fbpx

GBM Awareness Day is July 17. Join us in raising awareness all month long! Learn more.

Research Funding & Impact

The ABTA has contributed over $36.5 million to brain tumor research since we began funding research in 1974.

2024 ABTA-Funded Research Projects

Guided by our mission, and advised by a multidisciplinary team of experts, the ABTA is dedicated to funding research that has the potential to change the lives of people affected by brain tumors. We also aim to “seed the field” with promising up-and-coming researchers in the brain tumor space.

The ABTA awarded $1,083,000 in grants to 24 new projects

The 2024 funded projects span multiple tumor types

  • Glioblastomas
  • Astrocytomas & Oligodendrogliomas
  • Meningiomas
  • Brain Metastasis
  • Choroid Plexus Carcinoma
  • Brain Stem Gliomas

Research areas of focus

  • Immunology/Immunotherapy
  • Drug Therapies/Experimental Design
  • Epigenetics 
  • DNA Damage/Repair Mechanisms
  • Biomarkers
  • Invasion/Motility

2024 Newly Awarded Grants

Research Collaboration Grants

The Research Collaboration Grant is a two-year, $200,000 grant, to support interdisciplinary team science projects that combine resources to streamline accelerate progress in the brain tumor field.

Jacques Lux, PhD

Jacques Lux, PhD

Co-PI: Wen Jiang, MD, PhD

Institution: University of Texas Southwestern Medical Center

Co-PI Institution: University of Texas M.D. Anderson Cancer Center

Tribute: In honor of Joel A. Gingras

Project: Engineering a Targeted, Acoustically Responsive Image-Guided GBM Immunotherapy

Co-PI: Wen Jiang, MD, PhD Co-PI: Wen Jiang, MD, PhD

While immunotherapies such as immune checkpoint blockade have revolutionized cancer treatments, they have not been successful clinically in extending the survival of patients with glioblastoma multiforme (GBM). This project aims to address this problem by developing a novel and clinically relevant ultrasound-guided strategy to boost the patient’s immune response against GBM. We have developed a new technology that we termed MUSIC (Microbubble-assisted Ultrasound-guided Immunotherapy of Cancer) to deliver a naturally occurring immune-boosting molecule into immune cells. We propose to further improve a second generation dual functional MUSIC (dMUSIC) system that will generate tumor-recognizing immune cells to produce durable antitumor immunity against GBM. We have reengineered microbubbles (MBs), which have been clinically used as ultrasound contrast agents for more than two decades, to carry and deliver both drug and tumor antigens to immune cells. Upon ultrasound exposure, MBs implode to open temporary holes into the targeted cells and deliver their cargo. Therefore, both drug and antigen are delivered when and where we want it by applying ultrasound on the brain tumor. As MBs are clinically approved with excellent safety profiles and ultrasound scanners are ubiquitous in hospitals, the results generated could help rapidly translate this image-guided cancer immunotherapy strategy into the clinic. 

Pavithra Viswanath, PhD

Pavithra Viswanath, PhD

Co-PI: Peng Zhang, PhD

Institution: University of California, San Francisco

Co-PI Institution: Northwestern University

Tribute: Partially supported by BrainUp

Project: Targeting and Imaging Immunosuppressive Metabolic Alterations in Glioblastomas
Co-PI: Peng Zhang, PhD
Co-PI: Peng Zhang, PhD

Co-PI: Wen Jiang, MD, PhD

Glioblastoma is the deadliest form of brain cancer in adults. Tumor-associated macrophages (TAMs) are natural immune cells within the tumor that actively promote tumor growth and immune evasion. Our studies indicate that, unlike normal macrophages, TAMs consume glucose to produce lactate, which alters gene expression and encourages tumor growth. Lactate then needs to be exported out of the cell to prevent an increase in acidity within the cell. We show that blocking lactate export by targeting a molecule called SLC16A3 that removes lactate, eliminates immunosuppressive tumor-associated macrophages and makes tumors more vulnerable to immunotherapy. We also find that a novel imaging technique called deuterium metabolic imaging allows us to tell whether the tumor is responding to therapy within days of treatment. Based on these findings, we will determine if SLC16A3 is an promising immunometabolic target and if deuterium metabolic imaging can be used to track treatment response in mouse models of glioblastoma. Our studies will lay the foundation for translation of this innovative therapeutic and imaging strategy to glioblastoma patients.  

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.

Adam Grippin, MD, PhD

Institution: University of Texas M.D. Anderson Cancer Center

Mentors: Wen Jiang, MD, PhD

Tribute: In memory of Stephanie Lee Kramer

Project: In Situ Generation of GD20 CAR Macrophages to Treat K3K27M Diffuse Midline Glioma

Diffuse intrinsic pontine glioma is a uniformly fatal form of brain cancer that is resistant to all current therapies. Here, we propose a novel technique that involves transforming a type of immune cell, called macrophages, from their usual role of suppressing immune responses into aggressive cancer cell hunters. By combining the powers of nanotechnology, mRNA therapeutics, and immunotherapy, we aim to reprogram these cells directly within the body using specially engineered microscopic vesicles, which are small cellular sacs, carrying the mRNA instructions needed to destroy tumors. Unlike traditional treatments, this innovative strategy holds great promise for a broader and more lasting defense against cancer by also helping the body’s immune system recognize and respond to cancer more effectively. 

Mark Youngblood, MD, PhD

Mark Youngblood, MD, PhD

Institution: Northwestern University

Mentor: Adam Sonabend, MD

Tribute: Fully supported by Tap Cancer Out

Project: Longitudinal Characterization of Glioblastoma Exosomes During Immunotherapy

Glioblastoma is the most deadly form of brain cancer in adults, and is in need of innovative new approaches to improve patient outcomes. Minimally-invasive blood tests could provide doctors with important details about how a glioblastoma changes over time in response to therapy, ensuring that the best treatments are utilized at the right time. Immunotherapy, which harnesses the power of the human immune system to fight glioblastoma, offers a powerful new approach in treating these tumors. This project will investigate a blood-based test that tracks a patient’s response to immunotherapy in glioblastoma, aiming to identify those who will benefit from use of this treatment method. If successful, our tool will shift how therapeutic decisions are made in patients, and improve outcomes by matching each glioblastoma with the most effective treatment type.  

Discovery Grant

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.

Toshio Hara, PhD

Institution: University of Michigan

Mentor: Pedro Lowenstein, MD, PhD

Tribute: In memory of George Surgent

Project: Understanding Glioblastoma Invasion Using Single-Cell Spatial Analyses

Glioblastoma is the most common and lethal form of intracranial tumor. It accounts for approximately 48% of the 24,500 new cases of malignant primary brain tumors diagnosed each year in children and adults in the United States. Glioblastoma is capable of infiltrating the brain, and missing even a few cells leads to high recurrence after surgical resection. There is a major gap in our knowledge as to what is the molecular identity of such invading tumor cells. To overcome this gap in our knowledge, we use a technology called single-cell RNA sequencing to measure gene expression of individual tumor cells that invade and hide in the brain. Evidence from this approach reveals the ability of tumor cells to switch their gene expression identities, by turning them on and off, along with their location. We hypothesize that the signals sent out from their surrounding environment can be recognized by invading tumor cells and determine their identities and functions. By controlling such environmental signals, the goal of this proposal is to perturb gene expression of glioblastoma cells to disrupt their abnormal behavior. Our proposed works hope to have a major impact in providing new strategies to control invasiveness, which could lead to significantly increased recurrence-free survival of glioblastoma patients–things that are not possible with the current treatment. 

