1
|
Du X, Li M, Huan C, Lv G. Dendritic cells in liver transplantation immune response. Front Cell Dev Biol 2023; 11:1277743. [PMID: 37900282 PMCID: PMC10606587 DOI: 10.3389/fcell.2023.1277743] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/15/2023] [Accepted: 09/27/2023] [Indexed: 10/31/2023] Open
Abstract
Dendritic cells (DCs) are the most powerful antigen presenting cells (APCs), they are considered one of the key regulatory factors in the liver immune system. There is currently much interest in modulating DC function to improve transplant immune response. In liver transplantation, DCs participate in both the promotion and inhibition of the alloreponse by adopting different phenotypes and function. Thus, in this review, we discussed the origin, maturation, migration and pathological effects of several DC subsets, including the conventional DC (cDC), plasmacytoid DC (pDC) and monocyte-derived DC (Mo-DC) in liver transplantation, and we summarized the roles of these DC subsets in liver transplant rejection and tolerance. In addition, we also outlined the latest progress in DC-based related treatment regimens. Overall, our discussion provides a beneficial resource for better understanding the biology of DCs and their manipulation to improve the immune adaptability of patients in transplant status.
Collapse
Affiliation(s)
- Xiaodong Du
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Mingqian Li
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| | - Chen Huan
- Center of Infectious Diseases and Pathogen Biology, Institute of Virology and AIDS Research, Key Laboratory of Organ Regeneration and Transplantation of The Ministry of Education, The First Hospital of Jilin University, Changchun, China
| | - Guoyue Lv
- Department of Hepatobiliary and Pancreatic Surgery, General Surgery Center, The First Hospital of Jilin University, Changchun, China
| |
Collapse
|
2
|
Sönmez MG, Sönmez LÖ. New treatment modalities with vaccine therapy in renal cell carcinoma. Urol Ann 2019; 11:119-125. [PMID: 31040593 PMCID: PMC6476201 DOI: 10.4103/ua.ua_166_17] [Citation(s) in RCA: 8] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/31/2017] [Accepted: 01/06/2019] [Indexed: 12/21/2022] Open
Abstract
The aim of implementing vaccine therapy is to activate immune response against malignant cells by overcoming the tolerance triggered by the tumor. These treatments are effective using the immune response against cancer. Not every type of cancer is suitable for vaccine therapies. For a vaccine therapy to be implemented, cancer should be immunogenic and contain tissue-specific proteins, should have a slow progression, and treatments should be feasible. For that reason, studies regarding urological cancers are mostly focused on the kidneys and the prostate. Vaccine therapies used in renal cell carcinoma (RCC) can be categorized under the following titles: autologous tumor cells, dendritic cells, genetically modified tumor cells, and protein/peptide. Although there are old studies on the implementation of vaccine therapies in RCC, researches have only been intensified recently. In addition to their effective potential for lengthening general survival, decreasing tumor burden and cancer development in long term, vaccine treatments are especially effective in metastatic RCC patients. We think that vaccine treatments would be applied more in near future since RCC are immunogenic. In this compilation, we will discuss vaccine therapies used in RCC, which urologists are not so familiar with, in the light of the up-to-date literature.
Collapse
Affiliation(s)
- Mehmet Giray Sönmez
- Department of Urology, Meram Medical Faculty, Necmettin Erbakan University, Konya, Turkey
| | - Leyla Öztürk Sönmez
- Department of Physiology, Selcuklu Medical School, Selcuk University, Konya, Turkey
| |
Collapse
|
3
|
Brookman-May S, May M, Gilfrich C, Ferdinand Wieland W, Burger M. Can vaccination or tyrosine kinase inhibitor therapy play a role in the adjuvant treatment of renal cell carcinoma? Expert Rev Anticancer Ther 2014; 10:813-23. [DOI: 10.1586/era.10.55] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
4
|
Bockorny B, Dasanu CA. Intrinsic immune alterations in renal cell carcinoma and emerging immunotherapeutic approaches. Expert Opin Biol Ther 2013; 13:911-25. [PMID: 23586712 DOI: 10.1517/14712598.2013.778970] [Citation(s) in RCA: 15] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/20/2023]
Abstract
INTRODUCTION Individuals affected by kidney cancer present a variety of immune abnormalities including cellular immune dysfunction, cytokine alterations and antigen presentation defects. On the other hand, spontaneous remissions are seen in up to 4% of renal cell carcinoma (RCC) patients and they are thought to occur via immune mechanisms. AREAS COVERED The authors comprehensively review the immune abnormalities in RCC patient and describe the kidney cancer immunotherapy candidates that are most advanced in their clinical development. Most relevant publications were identified through searching the PubMed database; the obtained information was thoroughly analyzed and synthesized. EXPERT OPINION As cure in advanced RCC cannot be accomplished with the current therapy standards such as tyrosine kinase inhibitors and mammalian target of rapamycin inhibitors, new treatment strategies are being sought. Enhancing the immune system represents an appealing avenue for kidney cancer therapy. Disappointingly, high-dose interleukin-2 and interferon-α cause severe toxicity and produce a questionable clinical benefit. The authors postulate that the 'durable responses' seen with these agents in only a handful of RCC patients represent spontaneous remissions. Promising immune strategies in RCC such as anti-cytotoxic T-lymphocyte-associated protein antibodies, anti-programmed cell death 1 (PD1)/PD1 ligand and tumor vaccines may expand the existing options for kidney cancer in future years.
