Cargando…

Mouse Preclinical Cancer Immunotherapy Modeling Involving Anti-PD-1 Therapies Reveals the Need to Use Mouse Reagents to Mirror Clinical Paradigms

SIMPLE SUMMARY: Immune checkpoint inhibition has revolutionized clinical cancer care. As clinical use of these inhibitors increases, the ability to study the effects of these therapies in preclinical models becomes more important. This study highlights the need for using species appropriate reagents...

Descripción completa

Detalles Bibliográficos
Autores principales: Monjazeb, Arta M., Wang, Ziming, Vick, Logan V., Dunai, Cordelia, Minnar, Christine, Khuat, Lam T., Murphy, William J.
Formato: Online Artículo Texto
Lenguaje:English
Publicado: MDPI 2021
Materias:
Acceso en línea:https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7916633/
https://www.ncbi.nlm.nih.gov/pubmed/33578798
http://dx.doi.org/10.3390/cancers13040729
_version_ 1783657522356289536
author Monjazeb, Arta M.
Wang, Ziming
Vick, Logan V.
Dunai, Cordelia
Minnar, Christine
Khuat, Lam T.
Murphy, William J.
author_facet Monjazeb, Arta M.
Wang, Ziming
Vick, Logan V.
Dunai, Cordelia
Minnar, Christine
Khuat, Lam T.
Murphy, William J.
author_sort Monjazeb, Arta M.
collection PubMed
description SIMPLE SUMMARY: Immune checkpoint inhibition has revolutionized clinical cancer care. As clinical use of these inhibitors increases, the ability to study the effects of these therapies in preclinical models becomes more important. This study highlights the need for using species appropriate reagents to properly evaluate the efficacy and toxicity of cancer immunotherapy in mouse models. ABSTRACT: Immune checkpoint inhibition (ICI) has emerged as one of the most powerful tools to reverse cancer induced immune suppression. Monoclonal antibodies (mAbs) targeting programmed cell death 1/programmed cell death ligand 1(PD-1/PD-L1) are FDA-approved and their clinical use is rapidly expanding. As opposed to the clinical paradigm, which can result in significant responses and toxicities, it has been difficult to reproduce these effects preclinically using mouse models. In large part, this is due to models, which employ rapidly growing ex vivo cultured transplantable tumor cell lines engrafted into young naïve inbred laboratory mice. However, another issue concerns the use and repeated application of xenogeneic reagents in mice (i.e., rat or hamster mAbs directed against mouse antigens at variance with clinical use of human or humanized mAbs). Building on our previous studies demonstrating that repeated administration of commonly used xenogeneic anti-PD-1 mAbs derived from both rat and hamster can induce fatal hypersensitivity in some tumor-bearing mice, we sought to compare these result with the effects of a mouse anti-mouse PD-1 mAb. Application of a murine anti-mouse PD-1 (clone: MuDX400) did not result in lethal anaphylaxis in the 4T1 tumor model. It also displayed superior antitumor effects in this and other tumor models, as it did not induce neutralizing antibody responses against the anti-PD-1 mAb, such as were observed when using xenogeneic anti-PD1 mAbs. These results demonstrate that more accurate preclinical modeling necessitates the use of mouse reagents mirroring the clinical scenario to ascertain long-term effects or toxicities, while avoiding xenogeneic responses, which do not occur clinically. Furthermore, these studies suggest a direct mechanism, whereby preclinical murine studies have often failed to recapitulate the clinical efficacy and toxicity of single agent checkpoint inhibition.
format Online
Article
Text
id pubmed-7916633
institution National Center for Biotechnology Information
language English
publishDate 2021
publisher MDPI
record_format MEDLINE/PubMed
spelling pubmed-79166332021-03-01 Mouse Preclinical Cancer Immunotherapy Modeling Involving Anti-PD-1 Therapies Reveals the Need to Use Mouse Reagents to Mirror Clinical Paradigms Monjazeb, Arta M. Wang, Ziming Vick, Logan V. Dunai, Cordelia Minnar, Christine Khuat, Lam T. Murphy, William J. Cancers (Basel) Article SIMPLE SUMMARY: Immune checkpoint inhibition has revolutionized clinical cancer care. As clinical use of these inhibitors increases, the ability to study the effects of these therapies in preclinical models becomes more important. This study highlights the need for using species appropriate reagents to properly evaluate the efficacy and toxicity of cancer immunotherapy in mouse models. ABSTRACT: Immune checkpoint inhibition (ICI) has emerged as one of the most powerful tools to reverse cancer induced