FUNDING CHILDHOOD CANCER RESEARCH
B+ Scientific Advisory Board
Part of our mission is to provide childhood cancer funding. We are humbled and appreciative to have such a distinguished panel of world-class pediatric oncology clinicians and researchers on The B+ Foundation Scientific Advisory Board. Upon the recommendations of this esteemed group, The B+ Foundation looks forward to continuing to play a very active role in childhood cancer research funding.
The members of the B+ Scientific Advisory Board are:
Dr. Peter C. Adamson
Peter C. Adamson, MD is Global Development Therapeutic Area Head, Oncology and Pediatric Innovation at Sanofi. Dr. Adamson leads global cancer drug development for Sanofi and is also working across therapeutic areas to further pediatric drug development efforts. Prior to this, Dr. Adamson served as Chair of the Children’s Oncology Group (COG), a National Cancer Institute supported international consortium of more than 220 childhood centers. Dr. Adamson, currently Emeritus Professor of Pediatrics and Pharmacology at the Perelman School of Medicine, is Board Certified in Pediatric Hematology/Oncology and Clinical Pharmacology. He was appointed by President Obama to, and continues to serve on, the National Cancer Advisory Board (NCAB), and also served on the Blue-Ribbon Panel for the Beau Biden National Cancer Moonshot Initiative.
Dr. Steven Dubois
Dr. DuBois completed medical school and pediatric training at the University of California, San Francisco (UCSF). He completed pediatric oncology training at Dana-Farber / Boston Children’s Hospital and obtained a Master of Science in Epidemiology from the Harvard School of Public Health. He is currently a Professor of Pediatrics at Harvard Medical School. He is the Director of Experimental Therapeutics at Dana-Farber / Boston Children’s Cancer and Blood Disorders Center where he leads a program designed to bring new targeted therapies to children with cancer.
Dr. DuBois leads an active clinical and translational research program focused on patients with advanced neuroblastoma and Ewing sarcoma. He conducts clinical trials of novel targeted agents relevant to these diseases, including national phase 1, 2, and 3 clinical trials. He also studies new biomarkers that improve our understanding of the biology of pediatric solid tumors and of the pharmacodynamic effects of targeted therapies.
Dr. DuBois has served on a number of national committees, including the Children’s Oncology Group (COG) Neuroblastoma Steering Committee, COG Bone Tumor Committee, the American Society of Clinical Oncology (ASCO) Scientific Program Committee, and the ACCELERATE Scientific Steering Committee.
Dr. Elizabeth Fox
Elizabeth Fox, MD, MSCR is the Senior Vice President, Clinical Research Administration, St Jude Children’s Research Hospital and Associate Director of Clinical Research, St Jude Comprehensive Cancer Center where she leads regulatory affairs and clinical trial operations as well clinical research prioritization and strategy. Prior to joining St Jude in 2020, she was a professor of Pediatrics at the Perelman School of Medicine at the University of Pennsylvania and Director of Developmental Therapeutics in Oncology at the Children’s Hospital of Philadelphia. Nationally, she is the chair of Developmental Therapeutics for the Children’s Oncology Group and the UM1 funded Pediatric Early Phase Clinical Trials Network. She is the Vice-Chair of ACCELERATE, the international multi stakeholder platform to advance drug development for children and young adults with cancer. Dr. Fox has expertise in quantitative clinical and pre-clinical pharmacology, pediatric clinical oncology, and clinical research trial design including response and toxicity biomarker endpoint development. Her professional mission is to utilize clinical research to develop the most promising therapies for children and mentor clinician-scientists and lead a clinical research workforce to foster innovation to improve the outcome for children with catastrophic disease.
Dr. Rebecca Gardner
Rebecca Gardner, MD is a pediatric oncologist who has spent the past two decades focusing on T cell-based therapeutics for pediatric malignancies with expertise in the academic medical center in bringing products from the bench to the bedside, with a strong interest in reverse translation. While in Seattle, she oversaw the clinical development group responsible for 15 active INDs of CAR T cells for pediatric malignancies with indications across both liquid and solid tumors and the treatment of over 500 patients. She is currently an Associate Member at St Jude Children’s Research Hospital and continues to work on the clinical development of novel cell therapies, with special attention on how to integrate immunotherapy along the compendium of treatments for pediatric cancers. In addition to the development of novel cell therapies, she has a keen interest in providing access to cell therapies with a focus on promising academic products that do not have a current pathway to commercialization.
