Cargando…

Nanobody-mediated SPECT/CT imaging reveals the spatiotemporal expression of programmed death-ligand 1 in response to a CD8(+) T cell and iNKT cell activating mRNA vaccine

Rationale: Although promising responses are obtained in patients treated with immune checkpoint inhibitors targeting programmed death ligand 1 (PD-L1) and its receptor programmed death-1 (PD-1), only a fraction of patients benefits from this immunotherapy. Cancer vaccination may be an effective appr...

Descripción completa

Detalles Bibliográficos
Autores principales: Ertveldt, Thomas, Meulewaeter, Sofie, De Vlaeminck, Yannick, Olarte, Oscar, Broos, Katrijn, Van Calenbergh, Serge, Bourgeois, Stephanie, Deprez, Joke, Heremans, Yves, Goyvaerts, Cleo, Staels, Willem, De Smedt, Stefaan, Dewitte, Heleen, Devoogdt, Nick, Keyaerts, Marleen, Verbeke, Rein, Barbé, Kurt, Lentacker, Ine, Breckpot, Karine
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Ivyspring International Publisher 2023
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10614673/
https://www.ncbi.nlm.nih.gov/pubmed/37908728
http://dx.doi.org/10.7150/thno.85106
_version_ 1785129071325216768
author Ertveldt, Thomas
Meulewaeter, Sofie
De Vlaeminck, Yannick
Olarte, Oscar
Broos, Katrijn
Van Calenbergh, Serge
Bourgeois, Stephanie
Deprez, Joke
Heremans, Yves
Goyvaerts, Cleo
Staels, Willem
De Smedt, Stefaan
Dewitte, Heleen
Devoogdt, Nick
Keyaerts, Marleen
Verbeke, Rein
Barbé, Kurt
Lentacker, Ine
Breckpot, Karine
author_facet Ertveldt, Thomas
Meulewaeter, Sofie
De Vlaeminck, Yannick
Olarte, Oscar
Broos, Katrijn
Van Calenbergh, Serge
Bourgeois, Stephanie
Deprez, Joke
Heremans, Yves
Goyvaerts, Cleo
Staels, Willem
De Smedt, Stefaan
Dewitte, Heleen
Devoogdt, Nick
Keyaerts, Marleen
Verbeke, Rein
Barbé, Kurt
Lentacker, Ine
Breckpot, Karine
author_sort Ertveldt, Thomas
collection PubMed
description Rationale: Although promising responses are obtained in patients treated with immune checkpoint inhibitors targeting programmed death ligand 1 (PD-L1) and its receptor programmed death-1 (PD-1), only a fraction of patients benefits from this immunotherapy. Cancer vaccination may be an effective approach to improve the response to immune checkpoint inhibitors anti-PD-L1/PD-1 therapy. However, there is a lack of research on the dynamics of PD-L1 expression in response to cancer vaccination. Methods: We performed non-invasive whole-body imaging to visualize PD-L1 expression at different timepoints after vaccination of melanoma-bearing mice. Mice bearing ovalbumin (OVA) expressing B16 tumors were i.v. injected with the Galsome mRNA vaccine: OVA encoding mRNA lipoplexes co-encapsulating a low or a high dose of the atypical adjuvant α-galactosylceramide (αGC) to activate invariant natural killer T (iNKT) cells. Serial non-invasive whole-body immune imaging was performed using a technetium-99m ((99m)Tc)-labeled anti-PD-L1 nanobody, single-photon emission computerized tomography (SPECT) and X-ray computed tomography (CT) images were quantified. Additionally, cellular expression of PD-L1 was evaluated with flow cytometry. Results: SPECT/CT-imaging showed a rapid and systemic upregulation of PD-L1 after vaccination. PD-L1 expression could not be correlated to the αGC-dose, although we observed a dose-dependent iNKT cell activation. Dynamics of PD-L1 expression were organ-dependent and most pronounced in lungs and liver, organs to which the vaccine was distributed. PD-L1 expression in lungs increased immediately after vaccination and gradually decreased over time, whereas in liver, vaccination-induced PD-L1 upregulation was short-lived. Flow cytometric analysis of these organs further showed myeloid cells as well as non-immune cells with elevated PD-L1 expression in response to vaccination. SPECT/CT imaging of the tumor demonstrated that the expression of PD-L1 remained stable over time and was overall not affected by vaccination although flow cytometric analysis at the cellular level demonstrated changes in PD-L1 expression in various immune cell populations following vaccination. Conclusion: Repeated non-invasive whole-body imaging using (99m)Tc-labeled anti-PD-L1 nanobodies allows to document the dynamic nature of PD-L1 expression upon vaccination. Galsome vaccination rapidly induced systemic upregulation of PD-L1 expression with the most pronounced upregulation in lungs and liver while flow cytometry analysis showed upregulation of PD-L1 in the tumor microenvironment. This study shows that imaging using nanobodies may be useful for monitoring vaccine-mediated PD-L1 modulation in patients and could provide a rationale for combination therapy. To the best of our knowledge, this is the first report that visualizes PD-L1 expression upon cancer vaccination.
