Skip to main content
Clinical and Developmental Immunology logoLink to Clinical and Developmental Immunology
. 2011 Jan 3;2010:284581. doi: 10.1155/2010/284581

Immunotherapy for Renal Cell Carcinoma

Momoe Itsumi 1, Katsunori Tatsugami 1,*
PMCID: PMC3022170  PMID: 21253521

Abstract

Immunotherapy plays a significant role in the management of renal cell carcinoma (RCC) patients with metastatic disease because RCC is highly resistant to both chemotherapy and radiation therapy. Many reports illustrate various approaches to the treatment of RCC, such as cytokine-, antigen- or dendritic cell- (DC-) based immunotherapy, and the safety and effectiveness of immunotherapy have been highlighted by multiple clinical trials. Although antitumor immune responses and clinically significant outcomes have been achieved in these trials, the response rate is still low, and very few patients show long-term clinical improvement. Recently, the importance of immune regulation by antigen-presenting cells (APC) and regulatory T cells (Treg cells) has also been discussed. The authors outline the principles of cell-mediated tumor immunotherapy and discuss clinical trials of immunotherapy for RCC.

1. Introduction

Renal cell carcinoma (RCC), a glandular carcinoma, accounts for approximately 85%–95% of adult malignant kidney cancer cases [1]. Patients with advanced or metastatic disease have a poor prognosis, with a 5-year survival rate of less than 15%. Surgical treatment is effective, even in patients with advanced or metastatic RCC, because of its high resistance to chemotherapy and radiation therapy. Immunotherapy using interferon (IFN)-α and/or interleukin (IL)-2 has shown promising anti-tumor activity in RCC [24]. However, these cytokines have a positive effect in only 10%–20% of cases [5]. Like melanoma, RCC is classed as an immunogenic tumor based on its response rate to immunotherapy, the incidence of spontaneous regression, and the high level of tumor T cell infiltration. Despite its immunogenicity, only a few CD8+ cytotoxic-T-lymphocytes (CTLs), which can efficiently eliminate RCC cells, have been isolated [6]. This is in line with the small number of RCC-associated antigens that have so far been identified, thereby limiting the trials of candidate vaccines in these patients [7, 8].

Recently, tumor immunotherapy using DC has been shown to have therapeutic potential for malignant tumors. Moreover, nonmyeloablative stem cell transplantation (NST), which was developed for the treatment of leukemia, is effective against RCC [9, 10] and other solid tumors [11]. In this review, we discuss the current status of cell-mediated tumor-specific and nonspecific immunotherapy for RCC.

2. Tumor-Specific and Non-Specific Immunotherapy

In vivo studies show that cellular immunity mediated by T cells, natural killer (NK) cells or NK T cells plays a central role in the eradication of tumors. Since 1980, many attempts have been made to administer anti-tumor cells to cancer patients. In the late 1980s, human tumor antigens were identified and tumor-specific cellular immunity mediated via these tumor antigens received a lot of attention. Also, the administration of cytokines that activate cellular anti-tumor responses, including those mediated by T cells and NK cells, has been the subject of much research. It is thought that IFN-α induces Th1 cytokine production, thus promoting anti-tumor activity by cells that elicit cytotoxicity by acting directly on the tumor [12].

IL-2 is a growth/differentiation factor for NK cells and T cells, which induces and maintains the cytotoxicity, both these cell types [13]. Because cytokine treatment induces nonspecific anti-tumor activity, it is known as nonspecific immunotherapy.

In 1984, Mule et al. reported lymphokine-activated killer (LAK) cell treatment of tumors using inducible cultured cells [14]. Culturing immune cells isolated from a cancer patient's peripheral blood, or excised tumor tissue, with IL-2 causes them to differentiate into LAK cells. Since the second half of the 1980s, treatment using LAK cells has been attempted in several facilities [15, 16]. However, because the treatment method causes severe side effects, it was never established as an effective treatment method. LAK cells have no tumor specificity because they are induced in culture in response to IL-2 alone and not by tumor antigens. Thus, it was thought that the adoptive transfer of LAK cells might result in damage to normal host cells in vivo.

Since Van Der Bruggen et al. identified tumor antigens that were specifically recognized by T cells in a melanoma-bearing patient [17], research became more focused on tumor-specific immunotherapy. Though LAK cells, CTLs, macrophages, NK cells and NKT cells are all involved in host immune response against tumors, CTLs are now thought to be one of the most important factors responsible for anti-tumor immunity.

3. Immunotherapy Using Inactivated Tumor Cells and Gene Modified Tumor Vaccines (GMTV)

Immunotherapy using inactivated tumor cells or tumor lysates is based on the idea that tumor cells express antigens that induce anti-tumor immune responses [1822] (Table 1). Because immunotherapy using tumor cells is relatively straightforward, Jocham et al. undertook a large-scale randomized controlled trial and reported that the “nonreplaced phase” after surgery for kidney cancer was extended by an autologous tumor vaccine [20]. The percentage of vaccinated patients showing no disease progression 5 years after treatment was 77.4% compared with 67.8% of the controls.

Table 1.

Immunotherapy using inactivated tumor cells and a gene modified tumor vaccine (GMTV).