Dr. Douglas Hawkins
Douglas S. Hawkins, MD, is the Group Chair of the Children’s Oncology Group (COG). COG is the world’s largest organization devoted exclusively to childhood and adolescent cancer research. COG unites over 10,000 experts in childhood cancer at more than 200 leading children’s hospitals, universities, and cancer centers across North America, Australia, and New Zealand in the fight against childhood cancer. Dr. Hawkins is also a clinician at Seattle Children’s Hospital and Professor of Pediatrics at the University of Washington School of Medicine.
Prior to becoming COG Group Chair in 2020, Dr. Hawkins was the Chair of the COG Soft Tissue Sarcoma Committee, overseeing the conduct of biology studies and clinical trials for rhabdomyosarcoma and other soft tissue sarcomas across North America. He was a member of the COG Bone Tumor Steering Committee. Dr. Hawkins had focused on clinical research, particularly in the treatment of pediatric sarcomas. He was the chair of two COG clinical trials, one for Ewing sarcoma and another for rhabdomyosarcoma. He chairs the international EURO EWING Consortium External Advisory Board and also chairs the international Frontline and Relapse in RhabdoMyoSarcoma Study (FaR-RMS) Data Monitoring Committee.
Dr. Andy Kolb
Andy Kolb, MD received his undergraduate education at the University of Pennsylvania and his medical degree from Jefferson Medical College. After a residency in pediatrics at St. Christopher’s Hospital for Children, Dr. Kolb completed his fellowship training at Memorial Sloan-Kettering Cancer Center. He now serves as the Director of the Nemours Center for Cancer and Blood Disorders in Wilmington, DE. Dr. Kolb is a clinician scientist primarily focused in the laboratory and in the clinic on the efficient and effective translation of novel therapies into children. He is a founding member of the National Cancer Institute funded Pediatric Preclinical Testing Program and has successfully completed preclinical evaluations of numerous compounds and aided in the translation of these agents into clinical trials. In exploring the mechanism of action of targeted compounds, Dr. Kolb has developed an expertise in proteomics and cell signaling. Dr. Kolb serves within the Children’s Oncology Group (COG) as Chair of the Myeloid Disease Committee, Member of the Scientific Council, and Member of the Bone Tumor Committee. Through this work, Dr. Kolb has also developed expertise and experience in collaborative science, resource stewardship, clinical research development, clinical trial design and implementation, and in the necessities of young investigator development.
Dr. A. Thomas Look
A. Thomas Look, MD, is a Professor of Pediatrics at Harvard Medical School and Vice Chair for Research in the Department of Pediatric Oncology at the Dana-Farber Cancer Institute, as well as co-leader of the Dana-Farber/Harvard Cancer Center’s Leukemia Program. Over the past three decades, Dr. Look has published multiple peer-reviewed papers about the molecular basis of cancer and the application of molecular genetic findings to improve the treatment of childhood malignancies, particularly T-cell acute leukemia, myelodysplastic syndrome and neuroblastoma. He moved from St Jude Children’s Research Hospital to Dana-Farber Cancer Institute in 1999 specifically to establish a research program in the zebrafish model, to conduct genetic studies aimed at the identification of novel targets for cancer therapy, and he is now internationally recognized as a leader in this field.
His initial work led to the first transgenic model of leukemia in the zebrafish, paving the way for small-molecule drug and targeted genetic modifier experiments in a vertebrate disease model. More recently, his laboratory has developed the first zebrafish transgenic model of childhood neuroblastoma, opening up the opportunity to apply the powerful genetic technology available in the zebrafish to identify new molecular targets for therapy in this devastating childhood tumor.
He is the principal investigator on several NIH-funded grants, including a Program Project focusing on T-ALL pathogenesis. He has won numerous awards, including the Allison Eberlein Award for Childhood Leukemia Research, the Award for Excellence from the American Academy of Pediatrics, the Pediatric Oncology Lectureship of the American Society of Clinical Oncology, the ASPHO Frank A. Oski Memorial Lectureship Award of the American Society of Pediatric Hematology and Oncology, and he is a Fellow of the American Association for the Advancement of Science.
Dr. Look received his MD degree and postgraduate training in Pediatrics from the University of Michigan, and his fellowship training in Pediatric Oncology at St. Jude Children’s Research Hospital. Prior to his appointment at Harvard, he was a professor at the University of Tennessee College of Medicine.
Dr. Julie R. Park
Julie R. Park, MD is attending physician at Seattle Children’s Hospital, professor in pediatrics at the University of Washington School of Medicine and associate in the Clinical Research Division at Fred Hutchinson Cancer Research Center (FHCRC). She is director of the pediatric hematology-oncology fellowship at the University of Washington.