Tanner Johanns, MD, PhD

Tanner Johanns, MD, PhD

Institution: Washington University in St. Louis

Tribute: Fully supported by an Anonymous Family Foundation

Project: Validating Novel Models to Study GZMK T cells in Brain Tumors

Glioblastoma is the most common and deadliest brain tumor in adults; therefore new, effective treatment options are needed to improve outcomes for patients diagnosed with this disease. Immunotherapies are treatments that aim to improve the patient’s own immune response against cancer cells and offer many ways in which the immune system can be activated. However, immunotherapies to date have not been effective in patients with glioblastoma, and it is not clear why. We have recently observed that the most common cancer-fighting immune cell, called a CD8 T cell, expresses an enzyme called granzyme K. Not much is known about granzyme K or the CD8 T cells that express granzyme K, but the high prevalence of these cells suggests they may play an important role. Unfortunately, there are no mouse models available that allow us to perform in-depth studies to learn more about how these granzyme K cells develop, what role granzyme K or granzyme K-expressing cells play in protection against glioblastoma, or how these cells can be targeted to improve outcomes for patients with brain tumors. Successful completion of this project will result in the generation of two new mouse models that can be used to address these key questions. 

Chrystian Janqueira Alves, PhD

Chrystian Junqueira Alves, PhD

Institution: Icahn School of Medicine at Mount Sinari

Tribute: In honor of Charles “Chip” McKinley Greenlee

Project: Manipulating Membrane Surface Charges to Curb Glioblastoma Invasiveness

Glioblastoma (GBM), the most aggressive brain tumor, is notorious for wide infiltration in the brain, making complete surgical resection impossible. Traditionally, scientists have focused on understanding the molecular factors that drive GBM migration, but how tumor cells navigate physical barriers such as confined space in the brain is not well understood. Using cutting-edge microdevices that mimic the narrow passages in the brain, we can obtain live images of how GBM cells navigate through tight spaces. Remarkably, we observed that GBM cells like to migrate through tight constrictions, illustrating the astounding ability of GBM cells to respond and adapt to physical surroundings. Here, I will dive deeper into the biomechanical flexibility behind this confined migration, by focusing on two key aspects: i) changes in the electric charge of inner membrane surface that organize the internal framework of the cells; ii) regulation of this charge by Plexin-B2, a molecule established as modulator of cell biomechanics that is usurped by GBM cells to gain invasiveness . Functional blockade of Plexin-B2 with nanobodies will be tested to develop novel translational strategies to curb GBM invasion. 

Gilbert Rahme, PhD

Gilbert Rahme, PhD

Institution: SUNY at Stony Brook

Mentor: Styliani-Anna E. Tsirka, PhD

Tribute: Fully supported by The Karl Schmidt Oligodendroglioma Research Fund

Project: Dissecting Epigenetic Lesions that Drive IDH-mutant Gliomas

Diffuse gliomas are serious brain tumors with no cure. To better treat patients with diffuse gliomas, we need to understand how these tumors grow. Many diffuse gliomas start because of mutations, or changes, in a gene called IDH1. These mutations can damage special parts of the DNA structure called CTCF sites. CTCF sites act like guards, deciding how different bits of the DNA interact with each other. We’ve already found one damaged CTCF guard near a gene called PDGFRA, which is linked to tumor growth. Now, we’re looking at damaged guards near other genes to see how they might help tumors grow. Understanding how damaged CTCF guards affect tumor growth could help us find new ways to treat gliomas. 

David Raleigh, MD, PhD

Institution: University of California, San Francisco

Tribute: Fully supported by An Anonymous Family Foundation

Project: Mechanistic and Functional Validation of Meningioma Radiotherapy Response Genes

The most common brain tumor grows from the lining that surrounds the brain and is called a meningioma. These tumors are often slow growing and can often be cured with surgery, but many meningiomas require additional treatment with radiotherapy to block or slow their growth. There are no effective medical therapies to treat meningiomas that are resistant to surgery and radiotherapy. This project will study how genes influence meningioma responses to radiotherapy. Using what we have learned from studying genes in meningiomas from patients and genes in meningioma cells in our lab, we hope to find drug targets that could be used to improve treatment options and clinical outcomes for patients with meningiomas. Our studies will make use of both conventional and cutting-edge techniques that will allow us to study how each gene influences meningioma responses to radiotherapy in individual cells. These technical advances that we developed in our lab will facilitate previously impossible investigations of many dozens of genes at once, which will significantly increase our bandwidth and allow us to complete this high-risk project in the allotted year of funding. Ultimately, we hope to test the genes that are most important for radiotherapy responses in cells in this project as part of mouse studies in future projects, and then in human patients in clinical trials. 

Artak Tovmasyan, PhD

Artak Tovmasyan, PhD

Institution: Ivy Brain Tumor Center, Barrow Neurological Institute

Mentor: Nader Sanai, MD

Tribute: In memory of Brian Bouts

Project: Enhancing Radiotherapy Efficacy by Modulating Glioblastoma's Redox Environment

Glioblastoma (GBM) is the most aggressive primary malignant brain tumor, with a median survival at diagnosis of 12-16 months. The current standard treatment involves maximal surgical resection followed by radiotherapy, which delays tumor progression and extends patient survival. However, the tumor always re-emerges aggressively, leading to patient death. Novel therapeutic approaches are urgently needed to enhance the tumor response to radiotherapy. Yet, developing such agents poses significant challenges, notably due to their limited ability to cross the blood-brain barrier and selectively sensitize brain tumors to radiation while sparing normal tissue. In this proposal, a novel molecule, MnP3, is proposed as a potential radio-sensitizer aimed at improving the survival outcomes for GBM patients. MnP3 is designed to overcome previous concerns regarding brain penetration and toxicity associated with earlier generation analogs by modifying specific molecular features responsible for adverse effects. MnP3 possesses significant brain targeting and improved safety profile. Our initial findings demonstrate that MnP3 enhances the sensitivity of brain tumors to radiation in animal models while protecting normal brain from radiation-induced side effects. Through this application, we intend to validate these promising preliminary results and investigate the role of the immune system in the therapeutic benefit offered by MnP3. Successful completion of this project will warrant further clinical development of MnP3 for GBM patients. 