format Online
Article
Text
id pubmed-10614673
institution National Center for Biotechnology Information
language English
publishDate 2023
publisher Ivyspring International Publisher
record_format MEDLINE/PubMed
spelling pubmed-106146732023-10-31 Nanobody-mediated SPECT/CT imaging reveals the spatiotemporal expression of programmed death-ligand 1 in response to a CD8(+) T cell and iNKT cell activating mRNA vaccine Ertveldt, Thomas Meulewaeter, Sofie De Vlaeminck, Yannick Olarte, Oscar Broos, Katrijn Van Calenbergh, Serge Bourgeois, Stephanie Deprez, Joke Heremans, Yves Goyvaerts, Cleo Staels, Willem De Smedt, Stefaan Dewitte, Heleen Devoogdt, Nick Keyaerts, Marleen Verbeke, Rein Barbé, Kurt Lentacker, Ine Breckpot, Karine Theranostics Research Paper Rationale: Although promising responses are obtained in patients treated with immune checkpoint inhibitors targeting programmed death ligand 1 (PD-L1) and its receptor programmed death-1 (PD-1), only a fraction of patients benefits from this immunotherapy. Cancer vaccination may be an effective approach to improve the response to immune checkpoint inhibitors anti-PD-L1/PD-1 therapy. However, there is a lack of research on the dynamics of PD-L1 expression in response to cancer vaccination. Methods: We performed non-invasive whole-body imaging to visualize PD-L1 expression at different timepoints after vaccination of melanoma-bearing mice. Mice bearing ovalbumin (OVA) expressing B16 tumors were i.v. injected with the Galsome mRNA vaccine: OVA encoding mRNA lipoplexes co-encapsulating a low or a high dose of the atypical adjuvant α-galactosylceramide (αGC) to activate invariant natural killer T (iNKT) cells. Serial non-invasive whole-body immune imaging was performed using a technetium-99m ((99m)Tc)-labeled anti-PD-L1 nanobody, single-photon emission computerized tomography (SPECT) and X-ray computed tomography (CT) images were quantified. Additionally, cellular expression of PD-L1 was evaluated with flow cytometry. Results: SPECT/CT-imaging showed a rapid and systemic upregulation of PD-L1 after vaccination. PD-L1 expression could not be correlated to the αGC-dose, although we observed a dose-dependent iNKT cell activation. Dynamics of PD-L1 expression were organ-dependent and most pronounced in lungs and liver, organs to which the vaccine was distributed. PD-L1 expression in lungs increased immediately after vaccination and gradually decreased over time, whereas in liver, vaccination-induced PD-L1 upregulation was short-lived. Flow cytometric analysis of these organs further showed myeloid cells as well as non-immune cells with elevated PD-L1 expression in response to vaccination. SPECT/CT imaging of the tumor demonstrated that the expression of PD-L1 remained stable over time and was overall not affected by vaccination although flow cytometric analysis at the cellular level demonstrated changes in PD-L1 expression in various immune cell populations following vaccination. Conclusion: Repeated non-invasive whole-body imaging using (99m)Tc-labeled anti-PD-L1 nanobodies allows to document the dynamic nature of PD-L1 expression upon vaccination. Galsome vaccination rapidly induced systemic upregulation of PD-L1 expression with the most pronounced upregulation in lungs and liver while flow cytometry analysis showed upregulation of PD-L1 in the tumor microenvironment. This study shows that imaging using nanobodies may be useful for monitoring vaccine-mediated PD-L1 modulation in patients and could provide a rationale for combination therapy. To the best of our knowledge, this is the first report that visualizes PD-L1 expression upon cancer vaccination. Ivyspring International Publisher 2023-10-09 /pmc/articles/PMC10614673/ /pubmed/37908728 http://dx.doi.org/10.7150/thno.85106 Text en © The author(s) https://creativecommons.org/licenses/by/4.0/This is an open access article distributed under the terms of the Creative Commons Attribution License (https://creativecommons.org/licenses/by/4.0/). See http://ivyspring.com/terms for full terms and conditions.
