101
|
Turner MS, McKolanis JR, Ramanathan RK, Whitcomb DC, Finn OJ. Mucins in gastrointestinal cancers. CANCER CHEMOTHERAPY AND BIOLOGICAL RESPONSE MODIFIERS 2003; 21:259-74. [PMID: 15338749 DOI: 10.1016/s0921-4410(03)21012-7] [Citation(s) in RCA: 10] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/02/2023]
Abstract
The mucin family has been under study by molecular biologists, biochemists, pathologists and immunologists interested in cancer because of the role these molecules can play in the diagnosis and treatment of cancer. Immense knowledge has been accumulated, but the high speed of progress in the laboratory has not been matched by the progress towards applying this knowledge in the clinic. For example, specific knowledge of cancer-associated changes in the expression and glycosylation of various mucins, which can aid in the diagnosis as well as prognosis of GI cancers, has not yet led to the use of a panel of anti-mucin antibodies as a standard diagnostic tool. Similarly, many more opportunities exist for using mucin-based therapies than are currently being considered in the clinic. This chapter aimed to highlight some of these opportunities and to interest clinician scientists in exploring them in the near future.
Collapse
Affiliation(s)
- Michael S Turner
- Department of Immunology, University of Pittsburgh School of Medicine, Cancer Center, PA 15261, USA
| | | | | | | | | |
Collapse
|
102
|
Becker M, Nitsche A, Neumann C, Aumann J, Junghahn I, Fichtner I. Sensitive PCR method for the detection and real-time quantification of human cells in xenotransplantation systems. Br J Cancer 2002; 87:1328-35. [PMID: 12439725 PMCID: PMC2408903 DOI: 10.1038/sj.bjc.6600573] [Citation(s) in RCA: 93] [Impact Index Per Article: 4.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2002] [Revised: 08/05/2002] [Accepted: 08/15/2002] [Indexed: 11/20/2022] Open
Abstract
The sensitive detection of human cells in immunodeficient rodents is a prerequisite for the monitoring of micrometastasis of solid tumours, dissemination of leukaemic cells, or engraftment of haematological cells. We developed a universally applicable polymerase chain reaction method for the detection of a human-specific 850-bp fragment of the alpha-satellite DNA on human chromosome 17. The method allows the detection of one human cell in 10(6) murine cells and could be established as both, a conventional DNA polymerase chain reaction-assay for routine screening, and a quantitative real-time polymerase chain reaction-assay using TaqMan-methodology. It was applied to the following xenotransplantation systems in SCID and NOD/SCID mice: (1) In a limiting dilution assay, cells of the MDA-MB 435 breast carcinoma were injected into the mammary fat pad of NOD/SCID mice. It could be shown that 10 cells mouse(-1) were sufficient to induce a positive polymerase chain reaction signal in liver and lung tissue 30 days after transplantation as an indicator for micrometastasis. At this time a palpable tumour was not yet detectable in the mammary fat pad region. (2) Cells of a newly established human acute lymphatic leukaemia were administered intraperitoneally to SCID mice. These cells apparently disseminated and were detectable as early as day 50 in the peripheral blood of living mice, while the leukaemia manifestation was delayed by day 140. (3) In a transplantation experiment using mature human lymphocytes we wanted to standardise conditions for a successful survival of these cells in NOD/SCID mice. It was established that at least 5 x 10(7) cells given intravenously were necessary and that the mice had to be conditioned by 2 Gy body irradiation to get positive polymerase chain reaction bands in several organs. (4) Engraftment studies with blood stem cells originating from cytapheresis samples of tumour patients or from cord blood were undertaken in NOD/SCID mice in order to define conditions of successful engraftment and to use this model for further optimisation strategies. The polymerase chain reaction method presented allowed a reliable prediction of positive engraftment and agreed well with the results of immunohistochemical or FACS analysis. All together, the polymerase chain reaction method developed allows a sensitive and reliable detection of low numbers of human cells in immunodeficient hosts. In combination with real-time (TaqMan) technique it allows an exact quantification of human cells. As this method can be performed with accessible material of living animals, follow up studies for the monitoring of therapeutic interventions are possible in which the survival time of mice as evaluation criteria can be omitted.
Collapse
Affiliation(s)
- M Becker
- Max-Delbrück-Center for Molecular Medicine, Robert-Rössle-Strasse 10, 13092 Berlin, Germany
| | | | | | | | | | | |
Collapse
|
103
|
Wykes M, MacDonald KPA, Tran M, Quin RJ, Xing PX, Gendler SJ, Hart DNJ, McGuckin MA. MUC1 epithelial mucin (CD227) is expressed by activated dendritic cells. J Leukoc Biol 2002. [DOI: 10.1189/jlb.72.4.692] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- M. Wykes
- Dendritic Cell, Mater Medical Research Institute, Mater Misericordiae Hospitals, South Brisbane, Australia and
| | - K. P. A. MacDonald
- Dendritic Cell, Mater Medical Research Institute, Mater Misericordiae Hospitals, South Brisbane, Australia and
| | - M. Tran
- Cancer Characterisation Laboratories, Mater Medical Research Institute, Mater Misericordiae Hospitals, South Brisbane, Australia
| | - R. J. Quin
- Cancer Characterisation Laboratories, Mater Medical Research Institute, Mater Misericordiae Hospitals, South Brisbane, Australia
| | - P. X. Xing
- Austin Research Institute, Heidelberg, Australia; and
| | | | - D. N. J. Hart
- Dendritic Cell, Mater Medical Research Institute, Mater Misericordiae Hospitals, South Brisbane, Australia and
| | - M. A. McGuckin
- Cancer Characterisation Laboratories, Mater Medical Research Institute, Mater Misericordiae Hospitals, South Brisbane, Australia
| |
Collapse
|
104
|
Evangelou A, Letarte M, Marks A, Brown TJ. Androgen modulation of adhesion and antiadhesion molecules in PC-3 prostate cancer cells expressing androgen receptor. Endocrinology 2002; 143:3897-904. [PMID: 12239101 DOI: 10.1210/en.2002-220156] [Citation(s) in RCA: 28] [Impact Index Per Article: 1.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/17/2023]
Abstract
The metastatic spread of cancer cells involves a complex process of detachment via antiadhesion molecules and attachment and migration through adhesion. In the prostate, androgens are generally thought to contribute to the development and progression of prostate cancer by promoting cell proliferation and survival through poorly defined mechanisms. We have reported previously that PC-3 prostate cancer cells, which are unresponsive to androgens, show androgen-dependent detachment and ultimately apoptosis when stably transfected with a full-length human androgen receptor (AR) cDNA. We now demonstrate that treatment of these cells with 5alpha-dihydrotestosterone (DHT) for 24 or 48 h increased the expression of antiadhesion mucin MUC-1 at the cell surface as detected by flow cytometry with two independent antibodies. This increase in protein was concordant with up-regulation of MUC-1 mRNA in the AR-transfected PC-3 sublines, as determined by quantitative RT-PCR. Treatment with DHT for 48 h also down-regulated the cell surface expression of alpha2beta1-integrin but having little effect on the levels of alpha3beta1- and alpha5beta1-integrins. Androgen also decreased, in a dose-dependent manner, the adhesion of AR-transfected PC-3 cells to collagen type I, which was shown to be specifically inhibited by blocking antibody to alpha2beta1-integrin. The present data demonstrate that DHT can modulate expression of adhesion and antiadhesion molecules and suggest that this effect of androgen might contribute to prostate cancer progression.
Collapse
Affiliation(s)
- Andreas Evangelou
- Cancer and Blood Research Program, The Hospital for Sick Children, Toronto, Ontario, Canada M5G 1X8
| | | | | | | |
Collapse
|
105
|
Müller S, Hanisch FG. Recombinant MUC1 probe authentically reflects cell-specific O-glycosylation profiles of endogenous breast cancer mucin. High density and prevalent core 2-based glycosylation. J Biol Chem 2002; 277:26103-12. [PMID: 12000758 DOI: 10.1074/jbc.m202921200] [Citation(s) in RCA: 111] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
Knowledge about the O-linked glycan chains of tumor-associated MUC1 is primarily based on enzymatic and immunochemical evidence. To obtain structural information and to overcome limitations by the scarcity of endogenous mucin, we expressed a recombinant glycosylation probe corresponding to six MUC1 tandem repeats in four breast cancer cell lines. Comparative analyses of the O-glycan profiles were performed after hydrazinolysis and normal phase chromatography of 2-aminobenzamide-labeled glycans. Except for a general reduction in the O-glycan chain lengths and a high density glycosylation, no common structural pattern was revealed. T47D fusion protein exhibits an almost complete shift from core 2 to core 1 expression with a preponderance of sialylated glycans. By contrast, MCF-7, MDA-MB231, and ZR75-1 cells glycosylate the MUC1 repeat peptide preferentially with core 2-based glycans terminating mostly with alpha 3-linked sialic acid (MDA-MB231, ZR75-1) or alpha 2/3-linked fucose (MCF-7). Endogenous MUC1 from T47D and MCF-7 cell supernatants revealed almost identical O-glycosylation profiles compared with the respective recombinant probes, indicating that the fusion proteins reflected the authentic O-glycan profiles of the cells. The structural patterns in the majority of cells under study are in conflict with biosynthetic models of MUC1 O-glycosylation in breast cancer, which claim that the truncation of normal core 2-based polylactosamine structures to short sialylated core 1-based glycans is due to the reduced activity of core 2-forming beta 6-N-acetylglucosaminyltransferases and/or to overexpression of competitive alpha 3- sialyltransferase.
Collapse
Affiliation(s)
- Stefan Müller
- Institute of Biochemistry II, Medical Faculty of the University, Joseph-Stelzmann-Str. 52, Köln D-50931, Germany.
| | | |
Collapse
|
106
|
Evaluation of a New Serum Testing Method for Detection of Prostate Cancer. J Urol 2002. [DOI: 10.1097/00005392-200207000-00022] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
107
|
Seabury CA, Calenoff E, Ditlow C, Bux S, Clarke H, Issa M, Marshall F, Petros J. Evaluation of a New Serum Testing Method for Detection of Prostate Cancer. J Urol 2002. [DOI: 10.1016/s0022-5347(05)64838-0] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022]
Affiliation(s)
- Charles A. Seabury
- From Emory University, Atlanta, Georgia, Northwestern University, Chicago, Illinois, and West Virginia University, Morgantown, West Virginia
| | - Emanuel Calenoff
- From Emory University, Atlanta, Georgia, Northwestern University, Chicago, Illinois, and West Virginia University, Morgantown, West Virginia
| | - Charles Ditlow
- From Emory University, Atlanta, Georgia, Northwestern University, Chicago, Illinois, and West Virginia University, Morgantown, West Virginia
| | - Sajit Bux
- From Emory University, Atlanta, Georgia, Northwestern University, Chicago, Illinois, and West Virginia University, Morgantown, West Virginia
| | - Harry Clarke
- From Emory University, Atlanta, Georgia, Northwestern University, Chicago, Illinois, and West Virginia University, Morgantown, West Virginia
| | - Muta Issa
- From Emory University, Atlanta, Georgia, Northwestern University, Chicago, Illinois, and West Virginia University, Morgantown, West Virginia
| | - Fray Marshall
- From Emory University, Atlanta, Georgia, Northwestern University, Chicago, Illinois, and West Virginia University, Morgantown, West Virginia
| | - John Petros
- From Emory University, Atlanta, Georgia, Northwestern University, Chicago, Illinois, and West Virginia University, Morgantown, West Virginia
| |
Collapse
|
108
|
Abstract
Over the last decade, there has been a rapid expansion in the field of tumour immunology. There is now convincing evidence that both the cellular and humoral arms of the immune system are capable of interacting with tumour cells. The most significant advances have been in our understanding of cellular responses and the complex events that lead to T-lymphocyte activation, as well as in the identification of tumour antigens recognised by T-lymphocytes. This knowledge has led to the development of anticancer immunotherapies designed to produce tumour antigen-specific T-cell responses, adding to the earlier antibody or whole-cell vaccine approaches. In addition, new methods have been developed to quantify antigen-specific T-cell responses, and the emergent field of recombinant gene technology has led to an increasing number of novel methods for vaccine delivery. This review will explore these advances, as well as possible future directions, with an emphasis on colorectal cancer.
