151
|
Abstract
Adoptive transfer of antigen-specific T cells is a promising approach for preventing progressive viral infections in immunosuppressed hosts. By contrast, effective T-cell therapy of malignant disease has proven to be much more difficult to achieve. This, in part, reflects the difficulty of isolating high avidity T cells specific for tumor-associated antigens, many of which are self-antigens that have induced some level of tolerance in the host. Even when tumor-reactive T cells can be isolated, the ability of these cells to survive in vivo and traffic to tumor sites is often impaired. Additionally, most tumors employ multiple mechanisms to escape T-cell recognition, including interference in antigen presentation, secretion of inhibitory factors and recruitment of regulatory or immunosuppressive cells. The genetic modification of T cells prior to transfer provides a potential means to overcome many of these obstacles and enhance the efficacy of T-cell therapy. This review article discusses the rationale for genetic modification of T cells, the critical steps involved in gene transfer, and potential advantages and disadvantages of strategies that are now being examined to engineer improved effector T cells for the treatment of human infectious and malignant disease.
Collapse
Affiliation(s)
- Carolina Berger
- Fred Hutchinson Cancer Research Center, Program in Immunology, Seattle, WA 98109-1024, USA.
| | | | | | | |
Collapse
|
152
|
Hu W, Davis JJ, Zhu H, Dong F, Guo W, Ang J, Peng H, Guo ZS, Bartlett DL, Swisher SG, Fang B. Redirecting adaptive immunity against foreign antigens to tumors for cancer therapy. Cancer Biol Ther 2007; 6:1773-9. [PMID: 17986853 DOI: 10.4161/cbt.6.11.4855] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/19/2022] Open
Abstract
Immunotherapy for cancer is often limited by weak immunogenicity of tumor antigens. However, immune systems are usually strong and effective against foreign invading antigens. To test whether the destructive effect of adaptive immunity against foreign antigens can be redirected to tumors for cancer therapy, we immunized mice with adenovector expressing LacZ (Ad/CMV-LacZ). Subcutaneous syngeneic tumors were then established in the immunized animals or in naïve animals. The immune response against adenovirus or LacZ was redirected to tumors by intratumoral injection of Ad/CMV-LacZ. We found that immunization and treatment with the adenovector dramatically reduced the tumor growth rate compared with intratumoral administration of adenovector in naïve mice. Complete tumor regression was observed in about 50% of the immunized animals but not in the naïve animals. Similar effects were observed when oncolytic vaccinia virus was used to immunize and treat tumors. Lymphocyte infiltration in tumors was dramatically increased in the immunized group when compared with other groups. Moreover, immunity against parental tumor cells was induced in the animals cured with immunization and treatment with Ad/CMV-LacZ, as evidenced by the lack of tumor growth when the mice were challenged with parental tumor cells. Taken together, these results suggest that redirecting adaptive immunity against foreign antigens is a potential approach for anticancer therapy and that pre-existing immunity could enhance virotherapy against cancers.
Collapse
Affiliation(s)
- Wenxian Hu
- Department of Thoracic and Cardiovascular Surgery, The University of Texas MD Anderson Cancer Center, Houston, Texas 77030, USA
| | | | | | | | | | | | | | | | | | | | | |
Collapse
|
153
|
Ivanov VN, Zhou H, Hei TK. Sequential treatment by ionizing radiation and sodium arsenite dramatically accelerates TRAIL-mediated apoptosis of human melanoma cells. Cancer Res 2007; 67:5397-407. [PMID: 17545621 PMCID: PMC4378527 DOI: 10.1158/0008-5472.can-07-0551] [Citation(s) in RCA: 24] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Melanoma is the most lethal form of skin cancer. There is a lack of effective treatments for individuals with advanced disease. Many melanomas exhibit high levels of radioresistance. The direct consequence of gamma-irradiation for most melanoma cells is growth arrest at the G2-M phase of cell cycle. However, radiation-induced signaling pathways may affect numerous additional targets in cancer cells. We show in the present study that gamma-irradiation, as well as alpha-particle exposure, dramatically increases the susceptibility of melanoma cells to recombinant tumor necrosis factor-related apoptosis-inducing ligand (TRAIL)-mediated apoptosis via up-regulation of surface TRAIL-receptor 1/receptor 2 (DR4/DR5) levels and to Fas ligand-mediated apoptosis via up-regulation of surface Fas levels. Additionally, increased dynamin-2 expression after irradiation is critically important in the translocation of death receptor to the cell surface. Moreover, sodium arsenite treatment may up-regulate expression of endogenous TRAIL and induces its translocation to cell surface and further down-regulates cFLIP levels in melanoma cells. We have evaluated the effects of sequential gamma-irradiation and arsenite treatment of melanoma cells for the induction of death signaling. Such treatment results in an efficient TRAIL-mediated apoptosis via a paracrine mechanism. These data highlight the efficacy of combined modality treatment involving radiation and arsenite in clinical management of this often fatal form of skin cancer.
Collapse
Affiliation(s)
- Vladimir N Ivanov
- Center for Radiological Research, College of Physicians and Surgeons, Columbia University, New York, New York 10032, USA.
| | | | | |
Collapse
|
154
|
Fingleton B, Carter KJ, Matrisian LM. Loss of functional Fas ligand enhances intestinal tumorigenesis in the Min mouse model. Cancer Res 2007; 67:4800-6. [PMID: 17510409 DOI: 10.1158/0008-5472.can-06-4473] [Citation(s) in RCA: 20] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Fas ligand (FasL/CD95L), a member of the tumor necrosis factor family, interacts with a specific receptor Fas, ultimately leading to cell death. Tumor expression of FasL has been proposed to aid in immune evasion through a "Fas counterattack" mechanism but has also been described as a proinflammatory factor. Here, we tested the role of FasL in a mouse model of spontaneous tumor development. We used the Min mouse in which multiple benign polyps develop in the intestine due to a mutation in the Apc tumor suppressor gene. Mutant mice deficient in functional FasL, termed gld/gld, were crossed to Min mice to generate tumor-prone animals lacking functional FasL. Comparison of FasL-deficient versus proficient Min mice revealed a significant increase in polyp number in the gld/gld mice. We next assessed immune cell infiltration into adenomas. There was no difference in the number of either lymphocytes or macrophages; however, the number of tumor-infiltrating neutrophils was 3-fold lower in the gld/gld specimens compared with controls. Neutrophil migration in vitro was stimulated by wild-type but not mutant FasL. In a nontumor-bearing colitis model in vivo, neutrophil recruitment to the intestine was also reduced in gld/gld mice. Although the Fas counterattack hypothesis suggests that the absence of FasL would result in increased immune-mediated tumor elimination, the opposite is true in the Min model with lack of functional FasL associated with reduced neutrophil influx and increased tumor development. Thus, the proinflammatory rather than counterattack role of tumor FasL is more relevant.
Collapse
Affiliation(s)
- Barbara Fingleton
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA.
| | | | | |
Collapse
|
155
|
Hanson MGV, Özenci V, Carlsten MCV, Glimelius BL, Frödin JEA, Masucci G, Malmberg KJ, Kiessling RVR. A short-term dietary supplementation with high doses of vitamin E increases NK cell cytolytic activity in advanced colorectal cancer patients. Cancer Immunol Immunother 2007; 56:973-84. [PMID: 17143612 PMCID: PMC11030861 DOI: 10.1007/s00262-006-0261-4] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/12/2006] [Accepted: 11/14/2006] [Indexed: 01/22/2023]
Abstract
Cancer patients with advanced disease display signs of immune suppression, which constitute a major obstacle for effective immunotherapy. Both T cells and NK cells are affected by a multitude of mechanisms of which the generation of reactive oxygen species is of major importance. Therefore, we hypothesized that two weeks of high-dose treatment with the anti-oxidant vitamin E may enhance NK cell function in cancer patients by protecting from oxidative stress. Seven patients with colorectal cancer (Dukes stage C and D) received a daily dose of 750 mg of vitamin E during a period of two weeks and the function, phenotype and receptor expression of NK cells were analyzed. The short-term vitamin E treatment significantly improved NK cell cytolytic activity in six out of the seven patients analyzed. The increased NK cell activity in patients' PBMC was not due to increased numbers of NK cells or an increase in the proportion of the CD56(dim) NK cell subpopulation. Furthermore, neither an increased perforin expression nor an enhanced ability of NK cells to produce IFN-gamma was observed as a result of vitamin E treatment. Finally, vitamin E treatment was associated with a minor, but consistent, induction of NKG2D expression in all patients analyzed. In conclusion, this pilot study demonstrates that vitamin E may boost NK cell function in patients with colorectal cancer. Further studies are warranted to explore the potential of vitamin E as an adjuvant for immunotherapy against cancer and to determine the underlying mechanism(s) behind vitamin E induced NK cell activation.
Collapse
Affiliation(s)
- Mikael G. V. Hanson
- Department of Oncology and Pathology, Immune and Gene Therapy Laboratory, Cancer Center Karolinska, Karolinska Institutet, R8:01, Karolinska Hospital, 171 76 Stockholm, Sweden
| | - Volkan Özenci
- Department of Oncology and Pathology, Immune and Gene Therapy Laboratory, Cancer Center Karolinska, Karolinska Institutet, R8:01, Karolinska Hospital, 171 76 Stockholm, Sweden
| | - Mattias C. V. Carlsten
- Department of Oncology and Pathology, Immune and Gene Therapy Laboratory, Cancer Center Karolinska, Karolinska Institutet, R8:01, Karolinska Hospital, 171 76 Stockholm, Sweden
- Department of Medicine, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Bengt L. Glimelius
- Department of Oncology and Pathology, Immune and Gene Therapy Laboratory, Cancer Center Karolinska, Karolinska Institutet, R8:01, Karolinska Hospital, 171 76 Stockholm, Sweden
- Department of Oncology, Radiology and Clinical Immunology Akademiska sjukhuset Uppsala, Uppsala, Sweden
| | - Jan-Erik A. Frödin
- Department of Oncology and Pathology, Immune and Gene Therapy Laboratory, Cancer Center Karolinska, Karolinska Institutet, R8:01, Karolinska Hospital, 171 76 Stockholm, Sweden
- Department of Oncology (Radiumhemmet), Karolinska University Hospital Solna, Stockholm, Sweden
| | - Giuseppe Masucci
- Department of Oncology and Pathology, Immune and Gene Therapy Laboratory, Cancer Center Karolinska, Karolinska Institutet, R8:01, Karolinska Hospital, 171 76 Stockholm, Sweden
| | - Karl-Johan Malmberg
- Department of Medicine, Center for Infectious Medicine, Karolinska Institutet, Karolinska University Hospital Huddinge, Stockholm, Sweden
| | - Rolf V. R. Kiessling
- Department of Oncology and Pathology, Immune and Gene Therapy Laboratory, Cancer Center Karolinska, Karolinska Institutet, R8:01, Karolinska Hospital, 171 76 Stockholm, Sweden
| |
Collapse
|
156
|
Fearfield LA, Larkin JMG, Rowe A, A'Hern R, Fisher C, Francis N, MacKie R, McCann B, Gore ME, Bunker CB. Expression of p16, CD95, CD95L and Helix pomatia agglutinin in relapsing and nonrelapsing very thin melanoma. Br J Dermatol 2007; 156:440-7. [PMID: 17300231 DOI: 10.1111/j.1365-2133.2006.07581.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Abstract
BACKGROUND The incidence of malignant melanoma is increasing worldwide and patients are being diagnosed earlier with thinner primary lesions. Most patients with very thin melanoma (Breslow thickness < 0.76 mm) are cured by surgery but 2-18% relapse locally or with distant metastases. OBJECTIVES The objective of this study was to establish potential new prognostic markers in very thin melanoma. METHODS We identified a group of subjects with relapsing very thin primary cutaneous melanoma and a matched control group who had not relapsed. We investigated the expression of p16, Helix pomatia agglutinin (HPA), CD95 and CD95 ligand (CD95L) by immunohistochemistry on paraffin-embedded tissue sections from the subject group, their subsequent metastases and the control group. RESULTS Reduced p16 expression was significantly associated with relapse in very thin melanoma (P = 0.0129). Loss of p16 expression was also found in 76% of metastases. There was no significant association between HPA, CD95 or CD95L expression and subsequent relapse. CONCLUSIONS This work is the first to show a significant loss of p16 in relapsing very thin melanoma.
