Published cancer vaccine trials reporting on Wilms' tumor 1–targeted active specific immunotherapy in patients with hematological malignancies or solid tumors are reviewed. An untapped potential for inducing cancer immunity with minimal side effects is revealed.
Learning Objectives
After completing this course, the reader will be able to:
Explain the role of the Wilm's tumor protein 1 (WT1) as a tumor antigen in peptide- and dendritic cell-based cancer immunotherapy trials.
Describe the immune responses elicited by WT1-based cancer vaccines and their potential for creating clinical responses in a majority of evaluable cancer patients
This article is available for continuing medical education credit at CME.TheOncologist.com
Abstract
There is a growing body of evidence that Wilms' tumor protein 1 (WT1) is a promising tumor antigen for the development of a novel class of universal cancer vaccines. Recently, in a National Cancer Institute prioritization project, WT1 was ranked first in a list of 75 cancer antigens. In this light, we exhaustively reviewed all published cancer vaccine trials reporting on WT1-targeted active specific immunotherapy in patients with hematological malignancies and solid tumors. In all clinical trials, vaccine-induced immunological responses could be detected. Importantly, objective clinical responses (including stable disease) were observed in 46% and 64% of evaluable vaccinated patients with solid tumors and hematological malignancies, respectively. Immunogenicity of WT1-based cancer vaccines was demonstrated by the detection of a specific immunological response in 35% and 68% of evaluable patients with solid tumors and hematological malignancies, respectively. In order to become part of the armamentarium of the modern oncologist, it will be important to design WT1-based immunotherapies applicable to a large patient population, to standardize vaccination protocols enabling systematic review, and to further optimize the immunostimulatory capacity of the vaccine components. Moreover, improved immunomonitoring tools that reveal clinically relevant T-cell responses will further shape the ideal WT1 immunotherapy strategy. In conclusion, the clinical results obtained so far in WT1-targeted cancer vaccine trials reveal an untapped potential for inducing cancer immunity with minimal side effects and hold promise for a new adjuvant treatment against residual disease and against cancer relapse.
Introduction
Wilms' tumor protein 1 (WT1) is a promising tumor antigen for the development of a novel class of universal cancer vaccines. In this review, we focus solely on cancer vaccines targeting WT1 as an antigenic target for active specific immunotherapy. In early publications, WT1 was described as a tumor suppressor gene [1–4], but afterward it became clear that it also can act as an oncogene [5, 6]. WT1 is a transcription factor and is involved in cell proliferation, differentiation, as well as apoptosis and organ development [7–10]. Several features of this gene make it a promising target for immunotherapy. First, it is highly expressed in several types of hematological malignancies as well as in solid tumors (Table 1) [11–50]. Growth inhibition could be shown in leukemic and solid tumor cells using treatment with WT1 antisense oligonucleotides [11, 51–53]. WT1 has a negative influence on differentiation, but promotes proliferation of progenitor cells [53, 54]. Osaka et al. [6] showed that cells with high levels of WT1 had a stronger tendency to develop into leukemias. WT1 protein is an immunogenic target and exhibits high T-cell antigenicity, as shown by several groups [55–59]. WT1-specific T cells as well as IgG anti-WT1 antibodies have been demonstrated in cancer patients [12, 60–67]. Loss of WT1 expression leads to cessation of proliferation or death of the cancer cells. Therefore, the risk for tumor immune escape resulting from emergence of antigen loss variants is believed to be very small [11, 13, 51]. WT1 is also expressed in a small number of normal tissues, like gonads (testis, ovary), kidney, spleen, and bone marrow [14, 68–70]. Despite expression in normal tissues, there are no reports so far indicating autoimmune reactions in mice or humans after WT1-targeted immunotherapy [55, 57–59, 71, 72]. Because WT1 is a self antigen, it is believed that high-affinity T cells against this antigen are deleted from the repertoire by clonal deletion in the thymus (reviewed by Wiegers et al. [73]). Several mechanisms have been proposed to explain the finding that WT1-specific cytotoxic T lymphocytes (CTLs) could kill leukemic cells, but not WT1-expressing normal cells. A first explanation is that the level of WT1 expression in normal cells is too low to be recognized by low-affinity WT1-specific T cells. Hence, only WT1-overexpressing tumor cells could be the target of those low-affinity T cells. However, this hypothesis has been challenged by the data of Hosen et al. [70], who showed that, at the single-cell level, WT1 levels are similar between normal CD34+ progenitors and their leukemic counterparts. Other possible mechanisms include a lower major histocompatibility complex (MHC) class I expression level in normal cells than in tumor cells; weak, if any, WT1 processing and presentation on MHC class I molecules in normal cells; and absent or weak expression of costimulatory molecules on normal WT1-expressing cells.
Table 1.
Overexpression of WT1 in solid tumors and hematological malignancies
Abbreviations: RT-PCR, reverse transcription polymerase chain reaction; WT1, Wilms' tumor 1.
WT1-Targeted Cancer Vaccine Trials: What Have We Learned?
In the last decade, several WT1-based vaccines have been tested in early-phase clinical trials. Recently, a working party from the National Cancer Institute (NCI) performed a large-scale prioritization study of cancer antigens and produced a priority list of 75 promising tumor-associated antigens based on (a) therapeutic function, (b) immunogenicity, (c) role in oncogenicity, (d) specificity, (e) expression levels and percentage of antigen-positive cells, (f) expression in stem cells, (g) number of patients with antigen-positive cancers, (h) number of antigenic epitopes, and (i) cellular localization of antigen expression [74]. Interestingly, WT1 headed this list, supporting translational research to further design WT1-based cancer vaccines. Here, we review and discuss a list of 21 published clinical trials that used WT1 as immunogenic target. Each trial has unique features in terms of tumor type, patient inclusion criteria, nature of the WT1-derived immunogen, type of adjuvant, and route and frequency of administration. Nevertheless we have tried to carry out a systematic review of these trials in order to address a set of parameters impacting clinical outcome and immunogenicity. In addition, several important differences among the trials that may have had an impact on both the immune responses and the clinical effects are discussed.
Tumor Type and Adverse Events
Because WT1 expression has been documented in a wide variety of solid and hematological tumors (Table 1), many cancer types have been the subject of early-phase clinical trials using WT1-based vaccines (Table 2) [75–95]. In summary, seven trials included patients with different types of solid tumor, two trials reported on patients with either solid or hematological malignancies, and 12 trials focused on patients with hematological cancers.
Table 2.
Overview of clinical trials investigating active specific immunotherapy targeting WT1 as a principal tumor antigen
Table 2a.
