4.7 Letter

Polymorphonuclear myeloid-derived suppressor cells impair the anti-tumor efficacy of GD2.CAR T-cells in patients with neuroblastoma

Journal

JOURNAL OF HEMATOLOGY & ONCOLOGY
Volume 14, Issue 1, Pages -

Publisher

BMC
DOI: 10.1186/s13045-021-01193-0

Keywords

Neuroblastoma; Polymorphonuclear myeloid-derived suppressor cells; GD2; CAR T-cells; Clinical response; T-cell functionality; Long-term response

Funding

  1. Associazione Italiana Ricerca sul Cancro (AIRC) [21724, 19920]
  2. Special Program Metastatic disease: the key unmet need in oncology 5 per mille [21147, 17184]
  3. Ministero della SaluteRicerca Corrente 2020
  4. Ministero della Salute [GR-2018-12365485, RF-2016-02364388, RCR-2019-23669115]
  5. Ministero dell'Istruzione, Universita e Ricerca-MIUR, Project PRIN 2017 [2017WC8499]
  6. Elterninitiative leukamie-und tumorkranker Kinder Wurzburg e.V.
  7. AIRC
  8. European Union's Horizon 2020 research and innovation program under the Marie Sklodowska-Curie Grant [800924]
  9. Aktion Regenbogen fur leukamie-und tumorkranke Kinder Main-Tauber e.V.
  10. AIFAAgenzia Italiana del Farmaco [2016-02364631]

Ask authors/readers for more resources

This study developed a third-generation GD2-specific CAR T-cell construct that showed marked tumor reduction and complete remission, but relapses were common. It identified an immune mechanism of resistance involving PMN-MDSC in NB patients after treatment with GD2.CAR T-cells, suggesting the need to further optimize CAR T-cells and clinical trials to target elements of the tumor microenvironment.
The outcome of patients affected by high-risk or metastatic neuroblastoma (NB) remains grim, with >= 50% of the children experiencing relapse or progression of the disease despite multimodal, intensive treatment. In order to identify new strategies to improve the overall survival and the quality of life of these children, we recently developed and optimized a third-generation GD2-specific chimeric antigen receptor (CAR) construct, which is currently under evaluation in our Institution in a phase I/II clinical trial (NCT03373097) enrolling patients with relapsed/refractory NB. We observed that our CAR T-cells are able to induce marked tumor reduction and even achieve complete remission with a higher efficiency than that of other CAR T-cells reported in previous studies. However, often responses are not sustained and relapses occur. Here, we demonstrate for the first time a mechanism of resistance to GD2.CAR T-cell treatment, showing how polymorphonuclear myeloid-derived suppressor cells (PMN-MDSC) increase in the peripheral blood (PB) of NB patients after GD2.CAR T-cell treatment in case of relapse and loss of response. In vitro, isolated PMN-MDSC demonstrate to inhibit the anti-tumor cytotoxicity of different generations of GD2.CAR T-cells. Gene-expression profiling of GD2.CAR T-cells conditioned with PMN-MDSC shows downregulation of genes involved in cell activation, signal transduction, inflammation and cytokine/chemokine secretion. Analysis of NB gene-expression dataset confirms a correlation between expression of these genes and patient outcome. Moreover, in patients treated with GD2.CAR T-cells, the frequency of circulating PMN-MDSC inversely correlates with the levels of GD2.CAR T-cells, resulting more elevated in patients who did not respond or lost response to the treatment. The presence and the frequency of PMN-MDSC in PB of high-risk and metastatic NB represents a useful prognostic marker to predict the response to GD2.CAR T-cells and other adoptive immunotherapy. This study underlines the importance of further optimization of both CAR T-cells and clinical trial in order to target elements of the tumor microenvironment.

Authors

I am an author on this paper
Click your name to claim this paper and add it to your profile.

Reviews

Primary Rating

4.7
Not enough ratings

Secondary Ratings

Novelty
-
Significance
-
Scientific rigor
-
Rate this paper

Recommended

No Data Available
No Data Available