1
|
Tian L, Wang S, Jiang S, Liu Z, Wan X, Yang C, Zhang L, Zheng Z, Wang B, Li L. Luteolin as an adjuvant effectively enhances CTL anti-tumor response in B16F10 mouse model. Int Immunopharmacol 2021; 94:107441. [PMID: 33611060 DOI: 10.1016/j.intimp.2021.107441] [Citation(s) in RCA: 22] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/21/2020] [Revised: 01/19/2021] [Accepted: 01/25/2021] [Indexed: 12/13/2022]
Abstract
Luteolin, a naturally found dietary flavonoid, has a wide range of beneficial biological effects, including effects against tumors and oxidants. Studies proved that luteolin can modulate immune responses. In this study, we investigated the function of luteolin as an antitumor vaccine adjuvant (to treat malignant melanoma) in vitro and in vivo. We found that Luteolin may activated the PI3K-Akt pathways in APCs (Antigen Presenting Cells), induced the activation of APCs, enhanced CTL (Cytotoxic T Lymphocyte) responses, and inhibited tolerogenic T cells. To prove the role of CD8+T cells in immune process, we sorted the CD8+T cells from the immunized mice and transferred them to the B16F10 tumor-bearing mice, the result showed that the survival rate was improved. We also observed that in the mice immunized with Luteolin as an adjuvant, the tumor growth was significantly reduced. Taken together, the result demonstrated that luteolin showed promising properties as a vaccine adjuvant for treating malignant melanoma.
Collapse
Affiliation(s)
- Le Tian
- School of Basic Medical, Qingdao University, Qingdao, China
| | - Shuang Wang
- School of Basic Medical, Qingdao University, Qingdao, China
| | - Shasha Jiang
- Department of Pathogenic Biology, Qingdao University, Qingdao, China
| | - Zeyuan Liu
- Department of Special Medicine, Qingdao University, Qingdao, China
| | - Xueqi Wan
- School of Basic Medical, Qingdao University, Qingdao, China
| | - Chaochao Yang
- School of Basic Medical, Qingdao University, Qingdao, China
| | - Li Zhang
- School of Basic Medical, Qingdao University, Qingdao, China
| | - Zheng Zheng
- School of Basic Medical, Qingdao University, Qingdao, China
| | - Bin Wang
- Department of Pathogenic Biology, Qingdao University, Qingdao, China
| | - Ling Li
- School of Basic Medical, Qingdao University, Qingdao, China.
| |
Collapse
|
2
|
Oh J, Barve M, Matthews CM, Koon EC, Heffernan TP, Fine B, Grosen E, Bergman MK, Fleming EL, DeMars LR, West L, Spitz DL, Goodman H, Hancock KC, Wallraven G, Kumar P, Bognar E, Manning L, Pappen BO, Adams N, Senzer N, Nemunaitis J. Phase II study of Vigil® DNA engineered immunotherapy as maintenance in advanced stage ovarian cancer. Gynecol Oncol 2016; 143:504-510. [DOI: 10.1016/j.ygyno.2016.09.018] [Citation(s) in RCA: 56] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/27/2016] [Revised: 09/15/2016] [Accepted: 09/19/2016] [Indexed: 12/19/2022]
|
3
|
Zhou J, Ma P, Li J, Song W. Comparative analysis of cytotoxic T lymphocyte response induced by dendritic cells pulsed with recombinant adeno-associated virus carrying α-fetoprotein gene or cancer cell lysate. Mol Med Rep 2014; 11:3174-80. [PMID: 25484119 DOI: 10.3892/mmr.2014.3059] [Citation(s) in RCA: 6] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2014] [Accepted: 11/21/2014] [Indexed: 12/13/2022] Open
Abstract
Hepatocellular carcinoma (HCC) is one of the most common and difficult to treat types of cancer worldwide. Antigen‑targeted immunotherapy has the potential to be a novel and effective adjuvant for use in HCC. In the present study, recombinant adeno‑associated virus carrying the α‑fetoprotein gene (rAAV/AFP) and cancer cell lysates were used to pulse antigen‑presenting dendritic cells (DCs) in order to stimulate a cytotoxic T lymphocyte (CTL) response against HCC. rAAV/AFP‑pulsed and cancer cell lysate‑pulsed DCs resulted in a mature DC phenotype with high expression of major histocompatibility complex (MHC) class I, MHC class II, CD80, CD83 and CD86 molecules. However, rAAV/AFP‑pulsed DCs exhibited superiority over cancer cell lysate‑pulsed DCs in terms of stimulating proliferation of T cells, activating T cells to secret interferon‑γ (IFN‑γ) and inducing an AFP‑specific MHC class I‑restricted CTL response. The current data suggest that pulsing of DCs using rAAV/AFP is more effective than the cancer cell lysate‑pulsing technique, and that this technique may be used for the development of immunotherapy in AFP‑positive HCC.
Collapse
Affiliation(s)
- Jun Zhou
- Department of Oncology, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Ping Ma
- Department of Opthalmology, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Jun Li
- Department of Oncology, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| | - Wei Song
- Department of Oncology, Provincial Hospital Affiliated to Shandong University, Jinan, Shandong 250021, P.R. China
| |
Collapse
|
4
|
Zehn D, Roepke S, Weakly K, Bevan MJ, Prlic M. Inflammation and TCR signal strength determine the breadth of the T cell response in a bim-dependent manner. THE JOURNAL OF IMMUNOLOGY 2013; 192:200-5. [PMID: 24273000 DOI: 10.4049/jimmunol.1302289] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Generating a diverse T cell memory population through vaccination is a promising strategy to overcome pathogen epitope variability and tolerance to tumor Ags. The effector and memory pool becomes broad in TCR diversity by recruiting high- and low-affinity T cells. We wanted to determine which factors dictate whether a memory T cell pool has a broad versus focused repertoire. We find that inflammation increases the magnitude of low- and high-affinity T cell responses equally well, arguing against a synergistic effect of TCR and inflammatory signals on T cell expansion. We dissect the differential effects of TCR signal strength and inflammation and demonstrate that they control effector T cell survival in a bim-dependent manner. Importantly, bim-dependent cell death is overcome with a high Ag dose in the context of an inflammatory environment. Our data define the framework for the generation of a broad T cell memory pool to inform future vaccine design.
Collapse
Affiliation(s)
- Dietmar Zehn
- Swiss Vaccine Research Institute, 1066 Epalinges, Switzerland
| | | | | | | | | |
Collapse
|
5
|
Mohan T, Verma P, Rao DN. Novel adjuvants & delivery vehicles for vaccines development: a road ahead. Indian J Med Res 2013; 138:779-95. [PMID: 24434331 PMCID: PMC3928709] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/01/2013] [Indexed: 11/29/2022] Open
Abstract
The pure recombinant and synthetic antigens used in modern day vaccines are generally less immunogenic than older style live/attenuated and killed whole organism vaccines. One can improve the quality of vaccine production by incorporating immunomodulators or adjuvants with modified delivery vehicles viz. liposomes, immune stimulating complexes (ISCOMs), micro/nanospheres apart from alum, being used as gold standard. Adjuvants are used to augment the effect of a vaccine by stimulating the immune system to respond to the vaccine, more vigorously, and thus providing increased immunity to a particular disease. Adjuvants accomplish this task by mimicking specific sets of evolutionary conserved molecules which include lipopolysaccharides (LPS), components of bacterial cell wall, endocytosed nucleic acids such as dsRNA, ssDNA and unmethylated CpG dinucleotide containing DNA. This review provides information on various vaccine adjuvants and delivery vehicles being developed to date. From literature, it seems that the humoral immune responses have been observed for most adjuvants and delivery platforms while viral-vector, ISCOMs and Montanides have shown cytotoxic T-cell response in the clinical trials. MF59 and MPL® have elicited Th1 responses, and virus-like particles (VLPs), non-degradable nanoparticle and liposomes have also generated cellular immunity. Such vaccine components have also been evaluated for alternative routes of administration with clinical success reported for intranasal delivery of viral-vectors and proteosomes and oral delivery of VLP vaccines.
Collapse
Affiliation(s)
- Teena Mohan
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - Priyanka Verma
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| | - D. Nageswara Rao
- Department of Biochemistry, All India Institute of Medical Sciences, New Delhi, India
| |
Collapse
|
6
|
Mir SA, Sharma S. Role of MHC class Ib molecule, H2-M3 in host immunity against tuberculosis. Vaccine 2013; 31:3818-25. [PMID: 23628242 DOI: 10.1016/j.vaccine.2013.04.005] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/03/2012] [Revised: 03/22/2013] [Accepted: 04/01/2013] [Indexed: 01/21/2023]
Abstract
The MHC class I family comprises both classical (class Ia) and non-classical (class Ib) members. While the prime function of classical MHC class I molecules (MHC class Ia) is to present peptide antigens to pathogen-specific cytotoxic T cells, non-classical MHC-I (MHC class Ib) antigens perform diverse array of functions in both innate and adaptive immunity. Vaccines against intracellular pathogens such as Mycobacterium tuberculosis need to induce strong cellular immune responses. Recent studies have shown that MHC class I molecules play an important role in the protective immune response to M. tuberculosis infection. Both MHC Ia-restricted and MHC class Ib-restricted M. tuberculosis -reactive CD8(+) T cells have been identified in humans and mice, but their relative contributions to immunity is still uncertain. Unlike MHC class Ia-restricted CD8(+) T cells, MHC class Ib-restricted CD8(+) T cells are constitutively activated in naive animals and respond rapidly to infection challenge, hence filling the temporal gap between innate and adaptive immunity. The present review article summarizes the general host immunity against M. tuberculosis infection highlighting the possible role of MHC class Ib molecule, H2-M3 and their ligands (N-formylated peptides) in protection against tuberculosis.
Collapse
Affiliation(s)
- Shabir Ahmad Mir
- Department of Biochemistry, Postgraduate Institute of Medical Education & Research, Chandigarh 160012, India
| | | |
Collapse
|
7
|
Hong S, Li H, Qian J, Yang J, Lu Y, Yi Q. Optimizing dendritic cell vaccine for immunotherapy in multiple myeloma: tumour lysates are more potent tumour antigens than idiotype protein to promote anti-tumour immunity. Clin Exp Immunol 2013; 170:167-77. [PMID: 23039887 DOI: 10.1111/j.1365-2249.2012.04642.x] [Citation(s) in RCA: 33] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Abstract
Dendritic cells (DCs) are the most potent antigen-presenting cells and are the mediators of T cell immunity. Many investigators have explored the potential of using DCs as a vaccine for tumour-derived antigens in immunotherapy of B cell malignancies, and the results have been disappointing. To search for better tumour antigens to improve the efficacy of DC-based immunotherapy in myeloma, we evaluated and compared the efficacy of the vaccination of DCs pulsed with idiotype (Id) or tumour lysate in the 5TGM1 myeloma mouse model. Our results showed that Id- or tumour lysate-pulsed DC vaccines protected mice efficiently against developing myeloma, retarded tumour growth, induced tumour regression against established tumour and protected surviving mice from tumour rechallenge. The therapeutic responses were associated with an induction of strong humoral immune responses, including anti-Id or anti-lysate antibodies, and cellular immune responses including myeloma-specific CD8(+) cytotoxic T lymphocytes, CD4(+) type 1 T helper cells and memory T cells in mice receiving Id- or tumour lysate-pulsed DC vaccines. In addition, our studies showed that tumour lysate-pulsed DCs were more potent vaccines than the Id-pulsed DC vaccines to promote anti-tumour immunity in the model. This information will be important for improving the strategies of DC-based immunotherapy for patients with myeloma and other B cell tumours.
