2025 Research Grant Recipients
The ABTA Research Program oversees the competitive, peer-reviewed grant process to ensure funding goes to the most promising brain tumor research. Once awarded, we track each project’s progress and long-term outcomes to measure impact and advance discoveries in the field.
2025 ABTA-Funded Research Projects
The ABTA awarded more than $1.3 million towards 30 grants
The 2025 funded projects span multiple tumor types
- Glioblastoma
- Medulloblastoma
- Metastatic Brain Tumors
- Malignant Glioma
- Diffuse Midline Gliomas
Research areas of focus
- Immunology/Immunotherapy
- Drug Therapies/Experimental Therapeutics
- Epigenetics
- Biomarkers
- Radiation Therapy
- Proteomics
Basic Research Fellowship
The Basic Research Fellowship is a two-year, $100,000 grant, awarded to post-doctoral fellows who are mentored by established and nationally-recognized experts in the neuro-oncology field.
Ahmed Gad, PhD
Institution: Baylor College of Medicine
Mentor: Nabil Ahmed, MD
Tribute: In memory of Kaitlyn Berg
Chimeric antigen receptor T cell (CAR-T) therapy is an innovative treatment that engineers a patient’s own immune cells to find and destroy cancer. While this approach has succeeded in some blood cancers, brain tumor trials have shown only limited tumor control, along with significant side effects.
One major challenge in brain tumors is that drug targets vary widely across tumor types. For instance, the expression levels of HER2, a common target in solid tumors, in glioblastoma are often too low for CAR-T to detect, while in ependymoma and breast cancer brain metastases, they can be so high that they overstimulate and exhaust CAR-T. Many targets, including HER2, are also found at low levels in vital organs like the lungs, raising the risk of toxicity.
Unlike chemical drugs that can be dosed precisely, CAR-T are living drugs whose activity is difficult to adjust once infused. This project aims to develop a “remote control” (RC) system that can fine tune CAR-T activity after infusion in brain cancer patients. Our RC system repurposes two FDA-approved drugs: danoprevir, to boost CAR-T activity when tumors are hard to detect, and grazoprevir, to reduce or completely halt CAR-T activity when overactivation or attacks on healthy tissues occur.
By enabling precise and remote tuning of CAR-T activity, this approach could improve both the efficacy and safety of treating brain cancers.
Michael Kilian, PhD
Institution: Brigham and Women’s Hospital
Mentor: Francisco Quintana, PhD
Tribute: Co-funded by StacheStrong
Glioblastoma (GBM) is an aggressive primary tumor of the central nervous system, with limited available treatment options due to the heterogeneity of tumor cells and their ability to evade the immune response. The meninges are the membrane layers that cover and protect the brain. The dura, the outermost of the three meningeal membranes, has recently been identified as important site for immune cell activation in the setting of neuroinflammation like multiple sclerosis. However, the contribution of the dura in the immune response against brain tumors, like GBM, is still unknown. This project aims at identifying anti-tumor immunity-promoting or inhibiting cell-cell interactions in recently identified immune cell clusters in the dura using a novel technology called RABID-Seq a barcoding method that allows to study cell communication in vivo, spatial sequencing technologies to study cells in a spatial context as well as functional studies and human GBM tissue. We hypothesize that the dura is a unique priming location for tumor-reactive T cells augmenting anti-tumor functions. The basic understanding of meningeal driven anti-tumor immune responses can potentially lead to novel treatment approaches like tissue-specific delivery of immunostimulatory factors or the genetic modifications of cellular tumor-targeting products like CAR-T cells.
Kavita Rawat, PhD
Institution: University of Pennsylvania
Mentor: Dolores Hambardzumyan, PhD
Tribute: In honor of Joel A. Gingras Jr.
