101
|
Enk AH, Becker JC, Schuler G. Immunotherapy of malignant melanoma - Basic principles and novel therapeutic approaches. J Dtsch Dermatol Ges 2006. [DOI: 10.1111/j.1610-0387.2005.05038.x-i1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
102
|
Young MRI. Cytokine-containing gelfoam implants at a postsurgical tumor excision site to stimulate local immune reactivity. Int J Cancer 2006; 119:133-8. [PMID: 16450395 DOI: 10.1002/ijc.21806] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
We previously demonstrated increased numbers of CD34(+) progenitor cells in the peripheral blood of tumor bearers. Also demonstrated was the feasibility of chemoattracting these cells by sponge implants containing VEGF. The present study used a murine Lewis lung carcinoma (LLC) model to test if CD34(+) cells that are chemoattracted to a tumor excision site can be differentiated in situ into dendritic cells and whether this leads to increased local immune reactivity. After surgically excising established LLC tumors, mice received at the excision site gelatin sponge implants containing VEGF to chemoattract CD34(+) cells, and/or GM-CSF plus SCF to induce CD34(+) cell differentiation into dendritic cells. In some studies, lysates of GFP-transfected LLC cells (LLC(GFP)) were also included in the implants as a source of tumor antigen. After 2 weeks, implants and local lymph nodes were removed and analyzed. Implants containing VEGF, GM-CSF/SCF or VEGF/GM-CSF/SCF had a higher proportion of CD34(+) cells compared to control implants. However, the number of dendritic cells was higher in implants containing GM-CSF/SCF or VEGF/GM-CSF/SCF than those containing either VEGF or diluent. Regional lymph node from mice containing GM-CSF/SCF or VEGF/GM-CSF/SCF implants showed increased dendritic cell levels. However, when lysates from LLC(GFP) were added to the implants, the highest proportion of dendritic cells associated with GFP was in lymph nodes of mice containing GM-CSF/SCF implants. Lymph node cells from mice with GM-CSF/SCF or VEGF/GM-CSF/SCF had a higher level of proliferation and IFN-gamma secretion in response to in vitro LLC lysate challenge, with the greatest response being from lymph node cells of mice with GM-CSF/SCF implants. These results suggest the feasibility of using GM-CSF/SCF-containing implants to increase dendritic cell levels, uptake of tumor antigens, trafficking to lymph nodes and stimulation of immune reactivity at tumor excision sites with residual tumor.
Collapse
Affiliation(s)
- M Rita I Young
- Department of Research Services, Ralph H. Johnson V.A. Medical Center, Charleston, SC 29401, USA.
| |
Collapse
|
103
|
Jack A, Boyes C, Aydin N, Alam K, Wallack M. The treatment of melanoma with an emphasis on immunotherapeutic strategies. Surg Oncol 2006; 15:13-24. [PMID: 16815006 DOI: 10.1016/j.suronc.2006.05.003] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
Melanoma continues to be one of the most difficult to treat of all solid tumors. Many new advances have been made in the surgical management of melanoma, including new guidelines for margins of excision, as well as sentinel node biopsy for the diagnosis of lymph node micrometastases. The search continues for an effective adjuvant melanoma treatment that can prevent local and distant recurrences. Melanoma is one of the most immunogenic of all tumors, and several clinical trials testing the immunotherapy of melanoma have been conducted, including trials in interferon, interleukin-2, and melanoma vaccines. Here we discuss many of the recent clinical trials in the surgical management of melanoma, in addition to the advances that have been made in the field of immunotherapy. A new second-generation melanoma vaccine, DC-MelVac (patent # 11221/5), has recently been granted FDA approval for Phase I clinical trials and will be introduced in this review.
Collapse
Affiliation(s)
- Angela Jack
- Surgery Research Laboratory, Department of Surgery, Saint Vincent's Catholic Medical Centers/New York Medical College, 153 West 11th Street, Cronin Building, Room 667, New York, NY 10011, USA
| | | | | | | | | |
Collapse
|
104
|
Abstract
Novel adjuvant therapies are urgently needed to complement the existing treatment options for breast cancer. The advent of the use of dendritic cells (DCs) for cancer immunotherapy provides a unique opportunity to overcome the relative non-immunogenic property of breast tumours and address the underlying immunodeficiency. To date, the success of this approach has been limited, possibly due to the targeting of specific tumour antigens that rapidly mutate and, thus, become undetectable to the immune system. A more efficient approach would include preparations encompassing multiple antigens, such as those provided by loading of whole tumour cells or tumour RNA. It is proposed that targeting mammary stem cells responsible for resistance to chemo/immunotherapy, through the expression of a broad array of wild-type and mutated tumour antigens in the context of DCs, will become a mainstay for immunotherapy of breast cancer.
Collapse
Affiliation(s)
- Alberto Pinzon-Charry
- Dendritic Cell and Cancer Laboratory, Queensland Institute of Medical Research, Brisbane 4006, Australia
| | | | | |
Collapse
|
105
|
Loveland BE, Zhao A, White S, Gan H, Hamilton K, Xing PX, Pietersz GA, Apostolopoulos V, Vaughan H, Karanikas V, Kyriakou P, McKenzie IFC, Mitchell PLR. Mannan-MUC1-pulsed dendritic cell immunotherapy: a phase I trial in patients with adenocarcinoma. Clin Cancer Res 2006; 12:869-77. [PMID: 16467101 DOI: 10.1158/1078-0432.ccr-05-1574] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022]
Abstract
PURPOSE Tumor antigen-loaded dendritic cells show promise for cancer immunotherapy. This phase I study evaluated immunization with autologous dendritic cells pulsed with mannan-MUC1 fusion protein (MFP) to treat patients with advanced malignancy. EXPERIMENTAL DESIGN Eligible patients had adenocarcinoma expressing MUC1, were of performance status 0 to 1, with no autoimmune disease. Patients underwent leukapheresis to generate dendritic cells by culture ex vivo with granulocyte macrophage colony-stimulating factor and interleukin 4 for 5 days. Dendritic cells were then pulsed overnight with MFP and harvested for reinjection. Patients underwent three cycles of leukapheresis and reinjection at monthly intervals. Patients with clinical benefit were able to continue with dendritic cell-MFP immunotherapy. RESULTS Ten patients with a range of tumor types were enrolled, with median age of 60 years (range, 33-70 years); eight patients were of performance status 0 and two of performance status 1. Dendritic cell-MFP therapy led to strong T-cell IFNgamma Elispot responses to the vaccine and delayed-type hypersensitivity responses at injection sites in nine patients who completed treatments. Immune responses were sustained at 1 year in monitored patients. Antibody responses were seen in three patients only and were of low titer. Side effects were grade 1 only. Two patients with clearly progressive disease (ovarian and renal carcinoma) at entry were stable after initial therapy and went on to further leukapheresis and dendritic cell-MFP immunotherapy. These two patients have now each completed over 3 years of treatment. CONCLUSIONS Immunization produced T-cell responses in all patients with evidence of tumor stabilization in 2 of the 10 advanced cancer patients treated. These data support further clinical evaluation of this dendritic cell-MFP immunotherapy.
