1
|
Abstract
Dendritic cells (DCs) are the most powerful professional antigen-presenting cells and are unique in their capability to initiate, maintain and regulate the intensity of primary immune responses, including specific antitumor responses. Development of practical procedures to prepare sufficient numbers of functional human DCs in culture from the peripheral blood precursors, paved the way for clinical trials to evaluate various DC-based strategies in patients with malignant diseases. However, no definite conclusions regarding the clinical and even immunological efficacy of DC vaccination can be stated, despite the fact that 12 years have passed since the first clinical trial utilizing DCs in cancer patients. Many unanswered questions hamper the development of DC-based vaccines, including the source of DC preparation and protocols for DC generation, activation and loading with tumor antigens, source of tumor antigens, route of vaccine administration and methods of immunomonitoring. Fortunately, in spite of the many obstacles, DC vaccines continue to hold promise for cancer therapy.
Collapse
Affiliation(s)
- Hua Zhong
- Shanghai Jiao Tong University, Shanghai Chest Hospital, 241 Huaihai Road (w), Shanghai 200030, China.
| | | | | |
Collapse
|
2
|
Chen YZ, Ruan GX, Yao XL, Li LM, Hu Y, Tabata Y, Gao JQ. Co-transfection gene delivery of dendritic cells induced effective lymph node targeting and anti-tumor vaccination. Pharm Res 2013; 30:1502-12. [PMID: 23371516 DOI: 10.1007/s11095-013-0985-8] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/11/2012] [Accepted: 01/09/2013] [Indexed: 12/18/2022]
Abstract
PURPOSE Successful genetically engineered Dendritic Cell (DC) can enhance DC's antigen presentation and lymph node migration. The present study aims to genetically engineer a DC using an efficient non-viral gene delivery vector to induce a highly efficient antigen presentation and lymph node targeting in vivo. METHODS Spermine-dextran (SD), a cationic polysaccharide vector, was used to prepare a gene delivery system for DC engineering. Transfection efficiency, nuclear trafficking, and safety of the SD/DNA complex were evaluated. A vaccine prepared by engineering DC with SD/gp100, a plasmid encoding melanoma-associated antigen, was injected subcutaneously into mice to evaluate the tumor suppression. The migration of the engineered DCs was also evaluated in vitro and in vivo. RESULTS SD/DNA complex has a better transfection behavior in vitro than commercially purchased reagents. The DC vaccine co-transfected with plasmid coding CCR7, a chemokine receptor essential for DC migration, and plasmid coding gp100 displayed superior tumor suppression than that with plasmid coding gp100 alone. Migration assay demonstrated that DC transfected with SD/CCR7 can promote DC migration capacity. CONCLUSIONS The study is the first to report the application of nonviral vector SD to co-transfect DC with gp100 and CCR7-coding plasmid to induce both the capacity of antigen presentation and lymph node targeting.
Collapse
Affiliation(s)
- Yu-Zhe Chen
- Institute of Pharmaceutics, College of Pharmaceutical Sciences, Zhejiang University, 866 Yuhangtang Road, Hangzhou 310058 Zhejiang, People's Republic of China
| | | | | | | | | | | | | |
Collapse
|
3
|
Chen YZ, Yao XL, Ruan GX, Zhao QQ, Tang GP, Tabata Y, Gao JQ. Gene-carried chitosan-linked polyethylenimine induced high gene transfection efficiency on dendritic cells. Biotechnol Appl Biochem 2012; 59:346-52. [DOI: 10.1002/bab.1036] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/27/2012] [Accepted: 08/06/2012] [Indexed: 11/09/2022]
Affiliation(s)
- Yu-Zhe Chen
- Institute of Pharmaceutics; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou; Zhejiang; People's Republic of China
| | - Xing-Lei Yao
- Institute of Pharmaceutics; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou; Zhejiang; People's Republic of China
| | - Gui-Xin Ruan
- Institute of Pharmaceutics; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou; Zhejiang; People's Republic of China
| | - Qing-Qing Zhao
- Institute of Pharmaceutics; College of Pharmaceutical Sciences; Zhejiang University; Hangzhou; Zhejiang; People's Republic of China
| | - Gu-Ping Tang
- Institute of Chemical Biology and Pharmaceutical Chemistry; Zhejiang University; Hangzhou; Zhejiang; People's Republic of China
| | - Yasuhiko Tabata
- Department of Biomaterials; Field of Tissue Engineering; Institute for Frontier Medical Sciences; Kyoto University; Kyoto; Japan
| | | |
Collapse
|
4
|
Hertz AL, Beavo JA. Cyclic nucleotides and phosphodiesterases in monocytic differentiation. Handb Exp Pharmacol 2011:365-90. [PMID: 21695649 DOI: 10.1007/978-3-642-17969-3_16] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Monocytes are immune cells that can differentiate into a number of cell types including macrophages, dendritic cells, and osteoclasts upon exposure to various cytokines. The phenotypes of these differentiated cells are highly heterogeneous and their differentiation can be affected by the cyclic nucleotides, 3'-5'-cyclic adenosine monophosphate (cAMP) and 3'-5'-cyclic guanosine monophosphate (cGMP). The intracellular levels of cAMP and cGMP are controlled through regulation of production by adenylyl and guanylyl cyclases and through degradation by cyclic nucleotide phosphodiesterases (PDEs). PDE inhibition and subsequent changes in cyclic nucleotide levels can alter the final phenotype of a differentiating monocyte with regards to surface marker expression, gene expression, or changes in secreted chemokine and cytokine levels. The differentiation process itself can also be either inhibited or augmented by changes in cyclic nucleotide levels, depending on the system being studied and the timing of cyclic nucleotide elevation. This chapter explores the effects of PDE inhibition and increases in cGMP and cAMP on monocytic differentiation into osteoclasts, dendritic cells, and macrophages.
Collapse
Affiliation(s)
- Angie L Hertz
- Department of Pharmacology, School of Medicine, University of Washington, 357280, Seattle, WA 98125, USA.
| | | |
Collapse
|
5
|
Zhong Z, Wan Y, Shi S, Han J, Zhang Z, Sun X. Co-delivery of Adenovirus and Carmustine by Anionic Liposomes with Synergistic Anti-tumor Effects. Pharm Res 2011; 29:145-57. [DOI: 10.1007/s11095-011-0521-7] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2011] [Accepted: 06/27/2011] [Indexed: 10/18/2022]
|
6
|
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
|
7
|
Toll-like receptors, tissue injury, and tumourigenesis. Mediators Inflamm 2010; 2010. [PMID: 20871832 PMCID: PMC2943133 DOI: 10.1155/2010/581837] [Citation(s) in RCA: 54] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/02/2009] [Accepted: 08/06/2010] [Indexed: 02/07/2023] Open
Abstract
Toll-like receptors (TLRs) belong to a class of molecules known as pattern recognition receptors, and they are part of the innate immune system, although they modulate mechanisms that impact the development of adaptive immune responses. Several studies have shown that TLRs, and their intracellular signalling components, constitute an important cellular pathway mediating the inflammatory process. Moreover, their critical role in the regulation of tissue injury and wound healing process as well as in the regulation of apoptosis is well established. However, interest in the role of these receptors in cancer development and progression has been increasing over the last years. TLRs are likely candidates to mediate effects of the innate immune system within the tumour microenvironment. A rapidly expanding area of research regarding the expression and function of TLRs in cancer cells and its association with chemoresistance and tumourigenesis, and TLR-based therapy as potential immunotherapy in cancer treatment is taking place over the last years.
