The central hypothesis of this proposal is that the immune system of the cancer-bearing host can be induced to respond to tissue-specific (differentiation) autoantigens. We propose that immune responses to tissue-specific antigens can mediate tumor rejection. A corollary to this proposition is that tumor cells are normally unable to present antigens effectively to the immune system. As a model, we propose to study the response to gp75, an abundant tissue-specific antigen whose expression is absolutely restricted to normal melanocytes and melanoma cells. The gp75 antigen, the gene product of the brown locus, is a transmembrane glycoprotein expressed in melanosomes. The gp75 antigen can be recognized by the immune system, and thus is a potential target for immunotherapy of melanoma. In our initial studies, we will examine methods to immunize against gp75 in a syngeneic mouse system (C57BL mice) using: 1) gp75+ melanoma cells; 2) gp75 melanoma cells expressing transfected or transduced gp75 or truncated, unstable constructs of gp75; 3) gp75+ melanoma cells secreting interleukin-2 or interferon-gamma (transduced by retroviral vectors); 4) purified gp75 protein and peptide fragments of gp75; and 5) gp75 expressed by or presented by antigen-presenting cells, including macrophages and dendritic cells. We will model immune responses to tumor-specific antigens by introducing mutations into the gp75 gene to construct potentially immunogenic T cell and B cell epitopes. We propose that immune responses generated against immunogenic tumor-specific epitopes (e.g. against mutations) can lead to immune responses both against the tumor-specific determinant and against native, non-mutant differentiation antigens expressed by parental tumor cells. The ability of immune responses against gp75 or mutated gp75 to reject melanoma tumors will be investigated. We will use a transplantable melanoma tumor and melanomas developing de novo in a transgenic mouse system. These studies will give insights into immune recognition of differentiation and tumor-specific antigens expressed by cancer cells and provide strategies for construction of cancer vaccines.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
1R01CA056821-01
Application #
3201238
Study Section
Experimental Immunology Study Section (EI)
Project Start
1992-06-01
Project End
1995-05-31
Budget Start
1992-06-01
Budget End
1993-05-31
Support Year
1
Fiscal Year
1992
Total Cost
Indirect Cost
Name
Sloan-Kettering Institute for Cancer Research
Department
Type
DUNS #
064931884
City
New York
State
NY
Country
United States
Zip Code
10065
Oseledchyk, Anton; Ricca, Jacob M; Gigoux, Mathieu et al. (2018) Lysis-independent potentiation of immune checkpoint blockade by oncolytic virus. Oncotarget 9:28702-28716
Zamarin, Dmitriy; Ricca, Jacob M; Sadekova, Svetlana et al. (2018) PD-L1 in tumor microenvironment mediates resistance to oncolytic immunotherapy. J Clin Invest 128:1413-1428
Ribas, Antoni; Wolchok, Jedd D (2018) Cancer immunotherapy using checkpoint blockade. Science 359:1350-1355
Ricca, Jacob M; Oseledchyk, Anton; Walther, Tyler et al. (2018) Pre-existing Immunity to Oncolytic Virus Potentiates Its Immunotherapeutic Efficacy. Mol Ther 26:1008-1019
Zamarin, Dmitriy; Holmgaard, Rikke B; Ricca, Jacob et al. (2017) Intratumoral modulation of the inducible co-stimulator ICOS by recombinant oncolytic virus promotes systemic anti-tumour immunity. Nat Commun 8:14340
Dai, Peihong; Wang, Weiyi; Yang, Ning et al. (2017) Intratumoral delivery of inactivated modified vaccinia virus Ankara (iMVA) induces systemic antitumor immunity via STING and Batf3-dependent dendritic cells. Sci Immunol 2:
Budhu, Sadna; Schaer, David A; Li, Yongbiao et al. (2017) Blockade of surface-bound TGF-? on regulatory T cells abrogates suppression of effector T cell function in the tumor microenvironment. Sci Signal 10:
Holmgaard, Rikke B; Brachfeld, Alexandra; Gasmi, Billel et al. (2016) Timing of CSF-1/CSF-1R signaling blockade is critical to improving responses to CTLA-4 based immunotherapy. Oncoimmunology 5:e1151595
Holmgaard, Rikke B; Zamarin, Dmitriy; Lesokhin, Alexander et al. (2016) Targeting myeloid-derived suppressor cells with colony stimulating factor-1 receptor blockade can reverse immune resistance to immunotherapy in indoleamine 2,3-dioxygenase-expressing tumors. EBioMedicine 6:50-58
Zou, Weiping; Wolchok, Jedd D; Chen, Lieping (2016) PD-L1 (B7-H1) and PD-1 pathway blockade for cancer therapy: Mechanisms, response biomarkers, and combinations. Sci Transl Med 8:328rv4

Showing the most recent 10 out of 78 publications