Dionysios Watson, MD, PhD

Dionysios Watson, MD, PhD

Institution: Sylvester Comprehensive Cancer Center/University of Miami

Mentor: Antonio Iavarone, MD

Tribute: Fully supported by an Anonymous Family Foundation

Project: Acquisition of Glioblastoma Mitochondria Reprograms Tumor-Associated Astrocytes

Glioblastoma (GBM) is the most common primary malignant brain tumor. Despite standard therapy, it invariably recurs and becomes resistant to treatment with a long-term survival of <7%. There is growing appreciation that as GBM grows, it integrates into the brain. The resulting interactions with brain cells promotes the growth, persistence, and recurrence of these tumors. However, there remains limited understanding of the mechanisms of communication between GBM and the brain. Given the universal recurrence of GBM after standard therapy, there is an urgent need to elucidate these interactions to identify novel drug targets. We previously published that GBM cells form physical connections with astrocytes, the most abundant cells that support brain function. Through these connections, cancer cells acquire astrocyte mitochondria, sub-compartments of cells which are responsible for energy production and regulation of multiple cellular processes. Astrocyte-to-GBM mitochondria transfer resulted in reprogramming of metabolism and more aggressive tumors in preclinical models of GBM. In this proposal, we interrogate the effect of mitochondria transfer on astrocyte biology, an understudied aspect of this process. We propose this as a mechanism whereby GBM in turn reprograms its microenvironment to facilitate tumor growth and persistence. Understanding this poorly studied communication pathway will identify novel therapeutic opportunities for this devastating disease. 

Co-PI: Peng Zhang, PhD

Peng Zhang, PhD

Institution: Northwestern University 

Mentor: Maciej Lesniak, MD

Tribute: Fully supported by BrainUp

Project: Immunostimulating mRNA Therapeutics to Overcome Therapy Resistance in Gliomas

Therapy resistance is a major challenge in treating malignant brain tumors. Loss of tumor antigen, a peptide “tag” on tumor cell surface that could be recognized by immune cells, is a strategy tumor uses to escape from being attacked by immune system, causing failure of immune response and therapy. To better understand this, we have created a brain tumor model that is highly resistant to the clinically used standard treatments, such as radiotherapy and chemotherapy. We found that these resistant tumor cells have much lower “tumor tag” level than normal tumor cells. To overcome this, we propose to develop a new mRNA therapy to enhance the tumor antigen (“tag”) presentation, making these tumor cells more visible to immune cells for a better tumor killing. To do so, we have developed a lipid nanoparticle, a fat-like sac, to protect and deliver the mRNA molecules specifically to brain tumors. By testing a library of lipid nanoparticles made by different FDA-approved lipid components, we have identified the best formulation with the highest efficiency for targeting glioma cells. As mRNA-based drug and lipid nanoparticle technology have been proved to be feasible and successful in the development of COVID-19 vaccines during the pandemic, a rapid translation of our strategy to clinical usage is anticipated. Our work may develop a new therapeutic approach to boost the body’s immune system to attack brain tumors and improve the current clinical treatments for brain tumor patients. 

Julie Miller

Julie Miller, MD, PhD

Institution: Massachusetts General Hospital

Tribute: Fully supported by An Anonymous Family Foundation

Targeting Methionine Dependence in IDH-mutant Glioma

Isocitrate dehydrogenase (IDH) mutant gliomas are primary brain tumors that typically affect young and middle-aged adults. Despite treatment with radiation and chemotherapy, these tumors inevitably return, at which point effective therapies are limited. We are interested in discovering novel ways to attack IDH-mutant gliomas that are more effective and cause fewer side effects than current treatments. IDH is an important enzyme involved in metabolism, the biochemical reactions used by cells to create energy. Mutations in IDH cause changes in the way cells use nutrients and metabolites, leading to tumor formation. We believe that understanding how IDH mutations impact glioma metabolism will shed light on potential ways to take advantage of these metabolic changes to develop improved treatment strategies. In the laboratory, we recently discovered that restriction of the amino acid methionine is detrimental to IDH-mutant glioma growth. Interestingly, methionine restriction does not harm non-cancerous cells. The goal of this project is to examine methionine restriction in more detail to understand the metabolic process that are dependent on ample levels of methionine and to test if a low methionine diet can slow IDH-mutant glioma growth. By using tumor models derived from patients with gliomas, we hope our research will uncover new treatment options for IDH-mutant glioma, to eventually be tested in clinical trials.

Jack & Fay Netchin Medical Student Summer Fellowship

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. 

Lucy Chen, AB

Lucy Chen, AB

Institution: Johns Hopkins University School of Medicine

Mentor: Karisa Schreck, MD, PhD

Tribute: In honor of Paul Fabbri

Project: Identifying a Transcriptional Signature for MAPK-dependent BRAF-altered Gliomas

Gliomas are a group of tumors that come from the supporting cells in the brain. They can be deadly cancers, and most gliomas are incurable. Some can be treated with drugs targeted against certain cancer-causing mutations. One such mutation found in gliomas is in the protein BRAF. This mutation causes the protein to send signals telling cells to grow and divide out of control. Currently, there are several FDA-approved drugs that target mutated BRAF and how it affects other cells. However, cancer cells eventually become resistant to these targeted therapies and continue to grow. 
 
Previous studies have found gliomas become resistant to BRAF-targeted therapies by changing their expression of specific genes. However, no studies have been able to find a group of genes whose use can predict whether a glioma will be resistant. Here, we propose a project to identify specific gene sets that will indicate what outcome a patient will have during treatment. We will use a large set of patient-derived data from Caris Life Sciences, a tumor genetics company. Next, we will test this group of genes on data from 20 patients to see if they can predict resistance. If successful, these specific genes identified in this study could help doctors choose the best treatment for patients with BRAF mutations.  

Harrsha Congivaram, BS

Harrsha Congivaram, BS

Institution: Northwestern University Feinberg School of Medicine 

Mentor: Feng Yue, PhD 

Tribute: Fully supported by BrainUp

Project: Classifying the Effects of Methylation Signatures in Meningiomas

Meningiomas are the most common brain tumors and are generally considered benign. However, there are cases where meningiomas can become malignant and can possibly grow back, an event known as tumor recurrence. Currently, meningiomas are classified into three sub-groups with different degrees of risk for malignancy and recurrence, based on microscope cellular imaging. While the classification system has improved clinical decision-making, a significant number of cases that are classified as low-risk can recur. Recent classification efforts based by DNA methylation, an adjustment to the DNA that can inactivate a gene, showed improved predictive capabilities to demonstrate which tumors will most likely recur. However, these algorithms were developed using different DNA methylation platforms and methodologies, making it challenging to adopt such approaches in a clinical setting. Therefore, we propose to compare existing methylation datasets to develop a universal platform that can classify meningiomas regardless of the method used to collect DNA methylation profile. Such an algorithm would allow for wide clinical usage of methylation-based classification, improving clinical decision-making and patient outcomes. We plan to make this classifier available to clinicians around through an interactive web app. Furthermore, we will assess whether these subtypes have unique methylation patterns that can inform our understanding of biological pathways that drive tumor growth. 