Collapse
Affiliation(s)
- Bruno Bockorny
- University of Connecticut, Department of Medicine, 263 Farmington Avenue, Farmington, CT 06030-1235, USA.
| | | |
Collapse
|
5
|
Yoshimura K, Uemura H. Role of vaccine therapy for renal cell carcinoma in the era of targeted therapy. Int J Urol 2013; 20:744-55. [PMID: 23521119 DOI: 10.1111/iju.12147] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/18/2012] [Accepted: 02/21/2013] [Indexed: 12/28/2022]
Abstract
Renal cell carcinoma is the most common malignant tumor originating from the kidney. Compared with other solid tumors, it does not respond to traditional management modalities, such as chemotherapy and radiotherapy. However, it is well known that renal cell carcinoma represents one of the most immune-responsive cancers and several immunotherapeutic strategies have been investigated in the management of renal cell carcinoma with variable degrees of success. The development of immunotherapy with α-interferon or high-dose interleukin-2 is the best established treatment, and is associated with durable disease control. Although the lack of defined antigens in renal cell carcinoma has hindered more specific vaccine development, research regarding vaccination therapy has been of special interest for the treatment of renal cell carcinoma for more than 30 years. At present, there are three types of cell-based vaccines in renal cell carcinoma treatment: autologous tumor-cell vaccines, genetically modified tumor vaccines and dendritic cell-based vaccines. A further type is peptide-based vaccination with tumor-associated antigens as possible targets, such as carbonic anhydrase IX, survivin and telomerase that are overexpressed in renal cell carcinoma. In the present article, we review data from completed clinical trials of vaccine therapy, and discuss future trials to assess the current knowledge and future role of vaccine therapy for renal cell carcinoma in the era of recently developed targeted therapy.
Collapse
Affiliation(s)
- Kazuhiro Yoshimura
- Department of Urology, Faculty of Medicine, Kinki University, Osaka, Japan.
| | | |
Collapse
|
6
|
Brookman-May S, Burger M, Wieland WF, Rössler W, May M, Denzinger S. Vaccination therapy in renal cell carcinoma: current position and future options in metastatic and localized disease. Expert Rev Vaccines 2011; 10:837-52. [PMID: 21692704 DOI: 10.1586/erv.11.64] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
As renal cell carcinoma represents one of the most immune-responsive cancers, immunotherapy exhibits a suitable treatment basis. Beside nonspecific stimulation via cytokines, passive specific and active immunotherapy are also appropriate options to recognize and destroy tumor cells. For more than 30 years, research regarding vaccination therapy has been of special interest for the treatment of renal cell carcinoma. However, apart from occasional promising results in Phase I and II trials, vaccination therapy is still considered experimental in this tumor entity, especially owing to missing results from Phase III trials demonstrating clinical efficacy. In the present article, we review data from completed clinical trials of vaccination therapy and also discuss scheduled future trials, in order to assess the current position and possible future fields of application of vaccination therapy in renal cell carcinoma in the era of recently developed targeted therapies.
Collapse
Affiliation(s)
- Sabine Brookman-May
- University of Regensburg, Department of Urology, Caritas St. Josef Medical Center, Landshuter Strasse 65, 93053 Regensburg, Germany.
| | | | | | | | | | | |
Collapse
|
7
|
Alpizar YA, Chain B, Collins MK, Greenwood J, Katz D, Stauss HJ, Mitchison NA. Ten years of progress in vaccination against cancer: the need to counteract cancer evasion by dual targeting in future therapies. Cancer Immunol Immunother 2011; 60:1127-35. [PMID: 21479639 PMCID: PMC11028423 DOI: 10.1007/s00262-011-0985-7] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/09/2010] [Accepted: 01/31/2011] [Indexed: 12/19/2022]
Abstract
Although cancer immunology has made vigorous progress over the last decade, its future remains uncertain. Tumors have clearly proved subject to immune surveillance, leading to antigenic editing, and means of activating both T and B arms of the immune system have been devised. Therapeutic vaccination and monoclonal antibody therapy have so far proved disappointing, because tumors prove adept at evasion from immune control. Dual targeting could well counteract evasion, provided that the two targets are independent and are attacked simultaneously. This stage has nearly but not quite been reached in several forms of immunotherapy, particularly of B-cell cancers, although such treatment also carries hazards.
Collapse
Affiliation(s)
| | - Benjamin Chain
- Division of Infection and Immunity, University College London (UCL), London, UK
| | - Mary K. Collins
- Division of Infection and Immunity, University College London (UCL), London, UK
| | - John Greenwood
- UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL UK
| | - David Katz
- Division of Infection and Immunity, University College London (UCL), London, UK
| | - Hans J. Stauss
- UCL Institute of Ophthalmology, 11-43 Bath Street, London, EC1V 9EL UK
| | | |
Collapse
|
8
|
Gene carriers and transfection systems used in the recombination of dendritic cells for effective cancer immunotherapy. Clin Dev Immunol 2010; 2010:565643. [PMID: 21197274 PMCID: PMC3010860 DOI: 10.1155/2010/565643] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2010] [Accepted: 10/28/2010] [Indexed: 12/11/2022]
Abstract
Dendritic cells (DCs) are the most potent antigen-presenting cells. They play a vital role in the initiation of immune response by presenting antigens to T cells and followed by induction of T-cell response. Reported research in animal studies indicated that vaccine immunity could be a promising alternative therapy for cancer patients. However, broad clinical utility has not been achieved yet, owing to the low transfection efficiency of DCs. Therefore, it is essential to improve the transfection efficiency of DC-based vaccination in immunotherapy. In several studies, DCs were genetically engineered by tumor-associated antigens or by immune molecules such as costimulatory molecules, cytokines, and chemokines. Encouraging results have been achieved in cancer treatment using various animal models. This paper describes the recent progress in gene delivery systems including viral vectors and nonviral carriers for DC-based genetically engineered vaccines. The reverse and three-dimensional transfection systems developed in DCs are also discussed.
