Skip to main content
Top
Published in: Cancer Immunology, Immunotherapy 3/2019

Open Access 01-03-2019 | Original Article

A secreted PD-L1 splice variant that covalently dimerizes and mediates immunosuppression

Authors: Kathleen M. Mahoney, Sachet A. Shukla, Nikolaos Patsoukis, Apoorvi Chaudhri, Edward P. Browne, Arnon Arazi, Thomas M. Eisenhaure, William F. Pendergraft III, Ping Hua, Hung C. Pham, Xia Bu, Baogong Zhu, Nir Hacohen, Edward F. Fritsch, Vassiliki A. Boussiotis, Catherine J. Wu, Gordon J. Freeman

Published in: Cancer Immunology, Immunotherapy | Issue 3/2019

Login to get access

Abstract

Targeting immune checkpoint pathways, such as programmed death ligand-1 (PD-L1, also known as CD274 or B7-H1) or its receptor programmed cell death-1 (PD-1) has shown improved survival for patients with numerous types of cancers, not limited to lung cancer, melanoma, renal cell carcinoma, and Hodgkin lymphoma. PD-L1 is a co-inhibitory molecule whose expression on the surface of tumor cells is associated with worse prognosis in many tumors. Here we describe a splice variant (secPD-L1) that does not splice into the transmembrane domain, but instead produces a secreted form of PD-L1 that has a unique 18 amino acid tail containing a cysteine that allows it to homodimerize and more effectively inhibit lymphocyte function than monomeric soluble PD-L1. We show that recombinant secPD-L1 can dimerize and inhibit T-cell proliferation and IFN-gamma production in vitro. The secPD-L1 variant is expressed by malignant cells in vitro that also express high levels of full-length PD-L1. Transcriptomic analysis of gene expression across The Cancer Genome Atlas found the strongest association of secPD-L1 with full-length PD-L1, but also with subsets of immunologic genes, such as in myeloid-derived suppressor cells. Moreover, the splice variant is also expressed in normal tissues and within normal peripheral blood cells it is preferentially expressed in activated myeloid cells. This is the first report of a form of secreted PD-L1 that homodimerizes and is functionally active. SecPD-L1 may function as a paracrine negative immune regulator within the tumor, since secPD-L1 does not require a cell-to-cell interaction to mediate its inhibitory effect.
Appendix
Available only for authorised users
Literature
1.
go back to reference Mahoney KM et al (2016) A secreted PD-L1 splice variant expressed across tumor types inhibits lymphocyte function. J Immunother Cancer 4(Suppl 1):82 (Poster) Mahoney KM et al (2016) A secreted PD-L1 splice variant expressed across tumor types inhibits lymphocyte function. J Immunother Cancer 4(Suppl 1):82 (Poster)
2.
go back to reference Mahoney KM et al (2018) A secreted PD-L1 splice variant that covalently dimerizes and mediates immunosuppression. Kidney Cancer Symposium, November 3, 2018, Miami, Florida (oral presentation) Mahoney KM et al (2018) A secreted PD-L1 splice variant that covalently dimerizes and mediates immunosuppression. Kidney Cancer Symposium, November 3, 2018, Miami, Florida (oral presentation)
3.
go back to reference Freeman GJ et al (2000) Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med 192(7):1027–1034CrossRefPubMedPubMedCentral Freeman GJ et al (2000) Engagement of the PD-1 immunoinhibitory receptor by a novel B7 family member leads to negative regulation of lymphocyte activation. J Exp Med 192(7):1027–1034CrossRefPubMedPubMedCentral
4.
go back to reference Thompson RH et al (2006) Tumor B7-H1 is associated with poor prognosis in renal cell carcinoma patients with long-term follow-up. Cancer Res 66(7):3381–3385CrossRefPubMed Thompson RH et al (2006) Tumor B7-H1 is associated with poor prognosis in renal cell carcinoma patients with long-term follow-up. Cancer Res 66(7):3381–3385CrossRefPubMed
5.