Collapse
Affiliation(s)
- L A Fearfield
- Department of Dermatology, Imperial College School of Medicine (START Laboratories), Chelsea and Westminster Hospital, London, UK.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
157
|
Simons MP, Nauseef WM, Griffith TS. Neutrophils and TRAIL: insights into BCG immunotherapy for bladder cancer. Immunol Res 2007; 39:79-93. [DOI: 10.1007/s12026-007-0084-1] [Citation(s) in RCA: 32] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/30/1999] [Revised: 11/30/1999] [Accepted: 11/30/1999] [Indexed: 11/30/2022]
|
158
|
Abstract
Despite major advances in understanding the mechanisms leading to tumor immunity, a number of obstacles hinder the successful translation of mechanistic insights into effective tumor immunotherapy. Such obstacles include the ability of tumors to foster a tolerant microenvironment and the activation of a plethora of immunosuppressive mechanisms, which may act in concert to counteract effective immune responses. Here we discuss different strategies employed by tumors to thwart immune responses, including tumor-induced impairment of antigen presentation, the activation of negative costimulatory signals, and the elaboration of immunosuppressive factors. In addition, we underscore the influence of regulatory cell populations that may contribute to this immunosuppressive network; these include regulatory T cells, natural killer T cells, and distinct subsets of immature and mature dendritic cells. The current wealth of preclinical information promises a future scenario in which the synchronized blockade of immunosuppressive mechanisms may be effective in combination with other conventional strategies to overcome immunological tolerance and promote tumor regression.
Collapse
Affiliation(s)
- Gabriel A Rabinovich
- Division of Immunogenetics, Hospital de Clínicas José de San Martín, University of Buenos Aires, Buenos Aires, Argentina.
| | | | | |
Collapse
|
159
|
Abstract
Adoptive transfer of antigen-specific T lymphocytes is a powerful therapy for the treatment of opportunistic disease and some virus-associated malignancies such as Epstein-Barr virus-positive post-transplant lymphoproliferative disease. However, this strategy has been less successful in patients with nonviral cancers owing to their many and varied immune evasion mechanisms. These mechanisms include downregulation of target antigens and antigen-presenting machinery, secretion of inhibitory cytokines, and recruitment of regulatory immune cells to the tumor site. With increased understanding of the tumor microenvironment and the behavior and persistence of ex vivo-manipulated, adoptively transferred T cells, two novel approaches for increasing the efficacy of T cell therapy have been proposed. The first involves genetic modification of tumor-specific T cells to improve their biological function, for example by augmenting their ability to recognize tumor cells or their resistance to tumor-mediated immunosuppression. The second requires modifications to the host environment to improve the homeostatic expansion of infused T cells or to eliminate inhibitory T cell subsets. In this review, we discuss current, promising strategies to improve adoptive T cell therapy for the treatment of cancer.
Collapse
Affiliation(s)
- Ann M Leen
- Center for Cell and Gene Therapy, Department of Pediatrics, Baylor College of Medicine, The Methodist Hospital and Texas Children's Hospital, Houston, Texas 77030, USA
| | | | | |
Collapse
|
160
|
Critchley-Thorne RJ, Yan N, Nacu S, Weber J, Holmes SP, Lee PP. Down-regulation of the interferon signaling pathway in T lymphocytes from patients with metastatic melanoma. PLoS Med 2007; 4:e176. [PMID: 17488182 PMCID: PMC1865558 DOI: 10.1371/journal.pmed.0040176] [Citation(s) in RCA: 108] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 11/23/2006] [Accepted: 03/26/2007] [Indexed: 02/08/2023] Open
Abstract
BACKGROUND Dysfunction of the immune system has been documented in many types of cancers. The precise nature and molecular basis of immune dysfunction in the cancer state are not well defined. METHODS AND FINDINGS To gain insights into the molecular mechanisms of immune dysfunction in cancer, gene expression profiles of pure sorted peripheral blood lymphocytes from 12 patients with melanoma were compared to 12 healthy controls. Of 25 significantly altered genes in T cells and B cells from melanoma patients, 17 are interferon (IFN)-stimulated genes. These microarray findings were further confirmed by quantitative PCR and functional responses to IFNs. The median percentage of lymphocytes that phosphorylate STAT1 in response to interferon-alpha was significantly reduced (Delta = 16.8%; 95% confidence interval, 0.98% to 33.35%) in melanoma patients (n = 9) compared to healthy controls (n = 9) in Phosflow analysis. The Phosflow results also identified two subgroups of patients with melanoma: IFN-responsive (33%) and low-IFN-response (66%). The defect in IFN signaling in the melanoma patient group as a whole was partially overcome at the level of expression of IFN-stimulated genes by prolonged stimulation with the high concentration of IFN-alpha that is achievable only in IFN therapy used in melanoma. The lowest responders to IFN-alpha in the Phosflow assay also showed the lowest gene expression in response to IFN-alpha. Finally, T cells from low-IFN-response patients exhibited functional abnormalities, including decreased expression of activation markers CD69, CD25, and CD71; TH1 cytokines interleukin-2, IFN-gamma, and tumor necrosis factor alpha, and reduced survival following stimulation with anti-CD3/CD28 antibodies compared to controls. CONCLUSIONS Defects in interferon signaling represent novel, dominant mechanisms of immune dysfunction in cancer. These findings may be used to design therapies to counteract immune dysfunction in melanoma and to improve cancer immunotherapy.
Collapse
Affiliation(s)
- Rebecca J Critchley-Thorne
- Division of Hematology, Department of Medicine, Stanford University, Stanford, California, United States of America
| | - Ning Yan
- Division of Hematology, Department of Medicine, Stanford University, Stanford, California, United States of America
- Department of Statistics, Stanford University, Stanford, California, United States of America
| | - Serban Nacu
- Department of Statistics, Stanford University, Stanford, California, United States of America
| | - Jeffrey Weber
- Norris Comprehensive Cancer Center, University of Southern California, Los Angeles, California, United States of America
| | - Susan P Holmes
- Department of Statistics, Stanford University, Stanford, California, United States of America
| | - Peter P Lee
- Division of Hematology, Department of Medicine, Stanford University, Stanford, California, United States of America
| |
Collapse
|
161
|
Agarwal A, Mohanti BK, Das SN. Ex vivo triggering of T-cell-mediated immune responses by autologous tumor cell vaccine in oral cancer patients. Immunopharmacol Immunotoxicol 2007; 29:95-104. [PMID: 17464770 DOI: 10.1080/08923970701282742] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/23/2022]
Abstract
We investigated immunomodulatory activity of autologous tumor cell vaccine from oral cancer patients ex vivo by lymphoproliferation assay and two color flow cytometry. Vaccine treatment lead to 10-fold higher proliferation of lymphocytes compared with the untreated controls. A significant increase in CD69(+) and HLA-DR(+) T-cells was observed in vaccine pulsed cultures compared with untreated (p<0.0001) controls. The frequency of IFN-gamma and IL-2 expressing CD4(+)/CD8(+)T-cell subsets was significantly higher with a concomitant reduction in IL-4 and IL-10 expression in the vaccine-treated group (p<0.0001) compared with the untreated controls. Vaccine treatment further increased T-cell receptor Vbeta3, Vbeta5, and Vbeta8 usage. It seems that the autologous tumor cell vaccine triggers T-cell responses ex vivo, hence it may have a protective role in oral cancer patients.
Collapse
MESH Headings
- Adult
- Aged
- Antigens, CD/immunology
- Antigens, Differentiation, T-Lymphocyte/immunology
- CD4-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/immunology
- Cancer Vaccines/immunology
- Carcinoma, Squamous Cell/immunology
- Cells, Cultured
- Cytokines/immunology
- Female
- HLA-DR Antigens/immunology
- Humans
- Immunity, Cellular
- Lectins, C-Type
- Male
- Middle Aged
- Mouth Neoplasms/immunology
- Receptors, Antigen, T-Cell, alpha-beta/immunology
Collapse
Affiliation(s)
- Abhilasha Agarwal
- Department of Biotechnology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | | | | |
Collapse
|
162
|
Abstract
Death receptor ligands (FasL, TRAIL) activate apoptosis in cells expressing the cognate receptors. Evidence suggests that these ligands also deliver pro-inflammatory signals. In the tumor microenvironment, "Fas counterattack" mounted by tumors against immune cells is mediated by tumor-associated FasL. But death ligands crosslinking their receptors also induce inhibition of apoptosis and activation of the transcription factor, NFkappaB, with a subsequent burst of pro-inflammatory cytokine production and tumor growth promotion. NFkappaB, a key link between inflammation and cancer, regulates dual activities of death ligands, depending on molecular signals in the tumor microenvironment. This paper focuses on death ligands as an example of the extensive repertoire of strategies devised by tumors for escape from immune control.
Collapse
Affiliation(s)
- Theresa L Whiteside
- Department of Pathology, Immunology and Otolaryngology, University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, USA.
| |
Collapse
|
163
|
Aptsiauri N, Cabrera T, Garcia-Lora A, Lopez-Nevot MA, Ruiz-Cabello F, Garrido F. MHC Class I Antigens and Immune Surveillance in Transformed Cells. INTERNATIONAL REVIEW OF CYTOLOGY 2007; 256:139-89. [PMID: 17241907 DOI: 10.1016/s0074-7696(07)56005-5] [Citation(s) in RCA: 111] [Impact Index Per Article: 6.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/13/2022]
Abstract
MHC class I antigens play a crucial role in the interaction of tumor cells with the host immune system, in particular, in the presentation of peptides as tumor-associated antigens to cytotoxic lymphocytes (CTLs) and in the regulation of cytolytic activity of natural killer (NK) cells. In this review we discuss the role of MHC class I antigens in the recognition and elimination of transformed cells and in the generation of tumor immune escape routes when MHC class I losses occur in tumors. The different altered MHC class I phenotypes and their distribution in different human tumors are the main topic of this review. In addition, molecular defects that underlie MHC alterations in transformed cells are also described in detail. Future research directions in this field are also discussed, including the laboratory analysis of tumor MHC class I-negative variants and the possible restoration of MHC class I expression.