Collapse
|
9
|
Westermann J, Flörcken A, Willimsky G, van Lessen A, Kopp J, Takvorian A, Jöhrens K, Lukowsky A, Schönemann C, Sawitzki B, Pohla H, Frank R, Dörken B, Schendel DJ, Blankenstein T, Pezzutto A. Allogeneic gene-modified tumor cells (RCC-26/IL-7/CD80) as a vaccine in patients with metastatic renal cell cancer: a clinical phase-I study. Gene Ther 2010; 18:354-63. [DOI: 10.1038/gt.2010.143] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
10
|
Herbert N, Haferkamp A, Schmitz-Winnenthal HF, Zöller M. Concomitant tumor and autoantigen vaccination supports renal cell carcinoma rejection. THE JOURNAL OF IMMUNOLOGY 2010; 185:902-16. [PMID: 20548033 DOI: 10.4049/jimmunol.0902683] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
Efficient tumor vaccination frequently requires adjuvant. Concomitant induction of an autoimmune response is discussed as a means to strengthen a weak tumor Ag-specific response. We asked whether the efficacy of dendritic cell (DC) vaccination with the renal cell carcinoma Ags MAGE-A9 (MAGE9) and G250 could be strengthened by covaccination with the renal cell carcinoma autoantigen GOLGA4. BALB/c mice were vaccinated with DC loaded with MHC class I-binding peptides of MAGE9 or G250 or tumor lysate, which sufficed for rejection of low-dose RENCA-MAGE9 and RENCA-G250 tumor grafts, but only retarded tumor growth at 200 times the tumor dose at which 100% of animals will develop a tumor. Instead, 75-100% of mice prevaccinated concomitantly with Salmonella typhimurium transformed with GOLGA4 cDNA in a eukaryotic expression vector rejected 200 times the tumor dose at which 100% of animals will develop tumor. In a therapeutic setting, the survival rate increased from 20-40% by covaccination with S. typhimurium-GOLGA4. Autoantigen covaccination significantly strengthened tumor Ag-specific CD4(+) and CD8(+) T cell expansion, particularly in peptide-loaded DC-vaccinated mice. Covaccination was accompanied by an increase in inflammatory cytokines, boosted IL-12 and IFN-gamma expression, and promoted a high tumor Ag-specific CTL response. Concomitant autoantigen vaccination also supported CCR6, CXCR3, and CXCR4 upregulation and T cell recruitment into the tumor. It did not affect regulatory T cells, but slightly increased myeloid-derived suppressor cells. Thus, tumor cell eradication was efficiently strengthened by concomitant induction of an immune response against a tumor Ag and an autoantigen expressed by the tumor cell. Activation of autoantigen-specific Th cells strongly supports tumor-specific Th cells and thereby CTL activation.
Collapse
Affiliation(s)
- Nicolás Herbert
- Department of Tumor Cell Biology, University Hospital of Surgery, University of Heidelberg, Germany
| | | | | | | |
Collapse
|
11
|
Buchner A, Pohla H, Willimsky G, Frankenberger B, Frank R, Baur-Melnyk A, Siebels M, Stief CG, Hofstetter A, Kopp J, Pezzutto A, Blankenstein T, Oberneder R, Schendel DJ. Phase 1 trial of allogeneic gene-modified tumor cell vaccine RCC-26/CD80/IL-2 in patients with metastatic renal cell carcinoma. Hum Gene Ther 2010; 21:285-97. [PMID: 19788391 DOI: 10.1089/hum.2008.192] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Preclinical studies showed that the allogeneic tumor cell line RCC-26 displayed natural immunogenic potential that was enhanced through expression of CD80 costimulatory molecules and secretion of interleukin-2. Here we report the study of RCC-26/CD80/IL-2 cells in a phase 1 vaccine trial of renal cell carcinoma patients with metastatic disease (mRCC). Fifteen patients of the HLA-A*0201 allotype, with at least one metastatic lesion, were included. Irradiated vaccine cells were applied in increasing doses of 2.5, 10, and 40 x 10(6) cells over 22 weeks. Primary study parameters included safety and toxicity. Sequential blood samples were analyzed by interferon-gamma enzyme-linked immunospot assays to detect tumor antigen-associated (TAA) effector cells. The vaccine was well tolerated and the designated vaccination course was completed in 9 of 15 patients. Neither vaccine-induced autoimmunity nor systemic side effects were observed. Delayed-type hypersensitivity skin reactions were detected in 11 of 12 evaluated patients and were particularly strong in patients with prolonged survival. In parallel, vaccine-induced immune responses against vaccine or overexpressed TAA were detected in 9 of 12 evaluated patients. No tumor regressions occurred according to RECIST (Response Evaluation Criteria in Solid Tumors) criteria; however, median time to progression was 5.3 months and median survival was 15.6 months, indicating substantial disease stabilization. We conclude that vaccine use was safe and feasible in mRCC. Clinical benefits were limited in these patients with advanced disease; however, immune monitoring revealed vaccine-induced responses against multiple TAAs in the majority of study participants. These results suggest that this vaccine could be useful in combination therapies and/or minimal residual disease.
