437 results

Refine Your Search

Samantha Tauchmann
Oregon Health & Science University Cancer Institute

This study aims to explore how mutant SETBP1 affects histone methyltransferase complexes to drive leukemia-associated gene transcription. I will use biochemical, imaging, and epigenetic methods to assess the effects of SETBP1 mutations on complex formation, genomic localization, and function. I will evaluate if inhibitors can disrupt SETBP1-driven oncogenesis in human leukemia cell lines, hematopoietic cells, and patient samples to identify novel therapeutic targets in SETBP1-mutant leukemias.

Project Term: July 1, 2025 - June 30, 2028

Yue Wang

Children with Down Syndrome (DS) have a 30-fold increased risk of B-cell Acute Lymphoblastic Leukemia (B-ALL). We aim to identify the cells of origin in DS-B-ALL and define its unique features. Using scRNA-seq, we will create an immune cell atlas to study how trisomy 21 (T21) affects lymphopoiesis, and map the cellular and molecular heterogeneity in DS-B-ALL at disease onset and during relapse. These studies will help understand the B lymphoid defects in T21 and how they predispose to DS-B-ALL.

Project Term: July 1, 2025 - June 30, 2028

Qian Zhang
Sloan Kettering Institute for Cancer Research

Mutations affecting RNA splicing factors are the most common class of mutations in patients with myelodysplastic syndromes and related myeloid neoplasms. Although these mutations cause a gain of function, there are no treatments which selectively inhibit the enzymatic activity of the mutant spliceosome. To address this issue, here we have developed a new precision therapeutic that selectively target and eliminate cells carrying cancer-causing mutations affecting the RNA splicing factor U2AF1.

Project Term: July 1, 2025 - June 30, 2028

Michael Bern
Washington University in St. Louis

Approximately 25% of Acute Myeloid Leukemia (AML) patients are “Primary-Refractory” (P-R) and fail to go into remission with intensive induction chemotherapy. These patients have limited treatment options and overall survival <1 year. We will investigate mechanisms causing chemoresistance through multi-omic studies of a mouse model of P-R AML driven by Mecom overexpression. The goal of this project is to identify potential new therapeutic approaches for P-R AML patients.

Project Term: July 1, 2025 - June 30, 2028

Abhilash Barpanda
The Regents of the University of California, San Francisco

A lack of highly selective surface antigens for Acute Myeloid Leukemia (AML) immunotherapy is a major bottleneck in the development of both CAR T cells and T-cell engaging antibodies.  We aim to identify surface-exposed post-translational modifications (PTMs) unique to AML, using high-throughput LC-MS/MS based surfaceomics. By focusing on these distinct PTMs, we hope to develop precision immunotherapies that eliminate AML cells with minimal off-target effects, improving patient outcomes.

Project Term: July 1, 2025 - June 30, 2028

Alba Rodriguez-Meira
Dana-Farber Cancer Institute, Inc.

Clonal hematopoiesis (CH) often precedes AML development, yet the molecular basis of CH expansion and progression to AML remains a mystery. By deploying single-cell DNA methylation analysis of longitudinal human in-vitro and in-vivo CH models, I aim to identify DNA methylation defects promoting CH fitness advantage, specifically in response to chronic inflammation. This will facilitate the design of therapies to halt premalignant clonal expansions and ultimately prevent leukemic transformation.

Project Term: July 1, 2025 - June 30, 2027

Christopher Hergott
The Brigham and Women’s Hospital, Inc.

Clonal cytopenia of undetermined significance (CCUS) is a poorly understood precursor condition linking clonal hematopoiesis with myeloid malignancy. Motivated by human biobank data, I developed a novel murine model of neutropenic CCUS and found interleukin-17A to be necessary and sufficient to propel Tet2-deficient clonal outgrowth. The objectives of this project are to define the drivers of interleukin-17A liberation in neutropenic CCUS and the mechanism by which it hastens clonal progression.

Project Term: July 1, 2025 - June 30, 2028

Sweta Patel
University of Colorado Denver, Anschutz Medical Campus

Myelodysplastic syndrome (MDS) is a fatal disease with limited therapeutic opportunities. To increase survival rate, it is essential to identify therapeutic targets specific for MDS stem and progenitor cells (MDS-SC), the source of the disease. MDS-SC uniquely upregulate nicotinamide metabolism. We thus aim to understand its importance on MDS-SC function and survival using multi-omics analysis. Completion of the study will have identified a new treatment modality to improve MDS patient outcome.

Project Term: July 1, 2025 - June 30, 2027

Haley Newman
The Children's Hospital of Philadelphia

Outcomes for children with T-ALL and T-LL have improved, yet prognosis for children with relapsed disease is dismal. A critical gap remains in identifying high-risk patients in order to allocate novel targeted therapies or immunotherapies. Building on prior work, I will utilize comprehensive genomic profiling to examine the impact of genetic ancestry on tumor biology and survival outcomes. My goal is to improve risk stratification, guide targeted therapy, and reduce inequity in T-ALL/T-LL.

Project Term: July 1, 2025 - December 31, 2025

Xiaodi Wu
Sloan Kettering Institute for Cancer Research

In-frame mutations affecting the basic leucine zipper (bZIP) domain of C/EBPα characterize a distinct subset of acute myeloid leukemia with relatively favorable prognosis, though five-year overall survival remains roughly 60%. Using new mouse models, this project will identify targets of endogenous bZIP–mutated C/EBPα. We will test whether modulating mutant-specific targets alters disease course in mouse models and in patient-derived xenografts, thereby nominating new approaches to therapy.

Project Term: July 1, 2025 - June 30, 2028

Chen Xi
Joan & Sanford I. Weill Medical College of Cornell University

EZB lymphomas, driven by BCL2 translocations and EZH2 mutations, induce macrophages to adopt a supportive role, essential for maintaining the malignant phenotype. This reprogramming suppresses phagocytosis and promotes pro-tumor activation, mediated by immune synapse signaling and cytokine release. Our study aims to identify molecular pathways involved in this macrophage reprogramming and explore restoring anti-tumor functions as a therapeutic approach.

Project Term: July 1, 2025 - June 30, 2027

Dai Chihara
The University of Texas MD Anderson Cancer Center

Improving survival for patients with diffuse large B-cell lymphoma requires tailoring treatment to lymphoma genetic heterogeneity, addressing minimal residual disease (MRD), and bringing safer, effective therapies for frail patients who cannot tolerate aggressive regimens. I address these challenges with genomic subtype targeted treatment, bispecific antibody to eliminate MRD, and novel treatment for frail patients. My goal is to improve outcomes by bringing biomarkers to standard of care.

Project Term: July 1, 2025 - June 30, 2030