American Association for Cancer Research
Browse
mct-23-0359_supplementary_figure_7_supps7.png (397.69 kB)

Supplementary figure 7 from Design and Evaluation of Phosphonamidate-Linked Exatecan Constructs for Highly Loaded, Stable, and Efficacious Antibody–Drug Conjugates

Download (397.69 kB)
figure
posted on 2024-02-01, 08:40 authored by Saskia Schmitt, Paul Machui, Isabelle Mai, Sarah Herterich, Swetlana Wunder, Philipp Cyprys, Marcus Gerlach, Philipp Ochtrop, Christian P.R. Hackenberger, Dominik Schumacher, Jonas Helma, Annette M. Vogl, Marc-André Kasper

Exatecan, DXd, Enhertu and trastuzumab-LP5 induce immunogenic cell death. Immunogenic cell death induced by exatecan, DXd, trastuzumab-LP5 DAR 8 and Enhertu. HER2-positive SKBR-3 and N-87 cells were treated for indicated time points with 20 nM of respective free payloads and ADCs. As markers for immunogenic cell death, ATP and HMGB1 release into the supernatant as well as calreticulin exposure on the cell surface were determined. Therefore, cells were stained with live/dead stain and anti-calreticulin and analyzed by flow cytometry. ATP and HMGB1 release into the supernatant were measured by luminescence-based readouts. Graphs show means of n = 2 ± SEM.

Funding

Tubulis GmbH

History

ARTICLE ABSTRACT

Topoisomerase I (TOP1) Inhibitors constitute an emerging payload class to engineer antibody–drug conjugates (ADC) as next-generation biopharmaceutical for cancer treatment. Existing ADCs are using camptothecin payloads with lower potency and suffer from limited stability in circulation. With this study, we introduce a novel camptothecin-based linker–payload platform based on the highly potent camptothecin derivative exatecan. First, we describe general challenges that arise from the hydrophobic combination of exatecan and established dipeptidyl p-aminobenzyl-carbamate (PAB) cleavage sites such as reduced antibody conjugation yields and ADC aggregation. After evaluating several linker–payload structures, we identified ethynyl-phosphonamidates in combination with a discrete PEG24 chain to compensate for the hydrophobic PAB–exatecan moiety. Furthermore, we demonstrate that the identified linker–payload structure enables the construction of highly loaded DAR8 ADCs with excellent solubility properties. Head-to-head comparison with Enhertu, an approved camptothecin-based ADC, revealed improved target-mediated killing of tumor cells, excellent bystander killing, drastically improved linker stability in vitro and in vivo and superior in vivo efficacy over four tested dose levels in a xenograft model. Moreover, we show that ADCs based on the novel exatecan linker–payload platform exhibit antibody-like pharmacokinetic properties, even when the ADCs are highly loaded with eight drug molecules per antibody. This ADC platform constitutes a new and general solution to deliver TOP1 inhibitors with highest efficiency to the site of the tumor, independent of the antibody and its target, and is thereby broadly applicable to various cancer indications.