101
|
Grimm M, Gasser M, Bueter M, Strehl J, Wang J, Nichiporuk E, Meyer D, Germer CT, Waaga-Gasser AM, Thalheimer A. Evaluation of immunological escape mechanisms in a mouse model of colorectal liver metastases. BMC Cancer 2010; 10:82. [PMID: 20205946 PMCID: PMC2838824 DOI: 10.1186/1471-2407-10-82] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/03/2009] [Accepted: 03/07/2010] [Indexed: 11/10/2022] Open
Abstract
BACKGROUND The local and systemic activation and regulation of the immune system by malignant cells during carcinogenesis is highly complex with involvement of the innate and acquired immune system. Despite the fact that malignant cells do have antigenic properties their immunogenic effects are minor suggesting tumor induced mechanisms to circumvent cancer immunosurveillance. The aim of this study is the analysis of tumor immune escape mechanisms in a colorectal liver metastases mouse model at different points in time during tumor growth. METHODS CT26.WT murine colon carcinoma cells were injected intraportally in Balb/c mice after median laparotomy using a standardized injection technique. Metastatic tumor growth in the liver was examined by standard histological procedures at defined points in time during metastatic growth. Liver tissue with metastases was additionally analyzed for cytokines, T cell markers and Fas/Fas-L expression using immunohistochemistry, immunofluorescence and RT-PCR. Comparisons were performed by analysis of variance or paired and unpaired t test when appropriate. RESULTS Intraportal injection of colon carcinoma cells resulted in a gradual and time dependent metastatic growth. T cells of regulatory phenotype (CD4+CD25+Foxp3+) which might play a role in protumoral immune response were found to infiltrate peritumoral tissue increasingly during carcinogenesis. Expression of cytokines IL-10, TGF-beta and TNF-alpha were increased during tumor growth whereas IFN-gamma showed a decrease of the expression from day 10 on following an initial increase. Moreover, liver metastases of murine colon carcinoma show an up-regulation of FAS-L on tumor cell surface with a decreased expression of FAS from day 10 on. CD8+ T cells express FAS and show an increased rate of apoptosis at perimetastatic location. CONCLUSIONS This study describes cellular and macromolecular changes contributing to immunological escape mechanisms during metastatic growth in a colorectal liver metastases mouse model simulating the situation in human cancer.
Collapse
Affiliation(s)
- Martin Grimm
- Department of General Surgery, Molecular Oncology and Immunology, University of Wuerzburg Hospital, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
102
|
Liu B, Faia L, Hu M, Nussenblatt RB. Pro-angiogenic effect of IFNgamma is dependent on the PI3K/mTOR/translational pathway in human retinal pigmented epithelial cells. Mol Vis 2010; 10:3. [PMID: 20144242 PMCID: PMC2834660 DOI: 10.1186/1471-2415-10-3] [Citation(s) in RCA: 18] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/29/2009] [Accepted: 02/10/2010] [Indexed: 12/13/2022] Open
Abstract
Background Mutations in COL8A2 gene which encodes the collagen alpha-2 (VIII) chain have been identified in both familial and sporadic cases of Fuchs endothelial corneal dystrophy (FECD). Heterozygous mutations in the SLC4A11 gene are also known to cause late-onset FECD. Therefore we screened for COL8A2, SLC4A11 gene variants in Indian FECD patients. Methods Eighty patients with clinically diagnosed FECD and 100 age matched normal individuals were recruited. Genomic DNA was isolated from peripheral blood leukocytes. Mutations in COL8A2, SLC4A11 coding regions were screened using bi-directional sequencing. Fischer's exact test or Pearson's chi squared test were used to predict the statistical association of genotypes with the phenotype. Results Screening of COL8A2 gene revealed 2 novel c.1610G>A, c.1643A>G and 3 reported variations c.112G>A, c.464G>A and c.1485G>A. In SLC4A11 gene, novel c.1659C>T, c.1974C>T and reported c.405G>A, c.481A>C and c.639G>A variants were identified. However all the variations in both the genes were also present in unaffected controls. Conclusions This is the first study analysing COL8A2 gene in Indian patients with FECD. No pathogenic mutations were identified in COL8A2. Merely silent changes, which showed statistically insignificant association with FECD, were identified in the screening of SLC4A11 gene. These results suggest that COL8A2, SLC4A11 genes may not be responsible for FECD in patients examined in this study.
Collapse
Affiliation(s)
- Baoying Liu
- Laboratory of Immunology, National Eye Institute, National Institutes of Health, Bethesda, MD 20892, USA
| | | | | | | |
Collapse
|
103
|
|
104
|
Ostrand-Rosenberg S. CD4+T Lymphocytes: A Critical Component of Antitumor Immunity. Cancer Invest 2009. [DOI: 10.1081/cnv-67428] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/03/2022]
|
105
|
Ding Y, Seow SV, Huang CH, Liew LM, Lim YC, Kuo IC, Chua KY. Coadministration of the fungal immunomodulatory protein FIP-Fve and a tumour-associated antigen enhanced antitumour immunity. Immunology 2009; 128:e881-94. [PMID: 19740349 DOI: 10.1111/j.1365-2567.2009.03099.x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/28/2022] Open
Abstract
Fve is a fungal protein isolated from the golden needle mushroom Flammulina velutipes and has previously been reported to trigger immunological responses in both mouse and human lymphocytes. In this study, we evaluated the potential application of Fve as an adjuvant for tumour immunotherapy and examined the underlying mechanism(s). When the human papillomavirus (HPV)-16 E7 oncoprotein was used as a model antigen, mice coimmunized with HPV-16 E7 and Fve showed enhanced production of HPV-16 E7-specific antibodies as well as expansion of HPV-16 E7-specific interferon (IFN)-gamma-producing CD4(+) and CD8(+) T cells as compared with mice immunized with HPV-16 E7 alone. Tumour protection assays showed that 60% of mice coimmunized with HPV-16 E7 plus Fve, as compared with 20% of those immunized only with HPV-16 E7, remained tumour-free for up to 167 days after challenge with the tumour cells. Tumour therapeutic assays showed that HPV-16 E7 plus Fve treatment significantly prolonged the survival of tumour-bearing mice as compared with those treated only with HPV-16 E7. In vivo cell depletion and adoptive T-cell transfer assays showed that CD4(+) and CD8(+) T cells and IFN-gamma played critical roles in conferring the antitumour effects. Interestingly, Fve could stimulate the maturation of splenic dendritic cells in vivo and induce antigen-specific CD8(+) T-cell immune responses. In summary, Fve has potent adjuvant properties that enhance T helper type 1 antigen-specific humoral and cellular immune responses which confer strong antitumour effects. The use of Fve as an adjuvant could be an attractive alternative to the current vaccination strategy for cancer immunotherapy.
Collapse
Affiliation(s)
- Ying Ding
- Department of Paediatrics, Yong Loo Lin School of Medicine, National University of Singapore, Singapore City, Singapore
| | | | | | | | | | | | | |
Collapse
|
106
|
EpCAM, a human tumor-associated antigen promotes Th2 development and tumor immune evasion. Blood 2009; 113:3494-502. [PMID: 19188665 DOI: 10.1182/blood-2008-08-175109] [Citation(s) in RCA: 56] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/29/2022] Open
Abstract
Experimental tumor vaccination and adoptive T-cell therapies show that interferon-gamma (IFN-gamma)-producing CD4(+) T helper cells (Th1) can be highly effective in tumor prevention and therapy. Unexpectedly, first vaccine trials in humans revealed that tumor immune therapy may not only be protective, but, on the contrary, even promote tumor progression. Here, we analyzed T-cell immune responses to the epithelial cell adhesion molecule (EpCAM), one of the most common tumor-associated antigens (TAA) serving as immune target in colon cancer patients. Th-cell priming against EpCAM inevitably resulted in interleukin-4 (IL-4)-dominated Th2 responses, even under most stringent Th1-inducing conditions. These EpCAM-reactive Th2 cells rather promoted growth of EpCAM-expressing tumors. To analyze the role of IL-4 in tumor immune evasion, we generated EpCAM-reactive Th1 cells from IL-4.ko mice. These Th1 cells provided tumor-specific protection and established highly protective Th1 memory responses, even in naive BALB/c mice. Inhibition of tumor growth by Th1 cells resulted in intra-tumoral expression of cytokines of the IL-12 family and of IFN-gamma. Preventing activation-associated death of Th1 cells further increased intratumoral IFN-gamma expression and improved therapeutic efficacy. Thus, human TAA may promote tumor immune evasion by strongly favoring Th2 development.
Collapse
|
107
|
Song S, Liu C, Wang J, Zhang Y, You H, Wang Y, Liu F, Sun S. Vaccination with combination of Fit3L and RANTES in a DNA prime-protein boost regimen elicits strong cell-mediated immunity and antitumor effect. Vaccine 2008; 27:1111-8. [PMID: 19100302 DOI: 10.1016/j.vaccine.2008.11.095] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/24/2008] [Accepted: 11/22/2008] [Indexed: 12/21/2022]
Abstract
With accumulating evidence indicating the importance of cytotoxic T lymphocytes (CTLs) in the antitumor response, strategies are being pursued to elicit augmented CD8(+) T-cell responses against tumors with tumor vaccines. Here, we report the protective efficacy of vaccine-elicited antitumor immune responses with an aggressive HBc-expressing B16-HBc melanoma, which expressed HBc as a self and model antigen, tumor model. We demonstrated that the significantly better memory responses or marked inhibition on tumor growth could be achieved after coadministration of cytokine adjuvants RANTES and Flt3L in a DNA prime-protein boost regimen. Furthermore, the augmentation of DNA prime-protein boost regimens by cytokines gene was due to the improvement the immunopotency of DNA vaccine and subsequently the augmented Ag-specific and IFN-gamma mediating CD8(+) T-cell responses after protein boosting. Hence, this study demonstrates for the first time that combinatorial use of chemotactic and potent DC-specific growth factor molecules provides a useful strategy for enhancing antitumor responses.
Collapse
Affiliation(s)
- Shuxia Song
- Department of Molecular Biology and Key Lab of Laboratory Animal, Hebei Medical University, Shijiazhuang, PR China
| | | | | | | | | | | | | | | |
Collapse
|
108
|
Barber A, Zhang T, Megli CJ, Wu J, Meehan KR, Sentman CL. Chimeric NKG2D receptor-expressing T cells as an immunotherapy for multiple myeloma. Exp Hematol 2008; 36:1318-28. [PMID: 18599182 PMCID: PMC2638591 DOI: 10.1016/j.exphem.2008.04.010] [Citation(s) in RCA: 62] [Impact Index Per Article: 3.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/04/2008] [Revised: 04/14/2008] [Accepted: 04/22/2008] [Indexed: 11/25/2022]
Abstract
OBJECTIVE Most myeloma tumor cells from patients express NKG2D ligands. We have reported the development of a chimeric NKG2D receptor (chNKG2D), which consists of the NKG2D receptor fused to the CD3zeta chain. T cells expressing this receptor kill and produce cytokines in response to NKG2D-ligand+ tumor cells. Therefore, we investigated whether human chNKG2D T cells respond against human myeloma cells. MATERIALS AND METHODS ChNKG2D T cells were generated from healthy donors and myeloma patients. The effector phase of chNKG2D T cells was analyzed by cell-surface marker expression and human myeloma cell lines were tested for expression of NKG2D ligands. Lysis of myeloma cell lines and cytokine secretion by chNKG2D T cells was determined. ChNKG2D T cells grown in serum-free media, or cyropreserved, were assessed for effector cell functions. RESULTS Myeloma cell lines expressed NKG2D ligands. ChNKG2D T cells from healthy donors and myeloma patients lysed myeloma cells, and secreted proinflammatory cytokines when cultured with myeloma cells or patient bone marrow, but not with peripheral blood mononuclear cells or normal bone marrow. Lysis of myeloma cells was dependent on chNKG2D T-cell expression of NKG2D and perforin. Additionally, chNKG2D T cells upregulated CD45RO, did not express CD57, and maintained expression of CD27, CD62L, and CCR7, indicating that the T cells were at an early effector stage. Finally, we showed that chNKG2D T cells generated with serum-free media, or when cryopreserved, maintained effector functions. CONCLUSION ChNKG2D T cells respond to human myeloma cells and can be generated using clinically applicable cell culture techniques.
