1
|
ElTanbouly MA, Noelle RJ. Rethinking peripheral T cell tolerance: checkpoints across a T cell's journey. Nat Rev Immunol 2021; 21:257-267. [PMID: 33077935 DOI: 10.1038/s41577-020-00454-2] [Citation(s) in RCA: 134] [Impact Index Per Article: 33.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Accepted: 09/18/2020] [Indexed: 01/10/2023]
Abstract
Following their exit from the thymus, T cells are endowed with potent effector functions but must spare host tissue from harm. The fate of these cells is dictated by a series of checkpoints that regulate the quality and magnitude of T cell-mediated immunity, known as tolerance checkpoints. In this Perspective, we discuss the mediators and networks that control the six main peripheral tolerance checkpoints throughout the life of a T cell: quiescence, ignorance, anergy, exhaustion, senescence and death. At the naive T cell stage, two intrinsic checkpoints that actively maintain tolerance are quiescence and ignorance. In the presence of co-stimulation-deficient T cell activation, anergy is a dominant hallmark that mandates T cell unresponsiveness. When T cells are successfully stimulated and reach the effector stage, exhaustion and senescence can limit excessive inflammation and prevent immunopathology. At every stage of the T cell's journey, cell death exists as a checkpoint to limit clonal expansion and to terminate unrestrained responses. Here, we compare and contrast the T cell tolerance checkpoints and discuss their specific roles, with the aim of providing an integrated view of T cell peripheral tolerance and fate regulation.
Collapse
Affiliation(s)
- Mohamed A ElTanbouly
- Department of Microbiology and Immunology, Geisel School of Medicine, Norris Cotton Cancer Center, Dartmouth College, Hanover, NH, USA
| | - Randolph J Noelle
- Department of Microbiology and Immunology, Geisel School of Medicine, Norris Cotton Cancer Center, Dartmouth College, Hanover, NH, USA.
| |
Collapse
|
2
|
Knockdown of LncRNA MALAT1 contributes to cell apoptosis via regulating NF-κB/CD80 axis in neonatal respiratory distress syndrome. Int J Biochem Cell Biol 2018; 104:138-148. [PMID: 30243953 DOI: 10.1016/j.biocel.2018.09.009] [Citation(s) in RCA: 12] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/16/2018] [Revised: 09/16/2018] [Accepted: 09/18/2018] [Indexed: 01/28/2023]
Abstract
Neonatal respiratory distress syndrome (NRDS) is a leading cause of morbidity in premature newborns and is a common reason for admission to the neonatal intensive care unit (NICU). Recent studies found that the pathogenesis of NRDS is not simply lung immaturity. Apoptosis is an essential process for the development and maturation of the lungs. In this study, we report a critical role of lncRNA MALAT1 in regulating CD80 transcription in the NRDS-associated apoptosis via binding with NF-κB. We first showed MALAT1 and CD80 were highly expressed in the peripheral blood mononuclear cells of NRDS with infection exposure. Then we found MALAT1 expressions were significantly increased by the treatment of LPS. We confirmed knockdown of MALAT1 promoted cell viability by CCK-8 assays, cell apoptosis by flow cytometric assays and cytoskeleton destruction by immunocytochemistry. We confirmed CD80 expression level was associated with cell apoptosis by affecting PARP and caspase-3. Then we demonstrated knockdown of MALAT1 promoted CD80 transcription in A549 cells. Furthermore, we confirmed that MALAT1 downregulated transcriptional expression of CD80 by interfering with NF-κB activation and disrupting its binding efficiency with the CD80 promoter in the cell nucleus. In conclusion, we first identified lncRNA MALAT1 as an important prognosis maker for NRDS patients. Most significantly, this study then demonstrated a novel regulatory function of knocked-down MALAT1 on the transcriptional level of CD80 by enhancing the binding of NF-κB to CD80 promoter. Since the interaction between MALAT1 and CD80 plays an essential role in the cell apoptosis of NRDS, our findings demonstrate the possibility of using MALAT1 as therapeutic target for treatment of NRDS, and extend existing knowledge about the molecular mechanism that underlies NRDS pathogensis.
Collapse
|
3
|
Lacher MD, Bauer G, Fury B, Graeve S, Fledderman EL, Petrie TD, Coleal-Bergum DP, Hackett T, Perotti NH, Kong YY, Kwok WW, Wagner JP, Wiseman CL, Williams WV. SV-BR-1-GM, a Clinically Effective GM-CSF-Secreting Breast Cancer Cell Line, Expresses an Immune Signature and Directly Activates CD4 + T Lymphocytes. Front Immunol 2018; 9:776. [PMID: 29867922 PMCID: PMC5962696 DOI: 10.3389/fimmu.2018.00776] [Citation(s) in RCA: 16] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2017] [Accepted: 03/28/2018] [Indexed: 12/18/2022] Open
Abstract
Targeted cancer immunotherapy with irradiated, granulocyte–macrophage colony-stimulating factor (GM-CSF)-secreting, allogeneic cancer cell lines has been an effective approach to reduce tumor burden in several patients. It is generally assumed that to be effective, these cell lines need to express immunogenic antigens coexpressed in patient tumor cells, and antigen-presenting cells need to take up such antigens then present them to patient T cells. We have previously reported that, in a phase I pilot study (ClinicalTrials.gov NCT00095862), a subject with stage IV breast cancer experienced substantial regression of breast, lung, and brain lesions following inoculation with clinical formulations of SV-BR-1-GM, a GM-CSF-secreting breast tumor cell line. To identify diagnostic features permitting the prospective identification of patients likely to benefit from SV-BR-1-GM, we conducted a molecular analysis of the SV-BR-1-GM cell line and of patient-derived blood, as well as a tumor specimen. Compared to normal human breast cells, SV-BR-1-GM cells overexpress genes encoding tumor-associated antigens (TAAs) such as PRAME, a cancer/testis antigen. Curiously, despite its presumptive breast epithelial origin, the cell line expresses major histocompatibility complex (MHC) class II genes (HLA-DRA, HLA-DRB3, HLA-DMA, HLA-DMB), in addition to several other factors known to play immunostimulatory roles. These factors include MHC class I components (B2M, HLA-A, HLA-B), ADA (encoding adenosine deaminase), ADGRE5 (CD97), CD58 (LFA3), CD74 (encoding invariant chain and CLIP), CD83, CXCL8 (IL8), CXCL16, HLA-F, IL6, IL18, and KITLG. Moreover, both SV-BR-1-GM cells and the responding study subject carried an HLA-DRB3*02:02 allele, raising the question of whether SV-BR-1-GM cells can directly present endogenous antigens to T cells, thereby inducing a tumor-directed immune response. In support of this, SV-BR-1-GM cells (which also carry the HLA-DRB3*01:01 allele) treated with yellow fever virus (YFV) envelope (Env) 43–59 peptides reactivated YFV-DRB3*01:01-specific CD4+ T cells. Thus, the partial HLA allele match between SV-BR-1-GM and the clinical responder might have enabled patient T lymphocytes to directly recognize SV-BR-1-GM TAAs as presented on SV-BR-1-GM MHCs. Taken together, our findings are consistent with a potentially unique mechanism of action by which SV-BR-1-GM cells can act as APCs for previously primed CD4+ T cells.
Collapse
Affiliation(s)
| | - Gerhard Bauer
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Brian Fury
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Sanne Graeve
- BriaCell Therapeutics Corp., Berkeley, CA, United States
| | - Emily L Fledderman
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Tye D Petrie
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Dane P Coleal-Bergum
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Tia Hackett
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Nicholas H Perotti
- GMP Facility, Institute for Regenerative Cures, University of California, Davis (UCD), Sacramento, CA, United States
| | - Ying Y Kong
- Benaroya Research Institute at Virginia Mason, Seattle, WA, United States
| | - William W Kwok
- Benaroya Research Institute at Virginia Mason, Seattle, WA, United States
| | | | | | | |
Collapse
|
4
|
Evaluating the expression level of co-stimulatory molecules CD 80 and CD 86 in different types of colon polyps. Curr Res Transl Med 2017; 66:19-25. [PMID: 29277562 DOI: 10.1016/j.retram.2017.11.003] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/22/2017] [Revised: 10/14/2017] [Accepted: 11/02/2017] [Indexed: 01/08/2023]
Abstract
PURPOSE OF THE STUDY Co-stimulatory molecules CD80 and CD86 are the members of B7 family, which stimulate the T lymphocytes in response to the malignant colon polyps. However, the expression of these molecules is depressed in cancers. In the present study, the transcription levels of CD80 and CD86 genes in the colon polyps (Precancerous lesions) and its association with the clinical features were examined. PATIENTS AND METHODS Forty-nine biopsies samples from patients with the colorectal polyps and 10 healthy subjects were collected by the colonoscopy. Questionnaires including clinical and demographic data were filled for all cases. Using Real-time PCR, the mucosal mRNA expression levels of CD80 and CD86 genes were quantified. RESULTS Adenoma and hyperplastic polyps were reported in 69.3 and 30.7 percent of 49 patients, respectively. Unlike hyperplastic polyps, the expression of CD86 was increased in adenoma polyps compared to controls (RQ=2.75 vs. 0.837, respectively). The data from CD80 showed noticeable reduction about 0.31 and 0.11 in adenoma and hyperplastic polyps, respectively, in response to control group (RQ=0.729). Also, analyzing colon and rectum polyps depicted a marked increment in CD86 level, in contrast to CD80. CONCLUSION Examining the mRNA expression levels of CD80 and CD86 genes between colon polyps with the rectal polyps shows that the enhanced level of CD86 in adenoma samples could be considered as a valuable biomarker for distinguishing the adenoma from hyperplastic polyps and the masses located in the colon from the rectum.
Collapse
|
5
|
Hu J, Batth IS, Xia X, Li S. Regulation of NKG2D +CD8 + T-cell-mediated antitumor immune surveillance: Identification of a novel CD28 activation-mediated, STAT3 phosphorylation-dependent mechanism. Oncoimmunology 2016; 5:e1252012. [PMID: 28123894 DOI: 10.1080/2162402x.2016.1252012] [Citation(s) in RCA: 18] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/13/2016] [Revised: 10/10/2016] [Accepted: 10/19/2016] [Indexed: 01/06/2023] Open
Abstract
The natural killer (NK) group 2D (NKG2D) receptor, which displays on mouse and human NK cells, activates CD8+ T cells and small subsets of other T cells. NKG2D+CD8+ T cells play critical roles in both innate and adaptive immunity upon engagement with NKG2D ligands to eliminate tumor and infected cells. Despite the important role of NKG2D+CD8+ T cells in immune surveillance, the mechanisms of how NKG2D expression on CD8+ T cells is regulated remain poorly defined. We treated mouse and human CD8+ T cells with CD80 recombinant protein, plus a pharmacologic model with small molecular inhibitors to determine which signaling pathway leads to NKG2D regulation on CD8+T cells. This study revealed that CD28 activation gives rise to sustained NKG2D expression on both mouse and human CD8+ T cells in a signal transducer and activator of transcription 3 (STAT3) phosphorylation-dependent manner. Further, we found that CD28 activation stimulated sustained activation of the tyrosine kinase Lck, which recruits and triggers Janus kinase/STAT3 signaling to phosphorylate STAT3, and in turn increases NKG2D expression. Moreover, NKG2D induction on CD8+ T cells exerts cytolytic activity against target tumor cells in vitro, as well as significantly improves the antitumor therapeutic effects in vivo in an NKG2D-dependent manner. Taken together, these results elucidated a novel mechanism of NKG2D regulation by phosphorylated STAT3 (pSTAT3) on CD8+ T cells upon CD28 activation. This mechanism may shed light on the effectiveness of CD80-based, NKG2D-dependent antitumor immunotherapy.
