top of page

Elucidating the role of NKR-P1B in NK cell-mediated mammary tumor immunosurveillance 

Posted: December 21, 2023

Written by: Bigitha Bennychen

Edited by: Elizabeth Balint

rahim lab pic 2.png

In their work published in Oncoimmunology, Raghd Al Olabi and Dr Mir Munir Rahim elucidate the role of the inhibitory NKR-P1B receptor interaction with C-type lectin-related protein-b (Clr-b) ligand in Natural Killer (NK) cell-mediated mammary tumour immunosurveillance in MMTV-PyVT mice. 

​

NK cells are well known for the role they play in anti-cancer immune responses. NK cell functions are modulated by a plethora of activating and inhibitory cell surface receptors interacting with ligands expressed on the surface of target cells. NKR-P1B is a type of inhibitory receptor that recognises the Clr-b inhibitory ligand on the surface of target cells. Downregulation of inhibitory ligands, like Clr-b, on tumour cells can trigger NK cell activation in a process called the 'missing-self' response.

​

Given the role of NKR-P1B:Clr-b interactions in cancer immune evasion, Al Olabi et al. studied the role of this interaction in mammary tumour immunosurveillance using MMTV-PyVT mice bred on NKR-P1B deficient (Nkrp1b-/-) background. MMTV-PyVT wild-type (WT) mice remained tumour-free for a significantly longer period than the NKR-P1B-deficient mice, which may be due impaired NK cell-mediated missing-self response against Clr-b negative target cells in NKR-P1B-deficient mice. They also observed an increased frequency of EOMES- CD49a+ tumour-infiltrating NK cells in NKR-P1B deficient mice compared to the WT mice. Ex vivo analysis of EOMES- CD49a+ tumour-infiltrating NK cells demonstrated increased granzyme B expression, indicating an activated phenotype, but showed reduced expression of Ki67 in larger mammary tumours, indicating reduced proliferative capacity. Furthermore, EOMES- CD49a+ tumour-infiltrating NK cells showed upregulation of immune exhaustion markers, such as LAG-3 and PD-1, while exhibiting significant loss of markers associated with NK cell function, maturation, and homeostasis. Thus, the EOMES- tumour-infiltrating NK cells were associated with a dysfunctional state within NKR-P1B deficient MMTV-PyVT mice compared to WT mice.

​

Ultimately, Al Olabi et al revealed NKR-P1B:Clr-b interactions regulate anti-cancer responses and maintaining functional homeostasis in tumour-infiltrating NK cells. Further research investigating the mechanisms by which NK cell function is regulated in cancer can help realize the full potential of NK cells in cancer immunotherapy.

bottom of page