Collapse
Affiliation(s)
- Amorette Barber
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, NH
| | - Tong Zhang
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, NH
| | - Christina J. Megli
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, NH
| | - Jillian Wu
- Bone Marrow Transplant Program, Dartmouth Hitchcock Medical Center, Dartmouth Medical School and the Norris Cotton Cancer Center, Lebanon, NH
| | - Kenneth R. Meehan
- Bone Marrow Transplant Program, Dartmouth Hitchcock Medical Center, Dartmouth Medical School and the Norris Cotton Cancer Center, Lebanon, NH
| | - Charles L. Sentman
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, NH
| |
Collapse
|
109
|
Naschberger E, Croner RS, Merkel S, Dimmler A, Tripal P, Amann KU, Kremmer E, Brueckl WM, Papadopoulos T, Hohenadl C, Hohenberger W, Stürzl M. Angiostatic immune reaction in colorectal carcinoma: Impact on survival and perspectives for antiangiogenic therapy. Int J Cancer 2008; 123:2120-9. [PMID: 18697200 DOI: 10.1002/ijc.23764] [Citation(s) in RCA: 69] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/24/2022]
Abstract
Angiogenesis and inflammation are the 2 major stroma reactions in colorectal carcinoma (CRC). Guanylate binding protein-1 (GBP-1) is a key mediator of angiostatic effects of inflammation. Therefore, we hypothesized that GBP-1 may be a biomarker of intrinsic angiostasis associated with an improved outcome in CRC patients. GBP-1 was strongly expressed in endothelial cells and immune cells in the desmoplastic stroma of 32% of CRC as determined by immunohistochemical investigation of 388 sporadic CRC. Cancer-related 5-year survival was highly significant (p < 0.001) increased (16.2%) in patients with GBP-1-positive CRC. Multivariate analysis showed that GBP-1 is an independent prognostic factor indicating a reduction of the relative risk of cancer-related death by the half (p = 0.032). A comparative transcriptome analysis (22,215 probe sets) of GBP-1-positive (n = 12) and -negative (n = 12) tumors showed that particularly IFN-gamma-induced genes including the major antiangiogenic chemokines CXCL9, CXCL10 and CXCL11 were coexpressed with GBP-1. Altogether our findings indicated that GBP-1 may be a novel biomarker and an active component of a Th-1-like angiostatic immune reaction in CRC. This reaction may affect patient's response to antiangiogenic therapy and the identification of such tumors may provide a novel criterion for patient selection. Moreover, the induction of a Th-1-like angiostatic immune reaction may be a promising approach for the clinical treatment of CRC.
Collapse
Affiliation(s)
- Elisabeth Naschberger
- Division of Molecular and Experimental Surgery, Department of Surgery, University of Erlangen-Nuremberg, Schwabachanlage 10, Erlangen, Germany
| | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
110
|
Barber A, Zhang T, Sentman CL. Immunotherapy with chimeric NKG2D receptors leads to long-term tumor-free survival and development of host antitumor immunity in murine ovarian cancer. THE JOURNAL OF IMMUNOLOGY 2008; 180:72-8. [PMID: 18097006 DOI: 10.4049/jimmunol.180.1.72] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/07/2023]
Abstract
Ovarian cancer is one of the leading causes of cancer death in women and the development of novel therapies is needed to complement the standard treatment options such as chemotherapy and radiation. In this study, we show that treatment with T cells expressing a chimeric NKG2D receptor (chNKG2D) was able to lead to long-term, tumor-free survival in mice bearing established ovarian tumors. Tumor-free mice were able to reject a rechallenge with ovarian tumor cells 225 days after original tumor injection. In addition, chNKG2D T cell treatment induced specific host immune responses to ovarian tumor cells, including the development of both CD8+ and CD4+ T cell tumor-specific memory responses. The chNKG2D T cells reduced the ovarian tumor burden using both cytotoxic and cytokine-dependent pathways. Specifically, chNKG2D T cell expression of perforin, GM-CSF, and IFN-gamma were essential for complete antitumor efficacy.
Collapse
Affiliation(s)
- Amorette Barber
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, NH 03756, USA
| | | | | |
Collapse
|
111
|
Miller TJ, Honchel R, Espandiari P, Knapton A, Zhang J, Sistare FD, Hanig JP. The utility of the K6/ODC transgenic mouse as an alternative short term dermal model for carcinogenicity testing of pharmaceuticals. Regul Toxicol Pharmacol 2007; 50:87-97. [PMID: 18069108 DOI: 10.1016/j.yrtph.2007.10.011] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/24/2007] [Revised: 10/16/2007] [Accepted: 10/17/2007] [Indexed: 11/19/2022]
Abstract
The use of transgenic rodents may overcome many limitations of traditional cancer studies. Regulatory perspectives continue to evolve as new models are developed and validated. The transgenic mouse, K6/ODC, develops epidermal tumors when exposed to genotoxic carcinogens. In this study, K6/ODC mice were evaluated for model fitness and health robustness in a 36-week study to determine oncogenic risk of residual DNA in vaccines from neoplastic cell substrates. K6/ODC and C57BL/6 mice were treated with T24-H-ras expression plasmid, carrier vector DNA, or saline topically or by subcutaneous injection. One group of K6/ODC mice received 7,12-dimethylbenz-[a]anthracene [DMBA] dermally. Only DMBA-treated mice developed papillomas by six weeks, increasing in incidence to 25 weeks. By week 11, many K6/ODC mice showed severe dehydration and dermal eczema. By week 32, (6/8) surviving K6/ODC mice showed loss of mobility and balance. Microscopic evaluation of tissues revealed dermal/sebaceous gland hyperplasia, follicular dystrophy, splenic atrophy, and amyloid deposition/neutrophilic infiltration within liver, heart, and spleen, in all K6/ODC mice. Pathology was not detected in C57BL/6 mice. Progressive adverse health, decreased survival, and failure to develop papillomas to the H-ras plasmid suggest that K6/ODC mice may be an inappropriate alternative model for detection of oncogenic DNA and pharmaceutical carcinogenicity testing.
Collapse
Affiliation(s)
- T J Miller
- Division of Applied Pharmacology Research, Center for Drug Evaluation and Research, Food and Drug Administration, Silver Spring, MD 20993-0002, USA.
| | | | | | | | | | | | | |
Collapse
|
112
|
Zhang T, Barber A, Sentman CL. Chimeric NKG2D modified T cells inhibit systemic T-cell lymphoma growth in a manner involving multiple cytokines and cytotoxic pathways. Cancer Res 2007; 67:11029-36. [PMID: 18006849 DOI: 10.1158/0008-5472.can-07-2251] [Citation(s) in RCA: 67] [Impact Index Per Article: 3.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
In this study, the efficacy and mechanisms of chimeric NKG2D receptor (chNKG2D)-modified T cells in eliminating NKG2D ligand-positive RMA/Rae1 lymphoma cells were evaluated. Intravenous injection of RMA/Rae1 cells led to significant tumor formation in spleens and lymph nodes within 2 weeks. Adoptive transfer of chNKG2D-modified T cells after tumor injection significantly reduced tumor burdens in both spleens and lymph nodes, and prolonged the survival of tumor-bearing mice. Multiple treatments with chNKG2D T cells resulted in long-term tumor-free survival. Moreover, these long-term survivors were resistant to rechallenge with RMA tumor cells (NKG2D ligand-negative), and their spleen and lymph node cells produced IFN-gamma in response to RMA but not to other tumors in vitro, indicating immunity against RMA tumor antigens. ChNKG2D T cell-derived IFN-gamma and granulocyte-macrophage colony-stimulating factor, but not perforin (Pfp), tumor necrosis factor-related apoptosis-inducing ligand, or Fas ligand (FasL) alone were critical for in vivo efficacy. T cells deficient in both Pfp and FasL did not kill NKG2D ligand-positive RMA cells in vitro. Adoptive transfer of Pfp(-/-)FasL(-/-) chNKG2D T cells had reduced in vivo efficacy, indicating that chNKG2D T cells used both mechanisms to attack RMA/Rae1 cells. Taken together, these results indicate that chNKG2D T-cell-mediated therapeutic effects are mediated by both cytokine-dependent and cytotoxic mechanisms in vivo.
Collapse
Affiliation(s)
- Tong Zhang
- Department of Microbiology and Immunology, Dartmouth Medical School, Lebanon, New Hampshire 03756, USA
| | | | | |
Collapse
|
113
|
Dace DS, Chen PW, Niederkorn JY. CD4+ T-cell-dependent tumour rejection in an immune-privileged environment requires macrophages. Immunology 2007; 123:367-77. [PMID: 17944931 DOI: 10.1111/j.1365-2567.2007.02700.x] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/30/2022] Open
Abstract
Although intraocular tumours reside in an immune-privileged site, they can circumvent immune privilege and undergo rejection. Ocular tumour rejection typically follows one of two pathways. One pathway involves CD4+ T cells, delayed-type hypersensitivity (DTH), and culmination in ischemic necrosis of the tumour and phthisis (atrophy) of the eye. The second pathway is DTH-independent and does not inflict collateral injury to ocular tissues, and the eye is preserved. In this study, we used a well-characterized tumour, Ad5E1, to investigate the role of CD4+ T cells in the non-phthisical form of intraocular tumour rejection. It has been previously documented that CD4+ T cells and interferon (IFN)-gamma are necessary for rejection of these tumours in the eye. In this study, we demonstrate that CD4+ T cells can circumvent immune privilege and infiltrate intraocular Ad5E1 tumours. Following tumour rejection, CD4+ T cells from tumour rejector mice could be adoptively transferred to severe combined immunodeficiency (SCID) mice and protect them from intraocular Ad5E1 tumour growth. Tumour-specific CD4+ T cells produced IFN-gamma in response to Ad5E1 tumour antigens. Macrophages also contributed to rejection, as they were present in intraocular Ad5E1 tumours, and local depletion of macrophages resulted in progressive tumour growth. Ocular macrophages contributed to Ad5E1 tumour rejection, as Ad5E1 tumour rejection did not occur in macrophage-depleted SCID mice reconstituted with rejector CD4+ T cells. This demonstrates that macrophage and CD4+ T-cell co-operation is needed for non-phthisical rejection of intraocular tumours.
Collapse
Affiliation(s)
- Dru S Dace
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9057, USA
| | | | | |
Collapse
|
114
|
Abstract
Over the past 50 years, thalidomide has been a target of active investigation in both malignant and inflammatory conditions. Although initially developed for its sedative properties, decades of investigation have identified a multitude of biological effects that led to its classification as an immunomodulatory drug (IMiD). In addition to suppression of tumor necrosis factor-alpha (TNF-alpha), thalidomide effects the generation and elaboration of a cascade of pro-inflammatory cytokines that activate cytotoxic T-cells even in the absence of co-stimulatory signals. Furthermore, vascular endothelial growth factor (VEGF) and beta fibroblast growth factor (bFGF) secretion and cellular response are suppressed by thalidomide, thus antagonizing neoangiogenesis and altering the bone marrow stromal microenvironment in hematologic malignancies. The thalidomide analogs, lenalidomide (CC-5013; Revlimid) and CC-4047 (Actimid), have enhanced potency as inhibitors of TNF-alpha and other inflammatory cytokines, as well as greater capacity to promote T-cell activation and suppress angiogenesis. Both thalidomide and lenalidomide are effective in the treatment of multiple myeloma and myelodysplastic syndromes for which the Food and Drug Administration granted recent approval. Nonetheless, each of these IMiDs remains the subject of active investigation in solid tumors, hematologic malignancies, and other inflammatory conditions. This review will explore the pharmacokinetic and biologic effects of thalidomide and its progeny compounds.
Collapse
Affiliation(s)
- Magda Melchert
- Moffitt Cancer Center and Research Institute, 12902 Magnolia Drive, Tampa, FL 33612, USA.
| | | |
Collapse
|
115
|
Rudge G, Barrett SP, Scott B, van Driel IR. Infiltration of a mesothelioma by IFN-gamma-producing cells and tumor rejection after depletion of regulatory T cells. THE JOURNAL OF IMMUNOLOGY 2007; 178:4089-96. [PMID: 17371963 DOI: 10.4049/jimmunol.178.7.4089] [Citation(s) in RCA: 31] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/31/2022]
Abstract
Depletion of CD4+CD25+Foxp3+ regulatory T cells (CD25+ T(reg)) with an anti-CD25 Ab results in immune-mediated rejection of tolerogenic solid tumors. In this study, we have examined the immune response to a mesothelioma tumor in mice after depletion of CD25+ cells to elucidate the cellular mechanisms of CD25+ T(reg), a subject over which there is currently much conjecture. Tumor rejection was found to be primarily due to the action of CD8+ T cells, although CD4+ cells appeared to play some role. Depletion of CD25+ cells resulted in an accumulation in tumor tissue of CD4+ and CD8+ T cells and NK cells that were producing the potent antitumor cytokine IFN-gamma. Invasion of tumors by CD8+ T cells was partially dependent on the presence of CD4+ T cells. Although a significant increase in the proliferation and number of tumor-specific CD8+ T cells was observed in lymph nodes draining the tumor of anti-CD25-treated mice, this effect was relatively modest compared with the large increase in IFN-gamma-producing T cells found in tumor tissue, which suggests that the migration of T cells into tumor tissue may also have been altered. Depletion of CD25+ cells did not appear to modulate antitumor CTL activity on a per cell basis. Our data suggests that CD25+ T(reg) limit the accumulation of activated T cells producing IFN-gamma in the tumor tissue and, to a lesser extent, activation and/or rate of mitosis of tumor-specific T cells in lymph nodes.