Glioblastoma is the most common and aggressive type of brain cancer in adults. Glioblastoma is particularly hard difficult to treat, which leads to poor prognosis. For instance, with standard of care treatment, the median survival of glioblastoma patients is only 14.6 months. The immune system, which protects the body from infections and diseases, might also play a role in helping tumors grow as tumors are known to hijack the immune system for their own benefit. One of the key cells in the immune system is neutrophils, which are the body’s first line of defense against infections, helping to fight off bacteria and other harmful invaders. However, recent research shows that the aberrant activation of these cells might also support the growth and spread of cancer, including glioblastoma. In our study, we are investigating how neutrophils contribute to glioblastoma development and whether targeting these cells could help slow down or stop the glioblastoma progression. By understanding the relationship between neutrophils and glioblastoma, we hope to discover new ways to treat this challenging cancer and improve patient outcomes.
Amy Wisdom, MD, PhD
Institution: Massachusetts General Hospital
Mentor: Stefani Spranger, PhD
Tribute: Fully supported by Tap Cancer Out
Glioblastoma is the most aggressive type of brain cancer, with limited treatment options and poor survival rates. Unlike other cancers that respond to immunotherapy, glioblastomas suppress the immune system, making it difficult for the body to fight the tumor. My research focuses on understanding how glioblastomas evade immune detection by disrupting the function of key immune cells called dendritic cells, which are responsible for activating tumor-fighting T cells. I will use advanced mouse models to determine where and how these immune cells interact with glioblastomas, and how treatments like radiation and chemotherapy influence this process. By identifying the mechanisms that prevent the immune system from attacking glioblastoma, this study could reveal new ways to enhance immune-based therapies. Ultimately, this research aims to improve treatment strategies and offer new hope for patients battling this devastating disease.
Discovery Grants
A Discovery Grant is a one-year, $50,000 grant to support cutting-edge, innovative approaches that have the potential to change current diagnostic or treatment standards of care for either adult or pediatric brain tumors.
Nandini Acharya, PhD
Institution: The Ohio State University
Mentor: Monica Venere, PhD
Tribute: In honor of Charles “Chip” McKinley Greenlee
Glioblastoma (GBM) is the most aggressive brain cancer and has a median survival of only 12–15 months despite the standard of care. While immunotherapy has revolutionized cancer treatment, GBM remains highly resistant; thus, overcoming this resistance is crucial for improved patient outcomes.
Our research investigates an overlooked regulator of immune responses in GBM: nociceptors, the pain-sensing neurons. Though nociceptors have been shown to modulate immunity outside the central nervous system, their role in GBM remains unexplored. Our preliminary data show that nociceptors in GBM bearing mice are activated, and these tumor-activated nociceptors actively foster an immunosuppressive tumor microenvironment. Chemically disrupting nociceptors improve survival in GBM-bearing mice and enhance their response to immunotherapy. In this proposal, we will use single-nucleus RNA sequencing to map molecular changes in nociceptors and integrate this data with our previously generated single-cell RNA sequencing of immune cells from the meninges and tumor tissue of GBM-bearing mice. This will help reveal how GBM manipulates neuro-immune interactions to evade immune attacks.
These insights could lead to new treatment strategies, such as repurposing FDA-approved nociceptor-targeting drugs or identifying novel therapeutic targets to enhance immunotherapy. By disrupting the ability of GBM to hijack the nervous system, we aim to reshape the immune landscape, improve immunotherapy efficacy, and provide new hope for patients with GBM.