Karenna Groff, MEng

Institution: New York University Grossman School of Medicine

Mentor: Dimitris Placantonakis, MD, PhD

Tribute: Fully Supported by the Southeastern Brain Tumor Foundation

Project: Optimization of a CD97-Targeting Antibody-Drug Conjugate for Treatment of Glioblastoma

Glioblastoma (GBM) is the most common and lethal brain malignancy, with a median survival of only 14-16 months after current treatment options. Recent attempts to control tumors with various forms of immunotherapy have also failed. The reason behind GBM’s resistance to new therapies is complex and not fully understood. However, GBM stem-like cells (GSCs) have been shown to play a fundamental role in treatment resistance. Based upon this insight, we identified a therapeutic target: a cell surface receptor, called CD97, that mediates the ability of cells to make more GSCs, and is present in GBM cells but not in healthy brain tissue. We have developed a drug to target this receptor and selectively kill GBM cells. By directly injecting this drug into the tumors of mice with GBM, we demonstrate slower tumor growth and prolonged survival. During my summer fellowship, I will evaluate whether multiple doses of the drug further improve survival in mice compared to a single dose. I will also assess whether the drug synergizes with the current standard of care chemoradiotherapy. Our initial results suggest that this drug may offer an effective therapeutic option for GBM. The results of this study will determine the most effective delivery strategy for the drug, as well as whether it can be administered alongside the current standard of care: two critical steps in transitioning this work from the lab bench to the bedside.  

Joseph Inger, BS

Institution: Brown University

Mentor: Sean Lawler, PhD

Tribute: Fully supported by the Gladiator Project

Project: mGluR3 Modulation as a Combination Therapy

Glioblastoma, a particularly vicious brain cancer, is sadly incurable. While current treatments, such as temozolomide and tumor resection, provide some help, they ultimately fall short. This study investigates a promising new approach that could change treatment for glioblastoma patients. The key player in this study is VU6010572, an investigational medication originally developed for mental health conditions such as Major Depression. What makes this compound special is its ability to easily cross into the brain and decrease the activity of a signaling protein called mGluR3, without effecting any other proteins. We believe the increased activity of this protein plays a crucial role in glioblastoma’s ability to resist temozolomide, making it a great target and modifiable by VU6010572. The combination of these two drugs could be significantly more effective than either one alone. We will conduct experiments to see how well VU6010572 and temozolomide or other cancer medications work together against glioblastoma cells. If successful, this study could lead to a whole new way of treating glioblastoma and other brain tumors. Repurposing an existing investigational medication that could more easily be brought to the clinic, offers a glimmer of hope for patients battling this aggressive cancer. This research could pave the way for more effective treatment options, giving patients a fighting chance against this challenging disease.

Jill Jones

Jill Jones, BS

Institution: Boston Children’s Hospital

Mentor: Maria Lehtinen, PhD

Tribute: In memory of Rose Digangi

Project: Illuminating Choroid Plexus Carcinoma Vascularization in Real Time

Choroid plexus carcinoma (CPC) is a rare, aggressive brain cancer arising from the choroid plexus (ChP), a highly complex tissue within the brain’s ventricles that produces cerebrospinal fluid. CPC is often diagnosed in very young children, and the only hope for survival is usually total removal of the tumor through surgery. However, this surgery is often complicated due to large tumor size, invasion of nearby brain tissue, and complicated networks of blood vessels in CPC that bleed significantly in the operating room. Our group is interested in investigating mechanisms behind the elaborate vascular biology in these tumors. Using patient samples, our laboratory’s established mouse model of CPC, an array of molecular biology techniques, and high-resolution microscopy approaches, we aim to characterize vascular development in CPC at the micro (molecular) and macro (whole-animal) levels, combining cellular studies with novel imaging technology that can accurately track blood vessel development over time. Understanding how CPC vasculature differs from healthy ChP in this way should identify critical periods in the fast developmental time course of this disease (and hopefully other brain tumors) and reveal new targets for chemotherapy to “normalize” blood vessels before tumor surgery, enabling more young patients to undergo cure of this horrible cancer. 

Seth Meade, BSE

Seth Meade, BSE

Institution: Cleveland Clinic

Mentor: Jennifer Yu, MD, PhD

Tribute: In honor of Debbi Schaubman

Project: Prospective Surgical Study of High-Grade Glioma Electrical Activity & Progression

Glioblastoma (GBM) is the most aggressive and most common primary adult brain cancer. However, therapies to combat these diseases are limited. Current standard-of-care (SOC) treatment of surgery, radiotherapy, tumor treating fields, and chemotherapy has limited effectiveness leading to poor prognosis for many glioma patients (median survival of ~15-17 months). Neurosurgeons’ major concern when treating gliomas is the rapid growth of these tumors bother before and after they are removed. Small GBM stem cells within the brain surrounding the tumor are thought to drive this rapid growth and return of the tumor after resection. The inability of current treatment to prevent tumor growth highlights the need to better understand how GBM growth occurs. Recently, it was discovered that similar to how different brain regions can exchange electrical signals to communicate, GBM can electrically signal to the brain in a way that can drive tumor growth. Others have found inflammation can alter electrical activity of both gliomas and the brain. We are performing a novel surgical trial to measure electrical activity and inflammation across different areas along the surface of GBM tumors to better understand how electrical activity and inflammation affect communication between glioma and the brain driving the growth of GBM. This work will help inform future work to identify novel drugs and therapies that may hold promise to reduce tumor growth. 

Jonathan Mitchell, BS

Institution:

Mentor: Defne Bayik, PhD 

Tribute: Fully supported by BrainUp

Project: Evaluating the Role of Complement Protein 1a in Glioblastoma Immunity

Glioblastoma (GBM) remains an aggressive and highly challenging cancer to treat. Traditional therapies which focus on killing tumor cells have not greatly improved patient outcomes in the last decade. Activating T cells in the immune system has been an effective strategy to fight other cancers, but recruitment of tumor suppressive immune cells like tumor-associated macrophages (TAMs) and other myeloid cells by GBM protects tumors and facilitates growth. Our lab has discovered that some immune cells can promote tumors differently in males and females. Understanding more about how TAMs communicate with tumor cells in a sex-dependent manner is critical to developing new treatments capable of reversing this immunosuppressive environment. A specific protein, C1q, is produced by TAMs and has been linked to poor prognosis in GBM. We have shown that C1q is elevated in female models of GBM and that eliminating C1q prolongs survival in female mice. This proposed research will identify the role of C1q in immunosuppression in GBM in females by studying how it influences the tumor-protective behavior of TAMs. Additionally, this work will increase understanding of tumor-immune cell communication in a sex-dependent context. Ultimately, these results may identify C1q as a viable target for the development of new therapies that could be combined with existing immunotherapies to improve treatment response in GBM and increase survival in patients.   

Megan Parker, BS

Megan Parker, BS

Institution:

Mentor: Chetan Bettegowda, MD, PhD 

Tribute: In honor of Debbi Schaubman

Project: Detection and Characterization of Primary Brain Cancer by Flow Cytometry

Gliomas are the most common type of malignant tumors that originate in the brain. To diagnose and characterize gliomas, pathologists examine biopsies of these tumors under the microscope. While this is currently the gold standard of diagnosis, this method is limited by a high degree of subjectivity. Accurate characterization of gliomas is imperative for guiding therapy. Incomplete or inaccurate diagnoses can result in patients requiring a second biopsy or delays in treatment. 
 