Collapse
Affiliation(s)
- Natalia Aptsiauri
- Servicio de Análisis Clínicos, Hospital Universitario Virgen de las Nieves, Granada, Spain
| | | | | | | | | | | |
Collapse
|
164
|
Evaluation of Selected Indicators of Apoptosis in Patients with Thyroid Tumors. POLISH JOURNAL OF SURGERY 2007. [DOI: 10.2478/v10035-007-0014-x] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
|
165
|
Hargadon KM, Brinkman CC, Sheasley-O'neill SL, Nichols LA, Bullock TNJ, Engelhard VH. Incomplete differentiation of antigen-specific CD8 T cells in tumor-draining lymph nodes. THE JOURNAL OF IMMUNOLOGY 2006; 177:6081-90. [PMID: 17056534 DOI: 10.4049/jimmunol.177.9.6081] [Citation(s) in RCA: 47] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/29/2022]
Abstract
CD8 T cells lacking effector activity have been recovered from lymphoid organs of mice and patients with progressing tumors. We explored the basis for lack of effector activity in tumor-bearing mice by evaluating Ag presentation and CD8 T cell function in lymphoid organs over the course of tumor outgrowth. Early after tumor injection, cross-presentation by bone marrow-derived APC was necessary for T cell activation, inducing proliferation and differentiation into IFN-gamma-producing, cytolytic effectors. At later stages of outgrowth, tumor metastasized to draining lymph nodes. Both cross- and direct presentation occurred, but T cell differentiation induced by either modality was incomplete (proliferation without cytokine production). T cells within tumor-infiltrated nodes differentiated appropriately if Ag was presented by activated, exogenous dendritic cells. Thus, activated T cells lacking effector function develop through incomplete differentiation in the lymph nodes of late-stage tumor-bearing mice, rather than through suppression of previously differentiated cells.
Collapse
Affiliation(s)
- Kristian M Hargadon
- Department of Microbiology and Carter Immunology Center, University of Virginia Health System, Charlottesville, VA 22908, USA
| | | | | | | | | | | |
Collapse
|
166
|
Valenti R, Huber V, Filipazzi P, Pilla L, Sovena G, Villa A, Corbelli A, Fais S, Parmiani G, Rivoltini L. Human tumor-released microvesicles promote the differentiation of myeloid cells with transforming growth factor-beta-mediated suppressive activity on T lymphocytes. Cancer Res 2006; 66:9290-8. [PMID: 16982774 DOI: 10.1158/0008-5472.can-06-1819] [Citation(s) in RCA: 388] [Impact Index Per Article: 20.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Abstract
Human tumors constitutively release endosome-derived microvesicles, transporting a broad array of biologically active molecules with potential modulatory effects on different immune cells. Here, we report the first evidence that tumor-released microvesicles alter myeloid cell function by impairing monocyte differentiation into dendritic cells and promoting the generation of a myeloid immunosuppressive cell subset. CD14+ monocytes isolated from healthy donors and differentiated with interleukin (IL)-4 and granulocyte macrophage colony-stimulating factor in the presence of tumor-derived microvesicles turned into HLA-DR(-/low) cells, retaining CD14 expression and failing to up-regulate costimulatory molecules, such as CD80 and CD86. These phenotypic changes were paralleled by a significant release of different cytokines, including IL-6, tumor necrosis factor-alpha, and transforming growth factor-beta (TGF-beta), and a dose-dependent suppressive activity on activated T-cell-proliferation and cytolytic functions, which could be reversed by anti-TGF-beta-neutralizing antibodies. Microvesicles isolated from plasma of advanced melanoma patients, but not from healthy donors, mediated comparable effects on CD14+ monocytes, skewing their differentiation toward CD14+HLA-DR-/low cells with TGF-beta-mediated suppressive activity on T-cell-functions. Interestingly, a subset of TGF-beta-secreting CD14+HLA-DR- cells mediating suppressive activity on T lymphocytes was found to be significantly expanded in peripheral blood of melanoma patients compared with healthy donors. These data suggest the development in cancer patients of an immunosuppressive circuit by which tumors promote the generation of suppressive myeloid cells through the release of circulating microvesicles and without the need for cell-to-cell contact. Therapeutic interventions on the crucial steps of this pathway may contribute to restore tumor/immune system interactions favoring T-cell-mediated control of tumor growth in cancer patients.
Collapse
Affiliation(s)
- Roberta Valenti
- Unit of Immunotherapy of Human Tumors, Istituto Nazionale Tumori, Milan, Italy
| | | | | | | | | | | | | | | | | | | |
Collapse
|
167
|
Hong C, Lee H, Oh M, Kang CY, Hong S, Park SH. CD4+T Cells in the Absence of the CD8+Cytotoxic T Cells Are Critical and Sufficient for NKT Cell-Dependent Tumor Rejection. THE JOURNAL OF IMMUNOLOGY 2006; 177:6747-57. [PMID: 17082588 DOI: 10.4049/jimmunol.177.10.6747] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
NKT cells perform crucial roles in tumor surveillance, functioning as regulators of early host response. In this study, we have assessed the effects of NKT activation at the time of tumor Ag immunization, and have evaluated the contributions of CD4+ and CD8+ T cells in tumor rejection during adaptive immune response against live tumor cells. Our data indicate that CD4+ T cells play critical roles, not only in assisting CTL, but also in the orchestration of host response against the tumor. The CD4+ T cells were found to reject the transplanted tumor cells very efficiently under conditions in which the CTLs were removed either genetically, or via the action of anti-CD8 Ab in mice that had been immunized with tumor extracts and alpha-galactosylceramide. Immunization resulted in an NKT cell-dependent antitumor adaptive immune response, which was associated with both CD4+ T cells and cytokine IFN-gamma.
Collapse
MESH Headings
- Animals
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/transplantation
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/immunology
- Cell Line, Tumor
- Epitopes, T-Lymphocyte/administration & dosage
- Epitopes, T-Lymphocyte/immunology
- Galactosylceramides/administration & dosage
- Galactosylceramides/immunology
- Graft Rejection/immunology
- Immunity, Innate
- Killer Cells, Natural/immunology
- Killer Cells, Natural/transplantation
- Lymphocyte Activation/immunology
- Lymphopenia/immunology
- Melanoma, Experimental/immunology
- Melanoma, Experimental/prevention & control
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- Mice, Knockout
- Neoplasm Transplantation
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/prevention & control
- T-Lymphocyte Subsets/immunology
- T-Lymphocyte Subsets/transplantation
- T-Lymphocytes, Cytotoxic/immunology
Collapse
Affiliation(s)
- Changwan Hong
- School of Life Sciences and Biotechnology, Korea University, Seoul, Korea
| | | | | | | | | | | |
Collapse
|
168
|
Jung MK, Song HK, Kim KE, Hur DY, Kim T, Bang S, Park H, Cho DH. IL-18 enhances the migration ability of murine melanoma cells through the generation of ROI and the MAPK pathway. Immunol Lett 2006; 107:125-30. [PMID: 17014914 DOI: 10.1016/j.imlet.2006.08.004] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/11/2006] [Revised: 08/14/2006] [Accepted: 08/20/2006] [Indexed: 11/20/2022]
Abstract
Interleukin-18 (IL-18) has multiple effects on various cells that are involved in immune escape of murine melanoma cells and in the inflammatory responses. This study investigated the effect of IL-18 on the ability of murine melanoma cells to migrate by using B16F10 cells and the IL-18 antisense transfectants of B16F10 cells (the B16F10/IL-18 antisense transfectant). The B16F10 cells were more able to migrate than were the B16F10/IL-18 antisense transfectants. An exogenous IL-18 treatment improved the ability of the B16F10/IL-18 antisense transfectant cells to migrate, indicating that IL-18 enhanced the migration ability of melanoma cells. To determine the signaling mechanisms involved in IL-18-enhanced migration, we measured the ROI levels. It was found that the ROI levels were increased by IL-18, and an antioxidant, N-acetyl-l-cystein (NAC), blocked the effect of IL-18 on migration, suggesting the involvement of ROI in the signal transduction of IL-18-enhanced cell migration. IL-18-enhanced cell migration was also reduced by PD98059. In addition, the level of ERK1/2 phosphorylation was markedly increased by treating with exogenous IL-18 at 20 min. These results suggest that IL-18 enhances the ability of melanoma cells to migrate via the generation of ROI and the MAPK pathway.
Collapse
Affiliation(s)
- Min Kyung Jung
- Department of Life Science, Sookmyung Women's University, Chungpa-Dong 2-Ka, Yongsan-ku, Seoul 140-742, Republic of Korea
| | | | | | | | | | | | | | | |
Collapse
|
169
|
Devarapu SK, Sharma SC, Das SN. Triggering of T cell-mediated immune responses by allogenic tumor cell vaccine in patients with oral cancer. Immunopharmacol Immunotoxicol 2006; 28:387-95. [PMID: 16997788 DOI: 10.1080/08923970600927348] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/24/2022]
Abstract
We investigated the immunomodulatory activity of allogenic whole tumor cell vaccine in oral cancer patients in vitro by two-color flow cytometry. Vaccine treatment significantly increased the expression of CD69 and HLA-DR in CD3+ T-cell subsets. The frequency of Interferon-gamma and Interleukin (IL)-2 expressing CD4+/CD8+ T-cell subsets was significantly higher with a concomitant reduction in IL-4 and IL-10 expressing T-cells in the vaccine treated group as compared with the untreated controls. Vaccine treatment significantly increased T-cell receptor (TCR), Vbeta3, Vbeta5 and Vbeta8 usage. The results indicate that the allogenic whole tumor cell vaccine is able to trigger T-cell mediated immunity in patients with intraoral squamous cell carcinoma.
Collapse
MESH Headings
- Adult
- Aged
- Antigens, CD/analysis
- Antigens, Differentiation, T-Lymphocyte/analysis
- CD3 Complex/analysis
- CD4-Positive T-Lymphocytes/immunology
- CD4-Positive T-Lymphocytes/metabolism
- CD8-Positive T-Lymphocytes/immunology
- CD8-Positive T-Lymphocytes/metabolism
- Cancer Vaccines/administration & dosage
- Cancer Vaccines/immunology
- Female
- Flow Cytometry/methods
- HLA-DR Antigens/analysis
- Humans
- Interferon-gamma/analysis
- Interleukin-10/analysis
- Interleukin-2/analysis
- Interleukin-4/analysis
- Interleukins/analysis
- Interleukins/classification
- Jurkat Cells
- Lectins, C-Type
- Male
- Middle Aged
- Mouth Neoplasms/drug therapy
- Mouth Neoplasms/immunology
- Mouth Neoplasms/metabolism
- Neoplasms, Squamous Cell/drug therapy
- Neoplasms, Squamous Cell/immunology
- Neoplasms, Squamous Cell/metabolism
- Receptors, Antigen, T-Cell, alpha-beta/analysis
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
Collapse
Affiliation(s)
- Satish K Devarapu
- Department of Biotechnology, All India Institute of Medical Sciences, Ansari Nagar, New Delhi, India
| | | | | |
Collapse
|
170
|
Govedarović V, Radojević-Skodrić S, Mitrović D, Müller CA, Müller GA, Marković-Lipovski J. [Expression of Fas and Fas-L in renal cell carcinoma]. SRP ARK CELOK LEK 2006; 134:213-8. [PMID: 16972408 DOI: 10.2298/sarh0606213g] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/27/2022] Open
Abstract
INTRODUCTION The previous investigations revealed that Fas-L expression on tumor cells can be one of the reasons of tumor growth, or tumor regression, with or without activation of the immune response. OBJECTIVE The objective of our study was to investigate the expression of Fas and Fas-L in situ in normal human renal tissue as well as in different types of renal cell carcinoma (RCC) according to tumor grading. METHOD Expression of Fas and Fas-L was examined in 25 RCCs classified according to nuclear grades: G1-G3 and to cell type: 17 clear cells, 3 chromophilics (2 eosinophilics, 1 basophilic), 2 chromophobes and 3 spindle cells. Ten normal human kidneys were analyzed, too. Indirect immunoperoxidase technique was applied. Spread and intensity of staining of Fas and Fas-L molecules expression were scored semiquantitatively. RESULTS Distribution of Fas expression in these RCC was typically diffuse. However, Fas-L was almost completely absent in clear cell RCC. In 3 clear cell RCC, some tumor stromal cells exhibited strong expression of Fas-L. On the contrary, chromophilic, chromophobe and spindle cell RCCs grading from G2-G3, manifested variable combinations of Fas and Fas-L expression. CONCLUSION The most of clear cell type low grade RCCs manifested intensive and extensive expression of Fas and completely absence of Fas-L. However, RCCs of high grade malignancy belonging to the clear cell, eosinophilic, chromophobe or spindle cell types can have various combinations of Fas and Fas-L expression. It may probably lead to development of different mechanisms of avoidance of immune response to RCC.
