Stem cells from a patient’s own fat may have the potential to deliver new treatments directly into the brain after the surgical removal of a glioblastoma, according to researchers at Johns Hopkins. The investigators say mesenchymal stem cells (MSCs) have an unexplained ability to seek out damaged cells, such as those involved in cancer, and may provide clinicians a new tool for accessing difficult-to-reach parts of the brain where cancer cells can hide and proliferate. The researchers say harvesting MSCs from fat is less invasive and less expensive than getting them from bone marrow, a more commonly studied method.

“The biggest challenge in brain cancer is the migration of cancer cells. Even when we remove the tumor, some of the cells have already slipped away and are causing damage somewhere else,” says study leader Alfredo Quinones-Hinojosa, M.D., a professor of neurosurgery, oncology, and neuroscience at the Johns Hopkins University School of Medicine. “Building off our findings, we may be able to find a way to arm a patient’s own healthy cells with the treatment needed to chase down those cancer cells and destroy them. It’s truly personalized medicine.”

Dr. Quinones-Hinojosa and his colleagues purchased human MSCs derived from both fat and bone marrow, and also isolated and grew their own stem cell lines from fat removed from two patients. They found that commercial and primary culture AMSCs and commercial BMSCs demonstrated no statistically significant difference in their migration toward glioma conditioned media in vitro. There was statistically significant difference in the proliferation rate of both commercial AMSCs and BMSCs as compared to primary culture AMSCs, suggesting primary cultures have a slower growth rate than commercially available cell lines.

The scientists say these results suggest that a patient’s own fat cells might work as well as any to create cancer-fighting cells. MSCs, with their ability to home in on cancer cells, might be able to act as a delivery mechanism, bringing drugs, nanoparticles, or some other treatment directly to the cells.

Dr. Quinones-Hinojosa cautions that while further studies are under way, it will be years before human trials of MSC delivery systems can begin. Ideally, he says, if MSCs work, a patient with a glioblastoma would have some adipose tissue (fat) removed a short time before surgery. The MSCs in the fat would be drawn out and manipulated in the lab to carry drugs or other treatments. Then, after surgeons removed the brain tumor, they could deposit these treatment-armed cells into the brain in the hopes that they would seek out and destroy the cancer cells. Given the well-documented ability to harvest larger numbers of adipose MSCs under local anesthesia, adipose tissue may provide a more efficient source of MSCs for research and clinical applications, while minimizing patient morbidity during cell harvesting, he speculates.

“Essentially these MSCs are like a ‘smart’ device that can track cancer cells,” Dr. Quinones-Hinojosa says. He adds that it’s unclear why MSCs are attracted to glioblastoma cells, but they appear to have a natural affinity for sites of damage in the body, such as a wound. MSCs, whether derived from bone marrow or fat, have been studied in animal models to treat trauma, Parkinson’s disease, ALS, and other diseases.

Results of this Johns Hopkins proof-of-principle study are described online in the journal PLOS ONE, in a paper titled “Mesenchymal Stem Cells Derived from Adipose Tissue vs Bone Marrow: In Vitro Comparison of Their Tropism towards Gliomas”.

Previous articleSurvival on Mars Depends on Key Protein Features
Next articleMerck and Luminex Ink Alzheimer’s CDx Deal