NIH funding Archives - Sanford Burnham Prebys
Institute News

Acceleration by automation

AuthorGreg Calhoun
Date

September 5, 2024

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.

Humans have long been fascinated by automata, objects that can or appear to move and act of their own volition. From the golems of Jewish folklore to Pinocchio and Frankenstein’s Creature—among the subjects of many other tales—storytellers have long explored the potential consequences of creating beings that range from obedient robots to sentient saboteurs.

While the power of our imagination preceded the available technology for such feats of automation, many scientists and engineers throughout history succeeded in creating automata that were as amusing as they were examples of technical mastery. Three doll automata made by inventor Pierre Jaquet-Droz traveled around the world to delight kings and emperors by writing, drawing and playing music, and they now fascinate visitors to the Musée d’Art et d’Histoire of Neuchâtel, Switzerland.

While these more whimsical machinations can be found in collections from the House on the Rock in Spring Green, Wis., to the Hermitage Museum in Saint Petersburg, Russia, applications in certain forms of labor have made it so more modern automation is located in factories and workshops. There is no comparing the level of automation at research institutions to that of many manufacturing facilities more than 110 years since the introduction of the assembly line, nor should there be given the differing aims. However, the mechanization of certain tasks in the scientific process has been critical to increasing the accessibility of the latest biomedical research techniques and making current drug discovery methods possible.

researcher at work in Prebys Center

As a premier drug discovery center, the Conrad Prebys Center for Chemical Genomics team is well-versed in using automation to enable the testing of hundreds of thousands of chemicals to find new potential medicines.

“Genomic sequencing has become a very important procedure for experiments in many labs,” says Ian Pass, PhD, director of High-Throughput Screening at the Conrad Prebys Center for Chemical Genomics (Prebys Center) at Sanford Burnham Prebys. “Looking back just 20-30 years, the first sequenced human genome required the building of a robust international infrastructure and more than 12 years of active research. Now, with how we’ve refined and automated the process, I could probably have my genome sampled and sequenced in an afternoon.”

While many tasks in academic research labs require hands-on manipulation of pipettes, petri dishes, chemical reagents and other tools of the trade, automation has been a major factor enabling omics and other methods that process and sequence hundreds or thousands of samples to capture incredible amounts of information in a single experiment. Many of these sophisticated experiments would be simply too labor-intensive and expensive to conduct by hand.

Where some of the automation of yore would play a tune, enact a puppet show or tell a vague fortune upon inserting a coin, scientists now prepare samples for instruments equipped with advanced robotics, precise fluid handling technologies, cameras and integrated data analysis capabilities. Automation in liquid handling has enabled one of the biggest steps forward as it allows tests to be miniaturized. This not only results in major cost savings, but also it allows experiments to have many replicas, generating very high-quality, reliable data. These characteristics in data are a critical underpinning for ensuring the integrity of the scientific community’s findings and maintaining the public’s trust.

Ian Pass headshot

Ian Pass, PhD, is the director of High-Throughput Screening at the Conrad Prebys Center for Chemical Genomics.

“At their simplest, many robotic platforms amount to one or more arms that have a grip that can be programmed to move objects around,” explains Pass. “If a task needs to be repeated just a few times, then it probably isn’t worth the effort to deploy a robot. But, once that step needs to be repeated thousands of times at precise intervals, and handled the exact same way each time, then miniaturization and automation are the answers.”

As a premier drug discovery center, the Prebys Center team is well-versed in using automation to enable the testing of hundreds of thousands of chemicals to find new potential medicines. The center installed its first robotics platform, affectionately called “big yellow,” in the late 2000s to enable what is known as ultra-high-throughput screening (uHTS). Between 2009 and 2014, this robot was the workhorse for completing over 100 uHTS of a large chemical library. It generated tens of millions of data points as part of an initiative funded by the National Institutes of Health (NIH) called the Molecular Libraries Program that involved more than 50 research institutions across the US. The output of the program was the identification of hundreds of chemical probes that have been used to accelerate drug discovery and launch the field of chemical biology.

“Without automation, we simply couldn’t have done this,” says Pass. “If we were doing it manually, one experiment at a time, we’d still be on the first screen.”