Collapse
Affiliation(s)
- Bruce E Loveland
- Austin Research Institute, and Medical Oncology Unit, Austin Hospital, Heidelberg, Melbourne, Victoria, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
106
|
Abstract
Melanoma has been widely studied as a target for immunotherapy because it has been considered more susceptible to immune attack than other tumors and because of the relative ease with which melanoma cells can be adapted to in vitro culture. The availability of hundreds of melanoma cell lines for study has led to the identification of tumor antigens and the development of monoclonal antibodies and T cells against these antigens, revolutionizing the understanding of how the immune system sees and reacts to cancer. This article reviews the recent clinical results of trials exploring different immunotherapy strategies against melanoma.
Collapse
Affiliation(s)
- Petra Rietschel
- Department of Medicine, Memorial Sloan-Kettering Cancer Center, New York, NY 10021, USA
| | | |
Collapse
|
107
|
Abstract
The use of tumour vaccines is being explored as a means of generating effective antitumour immune responses in patients with cancer. Dendritic cells (DCs) are the most potent antigen-presenting cells that are essential for initiating primary immune responses. As such, DCs are being studied as a platform for the design of cancer vaccines. DCs loaded with tumour antigens or whole tumour cell derivatives stimulate tumour-specific immunity. A promising vaccine strategy involves the fusion of DCs with whole tumour cells. DC/tumour fusions express a broad array of tumour antigens, including those yet to be identified, in the context of DC-mediated costimulation. Animal models have demonstrated that vaccination with fusion cells is protective against tumour challenge and results in the regression of established metastatic disease. In vitro human studies have demonstrated that DC/tumour fusions potently stimulate antitumour immunity and lysis of autologous tumour cells. Vaccination of cancer patients with DC/tumour fusions is being studied in Phase I/II clinical trials. Preliminary results demonstrate that generation of a vaccine is feasible and that vaccination is associated with minimal toxicity. Immunological and clinical responses have been found in a subset of patients.
Collapse
Affiliation(s)
- Jacalyn Rosenblatt
- Hematologic Malignancy Bone Marrow Transplant Program, Beth Israel Deaconess Medical Center, 330 Brookline Avenue, KS 121, Boston, MA 02215, USA.
| | | | | |
Collapse
|
108
|
Bosma BM, Metselaar HJ, Mancham S, Boor PPC, Kusters JG, Kazemier G, Tilanus HW, Kuipers EJ, Kwekkeboom J. Characterization of human liver dendritic cells in liver grafts and perfusates. Liver Transpl 2006; 12:384-93. [PMID: 16498646 DOI: 10.1002/lt.20659] [Citation(s) in RCA: 46] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
It is generally accepted that donor myeloid dendritic cells (MDC) are the main instigators of acute rejection after organ transplantation. The aim of the present study was to characterize MDC in human donor livers using liver grafts and perfusates as a source. Perfusates were collected during ex vivo vascular perfusion of liver grafts pretransplantation. MDC, visualized in wedge biopsies by immunohistochemistry with anti-BDCA-1 monoclonal antibody (mAb), were predominantly observed in the portal fields. Liver MDC, isolated from liver wedge biopsies, had an immature phenotype with a low expression of CD80 and CD83. Perfusates were collected from 20 grafts; perfusate mononuclear cells (MNC) contained 1.5% (range, 0.3-6.6%) MDC with a viability of 97 +/- 2%. Perfusates were a rich source of hepatic MDC since 0.9 x 10(6) (range, 0.11-4.5 x 10(6)) MDC detached from donor livers during vascular perfusion pretransplantation. Perfusate MDC were used to further characterize hepatic MDC. Perfusate MDC expressed less DC-LAMP (P = 0.000), CD80 (P = 0.000), CD86 (P = 0.003), and CCR7 (P = 0.014) than mature hepatic lymph node (LN) MDC, and similar CD86 (P = 0.140) and CCR7 (P = 0.262) as and more DC-LAMP (P = 0.007) and CD80 (P = 0.002) than immature blood MDC. Perfusate MDC differed from blood MDC in producing significantly higher amounts of interleukin (IL)-10 in response to lipopolysaccharide (LPS), and in being able to stimulate allogeneic T-cell proliferation. In conclusion, human donor livers contain exclusively immature MDC that detach in high numbers from the liver graft during pretransplantation perfusion. These viable MDC have the capacity to stimulate allogeneic T-cells, and thus may represent a major player in the induction of acute rejection.
