101
|
Liechtenstein T, Dufait I, Lanna A, Breckpot K, Escors D. MODULATING CO-STIMULATION DURING ANTIGEN PRESENTATION TO ENHANCE CANCER IMMUNOTHERAPY. IMMUNOLOGY, ENDOCRINE & METABOLIC AGENTS IN MEDICINAL CHEMISTRY 2012; 12:224-235. [PMID: 22945252 PMCID: PMC3428911 DOI: 10.2174/187152212802001875] [Citation(s) in RCA: 36] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
One of the key roles of the immune system is the identification of potentially dangerous pathogens or tumour cells, and raising a wide range of mechanisms to eliminate them from the organism. One of these mechanisms is activation and expansion of antigen-specific cytotoxic T cells, after recognition of antigenic peptides on the surface of antigen presenting cells such as dendritic cells (DCs). However, DCs also process and present autoantigens. Therefore, antigen presentation has to occur in the appropriate context to either trigger immune responses or establishing immunological tolerance. This is achieved by co-stimulation of T cells during antigen presentation. Co-stimulation consists on the simultaneous binding of ligand-receptor molecules at the immunological synapse which will determine the type and extent of T cell responses. In addition, the type of cytokines/chemokines present during antigen presentation will influence the polarisation of T cell responses, whether they lead to tolerance, antibody responses or cytotoxicity. In this review, we will focus on approaches manipulating co-stimulation during antigen presentation, and the role of cytokine stimulation on effective T cell responses. More specifically, we will address the experimental strategies to interfere with negative co-stimulation such as that mediated by PD-L1 (Programmed cell death 1 ligand 1)/PD-1 (Programmed death 1) to enhance anti-tumour immunity.
Collapse
Affiliation(s)
- Therese Liechtenstein
- Division of Infection and Immunity. Rayne Institute. University College London. 5 University Street. WC1E 6JF. London. United Kingdom
| | - Ines Dufait
- Division of Infection and Immunity. Rayne Institute. University College London. 5 University Street. WC1E 6JF. London. United Kingdom
- Department of Physiology-Immunology. Medical School. Free University of Brussels. Laarbeeklaan 103. 1090 Jette. Belgium
| | - Alessio Lanna
- Division of Infection and Immunity. Rayne Institute. University College London. 5 University Street. WC1E 6JF. London. United Kingdom
| | - Karine Breckpot
- Department of Physiology-Immunology. Medical School. Free University of Brussels. Laarbeeklaan 103. 1090 Jette. Belgium
| | - David Escors
- Division of Infection and Immunity. Rayne Institute. University College London. 5 University Street. WC1E 6JF. London. United Kingdom
| |
Collapse
|
102
|
Li X, He Y. Caspase-2-dependent dendritic cell death, maturation, and priming of T cells in response to Brucella abortus infection. PLoS One 2012; 7:e43512. [PMID: 22927979 PMCID: PMC3425542 DOI: 10.1371/journal.pone.0043512] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/02/2012] [Accepted: 07/23/2012] [Indexed: 01/18/2023] Open
Abstract
Smooth virulent Brucella abortus strain 2308 (S2308) causes zoonotic brucellosis in cattle and humans. Rough B. abortus strain RB51, derived from S2308, is a live attenuated cattle vaccine strain licensed in the USA and many other countries. Our previous report indicated that RB51, but not S2308, induces a caspase-2-dependent apoptotic and necrotic macrophage cell death. Dendritic cells (DCs) are professional antigen presenting cells critical for bridging innate and adaptive immune responses. In contrast to Brucella-infected macrophages, here we report that S2308 induced higher levels of apoptotic and necrotic cell death in wild type bone marrow-derived DCs (WT BMDCs) than RB51. The RB51 and S2308-induced BMDC cell death was regulated by caspase-2, indicated by the minimal cell death in RB51 and S2308-infected BMDCs isolated from caspase-2 knockout mice (Casp2KO BMDCs). More S2308 bacteria were taken up by Casp2KO BMDCs than wild type BMDCs. Higher levels of S2308 and RB51 cells were found in infected Casp2KO BMDCs compared to infected WT BMDCs at different time points. RB51-infected wild type BMDCs were mature and activated as shown by significantly up-regulated expression of CD40, CD80, CD86, MHC-I, and MHC-II. RB51 induced the production of cytokines TNF-α, IL-6, IFN-γ and IL12/IL23p40 in infected BMDCs. RB51-infected WT BMDCs also stimulated the proliferation of CD4+ and CD8+ T cells compared to uninfected WT BMDCs. However, the maturation, activation, and cytokine secretion are significantly impaired in Casp2KO BMDCs infected with RB51 or Salmonella (control). S2308-infected WT and Casp2KO BMDCs were not activated and could not induce cytokine production. These results demonstrated that virulent smooth strain S2308 induced more apoptotic and necrotic dendritic cell death than live attenuated rough vaccine strain RB51; however, RB51, but not its parent strain S2308, induced caspase-2-mediated DC maturation, cytokine production, antigen presentation, and T cell priming.
Collapse
Affiliation(s)
- Xinna Li
- Unit for Laboratory Animal Medicine and Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
| | - Yongqun He
- Unit for Laboratory Animal Medicine and Department of Microbiology and Immunology, University of Michigan Medical School, Ann Arbor, Michigan, United States of America
- * E-mail:
| |
Collapse
|
103
|
Ellebaek E, Andersen MH, Svane IM, Straten PT. Immunotherapy for metastatic colorectal cancer: present status and new options. Scand J Gastroenterol 2012; 47:315-24. [PMID: 22214467 DOI: 10.3109/00365521.2012.640831] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Although no immunotherapeutic treatment is approved for colorectal cancer (CRC) patients, promising results from clinical trials suggest that several immunotherapeutic strategies may prove efficacious and applicable to this group of patients. This review describes the immunogenicity of CRC and presents the most interesting strategies investigated so far: cancer vaccination including antigen-defined vaccination and dendritic cell vaccination, chemo-immunotherapy, and adoptive cell transfer. Future treatment options as well as the possibility of combining existing therapies will be discussed along with the challenges presented by tumor escape mechanisms.
Collapse
Affiliation(s)
- Eva Ellebaek
- Department of Haematology, Center for Cancer Immune Therapy (CCIT), Copenhagen University Hospital Herlev, Herlev, Denmark.
| | | | | | | |
Collapse
|
104
|
Kumar N, Mishra J, Quazi SH. Training the Defense System for Modern-Day Warfare: The Horizons for Immunotherapy and Vaccines for Cancer. ACTA ACUST UNITED AC 2012; 1. [PMID: 25264543 DOI: 10.4172/2324-853x.1000e106] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Affiliation(s)
- Narendra Kumar
- Department of Pharmaceutical Sciences, College of Pharmacy Texas A &M Health Science Center Kingsville TX 78363
| | | | | |
Collapse
|
105
|
Yamanaka R, Kajiwara K. Dendritic cell vaccines. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2012; 746:187-200. [PMID: 22639169 DOI: 10.1007/978-1-4614-3146-6_15] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Despite progress in brain tumor therapy, the prognosis of malignant glioma patients remains dismal. Among the new treatments currently being investigated, immunotherapy is theoretically very attractive since it offers the potential for high tumor-specific cytotoxicity. Increasing numbers of reports demonstrate that systemic immunotherapy using dendritic cells is capable of inducing an antiglioma response. Therefore, dendritic cell-based immunotherapy could be a new treatment modality for patients with glioma. In this chapter, we will discuss the implications of these findings for glioma therapy, reviewing current literature on dendritic cell-based glioma immunotherapy. We will overview the role of dendritic cells in immunobiology, the central nervous system and tumor immunology, before outlining dendritic cell therapy results in clinical trials and future directions. Dendritic cell-based immunotherapy strategies appear promising as an approach to successfully induce an antitumor immune response in patients with glioma, where it seems to be safe and without major side effects. The development of methods for manipulating dendritic cells for the purpose of vaccination will enhance the clinical usefulness of these cells for biotherapy. Its efficacy should be further determined in randomized, controlled clinical trials.