Flow cytometry is a laboratory technique used to detect and measure characteristics of cells. This technique is well-established in the diagnosis of blood cancers. It has advantages over microscopic evaluation, including the ability to analyze more cells than can be observed under the microscope and the ability to objectively quantify key molecular factors that drive the tumor’s growth and behavior. Despite these advantages, a robust method to detect and characterize gliomas using flow cytometry has not yet been developed. 
 
In this project, we aim to develop such a method. We will assess if this method can add value to microscopy-based evaluation, especially for nondiagnostic brain biopsies. The successful completion of this project will offer an objective, comprehensive, and mechanistically-based method for characterizing brain biopsy specimens. That information, in turn, will enable clinicians to make more appropriate treatment decisions for each patient. 

Jorge Salcedo, BS

Jorge Salcedo, BS

Institution:

Mentor: Robert Prins, PhD 

Project: Role of cDCs in Melanoma Brain Metastases Treated with Immune Checkpoint Blockade

Melanoma brain metastases (MBM) continue to be a death sentence for those diagnosed with them. Upon diagnosis with metastatic melanoma, between 20-40% of patients will have metastases in their brain, and over 50% will develop MBM at some time in their disease course. Existing treatments like surgery and radiation yield modest outcomes, underscoring the need for more effective therapies. Despite advancements in immune checkpoint blockade (ICB) therapies allowing median survival to cross the 1-year mark, they remain bridled by patterns of resistance we have yet to understand. Our study aims to unravel the intricate dynamics of the immune system’s response to MBM, including identifying the key cells involved in activating the immune system in MBM and characterizing the mechanisms that promote and antagonize ICB efficacy. In particular, we are interested in studying the role of conventional dendritic cells (cDCs) in anti-MBM immunity after being activated by the original melanoma tumor outside the brain. cDCs are a type of immune cells that are a key part of the immune response in cancer. Using our mouse model of MBM, we will also examine the double-edged pro-immunologic and anti-immunologic effects of chronic exposure to a molecule called interferon (IFN) as it is produced in the tumor microenvironment and is important in modulating immune responses . This research aims to pave the way for innovative therapeutic strategies by decoding these complex interactions, potentially transforming MBM treatment outcomes and offering hope to patients facing this formidable health challenge.  

Ethan Schonfeld, MS

Ethan Schonfeld, MS

Institution:

Mentor: Michael Lim, MD 

Tribute: In memory of Jeffrey Michael Tomberlin

Project: Mechanisms Underlying Sustained Triple Cocktail Polymer Therapy for Glioblastoma

Immunotherapy represents a revolutionary approach that recruits the body’s own defenses to selectively attack cancer cells, and it has redefined the treatment and prognosis for numerous cancers. However, glioblastoma, the most aggressive form of brain cancer, has remained resistant to these immunotherapies. Several mechanisms of glioblastoma resistance to immunotherapy are known, and recent work has led to the development of therapies targeting these resistance mechanisms. Additionally, direct injection of immunotherapies into the tumor and/or its draining lymph nodes have shown efficacy while limiting the systemic side effects associated with conventional treatments. We seek to combine these recent advances in the field to bypass resistance as well as the serious side effects associated with current immunotherapies. By using a novel therapy that combines three promising immunotherapies into one supercharged treatment, we aim to enhance the body’s own immune response against glioblastoma. Central to our mission is to unravel how each drug rallies the immune system and how, in concert, they amplify this response to mount a more robust defense. The results of this project aim to advance the development of a state-of-the-art treatment for glioblastoma but may also inform the future of brain tumor treatment by determining how best to combine individual immunotherapies for the most effective treatment of a patient’s brain tumor. 

Suchet Taori, BA

Suchet Taori, BA

Institution:

Mentor: Jeremy Rich, MD 

Tribute: In honor of Debbi Schaubman

Project: Lactate Reprograms Glioblastoma Immune Escape Through Histone Lactylation

Glioblastoma (GBM), the most common and lethal primary brain cancer, can evade anti-tumor immune responses. Like other cancers, GBM also exhibits a metabolic reprogramming towards lactate production, a byproduct of cellular metabolism, via the Warburg Effect. However, the relationship between GBM metabolism and immune evasion remains unclear. In recent years, a novel role of lactate was first described, whereby lactate regulates gene expression through modification of gene expression (this specific modification is termed lactylation). To date, however, little is known regarding the function and molecular regulation of lactylation in cancer. Our preliminary work has demonstrated that lactate produced in the tumor can induce GBM lactylation-mediated reprogramming by upregulating various processes involved in immune suppression. Through my research, I aim to uncover the key regulators involved in the lactylation process, determine key pathways implicated in GBM immune resistance following lactylation, and determine the therapeutic efficacy of targeting lactylation with immunotherapies using state-of-the-art cellular and animal models. In my project, we are proposing a novel mechanism linking epigenetic reprogramming, tumor metabolism, and immune cell evasion in GBM. The proposed research could provide benefits for GBM and other cancers. 

2023 Ongoing & Recently Awarded Grants

Research Collaboration Grants

The Research Collaboration Grant is a two-year, $200,000 grant, to support interdisciplinary team science projects that combine resources to streamline accelerate progress in the brain tumor field.

Federico Gaiti, PhD

Federico Gaiti, PhD

Co-PI: Gelareh Zadeh, MD, PhD, FRCSCk

Institution: University Health Network

Co-PI Institution: University of Toronto

Tribute: In memory of George Surgent

Project: Epigenetic Evolution and Plasticity of Glioma Cell States

Co-PI: Gelereh Zadeh, MD, PhD, FRCSC Co-PI: Gelareh Zadeh, MD, PhD, FRCSC

Tumors continuously evolve into more aggressive and treatment-resistant forms. Brain tumors, such as gliomas, illustrate the dilemma of cancer evolution. Despite standard-of-care treatments, these tumors inevitably recur, with an average survival rate of about one year, thereby underscoring an urgent need for novel treatments.

Our preliminary findings suggest that these incurable brain tumors exhibit extensive diversity within and harbor multiple types of malignant cells, presenting a significant unmet therapeutic challenge. Therefore, identifying the key molecular factors that drive this cellular diversity and promote malignant transformation is a crucial goal for neuro-oncology and represents a prime opportunity for intervention. However, we currently lack a comprehensive understanding of the primary molecular factors that contribute to glioma diversity and malignant transformation.

In this project, we aim to develop and apply innovative genomic and experimental technologies to glioma samples collected at diagnosis and upon recurrence following chemotherapy and radiotherapy. This data will enhance our understanding of aggressive malignant transformation, tumor progression, and therapeutic resistance. These studies are expected to lead to novel treatment strategies for this devastating cancer, improving patient outcomes. Additionally, this work will generate new tools for investigating tumor evolution across other types of brain tumors.