immune suppression. Monoclonal antibodies (mAbs) targeting programmed cell death 1/programmed cell death ligand 1(PD-1/PD-L1) are FDA-approved and their clinical use is rapidly expanding. As opposed to the clinical paradigm, which can result in significant responses and toxicities, it has been difficult to reproduce these effects preclinically using mouse models. In large part, this is due to models, which employ rapidly growing ex vivo cultured transplantable tumor cell lines engrafted into young naïve inbred laboratory mice. However, another issue concerns the use and repeated application of xenogeneic reagents in mice (i.e., rat or hamster mAbs directed against mouse antigens at variance with clinical use of human or humanized mAbs). Building on our previous studies demonstrating that repeated administration of commonly used xenogeneic anti-PD-1 mAbs derived from both rat and hamster can induce fatal hypersensitivity in some tumor-bearing mice, we sought to compare these result with the effects of a mouse anti-mouse PD-1 mAb. Application of a murine anti-mouse PD-1 (clone: MuDX400) did not result in lethal anaphylaxis in the 4T1 tumor model. It also displayed superior antitumor effects in this and other tumor models, as it did not induce neutralizing antibody responses against the anti-PD-1 mAb, such as were observed when using xenogeneic anti-PD1 mAbs. These results demonstrate that more accurate preclinical modeling necessitates the use of mouse reagents mirroring the clinical scenario to ascertain long-term effects or toxicities, while avoiding xenogeneic responses, which do not occur clinically. Furthermore, these studies suggest a direct mechanism, whereby preclinical murine studies have often failed to recapitulate the clinical efficacy and toxicity of single agent checkpoint inhibition. MDPI 2021-02-10 /pmc/articles/PMC7916633/ /pubmed/33578798 http://dx.doi.org/10.3390/cancers13040729 Text en © 2021 by the authors. Licensee MDPI, Basel, Switzerland. This article is an open access article distributed under the terms and conditions of the Creative Commons Attribution (CC BY) license (http://creativecommons.org/licenses/by/4.0/).
spellingShingle Article
Monjazeb, Arta M.
Wang, Ziming
Vick, Logan V.
Dunai, Cordelia
Minnar, Christine
Khuat, Lam T.
Murphy, William J.
Mouse Preclinical Cancer Immunotherapy Modeling Involving Anti-PD-1 Therapies Reveals the Need to Use Mouse Reagents to Mirror Clinical Paradigms
title Mouse Preclinical Cancer Immunotherapy Modeling Involving Anti-PD-1 Therapies Reveals the Need to Use Mouse Reagents to Mirror Clinical Paradigms
title_full Mouse Preclinical Cancer Immunotherapy Modeling Involving Anti-PD-1 Therapies Reveals the Need to Use Mouse Reagents to Mirror Clinical Paradigms
title_fullStr Mouse Preclinical Cancer Immunotherapy Modeling Involving Anti-PD-1 Therapies Reveals the Need to Use Mouse Reagents to Mirror Clinical Paradigms
title_full_unstemmed Mouse Preclinical Cancer Immunotherapy Modeling Involving Anti-PD-1 Therapies Reveals the Need to Use Mouse Reagents to Mirror Clinical Paradigms
title_short Mouse Preclinical Cancer Immunotherapy Modeling Involving Anti-PD-1 Therapies Reveals the Need to Use Mouse Reagents to Mirror Clinical Paradigms
title_sort mouse preclinical cancer immunotherapy modeling involving anti-pd-1 therapies reveals the need to use mouse reagents to mirror clinical paradigms
topic Article
url https://www.ncbi.nlm.nih.gov/pmc/articles/PMC7916633/
https://www.ncbi.nlm.nih.gov/pubmed/33578798
http://dx.doi.org/10.3390/cancers13040729
work_keys_str_mv AT monjazebartam mousepreclinicalcancerimmunotherapymodelinginvolvingantipd1therapiesrevealstheneedtousemousereagentstomirrorclinicalparadigms
AT wangziming mousepreclinicalcancerimmunotherapymodelinginvolvingantipd1therapiesrevealstheneedtousemousereagentstomirrorclinicalparadigms
AT vickloganv mousepreclinicalcancerimmunotherapymodelinginvolvingantipd1therapiesrevealstheneedtousemousereagentstomirrorclinicalparadigms
AT dunaicordelia mousepreclinicalcancerimmunotherapymodelinginvolvingantipd1therapiesrevealstheneedtousemousereagentstomirrorclinicalparadigms
AT minnarchristine mousepreclinicalcancerimmunotherapymodelinginvolvingantipd1therapiesrevealstheneedtousemousereagentstomirrorclinicalparadigms
AT khuatlamt mousepreclinicalcancerimmunotherapymodelinginvolvingantipd1therapiesrevealstheneedtousemousereagentstomirrorclinicalparadigms
AT murphywilliamj mousepreclinicalcancerimmunotherapymodelinginvolvingantipd1therapiesrevealstheneedtousemousereagentstomirrorclinicalparadigms