Collapse
|
106
|
Kizjakina K, Bryson JM, Grandinetti G, Reineke TM. Cationic glycopolymers for the delivery of pDNA to human dermal fibroblasts and rat mesenchymal stem cells. Biomaterials 2011; 33:1851-62. [PMID: 22138032 DOI: 10.1016/j.biomaterials.2011.10.031] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/07/2011] [Accepted: 10/12/2011] [Indexed: 12/17/2022]
Abstract
Progenitor and pluripotent cell types offer promise as regenerative therapies but transfecting these sensitive cells has proven difficult. Herein, a series of linear trehalose-oligoethyleneamine "click" copolymers were synthesized and examined for their ability to deliver plasmid DNA (pDNA) to two progenitor cell types, human dermal fibroblasts (HDFn) and rat mesenchymal stem cells (RMSC). Seven polymer vehicle analogs were synthesized in which three parameters were systematically varied: the number of secondary amines (4-6) within the polymer repeat unit (Tr4(33), Tr5(30), and Tr6(32)), the end group functionalities [PEG (Tr4(128)PEG-a, Tr4(118)PEG-b), triphenyl (Tr4(107)-c), or azido (Tr4(99)-d)], and the molecular weight (degree of polymerization of about 30 or about 100) and the biological efficacy of these vehicles was compared to three controls: Lipofectamine 2000, JetPEI, and Glycofect. The trehalose polymers were all able to bind and compact pDNA polyplexes, and promote pDNA uptake and gene expression [luciferase and enhanced green fluorescent protein (EGFP)] with these primary cell types and the results varied significantly depending on the polymer structure. Interestingly, in both cell types, Tr4(33) and Tr5(30) yielded the highest luciferase gene expression. However, when comparing the number of cells transfected with a reporter plasmid encoding enhanced green fluorescent protein, Tr4(33) and Tr4(107)-c yielded the highest number of HDFn cells positive for EGFP. Interestingly, with RMSCs, all of the higher molecular weight analogs (Tr4(128)PEG-a, Tr4(118)PEG-b, Tr4(107)-c, Tr4(99)-d) yielded high percentages of cells positive for EGFP (30-40%).
Collapse
Affiliation(s)
- Karina Kizjakina
- Department of Chemistry and Macromolecules and Interfaces Institute, Virginia Tech, Blacksburg, VA 24061, USA
| | | | | | | |
Collapse
|
107
|
Abstract
The etiology and pathogenesis of gastric cancer (GC) are still elusive. Genetic factors, environmental factors and immune factors may play a role in the development of GC. While GC is progressing, tumor cells always induce immune cell apoptosis or immune system dysfunction to acquire immune privilege or immune evasion. It has been known that CD4+ T cells, CD8+ T cells, the B7 ligand family and dendritic cells are involved in these processes. GC immunotherapy, which can be antigen-specific or -nonspecific, has been an important part of adjuvant therapy. IL-2 is one of the most commonly used cytokines in GC-nonspecific immunotherapy and has been demonstrated to have certain curative effect. Administration of dendritic cell-based tumor vaccines will become a new approach for GC-specific immunotherapy.
Collapse
|
108
|
A mathematical model of tumor-immune interactions. J Theor Biol 2011; 294:56-73. [PMID: 22051568 DOI: 10.1016/j.jtbi.2011.10.027] [Citation(s) in RCA: 87] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/31/2010] [Revised: 07/01/2011] [Accepted: 10/19/2011] [Indexed: 12/31/2022]
Abstract
A mathematical model of the interactions between a growing tumor and the immune system is presented. The equations and parameters of the model are based on experimental and clinical results from published studies. The model includes the primary cell populations involved in effector T-cell mediated tumor killing: regulatory T cells, helper T cells, and dendritic cells. A key feature is the inclusion of multiple mechanisms of immunosuppression through the main cytokines and growth factors mediating the interactions between the cell populations. Decreased access of effector cells to the tumor interior with increasing tumor size is accounted for. The model is applied to tumors with different growth rates and antigenicities to gauge the relative importance of various immunosuppressive mechanisms. The most important factors leading to tumor escape are TGF-β-induced immunosuppression, conversion of helper T cells into regulatory T cells, and the limitation of immune cell access to the full tumor at large tumor sizes. The results suggest that for a given tumor growth rate, there is an optimal antigenicity maximizing the response of the immune system. Further increases in antigenicity result in increased immunosuppression, and therefore a decrease in tumor killing rate. This result may have implications for immunotherapies which modulate the effective antigenicity. Simulation of dendritic cell therapy with the model suggests that for some tumors, there is an optimal dose of transfused dendritic cells.
Collapse
|
109
|
Baek S, Kim CS, Kim SB, Kim YM, Kwon SW, Kim Y, Kim H, Lee H. Combination therapy of renal cell carcinoma or breast cancer patients with dendritic cell vaccine and IL-2: results from a phase I/II trial. J Transl Med 2011; 9:178. [PMID: 22013914 PMCID: PMC3213212 DOI: 10.1186/1479-5876-9-178] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/23/2011] [Accepted: 10/20/2011] [Indexed: 12/15/2022] Open
Abstract
Background Ten cancer patients (Six renal cell carcinoma and four breast cancer patients) were treated in a phase I/II study with a vaccine composed of autologous dendritic cells (DCs) and IL-2 to evaluate the DC vaccine-related toxicity and antigen-specific immune alteration. Methods Cancer patients were treated twice with autologous CD34+ hematopoietic stem cell-derived, GM-CSF/IFN-γ-differentiated DCs pulsed with autologous tumor lysate and KLH, by 4-week interval. Following each subcutaneous injection of therapeutic DCs, low-dose (200 MIU) IL-2 was introduced for 14 consecutive days as an immune adjuvant. To determine the DC vaccine-induced immunological alterations, the KLH-specific lymphocyte proliferation, number of IFN-γ secreting T cells (ELISPOT assay), NK activity and the cytokine modulation were measured. Results Cultured-DCs expressing HLA-DR, CD11c, CD83, and B7.1/B7.2 produced IL-12p70. After vaccination, the patients tolerated it. Clinical response was observed in one RCC patient as stable disease. However DC-vaccine related antigen-specific immune responses including peripheral blood lymphocyte proliferation and the number of IFN-r secreting cells were induced in six patients without clear correlation with clinical responses. Also NK activity was induced significantly in six patients after vaccination. DC vaccine-related decrease of TGF-β level or increase of IL-12p70 level and decline of CD4+CD25+ T cells were observed in three patients. However only in the RCC patient whose disease stabilized, combination of stimulatory as well as inhibitory immune alterations including induction of IFN-γ secreting T cell with reduction of CD4+ CD25+ T cell were correlated with clinical responses. Conclusion Data indicated that DC vaccine combined with IL-2 is well tolerated without major side effects. DC vaccine induced the specific immunity against introduced antigen. Combinatorial alterations of immunological parameters indicating antigen-specific immune induction along with reduction of inhibitory immunity were correlated with clinical responses in DC vaccine treated patients.
Collapse
Affiliation(s)
- Soyoung Baek
- Office of Biomedical Research, Samsung Medical Center, Sungkyunkwan University School of Medicine, Seoul, Korea
| | | | | | | | | | | | | | | |
Collapse
|
110
|
Rossowska J, Pajtasz-Piasecka E, Ryśnik O, Wojas J, Krawczenko A, Szyda A, Duś D. Generation of antitumor response by IL-2-transduced JAWS II dendritic cells. Immunobiology 2011; 216:1074-84. [DOI: 10.1016/j.imbio.2011.05.006] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/21/2010] [Revised: 03/16/2011] [Accepted: 05/07/2011] [Indexed: 11/16/2022]
|
111
|
Rong Y, Qin X, Jin D, Lou W, Wu L, Wang D, Wu W, Ni X, Mao Z, Kuang T, Zang YQ, Qin X. A phase I pilot trial of MUC1-peptide-pulsed dendritic cells in the treatment of advanced pancreatic cancer. Clin Exp Med 2011; 12:173-80. [PMID: 21932124 DOI: 10.1007/s10238-011-0159-0] [Citation(s) in RCA: 63] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/02/2011] [Accepted: 08/29/2011] [Indexed: 02/06/2023]
Abstract
The objectives of this study were to assess the toxicity and immunological response induced by the intra-dermal (i.d.) administration of MUC1-peptide-pulsed dendritic cells (DCs) in advanced pancreatic cancer patients. Patients with recurrent lesions or metastasis after surgery, and immunohistochemistry positive for MUC1 were treated in cohorts that received 3-6 × 10(6) DCs i.d. for three or four vaccines. Each vaccine was composed of autologus DCs pulsed with MUC1-peptide. Peripheral blood mononuclear cells (PBMCs) that harvested 2 weeks after the second immunization were compared with PBMCs obtained before treatment for immunological response. Serial ELISPOT assays of PBMCs for antitumor reactivity were performed. Three patients received all four vaccines, and four patients received three vaccines. These patients were evaluable for toxicity and immunological monitoring. There were no grade 3 or 4 toxicities associated with the vaccines or major evidence of autoimmunity. Interferon-γ and granzyme B ELISPOT assay reactivity increased significantly in 2 of 7 patients (P < 0.05). The administration of MUC1-peptide-pulsed DCs is non-toxic and capable of inducing immunological response to tumor antigen MUC1 in advanced pancreatic cancer patients. Additional studies are necessary to improve tumor rejection responses.