Collapse
|
171
|
Atre N, Thomas L, Mistry R, Pathak K, Chiplunkar S. Role of nitric oxide in heat shock protein induced apoptosis of gammadeltaT cells. Int J Cancer 2006; 119:1368-76. [PMID: 16619219 DOI: 10.1002/ijc.21966] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/10/2022]
Abstract
Activation induced cell death (AICD) has been proposed to serve as a mechanism to limit T lymphocyte proliferation induced by antigenic stimulation. Heat shock proteins (hsp60 and hsp70) expressed on oral tumor cells serve as ligands for peripheral blood gammadeltaT lymphocytes. Tumor cell lysis by gammadeltaT lymphocytes is mediated via recognition of hsp expressed on tumor cells. In the present study, we report that upon stimulation with hsp, gammadeltaT lymphocytes isolated from oral cancer patients undergo AICD as confirmed by DNA ploidy, annexin V staining and confocal microscopy. In cocultures of gammadeltaT lymphocytes and tumor cells, addition of antihsp60 and antihsp70 MAb, but not anti-Fas MAb (ZB4), inhibited DNA fragmentation of gammadeltaT lymphocytes. Flow cytometric analysis revealed a down regulation of Fas expression on gammadeltaT lymphocytes upon incubation with hsp60 and hsp70. Increased expression of iNOS was observed in hsp-stimulated gammadeltaT lymphocytes. Addition of monomethyl L-arginine monoacetate, competitive inhibitor of NOS, inhibited nitric oxide (NO) production and apoptosis of gammadeltaT lymphocytes induced by hsp60 and hsp70. The NO-induced apoptosis of gammadeltaT lymphocytes involves activation of caspase-9 and loss of mitochondrial membrane potential. The present study explains a novel strategy adopted by tumor cells to evade immune recognition by gammadeltaT lymphocytes.
Collapse
Affiliation(s)
- Nilangi Atre
- Immunology, Advanced Centre for Treatment, Research and Education in Cancer, Tata Memorial Centre, Navi Mumbai-410208, India
| | | | | | | | | |
Collapse
|
172
|
Neuber K, Eidam B. Expression of Fas ligand (CD95L) in primary malignant melanoma and melanoma metastases is associated with overall survival. Oncol Res Treat 2006; 29:361-5. [PMID: 16974112 DOI: 10.1159/000094355] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/19/2022]
Abstract
BACKGROUND There is increasing evidence that the Fas/Fas ligand (FasL) system is involved in tumor-mediated immune suppression. The purpose of this study was to investigate the effect of Fas (CD95) as well as FasL (CD95L) expression in primary malignant melanoma and melanoma metastases on overall survival (OS). PATIENTS AND METHODS 19 patients with metastatic malignant melanoma who were treated with different dacarbazine (DTIC)-based chemotherapy regimens were included in this study. From each patient, primary melanoma biopsies and biopsies from metastases were histologically evaluated. Immunohistology was performed with antibodies to Fas/CD95 and FasL/CD95L. Differences in OS were plotted using the Kaplan-Meier method and compared by the log rank test. RESULTS Fas/CD95 and FasL/CD95L expression was detected in 73.7 and 63.2% of primary melanomas, respectively. In metastases, expression of both Fas/CD95 (63.2%) and FasL/CD95L (47.4%) was markedly decreased. Presence of FasL/ CD95L expression in primary melanoma resulted in significantly (p = 0.024) prolonged OS compared with FasL/CD95L-negative high-risk primary melanomas. In contrast, loss of FasL/CD95L expression in melanoma metastases resected before chemotherapy was associated with significantly prolonged median survival (p = 0.0139). CONCLUSION Presence of FasL/CD95L expression in primary malignant melanoma and the loss of FasL/ CD95L expression in metastases seem to be positive prognostic factors.
Collapse
Affiliation(s)
- Karsten Neuber
- Klinik für Dermatologie und Venerologie, Kopf-und Hautzentrum, Universitätsklinikum Hamburg-Eppendorf, Germany.
| | | |
Collapse
|
173
|
Ohta A, Gorelik E, Prasad SJ, Ronchese F, Lukashev D, Wong MKK, Huang X, Caldwell S, Liu K, Smith P, Chen JF, Jackson EK, Apasov S, Abrams S, Sitkovsky M. A2A adenosine receptor protects tumors from antitumor T cells. Proc Natl Acad Sci U S A 2006; 103:13132-7. [PMID: 16916931 PMCID: PMC1559765 DOI: 10.1073/pnas.0605251103] [Citation(s) in RCA: 803] [Impact Index Per Article: 42.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
The A2A adenosine receptor (A2AR) has been shown to be a critical and nonredundant negative regulator of immune cells in protecting normal tissues from inflammatory damage. We hypothesized that A2AR also protects cancerous tissues by inhibiting incoming antitumor T lymphocytes. Here we confirm this hypothesis by showing that genetic deletion of A2AR in the host resulted in rejection of established immunogenic tumors in approximately 60% of A2AR-deficient mice with no rejection observed in control WT mice. The use of antagonists, including caffeine, or targeting the A2 receptors by siRNA pretreatment of T cells improved the inhibition of tumor growth, destruction of metastases, and prevention of neovascularization by antitumor T cells. The data suggest that effects of A2AR are T cell autonomous. The inhibition of antitumor T cells via their A2AR in the adenosine-rich tumor microenvironment may explain the paradoxical coexistence of tumors and antitumor immune cells in some cancer patients (the "Hellstrom paradox"). We propose to target the hypoxia-->adenosine-->A2AR pathway as a cancer immunotherapy strategy to prevent the inhibition of antitumor T cells in the tumor microenvironment. The same strategy may prevent the premature termination of immune response and improve the vaccine-induced development of antitumor and antiviral T cells. The observations of autoimmunity during melanoma rejection in A2AR-deficient mice suggest that A2AR in T cells is also important in preventing autoimmunity. Thus, although using the hypoxia-->adenosine-->A2AR pathway inhibitors may improve antitumor immunity, the recruitment of this pathway by selective drugs is expected to attenuate the autoimmune tissue damage.
Collapse
Affiliation(s)
- Akio Ohta
- *Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
- New England Inflammation and Tissue Protection Institute, Northeastern University, Boston, MA 02115
| | - Elieser Gorelik
- Department of Pathology and University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15213
| | - Simon J. Prasad
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Franca Ronchese
- Malaghan Institute of Medical Research, Wellington, New Zealand
| | - Dmitriy Lukashev
- *Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
- New England Inflammation and Tissue Protection Institute, Northeastern University, Boston, MA 02115
| | - Michael K. K. Wong
- Department of Pathology and University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15213
- Department of Medicine and Department of Pharmacology and Therapeutics, Roswell Park Cancer Institute, Buffalo, NY 14263
| | - Xiaojun Huang
- Department of Pathology and University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15213
| | - Sheila Caldwell
- **Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Kebin Liu
- **Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
- Department of Biochemistry and Molecular Biology, Medical College of Georgia, Augusta, GA 30912
| | - Patrick Smith
- *Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Jiang-Fan Chen
- Department of Neurology, Boston University School of Medicine, Boston, MA 02118; and
| | - Edwin K. Jackson
- Center for Clinical Pharmacology, Departments of Pharmacology and Medicine, University of Pittsburgh Cancer Institute, University of Pittsburgh, Pittsburgh, PA 15213
| | - Sergey Apasov
- *Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
| | - Scott Abrams
- **Laboratory of Tumor Immunology and Biology, Center for Cancer Research, National Cancer Institute, National Institutes of Health, Bethesda, MD 20892
| | - Michail Sitkovsky
- *Laboratory of Immunology, National Institute of Allergy and Infectious Diseases, National Institutes of Health, Bethesda, MD 20892
- New England Inflammation and Tissue Protection Institute, Northeastern University, Boston, MA 02115
- To whom correspondence should be addressed at:
New England Inflammation and Tissue Protection Institute, Northeastern University, 360 Huntington Avenue, 113 Mugar Life Sciences Building, Boston, MA 02115. E-mail:
| |
Collapse
|
174
|
Strauss G, Osen W, Knape I, Jacobsen EM, Müller SM, Debatin KM. Membrane-bound CD95 ligand expressed on human antigen-presenting cells prevents alloantigen-specific T cell response without impairment of viral and third-party T cell immunity. Cell Death Differ 2006; 14:480-8. [PMID: 16902496 DOI: 10.1038/sj.cdd.4402019] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
Genetically modified antigen-presenting cells (APC) represent an attractive strategy for in vitro immunomodulation. In the human system, APC expressing HLA-A1 and a membrane-bound form of CD95L (m-CD95L) were used for selective depletion of HLA-A1-specific T cells. In short-term assays, m-CD95L-expressing APC-induced apoptosis in activated T cells and the constitutive presence of m-CD95L and HLA-A1 expressing APC in long-term T cell cultures prevented the expansion of CD4(+) and CD8(+) HLA-A1-specific T cells and the development of HLA-A1-specific cytotoxicity. However, immunity towards third party, viral and bacterial antigens was maintained and T cells spared from depletion could be induced to develop cytotoxicity towards unrelated antigens. Interestingly, inhibition of HLA-A1-specific T cell response absolutely requires the coexpression of m-CD95L and HLA-A1 antigen on the same APC. Thus, m-CD95L expressing APC might be used in clinical settings to obtain tolerance induction in allogeneic transplantation systems or autoimmune diseases.
Collapse
Affiliation(s)
- G Strauss
- University Children's Hospital, Ulm, Germany
| | | | | | | | | | | |
Collapse
|
175
|
Zubieta MR, Furman D, Barrio M, Bravo AI, Domenichini E, Mordoh J. Galectin-3 expression correlates with apoptosis of tumor-associated lymphocytes in human melanoma biopsies. THE AMERICAN JOURNAL OF PATHOLOGY 2006; 168:1666-75. [PMID: 16651632 PMCID: PMC1606586 DOI: 10.2353/ajpath.2006.050971] [Citation(s) in RCA: 56] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/22/2022]
Abstract
The immune system recognizes diverse melanoma antigens. However, tumors can evade the immune response, therefore growing and progressing. It has been reported that galectin-3 and galectin-1 can induce apoptosis of activated lymphocytes. However, there is strong evidence indicating that the regulation of galectins function in the human tumor microenvironment is a complex process that is influenced by diverse biological circumstances. Here, we have investigated 33 biopsies (eight primary and 25 metastases) from 24 melanoma patients (15-72 years old) and describe the correlation between the expression of galectin-3 or galectin-1 and the level of apoptosis of tumor-associated lymphocytes using immunohistochemistry and an in situ nick translation assay. The range of galectin-3-positive tumor cells varied between 0% and 93% and that of galectin-1-positive tumor cells varied between 5% and 97%. In addition, 23 +/- 27% of tumor-associated lymphocytes were apoptotic. Although our results show a correlation between galectin-3 expression and apoptosis of tumor-associated lymphocytes, we could not find such correlation with galectin-1. Considering the complex process of cancer immunoediting, various interacting factors must be considered.
