Showing 1-20 of 420 results
Vanja Cabric M.D.
Funded: 07-01-2024 through 06-30-2026
Funding Type: St. Baldrick's Fellow
Institution Location: New York, NY
Institution: Memorial Sloan Kettering Cancer Center

Hepatoblastoma is the most common liver tumor diagnosed in early childhood, and new therapies are urgently needed to improve survival and reduce treatment related morbidity. Immunotherapy is a type of cancer treatment that harnesses the body's own immune system to target and attack cancer cells. While some immunotherapies have been very successful against certain tumor types in adult patients, they have been largely unsuccessful in treating pediatric tumors. This demonstrates how little we know about how the pediatric immune system responds to tumors. Using samples and models of hepatoblastoma, Dr. Cabric's research aims to identify the key immune cells involved in recognizing and responding to hepatoblastoma. Identifying the key immune cells involved in tumor immunity, and mechanisms that allow tumors to escape detection and deletion by the immune system, will allow us to find novel targets for future immunotherapies that work in children.

Sujatha Venkataraman Ph.D.
Funded: 10-01-2023 through 09-30-2025
Funding Type: Research Grant
Institution Location: Denver, CO
Institution: University of Colorado affiliated with Children's Hospital Colorado

Chimeric Antigen Receptor (CAR)-T cells have been clinically effective in patients with leukemias and lymphomas. Dr. Venkataraman’s goal is to bring similar success in treating a fatal brain tumor in children called DIPG (Diffuse Intrinsic Pontine Glioma). A major obstacle in treating brain tumors with CAR-T cell therapy is a lack of antigens which are tumor specific, or which are absent on normal vital tissues that can lead to off-target toxicities. To overcome this risk, Dr. Venkataraman and colleagues have successfully generated and tested the functionality of a novel “logic-gated” CAR-T cells targeting two distinct antigens, CD99 AND B7H3 that are highly expressed on DIPG but present singly on certain normal cells. This gated “AND” CAR-Ts will have full-activation against DIPG cells having both the antigens while sparing the single antigen expressing normal cells and will now investigate the safety, preclinical efficacy of these CAR-T cells against DIPG and evaluate its translational relevance to DIPG patients.

Mitchell Cairo M.D. 
Funded: 10-01-2023 through 09-30-2026
Funding Type: Research Grant
Institution Location: Valhalla, NY
Institution: New York Medical College affiliated with Maria Fareri Children's Hospital at Westchester Medical Center

Children, adolescents and young adults with recurrent or refractory Osteosarcoma have a very poor prognosis, with a dismal 6mo overall survival of less than 5%. Presumably, this poor prognosis is in large part secondary to the development of resistance to chemotherapy and radiation. More recent studies employing therapies that release and activate the patients’ immune cells, called T-cells, and even targeted T-cells have not improved this poor prognosis. Dr. Cairo proposes to investigate novel and innovative methods of combinatorial immunotherapy to circumvent known mechanisms of resistance. Together with colleagues, he proposes to investigate at the bench (in the laboratory) and in models with osteosarcoma alternative methods of combination immunotherapy including natural killer cells (NK) that we have been engineered in the laboratory to also circumvent mechanisms of resistance and to additionally express a single or dual target that are present on the osteosarcoma cells.

They further plan to investigate the efficacy of adding other immunotherapies to enhance the function and persistence of these targeted NK cells with antibodies, and two different NK activating cytokines. They will also investigate the optimal combination of this immunotherapy in children, adolescents and young adults with recurrent or refractory osteosarcoma to determine the safety and efficacy of this approach. Finally, Dr. Cario and team will determine what are the genetic and immune mechanisms of resistance after these novel combinatorial immunotherapy approaches utilizing state-of-the-art laboratory techniques. The goal of this grant is to develop novel combinatorial immunotherapy that will significantly increase the overall survival in children and adolescents with poor risk osteosarcoma.

