1
|
Affiliation(s)
- Stefan K Burgdorf
- Department of Surgical Gastroenterology Z2, Gentofte Hospital, University of Copenhagen, Denmark.
| | | | | |
Collapse
|
2
|
Sogo T, Kawahara M, Ueda H, Otsu M, Onodera M, Nakauchi H, Nagamune T. T cell growth control using hapten-specific antibody/interleukin-2 receptor chimera. Cytokine 2009; 46:127-36. [PMID: 19223197 DOI: 10.1016/j.cyto.2008.12.020] [Citation(s) in RCA: 29] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2008] [Revised: 12/10/2008] [Accepted: 12/29/2008] [Indexed: 11/27/2022]
Abstract
IL-2 is a cytokine that is essential for the expansion and survival of activated T cells. Although adoptive transfer of tumor-specific T cells with IL-2 is one of strategies for cancer immunotherapy, it is essential to replace IL-2 that exerts severe side effects in vivo. To solve this problem, we propose to use an antibody/IL-2R chimera, which can transduce a growth signal in response to a cognate antigen. We constructed two chimeras, in which ScFv of anti-fluorescein antibody was tethered to extracellular D2 domain of erythropoietin receptor and transmembrane/cytoplasmic domains of IL-2Rbeta or gamma chain. When the chimeras were co-expressed in IL-3-dependent pro-B cell line Ba/F3 and IL-2-dependent T cell line CTLL-2, gene-modified cells were selectively expanded in the absence of IL-3 and IL-2, respectively, by adding fluorescein-conjugated BSA (BSA-FL) as a cognate antigen. Growth assay revealed that the cells with the chimeras transduced a growth signal in a BSA-FL dose-dependent manner. Furthermore, STAT3, STAT5, ERK1/2 and Akt, which are hallmarks for IL-2R signaling, were all activated by the chimeras in CTLL-2 transfectant. We also demonstrated that the chimeras were functional in murine primary T cells. These results demonstrate that the antibody/IL-2R chimeras could substantially mimic the wild-type IL-2R and could specifically expand gene-modified T cells in the presence of the cognate antigen.
Collapse
Affiliation(s)
- Takahiro Sogo
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo 113-8656, Japan
| | | | | | | | | | | | | |
Collapse
|
3
|
Abstract
The ability to measure biochemical and molecular processes underlies progress in breast cancer biology and treatment. These assays have traditionally been performed by analysis of cell culture or tissue samples. More recently, functional and molecular imaging has allowed the in vivo assay of biochemistry and molecular biology, which is highly complementary to tissue-based assays. This review briefly describes different imaging modalities used in molecular imaging and then reviews applications of molecular imaging to breast cancer, with a focus on translational work. It includes sections describing work in functional and physiological tumor imaging, imaging gene product expression, imaging the tumor microenvironment, reporter gene imaging, and cell labeling. Work in both animal models and human is discussed with an eye towards studies that have relevance to breast cancer treatment in patients.
Collapse
Affiliation(s)
- David A Mankoff
- Seattle Cancer Care Alliance and University of Washington, Radiology, Seattle, WA 98109, USA.
| |
Collapse
|
4
|
Demirtzoglou FJ, Papadopoulos S, Zografos G. Cytolytic and Cytotoxic Activity of a Human Natural Killer Cell Line Genetically Modified to Specifically Recognize HER-2/neu Overexpressing Tumor Cells. Immunopharmacol Immunotoxicol 2008; 28:571-90. [PMID: 17190735 DOI: 10.1080/08923970601066971] [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: 10/23/2022]
Abstract
NK92 cells genetically engineered to recognize the HER-2/neu oncoprotein have been previously reported to lyse HER-2/neu positive tumor cell lines through direct cell to cell contact. In the present study we have transduced NK92 cells with a chimeric receptor gene composed of the HER-/neu specific scFv (FRP5) antibody fragment, joined to the peptide CD8 hinge region and the signaling CD3 zeta chain. NK92 cells expressing this chimeric receptor (NK92.HER-2/neu/zeta) specifically recognized and lysed HER-2/neu overexpressing tumor cell lines both in vitro and in preclinical tumor models in vivo. More important we demonstrate that NK92.HER-2/neu/zeta cells constitutively secrete high levels of soluble scFv which mediate strong tumor cytostatic effects by directly binding on cell surface HER-2/neu. Our data uncover an additional mechanism through which NK92.HER-2/neu/zeta cells mediate antitumor effects and further support their use in cell based therapeutics for the treatment of HER-2/neu expressing cancers.