Collapse
Affiliation(s)
- Jiemiao Hu
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Izhar S Batth
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Xueqing Xia
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center , Houston, TX, USA
| | - Shulin Li
- Division of Pediatrics, The University of Texas MD Anderson Cancer Center , Houston, TX, USA
| |
Collapse
|
6
|
Hawley TS, Linsley PS, Hawley RG. Co-expression of B7–1 with Interleukin-12 Enhances Vaccine-induced Antitumour Immunity in Experimental Myeloma. Hematology 2016; 3:365-74. [DOI: 10.1080/10245332.1998.11746410] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022] Open
Affiliation(s)
- Teresa S. Hawley
- Oncology Gene Therapy Program, The Toronto Hospital, Toronto, Ontario, Canada
| | - Peter S. Linsley
- Bristol-Meyers Squibb Pharmaceutical Research Institute, Seattle, Washington, USA
| | - Robert G. Hawley
- Oncology Gene Therapy Program, The Toronto Hospital, Toronto, Ontario, Canada
| |
Collapse
|
7
|
Patel JM, Vartabedian VF, Bozeman EN, Caoyonan BE, Srivatsan S, Pack CD, Dey P, D'Souza MJ, Yang L, Selvaraj P. Plasma membrane vesicles decorated with glycolipid-anchored antigens and adjuvants via protein transfer as an antigen delivery platform for inhibition of tumor growth. Biomaterials 2015; 74:231-44. [PMID: 26461116 DOI: 10.1016/j.biomaterials.2015.09.031] [Citation(s) in RCA: 28] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/04/2015] [Revised: 09/19/2015] [Accepted: 09/23/2015] [Indexed: 02/05/2023]
Abstract
Antigen delivered within particulate materials leads to enhanced antigen-specific immunity compared to soluble administration of antigen. However, current delivery approaches for antigen encapsulated in synthetic particulate materials are limited by the complexity of particle production that affects stability and immunogenicity of the antigen. Herein, we describe a protein delivery system that utilizes plasma membrane vesicles (PMVs) derived from biological materials such as cultured cells or isolated tissues and a simple protein transfer technology. We show that these particulate PMVs can be easily modified within 4 h by a protein transfer process to stably incorporate a glycosylphosphatidylinositol (GPI)-anchored form of the breast cancer antigen HER-2 onto the PMV surface. Immunization of mice with GPI-HER-2-modified-PMVs induced strong HER-2-specific antibody responses and protection from tumor challenge in two different breast cancer models. Further incorporation of the immunostimulatory molecules IL-12 and B7-1 onto the PMVs by protein transfer enhanced tumor protection and induced beneficial Th1 and Th2-type HER-2-specific immune responses. Since protein antigens can be easily converted to GPI-anchored forms, these results demonstrate that isolated plasma membrane vesicles can be modified with desired antigens along with immunostimulatory molecules by protein transfer and used as a vaccine delivery vehicle to elicit potent antigen-specific immunity.
Collapse
Affiliation(s)
- Jaina M Patel
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Vincent F Vartabedian
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Erica N Bozeman
- Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Brianne E Caoyonan
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Sanjay Srivatsan
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Christopher D Pack
- Metaclipse Therapeutics Corporation, 3175 Presidential Drive, Atlanta, GA 30340, USA
| | - Paulami Dey
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Martin J D'Souza
- Vaccine Nanotechnology Laboratory, College of Pharmacy, Mercer University, Atlanta, GA 30341, USA
| | - Lily Yang
- Department of Surgery, Emory University School of Medicine, Atlanta, GA 30322, USA
| | - Periasamy Selvaraj
- Department of Pathology and Laboratory Medicine, Emory University School of Medicine, Atlanta, GA 30322, USA.
| |
Collapse
|
8
|
Ya Z, Hailemichael Y, Overwijk W, Restifo NP. Mouse model for pre-clinical study of human cancer immunotherapy. CURRENT PROTOCOLS IN IMMUNOLOGY 2015; 108:20.1.1-20.1.43. [PMID: 25640991 PMCID: PMC4361407 DOI: 10.1002/0471142735.im2001s108] [Citation(s) in RCA: 17] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/11/2022]
Abstract
This unit describes protocols for developing tumors in mice, including subcutaneous growth, pulmonary metastases of B16 melanoma, and spontaneous melanoma in B-Raf V600E/PTEN deletion transgenic mouse models. Two immunization methods to prevent B16 tumor growth are described using B16.GM-CSF and recombinant vaccinia virus. A therapeutic approach is also included that uses adoptive transfer of tumor antigen-specific T cells. Methods including CTL induction, isolation, testing, and genetic modification of mouse T cells for adoptive transfer by using retrovirus-expressing genes of interest are provided. Additional sections, including growing B16 melanoma, enumerating pulmonary metastases, tumor imaging technique, and use of recombinant viruses for vaccination, are discussed together with safety concerns.
Collapse
MESH Headings
- Animals
- Antibodies/blood
- Antibodies/immunology
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/adverse effects
- Cancer Vaccines/immunology
- Cell Culture Techniques
- Cell- and Tissue-Based Therapy/adverse effects
- Cell- and Tissue-Based Therapy/methods
- Disease Models, Animal
- Enzyme-Linked Immunosorbent Assay
- Female
- Gene Transfer Techniques
- Genetic Vectors/genetics
- Immunization/methods
- Immunotherapy/adverse effects
- Immunotherapy/methods
- Male
- Melanoma, Experimental/diagnosis
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Melanoma, Experimental/therapy
- Mice
- Mice, Transgenic
- Molecular Imaging/methods
- Neoplasm Metastasis
- Neoplasms/diagnosis
- Neoplasms/etiology
- Neoplasms/immunology
- Neoplasms/therapy
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/metabolism
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/metabolism
- Transduction, Genetic
- Translational Research, Biomedical
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Zhiya Ya
- National Cancer Institute, Surgery Branch, Bethesda, Maryland
| | - Yared Hailemichael
- Department of Melanoma Medical Oncology-Research, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | - Willem Overwijk
- Department of Melanoma Medical Oncology-Research, Division of Cancer Medicine, The University of Texas MD Anderson Cancer Center, Houston, Texas
| | | |
Collapse
|
9
|
A functional variant at miR-132-3p, miR-212-3p, and miR-361-5p binding site in CD80 gene alters susceptibility to gastric cancer in a Chinese Han population. Med Oncol 2014; 31:60. [PMID: 24981235 DOI: 10.1007/s12032-014-0060-2] [Citation(s) in RCA: 24] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/06/2014] [Accepted: 05/29/2014] [Indexed: 12/22/2022]
Abstract
A number of single-nucleotide polymorphisms within the 3'-UTR of genes have been shown to relate to the occurrence of cancers. In this study, by using polymerase chain reaction-restriction fragment length analysis method, we determined an SNP rs1599795 in the 3'-UTR of CD80 gene in 183 gastric cancer patients and 348 healthy controls. Statistical analysis results showed that SNP rs1599795 genotypes were significantly correlated with the risk of gastric cancer. Compared with the AA homozygotes, the TA heterozygotes were significantly more prevalent in the patients (OR 1.44, 95 % CI 0.98-2.11) with a larger tumor size (P = 0.001), deeper infiltration (P = 1.5 × 10(-5)), higher possibility of lymph node metastasis (P = 0.003), and more in the late stage (TNM stage III and IV; P = 0.003); the TT homozygotes had larger tumor size (P = 0.001) and lower degree of differentiation (P = 2.2 × 10(-4)). Dual-luciferase reporter assays showed that miR-132-3p, miR-212-3p, and miR-361-5p inhibited the expression of CD80 through binding with the CD80 3'-UTR, and this inhibitory role of miR-132-3p, miR-212-3p, and miR-361-5p was impacted by rs1599795. Our findings have shown that the SNP rs1599795 in CD80 3'-UTR, through disrupting the regulatory role of miR-132-3p, miR-212-3p, and miR-361-5p in CD80 expression, contributed to the occurrence of gastric cancer.
Collapse
|
10
|
Low dose decitabine treatment induces CD80 expression in cancer cells and stimulates tumor specific cytotoxic T lymphocyte responses. PLoS One 2013; 8:e62924. [PMID: 23671644 PMCID: PMC3650049 DOI: 10.1371/journal.pone.0062924] [Citation(s) in RCA: 77] [Impact Index Per Article: 6.4] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/05/2012] [Accepted: 03/26/2013] [Indexed: 12/14/2022] Open
Abstract
Lack of immunogenicity of cancer cells has been considered a major reason for their failure in induction of a tumor specific T cell response. In this paper, we present evidence that decitabine (DAC), a DNA methylation inhibitor that is currently used for the treatment of myelodysplastic syndrome (MDS), acute myeloid leukemia (AML) and other malignant neoplasms, is capable of eliciting an anti-tumor cytotoxic T lymphocyte (CTL) response in mouse EL4 tumor model. C57BL/6 mice with established EL4 tumors were treated with DAC (1.0 mg/kg body weight) once daily for 5 days. We found that DAC treatment resulted in infiltration of IFN-γ producing T lymphocytes into tumors and caused tumor rejection. Depletion of CD8+, but not CD4+ T cells resumed tumor growth. DAC-induced CTL response appeared to be elicited by the induction of CD80 expression on tumor cells. Epigenetic evidence suggests that DAC induces CD80 expression in EL4 cells via demethylation of CpG dinucleotide sites in the promoter of CD80 gene. In addition, we also showed that a transient, low-dose DAC treatment can induce CD80 gene expression in a variety of human cancer cells. This study provides the first evidence that epigenetic modulation can induce the expression of a major T cell co-stimulatory molecule on cancer cells, which can overcome immune tolerance, and induce an efficient anti-tumor CTL response. The results have important implications in designing DAC-based cancer immunotherapy.
Collapse
|
11
|
Schweighoffer T. Molecular cancer vaccines: Tumor therapy using antigen-specific immunizations. Pathol Oncol Res 2012; 3:164-76. [PMID: 18470726 DOI: 10.1007/bf02899917] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/10/1997] [Accepted: 08/24/1997] [Indexed: 10/21/2022]
Abstract
Vaccination against tumors promises selective destruction of malignant cells by the host's immune system. Molecular cancer vaccines rely on recently identified tumor antigens as immunogens. Tumor antigens can be applied in many forms, as genes in recombinant vectors, as proteins or peptides representing T cell epitopes.Analysis of various aspects indicates some advantage for peptide-based vaccines over the other modalities. Further refinements and extensively monitored clinical trials are necessary to advance molecular cancer vaccines from concepts into powerful therapy.
Collapse
Affiliation(s)
- T Schweighoffer
- Department Cell Biology, Boehringer Ingelheim Research and Development, Dr. Boehringer-Gasse 5, A-l 120, Wien, Austria,
| |
Collapse
|
12
|
Wang S, Wang S, Yang T, Zhu F, Zhu J, Huang Y, Wu L, Chen L, Xu Z. CD40L-mediated inhibition of NF-kappaB in CA46 Burkitt lymphoma cells promotes apoptosis. Leuk Lymphoma 2009; 49:1792-9. [DOI: 10.1080/10428190802216723] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/21/2022]
|
13
|
Mir MA, Agrewala JN. Signaling through CD80: an approach for treating lymphomas. Expert Opin Ther Targets 2008; 12:969-79. [DOI: 10.1517/14728222.12.8.969] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
14
|
Overwijk WW, Restifo NP. B16 as a mouse model for human melanoma. ACTA ACUST UNITED AC 2008; Chapter 20:Unit 20.1. [PMID: 18432774 DOI: 10.1002/0471142735.im2001s39] [Citation(s) in RCA: 240] [Impact Index Per Article: 14.1] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
This unit details protocols for in vivo models of subcutaneous growth and pulmonary metastases of B16 melanoma. Therapeutic approaches include the use of B16.GM-CSF and rVVmTRP-1 to induce autoimmune vitiligo and tumor protection. The induction and use of gp 100-specific therapeutic cytotoxic T lymphocytes (CTL) are discussed. Methods are also included for CTL induction, isolation and testing, CTL maintenance, and adoptive transfer. Support protocols detail the testing of mouse sera for presence of MDA-specific antibodies by immunoblotting and ELISA, respectively. Additional sections, including growing B16 melanoma, enumerating pulmonary metastases, and use of recombinant viruses for vaccination, are discussed together with safety concerns.
