Antibody-induced erythrophagocyte reprogramming of Kupffer cells prevents anti-CD40 cancer immunotherapy-associated liver toxicity.

Details

Ressource 1Download: J Immunother Cancer 2023.pdf (19099.01 [Ko])
State: Public
Version: author
License: CC BY-NC 4.0
Serval ID
serval:BIB_255273ACC947
Type
Article: article from journal or magazin.
Collection
Publications
Institution
Title
Antibody-induced erythrophagocyte reprogramming of Kupffer cells prevents anti-CD40 cancer immunotherapy-associated liver toxicity.
Journal
Journal for immunotherapy of cancer
Author(s)
Pfefferlé M., Dubach I.L., Buzzi R.M., Dürst E., Schulthess-Lutz N., Baselgia L., Hansen K., Imhof L., Koernig S., Le Roy D., Roger T., Humar R., Schaer D.J., Vallelian F.
ISSN
2051-1426 (Electronic)
ISSN-L
2051-1426
Publication state
Published
Issued date
01/2023
Peer-reviewed
Oui
Volume
11
Number
1
Pages
e005718
Language
english
Notes
Publication types: Journal Article
Publication Status: ppublish
Abstract
Agonistic anti-CD40 monoclonal antibodies (mAbs) have emerged as promising immunotherapeutic compounds with impressive antitumor effects in mouse models. However, preclinical and clinical studies faced dose-limiting toxicities mediated by necroinflammatory liver disease. An effective prophylactic treatment for liver immune-related adverse events that does not suppress specific antitumor immunity remains to be found.
We used different mouse models and time-resolved single-cell RNA-sequencing to characterize the pathogenesis of anti-CD40 mAb induced liver toxicity. Subsequently, we developed an antibody-based treatment protocol to selectively target red blood cells (RBCs) for erythrophagocytosis in the liver, inducing an anti-inflammatory liver macrophage reprogramming.
We discovered that CD40 signaling in Clec4f <sup>+</sup> Kupffer cells is the non-redundant trigger of anti-CD40 mAb-induced liver toxicity. Taking advantage of the highly specific functionality of liver macrophages to clear antibody-tagged RBCs from the blood, we hypothesized that controlled erythrophagocytosis and the linked anti-inflammatory signaling by the endogenous metabolite heme could be exploited to reprogram liver macrophages selectively. Repeated low-dose administration of a recombinant murine Ter119 antibody directed RBCs for selective phagocytosis in the liver and skewed the phenotype of liver macrophages into a Hmox <sup>high</sup> /Marco <sup>high</sup> /MHCII <sup>low</sup> anti-inflammatory phenotype. This unique mode of action prevented necroinflammatory liver disease following high-dose administration of anti-CD40 mAbs. In contrast, extrahepatic inflammation, antigen-specific immunity, and antitumor activity remained unaffected in Ter119 treated animals.
Our study offers a targeted approach to uncouple CD40-augmented antitumor immunity in peripheral tissues from harmful inflammatoxicity in the liver.
Keywords
Mice, Animals, Kupffer Cells/metabolism, Antibodies, Monoclonal/therapeutic use, Antineoplastic Agents/therapeutic use, Immunotherapy/methods, Liver, Neoplasms, Immunity, Innate, Immunotherapy, Macrophages
Pubmed
Open Access
Yes
Funding(s)
SNF/Projects/310030_207418 SNF/Projects/310030_197823 SNF/Projects/310030_201202 SNF/Careers/323530_183984 OTHER//Fondation Carigest-Promex Stiftung für die Forschung
Create date
10/01/2023 14:18
Last modification date
11/03/2023 8:08
Usage data