Over the past 10 years the Center has shifted focus from discovering chemical probes to discovering drugs. Fortunately, much of the process is the same, but the scale of the experiments is even bigger, with screens of over 750,000 chemicals. To screen such large libraries, highly miniaturized arrays are used in which 1536 tests are conducted in parallel. Experiments are miniaturized to such an extent that hand pipetting is not possible and acoustic dispensing (i.e. sound waves) are used to precisely move the tiny amounts of liquid in a touchless, tipless automated process. In this way, more than 250,000 tests can be accomplished in a single day, allowing chemicals that bind to the drug target to be efficiently identified. Once the Prebys Center team identifies compounds that bind, these prototype drugs are then improved by the medicinal chemistry team, ultimately generating drugs with properties suitable for advancing to phase I clinical trials in humans.

Within the last year, the Prebys Center has retired “big yellow” and replaced it with three acoustic dispensing enabled uHTS robotic systems using 1536 well high-density arrays that can run fully independently.

“We used to use big yellow for just uHTP library screening, but now, with the new line up of robots, we use them for everything in the lab we can,” notes Pass. “It has really changed how we use automation to support and accelerate our science. Having multiple systems allows us to run simultaneous experiments and avoid scheduling conflicts. It also allows us to stay operational if one of the systems requires maintenance.”

One of the many drug discovery projects at the Prebys Center focuses on the national epidemic of opioid addiction. In 2021, fentanyl and other synthetic opioids accounted for nearly 71,000 of 107,000 fatal drug overdoses in the U.S. By comparison, in 1999 drug-involved overdose deaths totaled less than 20,000 among all ages and genders.

Like other addictive substances, opioids are intimately related to the brain’s dopamine-based reward system. Dopamine is a neurotransmitter that serves critical roles in memory, movement, mood and attention. Michael Jackson, PhD, senior vice president of Drug Discovery and Development at the Prebys Center and co-principal investigator Lawrence Barak, MD, PhD, at Duke University, have been developing a completely new class of drugs that works by targeting a receptor on neurons called neurotensin 1 receptor or NTSR1, that regulates dopamine release.

The researchers received a $6.3 million award from NIH and the National Institute on Drug Abuse (NIDA) in 2023 to advance their addiction drug candidate, called SBI-810, to the clinic. SBI-810 is an improved version of SBI-533, which previously had been shown to modulate NTSR1 signaling and demonstrated robust efficacy in mouse models of addiction without adverse side effects.

Michael Jackson profile photo

Michael Jackson, PhD, is the senior vice president of Drug Discovery and Development at the Conrad Prebys Center for Chemical Genomics.

Prebys Center researchers at work

The funding from the NIH and NIDA will be used to complete preclinical studies and initiate a Phase 1 clinical trial to evaluate safety in humans.

“The novel mechanism of action and broad efficacy of SBI-810 in preclinical models hold the promise of a truly new, first-in-class treatment for patients affected by addictive behaviors,” says Jackson.


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.
Institute News

Caroline Kumsta awarded $2.9M to study how short-term stress improves health and life expectancy

AuthorSusan Gammon
Date

July 11, 2024

By learning how small amounts of stress activate autophagy, researchers may create new approaches to combat age-related disease

Assistant Professor Caroline Kumsta, Ph.D., has been awarded a five-year, $2.9 million grant from the National Institute on Aging (NIA), part of the National Institutes of Health (NIH). The funding will advance research to better understand how the body’s cellular recycling system (autophagy) needs to be activated to produce long-term health benefits.

“This award will enable us to take a deeper dive into the fascinating concept of hormesis, where mild, sublethal stress leads to improved health and a longer lifespan,” says Kumsta. “Our goal with this grant is to learn more about how this is regulated, which may lead to healthier aging and improved treatments for age-related conditions.”

Like many researchers, Kumsta uses C. elegans—a tiny roundworm—as a model organism to reveal important lessons about aging and autophagy. C. elegans is a powerful tool for biological research because it shares many of the same anatomic and cell functions as humans, and their short lifespan (average 17 days) enables researchers to study how genes are activated and measure the effects in just two to three weeks.