To make a significant impact for kids fighting osteosarcoma, five funders have banded together with St. Baldrick’s to support this grant – The Helping Osteosarcoma Patients Everywhere (HOPE) Super grant supported by Battle Osteosarcoma, the Faris Foundation, the Zach Sobiech Osteosarcoma Fund of Children’s Cancer Research Fund, the Children’s Cancer Fund NY (supporting the Maria Fareri Children’s Hospital and New York Medical College) and Nationwide Children’s Hospital.

Iannis Aifantis Ph.D.
Funded: 09-01-2023 through 08-31-2025
Funding Type: Research Grant
Institution Location: New York, NY
Institution: New York University School of Medicine affiliated with NYU Langone Medical Center

CAR-T cell immunotherapies, treatments that use T cells constructed to recognize tumors and kill them, revolutionized how doctors treat children with B cell leukemia (B-ALL). These killer T cells recognize a specific protein expressed on the surface of the leukemic cells. Unfortunately, leukemia frequently relapses and often finds ways to "switch off" the expression of this protein, making T cells unable to track and kill them. This notion is called "antigen escape," as the tumor finds a way to escape the immune treatment. Dr. Aifantis plans to identify ways to avoid antigen escape by boosting the expression of the surface recognition protein. The study aims to validate such mechanisms in an organism using CAR-T cell models and sequencing patient cells. At the same time, Dr. Aifantis will design screens that will help identify surface antigen-specific regulators, so researchers can one day create combinatorial protocols using CAR-T cells and targeting specific antigen surface expression regulators.

Le Su Ph.D.
Funded: 07-01-2023 through 06-30-2026
Funding Type: St. Baldrick's Scholar
Institution Location: Jacksonville, AL
Institution: Jacksonville State University

Genes instruct cells to do their jobs through making specific proteins. In our body, all cells store this what-to-do manual in a set of higher-order structures called chromosomes. When chromosomes break off, the broken pieces sometimes exchange their places to build new chromosomes. These errors, known as translocations, could have no effect on our bodies, but in many cases they might cause problems as severe as cancer. Dr. Su's research focuses on learning how chromosomal translocations promote tumor formation in children and young adults, as well as looking for clinically useful approaches to correct their pathogenic activities and cure these deadly diseases.

Srividya Swaminathan Ph.D.
Funded: 07-01-2023 through 06-30-2026
Funding Type: St. Baldrick's Scholar
Institution Location: Duarte, CA
Institution: Beckman Research Institute of the City of Hope

Acute Lymphoblastic Leukemia (ALL) are aggressive cancers of B- and T- immune cells. ALL is most common in children but also affects adolescents and young adults. 90% of childhood ALL is curable. However, ~10% of children and ~30% of adolescents and young adults with ALL are not cured. To combat hard-to-treat ALL, Dr. Swaminathan will harness the body’s natural anti-cancer defense mechanism: a type of immune cell called a natural killer (NK) cell. He will also find defective NK cells in children with ALL. Those with fewer defective NK cells tend to survive longer and spend more of their lives free from disease compared to patients with high levels of abnormal NK cells. These findings will inform the development of NK cells as affordable therapies to cure pediatric ALL.

Ling Li Ph.D.
Funded: 07-01-2023 through 06-30-2025
Funding Type: Research Grant
Institution Location: Duarte, CA
Institution: Beckman Research Institute of the City of Hope

Each year, approximately 1000 Americans aged 20 years or younger are diagnosed with acute myeloid leukemia (AML). Currently, even the most effective targeted drug BCL2 inhibitor-venetoclax (VEN) cannot eradicate all leukemia cells. The remaining cells cause disease recurrence and result in a very low overall survival rate for AML patients. In preliminary studies, Dr. Li found that targeting an enzyme called ADSS2 promotes pediatric AML cells sensitivity to VEN induced mitochondrial apoptosis, resulting in a synthetic lethality. This study will ask how ADSS2 preserves AML cells fitness and test the effectiveness of a first-in-class ADSS2 inhibitor combined with VEN or other BCL2 family protein MCL1 inhibitor in models of AML. If successful, this could lead to a clinical trial with potential impact for childhood AML patients.