Collapse
Affiliation(s)
- Caroline L Smith
- Cancer Research UK Tumour Immunology Group, The Weatherall Institute of Molecular Medicine, University of Oxford, John Radcliffe Hospital, Headington, Oxford OX3 9DS, UK
| | | | | | | |
Collapse
|
109
|
Heukamp LC, van Hall T, Ossendorp F, Burchell JM, Melief CJM, Taylor-Papadimitriou J, Offringa R. Effective immunotherapy of cancer in MUC1-transgenic mice using clonal cytotoxic T lymphocytes directed against an immunodominant MUC1 epitope. J Immunother 2002; 25:46-56. [PMID: 11926165 DOI: 10.1097/00002371-200201000-00005] [Citation(s) in RCA: 12] [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
The tumor-associated autoantigen MUCI is intensively studied as a potential target for antigen-specific immunotherapy of cancer. Previous reports concerning experiments in preclinical murine tumor models have provided evidence supporting the feasibility of this approach. However, such studies have not been performed with clonal cytotoxic T lymphocyte populations displaying a highly defined MUC1 specificity. The authors demonstrate that the immunodominant MUC1-specific cytotoxic T lymphocyte response in C57BL/6 mice is directed against an H-2Kb-restricted epitope, MUC1(19-27), which is derived from the N-terminal signal sequence of the MUC1 protein. Processing of this epitope was independent of transporter of antigen presentation and proteasome function. Importantly, successful immunotherapy of MUC1-overexpressing tumors in MUC1-transgenic mice was not accompanied by damage to normal somatic MUC1-positive tissues, even when this involved the infusion of large numbers of clonal cytotoxic T lymphocyte that recognized the immunodominant MUC1 epitope. Although the risk for autoimmune pathology is limited, data indicate that immune tolerance in MUC1-positive subjects restricts the breadth of the MUC1-specific cytotoxic T lymphocyte repertoire that is available for recruitment to immunotherapeutic antitumor responses.
Collapse
MESH Headings
- ATP Binding Cassette Transporter, Subfamily B, Member 2
- ATP-Binding Cassette Transporters/physiology
- Amino Acid Sequence
- Animals
- Autoimmunity
- COS Cells
- Cysteine Endopeptidases/physiology
- Epitopes, T-Lymphocyte
- Histocompatibility Antigens Class I/immunology
- Immune Tolerance
- Immunodominant Epitopes
- Immunotherapy, Adoptive
- Mice
- Mice, Inbred C57BL
- Mice, Inbred CBA
- Mice, Transgenic
- Molecular Sequence Data
- Mucin-1/chemistry
- Mucin-1/immunology
- Multienzyme Complexes/physiology
- Neoplasms, Experimental/immunology
- Neoplasms, Experimental/therapy
- Proteasome Endopeptidase Complex
- T-Lymphocytes, Cytotoxic/immunology
Collapse
Affiliation(s)
- Lukas C Heukamp
- Imperial Cancer Research Fund, Breast Cancer Biology Group, Guy's Hospital, London, United Kingdom
| | | | | | | | | | | | | |
Collapse
|
110
|
Kao H, Marto JA, Hoffmann TK, Shabanowitz J, Finkelstein SD, Whiteside TL, Hunt DF, Finn OJ. Identification of cyclin B1 as a shared human epithelial tumor-associated antigen recognized by T cells. J Exp Med 2001; 194:1313-23. [PMID: 11696596 PMCID: PMC2195974 DOI: 10.1084/jem.194.9.1313] [Citation(s) in RCA: 98] [Impact Index Per Article: 4.1] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/09/2001] [Revised: 09/10/2001] [Accepted: 09/25/2001] [Indexed: 11/07/2022] Open
Abstract
We eluted peptides from class I molecules of HLA-A2.1(+) breast adenocarcinoma and loaded reverse phase high-performance liquid chromatography (HPLC) fractions onto dendritic cells to prime naive CD8(+) T cells. Fractions that supported growth of tumor-specific cytotoxic T lymphocytes were analyzed by nano-HPLC micro-ESI tandem mass spectrometry. Six HLA-A2.1-binding peptides, four 9-mers (P1-P4) differing in the COOH-terminal residue, and two 10-mers (P5 and P6) with an additional COOH-terminal alanine, were identified in one fraction. Peptide sequences were homologous to cyclin B1. We primed CD8(+) T cells from another HLA-A2.1(+) healthy donor with synthetic peptides and generated P4-specific responses. We also detected memory T cells specific for one or more of these peptides in patients with breast cancer and squamous cell carcinomas of the head and neck (SCCHN). T cells from one patient, restimulated once in vitro, could kill the tumor cell line from which the peptides were derived. Immunohistochemical analysis of tumor lines and tissue sections showed cyclin B1 overexpression and aberrant localization in the cytoplasm instead of the nucleus. Sequencing genomic DNA and cDNA corresponding to P1-P6 region showed that differences in COOH-terminal residues were not due to either DNA mutations or errors in transcription, suggesting a high error rate in translation of cyclin B1 protein in tumors.
Collapse
Affiliation(s)
- H Kao
- Department of Molecular Genetics and Biochemistry, University of Pittsburgh, Pittsburgh, PA 15261, USA
| | | | | | | | | | | | | | | |
Collapse
|
111
|
Croce MV, Isla-Larrain MT, Capafons A, Price MR, Segal-Eiras A. Humoral immune response induced by the protein core of MUC1 mucin in pregnant and healthy women. Breast Cancer Res Treat 2001; 69:1-11. [PMID: 11759823 DOI: 10.1023/a:1012220902991] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022]
Abstract
UNLABELLED Serum levels of MUC1 and antibodies (Abs) against MUC1 (IgG and IgM-MUC1) were evaluated in healthy women related to pregnancy and lactation status. A total of 149 serum samples were obtained from: nulliparous, primiparous pregnant, multiparous pregnant that have lactated, multiparous pregnant without lactation, multiparous non-pregnant actual lactating, multiparous non-pregnant that have lactated and finally, multiparous non-pregnant women without lactation. In all assays, we included pre- and post-serum samples belonging to a breast cancer patient vaccinated with a MUC1 derived peptide. CASA test was employed to measure MUC1 while IgG- and IgM-MUC1 serum Abs were evaluated with an ELISA using a 100 mer peptide as catcher. In all groups, mean IgM levels were higher than IgG mean values; when samples were grouped in pregnants versus non-pregnants, a significant difference was detected with both Abs, being raised in non-pregnants. When samples were grouped in lactating versus non-lactating a significant difference was detected with IgG-MUC1, being raised in lactating women while no significant difference was found with IgM-MUC1. The evaluation of serum MUC1 levels confirmed previous results since a significant difference between pregnant versus non-pregnant groups was found while lactating versus non-lactating samples did not. CONCLUSIONS (i) Increased MUC1 serum levels are apparently associated with pregnancy but not with lactation; (ii) MUC1 Abs are mainly associated with lactation and with non-pregnant status. These results may be considered a contribution on studies about protection against breast cancer induced by pregnancy and lactation.
Collapse
Affiliation(s)
- M V Croce
- Comisión de Investigaciones Científicas de la Provincia de Buenos Aires, Argentina
| | | | | | | | | |
Collapse
|
112
|
Snijdewint FG, von Mensdorff-Pouilly S, Karuntu-Wanamarta AH, Verstraeten AA, Livingston PO, Hilgers J, Kenemans P. Antibody-dependent cell-mediated cytotoxicity can be induced by MUC1 peptide vaccination of breast cancer patients. Int J Cancer 2001; 93:97-106. [PMID: 11391628 DOI: 10.1002/ijc.1286] [Citation(s) in RCA: 59] [Impact Index Per Article: 2.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/11/2022]
Abstract
Human polymorphic epithelial mucin (PEM, MUC1) is a high molecular weight transmembrane glycoprotein expressed on the apical cell surface of glandular epithelium and is over-expressed and hypo-glycosylated in adenocarcinomas. The extracellular part of the molecule consists mainly of a variable number of 20 amino acid repeats that contain cryptic epitopes exposed in malignancy. The objective of our study was to determine whether humanized MUC1 MAbs and Abs induced by vaccination of breast cancer patients with MUC1 peptides can effect an antibody-dependent cell-mediated cytotoxicity (ADCC). An in vitro assay has been set up in which the breast tumor cell line ZR-75-1 is used as target and PBMC of healthy donors as effector cells. Different target and effector cells, as well as various MUC1 MAbs were tested to optimize the efficacy of the in vitro assay. The humanized MAb HuHMFG-1, which recognizes the PDTR sequence in the MUC1 tandem repeat, induced a strong cell-mediated cytotoxicity. Nine MUC1-expressing tumor cell lines, including 3 bone marrow-derived cell lines, as well as 2 MUC1-transfected cell lines were susceptible to different extent to MUC1 Ab-dependent killing. Large variations in the killing capacity of PBMC from healthy donors were found. The NK cells were the essential effector cells for the MUC1 Ab-dependent killing. Plasma samples with induced high levels of MUC1 Ab were obtained from breast cancer patients repeatedly immunized with a KLH-conjugated 33-mer or 106-mer MUC1 tandem repeat. Pre- and post-vaccinated plasma samples of these patients were compared in the ADCC assay and it could be clearly demonstrated that the induced MUC1 Abs can effect tumor cell killing. MUC1 Ab-dependent cell-mediated tumor cell killing may occur in vivo and the ADCC assay can be applied to monitor MUC1 vaccination trials.
