The research interests of my laboratory are two-fold; they involve the identification and characterization of (1) mechanisms of T cell-mediated antitumor activity in vivo and (2) tumor escape mechanisms and their impact on metastatic development. We have established two mouse models---one is in a transplant setting involving experimental lung metastases, and the other is in a transgenic setting reflecting spontaneous development of primary and metastatic mammary carcinoma.The mechanisms of CD8+ cytotoxic T lymphocyte (CTL)-mediated tumor regression in vivo remain to be fully understood. If CTL do mediate tumor regression in vivo by direct cytotoxicity, this may occur via two major effector mechanisms involving the secretion of perforin/granzymes and/or engagement of Fas by FasL expressed by the activated CTL. Although the perforin pathway has been considered the dominant player, it is unclear whether Fas-mediated cytotoxicity is additionally required for optimal tumor rejection in vivo. Accordingly, we examined whether a Fas/FasL-dependent CTL effector mechanism was important for optimal tumor regression in vivo. To that end, we developed a syngeneic mouse model of CTL adoptive immunotherapy against experimental lung metastases in both minimal (i.e., day 3 tumor-bearing mice) and extensive (day 10 tumor-bearing mice) disease settings. The model consisted of the CMS4 sarcoma, a solid tumor of BALB/c (H-2d) origin, and CMS4-reactive CTL, previously produced in immune competent mice using an anti-CTLA4 mAb-based strategy. CTL adoptive transfer resulted in nearly the complete regression of 3-day established experimental lung metastases. More importantly, under conditions of extensive lung metastases, we showed for the first time that (a) CTL adoptive transfer can promote significant tumor regression, and (b) the mechanism(s) of tumor regression required execution of a FasL-dependent CTL effector pathway. We propose that under such physiologic conditions, multiple immune effector mechanisms, in addition to the perforin/granzyme pathway, are necessary to achieve maximal antitumor reactivity. From a positive standpoint, combination strategies that involve the use of pro-inflammatory cytokines, certain chemotherapeutic agents or radiation, for example, to further sensitize neoplastic cells to T cell attack via regulation of Fas expression and function, may have implications for improving vaccine and/or passive-based antitumor regimens. In fact, in collaborative studies, we have demonstrated in pre-clinical models that local irradiation of a subcutaneously established tumor combined with Ag-based active or passive immunotherapies enhanced the tumor rejection response significantly more so than either modality given alone. Moreover, the immune mechanism(s) of tumor rejection induced by radiation exposure occurred via Fas-based interactions.

Agency
National Institute of Health (NIH)
Institute
Division of Basic Sciences - NCI (NCI)
Type
Intramural Research (Z01)
Project #
1Z01BC009029-13
Application #
7292124
Study Section
(LTIB)
Project Start
Project End
Budget Start
Budget End
Support Year
13
Fiscal Year
2005
Total Cost
Indirect Cost
Name
Basic Sciences
Department
Type
DUNS #
City
State
Country
United States
Zip Code
Stewart, Trina J; Liewehr, David J; Steinberg, Seth M et al. (2009) Modulating the expression of IFN regulatory factor 8 alters the protumorigenic behavior of CD11b+Gr-1+ myeloid cells. J Immunol 183:117-28
Liu, Kebin; Caldwell, Sheila A; Greeneltch, Kristy M et al. (2006) CTL adoptive immunotherapy concurrently mediates tumor regression and tumor escape. J Immunol 176:3374-82
Lundqvist, Andreas; Abrams, Scott I; Schrump, David S et al. (2006) Bortezomib and depsipeptide sensitize tumors to tumor necrosis factor-related apoptosis-inducing ligand: a novel method to potentiate natural killer cell tumor cytotoxicity. Cancer Res 66:7317-25
Gelbard, Alexander; Garnett, Charlie T; Abrams, Scott I et al. (2006) Combination chemotherapy and radiation of human squamous cell carcinoma of the head and neck augments CTL-mediated lysis. Clin Cancer Res 12:1897-905
Liu, Kebin; Caldwell, Sheila A; Abrams, Scott I (2005) Cooperative disengagement of Fas and intercellular adhesion molecule-1 function in neoplastic cells confers enhanced colonization efficiency. Cancer Res 65:1045-54
Abrams, Scott I (2005) Positive and negative consequences of Fas/Fas ligand interactions in the antitumor response. Front Biosci 10:809-21
Chakraborty, Mala; Abrams, Scott I; Coleman, C Norman et al. (2004) External beam radiation of tumors alters phenotype of tumor cells to render them susceptible to vaccine-mediated T-cell killing. Cancer Res 64:4328-37
Abrams, Scott I (2004) Regulation of tumor progression by anti-neoplastic T cell responses. Cancer Biol Ther 3:140-6
Chakraborty, Mala; Abrams, Scott I; Camphausen, Kevin et al. (2003) Irradiation of tumor cells up-regulates Fas and enhances CTL lytic activity and CTL adoptive immunotherapy. J Immunol 170:6338-47
Caldwell, Sheila A; Ryan, Mary H; McDuffie, Elwood et al. (2003) The Fas/Fas ligand pathway is important for optimal tumor regression in a mouse model of CTL adoptive immunotherapy of experimental CMS4 lung metastases. J Immunol 171:2402-12

Showing the most recent 10 out of 16 publications