Jessica Tsai M.D., Ph.D.
Funded: 07-01-2023 through 06-30-2026
Funding Type: St. Baldrick's Scholar
Institution Location: Los Angeles, CA
Institution: Children's Hospital Los Angeles

Diffuse intrinsic pontine glioma (DIPG) is a deadly pediatric brain cancer, and there is a dire need to develop new therapeutic strategies to improve the terrible outcomes for these patients. Looking at genes that are turned on or off in a cancer can be helpful to figure out what is causing cancer growth. While looking at genes that are turned on in DIPG, Dr. Tsai found a gene called FOXR2 that is turned on at very high levels in a subset of DIPGs. FOXR2 is usually turned off, and turning on FOXR2 makes tumors grow very quickly. FOXR2 is actually capable of turning on an entire set of genes that are called ETS transcription factors (TFs). This is surprising as these genes have never been shown to be activated in DIPGs. Others have shown that ETS TFs can turn on the MAPK signaling pathway. Dr. Tsai also has found that FOXR2 is able to activate MAPK signaling. The goal is to determine exactly how FOXR2 and ETS TFs cooperate together to turn on MAPK signaling to make DIPGs grow. This grant was awarded at Dana-Farber Cancer Institute and transferred to Children's Hospital of Los Angeles.

A portion of this grant is generously supported by Griffin's Guardians, a St. Baldrick's partner. Griffin's Guardians was created by the Engles in memory of their son, Griffin. Their mission is to provide support and financial assistance to children battling cancer in Central New York, raise awareness about pediatric cancer and provide funding for research.

Emma Cantor M.D.
Funded: 07-01-2023 through 06-30-2024
Funding Type: St. Baldrick's Fellow
Institution Location: Los Angeles, CA
Institution: Children's Hospital Los Angeles

Osteosarcoma is the most common bone tumor in children yet the survival rate remains low, below 75%. Children who are born with or develop certain mutations or who have been exposed to radiation or chemotherapy are more likely to get this cancer. However, not enough is known about how osteosarcomas develop. To learn more, researchers must better understand how normal bone cells become osteosarcoma cells. Dr. Cantor and colleagues have previously seen that patients with this cancer have elevated serum levels of abnormal DNA sequences (repetitive element DNAs) that may affect how these cells behave. Dr. Cantor and colleagues are creating models that mimic the cancer formation process to define the factors that drive the production of these abnormal DNA sequences and the effects of such sequences on the osteosarcoma cell behavior. Through these studies, Dr. Cantor hopes to learn more about this previously unrecognized abnormality. Better understanding of this process may allow researchers to develop new therapeutic approaches for children with osteosarcoma.

Christopher Kuo M.D.
Funded: 07-01-2023 through 06-30-2025
Funding Type: St. Baldrick's Fellow
Institution Location: Los Angeles, CA
Institution: Children's Hospital Los Angeles

Ewing sarcoma (EwS) is a malignant cancer of bone and soft tissues that occurs mainly in children, adolescents and young adults. If the tumors spread, fewer than 1/3 will survive. For some pediatric cancers, recent progress has led to new treatments that use one's own immune system to target cancer cells. However, immunotherapy has not been successful for EwS because we don't know enough about how EwS tumor cells evade the immune system. The tumor microenvironment (TME) is an intricate ecosystem consists of cancer cells and the host's immune system. Dr. Kuo's project will focus on dissecting the TME of EwS, to understand how tumors develop. Using EwS tumors removed from pediatric patients during their cancer diagnosis and treatment, Dr. Kuo will use newly-developed techniques to map the TME and use a genetic model of EwS developed at CHLA to examine tumor/immune cell interactions in living tissue. The long-term goal of this work is to identify new treatment options for children with EwS.

This grant is funded by and named for The Shohet Family Fund for Ewing Sarcoma Research. In his freshman year of college, Noah was diagnosed with Ewing sarcoma. He endured many months of chemotherapy and had limb salvage surgery. Able to return to school, Noah had no evidence of disease for 2½ years until April 2018 when routine scans revealed he had relapsed. He passed away in May 2021 at the age of 25. Noah and his family were always passionate about the need for curative treatments for diseases of the AYA population. The Shohet family intends to raise funds for this Hero Fund in Noah's memory to find cures for Ewing sarcoma and to carry on his legacy of possibilities and hope.

