en-us Copyright (C) 2016 utsouthwestern.edu https://www.simmonscancercenter.org/ Simmons Cancer Center News Harold C. Simmons Comprehensive Cancer Center - Cancer Center Current News <![CDATA[Loss of vitamin C synthesis protects animals from schistosomiasis]]> Michalis Agathocleous, Ph.D.
Michalis Agathocleous, Ph.D., Assistant Professor in CRI and of Pediatrics, works with Ji Hyung Jun, Ph.D., Agathocleous Lab Senior Research Scientist at UT Southwestern.

DALLAS – Dec. 29, 2025 – Scientists at Children’s Medical Center Research Institute at UT Southwestern (CRI) have discovered a benefit of vitamin C deficiency: protection from a major parasitic disease. Their research suggests an explanation for the loss of the ability to synthesize vitamin C in some animals, including humans.

Michalis Agathocleous, Ph.D.
Michalis Agathocleous, Ph.D., is Assistant Professor in CRI and of Pediatrics at UT Southwestern.

Ascorbate, better known as vitamin C, is not required by most animals because they can synthesize it using a gene called L-Gulonolactone Oxidase (GULO). But GULO was lost in humans and some other species as they evolved, making ascorbate a vitamin – a necessary nutrient that must come from diet. Most scientists view this as a neutral trait loss because there have been no known benefits to vitamin C deficiency.

New CRI research published in the Proceedings of the National Academy of Sciences challenges this view by showing that losing the ability to synthesize vitamin C and becoming vitamin C deficient protects animals infected with schistosomes, a type of parasitic flatworm that needs vitamin C from its host to reproduce.

The research was conducted by the lab of Michalis Agathocleous, Ph.D., Assistant Professor in CRI and of Pediatrics, in collaboration with the labs of Jipeng Wang, Ph.D., Assistant Professor at Fudan University in Shanghai, and James J. Collins, Ph.D., Professor of Pharmacology at UT Southwestern and a Howard Hughes Medical Institute Investigator.

Vitamin C deficiency classically causes scurvy. Dr. Agathocleous discovered in 2017 that vitamin C deficiency promotes myeloid leukemia development, suggesting that the disadvantages of deficiency extend beyond scurvy into cancer development.

Other scientists have shown that vitamin C synthesis is an ancient metabolic pathway lost not only in some animals, but also in many parasites. Then in 2019, Drs. Wang and Collins discovered ascorbate was one of the vital elements necessary for schistosomes to lay eggs in a petri dish. 

Dr. Agathocleous said these discoveries led him to hypothesize that a host deficient in vitamin C could be protected from parasites that require vitamin C but cannot synthesize it.

Ji Hyung Jun, Ph.D.
Ji Hyung Jun, Ph.D., is a Senior Research Scientist in the Agathocleous Lab.

Ji Hyung Jun, Ph.D., Agathocleous Lab Senior Research Scientist, and CRI researchers studied normal mice, which can naturally synthesize ascorbate, compared with mice missing the Gulo gene. They found most normal mice infected with schistosomes died from schistosomiasis, but only 5% of mice without the Gulo gene died. Intermittent vitamin C intake reduced morbidity and mortality from schistosomiasis while preventing scurvy.

“Our work changed my view of vitamins. Vitamins have been studied for over a hundred years for their possible benefits, and vitamin deficiencies are, by definition, harmful,” Dr. Agathocleous said. “This research shows that having transient deficiency in a vitamin can be beneficial in an animal infected with a pathogen that requires the vitamin.”

Nearly 250 million people are affected with schistosomiasis, the disease caused when schistosomes penetrate human skin via contaminated water. Schistosomes live, sometimes for decades, in human blood vessels near the liver.

“We think the advantage of deficiency comes from the different timescales over which the worms need vitamin C versus the host,” Dr. Agathocleous said. “Worms lay eggs every day, whereas host disease due to deficiency takes months to develop. So, on balance, there is a benefit for an infected animal to be transiently deficient in vitamin C. 

“It is still possible that the loss of vitamin C synthesis was evolutionarily neutral, since we don’t have the tools to formally test for positive selection of GULO loss in ancient primates,” Dr. Agathocleous added. “But because schistosomiasis is so prevalent, and the survival benefit for infected animals is so strong, our results could explain why GULO was lost and ascorbate became a vitamin.”

Future Agathocleous Lab research will continue to investigate the role of vitamin C and effects of its deficiency in human diseases, including parasites and myeloid leukemia, a type of blood cancer that starts in the bone marrow and affects white blood cells. 

This research was funded by the Cancer Prevention and Research Institute of Texas (CPRIT), the American Society of Hematology, the Moody Foundation, The Welch Foundation, the National Institutes of Health, the National Key Research and Development Program of China, and the Fund of Fudan University and Cao’ejiang Basic Research.

Dr. Agathocleous is a CPRIT Scholar. He is also a member of the Cellular Networks in Cancer Research Program at the Harold C. Simmons Comprehensive Cancer Center at UT Southwestern.

Dr. Collins holds the Jan and Bob Bullock Distinguished Chair for Science Education and the Jane and Bud Smith Distinguished Chair in Medicine and is a Rita C. and William P. Clements, Jr. Scholar in Biomedical Research.

About CRI

Children’s Medical Center Research Institute at UT Southwestern (CRI) is a joint venture of UT Southwestern Medical Center and Children’s Medical Center Dallas. CRI’s mission is to perform transformative biomedical research to better understand the biological basis of disease. Located in Dallas, Texas, CRI is home to interdisciplinary groups of scientists and physicians pursuing research at the interface of regenerative medicine, cancer biology, and metabolism – relentless discovery toward the treatments of tomorrow.

X/Twitter | LinkedIn | Instagram | YouTube | Facebook

About UT Southwestern Medical Center

UT Southwestern, one of the nation’s premier academic medical centers, integrates pioneering biomedical research with exceptional clinical care and education. The institution’s faculty members have received six Nobel Prizes and include 24 members of the National Academy of Sciences, 25 members of the National Academy of Medicine, and 13 Howard Hughes Medical Institute Investigators. The full-time faculty of more than 3,200 is responsible for groundbreaking medical advances and is committed to translating science-driven research quickly to new clinical treatments. UT Southwestern physicians provide care in more than 80 specialties to more than 140,000 hospitalized patients, more than 360,000 emergency room cases, and oversee nearly 5.1 million outpatient visits a year.

]]>
https://www.utsouthwestern.edu/newsroom/articles/year-2025/dec-vitamin-c-synthesis-schistosomiasis.html Mon, 29 Dec 2025 08:32:00 -0600
<![CDATA[Stiffer colon could signal risk of early-onset colorectal cancer]]> 3D illustration shows tau proteins
This image represents the spatial analysis of an early-onset tumor. In the polygon above the “008” are fibroblasts (stained yellow). Above and below the polygon are nests of cancer. Note that the fibroblasts in this area are aligned, potentially creating a “highway” for the tumor cells to spread.

DALLAS – Dec. 23, 2025 – Increased stiffness of the colon, spurred by chronic inflammation, may encourage the development and progression of early-onset colorectal cancer (CRC), a study co-led by UT Southwestern Medical Center researchers suggests. The findings, published in Advanced Science, could lead to new ways to prevent and treat this deadly subset of CRC.

Emina Huang, M.D., M.B.A.
Emina Huang, M.D., M.B.A., is Professor of Surgery in the Division of Colon and Rectal Surgery and Executive Vice Chair of Research for Surgery at UT Southwestern. Dr. Huang is also Professor of Biomedical Engineering and in the Harold C. Simmons Comprehensive Cancer Center.

“We consider this study a significant advancement toward identifying those at risk of early-onset CRC and finding new ways to treat them,” said Emina Huang, M.D., M.B.A., Professor of Surgery in the Division of Colon and Rectal Surgery and Executive Vice Chair of Research for Surgery at UT Southwestern. She is also Professor of Biomedical Engineering and in the Harold C. Simmons Comprehensive Cancer Center.

UT Southwestern partnered with researchers from The University of Texas at Dallas on the study.

“This is the first study to highlight the key role of biomechanical forces in the pathogenesis of early-onset CRC,” said Jacopo Ferruzzi, Ph.D., Assistant Professor of Bioengineering at UT Dallas and Biomedical Engineering at UT Southwestern. “Our observations are consistent across multiple length scales and link connective tissue stiffening to altered biochemical signaling in cancer cells.”

CRCs that are not caused by genetic syndromes and that occur at an average age of over 50 are known as average-onset or sporadic CRCs. The incidence and deaths from average-onset CRC have decreased over the last three decades. At the same time, the incidence and deaths from CRCs that occur before age 50, known as early-onset CRCs, have risen dramatically during the same period. Early-onset CRC now comprises about 12% of all CRCs diagnosed in the U.S. since 2020.

Jacopo Ferruzzi, Ph.D.
Jacopo Ferruzzi, Ph.D., is Assistant Professor of Bioengineering at UT Dallas and Biomedical Engineering at UT Southwestern.

The reason for this rapid increase is unknown. Most research in this area has focused on lifestyle, excess weight, and environmental exposures that could potentially drive CRC by causing chronic intestinal inflammation. However, why chronic inflammation might lead to early-onset CRC has been unclear.

Dr. Huang explained that chronic inflammation can cause scarring, gradually increasing the stiffness of tissues over time. Such stiffness is known to drive development and progression in some other cancer types, such as breast and pancreatic cancers. She and her colleagues wondered whether a similar phenomenon might spur early-onset CRC.

To answer this question, researchers worked with intestinal tissue from patients who underwent surgery to remove their cancerous tumors at William P. Clements University Hospital and Parkland Health: 19 samples from patients with average-onset CRC and 14 from patients with early-onset CRC. Each sample included not only malignant tumors but also their noncancerous margins. Tests showed that both the tumors and the noncancerous tissue were significantly stiffer in samples from patients with early-onset CRC compared with those from patients with average-onset CRC. These findings suggest that an increase in stiffness may have preceded early-onset CRC development.

Searching for a reason for this increased rigidity, researchers examined the collagen in both sample types, a protein that increases in abundance and changes conformation with scarring. They found that collagen in the early-onset samples was denser, longer, more mature, and more aligned than those in the average-onset samples. Those factors underscore the role of scarring in early-onset CRC tissue.

When scientists compared gene activity in the two sample types, they saw a significant increase in the expression of genes associated with collagen metabolism, blood vessel formation, and inflammation in the early-onset CRC tissues, further reinforcing that scarring from chronic inflammation is responsible for tissue stiffness. Importantly, they also noticed an uptick in a molecular pathway responsible for mechanotransduction, a process in which cells convert mechanical forces into biochemical signals. This suggests that cells in the early-onset CRC samples might change their behavior based on the stiffness of their environment.

Not surprisingly, when the researchers grew CRC cell lines on substrates with various levels of rigidity, they found that the cells multiplied quicker on stiffer substrates and increased rigidity. Similarly, three-dimensional organoid models made from CRC cells grew bigger faster on stiffer substrates.

Together, Dr. Huang said, these findings suggest that a more rigid environment might cause CRC to initiate and grow in those who develop early-onset CRC. They also reinforce the idea that disrupting mechanotransduction molecular pathways in these cells could halt or reverse CRC initiation and growth, a strategy currently being explored for some other cancers. Developing diagnostic tests to assess intestinal stiffness could help identify those at risk of early-onset CRC, Dr. Huang added, much like colonoscopies have done for average-onset CRC.

A complete list of UTSW contributors can be found in the study.

Dr. Huang holds the Doyle L. Sharp, M.D. Distinguished Chair in Surgical Research. She is a member of the Cellular Networks in Cancer Research Program at Simmons Cancer Center.

This study was funded by the National Institutes of Health (R01 CA234307 and U01 CA214300), The University of Texas at Dallas Office of Research and Innovation through the CoBRA program, the Burroughs-Wellcome Trust, the American Society of Colon and Rectal Surgeons Resident Research Initiation Grant, The University of Texas at Dallas Bioengineering Research Award, the UT Southwestern Whole Brain Microscopy Facility, an Axioscan 7 Award, the Catherine and James McCormick Charitable Foundation supporting research in early-onset colorectal cancer, and a National Cancer Institute (NCI) Cancer Center Support Grant (P30 CA142543).

About UT Southwestern Medical Center    

UT Southwestern, one of the nation’s premier academic medical centers, integrates pioneering biomedical research with exceptional clinical care and education. The institution’s faculty members have received six Nobel Prizes and include 24 members of the National Academy of Sciences, 25 members of the National Academy of Medicine, and 13 Howard Hughes Medical Institute Investigators. The full-time faculty of more than 3,200 is responsible for groundbreaking medical advances and is committed to translating science-driven research quickly to new clinical treatments. UT Southwestern physicians provide care in more than 80 specialties to more than 140,000 hospitalized patients, more than 360,000 emergency room cases, and oversee nearly 5.1 million outpatient visits a year.

About Parkland Health

Parkland Health is one of the largest public hospital systems in the country. Premier services at the state-of-the-art Parkland Memorial Hospital include the Level I Rees-Jones Trauma Center, the only burn center in North Texas verified by the American Burn Association for adult and pediatric patients, and a Level III Neonatal Intensive Care Unit. The system also includes two on-campus outpatient clinics – the Ron J. Anderson, MD Clinic and the Moody Outpatient Center, as well as more than 30 community-based clinics and numerous outreach and education programs. By cultivating its diversity, inclusion, and health equity efforts, Parkland enriches the health and wellness of the communities it serves. For more information, visit parklandhealth.org.

]]>
https://www.utsouthwestern.edu/newsroom/articles/year-2025/dec-stiffer-intestines-early-onset-colorectal-cancer.html Tue, 23 Dec 2025 08:00:00 -0600
<![CDATA[State’s investment in cancer research has helped draw top talent to UTSW]]> Joshua Gruber, M.D., Ph.D. and Matteo Ligorio, M.D., Ph.D.
Joshua Gruber, M.D., Ph.D., (left) Assistant Professor of Internal Medicine, and Matteo Ligorio, M.D., Ph.D., Assistant Professor of Surgery, both joined UT Southwestern after being awarded First-Time, Tenure-Track Faculty Member grants from the state-funded Cancer Prevention and Research Institute of Texas.

DALLAS – Dec. 19, 2025 – Early in his career, Ralf Kittler, Ph.D., attracted the attention of academic leaders at UT Southwestern Medical Center with his studies of DNA transcription factors and their role in tumor growth and suppression. His promising cancer research earned him an invitation to relocate to Dallas, where a $2 million grant from the state-funded Cancer Prevention and Research Institute of Texas (CPRIT) would help create his own lab at UTSW and turbocharge his scientific investigations.

Arriving from the University of Chicago in 2009, Dr. Kittler was the first of more than 300 highly sought-after scientists who have been recruited to Texas through the state’s multimillion-dollar program to advance the understanding and treatment of cancer.

In the more than 15 years since then, Dr. Kittler has become an Associate Professor at UT Southwestern’s Eugene McDermott Center for Human Growth and Development and the Harold C. Simmons Comprehensive Cancer Center as well as in the Department of Pharmacology. And CPRIT has provided more than $250 million in financial support to add faculty at UT Southwestern, giving it a competitive edge to attract some of the world’s most dynamic and in-demand cancer researchers.

>Ralf Kittler, Ph.D. and Robert Bachoo, M.D.
Ralf Kittler, Ph.D., (left) Associate Professor at UT Southwestern’s Eugene McDermott Center for Human Growth and Development, and Robert Bachoo, M.D., Ph.D., Associate Professor of Neurology, have studied ways to treat glioblastoma, a tumor that affects the brain and spinal cord.

This investment also has contributed to the foundational growth and success of Simmons Cancer Center, one of 57 NCI-designated Comprehensive Cancer Centers in the country and the only one in North Texas. Today, Simmons Cancer Center has 277 faculty members across 37 academic departments, runs hundreds of active clinical trials, and supports five research programs and 14 disease-oriented teams. UT Southwestern is also ranked by U.S. News & World Report as one of the top 20 hospitals for cancer care in the nation.

“It was clear from the start that CPRIT would be transformative for cancer research at UT Southwestern,” Dr. Kittler said.

Statewide, CPRIT’s impact has been equally profound. It has funneled nearly $1 billion to academic institutions, research organizations, and biomedical companies to bring the best and brightest scientists and clinical investigators to Texas. And it was all done with one bold mission in mind: to make the state a global leader in the fight against cancer.

Steering the future of cancer therapy

Created with voter approval in 2007, CPRIT began with a $3 billion investment to accelerate cancer research, support screening and preventive services, develop therapies, and recruit top talent to make it all possible. In 2019, Texans overwhelmingly supported a constitutional amendment to continue CPRIT’s work and infuse another $3 billion into the program. CPRIT has since become the largest state cancer research investment in U.S. history and the second-largest cancer research and prevention program anywhere. 

Carlos L. Arteaga, M.D.
Carlos L. Arteaga, M.D., is Director of the Simmons Cancer Center and Associate Dean of Oncology Programs at UT Southwestern.

“CPRIT has invested millions of dollars in our effort to screen for, prevent, and fight cancer, moving us closer every day to breakthrough therapies and life-changing medicines,” said Carlos L. Arteaga, M.D., Director of the Simmons Cancer Center and Associate Dean of Oncology Programs at UT Southwestern. Dr. Arteaga, who joined UTSW as the Center’s director in 2017 with a $6 million CPRIT recruitment grant, is an internationally renowned physician-scientist who has led the development and approval of molecularly targeted therapies for breast cancer. In 2024, he was elected to the National Academy of Medicine, one of the highest honors in the fields of health and medicine.

Academic institutions across the state have successfully pursued some of the most accomplished researchers to bring to Texas. Investigators have come from every corner of the U.S. and abroad, including countries in Europe, South America, and Asia. And the grants are awarded to scientists of all levels, from first-time, tenure-track junior faculty to mid-level associate professors to established senior researchers.

Among the most recent high-profile hires at UT Southwestern is Stefan Gloeggler, Ph.D., Professor in the Advanced Imaging Research Center and of Biomedical Engineering, who was recruited from the Max Planck Institute of Multidisciplinary Sciences in Göttingen, Germany. Dr. Gloeggler is a pioneer in hyperpolarized magnetic resonance imaging (MRI) technology, which can be applied in studies of cancer metabolism to improve disease detection and treatment.

Daniel Addison, M.D., former Director of the Cardio-Oncology Program at The Ohio State University, also joined the faculty at UT Southwestern through a CPRIT Rising Star recruitment award. Dr. Addison is Associate Professor of Internal Medicine, Director of Translational Research in the Division of Cardiology, and Associate Director for Survivorship and Outcomes Research in the Simmons Cancer Center. His research on the link between cancer treatments and cardiovascular disease has led to multicenter clinical trials that aim to eliminate or reduce such heart complications.

Most recently, Shixuan Liu, Ph.D., Assistant Professor of Neuroscience in the Peter O’Donnell Jr. Brain Institute, was recruited to UT Southwestern this year from Stanford University with the help of a $2 million CPRIT Scholar grant. Her lab’s research focuses on decoding the molecular mechanisms of the seasonal clock and its cross-talk with circadian rhythms.

Many early-career researchers who were brought to UT Southwestern through CPRIT have continued their path to great academic success.

Matteo Ligorio, M.D., Ph.D., Assistant Professor of Surgery and in the Simmons Cancer Center, arrived at UT Southwestern in 2020 from Harvard after he was awarded a CPRIT First-Time, Tenure-Track Faculty Member grant. In October 2025, Nature Medicine published a one-of-a-kind study he co-led that shifted the paradigm on the understanding of how cancer kills. His findings suggest the ultimate cause of cancer death is not metastatic disease, but the invasion of tumors into major blood vessels that lead to blood clots and multi-organ failure. With this new discovery, he and his co-author Kelley Newcomer, M.D., Associate Professor of Internal Medicine at UT Southwestern, are now collaborating with other researchers from around the world to design clinical trials that can test potentially more effective cancer therapies.