Collapse
Affiliation(s)
- Alexander Buchner
- Department of Urology, Ludwig Maximilian University of Munich, 81377 Munich, Germany
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Vaccine Therapy in Patients with Renal Cell Carcinoma. Eur Urol 2009; 55:1333-42. [DOI: 10.1016/j.eururo.2009.01.043] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/29/2008] [Accepted: 01/22/2009] [Indexed: 11/20/2022]
|
13
|
Geiger C, Nössner E, Frankenberger B, Falk CS, Pohla H, Schendel DJ. Harnessing innate and adaptive immunity for adoptive cell therapy of renal cell carcinoma. J Mol Med (Berl) 2009; 87:595-612. [PMID: 19271159 DOI: 10.1007/s00109-009-0455-2] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/13/2008] [Revised: 02/18/2009] [Accepted: 02/20/2009] [Indexed: 12/22/2022]
Abstract
The development of immunotherapies for renal cell carcinoma (RCC) has been the subject of research for several decades. In addition to cytokine therapy, the benefit of various adoptive cell therapies has again come into focus in the past several years. Nevertheless, success in fighting this immunogenic tumor is still disappointing. RCC can attract a multitude of different effector cells of both the innate and adaptive immune system, including natural killer (NK) cells, gammadelta T cells, NK-like T cells, peptide-specific T cells, dendritic cells (DC), and regulatory T cells (Tregs). Based on intensive research on the biology and function of different immune cells, we now understand that individual cell types do not act in isolation but function within a complex network of intercellular interactions. These interactions play a pivotal role in the efficient activation and function of effector cells, which is a prerequisite for successful tumor elimination. This review provides a current overview of the diversity of effector cells having the capacity to recognize RCC. Aspects of the functions and anti-tumor properties that make them attractive candidates for adoptive cell therapies, as well as experience in clinical application are discussed. Improved knowledge of the biology of this immune network may help us to effectively harness various effector cells, placing us in a better position to develop new therapeutic strategies to successfully fight RCC.
Collapse
Affiliation(s)
- Christiane Geiger
- Institut für Molekulare Immunologie, Helmholtz Zentrum München, Deutsches Forschungszentrum für Gesundheit und Umwelt, Germany.
| | | | | | | | | | | |
Collapse
|
14
|
Butts CL, Bowers E, Horn JC, Shukair SA, Belyavskaya E, Tonelli L, Sternberg EM. Inhibitory effects of progesterone differ in dendritic cells from female and male rodents. ACTA ACUST UNITED AC 2009; 5:434-47. [PMID: 19108816 DOI: 10.1016/j.genm.2008.11.001] [Citation(s) in RCA: 49] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/02/2008] [Indexed: 10/21/2022]
Abstract
BACKGROUND Steroid hormones, such as progesterone, are known to have immunomodulatory effects. Our research group previously reported direct effects of progesterone on dendritic cells (DCs) from female rodents. Primarily affecting mature DC function, progesterone effects included inhibition of proinflammatory cytokine secretion, downregulation of cell surface marker (major histocompatibility complex class II, CD80) expression, and decreased T-cell proliferative capacity, and were likely mediated through progesterone receptor (PR) because the PR antagonist RU486 reversed these effects. OBJECTIVE The goal of this study was to assess differences in response to progesterone by DCs from female and male rodents. METHODS Using real-time reverse-transcriptase polymerase chain reaction, transcriptional expression of steroid hormone receptors was measured in immature bone marrow-derived DCs (BMDCs) from male and female rats. Expression of steroid hormone receptor protein was also assessed in these cells using flow cytometry and fluorescence microscopy. To evaluate functional differences between BMDCs from female and male rats in response to the steroid hormone progesterone, levels of secreted cytokines were measured using enzyme-linked immunosorbent assay. RESULTS Higher numbers of immature BMDCs from males expressed glucocorticoid receptor (GR) and androgen receptor (AR) proteins compared with females (males vs females, mean [SD]: GR = 68.75 [7.27] vs 43.61 [13.97], P = NS; AR = 75.99 [15.38] vs 8.25 [1.88], P = 0.002), whereas higher numbers of immature BMDCs from females expressed PR protein compared with males (females vs males: PR = 74.19 [12.11] vs 14.14 [4.55], P = 0.043). These differences were not found at the level of transcription (females vs males: GR = 0.088 vs 0.073, P = NS; AR = 0.076 vs 0.069, P = NS; PR = 0.075 vs 0.065, P = NS). Compared with those from females, mature BMDCs from males produced higher quantities of cytokines (tumor necrosis factor-alpha [TNF-alpha], interleukin [IL]-1beta, IL-10) (females vs males: TNF-alpha = 920.0 [79.25] vs 1100.61 [107.97], P = NS; IL-1beta = 146.60 [38.04] vs 191.10 [10.47], P = NS; IL-10 = 167.25 [4.50] vs 206.15 [23.48], P = NS). Conversely, BMDCs from females were more sensitive to progesterone, as indicated by a more dramatic reduction in proinflammatory cytokine secretion (females vs males, highest concentration of progesterone: TNF-alpha = 268.94 [28.59] vs 589.91 [100.98], P = 0.04; IL-1beta = 119.50 [10.32] vs 154.35 [6.22], P = NS). CONCLUSIONS These findings suggest that progesterone effects on DCs in rodents may be more pronounced in females than in males, and this is likely due to differences in PR protein expression. Our observations may help elucidate disparities in the incidence and severity of autoimmune disorders between females and males, and the role specific steroid hormones play in regulating immune responses.