Authors Vaccine Adjuvant Patients Duration of PFS/RFS Results
Galligioni Auto irrad tumor BCG 120 13 mo 5-year DFS 63%
(control 72%) P = .21
Schwaab Auto irrad tumor BCG, IFN-α, IFN-γ 14 3 MR, 5 SD, l PD
Dillman Auto irrad tumor BCG, IFN-α, IFN-β 25 2.4 mo median survival 33.4 mo,
GM-CSF, Cy 5-year survival 43%
Jocham Auto lysate None 379 47.8 mo 5-year PFS 77.4%
(control 67.8%) P = .02
Dudek Auto LMI None, Cy, Cy+IL-2 31 2.8 mo None: 5 SD, Cy: 4 SD,
Cy+ IL-2: 1PR 3 SD
May Auto lysate None 495 5 year,10 year OS: 80.6, 68.9%
(control 79.2, 62.1%) P = .066
Simons Auto irrad tumor None 16 1 PR
+ GM-CSF
Wittig Auto irrad tumor Oligonucleotides 10 1 CR, 1 PR, 1 MR, 2 SD, 5 PD
+ GM-CSF, IL-7
Antonia Auto irrad tumor IL-2 15 2 PR, 2 SD
+ B7.1 gene
Tani Auto irrad tumor None 6 1 SD, l MR
+ GM-CSF
Pizza Auto irrad tumor None 30 170.5 dy 1 CR, 4 PR, 9 SD
+ IL-2
Moiseyenko Auto irrad tumor None 3 mo 1 SD, l MR
+ tag7/PGPR-S gene 4
Fishman Auto irrad tumor IL-2 39 1 CR, 2 PR, 24 SD
+ B7.1 gene
Buchner Auto irrad tumor None 12 5.3 mo PFS 5.3 mo, OS 15.6 mo
+ B7.1, IL-2 gene

LMI: large multivalent immunogen, Cy: cyclophosphamide, DFS: disease-free survival, Os: overall survival, PR: partial response, MR: mixed response, SD: stable disease, PD: progressive disease, PFS: progression-free survival, RFS: recurrence-free survival.

Both cytokines and antigen-presenting cells are important for the induction of effective immune responses [23]. Thus, GMTV was used to introduce virus-expressing cytokines, or costimulatory molecules, into tumor cells (Table 1) [1822, 2433]. GMTV-immunotherapy introducing cytokine transgene, such as GM-CSF or IL-2, or costimulatory molecule transgene such as B7-1 into autologous irradiated tumors, has been carried out. However, these studies were disappointing in terms of a significant clinical response, such as tumor regression. Though the use of multiple tumor antigens should induce a greater immune response, one cannot rule out the possibility of unintentionally inhibiting anti-tumor immunity or of eliciting nonspecific immune responses.

4. Peptide-Based Immunotherapy

Since the development of the SEREX method, which enables the identification of tumor antigens from cDNA libraries, many peptide-based vaccination studies have been undertaken. Because the effective induction of anti-tumor immunity using single peptides is difficult, MHC class II peptides have been used along with adjuvants (Table 2) [3440]. HSPPC-96 (vitespen) is a heat shock protein. It is a peptide complex, in which the heat shock protein plays the role of an adjuvant. However, a recent randomized phase III study suggested that this complex did not improve recurrence-free survival rates [41]. Further studies are required to see whether antigen-specific T cells homogeneously induced by a single tumor antigen can be effective against a diverse population of tumor cells.

Table 2.

Peptide-based immunotherapy.

Authors Stage Vaccine Adjuvant Patients Duration of PFS/RFS Results
Uemura mRCC CA9-derived peptide Incomplete Freund's adjuvant 23 12.2 mo 3 PR, 6SD
Iiyama mRCC WT 1-peptide Incomplete Freund's adjuvant 3 2 SD
Suekane mRCC 4 different peptides None, IFN-α, IL-2 10 23 wk 6 SD
Wood cT1b-T4N0M0 or T ant N1-2 M0 HSPPC-96 (vitespen) None 728 1.9 yr No difference in recurrence-free survival
Jonasch mRCC HSPPC-96 (vitespen) None 60 65 dy 2 CR, 2 PR, 7 SD

mRCC: metastatic RCC, PADRE: pan-MHC class II binding peptide, Auto mDC: autologous mature DC, CR: complete response, PR: partial response, SD: stable disease, PFS: progression-free survival, RFS: recurrence-free survival.

5. DC-Based Immunotherapy

Antigens processed within the proteasome of tumor cells are presented on major histocompatibility antigen (MHC) class I molecules of tumor cell as tumor antigen peptides that CTLs recognize, thus triggering CTL-mediated cytotoxicity. However, CTLs are not activated by direct recognition of the antigens expressed by tumor cells; they need help from dendritic cells (DCs) and CD4+ helper T cells. To activate a CD8+ T cell to become a CTL, engagement of the T cell receptor with a peptide antigen presented by an MHC class I molecule is not enough. The T cell must also recognize a costimulatory molecule (e.g. CD80 or CD86) (Figure 1). Moreover, antigen presenting cells (APCs) are activated through their interaction with CD4+ T cells, and then they express various costimulatory molecules. DCs are the most well-known and efficient APCs and are present in various tissues, including lymphoid and nonlymphoid organs and the blood, where they take up both particulate and soluble antigens before migrating to the lymph nodes to induce immune responses. Subsequently, DCs present antigen to T cells in the lymph nodes and induce antigen-specific immune responses, including the induction of CTLs. DCs also present antigen to other cells, including NK cells.

Figure 1.