Kumsta’s lab has previously shown how brief exposure to heat shock (stress) early in life triggers autophagy, which is crucial for maintaining cellular health and function. They identified two key transcription factors, HLH-30/TFEB and HSF-1, proteins that help turn specific genes on or off, which play a significant role in regulating autophagy and are required for these long-term benefits.

“Next, we aim to pinpoint the exact timing and specific tissues where autophagy must be activated to achieve these long-term health benefits,” says Kumsta. “We will investigate how heat shock affects autophagy-related genes over time and uncover new regulators of HLH-30/TFEB.

“Our preliminary data suggest that certain autophagy genes maintain elevated transcript levels for several days post-heat shock, indicating a sustained beneficial effect. We will use cutting-edge techniques like single-cell RNA sequencing to identify these long-term transcriptional changes and determine their roles in promoting longevity and improved proteostasis,” adds Kumsta.

By understanding the precise spatiotemporal requirements for autophagy activation, Kumsta hopes to develop innovative strategies, such as heat therapy, to enhance cellular health during aging and treat age-related diseases.

The grant, awarded by the National Institute on Aging, is titled, “Hormetic regulation of Autophagy in Aging” R01 AG083373).

Institute News

Time to talk about aging research

AuthorGreg Calhoun
Date

February 29, 2024

Hundreds of scientists gather in San Diego and virtually to share knowledge on the science of aging

For scientists in San Diego and across the United States, March 6-7, 2024, is an important time to talk about developments in aging research. To kick off two scientific meetings on the subject, the NIH-funded San Diego Nathan Shock Center, a collaboration among the Salk Institute for Biological Studies, Sanford Burnham Prebys and the University of California San Diego, will host its 2024 symposium focused on the center’s primary research area, “The Heterogeneity of Aging,” on Wednesday, March 6 at the Salk Institute for Biological Studies in the Conrad T. Prebys Auditorium in La Jolla.

Just as people and organisms age at different rates, scientists have demonstrated that tissues also age at their own speeds – even some cells within tissues age at a unique pace. This phenomenon, known as heterogeneity of aging, is of great interest to researchers as it may hold clues for how to develop interventions that enable people to lead healthier lives as they age. to discuss this topic.

Caroline Kumsta, PhD, assistant professor in the Development, Aging and Regeneration Program at Sanford Burnham Prebys and associate dean of student affairs of the Institute’s Graduate School of Biomedical Sciences, will speak at the 2024 symposium about heterogeneity of aging within the process cells use to recycle or dispose of damaged DNA and other waste products. Kumsta recently coauthored a manuscript in Nature Aging that found new functions for genes involved in this waste management process, known as autophagy. Gaining a better understanding of autophagy is important as scientists have demonstrated that autophagy genes are responsible for prolonged life span in a variety of long-lived organisms. Kumsta received a pilot award from the San Diego Nathan Shock Center in 2022 to support her research on the subject.

“We’re excited to once again offer the La Jolla Aging Meeting on the next day, as we have found that many people like to attend both, and that both meetings help each other,” says Alessandra Sacco, PhD, cohost of both events, director of and professor in the Development, Aging and Regeneration Program at Sanford Burnham Prebys, and dean of the Institute’s Graduate School of Biomedical Sciences.

The 7th annual La Jolla Aging Meeting will be held on Thursday, March 7, also in Salk’s Conrad T. Prebys Auditorium. The meeting was organized by Sacco and Peter Adams, PhD, director of and professor in the Cancer Genome and Epigenetics Program at Sanford Burnham Prebys, and by Jan Karlseder, PhD, Donald and Darlene Shiley Chair, senior vice president and chief science officer at the Salk Institute. The event features mostly short talks from San Diego-based postdoctoral fellows and students researching the biology of aging. The meeting’s goal is to enable participants to meet other researchers and start new collaborations.

“The La Jolla Aging Meeting has more of a focus on early career development, so the events complement each other very well,” adds Sacco.