Collapse
|
176
|
Abstract
Transforming growth factor-beta (TGFbeta) signalling regulates cancer through mechanisms that function either within the tumour cell itself or through host-tumour cell interactions. Studies of tumour-cell-autonomous TGFbeta effects show clearly that TGFbeta signalling has a mechanistic role in tumour suppression and tumour promotion. In addition, factors in the tumour microenvironment, such as fibroblasts, immune cells and the extracellular matrix, influence the ability of TGFbeta to promote or suppress carcinoma progression and metastasis. The complex nature of TGFbeta signalling and crosstalk in the tumour microenvironment presents a unique challenge, and an opportunity to develop therapeutic intervention strategies for targeting cancer.
Collapse
Affiliation(s)
- Brian Bierie
- Department of Cancer Biology, Vanderbilt University Medical Center, Nashville, Tennessee 37232, USA
| | | |
Collapse
|
177
|
Lang K, Entschladen F, Weidt C, Zaenker KS. Tumor immune escape mechanisms: impact of the neuroendocrine system. Cancer Immunol Immunother 2006; 55:749-60. [PMID: 16435128 PMCID: PMC11030197 DOI: 10.1007/s00262-006-0126-x] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/31/2005] [Accepted: 01/04/2006] [Indexed: 12/24/2022]
Abstract
Tumor cells act upon, and react to both their proximate and more distant environment, the mechanisms by which this is achieved being both autocrine and paracrine in nature. This interaction, however, takes place not only between adjacent malignant cells, but also non-malignant cells such as those of the immune system, the latter also partaking in the modeling of the tumor environment. Although tumor cells descend from normal tissue cells and thus bear in classical immunological terms 'self signals', it is evident that the immune system is able to recognize tumor cells as a harassment for the body and in consequence tries to eliminate these cells. On the counterpart, tumor cells acquire various characteristics which allow them to evade this immunological surveillance, and have been collectively coined with the term "tumor escape mechanisms". This review will describe and summarize current understanding of tumor escape strategies, and also more closely elaborate on the modulatory role of the neuroendocrine system in the immune system-tumor cell interaction.
Collapse
Affiliation(s)
- Kerstin Lang
- Institute of Immunology, Witten/Herdecke University, Stockumer Str. 10, 58448 Witten, Germany.
| | | | | | | |
Collapse
|
178
|
Toyoshima T, Nakamura S, Kumamaru W, Kawamura E, Ishibashi H, Hayashida JN, Moriyama M, Ohyama Y, Sasaki M, Shirasuna K. Expression of tumor-associated antigen RCAS1 and its possible involvement in immune evasion in oral squamous cell carcinoma. J Oral Pathol Med 2006; 35:361-8. [PMID: 16762017 DOI: 10.1111/j.1600-0714.2006.00442.x] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
BACKGROUND RCAS1 (receptor-binding cancer antigen expressed on SiSo cells) is known to induce apoptosis in its receptor-positive cells. The authors investigated RCAS1 expression in oral squamous cell carcinoma (SCC) and its association with the apoptosis of tumor-infiltrating lymphocytes (TILs). METHODS In 130 patients with oral SCC, the expression of RCAS1 in tumor cells was immunohistochemically examined and the apoptosis of TILs was examined by Terminal Deoxynucleotidyltransferase-mediated dUTP Nick End Labeling (TUNEL) staining. RESULTS RCAS1 was detected both on the cytoplasm and the membrane of tumor cells in 41 of 130 cases (31.5%). Focusing on the expression at the invasive front interacting with host immune cells, RCAS1 was detected in 22 of 130 cases (16.9%). The percentage of TUNEL-positive TILs in cases with RCAS1-positive SCCs was significantly higher than in cases with RCAS1-negative SCCs (P < 0.0001). CONCLUSIONS RCAS1 can be expressed on oral SCC cells and may be involved in the tumor escape from the host immune system by inducing the apoptosis of TILs.
Collapse
Affiliation(s)
- T Toyoshima
- Section of Oral and Maxillofacial Surgery, Division of Maxillofacial Diagnostic and Surgical Sciences, Faculty of Dental Science, Kyushu University, 3-1-1 Maidashi, Higashi-ku, Fukuoka 812-8582, Japan
| | | | | | | | | | | | | | | | | | | |
Collapse
|
179
|
Hekimgil M, Cağirgan S, Pehlivan M, Doğanavşargil B, Tombuloğlu M, Soydan S. Immunohistochemical detection of CD 95 (Fas) & Fas ligand (Fas-L) in plasma cells of multiple myeloma and its correlation with survival. Leuk Lymphoma 2006; 47:271-80. [PMID: 16321857 DOI: 10.1080/10428190500286218] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/19/2023]
Abstract
Multiple myeloma (MM) is a malignant disease resulting from an uncontrolled proliferation of a neoplastic plasma cell clone in the bone marrow, which might also be induced by the loss of control on apoptosis. Fas ligand (Fas-L), a member of the tumor necrosis factor family, induces apoptosis mediated via its transmembrane death receptor Fas (Apo-1/CD95) antigen. In the present study, immunostaining was performed on the initial diagnostic bone marrow biopsies of 36 MM patients (1 stage I, 5 stage II, 30 stage III), to evaluate the distribution of Fas receptor and Fas-L on malignant plasma cells. Both Fas and Fas-L were positive in 13 cases and negative in 3, whereas 10 cases were Fas-negative, Fas-L-positive and 10 were Fas-positive, Fas-L-negative. Although no association was found between the expression of Fas receptor or Fas-L and overall survival, Fas-L positivity was significantly associated with a shorter event-free survival (p = 0.0335). In this study, it has been shown that the expression of Fas-L, in malignant plasma cells of myeloma patients significantly shortens the event-free survival, indicating that the defect in apoptosis might be associated with disease progression in MM.
Collapse
Affiliation(s)
- Mine Hekimgil
- Department of Pathology, Ege University Faculty of Medicine, Bornova, Izmir 35100, Turkey.
| | | | | | | | | | | |
Collapse
|
180
|
Flahaut M, Mühlethaler-Mottet A, Auderset K, Bourloud KB, Meier R, Popovic MB, Joseph JM, Gross N. Persistent inhibition of FLIP(L) expression by lentiviral small hairpin RNA delivery restores death-receptor-induced apoptosis in neuroblastoma cells. Apoptosis 2006; 11:255-63. [PMID: 16502263 DOI: 10.1007/s10495-006-3435-9] [Citation(s) in RCA: 22] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
Abstract
Neuroblastoma represents the most common and deadly solid tumour of childhood, which disparate biological and clinical behaviour can be explained by differential regulation of apoptosis. To understand mechanisms underlying death resistance in neuroblastoma cells, we developed small hairpin of RNA produced by lentiviral vectors as tools to selectively interfere with FLIP(L), a major negative regulator of death receptor-induced apoptosis. Such tools revealed highly efficient in interfering with FLIP(L) expression and function as they almost completely repressed endogenous and/or exogenously overexpressed FLIP(L) protein and fully reversed FLIP(L)-mediated TRAIL resistance. Moreover, interference with endogenous FLIP(L) and FLIP(S) significantly restored FasL sensitivity in SH-EP neuroblastoma cell line. These results reveal the ability of lentivirus-mediated shRNAs to specifically and persistently interfere with FLIP expression and support involvement of FLIP in the regulation of death receptor-mediated apoptosis in neuroblastoma cells. Combining such tools with other therapeutic modalities may improve treatment of resistant tumours such as neuroblastoma.
Collapse
Affiliation(s)
- M Flahaut
- Paediatric Oncology Research, Paediatric Department, University Hospital CHUV, CH-1011, Lausanne
| | | | | | | | | | | | | | | |
Collapse
|
181
|
Abstract
Adoptive transfer of antigen-specific T cells has been most effective in treating cytomegalovirus (CMV) disease and Epstein-Barr virus (EBV)-associated lymphoproliferative disease (LPD). Both of these diseases develop only during periods of acute immune suppression, and both involve highly immunogenic infected cells, and thus respond well to T cell therapies. In contrast, tumours that develop in the presence of a competent immune system evolve complex immune evasion strategies to avoid and subvert T cell-mediated killing. Therefore, even T cells that display potent cytotoxic activity against tumour cells in vitro may not be effective in vivo without altering the tumour:T cell balance in favour of the T cell. This review discusses several new areas of research aimed at improving adoptive T cell therapy for the treatment of cancer, including the genetic modification of antigen-specific T cells to allow them to perform better in vivo, and conditioning the host to improve in vivo expansion and function of transferred cells.
Collapse
Affiliation(s)
- Aaron E Foster
- Center for Cell and Gene Therapy, The Methodist Hospital and Texas Children's Hospital, Baylor College of Medicine, Houston, Texas 77030, USA
| | | |
Collapse
|
182
|
Walker DG, Chuah T, Rist MJ, Pender MP. T-cell apoptosis in human glioblastoma multiforme: Implications for immunotherapy. J Neuroimmunol 2006; 175:59-68. [PMID: 16631933 DOI: 10.1016/j.jneuroim.2006.03.006] [Citation(s) in RCA: 34] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/24/2005] [Revised: 03/06/2006] [Accepted: 03/06/2006] [Indexed: 12/28/2022]
Abstract
We used immunohistochemistry and flow cytometry to assess apoptosis in human glioblastoma multiforme (GBM). Our immunohistochemical study revealed apoptosis of glioma cells expressing glial fibrillary acidic protein and of CD3(+) T cells infiltrating GBM. To quantify and phenotype the apoptotic T cells, we performed flow cytometry on lymphocytes separated from GBM. The cells were stained with annexin-V-FLUOS/propidium iodide to identify apoptosis. We found that high proportions of both the CD4(+) and CD8(+) T cells were apoptotic. In particular, we found that T cells expressing Fas ligand (Fas-L, CD95L) were eight times more vulnerable to apoptosis than those not expressing Fas-L, which suggests that the T-cell apoptosis is induced by overactivation of the T-cell receptor, possibly in the absence of appropriate costimulation. Our results have implications for the design of immunotherapies for GBM.
Collapse
Affiliation(s)
- David G Walker
- Department of Neurosurgery, Royal Brisbane and Women's Hospital, c/- Post Office RBH, Herston, Q4029 Australia.
| | | | | | | |
Collapse
|
183
|
Selam B, Kayisli UA, Akbas GE, Basar M, Arici A. Regulation of FAS ligand expression by chemokine ligand 2 in human endometrial cells. Biol Reprod 2006; 75:203-9. [PMID: 16687653 DOI: 10.1095/biolreprod.105.045716] [Citation(s) in RCA: 21] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/01/2022] Open
Abstract
Human endometrium is a dynamic tissue under the influence of numerous hormones, growth factors, and cytokines interacting to maintain a balance of cellular growth, differentiation, and apoptosis. We have previously demonstrated that several factors including interleukin-8, extracellular matrix, and steroid hormones modulate FASLG, one of the apoptotic molecules, in human endometrium. Chemokine ligand 2 (CCL2), a monocyte chemoattractant and activating factor, is a cytokine involved in endometrial function. CCL2 is elevated in the peritoneal fluid of women with endometriosis. We hypothesize that increased levels of CCL2 in the endometriotic environment may upregulate FASLG expression in human endometrial stromal cells and induce a local immunotolerance in endometriosis. To test our hypothesis, we studied the in vitro regulation of FASLG expression and apoptosis by CCL2 in endometrial stromal cells. Western blot analysis revealed that CCL2 upregulated FASLG protein expression in cultured endometrial stromal cells. Based on semiquantitative RT-PCR analysis, CCL2 did not alter either FAS or FASLG mRNA expression in endometrial stromal cells. Immunocytochemistry results from the same cells treated with CCL2 demonstrated upregulation of FASLG protein expression. CCL2 did not change rate of apoptosis in endometrial stromal cells as evaluated by TUNEL assay. However, an increased apoptotic rate was detected in Jurkat (T lymphocytes) cells cocultured with endometrial stromal cells previously treated with CCL2. We speculate that increased FASLG expression by CCL2 may induce apoptosis of T lymphocytes and thus produce an immunotolerant environment for the development of ectopic implants.
