Immune checkpoint blockade enhances shared neoantigen-induced T-cell immunity directed against mutated calreticulin in myeloproliferative neoplasms

C Cimen Bozkus, V Roudko, JP Finnigan… - Cancer discovery, 2019 - AACR
C Cimen Bozkus, V Roudko, JP Finnigan, J Mascarenhas, R Hoffman, C Iancu-Rubin…
Cancer discovery, 2019AACR
Somatic frameshift mutations in the calreticulin (CALR) gene are key drivers of cellular
transformation in myeloproliferative neoplasms (MPN). All patients carrying these mutations
(CALR+ MPN) share an identical sequence in the C-terminus of the mutated CALR protein
(mut-CALR), with the potential for utility as a shared neoantigen. Here, we demonstrate that
although a subset of patients with CALR+ MPN develop specific T-cell responses against
the mut-CALR C-terminus, PD-1 or CTLA4 expression abrogates the full complement of …
Abstract
Somatic frameshift mutations in the calreticulin (CALR) gene are key drivers of cellular transformation in myeloproliferative neoplasms (MPN). All patients carrying these mutations (CALR+ MPN) share an identical sequence in the C-terminus of the mutated CALR protein (mut-CALR), with the potential for utility as a shared neoantigen. Here, we demonstrate that although a subset of patients with CALR+ MPN develop specific T-cell responses against the mut-CALR C-terminus, PD-1 or CTLA4 expression abrogates the full complement of responses. Significantly, blockade of PD-1 and CLTA4 ex vivo by mAbs and of PD-1 in vivo by pembrolizumab administration restores mut-CALR–specific T-cell immunity in some patients with CALR+ MPN. Moreover, mut-CALR elicits antigen-specific responses from both CD4+ and CD8+ T cells, confirming its broad applicability as an immunogen. Collectively, these results establish mut-CALR as a shared, MPN-specific neoantigen and inform the design of novel immunotherapies targeting mut-CALR.
Significance
Current treatment modalities for MPN are not effective in eliminating malignant cells. Here, we show that mutations in the CALR gene, which drive transformation in MPN, elicit T-cell responses that can be further enhanced by checkpoint blockade, suggesting immunotherapies could be employed to eliminate CALR+ malignant cells in MPN.
This article is highlighted in the In This Issue feature, p. 1143
AACR