Collapse
Affiliation(s)
- Yefei Rong
- Pancreatic Cancer Group, General Surgery Department, Zhongshan Hospital, Fudan University, Shanghai, China
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
112
|
Cancer vaccines. Any future? Arch Immunol Ther Exp (Warsz) 2011; 59:249-59. [PMID: 21644030 DOI: 10.1007/s00005-011-0129-y] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2010] [Accepted: 02/02/2011] [Indexed: 12/22/2022]
Abstract
The idea that vaccination can be used to fight cancer is not new. Approximately 100 years ago, researchers attempted to stimulate a tumor-specific, therapeutic immune response to tumors by injecting patients with cells and extracts from their own tumors, or tumors of the same type from different individuals. During the last decade, great efforts have been made to develop immunotherapeutic approaches for the treatment of malignant diseases as alternatives to traditional chemo- and radiotherapy. A quintessential goal of immunotherapy in cancer is treatment with vaccines that elicit potent anti-tumor immune responses without side effects. In this article, we have attempted to review some of the most problematic issues facing the development of cancer vaccines. With the prospect of immunosuppression, an ill-designed cancer vaccine can be more harmful than a no-benefit therapy. We have noted that "immunoediting" and "immunodominance" are the premier setbacks in peptide-based vaccines and therefore it appears necessary not only to manipulate the activity of a vast number of principal components but also to finely tune their concentrations in time and space. In the face of all these quandaries, it is at least doubtful that any reliable anti-cancer vaccine strategy will emerge in the near future.
Collapse
|
113
|
Optimizing DC vaccination by combination with oncolytic adenovirus coexpressing IL-12 and GM-CSF. Mol Ther 2011; 19:1558-68. [PMID: 21468000 PMCID: PMC3149171 DOI: 10.1038/mt.2011.29] [Citation(s) in RCA: 71] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
Dendritic cell (DC)-based vaccination is a promising strategy for cancer immunotherapy. However, clinical trials have indicated that immunosuppressive microenvironments induced by tumors profoundly suppress antitumor immunity and inhibit vaccine efficacy, resulting in insufficient reduction of tumor burdens. To overcome these obstacles and enhance the efficiency of DC vaccination, we generated interleukin (IL)-12- and granulocyte-macrophage colony-stimulating factor (GM-CSF)-coexpressing oncolytic adenovirus (Ad-ΔB7/IL12/GMCSF) as suitable therapeutic adjuvant to eliminate immune suppression and promote DC function. By treating tumors with Ad-ΔB7/IL12/GMCSF prior to DC vaccination, DCs elicited greater antitumor effects than in response to either treatment alone. DC migration to draining lymph nodes (DLNs) dramatically increased in mice treated with the combination therapy. This result was associated with upregulation of CC-chemokine ligand 21 (CCL21+) lymphatics in tumors treated with Ad-ΔB7/IL12/GMCSF. Moreover, the proportion of CD4+CD25+ T-cells and vascular endothelial growth factor (VEGF) expression was decreased in mice treated with the combination therapy. Furthermore, combination therapy using immature DCs also showed effective antitumor effects when combined with Ad-ΔB7/IL12/GMCSF. The combination therapy had a remarkable therapeutic efficacy on large tumors. Taken together, oncolytic adenovirus coexpressing IL-12 and GM-CSF in combination with DC vaccination has synergistic antitumor effects and can act as a potent adjuvant for promoting and optimizing DC vaccination.
Collapse
|
114
|
Abstract
Most cancers remain incurable. Introduction of novel therapeutic methods, including new cytostatic regimens and targeted therapies, such as monoclonal antibodies and tyrosine kinase inhibitors, have increased remission rates as well as improved patient survival, but the ability to cure many cancer patients remains elusive. It is thus necessary to further develop alternative strategies to improve patient prognosis. The majority of patients who respond to induction therapy inevitably relapse, mainly because of the proliferation of residual malignant cells that have escaped control by induction chemotherapy. Therefore the eradication of minimal residual disease may be crucial to prevent a relapse and achieve a long-term remission. It seems that an advantageous treatment option may be cellular immunotherapy with dendritic-cell vaccines which might induce long-term specific anticancer responses with immune memory cells, which could contribute to effective and lasting elimination of malignant cells.
Collapse
Affiliation(s)
- J Rolinski
- Department of Clinical Immunology, Medical University of Lublin, Clinic Hospital SPSK-4, Lublin, Poland.
| | | |
Collapse
|
115
|
Blank F, Stumbles P, von Garnier C. Opportunities and challenges of the pulmonary route for vaccination. Expert Opin Drug Deliv 2011; 8:547-63. [PMID: 21438741 DOI: 10.1517/17425247.2011.565326] [Citation(s) in RCA: 41] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
INTRODUCTION The respiratory tract is an attractive target for the delivery of vaccine antigens. Potential advantages of drug delivery by means of the pulmonary route include accessibility, non-invasiveness, ease of administration, and the possibility to reach an elaborate mucosal network of antigen-presenting cells. AREAS COVERED This review discusses current pulmonary vaccination strategies and their advantages and disadvantages. EXPERT OPINION To improve efficiency of vaccination and develop new strategies, a well-founded knowledge about composition and characterization of antigen-presenting cell populations throughout the respiratory tract is essential. In particular, respiratory tract dendritic cells, as key antigen-presenting cells in the lung, constitute an ideal target for vaccine delivery. Furthermore, particle size is a key factor when designing new inhalable vaccines, as size determines not only deposition in different respiratory tract compartments, but also how an antigen and its carrier will interact with lung tissue components and immune cells. An increased knowledge of different respiratory tract antigen-presenting cell populations and their interactions with other components of the immune system will enable new targeting strategies to improve the efficacy of pulmonary vaccination.
Collapse
Affiliation(s)
- Fabian Blank
- Bern University, Pulmonary Medicine, Department of Clinical Research, Murtenstrasse 50, CH-3010 Berne, Switzerland.
| | | | | |
Collapse
|
116
|
Tong J, Wu WN, Kong X, Wu PF, Tian L, Du W, Fang M, Zheng F, Chen JG, Tan Z, Gong F. Acid-sensing ion channels contribute to the effect of acidosis on the function of dendritic cells. THE JOURNAL OF IMMUNOLOGY 2011; 186:3686-92. [PMID: 21321108 DOI: 10.4049/jimmunol.1001346] [Citation(s) in RCA: 64] [Impact Index Per Article: 4.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Abstract
As an H(+)-gated subgroup of the degenerin/epithelial Na(+) channel family, acid-sensing ion channels (ASICs) were reported to be involved in various physiological and pathological processes in neurons. However, little is known about the role of ASICs in the function of dendritic cells (DCs). In this study, we investigated the expression of ASICs in mouse bone marrow-derived DCs and their possible role in the function of DCs. We found that ASIC1, ASIC2, and ASIC3 are expressed in DCs at the mRNA and protein levels, and extracellular acid can evoke ASIC-like currents in DCs. We also demonstrated that acidosis upregulated the expression of CD11c, MHC class II, CD80, and CD86 and enhanced the Ag-presenting ability of DCs via ASICs. Moreover, the effect of acidosis on DCs can be abolished by the nonsteroidal anti-inflammatory drugs ibuprofen and diclofenac. These results suggest that ASICs are involved in the acidosis-mediated effect on DC function.
Collapse
Affiliation(s)
- Jing Tong
- Department of Immunology, Tongji Medical College, Huazhong University of Science and Technology, Wuhan 430030, People's Republic of China
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
117
|
Park YS, Bae JH, Son CH, Lee KS, Kim W, Jung MH, Yang K, Kim SH, Kang CD. Cyclophosphamide potentiates the antitumor effect of immunization with injection of immature dendritic cells into irradiated tumor. Immunol Invest 2011; 40:383-99. [PMID: 21314288 DOI: 10.3109/08820139.2011.552141] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/27/2022]
Abstract
Growth of a tumor on the left flank was suppressed by direct injection of immature DCs (iDCs) into the irradiated tumor on the right thigh (IR/DC). This antitumor immune effect of IR/DC was enhanced by pretreatment with CTX (CTX+IR/DC) and this effect was related with increased number of tumor-specific IFN-γ secreting T cells and decreased ratio of CD4(+)CD25(+)/CD4(+) T cells. The treatment with CTX+IR/DC increased or decreased the levels of IL-2 or IL-10, respectively. These results demonstrated that antitumor effect of IR/DC could be augmented by pretreatment with low-dose CTX, suggesting a new antitumor therapeutic modality of chemoradioimmunotherapy.
Collapse
Affiliation(s)
- You-Soo Park
- Department of Biochemistry, Pusan National University School of Medicine, Yangsan 626-870, Korea
| | | | | | | | | | | | | | | | | |
Collapse
|
118
|
Balan S, Kale VP, Limaye LS. A large number of mature and functional dendritic cells can be efficiently generated from umbilical cord blood-derived mononuclear cells by a simple two-step culture method. Transfusion 2011; 50:2413-23. [PMID: 20497510 DOI: 10.1111/j.1537-2995.2010.02706.x] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
BACKGROUND Advances in the past two decades in dendritic cell (DC) biology paved the way to exploit them as a promising tool in cancer immunotherapy. The prerequisite for DC vaccine preparations is large-scale in vitro generations of homogeneous, mature, and functional DCs. Frequent improvements are being made in the existing in vitro DC production protocols to achieve this goal. In our previous study we reported a large-scale generation of mature, functional DCs from umbilical cord blood (UCB) CD34+ cells. Here we report that this method can be used for the efficient generation of DCs from UCB mononuclear cells (MNCs) and thus the hematopoietic stem cell isolation step is not essential. STUDY DESIGN AND METHODS MNCs or CD34+ cells isolated from the same cord blood (CB) samples were used for the generation of DCs. DCs were characterized for morphology, phenotype, and functional assays including antigen uptake, chemotaxis, and mixed leukocyte reaction. Similarly DCs generated from the MNCs of same fresh and frozen CB units were compared. RESULTS The morphologic, phenotypic, and functional characterization of the DCs generated from various sets show that they were comparable in nature irrespective of the starting population used. CONCLUSION We conclude that the CD34+ isolation step is not essential for the generation of mature, functional DCs and thus can be eliminated. More importantly, we show that DCs can be generated with equal efficiency from the MNCs of frozen CB units. Our culture method will be useful for exploiting the potential of UCB as an additional source for allogeneic DCs in the clinical settings.