Three members of Sanford Burnham Prebys will be presenting at the La Jolla Aging Meeting, including Xiao Tian, PhD, who recently joined the Institute as an assistant professor in the Degenerative Diseases Program. Tian focuses on epigenomic changes and deterioration that influence age-related diseases by studying the remarkable traits of naked mole rats: They rarely get cancer. They are resistant to some types of pain. They can survive up to 18 minutes without oxygen. And compared to their rodent peers, naked mole rats age very slowly. Tian’s goal is to unravel the molecular basis of aging and develop strategies to promote a healthier, more vital lifespan.

Last year, more than 400 academics, students and trainees at every career stage gathered in person and virtually from 20 states and several countries to participate in the San Diego Nathan Shock Center “The Heterogeneity of Aging” Symposium and the La Jolla Aging Meeting.

About the San Diego Nathan Shock Center
The San Diego Nathan Shock Center (SD-NSC), led by Gerald Shadel, PhD, Audrey Geisel Chair in Biomedical Science and professor in the Molecular and Cell Biology Laboratory at the Salk Institute, was established in the fall of 2020 with the overall goal of understanding the heterogeneity of aging in order to allow development of personalized interventions to increase the number of years of healthy life. To this end, the center provides three novel scientific Research Resource Cores to develop new human cell models of aging and enable the integrated analysis of molecular, cellular and tissue heterogeneity. The SD-NSC also supports and advocates basic biology of aging research in general through the development, training and mentoring activities of a Research Development Core and robust outreach efforts. All of these activities are accomplished via a consortium of three premier research institutions on the La Jolla Research Mesa: the Salk Institute for Biological Studies, Sanford Burnham Prebys and the University of California San Diego.

Alessandra Sacco serves as director of the SD-NSC Research Development Core and Peter Adams serves as codirector of the SD-NSC Heterogeneity of Aging Core.

Institute News

Three Sanford Burnham Prebys faculty receive promotions

AuthorMiles Martin
Date

June 30, 2022

Sanford Burnham Prebys is proud to announce the promotion of three of our faculty from assistant to associate professor. 

The promoted faculty, all from the Institute’s NCI-designated Cancer Center, include Ani Deshpande, PhD, Brooke Emerling, PhD and Charles Spruck, PhD

Ani Deshpande, PhD

Deshpande studies developmental processes in stem cells that get hijacked by cancer, focusing specifically on acute myeloid leukemia, one of the most common types of blood cancer. Earlier last year, Deshpande published a study with researchers at the National Institutes of Health (NIH) revealing that CRISPR gene editing can sometimes favor cells with cancer mutations, encouraging a cautious approach when using CRISPR therapies for certain cancers

Deshpande joined the Institute in 2015. Prior to that, he held positions at Memorial Sloan Kettering Cancer Center and Harvard Medical School.

Brooke Emerling, PhD

Emerling studies the metabolism of cancer cells, specifically how certain signaling proteins can contribute to the uninhibited growth typical of tumors. Emerling recently received a $2.3 million grant from the NIH to continue her work over the next four years.

Emerling joined the faculty at Sanford Burnham Prebys in 2016. Prior to that, she held positions at Weill Cornell Medicine and Harvard Medical School.

Charles Spruck, PhD 

Spruck develops new, effective, nontoxic treatments for patients with advanced cancers. Specifically, his recent studies have focused on the potential to treat cancer with viral mimicry, which tricks the body into thinking it has a viral infection, stimulating immune responses that can help the body fight cancer and improve the effects of other treatments. 

Spruck joined the Institute in 2010. Prior to that, he held positions at the Sidney Kimmel Cancer Center and Scripps Research.

Institute News

How misplaced DNA contributes to chronic illness

AuthorMiles Martin
Date

October 28, 2021

Though DNA is essential for life, it can also wreak havoc on our bodies as we age 

DNA is one of the essential building blocks of life, giving our cells instructions for virtually everything they do, but researchers at Sanford Burnham Prebys are investigating what happens to our cells when DNA ends up in places where it shouldn’t normally be, particularly as we age.

The answer – as described in their recent review in the journal Cell—is disease-causing inflammation. And the researchers hope that targeting this rogue DNA will lead to new therapeutic strategies for a range of age-related illnesses, including cancer, diabetes, rheumatoid arthritis, cardiovascular disease and neurodegenerative disorders.