Collapse
Affiliation(s)
- F G Snijdewint
- Department of Obstetrics and Gynaecology, Academic Hospital Vrije Universiteit, Amsterdam, The Netherlands.
| | | | | | | | | | | | | |
Collapse
|
113
|
Meijer SL, Dols A, Hu H, Jensen S, Poehlein CH, Chu Y, Winter H, Yamada J, Moudgil T, Wood WJ, Doran T, Justice L, Fisher B, Wisner P, Wood J, Vetto JT, Mehrotra R, Rosenheim S, Weinberg AD, Bright R, Walker E, Puri R, Smith JW, Urba WJ, Fox BA. Immunological and Molecular Analysis of the Sentinel Lymph Node: A Potential Approach to Predict Outcome, Tailor Therapy, and Optimize Parameters for Tumor Vaccine Development. J Clin Pharmacol 2001. [DOI: 10.1177/0091270001417012] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022]
Affiliation(s)
- S. L. Meijer
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute
- Departments of Surgery and Pathology, Providence Portland Medical Center
- Oregon Cancer Center and Department of Molecular Microbiology and Immunology, Oregon Health Sciences University
- Department of Biochemistry and Molecular Biology, Oregon Graduate Institute
| | - A. Dols
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute
- Departments of Surgery and Pathology, Providence Portland Medical Center
- Oregon Cancer Center and Department of Molecular Microbiology and Immunology, Oregon Health Sciences University
- Department of Biochemistry and Molecular Biology, Oregon Graduate Institute
| | - H‐M. Hu
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute
- Departments of Surgery and Pathology, Providence Portland Medical Center
- Oregon Cancer Center and Department of Molecular Microbiology and Immunology, Oregon Health Sciences University
- Department of Biochemistry and Molecular Biology, Oregon Graduate Institute
| | - S. Jensen
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute
- Departments of Surgery and Pathology, Providence Portland Medical Center
- Oregon Cancer Center and Department of Molecular Microbiology and Immunology, Oregon Health Sciences University
- Department of Biochemistry and Molecular Biology, Oregon Graduate Institute
| | - C. H. Poehlein
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute
- Departments of Surgery and Pathology, Providence Portland Medical Center
- Oregon Cancer Center and Department of Molecular Microbiology and Immunology, Oregon Health Sciences University
- Department of Biochemistry and Molecular Biology, Oregon Graduate Institute
| | - Y. Chu
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute
- Departments of Surgery and Pathology, Providence Portland Medical Center
- Oregon Cancer Center and Department of Molecular Microbiology and Immunology, Oregon Health Sciences University
- Department of Biochemistry and Molecular Biology, Oregon Graduate Institute
| | - H. Winter
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute
- Departments of Surgery and Pathology, Providence Portland Medical Center
- Oregon Cancer Center and Department of Molecular Microbiology and Immunology, Oregon Health Sciences University
- Department of Biochemistry and Molecular Biology, Oregon Graduate Institute
| | - J. Yamada
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute
- Departments of Surgery and Pathology, Providence Portland Medical Center
- Oregon Cancer Center and Department of Molecular Microbiology and Immunology, Oregon Health Sciences University
- Department of Biochemistry and Molecular Biology, Oregon Graduate Institute
| | - T Moudgil
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute
- Departments of Surgery and Pathology, Providence Portland Medical Center
- Oregon Cancer Center and Department of Molecular Microbiology and Immunology, Oregon Health Sciences University
- Department of Biochemistry and Molecular Biology, Oregon Graduate Institute
| | - W. J. Wood
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute
- Departments of Surgery and Pathology, Providence Portland Medical Center
- Oregon Cancer Center and Department of Molecular Microbiology and Immunology, Oregon Health Sciences University
- Department of Biochemistry and Molecular Biology, Oregon Graduate Institute
| | - T Doran
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute
- Departments of Surgery and Pathology, Providence Portland Medical Center
- Oregon Cancer Center and Department of Molecular Microbiology and Immunology, Oregon Health Sciences University
- Department of Biochemistry and Molecular Biology, Oregon Graduate Institute
| | - L. Justice
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute
- Departments of Surgery and Pathology, Providence Portland Medical Center
- Oregon Cancer Center and Department of Molecular Microbiology and Immunology, Oregon Health Sciences University
- Department of Biochemistry and Molecular Biology, Oregon Graduate Institute
| | - B. Fisher
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute
- Departments of Surgery and Pathology, Providence Portland Medical Center
- Oregon Cancer Center and Department of Molecular Microbiology and Immunology, Oregon Health Sciences University
- Department of Biochemistry and Molecular Biology, Oregon Graduate Institute
| | - P. Wisner
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute
- Departments of Surgery and Pathology, Providence Portland Medical Center
- Oregon Cancer Center and Department of Molecular Microbiology and Immunology, Oregon Health Sciences University
- Department of Biochemistry and Molecular Biology, Oregon Graduate Institute
| | - J. Wood
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute
- Departments of Surgery and Pathology, Providence Portland Medical Center
- Oregon Cancer Center and Department of Molecular Microbiology and Immunology, Oregon Health Sciences University
- Department of Biochemistry and Molecular Biology, Oregon Graduate Institute
| | - J. T. Vetto
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute
- Departments of Surgery and Pathology, Providence Portland Medical Center
- Oregon Cancer Center and Department of Molecular Microbiology and Immunology, Oregon Health Sciences University
- Department of Biochemistry and Molecular Biology, Oregon Graduate Institute
| | - R. Mehrotra
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute
- Departments of Surgery and Pathology, Providence Portland Medical Center
- Oregon Cancer Center and Department of Molecular Microbiology and Immunology, Oregon Health Sciences University
- Department of Biochemistry and Molecular Biology, Oregon Graduate Institute
| | - S. Rosenheim
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute
- Departments of Surgery and Pathology, Providence Portland Medical Center
- Oregon Cancer Center and Department of Molecular Microbiology and Immunology, Oregon Health Sciences University
- Department of Biochemistry and Molecular Biology, Oregon Graduate Institute
| | - A. D. Weinberg
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute
- Departments of Surgery and Pathology, Providence Portland Medical Center
- Oregon Cancer Center and Department of Molecular Microbiology and Immunology, Oregon Health Sciences University
- Department of Biochemistry and Molecular Biology, Oregon Graduate Institute
| | - R. Bright
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute
- Departments of Surgery and Pathology, Providence Portland Medical Center
- Oregon Cancer Center and Department of Molecular Microbiology and Immunology, Oregon Health Sciences University
- Department of Biochemistry and Molecular Biology, Oregon Graduate Institute
| | - E. Walker
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute
- Departments of Surgery and Pathology, Providence Portland Medical Center
- Oregon Cancer Center and Department of Molecular Microbiology and Immunology, Oregon Health Sciences University
- Department of Biochemistry and Molecular Biology, Oregon Graduate Institute
| | - R. Puri
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute
- Departments of Surgery and Pathology, Providence Portland Medical Center
- Oregon Cancer Center and Department of Molecular Microbiology and Immunology, Oregon Health Sciences University
- Department of Biochemistry and Molecular Biology, Oregon Graduate Institute
| | - J. W. Smith
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute
- Departments of Surgery and Pathology, Providence Portland Medical Center
- Oregon Cancer Center and Department of Molecular Microbiology and Immunology, Oregon Health Sciences University
- Department of Biochemistry and Molecular Biology, Oregon Graduate Institute
| | - W. J. Urba
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute
- Departments of Surgery and Pathology, Providence Portland Medical Center
- Oregon Cancer Center and Department of Molecular Microbiology and Immunology, Oregon Health Sciences University
- Department of Biochemistry and Molecular Biology, Oregon Graduate Institute
| | - B. A. Fox
- Robert W. Franz Cancer Research Center, Earle A. Chiles Research Institute
- Departments of Surgery and Pathology, Providence Portland Medical Center
- Oregon Cancer Center and Department of Molecular Microbiology and Immunology, Oregon Health Sciences University
- Department of Biochemistry and Molecular Biology, Oregon Graduate Institute
| |
Collapse
|
114
|
Noujaim AA, Schultes BC, Baum RP, Madiyalakan R. Induction of CA125-specific B and T cell responses in patients injected with MAb-B43.13--evidence for antibody-mediated antigen-processing and presentation of CA125 in vivo. Cancer Biother Radiopharm 2001; 16:187-203. [PMID: 11471484 DOI: 10.1089/10849780152389384] [Citation(s) in RCA: 85] [Impact Index Per Article: 3.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The murine monoclonal anti-CA125 antibody MAb-B43.13 has previously been administered as an immunoscintigraphic agent in order to monitor recurrence of ovarian cancer in patients, and a long-term follow-up demonstrated a survival benefit for these patients. The clinical benefit was initially attributed to the activation of the idiotypic network. The objective of this study was to investigate the role of CA125-MAb-B43.13 immune complex formation on the induction of CA125-specific immune responses. Analysis of patient serum samples from pharmacokinetic studies demonstrated that the antibody forms immune complexes with CA125 in circulation within 30 minutes of injection. Induction of humoral and cellular anti-CA125 responses correlated with the amount of circulating CA125 antigen present at time of antibody injection. Subsequent to the injection of MAb-B43.13, the patients generated anti-CA125 antibodies that were directed against various epitopes on the antigen and were not restricted to the specific epitope recognized by MAb-B43.13. The generation of CA125-specific B and T cell responses after MAb-B43.13 injection correlated with improved survival. The influence of circulating CA125 for the induction of CA125-specific immune responses and the multi-epitopic nature of the human anti-CA125 antibodies suggest that the majority of these antibodies were not induced via the idiotypic network but by the autologous antigen itself. Since antibody and T cell responses to CA125 were not present before injection of MAb-B43.13, it is hypothesized that complex formation of MAb-B43.13 with circulating antigen triggers the induction of CA125-specific immune responses.
Collapse
Affiliation(s)
- A A Noujaim
- AltaRex Corp., 1123 Dentistry-Pharmacy, University of Alberta, Edmonton, AB, T6G 2N8, Canada.
| | | | | | | |
Collapse
|
115
|
Pecher G, Harnack U, Günther M, Hummel M, Fichtner I, Schenk JA. Generation of an Immortalized Human CD4+ T Cell Clone Inhibiting Tumor Growth in Mice. Biochem Biophys Res Commun 2001; 283:738-42. [PMID: 11350045 DOI: 10.1006/bbrc.2001.4846] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
Tumor antigen-specific T cell clones represent a useful tool in tumor immunology; however, their long-term culture is limited. To generate an immortalized cytotoxic T cell clone against the human tumor antigen mucin, we exposed a previously generated T cell culture to Herpesvirus saimiri. We obtained an immortalized human CD4+ T cell clone, termed SITAM. Clonality of these cells was shown by analysis of the alpha/beta-T cell receptor (TCR) repertoire. Cytolytic activity was demonstrated against several mucin-expressing tumor cell lines and could not be detected against non-mucin-expressing cells. SITAM cells maintained their features stably for 2 years. Furthermore, growth of the tumor cell line Capan-2 in NOD/SCID mice was inhibited when SITAM cells were coinjected subcutaneously with tumor cells. SITAM cells provide an unlimited source of clonal T cells for analysis of tumor recognition and may be of help in TCR-targeted immunotherapy.