Mariella Filbin, MD, PhD
Institution: Dana-Farber Cancer Institute
Tribute: Co-funded by the Yuvaan Tiwari Foundation
Pediatric diffuse midline gliomas (DMG) are incurable brain cancers with a median survival of 9-12 months and no long-term survivors. To date, focal radiation remains the standard of care but improves survival by only a few months. Unfortunately, rapid radiation resistance ultimately arises in all DMG, resulting in lethal tumor recurrence in 100% of patients. Despite intense research efforts over the past four decades, there is still a lack of mechanistic understanding of the biology underlying DMG radiation resistance. While there is ample evidence that direct communication between normal brain cells (neurons) and cancer cells is critical to fuel growth of pediatric brain tumors; whether these important interactions contribute to treatment failure in DMG is currently not known. The innovative work outlined in this proposal will enable us to discover the biochemical mechanisms through which DMG cells exploit interactions with surrounding normal neurons to survive radiation-induced cell killing. If successful, these experiments have the potential to (i) completely transform our understanding of how resistance to radiation develops in DMG as well as other deadly brain tumors, and (ii) lead to the development of new therapeutic approaches to make radiation treatment more effective. Hence, this proposal addresses a critical need to understand the mechanisms underlying DMG radiation resistance and improve the efficacy of treatment for children diagnosed with DMG.
Luciano Garofano, PhD
Institution: University of Miami
Mentor: Antonio Iavarone, MD
Tribute: In memory of Brian Bouts
Glioblastoma (GBM) is an uncurable form of primary brain tumor with extremely poor prognosis. Despite current state-of-the-art therapy that includes surgery, irradiation and chemotherapy, all patients experience tumor progression, unfortunately. Numerous efforts have been made in the scientific community to design more appropriate therapeutic strategies. Some of these studies include computational approaches to sort tumor patients into subgroups of GBM, to understand the specific biological functions activated within these tumors and identify therapeutic targets. Single cell data allowed to discriminate tumor cells from the non-malignant cell components, and study how distinct tumor cell states interact within the tumor microenvironment. Moreover, proteomics analysis allowed for the discovery of kinases, proteins that specifically regulate tumor signaling, important for tumor growth and to repurpose current drugs for cancer therapy. Here, this study will examine the composition of the GBM ecosystem, dissect the biological mechanisms of GBM tumorigenesis and aggressiveness, and identify novel therapeutic opportunities for GBM. The development of innovative computational tools to understand the role of kinases in the reprogramming of GBM tumor-TME ecosystems is necessary to elucidate the mechanisms of therapeutic resistance.
Robyn Gartrell, MD
Institution: Johns Hopkins University School of Medicine
Tribute: Co-funded by StacheStrong
Atypical teratoid/rhabdoid tumors (AT/RT) are very tough brain tumors to treat. They’re aggressive and don’t respond well to chemotherapy, even at high doses. These treatments can make patients very sick and often they have to stay in the hospital. While radiation therapy can be used to target this tumor, patients with AT/RT are typically infants or toddlers under 3 years old and standard radiation at this age can cause delayed development. A new kind of radiation called FLASH has been shown to be safer for the brain than the usual type in tests on adult animals and even young healthy mice. FLASH has not yet been studied in very young mice in their first week of life. Since AT/RT mostly affects infants, it is essential to know how FLASH would affect this age group. Our study proposes to investigate how FLASH radiation therapy influences brain function and tumor control in AT/RT, aiming to shed light on a potential treatment approach for these challenging tumors while also preventing serious brain toxicity.
Filipe Pereira, PhD
Institution: Lund University
Tribute: Fully supported by an Anonymous Family Foundation
Cancer heterogeneity and the formation of an immunosuppressive tumor microenvironment (TME) are among the causes of failure of immunotherapy. Glioblastoma (GBM) is a highly heterogeneous tumor type, characterized by intra-tumoral and systemic immunosuppression.
Cellular reprogramming is emerging as a tumor-agnostic and immunosuppression-independent immunotherapy. My group has previously reprogrammed cancer cells into antigen-presenting conventional dendritic cells type 1 (cDC1). cDC1 are rare in tumor tissue but, even at low numbers, are sufficient to induce cancer cell death by the immune system. The current proposal aims to evaluate cDC1 reprogramming as a GBM immunotherapy. First, we will assess the feasibility of reprogramming GBM cells within the brain and characterize the immune mechanisms launched by cDC1-like cells in mouse models with different TME and immunogenicity profiles. Then, we will evaluate cDC1 reprogramming and associated immunogenicity in human patient-derived GBM organotypic cultures with an established immunosuppressive TME. Lastly, we will demonstrate the efficacy and safety of the cDC1 reprogramming approach in syngeneic models, validating it as a gene therapy for GBM. By connecting cellular reprogramming and immunotherapy in a novel gene therapy, our strategy has the potential to provide a personalized and effective treatment for GBM patients.