Dr. Park is an active member of the Children’s Oncology Group Consortium and as chair of the COG Neuroblastoma Scientific Committee provides leadership for the development of neuroblastoma clinical research within COG. Dr. Park’s primary research focus has been investigating novel therapies for the treatment of high-risk neuroblastoma, a rare but aggressive form of childhood cancer. She has conducted a multi-center clinical trial to determine the feasibility and toxicity of a novel induction chemotherapy regimen for high-risk neuroblastoma and has collaborated with local and national investigators to optimize the use of radiation therapy as part of treatment for neuroblastoma. Dr. Park’s work has led to her development of the current national randomized phase III trial within COG for treatment of newly diagnosed high-risk neuroblastoma. Dr. Park has ongoing collaborations with Dr. Michael Jensen and is currently the primary investigator on an early phase clinical trial that uses adoptive immunotherapy approaches to treat neuroblastoma. Dr. Park also leads the Advanced Therapeutics Program at Seattle Children’s Hospital and has steered Seattle Children’s into becoming a leading participant in the Phase I Consortium of COG and the New Approaches to Neuroblastoma Therapy Consortium. She has been actively involved in the development of novel chemotherapeutic agents that may block critical tumor cell pathways necessary for tumor cell growth and survival.
Dr. Michele Redell
Dr. Redell is an Associate Professor of Pediatrics at Baylor College of Medicine in Houston, TX. She earned her MD and PhD degrees through the Medical Scientist Training Program at the University of Washington. She did her residency in Pediatrics and her fellowship in Pediatric Hematology-Oncology at Baylor College of Medicine, where she has stayed on as faculty. She is a physician-scientist who treats children with leukemia and directs a translational research lab investigating mechanisms of chemotherapy resistance in pediatric acute myeloid leukemia (AML). Research projects in her lab are focused on understanding interactions between AML cells and the microenvironment that allow leukemia cells to survive, and identifying new ways to target chemoresistant AML cells. The lab studies potential new therapies using models that include elements of the bone marrow niche, such as stromal cells and cytokines. Dr. Redell’s group has a productive patient-derived xenograft (PDX) program with one of the largest collections of pediatric AML PDX models in the country. Because of this work, she serves as the Leukemia Program Lead for Baylor College of Medicine’s Patient-Derived Xenograft and Advanced In Vivo Models (PDX-AIM) Core Resource. Dr. Redell is active in the Children’s Oncology Group Myeloid Diseases Committee as the Vice Chair of Biology, a member of the steering committee, and a member of several clinical study committees.
Dr. Lewis Silverman
Dr. Silverman is at Columbia University Irving Medical Center. He leads the DFCI ALL Consortium, a multi-institutional clinical trials group focused on developing more effective and less toxic therapies for children and adolescents with newly diagnosed ALL. He is the Principal Investigator of an international Phase III trial in pediatric Philadelphia chromosome-positive ALL being conducted by the Children’s Oncology Group (COG) and the multi-national European EsPhALL group. Other leadership roles include serving on the COG Scientific Council and as Scientific Chair for the TACL Consortium, which conducts trial for children with relapsed and refractory leukemia and lymphoma.
Dr. Stephen Skapek
Stephen Skapek, MD holds the Distinguished Chair in Pediatric Oncology Research at the University of Texas Southwestern Medical Center, where he serves as the Chief of the Division of Hematology-Oncology in the Department of Pediatrics, and the Medical Director the Pauline Allen Gill Center for Cancer and Blood Disorders at Children’s Medical Center in Dallas. p>
Dr. Skapek graduated from the Duke University School of Medicine, completed his pediatric residency training at the Wilford Hall Medical Center at Lackland AFB in San Antonio, Texas, and completed fellowship training in pediatric hematology and oncology at the Harvard Medical School’s Dana Farber Cancer Institute and Boston Children’s Hospital. p>
After completing his training, Dr. Skapek has focused clinical work on caring for children with rhabdomyosarcoma and other soft tissue sarcomas, and he has carried out both laboratory-based research in cancer and developmental biology and clinical research through the Children’s Oncology Group, which he serves as a member of the Scientific Council and Executive Committee and also as vice-Chair of the Soft Tissue Sarcoma Committee.
