1
|
Jiang C, Li J, Zhang W, Zhuang Z, Liu G, Hong W, Li B, Zhang X, Chao CC. Potential association factors for developing effective peptide-based cancer vaccines. Front Immunol 2022; 13:931612. [PMID: 35967400 PMCID: PMC9364268 DOI: 10.3389/fimmu.2022.931612] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2022] [Accepted: 06/29/2022] [Indexed: 11/26/2022] Open
Abstract
Peptide-based cancer vaccines have been shown to boost immune systems to kill tumor cells in cancer patients. However, designing an effective T cell epitope peptide-based cancer vaccine still remains a challenge and is a major hurdle for the application of cancer vaccines. In this study, we constructed for the first time a library of peptide-based cancer vaccines and their clinical attributes, named CancerVaccine (https://peptidecancervaccine.weebly.com/). To investigate the association factors that influence the effectiveness of cancer vaccines, these peptide-based cancer vaccines were classified into high (HCR) and low (LCR) clinical responses based on their clinical efficacy. Our study highlights that modified peptides derived from artificially modified proteins are suitable as cancer vaccines, especially for melanoma. It may be possible to advance cancer vaccines by screening for HLA class II affinity peptides may be an effective therapeutic strategy. In addition, the treatment regimen has the potential to influence the clinical response of a cancer vaccine, and Montanide ISA-51 might be an effective adjuvant. Finally, we constructed a high sensitivity and specificity machine learning model to assist in designing peptide-based cancer vaccines capable of providing high clinical responses. Together, our findings illustrate that a high clinical response following peptide-based cancer vaccination is correlated with the right type of peptide, the appropriate adjuvant, and a matched HLA allele, as well as an appropriate treatment regimen. This study would allow for enhanced development of cancer vaccines.
Collapse
Affiliation(s)
- Chongming Jiang
- Department of Medicine, Baylor College of Medicine, Houston TX, United States
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, United States
- *Correspondence: Chongming Jiang, ; Cheng-Chi Chao,
| | - Jianrong Li
- Department of Medicine, Baylor College of Medicine, Houston TX, United States
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, United States
| | - Wei Zhang
- Institute of Super Cell, BGI-Shenzhen, Shenzhen, China
| | | | - Geng Liu
- Institute of Super Cell, BGI-Shenzhen, Shenzhen, China
| | - Wei Hong
- Department of Medicine, Baylor College of Medicine, Houston TX, United States
- Dan L Duncan Comprehensive Cancer Center, Baylor College of Medicine, Houston, TX, United States
- Institute for Clinical and Translational Research, Baylor College of Medicine, Houston, TX, United States
| | - Bo Li
- Institute of Super Cell, BGI-Shenzhen, Shenzhen, China
| | - Xiuqing Zhang
- Institute of Super Cell, BGI-Shenzhen, Shenzhen, China
| | - Cheng-Chi Chao
- Department of Pipeline Development, Biomap, Inc, San Francisco, CA, United States
- *Correspondence: Chongming Jiang, ; Cheng-Chi Chao,
| |
Collapse
|
2
|
Fabian KL, Storkus WJ. Immunotherapeutic Targeting of Tumor-Associated Blood Vessels. ADVANCES IN EXPERIMENTAL MEDICINE AND BIOLOGY 2017; 1036:191-211. [PMID: 29275473 DOI: 10.1007/978-3-319-67577-0_13] [Citation(s) in RCA: 13] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Pathological angiogenesis occurs during tumor progression and leads in the formation of an abnormal vasculature in the tumor microenvironment (TME). The tumor vasculature is disorganized, tortuous and leaky, resulting in high interstitial pressure and hypoxia in the TME, all of which are events that support tumor growth and survival. Given the sustaining role of the tumor vasculature, it has become an increasingly attractive target for the development of anti-cancer therapies. Antibodies, tyrosine kinase inhibitors and cancer vaccines that target pro-angiogenic factors, angiogenesis-associated receptors or tumor blood vessel-associated antigens continue to be developed and tested for therapeutic efficacy. Preferred anti-angiogenic protocols include those that "normalize" the tumor-associated vasculature which reduce hypoxia and improve tumor blood perfusion, resulting in tumor cell apoptosis, decreased immunosuppression, and enhanced effector immune cell infiltration/tumoricidal action within the TME.
Collapse
Affiliation(s)
- Kellsye L Fabian
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA.
| | - Walter J Storkus
- Department of Immunology, University of Pittsburgh School of Medicine, Pittsburgh, PA, USA
- Department of Dermatology, University of Pittsburgh Cancer Institute, Pittsburgh, PA, USA
| |
Collapse
|
3
|
Enhanced Dendritic Cell–based Immunotherapy Using Low-dose Cyclophosphamide and CD25-targeted Antibody for Transplanted Lewis Lung Carcinoma Cells. J Immunother 2015; 38:107-15. [DOI: 10.1097/cji.0000000000000068] [Citation(s) in RCA: 10] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
4
|
Lam JS, Belldegrun AS, Figlin RA. Advances in immune-based therapies of renal cell carcinoma. Expert Rev Anticancer Ther 2014; 4:1081-96. [PMID: 15606335 DOI: 10.1586/14737140.4.6.1081] [Citation(s) in RCA: 29] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
Renal cell carcinoma (RCC) is the most prevalent malignancy within the kidney, and over 40% of patients with RCC will die from their disease. Approximately a third of patients present with metastatic disease and a third of patients undergoing nephrectomy for clinically localized RCC will develop metastases. Metastatic RCC poses a therapeutic challenge due to its resistance to conventional modes of therapy such as chemotherapy and radiation therapy. While it is clear that strides have been made against metastatic RCC, the overall objective response rates of interleukin-2- and interferon-alpha-based immunotherapy remain at approximately 20%. Despite advances in biologic- and immune-based therapies, long-term survival for patients with metastatic RCC remains modest. Furthermore, the toxicity profile of these cytokine regimens is significant. Advances in the understanding of the nature of tumor antigens and their optimal presentation, and in the regulatory mechanisms that govern the immune system, have provided multiple novel immunotherapy intervention strategies with increased specificity and potentially fewer side effects. Such strategies are currently being tested in clinical trials.
Collapse
Affiliation(s)
- John S Lam
- Department of Urology, David Geffen School of Medicine at University of California-Los Angeles, Los Angeles, CA 90095, USA.
| | | | | |
Collapse
|
5
|
Zhao X, Bose A, Komita H, Taylor JL, Kawabe M, Chi N, Spokas L, Lowe DB, Goldbach C, Alber S, Watkins SC, Butterfield LH, Kalinski P, Kirkwood JM, Storkus WJ. Intratumoral IL-12 gene therapy results in the crosspriming of Tc1 cells reactive against tumor-associated stromal antigens. Mol Ther 2011; 19:805-14. [PMID: 21189473 PMCID: PMC3070096 DOI: 10.1038/mt.2010.295] [Citation(s) in RCA: 34] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/01/2010] [Accepted: 12/08/2010] [Indexed: 12/20/2022] Open
Abstract
HLA-A2 transgenic mice bearing established HLA-A2(neg) B16 melanomas were effectively treated by intratumoral (i.t.) injection of syngeneic dendritic cells (DCs) transduced to express high levels of interleukin (IL)-12, resulting in CD8(+) T cell-dependent antitumor protection. In this model, HLA-A2-restricted CD8(+) T cells do not directly recognize tumor cells and therapeutic benefit was associated with the crosspriming of HLA-A2-restricted type-1 CD8(+) T cells reactive against antigens expressed by stromal cells [i.e., pericytes and vascular endothelial cells (VEC)]. IL-12 gene therapy-induced CD8(+) T cells directly recognized HLA-A2(+) pericytes and VEC flow-sorted from B16 tumor lesions based on interferon (IFN)-γ secretion and translocation of the lytic granule-associated molecule CD107 to the T cell surface after coculture with these target cells. In contrast, these CD8(+) T effector cells failed to recognize pericytes/VEC isolated from the kidneys of tumor-bearing HHD mice. The tumor-associated stromal antigen (TASA)-derived peptides studied are evolutionarily conserved and could be recognized by CD8(+) T cells harvested from the blood of HLA-A2(+) normal donors or melanoma patients after in vitro stimulation. These TASA and their derivative peptides may prove useful in vaccine formulations against solid cancers, as well as, in the immune monitoring of HLA-A2(+) cancer patients receiving therapeutic interventions, such as IL-12 gene therapy.
Collapse
Affiliation(s)
- Xi Zhao
- Department of Dermatology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania 15213, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
6
|
Abstract
Enthusiasm for therapeutic cancer vaccines has been rejuvenated with the recent completion of several large, randomized phase III clinical trials that in some cases have reported an improvement in progression free or overall survival. However, an honest appraisal of their efficacy reveals modest clinical benefit and a frequent requirement for patients with relatively indolent cancers and minimal or no measurable disease. Experience with adoptive cell transfer-based immunotherapies unequivocally establishes that T cells can mediate durable complete responses, even in the setting of advanced metastatic disease. Further, these findings reveal that the successful vaccines of the future must confront: (i) a corrupted tumor microenvironment containing regulatory T cells and aberrantly matured myeloid cells, (ii) a tumor-specific T-cell repertoire that is prone to immunologic exhaustion and senescence, and (iii) highly mutable tumor targets capable of antigen loss and immune evasion. Future progress may come from innovations in the development of selective preparative regimens that eliminate or neutralize suppressive cellular populations, more effective immunologic adjuvants, and further refinement of agents capable of antagonizing immune check-point blockade pathways.