John Prensner, MD, PhD
Institution: University of Michigan
Mentor: Sriram Venneti, MD, PhD
Tribute: Co-funded by StacheStrong
Children diagnosed with diffuse midline glioma (DMG), including diffuse midline pontine glioma (DIPG), are faced with a dismal prognosis: over 90% of patients will die from disease within two years. Removal of these tumors by surgery is not possible, and so current treatment focuses on radiation therapy, which is not curative for patients. Our research has worked to unveil a new potential avenue for therapy for these children through investigation of the ‘dark’ proteome. This term refers to the thousands of small proteins generated by DMG/DIPG cells that have not been previously studied. We have pioneered cutting-edge methods to identify key cancer drivers from the dark proteome of medulloblastoma and other cancers. In this American Brain Tumor Association award, we will employ multiple patient-derived models of DMG/DIPG to define the dark proteome of associated with cancer gene mutations that cause DMG/DIPG in children. In addition, we will identify how radiation treatment – the standard therapy for children with DMG/DIPG – creates new opportunities for therapies based on the dark proteome. Overall, we expect this project to catalyze a new direction of research in DMG/DIPG by demonstrating the extent and relevance of the dark proteome, which may be used as a platform for future therapeutic developments.
Xueqin Sun, PhD
Institution: Sanford Burnham Prebys Medical Discovery Institute
Mentor: Charles Spruck, PhD
Tribute: Fully supported by an Anonymous Family Foundation
Glioblastoma (GBM) remains the most common and deadly type of primary brain cancer, leading to death of approximately half of all patients within just one year from the time of diagnosis and 95% within five years. We have discovered BRD8 protein as a novel and promising therapeutic candidate that, when targeted, could extend survival in about 71% of GBM cases—specifically those that still retain the tumor-suppressing protein p53. We further find that BRD8—the culprit for GBM aggressiveness, is kept at high levels by its partner protein, MRGBP. MRGBP shields BRD8 from being broken down by the proteasome—a machine that breaks down unneeded or erroneous proteins in the cell. This project aims to understand whether and how BRD8 is protected by MRGBP to block our body’s natural anti-tumor defense. We seek to answer this question using comprehensive methods from two aspects: 1) Is BRD8 protected by MRGBP from degradation; 2) Does BRD8 degradation restore the body’s natural anti-tumor ability? Successful completion of this innovative and timely project will provide insights into eradicating BRD8 using the proteasome; thereby, unleashes the intrinsic tumor suppression activity of p53 to halt GBM, paving the way for developing more effective therapies for people afflicted with this devastating malignancy.
Dong Wang, PhD
Institution: University of Colorado Denver
Mentor: Rajeev Vibhakar, MD, PhD
Tribute: Fully supported by an Anonymous Family Foundation
Medulloblastoma is the most common malignant brain tumor in children, and tumors driven by the MYC oncogene are particularly aggressive and resistant to standard treatments. We hypothesize that MYC, along with another key driver gene, OTX2, promotes tumor growth by increasing protein production, which relies on ribosomes—the structures in cells that build proteins. Our research has shown that a gene called CDK8 is essential for maintaining MYC and OTX2 activity and supporting ribosome production in these tumors. We will investigate whether blocking CDK8—either alone or in combination with other inhibitors that also regulate ribosome biogenesis—can slow tumor growth and improve treatment options for children with MYC-driven medulloblastoma. If successful, this work could lay the foundation for developing new therapies that may advance toward clinical trials.