go back to reference Finkelmeier F et al (2016) High levels of the soluble programmed death-ligand (sPD-L1) identify hepatocellular carcinoma patients with a poor prognosis. Eur J Cancer 59:152–159CrossRefPubMed Finkelmeier F et al (2016) High levels of the soluble programmed death-ligand (sPD-L1) identify hepatocellular carcinoma patients with a poor prognosis. Eur J Cancer 59:152–159CrossRefPubMed
6.
go back to reference Frigola X et al (2011) Identification of a soluble form of B7-H1 that retains immunosuppressive activity and is associated with aggressive renal cell carcinoma. Clin Cancer Res 17(7):1915–1923CrossRefPubMedPubMedCentral Frigola X et al (2011) Identification of a soluble form of B7-H1 that retains immunosuppressive activity and is associated with aggressive renal cell carcinoma. Clin Cancer Res 17(7):1915–1923CrossRefPubMedPubMedCentral
7.
go back to reference Wang L et al (2015) Serum levels of soluble programmed death ligand 1 predict treatment response and progression free survival in multiple myeloma. Oncotarget 6(38):41228–41236PubMedPubMedCentral Wang L et al (2015) Serum levels of soluble programmed death ligand 1 predict treatment response and progression free survival in multiple myeloma. Oncotarget 6(38):41228–41236PubMedPubMedCentral
8.
go back to reference Rossille D et al (2014) High level of soluble programmed cell death ligand 1 in blood impacts overall survival in aggressive diffuse large B-Cell lymphoma: results from a French multicenter clinical trial. Leukemia 28(12):2367–2375CrossRefPubMed Rossille D et al (2014) High level of soluble programmed cell death ligand 1 in blood impacts overall survival in aggressive diffuse large B-Cell lymphoma: results from a French multicenter clinical trial. Leukemia 28(12):2367–2375CrossRefPubMed
9.
go back to reference Okuma Y et al (2017) High plasma levels of soluble programmed cell death ligand 1 are prognostic for reduced survival in advanced lung cancer. Lung Cancer 104:1–6CrossRefPubMed Okuma Y et al (2017) High plasma levels of soluble programmed cell death ligand 1 are prognostic for reduced survival in advanced lung cancer. Lung Cancer 104:1–6CrossRefPubMed
11.
go back to reference Fang W et al (2014) EBV-driven LMP1 and IFN-gamma up-regulate PD-L1 in nasopharyngeal carcinoma: Implications for oncotargeted therapy. Oncotarget 5(23):12189–12202CrossRefPubMedPubMedCentral Fang W et al (2014) EBV-driven LMP1 and IFN-gamma up-regulate PD-L1 in nasopharyngeal carcinoma: Implications for oncotargeted therapy. Oncotarget 5(23):12189–12202CrossRefPubMedPubMedCentral
12.
go back to reference Green MR et al (2010) Integrative analysis reveals selective 9p24.1 amplification, increased PD-1 ligand expression, and further induction via JAK2 in nodular sclerosing Hodgkin lymphoma and primary mediastinal large B-cell lymphoma. Blood 116(17):3268–3277CrossRefPubMedPubMedCentral Green MR et al (2010) Integrative analysis reveals selective 9p24.1 amplification, increased PD-1 ligand expression, and further induction via JAK2 in nodular sclerosing Hodgkin lymphoma and primary mediastinal large B-cell lymphoma. Blood 116(17):3268–3277CrossRefPubMedPubMedCentral
13.
go back to reference Kataoka K et al (2016) Aberrant PD-L1 expression through 3′-UTR disruption in multiple cancers. Nature 534(7607):402–406CrossRefPubMed Kataoka K et al (2016) Aberrant PD-L1 expression through 3′-UTR disruption in multiple cancers. Nature 534(7607):402–406CrossRefPubMed
14.
go back to reference Taube JM et al (2012) Colocalization of inflammatory response with B7-h1 expression in human melanocytic lesions supports an adaptive resistance mechanism of immune escape. Sci Transl Med 4(127):127ra37CrossRefPubMedPubMedCentral Taube JM et al (2012) Colocalization of inflammatory response with B7-h1 expression in human melanocytic lesions supports an adaptive resistance mechanism of immune escape. Sci Transl Med 4(127):127ra37CrossRefPubMedPubMedCentral