Collapse
Affiliation(s)
| | - Nicholas Acquavella
- Center for Cancer Research, National Cancer Institute, National Institutes of Health
| | - Zhiya Yu
- Center for Cancer Research, National Cancer Institute, National Institutes of Health
| | - Nicholas P. Restifo
- Center for Cancer Research, National Cancer Institute, National Institutes of Health
| |
Collapse
|
7
|
Saha A, Chatterjee SK. Combination of CTL-associated antigen-4 blockade and depletion of CD25 regulatory T cells enhance tumour immunity of dendritic cell-based vaccine in a mouse model of colon cancer. Scand J Immunol 2010; 71:70-82. [PMID: 20384858 DOI: 10.1111/j.1365-3083.2009.02355.x] [Citation(s) in RCA: 50] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
Immune regulation has been shown to be involved in the progressive growth of some murine tumours. Interruption of immune regulatory pathways via CTL-associated antigen-4 (CTLA-4) blockade or removal of CD4(+) CD25(+) regulatory T (Treg) cells appears to be a promising strategy for cancer immunotherapy. In this study, we tested the hypothesis that the combination of CTLA-4 blockade and depletion of Treg cells would improve the potency of dendritic cell (DC)-based vaccine in a clinically relevant mouse model, which is transgenic for both carcinoembryonic antigen (CEA) and HLA-A2 for the treatment of colon carcinoma in a therapeutic setting. We found that administration of anti-CD25 antibody prior to vaccination or systemic administration of anti-CTLA-4 antibody with the vaccine improved tumour-free survival against CEA-expressing tumours compared with mice immunized with DC-based vaccine alone. However, the efficacy of the vaccine proved to be most effective when anti-CTLA-4 antibody was combined with Treg inhibition. This vaccination strategy dramatically improved the tumour-free survival and allowed the development of long-lasting immune responses. The combined vaccination strategy resulted in increased secretion of IFN-gamma and enhanced HLA-A2-restricted CEA-specific CTL responses. Furthermore, coadministration of anti-CD25 and anti-CTLA-4 antibodies along with the vaccine was effective against more advanced tumours. These results provide evidence that simultaneous blockade of T-cell regulatory pathways is a promising approach for the induction of therapeutic antitumour immunity against CEA(+) colon carcinoma.
Collapse
Affiliation(s)
- Asim Saha
- Department of Internal Medicine, University of Cincinnati Medical Center, Cincinnati, OH, USA.
| | | |
Collapse
|
8
|
Campoli M, Ferris R, Ferrone S, Wang X. Immunotherapy of malignant disease with tumor antigen-specific monoclonal antibodies. Clin Cancer Res 2010; 16:11-20. [PMID: 20028761 PMCID: PMC2802861 DOI: 10.1158/1078-0432.ccr-09-2345] [Citation(s) in RCA: 52] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
A few tumor antigen (TA)-specific monoclonal antibodies (mAb) have been approved by the Food and Drug Administration for the treatment of several major malignant diseases and are commercially available. Once in the clinic, mAbs have an average success rate of approximately 30% and are well tolerated. These results have changed the face of cancer therapy, bringing us closer to more specific and more effective biological therapy of cancer. The challenge facing tumor immunologists at present is represented by the identification of the mechanism(s) underlying the patients' differential clinical response to mAb-based immunotherapy. This information is expected to lead to the development of criteria to select patients to be treated with mAb-based immunotherapy. In the past, in vitro and in vivo evidence has shown that TA-specific mAbs can mediate their therapeutic effect by inducing tumor cell apoptosis, inhibiting the targeted antigen function, blocking tumor cell signaling, and/or mediating complement- or cell-dependent lysis of tumor cells. More recent evidence suggests that TA-specific mAb can induce TA-specific cytotoxic T-cell responses by enhancing TA uptake by dendritic cells and cross-priming of T cells. In this review, we briefly summarize the TA-specific mAbs that have received Food and Drug Administration approval. Next, we review the potential mechanisms underlying the therapeutic efficacy of TA-specific mAbs with emphasis on the induction of TA-specific cellular immune responses and their potential to contribute to the clinical efficacy of TA-specific mAb-based immunotherapy. Lastly, we discuss the potential negative effect of immune escape mechanisms on the clinical efficacy of TA-specific mAb-based immunotherapy.
Collapse
Affiliation(s)
- Michael Campoli
- Department of Dermatology, University of Colorado Health Science Center, Denver, Colorado, USA
| | | | | | | |
Collapse
|
9
|
Wrzesinski C, Paulos CM, Kaiser A, Muranski P, Palmer DC, Gattinoni L, Yu Z, Rosenberg SA, Restifo NP. Increased intensity lymphodepletion enhances tumor treatment efficacy of adoptively transferred tumor-specific T cells. J Immunother 2010; 33:1-7. [PMID: 19952961 PMCID: PMC3247626 DOI: 10.1097/cji.0b013e3181b88ffc] [Citation(s) in RCA: 203] [Impact Index Per Article: 13.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Lymphodepletion before adoptive cell transfer (ACT)-based immunotherapies can enhance anti-tumor responses by augmenting innate immunity, by increasing access to homeostatic cytokines, and by depressing the numbers of regulatory T cells and myeloid-derived suppressor cells. Although it is clear that high-dose total body irradiation given together with hematopoietic stem cell (HSC) transplantation effectively enhances ACT, the relationship between the intensity of lymphodepletion and tumor treatment efficacy has not been systematically studied. Using the pmel-1 mouse model of self/tumor-reactive CD8 T cells, we observed a strong correlation between the intensity of the conditioning regimen and the efficacy of ACT-based treatments using linear regression analysis. This was the case for preparative total body irradiation administered either as a single dose (R=0.97, P<0.001) or in fractionated doses (R=0.94, P<0.001). Increased amounts of preparative total body irradiation were directly correlated with progressively more favorable ratios of transferred tumor-reactive CD8 T cells toward endogenous cells with the potential for inhibitory activity including: CD4 cells (potentially T regulatory cells); Gr1 cells (which are capable of functioning as myeloid-derived suppressor cells); and endogenous CD8 and natural killer 1.1 cells (that can act as "sinks" for homeostatic cytokines in the postablative setting). With increasing ablation, we also observed elevated lipopolysaccharide levels in the sera and heightened levels of systemic inflammatory cytokines. Thus, increased intensity lymphodepletion triggers enhanced tumor treatment efficacy and the benefits of high-dose total body irradiation must be titrated against its risks.
Collapse
Affiliation(s)
- Claudia Wrzesinski
- Surgery Branch, National Cancer Institute, National Institute of Health, Bethesda MD
| | - Chrystal M. Paulos
- Surgery Branch, National Cancer Institute, National Institute of Health, Bethesda MD
| | - Andrew Kaiser
- Surgery Branch, National Cancer Institute, National Institute of Health, Bethesda MD
| | - Pawel Muranski
- Surgery Branch, National Cancer Institute, National Institute of Health, Bethesda MD
| | - Douglas C. Palmer
- Surgery Branch, National Cancer Institute, National Institute of Health, Bethesda MD
- Performed in partial fulfillment of a Ph.D. in Biochemistry (to D.C.P.) at the George Washington University, Washington, DC
| | - Luca Gattinoni
- Surgery Branch, National Cancer Institute, National Institute of Health, Bethesda MD
| | - Zhiya Yu
- Surgery Branch, National Cancer Institute, National Institute of Health, Bethesda MD
| | - Steven A. Rosenberg
- Surgery Branch, National Cancer Institute, National Institute of Health, Bethesda MD
| | - Nicholas P. Restifo
- Surgery Branch, National Cancer Institute, National Institute of Health, Bethesda MD
| |
Collapse
|
10
|
Hong B, Ren W, Song XT, Evel-Kabler K, Chen SY, Huang XF. Human suppressor of cytokine signaling 1 controls immunostimulatory activity of monocyte-derived dendritic cells. Cancer Res 2009; 69:8076-84. [PMID: 19789342 DOI: 10.1158/0008-5472.can-09-1507] [Citation(s) in RCA: 32] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Dendritic cell (DC)-based tumor vaccines have only achieved limited clinical efficacy, underscoring the limitation of stimulatory strategies to elicit effective cytotoxic T lymphocyte (CTL) responses against self-tumor-associated antigens. Here, we investigate the role of human suppressor of cytokine signaling 1 (SOCS1), a feedback inhibitor of the Janus-activated kinase/signal transducer and activator of transcription signaling pathway, in regulating antigen presentation by human DCs (hDC). We find that human SOCS1 (hSOCS1)-silenced DCs have an enhanced stimulatory ability to prime self-antigen-specific CTLs in vitro and in a severe combined immunodeficient-hu mouse model. Human CTLs activated by SOCS1-silenced DCs, but not wild-type DCs, have an active lytic activity to natural antigen-expressing tumor cells. We further find that the capacity of hDCs to prime CTLs is likely controlled by SOCS1-restricted production and signaling of proinflammatory cytokines, such as interleukin-12. These results indicate a critical role of hSOCS1 in negatively regulating the immunostimulatory capacity of DCs and imply a translational potential of this alternative SOCS1 silencing strategy to develop effective DC vaccines.