Collapse
Affiliation(s)
- S Hong
- Department of Lymphoma/Myeloma, Division of Cancer Medicine, and Center for Cancer Immunology Research, The University of Texas MD Anderson Cancer Center, Houston, TX 77030, USA
| | | | | | | | | | | |
Collapse
|
8
|
Abstract
The past decade has revealed that melanoma is comprised of multiple subclasses that can be categorized on the basis of key features, including the clinical stage of disease, the oncogenic molecular 'drivers', the anatomical location or the behaviour of the primary lesion and the expression of specific biomarkers. Although exercises in subclassification are not new in oncology, progress in this area has produced both conceptual and clinical breakthroughs, which, for melanoma, are unprecedented in the modern history of the disease. This Review focuses on these recent striking advances in the strategy of molecularly targeted approaches to the therapy of melanoma in humans.
Collapse
Affiliation(s)
- Keith T Flaherty
- Massachusetts General Hospital Cancer Center, 55 Fruit Street, Boston, Massachusetts 02114, USA
| | | | | |
Collapse
|
9
|
Immunologic monitoring of cellular responses by dendritic/tumor cell fusion vaccines. J Biomed Biotechnol 2011; 2011:910836. [PMID: 21541197 PMCID: PMC3085507 DOI: 10.1155/2011/910836] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/12/2010] [Revised: 11/11/2010] [Accepted: 02/27/2011] [Indexed: 12/22/2022] Open
Abstract
Although dendritic cell (DC)- based cancer vaccines induce effective antitumor activities in murine models, only limited therapeutic results have been obtained in clinical trials. As cancer vaccines induce antitumor activities by eliciting or modifying immune responses in patients with cancer, the Response Evaluation Criteria in Solid Tumors (RECIST) and WHO criteria, designed to detect early effects of cytotoxic chemotherapy in solid tumors, may not provide a complete assessment of cancer vaccines. The problem may, in part, be resolved by carrying out immunologic cellular monitoring, which is one prerequisite for rational development of cancer vaccines. In this review, we will discuss immunologic monitoring of cellular responses for the evaluation of cancer vaccines including fusions of DC and whole tumor cell.
Collapse
|
10
|
Koido S, Hara E, Homma S, Ohkusa T, Gong J, Tajiri H. Cancer immunotherapy by fusions of dendritic cells and tumor cells. Immunotherapy 2011; 1:49-62. [PMID: 20635973 DOI: 10.2217/1750743x.1.1.49] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022] Open
Abstract
Dendritic cells (DCs) are potent professional antigen-presenting cells and play a critical role in the induction of primary immune responses. DC-based vaccination represents a potentially powerful strategy for cancer immunotherapy. Thus, the use of cancer vaccines to eliminate residual tumor cells is a promising area of investigation. The immunotherapy of tumor antigen-loaded DCs has now been demonstrated in cancer patients and some clinical responses without any significant toxicity. Fusions of DCs and tumor cells represent an alternative but promising approach to overcome the inability of tumor antigens to induce a sustainable T-cell response. This review deals with recent progress in the immunotherapy of cancer with fusions of DCs and tumor cells.
Collapse
Affiliation(s)
- Shigeo Koido
- Department of Internal Medicine, The Jikei University, Tokyo, Japan.
| | | | | | | | | | | |
Collapse
|
11
|
Huang FP, Chen YX, To CKW. Guiding the “misguided” - functional conditioning of dendritic cells for the DC-based immunotherapy against tumours. Eur J Immunol 2010; 41:18-25. [DOI: 10.1002/eji.201040543] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/01/2010] [Revised: 10/06/2010] [Accepted: 11/15/2010] [Indexed: 12/15/2022]
|
12
|
Regulation of tumor immunity by tumor/dendritic cell fusions. Clin Dev Immunol 2010; 2010:516768. [PMID: 21048993 PMCID: PMC2964897 DOI: 10.1155/2010/516768] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/29/2010] [Accepted: 09/22/2010] [Indexed: 02/07/2023]
Abstract
The goal of cancer vaccines is to induce antitumor immunity that ultimately will reduce tumor burden in tumor environment. Several strategies involving dendritic cells- (DCs)- based vaccine incorporating different tumor-associated antigens to induce antitumor immune responses against tumors have been tested in clinical trials worldwide. Although DCs-based vaccine such as fusions of whole tumor cells and DCs has been proven to be clinically safe and is efficient to enhance antitumor immune responses for inducing effective immune response and for breaking T-cell tolerance to tumor-associated antigens (TAAs), only a limited success has occurred in clinical trials. This paper reviews tumor immune escape and current strategies employed in the field of tumor/DC fusions vaccine aimed at enhancing activation of TAAs-specific cytotoxic T cells in tumor microenvironment.
Collapse
|
13
|
Yi Q, Szmania S, Freeman J, Qian J, Rosen NA, Viswamitra S, Cottler-Fox M, Barlogie B, Tricot G, van Rhee F. Optimizing dendritic cell-based immunotherapy in multiple myeloma: intranodal injections of idiotype-pulsed CD40 ligand-matured vaccines led to induction of type-1 and cytotoxic T-cell immune responses in patients. Br J Haematol 2010; 150:554-64. [PMID: 20618329 PMCID: PMC2924470 DOI: 10.1111/j.1365-2141.2010.08286.x] [Citation(s) in RCA: 60] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Abstract
Vaccination with idiotype (Id) protein-pulsed dendritic cells (DCs) has been explored in multiple myeloma and the results have been disappointing. To improve the efficacy of DC vaccination in myeloma, we investigated the use of Id- and keyhole limpet haemocyanin (KLH)-pulsed, CD40 ligand-matured DCs administered intranodally. Nine patients with smouldering or stable myeloma without treatment were enrolled and DC vaccines were administered at weekly intervals for a total of four doses. Following vaccination, all patients mounted Id-specific gamma-interferon T-cell response. Interleukin-4 response was elicited in two, and skin delayed-type hypersensitivity reaction occurred in seven patients. More importantly, Id-specific cytotoxic T-cell responses were also detected in five patients. Most if not all patients mounted a positive T-cell response to KLH following vaccination. At 1-year follow-up, six of the nine patients had stable disease, while three patients had slowly progressive disease even during the vaccination period. At 5-year follow-up, four of the six patients continued with stable disease. No major side effects were noted. In summary, intranodal administration of Id-pulsed CD40 ligand-matured DCs was able to induce Id-specific T and B-cell responses in patients. Current efforts are geared towards breaking tumour-mediated immune suppression and improving clinical efficacy of this immunotherapy.
Collapse
Affiliation(s)
- Qing Yi
- Department of Lymphoma and Myeloma, The University of Texas M. D. Anderson Cancer Center, Houston, TX, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
14
|
You CX, Liu Y, Shi M, Cao M, Luo RC, Hermonat PL. Comparison of AAV/IL-7 autocrine (T cell) versus paracrine (DC) gene delivery for enhancing CTL stimulation and function. Cancer Immunol Immunother 2010; 59:779-87. [PMID: 20091029 PMCID: PMC11030548 DOI: 10.1007/s00262-009-0798-0] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/17/2009] [Accepted: 11/10/2009] [Indexed: 11/29/2022]
Abstract
Adoptive transfer of antigen-specific cytotoxic T lymphocyte (CTL) into patients holds promise in treating cancer. Such anti-cancer CTL are stimulated by professional antigen-presenting dendritic cells (DC). We hypothesize the gene delivery of various Th1-response cytokines, such as interleukin 7 (IL-7), should further enhance CTL stimulation and activity. However, the issue as to which cell type, DC (paracrine) or the T cell (autocrine), should express a particular Th1 cytokine gene for optimal CTL stimulation has never been addressed. We used adeno-associated virus-2 (AAV) to compare delivery of IL-7 and IL-2 genes into DC or T cells and to exogenous commercial cytokines for generating robust carcinoembryonic antigen (CEA)-specific CTL. AAV/IL-7 transduction of T cells (autocrine delivery) generated CTL with the highest killing capability. Consistent with this, AAV/IL-7 delivery generated T cell populations with the highest proliferation, highest interferon gamma expression, highest CD8(+):CD4(+) ratio, highest CD8(+), CD69(+) levels, and lowest CD4(+), CD25(+) (Treg) levels. These data are consistent with higher killing by the AAV/IL-7-altered CTL. These data strongly suggest that IL-7 autocrine gene delivery is optimal for CTL generation. These data also suggest Th1 cytokine autocrine versus paracrine delivery is an important issue for immuno-gene therapy and uncovers new questions into cytokine mechanism of action.
Collapse
Affiliation(s)
- Chang-Xuan You
- Department of Internal Medicine, University of Arkansas for Medical Sciences, Slot 532, 4301 West Markham St., Little Rock, AR 72205 USA
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Yong Liu
- Department of Internal Medicine, University of Arkansas for Medical Sciences, Slot 532, 4301 West Markham St., Little Rock, AR 72205 USA
- Department of Obstetrics and Gynecology, University of Arkansas for Medical Sciences, Little Rock, AR 72205 USA
| | - Min Shi
- Department of Internal Medicine, University of Arkansas for Medical Sciences, Slot 532, 4301 West Markham St., Little Rock, AR 72205 USA
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Maohua Cao
- Department of Internal Medicine, University of Arkansas for Medical Sciences, Slot 532, 4301 West Markham St., Little Rock, AR 72205 USA
| | - Rong-Cheng Luo
- Department of Oncology, Nanfang Hospital, Southern Medical University, Guangzhou, China
| | - Paul L. Hermonat
- Department of Internal Medicine, University of Arkansas for Medical Sciences, Slot 532, 4301 West Markham St., Little Rock, AR 72205 USA
- Department of Obstetrics and Gynecology, University of Arkansas for Medical Sciences, Little Rock, AR 72205 USA
| |
Collapse
|
15
|
Moser JM, Sassano ER, Leistritz DC, Eatrides JM, Phogat S, Koff W, Drake DR. Optimization of a dendritic cell-based assay for the in vitro priming of naïve human CD4+ T cells. J Immunol Methods 2009; 353:8-19. [PMID: 19925804 DOI: 10.1016/j.jim.2009.11.006] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/04/2009] [Revised: 09/18/2009] [Accepted: 11/11/2009] [Indexed: 10/20/2022]
Abstract
Methods to prime human CD4(+) T cells in vitro would be of significant value for the pre-clinical evaluation of vaccine candidates and other immunotherapeutics. However, to date, there is no reliable method for the induction of primary human T cell responses in the laboratory. Here, we optimized a culture strategy incorporating highly purified lymphocytes and dendritic cells, in the absence of any exogenous growth factors, for the in vitro sensitization of naïve CD4(+) T cells against a variety of protein antigens. This fully autologous approach, which was superior to the more traditional PBMC assay for supporting the induction of primary human T helper cell responses in culture, elicited effector cells capable of producing a variety of Th cytokines, including IFNgamma, TNFalpha, IL-2, IL-5, IL-17 and IL-21, and memory cells that could be restimulated multiple times with a specific antigen. Through simple modifications to this culture method, we evaluated the role of dendritic cell maturation state and regulatory T cells on the sensitization of naïve T helper cells, which highlights its utility for addressing basic questions of human immunobiology. Finally, using the formulated yellow fever vaccine, YF-VAX (R), we provide a proof-of-concept demonstration of the utility of the system for evaluating the T cell immunogenicity of vaccine candidates in a pre-clinical setting.
