Back to Search Start Over

Human CD34+-derived complete plasmacytoid and conventional dendritic cell vaccine effectively induces antigen-specific CD8+ T cell and NK cell responses in vitro and in vivo.

Authors :
van Eck van der Sluijs, Jesper
van Ens, Diede
Brummelman, Jolanda
Heister, Daan
Sareen, Aastha
Truijen, Lisa
van Ingen Schenau, Dorette S.
Heemskerk, Mirjam H. M.
Griffioen, Marieke
Kester, Michel G. D.
Schaap, Nicolaas P. M.
Jansen, Joop H.
van der Waart, Anniek B.
Dolstra, Harry
Hobo, Willemijn
Source :
Cellular & Molecular Life Sciences; Oct2023, Vol. 80 Issue 10, p1-19, 19p
Publication Year :
2023

Abstract

Allogeneic stem cell transplantation (alloSCT) can be curative for hemato-oncology patients due to effective graft-versus-tumor immunity. However, relapse remains the major cause of treatment failure, emphasizing the need for adjuvant immunotherapies. In this regard, post-transplantation dendritic cell (DC) vaccination is a highly interesting strategy to boost graft-versus-tumor responses. Previously, we developed a clinically applicable protocol for simultaneous large-scale generation of end-stage blood DC subsets from donor-derived CD34<superscript>+</superscript> stem cells, including conventional type 1 and 2 DCs (cDC1s and cDC2s), and plasmacytoid DCs (pDCs). In addition, the total cultured end-product (DC-complete vaccine), also contains non-end-stage-DCs (i.e. non-DCs). In this study, we aimed to dissect the phenotypic identity of these non-DCs and their potential immune modulatory functions on the potency of cDCs and pDCs in stimulating tumor-reactive CD8<superscript>+</superscript> T and NK cell responses, in order to obtain rationale for clinical translation of our DC-complete vaccine. The non-DC compartment was heterogeneous and comprised of myeloid progenitors and (immature) granulocyte- and monocyte-like cells. Importantly, non-DCs potentiated toll-like receptor-induced DC maturation, as reflected by increased expression of co-stimulatory molecules and enhanced cDC-derived IL-12 and pDC-derived IFN-α production. Additionally, antigen-specific CD8<superscript>+</superscript> T cells effectively expanded upon DC-complete vaccination in vitro and in vivo. This effect was strongly augmented by non-DCs in an antigen-independent manner. Moreover, non-DCs did not impair in vitro DC-mediated NK cell activation, degranulation nor cytotoxicity. Notably, in vivo i.p. DC-complete vaccination activated i.v. injected NK cells. Together, these data demonstrate that the non-DC compartment potentiates DC-mediated activation and expansion of antigen-specific CD8<superscript>+</superscript> T cells and do not impair NK cell responses in vitro and in vivo. This underscores the rationale for further clinical translation of our CD34<superscript>+</superscript>-derived DC-complete vaccine in hemato-oncology patients post alloSCT. [ABSTRACT FROM AUTHOR]

Details

Language :
English
ISSN :
1420682X
Volume :
80
Issue :
10
Database :
Complementary Index
Journal :
Cellular & Molecular Life Sciences
Publication Type :
Academic Journal
Accession number :
172236284
Full Text :
https://doi.org/10.1007/s00018-023-04923-4