Just this month, the Texas Academy of Medicine, Engineering, Science & Technology (TAMEST) named Yunsun Nam, Ph.D., Professor of Biochemistry and Biophysics at UT Southwestern, as the winner of the prestigious 2026 Edith and Peter O’Donnell Award in Biological Sciences for her scientific achievements. Dr. Nam was also recruited to UT Southwestern as a first-time, tenure-track faculty member. Arriving in Dallas in 2012, Dr. Nam is widely recognized for her research on the molecular interactions of RNA and modifying proteins.

Financially backed by CPRIT and UTSW, these impactful researchers have the funding they need to purchase leading-edge lab equipment and hire the necessary staff to continue their pursuit of cancer breakthroughs.

“The resources you have when you start your career as a principal investigator are vitally important,” Dr. Kittler said.

Since his arrival, UT Southwestern has recruited more than 90 other experts with CPRIT support specializing in a variety of cancers – from liver cancer to ocular cancer to breast cancer to leukemia — as well as biomedical engineers and stem cell researchers, all of whom have made significant contributions to science.

Dr. Kittler himself was the co-leader of an investigation into how lentiviruses can mutate oncoproteins and render cancer cells resistant to drug therapy. By understanding the mechanisms at play and how to manipulate them, Dr. Kittler’s findings may unlock the development of more effective and targeted cancer treatments.

“CPRIT triggered a rapid growth of resources, talent, and collaboration soon after its start,” Dr. Kittler said. “It has been a massive stimulus to our university and exceeded expectations.”

Discoveries that have a lasting impact

Sean J. Morrison, Ph.D. and Julia Phan
Sean J. Morrison, Ph.D., (left) founding director of Children’s Medical Center Research Institute at UT Southwestern, works with Julia Phan, Ph.D., a former graduate student researcher and current student in the Medical Scientist Training Program at UT Southwestern.

In 2011, Sean J. Morrison, Ph.D., was recruited with a $10 million CPRIT grant to become the founding director of Children’s Medical Center Research Institute at UT Southwestern (CRI).

The nonprofit institute is focused on pioneering research at the intersection of stem cells, cancer, and metabolism. Since CRI’s inception, the internationally recognized team of scientists has made significant discoveries that improved the understanding of the biological basis of diseases, including cancer.

Dr. Morrison’s research has redefined strategies for cancer treatment. His studies in melanoma showed that antioxidants can promote disease progression and led to studies that are attempting to develop new pro-oxidant therapies. His work also uncovered the role of the bone marrow microenvironment, where blood-forming stem cells are located, leading to new insights that improved the safety of bone marrow and stem cell transplantation.

“CPRIT has profoundly strengthened cancer research in Texas because it accelerates medical science in a way that is not replicated in other parts of the country, where funding is difficult to obtain,” said Dr. Morrison, Professor in CRI and of Pediatrics at UT Southwestern and a member of the National Academy of Sciences, the National Academy of Medicine, and the European Molecular Biology Organization. Since 2000, he has also been a Howard Hughes Medical Institute (HHMI) Investigator. “Texas is the only state, aside from California, to make a multibillion-dollar commitment to science and to renew that investment after the initial term,” he said.

Exceptional reputation and vision drive progress

At the core of UT Southwestern’s mission is the commitment to enhance lives by developing better treatments, cures, and preventive care – a common goal shared by all CPRIT scholars.

Joshua T. Mendell, M.D., Ph.D.
Joshua T. Mendell, M.D., Ph.D., Professor of Molecular Biology at UT Southwestern and a Howard Hughes Medical Institute Investigator, studies how microRNAs contribute to oncogenesis and tumor suppression.

Joshua T. Mendell, M.D., Ph.D., Professor of Molecular Biology at UT Southwestern, member of the Cellular Networks in Cancer Research Program in the Simmons Cancer Center, and an HHMI Investigator, was also recruited with CPRIT support in 2011, after discovering that microRNAs can be modulated to inhibit liver cancer in mouse models. At UT Southwestern, Dr. Mendell and his lab continue to investigate how these noncoding molecules contribute to oncogenesis and tumor suppression.

“Our goal is to advance our understanding of RNA biology and to discover new functions for RNAs, because these molecules play critical roles in normal biology and often go awry in cancer and other diseases,” said Dr. Mendell, who, in October 2025, was elected to the National Academy of Medicine.  “Because of this, there is a strong interest in developing medicines based on RNA – the most famous and successful example, in recent times, being the COVID-19 vaccine.”

In fact, it is the same field of research that was awarded the Nobel Prize in Physiology or Medicine in both 2023 and 2024.

“UT Southwestern has always recognized the value of basic science,” Dr. Mendell said. “The research conducted on this campus has repeatedly demonstrated how fundamental scientific discoveries can lead to new clinical innovations that impact the lives of patients. While UT Southwestern has grown since my arrival here, the institutional commitment to bold and collaborative research has also continued.”

Recent CPRIT grants support AI research, lung and cervical cancer prevention programs

CPRIT awards grants not only to recruit scientists to Texas but also to fund research and cancer prevention efforts. In November, CPRIT announced its latest round of awards, including nearly $15 million in grants to UT Southwestern faculty members for programs ranging from lung cancer screening and tobacco cessation to research related to artificial intelligence (AI) and advanced imaging. In addition, recruitment grants for the year totaled $10 million to attract tenure-track faculty members to UTSW.

A sampling of significant CPRIT grants funded to UTSW in 2025 include:

  • Nearly $3 million to Kevin Dean, Ph.D., Assistant Professor in the Lyda Hill Department of Bioinformatics, to establish the Cancer Cell Imaging Core (CCIC), a facility to visualize and analyze cancer cells in unprecedented detail.
  • Nearly $3 million to Guanghua Xiao, Ph.D., Professor in the Peter O’Donnell Jr. School of Public Health, the Department of Biomedical Engineering, and the Lyda Hill Department of Bioinformatics, to establish a Data Science and AI Core for Population Research (DAICOR) in Texas.
  • Nearly $2.5 million to Keith Argenbright, M.D., Director of the Moncrief Cancer Institute, Professor and member of the Population Science and Cancer Control Research Program in the Simmons Cancer Center, and Professor in the O’Donnell School of Public Health, to implement a cervical cancer screening program in North Texas.
  • Nearly $2.5 million to David Gerber, M.D., Professor of Internal Medicine and Co-Director of the Simmons Cancer Center’s Office of Education and Training, to advance existing integrated lung cancer screening and tobacco cessation programs – with efforts focused on populations in southern Dallas County.

Dr. Addison holds the Audre and Bernard Rapoport Chair in Cardiovascular Research.

Dr. Argenbright is a Distinguished Teaching Professor.

Dr. Arteaga holds the Annette Simmons Distinguished University Chair in Breast Cancer Research.

Dr. Gerber holds the David Bruton, Jr. Professorship in Clinical Cancer Research.

Dr. Kittler is the John L. Roach Scholar in Biomedical Research.

Dr. Mendell holds the Charles Cameron Sprague, M.D. Chair in Medical Science.

Dr. Morrison holds the Kathryne and Gene Bishop Distinguished Chair in Pediatric Research at Children’s Research Institute at UT Southwestern and the Mary McDermott Cook Chair in Pediatric Genetics.

Dr. Nam holds the Doris and Bryan Wildenthal Distinguished Chair in Medical Science and is a Southwestern Medical Foundation Scholar in Biomedical Research and a UT Southwestern Presidential Scholar.

Dr. Xiao holds the Mary Dees McDermott Hicks Chair in Medical Science.

About UT Southwestern Medical Center

UT Southwestern, one of the nation’s premier academic medical centers, integrates pioneering biomedical research with exceptional clinical care and education. The institution’s faculty members have received six Nobel Prizes and include 24 members of the National Academy of Sciences, 25 members of the National Academy of Medicine, and 13 Howard Hughes Medical Institute Investigators. The full-time faculty of more than 3,200 is responsible for groundbreaking medical advances and is committed to translating science-driven research quickly to new clinical treatments. UT Southwestern physicians provide care in more than 80 specialties to more than 140,000 hospitalized patients, more than 360,000 emergency room cases, and oversee nearly 5.1 million outpatient visits a year.

]]>
https://www.utsouthwestern.edu/newsroom/articles/year-2025/dec-cprit-recruitment-grants-cancer-research.html Fri, 19 Dec 2025 08:08:00 -0600
<![CDATA[UT Southwestern cell biologist to receive Maddox Award from TAMEST]]> Maralice Conacci-Sorrell, Ph.D. is the recipient of the 2026 Mary Beth Maddox Award and Lectureship award
Photo credit: UT Southwestern/TAMEST (Texas Academy of Medicine, Engineering, Science & Technology)

DALLAS – Dec. 18, 2025 – Maralice Conacci-Sorrell, Ph.D., Associate Professor of Cell Biology at UT Southwestern Medical Center, is the recipient of the 2026 Mary Beth Maddox Award and Lectureship from the Texas Academy of Medicine, Engineering, Science & Technology (TAMEST). Dr. Conacci-Sorrell is being honored for her pioneering research revealing how cancer cells harness nutrients to drive their growth and for creating targeted strategies to suppress untreatable cancers.

The Maddox Award, which recognizes women scientists in Texas who bring “new ideas and innovations to the fight against cancer,” was established in 2022 and named after former TAMEST Executive Director Mary Beth Maddox, who died from pancreatic cancer.

Since Dr. Conacci-Sorrell arrived at UT Southwestern in 2015, her research has uncovered how changes in cellular metabolism support uncontrolled growth in cancer and how these vulnerabilities can be targeted. Her work focuses on two fundamental processes: nutrient utilization and protein synthesis. By studying how cancer‑driving genes alter these pathways, she and her colleagues in the Sorrell Lab aim to identify strategies that disrupt tumor growth without harming normal tissues.

“Maralice has done highly innovative research in the cancer field and is an extraordinary educator and mentor,” said Steven Kliewer, Ph.D., Professor of Molecular Biology and Pharmacology at UTSW and a member of the National Academy of Sciences, who nominated Dr. Conacci-Sorrell for the award. “She is richly deserving of this honor.”

The Sorrell Lab discovered that certain cancers depend heavily on nutrients such as tryptophan to fuel their growth. In liver tumors, tryptophan can produce a metabolite that acts as a growth signal driving the cancer cells to multiply. Dr. Conacci-Sorrell and her colleagues showed in 2024 that removing this nutrient from the diet can halt tumor growth in mice, and adding the metabolite restored it – revealing a potential therapeutic target for liver cancer. In parallel, her studies on brain tumors revealed that blocking pyrimidine synthesis – the process cells use to make DNA and RNA building blocks – slows tumor growth even in drug-resistant forms of brain cancer.

“Dr. Conacci-Sorrell’s research has been pivotal in advancing our understanding of how cellular processes drive disease and uncovering strategies to address them,” said Helen Heslop, M.D., Mary Beth Maddox Award and Lectureship Committee Chair, Professor of Medicine and Pediatrics at Baylor College of Medicine, and member of the National Academy of Medicine. “Equally notable is her steadfast leadership and dedication to mentorship, cultivating pathways that open doors for the next generation of scientists.”

Dr. Conacci-Sorrell will be celebrated in February at the TAMEST 2026 Annual Conference: Pioneering Climate Innovations, where she will present her research and receive a $5,000 honorarium and award.

“I am honored to be recognized with women who are driving discovery in cancer biology and contributing to a stronger scientific community,” said Dr. Conacci-Sorrell, who has a secondary appointment in Children’s Medical Center Research Institute at UT Southwestern (CRI).

After the conference, Dr. Conacci-Sorrell will share her discoveries across the state during lectures at four TAMEST member institutions with National Cancer Institute-Designated Cancer Centers, including the Harold C. Simmons Comprehensive Cancer Center at UT Southwestern. Dr. Conacci-Sorrell is a member of the Simmons Cancer Center.

Dr. Conacci-Sorrell holds the John P. Perkins, Ph.D. Distinguished Professorship in Biomedical Science and is a Virginia Murchison Linthicum Scholar in Medical Research at UTSW. She won the Outstanding Educator Award for the Graduate School of Biomedical Sciences in 2023 and the Excellence in Postdoctoral Mentoring Award in 2024.

TAMEST, founded in 2004, comprises Texas-based members of the three National Academies (National Academy of Medicine, National Academy of Engineering, and National Academy of Sciences) and other honorific organizations. TAMEST includes more than 355 members, eight Nobel Laureates, and 23 member institutions. 

Dr. Kliewer joined TAMEST in 2015. He holds the Diana K. and Richard C. Strauss Distinguished Chair in Developmental Biology at UTSW.

About UT Southwestern Medical Center    

UT Southwestern, one of the nation’s premier academic medical centers, integrates pioneering biomedical research with exceptional clinical care and education. The institution’s faculty members have received six Nobel Prizes and include 24 members of the National Academy of Sciences, 25 members of the National Academy of Medicine, and 13 Howard Hughes Medical Institute Investigators. The full-time faculty of more than 3,200 is responsible for groundbreaking medical advances and is committed to translating science-driven research quickly to new clinical treatments. UT Southwestern physicians provide care in more than 80 specialties to more than 140,000 hospitalized patients, more than 360,000 emergency room cases, and oversee nearly 5.1 million outpatient visits a year.

]]>
https://www.utsouthwestern.edu/newsroom/articles/year-2025/dec-maddox-tamest.html Thu, 18 Dec 2025 08:21:00 -0600
<![CDATA[UT Southwestern biochemist to receive O’Donnell Award from TAMEST]]> TAMEST 2026 O’Donnell Award in Biological Sciences: Yunsun Nam, Ph.D.
Photo credit: Courtesy of TAMEST (Texas Academy of Medicine, Engineering, Science & Technology)

DALLAS – Dec. 11, 2025 – Yunsun Nam, Ph.D., Professor of Biochemistry and Biophysics at UT Southwestern Medical Center, will receive the 2026 Edith and Peter O’Donnell Award in Biological Sciences from the Texas Academy of Medicine, Engineering, Science & Technology (TAMEST) for her research into how RNAs and proteins interact at the molecular level. Her work has shed light on gene regulation, cancer biology, and RNA-based therapeutics.

TAMEST presents annual awards to recognize the achievements of early-career Texas investigators in the fields of science, medicine, engineering, and technological innovation. The O’Donnell Award comes with a $25,000 honorarium and an invitation to make a presentation before hundreds of TAMEST members. Dr. Nam is the 18th scientist at UT Southwestern to be honored with an O’Donnell Award since TAMEST launched the awards in 2006 and is one of five Texas-based researchers receiving the award this year.

Watch the TAMEST video about Dr. Nam

“I am grateful for this award because recognition like this keeps encouraging us to aim high and keep challenging ourselves,” said Dr. Nam, who holds the Doris and Bryan Wildenthal Distinguished Chair in Medical Science. She is also a member of the Harold C. Simmons Comprehensive Cancer Center and an Investigator in the Peter O’Donnell Jr. Brain Institute.

Only an estimated 2% of the human genome codes for proteins. Most of the genome is still transcribed into RNAs, many of which function as noncoding RNAs that play crucial roles in gene regulation. The Nam Lab is particularly interested in a family of noncoding RNAs known as microRNAs that modulate messenger RNA (mRNA) translation and play key roles in diseases including cancer. Using cutting-edge biochemistry and structural biology methods, Dr. Nam and her colleagues have produced a wealth of insights into how microRNAs are processed in cells, modified with chemical groups, and remodeled by different proteins to exert their effects.

Using cryo-electron microscopy, which allows scientists to image molecules at atomic resolution, the team determined the core structure of the Microprocessor protein complex, the processing enzyme that produces microRNAs by cleaving longer RNA pieces. Their research showed that this complex recognizes where to cut RNA based on structural motifs found in the longer segments, rather than specific RNA sequences as some researchers had assumed.

Their research also extends to other classes of protein enzymes that act on RNAs, such as RNA modification enzymes. They found that structural motifs determine where the METTL1-WDR4 protein complex places chemical modifications to regulate the stability and function of transfer RNAs. In contrast, their work on the METTL3-METTL14 complex showed that some proteins determine where to chemically modify mRNAs through sequence recognition.

Chemical modification performed by both complexes has been found to go awry in various cancers, Dr. Nam explained, suggesting these complexes and their interactions with RNA could eventually serve as targets for novel cancer therapies.

“Yunsun is a rising star in the study of RNA-protein interactions,” said Yuh Min Chook, Ph.D., Professor of Pharmacology and Biophysics at UTSW and recipient of the 2015 O’Donnell Award in Biological Sciences, who nominated Dr. Nam for her O’Donnell Award. “Her work on how proteins modify RNAs is very much basic science, and yet when these modification processes go awry, they lead to diseases like cancer and developmental disorders. The work in the Nam Lab thus provides a unique foundation for development of therapeutics to target these diseases.”

Dr. Nam came to UTSW in 2013, supported by a recruitment grant from the Cancer Prevention and Research Institute of Texas. Born in Korea and raised in Indonesia, she was inspired to become a scientist at 8 years old after reading a biography of Marie Curie she had borrowed from the library. She followed her dream to Harvard University, where she earned an undergraduate degree in biochemical sciences and a doctoral degree in biological chemistry and molecular pharmacology, and continued on for postdoctoral fellowships. She became interested in noncoding RNAs while working on her last postdoctoral research project, where she studied RNA recognition by Lin28, a stem cell factor and an oncogene.

The Edith and Peter O’Donnell Awards recognize rising star Texas researchers who are addressing the essential role science and technology play in society and whose work meets the highest standards of exemplary professional performance, creativity, and resourcefulness. The Edith and Peter O’Donnell Awards are made possible by the O’Donnell Awards Endowment Fund, established in 2005 through the generous support of several individuals and organizations.

This year’s recipients will be honored at the 2026 Edith and Peter O’Donnell Awards Ceremony on Feb. 3 and will present their research at the TAMEST 2026 Annual Conference: Pioneering Climate Innovations at the Kimpton Santo Hotel in San Antonio.

“The Edith and Peter O’Donnell Awards have shone a spotlight on Texas’ brightest emerging researchers who are pushing the boundaries of science and technology for the past 20 years,” said Edith and Peter O’Donnell Awards Committee Chair Margaret A. Goodell, Ph.D., Chair and Professor of Molecular and Cellular Biology at Baylor College of Medicine and a member of the National Academy of Medicine. “Each year, these awards celebrate not only exceptional individual achievement but also the profound impact that innovative research has on communities, industries, and our future. It is inspiring to witness the next generation of trailblazers making Texas a global leader in transformative discovery.”

Dr. Nam is a Southwestern Medical Foundation Scholar in Biomedical Research and a UT Southwestern Presidential Scholar. Dr. Chook holds the Alfred and Mabel Gilman Chair in Molecular Pharmacology, is a Eugene McDermott Scholar in Biomedical Research, and is a member of Simmons Cancer Center.

About UT Southwestern Medical Center    

UT Southwestern, one of the nation’s premier academic medical centers, integrates pioneering biomedical research with exceptional clinical care and education. The institution’s faculty members have received six Nobel Prizes and include 24 members of the National Academy of Sciences, 25 members of the National Academy of Medicine, and 13 Howard Hughes Medical Institute Investigators. The full-time faculty of more than 3,200 is responsible for groundbreaking medical advances and is committed to translating science-driven research quickly to new clinical treatments. UT Southwestern physicians provide care in more than 80 specialties to more than 140,000 hospitalized patients, more than 360,000 emergency room cases, and oversee nearly 5.1 million outpatient visits a year.

]]>
https://www.utsouthwestern.edu/newsroom/articles/year-2025/dec-nam-tamest.html Thu, 11 Dec 2025 07:00:00 -0600
<![CDATA[Key protein behind necroptotic cell death could drive new treatment strategies]]> A human cell undergoing plasma membrane rupture during necroptotic cell death
This image shows a human cell undergoing plasma membrane rupture during necroptotic cell death.

