cancer Archives - Sanford Burnham Prebys
Institute News

Curebound awards two grants to Sanford Burnham Prebys scientists

AuthorGreg Calhoun
Date

February 12, 2025

The San Diego-based philanthropic organization has awarded $43 million in cancer research to date

Curebound recently announced the awarding of 17 grants in December 2024 for a total of $8.25 million in funding to advance cancer research in 2024.

Two new grants will support cancer research conducted by scientists at Sanford Burnham Prebys. Since 2014, 32 Curebound grants have supported projects that included scientists at the institute.

A workaround for a tricky target

The TP53 gene contains the blueprint for constructing a protein called tumor protein p53. This protein is considered a tumor suppressor because it helps cells grow in a controlled manner.

When cell growth goes awry, however, the TP53 gene is a common culprit as the most frequently mutated gene in cancers. While this ubiquity has placed a bullseye on the mutated tumor protein p53 for aspiring drug developers, it has proven tricky to target directly.

Brooke Emerling, PhD, director of the Cancer Metabolism and Microenvironment Program, and her collaborators have shown that the growth of cancer cells with a mutated TP53 gene is dependent on lipid enzymes called phosphatidylinositol-5-phosphate 4-kinases (PI5P4K). Emerling and her collaborators have identified compounds that break down these enzymes.

The researchers have demonstrated the ability of these compounds to target and eliminate cancer cells with a mutated TP53 gene without harming normal cells. Curebound will support the team’s ongoing efforts to work around the difficult-to-target tumor protein p53 by instead targeting PI5P4K.

Next, the group plans to optimize the compounds that break down PI5P4K to develop cancer drugs that are strong candidates for future clinical trials.

Curebound collaborators: Patrick Kearney, PhD, director of Medicinal Chemistry in the Conrad Prebys Center for Chemical Genomics, and Eric Wang, PhD, assistant professor in the Cancer Molecular Therapeutics Program.

Boosting the immune system against lung cancer

The immune system is one of the main defenses of the human body to fend off harmful pathogens and invasive cells such as cancer. Among all white blood cells, a particular cell type, called a T cell, can directly kill cancer cells and therefore plays an essential role in building anti-tumor immune responses.

Immunotherapies that boost the anti-cancer capabilities of T cells have revolutionized the way we treat cancer, especially in blood cancers such as leukemia, lymphoma and myeloma. More recently, immunotherapies are rapidly advancing to become mainstream treatments for solid cancers as well.

Currently, however, less than a third of patients with lung cancer benefit from immunotherapies. Pandurangan Vijayanand, MD, PhD, the William K. Bowes Distinguished Professor at the La Jolla Institute for Immunology, discovered that certain T cells called cytotoxic T lymphocytes have molecular features associated with a robust immune response against lung cancer tumors. His work has identified new targets for lung cancer immunotherapy.

Curebound will support Vijayanand’s collaboration with Michael Jackson, PhD, senior vice president for Drug Discovery and Development in the Conrad Prebys Center for Chemical Genomics, to use this research to identify agents to boost tumor immune responses.

The research team’s work has the potential to identify a new class of immunotherapy drugs for patients with lung cancer.

Curebound collaborator: Changlu Liu, PhD, director of Receptor Pharmacology in the Conrad Prebys Center for Chemical Genomics.

Pandurangan Vijayanand

Pandurangan Vijayanand, MD, PhD, is the William K. Bowes Distinguished Professor at the La Jolla Institute for Immunology.

Institute News

A monster, MASH

AuthorGreg Calhoun
Date

January 28, 2025

Scientists show how the advanced form of fatty liver disease has monstrous effects on liver cancer risk

Liver cancer has proven to be a tough beast to tame. Experts expected rates of the cancer to decrease following the development of the hepatitis B vaccine in the 1980s, which reduced one of the major risk factors for the disease.

Research in Taiwan showed that its universal infant hepatitis B vaccination program led to young adults experiencing a 35.9% reduction in cases of hepatocellular carcinoma (HCC), the most common liver cancer.

Despite innovation leading to the world’s first cancer-preventing vaccine, incidence of HCC has been on the rise due to a spike in fatty liver disease over recent decades. Lifestyle factors such as high-calorie diets, excessive alcohol consumption and minimal exercise — along with genetic predispositions — can lead to problematic changes in the liver, heart and kidneys.