Crystal Wang M.D.
Funded: 07-01-2023 through 06-30-2025
Funding Type: St. Baldrick's Fellow
Institution Location: Palo Alto, CA
Institution: Stanford University affiliated with Lucile Packard Children’s Hospital

Cancer cells compete with the body for food. Some cancer cells use fat to grow, spread, and hide in the brain. When cancer cells hide in the brain, it is hard for chemotherapy reach them due to the blood brain barrier, which allows cancers to come back when they come out of hiding. Dr. Wang and colleagues are investigating how childhood leukemia uses fat to survive in the brain and how drugs that starve leukemia of fat can kill leukemia cells hiding in the brain.

Robert Lindquist M.D., Ph.D.
Funded: 07-01-2023 through 06-30-2025
Funding Type: St. Baldrick's Fellow
Institution Location: San Francisco, CA
Institution: University of California, San Francisco affiliated with UCSF Benioff Children's Hospital

Children with the brain tumor ependymoma have high relapse rates and poor long-term survival. Treatment options for ependymoma are limited and there is no known effective chemotherapy. Dr. Lindquist is working to make a new model of this tumor, to study how the tumor forms and grows, and to test new therapies in this model and patient-derived tumors. The ultimate goal is to identify new therapies that will extend the lives of children with ependymoma.

Waitman Aumann M.D.
Funded: 07-01-2023 through 06-30-2026
Funding Type: St. Baldrick's Scholar
Institution Location: Atlanta, GA
Institution: Emory University affiliated with Aflac Cancer Center, Children's Healthcare of Atlanta, Children's Healthcare of Atlanta at Egleston

Leukemia, a cancer of the blood and bone marrow, is the most common cancer in kids. Over the last 60 years, great strides have been made in treating children with leukemia, and today, most leukemias are curable. However, certain leukemias are difficult to treat and have a poor prognosis. In order to better treat cancers, researchers seek to better understand the pathways by which cancer cells develop in order to identify medicines that target proteins in these pathways. Dr. Aumann and colleagues study the fusion protein CALM-AF10 which is found in some leukemias, and found that these leukemias have increased expression of a protein called SIX1. Dr. Aumann is studying how the SIX1 protein makes blood cells turn into leukemia cells, and is using two small molecule inhibitors in combination with other chemotherapy as potential new treatments for this and other leukemias. The hope is that the these studies will clarify the role of SIX1 in CALM-AF10 and other leukemias.

Alexander Kabanov Ph.D, D.Sci.
Funded: 07-01-2023 through 06-30-2025
Funding Type: Research Grant
Institution Location: Chapel Hill, NC
Institution: University of North Carolina at Chapel Hill affiliated with UNC Children's Hospital

Dr. Kabanov and colleagues propose an entirely new way to treat medulloblastoma, the most common malignant pediatric brain tumor. Current treatment requires radiation followed by a year of chemotherapy, fails almost half the patients, and can leave survivors with lifelong brain injury. Tiny extracellular vesicles called exosomes produced by a type of immune cell called macrophages were discovered to travel from the bloodstream into brain tumors. Dr. Kabanov will load exosomes with an agonist of Toll-like receptor to reprogram medulloblastoma tumor associated myeloid cells and enhance their tumoricidal properties. If successful, the therapy will improve medulloblastoma treatment by replacing the current radiation and chemotherapy with the one that is less toxic and more effective.

The first year of this grant is funded by and named for the Strong & Courageous Hero Fund, established in honor of Jonah. It celebrates his survivorship from medulloblastoma and his goofy, loving, inclusive personality. This fund was inspired by Jonah’s desire to help other kids with cancer and supports research of brain tumors and the multitude of challenges facing survivors post treatment.