Oliver Jonas, PhD

Oliver Jonas, PhD

Co-PI: Shawn Hervey-Jumper, MD 

Institution: Brigham and Women’s Hospital 

Co-PI Institution: University of California, San Francisco

Tribute: Partially supported by StacheStrong

Project: Using In Situ Microdevices to Identify Effective Therapies in Brain Tumors

Shawn Hervey-Jumper, MD Co-PI: Shawn Hervey-Jumper, MD

This project utilizes an implantable microdevice to measure multiple drug responses directly within the tumor of glioblastoma patients. We have developed a miniaturized implant, roughly the size of a grain of rice, that is implanted into the patient, remains inside the tumor for 3 days, and is then removed during a standard surgical tumor resection. The implant releases microdoses of 20 different drugs into different regions of the tumor. After surgery, we examine the tumor response to each of the treatments independently using a range of cutting-edge spatial biology techniques. This study represents the first time that  measurements of multiple drug responses from inside the tumor of the same patient are realized. We expect that the resulting data set will allow us to systematically understand drug response and resistance pathways to established and novel treatment options. The goal is to rationally select highly effective combination treatment regimens that are based on each patient’s actual tumor biological response to the range of available drugs.

Derek Wainwright, PhD

Derek Wainwright, PhD

Co-PI: Pilar Sanchez-Gomez, PhD

Institution: Loyola University of Chicago

 Co-PI Institution: Instituto de Salud Carlos III 

Tribute: Partially supported by BrainUp 

Project: Maladaptive Effects of the Aged Brain in Older Adults with Glioblastoma

Pilar Sanchez-Gomez, PhD Co-PI: Pilar Sanchez-Gomez, PhD

Older adults ≥65 years of age represent more than 70% of all new glioblastoma (GBM) diagnoses. Older adults with GBM tend to experience significantly worse overall survival as compared to their younger GBM patient counterparts. However, the biological underpinnings for the dismal outcomes in older GBM patients is poorly understood. Our study aims to determine how the  older adult brain macroenvironment that’s, outside of the bulk GBM mass, contributes to worse outcomes  in older adults with GBM . The remarkable improvements in healthcare has led to a world-wide population that is increasingly entering an age range associated with unprecedented increases in cancer incidence that will have important economic and public health consequences. Historically however, most older adults with GBM have been excluded from participating in clinical trials. Thus, more precision medicine approaches that are specifically tailored to older adults are necessary for understanding how best to improve the outcomes in this underserved GBM patient population. As direct result of our studies, we believe that our discoveries will improve treatment-related outcomes for older adults with GBM. By understanding age-dependent factors that decrease survival, we also believe that our work will indirectly improve outcomes for middle-aged and younger GBM patients that also eventually suffer from the hyper-aging effects caused by GBM.

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.

Charuta Furey, MD

Charuta Furey, MD

Institution: Barrow Neurological Institute, St. Joseph Medical Center

Mentor: Nader Sanai, MD

Tribute: In honor of Joel A. Gingras, Jr.

Project: Tracking Glioma Evolution Through Longitudinal Cerebrospinal Fluid Liquid Biopsy

Glioblastoma (GBM) is the most common type of malignant brain tumor in adults and remains incurable: most patients die within two years of diagnosis.  Tracking glioblastoma evolution in response to experimental therapies is crucial to developing effective treatment breakthroughs. This study proposes a new method of liquid biopsy that involves collecting cerebrospinal fluid (CSF) from a reservoir in the tumor cavity during outpatient clinic visits.  This minimally invasive technique allows researchers to isolate and examine circulating tumor DNA and GBM cells over time, which can help understand how the tumor evolves and develops resistance to therapy.  If successful, this study will be the first to use serial CSF biopsies to prospectively monitor GBM evolution in response to a novel DNA Damage Response inhibitor.  Findings from this study will help us better understand GBM resistance mechanisms and can be leveraged to create better combination therapies.  Additionally, this minimally invasive liquid biopsy technique has potential clinical utility in helping us predict treatment response, differentiate pseudoprogression, and even catch recurrent disease before traditional MRI surveillance methods.

Juyeun Lee, PhD

Juyeun Lee, PhD

Institution: Cleveland Clinic 

Mentor: Justin Lathia, PhD

Tribute: Fully supported by Tap Cancer Out

Project: Differential T cell Mitochondrial Function Drives Sex Differences in Glioblastoma

Glioblastoma, the most aggressive primary brain tumor, affects more men than women, and men generally have worse outcomes. These differences between men and women are due in part to how the immune system responds to the brain tumor. Immunotherapy, which aims to enhance a patient’s own immune system to fight against the tumor, has worked well in some other types of cancer, however, they haven’t been successful in treating glioblastoma. Therefore, it is important to discover a new way to improve the current therapeutics for glioblastoma patients. Our laboratory found that male and female immune cells, specifically T cells which can kill cancer cells, responded differently to the cancer, with male T cells becoming dysfunctional more quickly than female cells.

To understand why male and female T cells are different, I want to examine the cells’ energy factories, called mitochondria, and how they work differently between men and women, as mitochondria function is crucial in regulating T cell function. I hypothesize that differences in mitochondria could lead to faster T cell dysfunction (also referred to as exhaustion) and contribute to faster cancer growth in men. To investigate this further, I will study how the mitochondria in T cells behave in male and female tumors. I will also test a new treatment that could help boost the mitochondria specifically in T cells. These studies could help develop new treatments that are tailored to men and women’s different immune responses to the cancer.

Jun Nishida, PhD

Jun Nishida, PhD

Institution: Dana-Farber Cancer Institute 

Mentor: Kornelia Polyak, MD, PhD

Tribute: Fully supported by Breast Cancer Research Foundation

Project: Topological Heterogeneity of Breast Cancer Brain Metastases

Brain metastasis is a challenging clinical problem leading to the shortest survival among metastatic breast cancer patients, yet therapeutic options are very limited. Brain metastases of different cancers preferentially grow in specific parts of the brain. For example, brain metastases of melanoma frequently occur in the frontal regions while those of breast cancer can form everywhere but are most likely found in the back of the brain. The significance of these differences has not been studied and all patients are treated the same way regardless of the location in the brain.
The levels of nutrients in the brain are very different from those of other organs and even within the brain each region can be different. Thus, cancer cells must adapt to this special environment to be able to form tumors. In my preliminary studies I identified candidate molecules in breast cancer cells that regulate adaptation to the brain, including proteins that can be therapeutic targets. In this proposal I will further investigate how differences in nutrient levels in different parts of the brain are preferred by cancer cells with different properties and how we can exploit this to improve the treatment of patients with brain metastases. The mechanisms I identified also play a role in the growth of pediatric gliomas. Thus, the successful completion of my proposed study will facilitate the understanding and treatment of not only brain metastatic cancer patients, but also children with highly aggressive brain tumors.