Figure 1

CTL induction by Apcs. Antigens are taken up and degraded into peptide fragments by antigen presenting cells (APC), such as immature DC. At some point on their path to the cell surface, newly synthesized MHC class II or I molecules bind the peptide antigen fragments and transport the peptides to the cell surface. CD8+ T cells recognizing the antigen expressed by weakly costimulatory cells become activated only in the presence of CD4+ T cells bound to the same APC. This happens via CD4+ T cells recognizing antigens presented by APCs and being triggered to induce increased levels of costimulatory activity by the antigen-presenting cell. The CD4+ T cells also produce increased amounts of IL-2, which drives CD8+ T cell proliferation. CD8+ T cells then become cytotoxic T lymphocytes (CTL).

Clinical trials of DC therapy are listed in Table 3 [23, 35, 36, 4259]. Although immunotherapy using DCs and nonautologous tumor cells seems to induce host immune cells to recognize tumor cells, there is still the possibility of alloreactive immune responses induced by nonself-antigens. Because nonautologous DCs (allo-DCs) may be attacked by the host immune system, immunotherapy using autologous-DCs (auto DCs) might be more effective in vivo. To date, all reports regarding DC treatment are of phase I/II trials incorporating different methodologies. Although delayed-type hypersensitivity reactions in response to tumor cell lysates or keyhole limpet hemocyanin (KLH) and the production of IFN-γ by antigen-specific lymphocytes were observed, the number of patients showing a positive clinical response was still low.

Table 3.

DC-based immunotherapy.

Authors Antigen DC Adjuvant Patients Duration of PFS/RFS Results
Oosterwijk-Wakka Auto lysate Auto imDC KLH/IL-2 12 8 SD, 4 PD
Marten Auto lysate Auto mDC KLH 15 1 PR, 7 SD, 7 PD
Holtl Auto & Allo lysate Auto mDC KLH 27 20.4 mo 2 CR, 1 PR, 7 SD, 17 PD
Azuma Auto lysate Auto imDC KLH 3 1 NC, 2 PD
Marten DC/auto tumor fusion Allo mDC 12 4 SD, 8 PD
Su tumor RNA Auto imDC 10 not evaluated
Gitliz Auto lysate Auto imDC 12 1 PR, 3 SD, 8 PD
Barbuto DC/auto tumor fusion Allo mDC 19 5.7 mo 30 R, 14 SD, 2 PD
Avigan DC/auto tumor fusion Auto imDC KLH 13 4.2 mo 5 SD, 8 PD
Pandha Allo lysate Auto imDC KLH 5 2 SD
Arroyo Auto lysate Auto mDC KLH 5 9.6 mo (5–16) 3 SD
Holtl Auto & Allo TuLy Allo mDC KLH/Cy 20 22.3 mo 2 MR, 3 SD, 15 PD
Wierecky MUC-1 peptide Auto mDC PADRE 20 10.8 mo (4–24) 1 CR, 2 MR, 2 PR, 5 SD, 10 PD
Bleumer CA9 peptide Auto mDC KLH CA9 class II peptide 6 6 PD
Wei DC/auto tumor fusion Auto mDC IL-2 10 7 mo (5–12) 1 PR, 3 SD, 6 PD
Matsumoto Auto lysate Auto mDC KLH 3 1 SD, 2 PD
Kim Auto lysate Auto mDC KLH 9 5.2 mo 1 PR, 5 SD, 3 PD
Berntsen Lysate or surviving and telomerase peptides Auto mDC IL-2 27 2.7 mo 13 SD, 14 PD
Tatsugami Auto TuLy Auto mDC IFN-α 7 7.8 mo 5 SD, 2 PD
Zbou DC/auto tumor fusion Allo mDC 10 1 PR, 6 SD, 3 PD

Cy: cyclophosphamide, PADRE: pan-MHC class II binding peptide, Auto mDC: autologous mature DC, Allo imDC: allogeneic immature DC CR: complete response, PR: partial response, MR: mixed response, SD: stable disease, OR: objective response, PD: progressive disease, PFS: progression-free survival, RFS: recurrence-free survival.

We also used IFN-α as an adjunctive agent for DC therapy. As previously noted, IFN-α induced an environment conducive to DC activation and enhanced migratory competence [60, 61]. We evaluated the efficacy of DC-therapy in combination with IFN-α in patients with advanced RCC. After 4 months of vaccinations, five patients had stable disease and two had progressive disease. In six patients, the time-to-progression was prolonged compared with that seen after previous cytokine treatment. Because cytokine combination therapy induces the proliferation and maintenance of DC-activated T cells, combination therapy using IL-2 is reasonable. However, Oosterwijk et al. reported that combination therapy with IL-2 plus DCs was no more effective than DCs alone [44]. Recently, it was reported that IL-2 participates in the maintenance of regulatory T cells (Tregs), which suppress immune responses [62]. Further study of the role of IL-2 in immunotherapy is required.

6. Nonmyeloablative Stem Cell Transplantation (NST)

Though NST was developed for the treatment of leukemia, it began to gain attention as a treatment for solid tumors. In 2000, Childs et al. performed NST on 19 renal carcinoma patients and reported a success rate 53%; three patients were in complete remission and seven patents were in partial remission. Previous reports have highlighted the important role played by cellular anti-tumor immunity, including that mediated by donor T cells in graft versus host disease (GVHD) and the graft versus tumor effect (GVT); the appearance of GVHD induced by transplantation of donor T cells is inversely correlated with the rate of tumor recurrence. Recurrence is especially high in T cell-depleted stem cell transplants, and the administration of donor lymphocytes effectively reduces the incidence of recurrence [63, 64]. Donor T cells induce GVHD/GVT against recipient antigens, including MHC molecules, minor histocompatibility antigens and tumor cell-specific antigens. An effective GVT response can be induced if the antigen distribution between normal cells and tumor cells can be identified, and if donor T cell responses against normal cells can be controlled. Thus, in NST, the mechanism by which tumor specific immunity is induced is very important, and a recent study attempted to address the question of how this response was activated [65].