Collapse
Affiliation(s)
- Belgin Selam
- Yale University School of Medicine, Department of Obstetrics and Gynecology, Division of Reproductive Endocrinology, New Haven, Connecticut 06520-8063, USA
| | | | | | | | | |
Collapse
|
184
|
Cavallo F, Curcio C, Forni G. Immunotherapy and immunoprevention of cancer: where do we stand? Expert Opin Biol Ther 2006; 5:717-26. [PMID: 15934846 DOI: 10.1517/14712598.5.5.717] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/21/2022]
Abstract
Although evolution has shaped the immune system to control microbial invasions, this does not necessarily mean that the immune system can not be triggered to eliminate tumour cells. The exploitation of the terrific potential of the immune system to recognise cell alterations and to selectively destroy large populations of neoplastic cells is a possibility made even more attractive by the advances in our understanding of the immune mechanisms and our ability to manipulate them. This review summarises the state of the different immunotherapy strategies available or in development today, and examines the future developments that hold out the promise of an effective control of cancer growth.
Collapse
Affiliation(s)
- Federica Cavallo
- University of Turin, Department of Clinical and Biological Sciences, Ospedale San Luigi Gonzaga, I-10043 Orbassano, Italy.
| | | | | |
Collapse
|
185
|
Salon C, Eymin B, Micheau O, Chaperot L, Plumas J, Brambilla C, Brambilla E, Gazzeri S. E2F1 induces apoptosis and sensitizes human lung adenocarcinoma cells to death-receptor-mediated apoptosis through specific downregulation of c-FLIP(short). Cell Death Differ 2006; 13:260-72. [PMID: 16052233 DOI: 10.1038/sj.cdd.4401739] [Citation(s) in RCA: 59] [Impact Index Per Article: 3.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/05/2023] Open
Abstract
E2F1 is a transcription factor that plays a well-documented role during S phase progression and apoptosis. We had previously postulated that the low level of E2F1 in primary lung adenocarcinoma contributes to their carcinogenesis. Here, we show that E2F1 triggers apoptosis in various lung adenocarcinoma cell lines by a mechanism involving the specific downregulation of the cellular FLICE-inhibitory protein short, leading to caspase-8 activation at the death-inducing signaling complex. Importantly, we also provide evidence that E2F1 sensitizes tumor as well as primary cells to apoptosis mediated by FAS ligand or tumor necrosis factor-related apoptosis-inducing ligand, and enhances the cytotoxic effect of T lymphocytes against tumor cells. Finally, we describe the specific overexpression of c-FLIP(S) in human lung adenocarcinomas with low level of E2F1. Overall, our data identify E2F1 as a critical determinant of the cellular response to death-receptor-mediated apoptosis, and suggest that its downregulation contributes to the immune escape of lung adenocarcinoma tumor cells.
Collapse
Affiliation(s)
- C Salon
- 1Groupe de Recherche sur le Cancer du Poumon, INSERM U578, Institut Albert Bonniot, La Tronche Cedex, France
| | | | | | | | | | | | | | | |
Collapse
|
186
|
Babatz J, Röllig C, Löbel B, Folprecht G, Haack M, Günther H, Köhne CH, Ehninger G, Schmitz M, Bornhäuser M. Induction of cellular immune responses against carcinoembryonic antigen in patients with metastatic tumors after vaccination with altered peptide ligand-loaded dendritic cells. Cancer Immunol Immunother 2006; 55:268-76. [PMID: 16034561 PMCID: PMC11031026 DOI: 10.1007/s00262-005-0021-x] [Citation(s) in RCA: 43] [Impact Index Per Article: 2.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/10/2005] [Accepted: 05/03/2005] [Indexed: 12/21/2022]
Abstract
PURPOSE Dendritic cells (DCs) are characterized by their extraordinary capacity to induce T-cell responses, providing the opportunity of DC-based cancer vaccination protocols. In the present study, we conducted a phase I/II clinical trial to determine the capability of DCs differentiated from immunomagnetically isolated CD14+ monocytes and pulsed with a carcinoembryonic antigen-derived altered peptide (CEAalt) to induce specific CD8+ T cells in cancer patients. EXPERIMENTAL DESIGN Nine patients with CEA-positive colorectal cancer (n=7) or lung cancer (n=2) were enrolled in this study. Autologous CD14+ monocytes were isolated by large-scale immunomagnetic separation and differentiated to mature DCs in sufficient numbers and at high purity. After incubation with the CEAalt peptide and keyhole limpet hemocyanin, DCs were administered to patients intravenously at dose levels of 1 x 10(7) and 5 x 10(7) cells. Patients received four immunizations every second week. RESULTS ELISPOT analysis revealed a vaccine-induced increase in the number of CEAalt peptide-specific Interferon (IFN)-gamma producing CD8+ T cells in five of nine patients and of CD8+ T lymphocytes recognizing the native CEA peptide in three of nine patients. In addition, CD8+ T lymphocytes derived from one patient exhibiting an immunological response after vaccination efficiently lysed peptide-loaded T2 cells and tumor cells. Immunization was well tolerated by all patients without severe signs of toxicity. CONCLUSION Vaccination with CEAalt-pulsed DCs derived from immunomagnetically isolated CD14+ monocytes efficiently expand peptide-specific CD8+ T lymphocytes in vivo and may be a promising alternative for cancer immunotherapy.
Collapse
Affiliation(s)
- Jana Babatz
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| | - Christoph Röllig
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| | - Bärbel Löbel
- Institut für Immunologie, Medizinische Fakultät, TU Dresden, Germany
| | - Gunnar Folprecht
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| | - Michael Haack
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| | - Heinrich Günther
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| | - Claus-Henning Köhne
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| | - Gerhard Ehninger
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| | - Marc Schmitz
- Institut für Immunologie, Medizinische Fakultät, TU Dresden, Germany
| | - Martin Bornhäuser
- Medizinische Klinik und Poliklinik I, Universitätsklinikum Dresden, Fetscherstr. 74, 01307 Dresden, Germany
| |
Collapse
|
187
|
Labarthe MC, Halanek N, Birchall L, Russell N, Desel C, Todryk S, Peters MJ, Lucas A, Falkenberg FW, Dalgleish AG, Whelan M, Ward SJ. The biological effects of syngeneic and allogeneic cytokine-expressing prophylactic whole cell vaccines and the influence of irradiation in a murine melanoma model. Cancer Immunol Immunother 2006; 55:277-88. [PMID: 16158275 PMCID: PMC11030598 DOI: 10.1007/s00262-005-0061-2] [Citation(s) in RCA: 9] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/15/2005] [Accepted: 06/22/2005] [Indexed: 10/25/2022]
Abstract
Allogeneic whole tumour cell vaccines are inherently practical compared with autologous vaccines. Cell lines are derived from allogeneic tumour, grown in bulk and then administered as a vaccine to the patient, following irradiation, which not only prevents any replication but also enhances antigen presentation. Protection is believed to occur through the presentation of antigens shared between the syngeneic and allogeneic tumours. Although cytokine-transfected tumour whole cell vaccines have been used clinically, little data is available comparing the effects of immunomodulatory cytokine-transfection directly on the same cells when used as both an allogeneic and autologous vaccine. To address this, weakly immunogenic B16-F10 (H-2b) murine melanoma was transfected to secrete either GM-CSF, IL-4 or IL-7. Prophylactic vaccination of both syngeneic C57/BL6 (H-2b) (B6) and allogeneic C3H/Hej (H-2k) (C3H) mice showed the effects of transfected cytokine varied between models. Both GM-CSF and IL-7 significantly (P<0.05) increased the levels of protection within syngeneic B6 mice, but had a diminished effect (P>0.05) within C3H allogeneic mice. Allogeneic B16-F10 cells and syngeneic K1735 cells generated CTL against K1735 suggesting cross-reactive immunity. Using cells labeled with fluorescent dye we demonstrate that irradiated vaccines, of either syngeneic or allogeneic origin, appear to generate potent immune responses and fragments of either vaccine remain at the injection site for up to 9 days. This study shows that protection can be enhanced in vivo by using transfected cytokine, but suggests that irradiated whole cell vaccines, of either tissue-type, are rapidly processed. This leads to the conclusion that the cytokine effects are transient and thus transfection with cytokine may be of limited long-term use in situ.
Collapse
Affiliation(s)
| | - Nicole Halanek
- Department of Oncology, St George’s Hospital Medical School, Cranmer Terrace, Tooting, London, SW17 ORE UK
| | - Lindsay Birchall
- Department of Oncology, St George’s Hospital Medical School, Cranmer Terrace, Tooting, London, SW17 ORE UK
| | - Nick Russell
- Onyvax Ltd, St George’s Hospital Medical School, Cranmer Terrace, Tooting, London, SW17 ORE UK
| | - Christiane Desel
- Onyvax Ltd, St George’s Hospital Medical School, Cranmer Terrace, Tooting, London, SW17 ORE UK
| | - Stephen Todryk
- Department of Biochemistry, Immune Regulation Research Group, Trinity College Dublin, Dublin 2, Ireland
| | - Marcus J. Peters
- Abteilung für Medizinische Mikrobiologie, Ruhr-Universitaet Bochum, Universitaetsstrasse 150, 44790 Bochum, Germany
| | - Aisha Lucas
- Abteilung für Medizinische Mikrobiologie, Ruhr-Universitaet Bochum, Universitaetsstrasse 150, 44790 Bochum, Germany
| | - Frank W. Falkenberg
- Abteilung für Medizinische Mikrobiologie, Ruhr-Universitaet Bochum, Universitaetsstrasse 150, 44790 Bochum, Germany
| | - Angus G. Dalgleish
- Department of Oncology, St George’s Hospital Medical School, Cranmer Terrace, Tooting, London, SW17 ORE UK
| | - Mike Whelan
- Onyvax Ltd, St George’s Hospital Medical School, Cranmer Terrace, Tooting, London, SW17 ORE UK
| | - Stephen John Ward
- Onyvax Ltd, St George’s Hospital Medical School, Cranmer Terrace, Tooting, London, SW17 ORE UK
| |
Collapse
|
188
|
Wang B, He J, Liu C, Chang LJ. An effective cancer vaccine modality: lentiviral modification of dendritic cells expressing multiple cancer-specific antigens. Vaccine 2006; 24:3477-89. [PMID: 16530303 PMCID: PMC1850619 DOI: 10.1016/j.vaccine.2006.02.025] [Citation(s) in RCA: 40] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/05/2005] [Revised: 01/25/2006] [Accepted: 02/06/2006] [Indexed: 11/29/2022]
Abstract
Viral modification of dendritic cells (DCs) may deliver a "danger signal" critical to the hypo-reactive DCs in cancer patients. Using three highly differentially expressed hepatoma tumor-associated antigens (TAAs): stem cell antigen-2 (Sca-2), glycoprotein 38 (GP38) and cellular retinoic acid binding protein 1 (RABP1), we explored the therapeutic potential of the DCs modified with lentiviral vectors (LVs). Preventive and therapeutic injection of the LV-TAA-DC vaccine into tumor-bearing mice elicited a strong anti-tumor response and extended survival, which was associated with tumor-specific interferon-gamma and cytotoxic T cell responses. In vivo elimination of the LV-TAA-DCs by a co-expressed thymidine kinase suicide gene abrogated the therapeutic effect. The modification of DCs with LVs encoding multiple TAAs offers a great opportunity in cancer immunotherapy.