Collapse
Affiliation(s)
- Sreekumar Balan
- National Centre for Cell Science, Pune University Campus, Pune, India
| | | | | |
Collapse
|
119
|
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
|
120
|
This month in APR. Arch Pharm Res 2010; 33:1699-701. [PMID: 21116771 DOI: 10.1007/s12272-010-1100-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
|
121
|
Choi Y, Kim CW. Antitumor effects of combined granulocyte macrophage colony stimulating factor and macrophage inflammatory protein-3 alpha plasmid DNA. Cancer Sci 2010; 101:2341-50. [PMID: 20804501 PMCID: PMC11158867 DOI: 10.1111/j.1349-7006.2010.01704.x] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022] Open
Abstract
Dendritic cells (DC) are critical for priming adaptive immune responses to foreign antigens. However, the feasibility of harnessing these cells in vivo to optimize the antitumor effects has not been fully explored. The authors investigated a novel therapeutic approach that involves delivering synergistic signals that both recruit and expand DC populations at sites of intratumoral injection. More specifically, the authors examined whether the co-administration of plasmids encoding the chemokine macrophage inflammatory protein-3 alpha (pMIP3α) and plasmid encoding the granulocyte macrophage colony stimulating factor (pGM-CSF; a DC-specific growth factor) can recruit, expand and activate large numbers of DC at sites of intratumoral injection. It was found that the administration of pGM-CSF and pMIP3α resulted in dramatic recruitment and expansion of DC at these sites and in draining lymph nodes. Furthermore, treatment with pGM-CSF and pMIP3α generated the strongest MUC1-associated CD8+ T-cell immune responses in draining lymph nodes and in tumors, produced the greatest antitumor effects and enhanced survival rates more than pcDNA3.1, pGM-CSF alone and pMIP3α alone. It was also found that pGM-CSF plus pMIP3α generated the strongest MUC1-associated CD4+ T-cell immune responses in draining lymph nodes and in tumors. The findings of the present study suggest that the recruitment and activation of DC in vivo due to the synergistic actions of pGM-CSF and pMIP3α presents a potentially feasible means of controlling immunogenic malignancies and provides a basis for the development of novel immunotherapeutic treatments.
Collapse
Affiliation(s)
- Yun Choi
- Department of Pathology, Seoul National University College of Medicine, Seoul, Korea
| | | |
Collapse
|
122
|
Ma J, Usui Y, Takeuchi M, Okunuki Y, Kezuka T, Zhang L, Mizota A, Goto H. Human uveal melanoma cells inhibit the immunostimulatory function of dendritic cells. Exp Eye Res 2010; 91:491-9. [DOI: 10.1016/j.exer.2010.06.025] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2009] [Revised: 06/25/2010] [Accepted: 06/29/2010] [Indexed: 12/11/2022]
|
123
|
Bürdek M, Spranger S, Wilde S, Frankenberger B, Schendel DJ, Geiger C. Three-day dendritic cells for vaccine development: antigen uptake, processing and presentation. J Transl Med 2010; 8:90. [PMID: 20920165 PMCID: PMC2955579 DOI: 10.1186/1479-5876-8-90] [Citation(s) in RCA: 37] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/12/2010] [Accepted: 09/28/2010] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND Antigen-loaded dendritic cells (DC) are capable of priming naïve T cells and therefore represent an attractive adjuvant for vaccine development in anti-tumor immunotherapy. Numerous protocols have been described to date using different maturation cocktails and time periods for the induction of mature DC (mDC) in vitro. For clinical application, the use of mDC that can be generated in only three days saves on the costs of cytokines needed for large scale vaccine cell production and provides a method to produce cells within a standard work-week schedule in a GMP facility. METHODS In this study, we addressed the properties of antigen uptake, processing and presentation by monocyte-derived DC prepared in three days (3d mDC) compared with conventional DC prepared in seven days (7d mDC), which represent the most common form of DC used for vaccines to date. RESULTS Although they showed a reduced capacity for spontaneous antigen uptake, 3d mDC displayed higher capacity for stimulation of T cells after loading with an extended synthetic peptide that requires processing for MHC binding, indicating they were more efficient at antigen processing than 7d DC. We found, however, that 3d DC were less efficient at expressing protein after introduction of in vitro transcribed (ivt)RNA by electroporation, based on published procedures. This deficit was overcome by altering electroporation parameters, which led to improved protein expression and capacity for T cell stimulation using low amounts of ivtRNA. CONCLUSIONS This new procedure allows 3d mDC to replace 7d mDC for use in DC-based vaccines that utilize long peptides, proteins or ivtRNA as sources of specific antigen.
Collapse
Affiliation(s)
- Maja Bürdek
- German Research Center for Environmental Health, Institute of Molecular Immunology, Marchioninistr, 25, 81377 München, Germany
| | | | | | | | | | | |
Collapse
|
124
|
Qu Y, Chen L, Pardee AD, Taylor JL, Wesa AK, Storkus WJ. Intralesional delivery of dendritic cells engineered to express T-bet promotes protective type 1 immunity and the normalization of the tumor microenvironment. THE JOURNAL OF IMMUNOLOGY 2010; 185:2895-902. [PMID: 20675595 DOI: 10.4049/jimmunol.1001294] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/30/2023]
Abstract
T-bet (Tbx21), a T-box transcription factor, has been previously identified as a master regulator of type 1 T cell polarization. We have also recently shown that the genetic engineering of human dendritic cells (DCs) to express human T-bet cDNA yields type 1-polarizing APCs in vitro (1). In the present study, murine CD11c(+) DCs were transduced with a recombinant adenovirus encoding full-length murine T-bets (DC.mTbets) and analyzed for their immunomodulatory functions in vitro and in vivo. Within the range of markers analyzed, DC.mTbets exhibited a control DC phenotype and were indistinguishable from control DCs in their ability to promote allogenic T cell proliferation in MLR in vitro. However, DC.mTbets were superior to control DCs in promoting Th1 and Tc1 responses in vitro via a mechanism requiring DC-T cell interaction or the close proximity of these two cell types and that can only partially be explained by the action of DC-elaborated IL-12p70. When injected into day 7 s.c. CMS4 sarcoma lesions growing in syngenic BALB/c mice, DC.mTbets dramatically slowed tumor progression (versus control DCs) and extended overall survival via a mechanism dependent on both CD4(+) and CD8(+) T cells and, to a lesser extent, asialoGM1(+) NK cells. DC.mTbet-based therapy also promoted superior tumor-specific Tc1 responses in the spleens and tumor-draining lymph nodes of treated animals, and within the tumor microenvironment it inhibited the accumulation of CD11b(+)Gr1(+) myeloid-derived suppressor cells and normalized CD31(+) vascular structures. These findings support the potential translational utility of DC.Tbets as a therapeutic modality in the cancer setting.
Collapse
Affiliation(s)
- Yanyan Qu
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, PA 15213, USA
| | | | | | | | | | | |
Collapse
|
125
|
Hammarfjord O, Wallin RPA. Dendritic cell function at low physiological temperature. J Leukoc Biol 2010; 88:747-56. [PMID: 20651299 DOI: 10.1189/jlb.0310155] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/16/2022] Open
Abstract
Compared with the stable core temperature, the skin temperature is lower and varies depending on ambient temperature and convection conditions. The function of DCs, which are plentiful in the skin at lower physiological temperatures, has not been reported. We show that DC performed some functions normally at 28°C, including phagocytosis and macropinocytosis. TLR-4 signaling via MAPK pathways was delayed at 28°C but reached normal levels, which may explain the observed slower kinetics of stimulated macropinocytosis and TNF production. TLR-4-induced NO production was compromised severely at 28°C. Collagen degradation and migration through matrigel-coated transwell inserts were decreased, but no effect on podosome number or DC migration through noncoated transwell filters was seen. Lowering the temperature differentially regulated functions associated with the role of DCs in adaptive immunity. LPS-induced up-regulation of CD86 was normal; however, CD40 up-regulation was suppressed after TLR-4 stimulation at 28°C. Nonactivated DC processed and presented antigen on MHC class II equally well at 28°C and 37°C. However, DCs that were loaded with antigens and stimulated with TLR ligand at 28°C were poor at activating T cells at 37°C compared with DCs that were activated and loaded with antigen at 37°C.