Collapse
|
8
|
Cai Q, Kublo L, Cumberland R, Gooding W, Baar J. Optimized systemic dosing with CpG DNA enhances dendritic cell-mediated rejection of a poorly immunogenic mammary tumor in BALB/c mice. Clin Transl Sci 2010; 2:62-6. [PMID: 20443869 DOI: 10.1111/j.1752-8062.2008.00073.x] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/15/2023] Open
Abstract
To model a clinical trial of dendritic cell (DC) therapy of a poorly immunogenic mammary tumor, we treated BALB/c mice bearing an established TS/A mammary tumor with lysate-pulsed DCs and CpG DNA. We observed that the dose of CpG DNA required to activate DCs in vitro was insufficient to mediate tumor rejection in vivo. We therefore undertook in vivo studies to identify an optimized dose of CpG DNA for tumor therapy, defined as the lowest and least frequently administered dose of CpG DNA that mediated complete tumor rejection. We show that one priming dose of 15 nanomoles and one booster dose of 10 nanomoles of CpG DNA given 7 days apart, respectively, with lysate-loaded DCs were sufficient to mediate complete tumor rejection in vivo. This dose of CpG DNA was 42-fold higher than that required to activate DCs in vitro but was not associated with any toxicity in mice. Also, the cured mice rejected a subsequent challenge with fresh TS/A tumor, and both CD4(+) and CD8(+) T cells were required for tumor rejection. We conclude that effective DC-based therapy of a poorly immunogenic TS/A tumor is enhanced by optimized dosing of CpG DNA. Our data have important implications for DC-based clinical trials of breast cancer immunotherapy.
Collapse
Affiliation(s)
- Quan Cai
- Hillman Cancer Center, University of Pittsburgh Cancer Institute, Pittsburgh, Pennsylvania, USA
| | | | | | | | | |
Collapse
|
9
|
Chiu YG, Bowers WJ, Lim ST, Ryan DA, Federoff HJ. Effects of herpes simplex virus amplicon transduction on murine dendritic cells. Hum Gene Ther 2010; 20:442-52. [PMID: 19199821 DOI: 10.1089/hum.2008.160] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022] Open
Abstract
The herpes simplex virus (HSV)-based amplicon is a versatile vaccine platform that has been preclinically vetted as a gene-based immunotherapeutic for cancer, HIV, and neurodegenerative disorders. Although it is well known that injection of dendritic cells (DCs) transduced ex vivo with helper virus-free HSV amplicon vectors expressing disease-relevant antigens induces antigen-specific immune responses, the cellular receptor(s) by which the amplicon virion gains entry into DCs, as well as the effects that viral vector transduction impinges on the physiological status of these cells, is less understood. Herein, we examine the effects of amplicon transduction on mouse bone marrow-derived DCs. We demonstrate that HSV-1 cellular receptors HveC and HveA are expressed on the cell surface of murine DCs, and that HSV amplicons transduce DCs at high efficiency (>90%) with minimal effects on cell viability. Transduction of dendritic cells with amplicons induces a transient DC maturation phenotype as represented by self-limited upregulation of MHCII and CD11c markers. Mature DCs are less sensitive to HSV amplicon transduction than immature DCs regarding DC-related surface marker maintenance. From this and our previous work, we conclude that HSV amplicons transduce DCs efficiently, but impart differential and transient physiological effects on mature and immature DC pools, which will facilitate fine-tuning of this vaccination platform and further exploit its potential in immunotherapy.
Collapse
Affiliation(s)
- Yahui Grace Chiu
- Division of Allergy, Immunology, and Rheumatology, Department of Medicine, University of Rochester School of Medicine and Dentistry, Rochester, NY 14642, USA
| | | | | | | | | |
Collapse
|
10
|
Abstract
Advances in the understanding of the immunoregulatory functions of dendritic cells (DCs) in animal models and humans have led to their exploitation as anticancer vaccines. Although DC-based immunotherapy has proven clinically safe and efficient to induce tumor-specific immune responses, only a limited number of objective clinical responses have been reported in cancer patients. These relatively disappointing results have prompted the evaluation of multiple approaches to improve the efficacy of DC vaccines. The topic of this review focuses on personalized DC-based anticancer vaccines, which in theory have the potential to present to the host immune system the entire repertoire of antigens harbored by autologous tumor cells. We also discuss the implementation of these vaccines in cancer therapeutic strategies, their limitations and the future challenges for effective immunotherapy against cancer.
Collapse
Affiliation(s)
- Nona Janikashvili
- Department of Pediatrics, Steele Children’s Research Center, Arizona 85724, USA
| | - Nicolas Larmonier
- Department of Pediatrics, Steele Children’s Research Center, Arizona 85724, USA
- Department of Immunobiology, BIO5 Institute & Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA
| | - Emmanuel Katsanis
- Department of Pediatrics, Steele Children’s Research Center, Arizona 85724, USA
- Department of Immunobiology, BIO5 Institute & Arizona Cancer Center, University of Arizona, Tucson, AZ 85724, USA
- University of Arizona, Department of Pediatrics, 1501 N Campbell Ave, PO Box 245073, Tucson, AZ 85724-85073, USA
| |
Collapse
|
11
|
Mann DL, Celluzzi CM, Hankey KG, Harris KM, Watanabe R, Hasumi K. Combining conventional therapies with intratumoral injection of autologous dendritic cells and activated T cells to treat patients with advanced cancers. Ann N Y Acad Sci 2009; 1174:41-50. [PMID: 19769735 DOI: 10.1111/j.1749-6632.2009.04934.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022]
Abstract
Dendritic cells (DCs) are potent antigen-presenting cells that have been used in cancer immunotherapy. To take advantage of the ability of DCs to acquire antigenic materials from their environment and generate primary as well as recall immune responses, 37 patients with advanced cancers were enrolled in a series of protocols based on direct intratumoral injection of immature DCs. To augment antigen uptake and antitumor immune response, DC injection was combined with radiotherapy or chemotherapy and/or injection of activated T cells. Treatments were well tolerated with no adverse reactions. Clinical responses were based on Response Evaluation Criteria in Solid Tumors, with the majority of patients showing stable disease. One of two patients who also received local radiation achieved a sustained complete response at injected and metastatic sites. The clinical responses observed in cancer patients with advanced disease suggest potential effectiveness of combination strategies and establish the basis for the current treatment protocol that is underway.
Collapse
Affiliation(s)
- Dean L Mann
- Department of Pathology, University of Maryland School of Medicine, Baltimore, MD 21201, USA.
| | | | | | | | | | | |
Collapse
|
12
|
Riley CL, Mathieu MG, Clark RE, McArdle SEB, Rees RC. Tumour antigen-targeted immunotherapy for chronic myeloid leukaemia: is it still viable? Cancer Immunol Immunother 2009; 58:1489-99. [PMID: 19259670 PMCID: PMC11029949 DOI: 10.1007/s00262-009-0675-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2008] [Accepted: 01/29/2009] [Indexed: 01/07/2023]
Abstract
In haematological cancers, malignant cells circulate in the blood and lymphatic system. This may make leukaemic cells easier to target by immunotherapy than in other types of cancer. Various immunotherapy strategies have been trialled in several leukaemias including chronic myeloid leukaemia (CML) and in general, these have been aimed at targeting tumour-associated antigens (TAA). There are numerous TAA expressed by CML patients including WT1, proteinase 3, BCR-ABL and HAGE amongst others. The immunogenicity of the CML-specific tumour antigen, BCR-ABL, has been the subject of much debate and its role in the development of the disease and its unique sequence spanning the breakpoint region make it an ideal target for immunotherapy. However, there are a limited number of immunogenic epitopes across the junctional region, which are restricted to only a few HLA types, namely A2, A3 and B7 (Clark et al. in Blood 98:2887-2893, 2001). The second CML-associated antigen is the helicase antigen HAGE, a cancer-testis antigen found to be over-expressed in more than 50% of myeloid leukaemias (Adams et al. in Leukaemia 16:2238-2242, 2002). Very little is known about the function of this antigen and its significance to CML. However, its membership of the DEAD-box family of ATP-dependent RNA helicases and the involvement of other members of this family in tumour cell proliferation (Eberle et al. in Br J Cancer 86:1957-1962, 2002; Yang et al. in Cell Signal 17:1495-504, 2005) suggest a crucial role in the RNA metabolism of tumour cells. For these reasons, HAGE also seems to be a good target for immunotherapy as it would be applicable for the majority of patients with CML. This review aims to discuss the potential of immunotherapy for the treatment of leukaemia, in particular CML, and the prospect of targeting three CML associated antigens: BCR, ABL and HAGE. During his career, Prof. Tony Dodi made a significant contribution in this area of leukaemia research, confirming the identity of immunogenic HLA-A3 and B7-restricted peptides as targets for CTL. Published, as a highlighted paper in Clark et al. (Blood 98:2887-2893, 2001), this study demonstrated the expression of MHC-peptide complexes on the surface of CML cells and the presence of tetramer-positive CTL activity in CML patients positive for these two HLA alleles. His drive and dedication for research excellence will be remembered by all who knew and worked with him.
