Back to Search Start Over

Dendritic Cell Vaccination in Metastatic Melanoma Turns 'Non-T Cell Inflamed' Into 'T-Cell Inflamed' Tumors

Authors :
Jenny Bulgarelli
Marcella Tazzari
Anna Maria Granato
Laura Ridolfi
Serena Maiocchi
Francesco de Rosa
Massimiliano Petrini
Elena Pancisi
Giorgia Gentili
Barbara Vergani
Filippo Piccinini
Antonella Carbonaro
Biagio Eugenio Leone
Giovanni Foschi
Valentina Ancarani
Massimo Framarini
Massimo Guidoboni
Source :
Frontiers in Immunology, Vol 10 (2019)
Publication Year :
2019
Publisher :
Frontiers Media S.A., 2019.

Abstract

Dendritic cell (DC)-based vaccination effectively induces anti-tumor immunity, although in the majority of cases this does not translate into a durable clinical response. However, DC vaccination is characterized by a robust safety profile, making this treatment a potential candidate for effective combination cancer immunotherapy. To explore this possibility, understanding changes occurring in the tumor microenvironment (TME) upon DC vaccination is required. In this line, quantitative and qualitative changes in tumor-infiltrating T lymphocytes (TILs) induced by vaccination with autologous tumor lysate/homogenate loaded DCs were investigated in a series of 16 patients with metastatic melanoma. Immunohistochemistry for CD4, CD8, Foxp3, Granzyme B (GZMB), PDL1, and HLA class I was performed in tumor biopsies collected before and after DC vaccination. The density of each marker was quantified by automated digital pathology analysis on whole slide images. Co-expression of markers defining functional phenotypes, i.e., Foxp3+ regulatory CD4+ T cells (Treg) and GZMB+ cytotoxic CD8+ T cells, was assessed with sequential immunohistochemistry. A significant increase of CD8+ TILs was found in post-vaccine biopsies of patients who were not previously treated with immune-modulating cytokines or Ipilimumab. Interestingly, along with a maintained tumoral HLA class I expression, after DC vaccination we observed a significant increase of PDL1+ tumor cells, which significantly correlated with intratumoral CD8+ T cell density. This observation might explain the lack of a significant concurrent cytotoxic reactivation of CD8+ T cell, as measured by the numbers of GZMB+ T cells. Altogether these findings indicate that DC vaccination exerts an important role in sustaining or de novo inducing a T cell inflamed TME. However, the strength of the intratumoral T cell activation detected in post-DC therapy lesions is lessened by an occurring phenomenon of adaptive immune resistance, yet the concomitant PDL1 up-regulation. Overall, this study sheds light on DC immunotherapy-induced TME changes, lending the rationale for the design of smarter immune-combination therapies.

Details

Language :
English
ISSN :
16643224
Volume :
10
Database :
Directory of Open Access Journals
Journal :
Frontiers in Immunology
Publication Type :
Academic Journal
Accession number :
edsdoj.316112fefd0b4ff3be1250becac7f51b
Document Type :
article
Full Text :
https://doi.org/10.3389/fimmu.2019.02353