Engulfing tumors with synthetic extracellular matrices for cancer immunotherapy.

TitleEngulfing tumors with synthetic extracellular matrices for cancer immunotherapy.
Publication TypeJournal Article
Year of Publication2009
AuthorsHori, Y, Stern, PJ, Hynes, RO, Irvine, DJ
JournalBiomaterials
Volume30
Issue35
Pagination6757-67
Date Published2009 Dec
ISSN1878-5905
KeywordsAlginates, Animals, Cancer Vaccines, Dendritic Cells, Extracellular Matrix, Hydrogels, Immunotherapy, Interleukin-15, Melanoma, Experimental, Mice, Mice, Inbred C57BL, Models, Biological, Neoplasms, Time Factors, Transplantation, Homologous
Abstract

Local immunotherapies are under investigation for the treatment of unresectable tumors and sites of solid tumor resection to prevent local recurrence. Successful local therapy could also theoretically elicit systemic immune responses against cancer. Here we explored the delivery of therapeutic dendritic cells (DCs), cytokines, or other immunostimulatory factors to tumors via the use of 'self-gelling' hydrogels based on the polysaccharide alginate, injected peritumorally around established melanoma lesions. Peritumoral injection of alginate matrices loaded with DCs and/or an interleukin-15 superagonist (IL-15SA) around 14-day established ova-expressing B16F0 murine melanoma tumors promoted immune cell accumulation in the peritumoral matrix, and matrix infiltration correlated with tumor infiltration by leukocytes. Single injections of IL-15SA-carrying gels concentrated the cytokine in the tumor site approximately 40-fold compared to systemic injection and enabled a majority of treated animals to suppress tumor growth for a week or more. Further, we found that single injections of alginate matrices loaded with IL-15SA and the Toll-like receptor ligand CpG or two injections of gels carrying IL-15SA alone could elicit comparable anti-tumor activity without the need for exogenous DCs. Thus, injectable alginate gels offer an attractive platform for local tumor immunotherapy, and facilitate combinatorial treatments designed to promote immune responses locally at a tumor site while limiting systemic exposure to potent immunomodulatory factors.

DOI10.1016/j.biomaterials.2009.08.037
Alternate JournalBiomaterials
PubMed ID19766305
PubMed Central IDPMC2788234
Grant ListEB007280 / EB / NIBIB NIH HHS / United States
R01 EB007280-02 / EB / NIBIB NIH HHS / United States
R01 EB007280-03 / EB / NIBIB NIH HHS / United States
U54-CA112967 / CA / NCI NIH HHS / United States
U54-CA126515 / CA / NCI NIH HHS / United States
/ / Howard Hughes Medical Institute / United States