(Continued)
Abbreviations: aa, amino acids; ALL, acute lymphoblastic leukemia; AML, acute myeloid leukemia; BM, bone marrow; CA125, cancer antigen 125; CMML, chronic myelomonocytic leukemia; CR, complete remission; CTL, cytotoxic T lymphocyte; DC, dendritic cell; DTH, delayed type hypersensitivity test; HLA, human leukocyte antigen; IAP, immunosuppressive acidic protein; IC, intracellular; ID, intradermal; IFN, interferon; IL, interleukin; KLH, keyhole limpet hemocyanin; M2Y, peptide in which Y was substituted for M at position 2; MDS, myelodysplastic syndrome; MDS-RAEB, MDS–refractory anemia with excess blasts; MLPC, mixed lymphocyte peptide culture; MM, multiple myeloma; ND, not done; NK, natural killer; NSCLC, non-small cell lung cancer; PADRE, pan HLA-DR T helper cell epitope; PB, peripheral blood; PD, progressive disease; PR, partial response; PR1, peptide of proteinase 3; SD, stable disease; TNF, tumor necrosis factor; Treg, regulatory T cell; WT1, Wilms' tumor 1.
No severe adverse events were reported, except for two patients with myelodysplastic syndrome (MDS) who showed severe leukopenia [75]. The explanation for this observation was that MDS originates from CD34+ hematopoietic stem cells and that leukopenia is induced following WT1 vaccination targeting myelodysplastic CD34+ cells. In one patient with chronic myelomonocytic leukemia, the dose of WT1 peptide was decreased and no leukopenia was observed [76]. Common adverse events are local inflammation reactions at the sites of injection. In one case, there was transient thrombocytopenia that spontaneously resolved after the vaccination cycle [77]. Thus, in general, vaccinations are well tolerated and safe. There were no reports of autoimmune reactions resulting from the expression of WT1 in some normal nonhematopoietic tissues.
Antigen Source and Human Leukocyte Antigen Restriction
Based on the WT1 antigen source, the clinical trials summarized here can be divided into four groups: (a) human leukocyte antigen (HLA)-restricted peptide vaccines, (b) non-HLA-restricted long peptides vaccines, (c) dendritic cell (DC) vaccines loaded with HLA-restricted peptide, and (d) DC vaccines loaded with mRNA encoding full-length WT1.
The majority of trials have used HLA-restricted WT1 peptides. Given the high prevalence of HLA-A*2402 and HLA-A*0201 in the Japanese and Caucasian populations, respectively, peptide vaccines restricted to those HLA haplotypes have been the subject of intensive investigation. One trial [75] focused on naturally occurring as well as modified HLA-A*2402-restricted WT1235–243 peptides, nine trials [75, 76, 78–80, 83, 86, 87, 89, 91] focused on the modified heteroclitic HLA-A*2402-restricted WT1235–243 peptide, one trial [84] focused on the natural HLA-A*2402-restricted WT1235–243 peptide, and another trial [85] focused on the natural HLA-A*0201-restricted WT1126–134 peptide. Three trials [81, 93, 94] used a combination of two HLA-A*0201-restricted peptides, a proteinase 3-derived peptide, PR1169–177, and a WT1126–134 peptide. Two reports [88, 90] described vaccination of cancer patients with four different WT1-derived peptides, of which one was a modified WT1126–134 peptide whereas the others were long WT1 peptides (19 or 22 amino acids) that had the purpose of broadening immunogenicity over several HLA types. Finally, four papers described the use of DCs as antigen-presenting cells to enhance the presentation of WT1 by loading DCs with a modified HLA-A*2402-restricted WT1235–243 peptide [82, 95] or by electroporating DCs with mRNA encoding the entire WT1 gene [77, 92].
Although the use of peptides is straightforward and cost-effective, it has several disadvantages. A major drawback is MHC restriction and thus dependency on the patient's HLA haplotype. By selecting one type of MHC class I molecule (e.g., HLA-A*0201 or HLA-A*2402), the eligible patient population is diminished. In addition, immunodominant peptides for only a limited number of MHC molecules are currently known. Short peptide vaccines are designed to predominantly boost the patient's CD8+ cytotoxic peptide-specific T cells, but cannot directly activate cognate CD4+ helper T cells, which are believed to be needed to provide help to cytotoxic T cells and to sustain immunity by inducing memory T-cell responses.
Several options are available to enhance and broaden the immunostimulatory capacity of peptide vaccines. For example, the group of Scheinberg [88, 90] addressed this issue by using three longer WT1 peptides (19–22 amino acids) harboring MHC class II epitopes combined with a modified short WT1126–134 peptide in which arginine (R) at position 1 was replaced by tyrosine (Y) residue. To enhance the binding affinity for an MHC class I molecule or for subsequent better recognition by the T-cell receptor, synthetically modified peptides, so-called heteroclitic peptides, can be used. These are variants of the naturally occurring peptides with the same MHC class I specificity but with enhanced binding and T-cell stimulatory capacities. As described above, 14 of 21 WT1 clinical trials used at least one modified peptide to stimulate the immune system of cancer patients. Whether or not this observation can be extrapolated to other heteroclitic peptide trials remains to be established, especially in view of the data outside the WT1 vaccination field by Lesterhuis et al. [96], who showed no superiority of the gp100 heteroclitic peptide over the natural peptide in colorectal cancer patients. The inclusion of multiple WT1 epitopes can be obtained by the use of whole WT1 protein, but this strategy is rather cumbersome because of the difficulty of manufacturing WT1 protein of a good manufacturing practice grade level. Moreover, protein vaccination is known to result in humoral rather than cellular immune responses, and loading of DCs with proteins would lead predominantly to a CD4+ T-helper response through MHC class II presentation in the absence of CD8+ cytotoxic T-cell induction. This method was not addressed in any of the trials summarized here. Other strategies to circumvent HLA restriction were exploited by our group [77] and a collaborating group [92] using DCs transfected with full-length WT1 mRNA either modified or not with an MHC class II–skewing sequence derived from dendritic cell lysosome-associated membrane glycoprotein. The latter strategy is designed to include all epitopes of the WT1 antigen that are tailored by the DCs to the patient's own MHC molecules, independently of pre-existing knowledge about the patient's HLA type. The use of mRNA has many other advantages, such as its clinical safety profile (i.e., no risk for integration into the host genome) and the possibility of transfecting DCs simultaneously with immune-stimulating molecules [97]. Our group already demonstrated that transfecting DCs using mRNA electroporation is a clinically safe, reproducible, and very efficient procedure resulting in a transient expression, processing, and presentation of the electroporated antigen [98–100].
Adjuvants
In general, vaccines, whether prophylactic or therapeutic, are administered together with an adjuvant to boost the innate immune system at the site of injection. Different adjuvants, cell based or not, are currently used in therapeutic WT1-based cancer vaccines to enhance the effect, the potency, and the longevity of the vaccination. Montanide ISA-51, also called incomplete Freund's adjuvant, is widely recognized as an effective and safe adjuvant for vaccination. In some cases, recombinant GM-CSF was added with the aim of obtaining a more profound effect and stronger immune stimulation [81, 85, 88, 90, 93]. In those trials, three patients were reported with allergy [88], hypersensitivity to GM-CSF [88], and transient chest pain [93], probably because of the addition of GM-CSF. Another adjuvant often used in cancer vaccine trials is keyhole limpet hemocyanin (KLH), a xenogenic and highly immunogenic protein. Similarly here, combinations with GM-CSF were used in a WT1 [85] and a tyrosinase peptide [101] vaccine trial. KLH is commonly used as a noncognate CD4+ T-helper antigen in peptide or DC vaccines. One group [92] applied an imiquimod cream (Aldara®; Meda AB, Solna, Sweden) at the site of injection to boost the immune system. Imiquimod is a Toll-like receptor 7 ligand and has been shown to promote the survival and maturation of DCs as well as enhance priming of T cells [102, 103]. Kuball et al. [94] used a combination of pan HLA-DR T-helper cell epitope, low-dose CpG7909, and Montanide ISA-51. Finally, zoledronate was used as an adjuvant by Kitawaki et al. [95] to promote the activation of tumor antigen–specific T cells by activation of interferon (IFN)-γ–producing Vγ9Vδ2 T cells and stimulation of interacting DCs.
