Skip to main content

Lund University Publications

LUND UNIVERSITY LIBRARIES

Resistance to anti-PD-1/anti-PD-L1 : galectin-3 inhibition with GB1211 reverses galectin-3-induced blockade of pembrolizumab and atezolizumab binding to PD-1/PD-L1

Mabbitt, Joseph ; Holyer, Ian D ; Roper, James A ; Nilsson, Ulf J LU ; Zetterberg, Fredrik R ; Vuong, Lynda ; Mackinnon, Alison C ; Pedersen, Anders and Slack, Robert J (2023) In Frontiers in Immunology 14.
Abstract

BACKGROUND: Galectin-3 (Gal-3) is a β-galactoside-binding lectin that is highly expressed within the tumor microenvironment of aggressive cancers and has been suggested to predict a poor response to immune checkpoint therapy with the anti-PD-1 monoclonal antibody pembrolizumab. We aimed to assess if the effect of Gal-3 was a result of direct interaction with the immune checkpoint receptor.

METHODS: The ability of Gal-3 to interact with the PD-1/PD-L1 complex in the absence and presence of blocking antibodies was assessed in in vitro biochemical and cellular assays as well as in an in vivo syngeneic mouse cancer model.

RESULTS: Gal-3 reduced the binding of the checkpoint inhibitors pembrolizumab (anti-PD-1) and... (More)

BACKGROUND: Galectin-3 (Gal-3) is a β-galactoside-binding lectin that is highly expressed within the tumor microenvironment of aggressive cancers and has been suggested to predict a poor response to immune checkpoint therapy with the anti-PD-1 monoclonal antibody pembrolizumab. We aimed to assess if the effect of Gal-3 was a result of direct interaction with the immune checkpoint receptor.

METHODS: The ability of Gal-3 to interact with the PD-1/PD-L1 complex in the absence and presence of blocking antibodies was assessed in in vitro biochemical and cellular assays as well as in an in vivo syngeneic mouse cancer model.

RESULTS: Gal-3 reduced the binding of the checkpoint inhibitors pembrolizumab (anti-PD-1) and atezolizumab (anti-PD-L1), by potentiating the interaction between the PD-1/PD-L1 complex. In the presence of a highly selective Gal-3 small molecule inhibitor (GB1211) the binding of the anti-PD-1/anti-PD-L1 therapeutics was restored to control levels. This was observed in both a surface plasmon resonance assay measuring protein-protein interactions and via flow cytometry. Combination therapy with GB1211 and an anti-PD-L1 blocking antibody reduced tumor growth in an in vivo syngeneic model and increased the percentage of tumor infiltrating T lymphocytes.

CONCLUSION: Our study suggests that Gal-3 can potentiate the PD-1/PD-L1 immune axis and potentially contribute to the immunosuppressive signalling mechanisms within the tumor microenvironment. In addition, Gal-3 prevents atezolizumab and pembrolizumab target engagement with their respective immune checkpoint receptors. Reversal of this effect with the clinical candidate GB1211 offers a potential enhancing combination therapeutic with anti-PD-1 and -PD-L1 blocking antibodies.

(Less)
Please use this url to cite or link to this publication:
author
; ; ; ; ; ; ; and
organization
publishing date
type
Contribution to journal
publication status
published
subject
keywords
Animals, Mice, Galectin 3, Antibodies, Blocking, Antibodies, Monoclonal, Humanized/pharmacology
in
Frontiers in Immunology
volume
14
article number
1250559
publisher
Frontiers Media S. A.
external identifiers
  • scopus:85170563796
  • pmid:37701441
ISSN
1664-3224
DOI
10.3389/fimmu.2023.1250559
language
English
LU publication?
yes
id
d1815052-e4a7-4dc3-9800-7e4184b6b6db
date added to LUP
2023-09-28 08:45:04
date last changed
2024-04-19 01:45:44
@article{d1815052-e4a7-4dc3-9800-7e4184b6b6db,
  abstract     = {{<p>BACKGROUND: Galectin-3 (Gal-3) is a β-galactoside-binding lectin that is highly expressed within the tumor microenvironment of aggressive cancers and has been suggested to predict a poor response to immune checkpoint therapy with the anti-PD-1 monoclonal antibody pembrolizumab. We aimed to assess if the effect of Gal-3 was a result of direct interaction with the immune checkpoint receptor.</p><p>METHODS: The ability of Gal-3 to interact with the PD-1/PD-L1 complex in the absence and presence of blocking antibodies was assessed in  in vitro biochemical and cellular assays as well as in an  in vivo syngeneic mouse cancer model. </p><p>RESULTS: Gal-3 reduced the binding of the checkpoint inhibitors pembrolizumab (anti-PD-1) and atezolizumab (anti-PD-L1), by potentiating the interaction between the PD-1/PD-L1 complex. In the presence of a highly selective Gal-3 small molecule inhibitor (GB1211) the binding of the anti-PD-1/anti-PD-L1 therapeutics was restored to control levels. This was observed in both a surface plasmon resonance assay measuring protein-protein interactions and via flow cytometry. Combination therapy with GB1211 and an anti-PD-L1 blocking antibody reduced tumor growth in an in vivo syngeneic model and increased the percentage of tumor infiltrating T lymphocytes. </p><p>CONCLUSION: Our study suggests that Gal-3 can potentiate the PD-1/PD-L1 immune axis and potentially contribute to the immunosuppressive signalling mechanisms within the tumor microenvironment. In addition, Gal-3 prevents atezolizumab and pembrolizumab target engagement with their respective immune checkpoint receptors. Reversal of this effect with the clinical candidate GB1211 offers a potential enhancing combination therapeutic with anti-PD-1 and -PD-L1 blocking antibodies.</p>}},
  author       = {{Mabbitt, Joseph and Holyer, Ian D and Roper, James A and Nilsson, Ulf J and Zetterberg, Fredrik R and Vuong, Lynda and Mackinnon, Alison C and Pedersen, Anders and Slack, Robert J}},
  issn         = {{1664-3224}},
  keywords     = {{Animals; Mice; Galectin 3; Antibodies, Blocking; Antibodies, Monoclonal, Humanized/pharmacology}},
  language     = {{eng}},
  publisher    = {{Frontiers Media S. A.}},
  series       = {{Frontiers in Immunology}},
  title        = {{Resistance to anti-PD-1/anti-PD-L1 : galectin-3 inhibition with GB1211 reverses galectin-3-induced blockade of pembrolizumab and atezolizumab binding to PD-1/PD-L1}},
  url          = {{http://dx.doi.org/10.3389/fimmu.2023.1250559}},
  doi          = {{10.3389/fimmu.2023.1250559}},
  volume       = {{14}},
  year         = {{2023}},
}