Specifically in the liver, growing deposits of fat in the tissue can lead over time to an advanced form of fatty liver disease marked by chronic inflammation and the accumulation of thickened scar tissue, a condition known as metabolic-associated steatohepatitis (MASH). MASH significantly increases a patient’s risk of developing HCC.

Debanjan Dhar, PhD, headshot outside

Debanjan Dhar, PhD, is an associate professor in the Cancer Genome and Epigenetics Program.

In a paper published January 1, 2025, in Nature, scientists at Sanford Burnham Prebys, the University of California San Diego, Curtin University, the University of Pennsylvania and The Liver Cancer Collaborative, demonstrated that MASH damages the DNA of liver cells. The study also linked these changes to the development of liver cancer.

Peter Adams profile photo in lab

Peter Adams, PhD, is the director of the Cancer Genome and Epigenetics Program.

“DNA damage from MASH causes liver cells to stop dividing and enter a zombie-like state called senescence,” said Debanjan Dhar, PhD, associate professor in the Cancer Genome and Epigenetics Program at Sanford Burnham Prebys and coauthor on the study. “This study’s results demonstrate that some of these cells later exit senescence and are likely to become cancerous due to their accumulation of damage and mutations.”

“In the future, we can apply what we’ve learned to study potential opportunities to prevent or repair DNA damage from MASH to reduce patients’ risk of developing liver cancer,” said Peter Adams, PhD, director of the Cancer Genome and Epigenetics Program at Sanford Burnham Prebys and coauthor on the study.


Michael Karin, PhD, Distinguished Professor in the Department of Pharmacology at the University of California San Diego School of Medicine, is the senior and corresponding author on the study.   

Li Gu, PhD, a former postdoctoral fellow in the Karin lab, shares first authorship of the study with visiting scientist Yahui Zhu. 

Additional authors include:

  • Marcos Teneche and Souradipta Ganguly from Sanford Burnham Prebys
  • Shuvro Nandi, Maiya Lee, Kosuke Watari, Breanna Bareng, Masafumi Ohira, Yuxiao Liu, Sadatsugu Sakane, Mojgan Hosseini, Tatiana Kisseleva, Ludmil Alexandrov, Consuelo Sauceda and David Gonzalez from the University of California San Diego
  • Rodrigo Carlessi and Janina Tirnitz-Parker from Curtin Universit
  • The Liver Cancer Collaborative
  • M. Celeste Simon from the University of Pennsylvania
Institute News

Bile may be key to immunotherapy effectiveness in liver cancer

AuthorGreg Calhoun
Date

January 17, 2025

Understanding the crucial ingredient in bile may unlock the potential of treatments that help patients’ immune systems eliminate cancer

Hepatocellular carcinoma (HCC) is the most common liver cancer and a growing threat to public health across the globe due to the rising rate of fatty liver disease.

Liver cancer is difficult to treat as it often causes few if any symptoms early on, so it tends to be diagnosed at later, more aggressive stages. While immunotherapies that supercharge patients’ immune systems have proven effective in some cancers, this approach has had limited success in patients suffering from HCC or other forms of the disease.

Scientists are investigating the unique qualities of different tissues that may explain why the effectiveness of immunotherapy varies depending on the location of a tumor. The liver is known to have a flexible immune system capable of defending itself when necessary while not overreacting to a constant flood of foreign materials from digesting food, including metabolic byproducts from bacteria residing in the gut microbiome.

Transplant surgeons see the unique properties of the liver’s immune system firsthand when transplanted livers are typically integrated by recipients with only a low dose of immunosuppressive drugs. This ability to maintain immune tolerance, however, may reduce the ability of the liver’s immune system to find and destroy cancer cells, even when that capability is enhanced by immunotherapy.

In a paper published January 9, 2025, in Science, scientists at Sanford Burnham Prebys, the Salk Institute, the University of California San Diego, Columbia University Irving Medical Center, Memorial Sloan Kettering Cancer Center and the Geisel School of Medicine at Dartmouth, found that a critical ingredient in bile hinders the liver’s immune response against cancer.

Bile is a fluid made by the liver that assists in breaking down fats during digestion. This function is made possible by steroidal acids known as bile acids. The scientists found an increased amount of bile acids in tumor samples from patients with HCC. The team also found that genes involved in creating bile acids were being transcribed to make proteins and enzymes at an abnormally high rate in human samples and in mice genetically modified to develop liver cancer.

The authors went on to remove genes related to bile acid construction to demonstrate that mice without these blueprints developed fewer, smaller tumors. In addition, the liver’s T cells — the primary anti-tumor immune cells — were able to dig deeper into tumors and persist for longer without the immunosuppressive effects of certain bile acids.