When the patient receives immunosuppressive treatment for GVHD, it might also cause suppression of the associated anti-tumor effects. In these patients, the differentiation of mononuclear cells into DCs is inhibited in vitro [66]. Therefore, when treating a patient with NST, one should bear in mind possible aggravation of the neoplasm by immunosuppressive therapy directed against GVHD.

7. Regulatory CD4+ T Cells and the Tumor

Recent research shows that CD4+ T cells constitutively expressing the IL-2 receptor α-chain (CD25) act in a regulatory capacity by suppressing the activation and function of other T cells [67]. Their physiological role is to protect the host against the development of autoimmunity by regulating immune responses against antigens expressed by normal tissues [68, 69]. Since tumor antigens are largely self-antigens, these so-called Treg cells may also prevent the tumor-bearing host from mounting an effective antitumor immune response. Previous studies have shown that elevated numbers of CD4+CD25+ Tregs can be found in patients with advanced cancer [70] and that high Treg frequencies are associated with reduced survival [71]. In our experiments into cytokine therapy for RCC patients, the number of CD4+ and FoxP3+Treg cells was significantly decreased after IFN-α treatment, and Treg cell levels before treatment correlated with the clinical response [72]. The important role of CD4+CD25+ Tregs in controlling tumor growth was further highlighted by the demonstration that depletion of Tregs using anti-CD25 antibodies evokes effective antitumor immunity in mice [73, 74]. Dannull et al. used a recombinant IL-2:diphtheria toxin conjugate (DAB389IL-2; also known as denileukin diftitox and ONTAK) to eliminate CD25-expressing Tregs in metastatic RCC patients, and reported that depletion of Tregs in RCC patients followed by vaccination with tumor RNA-transfected DCs led to improved stimulation of tumor-specific T cells compared with vaccination alone [75]. It will be critical to collect accurate information regarding Tregs to address the clinical efficacy of such strategies in cancer patients.

8. Conclusions

The use of immunotherapy using cultured cells, such as DCs, to treat large numbers of patients, and the conduction of large-scale studies are difficult because of the problems associated with the need for adequate culture facilities and appropriate culture techniques. Because of the complexity of the immune responses involved, it is difficult to evaluate the efficacy of immunotherapy compared with other treatments. However, as it is clear that the immune system plays a significant role in the control of tumors, continued analysis of the mechanisms involved in tumor immunity and the development of new immunotherapies are vital.