DALLAS – Dec. 10, 2025 – Researchers at UT Southwestern Medical Center have identified a protein that causes human cell membranes to break open in a form of inflammatory programmed cell death called necroptosis. Their findings, reported in Nature, could eventually lead to new treatments for a broad array of conditions that involve this phenomenon, including severe infections and sepsis, chronic inflammatory diseases such as Crohn’s disease, neurodegenerative diseases such as Alzheimer’s and amyotrophic lateral sclerosis (ALS), and several forms of cancer.

“Our study identifies a human-specific mediator of necroptotic membrane rupture, revealing a previously unknown, druggable control point in inflammatory cell death,” said study leader Ayaz Najafov, Ph.D., Assistant Professor of Internal Medicine in the Division of Digestive and Liver Diseases and in Children’s Medical Center Research Institute at UT Southwestern. Dr. Najafov is also a member of the Cellular Networks in Cancer Research Program in the Harold C. Simmons Comprehensive Cancer Center.

Ayaz Najafov, M.Sc., Ph.D.
Ayaz Najafov, Ph.D., is Assistant Professor of Internal Medicine in the Division of Digestive and Liver Diseases and in Children’s Medical Center Research Institute at UT Southwestern. Dr. Najafov is also a member of the Harold C. Simmons Comprehensive Cancer Center.

In humans and most other organisms, programmed cell death is necessary to shape tissues during development; eliminate old, damaged, infected, or unnecessary cells; or strike a balance between cell growth and death, among other functions, Dr. Najafov explained. When cells become inflamed through infection or chronic disease, they can undergo necroptosis, a form of programmed cell death in which a molecular cascade ultimately culminates in cell membrane rupture. This process releases signals that recruit immune cells to the dead cells to remove their debris and fight released bacteria or viruses.

In other forms of programmed cell death that also involve cell membrane rupture – such as apoptosis, pyroptosis, and ferroptosis – researchers have shown that a protein called NINJ1 is responsible for splitting open the cell membrane. However, NINJ1 doesn’t appear to be involved in necroptosis. Although previous studies have identified the preceding steps in the necroptosis molecular cascade, Dr. Najafov said, none had discovered a protein analogous to NINJ1 in this process.

Searching for that missing piece, Dr. Najafov and his colleagues used the gene editing tool CRISPR to eliminate individual genes in human cells that had been modified to produce an activated form of MLKL, the last known protein in the necroptosis molecular cascade. Producing this form of MLKL caused most of these cells to undergo necroptosis and burst open. The only exception was a cell clone in which CRISPR had inactivated the gene coding for a protein called SIGLEC12, which has parts that are strikingly similar to NINJ1.

When the researchers stimulated cells missing SIGLEC12 to undergo necroptosis, their cell membranes ballooned outward but didn’t rupture. Forcing cells to produce extra SIGLEC12 didn’t cause them to burst open either. A closer look showed that another protein called TMPRSS4 cuts off part of SIGLEC12, a process that seems to be key for activating it. Experiments using just this cleaved form of SIGLEC12 showed that it was sufficient to prompt cell membrane rupture.

Cells from many cancer types are less likely than healthy cells to undergo necroptosis, a factor thought to help them survive and grow. Dr. Najafov and his colleagues found that SIGLEC12 mutations, common in many cancer types, prevent this protein from being cleaved by TMPRSS4, thus stymieing SIGLEC12 function. They identified several other SIGLEC12 mutations found in the general population, which also prevent SIGLEC12 cleavage by TMPRSS4. Although the significance of these mutations isn’t known, they could affect sensitivity to infections and other inflammatory conditions, he said.

In the future, Dr. Najafov added, drugs that target SIGLEC12 or TMPRSS4 could be used to prevent necroptosis and treat conditions in which it’s a common feature.

Other UTSW researchers who contributed to this study are first author Hyunjin Noh, Ph.D., postdoctoral researcher, and Zeena Hashem, B.Sc., graduate student researcher.

This study was funded by the National Institute of General Medical Sciences (R35 GM146861) and a National Cancer Institute Cancer Center Support Grant (P30 CA142543).

About UT Southwestern Medical Center    

UT Southwestern, one of the nation’s premier academic medical centers, integrates pioneering biomedical research with exceptional clinical care and education. The institution’s faculty members have received six Nobel Prizes and include 24 members of the National Academy of Sciences, 25 members of the National Academy of Medicine, and 13 Howard Hughes Medical Institute Investigators. The full-time faculty of more than 3,200 is responsible for groundbreaking medical advances and is committed to translating science-driven research quickly to new clinical treatments. UT Southwestern physicians provide care in more than 80 specialties to more than 140,000 hospitalized patients, more than 360,000 emergency room cases, and oversee nearly 5.1 million outpatient visits a year.

]]>
https://www.utsouthwestern.edu/newsroom/articles/year-2025/dec-key-protein-necroptotic-cell-death.html Wed, 10 Dec 2025 08:42:00 -0600
<![CDATA[Findings may move science closer to growing organs in other species]]> Tiny human RNA molecules (shown as white dots) move from human cells into neighboring mouse cells, activating those cells’ antiviral defense system.
When human (green) and mouse (red) pluripotent stem cells are grown together, the human cells tend to die off through a process known as cell competition. UT Southwestern researchers discovered one of the main triggers: Tiny human RNA molecules (shown as white dots) move from human cells into neighboring mouse cells, activating those cells’ antiviral defense system. This causes the mouse cells to push out and eliminate the human cells.

DALLAS – Dec. 08, 2025 – Failure of human pluripotent stem cells (PSCs) to survive when grown with the PSCs of distantly related species occurs because of an innate immune reaction in the nonhuman cells, a study led by UT Southwestern Medical Center researchers suggests. The findings, published in Cell, could help researchers remove a key barrier to growing human organs in other species for transplant.

Jun Wu, Ph.D.
Jun Wu, Ph.D., is Associate Professor of Molecular Biology at UT Southwestern and a member of the Cecil H. and Ida Green Center for Reproductive Biology Sciences, the Harold C. Simmons Comprehensive Cancer Center, and the Hamon Center for Regenerative Science and Medicine. Dr. Wu is a Virginia Murchison Linthicum Scholar in Medical Research.

“Our ultimate goal is to use human PSCs to generate organs and tissues in animals to overcome the worldwide shortage of organ and tissue donors. This research uncovers a previously unrecognized role for RNA innate immunity in cell competition and interspecies chimerism that’s blocking us from reaching that objective,” said Jun Wu, Ph.D., Associate Professor of Molecular Biology at UT Southwestern and a New York Stem Cell Foundation (NYSCF)-Robertson Investigator.

Dr. Wu co-led the study with Yingying Hu, Ph.D., former Assistant Instructor in the Wu Lab, and Masahiro Sakurai, Ph.D., Research Scientist in the Wu Lab. Dr. Hu is currently a Senior Research Associate in the lab of Elizabeth Chen, Ph.D., Professor of Molecular Biology.

The Wu Lab is particularly interested in learning how to grow human cells with those of other species – research that could eventually lead to generating full human organs in animals and that sheds light on developmental processes in humans and other species. In 2021, Dr. Wu and his colleagues showed that when human PSCs were grown in lab dishes with the PSCs of distantly related species such as mice or rats, the human cells gradually died off while the other species’ cells thrived.

Why human PSCs were the “losers” in this co-culture competition was not fully understood, Dr. Wu explained. Although subsequent research showed it’s possible to help the human cells survive by genetically altering a molecular pathway responsible for programmed cell death, this tweak could cause problems in tissues and organs destined for transplant, he added. Thus, finding ways to mitigate this competitive process in the other species’ cells and embryos would be preferable.

Toward this end, Dr. Wu and his colleagues searched for any role the nonhuman cells might play in harming the human cells by growing mouse and human PSCs together in lab dishes and comparing the mouse cells’ gene expression activity with that of mouse cells grown without human cells. Their work revealed that a molecular cascade known as the retinoic acid-inducible gene I-like receptor (RLR) pathway was significantly more active in the co-cultured mouse cells compared with the mouse cells growing alone. This pathway is responsible for sensing foreign RNAs in cells – a consequence of some viral infections – and turning on immune activity to fight these invaders.

To determine if the RLR pathway was responsible for killing the co-cultured human cells, it was shut down in the mouse cells by turning off a key gene in the cascade responsible for producing the mitochondrial antiviral signaling protein, or MAVS, discovered at UTSW by Zhijian “James” Chen, Ph.D., Professor of Molecular Biology and in the Center for the Genetics of Host Defense. Significantly more human cells survived after this alteration, suggesting RNA innate immunity in the mouse cells was responsible for harming the human cells.

Further study revealed small amounts of human RNA in the co-cultured mouse cells and vice versa, suggesting the cells had exchanged RNA molecules. A closer look through microscopy suggested this exchange happened through tunneling nanotubes (TNTs), bridges formed by extensions of the cell membrane. When the researchers shut down TNT formation, more human cells survived. Notably, when human cells were injected into mouse embryos lacking MAVS, significantly more survived than those in mouse embryos with MAVS.

Dr. Wu said these findings offer multiple targets that scientists can use to increase the survival of human PSCs growing with PSCs or within embryos of other species – a step that brings growing human organs in animals closer to fruition.

Dr. Wu is a Virginia Murchison Linthicum Scholar in Medical Research. He is a member of the Cecil H. and Ida Green Center for Reproductive Biology Sciences, the Harold C. Simmons Comprehensive Cancer Center, and the Hamon Center for Regenerative Science and Medicine at UTSW. Dr. James Chen holds the George L. MacGregor Distinguished Chair in Biomedical Science.

A complete list of contributors can be found in the study. 

This study was funded by grants from the Cancer Prevention and Research Institute of Texas (RR170076), the New York Stem Cell Foundation, the National Institutes of Health (HD103627-01A1, not used for human-mouse chimera work), and The Welch Foundation (I-2261 and I-2088).

About UT Southwestern Medical Center    

UT Southwestern, one of the nation’s premier academic medical centers, integrates pioneering biomedical research with exceptional clinical care and education. The institution’s faculty members have received six Nobel Prizes and include 24 members of the National Academy of Sciences, 25 members of the National Academy of Medicine, and 13 Howard Hughes Medical Institute Investigators. The full-time faculty of more than 3,200 is responsible for groundbreaking medical advances and is committed to translating science-driven research quickly to new clinical treatments. UT Southwestern physicians provide care in more than 80 specialties to more than 140,000 hospitalized patients, more than 360,000 emergency room cases, and oversee nearly 5.1 million outpatient visits a year.

]]>
https://www.utsouthwestern.edu/newsroom/articles/year-2025/dec-growing-organs-other-species.html Mon, 08 Dec 2025 08:41:00 -0600
<![CDATA[Single-dose radiation before surgery can eradicate breast cancer]]> Two magnetic resonance imaging (MRI) scans
These two magnetic resonance imaging (MRI) scans were taken 10 months apart. On the left, the blue arrow points to the edge of a breast tumor, and the red arrow locates a biopsy clip, which appears as a black dot. The MRI on the right, which includes the biopsy clip, shows the tumor is gone after a single, targeted dose of radiation and antihormone therapy.

DALLAS – Nov. 17, 2025 – A single, targeted high dose of radiation delivered before other treatments could completely eradicate tumors in most women with early-stage, operable hormone-positive breast cancer, according to a study led by UT Southwestern Medical Center researchers. The findings, published in JAMA Network Open, could shift the paradigm for patients with the most common form of breast cancer, who typically undergo surgery before a regimen of radiation therapy.

Asal Rahimi, M.D.
Asal Rahimi, M.D., is Professor of Radiation Oncology, Associate Vice Chair for Program Development, and Medical Director of the Clinical Research Office at the Harold C. Simmons Comprehensive Cancer Center.

“This is a major advance in the field,” said study leader Asal Rahimi, M.D., Professor of Radiation Oncology, Associate Vice Chair for Program Development, and Medical Director of the Clinical Research Office at the Harold C. Simmons Comprehensive Cancer Center. “This treatment protocol provides patients a significant time savings, spares a lot of their tissue from irradiation, and allows them to still undergo any type of oncoplastic surgery they may choose, all while very effectively treating their disease.”

Like patients with other forms of cancer, those with breast cancer are typically treated with a combination of surgery to remove tumors, medications such as hormone blockers, chemotherapy, and radiation, often in that order. In addition, many patients choose to have breast reconstructive surgeries before radiation treatment.

Having targeted radiation prior to surgery has several benefits, including a more than 100-fold smaller volume of tissue being irradiated compared with whole breast radiation; one day of radiation compared with up to 6.5 weeks of radiation, creating a huge time savings for patients; and more options for patients seeking reconstructive surgery, explained Dr. Rahimi, who also serves as Chief of the Breast Radiation Oncology Service at UT Southwestern.

UT Southwestern researchers
These UT Southwestern researchers were involved in the study of a new treatment protocol for patients with early-stage, operable hormone-positive breast cancer.

Early-stage, hormone-positive breast cancer accounts for 60%-75% of all breast cancers. Seeking a more time-efficient way to treat these patients, Dr. Rahimi and her colleagues tested a strategy in which 44 patients started treatment with a single dose of targeted radiation. While typical radiation therapy protocols call for 1.8-2.67 Gy (a measure of radiation strength) per day for 16 to 33 days, the researchers divided the study participants into three groups and gave each patient a single dose of 30, 34, or 38 Gy. The volunteers then went on hormone-blocking drugs and waited a median of 9.8 months until they underwent surgery to remove any residual tumor tissue.

In 72% of study participants, the surgeons found no residual tumor left, indicating that patients had a “pathological complete response.” An additional 21% of patients had a “near complete response,” meaning that their cancer was more than 90% eliminated.

When the researchers further analyzed the results, they found that time to surgery was the best predictor of response. The longer patients waited to undergo surgery, the more likely their tumors were to disappear, regardless of the radiation dose or tumor size. These results were probably due to the time it takes cells to die or be removed by the immune system after radiation therapy, Dr. Rahimi explained.

Marilyn Leitch, M.D.
Marilyn Leitch, M.D., is Professor of Surgery at UT Southwestern. She holds the S.T. Harris Family Distinguished Chair in Breast Surgery, in Honor of A. Marilyn Leitch, M.D.

This new treatment protocol could hold significant advantages over the current gold standard, said Marilyn Leitch, M.D., Professor of Surgery, who holds the S.T. Harris Family Distinguished Chair in Breast Surgery, in Honor of A. Marilyn Leitch, M.D. For example, being able to wait to schedule surgery will allow patients to plan for the disruption it brings to their lives. The radiation course lasts a single day rather than weeks. Plus, in the future, this new approach may eliminate the need for surgery in some patients.

“Much of the current research in breast cancer is looking at ways to reduce the extent of surgery, radiation, and/or medical therapy that is required to completely treat early-stage breast cancer. It is very exciting to be part of innovative research that can improve the quality of life of our cancer patients and minimize the extent of treatment they require,” Dr. Leitch said.

The research team is currently enrolling patients in a phase two clinical trial called RAPS that is funded through a grant from the Cancer Prevention and Research Institute of Texas (CPRIT).

“If the results mirror the ones from this study, an initial targeted dose of radiation could become a new treatment option for patients with small, early-stage, hormone-positive breast cancer,” Dr. Leitch said.

Key collaborators involved in this study at UT Southwestern are Basak Dogan, M.D., Director of Breast Imaging Research and Professor of Radiology in the Breast Imaging Division; Prasanna Alluri, M.D., Ph.D., Assistant Professor of Radiation Oncology; and Sunati Sahoo, M.D., Professor of Pathology. A full list of contributors can be found in the published study.

Drs. Alluri, Dogan, Leitch, Rahimi, and Sahoo are members of the Simmons Cancer Center.

About UT Southwestern Medical Center    

UT Southwestern, one of the nation’s premier academic medical centers, integrates pioneering biomedical research with exceptional clinical care and education. The institution’s faculty members have received six Nobel Prizes and include 24 members of the National Academy of Sciences, 25 members of the National Academy of Medicine, and 13 Howard Hughes Medical Institute Investigators. The full-time faculty of more than 3,200 is responsible for groundbreaking medical advances and is committed to translating science-driven research quickly to new clinical treatments. UT Southwestern physicians provide care in more than 80 specialties to more than 140,000 hospitalized patients, more than 360,000 emergency room cases, and oversee nearly 5.1 million outpatient visits a year.

]]>
https://www.utsouthwestern.edu/newsroom/articles/year-2025/nov-single-dose-radiation.html Mon, 17 Nov 2025 08:43:00 -0600
<![CDATA[Metabolic hormone found to boost resilience against flu symptoms]]> Illustration of fibroblast growth factor 21 (FGF21), a hormone secreted by the liver.
FGF21, a stress-induced hormone that regulates whole-body metabolism, acts on the brain to protect against the hypothermia and weight loss caused by influenza infection. (Photo credit: Getty Images)

DALLAS – Nov. 13, 2025 – A hormone known for regulating energy balance also helps the body cope with influenza by triggering protective responses in the brain, a study led by UT Southwestern Medical Center researchers shows. The findings, published in the Proceedings of the National Academy of Sciences (PNAS), suggest that targeting this pathway could offer a new pharmacological approach for treating the flu.

Steven Kliewer, Ph.D.
Steven Kliewer, Ph.D., is Professor of Molecular Biology and Pharmacology at UT Southwestern. He is a member of the Harold C. Simmons Comprehensive Cancer Center and an Investigator in the Peter O’Donnell Jr. Brain Institute. Dr. Kliewer holds the Diana K. and Richard C. Strauss Distinguished Chair in Developmental Biology and is a member of the National Academy of Sciences.

“Our work demonstrates that FGF21, a stress-induced hormone that regulates whole-body metabolism, acts on the brain to protect against the hypothermia and weight loss caused by influenza infection,” said senior author Steven Kliewer, Ph.D., Professor of Molecular Biology and Pharmacology at UT Southwestern.

The study found that levels of fibroblast growth factor 21 (FGF21) rose in both humans and mice during flu infection. In mice, the hormone activated a brain region that regulates the noradrenergic nervous system, prompting heat production from tissues that help regulate body temperature in mice.

This thermogenic response helped stabilize body temperature and improved the response to flu infection. Mice lacking FGF21 or its receptor in these neurons recovered more slowly, while treatment with pharmacologic FGF21 improved recovery. The hormone did not change viral levels, indicating that it protects the body by mitigating the physiological stress of infection rather than directly targeting the virus. Collectively, these results suggest FGF21 could help the body respond more effectively to a range of infections, not just influenza. 

“For serious cases of influenza infection, the care is mostly supportive,” Dr. Kliewer said. “Our findings suggest a new pharmacological approach for treating the flu. Further studies are required to determine if these findings are applicable to other infections.”

Kartik Rajagopalan, M.D., Ph.D.
Kartik Rajagopalan, M.D., Ph.D., is Assistant Professor of Internal Medicine in the Division of Pulmonary and Critical Care Medicine and in Children’s Medical Center Research Institute at UT Southwestern.

The research builds on decades of work from the Mangelsdorf/Kliewer Lab at UTSW, which previously identified FGF21 as a hormone produced by the liver in response to metabolic stresses such as fasting and alcohol exposure. The new study extends that work to infection, showing that FGF21 uses the same liver-to-brain signaling pathway to help the body maintain metabolic balance during illness. 

“These findings demonstrate that the immune system is not the only critical part of the response to infection,” said corresponding author Kartik Rajagopalan, M.D., Ph.D., Assistant Professor of Internal Medicine in the Division of Pulmonary and Critical Care Medicine and in Children’s Medical Center Research Institute at UT Southwestern. “There are signals that are sent to the brain that reprogram metabolism for an optimal response.”

The work was a collaboration among UT Southwestern’s Departments of Pharmacology, Molecular Biology, and Internal Medicine, bringing together expertise in infection, endocrinology, and neuroscience. It also engaged trainees at multiple levels, including postdoctoral and clinical fellows, and incorporated human data showing that FGF21 levels rise during influenza infection.

“This project highlights the power of integrating basic and clinical research, which is a defining strength of UT Southwestern,” Dr. Kliewer said. “There are very few places where a project like this could have blossomed. We’re fortunate that UT Southwestern is one of them.” 