Collapse
Affiliation(s)
- Brenda M Bosma
- Department of Gastroenterology and Hepatology, Erasmus MC-University Medical Center, Rotterdam, The Netherlands
| | | | | | | | | | | | | | | | | |
Collapse
|
109
|
Kang Y, Xu CJ, Wu CQ, Liu XS, Zhong CP, Zhang XH, Qiao SY, Gu JR. A novel strategy to compensate the disadvantages of live vaccine using suicide-gene system and provide better antitumor immunity. Vaccine 2005; 24:2141-50. [PMID: 16368168 DOI: 10.1016/j.vaccine.2005.11.023] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/15/2005] [Revised: 11/03/2005] [Accepted: 11/04/2005] [Indexed: 12/09/2022]
Abstract
Fusing dendritic cells (DCs) with tumor cells is a powerful vaccine to increase tumor immunogenicity. To develop more effective and safer therapeutic vaccine, we fused rat bone marrow-derived DCs with ovarian tumor cell line NuTu-19 modified by suicide gene (HSV1-TK gene) to obtain live vaccine against ovarian cancer. Our data showed that immunization of rats with such live vaccine solicited stronger ovarian tumor-specific cytotoxic T lymphocyte responses and induced immunopreventive and immunotherapeutic effects against parental tumor cells in vivo. Live vaccine could be induced to death after ganciclovir administration in vitro and in vivo. Our researches suggest that live vaccine modified with suicide gene might be effective and controllable in the therapy of ovarian cancer.
Collapse
Affiliation(s)
- Yu Kang
- State Key Laboratory of Genetic Engineering, Obstetrics and Gynecology Hospital, Fudan University, Shanghai 200433, PR China
| | | | | | | | | | | | | | | |
Collapse
|
110
|
Abstract
Lymphoproliferative disorders, including follicular lymphoma (FL), multiple myeloma (MM) and chronic lymphatic leukaemia (CLL), are slowly progressive malignancies which remain incurable despite advances in therapy. Harnessing the immune system to recognise and destroy tumours is a promising new approach to treating these diseases. Dendritic cells (DC) are unique antigen-presenting cells that play a central role in the initiation and direction of immune responses. DC loaded ex vivo with tumour-associated antigens and administered as a vaccine have already shown promise in early clinical trials for a number of lymphoproliferative disorders, but the need for improvement is widely agreed. Recent advances in the understanding of basic DC biology and lessons from early clinical trials have provided exciting new insights into the generation of anti-tumour immune responses and the design of vaccine strategies. In this review we provide an overview of our current understanding of DC biology and their function in patients with lymphoproliferative disorders. We discuss the current status of clinical trials and new approaches to exploit the antigen presenting capacity of DC to design vaccines of the future.
Collapse
MESH Headings
- Cancer Vaccines/immunology
- Cancer Vaccines/therapeutic use
- Clinical Trials as Topic
- Dendritic Cells/immunology
- Dendritic Cells/pathology
- Humans
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/pathology
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Lymphoma, Follicular/immunology
- Lymphoma, Follicular/pathology
- Lymphoma, Follicular/therapy
- Lymphoproliferative Disorders/immunology
- Lymphoproliferative Disorders/pathology
- Lymphoproliferative Disorders/therapy
- Multiple Myeloma/immunology
- Multiple Myeloma/pathology
- Multiple Myeloma/therapy
- Vaccination
Collapse
Affiliation(s)
- Kristen J Radford
- Mater Medical Research Institute, Dendritic Cell Laboratory, South Brisbane, Queensland, Australia.
| | | | | |
Collapse
|
111
|
Abstract
Advances in cellular and molecular immunology have led to the development of strategies for effective augmentation of antitumour immune responses in cancer patients. This review focuses on the manipulation of T cell immunity either by active specific immunotherapy (ASI) using tumour vaccines, or by adoptive immunotherapy (ADI) with immune T cells. Such therapies offer exquisite specificity of tumour recognition based on the ability of the T cell to distinguish single amino acid differences in any protein from any compartment of the tumour cell. Examples are presented of clinical survival benefits for cancer patients by postoperative ASI with a modified autologous tumour vaccine of high quality. Furthermore, clinical studies employing ADI with T cells activated and expanded ex vivo have demonstrated 'proof of principle' that tumour-specific T cells are capable of mediating anticancer activity in vivo, as measured by regression of metastatic tumours. Translation of these findings into a standardised immunotherapy is, however, not easy and will require coordination and cooperation among academic, private and federal sectors.
Collapse
Affiliation(s)
- Volker Schirrmacher
- German Cancer Research Center, Division of Cellular Immunology, Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany.
| |
Collapse
|
112
|
Peng JC, Thomas R, Nielsen LK. Generation and Maturation of Dendritic Cells for Clinical Application Under Serum-Free Conditions. J Immunother 2005; 28:599-609. [PMID: 16224278 DOI: 10.1097/01.cji.0000175491.21099.04] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Monocyte-derived dendritic cells (MoDCs) in clinical use for cancer immunotherapy are ideally generated in serum-free medium (SFM) with inclusion of a suitable maturation factor toward the end of the incubation period. Three good manfacturing practice (GMP) grade SFMs (AIM-V, X-VIVO 15, and X-VIVO 20) were compared with RPMI-1640, supplemented with 10% fetal bovine serum or 10% human serum. DCs generated for 7 days in SFM were less mature and secreted less interleukin (IL) 12p70 and IL-10 than DCs generated in 10% serum. DC yield was comparable in SFMs, and a greater proportion of cells was viable after maturation. Toll-like receptor (TLR) ligands were compared for their ability to induce cytokine secretion under serum-free conditions in the presence of interferon (IFN) gamma. With the exception of Poly I:C, TLR ligands stimulated high levels of IL-10 secretion. High levels of IL-12p70 were induced by two TLR4-mediated stimuli, lipopolysaccharide and Ribomunyl, a clinical-grade bacterial extract. When T-cell responses were compared in allogeneic mixed leukocyte reaction, DCs stimulated with Ribomunyl induced higher levels of IFNgamma than DCs stimulated with the cytokine cocktail: tumor necrosis factor-alpha, IL-1beta, IL-6, and prostaglandin E2. In the presence of IL-10 neutralizing antibodies, DC IL-12p70 production and T-cell IFNgamma were increased in vitro. Similarly, DCs stimulated with Ribomunyl, IFNgamma, and anti-IL-10 induced high levels of tetanus toxoid-specific T-cell proliferation and IFNgamma secretion. Thus, MoDCs generated in SFM efficiently stimulate T-cell IFNgamma production after maturation in the presence of a clinical-grade TLR4 agonist and IL-10 neutralization.