“These findings underscore a new appreciation for the influence of bile acids on the liver’s immune system,” said Debanjan Dhar, PhD, associate professor in the Cancer Genome and Epigenetics Program at Sanford Burnham Prebys and coauthor on the study. More research is needed to test the potential use of drugs to directly inhibit certain bile acids or bile acid receptors as a therapeutic strategy to reduce liver cancer growth.

Debanjan Dhar, PhD, headshot outside

Debanjan Dhar, PhD, is an associate professor in the Cancer Genome and Epigenetics Program at Sanford Burnham Prebys.

Peter Adams profile photo in lab

Peter Adams, PhD, is the director of the Cancer Genome and Epigenetics Program at Sanford Burnham Prebys.

It may also be possible to achieve this effect through dietary changes that alter the microbiome and result in modified bile acid production. Based on their findings, the research team suggests that this could be done by using ursodeoxycholic acid, a bile acid that currently is used to treat an autoimmune condition called primary biliary cholangitis. The acid is found at high levels in bear bile, which has served for thousands of years as a treatment in traditional Chinese medicine.

“Given the safety profile of ursodeoxycholic acid and the limited effectiveness of immunotherapy on liver cancer, this study shows significant potential for testing this bile acid as a combination treatment for patients with HCC,” said Peter Adams, PhD, director of the Cancer Genome and Epigenetics Program at Sanford Burnham Prebys and coauthor on the study.


Susan Kaech, PhD, NOMIS Chair, professor and director of the NOMIS Center for Immunobiology and Microbial Pathogenesis at the Salk Institute is the senior and corresponding author on the study.   

Siva Karthik Varanasi, PhD, assistant professor at the UMass Chan Medical School and a former postdoctoral fellow in the Kaech lab at the Salk Institute, is first author on the manuscript. 

Additional authors include:

  • Souradipta Ganguly, Marcos G. Teneche and Aaron Havas, from Sanford Burnham Prebys
  • Dan Chen, Melissa A. Johnson, Kathryn Lande, Michael A. LaPorta, Filipe Araujo Hoffmann, Thomas H. Mann, Eduardo Casillas, Kailash C. Mangalhara, Varsha Mathew, Ming Sun, Yagmur Farsakoglu, Timothy Chen, Bianca Parisi, Shaunak Deota, H. Kay Chung, Satchidananda Panda, April E. Williams and Gerald S. Shadel, from the Salk Institute
  • Yingluo Liu, Cayla M. Miller, Jin Lee and Gen-Sheng Feng, from the University of California San Diego
  • Isaac J. Jensen and Donna L. Farber, from Columbia University Irving Medical Center
  • Andrea Schietinger from Memorial Sloan Kettering Cancer Center
  • Mark S. Sundrud from the Geisel School of Medicine at Dartmouth

Wolfram Goessling, MD, PhD, the Robert H. Ebert Associate Professor of Medicine and associate professor of Health Sciences and Technology at Harvard Medical School, authored a Perspective article on the new study in Science called, “Ena-bile-ing liver cancer growth.”

Institute News

AI-driven cancer prediction tool makes NIH director’s highlights for 2024

AuthorScott LaFee
Date

January 3, 2025

On April 18, 2024, first author Sanju Sinha, PhD, an assistant professor in the Cancer Molecular Therapeutics Program at Sanford Burnham Prebys, and colleagues published details about a new artificial intelligence-powered tool called PERCEPTION (PERsonalized Single-Cell Expression-Based Planning for Treatments In ONcology).

PERCEPTION was proof-of-concept that AI could be used to predict a cancer’s treatment responses from bulk RNA. Sinha and colleagues built AI models for 44 drugs approved by the FDA and found that their tool “predicted the success of targeted treatments against cell lines with a high degree of accuracy.”

The paper was among six specifically highlighted by Monica Bertagnolli, MD, in her blog as director of the National Institutes of Health.

Institute News

Protein superfamily crucial to the immune system experiences Broadway-style revival

AuthorGreg Calhoun
Date

November 19, 2024

More than 25 years after targeting a member of this superfamily of proteins led to groundbreaking treatments for several autoimmune diseases including rheumatoid arthritis and Crohn’s disease, San Diego scientists note a resurgence of interest in research to find related new drug candidates.

