Abstract

Abstract Multiple species of oncolytic viruses (OVs) have been shown to modulate the tumor microenvironment (TME) by increasing immune cell infiltration and activating stimulatory immune responses, leading to the induction of a tumor-specific immune response. Many engineered OVs achieve tumor specificity by either gene deletion or mutations and are then armed with immunomodulatory transgenes to promote anti-tumor immune responses. In an alternative approach, OVs derived from Codagenix’s codon/codon pair modification platform aim to leverage the natural immunostimulatory capacity of selected viral species for efficacy and take advantage of defects in innate immune sensing and apoptosis mechanisms in cancer cells as well as receptor overexpression for tumor selectivity. CodaLytic is a novel virotherapeutic derived from influenza virus strain A/California/07/2009, that is synthetically engineered to contain over 600 silent mutations in hemagglutinin and neuraminidase genes and is being developed as a novel immunotherapeutic for breast cancer. In the orthotopic EMT6 triple-negative breast cancer model, known for its moderate sensitivity to immunotherapies, intratumoral injection of 108 PFU three times a week for up to 4 weeks as a monotherapy led to significant tumor growth inhibition by 76% (p < 0.001 vs vehicle control), translating into a significant survival benefit with a 66% cure rate. Intravenous rechallenge of EMT6 long-term survivors led to a 27-fold reduction in lung nodule formation and a tumor-specific interferon-γ memory response was observed ex vivo in their splenocytes. Anti-tumor efficacy after CodaLytic treatment was accompanied by a change in the composition of the tumor immune infiltrate with significant increases in T, B and NK cells and increased gene expression of pathways and genes related to T cell effector function, dendritic cell activation, antigen presentation and chemoattraction. In immunotherapy-resistant orthotopic 4T1 tumors, combination of CodaLytic with a CTLA-4 inhibitory antibody, but not anti-PD-1 blockade – a combination that had demonstrated combination benefit in several other preclinical models – led to reduction in tumor growth by 75% (p < 0.0001 vs vehicle control and PD-1 combination group). Median overall survival improved from 22 to 30 days with addition of CTLA-4 blockade. Triple combination including PD-1 inhibition led to improved long-term survival beyond 50 days with 30% complete regressions. In summary, these preclinical data demonstrate CodaLytic’s ability to induce broad innate and adaptive changes in the breast cancer TME, resulting in anti-tumor efficacy and prolonged survival. Characterization of the tumor immune infiltrate after CTLA-4 combination and pharmacodynamic changes achieved after CodaLytic/checkpoint combination treatment in human breast cancer tumoroids will further support identification of correlates of efficacy with translational implications. Together with preclinical toxicology data and demonstrated clinical safety of this attenuated influenza virus after intranasal administration in healthy individuals, CodaLytic emerges as a promising novel viroimmunotherapeutic agent and is planned to enter a phase 1 clinical trial in early 2023. Citation Format: Marcin Stawowczyk, Yiwen Zhao, Katie Pfeffer, Juliana Tafrova, James Rodriguez, Chen Yang, Nusrat Jahan, Sybil A. Tasker, Steffen Mueller, J. Robert Coleman, Johanna K. Kaufmann. Preclinical development of CodaLytic™, a codon-modified influenza virus, as a novel virotherapeutic agent for breast cancer immunotherapy [abstract]. In: Proceedings of the 2022 San Antonio Breast Cancer Symposium; 2022 Dec 6-10; San Antonio, TX. Philadelphia (PA): AACR; Cancer Res 2023;83(5 Suppl):Abstract nr PD2-04.

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