Collapse
Affiliation(s)
- Judy C Peng
- Division of Chemical Engineering, University of Queensland, Brisbane, Queensland, Australia
| | | | | |
Collapse
|
113
|
Luiten RM, Kueter EWM, Mooi W, Gallee MPW, Rankin EM, Gerritsen WR, Clift SM, Nooijen WJ, Weder P, van de Kasteele WF, Sein J, van den Berk PCM, Nieweg OE, Berns AM, Spits H, de Gast GC. Immunogenicity, including vitiligo, and feasibility of vaccination with autologous GM-CSF-transduced tumor cells in metastatic melanoma patients. J Clin Oncol 2005; 23:8978-91. [PMID: 16260696 DOI: 10.1200/jco.2005.01.6816] [Citation(s) in RCA: 88] [Impact Index Per Article: 4.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/09/2023] Open
Abstract
PURPOSE To determine the feasibility, toxicity, and immunologic effects of vaccination with autologous tumor cells retrovirally transduced with the GM-CSF gene, we performed a phase I/II vaccination study in stage IV metastatic melanoma patients. PATIENTS AND METHODS Sixty-four patients were randomly assigned to receive three vaccinations of high-dose or low-dose tumor cells at 3-week intervals. Tumor cell vaccine preparation succeeded for 56 patients (88%), but because of progressive disease, the well-tolerated vaccination was completed in only 28 patients. We analyzed the priming of T cells against melanoma antigens, MART-1, tyrosinase, gp100, MAGE-A1, and MAGE-A3 using human leukocyte antigen/peptide tetramers and functional assays. RESULTS The high-dose vaccination induced the infiltration of T cells into the tumor tissue. Three of 14 patients receiving the high-dose vaccine showed an increase in MART-1- or gp100-specific T cells in the peripheral blood during vaccination. Six patients experienced disease-free survival for more than 5 years, and two of these patients developed vitiligo at multiple sites after vaccination. MART-1- and gp100-specific T cells were found infiltrating in vitiligo skin. Upon vaccination, the T cells acquired an effector phenotype and produced interferon-gamma on specific antigenic stimulation. CONCLUSION We conclude that vaccination with GM-CSF-transduced autologous tumor cells has limited toxicity and can enhance T-cell activation against melanocyte differentiation antigens, which can lead to vitiligo. Whether the induction of autoimmune vitiligo may prolong disease-free survival of metastatic melanoma patients who are surgically rendered as having no evidence of disease before vaccination is worthy of further investigation.
Collapse
Affiliation(s)
- Rosalie M Luiten
- Clinical Immunotherapy, Department of Medical Oncology, The Netherlands Cancer Institute, Plesmanlaan 121, 1066 CX Amsterdam, the Netherlands
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
114
|
Ali S, King GD, Curtin JF, Candolfi M, Xiong W, Liu C, Puntel M, Cheng Q, Prieto J, Ribas A, Kupiec-Weglinski J, van Rooijen N, Lassmann H, Lowenstein PR, Castro MG. Combined immunostimulation and conditional cytotoxic gene therapy provide long-term survival in a large glioma model. Cancer Res 2005; 65:7194-204. [PMID: 16103070 PMCID: PMC1242178 DOI: 10.1158/0008-5472.can-04-3434] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022]
Abstract
In spite of preclinical efficacy and recent randomized, controlled studies with adenoviral vectors expressing herpes simplex virus-1 thymidine kinase (HSV1-TK) showing statistically significant increases in survival, most clinical trials using single therapies have failed to provide major therapeutic breakthroughs. Because glioma is a disease with dismal prognosis and rapid progression, it is an attractive target for gene therapy. Preclinical models using microscopic brain tumor models (e.g., < or =0.3 mm3) may not reflect the pathophysiology and progression of large human tumors. To overcome some of these limitations, we developed a syngeneic large brain tumor model. In this model, administration of single therapeutic modalities, either conditional cytotoxicity or immunostimulation, fail. However, when various immunostimulatory therapies were delivered in combination with conditional cytotoxicity (HSV1-TK), only the combined delivery of fms-like tyrosine kinase ligand (Flt3L) and HSV1-TK significantly prolonged the survival of large tumor-bearing animals (> or =80%; P < or = 0.005). When either macrophages or CD4+ cells were depleted before administration of viral therapy, TK + Flt3L therapy failed to prolong survival. Meanwhile, depletion of CD8+ cells or natural killer cells did not affect TK + Flt3L efficacy. Spinal cord of animals surviving 6 months after TK + Flt3L were evaluated for the presence of autoimmune lesions. Whereas macrophages were present within the corticospinal tract and low levels of T-cell infiltration were detected, these effects are not indicative of an overt autoimmune disorder. We propose that combined Flt3L and HSV1-TK adenoviral-mediated gene therapy may provide an effective antiglioma treatment with increased efficacy in clinical trials of glioma.