Collapse
Affiliation(s)
- Cherié L Butts
- Section on Neuroendocrine Immunology & Behavior, National Institute of Mental Health/NIH, Bethesda, Maryland 20892, USA
| | | | | | | | | | | | | |
Collapse
|
15
|
Havranek EG, Labarthe MC, Ward S, Anderson CJ, Whelan MA, Pandha H. A novel murine model of allogeneic vaccination against renal cancer. BJU Int 2008; 101:1165-9. [PMID: 18399831 DOI: 10.1111/j.1464-410x.2008.07532.x] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
OBJECTIVES To develop a murine model for whole-cell allogeneic vaccination in renal cancer, as such vaccines aim to direct immune responses against patient tumour cells, due to shared antigens between the vaccine and tumour cells. MATERIALS AND METHODS A novel murine renal cell line, allogeneic to BALB/c, was developed from a C57BL/6 mouse by primary cell culture (RVIK). It was immortalized by HPV16 E6/E7 and transfected with ras in an attempt to improve its immunogenicity. The cell line was characterized and tested as a vaccine in a BALB/c tumour-protection model after subsequent tumour challenge with autologous RenCa tumour cells. RESULTS RVIK alone, with no ras induced cross-reactive immunity, providing a valid non-tumorigenic allogeneic whole-cell vaccine model for renal cancer. Ras transfection per se did not improve RVIK immunity. CONCLUSIONS RVIK is a novel immunogenic murine renal epithelial cell line, which confers protection when used as an allogeneic vaccine. It provides proof of principle for the effectiveness of allogeneic whole-cell vaccines and may therefore form the basis of a useful model of allogeneic vaccination to further optimize vaccination schedules, formulation and adjuvants for a clinical setting.
Collapse
|
16
|
Tang XD, Wan Y, Chen L, Chen T, Yu ST, Xiong Z, Fang DC, Liang GP, Yang SM. H-2Kb-restricted CTL epitopes from mouse heparanase elicit an antitumor immune response in vivo. Cancer Res 2008; 68:1529-37. [PMID: 18316618 DOI: 10.1158/0008-5472.can-07-5965] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The identification of CTL epitopes from tumor antigens is very important for the development of peptide-based, cancer-specific immunotherapy. Heparanase is broadly expressed in various advanced tumors and can serve as a universal tumor-associated antigen. Although several epitopes of heparanase antigen are known in humans, the corresponding knowledge in mice is still rather limited. The present study was designed to predict and identify the CTL epitopes in the mouse heparanase protein. For this purpose, H-2K(b)-restricted CTL epitopes were identified by using the following four-step procedure: (a) a computer-based epitope prediction from the amino acid sequence of mouse heparanase, (b) a peptide-binding assay to determine the affinity of the predicted epitopes with the H-2K(b) molecule, (c) the testing of the induction of CTLs toward various carcinoma cells expressing heparanase antigens and H-2K(b), and (d) the induction of immunoprotection and immunotherapy in vivo. The results showed that, of the tested peptides, effectors induced by peptides of mouse heparanase at residue positions 398 to 405 (LSLLFKKL; mHpa398) and 519 to 526 (FSYGFFVI; mHpa519) lysed three kinds of carcinoma cells expressing both heparanase and H-2K(b) (B16 melanoma cells, EL-4 lymphoma cells, and Lewis lung cancer cells). In vivo experiments indicated that mHpa398 and mHpa519 peptides offered the possibility of not only immunizing against tumors but also treating tumor-bearing hosts successfully. Our results suggest that the mHpa398 and mHpa519 peptides are novel H-2K(b)-restricted CTL epitopes capable of inducing heparanase-specific CTLs in vitro and in vivo. These epitopes may serve as valuable tools for the preclinical evaluation of vaccination strategies.
Collapse
Affiliation(s)
- Xu-Dong Tang
- Institute of Gastroenterology, Southwest Hospital, Third Military Medical University, Chongqing, PR China
| | | | | | | | | | | | | | | | | |
Collapse
|
17
|
Uemura H, De Velasco MA. Tumor vaccines in renal cell carcinoma. World J Urol 2008; 26:147-54. [DOI: 10.1007/s00345-008-0246-3] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2007] [Accepted: 02/19/2008] [Indexed: 11/30/2022] Open
|
18
|
Cai YG, Fang DC, Chen L, Tang XD, Chen T, Yu ST, Luo YH, Xiong Z, Wang DX, Yang SM. Dendritic cells reconstituted with a human heparanase gene induce potent cytotoxic T-cell responses against gastric tumor cells in vitro. Tumour Biol 2007; 28:238-46. [PMID: 17717429 DOI: 10.1159/000107584] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/25/2007] [Accepted: 04/04/2007] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND AND AIMS Dendritic cell-based tumor vaccination is a promising approach in the treatment of cancer. Strategies to modify dendritic cells (DCs) with tumor-associated antigens (TAAs) can elicit specific immune responses against tumors. Heparanase is overexpressed in gastric cancer, especially in invasive and metastatic cells, but is downregulated in differential normal tissue. Therefore, heparanase is a potential target in immunotherapy for patients with advanced gastric cancer who are not candidates for surgery. The present paper was designed to investigate the immune response of a heparanase gene-modified DC-based vaccine against gastric cancer cell lines in vitro. METHODS DCs from peripheral blood mononuclear cells of healthy HLA-A2-positive donors were transfected with recombinant adenovirus containing the full-length cDNA of heparanase (rAd-Hpa) to generate heparanase gene-modified DC vaccine. T lymphocytes from the same donors were repeatedly activated by genetically modified DC vaccine to generate heparanase-specific cytotoxicity T lymphocytes (CTLs). CTL-mediated cell lysis of gastric cancer cells lines (KATO-III and SGC-7901) was analyzed in vitro by a standard (51)Cr releasing assay. IFN-gamma secretion was measured by ELISA in heparanase-specific CTLs cocultured with those gastric cancer cell lines. RESULTS Our results showed that the expression of heparanase in DCs transfected with rAd-Hpa was significantly increased. Furthermore, DCs transfected with rAd-Hpa could induce heparanase-specific CTLs against HLA-matched and heparanase-positive gastric cancer cells in vitro, while there were no killing effects on autologous lymphocytes. Meanwhile, these rAd-Hpa-modified DCs could increase IFN-gamma secretion of effector cells when cocultured with KATO-III cells. CONCLUSIONS These findings demonstrate for the first time that the transduction of DCs with rAd-Hpa can induce CTLs that specifically lyse heparanase-positive gastric cancer cells and increase IFN-gamma secretion in an MHC-restricted fashion. Heparanase gene-modified DC vaccine offers a great opportunity for immunotherapy in patients with advanced gastric cancer and possibly also with other malignancies.