Route of Administration
The two most frequent administration routes in active WT1 immunotherapy trials are intradermal (12 trials) and s.c. (seven trials). One trial combined both routes [85] and one group vaccinated one part of the DC vaccine intradermally and another part i.v. [95]. Until now, there has been no consensus on what is the most optimal way to deliver a peptide- or DC-based cancer vaccine. In general, possible cancer vaccine injection routes include intradermal, s.c., i.v., intralymphatic, and intranodal injections. i.v. administration is not commonly used because the cells end up in the lungs, spleen, and liver and can only migrate secondarily to the lymph nodes. Furthermore, it was shown that i.v. delivery of antigen resulted in a humoral rather than a cellular response [104]. A major disadvantage of the intranodal route is the complexity and the difficult-to-standardize procedure of the injection in the lymph nodes.
Number and Frequency of Vaccinations
The frequency of vaccine administration ranged from weekly to monthly. The first clinical trial using WT1 peptide was reported with biweekly vaccinations [75]. Morita et al. [78] investigated the safety of weekly vaccinations regarding grade 3 or 4 toxicities. No severe toxicities were seen. In summary, patients were vaccinated on a weekly basis in eight trials [78–80, 83, 86, 87, 91, 92], whereas 10 other trials reported a biweekly scheme [75–77, 82, 84, 85, 89]. Two studies had an interval of 4 weeks between the first and second injections, followed by biweekly injections thereafter [88, 90]. In a first report, Rezvani et al. [81] injected patients once with peptides. In a subsequent trial [93], they vaccinated patients every 2 weeks for six vaccines with one booster vaccine 12 weeks thereafter. Briefly, the number of vaccines ranged from one to >64. After a first standard vaccination scheme according to the protocol (different in all studies), in many cases additional booster vaccinations were given with the patient's consent. Because there is no clear evidence for a standard booster scheme, every group independently decided on the interval between additional booster vaccinations. As evident from the above, there is no standardized treatment scheme in terms of frequency and number of vaccinations, resulting in a wide variety of vaccine intervals in early-phase trials.
Outcome
As expected, the clinical outcomes in the reported trials were diverse. Overall, an objective clinical response was defined as a reduction in the tumor mass (e.g., breast cancer, lung cancer), a decrease in the number of malignant cells (e.g., leukemic cells, myeloma cells), a decrease in a tumor marker, and stable disease for a prolonged period of time [105]. For acute myeloid leukemia (AML) patients in remission, the number of WT1 RNA transcripts in blood or bone marrow can be monitored and used as a minimal residual disease biomarker to assess the effect of vaccination and to predict incipient relapse [106]. In view of this, normalization of WT1 RNA to threshold levels after vaccination points to the induction of molecular remission [77].
Clinical responses were seen in all trials except for one, in which only a single AML patient was included [82]. In total, 158 patients were included in 21 clinical trials, of whom 75 had solid tumors and 83 had hematological malignancies (Table 3). Clinical responses postvaccination were observed in 78 of 146 evaluable patients, resulting in overall response rates of 45.8% for patients with solid tumors and 63.5% for patients with hematological tumors. In the solid tumor clinical trials, two patients obtained a complete remission, three had a partial remission, and five showed a decrease in tumor marker or tumor size. In patients with hematological malignancies, complete remission was reached in 13 patients and a decrease in tumor marker or tumor size was reached in 16 patients. Importantly, additional responses were seen if patients with stable disease were also included as clinical responders according to the new criteria for the evaluation of cancer immunotherapy recently put forward by Hoos et al. [105]. Twenty-three patients with solid tumors and 18 patients with hematological malignancies showed stable disease after tumor vaccination.
Table 3.
Overview of clinical and immunological responders in clinical trials investigating active specific immunotherapy targeting Wilms tumor 1 as a principal tumor antigen
Most trials also documented immunological responses after WT1 vaccination. T-cell responses were demonstrated using WT1–MHC class I tetramer staining, the presence of IFN-γ–producing or tumor necrosis factor α–producing T cells, and the presence of other activation molecules (CD107a, CXCR4). A few reports described the cytotoxicity of WT1-specific T cells in some patients [88–90]. When using one or more defined peptides, it is relatively straightforward to monitor peptide-specific T-cell responses. However, when using DCs loaded with full-length mRNA, it becomes more complex to delineate all the T-cell responses elicited by the vaccine because of the presentation of different epitopes presented by different HLA molecules in an MHC class I– and class II–restricted fashion. Detection of tetramer-positive T cells is also limited to known peptides in combination with the restricting HLA subtype. Thus, in fact, there are still major limitations to standardized immunomonitoring regarding the evaluation of immune responses generated by non-HLA–restricted vaccines. The scarcity of circulating tumor-specific T cells in the blood prompted several groups to investigate other techniques to detect vaccine-induced immune responses. Delayed-type hypersensitivity (DTH) skin reaction tests as a measure of in vivo cellular immune response were performed by several groups. In most cases, positive skin reactions were observed and linked to postvaccination immune activation [77, 79, 82, 88, 90, 95]. It would be interesting to further analyze these DTH sites for the presence of DTH-infiltrating lymphocytes and determine their antigen specificity and reactivity [107].
Whereas two clinical trials did not provide immunological monitoring data [78, 87], immune responses were observed in 17 of 19 trials. One trial [83], in glioblastoma multiforme patients, showed no increase in CTL frequencies in any of the 21 vaccinated patients, whereas another trial [94], in AML patients, showed no increase in WT1 tetramer-positive cells and could not demonstrate any reaction in DTH tests. For the 17 other trials, immunological responses could be demonstrated in 69 of 125 evaluable patients (17 patients with solid tumors and 52 hematological patients). Overall immunological response rates of 35.4% for patients with solid tumors and 67.5% for patients with hematological malignancies were obtained throughout the 19 clinical trials reporting on WT1-specific immunomonitoring (Table 3).
