Year 1

Primary brain tumors are among the most difficult cancers to treat. High-grade gliomas, the most common primary brain tumors in adults, remain incurable with current therapies. These devastating tumors present significant treatment challenges for several reasons: 1) surgical removal runs the risk of causing permanent neurologic damage and does not eliminate cancer cells that have migrated throughout the brain; 2) most anti-cancer drugs are prevented from entering the brain because of the presence of the blood-brain barrier, which often does not allow enough chemotherapy into the brain to kill the cancer cells; and 3) typically, the amount of chemotherapy that can be given to cancer patients is limited by intolerable or harmful side effects from these agents. If concentrated cancer therapies could be specifically localized to sites of tumor, damage to healthy tissues would be avoided.

The long-range goal of this research project is to develop a neural stem cell (NSC)-based treatment strategy that produces a potent, localized anti-tumor effect while minimizing toxic side effects. NSCs hold the promise of improved treatment for brain cancers because they have the natural ability to distribute themselves within a tumor, as well as seek out other sites of tumor in the brain. Because they can home to the tumor cells, NSCs may offer a new way to bring more chemotherapy selectively to brain tumor sites. After modifying the NSCs by transferring a therapeutic gene into them, NSCs can serve as vehicles to deliver anti-cancer treatment directly to the primary tumor, as well as potentially to malignant cells that have spread away from the original tumor site. With funding from CIRM, we are studying the ability of NSCs, that carry an activating protein called carboxylesterase (CE) to convert the chemotherapy agent CPT-11 (irinotecan) to its more potent form, SN-38, at sites of tumor in the brain.

During the first year of funding we have determined that 1) when administered directly into the brain or into a peripheral vein (intravenous injection) of mice with brain tumors, NSCs will travel to several different subtypes of gliomas; 2) we can engineer the NSCs to consistently produce high levels of more powerful forms of CE: rCE and hCE1m6; 3) glioma cells die when they are exposed to very low (nanomolar) concentrations of SN-38, and 4) although glioma cells survive when exposed to a relatively high concentration of CPT-11 alone, they do die when the same concentration of CPT-11 is administered in combination with either rCE or hCE1m6. These results suggest that the engineered NSCs are expressing relatively high levels of CE enzymes and that the CE enzymes are converting CPT-11 into SN-38. We have also been able to label our NSCs with iron particles, so that we can track their movement in real-time by magnetic resonance imaging (MRI), and follow their location and distribution in relation to the tumor.

All of our data thus far support the original hypothesis that effective, tumor-specific therapy for glioma patients can be developed using NSCs that express rCE or hCE1 and the prodrug CPT-11. During the second year of CIRM funding, we will further analyze our data to make a final determination regarding the best form of CE to develop towards clinical trials, and the best dose range and route of delivery of NSCs to achieve maximal tumor coverage. We will then begin our therapeutic studies and start discussions with the Food and Drug Administration, to define the safety studies necessary to obtain approval for testing this new treatment strategy in patients with brain tumors.