Dr. Sarah K. Tasian
Sarah K Tasian, MD is a pediatric oncologist and physician-scientist at the Children’s Hospital of Philadelphia (CHOP) and University of Pennsylvania School of Medicine focused upon development of molecularly-targeted therapeutics high-risk pediatric leukemias in the laboratory and via early-phase clinical trials. She is a graduate of the University of Notre Dame (BS, BA) and Baylor College of Medicine (MD), trained in Pediatrics at Seattle Children’s Hospital and in Pediatric Hematology-Oncology at University of California, San Francisco (UCSF), and is a current PhD student in the Clinical and Translational Oncology Programme at the Universiteit Utrecht in the Netherlands. She directs the Hematologic Malignancies Program at CHOP and is an internationally-recognized expert in pediatric ALL and AML. Her bench-to-bedside and bedside-back-to-bench translational laboratory research program focuses upon testing of small molecule inhibitors and chimeric antigen receptor (CAR) T cell immunotherapies in genetic subsets of childhood ALL, AML and JMML. Dr Tasian has leadership roles in the Children’s Oncology Group ALL executive committee and Myeloid Diseases committees (Vice Chair of Relapse), is the Leukemia Lymphoma Society PedAL/EuPAL consortium clinical trials leader, and leads or co-leads several national or international early phase clinical trials testing precision medicine therapies and immunotherapies in children with high-risk leukemias. p>
Apply for a B+ Research Grant
Our Fall 2025 Research Grant Application is now closed
Investigators interested in applying for a B+ Research Grant next year can read more about our guidelines below. Applications will open in June 2026.
Fall 2025 B+ Grant Awardees
Dr. Sahaja Acharya, M.D. – Johns Hopkins University School of Medicine, Baltimore, MD

In a FLASH: reducing toxicity and enhancing cure for pediatric brain tumors p>
Although treatment advancements have increased survival rates for pediatric brain tumors, side effects remain a major issue. Recalling new information (memory), promptly completing mental tasks (processing speed), and maintaining focus (attention) are particularly affected in patients treated with radiation therapy (RT). We will address this problem by investigating FLASH, a method of delivering RT quickly, which has been shown to reduce short- and long-term neurocognitive problems in young mice compared to conventional RT. Originally, FLASH was delivered using electrons and X-rays, but these methods cannot reach deep parts of the brain where most brain tumors are located. A new technique, pencil beam scanning proton therapy, uses protons to deliver FLASH (pFLASH) to treat brain tumors. While promising, this approach cannot be translated to clinical practice because the correct dose for curing tumors and the best delivery method are still unknown. We aim to characterize the toxicity, cognitive function, and survival after p-FLASH compared to conventional RT in healthy mice and in mice with metastatic medulloblastoma and atypical teratoid/rhabdoid tumor, both of which are aggressive pediatric brain tumors. Our study will define optimal radiation doses and schedules to protect cognition and effectively treat these aggressive pediatric brain tumors with the long-term goal of translating this new technique into clinical practice. p>
Dr. Nicole Anderson, Ph.D. – University of Mississippi Medical Center, Jackson, MS

Identify Genetic Drivers Metastasis Using an Established Zebrafish Model of High-Risk Neuroblastoma p>
Metastatic disease is difficult to treat in neuroblastoma (NB) and is the number one cause of death. Unfortunately, we know little about the mechanisms that control metastasis in NB due to the lack of an appropriate animal model. Patients are designated as high-risk (HR) for having MYCN-amplification or being over 18-months-of-age with metastatic disease. In addition, segmental chromosomal aberrations (SCA) are the most common genetic alteration in HR-NB, where large chromosomal regions are lost or gained disrupting 100s of genes. Determining which genes are critical to metastatic disease within a SCA is difficult, as many are passengers genes that are not important in disease pathology. The zebrafish is important in vivo model for defining the role of new genetic mutations in HR-NB. MYCN_TT zebrafish are unique NB animal model that spontaneously metastasize. Our preliminary data shows that MYCN_TT tumors evolve through copy number alterations (CNA), like HR-NB. We will perform whole genome sequencing to detect CNA in highly metastatic MYCN_TT zebrafish. Next, we will use a cross-species genomics approach to identify CNA shared in both metastatic MYCN_TT and human NB. CNAs shared between species separated by 450 million years of evolution are strong candidate drivers of metastatic disease in NB. Finally, we will test our top 5 driver CNA in human NB cell lines to see their impact on metastatic phenotypes (invasion and migration). p>
Dr. Darko Barisic, Ph.D. – Weill Medical College of Cornell University, New York, NY

Precision Epigenetic Therapies for Pediatric Burkitt Lymphoma p>
