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. Rebecca Gardner
Rebecca Gardner, MD is a pediatric oncologist and clinical researcher at Seattle Children’s Hospital and an associate professor at the University of Washington School of Medicine. Her area of research is focused on the development of cell-based immunotherapies for the treatment of pediatric malignancies, with a special focus on leukemia and lymphoma.
Dr. Gardner is the medical director for immunotherapy at Seattle Children’s. She has led the development of 9 CAR T cell trials for pediatric patients with leukemia and lymphoma. She is an active participant on national and international committees in the cell therapy field, focusing on the development and implementation of novel cell therapy products with improved efficacy and tolerability. She is also an active member of the Children’s Oncology Group’s Non-Hodgkin Lymphoma Committee and recently served on the NCCN guideline committee for pediatric mature B cell lymphoma.
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 and University of Pennsylvania School of Medicine who is interested in development of molecularly-targeted therapeutics for children with high-risk leukemias. She is a graduate of the University of Notre Dame (BS, BA) and Baylor College of Medicine (MD), and she trained in Pediatrics at Seattle Children’s Hospital and in Pediatric Hematology-Oncology at University of California, San Francisco (UCSF). She specialises in the clinical care of children with hematologic malignancies 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 kinase inhibitors and chimeric antigen receptor (CAR) T cell immunotherapies in genetic subsets of childhood ALL and AML. Dr Tasian has leadership roles in the Children’s Oncology Group (COG) ALL and Myeloid Diseases committees and Leukemia Lymphoma Society PedAL/EUpAL consortium, is the COG Developmental Therapeutics committee Vice-Chair of Biology for Hematologic Malignancies, and leads or co-leads several national or international early phase clinical trials testing precision medicine therapies in children with high-risk leukemias. p>
Apply for a B+ Research Grant
Our Fall 2024 Research Grant Application is closed.
Investigators interested in applying for a B+ Research Grant in 2025 can read more about our guidelines below.
Fall 2023 B+ Grant Awardees
Dr. Bruno Di Stefano, Ph.D. – Baylor College of Medicine, Houston, TX
Targeting RNA Condensates in Pediatric Myeloid Leukemia p>
Acute myeloid leukemia (AML), the second most common childhood leukemia, is an aggressive blood cancer driven by uncontrolled proliferation of immature white blood cells. Only 65% of children achieve long-term remission, despite intensive chemotherapy that results in harsh side effects and even death. We aim to discover alternative therapies by understanding what makes pediatric AML cells survive. Here, we found P-bodies to be crucial for pediatric AML cell survival. P-bodies are small particles within the cell that serve as reservoirs to store mRNAs and prevent them from guiding protein production. We discovered that a gene called DDX6, responsible for assembling P-bodies, is overexpressed in pediatric AML patients. Moreover, AML cells have more P-bodies compared to healthy blood cells. We further show that P-bodies are crucial for progression of pediatric AML. By isolating intact P-bodies from pediatric AML cells, we found that pediatric AML cells use them to hide mRNAs encoding key tumor suppressors, preventing them from exerting an anti-cancer effect. Therefore, targeting P-bodies could be a potential therapeutic approach for treating pediatric AML. Through our research, we aim to gain insights into how dysregulation of DDX6 and P-body assembly sustains pediatric AML. By understanding these mechanisms, we hope to develop more effective and targeted treatments, improving the lives of young patients and reducing the severe side effects associated with current therapies. p>
Dr. William Carroll, M.D. – New York University Grossman School of Medicine, New York, NY
The Role of S100A8/A9-IL6R in B-ALL Chemoresistance p>
In spite of dramatic improvements in outcome for B acute lymphoblastic leukemia (B-ALL) it remains a leading cause of cancer associated death in children and treatment is associated with short and long-term side effects. In an effort to understand how B-ALL cells evade therapy and result in relapse, we have studied genetic and epigenetic differences between blasts at diagnosis (sensitive) and relapse (resistant). Our work and results from others have identified key pathways that leukemia cells use to overcome treatment. Targeting these pathways will lead to more effective and potentially less toxic forms of treatment. We now have significant preliminary data showing that a majority of relapsed B-ALL cells express an inflammatory signature with overexpression of S100A8/A9 and IL6R that results in a protective niche within the bone marrow. The availability of agents already in clinical trials that block these pathways highlights the immediate clinical significance of this work. p>