Other UTSW researchers who contributed to this study are first author Wei Fan, Ph.D., a former postdoctoral researcher in the Mangelsdorf/Kliewer Lab; Yuan Zhang, Ph.D., Assistant Professor of Pharmacology; Laurent Gautron, Ph.D., Assistant Professor of Internal Medicine; Tadiwanashe Gwatiringa, Research Assistant and Lab Manager; and the late David Mangelsdorf, Ph.D., former Chair and Professor of Pharmacology. The human studies were performed by collaborators at Weill Cornell Medicine.

Dr. Kliewer holds the Diana K. and Richard C. Strauss Distinguished Chair in Developmental Biology and is a member of the National Academy of Sciences. Drs. Kliewer, Rajagopalan, and Gautron are Investigators in the Peter O’Donnell Jr. Brain Institute at UT Southwestern. Dr. Kliewer is also a member of the Harold C. Simmons Comprehensive Cancer Center.

This study was funded by the National Institutes of Health (K23HL151876, R01AG079937, and R01AA028473), the Robert A. Welch Foundation (I-1275), a Stony Wold-Herbert Fund Fellowship, and the Howard Hughes Medical Institute.

About UT Southwestern Medical Center    

UT Southwestern, one of the nation’s premier academic medical centers, integrates pioneering biomedical research with exceptional clinical care and education. The institution’s faculty members have received six Nobel Prizes and include 24 members of the National Academy of Sciences, 25 members of the National Academy of Medicine, and 13 Howard Hughes Medical Institute Investigators. The full-time faculty of more than 3,200 is responsible for groundbreaking medical advances and is committed to translating science-driven research quickly to new clinical treatments. UT Southwestern physicians provide care in more than 80 specialties to more than 140,000 hospitalized patients, more than 360,000 emergency room cases, and oversee nearly 5.1 million outpatient visits a year.

]]>
https://www.utsouthwestern.edu/newsroom/articles/year-2025/nov-metabolic-hormone-flu-symptoms.html Thu, 13 Nov 2025 15:05:00 -0600
<![CDATA[Nanovaccine shows great promise for treating HPV-related cancers]]> Scientist pipetting liquid into a cell culture plate
Researchers tested the effectiveness of a nanovaccine in the lab to deliver antigens to immune cells and trigger the body’s protective response. (Photo credit: Getty Images)

DALLAS – Nov. 07, 2025 – A nanoparticle vaccine designed to fight cancers induced by human papillomavirus (HPV) eradicated tumors in an animal model of late-stage metastatic disease, UT Southwestern Medical Center scientists report in a new study published in the Proceedings of the National Academy of Sciences (PNAS). The findings could ultimately lead to a new type of vaccine that would be used to treat a variety of cancers.

Jinming Gao, Ph.D.
Jinming Gao, Ph.D., is Professor in the Harold C. Simmons Comprehensive Cancer Center and of Biomedical Engineering, Cell Biology, Otolaryngology – Head & Neck Surgery, and Pharmacology at UT Southwestern. He holds the Elaine Dewey Sammons Distinguished Chair in Cancer Research, in Honor of Eugene P. Frenkel, M.D.

“Our study provides a safe and effective way to treat cancers that have spread or cannot be surgically removed,” said Jinming Gao, Ph.D., Professor in the Harold C. Simmons Comprehensive Cancer Center and of Biomedical Engineering, Cell Biology, Otolaryngology Head & Neck Surgery, and Pharmacology at UT Southwestern. “Creating a nanovaccine for systemic use for metastatic cancers is not easy due to potential toxicity, but we have overcome those challenges with this new therapy.”

Dr. Gao co-led the study with Shuang Chen, Ph.D., and Shuyue Ye, Ph.D., both postdoctoral researchers in the Gao Lab.

Researchers have been developing vaccines that activate the immune system to prevent various illnesses since the late 1700s. More recently, they have developed a growing number of therapeutic vaccines, which harness the immune system to manage or treat preexisting diseases, such as cancer. A nanovaccine uses tiny particles to encapsulate and deliver antigens to immune cells, triggering the body’s protective response.

HPV causes about 37,800 new cancer cases in the U.S. each year, a number that continues to grow. Although there is an effective vaccine to prevent HPV, a sexually transmitted infection, no therapeutic vaccines exist to treat HPV-related cancers. Such a vaccine would be used to treat patients with HPV-related cancers, such as cervical and head and neck cancers, that have spread or are in locations that are inaccessible to surgical interventions or where radiation therapy is not feasible, Dr. Gao explained. Few effective treatments currently exist for these disease subsets.

To develop a therapeutic vaccine against HPV-related cancers, Dr. Gao and his colleagues combined a polymer and a small-molecule drug that both activate stimulator of interferon genes (STING) – a protein that triggers immune activity – with a protein antigen called E7 derived from HPV. Together, these components formed nanoparticles about 25-30 nanometers in diameter (for comparison, 1 million nanometers equal 1 millimeter).

When the researchers examined mice that received the nanovaccine, they found it was taken up by the spleen, an organ that harbors immune cells for surveillance of foreign particles such as viruses. Nanoparticles that entered immune cells unraveled into their components, with the polymer and drug stimulating STING activity and the viral protein priming the immune system to fight against cells that carried it.

Tests showed that the nanovaccine eradicated both primary HPV-related tumors and metastatic cancer nodules that spread to other organs. In a mouse model of metastatic HPV-related lung cancer, 71% of animals that received the nanovaccine were still alive 60 days after treatment, while those that received immune checkpoint inhibitors – drugs that are considered the current gold standard for treating metastatic HPV-related cancers – died of their disease during this time. When the scientists combined the nanovaccine with the checkpoint therapy, 100% of the mice survived. The nanovaccine appeared safe, causing no organ damage, weight loss, or immune activity beyond that aimed at the cancers.

Dr. Gao said these results showcase the promise of this approach for treating HPV-related cancers and could be adapted to other cancer types by customizing the cancer-related protein targeted by the vaccine. He and his colleagues are continuing to test this approach in animal models with a plan to eventually conduct clinical trials in patients.

A fellow of the National Academy of Inventors, Dr. Gao holds 18 U.S. patents and 78 foreign patents in the fields of polymer biomaterials, nanoparticle drug delivery, tumor surgical imaging, and cancer immunotherapy. Thirteen of the patents have been licensed to biotech companies.

Other UTSW researchers who contributed to this study are Baran D. Sumer, M.D., Professor of Otolaryngology – Head & Neck Surgery and Division Chief of Head and Neck Surgery; Gang Huang, Ph.D., Assistant Professor in the Simmons Cancer Center and of Pharmacology; Qiang Feng, Ph.D., Assistant Professor in the Simmons Cancer Center and of Biomedical Engineering; Zhichen Sun, Ph.D., Senior Research Associate; and Maggie Wang, M.S., and Raymundo Pantoja, B.S., graduate student researchers.

Drs. Gao, Sumer, and Huang are members of the Simmons Cancer Center.

This study was funded by grants from the National Institutes of Health (U54 CA244719 and R01CA216839), the Cancer Prevention and Research Institute of Texas (RP220150), the Mendelson-Young Endowment in Cancer Therapeutics, and the National Cancer Institute (NCI) Cancer Center Support Grant (P30CA142543).

Dr. Gao and Dr. Sumer are co-founders, stockholders, scientific advisory board members, and royalty recipients of OncoNano Medicine Inc. Dr. Huang is a scientific adviser and royalty recipient of OncoNano Medicine Inc. UT Southwestern also receives licensing income from OncoNano Medicine.

Dr. Gao holds the Elaine Dewey Sammons Distinguished Chair in Cancer Research, in Honor of Eugene P. Frenkel, M.D.

About UT Southwestern Medical Center    

UT  Southwestern, one of the nation’s premier academic medical centers, integrates pioneering biomedical research with exceptional clinical care and education. The institution’s faculty members have received six Nobel Prizes and include 24 members of the National Academy of Sciences, 25 members of the National Academy of Medicine, and 13 Howard Hughes Medical Institute Investigators. The full-time faculty of more than 3,200 is responsible for groundbreaking medical advances and is committed to translating science-driven research quickly to new clinical treatments. UT Southwestern physicians provide care in more than 80 specialties to more than 140,000 hospitalized patients, more than 360,000 emergency room cases, and oversee nearly 5.1 million outpatient visits a year.

]]>
https://www.utsouthwestern.edu/newsroom/articles/year-2025/nov-nanovaccine-hpv-related-cancers.html Fri, 07 Nov 2025 10:12:00 -0600
<![CDATA[UTSW study identifies factors affecting survival of patients with metastatic breast cancer]]> Doctor examining senior woman
Identifying factors associated with survival in patients with metastatic breast cancer may help clinicians as they formulate treatment plans.
(Photo credit: Getty Images)

DALLAS – Oct. 22, 2025 – Researchers have identified factors associated with survival for patients initially diagnosed with metastatic breast cancer who were seen at UT Southwestern Medical Center and its affiliated sites. Their findings, published in Communications Medicine, list certain demographic and clinical characteristics to consider among the regional population when formulating treatment plans for individual patients.

“Understanding local risk factors and regional practice patterns can guide more nuanced multidisciplinary care, helping clinicians identify patients at risk for worse outcomes and provide more personalized management,” said Isaac Chan, M.D., Ph.D., Assistant Professor of Internal Medicine in the Division of Hematology and Oncology and a member of the Harold C. Simmons Comprehensive Cancer Center at UT Southwestern.

Isaac Chan, M.D., Ph.D.
Isaac Chan, M.D., Ph.D., is Assistant Professor of Internal Medicine in the Division of Hematology and Oncology and a member of the Harold C. Simmons Comprehensive Cancer Center at UT Southwestern.

Researchers elsewhere have built national datasets that include information from thousands of patients to search for insights into metastatic breast cancer to better understand which individuals are at risk for poor outcomes. However, Dr. Chan explained, such large numbers can obscure findings that may be specific to local populations.

To overcome this issue, he and his colleagues developed the Dallas Metastatic Cancer Study, a database that has tracked patients with metastatic disease treated at UT Southwestern and affiliated sites, including Parkland Health, since 2010. Pulling data for patients who were first diagnosed with metastatic breast cancer between 2010 and 2021, the researchers examined clinical and demographic features, searching for those that correlated with decreased length of survival.

Their findings showed that patients who were Black, had public insurance or no health insurance, had underlying metabolic diseases such as high blood pressure or diabetes, or had cancer that metastasized to specific organs, including the brain, liver, or lungs, tended to die earlier than those without these factors.

Why these variables are associated with reduced survival will be the focus of future research, Dr. Chan said. In the meantime, he added, doctors may be able to improve survival by keeping a closer eye on patients with these risk factors.

Dr. Chan, who is also Assistant Professor of Molecular Biology, co-led the study with former trainees Hannah Chang, M.D., a member of the Chan Lab who is now an Assistant Professor of Medical Oncology & Therapeutics Research at City of Hope, and Meng Cao, M.D., medical resident. This is the first published study of the Chan Lab’s Metastasis Research Program.

Other UTSW researchers who contributed to this study are Mir Lim, M.D., Ariana Weiss, M.D., Danielle Martinez, M.D., Giselle Uwera, M.D., Jonathan Ladner, M.D., Priscilla Okanlawon, M.D., Ruchita Iyer, M.D., and Luis Chinea, M.D., medical residents; Anna Moscowitz, M.D., and Sangeetha Reddy, M.D., Assistant Professors of Internal Medicine; Ang Gao, M.S., Biostatistical Consultant; Katherine Lei, B.A., medical student; Heather McArthur, M.D., Professor of Internal Medicine and Clinical Director of the Breast Cancer Program at Simmons Cancer Center; and Sakshi Mohta, B.S., and Shao-Po Huang, B.S., graduate student researchers.

Drs. Reddy and McArthur are also members of Simmons Cancer Center.

This study was funded by the National Institutes of Health (1K08CA270188-01A1), a METAvivor Early Career Investigator Award, a Susan G. Komen Career Catalyst Research Grant (1010879), a Mary Kay Ash Foundation Cancer Research Grant (11-23), a Robert J. & Claire Pasarow Foundation Award, and a National Cancer Institute Cancer Center Support Grant (P30CA142543).

About UT Southwestern Medical Center    

UT Southwestern, one of the nation’s premier academic medical centers, integrates pioneering biomedical research with exceptional clinical care and education. The institution’s faculty members have received six Nobel Prizes and include 24 members of the National Academy of Sciences, 25 members of the National Academy of Medicine, and 13 Howard Hughes Medical Institute Investigators. The full-time faculty of more than 3,200 is responsible for groundbreaking medical advances and is committed to translating science-driven research quickly to new clinical treatments. UT Southwestern physicians provide care in more than 80 specialties to more than 140,000 hospitalized patients, more than 360,000 emergency room cases, and oversee nearly 5.1 million outpatient visits a year.

About Parkland Health

Parkland Health is one of the largest public hospital systems in the country. Premier services at the state-of-the-art Parkland Memorial Hospital include the Level I Rees-Jones Trauma Center, the only burn center in North Texas verified by the American Burn Association for adult and pediatric patients, and a Level III Neonatal Intensive Care Unit. The system also includes two on-campus outpatient clinics – the Ron J. Anderson, MD Clinic and the Moody Outpatient Center, as well as more than 30 community-based clinics and numerous outreach and education programs. By cultivating its diversity, inclusion, and health equity efforts, Parkland enriches the health and wellness of the communities it serves. For more information, visit parklandhealth.org.

]]>
https://www.utsouthwestern.edu/newsroom/articles/year-2025/oct-metastatic-breast-cancer.html Wed, 22 Oct 2025 09:51:00 -0500
<![CDATA[UTSW Physiology Chair, molecular biologist elected to National Academy of Medicine]]> DALLAS – Oct. 20, 2025 – Duojia Pan, Ph.D., Chair and Professor of Physiology at UT Southwestern Medical Center, and Joshua Mendell, M.D., Ph.D., Professor of Molecular Biology, have been elected to the National Academy of Medicine (NAM), one of the highest honors in the fields of health and medicine.

With the elections, UT Southwestern has 25 members of the National Academy of Medicine – more than any other institution in Texas – along with 24 members of the National Academy of Sciences (NAS) and 13 Howard Hughes Medical Institute (HHMI) Investigators.

Dr. Pan is recognized for advancing the understanding of the molecular pathways that regulate tissue growth and homeostasis, while Dr. Mendell has led pioneering research into the functions of noncoding RNAs in both normal physiology and diseases such as cancer. Both investigators are members of the Harold C. Simmons Comprehensive Cancer Center, where their discoveries continue to shape innovative approaches to cancer treatment and deepen our understanding of tumor biology.

“The elections of Dr. Mendell and Dr. Pan to the National Academy of Medicine reflect the depth and significance of their scientific contributions to our understanding of cancer biology,” said Daniel K. Podolsky, M.D., President of UT Southwestern and a member of the NAM. “Dr. Mendell’s work has illuminated the role of microRNAs in tumor development, leading to promising therapeutic strategies, while Dr. Pan’s discoveries related to tumor suppressor genes have advanced the use of targeted inhibitors to control cancer growth. This recognition underscores the impact of their research for its potential to ultimately lead to more effective therapies.”

Joshua Mendell, M.D., Ph.D.

Joshua Mendell, M.D., Ph.D.
Joshua Mendell, M.D., Ph.D., is Vice Chair and Professor of Molecular Biology at UT Southwestern. He holds the Charles Cameron Sprague, M.D. Chair in Medical Science.

Charles Cameron Sprague, M.D. Chair in Medical Science

Dr. Mendell, an HHMI Investigator, joined UT Southwestern in 2011 from the Johns Hopkins University School of Medicine. He serves as Vice Chair in the Department of Molecular Biology and is a member of the Hamon Center for Regenerative Science and Medicine.

The Mendell Lab investigates fundamental aspects of post-transcriptional gene regulation, noncoding RNA regulation and function, and the roles of these pathways in normal physiology, cancer, and other diseases. In 2005, he and his colleagues uncovered the first example of a vertebrate transcription factor that regulates the expression of microRNAs (miRNAs), a type of noncoding RNA. This study was important for establishing the principle that miRNAs have been functionally integrated into core cancer pathways.

Dr. Mendell’s team further defined the roles of miRNAs in several critical oncogenic and tumor suppressor pathways. They have translated these findings into novel therapeutic approaches, most notably through demonstrating that systemic delivery of miRNAs potently suppresses the growth of tumors in mouse cancer models without toxicity. Most recently, Dr. Mendell and his colleagues have used high-throughput approaches to investigate RNA biology and post-transcriptional regulation, a strategy they are now applying to diverse problems in the laboratory.

Dr. Mendell earned his undergraduate degree in biology from Cornell University and his Ph.D. and M.D. from Johns Hopkins. Previous honors include the Paul Marks Prize for Cancer Research (2019) and the Edith and Peter O’Donnell Award in Medicine from the Texas Academy of Medicine, Engineering, Science and Technology (2016).

“I am deeply honored to be elected to the National Academy of Medicine and join the ranks of the many accomplished UT Southwestern faculty who have previously been recognized with this distinction,” Dr. Mendell said. “This would not have been possible without the amazing trainees and staff who have worked in my laboratory over the last 20 years, as well as the support of our Chair, Eric Olson, Ph.D., my colleagues in the Department of Molecular Biology, and the broader UT Southwestern community. I feel very fortunate to have an opportunity to lead a research team at this remarkable institution.”

Duojia Pan, Ph.D.

Duojia Pan, Ph.D.
Duojia Pan, Ph.D., is Chair and Professor of Physiology at UT Southwestern. He holds the Fouad A. and Val Imm Bashour Distinguished Chair in Physiology.

Fouad A. and Val Imm Bashour Distinguished Chair in Physiology

Dr. Pan, who is also a member of the NAS and an HHMI Investigator, first joined the UT Southwestern faculty in 1998. Recruited to Johns Hopkins in 2004, he returned to UT Southwestern in 2016 as Chair of the Department of Physiology.

Dr. Pan is best known for his foundational discoveries of the Hippo signaling pathway that controls animal tissue growth. Using the fruit fly Drosophila as a model, the Pan Lab made a series of discoveries that defined, in a stepwise manner, the key molecular events in the Hippo signaling pathway. Most recently, his lab revealed a surprising role for Hippo signaling in regulating cell aggregation and density in a close unicellular relative of animals.

In addition, the Pan Lab elucidated the molecular function of the Tsc1 and Tsc2 tumor suppressor genes, linking Tsc1/Tsc2 to Rheb and TOR signaling. This work provided the key molecular insight for the use of mTOR inhibitors in the treatment of tuberous sclerosis, a genetic disease that can lead to tumor development in multiple tissues.

Dr. Pan earned his undergraduate degree in biochemistry from Peking University in 1988 and his Ph.D. from the University of California, Los Angeles in 1993. He completed his postdoctoral training at the University of California, Berkeley. Previous honors include the Passano Award (2022) and the Paul Marks Prize for Cancer Research (2013).

“I am deeply honored and humbled to be elected to the National Academy of Medicine,” Dr. Pan said. “This is a recognition of the creativity, hard work, and team efforts of my laboratory over the last 27 years. Our work started at UT Southwestern when I was an Assistant Professor. I am extremely grateful for the superb scientific environment at UT Southwestern.”

Founded in 1970 as the Institute of Medicine, the NAM is one of three academies that make up the National Academies of Sciences, Engineering, and Medicine in the United States. Operating under the 1863 Congressional charter of the National Academy of Sciences, the National Academies are private, nonprofit institutions that work outside of government to provide objective advice on matters of science, technology, and health. 

For a complete list of NAM members at UTSW, please visit our Legacy of Excellence in Science & Medicine page.

Dr. Podolsky holds the Philip O’Bryan Montgomery, Jr., M.D. Distinguished Presidential Chair in Academic Administration and the Charles Cameron Sprague Distinguished Chair in Biomedical Science.