Collapse
Affiliation(s)
- Oscar Hammarfjord
- Center for Infectious Medicine, F59, Department of Medicine, Karolinska Institutet, Karolinska University Hospital, Huddinge, Stockholm, Sweden
| | | |
Collapse
|
126
|
Abstract
Therapeutic immunization leading to cancer regression remains a significant challenge. Successful immunization requires activation of adaptive immunity, including tumor specific CD4 T cells and CD8 T cells. Generally, the activation of T cells is compromised in patients with cancer because of immune suppression, loss of tumor antigen expression, and dysfunction of antigen-presenting cells. Antigen-presenting cells such as dendritic cells (DCs) are key for the induction of adaptive antitumor immune responses. Recently, attention has focused on novel adjuvants that enhance dendritic cell function and their ability to prime T cells. Agonists that target toll-like receptors are being used clinically either alone or in combination with tumor antigens and showing initial success both in terms of enhancing immune responses and eliciting antitumor activity. This review summarizes the application of these adjuvants to treat cancer and the potential for boosting responses in vivo.
Collapse
Affiliation(s)
- Nina Bhardwaj
- New York University School of Medicine, 522 First Avenue, Smilow Research Building, Room 1303, New York, NY 10016, Office: (212) 263-5814, Fax: (212) 263-6729,
| | | | | |
Collapse
|
127
|
Combined Treatment With Dendritic Cells and 5-fluorouracil Elicits Augmented NK Cell-mediated Antitumor Activity Through the Tumor Necrosis Factor-α Pathway. J Immunother 2010; 33:467-74. [DOI: 10.1097/cji.0b013e3181d36726] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
128
|
He S, Wang L, Wu Y, Li D, Zhang Y. CCL3 and CCL20-recruited dendritic cells modified by melanoma antigen gene-1 induce anti-tumor immunity against gastric cancer ex vivo and in vivo. J Exp Clin Cancer Res 2010; 29:37. [PMID: 20420712 PMCID: PMC2873423 DOI: 10.1186/1756-9966-29-37] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/10/2009] [Accepted: 04/27/2010] [Indexed: 12/23/2022] Open
Abstract
BACKGROUND To investigate whether dendritic cell (DC) precursors, recruited by injection of chemokine ligand 3 (CCL3) and CCL20, induce anti-tumor immunity against gastric cancer induced by a DC vaccine expressing melanoma antigen gene-1 (MAGE-1) ex vivo and in vivo. METHODS B6 mice were injected with CCL3 and CCL20 via the tail vein. Freshly isolated F4/80-B220-CD11c+ cells cultured with cytokines were analyzed by phenotype analysis and mixed lymphocyte reaction (MLR). For adenoviral (Ad)-mediated gene transduction, cultured F4/80-B220-CD11c+ cells were incubated with Ad-MAGE-1. Vaccination of stimulated DC induced T lymphocytes. The killing effect of these T cells against gastric carcinoma cells was assayed by MTT. INF-gamma production was determined with an INF-gamma ELISA kit. In the solid tumor and metastases model, DC-based vaccines were used for immunization after challenge with MFC cells. Tumor size, survival of mice, and number of pulmonary metastatic foci were used to assess the therapeutic effect of DC vaccines. RESULTS F4/80-B220-CD11c+ cell numbers increased after CCL3 and CCL20 injection. Freshly isolated F4/80-B220-CD11c+ cells cultured with cytokines were phenotyically identical to typical DC and gained the capacity to stimulate allogeneic T cells. These DCs were transduced with Ad-MAGE-1, which were prepared for DC vaccines expressing tumor antigen. T lymphocytes stimulated by DCs transduced with Ad-MAGE-1 exhibited specific killing effects on gastric carcinoma cells and produced high levels of INF-gamma ex vivo. In vivo, tumor sizes of the experimental group were much smaller than both the positive control group and the negative control groups (P < 0.05). Kaplan-Meier survival curves showed that survival of the experimental group mice was significantly longer than the control groups (P < 0.05). In addition, MAGE-1-transduced DCs were also a therapeutic benefit on an established metastatic tumor, resulting in a tremendous decrease in the number of pulmonary metastatic foci. CONCLUSIONS CCL3 and CCL20-recruited DCs modified by adenovirus-trasnsduced, tumor-associated antigen, MAGE-1, can stimulate anti-tumor immunity specific to gastric cancer ex vivo and in vivo. This system may prove to be an efficient strategy for anti-tumor immunotherapy.
Collapse
Affiliation(s)
- Songbing He
- Department of General Surgery, the First Affiliated Hospital of Soochow University, 215006 Suzhou, Jiangsu Province, China
| | - Liang Wang
- Department of General Surgery, the First Affiliated Hospital of Soochow University, 215006 Suzhou, Jiangsu Province, China
| | - Yugang Wu
- Department of General Surgery, the Third Affiliated Hospital of Soochow University, 213000 Changzhou, Jiangsu Province, China
| | - Dechun Li
- Department of General Surgery, the First Affiliated Hospital of Soochow University, 215006 Suzhou, Jiangsu Province, China
| | - Yanyun Zhang
- Institute of Health Science and Shanghai Institute of Immunology, Shanghai Institute for Biological Science, Chinese Academy of Science and Shanghai Jiao Tong University School of Medicine, 200025 Shanghai, China
| |
Collapse
|
129
|
|
130
|
Kalinski P, Wieckowski E, Muthuswamy R, de Jong E. Generation of stable Th1/CTL-, Th2-, and Th17-inducing human dendritic cells. Methods Mol Biol 2010; 595:117-33. [PMID: 19941108 DOI: 10.1007/978-1-60761-421-0_7] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/17/2022]
Abstract
Dendritic cells (DC) are the most potent inducers and regulators of immune responses, responsible for communication within immune system. The ability of DC to act both as the inducers of immune responses and as regulatory/suppressive cells led to the interest in their immunotherapeutic use in different disease types, ranging from cancer to autoimmunity, and as a tool to prevent the rejection of transplanted tissues and organs. Over the last years, several groups including ours have demonstrated the feasibility of obtaining monocyte-derived DC with different functions, by modulating the conditions and the duration of DC maturation. The current chapter provides a detailed protocol of generating type-1-, type-2-, and type-17-polarized DC for testing the cytokine-producing abilities of these cells and their effectiveness in inducing Th1, Th2, and Th17 responses of CD4(+) T cells and CTL responses of naïve and memory CD8(+) T cells.
Collapse
Affiliation(s)
- Pawel Kalinski
- Department of Surgery, University of Pittsburgh and University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| | | | | | | |
Collapse
|
131
|
Steiner JM, Sartor M, Sanchez AB, Messmer D, Freed A, Esener S, Messmer BT. DeNAno: Selectable deoxyribonucleic acid nanoparticle libraries. J Biotechnol 2009; 145:330-3. [PMID: 19963022 DOI: 10.1016/j.jbiotec.2009.12.002] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/07/2009] [Revised: 11/20/2009] [Accepted: 12/01/2009] [Indexed: 10/20/2022]
Abstract
DNA nanoparticles of approximately 250 nm were produced by rolling circle replication of circular oligonucleotide templates which results in highly condensed DNA particulates presenting concatemeric sequence repeats. Using templates containing randomized sequences, high diversity libraries of particles were produced. A biopanning method that iteratively screens for binding and uses PCR to recover selected particles was developed. The initial application of this technique was the selection of particles that bound to human dendritic cells (DCs). Following nine rounds of selection the population of particles was enriched for particles that bound DCs, and individual binding clones were isolated and confirmed by flow cytometry and microscopy. This process, which we have termed DeNAno, represents a novel library technology akin to aptamer and phage display, but unique in that the selected moiety is a multivalent nanoparticle whose activity is intrinsic to its sequence. Cell targeted DNA nanoparticles may have applications in cell imaging, cell sorting, and cancer therapy.
Collapse
Affiliation(s)
- Jason M Steiner
- Department of Electrical and Computer Engineering, University of California, San Diego, 9500 Gilman Dr, La Jolla, CA 92093, USA
| | | | | | | | | | | | | |
Collapse
|
132
|
Immunological factors relating to the antitumor effect of temozolomide chemoimmunotherapy in a murine glioma model. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2009; 17:143-53. [PMID: 19889936 DOI: 10.1128/cvi.00292-09] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
In this study, we investigated the potential of combined treatment with temozolomide (TMZ) chemotherapy and tumor antigen-pulsed dendritic cells (DCs) and the underlying immunological factors of TMZ chemoimmunotherapy with an intracranial GL26 glioma animal model. The combined treatment enhanced the tumor-specific immune responses and prolonged the survival more effectively than either single therapy in GL26 tumor-bearing animals. Apoptosis was induced in the tumors of the animals by the treatment with TMZ. Calreticulin (CRT) surface exposure was detected by immunofluorescence staining of TMZ-treated GL26 cells. TMZ chemotherapy increased tumor antigen cross-priming from tumor cells, leading to cross-priming of tumor antigen-specific CD4(+) T cells and CD8(+) T cells. This chemotherapy appeared to suppress the frequency of CD4(+) CD25(+) regulatory T cells (Treg). Moreover, this combined therapy resulted in an increase in the tumor infiltration of CD4(+) and CD8(+) T cells. Collectively, the findings of this study provide evidence that the combination of TMZ chemotherapy and treatment with DC-based vaccines leads to the enhancement of antitumor immunity through increased tumor-specific immune responses via the cross-priming of apoptotic tumor cell death mediated by CRT exposure and, in part, the suppression of Treg. Therefore, CRT exposure, regulatory T cells, and cross-priming by TMZ chemotherapy may be immunological factors related to the enhancement of the antitumor effects of chemoimmunotherapy in an experimental brain tumor model.