Collapse
MESH Headings
- Animals
- Antibodies, Monoclonal/biosynthesis
- Antibodies, Monoclonal/genetics
- Blotting, Western
- CD3 Complex/genetics
- Cell Line, Tumor
- Cell Proliferation
- Cell Survival
- Exotoxins/genetics
- Female
- Fluorescent Antibody Technique
- Humans
- Killer Cells, Natural/physiology
- Mice
- Mice, SCID
- Neoplasms/metabolism
- Neoplasms/pathology
- Organisms, Genetically Modified
- Receptor, ErbB-2/biosynthesis
- Receptor, ErbB-2/genetics
- Recombinant Fusion Proteins/genetics
- Single-Chain Antibodies
- T-Lymphocytes, Cytotoxic/physiology
- Transduction, Genetic
Collapse
Affiliation(s)
- F J Demirtzoglou
- Cancer Immunology and Immunotherapy Center, Saint Savas Cancer Hospital, Athens, Greece.
| | | | | |
Collapse
|
5
|
Sogo T, Kawahara M, Tsumoto K, Kumagai I, Ueda H, Nagamune T. Selective expansion of genetically modified T cells using an antibody/interleukin-2 receptor chimera. J Immunol Methods 2008; 337:16-23. [PMID: 18589435 DOI: 10.1016/j.jim.2008.05.003] [Citation(s) in RCA: 12] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/11/2007] [Revised: 04/25/2008] [Accepted: 05/06/2008] [Indexed: 10/22/2022]
Abstract
Although adoptive transfer of tumor-specific T cells is a plausible approach for cancer immunotherapy, the therapeutic application was hampered due to severe side effects caused by administration of high-dose interleukin (IL)-2, which was used for long-lasting maintenance of tumor-specific T cells in vivo. To solve this problem, here we propose to use an antibody/IL-2 receptor chimera, which can transduce a growth signal in response to a cognate antigen. As a model system, V(H) or V(L) region of anti-hen egg lysozyme (HEL) antibody HyHEL-10 was tethered to extracellular D2 domain of erythropoietin receptor and transmembrane/cytoplasmic domains of IL-2 receptor beta or gamma chain. When the pairs of chimeric receptors (V(H)-IL-2Rbeta and V(L)-IL-2Rgamma, or V(H)-IL-2Rgamma and V(L)-IL-2Rbeta) were expressed in IL-3-dependent pro-B cell line Ba/F3 and IL-2-dependent T cell line CTLL-2, the cognate antigen HEL induced selective expansion of gene-modified cells in the absence of IL-3 and IL-2, respectively. Growth assay revealed that the combination of V(H)-IL-2Rbeta and V(L)-IL-2Rgamma transduced a more stringent HEL-dependent growth signal, indicating some conformational effects of the chimeras. Furthermore, STAT3, STAT5 and ERK1/2, which are hallmarks for IL-2R signaling, were all activated by the antibody/IL-2R chimeras. These results clearly demonstrate that the antibody/IL-2R chimeras could substantially mimic the wild-type IL-2R signaling, suggesting the potential application in expansion of gene-modified T cells.