Murat Yildirim, PhD
Institution: Cleveland Clinic
Mentor: Justin Lathia, PhD
Tribute: In memory of Kaitlyn Berg
Glioblastoma (GBM) is the deadliest form of brain cancer, with most patients surviving only about a year after diagnosis. In addition to rapid tumor growth, GBM disrupts brain function, causing memory loss, movement problems, and seizures. These symptoms often appear late in the disease, making early detection and intervention critical. Our project will explore how GBM affects brain activity and behavior from its earliest stages, with the goal of developing better diagnostic tools and treatment strategies. In Aim 1, we will use advanced brain imaging techniques to track real-time brain activity and chemical signaling in mice before and after tumor formation. By monitoring pupil size, facial movements, and walking patterns, we will train artificial intelligence models to recognize early warning signs of GBM-related brain dysfunction. In Aim 2, we will test whether light-based therapies (optogenetics) can restore brain function in mice with GBM. By stimulating inhibitory brain circuits, we aim to correct the harmful changes GBM causes in brain networks. If successful, this approach could pave the way for new, targeted therapies to improve brain function in GBM patients. By combining early detection strategies with innovative treatments, this research aims to slow disease progression and improve patient outcomes, offering hope for a more effective fight against GBM.
Jack and Fay Netchin Summer Research Grant
A Medical Student Summer Fellowship is a three month, mentor-guided summer research experience, intended to motivate talented medical students to pursue a career in neuro-oncology research.
Stephanie Bean, BS
Institution:University of Texas Health Science Center at Houston
Mentor: Ji Young Yoo, PhD
Tribute: Fully Supported by BrainUp
Glioblastoma (GBM) is a highly aggressive and deadly type of brain cancer with limited effective treatments. One promising new therapy uses a genetically engineered herpes virus, called oncolytic herpes simplex virus-1 (oHSV), which has been approved by the FDA to treat melanoma. While some GBM patients respond well to this therapy, only a small subset of GBM patients experience long-term survival benefits because many tumors are naturally resistant. This highlights the need for new strategies to reprogram tumor cells to become more susceptible to virus therapy. Our research has identified a tumor-suppressive molecule in cells called miR-433-3p. This molecule helps slow cancer growth and can push cancer cells into a state called “senescence”, where they stop growing and become more vulnerable. Our project will investigate how miR-433-3p-causes senescence and how it may enhance the effectiveness of virus therapy by promoting immunogenic senolysis, a process that helps the immune system recognize and eliminate tumor cells. The successful completion of our study could lead to new treatments that combine miR-433-3p with virus therapy, offering hope for better outcomes and longer survival for patients with GBM.
Darwin Kwok, PhD
Institution: University of California, San Francisco
Mentor: Hideho Okada, MD, PhD
Tribute: In Memory of Jameson Taylor, GBM warrior and medical student
Gliomas, a challenging type of brain tumor, are difficult to treat due to their low mutation rates and variability within and between patients. Immunotherapy, which uses the body’s immune system to target tumor-specific markers, has been limited in these cases. Our research focuses on “neojunctions” (NJs), abnormal RNA splicing (processing) patterns unique to cancer cells that are consistently present throughout a tumor. These NJs can serve as targets for immune cells, offering a new way to treat tumors. While we’ve previously identified NJs capable of activating immune cells to kill cancer, it remains unclear how standard glioma treatments—radiation, temozolomide (TMZ), and mutant IDH inhibitors—affect NJ expression.
Our study aims to identify NJs that persist or emerge after these treatments using advanced RNA sequencing techniques. We will also develop and test engineered T-cells designed to recognize and destroy tumor cells expressing these treatment-specific NJs. By combining traditional treatments with these innovative immunotherapies, we hope to provide more effective, targeted options for glioma patients, especially for those with recurrent or treatment-resistant tumors. This work has the potential to transform glioma treatment and significantly improve patient outcomes.