Collapse
Affiliation(s)
- G Pecher
- AG Molecular Gene- and Immunotherapy, Humboldt-University Berlin, Charité Campus Mitte, Hessische Strasse 3-4, Berlin, 10115, Germany.
| | | | | | | | | | | |
Collapse
|
116
|
Feuerer M, Beckhove P, Bai L, Solomayer EF, Bastert G, Diel IJ, Pedain C, Oberniedermayr M, Schirrmacher V, Umansky V. Therapy of human tumors in NOD/SCID mice with patient-derived reactivated memory T cells from bone marrow. Nat Med 2001; 7:452-8. [PMID: 11283672 DOI: 10.1038/86523] [Citation(s) in RCA: 211] [Impact Index Per Article: 8.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
In an analysis of 84 primary-operated breast cancer patients and 11 healthy donors, we found that the bone marrow of most patients contained memory T cells with specificity for tumor-associated antigens. Patients' bone marrow and peripheral blood contained CD8+ T cells that specifically bound HLA/peptide tetramers. In short-term culture with autologous dendritic cells pre-pulsed with tumor lysates, patients' memory T cells from bone marrow (but not peripheral blood) could be specifically reactivated to interferon-gamma-producing and cytotoxic effector cells. A single transfer of restimulated bone-marrow T cells into NOD/SCID mice caused regression of autologous tumor xenotransplants associated with infiltration by human T cells and tumor-cell apoptosis and necrosis. T cells from peripheral blood showed much lower anti-tumor reactivity. Our findings reveal an innate, specific recognition of breast cancer antigens and point to a possible novel cancer therapy using patients' bone-marrow-derived memory T cells.
Collapse
MESH Headings
- Amino Acid Sequence
- Animals
- Antigens, Neoplasm/chemistry
- Apoptosis
- Bone Marrow Transplantation
- Breast Neoplasms/immunology
- Breast Neoplasms/pathology
- Breast Neoplasms/therapy
- Female
- HLA-A2 Antigen/metabolism
- Humans
- Immunologic Memory
- In Vitro Techniques
- Interferon-gamma/biosynthesis
- Lymphocyte Activation
- Lymphocytes, Tumor-Infiltrating/immunology
- Lymphocytes, Tumor-Infiltrating/pathology
- Mice
- Mice, Inbred NOD
- Mice, SCID
- Mucin-1/chemistry
- Mucin-1/immunology
- Necrosis
- Peptide Fragments/chemistry
- Peptide Fragments/immunology
- Receptor, ErbB-2/chemistry
- Receptor, ErbB-2/immunology
- T-Lymphocytes/immunology
- T-Lymphocytes/transplantation
- T-Lymphocytes, Cytotoxic/immunology
- Transplantation, Autologous
- Transplantation, Heterologous
Collapse
Affiliation(s)
- M Feuerer
- Division of Cellular Immunology, Tumor Immunology Program, German Cancer Research Center, Heidelberg, Germany
| | | | | | | | | | | | | | | | | | | |
Collapse
|
117
|
Sosman JA, Stiff P, Moss SM, Sorokin P, Martone B, Bayer R, van Besien K, Devine S, Stock W, Peace D, Chen Y, Long C, Gustin D, Viana M, Hoffman R. Pilot trial of interleukin-2 with granulocyte colony-stimulating factor for the mobilization of progenitor cells in advanced breast cancer patients undergoing high-dose chemotherapy: expansion of immune effectors within the stem-cell graft and post-stem-cell infusion. J Clin Oncol 2001; 19:634-44. [PMID: 11157013 DOI: 10.1200/jco.2001.19.3.634] [Citation(s) in RCA: 26] [Impact Index Per Article: 1.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE To evaluate whether administration of interleukin-2 (IL-2) with granulocyte colony-stimulating factor (G-CSF) improves mobilization of immune effector cells into the stem-cell graft of patients undergoing high-dose chemotherapy and autografting. PATIENTS AND METHODS We performed a trial of stem-cell mobilization with IL-2 and G-CSF in advanced breast cancer patients receiving high-dose chemotherapy with cyclophosphamide, thiotepa, and carboplatin and stem cells followed by IL-2. The trial defined immune, hematologic, and clinical effects of IL-2 in this setting. RESULTS Of 32 patients enrolled, nine received G-CSF alone for mobilization. Twenty-one of 23 patients mobilized with IL-2 plus G-CSF had stem cells collected with more mononuclear cells than those receiving G-CSF (19.3 v 10.4 x 10(8)/kg; P =.006), but fewer CD34(+) progenitor cells (6.9 v 22.0 x 10(6)/kg; P =.049). The IL-2 plus G-CSF-mobilized patients had greater numbers of activated T (CD3(+)/CD25(+)) cells (P =.009), natural killer (NK; CD56(+)) cells (P =.007), and activated NK (CD56 bright(+)) cells (P: =.039) than those patients mobilized with G-CSF. NK (P =.042) and lymphokine-activated killer (LAK) (P =.016) activity was increased in those mobilized with IL-2 + G-CSF, whereas G-CSF-mobilized patients had a decline in cytolytic activity. In the third week posttransplantation, immune reconstitution was superior in those mobilized with IL-2 plus G-CSF based on greater numbers of activated T cells (P =.003), activated NK cells (P =.04), and greater LAK activity (P =.003). The 16 of 21 IL-2 + G-CSF-mobilized patients with adequate numbers of stem cells (> 1.5 x 10(6) CD34(+) cells/kg) collected engrafted rapidly posttransplantation. CONCLUSION The results demonstrate that G-CSF + IL-2 can enhance the number and function of antitumor effector cells in a mobilized autograft without impairing the hematologic engraftment, provided that CD34 cell counts are more than 1.5 x 10(6) cells/kg. Mobilization of CD34(+) stem cells does seem to be adversely affected. In those mobilized with IL-2 and G-CSF, post-stem-cell immune reconstitution of antitumor immune effector cells was enhanced.
Collapse
Affiliation(s)
- J A Sosman
- Section of Hematology/Oncology, University of Illinois at Chicago College of Medicine, Chicago 60612, USA.
| | | | | | | | | | | | | | | | | | | | | | | | | | | | | |
Collapse
|
118
|
Morikane K, Tempero R, Sivinski CL, Kitajima S, Gendler SJ, Hollingsworth MA. Influence of organ site and tumor cell type on MUC1-specific tumor immunity. Int Immunol 2001; 13:233-40. [PMID: 11157856 DOI: 10.1093/intimm/13.2.233] [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: 11/13/2022] Open
Abstract
We investigated the influence of organ-specific parameters on tolerance and immunity to human MUC1. C57Bl/6 mice (wild-type) and C57Bl/6 transgenic for MUC1 (MUC1.Tg) were challenged in the pancreas with Panc02-MUC1, a C57Bl/6-syngeneic pancreatic cancer cell line expressing human MUC1. Wild-type mice produced immune responses to MUC1 when presented on tumor cells growing in the pancreas; however, the responses to tumors in the pancreas were less effective than responses produced by tumor challenge at the s.c. site. Tumor immunity specific for MUC1 was produced in wild-type mice by two different procedures: (i) s.c. immunization of wild-type mice with a low dose of Panc02-MUC1 or (ii) adoptive transfer of spleen and lymph node cells harvested from wild-type mice previously immunized s.c. with Panc02-MUC1. This demonstrates that immune responses to MUC1 presented at the s.c. site can be detected and adoptively transferred. MUC1.Tg mice were immunologically tolerant to MUC1; however, some immunological protection against orthotopic challenge with Panc02-MUC1 was conferred by adoptive transfer of CD4+ and CD8+ T cells from wild-type mice. These results show that it is more difficult to produce immune responses to tumors growing at the pancreatic site than the s.c. site. Panc02-MUC1 cells growing in the pancreas were accessible to the immune system, and immune responses evoked by s.c. presentation of this molecule in wild-type mice were effective in rejecting tumor cells in the pancreas of both wild-type and MUC1.Tg mice. No effective anti-tumor immune responses against MUC1 were produced in MUC1.Tg mice.
Collapse
Affiliation(s)
- K Morikane
- Eppley Institute for Research in Cancer and Allied Diseases, University of Nebraska Medical Center, 600 South 42nd Street, Omaha, NE 68198-6805, USA
| | | | | | | | | | | |
Collapse
|
119
|
Segura JA, Ruiz-Bellido MA, Arenas M, Lobo C, Márquez J, Alonso FJ. Ehrlich ascites tumor cells expressing anti-sense glutaminase mRNA lose their capacity to evade the mouse immune system. Int J Cancer 2000. [DOI: 10.1002/1097-0215(200002)9999:9999<::aid-ijc1046>3.0.co;2-l] [Citation(s) in RCA: 4] [Impact Index Per Article: 0.2] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
|
120
|
Gerloni M, Xiong S, Mukerjee S, Schoenberger SP, Croft M, Zanetti M. Functional cooperation between T helper cell determinants. Proc Natl Acad Sci U S A 2000; 97:13269-74. [PMID: 11069291 PMCID: PMC27214 DOI: 10.1073/pnas.230429197] [Citation(s) in RCA: 72] [Impact Index Per Article: 2.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/26/2000] [Accepted: 09/07/2000] [Indexed: 11/18/2022] Open
Abstract
The immune response to T helper (Th) cell determinants of a variety of antigens is often poor and limits severely the potential efficacy of current therapeutic measures through vaccination. Here, we report that an immunologically silent tumor determinant can be rendered immunogenic if linked with a dominant determinant of a parasite antigen, suggesting the existence of functional Th-Th cooperation in vivo. This phenomenon could be mimicked in part by signaling either through CD40 to the antigen-presenting cells or through OX40 to the tumor-determinant reactive T cells, with maximal effects obtained by combined anti-CD40 and anti-OX40 treatment in vivo. The data suggest that CD4 T cells reactive with a dominant determinant provide help to other CD4 T cells through up-regulating the costimulatory ability of antigen-presenting cells, in much the same way as help for CD8 cells. CD4 help for CD4 T cells represents a new immunological principle and offers new practical solutions for vaccine therapy against cancer and other diseases in which antigenic help is limiting.
