The University of Southampton
University of Southampton Institutional Repository

FcγRII (CD32) modulates antibody clearance in NOD SCID mice leading to impaired antibody-mediated tumor cell deletion

FcγRII (CD32) modulates antibody clearance in NOD SCID mice leading to impaired antibody-mediated tumor cell deletion
FcγRII (CD32) modulates antibody clearance in NOD SCID mice leading to impaired antibody-mediated tumor cell deletion
Background Immune compromised mice are increasingly used for the preclinical development of monoclonal antibodies (mAb). Most common are non-obese diabetic (NOD) severe combined immunodeficient (SCID) and their derivatives such as NOD SCID interleukin-2 γ-/- (NSG), which are attractive hosts for patient-derived xenografts. Despite their widespread use, the relative biological performance of mAb in these strains has not been extensively studied.

Methods Clinically relevant mAb of various isotypes were administered to tumor and non-tumor-bearing SCID and NOD SCID mice and the mAb clearance monitored by ELISA. Expression analysis of surface proteins in both strains was carried out by flow cytometry and immunofluorescence microscopy. Further analysis was performed in vitro by surface plasmon resonance to assess mAb affinity for Fcγ receptors (FcγR) at pH 6 and pH 7.4. NOD SCID mice genetically deficient in different FcγR were used to delineate their involvement.

Results Here, we show that strains on the NOD SCID background have significantly faster antibody clearance than other strains leading to reduced antitumor efficacy of clinically relevant mAb. This rapid clearance is dependent on antibody isotype, the presence of Fc glycosylation (at N297) and expression of FcγRII. Comparable effects were not seen in the parental NOD or SCID strains, demonstrating the presence of a compound defect requiring both genotypes. The absence of endogenous IgG was the key parameter transferred from the SCID as reconstituting NOD SCID or NSG mice with exogenous IgG overcame the rapid clearance and recovered antitumor efficacy. In contrast, the NOD strain was associated with reduced expression of the neonatal Fc Receptor (FcRn). We propose a novel mechanism for the rapid clearance of certain mAb isotypes in NOD SCID mouse strains, based on their interaction with FcγRII in the context of reduced FcRn.

Conclusions This study highlights the importance of understanding the limitation of the mouse strain being used for preclinical evaluation, and demonstrates that NOD SCID strains of mice should be reconstituted with IgG prior to studies of mAb efficacy.
antibodies, neoplasm, immunotherapy
1-11
Oldham, Robert J.
9b947e29-a28f-4b4a-a294-d8d70ff0ec07
Mockridge, C. Ian
0137bec1-e2f6-464f-b88b-61d2c31a94ea
James, Sonya
50bd303f-3c0e-4eea-8c01-defa7f25cb18
Duriez, Patrick J.
d8643461-b408-43eb-8900-63d4cd15f0d7
Chan, H.T. Claude
dd26be49-6344-432e-b458-8c644bf4828f
Cox, Kerry L.
2571bd91-cdad-4dea-b47e-b186aebb153f
Pitic, Vicentiu A.
09668404-1710-4e28-9bf4-8f2b19f31b1e
Glennie, Martin J.
566a8be9-c561-4533-8ef3-bcb6ded938bc
Cragg, Mark
ec97f80e-f3c8-49b7-a960-20dff648b78c
Oldham, Robert J.
9b947e29-a28f-4b4a-a294-d8d70ff0ec07
Mockridge, C. Ian
0137bec1-e2f6-464f-b88b-61d2c31a94ea
James, Sonya
50bd303f-3c0e-4eea-8c01-defa7f25cb18
Duriez, Patrick J.
d8643461-b408-43eb-8900-63d4cd15f0d7
Chan, H.T. Claude
dd26be49-6344-432e-b458-8c644bf4828f
Cox, Kerry L.
2571bd91-cdad-4dea-b47e-b186aebb153f
Pitic, Vicentiu A.
09668404-1710-4e28-9bf4-8f2b19f31b1e
Glennie, Martin J.
566a8be9-c561-4533-8ef3-bcb6ded938bc
Cragg, Mark
ec97f80e-f3c8-49b7-a960-20dff648b78c

Oldham, Robert J., Mockridge, C. Ian, James, Sonya, Duriez, Patrick J., Chan, H.T. Claude, Cox, Kerry L., Pitic, Vicentiu A., Glennie, Martin J. and Cragg, Mark (2020) FcγRII (CD32) modulates antibody clearance in NOD SCID mice leading to impaired antibody-mediated tumor cell deletion. Journal for Immunotherapy of Cancer, 8 (1), 1-11, [e000619]. (doi:10.1136/jitc-2020-000619).

