Abstract

Abstract cMET is an oncogenic receptor tyrosine kinase overexpressed in multiple types of solid tumors, including non-small cell lung cancer (NSCLC), gastric, head and neck, liver cancer and others. Several cMET-directed therapies including antibody-drug conjugates (ADCs) have been developed that may benefit a subset of patients with tumors that have high cMET expression, MET amplification, or dependency on cMET signaling. MYTX-011 is an investigational, pH-sensitive, vcMMAE-based ADC that is designed to potentially benefit not only patients whose tumors express high levels of cMET, but also a broader set of patients whose tumors express low to moderate levels of cMET. MYTX-011 is engineered to rapidly dissociate from cMET only at the acidic pH of endolysosomes. We have previously demonstrated that MYTX-011 drives increased internalization and cytotoxicity in tumor cells expressing moderate cMET levels compared to a matched parent non-pH sensitive ADC. MYTX-011 also demonstrated markedly superior efficacy in NSCLC xenografts with moderate cMET expression as compared to either the parent ADC or benchmark ADCs representing clinical-stage anti-cMET ADCs with vcMMAE and maytansinoid payloads. MYTX-011 was efficacious in NSCLC PDX models with moderate cMET expression of adenocarcinoma/squamous histology and in heterogeneous cMET-expressing EGFR mutant models. In these studies, we demonstrate that MYTX-011 exerts enhanced efficacy by delivering increased levels of payload to cMET+ tumors compared to the parent ADC. MYTX-011 treatment induced significantly increased levels of phospho-histone H3 (pHH3), a pharmacodynamic marker of MMAE cytotoxicity, in moderate cMET expressing NCI-H1975 NSCLC xenograft tumors compared to the parent ADC. Further, increased levels of free MMAE were detected in tumors from mice treated with MYTX-011 compared to a matched dose of the parent ADC, indicating that the pH dependent binding engineered into MYTX-011 translated to increased payload delivery to tumors in vivo. To explore the potential efficacy of MYTX-011 in other cMET-expressing tumor types, we tested the activity of MYTX-011 in a panel of cell lines derived from gastric and head and neck cancers. MYTX-011 demonstrated increased cMET-dependent cytotoxicity compared to the parent ADC. MYTX-011 was also highly active in cMET+ xenograft models derived from gastric (non-MET amplified), esophageal, and head and neck cancers. Together, these findings highlight the potential of MYTX-011 as a therapeutic candidate for treating a broader range of cMET-expressing malignancies. Citation Format: Deepak Kanojia, William Comb, William Israelsen, Federico Colombo, Lin Nie, Nimish Gera, Thomas Chittenden, Brian Fiske. MYTX-011: A pH-dependent anti-cMET ADC with increased payload delivery in vivo and potent activity against cMET-expressing tumors of various epithelial origins [abstract]. In: Proceedings of the American Association for Cancer Research Annual Meeting 2024; Part 1 (Regular Abstracts); 2024 Apr 5-10; San Diego, CA. Philadelphia (PA): AACR; Cancer Res 2024;84(6_Suppl):Abstract nr 1907.

Full Text
Published version (Free)

Talk to us

Join us for a 30 min session where you can share your feedback and ask us any queries you have

Schedule a call