Collapse
Affiliation(s)
- W W Overwijk
- National Cancer Institute, NIH, Bethesda, Maryland, USA
| | | |
Collapse
|
15
|
Dezfouli S, Hatzinisiriou I, Ralph SJ. Enhancing CTL responses to melanoma cell vaccines in vivo: synergistic increases obtained using IFNgamma primed and IFNbeta treated B7-1+ B16-F10 melanoma cells. Immunol Cell Biol 2007; 81:459-71. [PMID: 14636243 DOI: 10.1046/j.0818-9641.2003.01189.x] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Sequentially treating human melanoma cell lines by priming with interferon-gamma before adding interferon-beta was previously found to be the most efficient protocol for producing concurrently increased expression of the three surface antigens B7-1, intercellular adhesion molecule-1 and human histocompatibility leucocyte antigens Class I. The present study describes similar outcomes when the same sequential intercellular adhesion molecule-based protocol is applied to murine B16-F10 melanoma cells as well as preclinical studies using the B16-F10 model as a poorly immunogenic melanoma. Thus, treating B16-F10 cells or a highly expressing B7-1 transfected subline (B16-F10/B7-1 hi) by priming with interferon-gamma for 24 h before adding interferon-beta for a further 48 h (interferon-gamma 72/beta 48) increased expression of all three surface antigens, particularly major histocompatibility complex class I whose increased expression was sustained for several days. As a whole tumour cell vaccine, interferon-gamma 72/beta 48 treated B16-F10 cells produced greater levels of cytoxic T lymphocyte response compared to vaccines prepared from cells treated with a single type of interferon. Furthermore, B16-F10 cells expressing high levels of B7-1 and treated using the interferon-gamma 72/beta 48 protocol (interferon-gamma 72/beta 48-treated B16-F10/B7-1 hi) produced substantially increased cytoxic T lymphocyte responses with a fivefold greater synergy than the combined results of either interferon treated or B7-1 expressing cells tested individually. The resulting CD8+ cytoxic T lymphocyte showed greater specificity for B16-F10 cells with tenfold higher killing than for syngeneic EL-4 lymphoma cells. Killing proceeded via the perforin-mediated pathway. CTL responses were induced independent of CD4+ T helper cells. The majority of mice receiving interferon-gamma 72/beta 48-treated B16-F10/B7-1 hi vaccine in vivo remained tumour free after challenge with 5 x 105 live B16-F10 cells expressing intermediate B7-1 levels. The novel strategy described will help enhance vaccine potency when applied clinically to prepare whole cell based cancer vaccine therapies.
Collapse
Affiliation(s)
- Shala Dezfouli
- Department Biochemistry and Molecular Biology, School of Biomedical Science, Monash University, Clayton, Vic 3800, Australia
| | | | | |
Collapse
|
16
|
Ganesan PL, Alexander SI, Watson D, Logan GJ, Zhang GY, Alexander IE. Robust anti-tumor immunity and memory in Rag-1-deficient mice following adoptive transfer of cytokine-primed splenocytes and tumor CD80 expression. Cancer Immunol Immunother 2007; 56:1955-65. [PMID: 17549473 PMCID: PMC11030577 DOI: 10.1007/s00262-007-0339-7] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/21/2007] [Accepted: 04/27/2007] [Indexed: 12/22/2022]
Abstract
Successful immunotherapy of solid tumors has proven difficult to achieve. The aim of the current study was to further investigate the effects of peripheral CD80-mediated co-stimulation on the efficacy of polyclonal anti-tumor effector CTL in an adoptive transfer model. Splenocytes obtained from wild-type mice immunized with CD80-transduced EL4 tumor cells were expanded in vitro in the presence of either IL-12 or IL-15 and irradiated CD80-transduced EL4 tumor cells. Polyclonal CD8 T cells were the major subset in the effector population. Primed effector cells were adoptively transferred into immuno-deficient Rag-1-deficient mice which were then challenged with syngeneic vector-control or CD80-transduced EL4 tumor cells. Expression of CD80 enhanced the elimination of EL4 tumors and mouse survival. Both IL-12 and IL-15 cultured cells had enhanced cytotoxicity. Importantly, anti-tumor memory was maintained without tumor evasion following re-challenge with either CD80-transduced and vector-control EL4 cells. We also show, using antibody-mediated depletion, that endogenous NK cells present in Rag-1-deficient mice exert anti-EL4 tumor activity that is enhanced by CD80 expression. Collectively these data show that peripheral co-stimulation by tumor expression of CD80 results in enhanced anti-tumor efficacy of NK and polyclonal effector T cells, and suggest that TCR repertoire diversity helps protect against tumor escape and provides memory with resultant robust immunity to subsequent tumor challenge irrespective of CD80 status.
Collapse
Affiliation(s)
- Priyadarshini L. Ganesan
- Gene Therapy Research Unit, The Children’s Hospital at Westmead and Children’s Medical Research Institute, Westmead, NSW Australia
| | - Stephen I. Alexander
- The Centre for Kidney Research, The Children’s Hospital at Westmead, Westmead, NSW Australia
| | - Debbie Watson
- The Centre for Kidney Research, The Children’s Hospital at Westmead, Westmead, NSW Australia
| | - Grant J. Logan
- Gene Therapy Research Unit, The Children’s Hospital at Westmead and Children’s Medical Research Institute, Westmead, NSW Australia
| | - Geoff Y. Zhang
- The Centre for Kidney Research, The Children’s Hospital at Westmead, Westmead, NSW Australia
| | - Ian E. Alexander
- Gene Therapy Research Unit, The Children’s Hospital at Westmead and Children’s Medical Research Institute, Westmead, NSW Australia
- The University of Sydney Discipline of Paediatrics and Child Health, Locked Bag 4001, Westmead, NSW 2145 Australia
- Gene Therapy Research Unit, The Children’s Hospital at Westmead, Locked Bag 4001, Westmead, NSW 2145 Australia
| |
Collapse
|
17
|
Arina A, Murillo O, Hervás-Stubbs S, Azpilikueta A, Dubrot J, Tirapu I, Huarte E, Alfaro C, Pérez-Gracia JL, González-Aseguinolaza G, Sarobe P, Lasarte JJ, Jamieson A, Prieto J, Raulet DH, Melero I. The combined actions of NK and T lymphocytes are necessary to reject an EGFP+ mesenchymal tumor through mechanisms dependent on NKG2D and IFN gamma. Int J Cancer 2007; 121:1282-95. [PMID: 17520674 DOI: 10.1002/ijc.22795] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
Better understanding of the mechanisms that mediate spontaneous immune rejections ought to be important in the quest for improvements in immunotherapy of cancer. A set of intraperitoneal tumors of mesenchymal origin that had been chemically induced in ubiquitously expressing EGFP transgenic mice provided a model in which both T and NK cells were absolutely required for tumor rejection. Tumor cells were traceable because of being fluorescent and readily grafted in RAG1(-/-) immunodeficient mice, whereas they were rejected in a majority of syngeneic C57BL/6 and EGFP-transgenic mice. Tumor-cell clones with the highest EGFP expression tended to be rejected, but a direct involvement of EGFP as the antigen recognized for the immune rejections was ruled out. Rejections were absolutely dependent on NK cells as well as on CD4(+) and CD8(+) T lymphocytes according to selective depletion studies. Furthermore, CD8(+) and CD4(+) T lymphocytes as well as NK cells were detected in the inflammatory infiltrate that mediates tumor rejection along with some DC. The effects of IFN gamma, produced at the tumor site by T and NK lymphocytes, were only required at the malignant cell level and were necessary for tumor eradication. NK recognition of tumor cells was mediated by the NKG2D-activating receptor and blocking its function in vivo partially interfered with rejection. Therefore, complete rejection of these mesenchymal tumors requires a concerted set of activities including direct tumor-cell destruction and IFN gamma production that are mediated by both NK and T cells.
Collapse
Affiliation(s)
- Ainhoa Arina
- Gene Therapy Unit, Centro de Investigación Médica Aplicada, University of Navarra, Pamplona, Spain
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
18
|
Yie SM, Yang H, Ye SR, Li K, Dong DD, Lin XM. Expression of human leucocyte antigen G (HLA-G) is associated with prognosis in non-small cell lung cancer. Lung Cancer 2007; 58:267-74. [PMID: 17673327 DOI: 10.1016/j.lungcan.2007.06.011] [Citation(s) in RCA: 91] [Impact Index Per Article: 5.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/05/2007] [Revised: 05/17/2007] [Accepted: 06/19/2007] [Indexed: 11/18/2022]
Abstract
In a previous study, we demonstrated that human leucocyte antigen G (HLA-G) was aberrantly expressed in a majority of primary colorectal carcinomas, and that the detection of HLA-G expression had a strong and independent prognostic value in human colorectal cancer. In the current study, we look into whether the aberrant expression of HLA-G is also related to non-small cell lung cancer (NSCLC). The expression of HLA-G was investigated immunohistochemically in 106 patients with NSCLC. The correlation between HLA-G status and various clinicopathological parameters was analysed. As well, the level of HLA-G expression was also compared to the survival rate of patients with NSCLC. In total, we found that in 75% (79/106) of the primary site of NSCLC, an aberrant HLA-G expression was detected. However, this expression was not observed in the normal lung tissues. HLA-G expression in NSCLC was significantly correlated with lymph nodal metastasis, clinical stages of the disease, and host immune response (P = 0.0001, 0.0001, and 0.027, respectively). Patients with HLA-G positive tumours had a significantly shorter survival time than those with tumours that were HLA-G negative (P = 0.001). In addition, through multivariate analysis, HLA-G exhibited an independent prognostic factor (P = 0.01, relative risk 4.09; 95% confidence interval 1.40-11.9). All in all, our results indicate that the expression of HLA-G is a characteristic feature of NSCLC, and they suggest that immunostaining by anti-HLA-G antibodies may be a potentially useful prognostic indicator.
Collapse
Affiliation(s)
- Shang-mian Yie
- Chengdu Bioengineering Institute for Cancer Research, Chengdu, Sichuan, PR China.
| | | | | | | | | | | |
Collapse
|
19
|
Altin JG, Parish CR. Liposomal vaccines--targeting the delivery of antigen. Methods 2007; 40:39-52. [PMID: 16997712 DOI: 10.1016/j.ymeth.2006.05.027] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/01/2006] [Accepted: 05/05/2006] [Indexed: 01/05/2023] Open
Abstract
Vaccines that can prime the adaptive immune system for a quick and effective response against a pathogen or tumor cells, require the generation of antigen (Ag)-specific memory T and B cells. The unique ability of dendritic cells (DCs) to activate naïve T cells, implies a key role for DCs in this process. The generation of tumor-specific CD8(+) cytotoxic T cells (CTLs) is dependent on both T cell stimulation with Ag (peptide-MHC-complexes) and costimulation. Interestingly, tumor cells that lack expression of T cell costimulatory molecules become highly immunogenic when transfected to express such molecules on their surface. Adoptive immunotherapy with Ag-pulsed DCs also is a strategy showing promise as a treatment for cancer. The use of such cell-based vaccines, however, is cumbersome and expensive to use clinically, and/or may carry risks due to genetic manipulations. Liposomes are particulate vesicular lipid structures that can incorporate Ag, immunomodulatory factors and targeting molecules, and hence can serve as potent vaccines. Similarly, Ag-containing plasma membrane vesicles (PMV) derived from tumor cells can be modified to incorporate a T cell costimulatory molecule to provide both TCR stimulation, and costimulation. PMVs also can be modified to contain IFN-gamma and molecules for targeting DCs, permitting delivery of both Ag and a DC maturation signal for initiating an effective immune response. Our results show that use of such agents as vaccines can induce potent anti-tumor immune responses and immunotherapeutic effects in tumor models, and provide a strategy for the development of effective vaccines and immunotherapies for cancer and infectious diseases.