Collapse
Affiliation(s)
- Janice M Moser
- VaxDesign Corporation, 12612 Challenger Parkway, suite 365, Orlando, FL 32826, USA
| | | | | | | | | | | | | |
Collapse
|
16
|
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
|
17
|
Yu Y, Pilgrim P, Zhou W, Gagliano N, Frezza EE, Jenkins M, Weidanz JA, Lustgarten J, Cannon M, Bumm K, Cobos E, Kast WM, Chiriva-Internati M. rAAV/Her-2/neu Loading of Dendritic Cells for a Potent Cellular-Mediated MHC Class I Restricted Immune Response Against Ovarian Cancer. Viral Immunol 2008; 21:435-42. [DOI: 10.1089/vim.2008.0029] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023] Open
Affiliation(s)
- Yuefei Yu
- Division of Hematology and Oncology, Texas Tech University Health Sciences Center and the Southwest Cancer Treatment and Research Center, Lubbock, Texas
| | - Petra Pilgrim
- Division of Hematology and Oncology, Texas Tech University Health Sciences Center and the Southwest Cancer Treatment and Research Center, Lubbock, Texas
| | - Wei Zhou
- Division of Hematology and Oncology, Texas Tech University Health Sciences Center and the Southwest Cancer Treatment and Research Center, Lubbock, Texas
| | - Nicoletta Gagliano
- Division of Hematology and Oncology, Texas Tech University Health Sciences Center and the Southwest Cancer Treatment and Research Center, Lubbock, Texas
- Department of Human Morphology, University of Milan, Milan, Italy
| | - E. Eldo Frezza
- Surgery, Texas Tech University Health Sciences Center and the Southwest Cancer Treatment and Research Center, Lubbock, Texas
| | - Marjorie Jenkins
- Departments of Internal Medicine and Obstetrics and Gynecology, and the Laura W. Bush Institute for Women's Health and Center for Women's Health and Gender-Based Medicine, Texas Tech University Health Sciences Center, Amarillo
| | - Jon A. Weidanz
- Division of Hematology and Oncology, Texas Tech University Health Sciences Center and the Southwest Cancer Treatment and Research Center, Lubbock, Texas
- Department of Pharmaceutical Sciences, School of Pharmacy, Texas Tech University Health Sciences Center, Amarillo, Texas
| | - Joseph Lustgarten
- Department of Immunology and Cancer Center, Mayo Clinic, Scottsdale, Arizona
| | - Martin Cannon
- Department of Microbiology and Immunology, University of Arkansas for Medical Sciences, Little Rock, Arkansas
| | - Klaus Bumm
- Department of Otorhinolaryngology, Head and Neck Surgery, University of Erlangen-Nuremberg, FAU Medical School, Erlangen, Germany
| | - Everardo Cobos
- Division of Hematology and Oncology, Texas Tech University Health Sciences Center and the Southwest Cancer Treatment and Research Center, Lubbock, Texas
| | - W. Martin Kast
- Departments of Molecular Microbiology and Immunology and Obstetrics and Gynecology, Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California
- Kiromic, Inc., Lubbock, Texas
| | - Maurizio Chiriva-Internati
- Division of Hematology and Oncology, Texas Tech University Health Sciences Center and the Southwest Cancer Treatment and Research Center, Lubbock, Texas
- Kiromic, Inc., Lubbock, Texas
| |
Collapse
|
18
|
Gong J, Koido S, Calderwood SK. Cell fusion: from hybridoma to dendritic cell-based vaccine. Expert Rev Vaccines 2008; 7:1055-68. [PMID: 18767954 DOI: 10.1586/14760584.7.7.1055] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
The deployment of dendritic cell (DC) and tumor cell fusions is increasing in tumor immunotherapy. In animal and human studies, fusion cell vaccines have been shown to possess the elements essential for processing and presenting tumor antigens to host immune cells, for inducing effective immune response and for breaking T-cell tolerance to tumor-associated antigens. Moreover, fusion cell vaccines provide protection against challenge with tumor cells and mediate regression of established tumors. Despite these unique features of fusion cell vaccines and the observation of tumor eradication in animal studies, limited success has occurred in clinical trials. This article reviews the methods used for optimizing the preparation and selection of DC-tumor fusion cells and analyzes factors influencing the success or failure of fusion cell-mediated immunotherapy. In addition, we discuss the challenges facing effective fusion cell vaccine production, including factors in preparation, selection and quality control of fusion cell vaccines, as well as approaches for enhancing anti-tumor immunity.
Collapse
Affiliation(s)
- Jianlin Gong
- Department of Medicine, Boston University Medical School, Boston, MA 02118, USA.
| | | | | |
Collapse
|
19
|
Yu Y, Pilgrim P, Yan J, Zhou W, Jenkins M, Gagliano N, Bumm K, Cannon M, Milzani A, Dalle-Donne I, Kast WM, Cobos E, Chiriva-Internati M. Protective CD8+ T-cell responses to cytomegalovirus driven by rAAV/GFP/IE1 loading of dendritic cells. J Transl Med 2008; 6:56. [PMID: 18834548 PMCID: PMC2570669 DOI: 10.1186/1479-5876-6-56] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/31/2008] [Accepted: 10/05/2008] [Indexed: 12/26/2022] Open
Abstract
Background Recent studies demonstrate that recombinant adeno-associated virus (rAAV)-based antigen loading of dendritic cells (DCs) generates in vitro, significant and rapid cytotoxic T-lymphocyte (CTL) responses against viral antigens. Methods We used the rAAV system to induce specific CTLs against CVM antigens for the development of cytomegalovirus HCMV) gene therapy. As an extension of the versatility of the rAAV system, we incorporated immediate-early 1 (IE1), expressed in HCMV. Our rAAV vector induced a strong stimulation of CTLs directed against the HCMV antigen IE1. We then investigated the efficiency of the CTLs in killing IE1 targeted cells. Results A significant MHC Class I-restricted, anti-IE1-specific CTL killing was demonstrated against IE1 positive peripheral blood mononuclear cells (PBMC) after one, in vitro, stimulation. Conclusion In summary, single PBMC stimulation with rAAV/IE1 pulsed DCs induces strong antigen specific-CTL generation. CTLs were capable to lyse low doses of peptides pulsed into target cells. These data suggest that AAV-based antigen loading of DCs is highly effective for generating human CTL responses against HCMV antigens.
Collapse
Affiliation(s)
- Yuefei Yu
- Division of Hematology & Oncology, Texas Tech University Health Sciences Center and Southwest Cancer Treatment and Research Center, Lubbock, TX, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
20
|
You H, Liu Y, Cong M, Ping W, You C, Zhang D, Mehta JL, Hermonat PL. HBV genes induce cytotoxic T-lymphocyte response upon adeno-associated virus (AAV) vector delivery into dendritic cells. J Viral Hepat 2006; 13:605-12. [PMID: 16907847 DOI: 10.1111/j.1365-2893.2006.00734.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/12/2023]
Abstract
Hepatitis B virus (HBV) has been an increasing problem throughout the world and remains difficult to treat. But immunotherapeutic approaches offer new, effective treatments. Three recombinant adeno-associated virus (AAV) type 2 vectors, carrying one of the HBV S, C or X gene, were used to load (transduce) professional antigen-presenting dendritic cells (DC) for the purpose of stimulating cytotoxic T lymphocytes (CTL) in vitro. It was found that all three recombinant AAV/HBV antigen virus loaded DC at approximately 90% transduction efficiency. Most importantly, all three AAV-loaded DC stimulated rapid, antigen-specific and major histocompatibility complex (MHC)-restricted CTL. In vitro, these CTL killed (30-50%) synthetic antigen-positive autologous targets as well as HepG2 liver cell targets. In comparing the three antigens, it was found that AAV/HBV-C-derived CTL consistently had the highest killing efficiency. CTL derived from AAV/HBV-C-loaded DC also showed significantly higher killing of targets than that from bacterially generated C-protein-loaded DC. Further studies showed that AAV/HBV-C-derived CTL had higher interferon (IFN)-gamma. These data suggest that AAV/HBV antigen gene-loading of DC may be useful for immunotherapeutic protocols against HBV infection and that the HBV C antigen may be the most useful for this purpose.
Collapse
Affiliation(s)
- H You
- Liver Research Center, Beijing Friendship Hospital, Capital University of Medical Sciences, Beijing, China
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Abstract
GVAX is a granulocyte-macrophage colony-stimulating factor (GM-CSF) gene-transfected tumor cell vaccine. Original work with GM-CSF as a recombinant DNA protein (Leukine) involved proliferative stimulation of macrophages and neutrophils for the purpose of reducing hematopoietic toxicity related to dose-intensive chemotherapy. Following US Food and Drug Administration approval of Leukine several years ago, extensive preclinical results have demonstrated an immunostimulatory effect related to GM-CSF gene when transfected into tumor cells and used as a vaccine (GVAX). Tumor regression and prolonged survival was demonstrated in animal models. Toxicology with GVAX indicated no adverse effects, which enabled further testing in cancer patients. A small number of responses were demonstrated in Phase I trials in immunosensitive cancer patients (renal cell carcinoma and melanoma). However, a series of dramatic complete and durable responses in advanced non-small cell lung cancer patients, demonstrated in recent clinical trials, have generated interest in further development of this vaccine in nontraditional cancer disease types. The rationale of GVAX development and a summary of clinical results are reviewed.