Conclusions and Future Perspectives
Altogether, most studies showed some benefit (immunological, clinical, or both) for a sizable number of patients. Surrogate immunological markers for clinical benefit were only sporadically reported [75, 77] and remain to be confirmed in larger randomized controlled trials. Therefore, at present, it is difficult to predict which patients will benefit most from active immunotherapy and how one should discriminate responders from nonresponders before enrolling patients into a vaccine protocol. Recently Busse et al. [108] showed that immune escape in AML patients was not a result of mutation of WT1, loss of WT1 expression, or decreased expression of MHC class I molecules on tumor cells. Although the mechanisms behind tumor immune escape after immunotherapy are largely unknown, several hypotheses have been put forward. The short-term beneficial effect often observed with WT1 peptide vaccines might be explained by the induction of T-cell tolerance caused by short peptides [109]. The presence of regulatory T-cell populations or other immune inhibitory pathways could play a role in the outcome of treatment with therapeutic cancer vaccines. Also, the combination, type, and dose of adjuvants used in immunotherapy trials must be taken into account. Montanide ISA-51 was reported by Rezvani et al. [93] to have a negative effect on the long-term immune response. Moreover, the findings of Kuball et al. [94] demonstrated that certain combinations and doses of adjuvants could have detrimental effects on the activation of tumor-specific T cells and could induce negative effects such as T-cell deletion and anergy.
In conclusion, WT1-based cancer vaccines have been shown to be feasible and safe in patients with multiple tumor types. Furthermore, these vaccines elicited WT1-specific immune responses and showed promising clinical results in a majority of patients. These conclusions run parallel with a recent NCI report [74] on the prioritization of cancer antigens and justify the further development of WT1-targeted immunotherapies. Design of larger phase II trials as well as two-arm trials will establish optimal vaccination strategies and will eventually reveal the true potential of WT1 as a universal cancer vaccine target in the adjuvant setting.
Acknowledgments
This work was supported in part by research grants from the FWO-Vlaanderen, the Stichting tegen Kanker, the Vlaamse Liga tegen Kanker, the IWT-TBM program of the IWT Vlaanderen, the National Cancer Plan of Belgium, and the Methusalem financing program of the Flemish Government to the University of Antwerp. No additional external funding was received for this study. The funders had no role in study design, data collection and analysis, decision to publish, preparation, or approval of the manuscript.
Author Contributions
Conception/Design: Ann Van Driessche, Viggo F.I. Van Tendeloo, Zwi N. Berneman
Collection and/or assembly of data: Ann Van Driessche
Data analysis and interpretation: Ann Van Driessche, Viggo F.I. Van Tendeloo, Zwi N. Berneman
Manuscript writing: Ann Van Driessche, Viggo F.I. Van Tendeloo, Zwi N. Berneman
Final approval of manuscript: Ann Van Driessche, Viggo F.I. Van Tendeloo, Zwi N. Berneman
References
- 1.Call KM, Glaser T, Ito CY, et al. Isolation and characterization of a zinc finger polypeptide gene at the human chromosome 11 Wilms' tumor locus. Cell. 1990;60:509–520. doi: 10.1016/0092-8674(90)90601-a. [DOI] [PubMed] [Google Scholar]
- 2.Haber DA, Buckler AJ, Glaser T, et al. An internal deletion within an 11p13 zinc finger gene contributes to the development of Wilms' tumor. Cell. 1990;61:1257–1269. doi: 10.1016/0092-8674(90)90690-g. [DOI] [PubMed] [Google Scholar]
- 3.Gessler M, Poustka A, Cavenee W, et al. Homozygous deletion in Wilms tumours of a zinc-finger gene identified by chromosome jumping. Nature. 1990;343:774–778. doi: 10.1038/343774a0. [DOI] [PubMed] [Google Scholar]
- 4.Gessler M, König A, Bruns GA. The genomic organization and expression of the WT1 gene. Genomics. 1992;12:807–813. doi: 10.1016/0888-7543(92)90313-h. [DOI] [PubMed] [Google Scholar]
- 5.Menke AL, van der Eb AJ, Jochemsen AG. The Wilms' tumor 1 gene: Oncogene or tumor suppressor gene? Int Rev Cytol. 1998;181:151–212. doi: 10.1016/s0074-7696(08)60418-0. [DOI] [PubMed] [Google Scholar]
- 6.Osaka M, Koami K, Sugiyama T. WT1 contributes to leukemogenesis: Expression patterns in 7,12-dimethylbenz[a]anthracene (DMBA)-induced leukemia. Int J Cancer. 1997;72:696–699. doi: 10.1002/(sici)1097-0215(19970807)72:4<696::aid-ijc23>3.0.co;2-8. [DOI] [PubMed] [Google Scholar]
- 7.Drummond IA, Madden SL, Rohwer-Nutter P, et al. Repression of the insulin-like growth factor-II gene by the Wilms tumor suppressor WT1. Science. 1992;257:674–678. doi: 10.1126/science.1323141. [DOI] [PubMed] [Google Scholar]
- 8.Englert C, Hou X, Maheswaran S, et al. WT1 suppresses synthesis of the epidermal growth factor receptor and induces apoptosis. EMBO J. 1995;14:4662–4675. doi: 10.1002/j.1460-2075.1995.tb00148.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 9.Hewitt SM, Hamada S, McDonnell TJ, et al. Regulation of the proto-oncogenes bcl-2 and c-myc by the Wilms' tumor suppressor gene WT1. Cancer Res. 1995;55:5386–5389. [PubMed] [Google Scholar]
- 10.Goodyer P, Dehbi M, Torban E, et al. Repression of the retinoic acid receptor-alpha gene by the Wilms' tumor suppressor gene product, WT1. Oncogene. 1995;10:1125–1129. [PubMed] [Google Scholar]
- 11.Oji Y, Ogawa H, Tamaki H, et al. Expression of the Wilms' tumor gene WT1 in solid tumors and its involvement in tumor cell growth. Jpn J Cancer Res. 1999;90:194–204. doi: 10.1111/j.1349-7006.1999.tb00733.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 12.Gillmore R, Xue SA, Holler A, et al. Detection of Wilms' tumor antigen–specific CTL in tumor-draining lymph nodes of patients with early breast cancer. Clin Cancer Res. 2006;12:34–42. doi: 10.1158/1078-0432.CCR-05-1483. [DOI] [PubMed] [Google Scholar]