About UT Southwestern Medical Center    

UT Southwestern, one of the nation’s premier academic medical centers, integrates pioneering biomedical research with exceptional clinical care and education. The institution’s faculty members have received six Nobel Prizes and include 24 members of the National Academy of Sciences, 25 members of the National Academy of Medicine, and 13 Howard Hughes Medical Institute Investigators. The full-time faculty of more than 3,200 is responsible for groundbreaking medical advances and is committed to translating science-driven research quickly to new clinical treatments. UT Southwestern physicians provide care in more than 80 specialties to more than 140,000 hospitalized patients, more than 360,000 emergency room cases, and oversee nearly 5.1 million outpatient visits a year.

]]>
https://www.utsouthwestern.edu/newsroom/articles/year-2025/oct-nam-physiology-chair-molecular-biologist.html Mon, 20 Oct 2025 08:49:00 -0500
<![CDATA[Study examines biological causes of cancer deaths]]>

DALLAS – Oct. 16, 2025 – The ultimate cause of death from cancer may not be metastatic disease, as researchers have long surmised, but an infiltration of tumors into major blood vessels that cause blood clots and multiorgan failure, a one-of-a-kind clinical study led by UT Southwestern Medical Center suggests. These findings, published in Nature Medicine, could spur interventions that extend the lives of patients with advanced cancers.

Matteo Ligorio, M.D., Ph.D.
Matteo Ligorio, M.D., Ph.D., is Assistant Professor of Surgery and in the Harold C. Simmons Comprehensive Cancer Center at UT Southwestern.

“The big question we were trying to answer: What kills cancer patients? Why do they die one specific day rather than six months earlier or later?” said Matteo Ligorio, M.D., Ph.D., Assistant Professor of Surgery and in the Harold C. Simmons Comprehensive Cancer Center. Dr. Ligorio led the study along with Kelley Newcomer, M.D., Associate Professor of Internal Medicine at UT Southwestern, and Nicola Aceto, Ph.D., Professor of Molecular Oncology at ETH Zurich in Switzerland.

Cancer claims about 600,000 people in the U.S. each year. However, what actually ends their lives has been a mystery, Dr. Ligorio explained. Although scientists have long proposed that cancer mortality is caused by the spread of tumors throughout the body – a phenomenon known as metastasis – patients often live with metastatic disease for years, suggesting that this may not be what instigates the clinical decline that ultimately leads to death.

Some studies have shown that cancer patients are more likely to develop blood clots in their heart, liver, and lungs, indicating that the cardiovascular system is altered in advanced malignancies. But whether this factor contributes to their demise has been unknown.

Kelley Newcomer, M.D.
Kelley Newcomer, M.D., is Associate Professor of Internal Medicine at UT Southwestern.

To investigate this question, Drs. Newcomer and Ligorio analyzed a retrospective cohort of more than 100 patients with colorectal, lung, ovarian, liver, or pancreatic cancer who had died at William P. Clements Jr. University Hospital and Parkland Health and undergone routine autopsies. Dr. Newcomer then recruited 31 terminally ill patients who were in hospice: 21 with solid tumors and 10 with other conditions. Over the following weeks, she monitored and examined these patients. Dr. Newcomer and Dr. Ligorio’s clinical team also took blood samples whenever the patients reported a significant change in their health status or when their score worsened on an assessment called the Palliative Performance Scale, one of the most commonly used bedside tools to determine the status of patients in palliative care settings.

When these patients died – an average of about 38 days after they were enrolled in the study – Dr. Ligorio performed a modified autopsy on each. While normal autopsy procedures tend not to maintain the integrity of all major blood vessels, his altered protocol preserved them so he could examine their walls and interiors.

The modified autopsies revealed that, unlike the patients who died of other causes, those with cancer typically had tumors penetrating the walls and extending into the interiors of major blood vessels, including the portal vein, inferior vena cava, hepatic veins, and/or abdominal aorta. In several cases where CT scans were available, these vessel-invading growths were present in the weeks or months preceding death, suggesting that such lesions may be detectable on routine imaging.

In addition, blood samples taken during the visits in the follow-up period and analyzed by Dr. Aceto’s team at ETH Zurich revealed a sharp uptick in the number of cancer cells in the bloodstream just before death, strengthening the massive involvement of the cardiovascular system during disease progression.

Together, these findings led Dr. Ligorio to a new theory on what kills cancer patients: When tumors – either primary or metastatic – impinge upon major blood vessels, microscopic pieces of the tumors may break off and join the bloodstream, making blood more likely to clot. Clots that form through this process would restrict blood flow to organs, leading to multiorgan failure that ultimately causes death.

To help validate this idea, researchers examined CT imaging data from 1,250 cancer patients who died that was collected by Dr. Ligorio’s collaborators at the University of Lubeck and the University of Mainz in Germany. Dario Ghersi, M.D., Ph.D., Associate Professor at the University of Nebraska at Omaha, and William Gasper, Ph.D., a graduate student at the University of Nebraska at Omaha at the time of this research, co-led these analyses with Dr. Ligorio, Dr. Newcomer, and Dr. Aceto. They confirmed that most of these patients had tumors infiltrating major blood vessels, supporting this new theory of cancer progression.

“Surgery or radiation to treat tumors approaching large blood vessels could potentially transform how we diagnose, manage, and treat patients with cancers,” Dr. Newcomer said.

Drs. Newcomer and Ligorio thanked the patients and their families who generously agreed to participate in this study to advance the scientific understanding of cancer and support the development of new treatments. They also expressed gratitude to the three hospice organizations — Visiting Nurse Association of Texas, Faith Presbyterian Hospice, and Pathway Hospice — for their collaboration in this clinical study.

Dr. Ligorio and Dr. Newcomer are now designing clinical trials, along with Herbert J. Zeh III, M.D., Chair and Professor of Surgery at UTSW, to test these therapeutic approaches and determine whether targeting tumor-vessel infiltration can substantially extend survival, including in patients with advanced disease.

A full list of contributors and their disclosures can be found in the published study.

This study was funded by grants from the Cancer Prevention and Research Institute of Texas (RR200023), the National Cancer Institute (NCI) (5R37CA242070), the American-Italian Cancer Foundation Post-Doctoral Research Fellowship, the European Research Council (101001652), the strategic focus area of Personalized Health and Related Technologies at ETH Zurich (PHRT-960), the Swiss National Science Foundation (212183), the Swiss Cancer League (KLS-5636-08-2022), the ETH Zurich Lymphoma Challenge (LC-02-22), the ETH Zurich, and an NCI Cancer Center Support Grant (P30CA142543).  

About UT Southwestern Medical Center    

UT Southwestern, one of the nation’s premier academic medical centers, integrates pioneering biomedical research with exceptional clinical care and education. The institution’s faculty members have received six Nobel Prizes and include 24 members of the National Academy of Sciences, 23 members of the National Academy of Medicine, and 13 Howard Hughes Medical Institute Investigators. The full-time faculty of more than 3,200 is responsible for groundbreaking medical advances and is committed to translating science-driven research quickly to new clinical treatments. UT Southwestern physicians provide care in more than 80 specialties to more than 140,000 hospitalized patients, more than 360,000 emergency room cases, and oversee nearly 5.1 million outpatient visits a year.

About Parkland Health

Parkland Health is one of the largest public hospital systems in the country. Premier services at the state-of-the-art Parkland Memorial Hospital include the Level I Rees-Jones Trauma Center, the only burn center in North Texas verified by the American Burn Association for adult and pediatric patients, and a Level III Neonatal Intensive Care Unit. The system also includes two on-campus outpatient clinics – the Ron J. Anderson, MD Clinic and the Moody Outpatient Center, as well as more than 30 community-based clinics and numerous outreach and education programs. By cultivating its diversity, inclusion, and health equity efforts, Parkland enriches the health and wellness of the communities it serves. For more information, visit parklandhealth.org.

]]>
https://www.utsouthwestern.edu/newsroom/articles/year-2025/oct-biological-causes-cancer-deaths.html Thu, 16 Oct 2025 09:51:00 -0500
<![CDATA[Neurons in brain’s timekeeper might control nighttime hunger]]> A thin slice from the middle of a mouse brain
This image shows a thin slice from the middle of a mouse brain, with the region known as the suprachiasmatic nucleus (SCN) surrounded by a white box. The small green dots mark brain cells that react to the hunger hormone ghrelin. When the ghrelin-sensitive cells in the SCN are turned off during the mouse’s normal rest time, the animals snack less and lose weight.

DALLAS – Oct. 15, 2025 – Activating specific neurons in a part of the brain that serves as the body’s master circadian pacemaker caused mice to eat significantly more during a time of day when they would normally be at rest, a UT Southwestern Medical Center study shows. The findings, published in Cell Reports, could lead to new strategies to help people lose weight, including night shift workers who have a higher prevalence of obesity.

“We identify for the first time a distinct set of neurons in the brain that controls feeding and metabolism during one specific time of day and accounts for a small but not insignificant proportion of body weight,” said Jeffrey Zigman, M.D., Ph.D., Professor of Internal Medicine and Psychiatry at UT Southwestern. Dr. Zigman co-led the study with first author Omprakash Singh, Ph.D., a postdoctoral researcher in the Zigman Lab.

Jeffrey Zigman, M.D., Ph.D.
Jeffrey Zigman, M.D., Ph.D., is a Professor of Internal Medicine and Psychiatry, a member of the Center for Hypothalamic Research and the Harold C. Simmons Comprehensive Cancer Center, and an Investigator in the Peter O’Donnell Jr. Brain Institute at UT Southwestern. He holds the Kent and Jodi Foster Distinguished Chair in Endocrinology, in Honor of Daniel Foster, M.D.; the Mr. and Mrs. Bruce G. Brookshire Professorship in Medicine; and The Diana and Richard C. Strauss Professorship in Biomedical Research.

Researchers have long known that eating impacts body weight differently depending on when food is consumed, Dr. Zigman explained. For example, eating late at night is associated with greater weight gain than eating the same amount during the day. This effect is especially apparent in night shift workers, who are more frequently overweight or obese despite caloric intake similar to day workers.

These observations suggest specific circuits of neurons that affect feeding and metabolism might operate differently at various times of the day. Dr. Zigman, Dr. Singh, and their colleagues hypothesized that one such circuit might be in the suprachiasmatic nucleus (SCN), a part of the brain that sets circadian rhythms throughout the body based on light received through the eyes.

Previous research in the Zigman Lab showed that some SCN neurons are stimulated by ghrelin, a hormone that prompts feeding and slows metabolism to encourage weight gain. However, the significance of these findings had been unclear.

To better understand this population of SCN neurons, the researchers worked with mice genetically altered so the scientists could turn these neurons on and off. They found that if they turned on the neurons in the middle of the animals’ rest period – around 10 a.m., since mice are nocturnal – they ate more than two times as much as they usually do during this time. Turning the neurons off at this time reduced the already low amount of food typically consumed during this period.

Whether the neurons were on or off during other times of day or night had no effect on the rodents’ feeding behavior or weight. But turning the neurons off during their rest period for 15 straight days caused them to lose about 4.3% of their body weight, while mice with unaltered SCN neurons gained about 2.5%. These results suggest the activity of the ghrelin-stimulated SCN neurons is responsible for about 7% of body weight – a small but significant amount that could make a marked difference for overall health, Dr. Zigman said.

If these results also apply to humans, he added, they suggest that targeting the same population of neurons in the SCN could offer weight-loss benefits similar to those seen with some modern weight-loss drugs. This strategy could be especially beneficial for night shift workers and other groups to prevent or treat weight gain linked to nighttime eating.

Other UTSW researchers who contributed to this study are Kripa Shankar, Ph.D., Instructor in the Center for Human Nutrition and of Internal Medicine; Deepali Gupta, Ph.D., Instructor in the Peter O’Donnell Jr. Brain Institute and of Neuroscience; Luis Leon Mercado, Ph.D., Instructor of Internal Medicine; Sherri Osborne-Lawrence, M.S., Senior Research Scientist; Corine P. Richard, R.N.; Sepideh Sheybani-Deloui, Ph.D., and Salil Varshney, Ph.D., postdoctoral researchers; Soumya Kulkarni, B.S., Moyu Lyu, M.S., and Bingbing Li, B.S., graduate student researchers; Avi W. Burstein, high school student researcher; and Connor Lawrence, research assistant.

Dr. Zigman is a member of the Center for Hypothalamic Research and the Harold C. Simmons Comprehensive Cancer Center and an Investigator in the O’Donnell Brain Institute. He holds the Kent and Jodi Foster Distinguished Chair in Endocrinology, in Honor of Daniel Foster, M.D.; the Mr. and Mrs. Bruce G. Brookshire Professorship in Medicine; and The Diana and Richard C. Strauss Professorship in Biomedical Research.

About UT Southwestern Medical Center    

UT Southwestern, one of the nation’s premier academic medical centers, integrates pioneering biomedical research with exceptional clinical care and education. The institution’s faculty members have received six Nobel Prizes and include 24 members of the National Academy of Sciences, 23 members of the National Academy of Medicine, and 13 Howard Hughes Medical Institute Investigators. The full-time faculty of more than 3,200 is responsible for groundbreaking medical advances and is committed to translating science-driven research quickly to new clinical treatments. UT Southwestern physicians provide care in more than 80 specialties to more than 140,000 hospitalized patients, more than 360,000 emergency room cases, and oversee nearly 5.1 million outpatient visits a year.

]]>
https://www.utsouthwestern.edu/newsroom/articles/year-2025/oct-neurons-brain-timekeeper.html Wed, 15 Oct 2025 08:46:00 -0500
<![CDATA[AI can identify stroke types using clinical notes, study shows]]> A glowing AI symbol hovers above a detailed CPU and circuit board, connected by intricate pathways of light.
(Photo Credit: Getty Images)

DALLAS – Oct. 13, 2025 – Using only text from doctors’ notes and radiology reports, an artificial intelligence (AI) program known as GPT-4o reliably identified patients’ types of strokes, UT Southwestern Medical Center researchers found. Their study, published in Stroke, could eventually lead to new ways to help guide doctors’ medical decisions in real time and reduce the heavy workload necessary to report data to patient registries.

Ann Marie Navar, M.D., Ph.D.
Ann Marie Navar, M.D., Ph.D., is Associate Professor of Internal Medicine and in the Peter O’Donnell Jr. School of Public Health at UT Southwestern.

“Large language models (LLMs) that can decipher unstructured text are an emerging AI technology with immense potential in medical research. Our study provides proof that these LLMs can abstract medical diagnoses from medical notes as well as human chart abstractors,” said Ann Marie Navar, M.D., Ph.D., Associate Professor of Internal Medicine and in the Peter O’Donnell Jr. School of Public Health at UT Southwestern.

Dr. Navar co-led the study with Eric Peterson, M.D., M.P.H., Professor of Internal Medicine and in the O’Donnell School of Public Health, Vice Provost, and Senior Associate Dean for Clinical Research; and Dylan Owens, Ph.D., M.S., Postdoctoral Researcher.

Like most large academic medical centers, UTSW participates in several patient registries – systematic collections of data on specific conditions that researchers use for studies. One prominent example is the American Heart Association’s Get With The Guidelines-Stroke (GWTG-Stroke), a quality improvement initiative involving over 2,600 hospitals across the country. When patients are treated at one of these hospitals for stroke, trained nurses collect a wealth of information from their electronic health records, inputting the data into lengthy forms. This process requires an enormous amount of human labor.

Eric Peterson, M.D., M.P.H.
Eric Peterson, M.D., M.P.H., is Professor of Internal Medicine and in the O’Donnell School of Public Health, Vice Provost, and Senior Associate Dean for Clinical Research at UT Southwestern. He holds the Adelyn and Edmund M. Hoffman Distinguished Chair in Medical Science.

To decrease this burden, Drs. Owens, Navar, and Peterson wondered whether LLMs – a form of AI designed to understand and generate human language – could be used for the same purpose. They started with a simple question: Could an LLM accurately determine stroke type based only on “unstructured” data found in electronic health records, such as notes and reports?

The researchers tested this idea with GPT-4o, an LLM introduced this year with capabilities beyond the more commonly used ChatGPT. Using electronic health records for 4,123 patients hospitalized for stroke at UT Southwestern and Parkland Health between January 2019 and August 2023, the team evaluated three types of prompts asking the LLM to distinguish each patient’s stroke type. Zero-shot chain-of-thought prompts encouraged the model to break complex queries into smaller, logical steps using minimal human input; expert-guided prompts incorporated tips from neurologists and cardiologists; and instruction-based prompts steered the model to evaluate patients’ records using GWTG-Stroke registry guidelines.

The researchers compared the results they received from GPT-4o with those recorded in registry reports for these patients in GWTG-Stroke. They found that all three LLM prompt styles accurately distinguished between the two major types of stroke – hemorrhagic and ischemic – and between hemorrhagic subtypes. However, accuracy was lower for some ischemic subtypes, such as cryptogenic strokes. This lower reliability reflects real-world difficulty in classifying these subtypes, which tend to be diagnoses of exclusion, Dr. Owens explained.

Dylan Owens, Ph.D., M.S.
Dylan Owens, Ph.D., M.S., is a Postdoctoral Researcher at UT Southwestern.

Together, he said, the results suggest LLMs could be a useful tool for accurately abstracting some information from electronic health records for populating time-intensive registry forms and could be used to flag other data that need a closer look from human abstractors. Future research will focus on using LLMs to fill in other parts of registry forms, as well as the feasibility of using LLMs for clinical decision support – programs that aim to improve patient outcomes by delivering timely information to providers at the point of care.

Dr. Owens noted that UTSW researchers also have achieved success working with LLMs for other tasks such as matching patients with clinical trials, performing quality assessments while investigating opportunities for population health improvement, and automating extraction of clinical data for research.

Additional UTSW researchers who contributed to this study are Justin Rousseau, M.D., M.M.Sc., Associate Professor of Neurology and in the Peter O’Donnell Jr. Brain Institute and Deputy Chief Medical Informatics Officer for Neurosciences; Michael Dohopolski, M.D., Assistant Professor of Radiation Oncology and a member of the Harold C. Simmons Comprehensive Cancer Center; and Danh Q. Nguyen, M.D., Clinical Fellow.

Dr. Peterson holds the Adelyn and Edmund M. Hoffman Distinguished Chair in Medical Science.

This study was funded by UT Southwestern Medical Center and grants from the National Institutes of Health (5T32HL12524710 and UL11R003163).

About UT Southwestern Medical Center 

UT Southwestern, one of the nation’s premier academic medical centers, integrates pioneering biomedical research with exceptional clinical care and education. The institution’s faculty members have received six Nobel Prizes and include 24 members of the National Academy of Sciences, 23 members of the National Academy of Medicine, and 13 Howard Hughes Medical Institute Investigators. The full-time faculty of more than 3,200 is responsible for groundbreaking medical advances and is committed to translating science-driven research quickly to new clinical treatments. UT Southwestern physicians provide care in more than 80 specialties to more than 140,000 hospitalized patients, more than 360,000 emergency room cases, and oversee nearly 5.1 million outpatient visits a year.

About Parkland Health

Parkland Health is one of the largest public hospital systems in the country. Premier services at the state-of-the-art Parkland Memorial Hospital include the Level I Rees-Jones Trauma Center, the only burn center in North Texas verified by the American Burn Association for adult and pediatric patients, and a Level III Neonatal Intensive Care Unit. The system also includes two on-campus outpatient clinics – the Ron J. Anderson, MD Clinic and the Moody Outpatient Center, as well as more than 30 community-based clinics and numerous outreach and education programs. By cultivating its diversity, inclusion, and health equity efforts, Parkland enriches the health and wellness of the communities it serves. For more information, visit parklandhealth.org.

]]>
https://www.utsouthwestern.edu/newsroom/articles/year-2025/oct-ai-strokes.html Mon, 13 Oct 2025 09:29:00 -0500
<![CDATA[Kidney cancer drug shows promise against dangerous calcium imbalance caused by tumors]]> The hormone responsible for calcium induction in kidney cancer patients
PT2399/belzutifan disrupts HIF-2α binding to HIF-1β, thereby blocking its ability to bind DNA and stimulate the production of PTHrP, the hormone responsible for calcium induction in kidney cancer patients.