Collapse
Affiliation(s)
- Geordie Rudge
- Department of Biochemistry and Molecular Biology, Bio21 Molecular Science and Biotechnology Institute, University of Melbourne, Parkville, Australia
| | | | | | | |
Collapse
|
116
|
Teramoto K, Kontani K, Fujita T, Ozaki Y, Sawai S, Tezuka N, Fujino S, Itoh Y, Taguchi O, Kannagi R, Ogasawara K. Successful tumor eradication was achieved by collaboration of augmented cytotoxic activity and anti-angiogenic effects following therapeutic vaccines containing helper-activating analog-loaded dendritic cells and tumor antigen DNA. Cancer Immunol Immunother 2007; 56:331-42. [PMID: 16896967 PMCID: PMC11031089 DOI: 10.1007/s00262-006-0192-0] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/05/2006] [Accepted: 06/07/2006] [Indexed: 11/30/2022]
Abstract
We reported previously that pigeon cytochrome c-derived peptides (Pan-IA), which bind broad ranges of MHC class II molecules efficiently, activate T helper (Th) function in mice. In an experimental model, Pan-IA DNA vaccines augmented antitumor immunity in tumor antigen-immunized mice. To elicit more potent antitumor immunity and to eradicate tumors in a therapeutic setting, Pan-IA-loaded dendritic cells (DCs) were inoculated in combination with vaccines including ovalbumin (OVA) antigen DNA in tumor-bearing mice. Seventy percent of the immunized mice survived tumor-free for at least 4 months after treatment. In contrast, mice vaccinated with OVA DNA, either with or without naïve DCs, did not eliminate the tumors and died within 5 weeks. Only in mice vaccinated with OVA DNA and Pan-IA-loaded DCs were both cytotoxic and helper responses specific for OVA induced at the spleen and tumor sites as well as at the vaccination sites. Furthermore, accumulation of OVA-specific CD4(+) and CD8(+) T lymphocytes and interferon-gamma-mediated anti-angiogenesis were observed in the tumors of these mice. Thus, the combined vaccination primed both tumor-specific cytotoxicity and helper immunity resulting in augmented tumor lysis ability and anti-angiogenic effects. This is the first report to show that most established tumors were successfully eradicated by collaboration of potent antitumor immunity and anti-angiogenic effects by vaccination with tumor antigens and helper-activating analogs. This novel vaccination strategy is broadly applicable, regardless of identifying helper epitopes in target molecules, and contributes to the development of therapeutic cancer vaccines.
Collapse
Affiliation(s)
- Koji Teramoto
- Department of Surgery, Shiga University of Medical Science, Seta-tsukinowa, Otsu, 520-2192 Japan
| | - Keiichi Kontani
- Second Department of Surgery, Kagawa University Faculty of Medicine, 1750-1 Miki-cho, Kita-gun, 761-0793 Japan
| | - Takuya Fujita
- Department of Surgery, Shiga University of Medical Science, Seta-tsukinowa, Otsu, 520-2192 Japan
| | - Yoshitomo Ozaki
- Department of Surgery, Shiga University of Medical Science, Seta-tsukinowa, Otsu, 520-2192 Japan
| | - Satoru Sawai
- Department of Surgery, Shiga University of Medical Science, Seta-tsukinowa, Otsu, 520-2192 Japan
| | - Noriaki Tezuka
- Department of Surgery, Shiga University of Medical Science, Seta-tsukinowa, Otsu, 520-2192 Japan
| | - Shozo Fujino
- Department of Surgery, Shiga University of Medical Science, Seta-tsukinowa, Otsu, 520-2192 Japan
| | - Yasushi Itoh
- Department of Pathology, Shiga University of Medical Science, Seta-tsukinowa, Otsu, 520-2192 Japan
| | - Osamu Taguchi
- Division of Molecular Pathology, Aichi Cancer Center Research Institute, 1-1 Kanokoden, Chikusa, Nagoya, 464-8681 Japan
| | - Reiji Kannagi
- Division of Molecular Pathology, Aichi Cancer Center Research Institute, 1-1 Kanokoden, Chikusa, Nagoya, 464-8681 Japan
| | - Kazumasa Ogasawara
- Department of Pathology, Shiga University of Medical Science, Seta-tsukinowa, Otsu, 520-2192 Japan
| |
Collapse
|
117
|
Minuzzo S, Moserle L, Indraccolo S, Amadori A. Angiogenesis meets immunology: Cytokine gene therapy of cancer. Mol Aspects Med 2007; 28:59-86. [PMID: 17306360 DOI: 10.1016/j.mam.2006.12.008] [Citation(s) in RCA: 15] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/29/2006] [Accepted: 12/29/2006] [Indexed: 01/19/2023]
Abstract
Delivery of cytokine genes at the tumor site in pre-clinical models has been shown to recruit host inflammatory cells followed by inhibition of tumor growth. This local effect is often accompanied by systemic protection mediated by the immune system, mainly by CD8(+) T and NK cells. On this basis, cytokine gene-transduced tumor cells have widely been used as vaccines in clinical trials, which have shown good safety profiles and some local responses but substantial lack of systemic efficacy. Are these findings the end of the story? Possibly not, if major improvements will be attained in the coming years. These should be directed at the level of gene selection and delivery, in order to identify the optimal cytokine and achieve efficient and durable cytokine expression, and at the level of improving immune stimulation, i.e. by co-administration of co-stimulatory molecules including B7 and CD40, or boosting the expression of tumor antigens or MHC class I molecules. Interestingly, some of the cytokines which have shown encouraging anti-tumor activity, including IFNs, IL-4, IL-12 and TNF-alpha, are endowed with anti-angiogenic or vasculotoxic effects, which may significantly contribute to local tumor control. Therapeutic exploitation of this property may result in the design of novel approaches which, by maximizing immune-stimulating and anti-angiogenic effects, could possibly lead to starvation of established tumors in patients.
Collapse
Affiliation(s)
- Sonia Minuzzo
- Department of Oncology and Surgical Sciences, University of Padova, via Gattamelata 64, 35128 Padova, Italy
| | | | | | | |
Collapse
|
118
|
Abstract
A clear picture of the dynamic relationship between the host immune system and cancer is emerging as the cells and molecules that participate in naturally occurring antitumour immune responses are being identified. The interferons (IFNs) - that is, the type I IFNs (IFNalpha and IFNbeta) and type II IFN (IFNgamma) - have emerged as central coordinators of tumour-immune-system interactions. Indeed, the decade-old finding that IFNgamma has a pivotal role in promoting antitumour responses became the focus for a renewed interest in the largely abandoned concept of cancer immunosurveillance. More recently, type I IFNs have been found to have distinct functions in this process. In this Review, we discuss the roles of the IFNs, not only in cancer immunosurveillance but also in the broader process of cancer immunoediting.
Collapse
Affiliation(s)
- Gavin P Dunn
- Washington University School of Medicine, Department of Pathology and Immunology, Box 8118, 660 South Euclid Avenue, St. Louis, Missouri 63110, USA
| | | | | |
Collapse
|
119
|
Dace DS, Chen PW, Alizadeh H, Niederkorn JY. Ocular immune privilege is circumvented by CD4+ T cells, leading to the rejection of intraocular tumors in an IFN-{gamma}-dependent manner. J Leukoc Biol 2006; 81:421-9. [PMID: 17077163 DOI: 10.1189/jlb.0806489] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/30/2022] Open
Abstract
Although intraocular tumors reside in an immune-privileged site, they can circumvent immune privilege and undergo rejection, which typically follows one of two pathways. One pathway involves CD4(+) T cells, delayed-type hypersensitivity (DTH), and the culmination in ischemic necrosis of the tumor and phthisis (atrophy) of the eye. The second pathway is DTH-independent and does not inflict collateral injury to ocular tissues, and the eye is preserved. In this study, we used a well-characterized tumor, Ad5E1, to analyze the role of IFN-gamma in the nonphthisical form of intraocular tumor rejection. The results showed that IFN-gamma induced tumor cell apoptosis, inhibited tumor cell proliferation, and promoted rejection by inhibiting angiogenesis. Microarray analysis revealed that IFN-gamma induced up-regulation of five antiangiogenic genes and down-regulation of four proangiogenic genes in Ad5E1 tumor cells. Although IFN-gamma knockout (KO) mice have progressively growing intraocular tumors, IFN-gamma was not needed for the elimination of extraocular tumors, as all IFN-gamma KO mice rejected s.c. tumor inocula. This represents a heretofore unrecognized role for IFN-gamma in circumventing ocular immune privilege and eliminating intraocular tumors. The findings also reveal that some IFN-gamma-independent tumor rejection processes are excluded from the eye and may represent a new facet of ocular immune privilege.
Collapse
Affiliation(s)
- Dru S Dace
- Department of Ophthalmology, University of Texas Southwestern Medical Center, Dallas, TX 75390-9057, USA
| | | | | | | |
Collapse
|
120
|
Dirkx AEM, Oude Egbrink MGA, Wagstaff J, Griffioen AW. Monocyte/macrophage infiltration in tumors: modulators of angiogenesis. J Leukoc Biol 2006; 80:1183-96. [PMID: 16997855 DOI: 10.1189/jlb.0905495] [Citation(s) in RCA: 261] [Impact Index Per Article: 13.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/13/2022] Open
Abstract
The role of a tumor immune infiltrate in cancer progression and metastasis has been debated frequently. Although often considered to be associated with improved prognosis and leading to the enhanced survival of cancer patients, inflammatory cells have also been described to assist the tumor's capabilities to progress, proliferate, and metastasize. Tumor-associated macrophages (TAMs), for example, have been shown to be symbiotically related to tumor cells: Tumor cells recruit TAMs and provide them with survival factors, and TAMs in turn produce a variety of angiogenic factors in response to the tumor microenvironment. This review will describe the composition of an immune infiltrate in tumors and the angiogenic and angiostatic properties of the cells present. Special emphasis will be on the angiogenesis-associated activities of TAMs. The development of immunotherapy and gene therapy using TAMs to mediate tumor cytotoxicity or to deliver gene constructs will be discussed as well. As immunotherapy has so far not been as effective as anticipated, a combination therapy in which angiostatic agents are used as well is put forward as a novel strategy to treat cancer.
Collapse
Affiliation(s)
- Anita E M Dirkx
- Department of Pathology, University Hospital Maastricht, P.O. Box 5800, 6202 AZ Maastricht, The Netherlands
| | | | | | | |
Collapse
|
121
|
Jarnicki AG, Lysaght J, Todryk S, Mills KHG. Suppression of Antitumor Immunity by IL-10 and TGF-β-Producing T Cells Infiltrating the Growing Tumor: Influence of Tumor Environment on the Induction of CD4+ and CD8+ Regulatory T Cells. THE JOURNAL OF IMMUNOLOGY 2006; 177:896-904. [DOI: 10.4049/jimmunol.177.2.896] [Citation(s) in RCA: 312] [Impact Index Per Article: 16.4] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/08/2023]
|
122
|
Wolff JEA, Wagner S, Reinert C, Gnekow A, Kortmann RD, Kühl J, Van Gool SW. Maintenance treatment with interferon-gamma and low-dose cyclophosphamide for pediatric high-grade glioma. J Neurooncol 2006; 79:315-21. [PMID: 16645718 DOI: 10.1007/s11060-006-9147-8] [Citation(s) in RCA: 60] [Impact Index Per Article: 3.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/05/2005] [Accepted: 03/13/2006] [Indexed: 11/29/2022]
Abstract
BACKGROUND The prognosis of high-grade glioma in children is poor. PURPOSE Interferon-gamma may increase the immune surveillance of glioma cells. Earlier clinical evidence had shown that low dose cyclophosphamide (CPM) increased immune response. METHODS After induction treatment with simultaneous radiation and chemotherapy, patients were treated with individually increasing interferon-gamma (IFN-gamma) doses starting from 25 microg/m2/d s.c. increasing up to a maximum of 175 microg/m2/d within 7 weeks. Cyclophosphamide was given at 300 mg/m2 i.v. every 21 days. Forty pediatric glioma patients were enrolled (median age: 8.5 year, male: n = 22). Tumor locations included cerebral cortex (n = 8), basal ganglia (n = 4), brainstem (n = 24), cerebellum (n = 3), spinal cord (n = 1). Histologies were GBM (n = 14), AA (n = 14), LGG (n = 2, diffuse intrinsic pontine glioma). There was grade IV toxicity for thrombocytopenia (10%) and leucopenia (2.5%), grade III toxicity for central nervous (2.5%) and hepatic (5%) side effects, no toxic death. The observation time of the six surviving patients was: 1.2, 1.9, 4.2, 4.4, 4.6 and 4.7 years respectively. The median overall survival (1 year) was not significantly different from a historical control group (0.8 years). The survival of pontine gliomas appeared even inferior when compared to the previous protocol (n.s.). CONCLUSION Maintenance treatment with IFN-gamma and low dose CPM has no sufficient beneficial effect for the treatment of high-grade glioma.
