Self antigens, in the form of differentiation antigens, and altered self antigens, in the form of mutations, are recognized on melanoma and other cancers by the immune system. Although mutations are an accepted basis for tumor-specific antigens, we and others have shown that recognition of differentiation antigens is also relevant to immunity to cancer. Recognition of these antigens presents problems, especially how one can overcome immune tolerance. We have used a mouse melanoma model to show that mice can be tolerant to the tyrosinase family of differentiation antigens. Tolerance can temporarily be broken by immunization with xenogeneic orthologues of antigen. However, our preliminary results suggest that a CD4+ cell population and B cells suppress immune responses to melanoma.
Our first aim addresses the role of CD4+ and B cells n suppressing tumor immunity and memory responses, particularly to different action: antigens. Our next two aims turn to the consequences of immune recognition of mutations. We have developed methods to create combinatorial libraries of mutated differentiation antigens to address what types of mutations can lead to autoimmunity against the non-mutated parental antigen (aim 2) and what type of mutations can lead directly to tumor immunity (aim 3).
In aim 4, we will continue to create more relevant mouse models of melanoma that develop endogenous invasive melanomas to study tumor immunity against differentiation and mutant antigens.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA056821-15
Application #
7317812
Study Section
Experimental Immunology Study Section (EI)
Program Officer
Welch, Anthony R
Project Start
1992-06-01
Project End
2009-11-30
Budget Start
2007-12-01
Budget End
2008-11-30
Support Year
15
Fiscal Year
2008
Total Cost
$294,335
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:
Malandro, Nicole; Budhu, Sadna; Kuhn, Nicholas F et al. (2016) Clonal Abundance of Tumor-Specific CD4(+) T Cells Potentiates Efficacy and Alters Susceptibility to Exhaustion. Immunity 44:179-193
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

Showing the most recent 10 out of 78 publications