Collapse
Affiliation(s)
- Catherine L. Riley
- The John Van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Clifton Lane, Nottingham, NG11 8NS UK
| | - Morgan G. Mathieu
- The John Van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Clifton Lane, Nottingham, NG11 8NS UK
| | - Richard E. Clark
- Department of Haematology, Royal Liverpool University Hospital, Prescott Street, Liverpool, L7 8XP UK
| | - Stephanie E. B. McArdle
- The John Van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Clifton Lane, Nottingham, NG11 8NS UK
| | - Robert C. Rees
- The John Van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Clifton Lane, Nottingham, NG11 8NS UK
| |
Collapse
|
13
|
Inhibition of activation-induced death of dendritic cells and enhancement of vaccine efficacy via blockade of MINOR. Blood 2009; 113:2906-13. [PMID: 19164597 DOI: 10.1182/blood-2008-08-176354] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
Activation of dendritic cells (DCs) leads to cell maturation, which is accompanied by a regulated pattern of gene expression changes. Two significant and contradictory consequences of DC activation are that, although activation is necessary for maximal T-cell stimulation, it also leads to the initiation of gene expression that results ultimately in cell death. We have identified a gene, MINOR (mitogen-inducible nuclear orphan receptor), that becomes highly up-regulated on activation and whose expression leads to apoptosis in mature DCs. MINOR is a member of the Nur77 family of nuclear orphan receptors, which includes Nur77 and Nurr1. Although Nur77 and Nurr1 are expressed in macrophages and DCs, their expression levels do not change on DC activation. We thus tested the hypothesis that induction of MINOR would lead to an activation-induced cell death in DCs and that its inhibition would increase the lifespan of DCs and improve their vaccine efficacy. To block natural expression of MINOR by DCs, we generated a lentiviral vector that expresses a small interfering RNA. Our results indicate that blockade of MINOR expression dramatically decreases apoptosis in DCs and suggest that this approach may be a novel means to improve the potency of ex vivo-generated DC vaccines.
Collapse
|
14
|
Cai Q, Kublo L, Cumberland R, Gooding W, Baar J. Optimized Systemic Dosing with CpG DNA Enhances Dendritic Cell-Mediated Rejection of a Poorly Immunogenic Mammary Tumor in BALB/c Mice. Clin Transl Sci 2009. [DOI: 10.1111/j.1752-8062.2009.00073.x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
|
15
|
Huarte E, Cubillos-Ruiz JR, Nesbeth YC, Scarlett UK, Martinez DG, Buckanovich RJ, Benencia F, Stan RV, Keler T, Sarobe P, Sentman CL, Conejo-Garcia JR. Depletion of dendritic cells delays ovarian cancer progression by boosting antitumor immunity. Cancer Res 2008; 68:7684-91. [PMID: 18768667 PMCID: PMC2742361 DOI: 10.1158/0008-5472.can-08-1167] [Citation(s) in RCA: 97] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
Dendritic cells (DC) and cytokines that expand myeloid progenitors are widely used to treat cancer. Here, we show that CD11c(+)DEC205(+) DCs coexpressing alpha-smooth muscle actin and VE-cadherin home to perivascular areas in the ovarian cancer microenvironment and are required for the maintenance of tumor vasculature. Consequently, depletion of DCs in mice bearing established ovarian cancer by targeting different specific markers significantly delays tumor growth and enhances the effect of standard chemotherapies. Tumor growth restriction was associated with vascular apoptosis after DC ablation followed by necrosis, which triggered an antitumor immunogenic boost. Our findings provide a mechanistic rationale for selectively eliminating tumor-associated leukocytes to promote antitumor immunity while impeding tumor vascularization and to develop more effective DC vaccines based on a better understanding of the tumor microenvironment.
Collapse
Affiliation(s)
- Eduardo Huarte
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, NH 03756, USA
| | - Juan R. Cubillos-Ruiz
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, NH 03756, USA
| | - Yolanda C. Nesbeth
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, NH 03756, USA
| | - Uciane K. Scarlett
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, NH 03756, USA
| | - Diana G. Martinez
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, NH 03756, USA
| | - Ronald J. Buckanovich
- Department of Internal Medicine, University of Michigan, Ann Arbor, MI 48109, USA
- Department of Obstetrics and Gynecology, University of Michigan, Ann Arbor, MI 48109, USA
| | - Fabian Benencia
- Department of Biomedical Sciences, Ohio University, Athens, OH 45701, USA
| | - Radu V. Stan
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, NH 03756, USA
- Department of Pathology, Dartmouth Medical School, Lebanon, NH 03756, USA
| | - Tibor Keler
- Celldex Therapeutics, Phillipsburg, NJ 08865, USA
| | - Pablo Sarobe
- CIMA, University of Navarra, Pamplona, Navarra 31008, Spain
| | - Charles L. Sentman
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, NH 03756, USA
| | - Jose R. Conejo-Garcia
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, NH 03756, USA
- Department of Medicine, Dartmouth Medical School, Lebanon, NH 03756, USA
| |
Collapse
|
16
|
Yoon TJ, Kim JY, Kim H, Hong C, Lee H, Lee CK, Lee KH, Hong S, Park SH. Anti-tumor immunostimulatory effect of heat-killed tumor cells. Exp Mol Med 2008; 40:130-44. [PMID: 18305406 DOI: 10.3858/emm.2008.40.1.130] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
As a part of our ongoing search for a safe and efficient anti-tumor vaccine, we attempted to determine whether the molecular nature of certain tumor antigens would influence immune responses against tumor cells. As compared with freeze-thawed or formaldehyde-fixed tumor antigens, heat-denatured tumor antigens elicited profound anti-tumor immune responses and greatly inhibited the growth of live tumor cells. The heat-denatured tumor antigens induced a substantial increase in the anti-tumor CTL response in the absence of any adjuvant material. This response appears to be initiated by strong activation of the antigen-presenting cells, which may recognize heat-denatured protein antigens. Upon recognition of the heat-denatured tumor antigens, macrophages and dendritic cells were found to acutely upregulate the expression of co-stimulatory molecules such as B7.2, as well as the secretion of inflammatory cytokines such as IL-12 and TNF-alpha. The results of this study indicate that heat-denatured tumor extracts might elicit protective anti-tumor adaptive immune responses and also raise the possibility that a safe and efficient adjuvant-free tumor vaccine might be developed in conjunction with a dendritic cell-based tumor vaccine.
Collapse
|
17
|
Dörrie J, Schaft N, Müller I, Wellner V, Schunder T, Hänig J, Oostingh GJ, Schön MP, Robert C, Kämpgen E, Schuler G. Introduction of functional chimeric E/L-selectin by RNA electroporation to target dendritic cells from blood to lymph nodes. Cancer Immunol Immunother 2008; 57:467-77. [PMID: 17768622 PMCID: PMC11041385 DOI: 10.1007/s00262-007-0385-1] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/09/2007] [Accepted: 07/30/2007] [Indexed: 10/22/2022]
Abstract
BACKGROUND Inefficient migration of dendritic cells (DC) to regional lymph nodes (LN) upon intracutaneous injection is a major obstacle for effective DC vaccination. Intravenous vaccination is unfavorable, because DC cannot migrate directly from the blood into LN. METHODS To enable human monocyte-derived (mo)DC to enter LN directly from the blood, we manipulated them by RNA electroporation to express a human chimeric E/L-selectin (CD62E/CD62L) protein, which binds to peripheral node addressin expressed on high endothelial venules. RESULTS Transfection efficiency exceeded 95%, and high E/L-selectin surface expression was detected for >48 h. E/L-selectin RNA-transfected DC displayed an identical mature DC phenotype as mock-transfected DC. Furthermore, E/L-selectin-transfected DC maintained their normal CCR7-mediated migration capacity, and their ability to prime and expand functional cytotoxic T cells recognizing MelanA. Most importantly, E/L-selectin-RNA-transfected DC gained the capability to attach to and roll on sialyl-Lewis(X) in vitro. OUTLOOK The presented strategy can be readily translated into the clinic, as it involves no stable genetic manipulation or viral transformation, and allows targeting of a large number of LN.
