2020
DOI: 10.1158/1078-0432.ccr-19-3275
|View full text |Cite
|
Sign up to set email alerts
|

A Novel GUCY2C-CD3 T-Cell Engaging Bispecific Construct (PF-07062119) for the Treatment of Gastrointestinal Cancers

Abstract: Purpose: Gastrointestinal cancers remain areas of high unmet need despite advances in targeted and immunotherapies. Here, we demonstrate potent, tumor-selective efficacy with PF-07062119, a T-cell engaging CD3 bispecific targeting tumors expressing Guanylyl Cyclase C (GUCY2C), which is expressed widely across colorectal cancer and other gastrointestinal malignancies. In addition, to address immune evasion mechanisms, we explore combinations with immune checkpoint blockade agents and with antiangiogenesis thera… Show more

Help me understand this report
View preprint versions

Search citation statements

Order By: Relevance

Paper Sections

Select...
4
1

Citation Types

0
29
0

Year Published

2020
2020
2024
2024

Publication Types

Select...
6
2

Relationship

2
6

Authors

Journals

citations
Cited by 34 publications
(34 citation statements)
references
References 41 publications
0
29
0
Order By: Relevance
“…TCB-induced T-cell activation is has been shown to upregulate PD-1 expression on T-cells and induce PD-L1 expression on tumor cells (IFNg driven) (8,9,(25)(26)(27)(28)(29). This may lead to adaptive immune resistance mechanisms related to the TCB mode of action, similar to what has been described for checkpoint inhibition (30,31).…”
Section: Introductionmentioning
confidence: 77%
See 1 more Smart Citation
“…TCB-induced T-cell activation is has been shown to upregulate PD-1 expression on T-cells and induce PD-L1 expression on tumor cells (IFNg driven) (8,9,(25)(26)(27)(28)(29). This may lead to adaptive immune resistance mechanisms related to the TCB mode of action, similar to what has been described for checkpoint inhibition (30,31).…”
Section: Introductionmentioning
confidence: 77%
“…A clear upregulation of both PD-1 (on CD4 and CD8 T-cells) as well as PD-L1 (on tumor cells and CD4 and CD8 T-cells) was detected in response to CEA-TCB treatment, indicative of the PD-1/PD-L1 axis being one of the adaptive resistance mechanisms related to TCB activity (8,9,(25)(26)(27)(28)(29). Combination of TCBs with anti-PD-L1 blocking antibody (in different tumor and mouse models and using different TCBs targeting both CEA (solid tumors) and CD20 (hematological malignancies) consistently translated into superior anti-tumor efficacy and stronger tumor growth inhibition when compared to either agent given as monotherapy.…”
Section: Discussionmentioning
confidence: 99%
“…After formation of the cytolytic synapse, the T-cells produce perforin and granzyme B, leading to apoptosis of the tumor cells. Activation of T-cells leads to transient release of cytokines, which engages other immune cells and broadens the immune response against the tumor tissue leading to conversion of a noninflamed (cold) to an inflamed (hot) tumor environment, infiltration and proliferation of T-cells, and serial killing of tumor cells (10)(11)(12)(13). After recent clinical successes of bispecific T-cell engagers with a short half-life (BiTE) for the treatment of hematologic malignancies (14,15), the next generation of T-cell engagers incorporating half-life extension for increased dosing convenience for patients for the treatment of solid tumors is emerging (13,(16)(17)(18).…”
Section: Introductionmentioning
confidence: 99%
“…[16][17][18] Expression of GUCY2C is confined to the luminal surfaces of healthy intestinal epithelium, and because tumors have disrupted tight junction architecture, GUCY2C may offer a promising target antigen for tumor-specific activation of systemically administered T cell redirection therapies. [19][20][21] GUCY2C cell-surface expression is reported to be higher on moderately to well-differentiated tumors compared to more poorly differentiated tumors. 21 We therefore sought to develop an anti-GUCY2C/anti-CD3ε T-BsAb that demonstrated potent in vitro cytotoxicity (EC 50 below 1 nM) and in vivo single-agent efficacy below 1 mg/kg in preclinical models, good tolerability in cynomolgus monkeys at the predicted human efficacious dose, a pharmacokinetic (PK) profile compatible with dosing every three weeks, and developability characteristics supporting clinical development.…”
Section: Introductionmentioning
confidence: 99%
“…[19][20][21] GUCY2C cell-surface expression is reported to be higher on moderately to well-differentiated tumors compared to more poorly differentiated tumors. 21 We therefore sought to develop an anti-GUCY2C/anti-CD3ε T-BsAb that demonstrated potent in vitro cytotoxicity (EC 50 below 1 nM) and in vivo single-agent efficacy below 1 mg/kg in preclinical models, good tolerability in cynomolgus monkeys at the predicted human efficacious dose, a pharmacokinetic (PK) profile compatible with dosing every three weeks, and developability characteristics supporting clinical development.…”
Section: Introductionmentioning
confidence: 99%