Collapse
|
133
|
Zhou J, Weng D, Zhou F, Pan K, Song H, Wang Q, Wang H, Wang H, Li Y, Huang L, Zhang H, Huang W, Xia J. Patient-derived renal cell carcinoma cells fused with allogeneic dendritic cells elicit anti-tumor activity: in vitro results and clinical responses. Cancer Immunol Immunother 2009; 58:1587-97. [PMID: 19221746 PMCID: PMC11030900 DOI: 10.1007/s00262-009-0668-9] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/04/2008] [Accepted: 01/21/2009] [Indexed: 10/21/2022]
Abstract
Renal cell carcinoma (RCC) has been shown to be susceptible to immunotherapeutic treatment strategies. In the present study, patient-derived tumor cells were fused with allogeneic dendritic cells (DC) to elicit anti-tumor activity against RCC. DC from HLA-A2+ healthy donors were fused with primary RCC cells from ten patients. Phenotype of fusion cells were characterized by flow cytometer and confocal microscopy. In vitro, T cell proliferation, IFN-gamma secretion and cytotoxic T lymphocytes (CTL) activity elicited by allogeneic DC/RCC fusion cells were assessed. Clinically, ten patients were vaccinated with allogeneic DC/RCC fusion vaccine. The adverse effects and toxicity were observed. The clinical response was evaluated by CT scans. After fusion, the created hybrids expressed both tumor associated antigen and DC-derived molecules and could stimulate the proliferation and IFN-gamma secretion of T cells as well as elicit strong CTL activity against RCC cells in vitro. In vivo, no serious adverse effects, toxicity, or signs of autoimmune disease were observed after vaccination therapy. Percentage of T lymphocyte subsets in peripheral blood of patients was increased significantly. One of ten patients exhibited a partial response with regression of lung metastases. Six patients showed stable disease with stabilization of previously progressive disease (follow up 1.5 years). The PR and SD responses, exhibited by 7/10 patients who received the allogeneic DC/RCC fusion vaccine treatment, suggest that this approach is safe and can elicit immunological responses in a significant portion of patients with RCC.
Collapse
Affiliation(s)
- Jun Zhou
- State Key Laboratory of Oncology in Southern China, 510060 Guangzhou, People’s Republic of China
- Biotherapy Center, Cancer Center, Sun Yat-sen University, 651 Dongfeng Road East, 510060 Guangzhou, People’s Republic of China
| | - Desheng Weng
- State Key Laboratory of Oncology in Southern China, 510060 Guangzhou, People’s Republic of China
- Biotherapy Center, Cancer Center, Sun Yat-sen University, 651 Dongfeng Road East, 510060 Guangzhou, People’s Republic of China
| | - Fangjian Zhou
- Department of Urology, Cancer Center, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Ke Pan
- State Key Laboratory of Oncology in Southern China, 510060 Guangzhou, People’s Republic of China
- Biotherapy Center, Cancer Center, Sun Yat-sen University, 651 Dongfeng Road East, 510060 Guangzhou, People’s Republic of China
| | - Haifeng Song
- State Key Laboratory of Oncology in Southern China, 510060 Guangzhou, People’s Republic of China
- Biotherapy Center, Cancer Center, Sun Yat-sen University, 651 Dongfeng Road East, 510060 Guangzhou, People’s Republic of China
| | - Qijing Wang
- State Key Laboratory of Oncology in Southern China, 510060 Guangzhou, People’s Republic of China
- Biotherapy Center, Cancer Center, Sun Yat-sen University, 651 Dongfeng Road East, 510060 Guangzhou, People’s Republic of China
| | - Huan Wang
- Biotherapy Center, Cancer Center, Sun Yat-sen University, 651 Dongfeng Road East, 510060 Guangzhou, People’s Republic of China
| | - Hui Wang
- State Key Laboratory of Oncology in Southern China, 510060 Guangzhou, People’s Republic of China
- Department of Urology, Cancer Center, Sun Yat-sen University, Guangzhou, People’s Republic of China
| | - Yongqiang Li
- State Key Laboratory of Oncology in Southern China, 510060 Guangzhou, People’s Republic of China
- Biotherapy Center, Cancer Center, Sun Yat-sen University, 651 Dongfeng Road East, 510060 Guangzhou, People’s Republic of China
| | - Lixi Huang
- State Key Laboratory of Oncology in Southern China, 510060 Guangzhou, People’s Republic of China
- Biotherapy Center, Cancer Center, Sun Yat-sen University, 651 Dongfeng Road East, 510060 Guangzhou, People’s Republic of China
| | - Huakun Zhang
- State Key Laboratory of Oncology in Southern China, 510060 Guangzhou, People’s Republic of China
- Biotherapy Center, Cancer Center, Sun Yat-sen University, 651 Dongfeng Road East, 510060 Guangzhou, People’s Republic of China
| | - Wei Huang
- State Key Laboratory of Oncology in Southern China, 510060 Guangzhou, People’s Republic of China
- Biotherapy Center, Cancer Center, Sun Yat-sen University, 651 Dongfeng Road East, 510060 Guangzhou, People’s Republic of China
| | - Jianchuan Xia
- State Key Laboratory of Oncology in Southern China, 510060 Guangzhou, People’s Republic of China
- Biotherapy Center, Cancer Center, Sun Yat-sen University, 651 Dongfeng Road East, 510060 Guangzhou, People’s Republic of China
| |
Collapse
|
134
|
Balan S, Kale VP, Limaye LS. A simple two-step culture system for the large-scale generation of mature and functional dendritic cells from umbilical cord blood CD34+ cells. Transfusion 2009; 49:2109-21. [DOI: 10.1111/j.1537-2995.2009.02231.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022]
|
135
|
Yang BB, Jiang H, Chen J, Zhang X, Ye JJ, Cao J. Dendritic cells pulsed with GST-EGFR fusion protein: effect in antitumor immunity against head and neck squamous cell carcinoma. Head Neck 2009; 32:626-35. [PMID: 19787789 DOI: 10.1002/hed.21233] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023] Open
Abstract
BACKGROUND Overexpression of epidermal growth factor receptor (EGFR) is common in head and neck squamous cell carcinoma (HNSCC). Targeting EGFR is an effective approach to treat EGFR-positive HNSCC. However, the clinical benefits of the present EGFR-targeting agents are still limited in HNSCC patients. METHODS Recombinant glutathione-S-transferase (GST)-EGFR fusion protein was produced and purified. Dendritic cells (DCs) of C3H mice were pulsed with fusion protein. Mice were challenged with HNSCC cells before or after vaccination with these DCs, and the cytotoxic T-lymphocyte (CTL) response, interferon-gamma (IFN-gamma) secretion, tumor growth, and survival of mice were assessed. RESULTS Significant in vitro and in vivo antitumor activities were observed for mice immunized with DCs pulsed with GST-EGFR fusion protein, compared with the control groups (p < .05). CONCLUSION The DCs pulsed with GST-EGFR fusion protein can provide not only preventive but also therapeutic antitumor activities against HNSCC in the animal model.
Collapse
Affiliation(s)
- Bei-Bei Yang
- Department of Otolaryngology, 2nd Affiliated Hospital, College of Medicine, Zhejiang University, Zhejiang, China
| | | | | | | | | | | |
Collapse
|
136
|
Therapeutic and tumor-specific immunity induced by combination of dendritic cells and oncolytic adenovirus expressing IL-12 and 4-1BBL. Mol Ther 2009; 18:264-74. [PMID: 19738604 PMCID: PMC2839296 DOI: 10.1038/mt.2009.205] [Citation(s) in RCA: 94] [Impact Index Per Article: 5.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/11/2022] Open
Abstract
Recently, gene-based cytokine treatment has been actively pursued as a new promising approach in treating cancer. In an effort to augment the efficiency of antitumor effect by cytokine-mediated immunotherapy, we selected both interleukin (IL)-12 and 4-1BB ligand (4-1BBL) as suitable cytokines to fully activate the type-1 immune response. Coexpression of IL-12 and 4-1BBL mediated by oncolytic adenovirus (Ad) greatly enhanced the antitumor effect. Further, synergistic enhancement in interferon (IFN)-gamma levels were seen in mice treated with oncolytic Ad expressing both IL-12 and 4-1BBL. Next, to improve the overall antitumor immune response, we coadministered IL-12- and 4-1BBL-coexpressing oncolytic Ad with dendritic cells (DCs). Combination treatment of IL-12- and 4-1BBL-coexpressing oncolytic Ad and DCs elicited greater antitumor and antimetastatic effects than either treatment alone. Moreover, enhanced type-1 antitumor immune response and higher migratory abilities of DCs in tumors were also observed in the combination arms. The nature of the enhanced antitumor immune response seems to be mediated through the enhanced cytolytic activity of cytotoxic T lymphocytes (CTLs) and IFN-gamma-releasing immune cells. Taken together, these data highlight the potential therapeutic benefit of combining IL-12- and 4-1BBL-coexpressing oncolytic Ad with DCs and warrants further evaluation in the clinic.
