Tumor-associated CD4+ and CD8+ T cells with an effector memory phenotype (Tem) are present within the microenvironment of human non-small cell lung tumors and ovarian carcinomas, but fail to control tumor progression. Our data have established that the non-responsiveness of these cells to the tumor is due in part to their failure to respond to activation signals via the T cell receptor (TCR). By elucidating the site of the arrest in the TCR pathway and by defining the molecular and cellular events that initiate this arrest it should be possible to design and test strategies to re-activate the Tem in situ. The local re-activation of the Tem within the treated tumor microenvironment is expected to result in, (a) T cell mediated killing of tumor cells in situ, (b) release of tumor antigens into the circulation, (c) generation of a systemic anti-tumor immunity, and (d) T cell recognition and eradication of existing tumors at sites that are adjacent to or distant from the initially treated tumor site. Using a combination of multispectral imaging flow cytometry, confocal microscopy, western blot and rtPCR our first aim is to determine where in the TCR pathway the transduction signal is blocked. In a related second aim the cells and molecules that are causally linked to triggering the TCR arrest are determined by cell depletion and add back protocols and by monitoring the effects of selected molecules on the initiation of this regulatory signaling checkpoint. Our preliminary studies have localized the site of the signaling checkpoint to occur somewhere upstream of PLC-3 and the TCR signal arrest has been causally linked to TGF-21 thereby demonstrating the feasibility and viability of our experimental protocols. The results obtained from these mechanistic studies will be utilized in aims 2 and 3 to identify biologically active factors that act directly or indirectly on the T cells to reverse their non-responsiveness, and to develop and test liposome formulations that are designed to deliver these factors in a local and sustained fashion in vivo. In the final aim the therapeutic efficacy of each factor for re-activating Tem in situ and for inducing a local and systemic anti-tumor response is evaluated. The latter is to be accomplished using an established xenograft model in which non- disrupted pieces of human tumor are surgically implanted into SCID or NOD-SCID/IL2 receptor ? chain null mice. In these xenografts the tumor microenvironment is preserved and the tumor- associated leukocytes remain viable and predictably responsive to cytokine signals for prolonged periods. Following the inoculation of the factor loaded liposomes into the xenografts tumor killing and Tem response patterns are monitored to determine the therapeutic efficacy of each liposomal preparation. These studies are expected to lay the foundation for the design of strategies that can be used to enhance the efficacy of our current cancer vaccination clinical trials. Memory T cells that are present within and surrounding cancer patients'tumors have the potential to recognize and kill tumor cells, but they often fail to control the growth and spread of the cancer cells in these patients. This failure is due in part to a non-responsive or quiescent state of the memory cells. We are investigating the reason for this quiescent state, designing strategies to reverse this non-responsiveness, and testing the therapeutic benefits of re-activating the tumor-associated memory T cells.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA131407-02
Application #
7544973
Study Section
Transplantation, Tolerance, and Tumor Immunology (TTT)
Program Officer
Mccarthy, Susan A
Project Start
2007-12-20
Project End
2012-11-30
Budget Start
2008-12-01
Budget End
2009-11-30
Support Year
2
Fiscal Year
2009
Total Cost
$345,888
Indirect Cost
Name
State University of New York at Buffalo
Department
Microbiology/Immun/Virology
Type
Schools of Medicine
DUNS #
038633251
City
Buffalo
State
NY
Country
United States
Zip Code
14260
Shenoy, Gautam N; Loyall, Jenni; Berenson, Charles S et al. (2018) Sialic Acid-Dependent Inhibition of T Cells by Exosomal Ganglioside GD3 in Ovarian Tumor Microenvironments. J Immunol 201:3750-3758
Shenoy, Gautam N; Loyall, Jenni; Maguire, Orla et al. (2018) Exosomes Associated with Human Ovarian Tumors Harbor a Reversible Checkpoint of T-cell Responses. Cancer Immunol Res 6:236-247
Kelleher Jr, Raymond J; Balu-Iyer, Sathy; Loyall, Jenni et al. (2015) Extracellular Vesicles Present in Human Ovarian Tumor Microenvironments Induce a Phosphatidylserine-Dependent Arrest in the T-cell Signaling Cascade. Cancer Immunol Res 3:1269-78
Fathallah, Anas M; Bankert, Richard B; Balu-Iyer, Sathy V (2013) Immunogenicity of subcutaneously administered therapeutic proteins--a mechanistic perspective. AAPS J 15:897-900
Gaitonde, Puneet; Ramakrishnan, Radha; Chin, Jamie et al. (2013) Exposure to factor VIII protein in the presence of phosphatidylserine induces hypo-responsiveness toward factor VIII challenge in hemophilia A mice. J Biol Chem 288:17051-6
Simpson-Abelson, Michelle R; Loyall, Jenni L; Lehman, Heather K et al. (2013) Human ovarian tumor ascites fluids rapidly and reversibly inhibit T cell receptor-induced NF-?B and NFAT signaling in tumor-associated T cells. Cancer Immun 13:14
Yokota, Sandra J; Facciponte, John G; Kelleher Jr, Raymond J et al. (2013) Changes in ovarian tumor cell number, tumor vasculature, and T cell function monitored in vivo using a novel xenograft model. Cancer Immun 13:11
Bernstein, Joel M; Lehman, Heather; Lis, Maciej et al. (2012) Humanized mouse model used to monitor MUC gene expression in nasal polyps and to preclinically evaluate the efficacy of montelukast in reducing mucus production. Ann Otol Rhinol Laryngol 121:307-16
Lehman, Heather K; Simpson-Abelson, Michelle R; Conway Jr, Thomas F et al. (2012) Memory T cells in the chronic inflammatory microenvironment of nasal polyposis are hyporesponsive to signaling through the T cell receptor. J Assoc Res Otolaryngol 13:423-35
Bankert, Richard B; Balu-Iyer, Sathy V; Odunsi, Kunle et al. (2011) Humanized mouse model of ovarian cancer recapitulates patient solid tumor progression, ascites formation, and metastasis. PLoS One 6:e24420

Showing the most recent 10 out of 14 publications