The long-term objective of this research is to dissect the tumor-specific antigenicity into its multiple separate components, so that we can determine the functional and molecular relationship of the antigens. To do this, tumor-specific T-cell clones and monoclonal antibodies will be used to select for tumor variants that express defined antigenic components. By consecutive cycles of selection for variants, the size of the antigenic repertoire of independently segregating unique tumor antigens will be determined. The frequency of antigen loss variants will be analyzed, and it will be established by fluctuation and analysis whether antigen loss is due to mutation or regulation. Monoclonal antibodies will be generated which have unique specificity for variants expressing T-cell-defined components. If a cytolytic or helper T-cell clone and a monoclonal antibody recognize the same unique antigen, then either of the two probes should select For variants that have specifically lost sensitivity to both probes. If possible, the T-cell recognition of the antibody-defined unique tumor antigen will be confirmed using L cells that have been transfected with genes coding for the unique tumor-specific antigen. Finally, the nature and molecular independence of the unique tumor antigens recognized by the monoclonal antibodies will be determined by immune precipitation and two-dimensional gel electrophoresis to indicate whether the multiple unique antigens belong to a family of related molecules. The hierarchy of response to multiple independent tumor antigens on an individual cancer cell means that the host only responds to one or a few of the multiple antigens. Therefore, the reasons for the hierarchical recognition of immunodominant tumor antigens will be examined. A clinical objective is to devise effective immunotherapy against cancer. Immune resistance should be increased if ways can be found to induce the host to respond to the recessive antigens or if a combination of monoclonal antibodies and/or T cells can be given passively (in analogy with the use of combination chemotherapy). (AG)

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA037156-02
Application #
3174901
Study Section
Immunobiology Study Section (IMB)
Project Start
1984-07-01
Project End
1987-06-30
Budget Start
1985-07-01
Budget End
1986-06-30
Support Year
2
Fiscal Year
1985
Total Cost
Indirect Cost
Name
University of Chicago
Department
Type
Schools of Medicine
DUNS #
225410919
City
Chicago
State
IL
Country
United States
Zip Code
60637
Harris, Daniel T; Hager, Marlies V; Smith, Sheena N et al. (2018) Comparison of T Cell Activities Mediated by Human TCRs and CARs That Use the Same Recognition Domains. J Immunol 200:1088-1100
Arina, Ainhoa; Karrison, Theodore; Galka, Eva et al. (2017) Transfer of Allogeneic CD4+ T Cells Rescues CD8+ T Cells in Anti-PD-L1-Resistant Tumors Leading to Tumor Eradication. Cancer Immunol Res 5:127-136
Kammertoens, Thomas; Friese, Christian; Arina, Ainhoa et al. (2017) Tumour ischaemia by interferon-? resembles physiological blood vessel regression. Nature 545:98-102
Posey Jr, Avery D; Schwab, Robert D; Boesteanu, Alina C et al. (2016) Engineered CAR T Cells Targeting the Cancer-Associated Tn-Glycoform of the Membrane Mucin MUC1 Control Adenocarcinoma. Immunity 44:1444-54
Arina, Ainhoa; Idel, Christian; Hyjek, Elizabeth M et al. (2016) Tumor-associated fibroblasts predominantly come from local and not circulating precursors. Proc Natl Acad Sci U S A 113:7551-6
Leisegang, Matthias; Engels, Boris; Schreiber, Karin et al. (2016) Eradication of Large Solid Tumors by Gene Therapy with a T-Cell Receptor Targeting a Single Cancer-Specific Point Mutation. Clin Cancer Res 22:2734-43
Blankenstein, Thomas; Leisegang, Matthias; Uckert, Wolfgang et al. (2015) Targeting cancer-specific mutations by T cell receptor gene therapy. Curr Opin Immunol 33:112-9
Binder, David C; Schreiber, Hans (2014) Dual blockade of PD-1 and CTLA-4 combined with tumor vaccine effectively restores T-cell rejection function in tumors--letter. Cancer Res 74:632; discussion 635
Arina, Ainhoa; Schreiber, Karin; Binder, David C et al. (2014) Adoptively transferred immune T cells eradicate established tumors despite cancer-induced immune suppression. J Immunol 192:1286-93
Binder, David C; Engels, Boris; Arina, Ainhoa et al. (2013) Antigen-specific bacterial vaccine combined with anti-PD-L1 rescues dysfunctional endogenous T cells to reject long-established cancer. Cancer Immunol Res 1:123-33

Showing the most recent 10 out of 65 publications