Collapse
Affiliation(s)
- Jan Dörrie
- Department of Dermatology, University Hospital Erlangen, Hartmannstr. 14, 91052 Erlangen, Germany
| | - Niels Schaft
- Department of Dermatology, University Hospital Erlangen, Hartmannstr. 14, 91052 Erlangen, Germany
| | - Ina Müller
- Department of Dermatology, University Hospital Erlangen, Hartmannstr. 14, 91052 Erlangen, Germany
| | - Verena Wellner
- Department of Dermatology, University Hospital Erlangen, Hartmannstr. 14, 91052 Erlangen, Germany
| | - Tanja Schunder
- Department of Dermatology, University Hospital Erlangen, Hartmannstr. 14, 91052 Erlangen, Germany
| | - Jens Hänig
- Department of Dermatology, University Hospital Erlangen, Hartmannstr. 14, 91052 Erlangen, Germany
| | - Gertie J. Oostingh
- Rudolf Virchow Center, DFG Research Center for Experimental Biomedicine
and Department of Dermatology, University of Würzburg, Würzburg, Germany
| | - Michael P. Schön
- Rudolf Virchow Center, DFG Research Center for Experimental Biomedicine
and Department of Dermatology, University of Würzburg, Würzburg, Germany
| | | | - Eckhart Kämpgen
- Department of Dermatology, University Hospital Erlangen, Hartmannstr. 14, 91052 Erlangen, Germany
| | - Gerold Schuler
- Department of Dermatology, University Hospital Erlangen, Hartmannstr. 14, 91052 Erlangen, Germany
| |
Collapse
|
18
|
Abstract
Cell-based therapies may gain future importance in defeating different kinds of diseases, including cancer, immunological disorders, neurodegenerative diseases, cardiac infarction and stroke. In this context, the noninvasive localization of the transplanted cells and the monitoring of their migration can facilitate basic research on the underlying mechanism and improve clinical translation. In this chapter, different ways to label and track cells in vivo are described. The oldest and only clinically established method is leukocyte scintigraphy, which enables a (semi)quantitative assessment of cell assemblies and, thus, the localization of inflammation foci. Noninvasive imaging of fewer or even single cells succeeds with MRI after labeling of the cells with (ultrasmall) superparamagentic iron oxide particles (SPIO and USPIO). However, in order to gain an acceptable signal-to-noise ratio, at a sufficiently high spatial resolution of the MR sequence to visualize a small amount of cells, experimental MR scanners working at high magnetic fields are usually required. Nevertheless, feasibility of clinical translation has been achieved by showing the localization of USPIO-labeled dendritic cells in cervical lymph nodes of patients by clinical MRI.Cell-tracking approaches using optical methods are important for preclinical research. Here, cells are labeled either with fluorescent dyes or quantum dots, or transfected with plasmids coding for fluorescent proteins such as green fluorescent protein (GFP) or red fluorescent protein (RFP). The advantage of the latter approach is that the label does not get lost during cell division and, thus, makes imaging of proliferating transplanted cells (e.g., tumor cells) possible. In summary, there are several promising options for noninvasive cell tracking, which have different strengths and limitations that should be considered when planning cell-tracking experiments.
Collapse
Affiliation(s)
- Fabian Kiessling
- Abteilung Medizinische Physik in der Radiologie, Deutsches Krebsforschungszentrum, Im Neuenheimer Feld 280, 69120 Heidelberg.
| |
Collapse
|
19
|
Monsurrò V, Marincola FM. Gene profiling for the prediction of tumor response to treatment: the case of immunotherapy. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2007; 593:86-94. [PMID: 17265719 DOI: 10.1007/978-0-387-39978-2_9] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 05/13/2023]
Affiliation(s)
- Vladia Monsurrò
- Department of Tranfusion Medicine, Immunogenetics Section, National Institutes of Health, Bethesda, Maryland 20892, USA
| | | |
Collapse
|
20
|
Garay RP, Viens P, Bauer J, Normier G, Bardou M, Jeannin JF, Chiavaroli C. Cancer relapse under chemotherapy: why TLR2/4 receptor agonists can help. Eur J Pharmacol 2007; 563:1-17. [PMID: 17383632 DOI: 10.1016/j.ejphar.2007.02.018] [Citation(s) in RCA: 90] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2006] [Revised: 02/07/2007] [Accepted: 02/08/2007] [Indexed: 01/16/2023]
Abstract
Liver or lung metastases usually relapse under chemotherapy. Such life-threatening condition urgently needs new, systemic anticancer compounds, with original and efficient mechanisms of action. In B16 melanoma mice treated with cyclophosphamide, D'Agostini et al. [D'Agostini, C., Pica, F., Febbraro, G., Grelli, S., Chiavaroli, C., Garaci, E., 2005. Antitumour effect of OM-174 and Cyclophosphamide on murine B16 melanoma in different experimental conditions. Int. Immunopharmacol. 5, 1205-1212.] recently found that OM-174, a chemically defined Toll-like receptor(TLR)2/4 agonist, reduces tumor progression and prolongs survival. Here we review 149 articles concerning molecular mechanisms of TLR2/4 agonists, alone or in combination with chemotherapy. It appears that TLR2/4 agonists induce a well controlled tumor necrosis factor-alpha (TNF-alpha) secretion, at plasma levels known to permeabilize neoangiogenic tumor vessels to the passage of cytotoxic drugs. Moreover, TLR2/4 agonists induce inducible nitric oxide synthase (iNOS) expression, and nitric oxide is able to induce apoptosis of chemotherapy-resistant tumor cell clones. Finally, TLR2/4-stimulation activates dendritic cell traffic and its associated tumor-specific, cytotoxic T-cell responses. Therefore, parenteral TLR2/4 agonists seem promising molecules to prolong survival in cancer patients who relapse under chemotherapy.
Collapse
|
21
|
Kim R, Emi M, Tanabe K. Functional roles of immature dendritic cells in impaired immunity of solid tumour and their targeted strategies for provoking tumour immunity. Clin Exp Immunol 2007; 146:189-96. [PMID: 17034569 PMCID: PMC1942049 DOI: 10.1111/j.1365-2249.2006.03215.x] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
Dendritic cells play a crucial role in initiating tumour immunity as well as in the immune response for invading foreign pathogens such as bacteria and viruses. For bacterial and viral infections, the immature dendritic cells (iDCs) residing in peripheral tissues are efficiently activated and matured by pathogen signals for performing the immune response. In contrast, for self-antigens, the naive T cells are not activated by iDCs but proceed to anergy/deletion, and the generation of regulatory T cells for immune tolerance. The induction of immune response and tolerance is regulated strictly by iDCs as the sensor for homeostasis of immune response in the host. Despite the identification of some tumour antigens, tumour immunity is not provoked successfully. Even though there are some critical obstacles to inhibit effective tumour immunity, tumour cells are able to exploit the functional roles of iDCs for tumour progression, which are induced by tumour-derived soluble factors such as vascular endothelial growth factor (VEGF) and functionally modulated in the microenvironment. The iDCs still remain as the critical target for provoking tumour immunity. In this review, the functional roles of tumour-associated iDCs and the strategy for targeting iDCs in effective tumour immunity for the cancer patient are discussed.