15.
go back to reference Frigola X et al (2012) Soluble B7-H1: differences in production between dendritic cells and T cells. Immunol Lett 142(1–2):78–82CrossRefPubMed Frigola X et al (2012) Soluble B7-H1: differences in production between dendritic cells and T cells. Immunol Lett 142(1–2):78–82CrossRefPubMed
16.
go back to reference Chen Y et al (2011) Development of a sandwich ELISA for evaluating soluble PD-L1 (CD274) in human sera of different ages as well as supernatants of PD-L1 + cell lines. Cytokine 56(2):231–238CrossRefPubMed Chen Y et al (2011) Development of a sandwich ELISA for evaluating soluble PD-L1 (CD274) in human sera of different ages as well as supernatants of PD-L1 + cell lines. Cytokine 56(2):231–238CrossRefPubMed
19.
go back to reference Zerdes I et al (2018) Genetic, transcriptional and post-translational regulation of the programmed death protein ligand 1 in cancer: biology and clinical correlations. Oncogene 37(34):4639–4661CrossRefPubMedPubMedCentral Zerdes I et al (2018) Genetic, transcriptional and post-translational regulation of the programmed death protein ligand 1 in cancer: biology and clinical correlations. Oncogene 37(34):4639–4661CrossRefPubMedPubMedCentral
21.
go back to reference Zhumabayeva B, Chenchik A, Siebert PD (1999) RecA-mediated affinity capture: a method for full-length cDNA cloning. Biotechniques 27(4):834–836 (838, 840 passim) CrossRefPubMed Zhumabayeva B, Chenchik A, Siebert PD (1999) RecA-mediated affinity capture: a method for full-length cDNA cloning. Biotechniques 27(4):834–836 (838, 840 passim) CrossRefPubMed
22.
go back to reference Brown JA et al (2003) Blockade of programmed death-1 ligands on dendritic cells enhances T cell activation and cytokine production. J Immunol 170(3):1257–1266CrossRefPubMed Brown JA et al (2003) Blockade of programmed death-1 ligands on dendritic cells enhances T cell activation and cytokine production. J Immunol 170(3):1257–1266CrossRefPubMed
23.
go back to reference Mahoney KM et al (2015) PD-L1 antibodies to its cytoplasmic domain most clearly delineate cell membranes in immunohistochemical staining of tumor cells. Cancer Immunol Res 3:1308–1315CrossRefPubMedPubMedCentral Mahoney KM et al (2015) PD-L1 antibodies to its cytoplasmic domain most clearly delineate cell membranes in immunohistochemical staining of tumor cells. Cancer Immunol Res 3:1308–1315CrossRefPubMedPubMedCentral
24.
go back to reference Van Allen EM et al (2015), Genomic correlates of response to CTLA4 blockade in metastatic melanoma. Science 350:aad8366CrossRef Van Allen EM et al (2015), Genomic correlates of response to CTLA4 blockade in metastatic melanoma. Science 350:aad8366CrossRef
26.
go back to reference Zhu B et al (2007) In-fusion assembly: seamless engineering of multidomain fusion proteins, modular vectors, and mutations. Biotechniques 43(3):354–359CrossRefPubMed Zhu B et al (2007) In-fusion assembly: seamless engineering of multidomain fusion proteins, modular vectors, and mutations. Biotechniques 43(3):354–359CrossRefPubMed
27.
go back to reference Patsoukis N et al (2015) PD-1 alters T-cell metabolic reprogramming by inhibiting glycolysis and promoting lipolysis and fatty acid oxidation. Nat Commun 6:6692CrossRefPubMed Patsoukis N et al (2015) PD-1 alters T-cell metabolic reprogramming by inhibiting glycolysis and promoting lipolysis and fatty acid oxidation. Nat Commun 6:6692CrossRefPubMed
28.
go back to reference Latchman Y et al (2001) PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat Immunol 2(3):261–268CrossRefPubMed Latchman Y et al (2001) PD-L2 is a second ligand for PD-1 and inhibits T cell activation. Nat Immunol 2(3):261–268CrossRefPubMed