Collapse
Affiliation(s)
- Takahiro Sogo
- Department of Chemistry and Biotechnology, School of Engineering, The University of Tokyo, 7-3-1 Hongo, Bunkyo-ku, Tokyo, Japan
| | | | | | | | | | | |
Collapse
|
6
|
Savai R, Schermuly RT, Pullamsetti SS, Schneider M, Greschus S, Ghofrani HA, Traupe H, Grimminger F, Banat GA. A combination hybrid-based vaccination/adoptive cellular therapy to prevent tumor growth by involvement of T cells. Cancer Res 2007; 67:5443-53. [PMID: 17545626 DOI: 10.1158/0008-5472.can-06-3677] [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: 11/16/2022]
Abstract
Cancer immunotherapy with dendritic cell-tumor cell fusion hybrids induces polyclonal stimulation against a variety of tumor antigens, including unknown antigens. Hybrid cells can prime CTLs, which subsequently develop antitumor responses. The aim of this study was to enhance the known antitumor effect of hybrid vaccination (HC-Vacc) and hybrid-primed adoptive T-cell therapy (HC-ACT) using the poorly immunogenic Lewis lung carcinoma (LLC1) model. The strategy used was a combination of a double HC-Vacc alternating with HC-ACT (HC-Vacc/ACT). Using flat-panel volumetric computer tomography and immunohistochemistry, we showed a significant retardation of tumor growth (85%). In addition, a significant delay in tumor development, a reduction in the number of pulmonary metastases, and increased survival times were observed. Furthermore, the tumors displayed significant morphologic changes and increased apoptosis, as shown by up-regulation of gene expression of the proapoptotic markers Fas, caspase-8, and caspase-3. The residual tumor masses seen in the HC-Vacc/ACT-treated mice were infiltrated with CD4+ and CD8+ lymphocytes and showed elevated IFNgamma expression. Moreover, splenic enlargement observed in HC-Vacc/ACT-treated mice reflected the increased functionality of T cells, as also indicated by increased expression of markers for CTL activation, differentiation, and proliferation (Cd28, Icosl, Tnfrsf13, and Tnfsf14). Our findings indicate that the combination therapy of dendritic cell-tumor cell HC-Vacc/ACT is a very effective and a promising immunotherapeutic regimen against poorly immunogenic carcinomas.
Collapse
MESH Headings
- Animals
- Apoptosis/genetics
- Cancer Vaccines/immunology
- Cancer Vaccines/pharmacology
- Carcinoma, Lewis Lung/genetics
- Carcinoma, Lewis Lung/immunology
- Carcinoma, Lewis Lung/pathology
- Carcinoma, Lewis Lung/therapy
- Cell Fusion/methods
- Dendritic Cells/cytology
- Dendritic Cells/immunology
- Female
- Gene Expression
- Hybrid Cells/immunology
- Immunotherapy, Adoptive/methods
- Lung Neoplasms/secondary
- Lymphocyte Activation
- Melanoma, Experimental/genetics
- Melanoma, Experimental/immunology
- Melanoma, Experimental/pathology
- Melanoma, Experimental/therapy
- Mice
- Mice, Inbred BALB C
- Mice, Inbred C57BL
- T-Lymphocytes/immunology
- T-Lymphocytes, Cytotoxic/immunology
Collapse
Affiliation(s)
- Rajkumar Savai
- Department of Hematology, Universitätsklinikum Giessen und Marburg GmbH, Giessen, Germany
| | | | | | | | | | | | | | | | | |
Collapse
|
7
|
Shilyansky J, Jacobs P, Doffek K, Sugg SL. Induction of cytolytic T lymphocytes against pediatric solid tumors in vitro using autologous dendritic cells pulsed with necrotic primary tumor. J Pediatr Surg 2007; 42:54-61; discussion 61. [PMID: 17208541 DOI: 10.1016/j.jpedsurg.2006.09.008] [Citation(s) in RCA: 11] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