Collapse
Affiliation(s)
- R Kim
- International Radiation Information Center, Research Institute for Radiation Biology and Medicine, Hiroshima University, Hiroshima, Japan.
| | | | | |
Collapse
|
22
|
Altin JG, Parish CR. Liposomal vaccines--targeting the delivery of antigen. Methods 2007; 40:39-52. [PMID: 16997712 DOI: 10.1016/j.ymeth.2006.05.027] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2006] [Accepted: 05/05/2006] [Indexed: 01/05/2023] Open
Abstract
Vaccines that can prime the adaptive immune system for a quick and effective response against a pathogen or tumor cells, require the generation of antigen (Ag)-specific memory T and B cells. The unique ability of dendritic cells (DCs) to activate naïve T cells, implies a key role for DCs in this process. The generation of tumor-specific CD8(+) cytotoxic T cells (CTLs) is dependent on both T cell stimulation with Ag (peptide-MHC-complexes) and costimulation. Interestingly, tumor cells that lack expression of T cell costimulatory molecules become highly immunogenic when transfected to express such molecules on their surface. Adoptive immunotherapy with Ag-pulsed DCs also is a strategy showing promise as a treatment for cancer. The use of such cell-based vaccines, however, is cumbersome and expensive to use clinically, and/or may carry risks due to genetic manipulations. Liposomes are particulate vesicular lipid structures that can incorporate Ag, immunomodulatory factors and targeting molecules, and hence can serve as potent vaccines. Similarly, Ag-containing plasma membrane vesicles (PMV) derived from tumor cells can be modified to incorporate a T cell costimulatory molecule to provide both TCR stimulation, and costimulation. PMVs also can be modified to contain IFN-gamma and molecules for targeting DCs, permitting delivery of both Ag and a DC maturation signal for initiating an effective immune response. Our results show that use of such agents as vaccines can induce potent anti-tumor immune responses and immunotherapeutic effects in tumor models, and provide a strategy for the development of effective vaccines and immunotherapies for cancer and infectious diseases.
Collapse
Affiliation(s)
- Joseph G Altin
- School of Biochemistry and Molecular Biology, Faculty of Science, The Australian National University, Canberra, ACT 0200, Australia.
| | | |
Collapse
|
23
|
Abstract
There is overwhelming evidence that the human immune system can keep in check the growth of autologous tumors. Yet, this phenomenon is rare and most often tumors survive striking a balance with the host's immune system. The well-documented coexistence of immune cells that can recognize cancer and their targets within the same host is reminiscent of chronic allograft rejection well-controlled by immune suppression or of a lingering tissue-specific autoimmune reaction. In this review, we argue that autologous tumor rejection represents a distinct form of tissue-specific rejection similar to acute allograft rejection or to flares of autoimmunity. Here we discuss similarities within the biology of these phenomena that may converge into a common immunological constant of rejection. The purpose is to simplify the basis of immune rejection to its bare bones critically dissecting the significance of those components proposed by experimental models as harbingers of this final outcome.
Collapse
Affiliation(s)
- Ena Wang
- Immunogenetics Section, The Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, Maryland, USA
| | | | | |
Collapse
|
24
|
McBride S, Hoebe K, Georgel P, Janssen E. Cell-Associated Double-Stranded RNA Enhances Antitumor Activity through the Production of Type I IFN. THE JOURNAL OF IMMUNOLOGY 2006; 177:6122-8. [PMID: 17056539 DOI: 10.4049/jimmunol.177.9.6122] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Abstract
The efficacy of tumor cell vaccination largely depends on the maturation and activation status of the dendritic cell. Here we investigated the ability of soluble and tumor cell-associated dsRNA to serve as an adjuvant in the induction of protective adaptive antitumor responses. Our data showed that cell-associated dsRNA, but not soluble dsRNA, enhanced both tumor-specific CD8(+) and CD4(+) T cell responses. The cell-associated dsRNA increased the clonal burst of tumor-specific CD8(+) T cells and endowed them with an enhanced capacity for expansion upon a secondary encounter with tumor Ags, even when the CD8(+) T cells were primed in the absence of CD4(+) T cell help. The adjuvant effect of cell-associated dsRNA was fully dependent on the expression of TLR3 by the APCs and their subsequent production of type I IFNs, as the adjuvant effect of cell-associated dsRNA was completely abrogated in mice deficient in TLR3 or type I IFN signaling. Importantly, treatment with dsRNA-associated tumor cells increased the number of tumor-infiltrating lymphocytes and enhanced the survival of tumor-bearing mice. The data from our studies suggest that using cell-associated dsRNA as a tumor vaccine adjuvant may be a suitable strategy for enhancing vaccine efficacy for tumor cell therapy in cancer patients.
Collapse
Affiliation(s)
- Sara McBride
- La Jolla Institute for Allergy and Immunology, Developmental Immunology 1B, 9420 Athena Circle, La Jolla, CA 92037, USA
| | | | | | | |
Collapse
|
25
|
Fushimi T, O'Connor TP, Crystal RG. Adenoviral gene transfer of stromal cell-derived factor-1 to murine tumors induces the accumulation of dendritic cells and suppresses tumor growth. Cancer Res 2006; 66:3513-22. [PMID: 16585175 DOI: 10.1158/0008-5472.can-05-1493] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
The human CXC chemokine, stromal cell-derived factor 1 (SDF-1alpha), is known to function in vitro as a chemotactic factor for lymphocytes, monocytes, and dendritic cells. In the context that dendritic cells are powerful antigen-presenting cells, we hypothesized that adenoviral gene transfer of SDF-1alpha to tumors might inhibit growth of preexisting tumors through attracting dendritic cells to the tumor. AdSDF-1alpha mediated the expression of SDF-1alpha mRNA and protein in A549 cells in vitro, and the supernatant of the AdSDF-1alpha-infected A549 cells showed chemotactic activity for dendritic cells. When syngeneic murine CT26 colon carcinoma tumors (BALB/c) and B16 melanoma and Lewis lung cell carcinoma (C57Bl/6) were injected with AdSDF-1alpha (5 x 10(8) plaque-forming units), there was an accumulation of dendritic cells and CD8(+) cells within the tumor and significant inhibition of tumor growth compared with tumors injected with PBS or AdNull (control vector). The injection of AdSDF-1alpha into tumors induced the inflammatory enlargement and the accumulation of dendritic cells in the draining lymph node. Intratumoral AdSDF-1alpha administration elicited tumor-specific CTLs and adoptive transfer of splenocytes from AdSDF-1alpha-treated mice resulted in the elongation of survival after tumor challenge. Interestingly, in wild-type and CD4(-/-) mice but not in CD8(-/-) mice, AdSDF-1alpha inhibited the growth of the tumor. These observations suggest that adenoviral gene transfer of SDF-1alpha may be a useful strategy to accumulate dendritic cells in tumors and evoke antitumor immune responses to inhibit tumor growth.
Collapse
MESH Headings
- Adenocarcinoma/genetics
- Adenocarcinoma/immunology
- Adenoviridae/genetics
- Animals
- Carcinoma, Lewis Lung/genetics
- Carcinoma, Lewis Lung/immunology
- Cell Growth Processes/immunology
- Chemokine CXCL12
- Chemokines, CXC/genetics
- Chemokines, CXC/immunology
- Colonic Neoplasms/genetics
- Colonic Neoplasms/immunology
- Dendritic Cells/cytology
- Dendritic Cells/immunology
- Female
- Gene Transfer Techniques
- Genetic Vectors/genetics
- Immunity, Cellular/genetics
- Immunity, Cellular/immunology
- Lymph Nodes/immunology
- Melanoma, Experimental/genetics
- Melanoma, Experimental/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/immunology
Collapse
Affiliation(s)
- Toshiaki Fushimi
- Division of Pulmonary and Critical Care Medicine, Weill Medical College of Cornell University, 515 East 71st Street, New York, NY 10021, USA
| | | | | |
Collapse
|
26
|
Fukui M, Ueno K, Suehiro Y, Hamanaka Y, Imai K, Hinoda Y. Anti-tumor activity of dendritic cells transfected with mRNA for receptor for hyaluronan-mediated motility is mediated by CD4+ T cells. Cancer Immunol Immunother 2006; 55:538-46. [PMID: 16025267 PMCID: PMC11030997 DOI: 10.1007/s00262-005-0027-4] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/15/2005] [Accepted: 05/05/2005] [Indexed: 11/29/2022]
Abstract
Receptor for hyaluronan-mediated motility (RHAMM) is overexpressed in various tumors with high frequency, and was recently identified as an immunogenic antigen by serologic screening of cDNA expression libraries. In this study, we explored whether RHAMM is a potential target for dendritic cell (DC) immunotherapy. We constructed a plasmid for transduction of in vitro-transcribed mRNAs into DCs to efficiently transport the intracellular protein RHAMM into MHC class II compartments by adding a late endosomal/lysosomal sorting signal to the RHAMM gene. Immunization of mice with modified RHAMM mRNA-transfected DCs (DC/RHAMM) induced killing activity against RHAMM-positive tumor cells in splenocytes. To examine whether CD4(+) and/or CD8(+) T cells were required for this antitumor immunity, an anti-CD4 or anti-CD8 antibody was administered to mice after immunization with DC/RHAMM. Depletion of CD4(+) T cells significantly diminished the induction of tumor cell-killing activity in splenocytes, whereas CD8(+) T cell depletion had no effect. We then investigated the therapeutic effect of DC/RHAMM in a 3-day tumor model of EL4. DC/RHAMM was administered to mice on days 3, 7 and 10 after EL4 tumor inoculation. The treatment markedly inhibited tumor growth compared to control DCs. Moreover, antibody-mediated depletion of CD4(+) T cells completely abrogated the therapeutic effect of DC/RHAMM, whereas depletion of CD8(+) T cells had no effect. The results of this preclinical study indicate that DCs transfected with a modified RHAMM mRNA targeted to MHC class II compartments can induce CD4(+) T cell-mediated antitumor activity in vivo.