“Age is the primary risk factor for all of these diseases, but they share another risk factor – chronic inflammation,” says first author Karl Miller, PhD, a postdoctoral researcher in the lab of Peter Adams, PhD, Sanford Burnham Prebys. “We’re trying to understand the underlying processes behind this inflammation so we can potentially treat all these age-related diseases together”

Typically, cells have DNA safely sequestered in their nucleus and in the mitochondria, where the DNA can do its job without interfering with the rest of the cells’ activities. When cells detect DNA in other areas, they unleash a series of biochemical responses designed to protect the cell from invaders. This response is a component of the innate immune system, our body’s first line of defense against infection.

Scientists have known about this system for decades, but until recently it was mostly thought to respond to foreign DNA, such as during a bacterial or viral infection. However, over the last decade, researchers have discovered that pieces of our own DNA, called endogenous cytoplasmic DNA, can escape from the nucleus or mitochondria and trigger this inflammatory response in our own cells, even in the absence of infection. The resulting ‘sterile’ inflammation can accumulate over time, contributing to a range of age-related diseases in all systems of the body.

But this inflammation is not without its upsides. Cytoplasmic DNA is actually an important short-term protective strategy against cancer formation. The inflammation can alert the immune system at the first sign of cancer, preventing its formation. But over the long term, the sterile inflammation caused by cytoplasmic DNA is also thought to contribute to cancer risk. In fact, we’ve only been able to observe the damage associated with sterile inflammation because people are now living long enough to experience it. 

“Systems like this exist because they’re beneficial in youth, but as we age, they break down,” says Miller. “100 years ago, a lot more people died from infectious diseases early in life. Over time, we’ve become better and better at treating these acute infections, and we’re living much longer. It’s in this later period in life that we see chronic diseases emerging that used to be much less common.”

Miller’s review describes four different types of cytoplasmic DNA fragments, classified according to when and how they appear. Some arise from the nucleus during mistakes in cell division. Others emerge because of errors in DNA repair or replication. Some even escape from mitochondria—energy-producing parts of the cell that have their own separate DNA. Others still are of unknown origin.

“They all look similar under a microscope, and they all can cause similar effects. That’s one of the major problems in this field. The benefit of studying how the different types emerge is that it gives us more points to target for therapeutics,” says Miller. 

In the Adams Lab, Miller and his colleagues look specifically at cytoplasmic chromatin fragments, one of the four types of cytoplasmic DNA. These fragments appear in the cell when the membrane surrounding the nucleus is weakened by senescence, a cellular stress response. Senescence is also associated with aging. 

“We’ve shown how this pathway works in mice, and now we’re actually moving forward with therapeutic applications for humans by doing drug screening to find compounds that can target it,” adds Miller. 

And while there is still a lot of work left for the researchers, their progress is encouraging. Adams, senior author on the Cell review, was recently awarded a $13 million grant by the NIH to study the effects of aging, including the role of cytoplasmic DNA, on the progression of liver cancer. 

“We like to call what we’re doing here ‘increasing the healthspan’, as opposed to the lifespan,” says Miller. “We’re hoping to maximize the healthy period of people’s lives.” 

Institute News

How Sanford Burnham Prebys is helping map the brain

AuthorMiles Martin
Date

October 11, 2021

By joining forces with hundreds of researchers across the country, a team from the Chun Lab at Sanford Burnham Prebys are working to create a comprehensive map of the human brain, in the hopes of leveraging that knowledge to better treat brain disorders.

Researchers in the lab of Sanford Burnham Prebys professor Jerold Chun, MD, PhD, have helped the NIH create a cellular atlas of the motor cortex – the area of the brain responsible for movement. Their work, published recently in the journal Nature, is the flagship paper for the NIH’s BRAIN initiative, a massive multi-institution project to unravel the mysteries of the human brain.

“There are hundreds of billions of cells in the brain, and identifying and classifying all the different types of brain cells is just too big a job for any single lab,” says Chun, who is a coauthor on the study. “Similar to efforts in particle physics, hundreds of neuroscientists have now come together and it’s really exciting for us to be part of this major effort.”