Collapse
|
22
|
He Y, Zhang J, Donahue C, Falo LD. Skin-derived dendritic cells induce potent CD8(+) T cell immunity in recombinant lentivector-mediated genetic immunization. Immunity 2006; 24:643-56. [PMID: 16713981 PMCID: PMC3077717 DOI: 10.1016/j.immuni.2006.03.014] [Citation(s) in RCA: 157] [Impact Index Per Article: 8.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/05/2005] [Revised: 02/16/2006] [Accepted: 03/01/2006] [Indexed: 11/17/2022]
Abstract
The skin contains readily accessible dendritic cells (DCs) with potent antigen presentation function and functional plasticity enabling the integration of antigen specificity with environmentally responsive immune control. Recent studies challenge the established paradigm of cutaneous immune function by suggesting that lymph node-resident DCs, rather than skin-derived DCs (sDCs), are responsible for eliciting T cell immunity against cutaneous pathogens including viral vectors. We show that cutaneous delivery of lentivirus results in direct transfection of sDCs and potent and prolonged antigen presentation. Further, sDCs are the predominant antigen-presenting cells for the induction of potent and durable CD8(+) T cell immunity. These results support the classical paradigm of cutaneous immune function and suggest that antigen presentation by sDCs contributes to the high potency of lentivector-mediated genetic immunization.
Collapse
Affiliation(s)
- Yukai He
- Department of Dermatology, University of Pittsburgh School of Medicine, Suite 145 Lothrop Hall, 190 Lothrop Street, Pittsburgh, Pennsylvania 15213
- Department of Immunology, University of Pittsburgh School of Medicine, Suite 145 Lothrop Hall, 190 Lothrop Street, Pittsburgh, Pennsylvania 15213
| | - Jiying Zhang
- Department of Dermatology, University of Pittsburgh School of Medicine, Suite 145 Lothrop Hall, 190 Lothrop Street, Pittsburgh, Pennsylvania 15213
| | - Cara Donahue
- Department of Dermatology, University of Pittsburgh School of Medicine, Suite 145 Lothrop Hall, 190 Lothrop Street, Pittsburgh, Pennsylvania 15213
| | - Louis D. Falo
- Department of Dermatology, University of Pittsburgh School of Medicine, Suite 145 Lothrop Hall, 190 Lothrop Street, Pittsburgh, Pennsylvania 15213
| |
Collapse
|
23
|
Jouanneau E, Poujol D, Gulia S, Le Mercier I, Blay JY, Belin MF, Puisieux I. Dendritic cells are essential for priming but inefficient for boosting antitumour immune response in an orthotopic murine glioma model. Cancer Immunol Immunother 2006; 55:254-67. [PMID: 16133115 PMCID: PMC11030922 DOI: 10.1007/s00262-005-0040-7] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/20/2004] [Accepted: 06/06/2005] [Indexed: 12/27/2022]
Abstract
The prognosis of malignant gliomas remains dismal and alternative therapeutic strategies are required. Immunotherapy with dendritic cells (DCs) pulsed with tumour antigens emerges as a promising approach. Many parameters influence the efficacy of DC-based vaccines and need to be optimised in preclinical models. The present study compares different vaccine schedules using DCs loaded with tumour cell lysate (DC-Lysate) for increasing long-term survival in the GL26 orthotopic murine glioma model, focusing on the number of injections and an optimal way to recall antitumour immune response. Double vaccination with DC-Lysate strongly prolonged median survival compared to unvaccinated animals (mean survival 87.5 days vs. 25 days; p < 0.0001). In vitro data showed specific cytotoxic activity against GL26. However, late tumour relapses frequently occurred after 3 months and only 20% of mice were finally cured at 7 months. While one, two or three DC injections gave identical survival, a boost using only tumour lysate after initial DC-Lysate priming dramatically improved long-term survival in vaccinated mice, compared to the double DC-Lysate group, with 67.5% of animals cured at 7 months (p < 0.0001). In vitro data showed better specific CTL response and also the induction of specific anti-GL26 antibodies in the DC-Lysate/Lysate group, which mediated Complement Dependent Cytotoxicity. These experimental data may be of importance for the design of clinical trials that currently use multiple DC injections.
Collapse
Affiliation(s)
- E. Jouanneau
- Neurosurgery department, Neurological and Neurosurgical Hospital Pierre Wertheimer, Claude Bernard University, Lyon, France
- INSERM U590 “Oncogenesis and tumour progression”, Cancer Center Léon Bérard, Claude Bernard University, Lyon, France
| | - D. Poujol
- INSERM U590 “Oncogenesis and tumour progression”, Cancer Center Léon Bérard, Claude Bernard University, Lyon, France
| | - S. Gulia
- INSERM U590 “Oncogenesis and tumour progression”, Cancer Center Léon Bérard, Claude Bernard University, Lyon, France
| | - I. Le Mercier
- INSERM U590 “Oncogenesis and tumour progression”, Cancer Center Léon Bérard, Claude Bernard University, Lyon, France
| | - J. Y. Blay
- INSERM U590 “Oncogenesis and tumour progression”, Cancer Center Léon Bérard, Claude Bernard University, Lyon, France
| | - M. F. Belin
- U433, U.F.R. Laennec, Claude Bernard University, Lyon, France
| | - I. Puisieux
- INSERM U590 “Oncogenesis and tumour progression”, Cancer Center Léon Bérard, Claude Bernard University, Lyon, France
| |
Collapse
|
24
|
Shimamura H, Sunamura M, Tsuchihara K, Egawa S, Takeda K, Matsuno S. Irradiated pancreatic cancer cells undergo both apoptosis and necrosis, and could be phagocytized by dendritic cells. Eur Surg Res 2006; 37:228-34. [PMID: 16260873 DOI: 10.1159/000087868] [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: 12/06/2004] [Accepted: 05/25/2005] [Indexed: 11/19/2022]
Abstract
The interaction of immature dendritic cells (DC) with irradiated pancreatic cancer cells was examined. Flow cytometric analysis using annexin V and propidium iodide revealed that ionizing radiation (25-35 Gy X-ray) induced both apoptosis and necrosis in pancreatic cancer cell lines. After irradiation, PK-1 and Panc-1 cells were likely to undergo necrosis, whereas MIAPaCa-2 cells underwent apoptosis. When DiO-stained immature DCs were co-incubated with DiI-stained irradiated MIAPaCa-2, it was observed under fluorescent microscopy that DCs phagocytized dead tumor cells as early as 4 h after co-incubation. The DCs' phagocytosis of irradiated tumor cells was also confirmed by flow cytometry. These results suggest that irradiated pancreatic cancer cells, which undergo both apoptosis and necrosis, could be a good source of tumor-associated antigens for cross-presentation by DCs.
Collapse
Affiliation(s)
- H Shimamura
- Department of Gastroenterological Surgery, Tohoku University Graduate School of Medicine, Sendai, Japan
| | | | | | | | | | | |
Collapse
|
25
|
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
|
26
|
Bang S, Kim HS, Choo YS, Park SW, Chung JB, Song SY. Differences in immune cells engaged in cell-mediated immunity after chemotherapy for far advanced pancreatic cancer. Pancreas 2006; 32:29-36. [PMID: 16340741 DOI: 10.1097/01.mpa.0000191651.32420.41] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
OBJECTIVES Little is known about the effects of chemotherapy on the immunity of cancer patient or the ideal timing for immunotherapy combined with chemotherapy. To address these questions, we evaluated the effect of gemcitabine and cisplatin combination chemotherapy on the immunity of pancreatic cancer patients. METHODS Thirteen patients with far advanced pancreatic cancer were enrolled and 7 sex- and age-matched healthy volunteers were included as a control group. RESULTS Compared with healthy controls, the amounts of peripheral blood mononuclear cells, dendritic cells (DCs), natural killer cells, CD4, and CD8 T cells were reduced. With this numerical suppression, NK cell cytotoxicity to K562 leukemia cells was also significantly impaired (7.7% +/- 4.9% versus 21.7% +/- 7.9% of DNA loss; P = 0.016). Serum concentrations of VEGF and interleukin-10 (IL-10) were higher than the control group (192.1 +/- 114.7 versus 50.8 +/- 39.5 pg/mL of vascular endothelial growth factor (VEGF) and 122 +/- 68.9 versus 111.4 +/- 37.4 pg/mL of IL-10). After 1 cycle of gemcitabine and cisplatin chemotherapy, the impaired immunity of patients with pancreatic cancer was restored. Specifically, the recovery of DCs occurred rapidly and exceeded the value of healthy controls. Levels of the immunosuppressive cytokines, IL-10 and VEGF, gradually decreased during chemotherapy. CONCLUSIONS Systemic chemotherapy seems to be beneficial for restoring the impaired immunity of patients with pancreatic cancer, and one of the ideal times to collect DCs for immunotherapy is after completing each cycle of chemotherapy.
Collapse
Affiliation(s)
- Seungmin Bang
- Institute of Gastroenterology, Division of Gastroenterology, Department of Internal Medicine, Yonsei University College of Medicine, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
27
|
Miyazaki J, Tsuzuki Y, Matsuzaki K, Hokari R, Okada Y, Kawaguchi A, Nagao S, Itoh K, Miura S. Combination therapy with tumor-lysate pulsed dendritic cells and antiangiogenic drug TNP-470 for mouse pancreatic cancer. Int J Cancer 2005; 117:499-505. [PMID: 15906361 DOI: 10.1002/ijc.21202] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Most cases of pancreatic cancer are inoperable when diagnosed. Since immunotherapy and antiangiogenic therapy have been reported to be promising for pancreatic cancer, we examined whether the combination of immunotherapy with dendritic cells (DCs) and the antiangiogenic drug TNP-470 induces tumor regression. Syngeneic mouse pancreatic adenocarcinoma cells were orthotopically inoculated into C57/BL6 mice. DCs with or without tumor lysate (TL) were administered i.p. at 4 and 5 weeks. TNP-470 was injected s.c. into tumor-bearing mice every other day from 4 weeks to 6 weeks. We compared anticancer effects in 6 groups: NT (no treatment), DC/TL- (DCs without TL), DC/TL+ (DCs pulsed with TL), TNP (TNP-470 alone), DC/TL-TNP (DC/TL- plus TNP-470) and DC/TL+TNP (DC/TL+ plus TNP-470). We measured tumor volume, mean vascular density (MVD) and vessel diameter by FITC-dextran using an intravital microscope; degrees of proliferation and apoptosis of cancer cells by PCNA and TUNEL; infiltrating lymphocytes and expression levels of VEGF and MMP-9 by immunohistochemistry and immunoblotting. Tumor volume and MVD were significantly suppressed in the treatment groups with prolonged survival rate, especially in the DC/TL+TNP group. There were no significant differences in apoptosis among the 6 groups except DC/TL+. The number of infiltrating CD4+ cells in the DC/TL+ group was higher than that in the NT group. VEGF expression was significantly suppressed in the treatment groups containing TNP-470, and MMP-9 was also suppressed in the groups containing DC/TL+. Our data suggested that TL-pulsed DCs combined with TNP-470 induced regression of mouse pancreatic cancer, possibly through induction of immune responses and suppression of angiogenesis.