Collapse
Affiliation(s)
- Mikiko Fukui
- Department of Clinical Laboratory Science, Yamaguchi University School of Medicine, 1-1-1, Minami-Kogushi, Ube, Yamaguchi, 755-8505 Japan
| | - Koji Ueno
- Department of Clinical Laboratory Science, Yamaguchi University School of Medicine, 1-1-1, Minami-Kogushi, Ube, Yamaguchi, 755-8505 Japan
| | - Yutaka Suehiro
- Department of Clinical Laboratory Science, Yamaguchi University School of Medicine, 1-1-1, Minami-Kogushi, Ube, Yamaguchi, 755-8505 Japan
| | - Yuichiro Hamanaka
- Department of Clinical Laboratory Science, Yamaguchi University School of Medicine, 1-1-1, Minami-Kogushi, Ube, Yamaguchi, 755-8505 Japan
| | - Kohzoh Imai
- Department of First Internal Medicine, Sapporo Medical University, Sapporo, Japan
| | - Yuji Hinoda
- Department of Clinical Laboratory Science, Yamaguchi University School of Medicine, 1-1-1, Minami-Kogushi, Ube, Yamaguchi, 755-8505 Japan
| |
Collapse
|
27
|
Legler DF, Krause P, Scandella E, Singer E, Groettrup M. Prostaglandin E2 is generally required for human dendritic cell migration and exerts its effect via EP2 and EP4 receptors. THE JOURNAL OF IMMUNOLOGY 2006; 176:966-73. [PMID: 16393982 DOI: 10.4049/jimmunol.176.2.966] [Citation(s) in RCA: 175] [Impact Index Per Article: 9.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Subscribe] [Scholar Register] [Indexed: 01/06/2023]
Abstract
The control of dendritic cell (DC) migration is pivotal for the initiation of cellular immune responses. In this study, we demonstrate that the migration of human monocyte-derived (Mo)DCs as well as of ex vivo peripheral blood DCs toward CCL21, CXCL12, and C5a is stringently dependent on the presence of the proinflammatory mediator PGE2, although DCs expressed CXCR4 and C5aR on their surface and DC maturation was accompanied by CCR7 up-regulation independently of PGE2. The necessity of exogenous PGE2 for DC migration is not due to the suppression of PGE2 synthesis by IL-4, which is used for MoDC differentiation, because maturation-induced endogenous production of PGE2 cannot promote DC migration. Surprisingly, PGE2 was absolutely required at early time points of maturation to enable MoDC chemotaxis, whereas PGE2 addition during terminal maturation events was ineffective. In contrast to mouse DCs, which exclusively rely on EP4 receptor triggering for migration, human MoDCs require a signal mediated by EP2 or EP4 either alone or in combination. Our results provide clear evidence that PGE2 is a general and mandatory factor for the development of a migratory phenotype of human MoDCs as well as for peripheral blood myeloid DCs.
Collapse
MESH Headings
- Base Sequence
- Cell Differentiation
- Cell Movement/drug effects
- Cell Movement/physiology
- Chemotaxis, Leukocyte/drug effects
- Chemotaxis, Leukocyte/physiology
- DNA, Complementary/genetics
- Dendritic Cells/cytology
- Dendritic Cells/drug effects
- Dendritic Cells/immunology
- Dendritic Cells/physiology
- Dinoprostone/biosynthesis
- Dinoprostone/pharmacology
- Dinoprostone/physiology
- Gene Expression/drug effects
- Granulocyte-Macrophage Colony-Stimulating Factor/pharmacology
- Humans
- In Vitro Techniques
- Interleukin-13/pharmacology
- Interleukin-4/pharmacology
- Membrane Proteins/genetics
- Receptor, Anaphylatoxin C5a
- Receptors, CCR7
- Receptors, CXCR4/genetics
- Receptors, Chemokine/genetics
- Receptors, Complement/genetics
- Receptors, Prostaglandin E/physiology
- Receptors, Prostaglandin E, EP2 Subtype
- Receptors, Prostaglandin E, EP4 Subtype
- Recombinant Proteins
- Signal Transduction
Collapse
|
28
|
Abstract
Melanoma incidence increases and conventional antitumor therapies are often ineffective, encouraging the design of novel therapies. Several lines of evidence support the notion of an immunological control of melanoma growth. Based on this information, active immunotherapy (vaccination) and adoptive immunotherapy trials (T cell therapy) were conducted in metastatic melanoma patients. The proof of principle of effective immunotherapy was brought up by pionnering trials using tumor infiltrated lymphocytes in lymphodepleted recipients or anti-CTLA4 Ab leading to tumor eradication but also autoimmune diseases. With the identification and characterization of tumor antigens recognized by cytotoxic T lymphocytes, the utilization of tumor rejection antigens along with adjuvants become available as tumor vaccines. The last five years have witnessed the emergence of dendritic cell based-vaccines that were efficient in priming and/or boosting T cell responses in normal volunteers and patients. This review highlights preclinical bases of cancer vaccines, their clinical development and discusses their limits. Correlations between immunomonitoring and tumor regressions await larger trials.
Collapse
Affiliation(s)
- François Ghiringhelli
- Inserm U.517, Faculté de Médecine et Centre Régional de Lutte contre le Cancer Georges-Francois Leclerc, 7, boulevard Jeanne d'Arc, 21079 Dijon Cedex, France.
| | | |
Collapse
|
29
|
Abstract
The formulation of therapeutic strategies to enhance immune-mediated tumor destruction is a central goal of cancer immunology. Substantive progress toward delineating the mechanisms involved in innate and adaptive tumor immunity has improved the prospects for crafting efficacious treatments. Schemes under active clinical evaluation include cancer vaccines, monoclonal antibodies, recombinant cytokines, and adoptive cellular infusions. While these manipulations increase tumor immunity in many patients, the majority still succumbs to progressive disease. Detailed analysis of subjects on experimental protocols together with informative studies of murine tumor models have begun to clarify the parameters that determine therapeutic activity and resistance. These investigations have highlighted efficient dendritic cell activation and inhibition of negative immune regulation as central pathways for intervention. This review discusses the development of genetically modified whole tumor cell vaccines and antibody-blockade of cytotoxic T lymphocyte associated antigen-4 (CTLA-4) as immunotherapies targeting these key control points. Early-stage clinical testing raises the possibility that combinatorial approaches that augment dendritic cell-mediated tumor antigen presentation and antagonize negative immune regulation may accomplish significant tumor destruction without the induction of serious autoimmune disease.
