RIVERSIDE, Calif. — In a significant stride toward combating metastatic cancer, Armida Labs, Inc., a University of California, Riverside startup, has secured a $2.25 million Small Business Innovation Research (SBIR) Phase II grant from the National Cancer Institute, part of the National Institutes of Health. This substantial funding advancement aims to accelerate the preclinical studies essential for an Investigational New Drug (IND) application and ultimately progress Targefrin, an innovative anti-metastatic therapeutic candidate, toward human clinical trials.
Targefrin represents a groundbreaking approach in targeting EphA2, or ephrin type-A receptor 2, a receptor tyrosine kinase that is markedly overexpressed in a variety of cancers including pancreatic, prostate, lung, breast, ovarian, and colorectal malignancies. EphA2’s aberrant expression facilitates cancer metastasis by promoting cancer cell migration from the primary tumor and enabling colonization of distant tissues. The ability of Targefrin to selectively degrade EphA2 offers a paradigm shift in controlling tumor progression and metastasis.
The molecule known as Targefrin was originally discovered in the laboratory of Maurizio Pellecchia, a renowned professor at the UCR School of Medicine and one of Armida Labs’ co-founders. Pellecchia’s pioneering work led to the conceptualization of Targefrin as a dimeric peptide mimetic designed to engage EphA2 in a unique mechanism of action. The IND-enabling studies will be overseen by Carlo Baggio, Armida Labs’ co-founder and chief technology officer, who serves as principal investigator for the grant.
Baggio highlighted that the laboratory’s multi-year focus on EphA2 has culminated in Targefrin’s evolution. “EphA2 is a key driver of pancreatic cancer aggressiveness,” he noted, “and elevated EphA2 expression is associated with dismal patient prognosis. Targefrin’s mechanism — effectively degrading EphA2 — has the potential to convert aggressive tumors into less invasive phenotypes.” The successful Phase I SBIR award of $600,000 laid the foundation for this current expanded research endeavor.
Delving into the biochemistry of Targefrin reveals a sophisticated molecular design inspired by nature’s own regulatory systems. The molecule is a dimeric peptide mimetic, structurally engineered to emulate ephrins—natural ligands of the EphA2 receptor. Through dimerization, Targefrin induces receptor clustering, a biological process that triggers internalization and degradation of EphA2 from the cancer cell surface. This targeted receptor downregulation disrupts the pro-oncogenic signaling cascade that EphA2 mediates in its ligand-independent, overexpressed state.
EphA2’s dualistic nature in cancer biology is critical to understanding Targefrin’s therapeutic rationale. Under normal physiological conditions, EphA2 interactions with ephrin ligands suppress tumorigenesis by inhibiting cell migration and proliferation. However, in many solid tumors, including pancreatic and prostate cancers, EphA2 is upregulated independently of its ligands, converting it into a pro-metastatic oncoprotein. This ligand-independent EphA2 promotes cellular motility, invasion, and establishment of metastatic niches, representing a formidable obstacle in cancer therapy. Targefrin’s ability to restore balance by mimicking ligand binding and triggering receptor degradation directly counteracts this pathological state.
The development pipeline for Targefrin involved an iterative chemical design process, meticulously refining the molecule to optimize its affinity and selectivity for EphA2. Each chemical modification sought to enhance pharmacodynamic properties while maintaining specificity, a critical factor for minimizing off-target effects and improving therapeutic indices. This rational drug design process exemplifies the intersection of medicinal chemistry and molecular biology driving next-generation cancer therapeutics.
Looking forward, Armida Labs is committed to advancing Targefrin through rigorous IND-enabling studies using the current SBIR funding. The primary focus is pancreatic cancer, one of the most lethal and treatment-resistant malignancies worldwide, underscoring the urgent need for novel therapeutic agents. Beyond pancreatic cancer, the scope of Targefrin’s application may extend to a wide range of EphA2-driven tumors, potentiating broad clinical impact.
The research team is optimistic about the translational potential of Targefrin and the implications for targeted therapy in metastatic disease. Pellecchia expressed enthusiasm: “Coming from an academic laboratory setting to startup verification embodies the spirit of translational medicine. With this funding, we are poised to move this innovative molecule closer to clinical reality.” He emphasized that additional support through fundraising will be vital to propel the molecule into early phase clinical trials.
Technical insights into Targefrin’s mode-of-action illustrate its distinction from traditional small-molecule inhibitors or monoclonal antibodies targeting receptor tyrosine kinases. Instead of competitive inhibition, Targefrin leverages induced receptor dimerization to hijack the natural receptor downregulation pathway, effectively removing the pro-tumorigenic receptor from the cancer cell architecture. This biological elegance represents an advanced modality in therapeutic design, addressing the unmet challenge of targeting overexpressed receptors in a ligand-deficient microenvironment.
The advancement of Targefrin is emblematic of a broader trend in oncology drug development focusing on protein degradation technologies, including proteolysis targeting chimeras (PROTACs) and similar modalities. Targefrin contributes to this evolving landscape by utilizing receptor dimerization-induced degradation, a less explored but highly promising avenue. If successful, this strategy may inspire novel approaches for other receptor-driven cancers.
Beyond the molecular frontiers, the initiative by Armida Labs underscores the critical role of academic-industry partnerships and government-funded innovation programs in accelerating cancer drug discovery. The SBIR grants exemplify how carefully structured funding mechanisms can bridge early-stage scientific discoveries with translational development, fostering the pipeline for novel therapeutics tailored to pressing clinical needs.
As Armida Labs moves forward, the oncology community will keenly watch the outcomes of these preclinical studies. The potential to transform metastatic cancer treatment could herald a new era where therapeutics not only inhibit tumor growth but also dismantle the cellular mechanisms facilitating metastasis, improving survival and quality of life for countless patients worldwide.
Subject of Research: Anti-metastatic therapy targeting EphA2 receptor in cancer using a dimeric peptide mimetic.
Article Title: Not provided.
News Publication Date: Not provided.
Web References:
- Armida Labs: http://armida-labs.com/
- UCR School of Medicine: https://profiles.ucr.edu/app/home/profile/maurizio
- Center for Molecular and Translational Medicine: https://molmed.ucr.edu/
- Armida Labs Team: https://www.armidalabs.com/armida-labs-team/carlo-baggio-ph-d
- UCR homepage: http://www.ucr.edu/
Image Credits: Armida Labs, Inc.
Keywords: Targefrin, EphA2, anti-metastatic agent, pancreatic cancer, receptor degradation, peptide mimetic, cancer metastasis, preclinical studies, SBIR grant, National Cancer Institute, molecular targeted therapy, peptide dimerization.