Collapse
Affiliation(s)
- Joseph G Altin
- School of Biochemistry and Molecular Biology, Faculty of Science, The Australian National University, Canberra, ACT 0200, Australia.
| | | |
Collapse
|
20
|
Lee YS, Kim JH, Choi KJ, Choi IK, Kim H, Cho S, Cho BC, Yun CO. Enhanced antitumor effect of oncolytic adenovirus expressing interleukin-12 and B7-1 in an immunocompetent murine model. Clin Cancer Res 2006; 12:5859-68. [PMID: 17020994 DOI: 10.1158/1078-0432.ccr-06-0935] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE We investigated whether an armed viral platform, where lytic property of a viral infection is coupled to viral-mediated delivery of therapeutic genes, could increase the therapeutic potential of a viral-based therapy. EXPERIMENTAL DESIGN We generated interleukin (IL)-12-expressing oncolytic adenovirus (YKL-IL-12) and IL-12- and B7-1-expressing (YKL-IL12/B7) oncolytic adenovirus. Therapeutic efficacy of these newly engineered adenoviruses was then evaluated in vivo using an immunocompetent mouse bearing murine melanoma B16-F10 tumors. Overall survival was assessed with the Kaplan-Meier method. The induction of immune cell cytotoxicity was assessed by CTL assay, IFN-gamma enzyme-linked immunospot assay, and immunohistochemical studies. RESULTS YKL-IL12/B7 oncolytic adenovirus, expressing both IL-12 and B7-1, showed a higher incidence of complete tumor regression compared with the analogous oncolytic adenovirus, YKL-1, or IL-12-expressing, YKL-IL12. Significant survival advantage was also seen in response to YKL-IL12/B7. Moreover, IL-12 and IFN-gamma levels produced in tumors treated with YKL-IL12/B7 was significantly greater than those treated with YKL-IL12. The enhanced survival advantage was mediated by the induction of immune cell cytotoxicity. In agreement with these results, massive infiltration of CD4(+) and CD8(+) T cells into tissues surrounding the necrotic area of the tumor was observed following in situ delivery of YKL-IL12/B7. CONCLUSION Combination of oncolysis and the enhancement of antitumor immune response by oncolytic adenovirus expressing both IL-12 and B7-1 elicits potent antitumor effect and survival advantage.
Collapse
Affiliation(s)
- Young-Sook Lee
- Brain Korea 21 Project for Medical Science, Institute for Cancer Research, Yonsei Cancer Center, Yonsei University College of Medicine, 134 Shinchon-Dong, Seodaemun-Gu, Seoul 120-752, Korea
| | | | | | | | | | | | | | | |
Collapse
|
21
|
Chiang EY, Stroynowski I. The role of structurally conserved class I MHC in tumor rejection: contribution of the Q8 locus. THE JOURNAL OF IMMUNOLOGY 2006; 177:2123-30. [PMID: 16887971 DOI: 10.4049/jimmunol.177.4.2123] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
The mouse multimember family of Qa-2 oligomorphic class I MHC genes is continuously undergoing duplications and deletions that alter the number of the two "prototype" Qa-2 sequences, Q8 and Q9. The frequent recombination events within the Q region lead to strain-specific modulation of the cumulative Qa-2 expression levels. Q9 protects C57BL/6 hosts from multiple disparate tumors and functions as a major CTL restriction element for shared tumor-associated Ags. We have now analyzed functional and structural properties of Q8, a class I MHC that differs significantly from Q9 in the peptide-binding, CTL-interacting alpha(1) and alpha(2) regions. Unexpectedly, we find that the extracellular domains of Q8 and Q9 act similarly during primary and secondary rejection of tumors, are recognized by cross-reactive antitumor CTL, have overlapping peptide-binding motifs, and are both assembled via the transporter associated with the Ag processing pathway. These findings suggest that shared Ag-presenting functions of the "odd" and "even" Qa-2 loci may contribute to the selective pressures shaping the haplotype-dependent quantitative variation of Qa-2 protein expression.
Collapse
Affiliation(s)
- Eugene Y Chiang
- Center for Immunology, Department of Microbiology, University of Texas Southwestern Medical Center, 5323 Harry Hines Boulevard, Dallas, TX 75390, USA
| | | |
Collapse
|
22
|
Grant JF, Iwasawa T, Sinn HW, Siemens DR, Griffith TS, Takacs EB, Ratliff TL. Induction of protective immunity to RM-1 prostate cancer cells with ALVAC-IL-2/IL-12/TNF-α combination therapy. Int J Cancer 2006; 119:2632-41. [PMID: 16991124 DOI: 10.1002/ijc.22220] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Human prostate cancers characteristically express low levels of major histocompatibility complex (MHC) Class I, which makes it challenging to induce protective antitumor responses involving T cells. Here we demonstrate that a whole cell tumor vaccine can induce protective T cell immunity to a low MHC Class I-expressing mouse prostate cancer cell line, RM-1. ALVAC recombinant canarypox viruses encoding interleukin-2, interleukin-12 and tumor necrosis factor-alpha were used to create therapeutic vaccines in 2 different ways. The RM-1 cells were pre-infected in vitro with the viruses prior to injection (pre-infection vaccine) or the RM-1 cells were injected alone, followed by the viruses (separate injection vaccine). The vaccines were each tested subcutaneously or intradermally. The pre-infection vaccine resulted in 100% clearance of primary tumors, whereas intradermal delivery of the separate injection vaccine cleared 40-60% of primary tumors. Despite the highly efficient primary tumor clearance by the pre-infection vaccine, only the separate injection vaccine generated protection upon rechallenge. Tumor-free survival induced by the separate injection vaccine required natural killer (NK) cells, CD4(+), and CD8(+) T cells. None of these cells alone were sufficient to induce tumor-free survival to the primary challenge, demonstrating an important cooperativity between NK cells and T cells. Secondary clearance of tumors also required NK and CD8(+) T cells, but not CD4(+) T cells. We report for the first time the generation of T cell immunity to the RM-1 prostate cancer cell line, demonstrating that it is possible to generate protective T cell immunity to a MHC I-low expressing tumor.
Collapse
Affiliation(s)
- Julieann F Grant
- Medical Scientist Training Program, University of Iowa, Iowa City, IA 52242-1089, USA
| | | | | | | | | | | | | |
Collapse
|
23
|
Schlom J, Tsang KY, Kantor JA, Abrams SI, Zaremba S, Greiner J, Hodge JW. Cancer vaccine development. Expert Opin Investig Drugs 2005; 7:1439-52. [PMID: 15992042 DOI: 10.1517/13543784.7.9.1439] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
A new era involving the evaluation of recombinant cancer vaccines has begun with the concurrent emergence of insights and technologies in the fields of molecular biology and immunology. These advances include: The identification and cloning of an array of genes associated with the neoplastic process, such as oncogenes, suppressor genes, genes encoding oncofoetal antigens and tissue-lineage determinants. The development of a variety of viral and bacterial vectors to deliver and present gene products. The identification of numerous T-cell costimulatory molecules and an understanding of their mode of action. The cloning and analysis of the modes of action of an array of cytokines and other immunomodulatory molecules. More sophisticated knowledge of the mode(s) of antigen presentation and T-cell activation. One current challenge in cancer therapy is the delineation of strategies toward the rational design and implementation of recombinant vaccines that will be of therapeutic benefit to cancer patients and/or members of groups at high risk for specific neoplasias. Numerous concepts are emerging in this regard. The study of immunologic intervention using laboratory animal models demonstrates that no one approach will prevail for all cancer types or, perhaps, for the various stages of the neoplastic process of a given tumour type. The immunological role(s) of CD8+, CD4+, natural killer and other cell types, as well as the roles of antibodies, must all be taken into consideration. This article reviews some of the strategies currently undergoing evaluation toward the development of recombinant vaccines for several carcinoma types.
Collapse
Affiliation(s)
- J Schlom
- Laboratory of Tumor Immunology and Biology, National Cancer Institute, National Institutes of Health, 10 Center Drive, Building 10, Room 8B07, Bethesda, MD 20892-1750, USA
| | | | | | | | | | | | | |
Collapse
|
24
|
Perry MJ, Todryk SM, Dalgleish AG. The role of herpes simplex virus thymidine kinase in the treatment of solid tumours. Expert Opin Investig Drugs 2005; 8:777-85. [PMID: 15992130 DOI: 10.1517/13543784.8.6.777] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
Abstract
Suicide gene therapy is the approach whereby the genetic alteration of a cell renders it susceptible to an otherwise non-toxic prodrug. Suicide gene therapy for solid tumours has progressed rapidly since the concept was originally described: nearly all tumour types have been explored, with some, such as glioma, melanoma and colon cancer frequently used experimentally. The exciting aspect of suicide gene therapy is the bystander effect, the phenomenon whereby there is extended tumour death when only a small fraction is transfected with the suicide gene. This phenomenon implies that there is a reduced need to target specifically all tumour cells, as the effect mechanism itself carries out this function. The bystander effect mode of action has not yet been fully characterised, but the role of gap junctions and the immune system are implicated as the main instruments in its potentiation. This approach is also amenable to pharmacological intervention, which may help to optimise parameters prior to commencing suicide gene therapy. Clinical trails have already commenced using this form of treatment and results are eagerly awaited.
Collapse
Affiliation(s)
- M J Perry
- Department of Urology, St George's Hospital Medical School, Cranmer Terrace, London SW17 0QT, UK.
| | | | | |
Collapse
|
25
|
Konishi J, Yamazaki K, Azuma M, Kinoshita I, Dosaka-Akita H, Nishimura M. B7-H1 expression on non-small cell lung cancer cells and its relationship with tumor-infiltrating lymphocytes and their PD-1 expression. Clin Cancer Res 2005; 10:5094-100. [PMID: 15297412 DOI: 10.1158/1078-0432.ccr-04-0428] [Citation(s) in RCA: 546] [Impact Index Per Article: 27.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
PURPOSE B7-H1/PD-L1 (B7-H1) and B7-DC/PD-L2 (B7-DC) are ligands for the receptor PD-1, which is known to negatively regulate T-cell activation. In the present study, we investigated the expression of B7-H1 and B7-DC in tumor specimens of non-small cell lung cancer and their relationships with clinicopathological variables and postoperative survival. Furthermore, we examined the correlation between B7-H1 expression on tumor cells and the number of tumor-infiltrating lymphocytes (TILs) or PD-1 expression on TILs. EXPERIMENTAL DESIGN The expression of B7-H1 and B7-DC in 52 surgically resected specimens of non-small cell lung cancer was evaluated immunohistochemically. RESULTS Expression of B7-H1 and B7-DC was focally observed in all non-small cell lung cancer tumor specimens. No relationship was found between the expression of B7-H1 or B7-DC and clinicopathological variables or postoperative survival. However, in the same sections evaluated, significantly fewer TILs were identified in B7-H1-positive tumor regions than in B7-H1-negative tumor regions in a subset of five patients (P = 0.01). Moreover, the percentage of TILs expressing PD-1 was significantly lower in B7-H1-positive tumor regions than in B7-H1-negative tumor regions (P = 0.02). CONCLUSIONS The expression of B7-H1 on tumor cells in local areas reciprocally correlated with the number of TILs, and this may contribute to negative regulation in antitumor immune responses in non-small cell lung cancer.
