Abstract

Melanoma is a carcinoma known to evade the host immune defenses via the downregulation of the immune response. One of the molecules involved in this mechanism is programmed cell death ligand 1 (PD-L1), which interacts with its receptor, programmed cell death protein 1 (PD-1), expressed on T cells, leading to a reduction in cytokine release and cytotoxic activity, as well as a halt in T-cell proliferation. The approved therapeutic monoclonal antibodies, such as pembrolizumab, target the PD-1/PD-L1 interaction and are revolutionizing cancer treatments. We developed an assay that provides a quantitative readout of PD-1/PD-L1 interactive states between cell membranes of human immune cells (peripheral blood mononuclear cells, PBMCs) and PD-L1-expressing samples. For this purpose, cell membrane microarrays (CMMAs) were developed from membranes isolated from a HT144 cell line and melanoma samples, and PD-L1 expression was quantified using immunofluorescence (IF). CMMAs were incubated with cell membranes of PBMCs expressing PD-1, and the interaction with PD-L1 was quantified by time-resolved Förster resonance energy transfer, in the presence and absence of pembrolizumab as a blocking drug. The developed assay was able to quantify the PD-1/PD-L1 interaction, and this engagement was disrupted in the presence of the blocking antibody. This demonstrates the potential of the method to analyze monoclonal antibody drugs, as well as the functional states of immune checkpoint regulators. Furthermore, our findings provide evidence to support the future implementation of this methodology for both drug discovery and immune system monitoring in cancer, transplantation, and inflammatory and autoimmune diseases.

Highlights

  • In order to study the programmed death receptor 1 (PD-1)/programmed cell death ligand 1 (PD-L1) interactions between PD-1 expressed in PBMCs and PD-L1 in HT114 cells and melanoma samples, cell membrane microarrays (CMMAs) were developed, and the PD-L1 expression was quantified by IF

  • PD-1/PD-L1 interactions were assessed by incubating CMMAs with PBMC membranes in the absence and presence of a saturating concentration of the blocking antibody, pembrolizumab, using Immune-Amplified Förster Resonance Energy Transfer (iFRET) and the developed protocol (Figure 1)

  • Human PD-L1 expression was assessed in the samples immobilized in the CMMAs, using an anti-hPD-L1 IgG labeled with a fluorescent secondary antibody, and the signal intensity was quantified

Read more

Summary

Introduction

Protein–protein interactions are important signaling events in pathologies such as autoimmune disease, inflammation, transplant rejection, and cancer. Non-smallcell lung carcinoma (NSCLC), and clear-cell renal cell carcinoma (ccRCC) are amongst a subset of carcinomas known to evade the host immune defenses via the manipulation of the immune system, leading to the downregulation of the immune response against the tumor. Key players in developing this adaptive immune resistance are programmed death receptor 1 (PD-1) and its complementary ligand, programmed death ligand 1 (PD-L1). PD-1 is a single-pass type I membrane protein with 288 residues, which is expressed on

Methods
Results
Discussion
Conclusion
Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call