Collapse
Affiliation(s)
- Bangxing Hong
- Departments of Molecular and Human Genetics, Baylor College of Medicine, Center for Cell and Gene Therapy, Houston, Texas, USA
| | | | | | | | | | | |
Collapse
|
11
|
Riley CL, Mathieu MG, Clark RE, McArdle SEB, Rees RC. Tumour antigen-targeted immunotherapy for chronic myeloid leukaemia: is it still viable? Cancer Immunol Immunother 2009; 58:1489-99. [PMID: 19259670 PMCID: PMC11029949 DOI: 10.1007/s00262-009-0675-x] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2008] [Accepted: 01/29/2009] [Indexed: 01/07/2023]
Abstract
In haematological cancers, malignant cells circulate in the blood and lymphatic system. This may make leukaemic cells easier to target by immunotherapy than in other types of cancer. Various immunotherapy strategies have been trialled in several leukaemias including chronic myeloid leukaemia (CML) and in general, these have been aimed at targeting tumour-associated antigens (TAA). There are numerous TAA expressed by CML patients including WT1, proteinase 3, BCR-ABL and HAGE amongst others. The immunogenicity of the CML-specific tumour antigen, BCR-ABL, has been the subject of much debate and its role in the development of the disease and its unique sequence spanning the breakpoint region make it an ideal target for immunotherapy. However, there are a limited number of immunogenic epitopes across the junctional region, which are restricted to only a few HLA types, namely A2, A3 and B7 (Clark et al. in Blood 98:2887-2893, 2001). The second CML-associated antigen is the helicase antigen HAGE, a cancer-testis antigen found to be over-expressed in more than 50% of myeloid leukaemias (Adams et al. in Leukaemia 16:2238-2242, 2002). Very little is known about the function of this antigen and its significance to CML. However, its membership of the DEAD-box family of ATP-dependent RNA helicases and the involvement of other members of this family in tumour cell proliferation (Eberle et al. in Br J Cancer 86:1957-1962, 2002; Yang et al. in Cell Signal 17:1495-504, 2005) suggest a crucial role in the RNA metabolism of tumour cells. For these reasons, HAGE also seems to be a good target for immunotherapy as it would be applicable for the majority of patients with CML. This review aims to discuss the potential of immunotherapy for the treatment of leukaemia, in particular CML, and the prospect of targeting three CML associated antigens: BCR, ABL and HAGE. During his career, Prof. Tony Dodi made a significant contribution in this area of leukaemia research, confirming the identity of immunogenic HLA-A3 and B7-restricted peptides as targets for CTL. Published, as a highlighted paper in Clark et al. (Blood 98:2887-2893, 2001), this study demonstrated the expression of MHC-peptide complexes on the surface of CML cells and the presence of tetramer-positive CTL activity in CML patients positive for these two HLA alleles. His drive and dedication for research excellence will be remembered by all who knew and worked with him.
Collapse
Affiliation(s)
- Catherine L. Riley
- The John Van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Clifton Lane, Nottingham, NG11 8NS UK
| | - Morgan G. Mathieu
- The John Van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Clifton Lane, Nottingham, NG11 8NS UK
| | - Richard E. Clark
- Department of Haematology, Royal Liverpool University Hospital, Prescott Street, Liverpool, L7 8XP UK
| | - Stephanie E. B. McArdle
- The John Van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Clifton Lane, Nottingham, NG11 8NS UK
| | - Robert C. Rees
- The John Van Geest Cancer Research Centre, School of Science and Technology, Nottingham Trent University, Clifton Lane, Nottingham, NG11 8NS UK
| |
Collapse
|
12
|
Cryopreservation of Monocytes Is Superior to Cryopreservation of Immature or Semi-mature Dendritic Cells for Dendritic Cell-based Immunotherapy. J Immunother 2009; 32:638-54. [DOI: 10.1097/cji.0b013e3181a5bc13] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
|
13
|
Passoni L, Gambacorti-Passerini C. ALK a Novel Lymphoma-associated Tumor Antigen for Vaccination Strategies. Leuk Lymphoma 2009; 44:1675-81. [PMID: 14692518 DOI: 10.1080/1042819031000099625] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/27/2022]
Abstract
The discovery of Tumor Associated Antigens (TAAs) demonstrated that tumor cells can be specifically recognized by the immune system raising the hypothesis that tumors express antigens that Cytotoxic T Lymphocytes (CTLs) can potentially attack. The identification of immunogenic epitopes led to their use as targets to mediate the specific clearance of neoplastic cells by TAA targeting strategies such as vaccination strategies. One of the critical issues in the development of efficient vaccination protocols is the identification of the appropriate TAAs. The TAA should be effective as a "tumor rejection antigen" able to induce an immune response that will affect tumor growth. A distinct pathologic entity characterized by the expression of the Anaplastic Lymphoma Kinase (ALK) protein and named "ALKoma" has recently emerged within the heterogeneous group of CD30+ Anaplastic Large Cell Lymphoma (ALCL). ALK is a receptor tyrosine kinase whose expression is normally restricted to a few scattered cells in the nervous system. Its pathological expression in lymphoma cells is due to a chromosomal translocation that leads to the formation of an ALK-derived oncogenic fusion proteins. ALK fusion proteins ectopically over-expressed and constitutively activated in lymphoid cells play a key role in the neoplastic transformation by the aberrant phosphorylation of intracellular substrates that likely contributes to the molecular pathogenesis of ALCL. The high level of ALK expression in lymphoma cells and its direct role in lymphomagenesis, combined with the fact that normal ALK is expressed at low levels in the immune privileged nervous system, makes ALK an ideal lymphoma-specific target for immunotherapy of ALK+ALCL.
Collapse
Affiliation(s)
- Lorena Passoni
- Oncogenic Fusion Genes and Proteins Unit, Istituto Nazionale per lo Studio e la Cura dei Tumori, Milan, Italy.
| | | |
Collapse
|
14
|
Mutual helper effect in copulsing of dendritic cells with 2 antigens: a novel approach for improvement of dendritic-based vaccine efficacy against tumors and infectious diseases simultaneously. J Immunother 2009; 32:325-32. [PMID: 19342973 DOI: 10.1097/cji.0b013e31819aa31e] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
To develop an efficient dendritic cell (DC)-based immunotherapy protocol, we examined whether simultaneous pulsing of DCs with a given antigen and a third-party antigen could enhance their antigen presentation capacity. Purified splenic DCs of Balb/c mice were pulsed separately with immunoglobulin G, ovalbumin, conalbumin, P15 peptide of Mycobacterium tuberculosis, and prostate-specific antigen or double combinations of the aforementioned antigens. In some settings, DCs pulsed with 1 antigen were mixed equally with those pulsed with another antigen. Antigen-pulsed DCs were injected into the footpad of syngeneic mice and proliferation of whole, CD4 and CD8 depleted lymph node cells was measured after restimulation with cognate antigen. Antigen-specific production of interferon-gamma (IFNgamma) was tested in culture supernatants. Frequency of responding lymph node cells was determined by IFNgamma enzyme-linked immunosorbent spot assay. Our results showed that copulsing of DCs with 2 unrelated antigens increased the capacity of DCs to induce antigen-specific T-cell proliferation against both antigens up to 16-fold. Injection of 2 populations of DCs each pulsed with a different antigen, increased proliferation of primed T cells significantly as well. Both CD4 and CD8 depleted populations showed vigorous proliferative response in copulsing system. In addition, copulsing of DCs with 2 antigens resulted in higher frequency of antigen-specific responding cells and significantly more IFNgamma production. Our results clearly showed that unrelated peptides and proteins could be used to enhance efficacy of DC-based vaccines and in this system, each antigen served to help the other one, a condition that we termed as "mutual helper effect."
Collapse
|
15
|
Hsiao YW, Liao KW, Chung TF, Liu CH, Hsu CD, Chu RM. Interactions of host IL-6 and IFN-gamma and cancer-derived TGF-beta1 on MHC molecule expression during tumor spontaneous regression. Cancer Immunol Immunother 2008; 57:1091-104. [PMID: 18259750 PMCID: PMC11029876 DOI: 10.1007/s00262-007-0446-5] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/07/2007] [Accepted: 12/18/2007] [Indexed: 12/18/2022]
Abstract
Many tumors down-regulate major histocompatibility complex (MHC) antigen expression to evade host immune surveillance. However, there are very few in vivo models to study MHC antigen expression during tumor spontaneous regression. In addition, the roles of transforming growth factor betal (TGF-beta1), interferon gamma (IFN-gamma), and interleukin (IL)-6 in modulating MHC antigen expression are ill understood. We previously reported that tumor infiltrating lymphocyte (TIL)-derived IL-6 inhibits TGF-beta1 and restores natural killing (NK) activity. Using an in vivo canine-transmissible venereal tumor (CTVT) tumor model, we presently assessed IL-6 and TGF-beta involvement associated with the MHC antigen expression that is commonly suppressed in cancers. IL-6, IFN-gamma, and TGF-beta1, closely interacted with each other and modulated MHC antigen expression. In the presence of tumor-derived TGF-beta1, host IFN-gamma from TIL was not active and, therefore, there was low expression of MHC antigen during tumor progression. TGF-beta1-neutralizing antibody restored IFN-gamma-activated MHC antigen expression on tumor cells. The addition of exogenous IL-6 that has potent anti-TGF-beta1 activity restored IFN-gamma activity and promoted MHC antigen expression. IFN-gamma and IL-6 in combination acted synergistically to enhance the expression of MHC antigen. Thus, the three cytokines, IL-6, TGF-beta1, and IFN-gamma, closely interacted to modulate the MHC antigen expression. Furthermore, transcription factors, including STAT-1, STAT-3, IRF-1, NF-kappaB, and CREB, were significantly elevated after IL-6 and IFN-gamma treatment. We conclude that the host IL-6 derived from TIL works in combination with host IFN-gamma to enhance MHC molecule expression formerly inhibited by TGF-beta1, driving the tumor toward regression. It is suggested that the treatment of cancer cells that constitutively secrete TGF-beta1 should incorporate anti-TGF-beta activity. The findings in this in vivo tumor regression model have potential applications in cancer immunotherapy.
