Melanosomal differentiation antigens are a family of proteins uniquely expressed on melanocytes and melanoma cells. As such, they represent rational and convenient surrogate markers to measure immune responses to melanoma in both animal model systems and human patients on clinical trials of innovative immunotherapeutic strategies. They also are very attractive targets for immunotherapies given their restricted expression on normal cells. Given the progress made during the prior funding period, we have proposed three specific aims. The first is to continue our focus on GITR (glucocorticoid-induced TNF-receptor family related protein) as a novel target of agonist immunotherapeutic strategies. We defined a novel mechanism underlying GITR-induced immune modulation with regulatory T cells (Treg) losing lineage commitment and a favorable change in the Treg:T effector cell ratio. This will be applied to a first-in-human clinical trial of an agonist monoclonal antibody to GITR, which we are currently leading as well as a Phase I trial of the Merck anti-GITR antibody. We will further these biomarker investigations by exploring the expression of immune regulatory molecules on tumor cells. We also plan to further investigate the basis for sub-optimal response at later timepoints (refractory vs. responding tumors) using assessment of these biomarkers over time.
In Aim 2, we will continue our efforts to identify the most impactful and relevant combinations of immune modulators by studying the effects of dual costimulation with agonist antibodies against the TNFR superfamily receptors OX40 and GITR in transplantable and spontaneous mouse models of melanoma, using immunity to melanosomal antigens as an outcome measure. This is directly applicable to clinical trials as we have ongoing collaborations with industry partners to make clinical grade reagents available for further trials.
In Aim 3, we will have a unique opportunity to directly compare two cell-based immunotherapeutic strategies, chimeric antigen receptor T cells (CAR+) and T cells bearing a transgenic T cell receptor for the same antigen that the CAR+ cells recognize (TRP1, a melanosomal antigen). We will compare different host cells as well as antigen receptors to define an optimal cell therapy to combine with the antibody combination defined in prior Aims. Our overall goal is to use immunity to melanosomal antigens to identify the most potent immunotherapeutic strategies for melanoma that can be brought into clinical trials.

Public Health Relevance

Despite significant advances in the past 5 years, melanoma continues to be a pervasive public health problem and immunotherapy represents the most feasible way to achieve durable control of this otherwise fatal illness. We propose novel and translational cellular and antibody-based strategies to optimize immune response to target molecules on melanoma, known as differentiation antigens. Some of these strategies are currently being tested in clinical trials in melanoma and other cancers while other data from this project will inform the design of future studies.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
2R01CA056821-22A1
Application #
9103715
Study Section
Cancer Immunopathology and Immunotherapy Study Section (CII)
Program Officer
Yovandich, Jason L
Project Start
1992-06-01
Project End
2021-03-31
Budget Start
2016-04-01
Budget End
2017-03-31
Support Year
22
Fiscal Year
2016
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:
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