References

  • 1.Mulders PFA, Brouwers AH, Hulsbergen-van Der Kaa CA, Van Lin ENJTH, Osanto S, De Mulder PHM. Guideline ’Renal cell carcinoma. Nederlands Tijdschrift voor Geneeskunde. 2008;152(7):376–380. [PubMed] [Google Scholar]
  • 2.Alexandrescu DT, Dasanu CA. Kidney cancer therapy: new perspectives and avenues. Expert Opinion on Pharmacotherapy. 2006;7(18):2481–2493. doi: 10.1517/14656566.7.18.2481. [DOI] [PubMed] [Google Scholar]
  • 3.Yang JC, Childs R. Immunotherapy for renal cell cancer. Journal of Clinical Oncology. 2006;24(35):5576–5583. doi: 10.1200/JCO.2006.08.3774. [DOI] [PubMed] [Google Scholar]
  • 4.Motzer RJ, Bukowski RM. Targeted therapy for metastatic renal cell carcinoma. Journal of Clinical Oncology. 2006;24(35):5601–5608. doi: 10.1200/JCO.2006.08.5415. [DOI] [PubMed] [Google Scholar]
  • 5.Leibovich BC, Han KR, Bui MHT, et al. Scoring algorithm to predict survival after nephrectomy and immunotherapy in patients with metastatic renal cell carcinoma: a stratification tool for prospective clinical trials. Cancer. 2003;98(12):2566–2575. doi: 10.1002/cncr.11851. [DOI] [PubMed] [Google Scholar]
  • 6.Finke JH, Rayman P, Edinger M, et al. Characterization of a human renal cell carcinoma specific cytotoxic CD8 T cell line. Journal of Immunotherapy. 1992;11(1):1–11. doi: 10.1097/00002371-199201000-00001. [DOI] [PubMed] [Google Scholar]
  • 7.Brouwenstijn N, Gaugler B, Krüse KM, et al. Renal-cell carcinoma-specific lysis by cytotoxic T-lymphocyte clones isolated from peripheral blood lymphocytes and tumor-infiltrating lymphocytes. International Journal of Cancer. 1996;68(2):177–182. doi: 10.1002/(SICI)1097-0215(19961009)68:2<177::AID-IJC6>3.0.CO;2-V. [DOI] [PubMed] [Google Scholar]
  • 8.Gaudin C, Kremer F, Angevin E, Scott V, Triebel F. A hsp70-2 mutation recognized by CTL on a human renal cell carcinoma. Journal of Immunology. 1999;162(3):1730–1738. [PubMed] [Google Scholar]
  • 9.Childs R, Chernoff A, Contentin N, et al. Regression of metastatic renal-cell carcinoma after nonmyeloablative allogeneic peripheral-blood stem-cell transplantation. New England Journal of Medicine. 2000;343(11):750–758. doi: 10.1056/NEJM200009143431101. [DOI] [PubMed] [Google Scholar]
  • 10.Rini BI, Zimmerman T, Stadler WM, Gajewski TF, Vogelzang NJ. Allogeneic stem-cell transplantation of renal cell cancer after nonmyeloablative chemotherapy: feasibility, engraftment, and clinical results. Journal of Clinical Oncology. 2002;20(8):2017–2024. doi: 10.1200/JCO.2002.08.068. [DOI] [PubMed] [Google Scholar]
  • 11.Slavin S, Nagler A, Naparstek E, et al. Nonmyeloablative stem cell transplantation and cell therapy as an alternative to conventional bone marrow transplantation with lethal cytoreduction for the treatment of malignant and nonmalignant hematologic diseases. Blood. 1998;91(3):756–763. [PubMed] [Google Scholar]
  • 12.Belardelli F, Ferrantini M, Proietti E, Kirkwood JM. Interferon-alpha in tumor immunity and immunotherapy. Cytokine and Growth Factor Reviews. 2002;13(2):119–134. doi: 10.1016/s1359-6101(01)00022-3. [DOI] [PubMed] [Google Scholar]
  • 13.Margolin KA. Interleukin-2 in the treatment of renal cancer. Seminars in Oncology. 2000;27(2):194–203. [PubMed] [Google Scholar]
  • 14.Mule JJ, Shu S, Schwarz SL, Rosenberg SA. Adoptive immunotherapy of established pulmonary metastases with LAK cells and recombinant interleukin-2. Science. 1984;225(4669):1487–1489. doi: 10.1126/science.6332379. [DOI] [PubMed] [Google Scholar]
  • 15.Negrier S, Philip T, Stoter G, et al. Interleukin-2 with or without LAK cells in metastatic renal cell carcinoma: a report of a European multicentre study. European Journal of Cancer and Clinical Oncology. 1989;25(supplement 3):S21–S28. [PubMed] [Google Scholar]
  • 16.Kuebler JP, Whitehead RP, Ward DL, Hemstreet GP, Bradley EC, DeKernion JB. Treatment of metastatic renal cell carcinoma with recombinant interleukin- 2 in combination with vinblastine or lymphokine-activated killer cells. Journal of Urology. 1993;150(3):814–820. doi: 10.1016/s0022-5347(17)35620-3. [DOI] [PubMed] [Google Scholar]
  • 17.Van Der Bruggen P, Traversari C, Chomez P, et al. A gene encoding an antigen recognized by cytolytic T lymphocytes on a human melanoma. Science. 1991;254(5038):1643–1647. doi: 10.1126/science.1840703. [DOI] [PubMed] [Google Scholar]
  • 18.Galligioni E, Quaia M, Merlo A, et al. Adjuvant immunotherapy treatment of renal carcinoma patients with autologous tumor cells and bacillus Calmette-Guèrin: five-year results of a prospective randomized study. Cancer. 1996;77(12):2560–2566. doi: 10.1002/(SICI)1097-0142(19960615)77:12<2560::AID-CNCR20>3.0.CO;2-P. [DOI] [PubMed] [Google Scholar]
  • 19.Schwaab T, Heaney JA, Schned AR, et al. A randomized phase II trial comparing two different sequence combinations of autologous vaccine and human recombinant interferon γ and human recombinant interferon α2B therapy in patients with metastatic renal cell carcinoma: clinical outcome and analysis of immunological parameters. Journal of Urology. 2000;163(4):1322–1327. doi: 10.1016/s0022-5347(05)67771-3. [DOI] [PubMed] [Google Scholar]