Collapse
|
137
|
Inoue M, Senju S, Hirata S, Irie A, Baba H, Nishimura Y. An in vivo model of priming of antigen-specific human CTL by Mo-DC in NOD/Shi-scid IL2rγnull (NOG) mice. Immunol Lett 2009; 126:67-72. [DOI: 10.1016/j.imlet.2009.08.001] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2009] [Revised: 07/28/2009] [Accepted: 08/02/2009] [Indexed: 10/20/2022]
|
138
|
Cruz M, Enes M, Pereira M, Dourado M, Sarmento Ribeiro AB. [Experimental models in oncology: contribution of cell culture on understanding the biology of cancer]. REVISTA PORTUGUESA DE PNEUMOLOGIA 2009; 15:669-82. [PMID: 19547898 DOI: 10.1016/s0873-2159(15)30163-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022] Open
Abstract
In the beginning of the 20th century, tissue culture was started with the aim of studying the behaviour of animal cells in normal and stress conditions. The cell study at molecular level depends on their capacity of growing and how they can be manipulated in laboratory. In vitro cell culture allows us the possibility of studying biological key processes, such as growth, differentiation and cell death, and also to do genetic manipulations essential to the knowledge of structure and genes function. Human stem cells culture provides strategies to circumvent other models' deficiencies. It seems that cancer stem cells remain quiescent until activation by appropriated micro-environmental stimulation. Several studies reveal that different cancer types could be due to stem cell malignant transformations. Removal of these cells is essential to the development of more effective cancer therapies for advanced disease. On the other hand, dendritic cells modified in culture may be used as a therapeutic vaccine in order to induce tumour withdraw.
Collapse
Affiliation(s)
- Mariana Cruz
- Faculdade de Medicina da Universidade de Coimbra, Coimbra, Portugal
| | | | | | | | | |
Collapse
|
139
|
Spleen migrating dendritic cells primed with CC531 colon cancer antigen and LPS - is it a method to compromise liver metastases? Surg Oncol 2009; 19:e85-94. [PMID: 19665370 DOI: 10.1016/j.suronc.2009.07.003] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/23/2008] [Revised: 07/10/2009] [Accepted: 07/14/2009] [Indexed: 11/24/2022]
Abstract
The anti-tumor vaccination is burdened by low recruitment rate of intravenously administered in vitro primed DC in liver metastases and lack of supplying them continuously in large numbers. Therefore, it seemed rational to create a model of in vivo vaccination with specifically primed splenic DC and cytotoxic T lymphocytes being continuously supplied to the liver vascular bed. The question we raised was whether anti-tumor immunized splenic DC flowing to liver metastases could adhere to and be cytotoxic to tumor cells. We immunized rats with CC531 tumor cells and stimulated them with Escherichia coli LPS. Subsequently, spleen DC-enriched population was isolated, its activation by LPS, adherence to CC531 cells and cytotoxicity were measured. Spleen cells home to the liver reaching it via splenic vein. These cells can be retrieved by simple washout of liver sinusoids (liver sinusoidal washout cells - LSWC). Their adherence to and cytotoxicity against CC531 cells were evaluated. Moreover, in vitro adherence of splenic DC-enriched cells and LSWC to CC531 liver tumor sections was measured. We found that in vivo immunization of splenic population containing DC, NK cells and lymphocytes with CC531 cells and stimulation with LPS activated these cells but did not significantly increase the cytotoxicity against CC531 cells. There was also no increase in cytotoxicity of LSWC. Adhesion of splenic DC and LWSC to liver CC531 metastases on cryosections was higher than to the adjacent liver tissue. However, it was more expressed on tumor stromal than neoplastic cells. The level of splenic Treg cells down-regulating immune response was found only slightly increased after immunization. Taken together, in the model of in vivo immunization against CC531 cells, low level of spleen DC and spleen-derived LSWC cytotoxicity as well as adherence rate to tumor cells were observed. More effective methods of immunizing splenic DC overcoming the suppressive mechanisms should be looked for.
Collapse
|
140
|
Improvement of a dendritic cell-based therapeutic cancer vaccine with components of Toxoplasma gondii. CLINICAL AND VACCINE IMMUNOLOGY : CVI 2009; 16:1393-8. [PMID: 19656994 DOI: 10.1128/cvi.00199-09] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/31/2023]
Abstract
The use of dendritic cells (DCs) as a cellular adjuvant is a promising approach to the immunotherapy of cancer. It has previously been demonstrated that DCs pulsed ex vivo with Toxoplasma gondii antigens trigger a systemic Th1-biased specific immune response and induce protective and specific antitoxoplasma immunity. In the present study, we demonstrate that tumor antigen-pulsed DCs matured in the presence of Toxoplasma gondii components induce a potent antitumor response in a mouse model of fibrosarcoma. Bone-marrow derived DCs (BMDCs) were cultured in the presence of granulocyte-macrophage colony-stimulating factor and interleukin-4. After 5 days, tumor lysates with or without the T. gondii lysate were added to the culture for another 2 days. The cytokine production in the BMDC culture and the coculture supernatants of DCs and splenic cells was evaluated. For immunization, 7 days after tumor challenge, different groups of BALB/c mice received different kinds of DCs subcutaneously around the tumor site. Tumor growth was monitored, and 2 weeks after DC immunotherapy, the cytotoxic activity and the infiltration of CD8(+) T cells were monitored in different groups. According to the findings, immunotherapy with T. gondii-matured DCs led to a significant increase in the activity of cytotoxic T cells and decreased the rate of growth of the tumor in immunized animals. Immature DCs did not cause any change in cytotoxic activity or the tumor growth rate compared to that in the healthy controls. The current study suggests that a specific antitumor immune response can be induced by DCs matured with T. gondii components and provide the basis for the use of T. gondii in DC-targeted clinical therapies.
Collapse
|
141
|
Application of Interleukin-12 Expressing Dendritic Cells for the Treatment of Animal Model of Leukemia. Exp Biol Med (Maywood) 2009; 234:952-60. [DOI: 10.3181/0805-rm-165] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
Residual cancer cells appearing in blood circulation reduce the effects of radiotherapy or chemotherapy in cancer patients. It has been well documented that cultured dendritic cells can be used as a powerful tool to induce immune response. In this study, we administered different manipulations of dendritic cells (DCs), including DCs pulsed with tumor cell lysate (TCL), transfected with adenoviral IL-12 vector (AdIL-12) and transfected with AdIL-12 after being pulsed with TCL, to determine whether improved DCs based immunotherapy can specifically suppress the metastasis of tumor cells. The results demonstrated that administration of engineered DCs that transfected with AdIL-12 after being pulsed with TCL to mice with leukemia had a better protective effect than that of DCs either pulsed with TCL or transfected with AdIL-12. Moreover, depletion of CD8+ cells in the engineered DCs administered leukemia mice reduced the protective effect. These results suggest that DCs modified with TCL and AdIL-12 can prolong survival time by enhancing the activity of cytotoxic T cells. Although more studies on the mechanisms are needed, cytokine genes engineered DCs provide a promising therapeutic potential on the murine model of leukemia.
Collapse
|
142
|
Intratumoral dendritic cells and chemoradiation for the treatment of murine squamous cell carcinoma. J Immunother 2009; 31:885-95. [PMID: 18832999 DOI: 10.1097/cji.0b013e3181880f1e] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
Dendritic cells are potent antigen-presenting cells that have been shown to have significant antitumor effects in vitro and in vivo. However, the therapeutic efficacy of dendritic cells as an immunotherapeutic treatment has been limited by both immunologic tolerance and active immunosuppression in the tumor microenvironment. To address this problem, we examined the ability of concurrent systemic chemotherapy and local, fractionated radiation to augment intratumoral dendritic cell injections in a mouse model of squamous cell carcinoma. Intratumoral injections of dendritic cells alone did not have a significant antitumor effect in mice with squamous cell carcinoma flank tumors, but the addition of chemoradiation resulted in significant tumor regression. Concurrent chemoradiation alone resulted in slower tumor growth, but no complete tumor regressions. The combination of chemoradiation and intratumoral dendritic cell injections resulted in improved survival and complete tumor regression in 30% mice. Mice with complete tumor regression were partially resistant to the repeat challenge with relevant tumor 60 days after treatment. These findings were partially dependent on the presence of CD4 T cells, CD8 T cells, and natural killer cells. Chemoradiation may augment intratumoral dendritic cell injections through increased intratumoral apoptosis and decreased intratumoral regulatory T cells. This work suggests a possible role for the use of intratumoral dendritic cell therapy with more traditional chemoradiation strategies.