Collapse
Affiliation(s)
- F Stephen Hodi
- Department of Medical Oncology, Dana-Farber Cancer Institute, Boston, Massachusetts, USA
| | | |
Collapse
|
30
|
Altin JG, van Broekhoven CL, Parish CR. Targeting dendritic cells with antigen-containing liposomes: antitumour immunity. Expert Opin Biol Ther 2005; 4:1735-47. [PMID: 15500402 DOI: 10.1517/14712598.4.11.1735] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Dendritic cells (DCs) are antigen-presenting cells that play an important role in the body's immune defence against cancer. Strategies using antigen-primed DCs as tumour vaccines show promise in patients, but the approach is cumbersome to use clinically. Soluble tumour antigens can be targeted to DCs in vivo, but this often induces antigenic tolerance rather than immunity. Liposomes are vesicular lipid structures with adjuvant-like properties. Importantly, liposomes can encapsulate antigen and immunomodulatory factors, thus serving as potent delivery vehicles. Different strategies are being explored to target liposomal antigens to DCs in vivo. One approach has employed single-chain antibody fragments to the DC surface molecules CD11c and DEC-205, attached to the vesicle surface by metal-chelating linkage, to target liposomal membranes containing antigen and either interferon-gamma or lipopolysaccharide to DCs. Such membranes induce dramatic antitumour responses and immunotherapeutic effects when used as a vaccine in the murine tumour model B16-OVA melanoma. Liposomal targeting of antigen and maturation signals directly to DCs in vivo, therefore, represents a much simpler strategy for cancer immunotherapy than antigen loading DCs ex vivo.
Collapse
Affiliation(s)
- Joseph G Altin
- The Australian National University, School of Biochemistry and Molecular Biology, Faculty of Science, Canberra, ACT 0200, Australia.
| | | | | |
Collapse
|
31
|
Tobiásová-Czetoová Z, Palmborg A, Lundqvist A, Karlsson G, Adamson L, Bartůnková J, Masucci G, Pisa P. Effects of human plasma proteins on maturation of monocyte-derived dendritic cells. Immunol Lett 2005; 100:113-9. [PMID: 16154491 DOI: 10.1016/j.imlet.2005.03.009] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2004] [Revised: 02/22/2005] [Accepted: 03/02/2005] [Indexed: 10/25/2022]
Abstract
Dendritic cells (DC) are a promising tool for vaccine therapy due to their unique properties as antigen presenting cells and their ability to prime naïve T cells. Increasing evidence suggests that maturation stage of DC critically influences the fate of the immune response. Generation of monocyte-derived DC for clinically applicable immunotherapy requires the use of well-defined components and stringent culture conditions. An alternative strategy is to use human autologous serum. However, its constituents are not stable and reflect the inflammatory condition of the donor. In order to investigate whether DC properties are influenced by proteins present in the plasma, we matured human monocyte-derived DC with four main plasma components: fibrinogen, fibronectin, plasminogen or C-reactive protein. These purified proteins were added at various concentrations on day 6 after the initial differentiation induced by IL-4 and GM-CSF. The maturation was assessed by phenotyping of maturation-associated marker (CD83) and co-stimulatory molecule CD86 as well as IL-12 production. Functional properties of DC were assessed by endocytic activity and mixed leukocyte culture. Our results indicate that fibrinogen had DC-maturation effect comparable to poly-I:C, TNF-alpha and PGE(2) as a positive control, but it failed to induce IL-12 production. The other plasma proteins had no effect on DC maturation. CRP at high concentration had rather inhibitory effect on DC induced lymphocyte function. We conclude that none of the tested plasma components and acute phase proteins sufficiently induce fully competent mature DC. This finding is important for the preparation of human DC-based vaccines supplemented by autologous sera.
Collapse
Affiliation(s)
- Z Tobiásová-Czetoová
- Department of Immunology, 2nd Medical Faculty and Faculty Hospital Motol, V Uvalu 84, 156 00 Prague 5, Czech Republic
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Viehl CT, Tanaka Y, Chen T, Frey DM, Tran A, Fleming TP, Eberlein TJ, Goedegebuure PS. Tat mammaglobin fusion protein transduced dendritic cells stimulate mammaglobin-specific CD4 and CD8 T cells. Breast Cancer Res Treat 2005; 91:271-8. [PMID: 15952060 DOI: 10.1007/s10549-005-0450-4] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Proteins can be efficiently introduced into cells when fused to a protein transduction domain, such as Tat from the human immunodeficiency virus. We recently reported that dendritic cells transduced with a Tat fusion protein containing the extracellular domain of Her2/neu (Tat-Her2/neu) induced CD8 cytotoxic T lymphocytes (CTL) that specifically lysed Her2/neu-expressing breast and ovarian cancer cells. In the current study we further investigated the mechanism of protein transduction, utilizing the breast cancer-associated protein, mammaglobin-A, which is expressed in about 80% of breast cancers. Using a Tat-mammaglobin fusion protein, we tested the ability of Tat-mammaglobin transduced dendritic cells to stimulate antigen-specific CD4 and CD8 T cells. Low levels of serum considerably improved protein transduction as determined by Western blot, and also improved presentation of antigenic peptide as evidenced by functional studies using antigen-specific T cells. Confocal microscope analyses of antigen-presenting cells (APC) incubated with Tat-mammaglobin showed localized distribution in addition to diffuse distribution in the cytosol. In contrast, mammaglobin lacking Tat showed only a localized distribution. Simultaneous incubation with both proteins resulted in overlapping localized distributions, suggesting Tat fusion proteins are processed through both the MHC class I and class II pathways. Indeed, stimulation of T cells with Tat-mammaglobin transduced dendritic cells led to an expansion of mammaglobin-specific CD4 T helper-1 lymphocytes along with CD8 CTL. We conclude that Tat-mammaglobin transduced dendritic cells can induce both CD4 and CD8 mammaglobin-specific T cells. These findings could be further exploited for the development of a mammaglobin-based vaccine for breast cancer.
Collapse
Affiliation(s)
- Carsten T Viehl
- Department of Surgery, Washington University School of Medicine, Saint Louis, MO 63110, USA
| | | | | | | | | | | | | | | |
Collapse
|
33
|
Moran TP, Collier M, McKinnon KP, Davis NL, Johnston RE, Serody JS. A Novel Viral System for Generating Antigen-Specific T Cells. THE JOURNAL OF IMMUNOLOGY 2005; 175:3431-8. [PMID: 16116238 DOI: 10.4049/jimmunol.175.5.3431] [Citation(s) in RCA: 39] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Dendritic cell (DC)-based vaccines are increasingly used for the treatment of patients with malignancies. Although these vaccines are typically safe, consistent and lasting generation of tumor-specific immunity has been rarely demonstrated. Improved methods for delivering tumor Ags to DCs and approaches for overcoming tolerance or immune suppression to self-Ags are critical for improving immunotherapy. Viral vectors may address both of these issues, as they can be used to deliver intact tumor Ags to DCs, and have been shown to inhibit the suppression mediated by CD4+CD25+ regulatory T cells. We have evaluated the potential use of Venezuelan equine encephalitis virus replicon particles (VRPs) for in vitro Ag delivery to human monocyte-derived DCs. VRPs efficiently transduced immature human DCs in vitro, with approximately 50% of immature DCs expressing a vector-driven Ag at 12 h postinfection. VRP infection of immature DCs was superior to TNF-alpha treatment at inducing phenotypic maturation of DCs, and was comparable to LPS stimulation. Additionally, VRP-infected DC cultures secreted substantial amounts of the proinflammatory cytokines IL-6, TNF-alpha, and IFN-alpha. Finally, DCs transduced with a VRP encoding the influenza matrix protein (FMP) stimulated 50% greater expansion of FMP-specific CD8+ CTL when compared with TNF-alpha-matured DCs pulsed with an HLA-A*0201-restricted FMP peptide. Thus, VRPs can be used to deliver Ags to DCs resulting in potent stimulation of Ag-specific CTL. These findings provide the rationale for future studies evaluating the efficacy of VRP-transduced DCs for tumor immunotherapy.