Stella Davies MBBS, Ph.D.
Funded: 07-01-2023 through 06-30-2025
Funding Type: Research Grant
Institution Location: Cincinnati, OH
Institution: Cincinnati Children's Hospital Medical Center affiliated with University of Cincinnati College of Medicine

Children receiving bone marrow transplant can have serious complication such as bloodstream infections and graft versus host disease and some children die of these complications. Alteration of the bacteria in the gut by treatments including antibiotics is an important cause of these complications. In a previous study Dr. Davies and colleagues have tested the use of human milk to help keep gut bacteria healthy in very young children and found that this treatment worked. They are now studying a purified sugar from human milk, 2-FL that can be given easily as a medicine. Dr. Davies will also test a novel rapid urine test and a blood test to assess health of the gut bacteria during the study. Current tests require a stool sample and can take a long time. This trial will generate the data needed to perform a large-scale multi-center randomized clinical trial that will best prove how well this treatment works.

This grant is generously supported by the Rays of Hope Hero Fund which honors the memory of Rayanna Marrero. She was a happy 3-year-old when she was diagnosed with Acute Lymphoblastic Leukemia (ALL). She successfully battled ALL, but a treatment induced secondary cancer claimed her life at age eight. Rayanna had an amazing attitude and loved life. She, like so many kids facing childhood cancer, did not allow it to define who she was. This Hero Fund aspires to give hope to kids fighting cancer through research.

Amanda Campbell M.D., Ph.D.
Funded: 07-01-2023 through 06-30-2025
Funding Type: St. Baldrick's Fellow
Institution Location: Columbus, OH
Institution: Nationwide Children's Hospital affiliated with The Research Institute at Nationwide

The goal of this project is to engineer immune cells to target cancer, particularly a type of pediatric cancer called acute myeloid leukemia (AML). AML cells develop strategies to escape surveillance by the immune system. Despite current therapies, cancer cells are able to survive and progress. Natural killer (NK) cells play an important role in the immune response to cancer by recognizing and killing tumor cells. NK cell activity is regulated by activating and inhibitory receptors. Tumor cells express proteins that provide inhibitory signals to NK cells, blocking NK cell anti-tumor functions and allowing for tumor escape. Dr. Campbell and colleagues propose to tip the balance in favor of immune cell activation by knocking out a key NK cell inhibitory receptor, TIGIT. Dr. Campbell hypothesizes that eliminating NK cell TIGIT expression will remove inhibitory "brakes" on NK cell activation and enhance anti-tumor activity. The purpose of this study is to develop an effective cellular therapy for pediatric AML.

Jennifer Kalish M.D., Ph.D.
Funded: 07-01-2023 through 06-30-2025
Funding Type: Research Grant
Institution Location: Philadelphia, PA
Institution: The Children's Hospital of Philadelphia affiliated with University of Pennsylvania

Beckwith-Wiedemann Syndrome (BWS) is a cancer predisposition syndrome and patients with BWS have a significantly increased risk of developing hepatoblastomas. The same genetic changes on chromosome 11 that cause BWS are found in 40% of hepatoblastomas. Dr. Kalish has previously shown that noncancerous liver and HB tissue from patients with BWS have distinct molecular signatures, suggesting the events that set up patients with BWS for HB are due to these changes on chromosome 11. Using the largest BWS collection of tissues worldwide, Dr. Kalish and colleagues will study the specific features of BWS and nonBWS liver cells and HB cells to determine how the changes of chromosome 11 lead to HB. Cell models derived from liver tissue will be used to test how these changes cause the transition from normal liver to HB. This work is a critical step in developing targeted therapies for patients with HB.

Anand Bhagwat M.D., Ph.D.
Funded: 07-01-2023 through 06-30-2025
Funding Type: St. Baldrick's Fellow
Institution Location: Philadelphia, PA
Institution: The Children's Hospital of Philadelphia affiliated with University of Pennsylvania

Acute Myeloid Leukemia (AML) is a blood cancer that sadly takes the lives of many children each year, and major efforts are being made to save these lives. One idea has been to teach the patient's body to fight off the AML like it would fight off an infection. This strategy alters the patient's immune system by making CAR-T cells, which are cells that fight cancer. CAR-T cells have been successful in curing patients with another similar type of blood cancer, but when it was tried in patients with AML, the approach was less successful. Dr. Anand's project is to understand why it didn't work as well so that further improvements that lead to cures for kids with AML can be made.

