Cargando…

Sources of inter-individual variability leading to significant changes in anti-PD-1 and anti-PD-L1 efficacy identified in mouse tumor models using a QSP framework

While anti-PD-1 and anti-PD-L1 [anti-PD-(L)1] monotherapies are effective treatments for many types of cancer, high variability in patient responses is observed in clinical trials. Understanding the sources of response variability can help prospectively identify potential responsive patient populati...

Descripción completa

Detalles Bibliográficos
Autores principales: Leete, Jessica C., Zager, Michael G., Musante, Cynthia J., Shtylla, Blerta, Qiao, Wenlian
Formato: Online Artículo Texto
Lenguaje:English
Publicado: Frontiers Media S.A. 2022
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9760747/
https://www.ncbi.nlm.nih.gov/pubmed/36545310
http://dx.doi.org/10.3389/fphar.2022.1056365
_version_ 1784852547939336192
author Leete, Jessica C.
Zager, Michael G.
Musante, Cynthia J.
Shtylla, Blerta
Qiao, Wenlian
author_facet Leete, Jessica C.
Zager, Michael G.
Musante, Cynthia J.
Shtylla, Blerta
Qiao, Wenlian
author_sort Leete, Jessica C.
collection PubMed
description While anti-PD-1 and anti-PD-L1 [anti-PD-(L)1] monotherapies are effective treatments for many types of cancer, high variability in patient responses is observed in clinical trials. Understanding the sources of response variability can help prospectively identify potential responsive patient populations. Preclinical data may offer insights to this point and, in combination with modeling, may be predictive of sources of variability and their impact on efficacy. Herein, a quantitative systems pharmacology (QSP) model of anti-PD-(L)1 was developed to account for the known pharmacokinetic properties of anti-PD-(L)1 antibodies, their impact on CD8(+) T cell activation and influx into the tumor microenvironment, and subsequent anti-tumor effects in CT26 tumor syngeneic mouse model. The QSP model was sufficient to describe the variability inherent in the anti-tumor responses post anti-PD-(L)1 treatments. Local sensitivity analysis identified tumor cell proliferation rate, PD-1 expression on CD8(+) T cells, PD-L1 expression on tumor cells, and the binding affinity of PD-1:PD-L1 as strong influencers of tumor growth. It also suggested that treatment-mediated tumor growth inhibition is sensitive to T cell properties including the CD8(+) T cell proliferation half-life, CD8(+) T cell half-life, cytotoxic T-lymphocyte (CTL)-mediated tumor cell killing rate, and maximum rate of CD8(+) T cell influx into the tumor microenvironment. Each of these parameters alone could not predict anti-PD-(L)1 treatment response but they could shift an individual mouse’s treatment response when perturbed. The presented preclinical QSP modeling framework provides a path to incorporate potential sources of response variability in human translation modeling of anti-PD-(L)1.
format Online
Article
Text
id pubmed-9760747
institution National Center for Biotechnology Information
language English
publishDate 2022
publisher Frontiers Media S.A.
record_format MEDLINE/PubMed
spelling pubmed-97607472022-12-20 Sources of inter-individual variability leading to significant changes in anti-PD-1 and anti-PD-L1 efficacy identified in mouse tumor models using a QSP framework Leete, Jessica C. Zager, Michael G. Musante, Cynthia J. Shtylla, Blerta Qiao, Wenlian Front Pharmacol Pharmacology While anti-PD-1 and anti-PD-L1 [anti-PD-(L)1] monotherapies are effective treatments for many types of cancer, high variability in patient responses is observed in clinical trials. Understanding the sources of response variability can help prospectively identify potential responsive patient populations. Preclinical data may offer insights to this point and, in combination with modeling, may be predictive of sources of variability and their impact on efficacy. Herein, a quantitative systems pharmacology (QSP) model of anti-PD-(L)1 was developed to account for the known pharmacokinetic properties of anti-PD-(L)1 antibodies, their impact on CD8(+) T