Collapse
Affiliation(s)
- Ya-Wen Hsiao
- Department of Veterinary Medicine, National Taiwan University, Taipei, Taiwan, ROC
- Institute of Biomedical Sciences, Academia Sinica, Taipei, Taiwan, ROC
| | - Kuang-Wen Liao
- Department of the Biological Science and Technology, National Chiao Tung University, Hsin-Chu, Taiwan, ROC
| | - Tien-Fu Chung
- Department of Veterinary Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Chen-Hsuan Liu
- Department of Veterinary Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Chia-Da Hsu
- Department of Veterinary Medicine, National Taiwan University, Taipei, Taiwan, ROC
| | - Rea-Min Chu
- Department of Veterinary Medicine, National Taiwan University, Taipei, Taiwan, ROC
- Department of Veterinary Medicine, Animal Cancer Research Center, 1 Roosevelt Road, Section 4, Taipei, 106 Taiwan, ROC
| |
Collapse
|
16
|
Ridolfi L, Petrini M, Fiammenghi L, Riccobon A, Ridolfi R. Human embryo immune escape mechanisms rediscovered by the tumor. Immunobiology 2008; 214:61-76. [PMID: 19159828 DOI: 10.1016/j.imbio.2008.03.003] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/01/2007] [Revised: 02/29/2008] [Accepted: 03/10/2008] [Indexed: 01/05/2023]
Abstract
Towards the end of the 1990s, the two opposing theories on immunosurveillance and immunostimulation were extensively studied by researchers in an attempt to understand the complex mechanisms that regulate the relation between tumors and the host's immune system. Both theories probably have elements that would help us to comprehend how the host can induce anti-tumor clinical responses through stimulation of the immune system and which could also give us a deeper insight into the mechanisms of tumor immunosuppression. The model that most resembles the behavior of tumor cells in terms of growth, infiltration and suppression of the immune system of the environment in which they live is undoubtedly that of the embryonic cell. The fetus behaves like an allogenic transplant within the mother's body, using every means it has to escape from and defend itself against the mother's immune system. The majority of these mechanisms are the same as those found in tumor cells: antigenic loss, lack of expression of classic HLA-I molecules, production of immunosuppressive cytokines, induction of lack of expression of co-stimulatory molecules in antigen presenting cells, and induction of apoptosis in infiltrating lymphocytes, with activation of a type Th2 regulatory lymphocyte response. A careful and comparative study of key mechanisms capable of triggering tolerance or cytotoxicity in both embryonic and tumor cells could prove immensely valuable in designing new strategies for anti-tumor immunotherapy.
Collapse
Affiliation(s)
- Laura Ridolfi
- Immunotherapy and Somatic Cell Therapy Unit, Istituto Scientifico Romagnolo per lo Studio e la Cura dei Tumori, Via Maroncelli 40, Meldola 47014, Italy.
| | | | | | | | | |
Collapse
|
17
|
Song S, Wang Y, Zhang Y, Wang F, He Y, Ren D, Guo Y, Sun S. Augmented induction of CD8+ cytotoxic T-cell response and antitumor effect by DCs pulsed with virus-like particles packaging with CpG. Cancer Lett 2007; 256:90-100. [PMID: 17656012 DOI: 10.1016/j.canlet.2007.06.004] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2007] [Revised: 06/02/2007] [Accepted: 06/05/2007] [Indexed: 11/28/2022]
Abstract
The present study aims at establishing a novel vaccine procedure based on HBc-VLP-pulsed DCs. Immature mice BMDCs could capture HBc-VLP or HBc-VLP packaging CpG efficiently and present the antigen to syngeneic mice spleen T cells in vitro. Immunization with DCs showed that compared to VLP-pulsed DCs, VLP packaging CpG-pulsed DCs elicit stronger T-cell responses in vivo, as measured by both intracellular production of IFN-gamma and in vivo killing assays by Ag-specific T cells. In the B16-pIR-HH tumor therapy model, the growth of established tumors was significantly inhibited by single immunization of DCs pulsed with HBc-VLP packaged with CpG, resulting in significantly longer survival of immunized animals and strikingly, high frequencies (>10% of CD8(+) cells) of protective CTL could be induced and maintained. The mice immunized with DCs treated with HBc-VLP, however, trigger an antitumor effect at the early phase of vaccination, after 20 days of tumor injection, the tumor growth inhibition of VLP-pulsed DCs vaccination was decreased gradually and the fact could be interpreted by the decreasing number of antigen-specific CD8(+) T-cell and IFN-gamma(+)-producing CD8(+) T cell. This study therefore shows that the use of HBc-VLP packaging CpG-pulsed DCs could facilitate the development of effective T-cell-based vaccines.
Collapse
Affiliation(s)
- Shuxia Song
- Department of Medical Genetics, Second Military Medical University, 800 Xiang Yin Road, Shanghai 200433, PR China.
| | | | | | | | | | | | | | | |
Collapse
|
18
|
Durso RJ, Andjelic S, Gardner JP, Margitich DJ, Donovan GP, Arrigale RR, Wang X, Maughan MF, Talarico TL, Olmsted RA, Heston WDW, Maddon PJ, Olson WC. A Novel Alphavirus Vaccine Encoding Prostate-Specific Membrane Antigen Elicits Potent Cellular and Humoral Immune Responses. Clin Cancer Res 2007; 13:3999-4008. [PMID: 17606734 DOI: 10.1158/1078-0432.ccr-06-2202] [Citation(s) in RCA: 55] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE Prostate-specific membrane antigen (PSMA) is an attractive target for active immunotherapy. Alphavirus vaccines have shown promise in eliciting immunity to tumor antigens. This study investigated the immunogenicity of alphavirus vaccine replicon particles (VRP) that encode PSMA (PSMA-VRP). EXPERIMENTAL DESIGN Cells were infected with PSMA-VRP and evaluated for PSMA expression and folate hydrolase activity. Mice were immunized s.c. with PSMA-VRP or purified PSMA protein. Sera, splenocytes, and purified T cells were evaluated for the magnitude, durability, and epitope specificity of the anti-PSMA response. Antibodies were measured by flow cytometry, and cellular responses were measured by IFN-gamma enzyme-linked immunospot and chromium release assays. Cellular responses in BALB/c and C57BL/6 mice were mapped using overlapping 15-mer PSMA peptides. A Good Laboratory Practice-compliant toxicology study was conducted in rabbits. RESULTS PSMA-VRP directed high-level expression of active PSMA. Robust T-cell and B-cell responses were elicited by a single injection of 2 x 10(5) infectious units, and responses were boosted following repeat immunizations. Anti-PSMA responses were detected following three immunizations with 10(2) infectious units and increased with increasing dose. PSMA-VRP was more immunogenic than adjuvanted PSMA protein. Responses to PSMA-VRP were characterized by Th-1 cytokines, potent CTL activity, and IgG2a/IgG2b antibodies. T-cell responses in BALB/c and C57BL/6 mice were directed toward different PSMA peptides. Immunogenic doses of PSMA-VRP were well tolerated in mice and rabbits. CONCLUSIONS PSMA-VRP elicited potent cellular and humoral immunity in mice, and specific anti-PSMA responses were boosted on repeat dosing. PSMA-VRP represents a promising approach for immunotherapy of prostate cancer.
Collapse
Affiliation(s)
- Robert J Durso
- Progenics Pharmaceuticals, Inc., Tarrytown, New York 10591, USA
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
19
|
Hervé P. [Therapeutic applications and promises of cellular and tissue engineering. What strategic choice for the Etablissement Français du Sang?]. Transfus Clin Biol 2007; 14:3-9. [PMID: 17485235 DOI: 10.1016/j.tracli.2007.04.004] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
A new medical field, known as regeneration medicine, is developing and attracting more and more researchers and practitioners. Whereas hematopoietic cell-based therapies have already proven their efficacy in numerous--malignant or not--diseases, non-hematopoietic cell-based therapies have not. They can be useful to dozens, if not hundreds, of patients with various disorders, such as cardiopathy, diabetes, some types of cancer, osteoarticular and neurodegenerative disorders. In these fields, numerous clinical applications are possible for mesenchymal stem cells. Cell and tissue (corneas, bone, skin) therapy products require the definition of pharmaceutical standards with new European requirements in terms of quality and safety. The legitimacy of the Etablissement Français du Sang (EFS) in cell and tissue engineering activities is established, it is recognized by most specialists and by regulatory authorities and has been asserted by the orientations of its "contrat d'objectifs et de moyens". An independent committee has been set up by the EFS President to define an EFS-specific strategy. This committee made up of qualified specialists was required to draw up a rational organization plan for these activities, in order for EFS to be in a position to produce cells and tissues according to pharmaceutical standards. The committee proposals are based on economic data and an inventory of existing cell and tissue engineering activities. Public/private partnerships are required and efforts must focus towards the industrial valorization of EFS expertise in R&D activities and staff know-how. Implementing such a new organization requires national management and the cooperation of institutional actors (university hospitals, cancer treatment centers, universities). For the success of this approach, EFS personnel must be convinced of its legitimacy and new skills must be encouraged. With its numerous assets, EFS can be ambitious and assert itself as a major actor in cell and tissue engineering in Europe.
