor cells. Cancer Immunol Immunother 1999;48:118–122.

    Fong L, Brockstedt D, Benike C et al. Dendritic cells injected via different routes induce immunity in cancer patients. J Immunol 2001;166:4254–4359.

    Martin-Fontecha A, Sebastiani S, Hopken UE et al. Regulation of dendritic cell migration to the draining lymph node: impact on T lymphocyte traffic and priming. J Exp Med 2003;198:615–621.

    Maier T, Tun-Kyi A, Tassis A et al. Vaccination of patients with cutaneous T-cell lymphoma using intranodal injection of autologous tumorlysate-pulsed dendritic cells. Blood 2003;102:2338–2344.

    Gilliet M, Kleinhans M, Lantelme E et al. Intranodal injection of semimature monocyte-derived dendritic cells induces T helper type 1 responses to protein neoantigen. Blood 2003;102:36–42.

    Bedrosian I, Mick R, Xu S et al. Intranodal administration of peptide-pulsed mature dendritic cell vaccines results in superior CD8+ T-cell function in melanoma patients. J Clin Oncol 2003;21:3826–3835.

    Marten A, Flieger D, Renoth S et al. Therapeutic vaccination against metastatic renal cell carcinoma by autologous dendritic cells: preclinical results and outcome of a first clinical phase I/II trial. Cancer Immunol Immunother 2002;51:637–644.

    O’Rourke MG, Johnson M, Lanagan C et al. Durable complete clinical responses in a phase I/II trial using an autologous melanoma cell/dendritic cell vaccine. Cancer Immunol Immunother 2003;52:387–395.

    Zehntner S, Townsend W, Parkes J et al. Tumor metastasis biopsy as a surrogate marker of response to melanoma immunotherapy. Pathology 1999;31:116–122.

    Housseau F, Lindsey KR, Oberholtzer SD et al. Quantitative real-time RT-PCR as a method for monitoring T lymphocyte reactivity to full-length tyrosinase protein in vaccinated melanoma patients. J Immunol Methods 2002;266:87–103.

    Thurner B, Haendle I, Roder C et al. Vaccination with mage-3A1 peptide-pulsed mature, monocyte-derived dendritic cells expands specific cytotoxic T cells and induces regression of some metastases in advanced stage IV melanoma. J Exp Med 1999;190:1669–1678.

    Lanzavecchia A, Sallusto F. Progressive differentiation and selection of the fittest in the immune response. Nat Rev Immunol 2002;2:982–987.

    Smithers M, O’Connell K, MacFadyen S et al. Clinical response after intradermal immature dendritic cell vaccination in metastatic melanoma is associated with immune response to particulate antigen. Cancer Immunol Immunother 2003;52:41–52.

    Schuler-Thurner B, Schultz ES, Berger TG et al. Rapid induction of tumor-specific type 1 T helper cells in metastatic melanoma patients by vaccination with mature, cryopreserved, peptide-loaded monocyte-derived dendritic cells. J Exp Med 2002;195:1279–1288.

    Holtl L, Zelle-Rieser C, Gander H et al. Immunotherapy of metastatic renal cell carcinoma with tumor lysate-pulsed autologous dendritic cells. Clin Cancer Res 2002;8:3369–3376.

    Lin CL, Lo WF, Lee TH et al. Immunization with Epstein-Barr Virus (EBV) peptide-pulsed dendritic cells induces functional CD8+ T-cell immunity and may lead to tumor regression in patients with EBV-positive nasopharyngeal carcinoma. Cancer Res 2002; 62:6952–6958.

    Maris M, Storb R. The transplantation of hematopoietic stem cells after non-myeloablative conditioning: a cellular therapeutic approach to hematologic and genetic diseases. Immunol Res 2003;28:13–24.

    Bethge WA, Hegenbart U, Stuart MJ et al. Adoptive immunotherapy with donor lymphocyte infusions after allogeneic hematopoietic cell transplantation following nonmyeloablative conditioning. Blood 2004;103: 790– 795.

    Carella AM, Cavaliere M, Lerma E et al. Autografting followed by nonmyeloablative immunosuppressive chemotherapy and allogeneic peripheral-blood hematopoietic stem-cell transplantation as treatment of resistant Hodgkin’s disease and non-Hodgkin’s lymphoma. J Clin Oncol 2000;18:3918–3924.

    Childs R, Chernoff A, Contentin N et al. Regression of metastatic renal-cell carcinoma after nonmyeloablative allogeneic peripheral-blood stem-cell transplantation. N Engl J Med 2000;343:750–758.

    Ueno NT, Cheng YC, Rondon G et al. Rapid induction of complete donor chimerism by the use of a reduced-intensity conditioning regimen composed of fludarabine and melphalan in allogeneic stem cell transplantation for metastatic solid tumors. Blood 2003;102: 3829–3836.

    Brossart P, Spahlinger B, Grunebach F et al. Induction of minor histocompatiblity antigen HA-1-specific cytotoxic T cells for the treatment of leukemia after allogeneic stem cell transplantation. Blood 1999;94: 4374–4376.

    Marten A, Renoth S, Heinicke T et al. Allogeneic dendritic cells fused with tumor cells: preclinical results and outcome of a clinical phase I/II trial in patients with metastatic renal cell carcinoma. Hum Gene Ther 2003; 14:483–494.

    Marten A, Flieger D, Renoth S et al. Therapeutic vaccination against metastatic renal cell carcinoma by autologous dendritic cells: preclinical results and outcome of a first clinical phase I/II trial. Cancer Immunol Immunother 2002;51:637–644.

    Oosterwijk-Wakka JC, Tiemessen DM, Bleumer I et al. Vaccination of patients with metastatic renal cell carcinoma with autologous dendritic cells pulsed with autologous tumor antigens in combination with interleukin-2: a phase 1 study. J Immunother 2002;25: 500–508.

    Krause SW, Neumann C, Soruri A et al. The treatment of patients with disseminated malignant melanoma by vaccination with autologous cell hybrids of tumor cells and dendritic cells. J Immunother 2002;25:421–428.

    Morse MA, Nair SK, Mosca PJ et al. Immunotherapy with autologous, human dendritic cells transfected with carcinoembryonic antigen mRNA. Cancer Invest 2003;21:341–349.

    Sadanaga N, Nagashima H, Mashino K et al. Dendritic cell vaccination with MAGE peptide is a novel therapeutic approach for gastrointestinal carcinomas. Clin Cancer Res 2001;7:2277–2284.

    Pecher G, Haring A, Kaiser L et al. Mucin gene (MUC1) transfected dendritic cells as vaccine: results of a phase I/II clinical trial. Cancer Immunol Immunother 2002;51:669–673.

    Chang AE, Redman BG, Whitfield JR et al. A phase I trial of tumor lysate-pulsed dendritic cells in the treatment of advanced cancer. Clin Cancer Res 2002;8: 1021–1032.

    Hernando JJ, Park TW, Kubler K et al. Vaccination with autologous tumour antigen-pulsed dendritic cells in advanced gynaecological malignancies: clinical and immunological evaluation of a phase I trial. Cancer Immunol Immunother 2002;51:45–52.

    Geiger JD, Hutchinson RJ, Hohenkirk LF et al. Vaccination of pediatric solid tumor patients with tumor lysate-pulsed dendritic cells can expand specific T cells and mediate tumor regression. Cancer Res 2001;61: 8513–8519.

    Kikuchi T, Akasaki Y, Irie M et al. Results of a phase I clinical trial of vaccination of glioma patients with fusions of dendritic and glioma cells. Cancer Immunol Immunother 2001;50:337–344.(Alessio Nencionia,b, Pete)