Collapse
Affiliation(s)
- Yong-Guo Cai
- Institute of Gastroenterology of PLA, Southwest Hospital, Third Military Medical University, Chongqing, China
| | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Frankenberger B, Noessner E, Schendel DJ. Immune suppression in renal cell carcinoma. Semin Cancer Biol 2007; 17:330-43. [PMID: 17656104 DOI: 10.1016/j.semcancer.2007.06.004] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2007] [Revised: 04/10/2007] [Accepted: 06/07/2007] [Indexed: 12/13/2022]
Abstract
The clear evidence that tumor-infiltrating lymphocytes with anti-tumor activity exist in situ raises the question why renal cell carcinomas (RCCs) progress in vivo. A complex array of factors and pathways has been identified that impinges on innate and adaptive effector cells thereby inhibiting their activity against RCCs. The current picture of suppressive mechanisms that contribute to the failure of the immune system to control RCCs is reviewed here. Understanding these complex host-tumor interactions has broad implications for successful application of cytokine therapy and other forms of immunotherapy for RCC.
Collapse
Affiliation(s)
- Bernhard Frankenberger
- Institute of Molecular Immunology, GSF-National Research Center for Environment and Health, Marchioninistrasse 25, 81377 Munich, Germany
| | | | | |
Collapse
|
20
|
Ranieri E, Gigante M, Storkus WJ, Gesualdo L. Translational mini-review series on vaccines: Dendritic cell-based vaccines in renal cancer. Clin Exp Immunol 2007; 147:395-400. [PMID: 17302887 PMCID: PMC1810504 DOI: 10.1111/j.1365-2249.2006.03305.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/03/2023] Open
Abstract
Renal cancer is a relatively uncommon solid tumor, accounting for about 3% of all adult malignancies, however this rate incidence is rising. The most common histological renal cell carcinoma (RCC) subtype is clear cell carcinoma that makes up approximately 70-80% of all renal neoplasms and appears to be the only histological subtype that is responsive to immunotherapeutic approaches with any consistency. Therefore, it has been hypothesized that immune-mediated mechanisms play important roles in limiting tumor growth and that dendritic cells (DC), the most potent APC in the body, and T cells are the dominant effector cells that regulate tumor progression in situ. In this context, the development of clinically effective DC-based vaccines is a major focus for active specific immunotherapy in renal cancer. In the current review we have not focused on the results of recently published RCC clinical trials, as several excellent reviews have already performed this function. Instead, we turned our attention to how the perception and practical application of DC-based vaccinations are evolving.
Collapse
Affiliation(s)
- E Ranieri
- Clinical Pathology, Department of Biomedical Sciences, University of Foggia, Italy.
| | | | | | | |
Collapse
|
21
|
Zobywalski A, Javorovic M, Frankenberger B, Pohla H, Kremmer E, Bigalke I, Schendel DJ. Generation of clinical grade dendritic cells with capacity to produce biologically active IL-12p70. J Transl Med 2007; 5:18. [PMID: 17430585 PMCID: PMC1858682 DOI: 10.1186/1479-5876-5-18] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/13/2006] [Accepted: 04/12/2007] [Indexed: 02/07/2023] Open
Abstract
Background For optimal T cell activation it is desirable that dendritic cells (DCs) display peptides within MHC molecules as signal 1, costimulatory molecules as signal 2 and, in addition, produce IL-12p70 as signal 3. IL-12p70 polarizes T cell responses towards CD4+ T helper 1 cells, which then support the development of CD8+ cytotoxic T lymphocytes. We therefore developed new maturation cocktails allowing DCs to produce biologically active IL-12p70 for large-scale cancer vaccine development. Methods After elutriation of leukapheresis products in a closed bag system, enriched monocytes were cultured with GM-CSF and IL-4 for six days to generate immature DCs that were then matured with cocktails, containing cytokines, interferon-gamma, prostaglandin E2, and a ligand for Toll-like receptor 8, with or without poly (I:C). Results Mature DCs expressed appropriate maturation markers and the lymph node homing chemokine receptor, CCR7. They retained full maturity after culture for two days without maturation cocktails and following cryopreservation. TLR ligand stimulation induced DCs capable of secreting IL-12p70 in primary cultures and after one day of coculture with CD40L-expressing fibroblasts, mimicking an encounter with T cells. DCs matured with our new cocktails containing TLR8 ligand, with or without poly (I:C), induced alloresponses and stimulated virus-specific T cells after peptide-pulsing. DCs matured in cocktails containing TLR8 ligand without poly (I:C) could also be loaded with RNA as a source of antigen, whereas DCs matured in cocktails containing poly (I:C) were unable to express proteins following RNA transfer by electroporation. Conclusion Our new maturation cocktails allowed easy DC harvesting, stable maturation and substantial recoveries of mature DCs after cryopreservation. Our procedure for generating DCs is easily adaptable for GMP-compliance and yields IL-12p70-secreting DCs suitable for development of cancer vaccines using peptides or RNA as sources of immunizing antigens.