Rakesh Trivedi, PhD

Rakesh Trivedi, PhD

Institution: University of Texas M.D. Anderson Cancer Center

Mentor: Krishna Bhat, PhD

Tribute: Fully supported by Tap Cancer Out

Project: Epigenetic Biomarkers Identification in Brain Tumors Using Liquid Biopsy Approach

The most aggressive form of primary malignant brain tumor is glioblastoma, with a 5-year survival rate of less than 5%. Monitoring glioblastoma progression and treatment response with the current brain scanning methods such as magnetic resonance imaging (MRI) is extremely challenging and does not accurately reflect tumor burden. Furthermore, MRI is not a routine procedure due to the high costs involved. The alternative detection methods used in systemic cancers such as tissue biopsies are not possible in brain cancer without putting the patient’s life at risk.

This proposal aims at addressing these challenges by developing a minimally invasive liquid biopsy procedure that can be used for longitudinal monitoring of glioblastoma progression. The diagnostic power of circulating DNA released by brain tumor cells in the blood of glioblastoma patients will be evaluated using specific modifications of circulating DNA to precisely detect tumor burden. The development of such innovative, cost-effective, and safe techniques will transform glioblastoma patient management and provides a convenient way of monitoring tumor progression and treatment response. We eventually hope to develop an assay integrated with standard imaging techniques for minimally invasive monitoring of glioblastoma.

Discovery Grant

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.

Theresa Barberi, PhD

Theresa Barberi, PhD

Institution: Johns Hopkins University School of Medicine

Mentor: Alan Friedman, MD 

Tribute: In memory of Kelli McLaughlin

Project: Genetically Locked Pro-Inflammatory Myeloid Cells as Glioblastoma Immunotherapy

Glioblastoma is a highly aggressive brain cancer, with less than 5% survival at five years. NF-kB p50, or simply “p50”, is a protein whose absence renders myeloid cells, white blood cells that are part of the immune system, more inflammatory due to its inhibitory effects on genes that control inflammation. We found that immature myeloid cells (IMCs) lacking p50 are active against glioblastoma in mouse models. Once infused, these “p50-IMCs” travel to tumors, develop into mature myeloid cells, and activate other immune system cells (including T cells) to fight the cancer.

STAT6 induces anti-inflammatory genes. We expect that myeloid cells lacking both p50 and STAT6 will be even more inflammatory than cells only lacking p50 and thereby will possess an even greater ability to cure glioblastoma. We will compare the ability of IMCs lacking p50 only, STAT6 only, and cells lacking both p50 and STAT6 (called a p50/STAT6 double knockout) to eliminate glioblastoma tumors in mice. We will also monitor mice for any toxicities; we have seen none thus far with p50-IMCs. In addition, we will develop p50/STAT6 double knockout in human cells and characterize them to learn if they are suitable for use in patients.

Upon completion of these studies, we anticipate rapidly evaluating our novel p50/STAT6 double knockout immature myeloid cell immunotherapy in clinical trials with the goal of increasing rates of cure and reducing toxicities of therapy for patients with glioblastoma and other high-grade brain cancers.

Defne Bayik, PhD

Defne Bayik, PhD

Institution: Sylvester Comprehensive Cancer Center, University of Miami Health Systems

Mentor: Antonio Iavarone, MD

Tribute: Fully supported by an Anonymous Family Foundation

Project: Gamma-aminobutyric Acid Regulates Glioblastoma Immunity in a Sex-specific Manner

The crosstalk between the tumor cells and the surrounding noncancerous cells drives the aggressiveness of glioblastoma (GBM), which is the most frequent and deadly malignant brain tumor. The treatment strategies aiming to kill tumor cells have not markedly improved patient outcomes over the past few decades. Therefore, understanding the complex interactions between tumors and their environment is important for the development of more effective anti-cancer therapies. One promising strategy is to educate the immune system to fight against tumors. However, tumors employ multiple mechanisms to recruit inhibitory immune cells, such as myeloid-derived suppressor cells (MDSCs), that would support the growth of the tumors.

We previously reported that different types of MDSCs play a role in the progression of GBM in males versus females. Recently, we observed that γ-aminobutyric acid (GABA), a critical modulator of neurons, can also act as a potential regulator of female-specific MDSC activity. In this project, we aim to understand how GABA reprograms MDSCs in females, and whether we can therapeutically target the pathways activated by GABA to reverse MDSC-mediated tumor growth. This project will identify new therapeutic opportunities for GBM that account for differences between males and females. Given that several GABA pathway inhibitors are commonly used in patients with mood disorders, repurposing these drugs can have an immediate impact on GBM outcomes.

Phedias Diamandis, MD, PhD 

Phedias Diamandis, MD, PhD 

Institution: University Health Network

Tribute: Fully supported by An Anonymous Family Foundation

Project: Consumer-Grade Electroencephalography for Remote Monitoring of Tumor Evolution

Cancerous brain tumors are challenging to detect and treat partly owing to the lack of early objective patient symptoms. In other cancer types (e.g. breast/colon) early detection through active monitoring has proven effective in improving patient outcomes. These approaches are, however, difficult to implement for less common cancers like glioblastoma (the most common/aggressive brain cancer). Recently, the application of advanced analytical methods like electroencephalography (EEG), a technique that allows monitoring of the brain’s electrical activity using external scalp electrodes, has re-emerged as a powerful clinical tool to non-invasively monitor/diagnose brain disease. Similar to the use of smartwatch devices to detect abnormal heart rhythms in the general population, it may now also be possible to use the growing number of consumer-grade EEG devices, commonly used for meditation and focus, to remotely detect brain tumors earlier in their evolution. Early detection could afford more effective treatments/outcomes. Our group has begun developing software to capture and analyze EEG data directly from patients/participants’ homes. This study is to determine how feasible/reliable remote EEG data collection is and if we can detect activity changes in patients with brain tumors, as their disease evolves/progresses. If successful, this EEG-based strategy bears potential value as a remote screening tool for early detection of brain tumors, in both the at-risk/general population. 

Siddharthra Mitra, PhD

Siddharthra Mitra, PhD

Institution: University of Colorado Denver

Tribute: In memory of Katie Monson

Project: Chimeric Antigen Receptor (CAR) Expressing Phagocytes for Targeting Glioblastoma

Brain tumors are among the deadliest cancers in humans. In adult Glioblastoma Multiforme (GBM) the average survival time is 12-18 months – only 25% of glioblastoma patients survive more than one year, and only 5% of patients survive more than five years. Similarly, pediatric high-grade gliomas are among the most deadly childhood cancer, with <20% of kids living past five years of diagnosis. One way to target brain tumor cells is by using the body’s own immune system against the tumor. One of the main immune cells that you find in your body is cells called macrophages, which essentially are like scavengers that go around eating dead or dying cells, infective bacteria and cells which have been infected with viruses. Cancer cells are known to protect themselves using proteins that tell immune cells not to attack them. Previously, we have published a “don’t eat me” signal on the surface of all the tumor cells, but if you block that “don’t eat me” signal, these macrophages can eat the tumor cells.

In this proposal, we will engineer these macrophages to carry homing devices on their surface that can direct their attack specifically on tumor cells, guided by the signals on the surface of the tumor cells. CAR T cell therapy, a form of immunotherapy that uses specially altered T cells – a part of the immune system that fights diseases, including cancer involves the collection of a patient’s T cells and genetically reprogramming them in the lab to recognize markers on specific cell types in the body. Engineering a similar protein-based homing device on the surface of macrophages will give them the ability to hunt out tumors and eat them up.

