Return to search

Innate immune receptor signaling induces transient melanoma dedifferentiation while preserving immunogenicity

Background Immune-stimulatory
agents, like agonists
of the innate immune receptor RIG-I,
are currently tested
in clinical trials as an intratumoral treatment option for
patients with unresectable melanoma, aiming to enhance
anti-tumor
T cell responses. Switching of melanoma
toward a dedifferentiated cell state has recently been
linked to T cell and therapy resistance. It remains to be
determined whether RIG-I
agonists affect melanoma
differentiation, potentially leading to T cell resistance.
Methods Patient metastases-derived
melanoma cell
lines were treated with the synthetic RIG-I
agonist
3pRNA, and effects on tumor cell survival, phenotype
and differentiation were determined. Transcriptomic
data sets from cell lines and metastases were analyzed
for associations between RIG-I
(DDX58) and melanoma
differentiation markers and used to define signaling
pathways involved in RIG-I-
driven
dedifferentiation. The
impact of 3pRNA-induced
melanoma dedifferentiation on
CD8 T cell activation was studied in autologous tumor T
cell models.
Results RIG-I
activation by 3pRNA induced apoptosis in
a subpopulation of melanoma cells, while the majority
of tumor cells switched into a non-proliferative
cell
state. Those persisters displayed a dedifferentiated cell
phenotype, marked by downregulation of the melanocytic
lineage transcription factor MITF and its target genes,
including melanoma differentiation antigens (MDA).
Transition into the MITFlow/MDAlow cell state was JAK-dependent,
with some cells acquiring nerve growth factor
receptor expression. MITFlow/MDAlow persisters switched
back to the proliferative differentiated cell state when RIG-I
signaling declined. Consistent with our in vitro findings,
an association between melanoma dedifferentiation and
high RIG-I
(DDX58) levels was detected in transcriptomic
data from patient metastases. Notably, despite their
dedifferentiated cell phenotype, 3pRNA-induced
MITFlow/
MDAlow persisters were still efficiently targeted by
autologous CD8 tumor-infiltrating
T lymphocytes (TILs).
Conclusions Our results demonstrate that RIG-I
signaling
in melanoma cells drives a transient phenotypic switch
toward a non-proliferative
dedifferentiated persister cell
state. Despite their dedifferentiation, those persisters are
highly immunogenic and sensitive toward autologous TILs,
challenging the concept of melanoma dedifferentiation as
a general indicator of T cell resistance. In sum, our findings
support the application of RIG-I
agonists as a therapeutic
tool for the generation of long-term
clinical benefit in non-resectable
melanoma.

Identiferoai:union.ndltd.org:DRESDEN/oai:qucosa:de:qucosa:86621
Date27 July 2023
CreatorsThier, Beatrice, Zhao, Fang, Stupia, Simone, Brüggemann, Alicia, Koch, Johannes, Schulze, Nina, Horn, Susanne, Coch, Christoph, Hartmann, Gunter, Sucker, Antje, Schadendorf, Dirk, Paschen, Annette
PublisherBioMed Central
Source SetsHochschulschriftenserver (HSSS) der SLUB Dresden
LanguageEnglish
Detected LanguageEnglish
Typeinfo:eu-repo/semantics/publishedVersion, doc-type:article, info:eu-repo/semantics/article, doc-type:Text
Rightsinfo:eu-repo/semantics/openAccess
Relation2051-1426, e003863

Page generated in 0.0021 seconds