Burkitt lymphoma is one of the fastest growing childhood cancers, yet children who relapse have very limited treatment options. This project focuses on a newly discovered weakness in Burkitt lymphoma: mutations in the BAF complex, a group of proteins that normally control how DNA is packaged and which genes are turned on or off. More than half of children with Burkitt lymphoma carry these mutations, which change how immune cells develop and make them more likely to become cancerous. We hypothesize that when these genetic changes co-occur together with chronic inflammation, such as from virus infection or even diet-related inflammation, immune cells are prone to enter a dangerous cycle of repeated growth and mutation. This sets the stage for lymphoma to form and progress. We have identified a new drug that blocks the altered BAF complex and selectively kills Burkitt lymphoma cells with these mutations, while sparing normal cells. In this project we will use advanced models and engineered pediatric lymphoma cells to learn exactly how BAF mutations and inflammation cooperate to drive cancer. We will also test whether combining BAF inhibitors with anti-inflammatory or immune-based therapies can more effectively eliminate tumors. The goal is to create precision treatments for children with Burkitt lymphoma, especially those with relapsed or high-risk disease. We aim to develop safer and more effective therapies that improve survival and quality of life for young patients. p>
Dr. Mark Chiang, M.D., Ph.D. – The Regents of the University of Michigan, Ann Arbor, MI

Deciphering mutually exclusive T-ALL and neurodevelopmental mutations in the ARD domain of the ZMIZ1 coactivator p>
There is an urgent need to find new drugs for ETP leukemia because the usual ones don’t work. It is also a large subtype, comprising 20% of pediatric T-ALL patients. Little is known about ETP cancer since it was recently defined. When scientists try to find a cure for a cancer, a popular idea is to find a drug that blocks a specific genetic mutation. But this approach would be inefficient for ETP leukemia because each mutation is rare. To overcome this challenge, our vision is to target the cell state that is shared by all ETP cancers rather than a specific mutation. What gives an ETP cell its state of "ETP-ness"? The answer is a genetic circuit inside cells that we call the "ETP gene network". This network turns on stem cell genes. Recently, my lab reported that the ZMIZ1 protein activates this network. We examined a part of ZMIZ1 called the ARD that is mutated in T-ALL and a neurodevelopmental disorder. Our initial work modeling these mutations suggests that the mutant ARD protects ZMIZ1 from being degraded and helps it rope together several control centers in the ETP gene network to construct "super control centers" with extraordinarily high activity. In our B+ project, we will learn how these mutations protect ZMIZ1 in order to find ways to overcome this protection and degrade ZMIZ1. Importantly, ZMIZ1 is not essential for health. Therefore, drugs that degrade ZMIZ1 might be a new kind of therapy that avoids the toxic effects of chemotherapy and help all ETP patients. p>
Dr. Roshni Dasgupta, M.D. – Cincinnati Children's Hospital Medical Center, Cincinnati, OH

Development and Validation of Patient-Reported Outcome Measures for Pediatric Cancer Surgery p>
The diagnosis of cancer in a child or adolescent often requires chemotherapy and surgical procedures for treatment. The best patient outcomes cannot be achieved simply by looking at traditional medical history, lab tests, and imaging findings. In previous studies, families of children that have received cancer surgery have told us that they wished their care providers communicated the options for surgery thoroughly and listened more carefully. Understanding a child’s quality of life and the impact that the treatment of a childhood cancer has on the patient and family is essential to the obtain the best outcomes from surgery and overall treatment. When children undergo surgery for cancer, it's not just the medical outcomes that matter—how they feel and recover after surgery is equally important. This project aims to create a system that listens to what young patients and their families have to say about their experiences before and after surgery. By working directly with patients, we can make better decisions about their care, helping them recover more quickly and feel better overall. This project will support efforts to develop a tool that doctors can use to gather patient and family feedback, making sure every child receives care tailored to their unique needs and experiences. This will help us understand where there may be problems in the patient’s treatment journey and allow health care professionals to fix the issues before they impact long-term outcomes of treatment. p>
Dr. Eran Elinav, M.D., Ph.D. – Weizmann Institute of Science, Israel

Linking the Gut Microbiome to Chemotherapy Toxicity in Pediatric ALL p>
Childhood acute lymphoblastic leukemia (ALL) is the most common type of cancer in children. While treatments have improved survival rates, many children experience severe adverse effects from chemotherapy, such as life-threatening infections or damage to vital organs. Current methods rely on trial-and-error approaches, while identifying which children are at higher risk for these complications before treatment starts constitutes a significant unmet clinical challenge. In this project, we will harness the gut microbiome, the community of bacteria inhabiting our gut, to predict which children are more likely to experience severe treatment-realted side effects. Together with a leading tertiary cancer center, we will collect host and microbiome data from 250 children newly diagnosed with ALL. Using AI, we will analyze the data to find patterns linked to adverse events and develop personalized AI-based prediction models for leukemia-related adverse effects. Our initial results show that utilization of personalized microbiome data from children prior to the beginning of chemotherapy, and its analysis using AI, enables robust predictability of a variety of chemotherapy-induced adverse events. To understand if and how the gut microbiome contributes to chemotherapy-induced adverse effects in ALL, we will transplant gut bacteria from patients impacted by chemotherapy-induced toxicities into germ-free mice and mechanistically decode the effect of gut commensals on these adverse effects.