Thomas Lai, BS
Institution: University of California, Los Angeles
Mentor: Robert Prins, PhD
Tribute: In Honor of Debbi Schaubman
Glioblastoma (GBM) is an aggressive brain cancer with poor survival rates despite standard treatments like surgery, radiation, and chemotherapy. While immunotherapy—treatments that use the immune system to fight cancer—has revolutionized care for some cancers, its success in GBM has been limited. Recent findings suggest that high levels of Type I Interferon (IFN-I), a signaling molecule crucial for activating immune responses, may suppress the immune system in GBM, creating resistance to these therapies. Patients with elevated IFN-I activity in their blood often have worse outcomes and respond poorly to immune-based treatments.
This project seeks to develop a rapid blood test to predict which GBM patients are most likely to benefit from immunotherapy. Using samples from clinical trials, we will evaluate how immune cells react to IFN-I and whether this reaction can serve as a biomarker for treatment response. We will also analyze routine blood test data, such as white blood cell counts, to identify additional indicators of IFN-I activity.
Our goal is to create a rapid, low-cost diagnostic tool to guide personalized treatment strategies, ensuring that patients receive therapies most likely to benefit them. This research could transform GBM care by improving treatment outcomes and offering new hope for patients battling this devastating disease.
Vishva Natarajan, MS
Institution: Dartmouth-Hitchcock Clinic
Mentor: Jennifer Hong, MD
Tribute: In Memory of Kaitlyn Berg
Brain tumor-related epilepsy causes debilitating seizures and is the leading cause of disability in patients with brain tumors. These seizures are often difficult to treat and may even worsen tumor growth. Recent studies suggest that brain tumor-related epilepsy might be linked to damage in brain structures called perineuronal nets. Studying perineuronal nets in humans involves applying chemicals to visualize their presence in precious human brain tissue samples, a process that is currently expensive, time-consuming, and destructive to the tissue. Here we introduce VirtualPNN, an artificial intelligence model that will be capable of highlighting the presence of perineuronal nets in brain tissue images without physically processing the tissue itself. By saving human brain tumor tissue specimens and allowing scientists to prioritize certain samples for more advanced analyses, VirtualPNN will enable the rapid and efficient study of perineuronal nets and their role in brain tumor-related epilepsy. By being free to use and openly available, VirtualPNN will help democratize brain tumor research and inform the design of even more advanced tools for digitally visualizing perineuronal nets. Ultimately, VirtualPNN could be used to develop a tool that will help doctors rapidly predict seizure risks from minimally processed brain tumor biopsies, paving the way for new diagnostic methods that will guide clinicians’ approach to therapy and advance personalized care for patients.
Shree Pari, BS
Institution: Washington University School of Medicine in St. Louis
Mentor: Bhuvic Patel, MD
Tribute: In Honor of Paul Fabbri
Meningiomas are the most common type of brain tumor and cause significant disability and death each year. More aggressive forms of meningiomas—classified as WHO grade 2 and grade 3—are difficult to treat because they are more likely to recur after surgery, even when followed by standard radiation therapy. The primary goal for surgical resection of meningiomas is to completely remove the tumor tissue while preserving normal brain tissue. However, in many aggressive meningiomas, the tumor invades into the surrounding normal brain tissue, making total surgical resection nearly impossible. It is also difficult to properly diagnose tumors that have invaded normal brain tissue even with careful microscopic inspection of the tumor, as tumor invasion is not always apparent on the microscope slide. This project aims to deepen our understanding of how and why high grade meningiomas invade normal brain tissue, and to improve our ability to accurately diagnose brain invasive meningiomas. These insights will guide the development of new treatment strategies and lead to more effective, personalized care for patients with meningioma.