Collapse
Affiliation(s)
- Jun Konishi
- First Department of Medicine, Hokkaido University School of Medicine, Sapporo, Japan
| | | | | | | | | | | |
Collapse
|
26
|
Tseng SH, Chen Y, Chang CJ, Tai KF, Lin SM, Hwang LH. Induction of T-Cell Apoptosis in Rats by Genetically Engineered Glioma Cells Expressing Granulocyte-Macrophage Colony-Stimulating Factor and B7.1. Clin Cancer Res 2005; 11:1639-49. [PMID: 15746069 DOI: 10.1158/1078-0432.ccr-04-1366] [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] [Indexed: 11/16/2022]
Abstract
PURPOSE To evaluate antitumor effects on intracerebral gliomas of genetically engineered tumor vaccines expressing granulocyte-macrophage colony-timulating factor (GM-CSF), B7.1, or both (combination). EXPERIMENTAL DESIGN A rat glioma cell line, RT-2, was engineered with a retroviral vector to express GM-CSF, B7.1, or combination. Tumorigenicity of engineered cells and therapeutic effects of s.c. given irradiated or live tumor vaccines on parental intracerebral gliomas were studied. Immune cell infiltration induced at vaccine and tumor sites was examined by histologic and immunohistochemical staining. Apoptosis of T cells from vaccine sites was analyzed with fluorescence-activated cell sorting. RESULTS Engineered RT-2 cells exhibited reduced s.c. tumorigenicity in rats with reduced tumor growth and prolonged animal survival time compared with control rats. Rats with intracerebral gliomas s.c. treated with irradiated or live GM-CSF-expressing vaccines had 60% and 100% survival rates, respectively, significantly better than the control groups (P < 0.05). In contrast, rats treated with vaccines expressing B7.1 or the combination had no or mild therapeutic effects. Studies revealed less T-cell infiltration at both vaccine and tumor sites in rats treated with vaccines expressing B7.1 or the combination than in rats treated with a vaccine expressing GM-CSF. Cell sorting analyses revealed higher proportions of apoptotic T cells at vaccine sites of rats treated with the combination than those treated with vaccine expressing GM-CSF. CONCLUSIONS Combination of GM-CSF- and B7.1-expressing tumor vaccines exerted no synergistic, or even worse, therapeutic effects on gliomas compared with single GM-CSF-secreting tumor vaccine. The worse therapeutic effects of the GM-B7.1-expressing tumor vaccine than the GM-CSF-expressing tumor vaccine were related to the reduced T-cell amount and increased T-cell apoptosis in the former.
Collapse
Affiliation(s)
- Sheng-Hong Tseng
- Department of Surgery, National Taiwan University Hospital, College of Medicine, 7 Chung-Shan South Road, Taipei 100, Taiwan
| | | | | | | | | | | |
Collapse
|
27
|
Zheng P, Liu Y. Co-stimulatory molecules B7-1 and B7-2 as experimental therapeutic targets. ACTA ACUST UNITED AC 2005. [DOI: 10.1517/14728222.3.1.93] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022]
|
28
|
Sakate Y, Yashiro M, Tanaka H, Sunami T, Kosaka K, Hirakawa K. Usefulness of inhibiting the lymph node metastasis in human gastric carcinoma by B7-1 gene transfection. J Surg Res 2004; 122:89-95. [PMID: 15522320 DOI: 10.1016/j.jss.2004.06.011] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/16/2004] [Indexed: 11/18/2022]
Abstract
INTRODUCTION Lymph node metastasis is one of the crucial prognostic factors in gastric cancer. We have reported that ICAM-1 gene transfection was effective against lymph node metastases of gastric cancer. B7-1, one of the co-stimulatory factors, was reported to induce cytotoxic T lymphocytes when using melanoma and bladder cancer cell lines, as well as ICAM-1. In this study, we investigated the inhibitory effect of B7-1 on lymph node metastasis by B7-1 gene transfection into gastric cancer cells. MATERIALS AND METHODS We transfected B7-1 genes into a gastric cancer cell line (OCUM-2MLN) and analyzed the effect of B7-1 transduction on lymph node metastasis, the in vitro adhesiveness and cytotoxicity assay of mononuclear lymphocytes to cancer cells and lymph node metastatic ability after orthotopic implantation of gastric cancer cells in vivo. RESULTS We revealed that mononuclear lymphocytes showed significantly stronger adherence and cytotoxicity to B7-1 transfected cells (2MLN/B7) than its parent OCUM-2MLN cells. The tumor growth rate of 2MLN/B7 xenograft was significantly slower than OCUM-2MLN xenograft in nude mice. In orthotopic implantation experiments for nude mice, 2MLN/B7 cells in stomach developed significantly less lymph node metastasis than OCUM-2MLN cells. Histologic findings showed that leukocytes were intensively infiltrated in both the 2MLN/B7 tumors and its metastatic lesions, however, were scarcely observed in the lesions associated with 2MLN cells. CONCLUSION B7-1 may play an important role in inhibiting lymph node metastasis by the mechanism of enhanced immunogenicity, and that B7-1 gene transduction might be effective against lymph node metastases of gastric cancer.
Collapse
Affiliation(s)
- Yoji Sakate
- Department of Surgical Oncology, Osaka City University Graduate School of Medicine, 1-4-3 Asahi-machi, Abeno-ku, Osaka 545-8585, Japan
| | | | | | | | | | | |
Collapse
|
29
|
Pizzoferrato E. B7-2 expression above a threshold elicits anti-tumor immunity as effective as interleukin-12 and prolongs survival in murine B-cell lymphoma. Int J Cancer 2004; 110:61-9. [PMID: 15054869 DOI: 10.1002/ijc.20085] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
The costimulatory molecule, B7-2, is expressed by various lymphomas, but this level of expression is not sufficient to generate effective anti-tumor immunity in vivo. To determine whether up-regulated expression of the costimulatory molecule, B7-2, leads to more effective anti-tumor immunity in vivo, the A20 murine model of B-cell lymphoma was used. A20 tumor cells express major histocompatibility complex (MHC) I and II molecules and moderate constitutive levels of B7-2. While B7-1 and B7-2 have been introduced into tumor cells lacking these molecules, studies have not been conducted to determine whether tumors that constitutively express B7-1 or B7-2 can be made more immunogenic by increasing the expression of these molecules. In this report, A20/B7-2 transfectants expressing greater levels of B7-2 were rejected in syngeneic mice, and systemic immunity against the A20 parental cells was generated. Treatment with the A20/B7-2 variant cells significantly improved the survival of tumor-bearing mice. Coinjection with IL-12 secreting variants did not further augment the anti-tumor immunity observed for B7-2 therapy alone. Both CD8(+) T cells and natural killer (NK) cells mediated the anti-tumor immune response observed in A20/B7-2 immunized mice. In mice that developed tumors after immunization with the A20/B7-2 variant cells, resected tumor cells were shown to express lower levels of B7-2 than the transfected variants. These results suggest that the level of costimulation is important for the generation of anti-tumor immunity and for host survival. In addition, tumors appear to be able to evade the immune response by downregulating the expression of B7-2 below a threshold level.
Collapse
Affiliation(s)
- Eva Pizzoferrato
- Department of Surgery, Hillman Cancer Center, University of Pittsburgh, 5117 Centre Avenue, Pittsburgh, PA 15213, USA.
| |
Collapse
|
30
|
Tykocinski ML, Chen A, Huang JH, Weber MC, Zheng G. New designs for cancer vaccine and artificial veto cells: an emerging palette of protein paints. Immunol Res 2004; 27:565-74. [PMID: 12857999 DOI: 10.1385/ir:27:2-3:565] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Antigen-presenting cells (APC) can be refaced with "protein paints" that change the appearance of their T cell-oriented trans signal arrays. Our group has developed three categories of protein paints suitable for this kind of APC engineering: artificial glycosylphosphatidylinositol (GPI) proteins, palmitated-protein A:Fc*1 fusion protein conjugates, and trans signal converter proteins. Protein paints have been devised with either immune enhancement or suppression in mind. Costimulator * GPI and palmitated-protein A costimulator * Fcgamma1 conjugates can be used to augment the immune-activating potential of tumor cells. Alternatively, protein paints can be designed to transform APC into artificial veto cells, in essence creating Trojan horses capable of inhibiting pathogenic T cells. Trans signal converter proteins (TSCP) have been devised for this purpose. Our first paradigmatic inhibitory TSCP, CTLA-4 * Fas ligand, binds to APC, and in so doing, simultaneously blocks B7 costimulation (via CTLA-4) and sends inhibitory trans signals (via Fas ligand) to T cells with dramatic efficacy. Protein transfer offers a number of advantages over gene transfer in facilitating quantitative and combinatorial protein expression and simplifying in vivo applications; the palette of protein paints with immunotherapeutic potential will undoubtedly continue to evolve.
Collapse
Affiliation(s)
- Mark L Tykocinski
- Department of Pathology and Laboratory Medicine, University of Pennsylvania, Philadelphia, PA 19104, USA.
| | | | | | | | | |
Collapse
|
31
|
Matte CC, Cormier J, Anderson BE, Athanasiadis I, Liu J, Emerson SG, Pear W, Shlomchik WD. Graft-versus-leukemia in a retrovirally induced murine CML model: mechanisms of T-cell killing. Blood 2004; 103:4353-61. [PMID: 14982874 DOI: 10.1182/blood-2003-10-3735] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The graft-versus-leukemia (GVL) effect, mediated by donor T cells, has revolutionized the treatment of leukemia. However, effective GVL remains difficult to separate from graft-versus-host disease (GVHD), and many neoplasms are GVL resistant. Murine studies aimed at solving these problems have been limited by the use of leukemia cell lines with limited homology to human leukemias and by the absence of loss-of-function leukemia variants. To address these concerns, we developed a GVL model against murine chronic-phase chronic myelogenous leukemia (mCP-CML) induced with retrovirus expressing the bcr-abl fusion cDNA, the defining genetic abnormality of chronic-phase CML (CP-CML). By generating mCP-CML in gene-deficient mice, we have studied GVL T-cell effector mechanisms. mCP-CML expression of Fas or tumor necrosis factor (TNF) receptors is not required for CD8-mediated GVL. Strikingly, maximal CD4-mediated GVL requires cognate interactions between CD4 cells and mCP-CML cells as major histocompatibility complex-negative (MHC II(-/-)) mCP-CML is relatively GVL resistant. Nevertheless, a minority of CD4 recipients cleared MHC II(-/-) mCP-CML; thus, CD4 cells can also kill indirectly. CD4 GVL did not require target Fas expression. These results suggest that CPCML's GVL sensitivity may in part be explained by the minimal requirements for T-cell killing, and GVL-resistance may be related to MHC II expression.