In 1998, the same year “Footloose” debuted on Broadway, REMICADE® (infliximab) was approved by the FDA for the treatment of Crohn’s disease. This was the first monoclonal antibody ever used to treat a chronic condition, and it upended the treatment of Crohn’s disease.

Research published in February 2024 demonstrated better outcomes for patients receiving infliximab or similar drugs right after diagnosis rather than in a “step up” fashion after trying other more conservative treatments such as steroids.

Infliximab and ENBREL® (etanercept) — also approved in 1998 to treat rheumatoid arthritis — were the first FDA-approved tumor necrosis factor-α (TNF) inhibitors. TNF is part of a large family of signaling proteins known to play a key role in developing and coordinating the immune system.

The early success of infliximab and etanercept generated excitement among researchers and within the pharmaceutical industry at the possibility of targeting other members of this protein family. They were interested in finding new protein-based (biologics) drugs to alter inflammation that underlies the destructive processes in autoimmune diseases.

As “Footloose” made it back to Broadway in 2024 for the first time since its initial run, therapies targeting the TNF family are in the midst of their own revival. Carl Ware, PhD, a professor in the Immunity and Pathogenesis Program at Sanford Burnham Prebys, and collaborators at the La Jolla Institute for Immunology and biotechnology company Inhibrx, report in Nature Reviews Drug Discovery that there is a resurgence of interest and investment in these potential treatments.

“Many of these signaling proteins or their associated receptors are now under clinical investigation,” said Ware. “This includes testing the ability to target them to treat autoimmune and inflammatory diseases, as well as cancer.”

Today, there are seven FDA-approved biologics that target TNF family members to treat autoimmune and inflammatory diseases. There also are three biologics and two chimeric antigen receptor (CAR)-T cell-based therapies targeting TNF members for the treatment of cancer. This number is poised to grow as Ware and his colleagues report on the progress of research and many clinical trials to test new drugs in this field and repurpose currently approved drugs for additional diseases.

“The anticipation levels are high as we await the results of the clinical trials of these first-, second- and — in some cases — third-generation biologics,” said Ware.

Ware and his coauthors also weighed in on the challenges that exist as scientists and drug companies develop therapies targeting the TNF family of proteins, as well as opportunities presented by improvements in technology, computational analysis and clinical trial design.

Portrait of Carl Ware

Carl Ware, PhD, is a professor in the Immunity and Pathogenesis Program at Sanford Burnham Prebys.

“There are still many hurdles to get over before we truly realize the potential of these drugs,” noted Ware. “This includes the creation of more complex biologics that can engage several different proteins simultaneously, and the identification of patient subpopulations whose disease is more likely to depend on the respective proteins being targeted.

“It will be important for researchers to use computational analysis of genetics, biomarkers and phenotypic traits, as well as animal models that mimic these variables. This approach will likely lead to a better understanding of disease mechanisms for different subtypes of autoimmune conditions, inflammatory diseases, and cancer, enabling us to design better clinical trials where teams can identify the appropriate patients for each drug.”

Institute News

The implastic nature of plastic culture

AuthorScott LaFee
Date

November 4, 2024

There is an art (and science) to creating cell culture models that reflect the complexities of disease. Such models have long been indispensable to parsing the underlying mechanisms of pathology and to preclinical drug discovery.

But art, writes Kevin Tharp, PhD, assistant professor in the Cancer Metabolism and Microenvironment Program, doesn’t always imitate life — at least not when it comes to finding effective cancer therapeutics.

“Just like a machine-learning algorithm trained on irrelevant datasets, efforts to discover anticancer therapeutics are limited by the models we use,” Tharp writes in the British Journal of Pharmacology. “Our drug discovery pipeline works incredibly well but is applied to models that poorly recapitulate in vivo physiology. This may be why drug discovery approaches efficiently identify drugs that work in the context tested and yet often fail to translate into clinical success.”

It’s a case of there’s no place like home. Cancer cell models are cultured on plastic in two-dimensions with limited or no diversity of neighbors. Cancer cells in vivo reside in three dimensions, with dynamic and complex interactions with neighboring cells and surroundings, i.e., the tumor microenvironment.

It’s like growing up on Disneyland’s Main Street versus a real-world urban city. Cultured cancer cells simply don’t look or behave exactly the same as cancer cells in an actual  tumor. Nor do the investigational molecules being tested as potential therapies.

Tharp suggests a multi-pronged approach: Initially culture target cells using conventional methods, then transfer the cells to new culture formats that enforce distinct, non-genomic cytoskeleton architectures and expression patterns that more closed mimic real life.