Collapse
Affiliation(s)
- Sumia Ali
- Molecular Medicine and Gene Therapy Unit, University of Manchester, Manchester, United Kingdom
| | - Gwendalyn D. King
- Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, and Departments of Medicine and Molecular and Medical Pharmacology
| | - James F. Curtin
- Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, and Departments of Medicine and Molecular and Medical Pharmacology
| | - Marianela Candolfi
- Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, and Departments of Medicine and Molecular and Medical Pharmacology
| | - Weidong Xiong
- Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, and Departments of Medicine and Molecular and Medical Pharmacology
| | - Chunyan Liu
- Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, and Departments of Medicine and Molecular and Medical Pharmacology
| | - Mariana Puntel
- Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, and Departments of Medicine and Molecular and Medical Pharmacology
| | - Queng Cheng
- Department of Medicine, School of Medicine, University of Navarra, Pamplona, Spain
| | - Jesus Prieto
- Department of Medicine, School of Medicine, University of Navarra, Pamplona, Spain
| | | | - Jerzy Kupiec-Weglinski
- Surgery, David Geffen School of Medicine, University of California Los Angeles, Los Angeles, California
| | | | - Hans Lassmann
- Institute of Brain Research, Department of Neuroimmunology, University of Vienna, Vienna, Austria
| | - Pedro R. Lowenstein
- Molecular Medicine and Gene Therapy Unit, University of Manchester, Manchester, United Kingdom
- Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, and Departments of Medicine and Molecular and Medical Pharmacology
| | - Maria G. Castro
- Molecular Medicine and Gene Therapy Unit, University of Manchester, Manchester, United Kingdom
- Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, and Departments of Medicine and Molecular and Medical Pharmacology
- Requests for reprints: Maria G. Castro, Gene Therapeutics Research Institute, Cedars-Sinai Medical Center, Davis Building, Room R5090, 8700 Beverly Boulevard, Los Angeles, CA 90048. Phone: 310-423-7303; Fax: 310-423-7308; E-mail:
| |
Collapse
|
115
|
Lennerz V, Fatho M, Gentilini C, Frye RA, Lifke A, Ferel D, Wölfel C, Huber C, Wölfel T. The response of autologous T cells to a human melanoma is dominated by mutated neoantigens. Proc Natl Acad Sci U S A 2005; 102:16013-8. [PMID: 16247014 PMCID: PMC1266037 DOI: 10.1073/pnas.0500090102] [Citation(s) in RCA: 337] [Impact Index Per Article: 16.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Our understanding of pathways leading to antitumor immunity may depend on an undistorted knowledge of the primary antigenic targets of patients' autologous T cell responses. In the melanoma model derived from patient DT, we applied cryopreserved short-term autologous mixed lymphocyte-tumor cell cultures (MLTCs) in combination with an IFN-gamma enzyme-linked immunospot (ELISPOT) assay to cDNA expression screening. We identified three previously unknown peptides processed from melanosomal proteins tyrosinase (presented by HLA-A(*)2601 and -B(*)3801) and gp100 (presented by HLA-B(*)07021) and five neoantigens generated by somatic point mutations in the patient's melanoma. The mutations were found in the genes SIRT2, GPNMB, SNRP116, SNRPD1, and RBAF600. Peptides containing the mutated residues were presented by HLA-A(*)03011, -B(*)07021, and -B(*)3801. Mutation-induced functional impairment was so far demonstrated for SIRT2. Within MLTC responder populations that were independently expanded from the patient's peripheral blood lymphocytes of different years, T cells against mutated epitopes clearly predominated. These results document a high degree of individuality for the cellular antitumor response and support the need for individualizing the monitoring and therapeutic approaches to the primary targets of the autologous T cell response, which may finally lead to a more effective cancer immunotherapy.
Collapse
Affiliation(s)
- Volker Lennerz
- Department of Medicine, Hematology/Oncology, Johannes Gutenberg University, Langenbeckstrasse 1, D-55101 Mainz, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
116
|
Suresh K, Rodriguez-Lecompte JC, Gauldie J, Foley R. Recent advances in immunotherapy of B-CLL using ex vivo modified dendritic cells. ACTA ACUST UNITED AC 2005; 10:189-203. [PMID: 16019468 DOI: 10.1080/10245330500094870] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Abstract
Chronic lymphocytic leukemia (CLL) results from the relentless accumulation of small mature, slowly dividing, monoclonal B-lymphocytes. The clinical course is heterogeneous, some patients with aggressive form of the disease progressing rapidly with early death while others exhibit a more stable, possibly, non-progressing indolent type of the disease lasting many years. Despite progress in modern treatment modalities, relapse invariably occurs and disease still remains incurable. The clinical management of CLL is therefore challenging and considerable effort has been directed towards novel therapeutic strategies aimed at reducing minimal residual disease which can increase remission duration. Recent insight into the role of dendritic cells (DCs) as pivotal antigen presenting cells that initiate immune responses may provide the basis for generating more specific and effective immune responses. Ex-vivo modified and monocyte-derived DCs represents a promising approach within the context of CLL. However, understanding the relationship between DCs and the cellular immune response is crucial in devising strategies for manipulating immune responses. After a brief survey of general properties of DCs, this review focuses on the different approaches exploiting monocyte-derived DCs in CLL, which may help to design novel strategies for phase-I clinical trials.
Collapse
MESH Headings
- Cells, Cultured
- Clinical Trials, Phase I as Topic
- Dendritic Cells/cytology
- Dendritic Cells/immunology
- Dendritic Cells/transplantation
- Humans
- Immunity, Cellular/immunology
- Immunotherapy, Adoptive/methods
- Leukemia, Lymphocytic, Chronic, B-Cell/immunology
- Leukemia, Lymphocytic, Chronic, B-Cell/therapy
- Monocytes/cytology
- Monocytes/immunology
- Monocytes/transplantation
Collapse
Affiliation(s)
- Kalathil Suresh
- Department of Pathology and Molecular Medicine, McMaster University, Hamilton, Ont., Canada, L8N 3Z5 2
| | | | | | | |
Collapse
|
117
|
Schenk-Braat EAM, Bangma CH. Immunotherapy for superficial bladder cancer. Cancer Immunol Immunother 2005; 54:414-23. [PMID: 15565330 PMCID: PMC11033020 DOI: 10.1007/s00262-004-0621-x] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2004] [Accepted: 09/13/2004] [Indexed: 11/28/2022]
Abstract
The treatment of superficial bladder cancer requires adjuvant therapies besides transurethral resection because of a high recurrence rate after this standard treatment alone. Current adjuvant therapies involve intravesical chemotherapy for patients at low and intermediate risk for recurrence and progression, and intravesical bacillus Calmette-Guérin for patients at intermediate and high risk. However, these adjuvant therapies fail in a significant number of patients, dictating the need for new and improved adjuvant treatment modalities for superficial bladder cancer. Immunotherapy aiming at the modulation of the immune system of the patient is a promising alternative adjuvant. This review discusses the current status of the clinical development of various immunotherapy approaches for superficial bladder cancer, including passive immunotherapy, immune stimulants, immunogene therapy and cancer vaccination.