Collapse
MESH Headings
- Animals
- Antigens, CD/immunology
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/transplantation
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/transplantation
- Disease Models, Animal
- Epitopes/immunology
- Female
- Fusion Proteins, bcr-abl/genetics
- Graft vs Leukemia Effect/immunology
- Leukemia, Myelogenous, Chronic, BCR-ABL Positive/immunology
- Male
- Mice
- Mice, Inbred AKR
- Mice, Inbred BALB C
- Mice, Inbred C3H
- Mice, Mutant Strains
- Receptor, Nerve Growth Factor/immunology
- Receptors, Tumor Necrosis Factor/immunology
- Receptors, Tumor Necrosis Factor, Type I
- Receptors, Tumor Necrosis Factor, Type II
- Retroviridae/genetics
- fas Receptor/immunology
Collapse
Affiliation(s)
- Catherine C Matte
- Yale University School of Medicine, Section of Medical Oncology, PO Box 208032, New Haven, CT 06520, USA
| | | | | | | | | | | | | | | |
Collapse
|
32
|
Abstract
Natural killer (NK) cells are lymphocytes that were first identified for their ability to kill tumor cells without deliberate immunization or activation. Subsequently, they were also found to be able to kill cells that are infected with certain viruses and to attack preferentially cells that lack expression of major histocompatibility complex (MHC) class I antigens. The recent discovery of novel NK receptors and their ligands has uncovered the molecular mechanisms that regulate NK activation and function. Several activating NK cell receptors and costimulatory molecules have been identified that permit these cells to recognize tumors and virus-infected cells. These are modulated by inhibitory receptors that sense the levels of MHC class I on prospective target cells to prevent unwanted destruction of healthy tissues. In vitro and in vivo, their cytotoxic ability can be enhanced by cytokines, such as interleukin (IL)-2, IL-12, IL-15 and interferon alpha/beta (IFN-alpha/beta). In animal studies, they have been shown to play a critical role in the control of tumor growth and metastasis and to provide innate immunity against infection with certain viruses. Following activation, NK cells release cytokines and chemokines that induce inflammatory responses; modulate monocyte, dendritic cells, and granulocyte growth and differentiation; and influence subsequent adaptive immune responses. The underlining mechanism of discriminating tumor cells and normal cells by NK cells has provided new insights into tumor immunosurveillance and has suggested new strategies for the treatment of human cancer.
Collapse
Affiliation(s)
- Jun Wu
- Shanghai Gnomics, Inc., and Chinese National Genome Center, Shanghai, China
| | | |
Collapse
|
33
|
Sartoris S, Testi MG, Stefani E, Chignola R, Guerriero C, Matucci A, Cestari T, Scarpa A, Riviera AP, Zanoni G, Tridente G, Andrighetto G. Induction of an antitumour adaptive immune response elicited by tumour cells expressing de novo B7-1 mainly depends on the anatomical site of their delivery: the dose applied regulates the expansion of the response. Immunology 2004; 110:474-81. [PMID: 14632645 PMCID: PMC1783082 DOI: 10.1111/j.1365-2567.2003.01760.x] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
De novo expression of costimulatory molecules in tumours generally increases their immunogenicity, but does not always induce a protective response against the parental tumour. This issue was addressed in the mouse Sp6 hybridoma model, comparing different immunization routes (subcutaneous, intraperitoneal and intravenous) and doses (0.5 x 10(6) and 5 x 10(6) cells) of Sp6 cells expressing de novo B7-1 (Sp6/B7). The results can be summarized as follows. First, de novo expression of B7-1 rendered Sp6 immunogenic, as it significantly reduced the tumour incidence to < or =15% with all delivery routes and doses tested, whereas wild-type Sp6 was invariably tumorigenic (100% tumour incidence). Second, long-lasting protection against wild-type Sp6 was mainly achieved when immunization with Sp6/B7 was subcutaneous: a dose of 0.5 x 10(6) Sp6/B7 cells elicited protection that was confined to sites in the same anatomical quarter as the immunizing injection. Repeated injections of the same dose extended protection against wild-type Sp6 to other anatomical districts, as well as a single injection of a 10-fold higher dose (5 x 10(6) cells). Finally, Sp6-specific cytotoxic T-lymphocyte activity was detected in draining lymph nodes, and the splenic expansion of Sp6-specific cytotoxic T-lymphocyte precursors quantitatively correlated with the dose of antigen. We conclude that activation of a protective immune response against Sp6 depends on the local environment where the immunogenic form of the 'whole tumour cell antigen' is delivered. The antigen dose regulates the anatomical extent of the protective response.
Collapse
Affiliation(s)
- Silvia Sartoris
- Dipartimento di Patologia, Sezione di Immunologia, Università di Verona, Verona, Italy.
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
34
|
Morioka J, Kajiwara K, Yoshikawa K, Ideguchi M, Uchida T, Suzuki M. Vaccine Therapy for Murine Glioma Using Tumor Cells Genetically Modified to Express B7.1. Neurosurgery 2004; 54:182-9; discussion 189-90. [PMID: 14683556 DOI: 10.1227/01.neu.0000097517.22018.3c] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/13/2003] [Accepted: 09/03/2003] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE In a syngeneic mouse brain tumor model, we tested the hypothesis that vaccination with tumor cells genetically modified to express B7.1 molecules induces tumor-specific T cells and immunological antitumor effects. METHODS Malignant glioma cells (RSV-MG) derived from a C3H/He mouse induced by Schmidt-Ruppin Rous sarcoma virus (RSV) were infected with an adenovirus encoding the B7.1 gene (AdB7). To investigate the effects of B7.1 expression on the tumorigenicity of RSV-MG cells, infected cells were implanted subcutaneously into C3H/He mice. The C3H/He mice were vaccinated with AdB7 transfectants injected subcutaneously and 2 weeks later were challenged intracerebrally with wild-type RSV-MG cells to determine whether or not the expression of B7.1 would enhance the immunogenicity of RSV-MG cells. RESULTS Immunocytochemistry confirmed the expression of B7.1 and major histocompatibility complex Class I antigen on the infected cells. The growth of subcutaneous tumors was markedly retarded in the AdB7 group, whereas tumors had formed and progressively increased in size in the other control groups. In the vaccine experiments, the mice immunized with AdB7 transfectants survived longer than did the mice of the other groups, and a significant difference in survival times was noted. Immunocytochemistry revealed that brain tumors in mice previously vaccinated with AdB7 infectants had been infiltrated by a larger number of CD3(+) lymphocytes and that these CD3(+) lymphocytes contained not only CD4(+) and CD8(+) T cells but also CD25(+)-activated T cells. In addition, a cytotoxicity assay confirmed that vaccination with the AdB7 transfectants induced tumor-specific cytotoxicity. CONCLUSION These results demonstrate the therapeutic potential of vaccination with tumor cells expressing B7.1 for the treatment of malignant glioma.
Collapse
Affiliation(s)
- Jun Morioka
- Department of Neurosurgery, Yamaguchi University School of Medicine, Yamaguchi, Japan
| | | | | | | | | | | |
Collapse
|
35
|
Chen XL, Cao XD, Kang AJ, Wang KM, Su BS, Wang YL. In situ expression and significance of B7 costimulatory molecules within tissues of human gastric carcinoma. World J Gastroenterol 2003; 9:1370-3. [PMID: 12800259 PMCID: PMC4611819 DOI: 10.3748/wjg.v9.i6.1370] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 02/06/2023] Open
Abstract
AIM: To explore the role and significance of costimulatory molecules B7H1, B7H2 and ICOS within tissues of human gastric carcinoma and the possible mechanisms in tumor escape.
METHODS: mRNA expressions of costimulatory molecules including B7H1, B7H2, ICOS and B7-1 in tissues of human gastric carcinoma were investigated by in situ hybridization using digoxigenin-labeled oligonucleotide-probes. The tissue of chronic gastric ulcer was used as a control. All data were analyzed by SPSS statistic software.
RESULTS: At the site of gastric carcinoma, mRNA expression levels of B7H1, B7H2 and ICOS were much higher than that of B7-1. Their mRNA positive expression indexes were 0.512 ± 0.333, 0.812 ± 0.454, 0.702 ± 0.359 and 0.293 ± 0.253, respectively. The positively stained cells were mainly tumor infiltrating lymphocytes (TILs), and some tumor cells. The difference between them was greatly significant P < 0.005. The mRNA expression levels of four molecules were not correlated to the pathological grade and matastasis of gastric carcinoma.
CONCLUSION: ICOS-B7H costimulatory pathway may be predominant at the site of gastric carcinoma. B7-1mRNA might be the basis of ICOS-B7H interaction. ICOS-B7H interaction induces the production of IL-10 which inhibits the antitumor immune responses. Therefore, it is supposed that ICOS-B7H costimulatory pathway may be involved in the negative regulation of cell-mediated immune responses.
Collapse
Affiliation(s)
- Xiao-Li Chen
- Department of Pathology, Second Hospital of Xi'an Jiaotong University, Xi'an 710004, Shaanxi Province, China.
| | | | | | | | | | | |
Collapse
|
36
|
Abstract
Prostate cancer is the second leading cause of cancer death in the US, largely because of the limitations of our current therapeutic options, especially once the cancer has metastasized. Investigators have long sought new therapeutic modalities such as angiogenesis inhibitors, vaccines, and gene therapy, among others. It appears that a combination approach will be required to cure the majority of malignancies. Immunotherapy for prostate cancer appears feasible and a likely therapeutic modality in the armamentarium. Unfortunately, further research in basic immunology and the interaction of the immune system with other forms of therapy is needed. Many obstacles exist in immunotherapy, including vector design, tumouricidal specificity, and tumor evasion, which will have to be overcome in order to realize the maximum therapeutic benefit from this treatment modality.
Collapse
Affiliation(s)
- Joseph M Kaminski
- Department of Radiology, Medical College of Georgia, Augusta, GA 30912, USA
| | | | | | | | | |
Collapse
|
37
|
Kaufman HL, Cheung K, Haskall Z, Horig H, Hesdorffer C, Panicali D, DeRaffele G, Spanknebel K. Clinical protocol. Intra-Lesional rF-B7.1 versus rF-TRICOM vaccine in the treatment of metastatic cancer. Hum Gene Ther 2003; 14:803-27. [PMID: 12804142 DOI: 10.1089/104303403765255183] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Affiliation(s)
- Howard L Kaufman
- Department of Surgery, Columbia Presbyterian Medical Center, New York, NY 10032, USA.
| | | | | | | | | | | | | | | |
Collapse
|
38
|
Airoldi I, Lualdi S, Bruno S, Raffaghello L, Occhino M, Gambini C, Pistoia V, Corrias MV. Expression of costimulatory molecules in human neuroblastoma. Evidence that CD40+ neuroblastoma cells undergo apoptosis following interaction with CD40L. Br J Cancer 2003; 88:1527-36. [PMID: 12771917 PMCID: PMC2377102 DOI: 10.1038/sj.bjc.6600951] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022] Open
Abstract
Tumour cells display low to absent expression of costimulatory molecules. Here, we have investigated the expression of costimulatory molecules (CD40, CD80, CD86, PD-1L, B7H2, OX40L and 4-1BBL) in human neuroblastoma (NB) cells, since virtually no information is available on this issue. Both established NB cell lines and primary tumours were tested by RT-PCR and flow cytometry. Neuroblastoma cell lines expressed the transcripts of all costimulatory molecule genes, but not the corresponding proteins. Culture of NB cell lines with human recombinant (r)IFN-gamma induced surface expression of CD40 in half of them. Primary NB cells showed CD40, CD80, CD86, OX40L, 4-1BBL, but not PD-1L and B7H2, mRNA expression. Surface CD40 was consistently detected on primary NB cells by flow cytometry. Interferon-gamma gene-transfected NB cells expressed constitutively surface CD40 and were induced into apoptosis by incubation with rCD40L through a caspase-8-dependent mechanism. CD40 may represent a novel therapeutic target in NB.