Collapse
Affiliation(s)
- Timothy P Moran
- Department of Microbiology and Immunology, University of North Carolina, Chapel Hill 27599-7295, USA
| | | | | | | | | | | |
Collapse
|
34
|
|
35
|
Groh V, Li YQ, Cioca D, Hunder NN, Wang W, Riddell SR, Yee C, Spies T. Efficient cross-priming of tumor antigen-specific T cells by dendritic cells sensitized with diverse anti-MICA opsonized tumor cells. Proc Natl Acad Sci U S A 2005; 102:6461-6. [PMID: 15824323 PMCID: PMC1088382 DOI: 10.1073/pnas.0501953102] [Citation(s) in RCA: 68] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/31/2022] Open
Abstract
Dendritic cells (DCs) have the capacity to prime tumor-specific T cell responses and are considered as potentially effective vaccines for immunotherapy of cancer. Critical parameters in the development of DC vaccines are the source of tumor antigen (TA) and the mode of DC-loading. Whole tumor cells contain complex assortments of TA, which has been exploited to enhance cross-presentation to CD8 T cells by DCs loaded with anti-syndecan mAb-opsonized myeloma cells. This approach may be broadly improved by targeting the MHC class I chain-related protein A (MICA), which is frequently and abundantly expressed on most if not all types of epithelial cancers but not in normal tissues except intestinal mucosa. Loading of DC with anti-MICA mAb-coated breast, melanoma, or ovarian tumor lines or uncultured ovarian cancer cells efficiently promoted TA cross-presentation and priming of multivalent anti-tumor CD8 and CD4 T cell responses. These were of substantially greater breadth and magnitude than those of T cells primed by peptide-pulsed or apoptotic tumor cell-loaded DCs. These results may advance DC vaccine development and provide a platform for adoptive T cell therapy and TA discovery. These results further suggest that antibody targeting of MICA might be applicable to elicit T cell immunity against tumors of diverse tissue origins in cancer patients.
Collapse
Affiliation(s)
- Veronika Groh
- Clinical Research Division, Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, Seattle, WA 98109, USA.
| | | | | | | | | | | | | | | |
Collapse
|
36
|
Panelli MC, Wang E, Monsurrò V, Jin P, Zavaglia K, Smith K, Ngalame Y, Marincola FM. Vaccination with T cell-defined antigens. Expert Opin Biol Ther 2005; 4:697-707. [PMID: 15155161 DOI: 10.1517/14712598.4.5.697] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Tumour immunology encompasses a broad array of biological phenomena including interactions between neoplastic cells and the innate and adaptive immune response. Among immune cells, T cells have taken the centre stage because they can be easily demonstrated to specifically recognise autologous cancer cells. As most tumour-associated antigens are intracellular proteins, T cells appear to be the most suitable tool for cancer-specific attack, as antibodies do not cross the cell membrane and the innate immune response lacks the same level of specificity. Finally, the relative ease in which T cells can be educated through antigen-specific immunisation to recognise cancer cells has elevated them to an even higher stature. In this review, it will be argued that T cells represent a unique anticancer agent, characterised by absolute specificity. Although other therapeutic modalities (antibody-based) have been effectively implemented, a comparison of T cell-based approaches with other modalities goes beyond the purposes of this review and will not be included in the discussion. However, it is obvious that the role of the T cell is limited and other components of the immune response (effector mononuclear phagocytes, natural killer cells, cytokines, chemokines, soluble factors), genetic background and tumour heterogeneity are likely to be necessary for the completion of cancer rejection.
Collapse
Affiliation(s)
- Monica C Panelli
- Immunogenetics Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | |
Collapse
|
37
|
Wang E, Panelli MC, Marincola FM. Understanding the response to immunotherapy in humans. ACTA ACUST UNITED AC 2005; 27:105-17. [PMID: 15666150 DOI: 10.1007/s00281-004-0198-7] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/23/2004] [Accepted: 12/15/2004] [Indexed: 01/08/2023]
Abstract
Whether the efforts of the last decade aimed at the development of vaccines against tumor-specific antigens encountered success or failure is a matter of expectations. On the bright side, we could optimistically observe that anti-cancer-vaccines stand as an outstanding example of the successful implementation of modern biotechnology tools for the development of biologically sound therapeutics. In particular, vaccines against melanoma (the prototype model of tumor immunology in humans) can reproducibly induce cytotoxic T cell (CTL) responses exquisitely specific for cancer cells. This achievement trespasses the specificity of any other anti-cancer therapy. The skeptics, on the other end, might point out that immunization only rarely leads to cancer regression, labeling, therefore, this approach is ineffective. In our opinion this judgment stems from the naïve expectation that CTL induction is sufficient for an effective immune response. Here we propose that more needs to be understood about the mechanisms required for the induction of a therapeutically relevant immune response in humans. In particular, we will discuss the variables related to cancer heterogeneity, the weight of individual patients' polymorphism(s), the role of the T cell activation and differentiation and, finally, the complex relationship between immune and cancer cells within the tumor microenvironment.
Collapse
Affiliation(s)
- Ena Wang
- Immunogenetics Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bethesda, MD 20892-1184, USA
| | | | | |
Collapse
|
38
|
Driessens G, Hamdane M, Cool V, Velu T, Bruyns C. Highly Successful Therapeutic Vaccinations Combining Dendritic Cells and Tumor Cells Secreting Granulocyte Macrophage Colony-stimulating Factor. Cancer Res 2004; 64:8435-42. [PMID: 15548715 DOI: 10.1158/0008-5472.can-04-0774] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In an attempt to induce potent immune antitumor activities, we investigated, within the rat 9L gliosarcoma model, distal therapeutic vaccinations associating three therapies: dendritic cell vaccination, intratumoral granulocyte macrophage colony-stimulating factor (GM-CSF) gene transfer, and tumor apoptosis induction. Vaccines of dendritic cells coinjected with processed GM-CSF secreting 9L cells induced systemic responses, resulting in the complete regression of distant preimplanted 9L tumor masses in, with the best strategy, 94% of male rats. All of the cured rats developed a long-term resistance to a rechallenge with parental cells. The curative responses were correlated with the detection of elevated specific cytotoxic activities and a CD4+, CD8+ T cell-, and natural killer (NK) cell-mediated IFN-gamma production. The survival rate of the rat seemed more directly linked to the amount of GM-CSF secreted by the transduced tumor cells, which in turn depended on the toxicity of the apoptosis-inducing treatment, than to the level of apoptosis induced. Unexpectedly, alive GM-CSF secreting 9L cells became apoptotic when injected in vivo. Thus we documented the positive role of apoptosis in the induction of therapeutic antitumor responses by comparing, at equal GM-CSF exogenous supply, the effects of dendritic cells coinjected with apoptotic or necrotic 9L cells. The data showed the superior therapeutic efficiency of combined vaccines containing apoptotic tumor cells. In conclusion, vaccinations with dendritic cells associated with apoptotic tumor cells secreting GM-CSF show a very high therapeutic potency that should show promise for the treatment of human cancer.
Collapse
Affiliation(s)
- Gregory Driessens
- Interdisciplinary Research Institute (IRIBHM), Faculty of Medicine, Université Libre de Bruxelles, Bruxelles, Belgium
| | | | | | | | | |
Collapse
|
39
|
Panelli MC, Wang E, Monsurrò V, Jin P, Zavaglia K, Smith K, Ngalame Y, Marincola FM. Overview of melanoma vaccines and promising approaches. Curr Oncol Rep 2004; 6:414-20. [PMID: 15291987 DOI: 10.1007/s11912-004-0069-3] [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: 11/28/2022]
Abstract
It is difficult to envision anything better than melanoma vaccines to exemplify the effectiveness of modern biotechnology in developing biologically rational therapeutics. Melanoma vaccines can reproducibly induce cytotoxic T lymphocyte (CTL) responses better than any other anticancer therapy. Anticancer vaccines have been labeled by some as ineffective for the simple reason that they only rarely lead to cancer regression. This oxymoron stems from the naïve expectation that CTLs are all that is needed to reject cancer. Little is known about requirements for CTL localization and effector function within the tumor microenvironment. In the future, more attention should be given to events downstream of immunization (afferent arm of immune response) to identify combination therapies likely to facilitate localization and activation of CTL at the receiving end (efferent arm).
Collapse
Affiliation(s)
- Monica C Panelli
- Immunogenetics Section, Department of Transfusion Medicine, Clinical Center, National Institutes of Health, Bldg 10, R-1C711, 9000 Rockville Pike, Bethesda, MD 20892, USA
| | | | | | | | | | | | | | | |
Collapse
|
40
|
Marincola FM, Ferrone S. Immunotherapy of melanoma: the good news, the bad ones and what to do next. Semin Cancer Biol 2004; 13:387-9. [PMID: 15001156 DOI: 10.1016/j.semcancer.2003.09.002] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
|