Collapse
Affiliation(s)
- Patrick Hervé
- Siège de l'EFS, 20, avenue du Stade-de-France, 93218 La Plaine Saint-Denis, France.
| |
Collapse
|
20
|
Soilleux EJ. Immune responses to tumours: current concepts and applications. PROGRESS IN PATHOLOGY 2007:163-198. [DOI: 10.1017/cbo9780511545955.008] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/06/2025]
|
21
|
Mensah GA, Catravas JD, Engelgau MM, Hooper WC, Madeddu P, Ryan US, She JX, Reed E, Vinicor F, Yacoub MH. Vascular endothelium summary statement VI: Research directions for the 21st century. Vascul Pharmacol 2006; 46:330-2. [PMID: 17222586 DOI: 10.1016/j.vph.2006.10.016] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/06/2006] [Accepted: 10/06/2006] [Indexed: 11/17/2022]
Abstract
Effective translation of research advances from the bench to clinical and public health practice at the bedside and in the community at large represents an important step in the health research discovery enterprise. Increasingly, the gap in translating these advances into practice is being recognized. Successfully addressing this translational gap for the prevention and control of chronic diseases will require the development of novel, innovative, and, if necessary, nontraditional approaches. Participants in the 8th International Conference on Vascular Endothelium discussed a variety of novel approaches that have significant promise. Three of these approaches-vaccine development, genomics and proteomics, and tissue engineering-are highlighted in this position statement and strategies for public health practice and research are suggested.
Collapse
Affiliation(s)
- George A Mensah
- National Center for Chronic Disease Prevention and Health Promotion, Centers for Disease Control and Prevention, (CDC), Mailstop K-40, 4770 Buford Highway NE, Atlanta, GA 30341-3717, USA.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
22
|
Shi Y, Roberts AI, Sun E. Dying T lymphocytes call for the death of tumor cells. Cell Res 2006; 16:679-80. [PMID: 16915292 DOI: 10.1038/sj.cr.7310087] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Affiliation(s)
- Yufang Shi
- Department of Molecular Genetics, Microbiology and Immunology, University of Medicine and Dentistry of New Jersey, Robert Wood Johnson Medical School, Piscataway, NJ 08854, USA
| | | | | |
Collapse
|
23
|
Babatz J, Röllig C, Löbel B, Folprecht G, Haack M, Günther H, Köhne CH, Ehninger G, Schmitz M, Bornhäuser M. Induction of cellular immune responses against carcinoembryonic antigen in patients with metastatic tumors after vaccination with altered peptide ligand-loaded dendritic cells. Cancer Immunol Immunother 2006; 55:268-76. [PMID: 16034561 PMCID: PMC11031026 DOI: 10.1007/s00262-005-0021-x] [Citation(s) in RCA: 49] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2005] [Accepted: 05/03/2005] [Indexed: 12/21/2022]
Abstract
PURPOSE Dendritic cells (DCs) are characterized by their extraordinary capacity to induce T-cell responses, providing the opportunity of DC-based cancer vaccination protocols. In the present study, we conducted a phase I/II clinical trial to determine the capability of DCs differentiated from immunomagnetically isolated CD14+ monocytes and pulsed with a carcinoembryonic antigen-derived altered peptide (CEAalt) to induce specific CD8+ T cells in cancer patients. EXPERIMENTAL DESIGN Nine patients with CEA-positive colorectal cancer (n=7) or lung cancer (n=2) were enrolled in this study. Autologous CD14+ monocytes were isolated by large-scale immunomagnetic separation and differentiated to mature DCs in sufficient numbers and at high purity. After incubation with the CEAalt peptide and keyhole limpet hemocyanin, DCs were administered to patients intravenously at dose levels of 1 x 10(7) and 5 x 10(7) cells. Patients received four immunizations every second week. RESULTS ELISPOT analysis revealed a vaccine-induced increase in the number of CEAalt peptide-specific Interferon (IFN)-gamma producing CD8+ T cells in five of nine patients and of CD8+ T lymphocytes recognizing the native CEA peptide in three of nine patients. In addition, CD8+ T lymphocytes derived from one patient exhibiting an immunological response after vaccination efficiently lysed peptide-loaded T2 cells and tumor cells. Immunization was well tolerated by all patients without severe signs of toxicity. CONCLUSION Vaccination with CEAalt-pulsed DCs derived from immunomagnetically isolated CD14+ monocytes efficiently expand peptide-specific CD8+ T lymphocytes in vivo and may be a promising alternative for cancer immunotherapy.
Collapse
Affiliation(s)
- Jana Babatz
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| | - Christoph Röllig
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| | - Bärbel Löbel
- Institut für Immunologie, Medizinische Fakultät, TU Dresden, Germany
| | - Gunnar Folprecht
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| | - Michael Haack
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| | - Heinrich Günther
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| | - Claus-Henning Köhne
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| | - Gerhard Ehninger
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| | - Marc Schmitz
- Institut für Immunologie, Medizinische Fakultät, TU Dresden, Germany
| | - Martin Bornhäuser
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| |
Collapse
|
24
|
Guo ZS, Hong JA, Irvine KR, Chen GA, Spiess PJ, Liu Y, Zeng G, Wunderlich JR, Nguyen DM, Restifo NP, Schrump DS. De novo induction of a cancer/testis antigen by 5-aza-2'-deoxycytidine augments adoptive immunotherapy in a murine tumor model. Cancer Res 2006; 66:1105-13. [PMID: 16424047 PMCID: PMC2242843 DOI: 10.1158/0008-5472.can-05-3020] [Citation(s) in RCA: 122] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023]
Abstract
Recent studies suggest that immunotherapy targeting specific tumor-associated antigens (TAAs) may be beneficial in cancer patients. However, most of these TAAs are tumor type specific and heterogeneous among patients, thus limiting their applications. Here, we describe the de novo induction of a cancer/testis antigen (CTA) for immunotherapy of tumors of various histologies. The murine CTA P1A, normally expressed only in a few tumor lines, could be induced de novo in all P1A-negative cancer lines of eight histologic origins in vitro and in various murine xenografts by systemic administration of 5-aza-2'-deoxycytidine. The induction of P1A expression correlated strongly with demethylation of the CpG island in the promoter region of this gene. The induced antigen was processed and presented properly for recognition by H-2L(d)-restricted P1A-specific CTLs. The combination of a demethylating agent and adoptive transfer of P1A-specific CTL effectively treated lung metastases in syngeneic mice challenged with P1A-negative 4T1 mammary carcinoma cells. These data show a novel strategy of combined chemoimmunotherapy of cancer targeting a CTA induced de novo in a broad range of tumor histologies, and support further evaluation of chromatin-remodeling agents for human cancer therapy.
Collapse
Affiliation(s)
- Z Sheng Guo
- Thoracic Oncology Section and Tumor Immunology Section, Surgery Branch, National Cancer Institute/NIH, Bethesda, MD 20892, USA.
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
25
|
Olver S, Groves P, Buttigieg K, Morris ES, Janas ML, Kelso A, Kienzle N. Tumor-Derived Interleukin-4 Reduces Tumor Clearance and Deviates the Cytokine and Granzyme Profile of Tumor-Induced CD8+ T Cells. Cancer Res 2006; 66:571-80. [PMID: 16397274 DOI: 10.1158/0008-5472.can-05-1362] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
An interleukin (IL)-4-containing tumor environment is reported to be beneficial for immune clearance of tumor cells in vivo; however, the effect of IL-4 on the effector CD8+ T cells contributing to tumor clearance is not well defined. We have used the immunogenic HLA-CW3-expressing P815 (P.CW3) mastocytoma and investigated whether IL-4 expression by the tumor affects tumor clearance and, if so, whether it alters the tumor-induced Vbeta10+ CD8+ T-cell response. P.CW3 were stably transfected with IL-4 or the empty control vector, and independent cell lines were injected i.p. into syngeneic DBA/2 mice. After apparent clearance of primary tumors over 12 to 15 days, secondary tumors arose that lacked surface expression and H-2-restricted antigen presentation of CW3 in part due to the loss of the HLA-CW3 expression cassette. Surprisingly, mice that received IL-4-producing tumor cells showed delayed primary tumor clearance and were significantly more prone to develop secondary tumors compared with mice receiving control tumor cells. Tumor clearance was dependent on CD8+ T cells. The IL-4-secreting P.CW3 tumor cells led to markedly higher mRNA expression of IL-4 and granzyme A and B but no differences in IFN-gamma and IL-2 production, cell proliferation, or ex vivo CTL activity in primary Vbeta10+ CD8+ T cells when compared with the control tumor cells. We concluded that tumor-derived IL-4 selectively changed the quality of the tumor-induced CD8+ T-cell response and resulted in unexpected negative effects on tumor clearance. These data bring into question the delivery of IL-4 to the tumor environment for improving tumor immunotherapy.
Collapse
Affiliation(s)
- Stuart Olver
- Cooperative Research Centre for Vaccine Technology, Brisbane, Queensland, Australia
| | | | | | | | | | | | | |
Collapse
|
26
|
Demaria S, Bhardwaj N, McBride WH, Formenti SC. Combining radiotherapy and immunotherapy: a revived partnership. Int J Radiat Oncol Biol Phys 2005; 63:655-66. [PMID: 16199306 PMCID: PMC1489884 DOI: 10.1016/j.ijrobp.2005.06.032] [Citation(s) in RCA: 281] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/29/2005] [Revised: 06/28/2005] [Accepted: 06/30/2005] [Indexed: 02/07/2023]
Abstract
Ionizing radiation therapy (RT) is an important local modality for the treatment of cancer. The current rationale for its use is based largely on the ability of RT to kill the cancer cells by a direct cytotoxic effect. Nevertheless, considerable evidence indicates that RT effects extend beyond the mere elimination of the more radiosensitive fraction of cancer cells present within a tumor at the time of radiation exposure. For instance, a large body of evidence is accumulating on the ability of RT to modify the tumor microenvironment and generate inflammation. This might have far-reaching consequences regarding the response of a patient to treatment, especially if radiation-induced tumor cell kill were to translate into the generation of effective antitumor immunity. Although much remains to be learned about how radiation can impact tumor immunogenicity, data from preclinical studies provide the proof of principle that different immunotherapeutic strategies can be combined with RT to enhance antitumor effects. Conversely, RT could be a useful tool to combine with immunotherapy. This article will briefly summarize what is known about the impact of RT on tumor immunity, including tumor-associated antigens, antigen-presenting cells, and effector mechanisms. In addition, the experimental evidence supporting the contention that RT can be used as a tool to induce antitumor immunity is discussed, and a new approach to radioimmunotherapy of cancer is proposed.
