Sort:
Open Access Original Article Issue
Blockade of CD300A enhances the ability of human NK cells to lyse hematologic malignancies
Cancer Biology & Medicine 2024, 21 (4): 331-346
Published: 29 February 2024
Abstract PDF (1.5 MB) Collect
Downloads:12
Objective

The human cluster of differentiation (CD)300A, a type-Ⅰ transmembrane protein with immunoreceptor tyrosine-based inhibitory motifs, was investigated as a potential immune checkpoint for human natural killer (NK) cells targeting hematologic malignancies (HMs).

Methods

We implemented a stimulation system involving the CD300A ligand, phosphatidylserine (PS), exposed to the outer surface of malignant cells. Additionally, we utilized CD300A overexpression, a CD300A blocking system, and a xenotransplantation model to evaluate the impact of CD300A on NK cell efficacy against HMs in in vitro and in vivo settings. Furthermore, we explored the association between CD300A and HM progression in patients.

Results

Our findings indicated that PS hampers the function of NK cells. Increased CD300A expression inhibited HM lysis by NK cells. CD300A overexpression shortened the survival of HM-xenografted mice by impairing transplanted NK cells. Blocking PS–CD300A signals with antibodies significantly amplified the expression of lysis function-related proteins and effector cytokines in NK cells, thereby augmenting the ability to lyse HMs. Clinically, heightened CD300A expression correlated with shorter survival and an “exhausted” phenotype of intratumoral NK cells in patients with HMs or solid tumors.

Conclusions

These results propose CD300A as a potential target for invigorating NK cell-based treatments against HMs.

Open Access Original Article Issue
Trispecific killer engager 161519 enhances natural killer cell function and provides anti-tumor activity against CD19-positive cancers
Cancer Biology & Medicine 2020, 17 (4): 1026-1038
Published: 15 November 2020
Abstract PDF (2 MB) Collect
Downloads:3
Objective

Natural killer (NK) cells have gained considerable attention due to their potential in treating “cold tumors,” and are therefore considered as one of the new strategies for curing cancer, by using worldwide development of their new possibilities and interventions with NK cell-related therapeutic products.

Methods

We constructed a trispecific killer engager (TriKE) consisting of anti-CD16, IL-15, and anti-CD19. This TriKE was designed to attract CD19+ tumor cells to CD16+ NK cells, whereas IL-15 sustained the proliferation, development, and survival of NK cells.

Results

Treatment with 161519 TriKE in the presence of CD19+ targets upregulated expression of CD69, CD107a, TRAIL, IFN-γ, and TNF-α in NK cells, and significantly improved the proliferation and cytotoxicity of NK cells. NK cells “armed” with 161519 TriKE showed stronger cytolysis against CD19+ targets compared with that of “unarmed” NK cells. A preclinical model of B-cell lymphoma in human peripheral blood mononuclear cell-reconstituted xenograft mice showed significant inhibition of tumor growth and prolonged overall survival after treatment with 161519 TriKE, when compared with that in control mice or mice treated with 1619 BiKE. Combined use of IL-2 was a more effective treatment with 1619 BiKE, when compared with that using 161519 TriKE.

Conclusions

The newly generated 161519 TriKE enhanced the proliferation, activation, cytokine secretion, and cytotoxicity of NK cells in the presence of CD19+ tumor cells. The 161519 TriKE aided inhibition of tumor growth and prolonged the overall survival of murine xenografts, and could be used to treat CD19-positive cancers.

Total 2