Collapse
Affiliation(s)
- Junichi Miyazaki
- Second Department of Internal Medicine, National Defense Medical College, Saitama, Japan
| | | | | | | | | | | | | | | | | |
Collapse
|
28
|
Yamanaka R, Honma J, Tsuchiya N, Yajima N, Kobayashi T, Tanaka R. Tumor lysate and IL-18 loaded dendritic cells elicits Th1 response, tumor-specific CD8+ cytotoxic T cells in patients with malignant glioma. J Neurooncol 2005; 72:107-13. [PMID: 15925989 DOI: 10.1007/s11060-004-3550-9] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
In this study, we demonstrate that tumor lysate-loaded dendritic cells can elicit a specific CD8+ cytotoxic T lymphocyte response against autologous tumor cells in patients with malignant glioma. CTL from three of five patients expressed strong cytolytic activity against autologous glioma cells, did not lyse autologous lymphoblasts and were variably cytotoxic against the LAK-sensitive cell line Daudi. Also, DCs pulsed normal brain lysate failed to induce cytolytic activity against autologous glioma cells, suggesting the lack of autoimmune response. Two of five patients CD8+ T cells expressed a modest cytotoxicity against autologous glioma cells. CD8+ T cells isolated during these ineffective primings secreted large amounts of IL-10, less amounts of IFN-gamma as detected by ELISA, Type 2 bias in the CD8+ T cell response accounts for the lack of cytotoxic effector function from these patients. Cytotoxicity against autologous glioma cells could be significantly inhibited by anti-HLA class I antibody. These data demonstrate that tumor lysate-loaded DC can be an effective tool in inducing glioma-specific CD8+ CTL able to kill autologous glioma cells in vitro. However, high levels of tumor specific tolerance in some patients may account for a significant barrier to therapeutic vaccination. Moreover, cytotoxic responses were augmented by transfecting DC with the gene for IL-18. For all five patients, CD8+T cells treated with IL18 transfected DC produced Th1 response. These results may have important implications for the treatment of malignant glioma patients with immunotherapy. DCs loaded with total tumor lysate and IL-18 may represent a method for inducing Th1 immunoresponses against the entire repertoire of glioma antigens.
Collapse
Affiliation(s)
- Ryuya Yamanaka
- Department of Neurosurgery, Brain Research Institute, Niigata University, Asahimachi-dori 1-757, Niigata City, 951-8585, Japan.
| | | | | | | | | | | |
Collapse
|
29
|
Abstract
The field of DNA vaccines can trace its inception to two papers which demonstrated that administration of plasmid DNA vectors expressing proteins resulted in expression in situ. Thereafter, the possible application of this technique to vaccine development was demonstrated through the induction of antibody responses in mice against a foreign protein, cellular immune responses against a viral antigen and protective efficacy in an infectious disease challenge model. Subsequently, the general utility of DNA vaccines in animal models of infectious and non-infectious disease has been established (for review, see [5]). Initially, most efforts were directed toward demonstration of effectiveness in particular disease models. Recently, however, more attention has been paid to gaining a better understanding of some of the underlying mechanisms of DNA vaccines. This review will focus on this new information and discuss it in the context of how it could benefit the development of more effective DNA vaccines.
Collapse
Affiliation(s)
- M Selby
- Vaccines Research, Chiron Corporation, Emeryville, CA 94608, USA
| | | | | |
Collapse
|
30
|
Eager R, Nemunaitis J. GM-CSF Gene-Transduced Tumor Vaccines. Mol Ther 2005; 12:18-27. [PMID: 15963916 DOI: 10.1016/j.ymthe.2005.02.012] [Citation(s) in RCA: 83] [Impact Index Per Article: 4.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2004] [Revised: 02/04/2005] [Accepted: 02/05/2005] [Indexed: 10/25/2022] Open
Abstract
GVAX is a GM-CSF gene-transduced tumor vaccine. Expression of the GM-CSF gene within either autologous or allogeneic tumor cell populations has demonstrated evidence of immune stimulation in patients and evidence of antitumor activity particularly in prostate cancer and non-small-cell lung cancer. Results of preclinical studies justify clinical investigation. A summary of clinical results is presented.
Collapse
Affiliation(s)
- Robert Eager
- Mary Crowley Medical Research Center/Texas Oncology PA, Dallas, TX 75201, USA
| | | |
Collapse
|
31
|
Cheng WF, Lee CN, Su YN, Chang MC, Hsiao WC, Chen CA, Hsieh CY. Induction of human papillomavirus type 16-specific immunologic responses in a normal and an human papillomavirus-infected populations. Immunology 2005; 115:136-49. [PMID: 15819706 PMCID: PMC1782130 DOI: 10.1111/j.1365-2567.2005.02126.x] [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] [Indexed: 11/26/2022] Open
Abstract
Human papillomavirus (HPV) infection, especially with the oncogenic genotypes, is the most important risk factor for developing cervical cancer. We focused on generating HPV16 E7-specific cytotoxic CD8(+) T lymphocytes and evaluating HPV16 E7-specific immune responses in HPV16-infected and uninfected populations. Peripheral blood mononuclear cells (PBMCs) were first collected from an uninfected group with an human lymphocyte antigen (HLA) A2 haplotype (four volunteers). Mature monocyte-derived dendritic cells (DCs) were generated from the PBMCs and pulsed with one of two HLA-A2-restricted E7 peptides, aa 11-20 [YMLDLQPETT] and aa 86-93 [TLGIVCPI], as antigen presenting cells. The autologous naive or cultured PBMCs were then cultured with peptide-pulsed DCs to detect the HPV16 E7-specific immune responses by a variety of techniques such as enzyme-linked immunosorbent assay (ELISA), enzyme-linked immunospot (ELISPOT) assay and cytotoxic T lymphocyte assay. Interferon-gamma (IFN-gamma) from E7-specific cytotoxic CD8(+) T lymphocytes stimulated with the respective peptide was detected by ELISA. Using ELISPOT analysis, a marked increase in the number of IFN-gamma-secreting CD8(+) E7-specific lymphocytes was observed following peptide stimulation. Cultured CD8(+) T lymphocytes were highly cytotoxic against the CaSki cells. PBMCs were then collected from an HPV16-infected population of the HLA-A2 haplotype, including four persons of HPV16 infection only, four with cervical intraepithelial neoplasia (CIN) lesions, and four cervical cancer patients. We then compared the immunologic responses to E7 between HPV16-infected and uninfected populations by ELISA and ELISPOT assay. The E7-specific immunologic responses of the HPV16-infected populations were significantly higher than those of the uninfected population. In addition, persons with an HPV16 infection only or those with CIN lesions generated higher E7-specific immunologic responses than cervical cancer patients. Our results demonstrate methods for evaluating E7-specific immunologic responses and reflect the biological responses of HPV16-infected people during different periods of cervical disease.
Collapse
Affiliation(s)
- Wen-Fang Cheng
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, National Taiwan UniversityTaipei, Taiwan
| | - Chien-Nan Lee
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, National Taiwan UniversityTaipei, Taiwan
| | - Yi-Ning Su
- Department of Medical Genetics, National Taiwan University Hospital, National Taiwan UniversityTaipei, Taiwan
| | - Ming-Cheng Chang
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, National Taiwan UniversityTaipei, Taiwan
| | - Wen-Chun Hsiao
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, National Taiwan UniversityTaipei, Taiwan
| | - Chi-An Chen
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, National Taiwan UniversityTaipei, Taiwan
| | - Chang-Yao Hsieh
- Department of Obstetrics and Gynecology, National Taiwan University Hospital, National Taiwan UniversityTaipei, Taiwan
| |
Collapse
|
32
|
Tanaka Y, Koido S, Ohana M, Liu C, Gong J. Induction of impaired antitumor immunity by fusion of MHC class II-deficient dendritic cells with tumor cells. THE JOURNAL OF IMMUNOLOGY 2005; 174:1274-80. [PMID: 15661883 DOI: 10.4049/jimmunol.174.3.1274] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
To dissect the role of Ag presentation through MHC class I and/or II pathways by dendritic cell (DC)-tumor fusion cells, we have created various types of DC-tumor fusion cells by alternating fusion cell partners. Fusions of MC38/MUC1 carcinoma cells with DC from wild-type (WT-DC), MHC class I knockout (IKO-DC), class II knockout (IIKO-DC), or class I and II knockout (I/IIKO-DC) mice created WTDC-fusion cells (FC), IKO-FC, IIKO-FC, and I/IIKO-FC, respectively. MHC class II- and MUC1-positive fusion cells were constructed by fusion of B16/MUC1 melanoma cells with IKO-DC (IKO/B16-FC). Immunization of MUC1 transgenic mice with 5 x 10(5) WTDC-FC, IKO-FC, IIKO-FC, or I/IIKO-FC provided 100, 91.7, 61.5, and 15.4% protection, respectively, against tumor challenge with MC38/MUC1 cells. In contrast, all mice immunized with irradiated MC38/MUC1 tumor cells or WT-DC developed tumors. One group of mice was immunized with 5 x 10(5) IKO/B16-FC and then challenged with B16/Ia(+)/MUC1 on one flank and MC38/MUC1 on the other flank. Immunization of these mice with IKO/B16-FC resulted in 100 and 78.6% protection against B16/Ia(+)/MUC1 and MC38/MUC1 tumor challenge, respectively. The antitumor immunity induced by immunization with IKO/B16-FC was able to inhibit the growth of MHC class II-negative tumor. In addition, in vivo results correlated with the induction of Ag-specific CTL. Collectively, the data indicate that MHC class II Ag presentation targeting activation of CD4 T cells is indispensable for antitumor immunity.
Collapse
MESH Headings
- Animals
- Antigen Presentation/genetics
- Antigen Presentation/immunology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD4-Positive T-Lymphocytes/pathology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/pathology
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/genetics
- Cancer Vaccines/immunology
- Cell Fusion/methods
- Cell Line, Tumor
- Cells, Cultured
- Dendritic Cells/immunology
- Dendritic Cells/metabolism
- Dendritic Cells/pathology
- Dendritic Cells/transplantation
- Histocompatibility Antigens Class I/administration & dosage
- Histocompatibility Antigens Class I/biosynthesis
- Histocompatibility Antigens Class II/administration & dosage
- Histocompatibility Antigens Class II/biosynthesis
- Histocompatibility Antigens Class II/genetics
- Histocompatibility Antigens Class II/immunology
- Interferon-gamma/antagonists & inhibitors
- Lymphocyte Activation/genetics
- Lymphocyte Activation/immunology
- Melanoma, Experimental
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, Transgenic
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/pathology
- Neoplasms, Experimental/prevention & control
- T-Lymphocytes/immunology
- T-Lymphocytes/pathology
- T-Lymphocytes, Cytotoxic/immunology
Collapse
Affiliation(s)
- Yasuhiro Tanaka
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02118, USA
| | | | | | | | | |
Collapse
|
33
|
Liu Y, Chiriva-Internati M, You C, Luo R, You H, Prasad CK, Grizzi F, Cobos E, Klimberg VS, Kay H, Mehta JL, Hermonat PL. Use and specificity of breast cancer antigen/milk protein BA46 for generating anti-self-cytotoxic T lymphocytes by recombinant adeno-associated virus-based gene loading of dendritic cells. Cancer Gene Ther 2005; 12:304-312. [PMID: 15565181 DOI: 10.1038/sj.cgt.7700785] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/15/2003] [Indexed: 12/17/2022]
Abstract
Antigen-targeted immunotherapy is an emerging treatment for breast cancer. However, useful breast cancer antigens are only found in a subset of cancer patients. BA46, also known as lactadherin, is a membrane-associated glycoprotein that is expressed in most breast cancer cells but not in general hematopoietic cell populations. Moreover, it is much more difficult to generate CTLs against self-antigens. We wished to determine if the use of recombinant adeno-associated virus (rAAV) type 2 vectors for gene-loading of dendritic cells (DCs) could generate rapid, effective cytotoxic T lymphocytes (CTLs) against BA46. We were able to demonstrate that AAV/BA46/Neo-loading of DCs resulted in: (1) BA46 expression in DCs, (2) chromosomal integration of the AAV/BA46/Neo vector within DCs, (3) strong, rapid BA46-specific, MHC class I-restricted CTLs in only 1 week, (4) T-cell populations with significant interferon-gamma (IFN-gamma) expression but low IL-4 expression, (5) high CD80 and CD86 expression in DCs, and (6) high CD8:CD4 and CD8:CD56 T cell ratios. These data suggest that rAAV-loading of DCs may be useful for immunotherapeutic protocols against self-antigens in addition to viral antigens and that the BA46 antigen is potentially appropriate for cell-mediated immunotherapeutic protocols addressing ductal breast cancer.