Collapse
Affiliation(s)
- Sandra Demaria
- Departments of Pathology and
- Address correspondence and reprint requests to: Sandra Demaria, M. D., Department of Pathology, MSB-563, New York University School of Medicine, 550 First Avenue, New York, NY 10016. Tel: (212) 263-7308; Fax: (212) 263-8211; e-mail:
| | - Nina Bhardwaj
- NYU Cancer Institute, New York University School of Medicine, New York, NY 10016
| | - William H. McBride
- Department of Radiation Oncology, Experimental Division, University of California at Los Angeles School of Medicine, Los Angeles, CA 90095, USA
| | | |
Collapse
|
27
|
Dziadek S, Hobel A, Schmitt E, Kunz H. Eine vollsynthetische Vakzine aus einem tumorassoziierten Glycopeptid-Antigen und einem T-Zell-Epitop zur Induktion einer hochspezifischen humoralen Immunantwort. Angew Chem Int Ed Engl 2005. [DOI: 10.1002/ange.200501594] [Citation(s) in RCA: 52] [Impact Index Per Article: 2.6] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
|
28
|
Dziadek S, Hobel A, Schmitt E, Kunz H. A Fully Synthetic Vaccine Consisting of a Tumor-Associated Glycopeptide Antigen and a T-Cell Epitope for the Induction of a Highly Specific Humoral Immune Response. Angew Chem Int Ed Engl 2005; 44:7630-5. [PMID: 16247815 DOI: 10.1002/anie.200501594] [Citation(s) in RCA: 114] [Impact Index Per Article: 5.7] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Affiliation(s)
- Sebastian Dziadek
- Institut für Organische Chemie, Johannes Gutenberg-Universität Mainz, Mainz, Germany
| | | | | | | |
Collapse
|
29
|
Zimmermann VS, Benigni F, Mondino A. Immune surveillance and anti-tumor immune responses: an anatomical perspective. Immunol Lett 2005; 98:1-8. [PMID: 15790503 DOI: 10.1016/j.imlet.2004.09.005] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/10/2004] [Revised: 09/06/2004] [Accepted: 09/10/2004] [Indexed: 11/15/2022]
Abstract
The development of adaptive immune responses against infectious agents relies on the initiation of antigen specific immune responses in secondary lymphoid organs and on the migration of effector cells at the site of infection. Similarly, the development of anti-tumor immunity depends on the recognition of tumor-derived antigens by specific lymphocytes in the context of the lymphoid tissues and on the re-localisation of the cells to the site of cell transformation. Here, we will review the preclinical studies, which have defined the spatial and temporal organisation of anti-tumor immunity, and discuss the implications of these findings in active immunotherapy.
Collapse
Affiliation(s)
- Valerie S Zimmermann
- Cancer Immunotherapy and Gene Therapy Program, San Raffaele Scientific Institute, DIBIT, Via Olgettina 58, 20132 Milan, Italy
| | | | | |
Collapse
|
30
|
Renneson J, Salio M, Mazouz N, Goldman M, Marchant A, Cerundolo V. Mature dendritic cells differentiated in the presence of interferon-beta and interleukin-3 prime functional antigen-specific CD8 T cells. Clin Exp Immunol 2005; 139:468-75. [PMID: 15730392 PMCID: PMC1809309 DOI: 10.1111/j.1365-2249.2005.02700.x] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 11/03/2004] [Indexed: 11/28/2022] Open
Abstract
Dendritic cell (DC)-based immunization represents a promising approach for the immunotherapy of cancer. The optimal conditions required to prepare DCs remain to be defined. Monocytes incubated in the presence of interferon (IFN)-beta and interleukin (IL)-3 give rise to a distinct type of DCs (IFN-beta/IL-3 DCs) that are particularly efficient at eliciting IFN-gamma and IL-5 production by allogeneic helper T cells. We assessed the capacity of this new type of DCs to prime antigen-specific naive CD8(+) T cells and compared them to the conventional DCs differentiated in the presence of granulocyte-macrophage colony stimulating factor (GM-CSF) and IL-4 (GM-CSF/IL-4 DCs). We demonstrate that IFN-beta/IL-3 DCs matured by TLR3 or CD40 ligation efficiently prime Melan-A(26-35)-specific CD8(+) T cells in vitro, at a similar level as GM-CSF/IL-4 DCs. Activated antigen-specific CD8(+) T cells produced IFN-gamma and displayed potent cytotoxic activity against peptide-pulsed target cells. Expansion of CD8(+) T cell numbers was generally higher following priming with CD40-L than with polyinosinic-polycytidylic acid (poly I:C) matured DCs. Cytolytic activity was induced by both maturing agents. These data indicate that IFN-beta/IL-3 DCs represent a promising cell population for the immunotherapy of cancer.
Collapse
Affiliation(s)
- J Renneson
- Cancer Research Tumour Immunology Unit, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Oxford, UK
| | | | | | | | | | | |
Collapse
|
31
|
Abstract
Cancer vaccines - a dream or a reality? There is no doubt that a time will come when this new approach to cancer treatment will provide opportunities that will be both complementary and synergistic with existing treatments. Novelty is often linked to risk, and if there is a given medical field that desperately needs therapeutic improvement, cancer is it. Recent advances on the understanding of the functioning and manipulation of the immune system and on the host-tumour relationship, and on tumour escape, will soon pay off. However, the development of cancer vaccines is not an easy undertaking and the multitude of obstacles are presented and reviewed in this article. The cancer vaccine 'detractors' that are arguing negatively on the chances of success are, to this date, correct. Nevertheless, a parallel could be drawn from the monoclonal antibodies, which have suffered from a long and difficult history, to hold promise in man. Recent technological progress has once again demonstrated that one can eventually bypass the commonly accepted barriers, as there are now monoclonal-based products currently helping patients. The entire oncology community is eagerly awaiting the next chapter in the cancer vaccine story.
Collapse
|
32
|
Demaria S, Kawashima N, Yang AM, Devitt ML, Babb JS, Allison JP, Formenti SC. Immune-Mediated Inhibition of Metastases after Treatment with Local Radiation and CTLA-4 Blockade in a Mouse Model of Breast Cancer. Clin Cancer Res 2005. [DOI: 10.1158/1078-0432.728.11.2] [Citation(s) in RCA: 372] [Impact Index Per Article: 18.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Abstract
Purpose: Ionizing radiation therapy (RT) is an important component in the management of breast cancer. Although the primary tumor can be successfully treated by surgery and RT, metastatic breast cancer remains a therapeutic challenge. Here we tested the hypothesis that the combination of RT to the primary tumor with CTLA-4 blockade can elicit antitumor immunity inhibiting the metastases.
Experimental Design: The poorly immunogenic metastatic mouse mammary carcinoma 4T1 was used as a model. Mice were injected s.c. with 4T1 cells, and treatment was started 13 days later when the primary tumors measured 5 mm in average diameter. Mice were randomly assigned to four treatment groups receiving: (1) control IgG (IgG), (2) RT + IgG, (3) 9H10 monoclonal antibody against CTLA-4, (4) RT + 9H10. RT was delivered to the primary tumor by one or two fractions of 12 Gy. 9H10 and IgG were given i.p. thrice after RT.
Results: Consistent with the fact that 4T1 is poorly immunogenic, 9H10 alone did not have any effect on primary tumor growth or survival. RT was able to delay the growth of the primary irradiated tumor, but in the absence of 9H10 survival was similar to that of control mice. In contrast, mice treated with RT + 9H10 had a statistically significant survival advantage. The increased survival correlated with inhibition of lung metastases formation and required CD8+ but not CD4+ T cells.
Conclusions: The combination of local RT with CTLA-4 blockade is a promising new immunotherapeutic strategy against poorly immunogenic metastatic cancers.
Collapse
Affiliation(s)
| | | | | | | | - James S. Babb
- 3Radiology, New York University School of Medicine, New York, New York; and
| | - James P. Allison
- 4Howard Hughes Medical Institute, University of California at Berkeley, Berkeley, California
| | | |
Collapse
|
33
|
Tiwari R, Geliebter J, Lucchese A, Mittelman A, Kanduc D. Computational peptide dissection of Melan-a/MART-1 oncoprotein antigenicity. Peptides 2004; 25:1865-71. [PMID: 15501517 DOI: 10.1016/j.peptides.2004.07.004] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 05/14/2004] [Revised: 07/14/2004] [Accepted: 07/14/2004] [Indexed: 12/22/2022]
Abstract
We have mapped the linear antigenic determinant of a commercial MAb raised in the mouse against the melanoma-associated-antigen Melan-A/MART-1. The B cell epitope on the Melan-A/MART-1 oncoprotein is located in the 15-mer amino acid sequence 101-115 PPAYEKLSAEQSPPP, within residues 102-106. The definition of the antigenic sequence on Melan-A/MART-1 oncoprotein was reached following analyses of MHC II binding potential and similarity level to the mouse proteome, that put into evidence the 15-mer amino acid sequence 101-115 PPAYEKLSAEQSPPP as the top scoring peptide in binding H2-A(d) molecules and the epitopic sequence residues 102-106 (i.e., the peptide sequence PAYEK) as having low-similarity level to the mouse proteome. Dot-blot epitope mapping immunoassay identified proline residue 102 as critical, based on its effect on antibody recognition. The present study adds to previous companion reports in validating the hypothesis that low-similarity to the host's proteome and binding potential to MHC II molecules are essential concurring factors in the modulation of the pool of epitopic sequences.
