Development and Cancer
54 members representing 18 departments at UT Southwestern
The Development and Cancer (DC) Program explores human cancer through the study of topics ranging from cell growth, fate, and differentiation to organism growth and pathophysiology to organ development—highlighting the importance of aberrant developmental programs in cancer pathogenesis. It also exploits new cellular and organismal models to understand cancer initiation and progression. Merging such tools with cutting-edge biochemistry, high-throughput screening, and genomics provides unparalleled opportunities to drive fundamental discovery toward clinical applications.
The overall goals of the DC Program are to foster collaborative research at the intersection of developmental and cancer biology and to move these discoveries toward the clinic to improve the outcomes of cancer patients in the SCCC catchment area and beyond. DC Program members also lead ground-breaking research in childhood cancers, where normal developmental processes are known to be derailed. There is a focus on priority cancers in the SCCC catchment area—lung, liver, colorectal, and kidney cancers. The DC Program’s overarching hypothesis is that the investigation of fundamental developmental processes will shed new light on the molecular underpinnings of human cancer and uncover new therapeutic strategies.
Systematic Analyses of Stem Cell Biology, Regeneration, and Cancer
Identification of noncoding RNAs that contribute to cancer pathogenesis
Cellular senescence is an irreversible state of cell-cycle arrest induced by oncogene activation, telomere shortening, or DNA damage. This anti-proliferative response is an important tumor suppressor mechanism. While the proteins that mediate senescence, such as p53 and RB, have been highly studied, the role of noncoding RNAs in senescence remains poorly understood. To address this knowledge gap, Joshua Mendell's, Ph.D., laboratory conducted a genome-wide CRISPRi screen in human cells, revealing an unexpected and essential role for the small nucleolar RNA (snoRNA) SNORA13 in senescence. Mendell discovered that SNORA13 promotes senescence by increasing the abundance of non-ribosome associated ribosomal proteins, which activates p53 in the presence of senescence-inducing stress. These findings suggest that delivery of SNORA13 could therapeutically induce senescence in cancer cells.
Exploration of Cell Fate, Cell Differentiation, and Organogenesis
Understanding how fat cells modulate cancer risk
How fat promotes cancer is a central question in cancer biology. Philipp Scherer's, Ph.D., laboratory recently reported that adipocytes from tumor-invasive mammary fat undergo de-differentiation to fibroblast-like precursor cells that integrate into the tumor microenvironment. This showed that the metabolic interplay between tumor cells and adipocytes induces an adipocyte mesenchymal transition that contributes to a tumor-promoting microenvironment. These insights introduce new ways of manipulating cells in the tumor microenvironment in patients with obesity-associated cancers, a research priority in the SCCC catchment area.
Analyzing Cell-Cell and Cell-Stroma Interactions
Understanding how the gut microbiome influences antitumor immunity
While the gut microbiome influences most aspects of human health, how it impacts cancer treatment is unclear. Andrew Koh's, M.D., laboratory demonstrated that immune checkpoint blockade induces the relocation of gut microbiota to lymphoid organs and melanomas. This microbiota promoted stronger immune responses against cancer, and antibiotic treatment that depletes gut microbiota reduced T cell responses and limited the effectiveness of immune checkpoint inhibitors for cancer. These findings revealed a key mechanism by which gut microbiota promote anticancer immunity outside of the gut and led to a multicenter Phase I/II trial testing immunotherapy combinations in children with solid tumors.
Understanding the genetic basis of immunotherapy responses in cancer
One of the critical shortcomings of genetically engineered mouse models of cancer is that they do not generate abundant somatic mutations, which are thought to induce antigen production and stimulate immune responses. Esra Akbay's, Ph.D., laboratory developed novel murine cancer models with extremely high mutation levels. These cancers were highly sensitive to immune checkpoint blockade and led to the insight that specific cancer genes such as TP53 can dramatically influence immunogenicity. This work allowed for the identification of critical factors that explain immunotherapy’s effectiveness in the clinic. Using these approaches, the Akbay lab showed that STING agonists and telomerase inhibitors are small molecules that can enhance immunotherapy in lung cancer, two strategies that are being tested in clinical trials.
Notable Publications
O'Donnell, K.A. et al. The Integrated Stress Response Pathway Coordinates Translational Control of Multiple Immune Checkpoints in Lung Cancer. Cancer Res. 2025; doi: 10.1158/0008-5472.CAN-24-3844. Online ahead of print.PMID: 40327600
Zhu, H. et al. Positive selection of somatically mutated clones identifies adaptive pathways in metabolic liver disease. Cell 2023; 186(9):1968-1984.e20. PMCID: PMC10321862.
Morrison, S.J. et al. Non-selective β- adrenergic receptor inhibitors impair hematopoietic regeneration in mice and humans after hematopoietic cell transplants. Cancer Discov 2024; PubMed PMID: 39786370.
Carroll, T.J. et al. Deletion of Lats1/2 in adult kidney epithelia leads to renal cell carcinoma. J. Clin Invest 2021; 131(11). DOI: 10.1172/JCI144108. PubMed PMID: 34060480; PMCID: PMC8159698.
Olson, E.N., Lui, N. et al. TWIST2-mediated chromatin remodeling promotes fusion-negative rhabdomyosarcoma. Sci Adv 2023 ; 9(17): eade8184. PMCID: PMC10146891.
Leadership and Contact

