An autologous cell therapy based on our
T cell Antigen Coupler (TAC) platform.

Unlike conventional chimeric
receptors, our lead program co-opts
natural T cell biology and functions.

Unlike conventional chimeric receptors, our lead program co-opts natural T cell biology and functions.
T cell activation and signaling via the endogenous T cell receptor (TCR)

TAC101-CLDN18.2 is under development for targeting relapsed or refractory CLDN18.2-positive solid tumors.

Claudin18.2 (CLDN18.2) is a novel, highly selective marker protein that is exclusively expressed in differentiated gastric mucosal membrane epithelial cells. While expression of CLDN18.2 is limited in healthy tissues, aberrant expression is frequently detected during the occurrence and development of a variety of malignant tumors. Upon malignant transformation of gastric epithelial tissue, cellular disruptions lead to exposure of CLDN18.2 epitopes, and CLDN18.2 becomes a highly expressed, highly selective, and stable marker of specific tumor tissues.

Claudin18.2 Cancers

An emerging, recently validated therapeutic target.

Despite being the foundation of treatment for advanced gastric and gastroesophageal tumors, traditional chemotherapy is associated with significant toxicities and subject to considerable limitations in terms of therapeutic outcomes. Advances in precision medicine have generated renewed interest in targeted and immunotherapeutic approaches to managing advanced gastric and gastroesophageal cancers, offering new avenues of hope for patients. Based on its expression pattern in malignant tissues, CLDN18.2 has emerged as a target of interest for new therapeutics.

With numerous completed and ongoing clinical trials testing various targeted treatment approaches, there is a growing body of clinical evidence demonstrating that therapies targeting CLDN18.2 can potentially lead to improved response, disease control and survival rates relative to standard chemotherapy regimens. However, to date there are no approved CLDN18.2 targeted treatments, creating a significant unmet need for patients with advanced gastric and gastroesophageal cancer. Our TAC technology has the potential to address that need, producing strong preclinical data in multiple models of CLDN18.2-overexpressing cancers.

A truly scalable technology for automating autologous cell therapy manufacturing.

Given the current challenges of existing autologous therapies, we have entered a risk-sharing collaboration with Lonza to use their Cocoon™ Manufacturing System. We believe the Cocoon will enable us to deliver a high-quality TAC101-CLDN18.2 cell therapy products for each patient in need.