PURPOSE Effective and generally applicable methods for generating cancer vaccines in children have not been defined. Dendritic cells (DCs) are the most potent professional antigen-presenting cells capable of activating primary cytolytic T cells. We tested the ability of DCs generated from pediatric patients' peripheral blood monocytes and pulsed with a necrotic tumor to activate autologous tumor-specific cytolytic T cells. METHODS Tumor and peripheral blood cells were obtained from pediatric patients undergoing biopsy or resection for advanced solid tumors according to an institutional research board-approved protocol and after acquiring informed consent from them. To generate DCs, we treated peripheral blood monocytes with granulocyte-macrophage colony stimulating factor and interleukin (IL)-4. Maturation was induced with a cytokine cocktail (CC) containing tumor necrosis factor-alpha, IL-6, IL-1beta, and prostaglandin E2. The DC phenotype was assayed using flow cytometry. Tumor necrosis was induced by exposure to UV-B irradiation (1000 mJ). Dendritic cells pulsed with a UV-B-treated primary tumor and matured with CC were used to stimulate autologous peripheral blood lymphocytes weekly. Tumor-specific cytolytic activity was assayed using 4-hour 51Cr release. RESULTS Peripheral blood monocytes isolated from pediatric patients differentiated into immature DCs (CD14-, MHCII+ [major histocompatibility complex], CD80(low), CD86(low)) in the presence of granulocyte-macrophage colony stimulating factor and IL-4. Cytokine cocktail induced maturation of DCs, as characterized by increased expressions of MHCII, CD83, CD80, and CD86. Patients' peripheral blood lymphocytes stimulated in vitro with DCs loaded with a necrotic primary tumor and matured with CC specifically lysed autologous neuroblastoma in 7 of 9 patients. CONCLUSION Dendritic cells generated from the peripheral blood of children with advanced solid tumors and pulsed with a necrotic primary tumor undergo maturation and effectively stimulate autologous tumor-specific cytolytic T cells in vitro. We describe a simple method for generating a vaccine capable of activating cytotoxic T cells against pediatric solid tumors that does not require the genetic identification of tumor-associated antigens.
Collapse
Affiliation(s)
- Joel Shilyansky
- Division of Pediatric Surgery, Department of Surgery, Medical College of Wisconsin, Milwaukee, WI 53226, USA
| | | | | | | |
Collapse
|
8
|
Neves AA, Brindle KM. Assessing responses to cancer therapy using molecular imaging. Biochim Biophys Acta Rev Cancer 2006; 1766:242-61. [PMID: 17140737 DOI: 10.1016/j.bbcan.2006.10.002] [Citation(s) in RCA: 38] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 06/21/2006] [Revised: 10/12/2006] [Accepted: 10/16/2006] [Indexed: 01/09/2023]
Abstract
Tumor responses to therapy in the clinic are still evaluated primarily from non-invasive imaging measurements of reductions in tumor size. This approach, however, lacks sensitivity and can only give a delayed indication of a positive response to treatment. Major advances in our understanding of the molecular mechanisms responsible for cancer, combined with new targeted clinical imaging technologies designed to detect the molecular correlates of disease progression and response to treatment, are set to revolutionize our approach to the detection and treatment of the disease. We describe here the imaging technologies available to image tumor cell proliferation and migration, metabolism, receptor and gene expression, apoptosis and tumor angiogenesis and vascular function, and show how measurements of these parameters can be used to give early indications of positive responses to treatment or to detect drug resistance and/or disease recurrence. Special emphasis has been placed on those applications that are already used in the clinic and those that are likely to translate into clinical application in the near future or whose use in preclinical studies is likely to facilitate translation of new treatments into the clinic.