- 13.Oji Y, Miyoshi S, Maeda H, et al. Overexpression of the Wilms' tumor gene WT1 in de novo lung cancers. Int J Cancer. 2002;100:297–303. doi: 10.1002/ijc.10476. [DOI] [PubMed] [Google Scholar]
- 14.Inoue K, Ogawa H, Sonoda Y, et al. Aberrant overexpression of the Wilms tumor gene (WT1) in human leukemia. Blood. 1997;89:1405–1412. [PubMed] [Google Scholar]
- 15.Nakatsuka S, Oji Y, Horiuchi T, et al. Immunohistochemical detection of WT1 protein in a variety of cancer cells. Mod Pathol. 2006;19:804–814. doi: 10.1038/modpathol.3800588. [DOI] [PubMed] [Google Scholar]
- 16.Ueda T, Oji Y, Naka N, et al. Overexpression of the Wilms' tumor gene WT1 in human bone and soft-tissue sarcomas. Cancer Sci. 2003;94:271–276. doi: 10.1111/j.1349-7006.2003.tb01432.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 17.Silberstein GB, Van Horn K, Strickland P, et al. Altered expression of the WT1 Wilms tumor suppressor gene in human breast cancer. Proc Natl Acad Sci U S A. 1997;94:8132–8137. doi: 10.1073/pnas.94.15.8132. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 18.Loeb DM, Evron E, Patel CB, et al. Wilms' tumor suppressor gene (WT1) is expressed in primary breast tumors despite tumor-specific promoter methylation. Cancer Res. 2001;61:921–925. [PubMed] [Google Scholar]
- 19.Koesters R, Linnebacher M, Coy JF, et al. WT1 is a tumor-associated antigen in colon cancer that can be recognized by in vitro stimulated cytotoxic T cells. Int J Cancer. 2004;109:385–392. doi: 10.1002/ijc.11721. [DOI] [PubMed] [Google Scholar]
- 20.Oji Y, Yamamoto H, Nomura M, et al. Overexpression of the Wilms' tumor gene WT1 in colorectal adenocarcinoma. Cancer Sci. 2003;94:712–717. doi: 10.1111/j.1349-7006.2003.tb01507.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 21.Amini Nik S, Hohenstein P, Jadidizadeh A, et al. Upregulation of Wilms' tumor gene 1 (WT1) in desmoid tumors. Int J Cancer. 2005;114:202–208. doi: 10.1002/ijc.20717. [DOI] [PubMed] [Google Scholar]
- 22.Oji Y, Yano M, Nakano Y, et al. Overexpression of the Wilms' tumor gene WT1 in esophageal cancer. Anticancer Res. 2004;24:3103–3108. [PubMed] [Google Scholar]
- 23.Clark AJ, Dos Santos WG, McCready J, et al. Wilms tumor 1 expression in malignant gliomas and correlation of +KTS isoforms with p53 status. J Neurosurg. 2007;107:586–592. doi: 10.3171/JNS-07/09/0586. [DOI] [PubMed] [Google Scholar]
- 24.Ohno S, Dohi S, Ohno Y, et al. Immunohistochemical detection of WT1 protein in endometrial cancer. Anticancer Res. 2009;29:1691–1695. [PubMed] [Google Scholar]
- 25.Oji Y, Inohara H, Nakazawa M, et al. Overexpression of the Wilms' tumor gene WT1 in head and neck squamous cell carcinoma. Cancer Sci. 2003;94:523–529. doi: 10.1111/j.1349-7006.2003.tb01477.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 26.Menssen HD, Bertelmann E, Bartelt S, et al. Wilms' tumor gene (WT1) expression in lung cancer, colon cancer and glioblastoma cell lines compared to freshly isolated tumor specimens. J Cancer Res Clin Oncol. 2000;126:226–232. doi: 10.1007/s004320050037. [DOI] [PubMed] [Google Scholar]
- 27.Oji Y, Nakamori S, Fujikawa M, et al. Overexpression of the Wilms' tumor gene WT1 in pancreatic ductal adenocarcinoma. Cancer Sci. 2004;95:583–587. doi: 10.1111/j.1349-7006.2004.tb02490.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 28.Oji Y, Suzuki T, Nakano Y, et al. Overexpression of the Wilms' tumor gene WT1 in primary astrocytic tumors. Cancer Sci. 2004;95:822–827. doi: 10.1111/j.1349-7006.2004.tb02188.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 29.Oji Y, Miyoshi Y, Koga S, et al. Overexpression of the Wilms' tumor gene WT1 in primary thyroid cancer. Cancer Sci. 2003;94:606–611. doi: 10.1111/j.1349-7006.2003.tb01490.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 30.Devilard E, Bladou F, Ramuz O, et al. FGFR1 and WT1 are markers of human prostate cancer progression. BMC Cancer. 2006;6:272. doi: 10.1186/1471-2407-6-272. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 31.King JW, Thomas S, Corsi F, et al. IL15 can reverse the unresponsiveness of Wilms' tumor antigen-specific CTL in patients with prostate cancer. Clin Cancer Res. 2009;15:1145–1154. doi: 10.1158/1078-0432.CCR-08-1821. [DOI] [PubMed] [Google Scholar]
- 32.Campbell CE, Kuriyan NP, Rackley RR, et al. Constitutive expression of the Wilms tumor suppressor gene (WT1) in renal cell carcinoma. Int J Cancer. 1998;78:182–188. doi: 10.1002/(sici)1097-0215(19981005)78:2<182::aid-ijc11>3.0.co;2-d. [DOI] [PubMed] [Google Scholar]
- 33.Carpentieri DF, Nichols K, Chou PM, et al. The expression of WT1 in the differentiation of rhabdomyosarcoma from other pediatric small round blue cell tumors. Mod Pathol. 2002;15:1080–1086. doi: 10.1097/01.MP.0000028646.03760.6B. [DOI] [PubMed] [Google Scholar]
- 34.Harada Y, Nonomura N, Nishimura K, et al. WT1 gene expression in human testicular germ-cell tumors. Mol Urol. 1999;3:357–364. [PubMed] [Google Scholar]
- 35.Coosemans A, Amini-Nik S, Caluwaerts S, et al. Upregulation of Wilms' tumour gene 1 (WT1) in uterine sarcomas. Eur J Cancer. 2007;43:1630–1637. doi: 10.1016/j.ejca.2007.04.008. [DOI] [PubMed] [Google Scholar]
- 36.Menssen HD, Renkl HJ, Rodeck U, et al. Presence of Wilms-tumor gene (WT1) transcripts and the WT1 nuclear-protein in the majority of human acute leukemias. Leukemia. 1995;9:1060–1067. [PubMed] [Google Scholar]
- 37.Menssen HD, Renkl HJ, Rodeck U, et al. Detection by monoclonal antibodies of the Wilms' tumor (WT1) nuclear protein in patients with acute leukemia. Int J Cancer. 1997;70:518–523. doi: 10.1002/(sici)1097-0215(19970304)70:5<518::aid-ijc5>3.0.co;2-0. [DOI] [PubMed] [Google Scholar]
- 38.Patmasiriwat P, Fraizer G, Kantarjian H, et al. WT1 and GATA1 expression in myelodysplastic syndrome and acute leukemia. Leukemia. 1999;13:891–900. doi: 10.1038/sj.leu.2401414. [DOI] [PubMed] [Google Scholar]
- 39.Niegemann E, Wehner S, Kornhuber B, et al. WT1 gene expression in childhood leukemias. Acta Haematol. 1999;102:72–76. doi: 10.1159/000040973. [DOI] [PubMed] [Google Scholar]
