Postdoctoral Research Fellow with expertise in translational therapies and molecular techniques. Contributed to the development of anti-IL-7R therapies for leukemia, with a focus on reducing treatment toxicity.
· Propose new avenues for targeted therapy in B-cell Acute Lymphoblastic Leukemia (B-ALL) by extending translational studies of anti-IL-7R, initially developed for T-cell Acute Lymphoblastic Leukemia (T-ALL).
· Investigate IL-7Rα (CD127) expression on B-ALL cells at the mRNA and protein levels, examining its association with aggressive central nervous system disease and its role in driving B-ALL in preclinical models.
· Evaluate the potential of IL-7R-targeted therapy as an alternative to current chemotherapy regimens, which are associated with severe toxicity and relapse.
· Develop and apply methodologies from T-ALL research to assess antibody-dependent cellular cytotoxicity (ADCC) in a panel of B-ALL cell lines using NK cells.
· Plan and conduct in vitro and in vivo experiments with B-ALL patient-derived xenografts (PDXs) treated with anti-IL-7R therapy.
· Work with Allterum Therapeutics, Inc., which has developed 4A10 as an IND for a clinical trial in relapsed pediatric T-ALL patients.
· Conducted advanced research on Acute Lymphoblastic Leukemia (ALL) to understand the genetic and molecular basis of childhood leukemias.
· Utilized functional genomics technologies, including single-cell RNA sequencing (scRNA-seq), Cellular Indexing of Transcriptomes and Epitopes by Sequencing (CITE-seq), Assay for Transposase-Accessible Chromatin using sequencing (ATAC-seq), flow cytometry, gene knockdown/knockout using shRNA and CRISPR, and transduction of primary and cell lines with lentivirus/retrovirus, to investigate genes related to ALL.
· Investigated the cooperation between oncogenes and tumor suppressor genes in leukemogenesis, linking genetic mutations and cellular perturbations to identify therapeutic targets for childhood leukemia treatment.
· Received the NIH Fellows Award for Research Excellence (FARE) twice, in 2021 and 2023, in recognition of high-quality and impactful research contributions.
· Contributed to the development of mouse models, including humanized mice, for studying the pathogenesis of viral infections such as dengue, chikungunya, Mayaro, yellow fever, Zika, and coronaviruses, though the models were not fully completed before my departure.
· Conducted tests of potential therapies for these viral infections, contributing to advancements in treatment strategies.