Collapse
|
143
|
Mutual helper effect in copulsing of dendritic cells with 2 antigens: a novel approach for improvement of dendritic-based vaccine efficacy against tumors and infectious diseases simultaneously. J Immunother 2009; 32:325-32. [PMID: 19342973 DOI: 10.1097/cji.0b013e31819aa31e] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
To develop an efficient dendritic cell (DC)-based immunotherapy protocol, we examined whether simultaneous pulsing of DCs with a given antigen and a third-party antigen could enhance their antigen presentation capacity. Purified splenic DCs of Balb/c mice were pulsed separately with immunoglobulin G, ovalbumin, conalbumin, P15 peptide of Mycobacterium tuberculosis, and prostate-specific antigen or double combinations of the aforementioned antigens. In some settings, DCs pulsed with 1 antigen were mixed equally with those pulsed with another antigen. Antigen-pulsed DCs were injected into the footpad of syngeneic mice and proliferation of whole, CD4 and CD8 depleted lymph node cells was measured after restimulation with cognate antigen. Antigen-specific production of interferon-gamma (IFNgamma) was tested in culture supernatants. Frequency of responding lymph node cells was determined by IFNgamma enzyme-linked immunosorbent spot assay. Our results showed that copulsing of DCs with 2 unrelated antigens increased the capacity of DCs to induce antigen-specific T-cell proliferation against both antigens up to 16-fold. Injection of 2 populations of DCs each pulsed with a different antigen, increased proliferation of primed T cells significantly as well. Both CD4 and CD8 depleted populations showed vigorous proliferative response in copulsing system. In addition, copulsing of DCs with 2 antigens resulted in higher frequency of antigen-specific responding cells and significantly more IFNgamma production. Our results clearly showed that unrelated peptides and proteins could be used to enhance efficacy of DC-based vaccines and in this system, each antigen served to help the other one, a condition that we termed as "mutual helper effect."
Collapse
|
144
|
Boczkowski D, Lee J, Pruitt S, Nair S. Dendritic cells engineered to secrete anti-GITR antibodies are effective adjuvants to dendritic cell-based immunotherapy. Cancer Gene Ther 2009; 16:900-11. [PMID: 19498460 DOI: 10.1038/cgt.2009.39] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
A number of monoclonal antibodies (mAbs) have been studied for their ability to enhance immune responses. Although these antibodies are effective in pre-clinical and clinical studies, they are costly and have occasionally been associated with adverse effects such as autoimmunity and cytokine storm. Numerous studies have shown that treatment of mice with an agonistic mAb, clone DTA-1, targeting murine glucocorticoid-induced tumor necrosis factor receptor (GITR) results in enhanced immune responses in tumor-bearing animals. Herein, we evaluate the novel approach of transfecting dendritic cell (DC) with mRNA encoding the heavy and light chain of the anti-GITR mAb. We show the induction of significantly enhanced tumor immunity by vaccinating with a combination of anti-GITR-secreting DC and tumor antigen-presenting DC. This enhancement is comparable to that seen with systemically delivered mAb along with the antigen-presenting DC. Importantly, when anti-GITR was delivered using RNA-transfected DC, we observed no evidence of autoimmune hypopigmentation in any tumor-free mice. We also show enhanced induction of cytotoxic T-lymphocyte responses, which is only observed when the antigen-presenting and antibody-secreting DC are co-injected at the same site. To illustrate the broad utility of this strategy, we show that DC transfected with mRNA encoding GITR-ligand/Fc fusion protein is also an effective tumor vaccine adjuvant.
Collapse
Affiliation(s)
- D Boczkowski
- Department of Surgery, Duke University Medical Center, Durham, NC 27710, USA
| | | | | | | |
Collapse
|
145
|
Heo YJ, Son CH, Chung JS, Park YS, Son JH. The cryopreservation of high concentrated PBMC for dendritic cell (DC)-based cancer immunotherapy. Cryobiology 2009; 58:203-9. [DOI: 10.1016/j.cryobiol.2008.12.006] [Citation(s) in RCA: 16] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2008] [Revised: 12/04/2008] [Accepted: 12/05/2008] [Indexed: 12/31/2022]
|
146
|
Acetalated dextran is a chemically and biologically tunable material for particulate immunotherapy. Proc Natl Acad Sci U S A 2009; 106:5497-502. [PMID: 19321415 DOI: 10.1073/pnas.0901592106] [Citation(s) in RCA: 228] [Impact Index Per Article: 14.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Materials that combine facile synthesis, simple tuning of degradation rate, processability, and biocompatibility are in high demand for use in biomedical applications. We report on acetalated dextran, a biocompatible material that can be formed into microparticles with degradation rates that are tunable over 2 orders of magnitude depending on the degree and type of acetal modification. Varying the degradation rate produces particles that perform better than poly(lactic-co-glycolic acid) and iron oxide, two commonly studied materials used for particulate immunotherapy, in major histocompatibility complex class I (MHC I) and MHC II presentation assays. Modulating the material properties leads to antigen presentation on MHC I via pathways that are dependent or independent of the transporter associated with antigen processing. To the best of our knowledge, this is the only example of a material that can be tuned to operate on different immunological pathways while maximizing immunological presentation.
Collapse
|
147
|
Simon T, Fonteneau JF, Grégoire M. Dendritic cell preparation for immunotherapeutic interventions. Immunotherapy 2009; 1:289-302. [DOI: 10.2217/1750743x.1.2.289] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
Much effort has been made over the last decade to use dendritic cells (DCs) in vaccines to induce specific antitumor immune responses. However, the great hope provided by in vitro and in vivo preclinical investigations was not translated to the clinic in terms of clinical efficacy. Thus, one of the challenges resides in optimizing DC-based therapy to give maximum clinical efficacy while using manufacturing processes that enable quality control and scale-up of consistent products. In this article, we review DC biology and the DC-based clinical trials performed to date and focus on the DC maturation status compatible with the goals of cancer immunotherapy. We also highlight the different approaches used in these clinical studies, such as the DC types or subtypes used and their preparation. Finally, we discuss the immunological and clinical outcomes in treated patients, with emphasis on the strategies that could be used to improve DC-based vaccination.
Collapse
Affiliation(s)
- Thomas Simon
- INSERM U892, Institut de Biologie, 9 quai Moncousu, 44093 Nantes Cedex 01, France
| | | | - Marc Grégoire
- INSERM U892, Institut de Biologie, 9 quai Moncousu, 44093 Nantes Cedex 01, France
| |
Collapse
|
148
|
Ferguson AR, Nichols LA, Zarling AL, Thompson ED, Brinkman CC, Hargadon KM, Bullock TN, Engelhard VH. Strategies and challenges in eliciting immunity to melanoma. Immunol Rev 2009; 222:28-42. [PMID: 18363993 DOI: 10.1111/j.1600-065x.2008.00620.x] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
The ability of CD8+ T cells to recognize melanoma tumors has led to the development of immunotherapeutic approaches that use the antigens CD8+ T cells recognize. However, clinical response rates have been disappointing. Here we summarize our work to understand the mechanisms of self-tolerance that limit responses to currently utilized antigens and our approach to identify new antigens directly tied to malignancy. We also explore several aspects of the anti-tumor immune response induced by peptide-pulsed dendritic cells (DCs). DCs differentially augment the avidity of recall T cells specific for self-antigens and overcome a process of aberrant CD8+ T-cell differentiation that occurs in tumor-draining lymph nodes. DC migration is constrained by injection route, resulting in immune responses in localized lymphoid tissue, and differential control of tumors depending on their location in the body. We demonstrate that CD8+ T-cell differentiation in different lymphoid compartments alters the expression of homing receptor molecules and leads to the presence of systemic central memory cells. Our studies highlight several issues that must be addressed to improve the efficacy of tumor immunotherapy.
Collapse
Affiliation(s)
- Andrew R Ferguson
- Beirne Carter Center for Immunology Research, Department of Microbiology, University of Virginia School of Medicine, Charlottesville, VA, USA
| | | | | | | | | | | | | | | |
Collapse
|
149
|
Non-erythroid activities of erythropoietin: Functional effects on murine dendritic cells. Mol Immunol 2009; 46:713-21. [DOI: 10.1016/j.molimm.2008.10.004] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/16/2008] [Accepted: 10/07/2008] [Indexed: 01/21/2023]
|
150
|
Sreevalsan T. Isolation of dendritic cells from human blood for in vitro interaction studies with fungal antigens. Methods Mol Biol 2009; 499:1-8. [PMID: 19152033 DOI: 10.1007/978-1-60327-151-6_1] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 05/27/2023]
Abstract
A method is described to generate dendritic cells (DCs) from human peripheral blood mononuclear cells (PBMCs). The procedure involves two major steps: (1) preparation of monocytes from human PBMCs and (2) in vitro differentiation of the monocytes into DCs by growth factors and cytokines. Cells obtained in this fashion are screened for the presence or absence of antigenic markers characteristic of DCs by flow cytometry.
Collapse
Affiliation(s)
- T Sreevalsan
- Department of Microbiology and Immunology, Georgetown University Medical Center, Washington, DC, USA
| |
Collapse
|