DALLAS – Oct. 09, 2025 – Elevated calcium levels in the blood – a complication of kidney cancers known as hypercalcemia – may be successfully treated with a class of medications called HIF-2α inhibitors developed by UT Southwestern Medical Center, a new study shows. The findings, published in Cancer Discovery by a team at UTSW, offer hope to patients who develop this condition.

About 10% of patients with advanced kidney cancer develop hypercalcemia, which can cause confusion, muscle spasms, and seizures and is associated with lower patient survival. It’s typically treated with drugs like bisphosphonates that reduce calcium release from bone; however, these drugs have side effects, including osteonecrosis of the jaw, fractures, and an opposing complication called hypocalcemia, when blood calcium levels become too low.

Arijit Mal, Ph.D.
Arijit Mal, Ph.D., is a postdoctoral researcher at UT Southwestern.

In their study, Arijit Mal, Ph.D., a postdoctoral researcher, and Bingqing Xie, Ph.D., Assistant Professor of Internal Medicine, along with senior investigator James Brugarolas, M.D., Ph.D., Professor of Internal Medicine in the Division of Hematology and Oncology and founding Director of the Kidney Cancer Program at the Harold C. Simmons Comprehensive Cancer Center at UT Southwestern, evaluated the potential of HIF-2α inhibitors to block hypercalcemia at its root.

Bingqing Xie, Ph.D.
Bingqing Xie, Ph.D., is Assistant Professor of Internal Medicine at UT Southwestern.

Hypercalcemia is frequently caused by a hormone produced by kidney tumors called parathyroid hormone-related protein (PTHrP), which raises blood calcium levels. A previous study by the Brugarolas Lab showed that PTHrP production in kidney cancer is regulated by HIF-2, which led the investigators to test the role of HIF-2α-blocking drugs in hypercalcemia.

HIF-2α-blocking drugs are the result of a long journey at UT Southwestern. In the 1990s, Steven McKnight, Ph.D., Professor of Biochemistry, and David Russell, Ph.D., Professor Emeritus of Molecular Genetics, identified HIF-2α, the key component of HIF-2. Subsequent studies by Richard Bruick, Ph.D., and Kevin Gardner, Ph.D., then at UTSW, identified a vulnerability in the structure that they exploited with a chemical obtained from the UTSW chemical library. These results led to the founding of Peloton Therapeutics, which developed a series of related HIF-2α inhibitors: PT2399 for animal studies and PT2977/belzutifan, which the Food and Drug Administration approved to treat kidney cancer in 2023.

James Brugarolas, M.D., Ph.D.
James Brugarolas, M.D., Ph.D., is Professor of Internal Medicine in the Division of Hematology and Oncology and founding Director of the Kidney Cancer Program at the Harold C. Simmons Comprehensive Cancer Center at UT Southwestern.

To determine whether inhibiting HIF-2α could treat hypercalcemia, researchers leveraged mice transplanted with human kidney tumors. They treated tumor-bearing mice that developed hypercalcemia with PT2399. Interestingly, the majority of the mice responded. Calcium levels dropped within just a few days of treatment onset, and symptoms, including weight loss, fatigue, and calcium deposition, subsided.

Additional studies showed that PT2399 prevents HIF-2α from binding to the gene that produces PTHrP, decreasing the amount of PTHrP made and explaining PT2399’s effects on hypercalcemia.

In a subsequent case study, a 63-year-old man with advanced clear cell renal cell carcinoma and elevated calcium was treated with PT2977/belzutifan. After treatment, PTHrP levels dropped, and calcium returned to normal levels within a few days without the side effects seen with standard therapies.

“Our study supports the systematic evaluation of HIF-2α inhibitors for kidney cancer patients with hypercalcemia,” Dr. Brugarolas said.

A full list of contributors can be found in the published study.

Dr. Brugarolas holds the Sherry Wigley Crow Cancer Research Endowed Chair in Honor of Robert Lewis Kirby, M.D., and is a member of the Cellular Networks in Cancer Research Program of the Simmons Cancer Center.

This study used kidney cancer tumor models developed by UTSW’s Kidney Cancer Program through a National Cancer Institute (NCI)-funded Specialized Program of Research Excellence award.

This study was funded by grants from the NCI (P50CA196516, R01CA245294, R01CA295997, P50CA196516, and P50CA070907), Department of Defense Congressionally Directed Medical Research Program’s Kidney Cancer Research Program (HT9425-25-1-0346), Cancer Prevention and Research Institute of Texas (RP230382, RP240516), National Institute of Diabetes and Digestive and Kidney Diseases through the UTSW Nutrition & Obesity Research Center (P30DK127984), and an NCI Cancer Center Support Grant (P30CA142543).

Disclosures: UT Southwestern and some of its researchers will receive financial compensation, through prior agreements with Peloton, based on belzutifan’s FDA approval.

About UT Southwestern Medical Center    

UT Southwestern, one of the nation’s premier academic medical centers, integrates pioneering biomedical research with exceptional clinical care and education. The institution’s faculty members have received six Nobel Prizes and include 24 members of the National Academy of Sciences, 23 members of the National Academy of Medicine, and 13 Howard Hughes Medical Institute Investigators. The full-time faculty of more than 3,200 is responsible for groundbreaking medical advances and is committed to translating science-driven research quickly to new clinical treatments. UT Southwestern physicians provide care in more than 80 specialties to more than 140,000 hospitalized patients, more than 360,000 emergency room cases, and oversee nearly 5.1 million outpatient visits a year.

]]>
https://www.utsouthwestern.edu/newsroom/articles/year-2025/oct-kidney-cancer-drug.html Thu, 09 Oct 2025 09:48:00 -0500
<![CDATA[UTSW discovery opens door to novel strategies for hard-to-treat cancers]]> Antigen-presenting cancer-associated fibroblasts change over time
This figure shows how antigen-presenting cancer-associated fibroblasts change over time, following two different paths.

DALLAS – Oct. 06, 2025 – UT Southwestern Medical Center researchers have identified two distinct populations of cells known as antigen-presenting cancer-associated fibroblasts (apCAFs) that appear to support the survival and growth of malignant tumors. Their findings, reported in Cancer Cell, could one day lead to new therapies for notoriously hard-to-treat cancers, including pancreatic cancer and advanced colorectal cancer (CRC) that has spread throughout the abdomen, known as peritoneal metastasis.

Huocong Huang, M.D., Ph.D.
Huocong Huang, M.D., Ph.D., is Assistant Professor of Surgery and Immunology and a member of the Harold C. Simmons Comprehensive Cancer Center at UT Southwestern.

“We have uncovered two groups of fibroblasts integrated within tumors and defined their spatial microenvironment that control the mechanism of cancer growth, metastasis, and treatment resistance, suggesting a new strategy to therapeutically target a tumor’s supportive tissue,” said Huocong Huang, M.D., Ph.D., Assistant Professor of Surgery and Immunology at UT Southwestern. Dr. Huang co-led the study with Alex Kim, M.D., Ph.D., Assistant Professor of Surgery. Both are members of the Harold C. Simmons Comprehensive Cancer Center at UT Southwestern.

Over the past two decades, researchers have had a growing understanding that cancerous tumors aren’t made only of cancer cells but instead are a heterogeneous mixture of many cell types. These include fibroblasts, which play vital roles in preserving tissue integrity, regulating inflammatory response, and facilitating wound repair. Although scientists initially thought that all cancer-associated fibroblasts (CAFs) were the same, advances in genetic profiling have shown that there are three subtypes.

In 2022, Dr. Huang’s team was among the researchers who discovered one of these subtypes, now known as apCAFs, in a mouse model of pancreatic cancer. Their findings showed that these cells express immune molecules on their surfaces and appear to regulate the activity of immune cells called T cells in tumors. However, little else was known about these cells, including whether they exist in human cancers, their origins in different cancer types, and where they tend to reside within tumors.

Alex Kim, M.D., Ph.D.
Alex Kim, M.D., Ph.D., is Assistant Professor of Surgery and a member of the Harold C. Simmons Comprehensive Cancer Center at UT Southwestern. He is a Eugene P. Frenkel, M.D. Scholar in Clinical Medicine and Director of the Peritoneal Surface Malignancy Program.

To learn more, Dr. Huang, Dr. Kim, and their UTSW colleagues combined information from multiple existing datasets of human cancer single-cell RNA sequencing (scRNA-seq). The technique allows researchers to analyze gene activity at the level of individual cells, making it possible to distinguish different cell types. Using this wealth of data – generated from more than 2.5 million cells in 532 samples across 15 different cancer types – they generated an atlas of cell types found within these tumors.

These results not only confirmed the existence of apCAFs in most of the cancer types – suggesting that they may be universally present in human cancers – but also showed a particularly large number of these cells in both pancreatic cancers and CRC peritoneal metastases. Notably, the big data analysis revealed that the apCAFs themselves aren’t a uniform population but instead form two distinct groups: cells that appear to come from the mesothelium, a tissue that lines body cavities and internal organs, and others that appear to come from bone marrow.

These two populations seem to perform separate roles in the tumors, Dr. Huang explained. Spatial analysis showed that the mesothelium-associated apCAFs tended to be located near cancer cells, and the bone marrow-associated apCAFs tended to be located near immune cells called lymphocytes. Their locations suggest that the two apCAF types could influence tumor behavior by interacting with different kinds of cells.

Additional experiments showed that both types of apCAFs produce a protein called secreted phosphoprotein 1 (SPP1), which facilitates the growth and spread of cancer and promotes chemotherapy resistance. When the researchers removed SPP1 through genetic manipulation in mouse models of primary pancreatic cancer and CRC peritoneal metastasis, tumors grew and migrated far more slowly and became more sensitive to chemotherapy.

Together, these results suggest that apCAFs could represent new targets for treating cancer and that SPP1 could be both a target and a biomarker that doctors might use to track cancer progression. This is particularly important for patients diagnosed with peritoneal metastases of colorectal cancer, where diagnostic and treatment options are completely lacking, resulting in very poor patient outcomes, Dr. Kim said. SPP1-inhibiting drugs are already being tested in clinical trials for other diseases, and their use could potentially be translated to these cancer settings, he added.

“Our findings could lead to big opportunities to make a huge impact for patients with limited or no treatment options,” said Dr. Kim, a Eugene P. Frenkel, M.D. Scholar in Clinical Medicine and Director of the Peritoneal Surface Malignancy Program.

Other UTSW researchers who contributed to the study are first author Xiongfeng Chen, Ph.D., postdoctoral researcher; Herbert J. Zeh III, M.D., Chair and Professor of Surgery; Patricio M. Polanco, M.D., Professor of Surgery, Director of Robotic Surgery Training, Co-Director of the Pancreatic Cancer Program, and Co-Director of the Pancreatic Cancer Prevention Clinic; Zhuan Zhou, Ph.D., Assistant Professor of Surgery; Luyu “Amber” Xie, Ph.D., Pharm.D., Assistant Professor in the Peter O’Donnell Jr. School of Public Health; Zeynep Yazgan, B.S., Research Technician; Yang Liu, Ph.D., Research Scientist; Bo Zhang, M.S., Lab Manager; Kailiang Qiao, Ph.D., and Yiyue Jia, Ph.D., postdoctoral researchers; and Shunheng Liu, undergraduate student assistant.

Drs. Zeh, Polanco, and Zhou are also Simmons Cancer Center members.

This study was funded by the National Institutes of Health (R00 CA252009) and a National Cancer Institute (NCI) Cancer Center Support Grant (P30CA142543).

About UT Southwestern Medical Center 

UT Southwestern, one of the nation’s premier academic medical centers, integrates pioneering biomedical research with exceptional clinical care and education. The institution’s faculty members have received six Nobel Prizes and include 24 members of the National Academy of Sciences, 23 members of the National Academy of Medicine, and 13 Howard Hughes Medical Institute Investigators. The full-time faculty of more than 3,200 is responsible for groundbreaking medical advances and is committed to translating science-driven research quickly to new clinical treatments. UT Southwestern physicians provide care in more than 80 specialties to more than 140,000 hospitalized patients, more than 360,000 emergency room cases, and oversee nearly 5.1 million outpatient visits a year.

]]>
https://www.utsouthwestern.edu/newsroom/articles/year-2025/oct-hard-to-treat-cancers.html Mon, 06 Oct 2025 08:05:00 -0500
<![CDATA[UTSW Research: Diagnosing Alzheimer’s, timed radiation therapy, and more]]> Smiling Senior Man With Dementia Standing Outside With Crutch Looking At Smiling Senior Woman Holding Him Close
(Photo credit: Getty Images)

Cerebrovascular Dynamics Index could help diagnose Alzheimer’s

The buildup of two pathological proteins, amyloid beta and phosphorylated tau, have long been considered hallmarks of Alzheimer’s disease. However, efforts to diagnose Alzheimer’s by searching for these proteins in spinal taps, PET imaging, and blood samples have drawbacks, including high cost and relatively low accuracy.

Research has shown that Alzheimer’s impairs vasomotor reactivity, in which the brain’s blood vessels dilate when carbon dioxide builds up in the blood. To determine whether this effect could be used to diagnose Alzheimer’s, two researchers from UT Southwestern Medical Center joined colleagues to develop the Cerebrovascular Dynamics Index (CDI). This noninvasive test uses Doppler ultrasound to measure blood flow velocity in some main arteries of the brain and near-infrared spectroscopy to measure oxygenation in the brain’s cortex.

Results from nearly 200 participants, published in Alzheimer’s & Dementia: Diagnosis, Assessment & Disease Monitoring, showed that the CDI distinguished among mild cognitive impairment, Alzheimer’s, and healthy individuals with better accuracy than existing methods. The authors say this test has significant promise to improve Alzheimer’s disease diagnosis and staging.

The study authors from UT Southwestern are Rong Zhang, Ph.D., Professor of Neurology, Biomedical Engineering, and Internal Medicine, an Investigator in the Peter O’Donnell Jr. Brain Institute, and Director of the Cerebrovascular Laboratory in the Institute for Exercise and Environmental Medicine at Texas Health Presbyterian Dallas; and Danilo Cardim, Ph.D., Instructor of Neurology.

Timed radiation extends survival for some non-small cell lung cancers

A subset of non-small cell lung cancer (NSCLC), the most common lung cancer type, has mutations in a molecule known as epidermal growth factor receptor (EGFR). Although EGFR-targeting drugs often work well initially to fight these cancers, most cases develop resistance and progress within two years. A study led by UT Southwestern researchers and published in eClinicalMedicine suggests that precisely timed radiation could be a useful additional treatment.

Results from a clinical trial involving 42 patients showed that highly targeted radiation therapy delivered eight weeks after drug initiation extended progression-free survival more than one year compared to outcomes seen with the drug alone. Patients on the new protocol also lived longer, with few additional side effects.

The study was led by Sawsan Rashdan, M.D., Associate Professor of Internal Medicine in the Division of Hematology and Oncology, and David Gerber, M.D., Professor of Internal Medicine and in the Peter O’Donnell Jr. School of Public Health. Dr. Rashdan is Director of Thoracic Medical Oncology Clinical Operations and a member of the Harold C. Simmons Comprehensive Cancer Center at UTSW. Dr. Gerber serves as Co-Director of Education and Training in the Simmons Cancer Center.

Investigating how airway cells respond to pathogens

Airway epithelial surfaces are the primary contact point for numerous infectious bacteria and viruses, including those that cause tuberculosis, measles, COVID-19, the common cold, and influenza. Rare epithelial microfold cells (M cells) can serve as an entry site and initiate early immune responses to these pathogens. Although M cells have been extensively studied in the intestines, little is known about their development and function in the airway.

To better understand these cells and how the body responds to airborne pathogens, UT Southwestern researchers and colleagues analyzed single cell gene expression to identify cell types in the adenoid – an airway immune organ where M cells are found – and defined their developmental trajectories and relationships. Their study in Mucosal Immunology identified 26 unique cell types and determined that airway M cells arise from progenitor club cells and have a distinct gene expression signature consistent with their ability to shuttle particles and pathogens from the airway surface to waiting immune cells.

The researchers also found a previously unknown cell type that appears to be primed by the immune molecule interferon to prevent early infection. This research could eventually lead to new treatments and vaccines for airway infections.

UTSW contributors to this study are first author Samuel Alvarez-Arguedas, Ph.D., Assistant Instructor of Internal Medicine in the Division of Infectious Diseases and Geographic Medicine; senior author Michael Shiloh, M.D., Ph.D., Professor of Internal Medicine in the Division of Infectious Diseases and Geographic Medicine and of Microbiology; Ron Mitchell, M.D., Professor of Otolaryngology-Head & Neck Surgery and Pediatrics; and John Lafin, Ph.D., Computational Biologist.

About UT Southwestern Medical Center

UT Southwestern, one of the nation’s premier academic medical centers, integrates pioneering biomedical research with exceptional clinical care and education. The institution’s faculty members have received six Nobel Prizes and include 24 members of the National Academy of Sciences, 23 members of the National Academy of Medicine, and 13 Howard Hughes Medical Institute Investigators. The full-time faculty of more than 3,200 is responsible for groundbreaking medical advances and is committed to translating science-driven research quickly to new clinical treatments. UT Southwestern physicians provide care in more than 80 specialties to more than 140,000 hospitalized patients, more than 360,000 emergency room cases, and oversee nearly 5.1 million outpatient visits a year.

]]>
https://www.utsouthwestern.edu/newsroom/articles/year-2025/sept-research-roundup.html Mon, 22 Sep 2025 06:00:00 -0500
<![CDATA[UTSW named Specialized Program of Research Excellence for liver cancer]]> Group of men posing for the camera in a lab setting
The principal investigators at UT Southwestern Medical Center involved in the Specialized Program of Research Excellence (SPORE) for liver cancer are (from left) Hao Zhu, M.D.; Amit Singal, M.D., M.S.; David Hsieh, M.D.; Yujin Hoshida, M.D., Ph.D.; Adam Yopp, M.D.; and Daolin Tang, M.D., Ph.D.

DALLAS – Sept. 17, 2025 – The Liver Tumor Program at UT Southwestern Medical Center’s Harold C. Simmons Comprehensive Cancer Center has been selected by the National Cancer Institute as a Specialized Program of Research Excellence (SPORE). Accompanied by a $12 million grant over five years, the SPORE aims to speed the development of new ways to treat and prevent liver cancer.

Dr. Hoshida and Dr. Singal
Yujin Hoshida, M.D., Ph.D., (left) Professor of Internal Medicine and Director of Liver Tumor Translational Research, will co-lead the liver cancer SPORE at UT Southwestern with Amit Singal, M.D., M.S., Professor of Internal Medicine and in the Peter O’Donnell Jr. School of Public Health as well as Medical Director of the Liver Tumor Program and Chief of Hepatology at UTSW.

“We’re quite honored to be chosen as a liver cancer SPORE. Our selection speaks to the quality of science and the potential for discovery and advances in clinical research and care at UT Southwestern,” said Amit Singal, M.D., M.S., Professor of Internal Medicine and in the Peter O’Donnell Jr. School of Public Health as well as Medical Director of the Liver Tumor Program and Chief of Hepatology at UTSW. Dr. Singal will co-lead the liver cancer SPORE with Yujin Hoshida, M.D., Ph.D., Professor of Internal Medicine and Director of Liver Tumor Translational Research.

Dr. Singal explained that the SPORE’s overarching goal is to transform innovative scientific discoveries from UTSW into precision interventions that reduce liver cancer incidence and mortality. The effort will have a special focus on hepatocellular carcinoma (HCC), which accounts for over 85% of liver cancer cases across the nation. More than 42,000 new cases of liver cancer, including HCC, are diagnosed every year in the U.S., and more than 30,000 Americans die of liver cancer annually. Cirrhosis from heavy alcohol use, metabolic dysfunction, viral hepatitis, and some genetic variations are key risk factors for HCC and other liver cancers.