Collapse
Affiliation(s)
- Johannes E A Wolff
- Department of Pediatrics, MD Anderson Cancer Center, Unit 87, University of Texas, 1515 Holcombe Blvd, Houston, TX 77030, USA.
| | | | | | | | | | | | | |
Collapse
|
123
|
Abstract
The prognostic significance of tumor-infiltrating lymphocytes (TILs) has been a longstanding topic of debate. In cases where TILs have improved patient outcome, T lymphocytes are recognized as the main effectors of antitumor immune responses. However, recent studies have revealed that a subset of CD4(+) T cells, referred to as CD4(+)CD25(+) regulatory T cells (Treg), may accumulate in the tumor environment and suppress tumor-specific T-cell responses, thereby hindering tumor rejection. Hence, predicting tumor behavior on the basis of an indiscriminate evaluation of tumor-infiltrating T cells may result in inconsistent prognostic accuracy. The presence of infiltrating CD4(+)CD25(+) Treg may be detrimental to the host defense against the tumor, while the presence of effector T lymphocytes, including CD8(+) T cells and non-regulatory CD4(+) helper T cells may be beneficial. Enhanced recruitment of antitumor effector T lymphocytes to tumor tissue in addition to inhibition of local Treg, may therefore be an ideal target for improving cancer immunotherapy. This article reviews the antitumor functions of T-lymphocytes, with special attention given to CD4(+) regulatory T-cells within the tumor environment.
Collapse
Affiliation(s)
- Ping Yu
- 1Department of Pathology and Committee on Immunology, University of Chicago, Chicago, IL 60637, USA.
| | | |
Collapse
|
124
|
Vincenzi B, Santini D, Russo A, Silletta M, Gavasci M, Battistoni F, Di Cuonzo G, Rocci L, Gebbia N, Tonini G. Angiogenesis modifications related with cetuximab plus irinotecan as anticancer treatment in advanced colorectal cancer patients. Ann Oncol 2006; 17:835-41. [PMID: 16507564 DOI: 10.1093/annonc/mdl031] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/20/2022] Open
Abstract
INTRODUCTION Angiogenesis has been correlated with increased invasion and metastases in a variety of human neoplasms. Inadequate inhibition of the growth of tumor microvessels by anticancer agents may result in treatment failure, rated clinically as progressive or stable disease. We designed this trial to investigate the modification of the vascular endothelial growth factor (VEGF) and interferon-gamma (IFN-gamma) in advanced colorectal cancer patients during treatment with a weekly combination of cetuximab plus irinotecan. MATERIALS AND METHODS Forty-five metastatic colorectal cancer patients were prospectively evaluated for circulating levels of VEGF and IFN-gamma during the treatment with cetuximab (initial dose of 400 mg/m(2), followed by weekly infusions of 250 mg/m(2)) plus weekly irinotecan (90 mg/m(2)). The circulating levels of the cytokines were assessed at the following time points: just before and at 1, 21, 50 and 92 days after the start of cetuximab plus irinotecan treatment. RESULTS Basal serum VEGF median levels were significantly decreased just at the first day (after the first treatment infusion (P = 0.016). The VEGF persisted at the following time points reaching the highest statistical significance 92 days after the first infusion (P < 0.0001). On the contrary, IFN-gamma values showed a statistical significant increase one day after the first infusion (P < 0.0001). This effect persisted 21 days after the treatment start (P = 0.001), but was no more evident at the following time points. Moreover, a linear regression model with variance analysis showed a significant negative correlation between VEGF and IFN-gamma values 1, 21 and 50 days after the treatment beginning (P = 0.002, 0.001 and 0.047, respectively). CONCLUSIONS This study suggests that a cetuximab may induce a modulation of VEGF circulating levels. The reduction of VEGF serum levels is a sudden and long lasting phenomenon. Moreover, in our study we identified a IFN-gamma increase, even if the specific role of this behavior remains to be investigated.
Collapse
Affiliation(s)
- B Vincenzi
- Medical Oncology, University Campus Bio-Medico, Rome, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
125
|
Blohm U, Potthoff D, van der Kogel AJ, Pircher H. Solid tumors “melt” from the inside after successful CD8 T cell attack. Eur J Immunol 2006; 36:468-77. [PMID: 16385625 DOI: 10.1002/eji.200526175] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
Adoptive transfer of tumor-specific T cells represents a promising approach for cancer immunotherapy. Here, we visualized the anti-tumor response of CD8 T cells from P14 TCR-transgenic mice specific for the model antigen GP33 by immunohistology. P14 T cells, adoptively transferred into tumor-bearing hosts, induced regression of established 3LL-A9(GP33) and MCA102(GP33) tumors that express GP33 as a tumor-associated model antigen. Strikingly, the visible effects of P14 T cell attack, such as the destruction of the tumor vasculature and accumulation of granulocytes, were predominantly detected inside the tumor mass. In regressing tumors, P14 T cells were found in the intact rim zone but not in central areas that were infiltrated with granulocytes and lacked CD31(+) endothelial cells. The rim of P14 T cell-treated tumors showed an increase in vascular density and decrease in hypoxia compared to untreated tumors. Hypoxic areas of tumors are known to exhibit decreased sensitivity to radiation therapy or chemotherapy. Thus, our data also imply that adoptive transfer of tumor-specific CD8 T cells might synergize with radiation therapy or chemotherapy in the elimination of solid tumors in vivo.
Collapse
MESH Headings
- Adoptive Transfer/methods
- Animals
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Antigens, Viral/genetics
- Antigens, Viral/immunology
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/pathology
- CD8-Positive T-Lymphocytes/transplantation
- Endothelial Cells/immunology
- Endothelial Cells/pathology
- Granulocytes/immunology
- Granulocytes/pathology
- Hypoxia/immunology
- Hypoxia/pathology
- Lymphocytic choriomeningitis virus/genetics
- Lymphocytic choriomeningitis virus/immunology
- Mice
- Mice, Transgenic
- Neoplasm Transplantation
- Neoplasms, Experimental/blood supply
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/pathology
- Neoplasms, Experimental/therapy
- Neovascularization, Pathologic/immunology
- Neovascularization, Pathologic/pathology
- Neovascularization, Pathologic/therapy
- Neutrophil Infiltration/genetics
- Neutrophil Infiltration/immunology
- Platelet Endothelial Cell Adhesion Molecule-1/immunology
- Radiotherapy
Collapse
Affiliation(s)
- Ulrike Blohm
- Institute of Medical Microbiology and Hygiene, Department of Immunology, University of Freiburg, Germany
| | | | | | | |
Collapse
|
126
|
Lee H, Baek S, Joe SJ, Pyo SN. Modulation of IFN-γ production by TNF-α in macrophages from the tumor environment: Significance as an angiogenic switch. Int Immunopharmacol 2006; 6:71-8. [PMID: 16332515 DOI: 10.1016/j.intimp.2005.08.010] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/07/2004] [Revised: 07/23/2005] [Accepted: 08/01/2005] [Indexed: 11/25/2022]
Abstract
BACKGROUND The role of macrophages in tumor angiogenesis has been known to influence in the production of angiogenic cytokines and growth factors including TNF-alpha. Recently, macrophages were also found to produce INF-gamma, which were found to be involved in angiogenic inhibition. Thus, the importance of macrophages in tumor angiogenesis might be the angiogenic switch. The hypothesis tested here is that TNF-alpha can modulate the INF-gamma production in macrophages in tumor environment as part of the tumor angiogenic switch. METHODS Macrophages in tumor environment were obtained from peritoneal cavity and s.c. grown tumor of C57BL/6 mice injected with B16F10 melanoma cell line for 6 and 11 days, respectively. Mac1+-macrophages were purified using magnetic beads (MACs; Milteny Biotech, Germany) and cultured with various concentrations of TNF-alpha at various time points at 37 degrees C. The supernatants were analyzed for IFN-gamma or VEGF by ELISA kit. RESULTS Residential macrophages from peritoneal cavity did not respond to LPS or TNF-alpha to produce INF-gamma. However, the cells from tumor environment produced IFN-gamma as well as VEGF. Upregulation of IFN-gamma production by the addition of LPS or TNF-alpha was observed in macrophages from the tumor bearing peritoneal cavity. RT-PCR analysis revealed external TNF-alpha-induced IFN-gamma gene expression in macrophages from tumor environment. CONCLUSION The overall data suggest that the macrophages in tumor environment might play an important role not only in angiogenic signal but also in anti-angiogenic signal by producing related cytokines. Moreover, TNF-alpha might be a key cytokine functioning as a tumor angiogenic switch.
Collapse
MESH Headings
- Animals
- Base Sequence
- Cell Line, Tumor
- Female
- In Vitro Techniques
- Interferon-gamma/biosynthesis
- Interferon-gamma/genetics
- Lipopolysaccharides/pharmacology
- Macrophages, Peritoneal/drug effects
- Macrophages, Peritoneal/immunology
- Melanoma, Experimental/blood supply
- Melanoma, Experimental/immunology
- Mice
- Mice, Inbred C57BL
- Neovascularization, Pathologic/etiology
- Neovascularization, Pathologic/immunology
- RNA, Messenger/genetics
- RNA, Messenger/metabolism
- RNA, Neoplasm/genetics
- RNA, Neoplasm/metabolism
- Signal Transduction
- Tumor Necrosis Factor-alpha/biosynthesis
- Tumor Necrosis Factor-alpha/pharmacology
- Vascular Endothelial Growth Factor A/biosynthesis
Collapse
Affiliation(s)
- Hyunah Lee
- The Cancer Center, Samsung Medical Center, School of Medicine, Sungkyunkwan University, 50 IL-Won Dong, Kang-Nam Gu, 135-710, Seoul, Korea.
| | | | | | | |
Collapse
|
127
|
Ferretti G, Fabi A, Carlini P, Papaldo P, Cordiali Fei P, Di Cosimo S, Salesi N, Giannarelli D, Alimonti A, Di Cocco B, D'Agosto G, Bordignon V, Trento E, Cognetti F. Zoledronic-acid-induced circulating level modifications of angiogenic factors, metalloproteinases and proinflammatory cytokines in metastatic breast cancer patients. Oncology 2005; 69:35-43. [PMID: 16088233 DOI: 10.1159/000087286] [Citation(s) in RCA: 103] [Impact Index Per Article: 5.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/18/2004] [Accepted: 01/10/2005] [Indexed: 12/29/2022]
Abstract
BACKGROUND To evaluate the modifications of circulating angiogenic factors, metalloproteinases and acute-phase cytokines after the first single zoledronic acid (ZA) intravenous infusion. EXPERIMENTAL DESIGN Eighteen consecutive breast cancer patients with bone metastases were evaluated for circulating levels of vascular endothelial growth factor (VEGF), basic fibroblast growth factor (bFGF), metalloproteinase 1 (MMP-1), metalloproteinase 2 (MMP-2), interleukins 1beta, 6 and 8 (IL-1beta, IL-6, IL-8), interferon gamma, tumor necrosis factor alpha (TNF-alpha) and transforming growth factor beta1 just before and 2 and 7 days after ZA infusion. RESULTS The MMP-2 basal value showed a statistically significant decrease 48 h after ZA (p = 0.01), being at 7 days higher than the day 2 value (p = 0.03). The VEGF basal value showed a statistically significant decrease 48 h after ZA infusion (p = 0.03), increasing above the basal level at 7 days (p = 0.07). The bFGF basal level almost significantly decreased 2 days after infusion (p = 0.06), being at 7 days higher than the basal value (p = 0.09). Comparing the day 2 values with basal ones, the linear regression model showed a significant positive correlation between IL-8 and bFGF (p = 0.02), IL-8 and TNF-alpha (p < 0.0001), bFGF and TNF-alpha (p = 0.01), MMP-1 and TNF-alpha (p = 0.02). CONCLUSIONS ZA could exert an antiangiogenic activity and inhibition of tumor cell bone invasiveness by a transient reduction of VEGF, bFGF and MMP-2 circulating levels after infusion.
Collapse
Affiliation(s)
- Gianluigi Ferretti
- Division of Medical Oncology A, Regina Elena Cancer Institute, Rome, Italy.