Collapse
Affiliation(s)
- Anke Zobywalski
- Institute of Molecular Immunology, GSF – National Research Center for Environment and Health, Munich, Germany
| | - Miran Javorovic
- Institute of Molecular Immunology, GSF – National Research Center for Environment and Health, Munich, Germany
| | - Bernhard Frankenberger
- Institute of Molecular Immunology, GSF – National Research Center for Environment and Health, Munich, Germany
| | - Heike Pohla
- Laboratory of Tumor Immunology, Ludwig Maximilian University, Munich, Germany
| | - Elisabeth Kremmer
- Institute of Molecular Immunology, GSF – National Research Center for Environment and Health, Munich, Germany
| | - Iris Bigalke
- Institute of Molecular Immunology, GSF – National Research Center for Environment and Health, Munich, Germany
| | - Dolores J Schendel
- Institute of Molecular Immunology, GSF – National Research Center for Environment and Health, Munich, Germany
- Clinical Cooperation Group "Immune Monitoring", GSF – National Research Center for Environment and Health, Munich, Germany
| |
Collapse
|
22
|
Abstract
Dendritic cell (DC) vaccines are an important experimental immunotherapy for renal cell carcinomas. DC vaccines have proven safe, but only minimal clinical efficacy has been observed to date. DC vaccine strategies reflect the continually evolving understanding of DC biology. The use of mature DCs is particularly important to avoid the induction of regulatory T cells. Better defined sources of immunizing antigens and more efficient antigen-loading will contribute to DC vaccines of better quality. Improved clinical efficacy may also be achieved using DCs that secrete biologically active IL-12, which fosters innate immunity and polarizes T helper type 1 responses that contribute to optimal antitumor immunity. Furthermore, combination therapies that treat systemic immune suppression will be crucial for obtaining improved clinical responses to DC vaccines in patients with advanced disease.
Collapse
Affiliation(s)
- Dolores J Schendel
- GSF National Research Center for Environment and Health, Institute of Molecular Immunology and the Clinical Cooperation Group Immune Monitoring, Marchioninistrasse 25, 81377 Munich, Germany.
| |
Collapse
|
23
|
Gouttefangeas C, Stenzl A, Stevanović S, Rammensee HG. Immunotherapy of renal cell carcinoma. Cancer Immunol Immunother 2007; 56:117-28. [PMID: 16676181 PMCID: PMC11030119 DOI: 10.1007/s00262-006-0172-4] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2006] [Accepted: 04/10/2006] [Indexed: 01/02/2023]
Abstract
Carcinomas of the kidney generally have a poor prognosis and respond minimally to classical radiotherapy or chemotherapy. Immunotherapy constitutes an interesting alternative to these established forms of treatment, and indeed, cytokine-based therapies have been used for many years, leading to favorable clinical responses in a small subset of patients. During the past few years, immunotherapeutical trials targeting renal cell tumor-associated antigens have also been reported, with diverse passive or active approaches using antibodies or aimed at activating tumor-directed T lymphocytes. The following review presents the results and the progress made in the field, including classical cytokine treatments, non-myeloablative stem cell transplantation and antigen specific-based trials, with special focus on T-cell studies. In consideration of the few specific molecular targets described so far for this tumor entity, current strategies which can lead to the identification of new relevant antigens will be discussed. Hopefully these will very soon contribute to an improvement in renal cell carcinoma specific immunotherapy and its evaluation.
Collapse
Affiliation(s)
- Cécile Gouttefangeas
- Institute for Cell Biology, Department of Immunology, Eberhard Karls University, Auf der Morgenstelle 15, 72076, Tubingen, Germany.
| | | | | | | |
Collapse
|
24
|
Tassi E, Facchinetti V, Seresini S, Borri A, Dell'antonio G, Garavaglia C, Casorati G, Protti MP. Peptidome from renal cell carcinoma contains antigens recognized by CD4+ T cells and shared among tumors of different histology. Clin Cancer Res 2006; 12:4949-57. [PMID: 16914584 DOI: 10.1158/1078-0432.ccr-06-0995] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Renal cell carcinoma (RCC) is considered immunogenic; nonetheless, rare tumor-associated antigens have been identified or are expressed in RCC. Peptidome (i.e., the total content of natural peptides of whole cells) from other tumors, such as melanoma, has proved to be immunogenic. The aims of this study were to determine whether peptidome from RCC is immunogenic and whether it contains tumor peptides shared among allogenic RCCs. EXPERIMENTAL DESIGN Autologous dendritic cells pulsed with RCC peptidome were used to activate in vitro CD4(+) T cells from healthy donors and a metastatic RCC patient. CD4(+) T-cell polyclonal lines and clones were characterized for tumor cell recognition by proliferation assay, killing activity, and cytokine secretion. RESULTS CD4(+) T-cell lines and clones recognized HLA-DR-matched allogenic RCC and, for the patient, the autologous tumor. RCC-reactive CD4(+) T cells showed a heterogeneous Th1 or Th0/Th2 pattern of cytokine secretion. Moreover, RCC-reactive CD4(+) T cells recognized also melanoma, colon carcinoma, cervical carcinoma, pancreas carcinoma, lung carcinoma, gastric carcinoma, and lymphoma cells but not autologous T-cell blasts. CONCLUSIONS Our results show that (a) the RCC peptidome contain antigens recognized by CD4(+) T cells and (b) shared among tumors of different histology and (c) it induces both Th1-type and Th2/Th0-type immune responses. These data support the use of the peptidome from allogenic RCC for specific immunotherapy in RCC and possibly in other neoplastic diseases. Moreover, the CD4(+) T-cell clones generated here are useful tools for tumor antigen identification.