Collapse
Affiliation(s)
- I Airoldi
- Laboratory of Oncology, G Gaslini Institute, Largo G Gaslini, 5, 16148 Genova, Italy
| | - S Lualdi
- Laboratory of Oncology, G Gaslini Institute, Largo G Gaslini, 5, 16148 Genova, Italy
| | - S Bruno
- Department of Experimental Medicine, Section of Human Anatomy, University of Genova, Genova, Italy
| | - L Raffaghello
- Laboratory of Oncology, G Gaslini Institute, Largo G Gaslini, 5, 16148 Genova, Italy
| | - M Occhino
- Laboratory of Oncology, G Gaslini Institute, Largo G Gaslini, 5, 16148 Genova, Italy
| | - C Gambini
- Service of Pathology, G Gaslini Institute, 16148 Genova, Italy
| | - V Pistoia
- Laboratory of Oncology, G Gaslini Institute, Largo G Gaslini, 5, 16148 Genova, Italy
- Laboratory of Oncology, G Gaslini Institute, Largo G Gaslini, 5, 16148 Genova, Italy. E-mail:
| | - M V Corrias
- Laboratory of Oncology, G Gaslini Institute, Largo G Gaslini, 5, 16148 Genova, Italy
| |
Collapse
|
39
|
Morioka J, Kajiwara K, Yoshikawa K, Ideguchi M, Uchida T, Ohmoto Y, Suzuki M. Adenovirus-mediated gene transfer of B7.1 induces immunological anti-tumor effects in a murine brain tumor. J Neurooncol 2002; 60:13-23. [PMID: 12416541 DOI: 10.1023/a:1020260822669] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
The purpose of the present study was to determine if adenovirus-mediated transfection of a syngeneic mouse brain tumor with the gene encoding B7.1 enhances immunogenicity against tumor. Malignant astrocytoma cells were transfected with adenoviral vectors carrying the B7.1 gene (AdB7). Immunocytochemical analysis confirmed the expression of B7.1 in vitro and in vivo. To investigate the effects of B7.1 expression on tumorigenicity of the malignant astrocytoma, mice were implanted intracerebrally with B7.1-transfected glioma cells. There was no significant difference in proliferation between B7.1-transfected cells and controls in vitro. Nevertheless, mice implanted with B7. 1-transfected cells survived significantly longer than those in the control groups. Immunocytochemical analysis of the tumors showed that there was infiltration of a number of CD8+ T-cells and CD25+ activated T-cells in the brain implanted with B7.1-transfected glioma cells. The results showed the possibility that adenovirus-mediated B7.1 gene transfection to a brain tumor induced activation of CD8+ cytotoxic T-lymphocytes.
Collapse
Affiliation(s)
- Jun Morioka
- Department of Neurosurgery, Yamaguchi University School of Medicine, Japan
| | | | | | | | | | | | | |
Collapse
|
40
|
Diefenbach A, Raulet DH. The innate immune response to tumors and its role in the induction of T-cell immunity. Immunol Rev 2002; 188:9-21. [PMID: 12445277 DOI: 10.1034/j.1600-065x.2002.18802.x] [Citation(s) in RCA: 151] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Recent genetic studies have resurrected the concept that the adaptive and innate immune systems play roles in tumor surveillance. Natural killer (NK) cells recognize many tumor cells but not normal self cells, and they are thought to aid in the elimination of nascent tumors. Two main strategies are employed by NK cells to recognize tumor targets. Many tumor cells down-regulate class I major histocompatibility complex (MHC) molecules, thus releasing the NK cell from the inhibition provided by class I MHC-specific inhibitory receptors ('missing self recognition'). More recently, it has become clear that a stimulatory receptor expressed by NK cells, T cells and macrophages (NKG2D) recognizes ligands (MHC class I chain related [MIC], H6O, retinoic acid early inducible [Rae1] and UL16 binding proteins [ULBP]) that are up-regulated on tumor cells and virally infected cells but are not expressed well by normal cells. Ectopic expression of these ligands on tumor cells leads to the potent rejection of the tumors in vivo. Importantly, mice that previously rejected the ligand+ tumor cells develop T-cell immunity to the parental (ligand-) tumor cells. The recognition of induced-self ligands as a strategy to recognize abnormal self sets a precedent for a new immune recognition strategy of the innate immune system.
Collapse
Affiliation(s)
- Andreas Diefenbach
- Department of Molecular and Cell Biology and Cancer Research Laboratory, University of California, Berkeley, CA 94720, USA
| | | |
Collapse
|
41
|
Ando H, Saio M, Tamakawa N, Ohe N, Nakayama T, Yu H, Kaku Y, Iwama T, Shinoda J, Sakai N, Takami T. Failure of B7.1-modified tumor to evoke full activation of CD8+ tumor-infiltrating lymphocytes in the central nervous system: prevention of parental tumor growth in the subcutaneous environment. J Neurosurg 2002; 97:432-40. [PMID: 12186473 DOI: 10.3171/jns.2002.97.2.0432] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT It is well known that the central nervous system (CNS) is an immunologically privileged site. To characterize CD8+ tumor-infiltrating lymphocytes (TILs) recovered from the CNS, the authors compared these cells with TILs recovered from subcutaneous tissue by using a B7.1 gene-modified tumor implantation model. METHODS The authors established a B7.1 gene-modified EL4 murine lymphoma cell line (EL4-B7.1) and implanted the cells into the CNS to observe the duration of tumor-free survival. Although EL4-B7.1 cells were completely rejected in a subcutaneous implantation model, 40% of animals died after the CNS implantation (all animals in which the parent tumor was implanted died within 16 days). Therefore, the authors isolated TILs from each implantation site and analyzed the expressions of activation antigens CD25 and CD69 by performing the anti-CD8 magnetic beads separation method and flow cytometric analysis. After implantation of the parent tumor, there was no difference in the number of TILs from each site (CD25 1.7-3.2%, CD69 21.9-34.3%). After implantation of the B7.1-modified tumor, the CD25-expressing TIL population from the subcutaneous site was 4.68 times higher than that from the CNS site (17.8% compared with 3.8%). Based on these findings, the authors used a mitomycin C-treated EL4-B7.1 subcutaneous vaccination with various protocols. Vaccination before tumor challenge was sufficient to prevent the development of the tumor. For animals with established tumor, the vaccination protocol was able to prolong host survival (p = 0.0053). CONCLUSIONS The data clearly demonstrate that the CNS environment fails to activate CD8+ TILs fully. These are the first data indicating in detail a difference between CD8+ TILs from the CNS and those from other sites based on a B7.1-modified tumor model.
Collapse
Affiliation(s)
- Hiromichi Ando
- Department of Neurosurgery, Gifu University School of Medicine, Japan
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
42
|
Kelly JM, Takeda K, Darcy PK, Yagita H, Smyth MJ. A role for IFN-gamma in primary and secondary immunity generated by NK cell-sensitive tumor-expressing CD80 in vivo. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:4472-9. [PMID: 11970991 DOI: 10.4049/jimmunol.168.9.4472] [Citation(s) in RCA: 50] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
We have investigated the primary and secondary immunity generated in vivo by a MHC class I-deficient tumor cell line that expressed CD80 (B7-1). CD80 expression enhanced primary NK cell-mediated tumor rejection in vivo and T cell immunity against secondary tumor challenge. CD80 expression enhanced primary NK cell-mediated tumor rejection, and both NK cell perforin and IFN-gamma activity were critical for the rejection of MHC class I-deficient RMA-S-CD80 tumor cells. This primary rejection process stimulated the subsequent development of specific CTL and Th1 responses against Ags expressed by the MHC class I-deficient RMA-S tumor cells. The development of effective secondary T cell immunity could be elicited by irradiated RMA-S-CD80 tumor cells and was dependent upon NK cells and IFN-gamma in the priming response. Our findings demonstrate a key role for IFN-gamma in innate and adaptive immunity triggered by CD80 expression on tumor cells.
Collapse
Affiliation(s)
- Janice M Kelly
- Cancer Immunology Program, Sir Donald and Lady Trescowthick Laboratories, Peter MacCallum Cancer Institute, East Melbourne, Victoria, Australia
| | | | | | | | | |
Collapse
|
43
|
van Mierlo GJD, den Boer AT, Medema JP, van der Voort EIH, Fransen MF, Offringa R, Melief CJM, Toes REM. CD40 stimulation leads to effective therapy of CD40(-) tumors through induction of strong systemic cytotoxic T lymphocyte immunity. Proc Natl Acad Sci U S A 2002; 99:5561-6. [PMID: 11929985 PMCID: PMC122809 DOI: 10.1073/pnas.082107699] [Citation(s) in RCA: 186] [Impact Index Per Article: 8.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Adequate spontaneous activation of tumor-specific T lymphocytes in tumor-bearing hosts is rare, despite the expression of tumor antigens that are potentially highly immunogenic. For example, failure of the immune system to raise competent responses against established tumors expressing the human adenovirus E1A-antigen allows this tumor to grow in immunocompetent mice. We show that systemic in vivo administration of agonistic anti-CD40 antibodies into tumor-bearing mice results in tumor eradication mediated by CD8(+) T cells. Treatment resulted in a strong expansion and systemic accumulation of E1A-specific CTL and depended on CD40 expression on host cells, as the tumor was CD40(-), and therapy failed in CD40-deficient mice. Local intratumoral administration of anti-CD40 mAb is equally effective in licensing strong, systemic CTL immunity, resulting in the clearance of distant tumor nodules. Our data indicate that the immune response after cancer-host interactions can be directed toward competence, leading to the cure of established tumors merely by delivery of a CD40-dependent "license to kill" signal.
Collapse
Affiliation(s)
- Geertje J D van Mierlo
- Department of Immunohematology and Bloodtransfusion, Leiden University Medical Center, P.O. Box 9600, 2300 RC, Leiden, The Netherlands
| | | | | | | | | | | | | | | |
Collapse
|
44
|
Pützer BM, Rödicker F, Hitt MM, Stiewe T, Esche H. Improved treatment of pancreatic cancer by IL-12 and B7.1 costimulation: antitumor efficacy and immunoregulation in a nonimmunogenic tumor model. Mol Ther 2002; 5:405-12. [PMID: 11945067 DOI: 10.1006/mthe.2002.0570] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022] Open
Abstract
Ductal pancreatic adenocarcinoma is one of the commonest and most lethal cancers in the Western world. Unfortunately, recent advances in diagnostics, staging, and therapy in pancreatic carcinoma have not resulted in significant improvements in long-term survival. We have previously shown that adenovirus (Ad)-mediated coexpression of interleukin-12 (IL-12) and the costimulatory molecule B7.1 is extremely efficient in inducing regression of highly immunogenic transplanted and nontransplanted tumors. Here, we examined the antitumor efficacy of IL-12- and B7.1-based immunotherapy against a nonimmunogenic murine model of ductal pancreatic cancer. Compared with AdIL-12 treatment alone, single intratumoral injection of AdIL-12/B7.1 led to a prolonged immune response and mediated complete regression in 80% of treated animals. After rechallenge with parental tumor cells, 70% of cured mice remained tumor-free, suggesting that protective immunity had been induced. The antitumoral response was associated with upregulation of H-2K(b) and Abcb2 expression, whereas other components of the proteasome (Abcb3, Psmb9, and Psmb8) were not affected. These data indicate that upregulation of the antigen presentation machinery by AdIL-12/B7.1 may be a therapeutic rationale for nonimmunogenic, therapy-resistant pancreatic cancer.