Institute News

Raising awareness of breast cancer research at Sanford Burnham Prebys

AuthorGreg Calhoun
Date

October 31, 2024

The October Science Connect Series event was themed around Breast Cancer Awareness Month and featured two cancer research experts.

The Sanford Burnham Prebys Wellness Ambassadors hosted a Science Connect event on Wednesday, October 30, 2024, featuring two faculty experts discussing their breast cancer research and its implications.

The Science Connect Series provides a forum for Sanford Burnham Prebys principal investigators to share their research with administrative personnel. Faculty members gain experience in communicating their science to a lay audience, and administrators gain a better understanding of research conducted at the institute so they can become better advocates and ambassadors of the shared mission to translate science into health.

Kelly Kersten, PhD, an assistant professor in the Cancer Metabolism and Microenvironment Program, opened the event by focusing on the importance of finding new treatments —such as immunotherapies — for the one-third of breast cancer patients that are diagnosed after the early stages of the disease when surgery is less effective.

The immune system is one of the main defenses of the human body to fend off harmful pathogens and invasive cells, such as cancer. Among all white blood cells, a particular cell type, called a T cell, can directly kill cancer cells and therefore plays an essential role in building anti-tumor immune responses.

Many types of cancer are confronted and infiltrated by T cells, only to be suppressed by the local tumor environment.

“While immunotherapies that boost the immune system have revolutionized the way we treat cancer, many patients do not respond to the treatments, and the mechanisms of resistance remain largely unclear,” said Kersten.

Kersten’s goal is to understand why T cells enter a state known as exhaustion and lose their tumor-killing capacity. This knowledge will help her team find potential future therapies that could prevent T-cell exhaustion and improve immunotherapies for cancer patients.

Kevin Tharp, PhD, also an assistant professor in the Cancer Metabolism and Microenvironment Program, shared that his lab’s focus is on how cancer cells adapt their metabolism to generate the energy needed to spread to other tissues through metastasis. He presented his team’s work with the Kersten lab on another aspect of potential resistance to immunotherapy in breast cancer.

Tharp and Kersten are studying the hypothesis that part of the reason why these therapies fail is due to tumor-associated fibrosis, the creation of a thick layer of fibrous collagen (like scar tissue) that acts as a barrier against the anti-tumor immune response. They published a paper on June 3, 2024, in Nature Cancer,  discussing how tumor-associated macrophages, a type of immune cell found abundantly in the tumor microenvironment, respond to the physical properties of fibrosis.

By synthesizing injury-associated collagens that facilitate wound closure, TAMs experience metabolic changes and generate metabolic byproducts that suppress the anti-tumor function of immune cells.

“The metabolic changes in the microenvironment present more of a challenge to anti-tumor responses than the physical barrier,” said Tharp. “Our study provides an alternative explanation for why anti-tumor immunity is impaired in fibrotic solid tumors.”

To follow up on these results, Tharp is collaborating with Sarah Blair, MD, a professor of surgery at the University of California San Diego, to fund and initiate a clinical trial testing the potential of dietary supplements to counteract the suppressive effects of TAM metabolic byproducts as an adjunct therapy to surgery.

Institute News

A Conversation About Aging and Cancer at Sanford Burnham Prebys 

AuthorGreg Calhoun
Date

October 24, 2024

Event recording now available for panel discussion with scientists held on October 9, 2024

David A. Brenner, MD, president and CEO of Sanford Burnham Prebys, welcomed attendees to the launch of a new community engagement program called “A Conversation About” in the institute’s Victor E. LaFave III Memorial Auditorium on October 9, 2024.

The initial panel discussion in the A Conversation About series focused on the connection between aging and cancer and included information about a current breast cancer research collaboration. A recording of the event is available online.

Reena Horowitz, the founder of Group of 12 and Friends at Sanford Burnham Prebys, provided introductory remarks. Brooke Emerling, PhD, director of the Cancer Metabolism and Microenvironment Program, moderated the discussion among three featured panelists:

  • Peter Adams, PhD, director of the Cancer Genome and Epigenetics Program, Sanford Burnham Prebys
  • Xiao Tian, PhD, assistant professor in the Degenerative Diseases Program, Sanford Burnham Prebys
  • Kay Yeung, MD, PhD, associate clinical professor in the Division of Hematology-Oncology, University of California San Diego Health

By bringing together community collaborators and clinicians with Sanford Burnham Prebys researchers, A Conversation About offers a unique perspective on how clinical research and practice can be used to inform fundamental and translational science.