- 40.Ozgen U, Anak S, Ozbek U, et al. WT1 gene expression in childhood acute leukemias. Acta Haematol. 2000;103:229–230. doi: 10.1159/000041057. [DOI] [PubMed] [Google Scholar]
- 41.Gaiger A, Linnerth B, Mann G, et al. Wilms' tumour gene (WT1) expression at diagnosis has no prognostic relevance in childhood acute lymphoblastic leukaemia treated by an intensive chemotherapy protocol. Eur J Haematol. 1999;63:86–93. doi: 10.1111/j.1600-0609.1999.tb01121.x. [DOI] [PubMed] [Google Scholar]
- 42.Bergmann L, Miething C, Maurer U, et al. High levels of Wilms' tumor gene (WT1) mRNA in acute myeloid leukemias are associated with a worse long-term outcome. Blood. 1997;90:1217–1225. [PubMed] [Google Scholar]
- 43.Bergmann L, Maurer U, Weidmann E. Wilms tumor gene expression in acute myeloid leukemias. Leuk Lymphoma. 1997;25:435–443. doi: 10.3109/10428199709039030. [DOI] [PubMed] [Google Scholar]
- 44.Schmid D, Heinze G, Linnerth B, et al. Prognostic significance of WT1 gene expression at diagnosis in adult de novo acute myeloid leukemia. Leukemia. 1997;11:639–643. doi: 10.1038/sj.leu.2400620. [DOI] [PubMed] [Google Scholar]
- 45.Inoue K, Sugiyama H, Ogawa H, et al. WT1 as a new prognostic factor and a new marker for the detection of minimal residual disease in acute leukemia. Blood. 1994;84:3071–3079. [PubMed] [Google Scholar]
- 46.Cilloni D, Gottardi E, De Micheli D, et al. Quantitative assessment of WT1 expression by real time quantitative PCR may be a useful tool for monitoring minimal residual disease in acute leukemia patients. Leukemia. 2002;16:2115–2121. doi: 10.1038/sj.leu.2402675. [DOI] [PubMed] [Google Scholar]
- 47.Tamaki H, Ogawa H, Ohyashiki K, et al. The Wilms' tumor gene WT1 is a good marker for diagnosis of disease progression of myelodysplastic syndromes. Leukemia. 1999;13:393–399. doi: 10.1038/sj.leu.2401341. [DOI] [PubMed] [Google Scholar]
- 48.Azuma T, Otsuki T, Kuzushima K, et al. Myeloma cells are highly sensitive to the granule exocytosis pathway mediated by WT1-specific cytotoxic T lymphocytes. Clin Cancer Res. 2004;10:7402–7412. doi: 10.1158/1078-0432.CCR-04-0825. [DOI] [PubMed] [Google Scholar]
- 49.Hatta Y, Takeuchi J, Saitoh T, et al. WT1 expression level and clinical factors in multiple myeloma. J Exp Clin Cancer Res. 2005;24:595–599. [PubMed] [Google Scholar]
- 50.Cilloni D, Messa F, Martinelli G, et al. WT1 transcript amount discriminates secondary or reactive eosinophilia from idiopathic hypereosinophilic syndrome or chronic eosinophilic leukemia. Leukemia. 2007;21:1442–1450. doi: 10.1038/sj.leu.2404670. [DOI] [PubMed] [Google Scholar]
- 51.Yamagami T, Sugiyama H, Inoue K, et al. Growth inhibition of human leukemic cells by WT1 (Wilms tumor gene) antisense oligodeoxynucleotides: Implications for the involvement of WT1 in leukemogenesis. Blood. 1996;87:2878–2884. [PubMed] [Google Scholar]
- 52.Algar EM, Khromykh T, Smith SI, et al. A WT1 antisense oligonucleotide inhibits proliferation and induces apoptosis in myeloid leukaemia cell lines. Oncogene. 1996;12:1005–1014. [PubMed] [Google Scholar]
- 53.Inoue K, Tamaki H, Ogawa H, et al. Wilms' tumor gene (WT1) competes with differentiation-inducing signal in hematopoietic progenitor cells. Blood. 1998;91:2969–2976. [PubMed] [Google Scholar]
- 54.Tsuboi A, Oka Y, Ogawa H, et al. Constitutive expression of the Wilms' tumor gene WT1 inhibits the differentiation of myeloid progenitor cells but promotes their proliferation in response to granulocyte-colony stimulating factor (G-CSF) Leuk Res. 1999;23:499–505. doi: 10.1016/s0145-2126(99)00037-5. [DOI] [PubMed] [Google Scholar]
- 55.Gao L, Bellantuono I, Elsässer A, et al. Selective elimination of leukemic CD34(+) progenitor cells by cytotoxic T lymphocytes specific for WT1. Blood. 2000;95:2198–2203. [PubMed] [Google Scholar]
- 56.Makita M, Hiraki A, Azuma T, et al. Antilung cancer effect of WT1-specific cytotoxic T lymphocytes. Clin Cancer Res. 2002;8:2626–2631. [PubMed] [Google Scholar]
- 57.Ohminami H, Yasukawa M, Fujita S. HLA class I-restricted lysis of leukemia cells by a CD8(+) cytotoxic T-lymphocyte clone specific for WT1 peptide. Blood. 2000;95:286–293. [PubMed] [Google Scholar]
- 58.Oka Y, Elisseeva OA, Tsuboi A, et al. Human cytotoxic T-lymphocyte responses specific for peptides of the wild-type Wilms' tumor gene (WT1) product. Immunogenetics. 2000;51:99–107. doi: 10.1007/s002510050018. [DOI] [PubMed] [Google Scholar]
- 59.Tsuboi A, Oka Y, Udaka K, et al. Enhanced induction of human WT1-specific cytotoxic T lymphocytes with a 9-mer WT1 peptide modified at HLA-A*2402-binding residues. Cancer Immunol Immunother. 2002;51:614–620. doi: 10.1007/s00262-002-0328-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 60.Rezvani K, Yong AS, Savani BN, et al. Graft-versus-leukemia effects associated with detectable Wilms tumor-1-specific T lymphocytes after allogeneic stem-cell transplantation for acute lymphoblastic leukemia. Blood. 2007;110:1924–1932. doi: 10.1182/blood-2007-03-076844. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 61.Rezvani K, Brenchley JM, Price DA, et al. T-cell responses directed against multiple HLA-A*0201-restricted epitopes derived from Wilms' tumor 1 protein in patients with leukemia and healthy donors: Identification, quantification, and characterization. Clin Cancer Res. 2005;11:8799–8807. doi: 10.1158/1078-0432.CCR-05-1314. [DOI] [PubMed] [Google Scholar]
- 62.Rezvani K, Grube M, Brenchley JM, et al. Functional leukemia-associated antigen-specific memory CD8(+) T cells exist in healthy individuals and in patients with chronic myelogenous leukemia before and after stem cell transplantation. Blood. 2003;102:2892–2900. doi: 10.1182/blood-2003-01-0150. [DOI] [PubMed] [Google Scholar]
- 63.Scheibenbogen C, Letsch A, Thiel E, et al. CD8 T-cell responses to Wilms tumor gene product WT1 and proteinase 3 in patients with acute myeloid leukemia. Blood. 2002;100:2132–2137. doi: 10.1182/blood-2002-01-0163. [DOI] [PubMed] [Google Scholar]