Collapse
Affiliation(s)
- M Gerloni
- The Department of Medicine and Cancer Center, University of California at San Diego, La Jolla CA 92093-0368, USA
| | | | | | | | | | | |
Collapse
|
121
|
Koido S, Kashiwaba M, Chen D, Gendler S, Kufe D, Gong J. Induction of antitumor immunity by vaccination of dendritic cells transfected with MUC1 RNA. JOURNAL OF IMMUNOLOGY (BALTIMORE, MD. : 1950) 2000; 165:5713-9. [PMID: 11067929 DOI: 10.4049/jimmunol.165.10.5713] [Citation(s) in RCA: 121] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 12/25/2022]
Abstract
Dendritic cells (DC) are potent APCs. In this study, murine bone marrow-derived DC were transfected with RNA encoding the MUC1 Ag that is aberrantly overexpressed in human breast and other carcinomas. The MUC1 RNA-transfected DC exhibited cell surface expression of MUC1 and costimulatory molecules. After injection at the base of the tail, the transfected DC were detectable in inguinal lymph nodes by dual immunochemical staining. Vaccination of wild-type mice with MUC1 RNA-transfected DC induced anti-MUC1 immune responses against MUC1-positive MC38/MUC1, but not MUC1-negative, tumor cells. Mice immunized with the transfected DC were protected against challenge with MC38/MUC1 tumor cells. Furthermore, mice with established MC38/MUC1 tumors were eliminated after receiving the vaccination. CTLs isolated from mice immunized with the transfected DC exhibited specific cytolytic activity against MC38/MUC1 tumor cells. In contrast to these findings, there was little if any anti-MUC1 immunity induced with the transfected DC in MUC1 transgenic (MUC1.Tg) mice. However, coadministration of the transfected DC and IL-12 reversed the unresponsiveness to MUC1 Ag in MUC1.Tg mice and induced MUC1-specific immune responses. These findings demonstrate that vaccination of DC transfected with MUC1 RNA and IL-12 reverses tolerance to MUC1 and induces immunity against MUC1-positive tumors.
Collapse
Affiliation(s)
- S Koido
- Dana-Farber Cancer Institute, Harvard Medical School, Boston, MA 02115, USA
| | | | | | | | | | | |
Collapse
|
122
|
Induction of cytotoxic T-lymphocyte responses in vivo after vaccinations with peptide-pulsed dendritic cells. Blood 2000. [DOI: 10.1182/blood.v96.9.3102] [Citation(s) in RCA: 375] [Impact Index Per Article: 15.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractVaccination of patients with cancer using dendritic cells (DCs) was shown to be effective for B-cell lymphoma and malignant melanoma. Here we provide evidence that patients with advanced breast and ovarian cancer can be efficiently vaccinated with autologous DCs pulsed with HER-2/neu– or MUC1-derived peptides. Ten patients were included in this pilot study. The DC vaccinations were well tolerated with no side effects. In 5 of 10 patients, peptide-specific cytotoxic T lymphocytes (CTLs) could be detected in the peripheral blood using both intracellular IFN-γ staining and 51Cr-release assays. The major CTL response in vivo was induced with the HER-2/neu–derived E75 and the MUC1-derived M1.2 peptide, which lasted for more than 6 months, suggesting that these peptides might be immunodominant. In addition, in one patient vaccinated with the MUC1-derived peptides, CEA- and MAGE-3 peptide-specific T-cell responses were detected after several vaccinations. In a second patient immunized with the HER-2/neu peptides, MUC1-specific T lymphocytes were induced after 7 immunizations, suggesting that antigen spreading in vivo might occur after successful immunization with a single tumor antigen. Our results show that vaccination of DCs pulsed with a single tumor antigen may induce immunologic responses in patients with breast and ovarian cancer. This study may be relevant to the design of future clinical trials of other peptide-based vaccines.
Collapse
|
123
|
Abstract
Vaccination of patients with cancer using dendritic cells (DCs) was shown to be effective for B-cell lymphoma and malignant melanoma. Here we provide evidence that patients with advanced breast and ovarian cancer can be efficiently vaccinated with autologous DCs pulsed with HER-2/neu– or MUC1-derived peptides. Ten patients were included in this pilot study. The DC vaccinations were well tolerated with no side effects. In 5 of 10 patients, peptide-specific cytotoxic T lymphocytes (CTLs) could be detected in the peripheral blood using both intracellular IFN-γ staining and 51Cr-release assays. The major CTL response in vivo was induced with the HER-2/neu–derived E75 and the MUC1-derived M1.2 peptide, which lasted for more than 6 months, suggesting that these peptides might be immunodominant. In addition, in one patient vaccinated with the MUC1-derived peptides, CEA- and MAGE-3 peptide-specific T-cell responses were detected after several vaccinations. In a second patient immunized with the HER-2/neu peptides, MUC1-specific T lymphocytes were induced after 7 immunizations, suggesting that antigen spreading in vivo might occur after successful immunization with a single tumor antigen. Our results show that vaccination of DCs pulsed with a single tumor antigen may induce immunologic responses in patients with breast and ovarian cancer. This study may be relevant to the design of future clinical trials of other peptide-based vaccines.
Collapse
|
124
|
Segura JA, Barbero LG, Márquez J. Ehrlich ascites tumour unbalances splenic cell populations and reduces responsiveness of T cells to Staphylococcus aureus enterotoxin B stimulation. Immunol Lett 2000; 74:111-5. [PMID: 10996385 DOI: 10.1016/s0165-2478(00)00208-x] [Citation(s) in RCA: 69] [Impact Index Per Article: 2.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Tumours must avoid host immune response to survive and proliferate; to achieve this purpose, tumours interact with cells of the immune system by means of tumour secreted factors. The alterations of splenic cell populations in mice bearing the Ehrlich ascites tumour have been studied. A rapid and acute response was observed, characterized by a decrease in both CD4 and CD8 T cells, and a transient increase in the number of B cells, which peaked 2 days after tumour inoculation. An increase in macrophage population and in the homing antigen CD18 was also detected. In vitro incubations of splenic cells with the Staphylococcus aureus enterotoxin B (SEB) showed that tumour induces a state of reduced responsiveness to stimulation of T cells, mainly affecting CD8 T cells, and a diminished IFN-gamma expression.
Collapse
Affiliation(s)
- J A Segura
- Departamento de Biología Molecular y Bioquímica, Facultad de Ciencias, Universidad de Málaga, 29071, Málaga, Spain.
| | | | | |
Collapse
|
125
|
Baldus SE, Hanisch FG. Biochemistry and pathological importance of mucin-associated antigens in gastrointestinal neoplasia. Adv Cancer Res 2000; 79:201-48. [PMID: 10818682 DOI: 10.1016/s0065-230x(00)79007-5] [Citation(s) in RCA: 44] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 01/21/2023]
Affiliation(s)
- S E Baldus
- Institute of Pathology, Medical Faculty, University of Cologne, Germany
| | | |
Collapse
|
126
|
Vaughan HA, Ho DW, Karanikas V, Sandrin MS, McKenzie IF, Pietersz GA. The immune response of mice and cynomolgus monkeys to macaque mucin 1-mannan. Vaccine 2000; 18:3297-309. [PMID: 10869775 DOI: 10.1016/s0264-410x(00)00143-2] [Citation(s) in RCA: 14] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/21/2022]
Abstract
Mice immunised with human epithelial mucin MUC1 coupled to oxidised mannan produce MUC1 specific MHC Class 1 restricted CD8(+) cytotoxic T cells and are completely protected from the development of MUC1(+) tumours; such therapy may be applicable to humans. In this light we describe pre-clinical studies in cynomolgus monkeys (Macaca fascicularis), to test the efficacy of mannan-MUC1 in higher primates. Monkey MUC1 genomic clones were isolated from a macaque library, peptides and fusion protein synthesised and mice and monkeys immunised with macaque MUC1-mannan. In mice CTL responses were induced (as has been found with human MUC1 mannan conjugates), but in contrast monkeys produced a humoral response, with no T cell proliferative, cytotoxic responses or CTLp found. In spite of the presence of anti-MUC1 auto-antibodies, there was no toxicity or induction of autoimmunity.
Collapse
Affiliation(s)
- H A Vaughan
- The Austin Research Institute, A&RMC, StudleyRoad, Vic 3084, Heidelberg, Australia
| | | | | | | | | | | |
Collapse
|
127
|
Kontani K, Taguchi O, Narita T, Hiraiwa N, Sawai S, Hanaoka J, Ichinose M, Tezuka N, Inoue S, Fujino S, Kannagi R. Autologous dendritic cells or cells expressing both B7‐1 and MUC1 can rescue tumor‐specific cytotoxic T lymphocytes from MUC1‐mediated apoptotic cell death. J Leukoc Biol 2000. [DOI: 10.1189/jlb.68.2.225] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022] Open
Affiliation(s)
- Keiichi Kontani
- Laboratory of Experimental Pathology, Aichi Cancer Center Research Institute, Nagoya, Japan
- Second Department of Surgery, Shiga University of Medical Science, Otsu, Japan
| | - Osamu Taguchi
- Laboratory of Experimental Pathology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Tatsuhiko Narita
- Laboratory of Experimental Pathology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Nozomu Hiraiwa
- Laboratory of Experimental Pathology, Aichi Cancer Center Research Institute, Nagoya, Japan
| | - Satoru Sawai
- Second Department of Surgery, Shiga University of Medical Science, Otsu, Japan
| | - Jun Hanaoka
- Second Department of Surgery, Shiga University of Medical Science, Otsu, Japan
| | - Masutaro Ichinose
- Second Department of Surgery, Shiga University of Medical Science, Otsu, Japan
| | - Noriaki Tezuka
- Second Department of Surgery, Shiga University of Medical Science, Otsu, Japan
| | - Shuhei Inoue
- Second Department of Surgery, Shiga University of Medical Science, Otsu, Japan
| | - Shozo Fujino
- Second Department of Surgery, Shiga University of Medical Science, Otsu, Japan
| | - Reiji Kannagi
- Laboratory of Experimental Pathology, Aichi Cancer Center Research Institute, Nagoya, Japan
| |
Collapse
|
128
|
Abstract
Standard treatments for adenocarcinoma of the prostate, such as surgery, hormones, radiation and chemotherapy, often achieve a clinical response, but this is usually short-lived. Prostate cancer frequently recurs and second-line therapies have a poor response rate. Many clinicians seem comfortable in limiting their philosophy of treating advanced recurrent disease merely to new regimens of failed therapies, such as combination chemotherapy. However, other medical researchers have chosen to pursue novel approaches, including immunotherapy, several of which are summarised in this review. Although ranging widely in antigen specificity, all attempt to exploit the body's natural antitumour immunity. Furthermore, all aim to stimulate immunity above a threshold level necessary for tumour regression or to induce stability in the face of progression. The goal of in vivo or ex vivo gene therapy is the modification of gene expression within an antigen-presented cell by the introduction of a vector, DNA, or RNA. Within that field, much progress has been made and is ongoing currently concerning gene delivery systems, target identification and characterisation. Comparatively, monoclonal antibodies are an established type of cancer immunotherapy. However, the more recent development of humanized or fully human antibodies, as well as novel moieties they can be coupled to, renews their prospects for clinical impact. Lastly, various cell-based therapies are the focus of several recent clinical studies demonstrating tumour regression or stabilisation. Immune cells, for example, T-lymphocytes and dendritic cells, have already demonstrated treatment benefit, as well as the ability to maintain an excellent quality of life for participants. Overall, there is a multitude of approaches being considered for the treatment of prostate cancer. The following review concentrates on those approaches that are currently in human or animal studies and have a specific emphasis on prostate cancer.