Collapse
Affiliation(s)
- Yong Liu
- Department of Internal Medicine, University of Arkansas for Medical Sciences, 4301 West Markham St., Little Rock, Arkansas 72205, USA
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Yoshida S, Tanaka R. Generation of a human leukocyte antigen-A24–restricted antitumor cell with the use of SART-1 peptide and dendritic cells in patients with malignant brain tumors. ACTA ACUST UNITED AC 2004; 144:201-7. [PMID: 15514588 DOI: 10.1016/j.lab.2004.06.003] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Dendritic cells (DCs) can be the principal initiators of antigen-specific immune responses. In this study, we attempted to generate cytotoxic T-lymphocytes (CTLs) using DCs pulsed with SART-1(254) peptide. Peripheral-blood mononuclear cells (PBMCs) and tumor-infiltrating mononuclear cells (TIMCs) were obtained from 11 patients with brain tumors expressing human leukocyte antigen (HLA)-A24. After stimulation with SART-1(254) peptide, CTLs showing over 15% were observed in one of 4 patients with gliomas and in 4 of 7 patients with metastatic brain tumors. Furthermore, exposure to DCs pulsed with SART-1(254) peptide increased the killing activity of these CTLs by 28.7% and 37.5%, respectively. We conclude that DCs pulsed with SART-1(254) peptide are effective in generating HLA-A24-restricted antitumor cells.
Collapse
Affiliation(s)
- Seiichi Yoshida
- Department of Neurosurgery, Niigata Cancer Hospital, Kawagishi-chou, Japan.
| | | |
Collapse
|
35
|
Gowans EJ, Jones KL, Bharadwaj M, Jackson DC. Prospects for dendritic cell vaccination in persistent infection with hepatitis C virus. J Clin Virol 2004; 30:283-90. [PMID: 15163415 PMCID: PMC4526278 DOI: 10.1016/j.jcv.2004.03.006] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Revised: 03/10/2004] [Accepted: 03/11/2004] [Indexed: 12/24/2022]
Abstract
Although hepatitis C virus (HCV) is classified in the Hepacivirus genus in the family Flaviviridae, it is unlike most of the other members of this family due to its propensity to cause persistent infections. This persistent infection eventually results in chronic liver disease, cirrhosis and hepatocellular carcinoma in a proportion of infected individuals. It has been difficult to examine correlates of clearance or persistence because most acute phase HCV infections are subclinical or result in symptoms which are non-specific; consequently, acute infections are not generally recognised and patients often present many years later with persistent infection and accompanying chronic liver disease. Nevertheless, seminal studies, performed during the acute phase, have identified a number of factors which are likely to influence the outcome of infection, although it is possible that the mechanism is multifactorial. One of these factors is impairment of dendritic cell function by a mechanism resulting from expression of an HCV protein(s) in these cells. This may be a major factor in the failure of the immune response to expand after HCV infection, leading to persistence. Nevertheless, it may be possible to overcome this defect by autologous transfusion of HCV antigen-loaded, mature dendritic cells and the purpose of this review is to highlight the need and general approaches for developing dendritic cell-based immunotherapy for HCV infection.
Collapse
Affiliation(s)
- Eric J Gowans
- Macfarlane Burnet Institute for Medical Research and Public Health, GPO Box 2284, Melbourne, VIC 3001, Australia.
| | | | | | | |
Collapse
|
36
|
Iinuma T, Homma S, Noda T, Kufe D, Ohno T, Toda G. Prevention of gastrointestinal tumors based on adenomatous polyposis coli gene mutation by dendritic cell vaccine. J Clin Invest 2004; 113:1307-17. [PMID: 15124022 PMCID: PMC398422 DOI: 10.1172/jci17323] [Citation(s) in RCA: 35] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/06/2002] [Accepted: 02/25/2004] [Indexed: 11/17/2022] Open
Abstract
Here we describe the effect of immunization with dendritic cells loaded with syngeneic tumor cells (DC/Ts) by polyethylene glycol treatment, on tumor development in adenomatous polyposis coli (APC) gene mutant mouse models, APC1309 and APC(Min-/+), in which adenomatous polyps of the gastrointestinal tracts develop with a high incidence. Treatment with DC/Ts prevented the development of gastrointestinal tumors, and coadministration of DC/Ts and IL-12 caused a further reduction in tumor incidence. Splenocytes from APC1309 mice treated with DC/Ts and IL-12 showed no cytotoxic activity toward the tumor cells, but serum antibody specific to them was detected. IgG from the treated mice exhibited cytotoxic activity against the tumor cells in vitro. Predominance of Th2 cell response over Th1 response was also suggested by ELISPOT assays in the treated mice. Depletion in vivo of CD4(+) T cells, not CD8(+) T cells, by the intraperitoneal administration of corresponding mAb's decreased the antitumor effect of DC/T inoculation. Immunofluorescence microscopic studies showed that Ig was attached to tumor cells in mice treated with DC/Ts and IL-12. These findings indicate that DC/T vaccination prevents tumor development through APC gene mutation and that its preventive effects are mediated by humoral antitumor immunity.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/metabolism
- CD4-Positive T-Lymphocytes/immunology
- Cancer Vaccines/immunology
- Carcinoma, Hepatocellular/pathology
- Carcinoma, Hepatocellular/therapy
- Cell Line, Tumor
- Dendritic Cells/immunology
- Gastrointestinal Neoplasms/pathology
- Gastrointestinal Neoplasms/prevention & control
- Genes, APC
- Interleukin-12/therapeutic use
- Melanoma, Experimental/pathology
- Melanoma, Experimental/therapy
- Mice
- Mice, Inbred C57BL
- Mice, Knockout
- Microscopy, Fluorescence
- Mutation
- T-Lymphocytes, Cytotoxic/immunology
- Th1 Cells/immunology
- Th1 Cells/metabolism
- Th2 Cells/immunology
- Th2 Cells/metabolism
Collapse
Affiliation(s)
- Toshio Iinuma
- Department of Internal Medicine, Jikei University School of Medicine, Minato-ku, Tokyo, Japan.
| | | | | | | | | | | |
Collapse
|
37
|
Gabrijel M, Repnik U, Kreft M, Grilc S, Jeras M, Zorec R. Quantification of cell hybridoma yields with confocal microscopy and flow cytometry. Biochem Biophys Res Commun 2004; 314:717-23. [PMID: 14741694 DOI: 10.1016/j.bbrc.2003.12.154] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/26/2022]
Abstract
The fusion of antigen presenting and cancer cells leads to the formation of hybrid cells, which are considered a potential vaccine for treating cancer. The quality assessment of hybrid cell vaccines is crucial for the introduction of this new treatment. Flow cytometry was the method used recently, since it is faster in comparison to classical microscopy. Here we describe a rapid confocal microscopy based approach to quantify hybrid cell yields. The extent of fusion rate was determined by confocal microscopy by counting dual fluorescent cells and by measuring the area of co-localized pixels. Results of both methods showed high degree of correlation. The same samples were also analyzed by flow cytometry. Fusion rates determined with both techniques showed significant correlation. In conclusion, using confocal microscopy we developed a sensitive and a rapid method to assess the yield of hybridomas in a large number of electrofused cells.
Collapse
Affiliation(s)
- Mateja Gabrijel
- Celica Biomedical Sciences Center, Stegne 21, 1000, Ljubljana, Slovenia
| | | | | | | | | | | |
Collapse
|
38
|
Kass R, Bellone S, Palmieri M, Canè S, Bignotti E, Henry-Tillman R, Hutchins L, Cannon MJ, Klimberg S, Santin AD. Restoration of tumor-specific HLA class I restricted cytotoxicity in tumor infiltrating lymphocytes of advanced breast cancer patients by in vitro stimulation with tumor antigen-pulsed autologous dendritic cells. Breast Cancer Res Treat 2004; 80:275-85. [PMID: 14503800 DOI: 10.1023/a:1024938215782] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
Breast tumor infiltrating lymphocytes (TIL) are enriched in tumor-specific cytotoxic T lymphocytes (CTL), and may represent a superior source of CTL compare to peripheral blood lymphocytes (PBL), for adoptive T cell immunotherapy of breast cancer. However, the immunocompetence of TIL and the possibility to consistently restore their tumor-specific lytic activity in vitro remains an open issue. In this study we evaluated the potential of tumor antigen-pulsed fully mature dendritic cell (DC) stimulation in restoring tumor-specific cytotoxicity in anergic TIL populations from advanced breast cancer patients. In addition we have compared tumor-specific T cell responses induced by tumor antigen-loaded DC stimulation of TIL to responses induced from PBL. Although TIL were consistently non-cytotoxic after isolation or culture in the presence of interleukin-2 (IL-2), in matched experiments from three consecutive patients, tumor-lysate-pulsed DC-stimulated CD8+ T cell derived from TIL were found to be significantly more cytotoxic than PBL (p < 0.05). In addition, cytotoxicity against autologous tumor cells was more significantly inhibited by an anti-HLA class I (W6/32) MAb in TIL compared to PBL (p < 0.05). CTL populations derived from TIL and PBL did not lyse autologous EBV-transformed lymphoblastoid cell lines, and showed negligible cytotoxicity against the NK-sensitive cell line K562. Furthermore, in both CD8+ T cell populations the majority of the tumor-specific CTL exhibited a Th1 cytokine bias (IFN-gamma(high)/IL-4(low)). Taken together, these data show that tumor lysate-pulsed mature DC can consistently restore tumor-specific lytic activity in non-cytotoxic breast cancer TIL. These results may have important implications for the treatment of chemotherapy resistant breast cancer with active or adoptive immunotherapy.