- 64.Gannagé M, Abel M, Michallet AS, et al. Ex vivo characterization of multiepitopic tumor-specific CD8 T cells in patients with chronic myeloid leukemia: Implications for vaccine development and adoptive cellular immunotherapy. J Immunol. 2005;174:8210–8218. doi: 10.4049/jimmunol.174.12.8210. [DOI] [PubMed] [Google Scholar]
- 65.Elisseeva OA, Oka Y, Tsuboi A, et al. Humoral immune responses against Wilms tumor gene WT1 product in patients with hematopoietic malignancies. Blood. 2002;99:3272–3279. doi: 10.1182/blood.v99.9.3272. [DOI] [PubMed] [Google Scholar]
- 66.Nicoli P, Defilippi I, Carturan S, et al. Detection of humoral immune responses against WT1 antigen in patients affected by different hematological malignancies. Acta Haematol. 2008;120:47–50. doi: 10.1159/000158576. [DOI] [PubMed] [Google Scholar]
- 67.Wu F, Oka Y, Tsuboi A, et al. Th1-biased humoral immune responses against Wilms tumor gene WT1 product in the patients with hematopoietic malignancies. Leukemia. 2005;19:268–274. doi: 10.1038/sj.leu.2403539. [DOI] [PubMed] [Google Scholar]
- 68.Huang A, Campbell CE, Bonetta L, et al. Tissue, developmental, and tumor-specific expression of divergent transcripts in Wilms tumor. Science. 1990;250:991–994. doi: 10.1126/science.2173145. [DOI] [PubMed] [Google Scholar]
- 69.Pelletier J, Schalling M, Buckler AJ, et al. Expression of the Wilms' tumor gene WT1 in the murine urogenital system. Genes Dev. 1991;5:1345–1356. doi: 10.1101/gad.5.8.1345. [DOI] [PubMed] [Google Scholar]
- 70.Hosen N, Sonoda Y, Oji Y, et al. Very low frequencies of human normal CD34+ haematopoietic progenitor cells express the Wilms' tumour gene WT1 at levels similar to those in leukaemia cells. Br J Haematol. 2002;116:409–420. doi: 10.1046/j.1365-2141.2002.03261.x. [DOI] [PubMed] [Google Scholar]
- 71.Oka Y, Udaka K, Tsuboi A, et al. Cancer immunotherapy targeting Wilms' tumor gene WT1 product. J Immunol. 2000;164:1873–1880. doi: 10.4049/jimmunol.164.4.1873. [DOI] [PubMed] [Google Scholar]
- 72.Tsuboi A, Oka Y, Ogawa H, et al. Cytotoxic T-lymphocyte responses elicited to Wilms' tumor gene WT1 product by DNA vaccination. J Clin Immunol. 2000;20:195–202. doi: 10.1023/a:1006637529995. [DOI] [PubMed] [Google Scholar]
- 73.Wiegers GJ, Kaufmann M, Tischner D, et al. Shaping the T-cell repertoire: A matter of life and death. Immunol Cell Biol. 2011;89:33–39. doi: 10.1038/icb.2010.127. [DOI] [PubMed] [Google Scholar]
- 74.Cheever MA, Allison JP, Ferris AS, et al. The prioritization of cancer antigens: A National Cancer Institute pilot project for the acceleration of translational research. Clin Cancer Res. 2009;15:5323–5337. doi: 10.1158/1078-0432.CCR-09-0737. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 75.Oka Y, Tsuboi A, Taguchi T, et al. Induction of WT1 (Wilms' tumor gene)-specific cytotoxic T lymphocytes by WT1 peptide vaccine and the resultant cancer regression. Proc Natl Acad Sci U S A. 2004;101:13885–13890. doi: 10.1073/pnas.0405884101. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 76.Kawakami M, Oka Y, Tsuboi A, et al. Clinical and immunologic responses to very low-dose vaccination with WT1 peptide (5 microg/body) in a patient with chronic myelomonocytic leukemia. Int J Hematol. 2007;85:426–429. doi: 10.1532/IJH97.06194. [DOI] [PubMed] [Google Scholar]
- 77.Van Tendeloo VF, Van de Velde A, Van Driessche A, et al. Induction of complete and molecular remissions in acute myeloid leukemia by Wilms' tumor 1 antigen-targeted dendritic cell vaccination. Proc Natl Acad Sci U S A. 2010;107:13824–13829. doi: 10.1073/pnas.1008051107. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 78.Morita S, Oka Y, Tsuboi A, et al. A phase I/II trial of a WT1 (Wilms' tumor gene) peptide vaccine in patients with solid malignancy: Safety assessment based on the phase I data. Jpn J Clin Oncol. 2006;36:231–236. doi: 10.1093/jjco/hyl005. [DOI] [PubMed] [Google Scholar]
- 79.Iiyama T, Udaka K, Takeda S, et al. WT1 (Wilms' tumor 1) peptide immunotherapy for renal cell carcinoma. Microbiol Immunol. 2007;51:519–530. doi: 10.1111/j.1348-0421.2007.tb03940.x. [DOI] [PubMed] [Google Scholar]
- 80.Tsuboi A, Oka Y, Nakajima H, et al. Wilms tumor gene WT1 peptide-based immunotherapy induced a minimal response in a patient with advanced therapy-resistant multiple myeloma. Int J Hematol. 2007;86:414–417. doi: 10.1007/BF02983998. [DOI] [PubMed] [Google Scholar]
- 81.Rezvani K, Yong AS, Mielke S, et al. Leukemia-associated antigen-specific T-cell responses following combined PR1 and WT1 peptide vaccination in patients with myeloid malignancies. Blood. 2008;111:236–242. doi: 10.1182/blood-2007-08-108241. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 82.Kitawaki T, Kadowaki N, Kondo T, et al. Potential of dendritic cell immunotherapy for relapse after allogeneic hematopoietic stem cell transplantation, shown by WT1 peptide- and keyhole limpet hemocyanin-pulsed, donor-derived dendritic-cell vaccine for acute myeloid leukemia. Am J Hematol. 2008;83:315–317. doi: 10.1002/ajh.21127. [DOI] [PubMed] [Google Scholar]
- 83.Izumoto S, Tsuboi A, Oka Y, et al. Phase II clinical trial of Wilms tumor 1 peptide vaccination for patients with recurrent glioblastoma multiforme. J Neurosurg. 2008;108:963–971. doi: 10.3171/JNS/2008/108/5/0963. [DOI] [PubMed] [Google Scholar]
- 84.Yasukawa M, Fujiwara H, Ochi T, et al. Clinical efficacy of WT1 peptide vaccination in patients with acute myelogenous leukemia and myelodysplastic syndrome. Am J Hematol. 2009;84:314–315. doi: 10.1002/ajh.21387. [DOI] [PubMed] [Google Scholar]
- 85.Keilholz U, Letsch A, Busse A, et al. A clinical and immunologic phase 2 trial of Wilms tumor gene product 1 (WT1) peptide vaccination in patients with AML and MDS. Blood. 2009;113:6541–6548. doi: 10.1182/blood-2009-02-202598. [DOI] [PubMed] [Google Scholar]