Collapse
Affiliation(s)
- M L Salgaller
- Northwest Biotherapeutics, Inc., 2203 Airport Way South, Suite 200, Seattle, WA 98134, USA.
| |
Collapse
|
129
|
Construction of a gene of the human tumor-associated antigen VNTR(MUC1) bound to streptavidin, its expression inEscherichia coli, and the study of properties of the hybrid protein. RUSSIAN JOURNAL OF BIOORGANIC CHEMISTRY 2000. [DOI: 10.1007/bf02758665] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.0] [Reference Citation Analysis] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 11/26/2022]
|
130
|
Pietersz GA, Li W, Osinski C, Apostolopoulos V, McKenzie IF. Definition of MHC-restricted CTL epitopes from non-variable number of tandem repeat sequence of MUC1. Vaccine 2000; 18:2059-71. [PMID: 10706970 DOI: 10.1016/s0264-410x(99)00515-0] [Citation(s) in RCA: 25] [Impact Index Per Article: 1.0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022]
Abstract
Mucin1 (MUC1) is expressed ubiquitously on breast cancer cells and is a potential target for the generation of cytotoxic T cells for vaccination against breast cancer. Thus far studies of the immunogenicity of MUC1 have used peptides from the variable number of tandem repeat (VNTR); mice so immunised can generate strong cellular and antibody responses to the VNTR of human MUC1. We now demonstrate that significant CTL and CTLp can be induced to other regions of MUC1. Using the whole native MUC1 molecule, the human milk fat globule membrane antigen (HMFG) linked to mannan, cytotoxic T cell precursors (CTLp) can be generated in BALB/c, C57BL/6, transgenic HLA-A*0201/K(b) and double transgenic HLA-A*0201/K(b)xhuman MUC1 (A2 K(b)MUC1) mice. By immunising with HMFG and testing selectively on (a) extracellular (non-VNTR); (b) VNTR and (c) intracellular peptides, it was shown that all three regions generated effective CTL. Further, the CTL responses to non-VNTR peptides were as strong as those generated to the VNTR. Epitope prediction algorithms were not particularly helpful to describe CTL epitopes: overlapping peptides had to be synthesised and tested to find the epitopes. Thus, for CTL generation, the whole HMFG molecule is a powerful immunogen when linked to mannan, especially as multiple peptide epitopes for presentation by many Class I molecules are contained within the one molecule. Furthermore, Class I restricted MUC1 CTL were generated in double transgenic A2 K(b)MUC1 mice by immunising with mannan-native mucin (HMFG), suggesting that tolerance to MUC1 can be overcome with mannan-HMFG.
Collapse
Affiliation(s)
- G A Pietersz
- The Austin Research Institute, Studley Rd, Heidelberg, Australia.
| | | | | | | | | |
Collapse
|
131
|
Bremers AJ, Parmiani G. Immunology and immunotherapy of human cancer: present concepts and clinical developments. Crit Rev Oncol Hematol 2000; 34:1-25. [PMID: 10781746 DOI: 10.1016/s1040-8428(99)00059-1] [Citation(s) in RCA: 55] [Impact Index Per Article: 2.2] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/29/2022] Open
Abstract
Immunotherapy of cancer is entering into a new phase of active investigation both at the pre-clinical and clinical level. This is due to the exciting developments in basic immunology and tumour biology that have allowed a tremendous increase in our understanding of mechanisms of interactions between the immune system and tumour cells. This review briefly summarizes the state of the art in basic tumour immunology before discussing the clinical applications of the new concepts in the clinical setting. Clinical approaches are diverse but can now be based on strong scientific rationales. The analysis of the available clinical results suggests that, despite some disappointments, there is room for optimism that both active immunotherapy (vaccination) and adoptive immunotherapy may soon become part of the therapeutic arsenal to combat cancer in a more efficient way.
Collapse
Affiliation(s)
- A J Bremers
- Unit of Immunotherapy of Human Tumours, Istituto Nazionale per lo Studio e la Cura dei Tumori, Via Venezian 1, 20133, Milan, Italy
| | | |
Collapse
|
132
|
von Mensdorff-Pouilly S, Verstraeten AA, Kenemans P, Snijdewint FG, Kok A, Van Kamp GJ, Paul MA, Van Diest PJ, Meijer S, Hilgers J. Survival in early breast cancer patients is favorably influenced by a natural humoral immune response to polymorphic epithelial mucin. J Clin Oncol 2000; 18:574-83. [PMID: 10653872 DOI: 10.1200/jco.2000.18.3.574] [Citation(s) in RCA: 164] [Impact Index Per Article: 6.6] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
PURPOSE Polymorphic epithelial mucin (PEM or MUC1) is being studied as a vaccine substrate for the immunotherapy of patients with adenocarcinoma. The present study analyzes the incidence of naturally occurring MUC1 antibodies in early breast cancer patients and relates the presence of these antibodies in pretreatment serum to outcome of disease. MATERIALS AND METHODS We measured immunoglobulin G (IgG) and immunoglobulin M (IgM) antibodies to MUC1 with an enzyme-linked immunoassay (PEM.CIg), which uses a MUC1 triple-tandem repeat peptide conjugated to bovine serum albumin, in pretreatment serum samples obtained from 154 breast cancer patients (52 with stage I disease and 102 with stage II) and 302 controls. The median disease-specific survival time of breast cancer patients was 74 months (range, 15 to 118 months). A positive test result was defined as MUC1 IgG or IgM antibody levels equal to or greater than the corresponding rounded-up median results obtained in the total breast cancer population. RESULTS A positive test result for both MUC1 IgG and IgM antibodies in pretreatment serum was associated with a significant benefit in disease-specific survival in stage I and II (P =.0116) breast cancer patients. Positive IgG and IgM MUC1 antibody levels had significant additional prognostic value to stage (P =.0437) in multivariate analysis. Disease-free survival probability did not differ significantly. However, stage II patients who tested positive for MUC1 IgG and IgM antibody and who relapsed had predominantly local recurrences or contralateral disease, as opposed to recurrences at distant sites in the patients with a negative humoral response (P =.026). CONCLUSION Early breast cancer patients with a natural humoral response to MUC1 have a higher probability of freedom from distant failure and a better disease-specific survival. MUC1 antibodies may control hematogenic tumor dissemination and outgrowth by aiding the destruction of circulating or seeded MUC1-expressing tumor cells. Vaccination of breast cancer patients with MUC1-derived (glyco)peptides in an adjuvant setting may favorably influence the outcome of disease.
Collapse
Affiliation(s)
- S von Mensdorff-Pouilly
- Departments of Obstetrics and Gynecology, Clinical Chemistry, Pathology, and Surgery, Academic Hospital Vrije Universiteit, Amsterdam, The Netherlands.
| | | | | | | | | | | | | | | | | | | |
Collapse
|
133
|
Abstract
Glycoproteins with O-glycosidically linked carbohydrate chains of complex structures and functions are found in secretions and on the cell surfaces of cancer cells. The structures of O-glycans are often unusual or abnormal in cancer, and greatly contribute to the phenotype and biology of cancer cells. Some of the mechanisms of changes in O-glycosylation pathways have been determined in cancer model systems. However, O-glycan biosynthesis is a complex process that is still poorly understood. The glycosyltransferases and sulfotransferases that synthesize O-glycans appear to exist as families of related enzymes of which individual members are expressed in a tissue- and growth-specific fashion. Studies of their regulation in cancer may reveal the connection between cancerous transformation and glycosylation which may help to understand and control the abnormal biology of tumor cells. Cancer diagnosis may be based on the appearance of certain glycosylated epitopes, and therapeutic avenues have been designed to attack cancer cells via their glycans.
Collapse
Affiliation(s)
- I Brockhausen
- Department of Biochemistry, University of Toronto, Oncology Research, Toronto Hospital, 67 College Street, Toronto, Ont., Canada.
| |
Collapse
|
134
|
Taylor-Papadimitriou J, Burchell J, Miles DW, Dalziel M. MUC1 and cancer. BIOCHIMICA ET BIOPHYSICA ACTA 1999; 1455:301-13. [PMID: 10571020 DOI: 10.1016/s0925-4439(99)00055-1] [Citation(s) in RCA: 347] [Impact Index Per Article: 13.3] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/10/2023]
Abstract
The MUC1 membrane mucin was first identified as the molecule recognised by mouse monoclonal antibodies directed to epithelial cells, and the cancers which develop from them. Cloning the gene showed that the extracellular domain is made up of highly conserved repeats of 20 amino acids, the actual number varying between 25 and 100 depending on the allele. Each tandem repeat contains five potential glycosylation sites, and between doublets of threonines and serines lies an immunodominant region which contains the epitopes recognised by most of the mouse monoclonal antibodies. The O-glycans added to the mucin produced by the normal breast are core 2 based and can be complex, while the O-glycans added to the breast cancer mucin are mainly core 1 based. This means that some core protein epitopes in the tandem repeat which are masked in the normal mucin are exposed in the cancer associated mucin. Since novel carbohydrate epitopes are also carried on the breast cancer mucin, the molecule is antigenically distinct from the normal breast mucin. (Changes in glycosylation in other epithelial cancers have been observed but are not so well documented.) Immune responses to MUC1 have been seen in breast and ovarian cancer patients and clinical studies have been initiated to evaluate the use of antibodies to MUC1 and of immunogens based on MUC1 for immunotherapy of these patients. The role of the carbohydrates in the immune response and in other interactions with the effector cells of the immune system is of particular interest and is discussed.
Collapse
|
135
|
Abstract
The adoptive transfer of tumor-infiltrating lymphocytes (TIL) along with interleukin (IL)-2 into autologous patients with cancer resulted in the objective regression of tumor, indicating that T cells play an important role in tumor regression. In the last few years, efforts have been made towards understanding the molecular basis of T-cell-mediated antitumor immunity and elucidating the molecular nature of tumor antigens recognized by T cells. Tumor antigens identified thus far could be classified into several categories: tissue-specific differentiation antigens, tumor-specific shared antigens and tumor-specific unique antigens. CD4+ T cells play a central role in orchestrating the host immune response against cancer, infectious diseases and autoimmune diseases, and we thus have attempted to identify major histocompatibility complex (MHC) class II-restricted tumor antigens as well. The identification of tumor rejection antigens provides new opportunities for the development of therapeutic strategies against cancer. This review will summarize the current status of MHC class I- and class II-restricted human tumor antigens, and their potential application to cancer treatment.
Collapse
Affiliation(s)
- R F Wang
- Surgery Branch, National Cancer Institute, NIH, Bethesda, MD 20814, USA.
| | | |
Collapse
|
136
|
Hiltbold EM, Alter MD, Ciborowski P, Finn OJ. Presentation of MUC1 tumor antigen by class I MHC and CTL function correlate with the glycosylation state of the protein taken Up by dendritic cells. Cell Immunol 1999; 194:143-9. [PMID: 10383817 DOI: 10.1006/cimm.1999.1512] [Citation(s) in RCA: 62] [Impact Index Per Article: 2.4] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/22/2022]
Abstract
We previously reported that the glycosylated MUC1 tumor antigen circulating as soluble protein in patients' serum is not processed by dendritic cells and does not elicit MHC-Class II-restricted T helper responses in vitro. In contrast, a long synthetic peptide from the MUC1 tandem repeat region is presented by Class II molecules, resulting in the initiation of T helper cell responses. Here we addressed the ability of dendritic cells to present various glycosylated or not glycosylated forms of MUC1 by MHC Class I. We found that three different forms of MUC1, ranging from glycosylated and underglycosylated protein to unglycosylated synthetic peptide, were able to elicit MUC1-specific, Class-I-restricted CTL responses. The efficiency of processing and the resulting strength of CTL activity were inversely correlated with the degree of glycosylation of the antigen. Furthermore, the more efficiently processed 100mer peptide primed a broader repertoire of CTL than the glycosylated protein.