Collapse
Affiliation(s)
- Ellen A M Schenk-Braat
- Department of Urology, Josephine Nefkens Institute, Room Be 362, PO Box 1738, 3000, DR Rotterdam, The Netherlands.
| | | |
Collapse
|
118
|
Grübe M, Melenhorst JJ, Barrett AJ. An APC for every occasion: induction and expansion of human Ag-specific CD4 and CD8 T cells using cellular and non-cellular APC. Cytotherapy 2005; 6:440-9. [PMID: 15512910 DOI: 10.1080/14653240410005230] [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: 10/26/2022]
Abstract
APC are used extensively to induce and expand Ag-specific T cells as well as to test their specificity and function. In the treatment of malignant and infectious diseases, APC are used to stimulate and expand Ag-specific T cells for adoptive transfer, or used directly in vivo to present Ag. The choice of APC to use depends on the particular application and on practical considerations, which include ease of production, availability, reproducibility and (for clinical use) established safety. The diversity of APC in use partly reflects the fact that no single technique of Ag presentation is ideal. For the clinician and laboratory worker alike the field can seem illogical and confusing. In this review we outline the functional requirements of APC for the induction of T cells, classify the APC in common use and describe their laboratory and clinical applications.
Collapse
Affiliation(s)
- M Grübe
- Hematology Branch, National Heart, Lung and Blood Institute, NIH, Bethesda, MD 20892, USA
| | | | | |
Collapse
|
119
|
Abstract
DC-based immunotherapy has shown early promise in clinical cancer trials, and efforts are being made to determine the optimal method of delivery of cells to achieve the best outcome. While it is accepted that mature DCs are required for stimulation of tumor-specific immunity, the route and frequency of injection and the optimal number of cells needed for clinical success are issues that are still being debated. To solve these questions controlled clinical trials are needed.
Collapse
Affiliation(s)
- S M Barratt-Boyes
- Department of Infectious Diseases and Microbiology, Graduate School of Public Health, University of Pittsburgh, Pennsylvania, Pittsburgh, USA
| | | |
Collapse
|
120
|
Affiliation(s)
- S B Ingram
- Mater Health Services, Raymond Terrace, South Brisbane, Queensland, Australia
| | | |
Collapse
|
121
|
Prasad SJ, Farrand KJ, Matthews SA, Chang JH, McHugh RS, Ronchese F. Dendritic cells loaded with stressed tumor cells elicit long-lasting protective tumor immunity in mice depleted of CD4+CD25+ regulatory T cells. THE JOURNAL OF IMMUNOLOGY 2005; 174:90-8. [PMID: 15611231 DOI: 10.4049/jimmunol.174.1.90] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Dendritic cell (DC)-based vaccination represents a promising approach to harness the specificity and potency of the immune system to combat cancer. Finding optimal strategies for tumor Ag preparation and subsequent pulsing of DC, as well as improving the immunogenicity of weak tumor Ags remain among the first challenges of this approach. In this report, we use a prophylactic vaccine consisting of DC loaded with whole, nonmanipulated B16-F10 melanoma cells that had been stressed by heat shock and gamma irradiation. Stressed B16-F10 cells underwent apoptosis and were internalized by bone marrow-derived DC during coculture. Surprisingly, coculture of DC with stressed B16-F10 undergoing apoptosis and necrosis did not induce DC maturation. However, a marked retardation in tumor growth was observed in C57BL/6 mice immunized using DC loaded with stressed B16-F10 cells and subsequently challenged with B16-F10 cells. Growth retardation was further increased by treating DC with LPS before in vivo administration. In vivo depletion studies revealed that both CD8(+) and CD4(+) T cells played a critical role in retarding tumor growth. In addition, treatment with anti-CD25 Ab to deplete CD4(+)CD25(+) regulatory T cells before DC vaccination considerably improved the effect of the vaccine and allowed the development of long-lived immune responses that were tumor protective. Our results demonstrate that depletion of regulatory T cells is an effective approach to improving the success of DC-based vaccination against weakly immunogenic tumors. Such a strategy can be readily applied to other tumor models and extended to therapeutic vaccination settings.
Collapse
Affiliation(s)
- Simon J Prasad
- Malaghan Institute of Medical Research, Wellington South, New Zealand
| | | | | | | | | | | |
Collapse
|
122
|
Weise JB, Maune S, Kabelitz D, Heiser A. [Significance of dendritic cells for the immunotherapy of tumors]. HNO 2005; 53:117-20. [PMID: 15657753 DOI: 10.1007/s00106-004-1201-2] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/25/2022]
Affiliation(s)
- J B Weise
- Klinik für Hals-, Nasen-,Ohrenheilkunde, Kopf- und Halschirurgie, Universitätsklinikum Schleswig-Holstein, Campus Kiel.