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
128
|
Dominiecki ME, Beatty GL, Pan ZK, Neeson P, Paterson Y. Tumor sensitivity to IFN-gamma is required for successful antigen-specific immunotherapy of a transplantable mouse tumor model for HPV-transformed tumors. Cancer Immunol Immunother 2005; 54:477-88. [PMID: 15750832 PMCID: PMC11032979 DOI: 10.1007/s00262-004-0610-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/03/2003] [Accepted: 08/04/2004] [Indexed: 11/24/2022]
Abstract
PURPOSE Many human tumors lose responsiveness to IFN-gamma, providing a possible mechanism for the tumor to avoid immune recognition and destruction. Here we investigate the importance of tumor responsiveness to IFN-gamma in the successful immunotherapy of TC1 tumors that were immortalized with human papillomavirus proteins E6 and E7. METHODS To investigate the role of IFN-gamma in vivo, we constructed a variant of TC1, TC1.mugR, that is unresponsive to IFN-gamma due to overexpression of a dominant negative IFN-gamma receptor. RESULTS Using recombinant Listeria monocytogenes that express HPV-16 E7 (Lm-LLO-E7) to stimulate an antitumor response, we demonstrate that sensitivity to IFN-gamma is required for therapeutic efficacy in that Lm-LLO-E7 induces regression of TC1 tumors but not TC1.mugR. In addition, we show that tumor sensitivity to IFN-gamma is not required for inhibition of tumor angiogenesis by Lm-LLO-E7 or for trafficking of CD4+ and CD8+ T cells to the tumor. However, it is required for penetration of lymphocytes into the tumor mass in vivo. CONCLUSIONS Our findings identify a role for IFN-gamma in immunity to TC1 tumors and show that loss of tumor responsiveness to IFN-gamma poses a challenge to antigen-based immunotherapy.
Collapse
MESH Headings
- Animals
- Antigens, Neoplasm/genetics
- Antigens, Neoplasm/immunology
- Bacterial Vaccines/therapeutic use
- Cancer Vaccines/immunology
- Cancer Vaccines/therapeutic use
- Cell Line, Tumor
- Cell Transformation, Viral
- Disease Models, Animal
- Female
- Interferon-gamma/physiology
- Listeria monocytogenes/genetics
- Listeria monocytogenes/immunology
- Lymphocytes, Tumor-Infiltrating/immunology
- Mice
- Mice, Inbred C57BL
- Neoplasm Transplantation
- Neoplasms, Experimental/drug therapy
- Neoplasms, Experimental/genetics
- Neoplasms, Experimental/immunology
- Oncogene Proteins, Viral/genetics
- Oncogene Proteins, Viral/immunology
- Papillomaviridae/genetics
- Papillomavirus E7 Proteins
- Papillomavirus Infections/drug therapy
- Papillomavirus Infections/genetics
- Papillomavirus Infections/immunology
- Receptors, Interferon/genetics
- Receptors, Interferon/metabolism
- T-Lymphocytes, Cytotoxic/immunology
- Transcriptional Activation
- Tumor Escape/immunology
- Uterine Cervical Neoplasms/virology
Collapse
Affiliation(s)
- Mary E. Dominiecki
- Department of Microbiology, University of Pennsylvania, 323 Johnson Pavilion, 3610 Hamilton Walk, Philadelphia, PA 19104-6076 USA
| | - Gregory L. Beatty
- Department of Microbiology, University of Pennsylvania, 323 Johnson Pavilion, 3610 Hamilton Walk, Philadelphia, PA 19104-6076 USA
| | - Zhen-Kun Pan
- Department of Microbiology, University of Pennsylvania, 323 Johnson Pavilion, 3610 Hamilton Walk, Philadelphia, PA 19104-6076 USA
| | - Paul Neeson
- Department of Microbiology, University of Pennsylvania, 323 Johnson Pavilion, 3610 Hamilton Walk, Philadelphia, PA 19104-6076 USA
| | - Yvonne Paterson
- Department of Microbiology, University of Pennsylvania, 323 Johnson Pavilion, 3610 Hamilton Walk, Philadelphia, PA 19104-6076 USA
| |
Collapse
|
129
|
Makar VR, Logani MK, Bhanushali A, Kataoka M, Ziskin MC. Effect of millimeter waves on natural killer cell activation. Bioelectromagnetics 2005; 26:10-9. [PMID: 15605409 DOI: 10.1002/bem.20046] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023]
Abstract
Millimeter wave therapy (MMWT) is being widely used for the treatment of many diseases in Russia and other East European countries. MMWT has been reported to reduce the toxic effects of chemotherapy on the immune system. The present study was undertaken to investigate whether millimeter waves (MMWs) can modulate the effect of cyclophosphamide (CPA), an anticancer drug, on natural killer (NK) cell activity. NK cells play an important role in the antitumor response. MMWs were produced with a Russian-made YAV-1 generator. The device produced modulated 42.2 +/- 0.2 GHz radiation through a 10 x 20 mm rectangular output horn. Mice, restrained in plastic tubes, were irradiated on the nasal area. Peak SAR at the skin surface and peak incident power density were measured as 622 +/- 100 W/kg and 31 +/- 5 mW/cm2, respectively. The maximum temperature elevation, measured at the end of 30 min, was 1 degrees C. The animals, restrained in plastic tubes, were irradiated on the nasal area. CPA injection (100 mg/kg) was given intraperitoneally on the second day of 3-days exposure to MMWs. All the irradiation procedures were performed in a blinded manner. NK cell activation and cytotoxicity were measured after 2, 5, and 7 days following CPA injection. Flow cytometry of NK cells showed that CPA treatment caused a marked enhancement in NK cell activation. The level of CD69 expression, which represents a functional triggering molecule on activated NK cells, was increased in the CPA group at all the time points tested as compared to untreated mice. However, the most enhancement in CD69 expression was observed on day 7. A significant increase in TNF-alpha level was also observed on day 7 following CPA administration. On the other hand, CPA caused a suppression of the cytolytic activity of NK cells. MMW irradiation of the CPA treated groups resulted in further enhancement of CD69 expression on NK cells, as well as in production of TNF-alpha. Furthermore, MMW irradiation restored CPA induced suppression of the cytolytic activity of NK cells. Our results show that MMW irradiation at 42.2 GHz can up-regulate NK cell functions.
Collapse
Affiliation(s)
- V R Makar
- Richard J Fox Center for Biomedical Physics, Temple University School of Medicine, Philadelphia, PA 19140, USA
| | | | | | | | | |
Collapse
|
130
|
Siracusano S, Niccolini B, Knez R, Tiberio A, Benedetti E, Bonin S, Ciciliato S, Pappagallo GL, Belgrano E, Stanta G. The Simultaneous Use of Telomerase, Cytokeratin 20 and CD4 for Bladder Cancer Detection in Urine. Eur Urol 2005; 47:327-33. [PMID: 15716196 DOI: 10.1016/j.eururo.2004.10.007] [Citation(s) in RCA: 33] [Impact Index Per Article: 1.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/08/2004] [Accepted: 10/08/2004] [Indexed: 11/20/2022]
Abstract
OBJECTIVE Because of the low sensitivity of urinary cytological diagnosis of urinary bladder carcinoma, new molecular diagnostic methods have been proposed. We decided to verify the expression of telomerase mRNA coding for the catalytic component (hTRT), cytokeratin 20 (CK20) and CD4 antigen mRNAs in urine as possible diagnostic tool. METHODS Evaluation of hTRT, CK20, CD4 mRNAs was performed in 50 ml of naturally voided urine of 205 patients of which 153 with bladder cancer (Tis, n = 11; TaGx, n = 4; TaG1, n = 25; TaG2, n = 26; TaG3, n = 8; T1G1, n = 16; T1G2, n = 17; T1G3, n = 20; T2G2, n = 6; T2G3, n = 13; T3G3, n = 7) and 52 controls. A quantitative expression of hTRT at mRNA level versus TRAP (telomeric repeat amplification protocol) assay was performed in 20 patients and 14 controls. The expression of RT-PCR for hTRT, CK20, CD4 versus urinary cytology was analysed in 44 patients with bladder cancer. Evaluating the three molecular markers together, the result was considered correct when at least two of the markers were positive, suspected when only one marker was positive and negative for diagnosis of tumour when all markers were negative. The performance of the diagnostic model resulted from the logistic analysis evaluated with receiver operating characteristics (ROC) curve analysis. RESULTS The sensitivity detected for each tumour marker was as follows: for hTRT 90.8%, for CK20 84.3% and for CD4 was 64.7%, while the specificity was 94.2% for CD4 and 78.8% for both hTRT and CK20. When a simultaneous evaluation of the three tumour markers was considered, 88.2% of the diagnoses were correct, 11.8% were suspected for tumour and none were mistaken. When compared with cytology, the simultaneous use of the three markers allowed reaching a correct diagnosis in 88% of the cases in comparison to 25% by urinary cytology. The sensitivity in the detection of bladder cancer was higher for hTRT at mRNA level in comparison with the enzymatic activity detection with TRAP (90% vs. 35%) while the specificity for both markers resulted very high (100%). CONCLUSIONS These data show that in the future the diagnostic improvement of urine based molecular markers for the detection of bladder cancer in the urine could improve the sensitivity of urinary cytology reducing the need of a cystoscopy.
Collapse
Affiliation(s)
- Salvatore Siracusano
- Department of Urology, Trieste University, Cattinara Hospital, Via Strada di Fiume 447, 34100 Trieste, Italy.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
131
|
Caras I, Grigorescu A, Stavaru C, Radu DL, Mogos I, Szegli G, Salageanu A. Evidence for immune defects in breast and lung cancer patients. Cancer Immunol Immunother 2004; 53:1146-52. [PMID: 15185014 PMCID: PMC11034324 DOI: 10.1007/s00262-004-0556-2] [Citation(s) in RCA: 79] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/19/2004] [Accepted: 04/23/2004] [Indexed: 10/26/2022]
Abstract
Immunosuppression is often identified in cancer patients. The aim of this study was to evaluate several immune parameters for patients with breast and lung cancer. Immunophenotyping analysis showed that the cancer patients investigated had significantly lower absolute numbers of peripheral blood lymphocytes than controls. The immunosuppression was more evident for the breast cancer subgroup. The most severe immune defect noticed was the marked impairment of IFN-gamma secretion. A shift toward the Th2 phenotype as revealed by assessment of intracellular level of IFN-gamma and IL-4 was also noticed. The secretion of proinflammatory cytokines IL-1beta and TNF-alpha in whole blood cultures was not impaired. Although the proportion of activated cells was slightly lower than in the control group, our results showed that both peripheral T lymphocytes and NK cells of cancer patients could be induced to express early activation marker CD69 after ex vivo mitogen stimulation. In conclusion, our study revealed several immune defects in cancer patients. This suggests that an appropriate immunotherapeutical approach might be used to restore compromised immune functions with beneficial effects on both antitumor and general immunity.
Collapse
Affiliation(s)
- Iuliana Caras
- Department of Immunology, National Institute for Research and Development in Microbiology and Immunology “Cantacuzino”, Bucharest, Romania
| | - A. Grigorescu
- Oncology Institute “Prof. Dr. Alex. Trestioreanu”, Bucharest, Romania
| | - Crina Stavaru
- Department of Immunology, National Institute for Research and Development in Microbiology and Immunology “Cantacuzino”, Bucharest, Romania
| | - D. L. Radu
- Department of Immunology, National Institute for Research and Development in Microbiology and Immunology “Cantacuzino”, Bucharest, Romania
| | - I. Mogos
- Oncology Institute “Prof. Dr. Alex. Trestioreanu”, Bucharest, Romania
| | - G. Szegli
- Department of Immunology, National Institute for Research and Development in Microbiology and Immunology “Cantacuzino”, Bucharest, Romania
| | - Aurora Salageanu
- Department of Immunology, National Institute for Research and Development in Microbiology and Immunology “Cantacuzino”, Bucharest, Romania
- Immunomodulators Group, Department of Immunology, Cantacuzino Institute, 103 Spl. Independentei, PO 050096, Bucharest, Romania
| |
Collapse
|
132
|
Mitra-Kaushik S, Harding J, Hess J, Schreiber R, Ratner L. Enhanced tumorigenesis in HTLV-1 tax-transgenic mice deficient in interferon-gamma. Blood 2004; 104:3305-11. [PMID: 15292059 DOI: 10.1182/blood-2004-01-0266] [Citation(s) in RCA: 44] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
The oncoprotein Tax of human T-cell leukemia virus type I (HTLV-1) is the major mediator of viral pathogenesis in infected individuals. Expression of Tax under the regulation of the human granzyme B promoter in mice results in a lymphoproliferative disorder resembling adult T-cell leukemia/lymphoma (ATL). Tax expression is associated with the production of high levels interferon-gamma (IFN-gamma) in HTLV-1-infected CD4(+) cells and Tax-transgenic tumors. We examined the role of IFN-gamma in tumorigenesis, by mating Tax-transgenic mice with a gene-specific knockout for IFN-gamma. IFN-gamma(-/-) Tax(+)-transgenic mice show accelerated tumor onset (median, 4 versus 6 months), dissemination (median, 5 versus 7 months), and death (median, 7 versus 10 months), compared with IFN-gamma(+/-) or IFN-gamma(+/+) Tax(+) mice. Pathologic and immunophenotypic characteristics of tumors from all genotypes are indistinguishable, except for enhanced interleukin 2 receptor-beta (IL-2Rbeta) and suppressed intercellular adhesion molecule-1 (ICAM-1) expression on tumors from IFN-gamma(-/-) Tax(+) transgenic mice. IFN-gamma(-/-) tumors demonstrate enhanced CD31 (platelet-endothelial CAM-1 [PECAM-1]) staining compared with those from IFN-gamma(+/-) or IFN-gamma(+/+) Tax(+) mice. Angiogenesis-specific cDNA microarray analysis identified 4 mediators of angiogenic growth differentially expressed in tumors from Tax(+)IFN-gamma(-/-) mice compared with Tax(+)IFN-gamma(+/+) littermates. As confirmed by reverse transcription-polymerase chain reaction (RT-PCR), loss of IFN-gamma results in down-regulation of tumor necrosis factor-alpha (TNF-alpha) and tissue inhibitor of matrix metalloproteinase-1 (TIMP-1) while up-regulating expression of vascular endothelial growth factor (VEGF) and tenascin C. These results provide insight into a possible mechanism by which IFN-gamma contributes to host resistance against HTLV-induced tumors through an angiostatic effect.