Collapse
Affiliation(s)
- E M Hiltbold
- University of Pittsburgh School of Medicine, Pittsburgh, Pennsylvania, 15261, USA
| | | | | | | |
Collapse
|
137
|
Identification of HLA-A2–Restricted T-Cell Epitopes Derived From the MUC1 Tumor Antigen for Broadly Applicable Vaccine Therapies. Blood 1999. [DOI: 10.1182/blood.v93.12.4309.412k19_4309_4317] [Citation(s) in RCA: 17] [Impact Index Per Article: 0.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/22/2022] Open
Abstract
The tumor-associated antigen MUC1 is overexpressed on various hematological and epithelial malignancies and is therefore a suitable candidate for broadly applicable vaccine therapies. It was demonstrated that major histocompatibility complex (MHC)-unrestricted cytotoxic T cells can recognize epitopes of the MUC1 protein core localized in the tandem repeat domain. There is increasing evidence now that MHC-restricted T cells can also be induced after immunization with the MUC1 protein or segments of the core tandem repeat. Using a computer analysis of the MUC1 amino acid sequence, we identified two novel peptides with a high binding probability to the HLA-A2 molecule. One of the peptides is derived from the tandem repeat region and the other is derived from the leader sequence of the MUC1 protein, suggesting that, in contrast to previous reports, the MUC1-directed immune responses are not limited to the extracellular tandem repeat domain. Cytotoxic T cells (CTL) were generated from several healthy donors by primary in vitro immunization using peptide-pulsed dendritic cells. The addition of a Pan-HLA-DR binding peptide PADRE as a T-helper epitope during the in vitro priming resulted in an increased cytotoxic activity of the MUC1-specific CTL and a higher production of cytokines such as interleukin-12 and interferon-γ in the cell cultures, demonstrating the importance of CD4 cells for an efficient CTL priming. The peptide induced CTL lysed tumors endogenously expressing MUC1 in an antigen-specific and HLA-A2–restricted fashion, including breast and pancreatic tumor cells as well as renal cell carcinoma cells, showing that these peptides are shared among many tumors. The use of MUC1-derived peptides could provide a broadly applicable approach for the development of dendritic cell-based vaccination therapies.
Collapse
|
138
|
Identification of HLA-A2–Restricted T-Cell Epitopes Derived From the MUC1 Tumor Antigen for Broadly Applicable Vaccine Therapies. Blood 1999. [DOI: 10.1182/blood.v93.12.4309] [Citation(s) in RCA: 203] [Impact Index Per Article: 7.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/20/2022] Open
Abstract
AbstractThe tumor-associated antigen MUC1 is overexpressed on various hematological and epithelial malignancies and is therefore a suitable candidate for broadly applicable vaccine therapies. It was demonstrated that major histocompatibility complex (MHC)-unrestricted cytotoxic T cells can recognize epitopes of the MUC1 protein core localized in the tandem repeat domain. There is increasing evidence now that MHC-restricted T cells can also be induced after immunization with the MUC1 protein or segments of the core tandem repeat. Using a computer analysis of the MUC1 amino acid sequence, we identified two novel peptides with a high binding probability to the HLA-A2 molecule. One of the peptides is derived from the tandem repeat region and the other is derived from the leader sequence of the MUC1 protein, suggesting that, in contrast to previous reports, the MUC1-directed immune responses are not limited to the extracellular tandem repeat domain. Cytotoxic T cells (CTL) were generated from several healthy donors by primary in vitro immunization using peptide-pulsed dendritic cells. The addition of a Pan-HLA-DR binding peptide PADRE as a T-helper epitope during the in vitro priming resulted in an increased cytotoxic activity of the MUC1-specific CTL and a higher production of cytokines such as interleukin-12 and interferon-γ in the cell cultures, demonstrating the importance of CD4 cells for an efficient CTL priming. The peptide induced CTL lysed tumors endogenously expressing MUC1 in an antigen-specific and HLA-A2–restricted fashion, including breast and pancreatic tumor cells as well as renal cell carcinoma cells, showing that these peptides are shared among many tumors. The use of MUC1-derived peptides could provide a broadly applicable approach for the development of dendritic cell-based vaccination therapies.
Collapse
|
139
|
Vaughan HA, Ho DW, Karanikas VA, Ong CS, Hwang LA, Pearson JM, McKenzie IF, Pietersz GA. Induction of humoral and cellular responses in cynomolgus monkeys immunised with mannan-human MUC1 conjugates. Vaccine 1999; 17:2740-52. [PMID: 10418926 DOI: 10.1016/s0264-410x(98)00493-9] [Citation(s) in RCA: 20] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/24/2022]
Abstract
Mice immunised with oxidised mannan conjugated to the human mucin 1 (MUC1), produce MHC Class 1 restricted CD8+ cytotoxic T-cells which eradicate MUC1 + tumours, indicating potential for the immunotherapy of MUC1 + cancers in humans. We now describe preclinical studies performed in cynomolgus monkeys immunised with human or murine MUC1 conjugated to oxidised mannan, where immune responses and toxicity were examined. High titred antibodies specific for MUC1 were produced, MUC1 specific CD4+ and CD8+ T-cell proliferative responses and specific cytotoxic precursor cells (CTLp) were found, but not MUC1 specific cytotoxic T-cells (CTL). There was no toxicity and monkeys can be immunised against human MUC1 with mannan-MUC1 conjugates, but a humoral response (Th2 type) predominates. The results contrast with those obtained in mice when a CTL response (Th1 type) predominates.
Collapse
Affiliation(s)
- H A Vaughan
- Austin Research Institute, Austin and Repatriation Medical Centre, Heidelberg, Vic., Australia
| | | | | | | | | | | | | | | |
Collapse
|
140
|
Miles DW, Taylor-Papadimitriou J. Therapeutic aspects of polymorphic epithelial mucin in adenocarcinoma. Pharmacol Ther 1999; 82:97-106. [PMID: 10341360 DOI: 10.1016/s0163-7258(99)00003-0] [Citation(s) in RCA: 22] [Impact Index Per Article: 0.8] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 10/18/2022]
Abstract
The gene MUC1 encodes a large membrane-associated glycoprotein, previously termed polymorphic epithelial mucin and now known as MUC1. The majority of the extracellular domain is made up of tandem repeats of 20 amino acids. In some epithelial malignancies, MUC1 is up-regulated, and as a result of changes in glycosyl and sialytransferases, the complex carbohydrate side chains are truncated, leading to exposure of novel peptide and carbohydrate epitopes. Cellular and humoral immune responses to MUC1 have been documented in malignant disease, and T-cell responses to MUC1 may not depend on presentation by the major histocompatibility complex. Several immunogens based on MUC1 are being investigated. These include cell lines expressing MUC1 given alone or fused with professional antigen-presenting cells and peptide epitopes, given either with conventional immunological adjuvants or coupled to mannan, which may target uptake into antigen-presenting cells. Cellular and humoral immune responses to these agents have been recorded in patients with advanced malignancy. Targeting of peptide epitopes may also be achieved using antibodies to MUC1 through induction of idiotypes and retrospective analyses in ovarian cancer have suggested a survival benefit for patients. The use of cDNA in coding MUC1 may allow endogenous processing of antigen. Phase I studies using vaccinia as a vector have been completed. Studies using carbohydrate antigens suggest that the ability to generate specific immune responses may influence survival of patients with metastatic epithelial malignancies. While examining the potential role of immunogens based on MUC1, it is also necessary to understand the nature of immunosuppression in patients with advanced malignancy in order to develop strategies to enhance the immunogenicity of potential cancer vaccines.
Collapse
Affiliation(s)
- D W Miles
- ICRF Immunotherapy Laboratory, Guy's Hospital, London, UK
| | | |
Collapse
|
141
|
|
142
|
Kilger E, Pecher G, Schwenk A, Hammerschmidt W. Expression of mucin (MUC-1) from a mini-Epstein-Barr virus in immortalized B-cells to generate tumor antigen specific cytotoxic T cells. J Gene Med 1999; 1:84-92. [PMID: 10738572 DOI: 10.1002/(sici)1521-2254(199903/04)1:2<84::aid-jgm21>3.0.co;2-q] [Citation(s) in RCA: 13] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/08/2022] Open
Abstract
BACKGROUND EBV immortalized B-cells can be used as antigen presenting cells (APC) to stimulate specific T-cell responses. Mini-Epstein-Barr virus (mini-EBV) plasmids contain all functional elements of Epstein-Barr virus (EBV) necessary to immortalize B-cells in vitro. These immortalized B-cells are incapable of releasing infectious virus in contrast to cells immortalized by wildtype EBV. In addition, mini-EBVs can be modified in E. coli to alter their genetic composition or adopt new genes. METHODS We constructed a mini-EBV plasmid carrying an expression cassette for the human tumor antigen mucin encoded by the gene MUC-1. Primary human B-cells were infected with the MUC-1 carrying mini-EBV plasmid packaged into an EBV coat and immortalized B-cell clones were expanded in vitro. These B-cells were analyzed by FACS analyses for the expression of mucin and co-stimulatory molecules and were subsequently used as antigen presenting cells (APC) to stimulate peripheral blood mononuclear cells from healthy donors. RESULTS Several B-cell lines were established that were shown to be free of helper virus or wildtype EBV. These B-cells expressed the relevant tumor-specific epitopes of mucin and the co-stimulatory ligands B7.1 and B7.2 necessary for efficient T-cell activation. Using the mucin expressing B-cells as antigen presenting cells (APC) mucin-epitope specific cytotoxic T-cells were established. CONCLUSIONS Virus-free B-cell lines expressing tumor-associated epitopes such as mucin or other antigens of interest provide an unlimited and safe source of APC to generate antigen specific T-cells which could be used for clinical trials in adoptive immune therapy or cancer vaccines.
