Cargando…

Agonist redirected checkpoint, PD1-Fc-OX40L, for cancer immunotherapy

Simultaneous blockade of immune checkpoint molecules and co-stimulation of the TNF receptor superfamily (TNFRSF) is predicted to improve overall survival in human cancer. TNFRSF co-stimulation depends upon coordinated antigen recognition through the T cell receptor followed by homotrimerization of t...

Descripción completa

Detalles Bibliográficos
Autores principales: Fromm, George, de Silva, Suresh, Johannes, Kellsey, Patel, Arpita, Hornblower, Josiah C., Schreiber, Taylor H.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: BioMed Central 2018
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6299665/
https://www.ncbi.nlm.nih.gov/pubmed/30563566
http://dx.doi.org/10.1186/s40425-018-0454-3
_version_ 1783381534903894016
author Fromm, George
de Silva, Suresh
Johannes, Kellsey
Patel, Arpita
Hornblower, Josiah C.
Schreiber, Taylor H.
author_facet Fromm, George
de Silva, Suresh
Johannes, Kellsey
Patel, Arpita
Hornblower, Josiah C.
Schreiber, Taylor H.
author_sort Fromm, George
collection PubMed
description Simultaneous blockade of immune checkpoint molecules and co-stimulation of the TNF receptor superfamily (TNFRSF) is predicted to improve overall survival in human cancer. TNFRSF co-stimulation depends upon coordinated antigen recognition through the T cell receptor followed by homotrimerization of the TNFRSF, and is most effective when these functions occur simultaneously. To address this mechanism, we developed a two-sided human fusion protein incorporating the extracellular domains (ECD) of PD-1 and OX40L, adjoined by a central Fc domain, termed PD1-Fc-OX40L. The PD-1 end of the fusion protein binds PD-L1 and PD-L2 with affinities of 2.08 and 1.76 nM, respectively, and the OX40L end binds OX40 with an affinity of 246 pM. High binding affinity on both sides of the construct translated to potent stimulation of OX40 signaling and PD1:PD-L1/L2 blockade, in multiple in vitro assays, including improved potency as compared to pembrolizumab, nivolumab, tavolixizumab and combinations of those antibodies. Furthermore, when activated human T cells were co-cultured with PD-L1 positive human tumor cells, PD1-Fc-OX40L was observed to concentrate to the immune synapse, which enhanced proliferation of T cells and production of IL-2, IFNγ and TNFα, and led to efficient killing of tumor cells. The therapeutic activity of PD1-Fc-OX40L in established murine tumors was significantly superior to either PD1 blocking, OX40 agonist, or combination antibody therapy; and required CD4+ T cells for maximum response. Importantly, all agonist functions of PD1-Fc-OX40L are independent of Fc receptor cross-linking. Collectively, these data demonstrate a highly potent fusion protein that is part of a platform, capable of providing checkpoint blockade and TNFRSF costimulation in a single molecule, which uniquely localizes TNFRSF costimulation to checkpoint ligand positive tumor cells. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (10.1186/s40425-018-0454-3) contains supplementary material, which is available to authorized users.
format Online
Article
Text
id pubmed-6299665
institution National Center for Biotechnology Information
language English
publishDate 2018
publisher BioMed Central
record_format MEDLINE/PubMed