| | | | | | | |
Collapse
|
123
|
Maraskovsky E, Sjölander S, Drane DP, Schnurr M, Le TTT, Mateo L, Luft T, Masterman KA, Tai TY, Chen Q, Green S, Sjölander A, Pearse MJ, Lemonnier FA, Chen W, Cebon J, Suhrbier A. NY-ESO-1 protein formulated in ISCOMATRIX adjuvant is a potent anticancer vaccine inducing both humoral and CD8+ t-cell-mediated immunity and protection against NY-ESO-1+ tumors. Clin Cancer Res 2004; 10:2879-90. [PMID: 15102697 DOI: 10.1158/1078-0432.ccr-03-0245] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
NY-ESO-1 is a 180 amino-acid human tumor antigen expressed by many different tumor types and belongs to the family of "cancer-testis" antigens. In humans, NY-ESO-1 is one of the most immunogenic tumor antigens and NY-ESO-1 peptides have been shown to induce NY-ESO-1-specific CD8(+) CTLs capable of altering the natural course of NY-ESO-1-expressing tumors in cancer patients. Here we describe the preclinical immunogenicity and efficacy of NY-ESO-1 protein formulated with the ISCOMATRIX adjuvant (NY-ESO-1 vaccine). In vitro, the NY-ESO-1 vaccine was readily taken up by human monocyte-derived dendritic cells, and on maturation, these human monocyte-derived dendritic cells efficiently cross-presented HLA-A2-restricted epitopes to NY-ESO-1-specific CD8(+) T cells. In addition, epitopes of NY-ESO-1 protein were also presented on MHC class II molecules to NY-ESO-1-specific CD4(+) T cells. The NY-ESO-1 vaccine induced strong NY-ESO-1-specific IFN-gamma and IgG2a responses in C57BL/6 mice. Furthermore, the NY-ESO-1 vaccine induced NY-ESO-1-specific CD8(+) CTLs in HLA-A2 transgenic mice that were capable of lysing human HLA-A2(+) NY-ESO-1(+) tumor cells. Finally, C57BL/6 mice, immunized with the NY-ESO-1 vaccine, were protected against challenge with a B16 melanoma cell line expressing NY-ESO-1. These data illustrate that the NY-ESO-1 vaccine represents a potent therapeutic anticancer vaccine.
Collapse
MESH Headings
- Adjuvants, Immunologic
- Animals
- Antigens, Neoplasm/chemistry
- Antineoplastic Agents/pharmacology
- CD4 Antigens/biosynthesis
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cancer Vaccines
- Cell Line, Tumor
- Disease Progression
- Dose-Response Relationship, Drug
- Enzyme-Linked Immunosorbent Assay
- Epitopes/chemistry
- Escherichia coli/metabolism
- HLA-A2 Antigen/chemistry
- Immunity, Cellular
- Immunoglobulin G/chemistry
- Immunohistochemistry
- Melanoma, Experimental
- Membrane Proteins/chemistry
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Transgenic
- Monocytes/metabolism
- Peptides/chemistry
- Phenotype
- Plasmids/metabolism
- RNA, Messenger/metabolism
- Recombinant Proteins/chemistry
- Reverse Transcriptase Polymerase Chain Reaction
- Time Factors
- Transfection
Collapse
Affiliation(s)
- Eugene Maraskovsky
- Ludwig Institute for Cancer Research, Austin and Repatriation Medical Centre, Melbourne, Victoria, Australia.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
124
|
Barbuto JAM, Ensina LFC, Neves AR, Bergami-Santos P, Leite KRM, Marques R, Costa F, Martins SC, Camara-Lopes LH, Buzaid AC. Dendritic cell-tumor cell hybrid vaccination for metastatic cancer. Cancer Immunol Immunother 2004; 53:1111-8. [PMID: 15185011 PMCID: PMC11032787 DOI: 10.1007/s00262-004-0551-7] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/14/2004] [Accepted: 04/13/2004] [Indexed: 01/19/2023]
Abstract
Dendritic cells are the most potent antigen-presenting cells, and the possibility of their use for cancer vaccination has renewed the interest in this therapeutic modality. Nevertheless, the ideal immunization protocol with these cells has not been described yet. In this paper we describe the preliminary results of a protocol using autologous tumor and allogeneic dendritic hybrid cell vaccination every 6 weeks, for metastatic melanoma and renal cell carcinoma (RCC) patients. Thirty-five patients were enrolled between March 2001 and March 2003. Though all patients included presented with large tumor burdens and progressive diseases, 71% of them experienced stability after vaccination, with durations up to 19 months. Among RCC patients 3/22 (14%) presented objective responses. The median time to progression was 4 months for melanoma and 5.7 months for RCC patients; no significant untoward effects were noted. Furthermore, immune function, as evaluated by cutaneous delayed-type hypersensitivity reactions to recall antigens and by peripheral blood proliferative responses to tumor-specific and nonspecific stimuli, presented a clear tendency to recover in vaccinated patients. These data indicate that dendritic cell-tumor cell hybrid vaccination affects the natural history of advanced cancer and provide support for its study in less advanced patients, who should, more likely, benefit even more from this approach.
Collapse
Affiliation(s)
- Jose Alexandre M Barbuto
- Departamento de Imunologia, ICB-USP, Av. Prof. Lineu Prestes, 1730, São Paulo, SP 05508-000, Brazil.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
125
|
Dead-cell-associated proteins are an important source of antigens for cross-presentation by dendritic cells. Nat Rev Immunol 2004. [DOI: 10.1038/nri1308-c1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
|
126
|
Affiliation(s)
- F O Nestle
- Department of Dermatology, University Hospital of Zurich, Zurich, Switzerland.
| | | |
Collapse
|
127
|
O'Neill DW, Adams S, Bhardwaj N. Manipulating dendritic cell biology for the active immunotherapy of cancer. Blood 2004; 104:2235-46. [PMID: 15231572 DOI: 10.1182/blood-2003-12-4392] [Citation(s) in RCA: 236] [Impact Index Per Article: 11.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/18/2022] Open
Abstract
Dendritic cells (DCs) are specialized antigen-presenting cells (APCs) that have an unequaled capacity to initiate primary immune responses, including tolerogenic responses. Because of the importance of DCs in the induction and control of immunity, an understanding of their biology is central to the development of potent immunotherapies for cancer, chronic infections, autoimmune disease, and induction of transplantation tolerance. This review discusses recent advances in DC research and the application of this knowledge toward new strategies for the clinical manipulation of DCs for cancer immunotherapy.