Collapse
Affiliation(s)
- Shibani Mitra-Kaushik
- Department of Internal Medicine, Washington University School of Medicine, 660 S Euclid Ave, Box 8069, St Louis, MO 63110, USA
| | | | | | | | | |
Collapse
|
133
|
Yang I, Kremen TJ, Giovannone AJ, Paik E, Odesa SK, Prins RM, Liau LM. Modulation of major histocompatibility complex Class I molecules and major histocompatibility complex—bound immunogenic peptides induced by interferon-α and interferon-γ treatment of human glioblastoma multiforme. J Neurosurg 2004; 100:310-9. [PMID: 15086239 DOI: 10.3171/jns.2004.100.2.0310] [Citation(s) in RCA: 65] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022]
Abstract
OBJECT Little is known about the quantitative modulation of major histocompatibility complex (MHC) Class I expression on human gliomas that is effected by interferons; even less is known about the immunogenic peptides that are accommodated in the peptide-binding motifs of MHC Class I alleles in these brain tumors. In this article the authors investigated the ability of interferon (IFN)alpha and IFNgamma to upregulate MHC Class I expression and to modulate acid-eluted Class I-bound peptides on human glioblastoma multiforme (GBM) cells in vitro. METHODS Early-passage primary human GBM cell cultures and U87MG GBM cells were incubated with varying doses of INFalpha or IFNgamma ranging between 0 and 2000 U/ml. Upregulation of MHC Class I expression was assayed by immunocytochemical analysis, flow cytometry, and Western blot analysis. Modulation of acid-eluted MHC Class I-bound peptides from the IFN-treated GBM cells was examined with the aid of mass spectroscopy. The in vitro expression of the MHC Class I molecule was upregulated by both IFNalpha and IFNgamma in a dose-dependent manner. Interferon-gamma exhibited a more potent effect on MHC Class I upregulation, peaking at 10 U/ml; whereas the effect of IFNalpha was less marked, reaching a plateau at 500 U/ml. In addition, a native peptide eluted from MHC Class I molecules of human GBM cells was identified and found to be consistently upregulated by IFN treatment. CONCLUSIONS Interferon-alpha and IFN-gamma can significantly upregulate the MHC Class I molecules that are expressed on the cell surface of human GBM cells as well as the potentially immunogenic peptides bound to the MHC. These results may help explain the molecular basis for increased immunogenicity with IFN treatment of human GBMs and might provide added insight into the design of future antitumor vaccines for human brain tumors.
Collapse
Affiliation(s)
- Isaac Yang
- UCLA Division of Neurosurgery, University of California at Los Angeles School of Medicine, Los Angeles, California 90095-6901, USA
| | | | | | | | | | | | | |
Collapse
|
134
|
Xiao BG, Huang YM, Link H. Dendritic cell vaccine design: strategies for eliciting peripheral tolerance as therapy of autoimmune diseases. BioDrugs 2004; 17:103-11. [PMID: 12641489 DOI: 10.2165/00063030-200317020-00003] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/02/2022]
Abstract
Dendritic cells (DC), as potent antigen-presenting cells (APC), constitute a complex system of cells that initiate and regulate immune responses that result in two opposite outcomes: immunity or tolerance. The fine regulation of these two distinct functions is not completely understood. After loading with antigen, DC exhibit the properties of both antigen and adjuvant, the functional components of vaccines. For a long time, attention has focused on the exceptional ability of DC as professional APC capable of eliciting T and B cell-mediated responses, and on their potential as immunotherapy in cancer. DC exhibit both heterogeneity and plasticity. On the one hand, distinct DC subsets exhibit distinct functions. On the other hand, DC functions can be altered by the cytokine environment or other factors. There is increasing evidence that DC could be used as a tool to induce peripheral tolerance. Because DC-based immunotherapy in autoimmune diseases depends on tolerogenic DC, discerning markers for tolerogenic DC is of great importance. Immature DC, plasmacytoid DC and interleukin-10-modified DC can mediate immune tolerance by inducing T-cell anergy or T-helper type 2 responses. Several possibilities exist for rational modulation of DC to achieve therapeutic tolerance against autoimmune diseases. The final goal is to create optimal prerequisites to use autologous DC that are prepared from the individual patient with autoimmune disease, to render such DC tolerogenic by exposure in vitro to factors that promote tolerogenicity, and to re-infuse these pretreated DC to the patient in order to treat the ongoing autoimmune disease and prevent its future exacerbation.
Collapse
Affiliation(s)
- Bao-Guo Xiao
- Experimental Neurology and Neuroimmunology Units, Department of Neurotec, Karolinska Institute, Huddinge University Hospital, SE-141 86 Stockholm, Sweden.
| | | | | |
Collapse
|
135
|
Nakakubo Y, Miyamoto M, Cho Y, Hida Y, Oshikiri T, Suzuoki M, Hiraoka K, Itoh T, Kondo S, Katoh H. Clinical significance of immune cell infiltration within gallbladder cancer. Br J Cancer 2003; 89:1736-42. [PMID: 14583778 PMCID: PMC2394404 DOI: 10.1038/sj.bjc.6601331] [Citation(s) in RCA: 76] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022] Open
Abstract
To investigate the pathophysiological significance of infiltrating antitumour immune cells, we evaluated the quantity of immune cell intratumoral infiltration in 110 surgically resected gallbladder specimens by immunohistochemistry. We examined 45 cases of gallbladder cancer and 65 cases of benign gallbladder diseases for CD4+ T cells, CD8+ T cells, natural killer cells (NKCs), and dendritic cells (DCs). High levels of CD4+ T cell, CD8+ T cell, NKC, and DC infiltration were recognised in 51.1% (23 out of 45), 37.8% (17 out of 45), 33.3% (15 out of 45), and 48.9% (22 out of 45) of cancer specimens, respectively. High numbers of infiltrating CD4+ and CD8+ T cells correlated with decreasing tumour invasion, and high numbers of infiltrating DCs correlated with decreasing lymph-node tumour metastasis. Furthermore, increased infiltration of CD4+ and CD8+ T cells and DCs exhibited a significant correlation with prolonged survival. NKC infiltration, however, did not correlate with any of the clinicopathological factors examined. Additionally, high levels of infiltration were not identified in specimens from benign diseases, consistent with the cancer-specific activity of CD4+ and CD8+ T cells and DCs. In this study, we demonstrate that CD4+ and CD8+ tumour-infiltrating lymphocyte and DCs, but not NKCs, are important factors in the accurate prognosis of survival after surgical removal of gallbladder adenocarcinoma.
Collapse
Affiliation(s)
- Y Nakakubo
- Department of Surgical Oncology, Division of Cancer Medicine, Hokkaido University, Graduate School of Medicine, N-15 W-7, Kita-ku, Sapporo 060-8638, Japan
| | - M Miyamoto
- Department of Surgical Oncology, Division of Cancer Medicine, Hokkaido University, Graduate School of Medicine, N-15 W-7, Kita-ku, Sapporo 060-8638, Japan
- Department of Surgical Oncology, Division of Cancer Medicine, Hokkaido University, Graduate School of Medicine, N-15 W-7, Kita-ku, Sapporo 060-8638, Japan. E-mail:
| | - Y Cho
- Department of Surgical Oncology, Division of Cancer Medicine, Hokkaido University, Graduate School of Medicine, N-15 W-7, Kita-ku, Sapporo 060-8638, Japan
| | - Y Hida
- Department of Surgical Oncology, Division of Cancer Medicine, Hokkaido University, Graduate School of Medicine, N-15 W-7, Kita-ku, Sapporo 060-8638, Japan
| | - T Oshikiri
- Department of Surgical Oncology, Division of Cancer Medicine, Hokkaido University, Graduate School of Medicine, N-15 W-7, Kita-ku, Sapporo 060-8638, Japan
| | - M Suzuoki
- Department of Surgical Oncology, Division of Cancer Medicine, Hokkaido University, Graduate School of Medicine, N-15 W-7, Kita-ku, Sapporo 060-8638, Japan
| | - K Hiraoka
- Department of Surgical Oncology, Division of Cancer Medicine, Hokkaido University, Graduate School of Medicine, N-15 W-7, Kita-ku, Sapporo 060-8638, Japan
| | - T Itoh
- Department of Pathology, Hokkaido University Hospital, N-14 W-5, Kita-ku, Sapporo 060-8648, Japan
| | - S Kondo
- Department of Surgical Oncology, Division of Cancer Medicine, Hokkaido University, Graduate School of Medicine, N-15 W-7, Kita-ku, Sapporo 060-8638, Japan
| | - H Katoh
- Department of Surgical Oncology, Division of Cancer Medicine, Hokkaido University, Graduate School of Medicine, N-15 W-7, Kita-ku, Sapporo 060-8638, Japan
| |
Collapse
|
136
|
Kowalczyk DW, Wlazlo AP, Giles-Davis W, Kammer AR, Mukhopadhyay S, Ertl HCJ. Vaccine-induced CD8+ T cells eliminate tumors by a two-staged attack. Cancer Gene Ther 2003; 10:870-8. [PMID: 14712313 DOI: 10.1038/sj.cgt.7700653] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
Data presented here demonstrate that vaccine-induced CD8(+) T cells can eliminate their specific tumor-target with a two-staged attack. First, they release interferon-gamma that results in growth arrest of the tumor cells via induction of antiangiogenic mediators. Then, during the latter stages of the immune response, CD8(+) effector T cells eradicate the remaining tumor cells through perforin-mediated lysis. A combination of these two mechanisms is highly effective in the described model, while either pathway alone fails to completely achieve tumor rejection.
Collapse
Affiliation(s)
- Dariusz W Kowalczyk
- Department of Cancer Immunology, Greatpoland Cancer Center, Garbary 15, 61-866 Poznan, Poland
| | | | | | | | | | | |
Collapse
|
137
|
Casares N, Arribillaga L, Sarobe P, Dotor J, Lopez-Diaz de Cerio A, Melero I, Prieto J, Borrás-Cuesta F, Lasarte JJ. CD4+/CD25+ Regulatory Cells Inhibit Activation of Tumor-Primed CD4+ T Cells with IFN-γ-Dependent Antiangiogenic Activity, as well as Long-Lasting Tumor Immunity Elicited by Peptide Vaccination. THE JOURNAL OF IMMUNOLOGY 2003; 171:5931-9. [PMID: 14634104 DOI: 10.4049/jimmunol.171.11.5931] [Citation(s) in RCA: 153] [Impact Index Per Article: 7.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
CD25(+) regulatory T (T reg) cells suppress the activation/proliferation of other CD4(+) or CD8(+) T cells in vitro. Also, down-regulation of CD25(+) T reg cells enhance antitumor immune responses. In this study, we show that depletion of CD25(+) T reg cells allows the host to induce both CD4(+) and CD8(+) antitumoral responses following tumor challenge. Simultaneous depletion of CD25(+) and CD8(+) cells, as well as adoptive transfer experiments, revealed that tumor-specific CD4(+) T cells, which emerged in the absence of CD25(+) T reg cells, were able to reject CT26 colon cancer cells, a MHC class II-negative tumor. The antitumoral effect mediated by CD4(+) T cells was dependent on IFN-gamma production, which exerted a potent antiangiogenic activity. The capacity of the host to mount this antitumor response is lost once the number of CD25(+) T reg cells is restored over time. However, CD25(+) T reg cell depletion before immunization with AH1 (a cytotoxic T cell determinant from CT26 tumor cells) permits the induction of a long-lasting antitumoral immune response, not observed if immunization is conducted in the presence of regulatory cells. A study of the effect of different levels of depletion of CD25(+) T reg cells before immunization with the peptide AH1 alone, or in combination with a Th determinant, unraveled that Th cells play an important role in overcoming the suppressive effect of CD25(+) T reg on the induction of long-lasting cellular immune responses.
