2015
DOI: 10.1080/19420862.2015.1007811
|View full text |Cite
|
Sign up to set email alerts
|

C-type lectin-like molecule-1 (CLL1)-targeted TRAIL augments the tumoricidal activity of granulocytes and potentiates therapeutic antibody-dependent cell-mediated cytotoxicity

Abstract: (2015) C-type lectin-like molecule-1 (CLL1)-targeted TRAIL augments the tumoricidal activity of granulocytes and potentiates therapeutic antibody-dependent cell-mediated cytotoxicity, mAbs, 7:2, 321-330,

Help me understand this report

Search citation statements

Order By: Relevance

Paper Sections

Select...
2
2
1

Citation Types

0
25
0

Year Published

2015
2015
2020
2020

Publication Types

Select...
5
2

Relationship

0
7

Authors

Journals

citations
Cited by 25 publications
(25 citation statements)
references
References 27 publications
(28 reference statements)
0
25
0
Order By: Relevance
“…To examine whether CLL1:TRAIL synergizes the anticancer effect of monoclonal antibodies, they co-treated FaDu (EGFR-expressing cells) and Ramos B cells (CD20-expressing cells) with CLL1:TRAIL-armed granulocytes and a relevant monoclonal antibody (cetuximab and rituximab, respectively). In both cases, apoptosis was increased significantly, indicating a potential role of antibody-dependent cell-mediated cytotoxicity in synergizing the anticancer activity of CLL1:TRAIL (47).…”
Section: Dual-targeting Carrier For Delivery Of Doctexal To Ovarian Cmentioning
confidence: 91%
See 1 more Smart Citation
“…To examine whether CLL1:TRAIL synergizes the anticancer effect of monoclonal antibodies, they co-treated FaDu (EGFR-expressing cells) and Ramos B cells (CD20-expressing cells) with CLL1:TRAIL-armed granulocytes and a relevant monoclonal antibody (cetuximab and rituximab, respectively). In both cases, apoptosis was increased significantly, indicating a potential role of antibody-dependent cell-mediated cytotoxicity in synergizing the anticancer activity of CLL1:TRAIL (47).…”
Section: Dual-targeting Carrier For Delivery Of Doctexal To Ovarian Cmentioning
confidence: 91%
“…Phagocytes express a surface antigen, called C-type lectin-like molecule-1 (CLL1), which can be exploited for targeted drug delivery to these cells. Wiersma and colleagues produced a fusion peptide composed of an anti-CLL1-scFv and sTRAIL to specifically deliver TRAIL to granulocytes (47). They found that the fusion peptide, called CLL1:TRAIL, was able to specifically bind to and increase the antitumor activity of granulocytes.…”
Section: Dual-targeting Carrier For Delivery Of Doctexal To Ovarian Cmentioning
confidence: 99%
“…Tumor necrosis factor related apoptosis inducing ligand (TRAIL) is a member of the tumor necrosis factor superfamily and plays a specific role in the induction of tumor cell apoptosis, but without any significant effects on normal tissues [ 3 5 ]. Currently there are few studies regarding the genetic regulation of TRAIL and IDD.…”
Section: Introductionmentioning
confidence: 99%
“…4,5 Previously, we demonstrated that direct arming of myeloid effector cells using a TRAIL fusion protein that binds to CLL-1 on granulocytes, enhanced the pro-apoptotic activity of such myeloid cells. 26 To assess whether potentiation of myeloid effector cell activity might also contribute to anti-PD-L1:TRAIL activity, we generated various myeloid effector cell populations, i.e., monocytes, M0, M1, and M2 macrophages, iDCs and mDCs. All of these effector cells expressed PD-L1 to a varying degree, with monocytes having lowest and mDCs having the highest expression (Fig.…”
Section: Anti-pd-l1:trail Converts Pd-l1-expressing Myeloid Cells Intmentioning
confidence: 99%
“…24 Furthermore, scFv-mediated display of TRAIL on the surface of T cells or granulocytes augments the cytolytic activity of these immune effector cells. 25,26 Based on this bi-functional TRAIL-based fusion protein format, we constructed and pre-clinically evaluated an anti-PD-L1:TRAIL fusion protein comprised of a PD-L1-blocking antibody fragment genetically fused to human soluble TRAIL. This anti-PD-L1:TRAIL fusion protein was designed to combine PD-L1 checkpoint inhibition with simultaneous TRAIL-mediated activation of cancer cell death.…”
Section: Introductionmentioning
confidence: 99%