Dr. Furqan Fazal, Ph.D. – Baylor College of Medicine, Houston, TX

Mapping Mitochondrial-localized mRNA Translational Rewiring in Radiation-resistant Pediatric DIPG p>
Diffuse intrinsic pontine glioma (DIPG) remains one of the most devastating pediatric cancers, with a median survival of less than one year and no effective therapies beyond radiotherapy. Despite decades of effort, chemotherapeutic interventions have failed to extend survival, in part due to intrinsic and acquired resistance to radiation. The urgent unmet clinical need for novel therapeutic strategies underscores the importance of uncovering fundamental biological mechanisms that enable DIPG survival under therapeutic stress. Mitochondria have emerged as central players in therapy resistance across many tumor types, and accumulating evidence suggests that DIPG is particularly dependent on mitochondrial oxidative phosphorylation (OXPHOS) for growth and survival. In particular, DIPG cells exhibit heightened OXPHOS activity and maintain metabolic flexibility that supports survival following radiation. Disruption of mitochondrial function can radiosensitize glioma cells, yet these approaches have shown limited translation to the clinic, in part because the systems-level rewiring of mitochondrial gene expression and local translation in DIPG remains poorly understood. This proposal addresses that gap by applying APEX-seq (Fazal et al., Cell 2019), an RNA proximity labeling approach we developed, to map the landscape of mitochondrial RNA localization and local translation in DIPG cells pre- and postradiotherapy. p>
Dr. Yuan Gao, Ph.D. – Case Western Reserve University - School of Medicine, Cleveland, OH

Dissecting Ewing Sarcoma On-Chip for Better Therapeutics p>
Ewing sarcoma is a rare and aggressive cancer that primarily affects the bones or soft tissues of children and young adults. For patients whose cancer has spread or returned, survival rates remain below 30% at five years. Current treatments like chemotherapy and radiation often fail in these high-risk cases. While new approaches like CAR T cell therapy have transformed care for some cancers, they’ve been ineffective in Ewing sarcoma, largely due to a poor understanding of how the tumor interacts with its surroundings, especially the bone marrow environment that supports tumor growth and immune evasion. To address this, we developed a 3D model called Sarcoma-on-a-Chip that mimics the human bone marrow. This miniaturized living system includes tumor cells along with immune cells, blood vessels, and bone-forming support cells. It enables us to study how Ewing sarcoma behaves in a realistic, human-like setting. Using this platform, we will 1)identify genes that help the tumor survive in the bone marrow using CRISPR gene-editing tools, and 2) investigate how the tumor suppresses immune responses. Early findings show that Ewing sarcoma disrupts immune cell function and creates an immune-suppressive environment. This project will uncover new vulnerabilities in Ewing sarcoma and guide the development of more effective therapies, with the ultimate goal of improving outcomes for children with this devastating cancer.
Dr. Gunda Georg, Ph.D. – Regents of the University of Minnesota - Twin Cities, Minneapolis, MN
Tumor-specific PROTAC development as a new therapeutic approach for osteosarcoma p>
This proposal addresses a need for the development of a safe and new therapeutic approach for treating an aggressive cancer, osteosarcoma. Osteosarcoma is one of the most prevalent bone cancers in children and young adults, with a 30% 5-year survival rate when pulmonary metastases are detected. Current standard of care for localized osteosarcoma includes surgery and chemotherapy, which can achieve long-term survival in only 60% of cases. No new approaches have been developed since the 1980s. To address this gap in development, we are developing a new therapeutic strategy against two well-established targets in osteosarcoma, BRD4 and the WEE1 kinase. While inhibitors have been developed, a limitation to inhibiting these proteins is the significant dose-limiting toxicities that patients experience. To overcome these toxic side effects, we are developing first-in-class tissue-specific molecules capable of degrading either the BRD4 protein or the WEE1 kinase in tumor tissue while sparing healthy cells. Our team has significant expertise in therapeutic development for both proteins and has already developed preliminary data supporting our ability to degrade the BRD4 protein in osteosarcoma cells and downregulate the cancer oncogene c-MYC, which is dysregulated in almost 50% of all cancers. These findings can also be extended to treating additional adult and pediatric cancers, including neuroblastoma. We anticipate this new approach will be more efficacious than existing inhibitors.
