What it does
We design mRNA with a toehold switch that detects cancer-specific RNA in liver cells. Once activated, it produces gasdermin, triggering targeted cancer cell death—offering precise, low-cost therapy for hepatocellular carcinoma.
Your inspiration
We knew that intravenously delivered mRNA, as used in CAR-T therapies, tends to accumulate and translate in the liver, reducing their effectiveness in other tissues. This liver accumulation led to decreased CAR-T therapy efficiency. Instead of avoiding it, we turned this disadvantage into an advantage. Inspired by this, we designed a targeted mRNA-based therapy for hepatocellular carcinoma (HCC)—the most common liver cancer. Using a toehold switch and gasdermin expression, we trigger therapeutic action only in cancerous liver cells. Our solution improves specificity, limits side effects, and may reduce costs.
How it works
Our therapy is based on mRNA technology, similar to that used in vaccines. We engineered a strand of mRNA that has two main functions. First, it contains the genetic instructions to produce a protein called gasdermin, which can create holes in cell membranes and lead to the death of cancer cells. However, to make sure this only happens in the right cells, we included a safety mechanism called a toehold switch. This switch keeps the mRNA inactive unless it detects a specific RNA sequence found only in liver cancer cells (HCC). When this sequence is present, the switch opens and the mRNA is translated into the gasdermin protein, which then triggers cell death. We deliver this mRNA into the body using lipid nanoparticles—tiny fat-based carriers that naturally accumulate in the liver. This delivery method, combined with the cancer-specific activation mechanism, ensures the therapy works only in cancerous liver cells, minimizing harm to healthy tissue.
Design process
Our design began with extensive literature research on riboswitches, especially toeholds in mammalian cells. We used the TrigGate server to design switches responsive to AFP mRNA, placing the Kozak sequence after the stem for increased stability. After running into server-side limitations, we developed a custom Python-Selenium tool to split the mRNA into overlapping 200-nt fragments, ensuring complete coverage. We optimized GC content, Kozak offset, loop/stem parameters, and filtered results using BLAST to exclude off-target effects. Best constructs expressing GFP or Gasdermin N were selected based on thermodynamic stability and structural features. Currently, we are cloning these sequences using MoClo. The toehold-GFP constructs will be tested in Hep-G2, SNU-449 and THLE-2 cell lines for fluorescence evaluation. Strongest toeholds will advance to cytotoxic mRNA assays in HepatoXCell, using IVT mRNA, with pyroptosis measured via Annexin V/PI flow cytometry, LDH, ATP, and IL-1β/IL-18 release. Nigericin and sorafenib serve as pyroptotic and therapeutic benchmarks. The results will guide final therapeutic candidate selection and inform safety/efficacy profiling in hepatocellular carcinoma models.
How it is different
Our therapy is fundamentally different from current HCC treatments like small-molecule drugs (e.g., sorafenib) or monoclonal antibodies (e.g., Tecentriq + Avastin). These options are not molecularly targeted, often cause side effects such as hepatotoxicity or fatigue, and tumors quickly develop resistance through alternative signaling pathways. Our solution uses mRNA with a toehold switch and expresses gasdermin to trigger pyroptosis only in cancerous liver cells. This offers high specificity, minimal off-target effects, and directly targets tumor microenvironment mechanisms. Unlike personalized therapies (e.g., TCR-T or neoantigen vaccines) developed by Geneos, Tvardi, or SCG, our design is scalable, cost-effective, and fast to deploy. Even mRNA leaders like Moderna and BioNTech do not use toehold switches or pyroptosis in their cancer pipelines. We introduce a novel therapeutic mechanism and a new class of targeted treatment for HCC.
Future plans
We are preparing to compete in the iGEM MIT competition this October. We continue to consult with doctors and scientists, and soon plan to engage with patients to better understand the people we aim to help. By then, we intend to finalize and select the most effective riboswitches based on lab testing. Next, we aim to secure further funding to move into preclinical testing on animal models. We are also developing our software to improve therapy personalization and expand its use to other cancer types. In the long term, we are considering both commercialization of the software and clinical translation of our therapeutic.
Awards
We qualified for the finals of iGEM, the world’s largest synthetic biology competition organized by MIT. The project was presented at seminars at IIMCB , WUT, and the 4EU+ Alliance Against Cancer at Sorbonne. We were awarded funding by Oxford Nanopore. Partnership: Bioforum, UW Incubator, Innovations Hub program.
Share this page on