Watch Event Recording

Institute News

Mammalian Genome Engineering Group holds 2024 symposium in San Diego

AuthorGreg Calhoun
Date

September 25, 2024

The four-day event included talks from experts from across North America and opportunities to discuss improving experimental methods and approaches to analyzing the resulting data.

Researchers convened at Sanford Burnham Prebys in La Jolla from September 12-15 to hear presentations from their peers and confer about the latest developments in modifying the genomes of mammalian animal models to advance biomedical research.

Anindya Bagchi, PhD, associate professor in the Institute’s Cancer Genome and Epigenetics Program, planned the 4th Mammalian Genome Engineering Symposium, which included 26 presentations from experts across the United States and Canada. Attendees asked many questions throughout, and numerous speakers commented on how valuable the conversation at the meeting was for refining planned experiments and considering new ideas and approaches.

“It was a truly enjoyable and thought-provoking meeting,” said Angela Liou, MD, an instructor in the Cancer Genome and Epigenetics Program at Sanford Burnham Prebys and pediatric oncologist and hematologist at Rady Children’s Hospital-San Diego. “It also was incredibly helpful in informing the next steps of my research project.”

“I’m so grateful for the invitation to attend this symposium,” said Praveen Raju, MD, PhD, the Nathan Gordon Chair in Neuro-Oncology and medical director of the Pediatric Neuro-Oncology Program at Rady Children’s Hospital-San Diego and director of the Pediatric Neuro-Oncology Program at the University of California San Diego School of Medicine.

Anindya Bagchi, PhD, headshot

Anindya Bagchi, PhD, is an associate professor in the Cancer Genome and Epigenetics Program.

“The presenters and attendees were welcoming and collaborative, and I certainly learned a lot.”

The symposium brings together the Mammalian Genome Engineering Group, which was formed by a small group of genome engineering enthusiasts including Bagchi, Nada Jabado, MD, PhD, professor of Pediatrics and Human Genetics at McGill University and a hematologist and oncologist at Montreal Children’s Hospital; David Largaespada, PhD, a professor of Pediatrics, Genetics, Cell Biology and Development at the University of Minnesota Medical School and the associate director for Basic Research in the Masonic Cancer Center; and Michael Taylor, MD, PhD, The Cyvia and Melvyn Wolff Chair of Pediatric Neuro-Oncology at Texas Children’s Cancer and Hematology Center and professor of Pediatrics (Hematology-Oncology) at Baylor College of Medicine.

The group is interested in developing functional models of genomic and epigenetic mutations associated with human diseases—especially cancers—that are difficult to recreate in animal models. The group’s first symposium was coordinated by Taylor in Napa, Calif., in 2014, followed by the 2nd symposium that was organized by Jabado in Montreal in 2015. After a hiatus, the group was revived in 2023 with the 3rd symposium hosted again by Taylor in Houston.

“We believe this symposium will, in the coming years, become a leading forum for discussing cutting-edge genomic and epigenomic approaches to tackle challenging genetic and epigenetic mutations,” said Bagchi. “These approaches are likely to become standard practice in the near future.”

The Sanford Burnham Prebys scientists that presented at the 4th Mammalian Genome Engineering Symposium were:

  • Bagchi, “Why are MYC-driven cancers so lethal?” 
  • Liou, “Investigating the deposition of H3.3K27M oncohistone and its effect on retrotransposon reactivation in H3K27M pediatric diffuse midline glioma” 
  • Ani Deshpande, PhD, associate professor in the Cancer Genome and Epigenetics Program and associate director of Diversity, Equity and Inclusion in the NCI-Designated Cancer Center, “Functional genomic approaches to identify selective dependencies in synovial sarcoma” 
  • Peter D. Adams, PhD, the director of the Cancer Genome and Epigenetics Program, “The role of aging in cancer” 
  • Lukas Chavez, PhD, associate professor in the Cancer Genome and Epigenetics Program, “Circular extrachromosomal DNA promotes tumor heterogeneity and enhancer rewiring” 
  • Jerold Chun, MD, PhD, professor in the Degenerative Diseases Program, “Genetic mosaicism and somatic gene recombination in the brain” 
  • Adarsh Rajesh, graduate student, Sanford Burnham Prebys, “CCND1-CDK6 complex inhibits DNA damage repair and promotes inflammation in senescence and the aged liver”

Additional speakers included:

  • Taylor, “Why does medulloblastoma love to be tetraploid and other nonsense”
  • Jabado, “Co-opting 3D structures to fuel tumorigenesis”
  • Tannishtha Reya, PhD, Herbert and Florence Irving Professor of Basic Science Research in Physiology and Cellular Biophysics, Columbia University, “New genetically engineered models to understand cancer heterogeneity and therapy resistance in pancreatic cancer”
  • Simona Dalin, PhD, postdoctoral fellow, Broad Institute of the Massachusetts Institute of Technology and Harvard University, “Contributions of perfect and imperfect homology to rearrangement formation in human and cancer genomes”
  • Alison M. Taylor, PhD, assistant professor of Pathology and Cell Biology, Columbia University, “Functional and computational approaches to uncover the consequences of chromosome arm aneuploidy in cancer”
  • Sean Eagan, PhD, senior scientist in the Cell Biology program at The Hospital for Sick Children, professor of Molecular Genetics, University of Toronto, “An update on Genetic analysis of 16q-syntenic block deletion in the mouse mammary gland – a tumor suppressor arm”
  • Claudia Kleinman, PhD, associate professor of Human Genetics, McGill University, investigator at the Lady Davis Institute for Medical Research, Jewish General Hospital, “Lineage programs and the 3D genome in pediatric brain tumors”
  • Branden Moriarity, PhD, associate professor of Pediatrics (Hematology and Oncology), University of Minnesota Medical School, “Next generation engineered immune effector cells for immunotherapy”
  • Beau Webber, PhD, associate professor of Pediatrics (Hematology and Oncology), University of Minnesota Medical School, “Building cancer in a dish: Sarcoma modeling using human pluripotent stem cells”
  • Sameer Agnihotri, PhD, associate professor of Neurological Surgery and director of the Brain Tumor Biology and Therapy Lab, University of Pittsburgh Medical Center Children’s Hospital of Pittsburgh, “Identifying genetic vulnerabilities by modeling Chromosome 9p loss”
  • Largaespada, “Loss of the polycomb repressor complex 2 (PRC2) alters the super-enhancer landscape, genome/epigenome stability, and therapeutic sensitivities of malignant peripheral nerve sheath tumors”
  • Teresa Davoli, PhD, assistant professor of Biochemistry and Molecular Pharmacology, New York University Langone Health, “Engineering chromosome specific aneuploidy by targeting human centromeres”
  • Rameen Beroukhim, MD, PhD, associate professor of Medicine, Dana-Farber Cancer Institute and Harvard Medical School, associate member of the Broad Institute of MIT and Harvard, “Detecting rearrangement signatures—naturally”
  • Quang Trinh, PhD, scientist, Ontario Institute for Cancer Research, “Perspectives and Challenges in PFA Integrative Analysis”
  • Taylor Gatesman, graduate student, University of Pittsburgh, “Genome Engineering: DREaming of and vCREating new models”
  • Joseph Skeate, PhD, postdoctoral fellow, University of Minnesota Medical School, “Targeted CAR integration and multiplex base editing in a single-step manufacturing process for enhanced cancer immunotherapies”
Institute News

Using machines to personalize patient care

AuthorGreg Calhoun
Date

July 30, 2024

Artificial intelligence (AI) and other computational techniques are aiding scientists and physicians in their quest to create treatments for individuals rather than populations

The Human Genome Project captured the public’s imagination with its global quest to better understand the genetic blueprint stored on the DNA within our cells. The project succeeded in delivering the first-ever sequence of the human genome while foreshadowing a future for medicine once considered to be science fiction. The project presaged the possibility that health care could be personalized based on clues within a patient’s unique genetic code.

Chavez lab

The Chavez Lab

While many more people have undergone genetic testing through consumer genealogy and health services such as 23andMe and Ancestry than through health care systems, genomic sequencing has influenced clinical care in some specialties. Personalized medicine—also known as precision medicine or genomic medicine—has been especially helpful for people suffering from rare diseases that historically have been difficult to diagnose and treat.

Scientists at Sanford Burnham Prebys are employing new technologies and expertise to test ways to improve diagnoses and customize treatments for many diseases based on unique characteristics within tumors, blood samples and other biopsies.

AI and other computational techniques are enabling patient samples to be rapidly analyzed and compared to data from vast numbers of individuals who have been treated for the same condition. Physicians can use AI and other tools to identify subtypes of cancers and other conditions, as well as improve selection of eligible candidates for clinical trials.