Allegra Petti, PhD

Allegra Petti, PhD

Institution: Massachusetts General Hospital

Tribute: Fully supported by an Anonymous Family Foundation

Project: Translating Multimodal Single-cell Genomics into Personalized Treatments for GBM

Glioblastoma (GBM), the most common primary malignant brain tumor in adults, is a devastating disease. Treatment for GBM has evolved little in recent decades, probably because this tumor type is notoriously complex: Each GBM tumor is heterogeneous with respect to genetics, tumor cell state, and immune microenvironment. In particular, a new technique called single-cell RNA-sequencing (scRNA-seq) has revealed that each GBM is a mixture of different types of tumor cells, each with different biological properties. Consequently, each tumor is essentially multiple diseases, and it will probably be necessary to treat each patient with drug combinations targeted to that individual’s tumor. Although scRNA-seq has enabled us to understand the complexity of GBM, it has not yet helped us treat the disease. This is partly because it is difficult to physically isolate and study the cell types discovered using scRNA-seq. Here, we propose a novel approach to isolating these GBM tumor cell populations, studying their therapeutic vulnerabilities, and identifying personalized drug combinations that target multiple cell populations simultaneously. Our approach is based on a new technology called CITE-seq, an extension of scRNA-seq that will enable us to define tumor cell populations and, in subsequent work, isolate them for further study using a technique called flow sorting.

Soma Sengupta, MD, PhD

Soma Sengupta, MD, PhD

Institution: University of North Carolina at Chapel Hill 

Tribute: Fully supported by an Anonymous Family Foundation

Project: Reprogramming Macrophages in Brain Metastatic Lung Cancer

In advanced cases, lung cancer that goes to the brain is a fatal disease. Treatment often uses radiosurgery, but this does not produce a durable response. We have a new drug (AM-101) that gets into the brain efficiently and can help the tumor microenvironment (the non-tumor cells and structural components in and around the tumor) become more favorable to treatments by increasing the population of an immune cell called a macrophage. What is unique is that the macrophage promoted by AM-101 kills cancer cells. We also know that AM-101 makes lung cancer cells more sensitive to radiation without side effects such as bone marrow toxicity. Once we complete these experiments, we intend to publish the data, and apply for R01 funding. 

Daniel Silver, PhD

Daniel Silver, PhD

Institution: Cleveland Clinic

Mentor: Justin Lathia, PhD

Tribute: In memory of Chip Greenlee

Project: Reactive Astrocytes Drive Disease Aggression in Glioblastoma

Glioblastoma (GBM) remains among the most lethal of human cancers despite ongoing efforts to understand the mechanisms used by the tumor cells that define this cancer. The challenge is that GBM is far more than a collection of cancer cells independent from the brain. GBM invades extensively throughout the organ where it intermingles with numerous brain cells. The field has detailed several processes by which nerves and immune cells support the growth of this tumor. However, even though GBM tumors infiltrate fields of astrocytes, the most abundant cell type in the brain, we know far less about GBM-astrocyte interactions. Most mouse models of GBM develop within a capsule formed by highly reactive astrocytes. However, astrocytes do not demonstrate this wall-building behavior in human GBM. We do not understand the molecular signals that shield invasive GBM cells from astrocyte encapsulation in the human brain, nor do we fully understand the mechanisms and effects that result from GBM-astrocyte interaction. In this project, we aim to clarify the functional consequences of astrocyte-GBM interactions. Doing so will help close this major knowledge gap in the field and, more importantly, this project will produce a roster of factors that we will then evaluate as potential therapeutic targets against this devastating cancer. More generally, emphasizing astrocyte-tumor interactivity and revealing the signals that govern this relationship will help shift our perspective towards a more complete understanding of GBM as a disease of the brain, not simply a disease that resides in the brain.

Elizabeth Sweeney, PhD

Elizabeth Sweeney, PhD

Institution: The George Washington University

Mentor: Conrad Russell Young Cruz, MD, PhD

Tribute: In memory of Stephanie Lee Kramer

Project: Personalized Nanoparticle-Mediated T Cell Therapy for Glioblastoma

Glioblastoma (GBM) is the most commonly diagnosed brain cancer in the United States, impacting ~12,000 people per year. Despite intense standards-of-care that include surgery, radiation, and chemotherapy, the prognosis for GBM remains dismal, with a relative five-year survival rate of 7.5%. In response to the urgent need for effective therapies for patients with GBM, we are proposing a fundamentally new and personalized therapeutic approach that uses nanotechnology to train a patient’s own immune cells to recognize and kill their tumor cells. Our strategy involves applying heat to GBM cells that have been removed during surgery, which kills them in a way that can optimally engage with immune cells. Then, the heat-killed GBM cells are added to a dish containing immune cells from the same patient, which educates the immune cells to recognize the tumor cells. These immune cells are grown in the laboratory and then infused back into the patient as therapy. This platform is personalized to a patient’s own unique tumor makeup and as such, it allows the immune cells to recognize multiple potential targets (known and undiscovered) particular to a given patient. If successful, this project will lay the foundation for a new process for developing better adoptive immune cell therapies for treating brain cancer. As the nanoparticle-based platform can be applied to any cancer type, we envision its utility in developing effective therapies for additional brain and solid tumors.

Nehalkumar Thakor, PhD

Nehalkumar Thakor, PhD

Institution: The University of Lethbridge

Tribute: Partially supported by Brain Tumor Foundation of Canada

Project: To Assess Eukaryotic Initiation Factor 5B as a Therapeutic Target in Glioblastoma

Glioblastoma (GBM) is one of the deadliest cancers with a dismal prognosis and survival rate. Frontline therapeutic agents are failing in the clinic and there is an urgent need to develop novel therapeutic intervention strategies for the treatment of patients with GBM tumors. Under the stress conditions found within tumors, the normal (canonical) process in cells of translating genetic messages (mRNA) into protein is reduced to conserve cellular energy. At the same time, the cells begin the non-canonical process of translating a subset of mRNAs to different proteins so that the cell can cope with the stress condition. This non-canonical process contributes to the resistance of cancer cells against therapeutic agents. Proteins in cells known as eIFs have been shown to regulate the early steps of non-canonical translation of mRNA to protein. We have identified a protein, eIF5B, that facilitates the translation of anti-death proteins and allows GBM cells to survive during high-stress conditions that would normally cause cell death. We propose to define the role of eIF5B in GBM cell survival, proliferation, and non-canonical translation using brain tumor-initiating cells from patients. These experiments will validate eIF5B as a therapeutic target for GBM. This proof-of-concept project will establish the preclinical rationale for targeting eIF5B for the therapeutic benefit of GBM patients.

Gina & Tim Abbas
Caregiver & Anaplastic Astrocytoma Survivor

Double Your Impact for Brain Tumor Families

Donate now and your gift will be matched thanks to the Will Hicks Charitable Association.