Kristen Park, BS
Institution: University of Pennsylvania
Mentor: Hongjun Song, PhD
Tribute: Fully Supported by Southeastern Brain Tumor Foundation
Glioblastoma (GBM), the most common primary brain tumor in adults, has a dire prognosis, with a median survival of less than 1.5 years and no significant advancements in therapy within the last two decades. The emerging field of cancer neuroscience reveals that the formation of direct connections, known as neural synapses, from non-cancerous brain cells onto GBM cells, plays a significant role in tumor growth and migration. These connections help the tumor grow and spread, but scientists still don’t fully understand how they affect the rest of the brain. Since GBM often develops in the brain’s outer layer (the cortex), it’s possible these tumor-neuron connections could disrupt brain function even in areas far from the tumor itself. I propose to extend previous work mapping neural inputs to cortically transplanted GBM, focusing on neuromodulatory brainstem areas that control processes like mood, sleep-wake cycles, and basic bodily function. The overall goal is to determine whether brainstem neurons and their associated circuitry are disrupted upon synapsing with tumor cells. Because GBM and brain activity can fuel each other in a vicious cycle, breaking any part of this loop, including hyperactivity of neural circuits, could not only improve neurocognitive symptoms but also limit tumor progression. Additionally, understanding the role of different neuromodulatory systems has the potential to reveal novel therapeutic targets. Ultimately, this research could change how we think about brain tumors, showing that they might cause symptoms in far-off brain areas without ever spreading there.
Gopi Patel, BS
Institution: University of Florida
Mentor: Jianping Huang, MD, PhD
Tribute: In Memory of Jim Topor
Glioblastoma (GBM) is one of the most aggressive and deadly brain tumors, with most patients surviving less than 15 months after diagnosis despite currently available treatment options. New therapies are urgently needed to improve outcomes for patients. Chimeric Antigen Receptor or CAR T-cell therapy, which uses a patient’s immune system to fight cancer, has worked well for blood cancers but has struggled to help patients with solid tumors like GBM because of challenges in delivering immune cells to the tumor and overcoming the tumor’s protective environment. To address these challenges, our team created a new therapy called 8R-70CAR. This therapy uses a signal produced by tumors, called IL-8, as a “GPS system” to guide immune cells directly to the tumor. It also targets a molecule called CD70, which is found in high levels on GBM tumors but not on normal cells, making it a safer and more effective treatment. In early preclinical studies, this approach completely eliminated tumors in multiple cancer models. This project is part of the first-ever clinical trial testing 8R-70CAR in patients with GBM. Early results show the therapy is safe and well-tolerated. As part of this study, I will analyze patient outcomes, such as tumor response, immune activity, and survival. The findings from this work may help inform the development of more effective treatments for GBM.
Christian Ramsoomair, BA
Institution: Miller School of Medicine of the University of Miami
Mentor: Danny Reinberg, PhD
Tribute: In Memory of Joe Smeeding
Diffuse midline glioma (DMG) is a devastating brain tumor in children that responds poorly to current treatments, including immunotherapy. DMG creates an environment in the brain that suppresses the immune system and keeps immune cells out, making it difficult for the body to fight the tumor. Researchers are developing new strategies to change this environment and make tumors more responsive to treatment by mimicking a viral infection in the tumor. Thus, this will “wake up” the immune system. Our research focuses on a novel approach to increase levels of a key molecule involved in immune activation — double stranded RNA (dsRNA). We discovered that an RNA-editing protein called ADAR is abnormally elevated in DMG, where it helps the tumor degrade immune-activating RNAs and evade immune responses. A drug called all-trans retinoic acid (ATRA) has shown promise in inhibiting ADAR, thereby making tumors more likely to respond to immunotherapy. In early experiments using patient-derived DMG cells, ATRA not only degraded ADAR but also slowed tumor growth. This project will investigate how ATRA works in DMG and whether it can trigger immune responses that, when combined with immunotherapy, can effectively target the tumor. This research has the potential to provide hope and lead to safer, more effective treatments for DMG patients.