Dr. Liron Grossmann, M.D. – The Sheba Fund for Health Service and Research, Israel
Epitranscriptomic mechanisms of therapy resistance in high-risk neuroblastoma p>
Neuroblastoma is a fatal childhood cancer with approximately 50% chance of recurrence despite multiple lines of aggressive therapies. Therefore, new therapies are urgently required to fight this devastating cancer. Our group has recently identified and characterized the malignant cells that survive chemotherapy in neuroblastoma patients. We found that neuroblastoma cancer cells may alter the levels of proteins which allow them to resist chemotherapy. They do so by chemically changing certain RNA molecules, which are derivatives of the DNA. The altered RNA molecules lead to changes of the these important proteins, which result in drug resistance. In this project, we ask: "how does chemotherapy change the chemical structure of RNA molecules in a way that allow neuroblastoma cells to survive instead of dying?". Together with our collaborators, who are world experts in RNA chemical modifications, we will use advanced technologies to study the differences in RNA chemistry before and after chemotherapy and identify the proteins that allow neuroblastoma cells to escape chemotherapy. If successful, our results may lead to the discovery of new therapies that prevent the RNA molecules from being chemically changed and kill the remaining neuroblastoma cells. Thus, our project has the potential of changing the way we currently treat children with neuroblastoma and improve their chances of surviving. p>
Dr. Hiroto Inaba, M.D., Ph.D. – St. Jude Children's Research Hospital, Inc., Memphis, TN
A PHASE 1 STUDY OF REVUMENIB, AZACITIDINE, AND VENETOCLAX AND IN PEDIATRIC AND YOUNG ADULT PATIENTS WITH REFRACTORY OR RELAPSED ACUTE MYELOID LEUKEMIA p>
Although survival rates for children with AML are greater than 60%, the outcome for patients with relapsed or refractory disease remains poor, with less than 40% of these patients becoming long term survivors. Therefore, novel approaches are urgently needed, both for the treatment of patients who have relapsed and for incorporation into treatment regimens for newly diagnosed patients. Because AML is a genetically and biologically diverse group of diseases, a rational approach is to develop and test novel therapies in patients who are predicted to respond to those agents. Exciting and extensive preclinical data strongly suggest that menin inhibitors will be active in patients with specific genetic alterations. Preclinical data also demonstrate that the combination of a menin inhibitor with a BCL2 inhibitor is synergistic, as is the combination of a BCL2 inhibitor with a hypomethylating agent. Our proposed RAVAML trial will be the first pediatric trial to test the combination of all three classes of agents, including revumenib (a menin inhibitor), azacitidine (a hypomethylating agent), and venetoclax (a BCL2 inhibitor). We strongly believe that this trial will facilitate the development of menin inhibitor therapy for children with AML and will help move this novel and effective therapy into the frontline setting, ultimately changing the treatment paradigm for children with AML. p>
Dr. Loretta Li, M.D. – Ann & Robert H. Lurie Children's Hospital of Chicago, Chicago, IL
Validating PRL2 as a New Therapeutic Target in T-ALL p>
Acute lymphoblastic leukemia (ALL) is the most common form of cancer in children and T-cell ALL (T-ALL) is a high risk subtype of this disease that accounts for 10-15% of pediatric cases. Changes in the PI3K/PTEN/AKT signaling pathway are often seen in T-ALL and this is associated with reduced disease free and overall survival. PTEN is a tumor suppressor gene that inhibits PI3K/AKT signaling, but its expression is often decreased in cancer. PRL2, in contrast, is a gene that is highly expressed in T-ALL cells. This gene can inhibit PTEN, thus promoting increased cell proliferation and survival. We hypothesize that PRL2 promotes T-ALL development by inhibiting the PTEN tumor suppressor pathway. In Aim 1, we will assess how PRL2 loss alone or in combination with PTEN loss affects overall survival in mouse models of T-ALL. We will also evaluate how increased or decreased PRL2 expression affects T-ALL cell growth and signaling through the PI3K/PTEN/AKT pathway. In Aim 2, we will assess the effectiveness of a PRL2 inhibitor against primary T-ALL patient samples and in mouse models of T-ALL. We will evaluate how chemical inhibition can affect T-ALL cell growth and signaling and determine whether treatment with a PRL2 inhibitor can prolong overall survival in human T-ALL cells grown in mice. Through this work, we hope to learn more about how PRL2 and PTEN contribute to T-ALL development and validate PRL2 as a new therapeutic target in T-ALL.
Dr. Yanxin Pei, M.D. – Children's Research Institute (CNMC), Washington, D.C.