Record type: Article

Abstract

Background Immune compromised mice are increasingly used for the preclinical development of monoclonal antibodies (mAb). Most common are non-obese diabetic (NOD) severe combined immunodeficient (SCID) and their derivatives such as NOD SCID interleukin-2 γ-/- (NSG), which are attractive hosts for patient-derived xenografts. Despite their widespread use, the relative biological performance of mAb in these strains has not been extensively studied.

Methods Clinically relevant mAb of various isotypes were administered to tumor and non-tumor-bearing SCID and NOD SCID mice and the mAb clearance monitored by ELISA. Expression analysis of surface proteins in both strains was carried out by flow cytometry and immunofluorescence microscopy. Further analysis was performed in vitro by surface plasmon resonance to assess mAb affinity for Fcγ receptors (FcγR) at pH 6 and pH 7.4. NOD SCID mice genetically deficient in different FcγR were used to delineate their involvement.

Results Here, we show that strains on the NOD SCID background have significantly faster antibody clearance than other strains leading to reduced antitumor efficacy of clinically relevant mAb. This rapid clearance is dependent on antibody isotype, the presence of Fc glycosylation (at N297) and expression of FcγRII. Comparable effects were not seen in the parental NOD or SCID strains, demonstrating the presence of a compound defect requiring both genotypes. The absence of endogenous IgG was the key parameter transferred from the SCID as reconstituting NOD SCID or NSG mice with exogenous IgG overcame the rapid clearance and recovered antitumor efficacy. In contrast, the NOD strain was associated with reduced expression of the neonatal Fc Receptor (FcRn). We propose a novel mechanism for the rapid clearance of certain mAb isotypes in NOD SCID mouse strains, based on their interaction with FcγRII in the context of reduced FcRn.

Conclusions This study highlights the importance of understanding the limitation of the mouse strain being used for preclinical evaluation, and demonstrates that NOD SCID strains of mice should be reconstituted with IgG prior to studies of mAb efficacy.

Text
JITC revised submission proof - Author's Original
Available under License Creative Commons Attribution.
Download (3MB)
Text
e000619.full - Version of Record
Download (1MB)

More information

Accepted/In Press date: 1 May 2020
e-pub ahead of print date: 17 June 2020
Published date: 17 June 2020
Additional Information: Funding Information: with Huntingdon Life Sciences from the MRC (1254288), Programme Grants from Bloodwise (12050) and Cancer Research UK (A24721) as well as CRUK center support C328/A25139. Competing interests MSC is a retained consultant for Bioinvent and has performed educational and advisory roles for Boehringer Ingelheim, Merck KGaA, Baxalta and GLG. He has received research funding from Bioinvent, Roche, Gilead, Iteos, UCB and GSK. Publisher Copyright: ©
Keywords: antibodies, neoplasm, immunotherapy

Identifiers

Local EPrints ID: 440722
URI: http://eprints.soton.ac.uk/id/eprint/440722
PURE UUID: 9927ac41-a0dc-487f-bb33-eb9d6f2fae28
ORCID for Mark Cragg: ORCID iD orcid.org/0000-0003-2077-089X

Catalogue record

Date deposited: 14 May 2020 16:31
Last modified: 17 Mar 2024 02:46

Export record

Altmetrics

Contributors

Author: Robert J. Oldham
Author: C. Ian Mockridge
Author: Sonya James
Author: Patrick J. Duriez
Author: H.T. Claude Chan
Author: Kerry L. Cox
Author: Vicentiu A. Pitic
Author: Martin J. Glennie
Author: Mark Cragg ORCID iD

Download statistics

Downloads from ePrints over the past year. Other digital versions may also be available to download e.g. from the publisher's website.

View more statistics

Atom RSS 1.0 RSS 2.0

Contact ePrints Soton: eprints@soton.ac.uk

ePrints Soton supports OAI 2.0 with a base URL of http://eprints.soton.ac.uk/cgi/oai2

This repository has been built using EPrints software, developed at the University of Southampton, but available to everyone to use.

We use cookies to ensure that we give you the best experience on our website. If you continue without changing your settings, we will assume that you are happy to receive cookies on the University of Southampton website.

×