Collapse
Affiliation(s)
- Bei Wang
- Department of Molecular Genetics and Microbiology Powell Gene Therapy Center and McKnight Brain Institute, University of Florida, College of Medicine, Gainesville, FL 32610-0266, USA
| | - Jin He
- Department of Molecular Genetics and Microbiology Powell Gene Therapy Center and McKnight Brain Institute, University of Florida, College of Medicine, Gainesville, FL 32610-0266, USA
| | - Chen Liu
- Department of Pathology, Immunology and Laboratory Medicine, University of Florida, College of Medicine, Gainesville, FL 32610-0266, USA
| | - Lung-Ji Chang
- Department of Molecular Genetics and Microbiology Powell Gene Therapy Center and McKnight Brain Institute, University of Florida, College of Medicine, Gainesville, FL 32610-0266, USA
- * Corresponding author. Tel.: +1 352 392 3315; fax: +1 352 392 3133. E-mail address: (L.-J. Chang)
| |
Collapse
|
189
|
Vianello F, Papeta N, Chen T, Kraft P, White N, Hart WK, Kircher MF, Swart E, Rhee S, Palù G, Irimia D, Toner M, Weissleder R, Poznansky MC. Murine B16 Melanomas Expressing High Levels of the Chemokine Stromal-Derived Factor-1/CXCL12 Induce Tumor-Specific T Cell Chemorepulsion and Escape from Immune Control. THE JOURNAL OF IMMUNOLOGY 2006; 176:2902-14. [PMID: 16493048 DOI: 10.4049/jimmunol.176.5.2902] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/18/2023]
Abstract
The chemokine, stromal-derived factor-1/CXCL12, is expressed by normal and neoplastic tissues and is involved in tumor growth, metastasis, and modulation of tumor immunity. T cell-mediated tumor immunity depends on the migration and colocalization of CTL with tumor cells, a process regulated by chemokines and adhesion molecules. It has been demonstrated that T cells are repelled by high concentrations of the chemokine CXCL12 via a concentration-dependent and CXCR4 receptor-mediated mechanism, termed chemorepulsion or fugetaxis. We proposed that repulsion of tumor Ag-specific T cells from a tumor expressing high levels of CXCL12 allows the tumor to evade immune control. Murine B16/OVA melanoma cells (H2b) were engineered to constitutively express CXCL12. Immunization of C57BL/6 mice with B16/OVA cells lead to destruction of B16/OVA tumors expressing no or low levels of CXCL12 but not tumors expressing high levels of the chemokine. Early recruitment of adoptively transferred OVA-specific CTL into B16/OVA tumors expressing high levels of CXCL12 was significantly reduced in comparison to B16/OVA tumors, and this reduction was reversed when tumor-specific CTLs were pretreated with the specific CXCR4 antagonist, AMD3100. Memory OVA-specific CD8+ T cells demonstrated antitumor activity against B16/OVA tumors but not B16/OVA.CXCL12-high tumors. Expression of high levels of CXCL12 by B16/OVA cells significantly reduced CTL colocalization with and killing of target cells in vitro in a CXCR4-dependent manner. The repulsion of tumor Ag-specific T cells away from melanomas expressing CXCL12 confirms the chemorepellent activity of high concentrations of CXCL12 and may represent a novel mechanism by which certain tumors evade the immune system.
Collapse
MESH Headings
- Animals
- Cancer Vaccines/immunology
- Cell Line, Tumor
- Cell Migration Inhibition
- Cell Proliferation
- Chemokine CXCL12
- Chemokines, CXC/biosynthesis
- Chemokines, CXC/genetics
- Chemokines, CXC/physiology
- Chemotaxis, Leukocyte/immunology
- Cytotoxicity, Immunologic
- Dose-Response Relationship, Immunologic
- Epitopes, T-Lymphocyte
- Immunotherapy, Adoptive
- Melanoma, Experimental/immunology
- Melanoma, Experimental/metabolism
- Melanoma, Experimental/pathology
- Melanoma, Experimental/therapy
- Mice
- Mice, Inbred C57BL
- Mice, Transgenic
- Ovalbumin/immunology
- Receptors, Antigen, T-Cell/genetics
- Receptors, CCR5/metabolism
- Receptors, CXCR4/physiology
- T-Lymphocytes/immunology
- T-Lymphocytes/metabolism
- T-Lymphocytes, Cytotoxic/immunology
Collapse
Affiliation(s)
- Fabrizio Vianello
- Partners AIDS Research Center, Infectious Diseases Division, and Massachusetts General Hospital Cancer Center, Charlestown, MA 02129, USA
| | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
190
|
Ivanov VN, Hei TK. Dual treatment with COX-2 inhibitor and sodium arsenite leads to induction of surface Fas Ligand expression and Fas-Ligand-mediated apoptosis in human melanoma cells. Exp Cell Res 2006; 312:1401-17. [PMID: 16487513 PMCID: PMC4376328 DOI: 10.1016/j.yexcr.2006.01.003] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/08/2005] [Revised: 12/12/2005] [Accepted: 01/09/2006] [Indexed: 11/18/2022]
Abstract
Most human melanomas express Fas receptor on the cell surface, and treatment with exogenous Fas Ligand (FasL) efficiently induces apoptosis of these cells. In contrast, endogenous surface expression of FasL is suppressed in Fas-positive melanomas. We report here the use of a combination of sodium arsenite, an inhibitor of NF-kappaB activation, and NS398, a cyclooxygenase-2 (COX-2) inhibitor, for restoration of the surface FasL expression. We observed a large increase of Fas-mediated apoptosis in Fas-positive melanomas. This was due to induction of FasL surface expression and increased susceptibility to Fas death signaling after arsenite and NS398 treatment. Furthermore, silencing COX-2 expression by specific RNAi also effectively increased surface FasL expression following arsenite treatment. Upregulation of the surface FasL levels was based on an increase in the efficiency of translocation to the cell surface and stabilization of FasL protein on the cell surface, rather than on acceleration of the FasL gene transcription. Data obtained demonstrate that the combination of arsenite with inhibitors of COX-2 may affect the target cancer cells via induction of FasL-mediated death signaling.
Collapse
Affiliation(s)
- Vladimir N Ivanov
- Center for Radiological Research, Columbia University College of Physicians and Surgeons, New York, NY 10032, USA.
| | | |
Collapse
|
191
|
Grandics P. The cancer stem cell: evidence for its origin as an injured autoreactive T cell. Mol Cancer 2006; 5:6. [PMID: 16478542 PMCID: PMC1386699 DOI: 10.1186/1476-4598-5-6] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/09/2006] [Accepted: 02/14/2006] [Indexed: 02/06/2023] Open
Abstract
This review explores similarities between lymphocytes and cancer cells, and proposes a new model for the genesis of human cancer. We suggest that the development of cancer requires infection(s) during which antigenic determinants from pathogens mimicking self-antigens are co-presented to the immune system, leading to breaking T cell tolerance. Some level of autoimmunity is normal and necessary for effective pathogen eradication. However, autoreactive T cells must be eliminated by apoptosis when the immune response is terminated. Apoptosis can be deficient in the event of a weakened immune system, the causes of which are multifactorial. Some autoreactive T cells suffer genomic damage in this process, but manage to survive. The resulting cancer stem cell still retains some functions of an inflammatory T cell, so it seeks out sites of inflammation inside the body. Due to its defective constitutive production of inflammatory cytokines and other growth factors, a stroma is built at the site of inflammation similar to the temporary stroma built during wound healing. The cancer cells grow inside this stroma, forming a tumor that provides their vascular supply and protects them from cellular immune response. As cancer stem cells have plasticity comparable to normal stem cells, interactions with surrounding normal tissues cause them to give rise to all the various types of cancers, resembling differentiated tissue types. Metastases form at an advanced stage of the disease, with the proliferation of sites of inflammation inside the body following a similar mechanism. Immunosuppressive cancer therapies inadvertently re-invigorate pathogenic microorganisms and parasitic infections common to cancer, leading to a vicious circle of infection, autoimmunity and malignancy that ultimately dooms cancer patients. Based on this new understanding, we recommend a systemic approach to the development of cancer therapies that supports rather than antagonizes the immune system.
Collapse
|
192
|
Ryan AE, Lane S, Shanahan F, O'Connell J, Houston AM. Fas ligand expression in human and mouse cancer cell lines; a caveat on over-reliance on mRNA data. J Carcinog 2006; 5:5. [PMID: 16457714 PMCID: PMC1373622 DOI: 10.1186/1477-3163-5-5] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/30/2005] [Accepted: 02/02/2006] [Indexed: 01/05/2023] Open
Abstract
Background During carcinogenesis, tumors develop multiple mechanisms for evading the immune response, including upregulation of Fas ligand (FasL/CD95L) expression. Expression of FasL may help to maintain tumor cells in a state of immune privilege by inducing apoptosis of anti-tumor immune effector cells. Recently this idea has been challenged by studies reporting that tumor cells of varying origin do not express FasL. In the present study, we aimed to comprehensively characterize FasL expression in tumors of both murine and human origin over a 72 hour time period. Methods RNA and protein was extracted from six human (SW620, HT29, SW480, KM12SM, HCT116, Jurkat) and three mouse (CMT93, CT26, B16F10) cancer cell lines at regular time intervals over a 72 hour time period. FasL expression was detected at the mRNA level by RT-PCR, using intron spanning primers, and at the protein level by Western Blotting and immunofluorescence, using a polyclonal FasL- specific antibody. Results Expression of FasL mRNA and protein was observed in all cell lines analysed. However, expression of FasL mRNA varied dramatically over time, with cells negative for FasL mRNA at many time points. In contrast, 8 of the 9 cell lines constitutively expressed FasL protein. Thus, cells can abundantly express FasL protein at times when FasL mRNA is absent. Conclusion These findings demonstrate the importance of complete analysis of FasL expression by tumor cells in order to fully characterize its biological function and may help to resolve the discrepancies present in the literature regarding FasL expression and tumor immune privilege.