Collapse
Affiliation(s)
- Brigitte M Pützer
- Center for Cancer Research and Cancer Therapy, Institute of Molecular Biology, University of Essen Medical School, Hufelandstr. 55, Essen, 45122, Germany.
| | | | | | | | | |
Collapse
|
45
|
LaBelle JL, Hanke CA, Blazar BR, Truitt RL. Negative effect of CTLA-4 on induction of T-cell immunity in vivo to B7-1+, but not B7-2+, murine myelogenous leukemia. Blood 2002; 99:2146-53. [PMID: 11877291 DOI: 10.1182/blood.v99.6.2146] [Citation(s) in RCA: 41] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
B7 molecules provide important costimulatory signals to T cells, and B7 genes have been introduced into B7-negative tumor cells to enhance their immunogenicity. However, the role of B7 molecules in inducing tumor immunity is controversial because of conflicting results and reports of differential signaling through the B7 molecules and their ligands CD28 and CTLA-4. In this study, we compared the effect of B7-1 (CD80) and B7-2 (CD86) on the induction of T-cell immunity to C1498, a murine myelogenous leukemia. When cultured with exogenous cytokines in vitro, C1498/B7-1 and C1498/B7-2 induced syngeneic CD8+ T cells to kill parental C1498. In vivo, C1498/B7-1 grew progressively after subcutaneous injection, whereas C1498/B7-2 completely regressed after transient growth in naive mice. Spontaneous rejection of C1498/B7-2 resulted in immunity to challenge doses of C1498 and C1498/B7-1. Antibody-depletion studies in vivo showed that CD8+ T cells rejected C1498/B7-2, whereas only natural killer cells affected the growth of C1498/B7-1. Two approaches were used to determine whether preferential interaction of B7-1 with CTLA-4 contributed to the failure of C1498/B7-1 to activate CD8+ T cells in vivo. First, CTLA-4 specific monoclonal antibody was used to block B7-1-CTLA-4 interaction. Second, CTLA-4 deletional mutant (-/-) bone marrow chimeras were used as tumor hosts. In both systems, there was a significant increase in the rate of rejection of C1498/B7-1 tumors. Resistance to C1498/B7-1 in CTLA-4(minus sign/minus sign) hosts was mediated by CD8+ T cells. Blocking or deletion of CTLA-4 did not affect the growth of parental C1498, indicating that B7-1 was important for the induction of CD8+ T-cell immunity in the absence of CTLA-4.
Collapse
MESH Headings
- Abatacept
- Animals
- Antigens, CD/genetics
- Antigens, CD/immunology
- Antigens, Differentiation/genetics
- Antigens, Differentiation/metabolism
- Antigens, Differentiation/pharmacology
- B7-1 Antigen/genetics
- B7-1 Antigen/immunology
- B7-1 Antigen/metabolism
- B7-2 Antigen
- CD8-Positive T-Lymphocytes/immunology
- CTLA-4 Antigen
- Cell Division/drug effects
- Cytotoxicity, Immunologic/drug effects
- Immunity, Cellular/drug effects
- Immunoconjugates
- Killer Cells, Natural/immunology
- Leukemia, Myeloid/immunology
- Leukemia, Myeloid/metabolism
- Leukemia, Myeloid/pathology
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/immunology
- Mice
- Mice, Knockout
- Protein Binding
- T-Lymphocytes/drug effects
- T-Lymphocytes/immunology
- Transfection
- Tumor Cells, Cultured/transplantation
Collapse
Affiliation(s)
- James L LaBelle
- Department of Microbiology and Molecular Genetics, Medical College of Wisconsin, 8701 Watertown Plank Road, Milwaukee, WI 53226, USA
| | | | | | | |
Collapse
|
46
|
Rodríguez-Calvillo M, Duarte M, Tirapu I, Berraondo P, Mazzolini G, Qian C, Prieto J, Melero I. Upregulation of natural killer cells functions underlies the efficacy of intratumorally injected dendritic cells engineered to produce interleukin-12. Exp Hematol 2002; 30:195-204. [PMID: 11882356 DOI: 10.1016/s0301-472x(01)00792-5] [Citation(s) in RCA: 18] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Injection of dendritic cells (DC) engineered with recombinant adenoviral vectors to produce interleukin-12 (IL-12) inside experimental murine tumors frequently achieves complete regressions. In such a system the function of CD8(+) T cells has been shown to be an absolute requirement, in contrast to observations made upon depletion of CD4(+) T cells, which minimally affected the outcome. The aim of this work was to study the possible involvement of natural killer (NK) cells in this setting. MATERIALS, METHODS, AND RESULTS Depletions with anti-AsialoGM1 antiserum showed only a small decrease in the proportion of complete regressions obtained that correlated with induction of NK activities in lymphatic tissues into which DC migrate, whereas combined depletions of CD4(+) and NK cells completely eliminated the antitumor effects. Likewise in vivo neutralization of interferon-gamma (IFN-gamma) also eliminated those therapeutic effects. Trying to define the cellular role played by NK cells in vivo, it was observed that injection of cultured DC inside the spleen of T- and B-cell-deficient (Rag1(-/-)) mice induced upregulation of NK activity only if DC had been adenovirally engineered to produce IL-12. In addition, identically transfected fibroblasts also activated NK cells, indicating that IL-12 transfection was the unique requirement. Equivalent human DC only activated in vitro the cytolytic and cytokine-secreting functions of autologous NK cells if transfected to express human IL-12. CONCLUSIONS Overall, these results point out an important role played by NK cell activation in the potent immunotherapeutic effects elicited by intratumoral injection of IL-12--secreting DC and that NK activation under these conditions is mainly, if not only, dependent on IL-12.
Collapse
|
47
|
Ferlazzo G, Tsang ML, Moretta L, Melioli G, Steinman RM, Münz C. Human dendritic cells activate resting natural killer (NK) cells and are recognized via the NKp30 receptor by activated NK cells. J Exp Med 2002; 195:343-51. [PMID: 11828009 PMCID: PMC2193591 DOI: 10.1084/jem.20011149] [Citation(s) in RCA: 751] [Impact Index Per Article: 32.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/05/2022] Open
Abstract
During the innate response to many inflammatory and infectious stimuli, dendritic cells (DCs) undergo a differentiation process termed maturation. Mature DCs activate antigen-specific naive T cells. Here we show that both immature and mature DCs activate resting human natural killer (NK) cells. Within 1 wk the NK cells increase two-- to fourfold in numbers, start secreting interferon (IFN)-gamma, and acquire cytolytic activity against the classical NK target LCL721.221. The DC-activated NK cells then kill immature DCs efficiently, even though the latter express substantial levels of major histocompatibility complex (MHC) class I. Similar results are seen with interleukin (IL)-2--activated NK cell lines and clones, i.e., these NK cells kill and secrete IFN-gamma in response to immature DCs. Mature DCs are protected from activated NK lysis, but lysis takes place if the NK inhibitory signal is blocked by a human histocompatibility leukocyte antigen (HLA)-A,B,C--specific antibody. The NK activating signal mainly involves the NKp30 natural cytotoxicity receptor, and not the NKp46 or NKp44 receptor. However, both immature and mature DCs seem to use a NKp30 independent mechanism to act as potent stimulators for resting NK cells. We suggest that DCs are able to control directly the expansion of NK cells and that the lysis of immature DCs can regulate the afferent limb of innate and adaptive immunity.
Collapse
Affiliation(s)
- Guido Ferlazzo
- Laboratorio di Immunoterapia Cellulare, Unità di Immunologia, Istituto Nazionale per la Ricerca sul Cancro, 16132 Genova, Italy.
| | | | | | | | | | | |
Collapse
|
48
|
Liu Y, Qureshi M, Xiang J. Antitumor immune responses derived from transgenic expression of CD40 ligand in myeloma cells. Cancer Biother Radiopharm 2002; 17:11-8. [PMID: 11915168 DOI: 10.1089/10849780252824028] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Tumor cells engineered to express immunogenes have been used for cancer vaccines to induce the antitumor immunity and study the antitumor immune mechanisms derived from the immunogene expression. In the present study, we engineered a mouse myeloma cell line J558 with a cloned CD40 ligand (CD40L) gene. We demonstrated that (i) the engineered J558/CD40L tumor cells expressing the CD40 ligand molecule lost their tumorigenicity in syngeneic mice, and (ii) the inoculation of J558/CD40L tumor cells further lead to the protective immunity against wild-type J558 tumors. In animal studies using T-cell subset depleted mice, we further showed that the primary rejection of J558/CD40L tumors did not require T cells, but was mainly mediated by NK cells, whereas the effector phase of the protective immunity is mediated by CD8+ T cells. In addition, our data, for the first time, showed that the inoculation of engineered J558/CD40L tumor cells is able to stimulate stronger activation of dendritic cells with enhanced expression of B7-1 and ICAM-1 molecules than the wild-type J558 tumor cells Taken together, we demonstrated the antitumor effect of engineered J558/CD40L tumor cells that is mediated by the activation of the host dendritic cells in vivo. Our data indicate that the introduction of co-stimulatory CD40 ligand molecule will be useful as a new strategy of immunogene therapy against tumors.
Collapse
Affiliation(s)
- Yongqing Liu
- Saskatoon Cancer Center, Department of Microbiology, University of Saskatchewan, Saskatoon, Saskatchewan S7N 0W0, Canada
| | | | | |
Collapse
|
49
|
Tokunaka K, Ohno N, Adachi Y, Miura NN, Yadomae T. Application of Candida solubilized cell wall beta-glucan in antitumor immunotherapy against P815 mastocytoma in mice. Int Immunopharmacol 2002; 2:59-67. [PMID: 11789670 DOI: 10.1016/s1567-5769(01)00148-5] [Citation(s) in RCA: 21] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022]
Abstract
This study was designed to evaluate the antitumor activity of CSBG, purified from the cell wall of Candida albicans IFO1385. First, as an effect of CSBG on P815 mastocytoma, significant prolonged survival and suppression of the tumor growth were observed. Second, the transfer of spleen cells from CSBG-sensitized BALB/c mice to CDF1 mice led to further suppression of tumor growth as well as P815-immunized spleen cells. Third, CSBG enhanced antitumor immunity in gene therapy using B7-1-transfected P815 cells. These results strongly suggest that CSBG enhances the host defense response to tumor due in part to an adjuvant effect.
Collapse
Affiliation(s)
- Kazuhiro Tokunaka
- Laboratory for Immunopharmacology of Microbial Products, School of Pharmacy, Tokyo University of Pharmacy and Life Science, Hachioji, Japan
| | | | | | | | | |
Collapse
|
50
|
Liu X, Bai XF, Wen J, Gao JX, Liu J, Lu P, Wang Y, Zheng P, Liu Y. B7H costimulates clonal expansion of, and cognate destruction of tumor cells by, CD8(+) T lymphocytes in vivo. J Exp Med 2001; 194:1339-48. [PMID: 11696598 PMCID: PMC2195972 DOI: 10.1084/jem.194.9.1339] [Citation(s) in RCA: 99] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/03/2000] [Revised: 07/13/2001] [Accepted: 07/24/2001] [Indexed: 01/03/2023] Open
Abstract
B7H/B7RP (hereby called B7H) is a new member of the B7 family of costimulatory molecules and interacts with inducible costimulatory molecule (ICOS). Its function for CD8 T cells has not been reported. We report here that expression of B7H on the tumor cells reduced tumorigenicity and induced immunity to subsequent challenge with parental tumor cells. The immune protection correlates with an enhanced cytotoxic T lymphocyte (CTL) response against P1A, the major tumor antigen expressed in the J558 tumor. To understand the mechanism of immune protection, we adoptively transferred transgenic T cells specific for tumor antigen P1A into mice that bore P1A-expressing tumors. We found that while the transgenic T cells divided faster in mice bearing the B7H(+) tumors, optimal B7H-induced clonal expansion of P1CTL required costimulation by B7-1 and B7-2 on the endogenous host antigen-presenting cells (APCs). Interestingly, when B7H(+) and B7H(-) tumors were coinjected, P1CTL selectively eliminated the B7H(+) tumor cells. Moreover, B7H expressed on the tumor cells made them highly susceptible to destruction by CTL in vivo, even if the CTL was administrated into mice with large tumor burdens. Tumors that recurred in the P1CTL-treated mice lost transfected B7H and/or H-2L(d), the class I molecule that presents the P1A peptide. Taken together, our results reveal that B7H costimulates clonal expansion of, and cognate destruction by CD8(+) T lymphocytes in vivo.
Collapse
Affiliation(s)
- X Liu
- Department of Pathology and Comprehensive Cancer Center, Ohio State University Medical Center, Columbus, OH 43210, USA
| | | | | | | | | | | | | | | | | |
Collapse
|