spellingShingle Research Paper
Ertveldt, Thomas
Meulewaeter, Sofie
De Vlaeminck, Yannick
Olarte, Oscar
Broos, Katrijn
Van Calenbergh, Serge
Bourgeois, Stephanie
Deprez, Joke
Heremans, Yves
Goyvaerts, Cleo
Staels, Willem
De Smedt, Stefaan
Dewitte, Heleen
Devoogdt, Nick
Keyaerts, Marleen
Verbeke, Rein
Barbé, Kurt
Lentacker, Ine
Breckpot, Karine
Nanobody-mediated SPECT/CT imaging reveals the spatiotemporal expression of programmed death-ligand 1 in response to a CD8(+) T cell and iNKT cell activating mRNA vaccine
title Nanobody-mediated SPECT/CT imaging reveals the spatiotemporal expression of programmed death-ligand 1 in response to a CD8(+) T cell and iNKT cell activating mRNA vaccine
title_full Nanobody-mediated SPECT/CT imaging reveals the spatiotemporal expression of programmed death-ligand 1 in response to a CD8(+) T cell and iNKT cell activating mRNA vaccine
title_fullStr Nanobody-mediated SPECT/CT imaging reveals the spatiotemporal expression of programmed death-ligand 1 in response to a CD8(+) T cell and iNKT cell activating mRNA vaccine
title_full_unstemmed Nanobody-mediated SPECT/CT imaging reveals the spatiotemporal expression of programmed death-ligand 1 in response to a CD8(+) T cell and iNKT cell activating mRNA vaccine
title_short Nanobody-mediated SPECT/CT imaging reveals the spatiotemporal expression of programmed death-ligand 1 in response to a CD8(+) T cell and iNKT cell activating mRNA vaccine
title_sort nanobody-mediated spect/ct imaging reveals the spatiotemporal expression of programmed death-ligand 1 in response to a cd8(+) t cell and inkt cell activating mrna vaccine
topic Research Paper
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC10614673/
https://www.ncbi.nlm.nih.gov/pubmed/37908728
http://dx.doi.org/10.7150/thno.85106
work_keys_str_mv AT ertveldtthomas nanobodymediatedspectctimagingrevealsthespatiotemporalexpressionofprogrammeddeathligand1inresponsetoacd8tcellandinktcellactivatingmrnavaccine
AT meulewaetersofie nanobodymediatedspectctimagingrevealsthespatiotemporalexpressionofprogrammeddeathligand1inresponsetoacd8tcellandinktcellactivatingmrnavaccine
AT devlaeminckyannick nanobodymediatedspectctimagingrevealsthespatiotemporalexpressionofprogrammeddeathligand1inresponsetoacd8tcellandinktcellactivatingmrnavaccine
AT olarteoscar nanobodymediatedspectctimagingrevealsthespatiotemporalexpressionofprogrammeddeathligand1inresponsetoacd8tcellandinktcellactivatingmrnavaccine
AT brooskatrijn nanobodymediatedspectctimagingrevealsthespatiotemporalexpressionofprogrammeddeathligand1inresponsetoacd8tcellandinktcellactivatingmrnavaccine
AT vancalenberghserge nanobodymediatedspectctimagingrevealsthespatiotemporalexpressionofprogrammeddeathligand1inresponsetoacd8tcellandinktcellactivatingmrnavaccine
AT bourgeoisstephanie nanobodymediatedspectctimagingrevealsthespatiotemporalexpressionofprogrammeddeathligand1inresponsetoacd8tcellandinktcellactivatingmrnavaccine
AT deprezjoke nanobodymediatedspectctimagingrevealsthespatiotemporalexpressionofprogrammeddeathligand1inresponsetoacd8tcellandinktcellactivatingmrnavaccine
AT heremansyves nanobodymediatedspectctimagingrevealsthespatiotemporalexpressionofprogrammeddeathligand1inresponsetoacd8tcellandinktcellactivatingmrnavaccine
AT goyvaertscleo nanobodymediatedspectctimagingrevealsthespatiotemporalexpressionofprogrammeddeathligand1inresponsetoacd8tcellandinktcellactivatingmrnavaccine
AT staelswillem nanobodymediatedspectctimagingrevealsthespatiotemporalexpressionofprogrammeddeathligand1inresponsetoacd8tcellandinktcellactivatingmrnavaccine
AT desmedtstefaan nanobodymediatedspectctimagingrevealsthespatiotemporalexpressionofprogrammeddeathligand1inresponsetoacd8tcellandinktcellactivatingmrnavaccine
AT dewitteheleen nanobodymediatedspectctimagingrevealsthespatiotemporalexpressionofprogrammeddeathligand1inresponsetoacd8tcellandinktcellactivatingmrnavaccine
AT devoogdtnick nanobodymediatedspectctimagingrevealsthespatiotemporalexpressionofprogrammeddeathligand1inresponsetoacd8tcellandinktcellactivatingmrnavaccine
AT keyaertsmarleen nanobodymediatedspectctimagingrevealsthespatiotemporalexpressionofprogrammeddeathligand1inresponsetoacd8tcellandinktcellactivatingmrnavaccine
AT verbekerein nanobodymediatedspectctimagingrevealsthespatiotemporalexpressionofprogrammeddeathligand1inresponsetoacd8tcellandinktcellactivatingmrnavaccine
AT barbekurt nanobodymediatedspectctimagingrevealsthespatiotemporalexpressionofprogrammeddeathligand1inresponsetoacd8tcellandinktcellactivatingmrnavaccine
AT lentackerine nanobodymediatedspectctimagingrevealsthespatiotemporalexpressionofprogrammeddeathligand1inresponsetoacd8tcellandinktcellactivatingmrnavaccine
AT breckpotkarine nanobodymediatedspectctimagingrevealsthespatiotemporalexpressionofprogrammeddeathligand1inresponsetoacd8tcellandinktcellactivatingmrnavaccine