Collapse
Affiliation(s)
- Rena Kass
- Department of General Surgery, University of Arkansas for Medical Sciences, Little Rock, AR 72205-7199, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
39
|
Santin AD, Bellone S, Palmieri M, Bossini B, Cane' S, Bignotti E, Roman JJ, Cannon MJ, Pecorelli S. Restoration of tumor specific human leukocyte antigens class I-restricted cytotoxicity by dendritic cell stimulation of tumor infiltrating lymphocytes in patients with advanced ovarian cancer. Int J Gynecol Cancer 2004; 14:64-75. [PMID: 14764031 DOI: 10.1111/j.1048-891x.2004.014175.x] [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] [Indexed: 12/24/2022] Open
Abstract
Despite the large number of potentially cytotoxic tumor-infiltrating (TIL) and tumor-associated (TAL) lymphocytes accumulated in the peritoneal cavity ascitic fluid and tumor tissue, advanced ovarian cancer is a progressive disease, suggesting that TIL and TAL populations eventually become functionally suppressed in vivo. Dendritic cells (DC) are the most powerful professional antigen presenting cells known in humans and recently, ovarian tumor antigen pulsed DC have been shown to elicit tumor specific human leukocyte antigens (HLA)-class I-restricted cytotoxicity from the peripheral blood of advanced ovarian cancer patients. In this study, we have evaluated the potential of tumor antigen-pulsed fully mature DC stimulation in restoring tumor-specific cytotoxicity in anergic TIL populations from advanced ovarian cancer patients. In addition, we have compared tumor-specific T-cell responses induced by tumor antigen-loaded DC in TIL to those induced in TAL and peripheral blood lymphocytes (PBL). DC stimulation induced powerful cytotoxicity against autologous tumor target cells in TIL-derived CD8+ T-cells from all patients tested, while autologous Epstein-Barr virus (EBV)-transformed lymphoblastoid cell lines (LCL) were not lysed. Killing of autologous tumor cells was higher by CD8+ T-cells from TIL compared to PBL and TAL (P < 0.01) and was more strongly inhibited by anti-HLA class I MAb (P < 0.05 compared to PBL and TAL). Phenotypically, all cytotoxic T lymphocyte (CTL) populations were CD3+/CD8+, with variable levels of CD56 expression. Finally, although a marked Type 1 cytokine bias [ie, interferon-gamma/interleukin-4 (IFN-gammahigh/IL-4low)] was observable in all DC-stimulated CD8+ T-cell populations, TIL derived CD8+ T-cells showed a higher percentage of IFN-gamma positive cells compared to TAL and PBL. Taken together, these data show that tumor lysate-pulsed DC can consistently restore strong CD8+ CTL responses from TIL against autologous ovarian cancer cells. DC-stimulated TIL may represent a superior source of tumor-specific CTL for adoptive T-cell immunotherapy for advanced ovarian cancer.
Collapse
Affiliation(s)
- A D Santin
- Department of Obstetrics and Gynecology, Division of Gynecologic Oncology, UAMS Medical Center, University of Arkansas for Medical Sciences, Little Rock, AR 72205-7199, USA.
| | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Irie M, Homma S, Komita H, Zeniya M, Kufe D, Ohno T, Toda G. Inhibition of spontaneous development of liver tumors by inoculation with dendritic cells loaded with hepatocellular carcinoma cells in C3H/HeNCRJ mice. Int J Cancer 2004; 111:238-45. [PMID: 15197777 DOI: 10.1002/ijc.20247] [Citation(s) in RCA: 19] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/04/2023]
Abstract
We attempted to prevent spontaneous development of liver tumors by s.c. inoculation with DCs loaded with syngeneic HCC cells in C3H/HeNCrj mice. A new cell line, MIH-2, was established from an HCC that had developed spontaneously in a C3H/HeNCrj mouse. Bone marrow-derived DCs were loaded with irradiated MIH-2 cells by treatment with PEG. Fluorescence microscopy and flow-cytometric analysis showed that about 45% of PEG-treated DCs and MIH-2 cells (DC/MIH-2) were DCs loaded with MIH-2 cells. Thirteen-month-old mice received inoculations of DC/MIH-2 (9 x 10(5)/mouse) 4 times at 6-day intervals and were killed at 16 months of age to assess liver tumors. The incidence of liver tumors in these mice was significantly lower than that in mice not receiving inoculations (p < 0.05) but similar to that in 13-month-old mice (the age at which inoculation started), indicating that inoculation inhibited the development of new tumors. Splenocytes from inoculated mice, but not those from uninoculated mice, showed cytotoxic activity against MIH-2 cells. Cytotoxic activity was not elicited by CD4(+) T cells, CD8(+) T cells, or DX5(+) cells isolated from splenocytes but was elicited by adherent cells, identified as CD11b(+) macrophages. CD4(+) T cells, but not CD8(+) T cells, from inoculated mice produced IFN-gamma by incubation with DC/MIH-2. Cytotoxicity by splenocytes was attenuated by anti-IFN-gamma antibody. Immunization with DCs loaded with syngeneic HCC cells induces CD4(+) T cells that produce IFN-gamma by response to antigen of HCC, which would lead to macrophage activation to kill liver tumor cells at an early stage.
Collapse
Affiliation(s)
- Masaki Irie
- Division of Gastroenterology and Hepatology, Department of Internal Medicine, Jikei University School of Medicine, Tokyo, Japan
| | | | | | | | | | | | | |
Collapse
|
41
|
Morisaki T, Matsumoto K, Onishi H, Kuroki H, Baba E, Tasaki A, Kubo M, Nakamura M, Inaba S, Yamaguchi K, Tanaka M, Katano M. Dendritic cell-based combined immunotherapy with autologous tumor-pulsed dendritic cell vaccine and activated T cells for cancer patients: rationale, current progress, and perspectives. Hum Cell 2003; 16:175-82. [PMID: 15147037 DOI: 10.1111/j.1749-0774.2003.tb00151.x] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Effective adoptive cancer immunotherapy depends on an ability to generate tumor-antigen-presenting cells and tumor-reactive effector lymphocytes and to deliver these effector cells to the tumor. Dendritic cells (DCs) are the most potent antigen-presenting cells, capable of sensitizing T cells to new and recall antigens. Many studies have shown that tumors express unique proteins that can be loaded on DCs to trigger an immune response. The current experimental and clinical statuses of adoptive transfer of tumor antigen-pulsed DCs and vaccine-primed activated T cells are summarized herein. Clinical trials of antigen-pulsed DCs have been conducted in patients with various types of cancer, including non-Hodgkin lymphoma, multiple myeloma, prostate cancer, renal cell carcinoma, malignant melanoma, colorectal cancer, and non-small cell lung cancer. These studies have shown that antigen-loaded DC vaccination is safe and promising for the treatment of cancer. In addition, tumor vaccine-primed T cells have been shown to induce antitumor activity in vivo. Several clinical studies are being conducted on the use of vaccine-primed T cells such as tumor-drainage lymph node. It is reasonable to consider using both tumor antigen-pulsed DCs and vaccine-primed lymphocytes as adjuvants. We are now investigating the use of autologous whole tumor antigen-pulsed DCs and the DC vaccine-primed activated lymphocytes in patients with multiple metastasis of solid tumors.
Collapse
Affiliation(s)
- Takashi Morisaki
- Department of Cancer Therapy and Research, Station for Collaborative Research, Graduate School of Medical Sciences, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Gillessen S, Naumov YN, Nieuwenhuis EES, Exley MA, Lee FS, Mach N, Luster AD, Blumberg RS, Taniguchi M, Balk SP, Strominger JL, Dranoff G, Wilson SB. CD1d-restricted T cells regulate dendritic cell function and antitumor immunity in a granulocyte-macrophage colony-stimulating factor-dependent fashion. Proc Natl Acad Sci U S A 2003; 100:8874-9. [PMID: 12847287 PMCID: PMC166406 DOI: 10.1073/pnas.1033098100] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
CD1d-restricted T cells contribute to tumor protection, but their precise roles remain unclear. Here we show that tumor cells engineered to secrete granulocyte-macrophage colony-stimulating factor induce the expansion of CD1d-restricted T cells through a mechanism that involves CD1d and macrophage inflammatory protein 2 expression by CD8 alpha-, CD11c+ dendritic cells (DCs). The antitumor immunity stimulated by vaccination with irradiated, granulocyte-macrophage colony-stimulating factor-secreting tumor cells was abrogated in CD1d- and J alpha 281-deficient mice, revealing a critical role for CD1d-restricted T cells in this response. The loss of antitumor immunity was associated with impaired tumor-induced T helper 2 cytokine production, although IFN-gamma secretion and cytotoxicity were preserved. DCs from immunized CD1d-deficient mice showed compromised maturation and function. Together, these results delineate a role for CD1d-restricted T cell-DC cross talk in the shaping of antitumor immunity.
Collapse
Affiliation(s)
- Silke Gillessen
- Department of Medical Oncology, Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
43
|
Kass R, Agha J, Bellone S, Palmieri M, Canè S, Bignotti E, Henry-Tillman R, Hutchins L, Cannon MJ, Klimberg S, Santin AD. In vitro induction of tumor-specific HLA class I-restricted CD8+ cytotoxic T lymphocytes from patients with locally advanced breast cancer by tumor antigen-pulsed autologous dendritic cells. J Surg Res 2003; 112:189-97. [PMID: 12888337 DOI: 10.1016/s0022-4804(03)00147-1] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
BACKGROUND Early dissemination of treatment-resistant tumor cells remains the major cause of metastatic recurrence and death in breast cancer patients. Dendritic cells (DCs) are the most powerful antigen-presenting cells, and recently DC-based vaccination has shown great promise for the treatment of human malignancies by immunological intervention. MATERIALS AND METHODS CD8+ T lymphocytes derived from peripheral blood mononuclear cells stimulated in vitro with autologous breast tumor antigen-pulsed DCs were tested for their ability to induce a HLA class I restricted cytotoxic T lymphocyte (CTL) response against autologous tumor cells. To correlate cytotoxic activity by CTL with T cell phenotype, two-color flow cytometric analysis of surface markers and intracellular cytokine expression was performed. RESULTS DC pulsed with breast tumor extracts consistently elicited a tumor-specific HLA class I restricted CTL response in vitro in three consecutive patients harboring locally advanced breast cancer. CTL expressed strong cytolytic activity against autologous tumor cells but did not lyse autologous Epstein Barr virus-transformed lymphoblastoid cell lines and showed variable cytotoxicity against the natural killer-sensitive cell line K-562. In all patients, two color flow cytometric analysis of surface markers and intracellular cytokine expression demonstrated that tumor-specific CTL exhibited an heterogeneous CD8+/CD56+ expression and a striking Th1 cytokine bias (IFNgamma(high)/IL-4 (low)). CONCLUSIONS Tumor lysate-pulsed DCs can consistently stimulate specific CD8+ CTLs able to kill autologous tumor cells in patients with locally advanced breast cancer in vitro. Tumor antigen-pulsed DC-based vaccinations may be appropriate for the treatment of residual and/or chemotherapy-resistant breast cancer refractory to standard salvage treatment modalities.