“I think we’ve gotten a lot better at precision diagnostics,” says Lukas Chavez, PhD, an assistant professor in the Cancer Genome and Epigenetics Program at Sanford Burnham Prebys. “In my work at Rady Children’s Hospital in cancer, we can characterize a tumor based on mutations, including predicting how quickly different tumors will spread. What we too often lack, however, are better treatment approaches or medicines. That will be the next generation of precision medicine.”

Sanju Sinha, PhD, an assistant professor in the Cancer Molecular Therapeutics Program at Sanford Burnham Prebys, is developing projects to help bridge the gap between precision diagnostics and treatment. He is partnering with the National Cancer Institute on a first-of-its-kind computational tool to systematically predict patient response to cancer drugs at single-cell resolution.

A study published in the journal  Nature Cancer discussed how the tool, called PERCEPTION, was successfully validated by predicting the response to individual therapies and combination treatments in three independent published clinical trials for multiple myeloma, breast and lung cancer.

Lukas Chavez, PhD

Lukas Chavez, PhD, is an assistant professor in the Cancer Genome and Epigenetics Program at Sanford Burnham Prebys.

In each case, PERCEPTION correctly stratified patients into responder and non-responder categories. In lung cancer, it even captured the development of drug resistance as the disease progressed, a notable discovery with great potential.

Sanju Sinha, PhD

Sanju Sinha, PhD, is an assistant professor in the Cancer Molecular
Therapeutics Program at Sanford Burnham Prebys.

“The ability to monitor the emergence of resistance is the most exciting part for me,” says Sinha. “It has the potential to allow us to adapt to the evolution of cancer cells and even modify our treatment strategy.”

While PERCEPTION is not yet ready for clinics, Sinha hopes that widespread adoption of this technology will generate more data, which can be used to further develop and refine the technology for use by health care providers.

In another project, Sinha is focused on patients being treated for potential cancers that may never progress into dangerous conditions warranting treatment and its accompanying side effects.

“Many women who are diagnosed with precancerous changes in the breast seek early treatment,” says Sinha. “Most precancerous cells never lead to cancer, so it may be that as many as eight of 10 women with this diagnosis are being overtreated, which is a huge issue.”

To try and counter this phenomenon, Sinha is training AI models on images of biopsied samples in conjunction with multi-omics sequencing data. His team’s goal is to develop a tool capable of predicting which patients’ cancers would progress based on the imaged samples alone.

“In the field of precancer, insurance does not cover the cost of computing this omics data,” says Sinha. “Health care systems do routinely generate histopathological slides from patient biopsies, so we feel that a tool leveraging these images could be a scalable and accessible solution.”

If Sinha’s team is successful, an AI tool integrated into clinics would predict whether precancerous cells would progress within the next 10 years to guide treatment decisions and how patients are monitored.

“With precision medicine, our hope is not to just treat patients with better drugs, but also to make sure that patients are not unnecessarily treated and made to bear needless costs and side effects that disrupt their quality of life.”


Programming in a Petri Dish, an 8-part series

How artificial intelligence, machine learning and emerging computational technologies are changing biomedical research and the future of health care

  • Part 1 – Using machines to personalize patient care. Artificial intelligence and other computational techniques are aiding scientists and physicians in their quest to prescribe or create treatments for individuals rather than populations.
  • Part 2 – Objective omics. Although the hypothesis is a core concept in science, unbiased omics methods may reduce attachments to incorrect hypotheses that can reduce impartiality and slow progress.
  • Part 3 – Coding clinic. Rapidly evolving computational tools may unlock vast archives of untapped clinical information—and help solve complex challenges confronting health care providers.
  • Part 4 – Scripting their own futures. At Sanford Burnham Prebys Graduate School of Biomedical Sciences, students embrace computational methods to enhance their research careers.
  • Part 5 – Dodging AI and computational biology dangers. Sanford Burnham Prebys scientists say that understanding the potential pitfalls of using AI and other computational tools to guide biomedical research helps maximize benefits while minimizing concerns.
  • Part 6 – Mapping the human body to better treat disease. Scientists synthesize supersized sets of biological and clinical data to make discoveries and find promising treatments.
  • Part 7 – Simulating science or science fiction? By harnessing artificial intelligence and modern computing, scientists are simulating more complex biological, clinical and public health phenomena to accelerate discovery.
  • Part 8 – Acceleration by automation. Increases in the scale and pace of research and drug discovery are being made possible by robotic automation of time-consuming tasks that must be repeated with exhaustive exactness.