- 86.Ohta H, Hashii Y, Yoneda A, et al. WT1 (Wilms tumor 1) peptide immunotherapy for childhood rhabdomyosarcoma: A case report. Pediatr Hematol Oncol. 2009;26:74–83. doi: 10.1080/08880010802435500. [DOI] [PubMed] [Google Scholar]
- 87.Ohno S, Kyo S, Myojo S, et al. Wilms' tumor 1 (WT1) peptide immunotherapy for gynecological malignancy. Anticancer Res. 2009;29:4779–4784. [PubMed] [Google Scholar]
- 88.Maslak PG, Dao T, Krug LM, et al. Vaccination with synthetic analog peptides derived from WT1 oncoprotein induces T-cell responses in patients with complete remission from acute myeloid leukemia. Blood. 2010;116:171–179. doi: 10.1182/blood-2009-10-250993. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 89.Narita M, Masuko M, Kurasaki T, et al. WT1 peptide vaccination in combination with imatinib therapy for a patient with CML in the chronic phase. Int J Med Sci. 2010;7:72–81. doi: 10.7150/ijms.7.72. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 90.Krug LM, Dao T, Brown AB, et al. WT1 peptide vaccinations induce CD4 and CD8 T cell immune responses in patients with mesothelioma and non-small cell lung cancer. Cancer Immunol Immunother. 2010;59:1467–1479. doi: 10.1007/s00262-010-0871-8. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 91.Hashii Y, Sato E, Ohta H, et al. WT1 peptide immunotherapy for cancer in children and young adults. Pediatr Blood Cancer. 2010;55:352–355. doi: 10.1002/pbc.22522. [DOI] [PubMed] [Google Scholar]
- 92.Coosemans A, Wölfl M, Berneman ZN, et al. Immunological response after therapeutic vaccination with WT1 mRNA-loaded dendritic cells in end-stage endometrial carcinoma. Anticancer Res. 2010;30:3709–3714. [PubMed] [Google Scholar]
- 93.Rezvani K, Yong AS, Mielke S, et al. Repeated PR1 and WT1 peptide vaccination in montanide-adjuvant fails to induce sustained high-avidity, epitope-specific CD8+ T cells in myeloid malignancies. Haematologica. 2011;96:432–440. doi: 10.3324/haematol.2010.031674. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 94.Kuball J, de Boer K, Wagner E, et al. Pitfalls of vaccinations with WT1-, proteinase3- and MUC1-derived peptides in combination with montanideISA51 and CpG7909. Cancer Immunol Immunother. 2011;60:161–171. doi: 10.1007/s00262-010-0929-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 95.Kitawaki T, Kadowaki N, Fukunaga K, et al. A phase I/IIa clinical trial of immunotherapy for elderly patients with acute myeloid leukaemia using dendritic cells co-pulsed with WT1 peptide and zoledronate. Br J Haematol. 2011;153:796–799. doi: 10.1111/j.1365-2141.2010.08490.x. [DOI] [PubMed] [Google Scholar]
- 96.Lesterhuis WJ, Schreibelt G, Scharenborg NM, et al. Wild-type and modified gp100 peptide-pulsed dendritic cell vaccination of advanced melanoma patients can lead to long-term clinical responses independent of the peptide used. Cancer Immunol Immunother. 2010;60:249–260. doi: 10.1007/s00262-010-0942-x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 97.Van Tendeloo VF, Ponsaerts P, Berneman ZN. mRNA-based gene transfer as a tool for gene and cell therapy. Curr Opin Mol Ther. 2007;9:423–431. [PubMed] [Google Scholar]
- 98.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. doi: 10.1182/blood.v98.1.49. [DOI] [PubMed] [Google Scholar]
- 99.Van Driessche A, Gao L, Stauss HJ, et al. Antigen-specific cellular immunotherapy of leukemia. Leukemia. 2005;19:1863–1871. doi: 10.1038/sj.leu.2403930. [DOI] [PubMed] [Google Scholar]
- 100.Van Driessche A, Van de Velde AL, Nijs G, et al. 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. doi: 10.1080/14653240902960411. [DOI] [PubMed] [Google Scholar]
- 101.Scheibenbogen C, Schadendorf D, Bechrakis NE, et al. Effects of granulocyte-macrophage colony-stimulating factor and foreign helper protein as immunologic adjuvants on the T-cell response to vaccination with tyrosinase peptides. Int J Cancer. 2003;104:188–194. doi: 10.1002/ijc.10961. [DOI] [PubMed] [Google Scholar]
- 102.Van Gool S, Maes W, Ardon H, et al. Dendritic cell therapy of high-grade gliomas. Brain Pathol. 2009;19:694–712. doi: 10.1111/j.1750-3639.2009.00316.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 103.Prins RM, Craft N, Bruhn KW, et al. The TLR-7 agonist, imiquimod, enhances dendritic cell survival and promotes tumor antigen-specific T cell priming: Relation to central nervous system antitumor immunity. J Immunol. 2006;176:157–164. doi: 10.4049/jimmunol.176.1.157. [DOI] [PubMed] [Google Scholar]
- 104.Dhodapkar MV, Bhardwaj N. Active immunization of humans with dendritic cells. J Clin Immunol. 2000;20:167–174. doi: 10.1023/a:1006681312249. [DOI] [PubMed] [Google Scholar]
- 105.Hoos A, Eggermont AM, Janetzki S, et al. Improved endpoints for cancer immunotherapy trials. J Natl Cancer Inst. 2010;102:1388–1397. doi: 10.1093/jnci/djq310. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 106.Cilloni D, Renneville A, Hermitte F, et al. Real-time quantitative polymerase chain reaction detection of minimal residual disease by standardized WT1 assay to enhance risk stratification in acute myeloid leukemia: A European LeukemiaNet study. J Clin Oncol. 2009;27:5195–5201. doi: 10.1200/JCO.2009.22.4865. [DOI] [PubMed] [Google Scholar]
- 107.de Vries IJ, Bernsen MR, Lesterhuis WJ, et al. Immunomonitoring tumor-specific T cells in delayed-type hypersensitivity skin biopsies after dendritic cell vaccination correlates with clinical outcome. J Clin Oncol. 2005;23:5779–5787. doi: 10.1200/JCO.2005.06.478. [DOI] [PubMed] [Google Scholar]
- 108.Busse A, Letsch A, Scheibenbogen C, et al. Mutation or loss of Wilms' tumor gene 1 (WT1) are not major reasons for immune escape in patients with AML receiving WT1 peptide vaccination. J Transl Med. 2010;8:5. doi: 10.1186/1479-5876-8-5. [DOI] [PMC free article] [PubMed] [Google Scholar]
- 109.Toes RE, Offringa R, Blom RJ, et al. Peptide vaccination can lead to enhanced tumor growth through specific T-cell tolerance induction. Proc Natl Acad Sci U S A. 1996;93:7855–7860. doi: 10.1073/pnas.93.15.7855. [DOI] [PMC free article] [PubMed] [Google Scholar]