spelling pubmed-62996652018-12-20 Agonist redirected checkpoint, PD1-Fc-OX40L, for cancer immunotherapy Fromm, George de Silva, Suresh Johannes, Kellsey Patel, Arpita Hornblower, Josiah C. Schreiber, Taylor H. J Immunother Cancer Research Article Simultaneous blockade of immune checkpoint molecules and co-stimulation of the TNF receptor superfamily (TNFRSF) is predicted to improve overall survival in human cancer. TNFRSF co-stimulation depends upon coordinated antigen recognition through the T cell receptor followed by homotrimerization of the TNFRSF, and is most effective when these functions occur simultaneously. To address this mechanism, we developed a two-sided human fusion protein incorporating the extracellular domains (ECD) of PD-1 and OX40L, adjoined by a central Fc domain, termed PD1-Fc-OX40L. The PD-1 end of the fusion protein binds PD-L1 and PD-L2 with affinities of 2.08 and 1.76 nM, respectively, and the OX40L end binds OX40 with an affinity of 246 pM. High binding affinity on both sides of the construct translated to potent stimulation of OX40 signaling and PD1:PD-L1/L2 blockade, in multiple in vitro assays, including improved potency as compared to pembrolizumab, nivolumab, tavolixizumab and combinations of those antibodies. Furthermore, when activated human T cells were co-cultured with PD-L1 positive human tumor cells, PD1-Fc-OX40L was observed to concentrate to the immune synapse, which enhanced proliferation of T cells and production of IL-2, IFNγ and TNFα, and led to efficient killing of tumor cells. The therapeutic activity of PD1-Fc-OX40L in established murine tumors was significantly superior to either PD1 blocking, OX40 agonist, or combination antibody therapy; and required CD4+ T cells for maximum response. Importantly, all agonist functions of PD1-Fc-OX40L are independent of Fc receptor cross-linking. Collectively, these data demonstrate a highly potent fusion protein that is part of a platform, capable of providing checkpoint blockade and TNFRSF costimulation in a single molecule, which uniquely localizes TNFRSF costimulation to checkpoint ligand positive tumor cells. ELECTRONIC SUPPLEMENTARY MATERIAL: The online version of this article (10.1186/s40425-018-0454-3) contains supplementary material, which is available to authorized users. BioMed Central 2018-12-18 /pmc/articles/PMC6299665/ /pubmed/30563566 http://dx.doi.org/10.1186/s40425-018-0454-3 Text en © The Author(s). 2018 Open AccessThis article is distributed under the terms of the Creative Commons Attribution 4.0 International License (http://creativecommons.org/licenses/by/4.0/), which permits unrestricted use, distribution, and reproduction in any medium, provided you give appropriate credit to the original author(s) and the source, provide a link to the Creative Commons license, and indicate if changes were made. The Creative Commons Public Domain Dedication waiver (http://creativecommons.org/publicdomain/zero/1.0/) applies to the data made available in this article, unless otherwise stated.
spellingShingle Research Article
Fromm, George
de Silva, Suresh
Johannes, Kellsey
Patel, Arpita
Hornblower, Josiah C.
Schreiber, Taylor H.
Agonist redirected checkpoint, PD1-Fc-OX40L, for cancer immunotherapy
title Agonist redirected checkpoint, PD1-Fc-OX40L, for cancer immunotherapy
title_full Agonist redirected checkpoint, PD1-Fc-OX40L, for cancer immunotherapy
title_fullStr Agonist redirected checkpoint, PD1-Fc-OX40L, for cancer immunotherapy
title_full_unstemmed Agonist redirected checkpoint, PD1-Fc-OX40L, for cancer immunotherapy
title_short Agonist redirected checkpoint, PD1-Fc-OX40L, for cancer immunotherapy
title_sort agonist redirected checkpoint, pd1-fc-ox40l, for cancer immunotherapy
topic Research Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC6299665/
https://www.ncbi.nlm.nih.gov/pubmed/30563566
http://dx.doi.org/10.1186/s40425-018-0454-3
work_keys_str_mv AT frommgeorge agonistredirectedcheckpointpd1fcox40lforcancerimmunotherapy
AT desilvasuresh agonistredirectedcheckpointpd1fcox40lforcancerimmunotherapy
AT johanneskellsey agonistredirectedcheckpointpd1fcox40lforcancerimmunotherapy
AT patelarpita agonistredirectedcheckpointpd1fcox40lforcancerimmunotherapy
AT hornblowerjosiahc agonistredirectedcheckpointpd1fcox40lforcancerimmunotherapy
AT schreibertaylorh agonistredirectedcheckpointpd1fcox40lforcancerimmunotherapy