Collapse
Affiliation(s)
- David W O'Neill
- New York University School of Medicine, 550 1st Ave, MSB 507, New York, NY 10016, USA
| | | | | |
Collapse
|
128
|
Abstract
Dendritic cells (DCs) are specialized antigen-presenting cells whose immunogenicity leads to the induction of antigen-specific immune responses. DCs can easily be generated ex vivo from peripheral blood monocytes or bone marrow/circulating hematopoietic stem cells cultured in the presence of cytokine cocktails. DCs have been used in numerous clinical trials to induce antitumor immune responses in cancer patients. The studies carried out to date have demonstrated that DCs pulsed with tumor antigens can be safely administered, and this approach produces antigen-specific immune responses. Clinical responses have been observed in a minority of patients. It is likely that either heavy medical pretreatment or the presence of large tumor burdens (or both) is among the causes that impair the benefits of vaccination. Hence, the use of DCs should be considered in earlier stages of disease such as the adjuvant setting. Prospective applications of DCs extend to their use in allogeneic adoptive immunotherapy to specifically target the graft versus tumor reaction. DCs continue to hold promise for cellular immunotherapy, and further investigation is required to determine the clinical settings in which patients will most benefit from the use of this cellular immune adjuvant.
Collapse
Affiliation(s)
- Alessio Nencioni
- Massachusetts Institute of Technology, Center for Cancer Research, Cambridge, USA
| | | |
Collapse
|
129
|
Svane IM, Pedersen AE, Johnsen HE, Nielsen D, Kamby C, Gaarsdal E, Nikolajsen K, Buus S, Claesson MH. Vaccination with p53-peptide-pulsed dendritic cells, of patients with advanced breast cancer: report from a phase I study. Cancer Immunol Immunother 2004; 53:633-41. [PMID: 14985857 PMCID: PMC11032806 DOI: 10.1007/s00262-003-0493-5] [Citation(s) in RCA: 90] [Impact Index Per Article: 4.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/20/2003] [Accepted: 12/02/2003] [Indexed: 11/26/2022]
Abstract
Peptides derived from over-expressed p53 protein are presented by class I MHC molecules and may act as tumour-associated epitopes. Due to the diversity of p53 mutations, immunogenic peptides representing wild-type sequences are preferable as a basis for a broad-spectrum p53-targeting cancer vaccine. Our preclinical studies have shown that wild-type p53-derived HLA-A2-binding peptides are able to activate human T cells and that the generated effector T cells are cytotoxic to human HLA-A2+, p53+ tumour cells. In this phase I pilot study, the toxicity and efficacy of autologous dendritic cells (DCs) loaded with a cocktail of three wild-type and three modified p53 peptides are being analysed in six HLA-A2+ patients with progressive advanced breast cancer. Vaccinations were well tolerated and no toxicity was observed. Disease stabilisation was seen in two of six patients, one patient had a transient regression of a single lymph node and one had a mixed response. ELISpot analyses showed that the p53-peptide-loaded DCs were able to induce specific T-cell responses against modified and unmodified p53 peptides in three patients, including two of the patients with a possible clinical benefit from the treatment. In conclusion, the strategy for p53-DC vaccination seems safe and without toxicity. Furthermore, indications of both immunologic and clinical effect were found in heavily pretreated patients with advanced breast cancer. An independent clinical effect of repeated administration of DCs and IL-2 can not of course be excluded; further studies are necessary to answer these questions.
Collapse
Affiliation(s)
- Inge Marie Svane
- Department of Oncology, Herlev University Hospital, Herlev, Denmark.
| | | | | | | | | | | | | | | | | |
Collapse
|
130
|
Cranmer LD, Trevor KT, Hersh EM. Clinical applications of dendritic cell vaccination in the treatment of cancer. Cancer Immunol Immunother 2004; 53:275-306. [PMID: 14648069 PMCID: PMC11032969 DOI: 10.1007/s00262-003-0432-5] [Citation(s) in RCA: 94] [Impact Index Per Article: 4.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/15/2003] [Accepted: 07/30/2003] [Indexed: 02/06/2023]
Abstract
Dendritic cell (DC) immunotherapy has shown significant promise in animal studies as a potential treatment for cancer. Its application in the clinic depends on the results of human trials. Here, we review the published clinical trials of cancer immunotherapy using exogenously antigen-exposed DCs. We begin with a short review of general properties and considerations in the design of such vaccines. We then review trials by disease type. Despite great efforts on the part of individual investigative groups, most trials to date have not yielded data from which firm conclusions can be drawn. The reasons for this include nonstandard DC preparation and vaccination protocols, use of different antigen preparations, variable means of immune assessment, and nonrigorous criteria for defining clinical response. While extensive animal studies have been conducted using DCs, optimal parameters in humans remain to be established. Unanswered questions include optimal cell dose, use of mature versus immature DCs for vaccination, optimal antigen preparation, optimal route, and optimal means of assessing immune response. It is critical that these questions be answered, as DC therapy is labor- and resource-intensive. Cooperation is needed on the part of the many investigators in the field to address these issues. If such cooperation is not forthcoming, the critical studies that will be required to make DC therapy a clinically and commercially viable enterprise will not take place, and this therapy, so promising in preclinical studies, will not be able to compete with the many other new approaches to cancer therapy presently in development. Trials published in print through June 2003 are included. We exclude single case reports, except where relevant, and trials with so many variables as to prevent interpretation about DC therapy effects.
Collapse
Affiliation(s)
- Lee D. Cranmer
- Department of Hematology and Oncology, The Arizona Cancer Center, University of Arizona/University Medical Center, 1515 N. Campbell Ave., Tucson, AZ 85724 USA
| | - Katrina T. Trevor
- Department of Hematology and Oncology, The Arizona Cancer Center, University of Arizona/University Medical Center, 1515 N. Campbell Ave., Tucson, AZ 85724 USA
| | - Evan M. Hersh
- Department of Hematology and Oncology, The Arizona Cancer Center, University of Arizona/University Medical Center, 1515 N. Campbell Ave., Tucson, AZ 85724 USA
| |
Collapse
|
131
|
|
132
|
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
|