Developing Human Cerebellar Organoid Models of Group 4 Medulloblastoma to Identify the Cell of Origin and Mechanisms that Support Oncogenesis p>
Our research will study the mysteries behind Group 4 Medulloblastoma (MB), a form of brain cancer commonly found in children. MB tumor cells originate from a specific set of cells in the developing brain that rely on interactions with other early normal brain cells to fuel their growth. To unravel the mysteries of Group 4 MB, we aim to 1) pinpoint the exact source of the tumor cells using miniature brain models known as organoids and 2) investigate which normal brain cells play a role in supporting the growth of these tumor cells. We will create a scenario where the tumor and normal brain cells interact and observe the outcomes closely. By successfully addressing these two aspects, we expect to gain crucial insights into the nature of Group 4 MB, potentially discovering novel avenues for therapeutic interventions. Our efforts are ultimately geared towards enhancing the prognosis and treatment strategies for children affected by Group 4 MB. p>
Dr. John Prensner, M.D., Ph.D. – University of Michigan Medical Center (Michigan Medicine), Ann Arbor, MI
Preclinical evaluation of omacetaxine as a novel therapeutic for ATRT p>
Children with atypical teratoid/rhabdoid tumor (ATRT), an aggressive brain cancer mainly afflicting infants and toddlers, frequently relapse and die from disease, despite receiving intensive treatment. As such, developing rational and effective therapeutic approaches for ATRT is a pressing clinical need. Unfortunately, extensive efforts to study ATRT have not shown genetic alterations that are readily targetable with therapeutic agents. Therefore, to achieve better treatments for children with ATRT, new therapeutic approaches based on the biology of this disease are needed. We have found that ATRT cells exhibit an exaggerated reliance on the cellular process of protein translation, which is how cancer cells convert genetic material (DNA and RNA) into functional proteins that perform cellular jobs. Moreover, protein translation is targetable, and one drug, named omacetaxine, is already used in adult cancer patients with leukemia. We have found that omacetaxine is highly effective at killing ATRT cells. The purpose of this grant is to evaluate this omacetaxine for the treatment of mouse models of ATRT. We hypothesize that treatment with omacetaxine will result in increased survival for mouse models of ATRT. Thus, successful completion of this proposal will provide the pre-clinical data necessary to support a clinical trial testing omacetaxine in children. Our overall goal is to translate the findings of this research into a clinical trial within the next 3-4 years. p>
Dr. Kristopher Sarosiek, Ph.D. – President and Fellows of Harvard College, Harvard T. H. Chan School of Public Health, Boston, MA
Targeting apoptosis to prevent cancer therapy-induced cardiovascular dysfunction in pediatric patients p>
Cancers commonly arising in children including leukemias, lymphomas and solid tumors of the nervous system are a leading cause of death and long-term disability. These cancers are typically treated with radiation and chemotherapies, which can control or even eradicate tumors in pediatric patients. However, these treatments can also severely damage cells within the developing heart and vascular system, causing life-long cardiovascular dysfunction including chronic heart failure and stroke. We recently discovered that cell death is regulated differently in our varying healthy tissues during development and early childhood. In fact, cells within the developing heart and vasculature are highly prone to dying via apoptosis, which may explain why many childhood cancer survivors experience cardiovascular disease. However, it is unclear which cells within the developing heart and vasculature are particularly at risk of dying in response to cancer therapies and how the loss of each cell type contributes to long-term dysfunction. Our proposal will build our understanding of how the regulation of apoptosis in young heart and vasculature affects both short-term therapy induced toxicity as well as long-term dysfunction. In addition, our studies will test the extent to which blocking cell death in healthy tissue will prevent therapy-induced dysfunction to enable the development of treatments that will reduce cardiovascular toxicity and improve the lives of pediatric cancer patients. p>
Dr. Palaniraja Thandapani, Ph.D. – University of Texas M.D. Anderson Cancer Center, Houston, TX
Dietary valine modulation to enhance therapies for high-risk T-ALL p>
Acute lymphoblastic leukemia (ALL) is the most common type of childhood cancer with more than 3000 children/adolescents under the age of 20 diagnosed with ALL each year in the United States. ALL is a disease that affects a type of white blood cells called lymphocytes that help the body fight infection and disease. ALL can be broadly divided into either B-ALL or T-ALL. B-ALL affects a type of lymphocytes called B-lymphocytes whereas T-ALL affects T lymphocytes. Historically children with T-ALL have worse prognosis than B-ALL. B-ALL also have better therapeutic options available including targeted and immunotherapies, whereas children with T-ALL are limited to therapies with well-documented long-term negative effects like chemotherapy, radiation therapy. Hence, better therapies that kill leukemic cells without harming normal cells is the need of the hour in T-ALL. In this proposal, we aim to evaluate a new therapeutic approach of nutrient deprivation to treat T-ALL grounded on our strong preliminary finding that T-ALL cells need high levels of the nutrient valine for their growth and survival. Our project investigates plausible different avenues of using this approach in combination with currently used chemotherapy and with new drugs in T-ALL treatment. The goal of our project is to identify therapy combinations that willm increase the efficiency of current treatments in killing the leukemia cells to prevent relapse and to provide new options for treatment of high-risk disease. p>
Dr. Norihiro Watanabe, Ph.D. – Baylor College of Medicine, Houston, TX
CAR gamma delta T cell-based multi-pronged immunotherapy for AML p>
Acute myeloid leukemia (AML) is the second most common type of leukemia in children and remains very deadly in patients with treatment-resistant disease. In other blood cancers, immunotherapy with patients’ immune cells (called T cells) that are genetically modified to express cancer-finding proteins called CARs has been very effective. However, extending this approach to AML is very difficult because cancerous cells frequently lose CAR targets thus becoming invisible to therapeutic cells. We will overcome this problem by utilizing a special subset of T cells, called gamma delta T cells. These cells can recognize and kill AML without a CAR, and therefore can be used to track down and eradicate cancer cells that lost the CAR target. Another advantage is that gamma delta T cells do not attack patients’ healthy tissue, and therefore can be generated from healthy individuals and infused into multiple patients. Here, we will establish a safe and effective CAR gamma delta T cell platform that we will manufacture from healthy donors and equip these cells with receptors to avoid immune rejection in AML patients. If successful, this will lead to a new therapeutic option available off-the-shelf for patients with difficult-to-treat AML. p>
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