Collapse
Affiliation(s)
- E Kilger
- GSF-National Research Center for Environment and Health, Institut für Klinische Molekularbiologie und Tumorgenetik, Munich, Germany
| | | | | | | |
Collapse
|
143
|
|
144
|
Wölfel T. Identification of Tumor Antigens Defined by Cytolytic T Lymphocytes and Therapeutic Implications. Gene Ther 1999. [DOI: 10.1007/978-3-0348-7011-5_20] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
|
145
|
Tempero RM, VanLith ML, Morikane K, Rowse GJ, Gendler SJ, Hollingsworth MA. CD4+ Lymphocytes Provide MUC1-Specific Tumor Immunity In Vivo That Is Undetectable In Vitro and Is Absent in MUC1 Transgenic Mice. THE JOURNAL OF IMMUNOLOGY 1998. [DOI: 10.4049/jimmunol.161.10.5500] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Subscribe] [Scholar Register] [Indexed: 01/01/2023]
Abstract
Abstract
A C57BL/6 mouse transgenic for human MUC1 (MUC1.Tg) was developed to evaluate MUC1-specific tumor immunity in an animal that expresses MUC1 as a normal self protein. Previous studies showed that MUC1.Tg mice, challenged with syngeneic tumors expressing MUC1 (B16.MUC1), developed progressively growing MUC1-positive tumors, whereas wild-type C57BL/6 (wt) mice developed MUC1-negative tumors at a significantly slower rate. The results of a limiting dilution CTL frequency assay were not informative, in that similar numbers of MUC1-specific CTL precursors (CTL) were detected in MUC1.Tg and wt mice. Tumor immunity in vivo was characterized by an adoptive transfer method to evaluate the degree of MUC1 or non-MUC1 tumor immunity in wt or MUC1.Tg mice. The results revealed that wt mice developed protective tumor immunity mediated by MUC1-specific CD4+ lymphocytes, while MUC1.Tg mice were functionally tolerant to MUC1 in vivo. The potential of adoptive immunotherapy to provide immunity to tumors expressing MUC1 and to produce undesirable autoimmunity in recipient MUC1.Tg mice expressing MUC1 as a self Ag was evaluated. Adoptive transfer of immune cells from wt mice primed in vivo with B16.MUC1 tumor cells into MUC1.Tg recipients resulted in significant increases in the survival of MUC1.Tg recipients compared with unmanipulated control MUC1.Tg mice challenged with B16.MUC1 tumor cells. This response was specific for MUC1 since control tumors developed at equivalent rates in recipient or control MUC1.Tg mice. No gross or histologic evidence of autoimmunity was observed in recipient MUC1.Tg mice, indicating that tumor immune responses mediated by MUC1-specific CD4+ lymphocytes spare nontransformed epithelia-expressing MUC1.
Collapse
Affiliation(s)
- Richard M. Tempero
- *Eppley Institute for Research in Cancer and Allied Disease, University of Nebraska Medical Center, Omaha, NE 68198; and
| | - Michelle L. VanLith
- *Eppley Institute for Research in Cancer and Allied Disease, University of Nebraska Medical Center, Omaha, NE 68198; and
| | - Keita Morikane
- *Eppley Institute for Research in Cancer and Allied Disease, University of Nebraska Medical Center, Omaha, NE 68198; and
| | | | | | - Michael A. Hollingsworth
- *Eppley Institute for Research in Cancer and Allied Disease, University of Nebraska Medical Center, Omaha, NE 68198; and
| |
Collapse
|
146
|
Chen XT, Sames D, Danishefsky SJ. Exploration of Modalities in Building α-O-Linked Systems through Glycal Assembly: A Total Synthesis of the Mucin-Related F1α Antigen. J Am Chem Soc 1998. [DOI: 10.1021/ja980724z] [Citation(s) in RCA: 57] [Impact Index Per Article: 2.1] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/28/2022]
Affiliation(s)
- Xiao-Tao Chen
- Contribution from the Department of Chemistry, Columbia University, New York, New York 10027; and Laboratory for Bioorganic Chemistry, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, New York 10021
| | - Dalibor Sames
- Contribution from the Department of Chemistry, Columbia University, New York, New York 10027; and Laboratory for Bioorganic Chemistry, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, New York 10021
| | - Samuel J. Danishefsky
- Contribution from the Department of Chemistry, Columbia University, New York, New York 10027; and Laboratory for Bioorganic Chemistry, Memorial Sloan-Kettering Cancer Center, 1275 York Avenue, New York, New York 10021
| |
Collapse
|
147
|
Wei WZ, Pauley R, Lichlyter D, Soule H, Shi WP, Calaf G, Russo J, Jones RF. Neoplastic progression of breast epithelial cells--a molecular analysis. Br J Cancer 1998; 78:198-204. [PMID: 9683293 PMCID: PMC2062906 DOI: 10.1038/bjc.1998.464] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Grants] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/08/2023] Open
Abstract
Molecular changes associated with breast cancer progression were characterized using the MCF-10F cell series. MCF-10F was established from fibrous mastectomy tissue of a patient without detectable cancer. In vitro treatment of MCF-10F cells with benzo(a)pyrene resulted in a transformed subclone MCF-10F-BP1 (BP1). Transfection of clone BP1 with T24-Hras resulted in the tumorigenic line MCF-10F-BP1-Tras (BP1-Tras). Using flow cytometry, the expression of HLA I, ERBB-2 and MUC-1 was found to be comparable in 'normal' MCF-10F, transformed BP1 and tumorigenic BP1-Tras cells. Glycosylated mucin is elevated in BP1 but reduced in BP1-Tras cells. Using mRNA differential display analysis, cDNA profiles of the 'normal', transformed and tumorigenic cell lines were strikingly similar, yet distinct and elevated expression of several common cDNA fragments was detected in BP1 and BP1-Tras when compared with MCF-10F cells. These fragments were cloned and sequenced. The sequences of clones T1-360 and C4-310 are homologous to two reported EST cDNA clones from human fetal tissue and were further characterized. Elevated expression of the genes corresponding to clones T1-360 and C4-310 was verified using Northern blotting. High-level expression of these genes was also detected in the breast cancer cell line MCF-7 that was derived from the pleural effusion of a patient with advanced breast cancer. Therefore, specific molecular changes associated with breast cancer development were identified and may be indicators of neoplastic progression.
Collapse
Affiliation(s)
- W Z Wei
- Karmanos Cancer Institute, Wayne State University, Detroit, MI 48201, USA
| | | | | | | | | | | | | | | |
Collapse
|
148
|
Abstract
Cellular and cytokine adjuvants, often immune effector cells and soluble factors, respectively, are supplemental and/or follow-up treatments of human origin for cancer patients who have unsatisfactory clinical responses to conventional chemotherapy, radiotherapy, and surgery. Since many human studies with these reagents are in their infancy, extensive data collection is only now being performed to determine which strategy provides the greatest therapeutic benefit. Research published in the literature since the genesis of this approach to cancer treatment is summarized in this report. Methodologies attempting to generate anticancer responses by provoking or enhancing the patient's own immune system are new compared with the other standard types of cancer treatment. Although a few encouraging human studies can be discussed, many of the most promising techniques are only now being transferred from the laboratory to the clinic. The administration of immune effector cells in combination with immunomodulators, such as interferons or interleukins, often enhances clinical outcome. The literature cited in this report indicate that immune-cell- and cytokine-based therapies hold promise in our attempts to improve the quality and duration of life in those with cancer. With each report reaching the literature, more effective clinical trials are being designed and implemented.
Collapse
Affiliation(s)
- M L Salgaller
- Pacific Northwest Cancer Foundation and Immunotherapeutics Division, Northwest Biotherapeutics, L.L.C., Seattle, Washington 98125, USA.
| | | |
Collapse
|
149
|
Munster DJ, Quin RJ, Bansal AS, Ward BG, McGuckin MA. Peritoneal fluid from ovarian cancer patients stimulates MUC1 epithelial mucin expression in ovarian cancer cell lines. Int J Cancer 1998; 76:393-8. [PMID: 9579577 DOI: 10.1002/(sici)1097-0215(19980504)76:3<393::aid-ijc17>3.0.co;2-c] [Citation(s) in RCA: 3] [Impact Index Per Article: 0.1] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/09/2022]
Abstract
The MUC1 epithelial mucin is a transmembrane glycoprotein that is frequently but variably over-expressed by adenocarcinomas. It is used as a diagnostic serum tumour marker and is a candidate target for tumour immunotherapy. Peritoneal fluid (PF) samples from ovarian cancer patients were investigated for their ability to modulate MUC1 expression in 6 ovarian cancer cell lines which showed a range from very low to high endogenous MUC1 expression. Cell lines were cultured in 20% PF for 4 days, fixed in situ and MUC1 assayed by ELISA. MUC1 expression was stimulated by some PF samples in 5 of 6 lines tested. MUC1 expression in the PE04 cell line (very low endogenous expression) was increased by 35 of 36 PFs tested (p < 0.05); stimulation varied between PFs but was greater than with 100 IU/mL hu-r-gamma-interferon. Western blotting confirmed the stimulation of MUC1 in PE04 cells and FACS showed an increase in the proportion of cells expressing MUC1. The active factor was partially purified by gel filtration and was shown to stimulate PE04 cells in a dose-dependent manner. Concentrations of IL1beta, IL4, IL6, IL8, IL10, TNF-alpha, TGF-beta and GM-CSF were often very high in PF and varied substantially between different PF samples but did not correlate with the degree of MUC1 stimulatory activity.
Collapse
Affiliation(s)
- D J Munster
- Department of Obstetrics and Gynaecology, The University of Queensland, Royal Brisbane Hospital, Herston, Australia
| | | | | | | | | |
Collapse
|
150
|
Krambovitis E, Hatzidakis G, Barlos K. Preparation of MUC-1 oligomers using an improved convergent solid-phase peptide synthesis. J Biol Chem 1998; 273:10874-9. [PMID: 9556562 DOI: 10.1074/jbc.273.18.10874] [Citation(s) in RCA: 24] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/06/2022] Open
Abstract
The sequentially repeating nature of the core mucin polypeptide chain MUC-1 on the surface of malignant cells makes it an excellent target for cancer immunotherapy. We describe a reliable and efficient method of synthesizing oligomers, up to five tandem repeats and oligomer heterotope derivatives with a 15-amino acid epitope from tetanus toxin using an improved convergent solid-phase peptide synthesis. The different oligomers were easily distinguishable by reverse-phase high pressure liquid chromatography, but they were poorly fixed and migrated with the same migration rate, irrespective of size, in electrophoretic studies. In contrast, the oligomer heterotopes exhibited size-dependent electrophoretic behavior but in high pressure liquid chromatography chromatograms the different heterotopes were eluted simultaneously in two peaks representing the L- and D-enantiomers of the derivatives. The oligomer heterotopes were recognized as antigens in Western blotting with a murine monoclonal antibody against the epitope APDTR. In enzyme immunoassay studies with the same antibody an increasing reactivity was observed against the larger oligomers and confirmed by inhibition assays as the MUC-1 pentamer was the most efficient inhibitor. These results support the suggestion that the pentamer attains a structure closer to the native conformation and is more immunogenic. In conclusion, large composite peptides can be reliably synthesized with the convergent solid-phase peptide strategy offering an attractive option to vaccine designing and development.
Collapse
Affiliation(s)
- E Krambovitis
- Department of Applied Biochemistry and Immunology, Institute of Molecular Biology and Biotechnology, P. O. Box 1527, Heraklion GR 71110, Crete, Greece.
| | | | | |
Collapse
|