cell activation and influx into the tumor microenvironment, and subsequent anti-tumor effects in CT26 tumor syngeneic mouse model. The QSP model was sufficient to describe the variability inherent in the anti-tumor responses post anti-PD-(L)1 treatments. Local sensitivity analysis identified tumor cell proliferation rate, PD-1 expression on CD8(+) T cells, PD-L1 expression on tumor cells, and the binding affinity of PD-1:PD-L1 as strong influencers of tumor growth. It also suggested that treatment-mediated tumor growth inhibition is sensitive to T cell properties including the CD8(+) T cell proliferation half-life, CD8(+) T cell half-life, cytotoxic T-lymphocyte (CTL)-mediated tumor cell killing rate, and maximum rate of CD8(+) T cell influx into the tumor microenvironment. Each of these parameters alone could not predict anti-PD-(L)1 treatment response but they could shift an individual mouse’s treatment response when perturbed. The presented preclinical QSP modeling framework provides a path to incorporate potential sources of response variability in human translation modeling of anti-PD-(L)1. Frontiers Media S.A. 2022-12-05 /pmc/articles/PMC9760747/ /pubmed/36545310 http://dx.doi.org/10.3389/fphar.2022.1056365 Text en Copyright © 2022 Leete, Zager, Musante, Shtylla and Qiao. https://creativecommons.org/licenses/by/4.0/This is an open-access article distributed under the terms of the Creative Commons Attribution License (CC BY). The use, distribution or reproduction in other forums is permitted, provided the original author(s) and the copyright owner(s) are credited and that the original publication in this journal is cited, in accordance with accepted academic practice. No use, distribution or reproduction is permitted which does not comply with these terms.
spellingShingle Pharmacology
Leete, Jessica C.
Zager, Michael G.
Musante, Cynthia J.
Shtylla, Blerta
Qiao, Wenlian
Sources of inter-individual variability leading to significant changes in anti-PD-1 and anti-PD-L1 efficacy identified in mouse tumor models using a QSP framework
title Sources of inter-individual variability leading to significant changes in anti-PD-1 and anti-PD-L1 efficacy identified in mouse tumor models using a QSP framework
title_full Sources of inter-individual variability leading to significant changes in anti-PD-1 and anti-PD-L1 efficacy identified in mouse tumor models using a QSP framework
title_fullStr Sources of inter-individual variability leading to significant changes in anti-PD-1 and anti-PD-L1 efficacy identified in mouse tumor models using a QSP framework
title_full_unstemmed Sources of inter-individual variability leading to significant changes in anti-PD-1 and anti-PD-L1 efficacy identified in mouse tumor models using a QSP framework
title_short Sources of inter-individual variability leading to significant changes in anti-PD-1 and anti-PD-L1 efficacy identified in mouse tumor models using a QSP framework
title_sort sources of inter-individual variability leading to significant changes in anti-pd-1 and anti-pd-l1 efficacy identified in mouse tumor models using a qsp framework
topic Pharmacology
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC9760747/
https://www.ncbi.nlm.nih.gov/pubmed/36545310
http://dx.doi.org/10.3389/fphar.2022.1056365
work_keys_str_mv AT leetejessicac sourcesofinterindividualvariabilityleadingtosignificantchangesinantipd1andantipdl1efficacyidentifiedinmousetumormodelsusingaqspframework
AT zagermichaelg sourcesofinterindividualvariabilityleadingtosignificantchangesinantipd1andantipdl1efficacyidentifiedinmousetumormodelsusingaqspframework
AT musantecynthiaj sourcesofinterindividualvariabilityleadingtosignificantchangesinantipd1andantipdl1efficacyidentifiedinmousetumormodelsusingaqspframework
AT shtyllablerta sourcesofinterindividualvariabilityleadingtosignificantchangesinantipd1andantipdl1efficacyidentifiedinmousetumormodelsusingaqspframework
AT qiaowenlian sourcesofinterindividualvariabilityleadingtosignificantchangesinantipd1andantipdl1efficacyidentifiedinmousetumormodelsusingaqspframework