Collapse
Affiliation(s)
- André A Neves
- Department of Biochemistry, University of Cambridge, Cambridge CB2 1GA, UK
| | | |
Collapse
|
9
|
Hicks AM, Riedlinger G, Willingham MC, Alexander-Miller MA, Von Kap-Herr C, Pettenati MJ, Sanders AM, Weir HM, Du W, Kim J, Simpson AJG, Old LJ, Cui Z. Transferable anticancer innate immunity in spontaneous regression/complete resistance mice. Proc Natl Acad Sci U S A 2006; 103:7753-8. [PMID: 16682640 PMCID: PMC1458507 DOI: 10.1073/pnas.0602382103] [Citation(s) in RCA: 115] [Impact Index Per Article: 6.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/18/2022] Open
Abstract
Spontaneous regression/complete resistance (SR/CR) mice resist very high doses of cancer cells that are lethal to WT mice even at low doses. In this study, we show that this resistance is mediated by rapid infiltration of leukocytes, mostly of innate immunity, in both primary and repeated challenges. Formation of rosettes with infiltrating natural killer cells, neutrophils, and macrophages was required for the subsequent destruction of cancer cells through rapid cytolysis. Highly purified natural killer cells, macrophages, and neutrophils from the SR/CR mice independently killed cancer cells in vitro. The independent killing activity by each subset of effector cells is consistent with the observation that the resistance was abolished by depleting total infiltrating leukocytes but not by depleting only one or two subsets of leukocytes. The resistance was completely transferable to WT recipient mice through SR/CR splenocytes, bone marrow cells, or enriched peritoneal macrophages, either for prevention against subsequent cancer challenges or eradication of established malignancy at distant sites.
Collapse
Affiliation(s)
| | | | | | | | - C. Von Kap-Herr
- Department of Pediatrics, Section on Medical Genetics, Wake Forest University School of Medicine, Winston-Salem, NC 27157; and
| | - Mark J. Pettenati
- Department of Pediatrics, Section on Medical Genetics, Wake Forest University School of Medicine, Winston-Salem, NC 27157; and
| | | | | | - Wei Du
- Departments of *Pathology
| | | | | | - Lloyd J. Old
- Ludwig Institute for Cancer Research, New York, NY 10158
- To whom correspondence may be addressed. E-mail:
or
| | - Zheng Cui
- Departments of *Pathology
- To whom correspondence may be addressed. E-mail:
or
| |
Collapse
|
10
|
Abstract
Cancer is a difficult target for any therapeutic strategy; therefore, there is a continuous search for new therapeutic modalities, for application either alone or in combination. In this regard, gene-based therapy is a new approach that offers hope of improved control of tumors. Intensive research to apply gene therapy for cancer treatment has led to identification of the most important technical and theoretical barriers that need to be overcome for clinical success. One of the central unresolved challenges remains the issue of specific and efficient delivery of genes to target cells or tissues, emphasizing the importance of the gene carrier. Along with different viral and non-viral vector systems, mammalian cells have also been considered as vehicles for delivery of anti-cancer therapeutics. The cell-based delivery approach was introduced as the first attempt to apply gene therapy to cancer treatment, and in general, has followed most of the ups and downs of gene therapy applications, progressing alongside new knowledge gained in this field. As a result, significant progress has been made in some aspects of the cell-based approach, while the development of other essential issues is only just gaining speed. It appears that the initial phase of development of cell-based protocols - the achievement of efficient ex vivo cell loading with therapeutics - has largely been fulfilled. However, the desired efficacy of cell-based strategies in general has not yet been reached, and specificity of tumor homing needs to be improved considerably. There is hope that advances in related scientific fields will promote the utilization of cells as powerful and versatile vehicles for cancer gene therapy.
Collapse
Affiliation(s)
- Larisa Pereboeva
- Division of Human Gene Therapy, Department of Medicine, The Gene Therapy Center, BMRII-572, University of Alabama at Birmingham, 901 19th Street S., Birmingham, AL 35294, USA.