  • 20.Jocham D, Richter A, Hoffmann L, et al. Adjuvant autologous renal tumour cell vaccine and risk of tumour progression in patients with renal-cell carcinoma after radical nephrectomy: phase III, randomised controlled trial. Lancet. 2004;363(9409):594–599. doi: 10.1016/S0140-6736(04)15590-6. [DOI] [PubMed] [Google Scholar]
  • 21.Dudek AZ, Mescher MF, Okazaki I, et al. Autologous large multivalent immunogen vaccine in patients with metastatic melanoma and renal cell carcinoma. American Journal of Clinical Oncology: Cancer Clinical Trials. 2008;31(2):173–181. doi: 10.1097/COC.0b013e3181573e6b. [DOI] [PubMed] [Google Scholar]
  • 22.Dillman R, Barth N, VanderMolen L, et al. Autologous tumor cell line-derived vaccine for patient-specific treatment of advanced renal cell carcinoma. Cancer Biotherapy and Radiopharmaceuticals. 2004;19(5):570–580. doi: 10.1089/cbr.2004.19.570. [DOI] [PubMed] [Google Scholar]
  • 23.Tatsugami K, Eto M, Harano M, et al. Dendritic cell therapy in combination with interferon-α for the treatment of metastatic renal cell carcinoma. International Journal of Urology. 2008;15(8):694–698. doi: 10.1111/j.1442-2042.2008.02088.x. [DOI] [PubMed] [Google Scholar]
  • 24.Antonia SJ, Seigne J, Diaz J, et al. Phase I trial of a B7-1 (CD80) gene modified autologous tumor cell vaccine in combination with systemic interleukin-2 in patients with metastatic renal cell carcinoma. Journal of Urology. 2002;167(5):1995–2000. [PubMed] [Google Scholar]
  • 25.May M, Kendel F, Hoschke B, et al. Adjuvant autologous tumour cell vaccination in patients with renal cell carcinoma: overall survival analysis with a follow-up period in excess of more than 10 yearsAdjuvante autologe Tumorvakzine beim Nierenzellkarzinom: gesamtüberlebensanalyse mit einem Nachbeobachtungszeitraum von mehr als 10 Jahren. Urologe A. 2009;48(9):1075–1083. doi: 10.1007/s00120-009-2044-y. [DOI] [PubMed] [Google Scholar]
  • 26.Buchner A, Pohla H, Willimsky G, et al. Phase 1 trial of allogeneic gene-modified tumor cell vaccine RCC-26/CD80/IL-2 in patients with metastatic renal cell carcinoma. Human Gene Therapy. 2010;21(3):285–297. doi: 10.1089/hum.2008.192. [DOI] [PubMed] [Google Scholar]
  • 27.Dillman RO, Barth NM, VanderMolen LA, et al. Treatment of kidney cancer with autologous tumor cell vaccines of short-term cell lines derived from renal cell carcinoma. Cancer Biotherapy and Radiopharmaceuticals. 2001;16(1):47–54. doi: 10.1089/108497801750096023. [DOI] [PubMed] [Google Scholar]
  • 28.Simons JW, Mikhak B. Ex vivo gene therapy using cytokine-transduced tumor vaccines: molecular and clinical pharmacology. Seminars in Oncology. 1998;25(6):661–676. [PubMed] [Google Scholar]
  • 29.Wittig B, Märten A, Dorbic T, et al. Therapeutic vaccination against metastatic carcinoma by expression-modulated and immunomodified autologous tumor cells: a first clinical phase I/II trial. Human Gene Therapy. 2001;12(3):267–278. doi: 10.1089/10430340150218404. [DOI] [PubMed] [Google Scholar]
  • 30.Fishman M, Hunter TB, Soliman H, et al. Phase II trial of B7-1 (CD-86) transduced, cultured autologous tumor cell vaccine plus subcutaneous interleukin-2 for treatment of stage IV renal cell carcinoma. Journal of Immunotherapy. 2008;31(1):72–80. doi: 10.1097/CJI.0b013e31815ba792. [DOI] [PubMed] [Google Scholar]
  • 31.Tani K, Azuma M, Nakazaki Y, et al. Phase I study of autologous tumor vaccines transduced with the GM-CSF gene in four patients with stage IV renal cell cancer in Japan: clinical and immunological findings. Molecular Therapy. 2004;10(4):799–816. doi: 10.1016/j.ymthe.2004.07.001. [DOI] [PubMed] [Google Scholar]
  • 32.Pizza G, De Vinci C, Lo Conte G, et al. Allogeneic gene-modified tumour cells in metastatic kidney cancer. Report II. Folia Biologica. 2004;50(6):175–183. [PubMed] [Google Scholar]
  • 33.Moiseyenko VM, Danilov AO, Baldueva IA, et al. Phase I/II trial of gene therapy with autologous tumor cells modified with tag7/PGRP-S gene in patients with disseminated solid tumors. Annals of Oncology. 2005;16(1):162–168. doi: 10.1093/annonc/mdi028. [DOI] [PubMed] [Google Scholar]
  • 34.Uemura H, Fujimoto K, Tanaka M, et al. A phase I trial of vaccination of CA9-derived peptides for HLA-A24-positive patients with cytokine-refractory metastatic renal cell carcinoma. Clinical Cancer Research. 2006;12(6):1768–1775. doi: 10.1158/1078-0432.CCR-05-2253. [DOI] [PubMed] [Google Scholar]
  • 35.Wierecky J, Mueller M, Brossart P. Dendritic cell-based cancer immunotherapy targeting MUC-1. Cancer Immunology, Immunotherapy. 2006;55(1):63–67. doi: 10.1007/s00262-005-0673-6. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 36.Bleumer I, Tiemessen DM, Oosterwijk-Wakka JC, et al. Preliminary analysis of patients with progressive renal cell carcinoma vaccinated with CA9-peptide-pulsed mature dendritic cells. Journal of Immunotherapy. 2007;30(1):116–122. doi: 10.1097/01.cji.0000211318.22902.ec. [DOI] [PubMed] [Google Scholar]