Patrick Grohar M.D., Ph.D.
Funded: 07-01-2023 through 06-30-2026
Funding Type: Research Grant
Institution Location: Philadelphia, PA
Institution: The Children's Hospital of Philadelphia affiliated with University of Pennsylvania

There is an urgent need for novel targeted therapy for osteosarcoma (OS). Molecular targeted therapy has yielded stunning response rates of >90% for specific targets such as NTRK gene fusions. In contrast, the mainstay of therapy for osteosarcoma has remained the same for more than 30 years and few targeted agents have successfully translated to the clinic for OS patients.

The challenge to develop targeted therapies for osteosarcoma is that the tumor has different driver mutations in different patients. However, 12-39% of tumors share a common amplification in a gene called MYC. In this project, Dr. Grohar and colleagues will consider this subset of osteosarcoma as a distinct entity. They are characterizing the role of MYC in the pathogenesis of osteosarcoma and determining how MYC makes osteosarcoma aggressive. Ultimately, they will identify compounds that will serve as clinical candidates for MYC-driven osteosarcoma. They will then determine if they are best translated to the clinic as single agents, in combination with chemotherapy, or as metastasis-targeted therapy.

Dr. Grohar has assembled a team with the necessary expertise. Chand Khanna is a disease expert in osteosarcoma, the Grohar lab has expertise in drug development for bone sarcomas, the Neamati and O'Keefe labs are experts in drug discovery/chemical biology/natural products. Filemon Dela Cruz is an expert in preclinical drug modeling, Ted Laetsch and Rashmi Chugh are experts in sarcoma clinical trial design, and Ethos (led by Chand Khanna) and Vuja De (led by David Warshawsky) are companies that will aid in the development of these compounds for the clinic. Together they will identify compounds that are specific and effective for MYC amplified osteosarcoma.

To make a significant impact for kids fighting osteosarcoma, five funding partners have banded together with St. Baldrick’s to support this grant – The Fight Osteosarcoma Together (FOT) Super Grant supported by Battle Osteosarcoma, CureSearch for Children’s Cancer, Michael and April Egge, The Osteosarcoma Collaborative, and the Zach Sobiech Osteosarcoma Fund of Children’s Cancer Research Fund.

Carl Allen M.D., Ph.D.
Funded: 07-01-2023 through 06-30-2025
Funding Type: Research Grant
Institution Location: Houston, TX
Institution: Baylor College of Medicine affiliated with Texas Children's Hospital, Vannie E. Cook Jr. Children's Cancer and Hematology Clinic

Pediatric Burkitt Lymphoma (BL) arises from errors during immune (B cell) development. Treatment failure is associated with dismal outcomes, and many pediatric BL survivors will suffer long-term toxicities from therapy, highlighting the need to explore opportunities to identify patients who may be cured with less intense therapies. Little is currently known about the biology of pediatric BL and clinical implications of specific mutations. Therefore, systematic analysis of tissue from children treated on clinical trials represents a unique opportunity to gain insights from valuable specimens to inform biologic risk-based chemotherapy and identify potential targeted therapeutic strategies. Dr. Allen will characterize intrinsic and acquired genetic factors that underlie pathogenesis and predict response to therapy in patients with pediatric BL who have completed treatment on COG clinical trials.

This grant is funded by and named for Jack's Pack - We Still Have His Back, a St. Baldrick's Hero Fund. Jack Klein was a ten year old who loved life, laughing and monkeys. During his illness, his community of family and friends near and far rallied around him under the moniker "Jack's Pack". Their slogan was "We have Jack's Back". After Jack succumbed to Burkitt's Lymphoma, his "pack" focused their energy and efforts to funding a cure...just as Jack would have wanted.