Dr. Brandon Hadland, M.D. – Fred Hutchinson Cancer Center, Seattle, WA

Dissecting CBFA2T3-GLIS2 Fusion-Driven Leukemogenesis and Therapeutic Resistance p>
Some types of childhood leukemia begin before birth, when early blood cells develop harmful genetic changes. One especially aggressive form, called CBFA2T3-GLIS2 acute megakaryocytic leukemia, is difficult to treat because it often resists chemotherapy and comes back after treatment. This project aims to understand how this leukemia starts and why it returns. Dr. Hadland’s team will use advanced lab models to mimic how the disease originates in early blood cells and transforms over time to frank leukemia. By studying these models, they hope to discover how leukemia cells grow, survive treatment, and change over time. The research has two main goals: first, to find ways to stop leukemia before it becomes dangerous, and second, to identify and eliminate the cells that survive treatment and cause relapse. The team will use powerful tools to track individual cells and uncover weak points that could be targeted with new therapies. This work could lead to better treatments and even ways to prevent leukemia in children who are at risk. In the future, it may be possible to detect genetic changes associated with these leukemias at birth and treat children early—before the disease ever develops.
Dr. Daniel Herranz, Ph.D. – Rutgers Biomedical and Health Sciences (Rutgers Health), New Brunswick, NJ

GLUL as a novel therapeutic target in pediatric T-ALL p>
T-cell acute lymphoblastic leukemia (T-ALL) is an aggressive hematologic malignancy that prominently affects kids. Despite recent advances, 20% of children still relapse and the prognosis of these cases remains extremely poor, highlighting the need to discover novel targeted therapeutic approaches. We previously demonstrated the therapeutic effects of targeting metabolic routes in leukemia. In this setting, our latest results have identified a novel metabolic regulator (GLUL) as an important player in T-ALL. Indeed, with genetic tricks by which we can remove GLUL from leukemias in mice, we have observed very significant therapeutic effects, curing 50% of the mice. However, there is nothing known about GLUL in T-ALL. Thus, here we will comprehensively dissect the effects of targeting GLUL in T-ALL using state-of-the-art techniques in human T-ALL cell lines, mouse primary leukemias in vivo and patient-derived xenografts. Integration of mouse and human data will allow us to understand how T-ALL cells respond to GLUL inhibition. Our results will help us design new combinations with current treatments that will result in stronger antileukemic effects and reduced relapses in the future. p>
Dr. Matthew Hocking, Ph.D. – The Children's Hospital of Philadelphia, Philadelphia, PA

Development of a Measure of Social Connectedness for Survivors of Pediatric Brain Tumors p>
Children treated for a brain tumor have significant problems with social connectedness that begin early in survivorship, persist through adulthood, and cause psychological and medical morbidities. Despite the significance of this issue, these youth are not routinely identified during clinical care due to a lack of validated measures evaluating social connectedness in this population. The constellation of social difficulties and factors contributing to them are unique to this population, increasing the need for a population-specific measure. Such a measure could be integrated into routine clinical care to identify those with social difficulties at an earlier stage to facilitate services to promote social connectedness. The objectives of this proposal are to develop and validate a measure of social connectedness difficulties in pediatric brain tumor survivors. An Advisory Board, consisting of pediatric neurooncology clinicians, experts in social challenges in other populations, and parents of pediatric brain tumor survivors will offer feedback at each phase. In Phase 1, we will develop an item bank for the measure. In Phase 2, we will evaluate the psychometric properties and factor structure of the measure in a sample of survivors recruited from the Children's Hospital of Philadelphia. In Phase 3, we will refine the measure based on Phase 2 data and Advisory Board feedback. This work will establish a measure specific to survivors and suitable for clinical use.