Collapse
Affiliation(s)
- Raj Tiwari
- Department of Microbiology and Immunology, New York Medical College, Valhalla, NY 10595, USA
| | | | | | | | | |
Collapse
|
34
|
Campoli M, Ferrone S. T-cell-based immunotherapy of melanoma: what have we learned and how can we improve? Expert Rev Vaccines 2004; 3:171-87. [PMID: 15056043 DOI: 10.1586/14760584.3.2.171] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
The lack of effective treatment for advanced stage melanoma by conventional therapies, such as radiation and chemotherapy, has highlighted the need to develop alternative therapeutic strategies. Among them, immunotherapy has attracted much attention because of the potential role played by immunological events in the clinical course of melanoma and the availability of well-characterized melanoma antigens to target melanoma lesions with immunological effector mechanisms. In recent years, T-cell-based immunotherapy has been emphasized, in part because of the disappointing results of the antibody-based trials conducted in the early 1980s, and in part because of the postulated major role played by T-cells in tumor growth control. In this review, the characteristics of antibody and T-cell-defined melanoma antigens will first be described, with emphasis on those used in clinical trials. Following a review of the current immunization and immunomonitoring strategies, the results from the T-cell-based immunotherapy clinical trials conducted to date will be reviewed.
Collapse
Affiliation(s)
- Michael Campoli
- Department of Immunology, Roswell Park Cancer Institute, Elm and Carlton Streets, Buffalo, NY 14263, USA.
| | | |
Collapse
|
35
|
Affiliation(s)
- David H Munn
- Department of Pediatrics, Institute of Molecular Medicine and Genetics, Medical College of Georgia, Augusta, GA 30912, USA.
| | | |
Collapse
|
36
|
Berzofsky JA, Terabe M, Oh S, Belyakov IM, Ahlers JD, Janik JE, Morris JC. Progress on new vaccine strategies for the immunotherapy and prevention of cancer. J Clin Invest 2004; 113:1515-25. [PMID: 15173875 PMCID: PMC419494 DOI: 10.1172/jci21926] [Citation(s) in RCA: 126] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023] Open
Abstract
In recent years, great strides in understanding and regulating the immune system have led to new hope for harnessing its exquisite specificity to destroy cancer cells without affecting normal tissues. This review examines the fundamental immunologic advances and the novel vaccine strategies arising from these advances, as well as the early clinical trials studying new approaches to treat or prevent cancer.
Collapse
Affiliation(s)
- Jay A Berzofsky
- Molecular Immunogenetics and Vaccine Research Section, Vaccine Branch, Center for Cancer Research, National Cancer Institute, NIH, Bethesda, Maryland 20892, USA.
| | | | | | | | | | | | | |
Collapse
|
37
|
Abstract
Therapeutic cancer vaccines treat disease by stimulating the body's immune system. They are a form of active immunotherapy with the goal of producing an immune response that involves the cellular and humoral components of the immune system. These two components appear to be complementary and work together to induce tumor regression and long-lasting immunity to the disease being treated. This article reviews the history of cancer vaccine development, autologous and allogeneic vaccines, vaccine targets, carrier proteins, adjuvants, and clinical trial data of studies evaluating cancer vaccines. Knowledge of this emerging cancer treatment option will enable oncology nurses to be informed about cancer vaccines and accurately provide information about them to patients.
Collapse
|
38
|
Vlad AM, Kettel JC, Alajez NM, Carlos CA, Finn OJ. MUC1 immunobiology: from discovery to clinical applications. Adv Immunol 2004; 82:249-93. [PMID: 14975259 DOI: 10.1016/s0065-2776(04)82006-6] [Citation(s) in RCA: 168] [Impact Index Per Article: 8.0] [Reference Citation Analysis] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/15/2022]
Affiliation(s)
- Anda M Vlad
- Department of Immunology, University of Pittsburgh School of Medicine Pittsburgh, Pennsylvania 15261, USA
| | | | | | | | | |
Collapse
|
39
|
Di Modugno F, Bronzi G, Scanlan MJ, Del Bello D, Cascioli S, Venturo I, Botti C, Nicotra MR, Mottolese M, Natali PG, Santoni A, Jager E, Nisticò P. Human Mena protein, a serex-defined antigen overexpressed in breast cancer eliciting both humoral and CD8+ T-cell immune response. Int J Cancer 2004; 109:909-18. [PMID: 15027125 DOI: 10.1002/ijc.20094] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Screening of a cDNA expression library from a primary breast tumor with the autologous patient serum led to the isolation of 6 cDNA clones corresponding to 3 different genes, including a novel gene that maps to chromosome 1 and encodes the human homologue of mouse Mena (hMena, cDNA clone RMNY-BR-55), a protein of the Ena/VASP family involved in the regulation of cell motility and adhesion. A cancer-restricted antibody response against hMena was demonstrated, since 18/93 cancer patient sera, the majority (10/52) from breast cancer, showed anti-hMena-specific IgG, while no antibodies were present in healthy donors. When hMena protein expression was analyzed by Western blot and immunohistochemistry, the antigen was overexpressed in the majority of breast cancer cell lines and in 75% of primary breast tumor lesions evaluated. Furthermore, when HLA-A2-restricted peptides from the hMena sequence were used to stimulate CD8+ T cells, an hMena-specific response was found in 9 out of 12 HLA-A2+ breast cancer patients. In 4 patients, this cell-mediated immune response was concomitant with antibody response to hMena. Furthermore, an hMena-specific T-cell line was established from an HLA-A2+ breast cancer patient whose primary tumor lesion overexpressed the hMena protein. The present findings highlight the emerging role that overexpression of cytoskeleton regulatory components may have in the induction of a specific antitumor immune response.
Collapse
Affiliation(s)
- Francesca Di Modugno
- Laboratory of Experimental Chemotherapy, Regina Elena Cancer Institute, Rome, Italy
| | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
40
|
Lu SY, Sui YF, Li ZS, Ye J, Dong HL, Qu P, Zhang XM, Wang WY, Li YS. Superantigen-SEA gene modified tumor vaccine for hepatocellular carcinoma: An in vitro study. World J Gastroenterol 2004; 10:53-7. [PMID: 14695768 PMCID: PMC4717078 DOI: 10.3748/wjg.v10.i1.53] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 12/15/2022] Open
Abstract
AIM: To construct an eukaryotic superantigen gene expression vector containing the recombinant gene of SEA and CD80 molecule transmembrane region (CD80TM), and to express staphylococcus enterotoxin A (SEA) on the membrane of hepatocellular carcinoma (HCC) cell to form a superantigen gene modified tumor vaccine for HCC.
METHODS: SEA and linker-CD80TM gene were amplified through PCR from plasmid containing cDNA of SEA and CD80. Gene fragments were then subcloned into the multiple cloning sites of retroviral vector pLXSN. Recombinant plasmid was transferred into HepG2 cells mediated with lipofectamine, positive clones were selected in culture medium containing G418. RT-PCR and indirect immunofluorescence studies confirmed that SEA was expressed specifically on HCC cell membrane. INFγ -ELISPOT study demonstrated that SEA protein was expressed on the membrane of HCC cells. Cytotoxicity of HepG2-SEA primed CTLs (SEA-T) was analyzed by 51Cr release assay. T cells cultured with rhIL-2 (IL-2-T) were used as control.
RESULTS: Restriction digestion and sequence analyses confirmed the correctness of length, position and orientation of inserted fusion genes. SEA was expressed on the surface of HepG2 cells, HepG2-SEA had strong stimulating effect on production of HepG2 specific CTL (P < 0.001). SEA-T had enhanced cytotoxicity to HepG2 cells (P < 0.05).
CONCLUSION: Tumor cell membrane expressed superantigen can be used to reinforce the immune effect of tumor cell vaccine for HCC, which provides a new method of the enhanced active immunotherapy for HCC.
Collapse
Affiliation(s)
- Shao-Ying Lu
- Department of Pathology, Fourth Military Medical University, Xi'an 710032, China.
| | | | | | | | | | | | | | | | | |
Collapse
|
41
|
Smith CL, Dunbar PR, Mirza F, Palmowski MJ, Shepherd D, Gilbert SC, Coulie P, Schneider J, Hoffman E, Hawkins R, Harris AL, Cerundolo V. Recombinant modified vaccinia Ankara primes functionally activated CTL specific for a melanoma tumor antigen epitope in melanoma patients with a high risk of disease recurrence. Int J Cancer 2004; 113:259-66. [PMID: 15386406 DOI: 10.1002/ijc.20569] [Citation(s) in RCA: 75] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Recombinant plasmid DNA and attenuated poxviruses are under development as cancer and infectious disease vaccines. We present the results of a phase I clinical trial of recombinant plasmid DNA and modified vaccinia Ankara (MVA), both encoding 7 melanoma tumor antigen cytotoxic T lymphocyte (CTL) epitopes. HLA-A*0201-positive patients with surgically treated melanoma received either a "prime-boost" DNA/MVA or a homologous MVA-only regimen. Ex vivo tetramer analysis, performed at multiple time points, provided detailed kinetics of vaccine-driven CTL responses specific for the high-affinity melan-A(26-35) analogue epitope. Melan-A26-35-specific CTL were generated in 2/6 patients who received DNA/MVA (detectable only after the first MVA injection) and 4/7 patients who received MVA only. Ex vivo ELISPOT analysis and in vitro proliferation assays confirmed the effector function of these CTL. Responses were seen in smallpox-vaccinated as well as vaccinia-naive patients, as defined by anti-vaccinia antibody responses demonstrated by ELISA assay. The observations that 1) CTL responses were generated to only 1 of the recombinant epitopes and 2) that the magnitude of these responses (0.029-0.19% CD8(+) T cells) was below the levels usually seen in acute viral infections suggest that to ensure high numbers of CTL specific for multiple recombinant epitopes, a deeper understanding of the interplay between CTL responses specific for the viral vector and recombinant epitopes is required.