Collapse
Affiliation(s)
- Rena Kass
- Department of Surgery, University of Arkansas for Medical Sciences, Little Rock, AR 72205, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
44
|
Weber M, Lange C, Günther W, Franz M, Kremmer E, Kolb HJ. Minor histocompatibility antigens on canine hemopoietic progenitor cells. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2003; 170:5861-8. [PMID: 12794111 DOI: 10.4049/jimmunol.170.12.5861] [Citation(s) in RCA: 23] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Adoptive immunotherapy with CTL against minor histocompatibility Ags (mHA) provides a promising way to treat leukemia relapse in allogeneic chimeras. Here we describe the in vitro generation of CTL against mHA in the dog. We tested their inhibitory effect on the growth of hemopoietic progenitor cells stimulated by hemopoietic growth factors in a 4-day suspension culture. CTL were produced by coculture of donor PBMC with bone marrow-derived dendritic cells (DCs). These DCs were characterized by morphology, high expression of MHC class II and CD1a, and the absence of the monocyte-specific marker CD14. Characteristically these cells stimulated allogeneic lymphocytes (MLR) and, after pulsing with a foreign Ag (keyhole limpet hemocyanin), autologous T cells. CTL were generated either ex vivo by coculture with DCs of DLA-identical littermates or in vivo by immunization of the responder with DCs obtained from a DLA-identical littermate. In suspension culture assays the growth of hemopoietic progenitor cells was inhibited in 53% of DLA-identical littermate combinations. In canine families mHA segregated with DLA as restriction elements. One-way reactivity against mHA was found in five littermate combinations. In two cases mHA might be Y chromosome associated, in three cases autosomally inherited alleles were detected. We conclude that CTL can be produced in vitro and in vivo against mHA on canine hemopoietic progenitor cells using bone marrow-derived DCs.
Collapse
Affiliation(s)
- Martin Weber
- Clinical Cooperative Group Hemopoietic Cell Transplantation, University of Munich, Munich, Germany.
| | | | | | | | | | | |
Collapse
|
45
|
Abstract
The development of biochemical and genetic schemes to characterize cancer antigens led to the recognition that malignant melanoma frequently evokes a host response. While the generation of brisk T-cell infiltrates in early stage disease is correlated with prolonged survival, host reactions in most cases are insufficient to impede tumor progression. One variable that may limit the potency of the host response against nascent melanoma is the mixture of cytokines present in the tumor microenvironment. In a murine melanoma model, we identified granulocyte-macrophage colony stimulating factor (GM-CSF) as the most potent molecule for augmenting tumor immunity following gene transfer into melanoma cells. Vaccination with irradiated melanoma cells engineered to secrete GM-CSF enhances host responses through improved tumor antigen presentation by recruited dendritic cells and macrophages. Melanoma-specific CD4(+) and CD8(+) T-cells, CD1d-restricted NKT-cells, and antibodies mediate tumor rejection. Initial testing of this immunization strategy in patients with metastatic melanoma revealed the consistent induction of cellular and humoral antitumor responses that provoked the extensive necrosis of distant metastases without significant toxicity.
Collapse
Affiliation(s)
- Glenn Dranoff
- Department of Adult Oncology, Dana-Farber Cancer Institute, Boston, MA 02115, USA.
| |
Collapse
|
46
|
Hodi FS, Mihm MC, Soiffer RJ, Haluska FG, Butler M, Seiden MV, Davis T, Henry-Spires R, MacRae S, Willman A, Padera R, Jaklitsch MT, Shankar S, Chen TC, Korman A, Allison JP, Dranoff G. Biologic activity of cytotoxic T lymphocyte-associated antigen 4 antibody blockade in previously vaccinated metastatic melanoma and ovarian carcinoma patients. Proc Natl Acad Sci U S A 2003; 100:4712-7. [PMID: 12682289 PMCID: PMC153621 DOI: 10.1073/pnas.0830997100] [Citation(s) in RCA: 748] [Impact Index Per Article: 34.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 02/19/2003] [Indexed: 12/12/2022] Open
Abstract
A large number of cancer-associated gene products evoke immune recognition, but host reactions rarely impede disease progression. The weak immunogenicity of nascent tumors contributes to this failure in host defense. Therapeutic vaccines that enhance dendritic cell presentation of cancer antigens increase specific cellular and humoral responses, thereby effectuating tumor destruction in some cases. The attenuation of T cell activation by cytotoxic T lymphocyte-associated antigen 4 (CTLA-4) further limits the potency of tumor immunity. In murine systems, the administration of antibodies that block CTLA-4 function inhibits the growth of moderately immunogenic tumors and, in combination with cancer vaccines, increases the rejection of poorly immunogenic tumors, albeit with a loss of tolerance to normal differentiation antigens. To gain a preliminary assessment of the biologic activity of antagonizing CTLA-4 function in humans, we infused a CTLA-4 blocking antibody (MDX-CTLA4) into nine previously immunized advanced cancer patients. MDX-CTLA4 stimulated extensive tumor necrosis with lymphocyte and granulocyte infiltrates in three of three metastatic melanoma patients and the reduction or stabilization of CA-125 levels in two of two metastatic ovarian carcinoma patients previously vaccinated with irradiated, autologous granulocyte-macrophage colony-stimulating factor-secreting tumor cells. MDX-CTLA4 did not elicit tumor necrosis in four of four metastatic melanoma patients previously immunized with defined melanosomal antigens. No serious toxicities directly attributable to the antibody were observed, although five of seven melanoma patients developed T cell reactivity to normal melanocytes. These findings suggest that CTLA-4 antibody blockade increases tumor immunity in some previously vaccinated cancer patients.
Collapse
Affiliation(s)
- F Stephen Hodi
- Department of Adult Oncology, Dana-Farber Cancer Institute and Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
47
|
Abstract
There is a renewed enthusiasm for therapeutic vaccination as a viable treatment for patients with cancer. Early tumor vaccines were comprised of whole tumor cells, fragments of tumor cells, or protein lysate from tumor cells. Limited results with these approaches led investigators to begin developing the next generation of cancer vaccines based on defined tumor-associated antigens (TAAs). Defining and characterizing TAAs for human cancer, development of new approaches for identifying TAAs, and novel strategies to deliver the antigens as potent therapeutic vaccines have all been the focus of intense research in the past decade and will continue to be the focus for decades to come.
Collapse
Affiliation(s)
- Jennifer D Lewis
- Department of Microbiology and Immunology and the Southwest Cancer Center, Texas Tech University Health Sciences Center, Lubbock, Texas 79430, USA
| | | | | |
Collapse
|
48
|
Mohty M, Olive D, Gaugler B. Leukemic dendritic cells: potential for therapy and insights towards immune escape by leukemic blasts. Leukemia 2002; 16:2197-204. [PMID: 12399962 DOI: 10.1038/sj.leu.2402710] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2002] [Accepted: 05/30/2002] [Indexed: 11/08/2022]
Abstract
Dendritic cells (DCs) are a system of potent antigen-presenting cells (APCs) specialized to initiate primary immune responses. DCs are considered important elements in the induction of specific antitumor cytotoxic effectors. At present, because of potential therapeutic implications, the critical role of DCs in cancer patients is under intensive investigation. Interactions between DCs and acute myeloid leukemia cells represent an attractive model for the study of DC physiology. Moreover, DCs can be a valuable therapeutic tool for the adjuvant treatment of leukemic patients. However, DC subsets in vivo may also be affected by leukemogenesis and may contribute to the escape of leukemia from immune control. The aim of this review is to shed further light on this paradoxical picture where the line between immune tolerance and immune defense is narrow.
Collapse
Affiliation(s)
- M Mohty
- Laboratoire d'Immunologie des Tumeurs, Institut Paoli-Calmettes, Université de la Méditerranée, Marseille, France
| | | | | |
Collapse
|
49
|
Ostler T, Ehl S. A cautionary note on experimental artefacts induced by fetal calf serum in a viral model of pulmonary eosinophilia. J Immunol Methods 2002; 268:211-8. [PMID: 12215389 DOI: 10.1016/s0022-1759(02)00191-6] [Citation(s) in RCA: 36] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
In BALB/c mice, sensitization with the attachment protein (G) of respiratory syncytial virus (RSV) leads to CD4(+) T cell-mediated lung eosinophilia during subsequent challenge with RSV. In this study, we originally intended to test whether activation of RSV-specific cytotoxic T cells by peptide-pulsed dendritic cells (DC) after G protein sensitization could prevent this eosinophilic response. Peptide-pulsed dendritic cells activated CTL, which could mediate protective immunity to RSV. However, DC vaccination aggravated, rather than prevented, pulmonary eosinophilia in G-sensitized mice and also enhanced weight loss upon RSV infection. This was accompanied by preferential pulmonary recruitment of CD4(+) T cells secreting IL-5. The same enhanced Th2-mediated eosinophilic response could be observed in mice that received unloaded dendritic cells and this response occurred even in the absence of prior G sensitization. Since both dendritic cells and RSV were grown in fetal calf serum (FCS)-containing medium, we suspected that FCS had provoked this response. Indeed, neither eosinophilia nor enhanced pathology were observed in mice treated with DC raised in mouse serum. This observation calls for meticulous controls for artefacts induced by fetal calf serum particularly in mouse models of allergic responses of the respiratory tract.
Collapse
Affiliation(s)
- Tobias Ostler
- Children's Hospital, University of Freiburg, Mathildenstrasse 1, Germany
| | | |
Collapse
|
50
|
Abstract
The crafting of genetic and biochemical techniques to identify cancer antigens yielded the unexpected discovery that immune recognition of tumors regularly accompanies cancer development. The failure of the host to suppress tumor formation or attenuate disease progression may thus reflect the limited immunogenicity of nascent tumors. One critical determinant of host immunity is the mixture of cytokines produced in the tumor microenvironment. We have compared a large number of secreted and surface molecules for their relative abilities to augment tumor immunity following gene transfer into cancer cells. In multiple murine models, granulocyte-macrophage colony stimulating factor (GM-CSF) proved to be the most potent immunostimulatory product. Vaccination with irradiated tumor cells engineered to secrete GM-CSF involves enhanced tumor antigen presentation by recruited dendritic cells (DCs) and macrophages; the coordinated functions of CD4+ and CD8+ T cells, CD1d-restricted NKT cells and antibodies mediate protective immunity. The evaluation of this vaccination strategy in patients with advanced melanoma revealed the consistent induction of cellular and humoral antitumor responses capable of effectuating substantial necrosis of distant metastases. The formulation of simplified methods for manufacturing autologous, GM-CSF-secreting tumor cells has enabled more extensive clinical testing in diverse patient settings.
Collapse
Affiliation(s)
- Glenn Dranoff
- Department of Adult Oncology, Dana-Farber Cancer Institute and Department of Medicine, Brigham and Women's Hospital and Harvard Medical School, Boston, MA 02115, USA.
| |
Collapse
|