  • 37.Iiyama T, Udaka K, Takeda S, et al. WT1 (Wilms’ tumor 1) peptide immunotherapy for renal cell carcinoma. Microbiology and Immunology. 2007;51(5):519–530. doi: 10.1111/j.1348-0421.2007.tb03940.x. [DOI] [PubMed] [Google Scholar]
  • 38.Suekane S, Nishitani M, Noguchi M, et al. Phase I trial of personalized peptide vaccination for cytokine-refractory metastatic renal cell carcinoma patients. Cancer Science. 2007;98(12):1965–1968. doi: 10.1111/j.1349-7006.2007.00631.x. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 39.Wood C, Srivastava P, Bukowski R, et al. An adjuvant autologous therapeutic vaccine (HSPPC-96; vitespen) versus observation alone for patients at high risk of recurrence after nephrectomy for renal cell carcinoma: a multicentre, open-label, randomised phase III trial. The Lancet. 2008;372(9633):145–154. doi: 10.1016/S0140-6736(08)60697-2. [DOI] [PubMed] [Google Scholar]
  • 40.Jonasch E, Wood C, Tamboli P, et al. Vaccination of metastatic renal cell carcinoma patients with autologous tumour-derived vitespen vaccine: clinical findings. British Journal of Cancer. 2008;98(8):1336–1341. doi: 10.1038/sj.bjc.6604266. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 41.Van Poppel H, Joniau S, Van Gool SW. Vaccine therapy in patients with renal cell carcinoma. European Urology. 2009;55(6):1333–1344. doi: 10.1016/j.eururo.2009.01.043. [DOI] [PubMed] [Google Scholar]
  • 42.Wei YC, Sticca RP, Li J, et al. Combined treatment of dendritoma vaccine and low-dose interleukin-2 in stage IV renal cell carcinoma patients induced clinical response: a pilot study. Oncology Reports. 2007;18(3):665–671. [PubMed] [Google Scholar]
  • 43.Kim JH, Lee Y, Bae YS, et al. Phase I/II study of immunotherapy using autologous tumor lysate-pulsed dendritic cells in patients with metastatic renal cell carcinoma. Clinical Immunology. 2007;125(3):257–267. doi: 10.1016/j.clim.2007.07.014. [DOI] [PubMed] [Google Scholar]
  • 44.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. Journal of Immunotherapy. 2002;25(6):500–508. doi: 10.1097/00002371-200211000-00006. [DOI] [PubMed] [Google Scholar]
  • 45.Märten 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 Immunology, Immunotherapy. 2002;51(11-12):637–644. doi: 10.1007/s00262-002-0324-0. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 46.Höltl L, Zelle-Rieser C, Gander H, et al. Immunotherapy of metastatic renal cell carcinoma with tumor lysate-pulsed autologous dendritic cells. Clinical Cancer Research. 2002;8(11):3369–3376. [PubMed] [Google Scholar]
  • 47.Azuma T, Horie S, Tomita K, et al. Dendritic cell immunotherapy for patients with metastatic renal cell carcinoma: university of Tokyo experience. International Journal of Urology. 2002;9(6):340–346. doi: 10.1046/j.1442-2042.2002.00477.x. [DOI] [PubMed] [Google Scholar]
  • 48.Märten 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. Human Gene Therapy. 2003;14(5):483–494. doi: 10.1089/104303403321467243. [DOI] [PubMed] [Google Scholar]
  • 49.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 Research. 2003;63(9):2127–2133. [PubMed] [Google Scholar]
  • 50.Gitlitz BJ, Belldegrun AS, Zisman A, et al. A pilot trial of tumor lysate-loaded dendritic cells for the treatment of metastatic renal cell carcinoma. Journal of Immunotherapy. 2003;26(5):412–419. doi: 10.1097/00002371-200309000-00004. [DOI] [PubMed] [Google Scholar]
  • 51.Barbuto JAM, Ensina LFC, Neves AR, et al. Dendritic cell-tumor cell hybrid vaccination for metastatic cancer. Cancer Immunology, Immunotherapy. 2004;53(12):1111–1118. doi: 10.1007/s00262-004-0551-7. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 52.Pandha HS, John RJ, Hutchinson J, et al. Dendritic cell immunotherapy for urological cancers using cryopreserved allogeneic tumour lysate-pulsed cells: a phase I/II study. BJU International. 2004;94(3):412–418. doi: 10.1111/j.1464-410X.2004.04922.x. [DOI] [PubMed] [Google Scholar]
  • 53.Arroyo JC, Gabilondo F, Llorente L, Meraz-Ríos MA, Sánchez-Torres C. Immune response induced in Vitro by CD16 and CD16 monocyte-derived dendritic cells in patients with metastatic renal cell carcinoma treated with dendritic cell vaccines. Journal of Clinical Immunology. 2004;24(1):86–96. doi: 10.1023/B:JOCI.0000018067.71622.fb. [DOI] [PubMed] [Google Scholar]
  • 54.Wierecky J, Müller MR, Wirths S, et al. Immunologic and clinical responses after vaccinations with peptide-pulsed dendritic cells in metastatic renal cancer patients. Cancer Research. 2006;66(11):5910–5918. doi: 10.1158/0008-5472.CAN-05-3905. [DOI] [PubMed] [Google Scholar]
  • 55.Matsumoto A, Haraguchi K, Takahashi T, et al. Immunotherapy against metastatic renal cell carcinoma with mature dendritic cells. International Journal of Urology. 2007;14(4):277–283. doi: 10.1111/j.1442-2042.2006.01723.x. [DOI] [PubMed] [Google Scholar]
  • 56.Berntsen A, Trepiakas R, Wenandy L, et al. Therapeutic dendritic cell vaccination of patients with metastatic renal cell carcinoma: a clinical phase 1/2 trial. Journal of Immunotherapy. 2008;31(8):771–780. doi: 10.1097/CJI.0b013e3181833818. [DOI] [PubMed] [Google Scholar]