29.
go back to reference Juneja VR et al (2017) PD-L1 on tumor cells is sufficient for immune evasion in immunogenic tumors and inhibits CD8 T cell cytotoxicity. J Exp Med 214(4):895–904CrossRefPubMedPubMedCentral Juneja VR et al (2017) PD-L1 on tumor cells is sufficient for immune evasion in immunogenic tumors and inhibits CD8 T cell cytotoxicity. J Exp Med 214(4):895–904CrossRefPubMedPubMedCentral
30.
go back to reference Sharpe AH, Pauken KE (2018) The diverse functions of the PD1 inhibitory pathway. Nat Rev Immunol 18(3):153–167CrossRefPubMed Sharpe AH, Pauken KE (2018) The diverse functions of the PD1 inhibitory pathway. Nat Rev Immunol 18(3):153–167CrossRefPubMed
31.
go back to reference Reck M et al (2016) Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung cancer. N Engl J Med 375(19):1823–1833CrossRefPubMed Reck M et al (2016) Pembrolizumab versus chemotherapy for PD-L1-positive non-small-cell lung cancer. N Engl J Med 375(19):1823–1833CrossRefPubMed
33.
go back to reference Kruger S et al (2017) Serum levels of soluble programmed death protein 1 (sPD-1) and soluble programmed death ligand 1 (sPD-L1) in advanced pancreatic cancer. Oncoimmunology 6(5):e1310358CrossRefPubMedPubMedCentral Kruger S et al (2017) Serum levels of soluble programmed death protein 1 (sPD-1) and soluble programmed death ligand 1 (sPD-L1) in advanced pancreatic cancer. Oncoimmunology 6(5):e1310358CrossRefPubMedPubMedCentral
34.
go back to reference Enninga EAL et al (2018) Immune checkpoint molecules soluble program death ligand 1 and galectin-9 are increased in pregnancy. Am J Reprod Immunol 79(2):e12795CrossRef Enninga EAL et al (2018) Immune checkpoint molecules soluble program death ligand 1 and galectin-9 are increased in pregnancy. Am J Reprod Immunol 79(2):e12795CrossRef
35.
go back to reference Chen Y et al (2017) sPD-L1 expression is associated with immunosuppression and infectious complications in patients with acute pancreatitis. Scand J Immunol 86(2):100–106CrossRefPubMed Chen Y et al (2017) sPD-L1 expression is associated with immunosuppression and infectious complications in patients with acute pancreatitis. Scand J Immunol 86(2):100–106CrossRefPubMed
36.
go back to reference Chen BJ et al (2013) PD-L1 expression is characteristic of a subset of aggressive B-cell lymphomas and virus-associated malignancies. Clin Cancer Res 19(13):3462–3473CrossRefPubMedPubMedCentral Chen BJ et al (2013) PD-L1 expression is characteristic of a subset of aggressive B-cell lymphomas and virus-associated malignancies. Clin Cancer Res 19(13):3462–3473CrossRefPubMedPubMedCentral
Metadata
Title
A secreted PD-L1 splice variant that covalently dimerizes and mediates immunosuppression
Authors
Kathleen M. Mahoney
Sachet A. Shukla
Nikolaos Patsoukis
Apoorvi Chaudhri
Edward P. Browne
Arnon Arazi
Thomas M. Eisenhaure
William F. Pendergraft III
Ping Hua
Hung C. Pham
Xia Bu
Baogong Zhu
Nir Hacohen
Edward F. Fritsch
Vassiliki A. Boussiotis
Catherine J. Wu
Gordon J. Freeman
Publication date
01-03-2019
Publisher
Springer Berlin Heidelberg
Published in
Cancer Immunology, Immunotherapy / Issue 3/2019
Print ISSN: 0340-7004
Electronic ISSN: 1432-0851
DOI
https://doi.org/10.1007/s00262-018-2282-1

Other articles of this Issue 3/2019

Cancer Immunology, Immunotherapy 3/2019 Go to the issue
Webinar | 19-02-2024 | 17:30 (CET)

Keynote webinar | Spotlight on antibody–drug conjugates in cancer

Antibody–drug conjugates (ADCs) are novel agents that have shown promise across multiple tumor types. Explore the current landscape of ADCs in breast and lung cancer with our experts, and gain insights into the mechanism of action, key clinical trials data, existing challenges, and future directions.

Dr. Véronique Diéras
Prof. Fabrice Barlesi
Developed by: Springer Medicine