Collapse
Affiliation(s)
- Noelia Casares
- Department of Internal Medicine, Medical School, University Clinic and Fundación para la Investigación Médica Aplicada, University of Navarra, Pamplona, Spain
| | | | | | | | | | | | | | | | | |
Collapse
|
138
|
Makar V, Logani M, Szabo I, Ziskin M. Effect of millimeter waves on cyclophosphamide induced suppression of T cell functions. Bioelectromagnetics 2003; 24:356-65. [PMID: 12820293 DOI: 10.1002/bem.10106] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/07/2022]
Abstract
The effects of low power electromagnetic millimeter waves (MWs) on T cell activation, proliferation, and effector functions were studied in BALB/c mice. These functions are important in T-lymphocyte mediated immune responses. The MW exposure characteristics were: frequency = 42.2 GHz; peak incident power density = 31 +/- 5 mW/cm(2), peak specific absorption rate (SAR) at the skin surface = 622 +/- 100 W/kg; duration 30 min daily for 3 days. MW treatment was applied to the nasal area. The mice were additionally treated with cyclophosphamide (CPA), 100 mg/kg, a commonly used immunosuppressant and anticancer drug. Four groups of animals were used in each experiment: naive control (Naive), CPA treated (CPA), CPA treated and sham exposed (CPA + Sham), and CPA treated and MW exposed (CPA + MW). MW irradiation of CPA treated mice significantly augmented the proliferation recovery process of T cells (splenocytes). A statistically significant difference (P <.05) between CPA and CPA + MW groups was observed when cells were stimulated with an antigen. On the other hand, no statistically significant difference between CPA and CPA-Sham groups was observed. Based on flow cytometry of CD4(+) and CD8(+) T cells, two major classes of T cells, we show that CD4(+) T cells play an important role in the proliferation recovery process. MW exposure restored the CD25 surface activation marker expression in CD4(+) T cells. We next examined the effector function of purified CD4(+) T cells by measuring their cytokine profile. No changes were observed after MW irradiation in interleukin-10 (IL-10) level, a Th2 type cytokine, while the level of interferon-gamma (IFN-gamma), a Th1 type cytokine was increased twofold. Our results indicate that MWs enhance the effector function of CD4(+) T cells preferentially, through initiating a Th1 type of immune response. This was further supported by our observation of a significant enhancement of tumor necrosis factor-alpha (TNF-alpha) production by peritoneal macrophage's in CPA treated mice. The present study shows MWs ameliorate the immunosuppressive effects of CPA by augmenting the proliferation of splenocytes, and altering the activation and effector functions of CD4(+) T cells.
Collapse
Affiliation(s)
- Vera Makar
- Richard J. Fox Center for Biomedical Physics, Temple University School of Medicine, Philadelphia, Pennsylvania 19140, USA
| | | | | | | |
Collapse
|
139
|
Flynn S, Stockinger B. Tumor and CD4 T-cell interactions: tumor escape as result of reciprocal inactivation. Blood 2003; 101:4472-8. [PMID: 12543861 DOI: 10.1182/blood-2002-10-3030] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/07/2023] Open
Abstract
This paper addresses the capacity of naive, effector, and memory CD4 T cells to control growth of a major histocompatibility complex (MHC) class II-positive B-cell lymphoma in vivo. To assess the role of T cells on their own without contributions by B cells, antibodies, or natural killer (NK) cells, we generated pure effector or memory CD4 T cells in Rag-/-gc-/- mice deficient in endogenous lymphocytes and NK cells. Lymphoma cells expressing a model antigen were injected into mice with T cells of cognate specificity that were either naive or in effector or resting memory state. Naive T cells were unable to prevent tumor growth, probably due to delay of efficient cross-presentation by dendritic cells. However, both effector and memory T cells, dependent on the amount of antigen available, controlled the tumor for a considerable period of time without the need for dendritic cell stimulation. Nevertheless, the tumor eventually grew uncontrolled in all cases. This was not because of a defect in T-cell homing to the tumor site or loss of MHC class II or costimulatory molecules by the tumor, but reflected mutual paralysis of T-cell responsiveness and antigen processing by tumor cells.
Collapse
Affiliation(s)
- Sarah Flynn
- Division of Molecular Immunology, National Institute for Medical Research, The Ridgeway, Mill Hill, London United Kingdom.
| | | |
Collapse
|
140
|
Abstract
CD4+ T cells have been shown to be able to affect tumor growth through both direct and indirect means. In addition, a requirement has been demonstrated for CD4+ T cells in the regulation and induction of T cell memory, and CD4+ suppressor T cells have been identified, stressing a role for CD4+ T cells in the induction and maintenance of antitumor immune responses. A review of the involvement of CD4+ T cells at different stages of tumor immunity is provided, and based on these data we discuss how CD4+ T cell response induction could be incorporated into tumor immunotherapy strategies.
Collapse
Affiliation(s)
- Markwin P Velders
- Cancer Immunology Program, Cardinal Bernardin Cancer Center, Loyola University Chicago, Maywood, Illinois 60153, USA
| | | | | | | |
Collapse
|
141
|
Abstract
The role of infectious agents in the development of cancer is well documented. The pathogenesis of various human neoplasms ranging from non-Hodgkin lymphoma (NHL) to cervical carcinoma frequently involves a chronic, most often viral, infection. At the same time, there is compelling evidence that certain acute infections result in the inhibition of neoplastic growth. The basis for this phenomenon is often thought to be concomitant anti-tumor immunity. Yet, experimental data supporting this hypothesis are scarce, and other non-immune anti-tumor factors could be involved. For instance, since virtually all aggressive tumors outstrip their blood supply, development of new vessels, or angiogenesis, is a limiting factor during neoplastic growth. In this review, we will discuss recent studies that implicate anti-angiogenesis in infection-mediated tumor suppression and suggest that this mechanism could also complement cytotoxic immunity arising from the use of cancer vaccines.
Collapse
Affiliation(s)
- Andrei Thomas-Tikhonenko
- Department of Pathobiology, University of Pennsylvania, 3800 Spruce Street, Philadelphia 19104-6051, USA.
| | | |
Collapse
|
142
|
Harada T, Miura NN, Adachi Y, Nakajima M, Yadomae T, Ohno N. IFN-gamma induction by SCG, 1,3-beta-D-glucan from Sparassis crispa, in DBA/2 mice in vitro. J Interferon Cytokine Res 2002; 22:1227-39. [PMID: 12581496 DOI: 10.1089/10799900260475759] [Citation(s) in RCA: 51] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/13/2022] Open
Abstract
Sparassis crispa Fr. in an edible mushroom recently cultivable in Japan. A branched beta-glucan from S. crispa (SCG) is a major 6-branched 1,3-beta-D-glucan showing antitumor activity. In this study, we examined interferon-gamma (IFN-gamma) induction by SCG from splenocytes in DBA/2 mice in vitro. In the splenocytes derived from almost all inbred strains of mice except for DBA/1 and DBA/2 mice, IFN-gamma production was not induced by SCG. The breeder and genders of DBA/2 mice showed no influence on IFN-gamma induction by SCG. On the other hand, the magnitude of IFN-gamma induction was lower in young mice than in their older counterparts. IFN-gamma was induced by SCG in adherent splenocytes, but IFN-gamma production was most significantly increased by SCG in instances involving coexistence of adherent and nonadherent splenocytes. In fact, inhibition of cell-cell contact reduced IFN-gamma induction by SCG. In addition, interleukin-12 p70 (IL-12p70) was induced by SCG in DBA/2 mice. It was suggested that soluble factors and cell-cell contact mediate synergistic effects on SCG-induced IFN-gamma production.
Collapse
Affiliation(s)
- Toshie Harada
- Laboratory for Immunopharmacology of Microbial Products, School of Pharmacy, Tokyo University of Pharmacy and Life Science, 1432-1 Horinouchi, Hachioji, Tokyo 192-0392, Japan
| | | | | | | | | | | |
Collapse
|
143
|
Haynes NM, Trapani JA, Teng MWL, Jackson JT, Cerruti L, Jane SM, Kershaw MH, Smyth MJ, Darcy PK. Rejection of syngeneic colon carcinoma by CTLs expressing single-chain antibody receptors codelivering CD28 costimulation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:5780-6. [PMID: 12421958 DOI: 10.4049/jimmunol.169.10.5780] [Citation(s) in RCA: 80] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
A new strategy to improve the therapeutic utility of redirected T cells for cancer involves the development of novel Ag-specific chimeric receptors capable of stimulating optimal and sustained T cell antitumor activity in vivo. Given that T cells require both primary and costimulatory signals for optimal activation and that many tumors do not express critical costimulatory ligands, modified single-chain Ab receptors have been engineered to codeliver CD28 costimulation. In this study, we have compared the antitumor potency of primary T lymphocytes expressing carcinoembryonic Ag (CEA)-reactive chimeric receptors that incorporate either TCR-zeta or CD28/TCR-zeta signaling. Although both receptor-transduced T cell effector populations demonstrated cytolysis of CEA(+) tumors in vitro, T cells expressing the single-chain variable fragment of Ig (scFv)-CD28-zeta chimera had a far greater capacity to control the growth of CEA(+) xenogeneic and syngeneic colon carcinomas in vivo. The observed enhanced antitumor activity of T cells expressing the scFv-CD28-zeta receptor was critically dependent on perforin and the production of IFN-gamma. Overall, this study has illustrated the ability of a chimeric scFv receptor capable of harnessing the signaling machinery of both TCR-zeta and CD28 to augment T cell immunity against tumors that have lost expression of both MHC/peptide and costimulatory ligands in vivo.
Collapse
MESH Headings
- Adenocarcinoma/genetics
- Adenocarcinoma/immunology
- Adenocarcinoma/prevention & control
- Adjuvants, Immunologic/administration & dosage
- Adjuvants, Immunologic/biosynthesis
- Adjuvants, Immunologic/genetics
- Animals
- Binding Sites, Antibody/genetics
- CD28 Antigens/administration & dosage
- CD28 Antigens/genetics
- CD28 Antigens/immunology
- Carcinoembryonic Antigen/administration & dosage
- Carcinoembryonic Antigen/biosynthesis
- Carcinoembryonic Antigen/genetics
- Carcinoembryonic Antigen/immunology
- Cells, Cultured
- Colonic Neoplasms/genetics
- Colonic Neoplasms/immunology
- Colonic Neoplasms/pathology
- Colonic Neoplasms/prevention & control
- Cytotoxicity, Immunologic/genetics
- Epitopes, T-Lymphocyte/genetics
- Epitopes, T-Lymphocyte/metabolism
- Graft Rejection/genetics
- Graft Rejection/immunology
- Growth Inhibitors/administration & dosage
- Growth Inhibitors/biosynthesis
- Growth Inhibitors/genetics
- Humans
- Immunoglobulin Variable Region/administration & dosage
- Immunoglobulin Variable Region/genetics
- Immunoglobulin Variable Region/immunology
- Immunotherapy, Adoptive/methods
- Interferon-gamma/deficiency
- Interferon-gamma/genetics
- Interferon-gamma/physiology
- Membrane Glycoproteins/deficiency
- Membrane Glycoproteins/genetics
- Membrane Glycoproteins/physiology
- Membrane Proteins/genetics
- Membrane Proteins/immunology
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Mice, SCID
- Perforin
- Pore Forming Cytotoxic Proteins
- Receptors, Antigen, T-Cell/genetics
- Receptors, Antigen, T-Cell/immunology
- Receptors, Immunologic/administration & dosage
- Receptors, Immunologic/biosynthesis
- Receptors, Immunologic/genetics
- Recombinant Fusion Proteins/administration & dosage
- Recombinant Fusion Proteins/biosynthesis
- Recombinant Fusion Proteins/genetics
- T-Lymphocytes, Cytotoxic/immunology
- T-Lymphocytes, Cytotoxic/metabolism
- T-Lymphocytes, Cytotoxic/transplantation
- Transplantation, Isogeneic
- Tumor Cells, Cultured
Collapse
Affiliation(s)
- Nicole M Haynes
- Cancer Immunology Program, Sir Donald and Lady Trescowthick Laboratories, Peter MacCallum Cancer Institute, St. Andrew's Place, East Melbourne, Victoria, Australia
| | | | | | | | | | | | | | | | | |
Collapse
|
144
|
Hural JA, Friedman RS, McNabb A, Steen SS, Henderson RA, Kalos M. Identification of naturally processed CD4 T cell epitopes from the prostate-specific antigen kallikrein 4 using peptide-based in vitro stimulation. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 169:557-65. [PMID: 12077288 DOI: 10.4049/jimmunol.169.1.557] [Citation(s) in RCA: 27] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/23/2023]
Abstract
Kallikrein (KLK)4 is a recently described member of the tissue kallikrein gene family that is specifically expressed in normal and prostate tumor tissues. The tissue-specific expression profile of this molecule suggests that it might be useful as a vaccine candidate against prostate cancer. To examine the presence of CD4 T cells specific for KLK4 in PBMC of normal individuals, a peptide-based in vitro stimulation protocol was developed that uses overlapping KLK4-derived peptides spanning the majority of the KLK4 protein. Using this methodology, three naturally processed CD4 epitopes derived from the KLK4 sequence are identified. These epitopes are restricted by HLA-DRB1*0404, HLA-DRB1*0701, and HLA-DPB1*0401 class II alleles. CD4 T cell clones specific for these epitopes are shown to efficiently and specifically recognize both recombinant KLK4 protein and lysates from prostate tumor cell lines virally infected to express KLK4. CD4 T cells specific for these KLK4 epitopes are shown to exist in PBMC from multiple male donors that express the relevant class II alleles, indicating that a CD4 T cell repertoire specific for KLK4 is present and potentially expandable in prostate cancer patients. The demonstration that KLK4-specific CD4 T cells exist in the peripheral circulation of normal male donors and the identification of naturally processed KLK4-derived CD4 T cell epitopes support the use of KLK4 in whole gene-, protein-, or peptide-based vaccine strategies against prostate cancer. Furthermore, the identification of naturally processed KLK4-derived epitopes provides valuable tools for monitoring preexisting and vaccine-induced responses to this molecule.