Dr. Jihye Paik, Ph.D. – Weill Medical College of Cornell University, New York, NY

Targeting ALT-Associated Vulnerabilities in Pediatric High-Grade Glioma p>
Pediatric high-grade gliomas are among the most aggressive childhood brain cancers, with few effective treatments and poor survival. Many of these tumors carry mutations in a gene called ATRX, which normally helps maintain chromosome ends, or telomeres. When ATRX is lost, the tumor cells switch on an alternative system called ALT (alternative lengthening of telomeres) to keep dividing. While this allows the cancer to grow, it also creates a weakness: these cells experience constant damage at their telomeres and must rely on backup repair pathways to survive. We discovered that ATRX-deficient, ALT-positive gliomas are especially dependent on a protein called FANCM, which helps repair damaged telomeres. When FANCM is blocked, ALT-driven tumor cells die, but healthy cells are largely unaffected. This makes FANCM a highly promising target for therapy. We have already identified small molecules that can mark FANCM for destruction inside tumor cells, a strategy known as targeted protein degradation. In this project, we will refine these FANCM degraders and test them in cell and mouse models of pediatric glioma. Our goal is to determine whether this approach can slow tumor growth and extend survival, without harming normal tissues. By exploiting a unique weakness created by ATRX loss, this research introduces a new treatment strategy for these otherwise “undruggable” pediatric brain tumors, with the potential to improve outcomes for children facing this devastating disease.
Dr. John Prensner, M.D., Ph.D. – The Regents of the University of Michigan, Ann Arbor, MI

Inhibition of the RNA translation factor, eIF4E, as a novel treatment strategy in Ewing Sarcoma p>
Ewing sarcoma is a rare, aggressive bone cancer mainly affecting children and young adults. Patients with metastatic or recurrent forms face poor prognoses, with 5-year survival rates dropping to 15-30 percent. Treatment involves aggressive chemotherapy and clinical trials exploring innovative therapies, though recent trials have not notably improved outcomes. Current research focuses on the cancer’s biological foundations, specifically the EWSR1-FLI1 fusion protein, which hijacks normal RNA processes to propagate the cancer. Our lab targets RNA translation, the process of making proteins from RNA, as a potential treatment avenue in pediatric cancers like Ewing sarcoma. Our preliminary data show that Ewing sarcoma cells are particularly sensitive to drugs impeding RNA translation. Key components of RNA processing are abundant in these cancer cells, presenting an opportunity for novel therapies. This proposal hypothesizes that blocking EIF4E, a critical component of RNA translation, could effectively treat Ewing sarcoma. Utilizing resources at the University of Michigan, we aim to test both existing and new drugs targeting EIF4E. Our research will involve assessing the efficacy of these drugs on Ewing sarcoma models and tumors in mice, as well as exploring the molecular reasons behind the cancer's susceptibility. This work seeks to deepen our understanding of Ewing sarcoma's cancer biology while identifying small molecule inhibitors suitable for clinical development.
Dr. Gengwen Tian, M.D., Ph.D. – Baylor College of Medicine, Houston TX

Enhancing GD2-CAR NKT anti-tumor efficacy against neuroblastoma by targeting BLIMP1 p>
My research focuses on training a special type of immune cell called a natural killer T cell (NKT) to recognize and kill neuroblastoma (NB) tumor cells without causing the toxic side effects associated with traditional chemotherapy. NB is the most common and hardest to treat pediatric solid tumor outside of the brain. We initiated the first trial in humans to treat patients with their own NKT cells engineered to express a chimeric antigen receptor (CAR) that targets NB cells and spares healthy cells. While the therapy was safe and generated some clinical responses, most did not last and the NKT cells did not persist long-term in the blood. Findings from recent research in my group showed that a protein called BLIMP1 impacts how long CAR-NKTs persist in the body and that when BLIMP1 expression is lower, CAR-NKTs tend to persist longer. However, we also found that when we completely remove BLIMP1 expression through genetic “knock-out,” CAR-NKTs become less effective at killing NB cells. Thus, my current research aims to use two unique strategies with the goal of improving CAR-NKT persistence without compromising their ability to kill NB cells.
ACCELERATE
In 2020, The Andrew McDonough B+ Foundation became a proud, lead supporter of ACCELERATE. ACCELERATE is a European-initiated platform that brings together all stakeholders – Industry (Pharma companies), Regulators (FDA in the USA and EMA in Europe), Academia (researchers), and Advocates (charities).
Objectives
1. ACCELERATE science-driven development of pediatric oncology drugs
2. FACILITATE international cooperation and collaboration between all stakeholders
3. IMPROVE early access to new anticancer drugs in development for children and adolescents
4. SET-UP long-term follow-up of children and adolescents exposed to new drugs