  • 57.Zhou J, Weng D, Zhou F, et al. Patient-derived renal cell carcinoma cells fused with allogeneic dendritic cells elicit anti-tumor activity: in vitro results and clinical responses. Cancer Immunology, immunotherapy . 2009;58(10):1587–1597. doi: 10.1007/s00262-009-0668-9. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 58.Höltl L, Ramoner R, Zelle-Rieser C, et al. Allogeneic dendritic cell vaccination against metastatic renal cell carcinoma with or without cyclophosphamide. Cancer Immunology, Immunotherapy. 2005;54(7):663–670. doi: 10.1007/s00262-004-0629-2. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 59.Avigan D, Vasir B, Gong J, et al. Fusion cell vaccination of patients with metastatic breast and renal cancer induces immunological and clinical responses. Clinical Cancer Research. 2004;10(14):4699–4708. doi: 10.1158/1078-0432.CCR-04-0347. [DOI] [PubMed] [Google Scholar]
  • 60.Luft T, Luetjens P, Hochrein H, et al. IFN-α enhances CD40 ligand-mediated activation of immature monocyte-derived dendritic cells. International Immunology. 2002;14(4):367–380. doi: 10.1093/intimm/14.4.367. [DOI] [PubMed] [Google Scholar]
  • 61.Padovan E, Spagnoli GC, Ferrantini M, Heberer M. IFN-α2a induces IP-10/CXCL10 and MIG/CXCL9 production in monocyte-derived dendritic cells and enhances their capacity to attract and stimulate CD8 effector T cells. Journal of Leukocyte Biology. 2002;71(4):669–676. [PubMed] [Google Scholar]
  • 62.Zhang H, Chua KS, Guimond M, et al. Lymphopenia and interleukin-2 therapy alter homeostasis of CD4 CD25 regulatory T cells. Nature Medicine. 2005;11(11):1238–1243. doi: 10.1038/nm1312. [DOI] [PubMed] [Google Scholar]
  • 63.Horowitz MM, Gale RP, Sondel PM, et al. Graft-versus-leukemia reactions after bone marrow transplantation. Blood. 1990;75(3):555–562. [PubMed] [Google Scholar]
  • 64.Kolb HJ, Mittermuller J, Clemm C, et al. Donor leukocyte transfusions for treatment of recurrent chronic myelogenous leukemia in marrow transplant patients. Blood. 1990;76(12):2462–2465. [PubMed] [Google Scholar]
  • 65.Harano M, Eto M, Iwai T, et al. Renal cancer treatment with low levels of mixed chimerism induced by nonmyeloablative regimen using cyclophosphamide in mice. Cancer Research. 2005;65(21):10032–10040. doi: 10.1158/0008-5472.CAN-05-0457. [DOI] [PubMed] [Google Scholar]
  • 66.Tatsugami K, Eto M, Harano M, et al. Dendritic-cell therapy after non-myeloablative stem-cell transplantation for renal-cell carcinoma. Lancet Oncology. 2004;5(12):750–752. doi: 10.1016/S1470-2045(04)01652-3. [DOI] [PubMed] [Google Scholar]
  • 67.Shevach EM. Certified professionals: CD4CD25 suppressor T cells. Journal of Experimental Medicine. 2001;193(11):F41–F45. doi: 10.1084/jem.193.11.f41. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 68.Jonuleit H, Schmitt E, Schuler G, Knop J, Enk AH. Induction of interleukin 10-producing, nonproliferating CD4 T cells with regulatory properties by repetitive stimulation with allogeneic immature human dendritic cells. Journal of Experimental Medicine. 2000;192(9):1213–1222. doi: 10.1084/jem.192.9.1213. [DOI] [PMC free article] [PubMed] [Google Scholar]
  • 69.Read S, Powrie F. CD4 regulatory T cells. Current Opinion in Immunology. 2001;13(6):644–649. doi: 10.1016/s0952-7915(01)00273-4. [DOI] [PubMed] [Google Scholar]
  • 70.Woo EY, Yeh H, Chu CS, et al. Cutting edge: regulatory T cells from lung cancer patients directly inhibit autologous T cell proliferation. Journal of Immunology. 2002;168(9):4272–4276. doi: 10.4049/jimmunol.168.9.4272. [DOI] [PubMed] [Google Scholar]
  • 71.Curiel TJ, Coukos G, Zou L, et al. Specific recruitment of regulatory T cells in ovarian carcinoma fosters immune privilege and predicts reduced survival. Nature Medicine. 2004;10(9):942–949. doi: 10.1038/nm1093. [DOI] [PubMed] [Google Scholar]
  • 72.Tatsugami K, Eto M, Naito S. Influence of immunotherapy with interferon-α on regulatory T cells in renal cell carcinoma patients. Journal of Interferon and Cytokine Research. 2010;30(1):43–48. doi: 10.1089/jir.2009.0014. [DOI] [PubMed] [Google Scholar]
  • 73.Shimizu J, Yamazaki S, Sakaguchi S. Induction of tumor immunity by removing CD25+CD4+ T cells: a common basis between tumor immunity and autoimmunity. Journal of Immunology. 1999;163(10):5211–5218. [PubMed] [Google Scholar]
  • 74.Onizuka S, Tawara I, Shimizu J, Sakaguchi S, Fujita T, Nakayama E. Tumor rejection by in vivo administration of anti-CD25 (interleukin-2 receptor α) monoclonal antibody. Cancer Research. 1999;59(13):3128–3133. [PubMed] [Google Scholar]
  • 75.Dannull J, Su Z, Rizzieri D, et al. Enhancement of vaccine-mediated antitumor immunity in cancer patients after depletion of regulatory T cells. Journal of Clinical Investigation. 2005;115(12):3623–3633. doi: 10.1172/JCI25947. [DOI] [PMC free article] [PubMed] [Google Scholar]

Articles from Clinical and Developmental Immunology are provided here courtesy of Wiley

RESOURCES