Collapse
Affiliation(s)
- Caroline L Smith
- Tumour Immunology Unit, Weatherall Institute of Molecular Medicine, Nuffield Department of Clinical Medicine, Oxford University, Oxford, OX3 9DS, UK
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Tan EM, Shi FD. Relative paradigms between autoantibodies in lupus and autoantibodies in cancer. Clin Exp Immunol 2003; 134:169-77. [PMID: 14616773 PMCID: PMC1808856 DOI: 10.1046/j.1365-2249.2003.02259.x] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Affiliation(s)
- E M Tan
- W M Keck Autoimmune Disease Center, Department of Molecular and Experimental Medicine, The Scripps Research Institute, La Jolla, CA 92037, USA.
| | | |
Collapse
|
43
|
Paczesny S, Ueno H, Fay J, Banchereau J, Palucka AK. Dendritic cells as vectors for immunotherapy of cancer. Semin Cancer Biol 2003; 13:439-47. [PMID: 15001163 DOI: 10.1016/j.semcancer.2003.09.008] [Citation(s) in RCA: 40] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
Dendritic cells (DCs) initiate and regulate immune responses. Numerous studies in mice showed that tumor antigens-loaded DCs are able to induce therapeutic and protective anti-tumor immunity. The immunogenicity of antigens delivered on DCs has now been demonstrated in cancer patients and some clinical responses without any significant toxicity have been observed. Nevertheless, many parameters of DC vaccination need to be established including: (1) the type of DCs, their maturation stage and stimuli; (2) the quality and the breadth of induced immune responses; (3) host-related factors, such as the extent of metastatic disease and myeloablation; and (4) efficacy as measured by the clinical outcome.
Collapse
Affiliation(s)
- Sophie Paczesny
- Baylor Institute for Immunology Research, 3434 Live Oak, Dallas, TX 75204, USA
| | | | | | | | | |
Collapse
|
44
|
Abstract
The current objective of our cancer programme is to develop an effective vaccine based on rationally designed T cell epitope analogues, for use in the adjuvant setting for non-small cell lung cancer (NSCLC) and colon cancer. Analogue epitopes, enhanced for either human leukocyte antigen (HLA) binding or T cell receptor (TCR) signalling, have been shown to be more effective at breaking immunological tolerance than cognate wild-type epitopes. Although encouraging early-phase clinical data has been obtained by others using a limited number of HLA-A2-restricted epitope analogues, the clinical benefits and immune correlates for vaccines comprised of multiple epitope analogues restricted by multiple HLA supertypes remains to be investigated. Clinical studies are currently being conducted on EP-2101, a prototype vaccine that delivers multiple HLA-A2-restricted analogue epitopes. In parallel, fixed anchor and heteroclitic analogues restricted by three other commonly expressed HLA supertypes are being identified. These analogues will be incorporated into future vaccines including optimised minigenes (epigenes) and tested in preclinical and clinical studies addressing various different cancer indications.
Collapse
Affiliation(s)
- John D Fikes
- Epimmune, Inc., 5820 Nancy Ridge Drive, San Diego, CA 92121, USA.
| | | |
Collapse
|
45
|
Teshima T, Reddy P, Liu C, Williams D, Cooke KR, Ferrara JLM. Impaired thymic negative selection causes autoimmune graft-versus-host disease. Blood 2003; 102:429-35. [PMID: 12663438 DOI: 10.1182/blood-2003-01-0266] [Citation(s) in RCA: 73] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
Animal models with impaired thymic negative selection do not always cause autoimmune diseases despite the development of an autoreactive T-cell repertoire. We investigated the requirements for the development of systemic autoimmune disease by using bone marrow chimeras that lacked expression of major histocompatibility complex (MHC) class II on thymic antigen-presenting cells (APCs), leading to impaired negative selection. We found that impaired negative selection mediated by absence of MHC class II, but not MHC class I, permitted the development of systemic autoimmune disease that is indistinguishable from acute graft-versus-host disease (GVHD). Thymectomy prevented disease, confirming the causal association of the thymus with its development. Adoptive transfer of CD4+ T cells caused GVHD in secondary hosts only when they were irradiated, and cotransfer of peripheral CD4+ and CD8+ T cells from naive mice prevented the disease. These results demonstrate that impaired thymic negative selection can cause lethal autoimmune disease indistinguishable from acute GVHD in the context of a proinflammatory milieu when peripheral regulatory mechanisms are absent.
Collapse
Affiliation(s)
- Takanori Teshima
- Department of Internal Medicine, University of Michigan Cancer Center, Ann Arbor, USA
| | | | | | | | | | | |
Collapse
|
46
|
Zöller M. Immunotherapy of cancer by active vaccination: does allogeneic bone marrow transplantation after non-myeloablative conditioning provide a new option? Technol Cancer Res Treat 2003; 2:237-60. [PMID: 12779354 DOI: 10.1177/153303460300200307] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
Abstract
The critical role of antigen-specific T cells in cancer immunotherapy has been amply demonstrated in many model systems. Though success of clinical trials still remains far behind expectation, the continuous improvement in our understanding of the biology of the immune response will provide the basis of optimized cancer vaccines and allow for new modalities of cancer treatment. This review focuses on the current status of active therapeutic vaccination and future prospects. The latter will mainly be concerned with allogeneic bone marrow cell transplantation after non-myeloablative conditioning, because it is my belief that this approach could provide a major breakthrough in cancer immunotherapy. Concerning active vaccination protocols the following aspects will be addressed: i) the targets of immunotherapeutic approaches; ii) the response elements needed for raising a therapeutically successful immune reaction; iii) ways to achieve an optimal confrontation of the immune system with the tumor and iv) supportive regimen of immunomodulation. Hazards which one is most frequently confronted with in trials to attack tumors with the inherent weapon of immune defense will only be briefly mentioned. Many question remain to be answered in the field of allogeneic bone marrow transplantation after non-myeloablative conditioning to optimize the therapeutic setting for this likely very powerful tool of cancer therapy. Current considerations to improve engraftment and to reduce graft versus host disease while strengthening graft versus tumor reactivity will be briefly reviewed. Finally, I will discuss whether tumor-reactive T cells can be "naturally" maintained during the process of T cell maturation in the allogeneic host. Provided this hypothesis can be substantiated, a T cell vaccine will meet a pool of virgin T cells in the allogeneically reconstituted host, which are tolerant towards the host, but not anergised towards tumor antigens presented by MHC molecules of the host.
Collapse
Affiliation(s)
- Margot Zöller
- Dept. of Tumor Progression & Immune Defense, German Cancer Research Center, Im Neuenheimer Feld 280, D-69120 Heidelberg, Germany.
| |
Collapse
|
47
|
Esslinger C, Chapatte L, Finke D, Miconnet I, Guillaume P, Lévy F, MacDonald HR. In vivo administration of a lentiviral vaccine targets DCs and induces efficient CD8+ T cell responses. J Clin Invest 2003. [DOI: 10.1172/jci200317098] [Citation(s) in RCA: 116] [Impact Index Per Article: 5.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/17/2022] Open
|
48
|
Abstract
Cutaneous squamous cell carcinoma is typically characterized by the over-expression of the tumour suppressor protein p53. Considerable evidence suggests that immune competence is important in the control of cutaneous SCC. We discuss the immunobiology of p53 and its relevance to cutaneous SCC, including the potential interaction with human papillomavirus.
Collapse
Affiliation(s)
- A P B Black
- University of Oxford, MRC Human Immunology Unit, Weatherall Institute of Molecular Medicine, The John Radcliffe Hospital, UK
| | | |
Collapse
|
49
|
Curtis JL, Punturieri A. Enhancing antitumor immunity perioperatively: a matter of timing, cooperation, and specificity. Am J Respir Cell Mol Biol 2003; 28:541-5. [PMID: 12707008 PMCID: PMC2640487 DOI: 10.1165/rcmb.f266] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022] Open
Affiliation(s)
- Jeffrey L Curtis
- Pulmonary and Critical Care Medicine Section, Medical Service, Department of Veterans Affairs Health Care System, Ann Arbor, MI 48105-2303, USA.
| | | |
Collapse
|
50
|
Abstract
There is reason to believe that the unfolding revolution in molecular biology and translational research will allow selective targeting of tumor cells, and radically change the way general practitioners and pediatric oncologists treat and follow children with cancer. This article highlights some of the most promising approaches being tested in the field. By learning about the underlying biology, the remaining hurdles, the projected timeline, and the possible impact of new therapies on the practice of pediatric oncology, health care professionals and patients should be better prepared for the future of pediatric oncology.
Collapse
Affiliation(s)
- Robert J Arceci
- Department of Oncology, Sidney Kimmel Comprehensive Cancer Center at Johns Hopkins, 1650 Orleans Street, Room 2M51, Baltimore, Maryland 21231, USA.
| | | |
Collapse
|