| | | |
Collapse
|
11
|
Wang J, Press OW, Lindgren CG, Greenberg P, Riddell S, Qian X, Laugen C, Raubitschek A, Forman SJ, Jensen MC. Cellular immunotherapy for follicular lymphoma using genetically modified CD20-specific CD8+ cytotoxic T lymphocytes. Mol Ther 2004; 9:577-86. [PMID: 15093188 DOI: 10.1016/j.ymthe.2003.12.011] [Citation(s) in RCA: 71] [Impact Index Per Article: 3.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 10/24/2003] [Accepted: 12/18/2003] [Indexed: 11/24/2022] Open
Abstract
Humoral immunotherapy using the monoclonal anti-CD20 antibody rituximab induces remissions in approximately 60% of patients with relapsed follicular lymphoma; however, most patients eventually relapse despite continued expression of CD20 on lymphoma cells. We have hypothesized that cellular immunotherapy targeting CD20(+) cells might provide a more effective mechanism for eliminating lymphoma cells than anti-CD20 antibodies and are therefore investigating the utility of cytotoxic T lymphocytes (CTL) genetically modified to target the CD20 antigen. Peripheral blood mononuclear cells were activated with anti-CD3 antibody (OKT3) and recombinant human interleukin-2 and electroporated with a plasmid containing a CD20-specific scFvFc:zeta chimeric T cell receptor gene and a neomycin phosphotransferase gene (neo(R)). Transfected cells were selected using the antibiotic G418 and cloned by limiting dilution. Using this approach, we have generated CD8(+) CTL clones with CD20-specific cytotoxicity, which specifically lysed CD20(+) target cells, including actual tumor cells from patients with follicular lymphoma, small lymphocytic lymphoma, splenic marginal zone lymphoma, diffuse large B cell lymphoma, and chronic lymphocytic leukemia. The CTL clones have been expanded to numbers sufficient for therapy ( approximately 10(9) cells). Our data indicate the feasibility of generating and expanding CD20-specific CTL and, for the first time, demonstrate that such CTL exhibit specific cytotoxicity against actual tumor cells isolated from patients with a variety of B lymphoid malignancies. In view of these promising findings, a Phase I clinical trial for relapsed follicular lymphoma is being initiated.
Collapse
MESH Headings
- Antigens, CD20/biosynthesis
- Antigens, CD20/metabolism
- Blotting, Southern
- Blotting, Western
- CD3 Complex/chemistry
- CD8-Positive T-Lymphocytes/cytology
- CD8-Positive T-Lymphocytes/metabolism
- Caspases/metabolism
- Cell Line, Tumor
- Cell Membrane/metabolism
- Cells, Cultured
- Chromium/metabolism
- Cloning, Molecular
- Electroporation
- Flow Cytometry
- Gene Transfer Techniques
- Genetic Therapy/methods
- Humans
- Immunotherapy/methods
- Interleukin-2/metabolism
- Kanamycin Kinase/metabolism
- Lymph Nodes/pathology
- Lymphoma/metabolism
- Lymphoma, Follicular/therapy
- Plasmids/metabolism
- Receptors, Antigen, T-Cell/metabolism
- T-Lymphocytes/metabolism
- Time Factors
- Transfection
- Transgenes
Collapse
Affiliation(s)
- Jinjuan Wang
- Fred Hutchinson Cancer Research Center, 1100 Fairview Avenue North, Seattle, WA 98109, USA
| | | | | | | | | | | | | | | | | | | |
Collapse
|
12
|
Knutson KL, Curiel TJ, Salazar L, Disis ML. Immunologic principles and immunotherapeutic approaches in ovarian cancer. Hematol Oncol Clin North Am 2003; 17:1051-73. [PMID: 12959191 DOI: 10.1016/s0889-8588(03)00064-9] [Citation(s) in RCA: 16] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/22/2023]
Abstract
Ovarian cancer is an immunogenic tumor, and numerous antigens have been identified in recent years. Several of these antigens are important in regulating tumor growth and may be ideal targets for the development of immune-based strategies. In the absence of immunologic intervention, tumors evade the immune system by several mechanisms, most notably tolerance and immunosuppression. As understanding of the immune response improves, strategies are being designed to circumvent T-cell tolerance to self-antigens through modulation of APC function. In addition, techniques are being developed to identify reverse ovarian cancer-induced immune evasion tactics. The type of the immune-based therapy to apply varies with disease burden. It is hoped that discoveries at the bench along with lessons learned in prior clinical trials soon will allow clinicians to develop rationally based immunologic strategies to treat and prevent ovarian cancer.
