We reported previously that murine tumor lysate-pulsed dendritic cells (TP-DC) could elicit tumor-specific CD4+ and CD8+ T cell reactivities in vitro and in vivo. TP-DC treatments could result in regression of well- established s.c. and lung metastases, which could be further enhanced by the systemic administration of low-dose IL-2. Although vaccine studies involving TP-DC have been performed, little, if any, information is available on the effects of phagocytic uptake of tumor lysate on DC biology and function. We have investigated gene expression pattern differences between unpulsed DC and TP-DC, using Affymetrix MG- U74Av2 oligonucleotide arrays, which contain ~12,000 genes and ESTs (expressed sequence tags). Upon 24 hr tumor lysate pulsing, the levels of 87 transcripts increased at least threefold while the levels of 121 transcripts were reduced by one-third or more, with accompanying p-values <0.01. Most of these genes encoded a repertoire of proteins important for DC effector functions including cytokines, chemokines and receptors, as well as antigen presentation, cell adhesion, and T cell activation molecules. Interestingly, we observed a high level of expression of a novel member of the class A scavenger receptor family, MARCO on both mouse and human DC. MARCO is thought to play an important role in the immune response by mediating binding and phagocytosis, but also in the formation of lamellipodia-like structures and of dendritic processes. We propose to to define the biology and potential therapeutic implication of TP-DC expressed MARCO. We hypothesize that modulation of MARCO expression will have substantial effects on TP-DC biology and function in vitro and in vivo. We propose the following Specific Aims: 1. To determine the effect of MARCO expression modulation on TP-DC function in vitro;2. To determine the effect of MARCO expression modulation on TP-DC trafficking in vivo;3. To determine the therapeutic efficacy of MARCO expression-modulated TP-DC on antitumor activity in mice. The experimental studies outlined in this renewal application are designed to continue our successful preclinical efforts to generate immunization strategies against cancer based on antigen-presenting DC. The findings could have significant translation to human clinical DC vaccine trials.

Agency
National Institute of Health (NIH)
Institute
National Cancer Institute (NCI)
Type
Research Project (R01)
Project #
5R01CA071669-12
Application #
7741683
Study Section
Cancer Immunopathology and Immunotherapy Study Section (CII)
Program Officer
Yovandich, Jason L
Project Start
1996-08-01
Project End
2011-11-30
Budget Start
2009-12-01
Budget End
2011-11-30
Support Year
12
Fiscal Year
2010
Total Cost
$375,750
Indirect Cost
Name
H. Lee Moffitt Cancer Center & Research Institute
Department
Type
DUNS #
139301956
City
Tampa
State
FL
Country
United States
Zip Code
33612
Komine, Hiroshi; Kuhn, Lisa; Matsushita, Norimasa et al. (2013) Examination of MARCO activity on dendritic cell phenotype and function using a gene knockout mouse. PLoS One 8:e67795
Matsushita, Norimasa; Komine, Hiroshi; Grolleau-Julius, Annabelle et al. (2010) Targeting MARCO can lead to enhanced dendritic cell motility and anti-melanoma activity. Cancer Immunol Immunother 59:875-84
Koike, Nobusada; Pilon-Thomas, Shari; Mule, James J (2008) Nonmyeloablative chemotherapy followed by T-cell adoptive transfer and dendritic cell-based vaccination results in rejection of established melanoma. J Immunother 31:402-12
Pilon-Thomas, Shari; Verhaegen, Monique; Kuhn, Lisa et al. (2006) Induction of anti-tumor immunity by vaccination with dendritic cells pulsed with anti-CD44 IgG opsonized tumor cells. Cancer Immunol Immunother 55:1238-46
Pilon-Thomas, Shari; Li, Wenbin; Briggs, Jon J et al. (2006) Immunostimulatory effects of CpG-ODN upon dendritic cell-based immunotherapy in a murine melanoma model. J Immunother 29:381-7
Asavaroengchai, W; Kotera, Y; Koike, N et al. (2004) Augmentation of antitumor immune responses after adoptive transfer of bone marrow derived from donors immunized with tumor lysate-pulsed dendritic cells. Biol Blood Marrow Transplant 10:524-33
Terando, Alicia; Roessler, Blake; Mule, James J (2004) Chemokine gene modification of human dendritic cell-based tumor vaccines using a recombinant adenoviral vector. Cancer Gene Ther 11:165-73
Grolleau, Annabelle; Misek, David E; Kuick, Rork et al. (2003) Inducible expression of macrophage receptor Marco by dendritic cells following phagocytic uptake of dead cells uncovered by oligonucleotide arrays. J Immunol 171:2879-88
Asavaroengchai, W; Kotera, Y; Mule, J J (2002) Tumor lysate-pulsed dendritic cells can elicit an effective antitumor immune response during early lymphoid recovery. Proc Natl Acad Sci U S A 99:931-6
Candido, K A; Shimizu, K; McLaughlin, J C et al. (2001) Local administration of dendritic cells inhibits established breast tumor growth: implications for apoptosis-inducing agents. Cancer Res 61:228-36

Showing the most recent 10 out of 19 publications