These risk factors are particularly common in Texas, which has one of the highest incidences of HCC in the country, Dr. Hoshida said. UTSW joins the Mayo Clinic and The University of Texas MD Anderson Cancer Center as liver cancer SPOREs across the nation.

“Our goal is to significantly improve liver cancer survival rates by refining the prevention and treatment of this disease through various new approaches,” he said.

The bulk of the SPORE grant will fund three research projects.

Reducing risk of developing HCC

The first, led by Drs. Singal and Hoshida, will test a promising “chemoprevention” strategy in people considered at high risk of developing HCC due to existing cirrhosis. Previous research in animal models at UTSW showed that a protein known as the epidermal growth factor receptor (EGFR) appears to foster cirrhosis-driven HCC development. By suppressing activity of this protein through an EGFR-inhibiting drug called erlotinib, researchers reduced the risk of HCC in these mouse models, a result mirrored in a recent phase one clinical trial in patients with cirrhosis. The UTSW team plans to test this strategy in a larger number of patients in a phase two trial, using a novel biomarker they discovered called a prognostic liver secretome signature as a proxy for HCC risk.

Preventing recurrence of HCC

The second project, led by Hao Zhu, M.D., Professor in Children’s Medical Center Research Institute at UT Southwestern and of Internal Medicine and Pediatrics, and David Hsieh, M.D., Associate Professor of Internal Medicine, will focus on preventing recurrence of HCC in cirrhosis patients who were previously treated. Tumors regrow within two years in about 50%-70% of these patients because the risk factors that caused the initial disease are still present, Dr. Zhu explained. Having extra chromosomes in liver cells – a condition called polyploidy – has been shown to be protective against developing HCC. Previous research in the Zhu Lab showed that reducing a protein known as anillin can induce polyploidy and reduce development of HCC. The researchers plan to test this anillin-targeting strategy in a phase one clinical trial.

Improving immunotherapy to prevent HCC recurrence

The third project, led by Daolin Tang, M.D., Ph.D., Professor of Surgery, and Adam Yopp, M.D., Professor of Surgery and Division Chief of Surgical Oncology and Surgical Director of the Liver Tumor Program, will focus on improving the efficacy of immunotherapy to prevent HCC recurrence. Although immunotherapy drugs – which harness the immune system to fight cancer – have shown promise in treating advanced stage HCC, their benefit in preventing recurrence after HCC tumors are surgically removed has been unclear. To boost their performance, the researchers plan to test these drugs in combination with a drug known as a telomerase reverse transcriptase (TERT) inhibitor. This agent has been shown to selectively stop HCC cells from multiplying and kill them while also activating a cancer-fighting subset of immune cells. A planned phase one b trial will test the safety and efficacy of the TERT inhibitor combined with immune checkpoint inhibitors in patients who will undergo surgery to remove HCC tumors.

Additional funding

Along with these projects, the SPORE grant will also fund a Developmental Research Program, which will provide seed funding to launch new high-risk, high-reward projects, and a Career Enhancement Program, which will aid early-career scientists and clinicians interested in translational liver cancer research. In addition, the grant will support three cores: an Administrative and Outreach Core, which will provide essential administrative services for the SPORE; a Biospecimen and Pathology Core, which will house patient-derived tissue and blood samples needed for research; and a Data Science Core, which will provide biostatistical and bioinformatic data analysis support to researchers.

“The successful funding of the liver SPORE is a major accomplishment of the Simmons Cancer Center. It represents an outstanding example of the multidisciplinary and collaborative science destined to have a major impact on the prevention and treatment of this lethal disease in the state of Texas and beyond as well as a testament to UTSW’s commitment for translational research,” said Carlos L. Arteaga, M.D., Director of the Simmons Cancer Center and Associate Dean of Oncology Programs at UT Southwestern.

UTSW is home to two other SPOREs for lung cancer and kidney cancer research.

Dr. Singal is a Dedman Family Scholar in Clinical Care and holds the Willis C. Maddrey, M.D. Distinguished Chair in Liver Disease. Dr. Hoshida holds the H. Ray and Paula Calvert Chair in Gastroenterology Oncology in Honor of Udit Verma, M.D. Dr. Zhu holds the Nancy B. and Jake L. Hamon Distinguished Chair in Therapeutic Oncology Research and is a Cancer Prevention and Research Institute of Texas Scholar in Cancer Research. Dr. Yopp holds The Occidental Chemical Chair in Cancer Research. Dr. Arteaga holds the Annette Simmons Distinguished University Chair in Breast Cancer Research.

Drs. Singal, Hoshida, Zhu, Hsieh, Tang, and Yopp are members of Simmons Cancer Center. Dr. Zhu is also co-leader of the Simmons Cancer Center Development and Cancer Research Program.

About UT Southwestern Medical Center

UT Southwestern, one of the nation’s premier academic medical centers, integrates pioneering biomedical research with exceptional clinical care and education. The institution’s faculty members have received six Nobel Prizes and include 24 members of the National Academy of Sciences, 23 members of the National Academy of Medicine, and 13 Howard Hughes Medical Institute Investigators. The full-time faculty of more than 3,200 is responsible for groundbreaking medical advances and is committed to translating science-driven research quickly to new clinical treatments. UT Southwestern physicians provide care in more than 80 specialties to more than 140,000 hospitalized patients, more than 360,000 emergency room cases, and oversee nearly 5.1 million outpatient visits a year.

]]>
https://www.utsouthwestern.edu/newsroom/articles/year-2025/sept-liver-cancer.html Wed, 17 Sep 2025 06:00:00 -0500
<![CDATA[Biomarkers linked to side effects from cancer immunotherapy]]> Nurse checking the vital signs of a patient at the hospital during a medical exam
Researchers at UT Southwestern have identified key features in blood linked to a higher risk of side effects among patients with cancer who are treated with immunotherapy, creating a path to potentially improve patient care in the future. (Photo credit: Getty Images)

DALLAS – Sept. 16, 2025 – A team led by UT Southwestern Medical Center scientists has identified early signals from the immune system that could help predict which cancer patients are most likely to develop harmful side effects from immunotherapy. The findings, published in the Journal for ImmunoTherapy of Cancer, offer a path toward tests to help doctors tailor care for at-risk patients.

David Gerber, M.D.
David Gerber, M.D., is Professor of Internal Medicine in the Division of Hematology and Oncology and in the Peter O’Donnell Jr. School of Public Health at UT Southwestern. He is also co-Director of the Office of Education and Training in the Harold C. Simmons Comprehensive Cancer Center. He holds the David Bruton, Jr. Professorship in Clinical Cancer Research.

“Through a multi-omic biomarker analysis, we identified a pre-existing but clinically silent proinflammatory state in patients with increased risk of immunotherapy toxicities,” said David Gerber, M.D., Professor of Internal Medicine in the Division of Hematology and Oncology and in the Peter O’Donnell Jr. School of Public Health at UT Southwestern. Dr. Gerber, who is also co-Director of the Office of Education and Training in the Harold C. Simmons Comprehensive Cancer Center, co-led the study with first author Shaheen Khan, Ph.D., who was previously Assistant Professor of Pathology at UT Southwestern.

Immune checkpoint inhibitors – drugs that enhance the immune system’s ability to attack cancer – have transformed treatment for many cancers and extended lives even in patients with advanced disease. But they can also spark reactions that damage healthy organs, sometimes causing serious and lasting complications. More than half of patients receiving immunotherapy experience side effects, which remain difficult to predict or diagnose.

By analyzing blood from 162 patients at UT Southwestern and Parkland Health before and after immunotherapy treatment, researchers identified three key features linked to higher risk: elevated levels of antibody-producing cells and autoantibodies, stronger activity from inflammatory molecules such as interferon-gamma, and heightened signals from another key inflammatory molecule known as tumor necrosis factor or TNF. Patients with these immune profiles were more likely to develop side effects once treatment began.

Mitchell von Itzstein, M.D.
Mitchell von Itzstein, M.D., is Assistant Professor of Internal Medicine in the Division of Hematology and Oncology at UT Southwestern and a member of the Harold C. Simmons Comprehensive Cancer Center.

“If prospectively verified, these findings potentially impact patients with any cancer types that are treated with immunotherapy,” said study co-author Mitchell von Itzstein, M.D., Assistant Professor of Internal Medicine in the Division of Hematology and Oncology at UT Southwestern. “Currently, immunotherapy is used to treat most cancers in advanced stages as well as some cancers in earlier stages.”

The study builds on more than a decade of work at the UT Southwestern Simmons Cancer Center, where Dr. Gerber and other researchers have developed an institutional registry of immunotherapy patients and biospecimens. With contributions from more than 800 patients cared for at UT Southwestern and Parkland Health, the registry has enabled one of the most comprehensive efforts to date to connect immune, genetic, and antibody changes with treatment complications.

While the results are promising, the authors noted further research is needed to confirm these biomarkers in larger and more diverse groups and translate them into tools for patient care.

Jeffrey A. SoRelle, M.D.
Jeffrey A. SoRelle, M.D., is Assistant Professor of Pathology and Pediatrics and a member of the Immunology Graduate Program at UT Southwestern.

“The prediction, diagnosis, treatment, and monitoring of immune-related side effects remain major clinical challenges,” said study co-author Jeffrey A. SoRelle, M.D., Assistant Professor of Pathology and Pediatrics and a member of the UTSW Immunology Graduate Program. “Identifying the molecular mechanisms behind these side effects may help predict which patients are at greatest risk and potentially provide guidance for the treatment of these toxicities.”

Dr. Gerber holds the David Bruton, Jr. Professorship in Clinical Cancer Research. Dr. von Itzstein is a member of the Simmons Cancer Center.

A full list of contributors and their disclosures can be found in the published study.

This study was funded by the National Institute of Allergy and Infectious Diseases (1U01AI156189-01 and K08AI155832), an American Cancer Society-Melanoma Research Alliance Team Award (MRAT-18-114-01-LIB), a V Foundation Robin Roberts Cancer Survivorship Award (DT2019-007), a Melanoma Research Alliance and Society for Immunotherapy of Cancer Young Investigator Award (619351), the University of Texas Lung Cancer Specialized Program of Research Excellence (SPORE) (P50CA070907-21), and a National Cancer Institute (NCI) Cancer Center Support Grant (P30CA142543).

About UT Southwestern Medical Center

UT Southwestern, one of the nation’s premier academic medical centers, integrates pioneering biomedical research with exceptional clinical care and education. The institution’s faculty members have received six Nobel Prizes and include 24 members of the National Academy of Sciences, 23 members of the National Academy of Medicine, and 13 Howard Hughes Medical Institute Investigators. The full-time faculty of more than 3,200 is responsible for groundbreaking medical advances and is committed to translating science-driven research quickly to new clinical treatments. UT Southwestern physicians provide care in more than 80 specialties to more than 140,000 hospitalized patients, more than 360,000 emergency room cases, and oversee nearly 5.1 million outpatient visits a year.

About Parkland Health

Parkland Health is one of the largest public hospital systems in the country. Premier services at the state-of-the-art Parkland Memorial Hospital include the Level I Rees-Jones Trauma Center, the only burn center in North Texas verified by the American Burn Association for adult and pediatric patients, and a Level III Neonatal Intensive Care Unit. The system also includes two on-campus outpatient clinics – the Ron J. Anderson, MD Clinic and the Moody Outpatient Center, as well as more than 30 community-based clinics and numerous outreach and education programs. By cultivating its diversity, inclusion, and health equity efforts, Parkland enriches the health and wellness of the communities it serves. For more information, visit parklandhealth.org.

]]>
https://www.utsouthwestern.edu/newsroom/articles/year-2025/sept-immunotherapy-biomarkers.html Tue, 16 Sep 2025 08:00:00 -0500
<![CDATA[UT Southwestern biochemist Steven McKnight, Ph.D., earns Lasker Award]]>

DALLAS – Sept. 11, 2025 – Steven McKnight, Ph.D., Professor of Biochemistry at UT Southwestern Medical Center, has been awarded the Albert Lasker Basic Medical Research Award for discoveries into the role of proteins of low sequence complexity and their influence on the dynamics of cell morphology and biological regulation.

Often called “America’s Nobels,” the Lasker Awards recognize significant advances in the understanding, diagnosis, treatment, cure, and prevention of human disease and are regarded as the country’s preeminent biomedical research prize. Since 1945, the Lasker Foundation has awarded more than 400 prizes.

Steven McKnight, Ph.D.
Steven McKnight, Ph.D.

Dr. McKnight’s recognition marks the second consecutive year and fifth time that a UT Southwestern scientist has earned a Lasker Award. With this honor, he joins Zhijian “James” Chen, Ph.D. (2024), Professor of Molecular Biology and Director of the Center for Inflammation Research, and Nobel Laureates Alfred Gilman, M.D., Ph.D. (1989), Michael Brown, M.D. (1985), and Joseph Goldstein, M.D. (1985), as UT Southwestern recipients.

Dr. McKnight’s studies – focused on proteins of low sequence complexity – have revealed how these disordered proteins can reversibly self-associate to control innumerable forms of dynamic cellular organization and aggregate in a manner that leads to neurologic and neurodegenerative disease.

“His work over the past three decades has exemplified our institution’s commitment to curiosity-driven research by advancing our understanding of cellular and molecular mechanisms, which ultimately inform new approaches to disease treatment,” said Daniel K. Podolsky, M.D., President of UT Southwestern. “We are thrilled to see the importance of his fundamental discoveries into the role of low complexity proteins in basic cellular functions recognized by this year’s Lasker Basic Medical Research Award.”

Scientist Snapshot

  • Steven McKnight, Ph.D.
  • Born: El Paso, Texas
  • Education: University of Texas, University of Virginia
  • Joined UTSW: In 1995 and appointed Chair of the Department of Biochemistry in 1996
  • Research focus: Gene regulation and intracellular signaling
  • Key discoveries: Proteins of low sequence complexity and their effects controlling the dynamics of cell morphology as well as a chemical inhibitor of the HIF2a protein now used as a treatment of kidney cancer.
  • Fast fact: Dr. McKnight dropped out of college and enlisted in the Army, where he served on an armored tank crew in Vietnam. He credits the military for instilling the discipline he needed to become a successful scientist.

Prior to the work that has garnered the Lasker Award, Dr. McKnight’s early studies of gene regulation led to the identification of the leucine zipper, a structural motif in transcription factors – proteins that regulate gene expression. This discovery helped clarify how the expression of cellular genes is turned on and off.

Teaming up with David Russell, Ph.D., Professor Emeritus and former Vice Provost and Dean of Basic Research, Dr. McKnight also discovered the HIF-2α transcription factor and identified its role in adapting cells and tissues to conditions of oxygen starvation. Working in collaboration with Richard Bruick, Ph.D., and synthetic chemists in the Biochemistry Department at UT Southwestern, Dr. McKnight discovered drug-like compounds capable of inhibiting the HIF-2α protein. In 2008, UT Southwestern licensed these chemicals to Peloton Therapeutics Inc., a Dallas-based biotechnology company founded by Dr. McKnight. Following extensive optimization and subsequent clinical trials, the chemical inhibitor of HIF-2α, designated belzutifan, was approved in 2021 by the Food and Drug Administration as a treatment for kidney cancer.

“I want to thank the Lasker Foundation for this great honor,” said Dr. McKnight, who holds the Distinguished Chair in Basic Biomedical Research at UT Southwestern. “I am also grateful to the colleagues and trainees who’ve worked with me over the years and to the leadership of UT Southwestern, who have created the environment for scientists to probe challenging and important questions.”

A former Howard Hughes Medical Institute Investigator, Dr. McKnight is a member of the National Academy of Sciences, the National Academy of Medicine, and the American Academy of Arts and Sciences. Other honors include the Robert A. Welch Award in Chemistry (2020), the Wiley Prize in Biomedical Sciences (2014), the National Institutes of Health Director’s Pioneer Award (2004), the Monsanto Award from the National Academy of Sciences (1991), and the Eli Lilly Award from the American Society for Microbiology (1989).

Low complexity domains: Structures and functions
(Credit: Lasker Foundation)

Dr. McKnight earned a bachelor’s degree in biology from the University of Texas at Austin, followed by a Ph.D. in biology from the University of Virginia. He did postdoctoral research at the Carnegie Institution of Washington before joining UT Southwestern in 1995. He is a member of the Harold C. Simmons Comprehensive Cancer Center.

Dr. McKnight shares the 2025 Albert Lasker Basic Medical Research Award with Dirk Görlich, Ph.D., a German biochemist who is director of the Max Planck Institute for Multidisciplinary Sciences. Dr. Görlich is also being honored for his work on proteins of low sequence complexity.

The Lasker Awards will be presented in New York on Sept. 19.

Dr. Brown is a Regental Professor and holds the W.A. (Monty) Moncrief Distinguished Chair in Cholesterol and Arteriosclerosis Research and the Paul J. Thomas Chair in Medicine.

Dr. Chen holds the George L. MacGregor Distinguished Chair in Biomedical Science.

Dr. Goldstein is a Regental Professor and holds the Julie and Louis A. Beecherl, Jr. Distinguished Chair in Biomedical Research and the Paul J. Thomas Chair in Medicine.

Dr. Podolsky holds the Philip O’Bryan Montgomery, Jr., M.D. Distinguished Presidential Chair in Academic Administration and the Charles Cameron Sprague Distinguished Chair in Biomedical Science.

About UT Southwestern Medical Center

UT Southwestern, one of the nation’s premier academic medical centers, integrates pioneering biomedical research with exceptional clinical care and education. The institution’s faculty members have received six Nobel Prizes and include 24 members of the National Academy of Sciences, 23 members of the National Academy of Medicine, and 13 Howard Hughes Medical Institute Investigators. The full-time faculty of more than 3,200 is responsible for groundbreaking medical advances and is committed to translating science-driven research quickly to new clinical treatments. UT Southwestern physicians provide care in more than 80 specialties to more than 140,000 hospitalized patients, more than 360,000 emergency room cases, and oversee nearly 5.1 million outpatient visits a year.

]]>
https://www.utsouthwestern.edu/newsroom/articles/year-2025/sept-lasker.html Thu, 11 Sep 2025 08:00:00 -0500
<![CDATA[UT Southwestern biochemist Zhijian ‘James’ Chen to receive 2026 Brinster Prize]]> Zhijian “James” Chen, Ph.D.
Zhijian “James” Chen, Ph.D., is one of the world’s top experts on innate immunity.

DALLAS – Sept. 9, 2025 – Zhijian “James” Chen, Ph.D., Professor in the Department of Molecular Biology at UT Southwestern Medical Center and one of the world’s top researchers on innate immunity, has been awarded the 2026 Elaine Redding Brinster Prize in Science or Medicine in recognition of his discovery of the cGAS enzyme and its role in immune response.

“I am extremely honored and humbled to be selected as the fifth recipient of the Elaine Redding Brinster Prize,” said Dr. Chen, who is a Howard Hughes Medical Institute Investigator and Director of the Center for Inflammation Research at UT Southwestern. “This prize is very special not only because Dr. Ralph Brinster is one of my scientific heroes but also because it epitomizes the key role of family support in the success of a scientist. I myself have benefited from the strong support of my family, as well as my colleagues and mentors. This prize is a recognition of the hard work and dedication of the researchers, trainees, and staff members in my lab who have contributed to the discoveries of cGAS and other molecules involved in our body’s immune defense.”

Scientist Snapshot

  • Zhijian “James” Chen, Ph.D.
  • Born: Fujian Province, China
  • Education: Fujian Normal University; State University of New York at Buffalo
  • Joined UTSW: In 1997, when he was recruited to the new Department of Molecular Biology
  • Research focus: Innate immunity
  • Key discovery: The DNA-sensing enzyme cGAS, which acts as a “burglar alarm” to trigger the body’s immune system when it detects a pathogen
  • Fast fact: Dr. Chen identified the first mitochondrial protein known to be involved in immune defense against microbial infections in 2005. He named it MAVS (mitochondrial antiviral signaling) in honor of his favorite basketball team, the Dallas Mavericks.