Collapse
Affiliation(s)
- Aideen E Ryan
- Department of Medicine, National University of Ireland Cork (NUIC), Clinical Science Building, Cork University Hospital, Wilton, Cork, Ireland
| | - Sinead Lane
- Department of Medicine, National University of Ireland Cork (NUIC), Clinical Science Building, Cork University Hospital, Wilton, Cork, Ireland
| | - Fergus Shanahan
- Department of Medicine, National University of Ireland Cork (NUIC), Clinical Science Building, Cork University Hospital, Wilton, Cork, Ireland
- Alimentary Pharmabiotic Centre, National University of Ireland Cork (NUIC), Ireland
| | - Joe O'Connell
- Department of Medicine, National University of Ireland Cork (NUIC), Clinical Science Building, Cork University Hospital, Wilton, Cork, Ireland
- Alimentary Pharmabiotic Centre, National University of Ireland Cork (NUIC), Ireland
| | - Aileen M Houston
- Department of Medicine, National University of Ireland Cork (NUIC), Clinical Science Building, Cork University Hospital, Wilton, Cork, Ireland
- Alimentary Pharmabiotic Centre, National University of Ireland Cork (NUIC), Ireland
| |
Collapse
|
193
|
Hiraoka K, Miyamoto M, Cho Y, Suzuoki M, Oshikiri T, Nakakubo Y, Itoh T, Ohbuchi T, Kondo S, Katoh H. Concurrent infiltration by CD8+ T cells and CD4+ T cells is a favourable prognostic factor in non-small-cell lung carcinoma. Br J Cancer 2006; 94:275-80. [PMID: 16421594 PMCID: PMC2361103 DOI: 10.1038/sj.bjc.6602934] [Citation(s) in RCA: 372] [Impact Index Per Article: 19.6] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 09/06/2005] [Revised: 11/07/2005] [Accepted: 11/30/2005] [Indexed: 01/14/2023] Open
Abstract
The purpose of this study was to clarify the relationship between the number of tumour-infiltrating T lymphocytes and the clinicopathological features and clinical outcome in patients with non-small-cell lung cancer (NSCLC). Tissue specimens from 109 patients who underwent surgical resection for NSCLC were immunohistochemically analysed for CD4 and CD8 expression. Patients were classified into two groups according to whether their tumours exhibited a 'high' or 'low' level of CD8(+) or CD4(+) lymphocyte infiltration. Although the level of infiltration by CD8(+) T cells alone had no prognostic significance, the survival rate for patients with both 'high' CD8(+) and 'high' CD4(+) T-cell infiltration was significantly higher than that for the other groups (log-rank test, P=0.006). Multivariate analysis indicated that concomitant high CD8(+) and high CD4(+) T-cell infiltration was an independent favourable prognostic factor (P=0.0092). In conclusion, the presence of high levels of both CD8(+) T cells and CD4(+) T cells is a significant indicator of a better prognosis for patients with NSCLC, and cooperation between these cell populations may allow a significantly more potent antitumour response than either population alone.
Collapse
Affiliation(s)
- K Hiraoka
- Surgical Oncology, Cancer Medicine, Division of Cancer Medicine, Hokkaido University Graduate School of Medicine, Kita-15, Nishi-7, Kita-ku, Sapporo 060-8638, Japan.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
194
|
Ibrahim R, Frederickson H, Parr A, Ward Y, Moncur J, Khleif SN. Expression of FasL in squamous cell carcinomas of the cervix and cervical intraepithelial neoplasia and its role in tumor escape mechanism. Cancer 2006; 106:1065-77. [PMID: 16456813 DOI: 10.1002/cncr.21697] [Citation(s) in RCA: 19] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022]
Abstract
BACKGROUND To date, several mechanisms have been described by which malignant cells escape from the immune system. One of these is through the expression of FasL. The authors hypothesized that the Fas/FasL interaction enables cervical carcinoma cells to induce apoptosis of the cells of the immune system and thereby escape from them. METHODS The authors tested the expression of FASL on the surface of cervical carcinoma tissues. Next, they stained the same cervical tissues with anti-human leukocyte common antigen and TUNEL to identify apoptotic cells. An in vitro functional assay was then done to test if the FASL expressed on the surface of cervical carcinoma cell lines was or was not responsible for inducing apoptosis in T-cells. Finally, they compared the expression of FASL on normal and dysplastic cervical tissues. RESULTS Ninety-four percent of the cervical carcinoma tissues the authors tested expressed FasL and the majority of the apoptotic cells in the specimens were leukocytes with very few tumor cells. In the in vitro functional assay, only the Fasl expressing cell line and not the Fasl negative cell line was able to induce apoptosis of the Fas-expressing Jurkat cells. On examining the normal cervical tissues, the authors found that the expression of Fasl was confined to the basal cell layer with loss of expression observed in the suprabasal layers, which made it an immune privileged site. Conversely, there was persistent expression of FasL in the dysplastic layers in cervical dysplasia and squamous cell carcinoma specimens. CONCLUSIONS The findings of the current study support the authors' hypothesis that persistent expression of FasL plays a role in the ability of cervical carcinoma cells to escape from the immune system.
Collapse
Affiliation(s)
- Ramy Ibrahim
- Cancer Vaccine Branch, National Cancer Institute, National Institutes of Health, Bethesda, Maryland 20889, USA
| | | | | | | | | | | |
Collapse
|
195
|
Abstract
Autoimmune disease is characterized by clinical symptoms mediated by adaptive (T cell and B cell) immune reactions towards autoantigen-expressing tissue. Here we discuss that autoimmune disease is often preceded by autoreactivity, meaning the priming of autoantigen-specific immune cells without relevant tissue damage. Recent experimental evidence has demonstrated that both the induction of autoreactivity and the conversion into autoimmune disease is controlled by the activation of the nonspecific innate immune system. Also, the "inflammatory status" of the target organ critically influences the onset of overt autoimmune disease.
Collapse
Affiliation(s)
- M Recher
- University Hospital Bruderholz, Institute of Internal Medicine, Switzerland.
| | | |
Collapse
|
196
|
Abstract
The intricate problems associated with the delivery and various unnecessary in vivo transitions of proteins and drugs needs to be tackled soon to be able to exploit the myriad of putative therapeutics created by the biotechnology boom. Nanomedicine is one of the most promising applications of nanotechnology in the field of medicine. It has been defined as the monitoring, repair, construction and control of human biological systems at the molecular level using engineered nanodevices and nanostructures. These nanostructured medicines will eventually turn the world of drug delivery upside down. PEGylation (i.e. the attachment of polyethylene glycol to proteins and drugs) is an upcoming methodology for drug development and it has the potential to revolutionise medicine by drastically improving the pharmacokinetic and pharmacodynamic properties of the administered drug. This article provides a total strategy for improving the therapeutic efficacy of various biotechnological products in drug delivery. This article also presents an extensive analysis of most of the PEGylated proteins, peptides and drugs, together with extensive clinical data. Nanomedicines and PEGylation, the latest offshoots of nanotechnology will definitely pave a way in the field of drug delivery where targeted delivery, formulation, in vivo stability and retention are the major challenges.
Collapse
Affiliation(s)
- Suphiya Parveen
- Laboratory of Nanomedicine, Institute of Life Sciences, Chandrasekharpur, Bhubaneswar, Orissa, India
| | | |
Collapse
|
197
|
Smyth MJ, Dunn GP, Schreiber RD. Cancer Immunosurveillance and Immunoediting: The Roles of Immunity in Suppressing Tumor Development and Shaping Tumor Immunogenicity. Adv Immunol 2006; 90:1-50. [PMID: 16730260 DOI: 10.1016/s0065-2776(06)90001-7] [Citation(s) in RCA: 576] [Impact Index Per Article: 30.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/12/2022]
Abstract
Cellular transformation and tumor development result from an accumulation of mutational and epigenetic changes that alter normal cell growth and survival pathways. For the last 100 years, there has been a vigorous debate as to whether the unmanipulated immune system can detect and eliminate such altered host derived cells despite the fact that cancer cells frequently express either abnormal proteins or abnormal levels of normal cellular proteins that function as tumor antigens. In this review, we discuss the current state of this argument and point out some of the recent key experiments demonstrating that immunity not only protects the host from cancer development (i.e., provides a cancer immunosurveillance function) but also can promote tumor growth, sometimes by generating more aggressive tumors. The terminology "cancer immunoediting" has been used to describe this dual host protective and tumor promoting action of immunity, and herein we summarize the ever-increasing experimental and clinical data that support the validity of this concept.
Collapse
Affiliation(s)
- Mark J Smyth
- Cancer Immunology Program, Peter MacCallum Cancer Centre, East Melbourne, 3002 Victoria, Australia
| | | | | |
Collapse
|
198
|
Abstract
The membrane-bound death ligands CD95L/FasL and TRAIL, which activate the corresponding death receptors CD95/Fas, TRAILR1 and TRAILR2, induce apoptosis in many tumour cells, but can also elicit an inflammatory response. This chapter focuses on the relevance of CD95L/FasL and TRAIL for the tumour surveillance function of natural killer cells and cytotoxic T-cells and discuss current concepts of utilizing these ligands in tumour therapy.
Collapse
Affiliation(s)
- Harald Wajant
- Department of Molecular Internal Medicine, Medical Polyclinic, University of Wuerzburg, Roentgenring 11, 97070 Wuerzburg, Germany
| |
Collapse
|
199
|
Gutierrez LS, Noria F, Finol H, Sun L, Castellino F, Pollard M. Fas Ligand Expression and Its Correlation with Apoptosis and Proliferation in Lobund-Wistar Prostate Carcinomas. Pathobiology 2005; 72:260-8. [PMID: 16374070 DOI: 10.1159/000089420] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/01/2004] [Accepted: 09/06/2005] [Indexed: 11/19/2022] Open
Abstract
OBJECTIVE The Fas (CD95) interaction with its receptor Fas ligand (FasL) is one of the main mechanisms of cell apoptosis. High expression of FasL has been consistently observed in a variety of human cancers. In this study, we evaluated FasL and its relationship with apoptosis and proliferation in Lobund-Wistar (L-W) cancers. The L-W rat strain develops spontaneous and induced adenocarcinomas in the anterior prostate and seminal vesicles. Although FasL expression has been observed in a subset of human prostate carcinomas, this multistage model allowed in vivo evaluation of subclones of malignant cells with a single genetic susceptibility. METHODS Apoptosis was evaluated in spontaneous, induced and transplanted tumors as well as metastasis using the terminal deoxynucleotidyl transferase-mediated dUTP nick end-labeling (TUNEL) technique and transmission electron microscopy. Proliferating cell nuclear antigen (PCNA) and FasL expression were detected using immunohistochemistry and analyzed according to the number of positive cells and intensity of staining using a semiquantitive method. RESULTS Apoptotic indexes were significantly higher in spontaneous tumors compared to induced (p < 0.008), transplanted tumors (p < 0.0112) and metastases (p < 0.009). TUNEL-positive cells were frequently observed in the leukocytic infiltrate of the stroma in transplanted carcinomas and metastases. These findings were confirmed by electron microscopy. FasL expression was not uniformly localized in L-W carcinomas and its highest expression was observed in transplanted tumors and metastasis (p < 0.005). Moreover, PCNA indices were directly correlated with cancers showing high FasL total scores (Hscores). CONCLUSIONS In this model, high FasL expression was associated with cells displaying low apoptotic indexes and high PCNA index. Therefore, analysis of FasL may have clinical relevance in detecting the malignant potential of prostate cancers.
Collapse
|
200
|
Hofbauer GFL, Hatta N, Daigle I, Hemmi S, Spanaus Schlapbach K, Willers J, Burg G, Simon HU, Dummer R. Fas Ligand Reduces Viability in Primary Melanoma Short-Term Cell Cultures More than in Metastatic Melanoma Short-Term Cell Cultures. Dermatology 2005; 211:318-24. [PMID: 16286739 DOI: 10.1159/000088500] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/15/2004] [Accepted: 01/15/2005] [Indexed: 11/19/2022] Open
Abstract
BACKGROUND Apoptotic pathway aberrations are reported as important tumor progression factors in melanoma. OBJECTIVE Effect of soluble Fas ligand (sFasL) and tumor necrosis factor-related apoptosis-inducing ligand (TRAIL) on short-term cultured melanoma cell viability from different stages of melanoma. RESULTS Recombinant human FasL reduced viability after 18 h in a dose-dependent manner in 4 of 5 cell cultures from primary tumors and 1 of 9 cell cultures from metastatic melanoma (67.5 vs. 96.4%, p = 0.007). DNA fragmentation on flow cytometry confirmed apoptosis. Incubation with TRAIL had no effect on melanoma cell viability. Immunohistochemistry showed Fas in 3 of 4 primary and in 6 of 7 metastatic lesions, no FasL in primary lesions, and FasL in 5 of 7 metastatic lesions. CONCLUSION Melanoma short-term cell cultures from primary tumors show decreased viability under FasL, but not TRAIL stimulation rather than short-term cell cultures derived from metastases.
Collapse
|