Collapse
Affiliation(s)
- John A Hural
- Corixa Corporation and Infectious Disease Research Institute, Seattle, WA 98104, USA
| | | | | | | | | | | |
Collapse
|
145
|
Dredge K, Marriott JB, Todryk SM, Muller GW, Chen R, Stirling DI, Dalgleish AG. Protective antitumor immunity induced by a costimulatory thalidomide analog in conjunction with whole tumor cell vaccination is mediated by increased Th1-type immunity. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2002; 168:4914-9. [PMID: 11994441 DOI: 10.4049/jimmunol.168.10.4914] [Citation(s) in RCA: 126] [Impact Index Per Article: 5.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
Thalidomide and its novel T cell costimulatory analogs (immunomodulatory drugs) are currently being assessed in the treatment of patients with advanced cancer. However, neither tumor-specific T cell costimulation nor effective antitumor activity has been demonstrated in vivo. In this study, we assessed the ability of an immunomodulatory drug (CC-4047/ACTIMID) to prime a tumor-specific immune response following tumor cell vaccination. We found that the presence of CC-4047 during the priming phase strongly enhanced antitumor immunity in the vaccinated group, and this correlated with protection from subsequent live tumor challenge. Protection was associated with tumor-specific production of IFN-gamma and was still observed following a second challenge with live tumor cells 60 days later. Furthermore, CD8(+) and CD4(+) splenocyte fractions from treated groups secreted increased IFN-gamma and IL-2 in response to tumor cells in vitro. Coculture of naive splenocytes with anti-CD3 mAb in the presence of CC-4047 directly costimulated T cells and increased Th1-type cytokines. Our results are the first to demonstrate that a costimulatory thalidomide analog can prime protective, long-lasting, tumor-specific, Th1-type responses in vivo and further support their ongoing clinical development as novel anti-cancer agents.
Collapse
MESH Headings
- Adjuvants, Immunologic/administration & dosage
- Animals
- Antigens, CD/biosynthesis
- Antineoplastic Combined Chemotherapy Protocols/immunology
- Antineoplastic Combined Chemotherapy Protocols/therapeutic use
- B7-1 Antigen/biosynthesis
- B7-2 Antigen
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/immunology
- Cells, Cultured
- Coculture Techniques
- Cytokines/biosynthesis
- Growth Inhibitors/administration & dosage
- Growth Inhibitors/immunology
- Histocompatibility Antigens Class I/biosynthesis
- Injections, Intraperitoneal
- Injections, Subcutaneous
- Interferon-gamma/biosynthesis
- Melanoma, Experimental
- Membrane Glycoproteins/biosynthesis
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Nude
- Neoplasm Transplantation/immunology
- Spleen/cytology
- Spleen/immunology
- Spleen/metabolism
- T-Lymphocyte Subsets/immunology
- Th1 Cells/immunology
- Thalidomide/administration & dosage
- Thalidomide/analogs & derivatives
- Tumor Cells, Cultured/immunology
- Tumor Cells, Cultured/metabolism
- Tumor Cells, Cultured/pathology
- Tumor Cells, Cultured/transplantation
Collapse
Affiliation(s)
- Keith Dredge
- Division of Oncology, Department of Oncology, Gastroenterology, Endocrinology, and Metabolism, St. George's Hospital Medical School, Cranmer Terrace, Tooting, London, United Kingdom SW17 0RE.
| | | | | | | | | | | | | |
Collapse
|
146
|
Abstract
Neoplastic cells form only one part of a complex network of cell types that make up a breast tumor. The normal cell types that make up the nonneoplastic components of tumors include fibroblasts, endothelium, and inflammatory cells, such as tumor associated macrophages (TAMs). TAMs have the potential to carry out both anti- and protumor activities In their antitumor role TAMs can present tumor antigens to cytotoxic T-cells and are capable of being directly cytotoxic to neoplastic cells. Conversely, TAMs are also able to promote tumor growth directly by secreting breast tumor mitogens, such as epidermal growth factor, and indirectly by stimulating tumor angiogenesis and metastasis. Recent studies have indicated that in breast cancers the protumor role of TAMs is dominant, and that TAMs may be executing a "wound healing" type of process in response to stimuli found in the tumor microenvironment, such as hypoxia. As such, TAMs may provide opportunities for future therapeutic interventions.
Collapse
Affiliation(s)
- Russell D Leek
- Cancer Research UK, Molecular Oncology Laboratories, University of Oxford, Weatherall Institute of Molecular Medicine, John Radcliffe Hospital, Headington, Oxford OX3 9DS, United Kingdom.
| | | |
Collapse
|
147
|
Abstract
BACKGROUND The growth of solid tumors and the formation of metastases depend on angiogenesis. Both tumor cells and host cells secrete a variety of factors to stimulate angiogenesis. METHODS We investigated the expression of angiogenic factors in gastric cancer in vitro and in vivo. RESULTS The expression of one of the angiogenic factors, vascular endothelial growth factor antigen, in gastric cancer cells can thus serve as a pertinent predictive factor for hematogenous invasion or metastasis, in addition to having prognostic value. The presence of micrometastasis in bone marrow was closely related to vascular endothelial growth factor positivity and microvessel density in the primary gastric cancer. In in vivo experiments antiangiogenic agents with cytotoxic anticancer drugs formed a highly effective modulator combination for the treatment of the Lewis lung carcinoma against primary and metastatic disease. CONCLUSIONS Antiangiogenic agents may thus be valuable for long-term administration to maintain tumor dormancy because drug resistance does not develop, and these agents have a sustained effect. As a target, antiangiogenic therapy may therefore be potentially able to prolong survival time of patients with gastric cancer.
Collapse
Affiliation(s)
- Yoshihiro Kakeji
- Department of Surgery and Science, Graduate School of Medical Sciences, Kyushu University, Fukuoka, Japan
| | | | | | | | | |
Collapse
|
148
|
Gunn GR, Zubair A, Peters C, Pan ZK, Wu TC, Paterson Y. Two Listeria monocytogenes vaccine vectors that express different molecular forms of human papilloma virus-16 (HPV-16) E7 induce qualitatively different T cell immunity that correlates with their ability to induce regression of established tumors immortalized by HPV-16. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2001; 167:6471-9. [PMID: 11714814 DOI: 10.4049/jimmunol.167.11.6471] [Citation(s) in RCA: 236] [Impact Index Per Article: 9.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/13/2023]
Abstract
Two recombinant Listeria monocytogenes (rLm) strains were produced that secrete the human papilloma virus-16 (HPV-16) E7 protein expressed in HPV-16-associated cervical cancer cells. One, Lm-E7, expresses and secretes E7 protein, whereas a second, Lm-LLO-E7, secretes E7 as a fusion protein joined to a nonhemolytic listeriolysin O (LLO). Lm-LLO-E7, but not Lm-E7, induces the regression of the E7-expressing tumor, TC-1, established in syngeneic C57BL/6 mice. Both recombinant E7-expressing rLm vaccines induce measurable anti-E7 CTL responses that stain positively for H-2D(b) E7 tetramers. Depletion of the CD8+ T cell subset before treatment abrogates the ability of Lm-LLO-E7 to impact on tumor growth. In addition, the rLm strains induce markedly different CD4+ T cell subsets. Depletion of the CD4+ T cell subset considerably reduces the ability of Lm-LLO-E7 to eliminate established TC-1 tumors. Surprisingly, the reverse is the case for Lm-E7, which becomes an effective anti-tumor immunotherapeutic in mice lacking this T cell subset. Ab-mediated depletion of TGF-beta and CD25+ cells improves the effectiveness of Lm-E7 treatment, suggesting that TGF-beta and CD25+ cells are in part responsible for this suppressive response. CD4+ T cells from mice immunized with Lm-E7 are capable of suppressing the ability of Lm-LLO-E7 to induce the regression of TC-1 when transferred to tumor-bearing mice. These studies demonstrate the complexity of L. monocytogenes-mediated tumor immunotherapy targeting the human tumor Ag, HPV-16 E7.
Collapse
MESH Headings
- Animals
- Antineoplastic Agents/immunology
- Bacterial Toxins
- Bacterial Vaccines/genetics
- Bacterial Vaccines/immunology
- CD4-Positive T-Lymphocytes/transplantation
- CD8-Positive T-Lymphocytes/immunology
- Cancer Vaccines/genetics
- Cancer Vaccines/immunology
- Cell Line, Transformed/immunology
- Cell Line, Transformed/pathology
- Cell Line, Transformed/virology
- Cytotoxicity, Immunologic/genetics
- Epitopes, T-Lymphocyte/immunology
- Gene Expression Regulation, Viral/immunology
- Genetic Vectors/chemical synthesis
- Genetic Vectors/immunology
- Heat-Shock Proteins/genetics
- Heat-Shock Proteins/immunology
- Hemolysin Proteins
- Humans
- Interferon-gamma/physiology
- Listeria monocytogenes/genetics
- Listeria monocytogenes/immunology
- Listeria monocytogenes/metabolism
- Lymphocyte Activation/genetics
- Mice
- Mice, Inbred C57BL
- Oncogene Proteins, Viral/biosynthesis
- Oncogene Proteins, Viral/genetics
- Oncogene Proteins, Viral/immunology
- Oncogene Proteins, Viral/metabolism
- Papillomaviridae/genetics
- Papillomaviridae/immunology
- Papillomavirus E7 Proteins
- Papillomavirus Vaccines
- Spleen/cytology
- Spleen/immunology
- T-Lymphocytes, Cytotoxic/immunology
- Transforming Growth Factor beta/immunology
- Transforming Growth Factor beta/metabolism
- Tumor Cells, Cultured/immunology
- Tumor Cells, Cultured/pathology
- Tumor Cells, Cultured/virology
- Vaccines, Synthetic/genetics
- Vaccines, Synthetic/immunology
Collapse
Affiliation(s)
- G R Gunn
- Department of Microbiology, University of Pennsylvania School of Medicine, Philadelphia, PA 19104, USA
| | | | | | | | | | | |
Collapse
|
149
|
Abstract
The interactions between cancer cells and their micro- and macroenvironment create a context that promotes tumour growth and protects it from immune attack. The functional association of cancer cells with their surrounding tissues forms a new 'organ' that changes as malignancy progresses. Investigation of this process might provide new insights into the mechanisms of tumorigenesis and could also lead to new therapeutic targets.
Collapse
Affiliation(s)
- M J Bissell
- Division of Life Sciences, Lawrence Berkeley National Laboratory, Berkeley, California 99720, USA.
| | | |
Collapse
|