Collapse
Affiliation(s)
- Keith L Knutson
- Tumor Vaccine Group, 1959 Northeast Pacific Street, Box 356527, HSB BB1321, University of Washington, Seattle, WA 98195, USA.
| | | | | | | |
Collapse
|
13
|
Gritzapis AD, Mamalaki A, Kretsovali A, Papamatheakis J, Belimezi M, Perez SA, Baxevanis CN, Papamichail M. Redirecting mouse T hybridoma against human breast and ovarian carcinomas: in vivo activity against HER-2/neu expressing cancer cells. Br J Cancer 2003; 88:1292-300. [PMID: 12698199 PMCID: PMC2747561 DOI: 10.1038/sj.bjc.6600888] [Citation(s) in RCA: 8] [Impact Index Per Article: 0.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022] Open
Abstract
Chimeric receptors comprising of the T-cell receptor-zeta cytoplasmic signalling chain fused to an extracellular ligand-binding domain of a single-chain antibody (scFv) have served as effective tools for redirecting cytotoxic T lymphocytes (CTL) against tumour cells. In this report, we constructed a chimeric scFv/zeta gene composed of the variable regions of an HER-2/neu-specific monoclonal antibody (MAb) joined to the TCR-zeta chain. The scFv(anti-HER-2/neu)/zeta chimeric gene was successfully expressed as a functional surface receptor in the MD.45 CTL hybridoma (MD.45-HER/zeta). More importantly, the scFv(anti-HER-2/neu)/zeta receptor was functionally active, since it triggered cytokine secretion by the MD.45-HER/zeta cells upon recognition of HER-2/neu-positive (+) tumour cell lines, or primary tumour cells from patients with HER-2/neu(+) cancers. The MD.45-HER/zeta-transduced cells also lysed HER-2/neu(+) target cells in vitro with high specificity. We tested the antitumour efficacy of scFv(anti-HER-2/neu)/zeta expressing MD.45 cells in severe combined immunodeficiency disease mice/human and murine tumour models. The adoptively transferred MD.45-HER/zeta cells both slowed significantly the growth of human FM3 melanoma or murine ALC leukaemic cells both transfected to express HER-2/neu. Our data demonstrate the feasibility of redirecting MD.45 CTL with the scFv(anti-HER-2/neu)/zeta chimeric receptor to respond specifically against HER-2/neu expressing tumour cells in vitro and in vivo. Moreover, they make it likely that T cells transduced with the same chimeric gene might be utilised in the treatment of patients with HER-2/neu(+) tumours.
Collapse
Affiliation(s)
- A D Gritzapis
- Saint Savas Cancer Hospital, Cancer Immunology and Immunotherapy Center, 171 Alexandras Ave, 11522 Athens, Greece
| | - A Mamalaki
- Department of Biochemistry, Hellenic Pasteur Institute, Athens, Greece
| | - A Kretsovali
- Foundation for Research and Technology, Institute of Molecular Biology and Biotechnology, Heraklion, Crete
| | - J Papamatheakis
- Foundation for Research and Technology, Institute of Molecular Biology and Biotechnology, Heraklion, Crete
| | - M Belimezi
- Department of Biochemistry, Hellenic Pasteur Institute, Athens, Greece
| | - S A Perez
- Saint Savas Cancer Hospital, Cancer Immunology and Immunotherapy Center, 171 Alexandras Ave, 11522 Athens, Greece
| | - C N Baxevanis
- Saint Savas Cancer Hospital, Cancer Immunology and Immunotherapy Center, 171 Alexandras Ave, 11522 Athens, Greece
- Saint Savas Cancer Hospital, Cancer Immunology and Immunotherapy Center, 171 Alexandras Ave, 11522 Athens, Greece. E-mail:
| | - M Papamichail
- Saint Savas Cancer Hospital, Cancer Immunology and Immunotherapy Center, 171 Alexandras Ave, 11522 Athens, Greece
| |
Collapse
|