The Brinster Prize is awarded annually by the Institute for Regenerative Medicine at the University of Pennsylvania to a researcher whose discovery has made a unique impact on biomedicine. It is supported by an endowment from the children of Elaine Redding Brinster and Ralph L. Brinster, V.M.D., Ph.D., the Richard King Mellon Professor of Reproductive Physiology at the University of Pennsylvania and a National Medal of Science recipient.

Announcing the award, Penn Medicine said Dr. Chen’s research has “opened up a new area of science by showing how our immune system spots harmful DNA from germs, like bacteria and viruses, and starts fighting them.”

Dr. Chen’s discoveries include MAVS, the first mitochondrial protein known to be involved in immunity against infections. The protein’s name both describes its function (mitochondrial antiviral signaling) and honors his favorite basketball team, the Dallas Mavericks. In 2012, his laboratory identified cGAS (cyclic GMP-AMP synthase), which triggers the innate immune system when it detects foreign DNA inside a cell. He and his colleagues are now studying the complex biochemical pathways by which cGAS works.

Dr. Chen’s research has been recognized with some of the most esteemed awards in science, including the Albert Lasker Basic Medical Research Award (2024), the Louisa Gross Horwitz Prize (2023), and the Breakthrough Prize in Life Sciences (2019).

Dr. Chen is a member of both the National Academy of Sciences and the National Academy of Medicine. At UTSW, he is a member of the Center for the Genetics of Host Defense as well as the Harold C. Simmons Comprehensive Cancer Center. He holds the George L. MacGregor Distinguished Chair in Biomedical Science.

The Brinster Prize will be presented March 18, 2026, as part of the daylong Ralph L. Brinster Symposium at Penn’s Philadelphia campus. The prize comes with an award of $200,000.

About UT Southwestern Medical Center

UT Southwestern, one of the nation’s premier academic medical centers, integrates pioneering biomedical research with exceptional clinical care and education. The institution’s faculty members have received six Nobel Prizes and include 24 members of the National Academy of Sciences, 23 members of the National Academy of Medicine, and 13 Howard Hughes Medical Institute Investigators. The full-time faculty of more than 3,200 is responsible for groundbreaking medical advances and is committed to translating science-driven research quickly to new clinical treatments. UT Southwestern physicians provide care in more than 80 specialties to more than 140,000 hospitalized patients, more than 360,000 emergency room cases, and oversee nearly 5.1 million outpatient visits a year.

]]>
https://www.utsouthwestern.edu/newsroom/articles/year-2025/sept-brinster-prize.html Tue, 09 Sep 2025 08:00:00 -0500
<![CDATA[Ribosome level changes detected in early brain development]]> This image shows a human brain organoid
This image shows a human brain organoid. Neuroepithelial cells are in red. Individual cells are labeled green to highlight the morphological changes they undergo as they differentiate into radial glia cells.

DALLAS – Aug. 04, 2025 – A team led by UT Southwestern Medical Center scientists has identified a specific stage of neurodevelopment when differentiating neural cells produce fewer ribosomes, which are responsible for making proteins. This subsequent drop in protein production, they report in Nature Cell Biology, helps explain why mutations that further affect ribosome production can cause neurodevelopmental disorders.

Michael Buszczak, Ph.D.
Michael Buszczak, Ph.D., is Professor of Molecular Biology and a member of the Development and Cancer Research Program in the Harold C. Simmons Comprehensive Cancer Center at UT Southwestern. He holds the Lillian B. and Tom B. Rhodes Professorship in Stem Cell Research and is an E.E. and Greer Garson Fogelson Scholar in Medical Research. 

“We find that ribosome levels decrease during neuroepithelial differentiation, a very early step in human brain development, making differentiating cells particularly vulnerable to changes in ribosome biogenesis during this time,” said Michael Buszczak, Ph.D., Professor of Molecular Biology at UT Southwestern.

Dr. Buszczak co-led the study with UT Southwestern collaborators Jun Wu, Ph.D., Associate Professor of Molecular Biology; Chunyang Ni, Ph.D., a former researcher in the Buszczak Lab who is now at Stanford University; and Yudong Wei, Ph.D., postdoctoral fellow in the Buszczak Lab. International collaborators were Barbara Vona, Ph.D., Group Leader of the Institute for Auditory Neuroscience at University Medical Center Gottingen in Germany, and Reza Maroofian, Ph.D., Research Fellow at University College London.

The researchers homed in on a group of neurodevelopmental disorders that share a set of features, including severe intellectual disability, low muscle tone, hearing and vision impairment, and small brain size. These disorders are linked to mutations in a gene known as AIRIM that plays a key role in generating ribosomes. But how the genetic mutations cause these features has been unknown.

Working with the Wu Lab – which specializes in creating model systems called organoids that replicate the development of organs – the researchers genetically manipulated cells carrying the genetic defects to revert into stem cells that formed brain organoids. For comparison, they did the same with defect-free cells.

Jun Wu, Ph.D.
Jun Wu, Ph.D., is Associate Professor of Molecular Biology at UT Southwestern and a Virginia Murchison Linthicum Scholar in Medical Research. 

They then tracked the organoids’ development. By day 15 – a crucial point in which specific descendants of stem cells called neuroepithelial cells transition into more specialized cells called radial glia – organoids made from mutated cells were smaller and more of their cells died.  

A closer look showed that cells in both types of organoids made fewer ribosomes during this time. However, organoids carrying mutations in AIRIM had even fewer ribosomes than normal. Subsequent experiments showed that this dearth of ribosomes in the mutated organoids led their cells to produce lower levels of certain proteins, particularly those involved in cell survival and cell differentiation.

By genetically or pharmaceutically prompting cells to increase the activity of mTOR – a protein that encourages protein production – the researchers “rescued” the cells carrying the genetic defects. These cells then formed organoids of the same size and similar protein production to those made of nonmutated cells. Dr. Buszczak suggested that a similar intervention could someday be used to treat some neurodevelopmental disorders caused by ribosome deficiencies in patients before they’re born, potentially sparing them from the symptoms of these neurodevelopmental conditions. He and his colleagues plan to investigate whether the symptoms of other neurodevelopmental disorders caused by genetic mutations in ribosome-related genes are connected to similar natural dips in ribosome production during development.

UTSW researchers who contributed to this study are Chao Xing, Ph.D., Professor in the Eugene McDermott Center for Human Growth and Development, the Lyda Hill Department of Bioinformatics, and the Peter O’Donnell Jr. School of Public Health; Matthew Sieber, Ph.D., Assistant Professor of Physiology; Yan Liu, Ph.D., and Ashwani Kumar, M.S., Computational Biologists; Yi Ding, Ph.D., Research Associate; Masahiro Sakurai, Ph.D., Research Scientist; Emily Ballard, B.S., graduate student researcher; and Leijie Li, Ph.D., and Shenlu Qin, Ph.D., postdoctoral researchers.

Dr. Buszczak holds the Lillian B. and Tom B. Rhodes Professorship in Stem Cell Research and is an E.E. and Greer Garson Fogelson Scholar in Medical Research. He is also a member of the Development and Cancer Research Program in the Harold C. Simmons Comprehensive Cancer CenterDr. Wu is a Virginia Murchison Linthicum Scholar in Medical Research. 

About UT Southwestern Medical Center 

UT Southwestern, one of the nation’s premier academic medical centers, integrates pioneering biomedical research with exceptional clinical care and education. The institution’s faculty members have received six Nobel Prizes and include 25 members of the National Academy of Sciences, 24 members of the National Academy of Medicine, and 14 Howard Hughes Medical Institute Investigators. The full-time faculty of more than 3,200 is responsible for groundbreaking medical advances and is committed to translating science-driven research quickly to new clinical treatments. UT Southwestern physicians provide care in more than 80 specialties to more than 140,000 hospitalized patients, more than 360,000 emergency room cases, and oversee nearly 5.1 million outpatient visits a year.

]]>
https://www.utsouthwestern.edu/newsroom/articles/year-2025/aug-ribosome-early-brain-development.html Mon, 04 Aug 2025 09:04:00 -0500
<![CDATA[UT Southwestern is best hospital in DFW for ninth straight year]]> Cancer malignant cells

DALLAS – July 29, 2025 – UT Southwestern Medical Center is the No. 1 hospital in Dallas-Fort Worth for the ninth consecutive year and ranks among the nation’s top hospitals for care in 12 specialties – the most of any hospital in Texas, according to U.S. News & World Report’s annual Best Hospitals list released today.

UT Southwestern ranks in the top 10 nationwide for Neurology & Neurosurgery and is among the top 25 in eight other specialties: Cancer; Cardiology, Heart & Vascular Surgery; Diabetes & Endocrinology; Ear, Nose & Throat; Geriatrics; Pulmonology & Lung Surgery; Rehabilitation; and Urology. In addition, UT Southwestern is rated “High Performing” in the great majority of evaluated conditions and procedures, from aortic valve surgery and hip fracture to back surgery (spinal fusion) and stroke care.

This recognition reflects the expertise, collaboration, and commitment to providing the very best care possible to our patients – a standard of excellence that extends across all we do, from educating future leaders in science and medicine to advancing discovery and providing exceptional care,” said Daniel K. Podolsky, M.D., President of UT Southwestern Medical Center.

A nurse giving a patient a shot
UT Southwestern is ranked among the nation's top hospitals for care in 12 specialties — the most of any hospital in Texas.

Among more than 4,400 hospitals reviewed by U.S. News, all data-driven specialties at UT Southwestern are nationally ranked. UTSW is among the top 50 nationwide in the following 12 specialties and continues to lead the state with the most nationally ranked specialties of any hospital in Texas:

UT Southwestern ranks No. 2 among all hospitals in Texas and is top-ranked in the state for Neurology & Neurosurgery and Geriatrics care. Among 22 procedures and conditions evaluated by U.S. News, UT Southwestern is designated High Performing in: abdominal aortic aneurysm repair; aortic valve surgery; back surgery (spinal fusion); chronic obstructive pulmonary disease (COPD); colon cancer surgery; diabetes; gynecological cancer surgery; heart arrhythmia; heart attack; heart failure; hip fracture; kidney failure; leukemia, lymphoma, and myeloma; lung cancer surgery; pneumonia; prostate cancer surgery; and stroke. 

Additionally, the Southwestern Health Resources network – which aligns the strengths of UT Southwestern with those of Texas Health Resources – has five of the nine top-ranked hospitals in Dallas-Fort Worth. In addition to UT Southwestern at No. 1, Texas Health Presbyterian Hospital Dallas ranked No. 5, Texas Health Harris Methodist Hospital Fort Worth ranked No. 6, Texas Health Presbyterian Hospital Plano ranked No. 7, and Texas Health Harris Methodist Hospital Southwest Fort Worth ranked No. 9. The patient-centered, clinically integrated network of 31 hospital locations and more than 7,000 physicians and other providers cares for millions of individuals across 16 counties in North Texas. Children’s Medical Center Dallas, where the UT Southwestern Pediatric Group practices, was rated among the nation’s best pediatric hospitals by U.S. News for 2024-25 and was the only pediatric hospital in North Texas ranked in all 11 specialties.

Growing to meet the needs of patients

UT Southwestern continues to expand its clinical footprint to meet the health care needs of patients in fast-growing North Texas.  

Construction is underway on a $177 million Radiation Oncology campus in Fort Worth. The 65,000-square-foot facility will include the city’s first MRI-guided precision radiation treatment.

Last fall, UT Southwestern and Children’s Health broke ground on a transformative $5 billion pediatric campus in Dallas’ Southwestern Medical District across from William P. Clements Jr. University Hospital, significantly expanding inpatient, surgical, and ambulatory capacity to meet the needs of one of the country’s fastest-growing and largest metropolitan areas. The new campus will serve as a collaborative center for innovation, academic research, training, and the advancement of lifesaving technologies.

Additionally, in recent years, UT Southwestern opened UT Southwestern Medical Center at RedBird to improve access to care for those living and working in southwestern Dallas County, as well as a regional medical center in Coppell and a nine-story Cancer Care Outpatient Building to serve patients of the Harold C. Simmons Comprehensive Cancer Center.  

Other recent national distinctions

  • Clements University Hospital was honored earlier this year for patient experience with Press Ganey’s Guardian of Excellence Award, and UTSW’s Multi-Specialty Clinic earned the Pinnacle of Excellence Award.
  • UT Southwestern Medical School is ranked in Tier 1 (top 16) for research and Tier 2 (top 50) for primary care by U.S. News in its 2025 Best Medical Schools rankings. The UT Southwestern Graduate School of Biomedical Sciences and the School of Health Professions also have nationally rated programs.
  • UT Southwestern is ranked No. 2 in the world for Endocrinology & Metabolism research and among the top 50 in seven other fields by U.S. News in its 2025-26 Best Global Universities report. Also, UT Southwestern is ranked No. 1 among global health care institutions by Nature Index for publishing high-quality research in biological sciences.

About UT Southwestern Medical Center   

UT Southwestern, one of the nation’s premier academic medical centers, integrates pioneering biomedical research with exceptional clinical care and education. The institution’s faculty members have received six Nobel Prizes and include 25 members of the National Academy of Sciences, 24 members of the National Academy of Medicine, and 14 Howard Hughes Medical Institute Investigators. The full-time faculty of more than 3,200 is responsible for groundbreaking medical advances and is committed to translating science-driven research quickly to new clinical treatments. UT Southwestern physicians provide care in more than 80 specialties to more than 140,000 hospitalized patients, more than 360,000 emergency room cases, and oversee nearly 5.1 million outpatient visits a year.

]]>
https://www.utsouthwestern.edu/newsroom/articles/year-2025/july-usnwr-best-hospital.html Tue, 29 Jul 2025 08:25:00 -0500
<![CDATA[UTSW builds AI-driven system to improve data collection ]]> Artificial Intelligence Machine Learning Large Language Model Technology
(Photo credit: Getty Images)

DALLAS – July 28, 2025 – A multidisciplinary team at UT Southwestern Medical Center has developed an artificial intelligence (AI)-enabled pipeline that can quickly and accurately extract relevant information from complex, free-text medical records. The team’s novel approach, featured in npj Digital Medicine, could dramatically reduce the time needed to create analysis-ready data for research studies.

David Hein, M.S.
David Hein, M.S., is a Data Scientist in the Lyda Hill Department of Bioinformatics at UT Southwestern.

“Constructing highly detailed, accurate datasets from free-text medical records is extremely time-consuming, often requiring extensive manual chart review,” said study first author David Hein, M.S., Data Scientist in the Lyda Hill Department of Bioinformatics at UT Southwestern. “Our study demonstrates one approach for creating AI-powered large language models (LLMs) that simplify the process of collecting and organizing medical data for analysis. By automating both data extraction and standardization through AI, we can make large-scale clinical research more efficient.”

To develop the pipeline, researchers used an AI-powered LLM to analyze over 2,200 kidney cancer pathology reports to evaluate the model’s ability to recognize and categorize distinct types of tumors. Through close collaboration with AI scientists, pathologists, clinicians, and statisticians, they refined the workflow through multiple rounds of testing, improving its handling of complex, nuanced information. Their findings were validated against existing electronic medical record (EMR) data to ensure reliability.

Payal Kapur, M.D.
Payal Kapur, M.D., is Professor of Pathology and Urology and a member of the Harold C. Simmons Comprehensive Cancer Center at UT Southwestern. She holds the Jan and Bob Pickens Distinguished Professorship in Medical Science, in Memory of Jerry Knight Rymer and Annette Brannon Rymer and Mr. and Mrs. W.L. Pickens.

The results were striking – 99% accuracy in identifying tumor types and 97% accuracy in detecting whether the cancer had metastasized.

“The biggest challenge in training AI to extract data from narrative reports is that clinicians use a wide range of open-ended terms to describe the same finding,” said study co-leader Payal Kapur, M.D., Professor of Pathology and Urology. “It’s not as simple as counting ‘yes-no’ results. Every report contains hundreds of details in narrative form. But with proper input and oversight, an AI model can efficiently review and categorize vast amounts of records with speed and accuracy.”

A final step included testing across a broader dataset of more than 3,500 internal kidney cancer pathology reports with similar results – a process facilitated by the high-quality, curated data and pipelines available through UT Southwestern’s Kidney Cancer Program.

“The key is collaborative teamwork across specialties to refine AI instructions and ensure accuracy,” said study co-author James Brugarolas, M.D., Ph.D., Director of the Kidney Cancer Program, Professor of Internal Medicine in the Division of Hematology and Oncology, and member of the Cellular Networks in Cancer Research Program of the Harold C. Simmons Comprehensive Cancer Center

James Brugarolas, M.D., Ph.D.
James Brugarolas, M.D., Ph.D., is Director of the Kidney Cancer Program, Professor of Internal Medicine in the Division of Hematology and Oncology, and member of the Cellular Networks in Cancer Research Program of the Harold C. Simmons Comprehensive Cancer Center at UT Southwestern. He holds the Sherry Wigley Crow Cancer Research Endowed Chair in Honor of Robert Lewis Kirby, M.D.

While this study focused on kidney cancer, the approach may have broader applications to other tumor types, the authors said. 

“There is no ‘one-size-fits-all’ model for medical data extraction,” said study co-leader Andrew Jamieson, Ph.D., Assistant Professor and Principal Investigator in the Lyda Hill Department of Bioinformatics. “But our study outlines key strategies that can help other researchers use AI-powered LLMs more effectively in their own specialties. We’re excited to continue refining this process and expanding AI’s role in medical research.”

Other UTSW researchers who contributed to the study are Bingqing Xie, Ph.D., Assistant Professor of Internal Medicine in the Division of Hematology and Oncology and Kidney Cancer Program; Joseph Vento, M.D., Assistant Professor of Internal Medicine in the Division of Hematology and Oncology; Lindsay Cowell, Ph.D., Professor, Peter O’Donnell Jr. School of Public Health and Department of Immunology; Scott Christley, Ph.D., Computational Biologist, O’Donnell School of Public Health; Ameer Hamza Shakur, Ph.D., Data Scientist/Machine Learning Engineer, Lyda Hill Department of Bioinformatics; Michael Holcomb, M.S., Lead Data Scientist, Lyda Hill Department of Bioinformatics; Alana Christie, M.S., Biostatistical Consultant, Simmons Cancer Center and Kidney Cancer Program; Neil Rakheja, student intern, Simmons Cancer Center; and AJ Jain, Ph.D. candidate, Biomedical Engineering.

Andrew Jamieson, Ph.D.
Andrew Jamieson, Ph.D., is Assistant Professor and Principal Investigator in the Lyda Hill Department of Bioinformatics at UT Southwestern.

Dr. Kapur holds the Jan and Bob Pickens Distinguished Professorship in Medical Science, in Memory of Jerry Knight Rymer and Annette Brannon Rymer and Mr. and Mrs. W.L. Pickens.

Dr. Brugarolas holds the Sherry Wigley Crow Cancer Research Endowed Chair in Honor of Robert Lewis Kirby, M.D.

Drs. Kapur, Brugarolas, and Cowell are members of the Simmons Cancer Center. 

The study was funded by a grant from the National Cancer Institute’s Kidney Cancer Specialized Program of Research Excellence (P50 CA196516) and an endowment from the Brock Fund for Medical Science Chair in Pathology.

About UT Southwestern Medical Center 

UT Southwestern, one of the nation’s premier academic medical centers, integrates pioneering biomedical research with exceptional clinical care and education. The institution’s faculty members have received six Nobel Prizes and include 25 members of the National Academy of Sciences, 24 members of the National Academy of Medicine, and 14 Howard Hughes Medical Institute Investigators. The full-time faculty of more than 3,200 is responsible for groundbreaking medical advances and is committed to translating science-driven research quickly to new clinical treatments. UT Southwestern physicians provide care in more than 80 specialties to more than 140,000 hospitalized patients, more than 360,000 emergency room cases, and oversee nearly 5.1 million outpatient visits a year.

]]>
https://www.utsouthwestern.edu/newsroom/articles/year-2025/july-ai-model-speeds-medical-data-extraction.html Mon, 28 Jul 2025 09:29:00 -0500