Collapse
Affiliation(s)
- Elena Tassi
- Tumor Immunology Unit, DIBIT, Department of Oncology, Scientific Institute H. San Raffaele, Milan, Italy
| | | | | | | | | | | | | | | |
Collapse
|
25
|
Chen L, Liang GP, Tang XD, Chen T, Cai YG, Fang DC, Yu ST, Luo YH, Yang SM. In vitro anti-tumor immune response induced by dendritic cells transfected with hTERT recombinant adenovirus. Biochem Biophys Res Commun 2006; 351:927-34. [PMID: 17097054 DOI: 10.1016/j.bbrc.2006.10.165] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/22/2006] [Accepted: 10/25/2006] [Indexed: 02/04/2023]
Abstract
Transduction with recombinant, replication-defective adenoviral (Ad) vectors encoding a transgene is an efficient method for gene transfer into human dendritic cells (DC). Several studies have demonstrated that epitopes of the human telomerase reverse transcriptase gene (hTERT) can produce CTLs specific for malignant tumors. In this study, we constructed an hTERT recombinant adenovirus (rAd-hTERT) using DNA recombination. We found that human dendritic cells transduced with rAd-hTERT could effectively induce hTERT-specific cytotoxic T lymphocytes (CTLs) in vitro against various tumor cell lines, which were hTERT-positive and HLA-A2 matched. We also found that these hTERT-specific CTLs could not lyse autologous lymphocytes with low telomerase activity. Further studies revealed that rAd-hTERT transduced DCs could increase secretion of IFN-gamma by effector cells when they were co-cultured with hTERT-positive and HLA-A2 matched tumor cell lines. These data suggest that an hTERT vaccine can induce anti-tumor immunity against various tumor cells expressing hTERT in a HLA-A2-restricted fashion in vitro. The transduction of DCs with rAd-hTERT offers a great opportunity in cancer immunotherapy.
Collapse
Affiliation(s)
- Ling Chen
- Institute of Gastroenterology of PLA, Southwest Hospital, Third Military Medical University, Chongqing 400038, China
| | | | | | | | | | | | | | | | | |
Collapse
|
26
|
Geiger C, Regn S, Weinzierl A, Noessner E, Schendel DJ. A generic RNA-pulsed dendritic cell vaccine strategy for renal cell carcinoma. J Transl Med 2005; 3:29. [PMID: 16045799 PMCID: PMC1188079 DOI: 10.1186/1479-5876-3-29] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2005] [Accepted: 07/26/2005] [Indexed: 11/18/2022] Open
Abstract
We present a generic dendritic cell (DC) vaccine strategy for patients with renal cell carcinoma (RCC) based on the use of RNA as a source of multiplex tumor-associated antigens (TAAs). Instead of preparing RNA from tumor tissue of each individual RCC patient, we propose to substitute RNA prepared from a well characterized highly immunogenic RCC cell line (RCC-26 tumor cells) as a generic source of TAAs for loading of DCs. We demonstrate here that efficient RNA transfer can be achieved using lipofection of immature DCs, which are subsequently matured with a cytokine cocktail to express high levels of MHC and costimulatory molecules as well as the chemokine receptor CCR7. Neither RNA itself nor the lipid component impacted on the phenotype or the cytokine secretion of mature DCs. Following RNA loading, DCs derived from HLA-A2-positive donors were able to activate effector-memory cytotoxic T lymphocytes (CTLs) specific for a TAA ligand expressed by the RCC-26 cell line. CTL responses to RNA-loaded DCs reached levels comparable to those stimulated directly by the RCC-26 tumor cells. Furthermore, DCs expressing tumor cell RNA primed naïve T cells, yielding T cell lines with cytotoxicity and cytokine secretion after contact with RCC tumor cells. RCC-26 cell lines are available as good manufacturing practice (GMP)-certified reagents enabling this source of RNA to be easily standardized and adapted for clinical testing. In addition, well defined immune monitoring tools, including the use of RNA expressing B cell lines, are available. Thus, this DC vaccine strategy can be directly compared with an ongoing gene therapy trial using genetically-engineered variants of the RCC-26 cell line as vaccines for RCC patients with metastatic disease.
Collapse
Affiliation(s)
- Christiane Geiger
- Institute of Molecular Immunology, GSF-National Research Center for Environment and Health, Munich, Germany
| | - Sybille Regn
- Institute of Molecular Immunology, GSF-National Research Center for Environment and Health, Munich, Germany
| | - Andreas Weinzierl
- Institute of Cell Biology, Department of Immunology, University of Tübingen, Tübingen, Germany
| | - Elfriede Noessner
- Institute of Molecular Immunology, GSF-National Research Center for Environment and Health, Munich, Germany
| | - Dolores J Schendel
- Institute of Molecular Immunology, GSF-National Research Center for Environment and Health, Munich, Germany
| |
Collapse
|