Robert Ruzic, MS
Institution: SUNY at Stony Brook
Mentor: Stella Tsirka, PhD
Tribute: In Memory of Rose DiGangi
Glioblastoma (GBM) is a highly aggressive type of brain tumor that is notoriously difficult to treat. Despite current treatments such as surgery, radiation, and chemotherapy, the tumor almost always recurs, posing significant challenges for further treatment. Patients diagnosed with GBM typically have a short life expectancy, with most surviving less than two years, and only a small percentage living longer. This grim prognosis is attributed to the ability of GBM cells to spread throughout the brain, resist treatment, and thrive in the tumor microenvironment. One mechanism through which these cells survive and proliferate is autophagy, a process where they recycle cellular components for energy. Led by Dr. Tsirka, scientists have been investigating a drug called lucanthone, which appears to inhibit autophagy and other survival mechanisms in GBM cells, including those resistant to standard treatments. The goal of this project is to determine whether lucanthone can effectively eradicate GBM cells obtained from patients’ tumors removed during surgery. These cells will be sourced from both newly diagnosed tumors and those that have recurred following treatment with drugs like temozolomide. This research holds promise for identifying novel approaches to treating this challenging cancer, which remains an area of significant unmet medical need.
James Trippett, MBE
Institution: University of California, San Francisco
Mentor: Manish Aghi, MD, PhD
Tribute: In Memory of Kaitlyn Berg
Glioblastoma (GBM) is an aggressive brain cancer known for its ability to grow quickly and resist treatment. Our project explores how to boost the body’s own immune system to fight GBM by examining two sources of immune cells: those traveling through the blood-brain barrier (BBB) and those originating in the skull bone marrow, which can reach the brain through special channels. We previously found that skull bone marrow cells can attack GBM when they are released into the tumor environment. However, their impact is limited if they cannot recruit enough T-cells, key fighters of the immune system, to help destroy cancer cells. To overcome this barrier, we will use focused ultrasound (FUS) to temporarily open the BBB, allowing more T-cells to enter the tumor. We will then combine this approach with a skull-injected medication (AMD3100) that frees more myeloid immune cells from the skull marrow. By studying how well these two strategies work together, we hope to discover a more effective way to get immune cells into the tumor, ultimately improving the body’s ability to control or eliminate GBM. If successful, this approach could point to new, more powerful immunotherapies for patients with brain cancer.
Alexander Wang, BS
Institution: Case Western Reserve University – School of Medicine
Mentor: Tyler Miller, MD, PhD
Tribute: Full Supported by the Gladiator Project
Dexamethasone (Dex) is a steroid that is used to reduce brain swelling in patients with brain tumors. While effective in managing symptoms, Dex weakens the immune system, increasing infection risk and impairing responses to immunotherapies, which use the body’s own immune system to attack the tumor and may be our best shot at curing patients with glioma. Specifically, we have found that Dex creates a population of highly immunosuppressive immune cells called myeloid-derived suppressor cells, which have been associated with resistance to immunotherapy and worse overall survival. We have found that induction of immunosuppressive myeloid cells by Dex persists long after discontinuation, raising questions about its impact on immunotherapy trials often conducted shortly after patients are weaned off the drug. This finding has motivated us to understand how long it takes for the immune system to recover sufficiently before they can effectively respond to immunotherapies, particularly for aggressive cancers like GBM. Thus, balancing Dex’s symptom-relieving benefits with its detrimental effects on immunotherapy is critical in managing patients where immunotherapy could be indicated, especially as most brain tumor patients receive Dex before and after surgery. By elucidating Dex’s role in immunosuppression, this research aims to play a role in optimizing the overall design of immunotherapy trials – ultimately enhancing survival and quality of life for patients with these devastating diseases.
Grants Awarded in Collaboration with Other Organizations
Metastatic Brain Tumor Collaborative CNS Metastasis Research Grants
CNS Metastasis Research Grants provide $50,000 over one-year to support researchers to conduct metastatic CNS tumors or leptomeningeal disease-focused projects that are applicable to at least two different primary cancers
Brain Tumor Funders Collaborative
In 2025 the Brain Tumor funders Collaborative (BTFC) awarded two $500,000 grants to support projects focused on Liquid Biopsy for Primary Brain Tumors