The NIH Brain Research through Advancing Innovative Neurotechnologies® (BRAIN) Initiative aims to revolutionize our understanding of the human brain to more effectively diagnose, treat and prevent neurological diseases and disorders. Since its launch in 2013, the BRAIN Initiative has awarded more than 900 grants to research institutions across the country, totaling $1.8 billion. 

Chun is one of the principal investigators of the BRAIN Initiative Cell Census Network (BICCN), a subset of the Initiative that aims to develop a database of all the brain cell types in humans, mice, and non-human primates.

“While the project is about exploring the brain, what we’re really interested in over the long term is the clinical applications,” says study coauthor Carter Palmer. “Understanding the nuances of the brain and how the trillions of neural connections really work is going to lead us to new targets for therapies and diagnostics so we can help people heal.” Palmer is a graduate student in Chun’s lab, alongside fellow co-authors Christine Liu and William Romanow.

There are over 150 billion cells in the average human brain and well over a thousand different cell types, depending on how you characterize them. With such a vast landscape to track, many different types of data are needed to develop a comprehensive atlas of the brain.

For their part, the Chun Lab provided single-cell transcriptomes for human brain cells, focusing on the motor cortex. Single cell transcriptomes provide a measure of how hundreds to thousands of genes are expressed in individual cells and can provide hints as to what functions those cells are serving. This process also provides a molecular definition of cell types, making it easier for researchers to identify and classify them.

“Looking at how genes are expressed gives us a wealth of information on what cells are doing, how they develop and how they’re interacting with other cells,” says Palmer. “And when our data feed into the data from other teams, we start to get a much clearer picture of what’s happening in the brain than has ever been possible.” 

Their flagship Nature paper is one of seventeen in a special edition of the journal, chronicling recent advances by hundreds of BICCN researchers. The team also contributed to a second paper in the issue, which expands on the first by comparing the motor cortex cells of humans, mice, and marmosets. These publications speak not only to the expertise of Chun and his colleagues, but to the power of collaborative, interdisciplinary work to achieve previously unheard-of research goals.

“Fifty years ago, a project like this would have been impossible, because we just didn’t have the technology or even basic knowledge to collaborate on such a large scale,” says Chun. “Huge initiatives like BRAIN are an important part of the future of scientific research, and we’re thrilled we were able to contribute to this milestone in neuroscience.” 

Institute News

Sanford Burnham Prebys drug enters Phase 1 study for the treatment of tobacco use disorder

AuthorSusan Gammon
Date

August 26, 2021

A drug discovered in the lab of Nicholas Cosford, PhD, professor and deputy director of the NCI-designated Cancer Center at Sanford Burnham Prebys, has entered a Phase 1 clinical study.

The compound, SBP-9330, targets a neuronal signaling pathway underlying addictive behaviors and would be a first-in-class oral therapeutic to help people quit smoking. 

The study is being funded by the National Institute on Drug Abuse (NIDA) at the National Institutes of Health (NIH) through a grant awarded to Sanford Burnham Prebys, the Department of Psychiatry, University of California San Diego, School of Medicine, and Camino Pharma, LLC, who will oversee activities related to the Phase 1 study.  

“Smoking continues to be the leading cause of preventable death in the US. Nearly 70% of adult smokers try to quit smoking, but only succeed less than 30% of the time, and often relapse after quitting,” says Cosford, who is also co-founder of Camino Pharma. “It has been 15 years since the U.S. Food and Drug Administration (FDA) last approved a therapeutic for this indication. We hope that SBP-9330 ultimately becomes a viable therapeutic option for smokers to quit for good.”

As a novel selective positive allosteric modulator of the metabotropic glutamate receptor 2 (mGlu2), SBP-9330 is designed to reduce levels of glutamate, a neurotransmitter linked to addiction and relapse behavior. Preclinical studies of SBP-9330, supported by a previous NIDA grant awarded to the same three institutions, demonstrated that the drug candidate reduces nicotine self-administration in animal models and is safe and well tolerated in preclinical safety and toxicology studies.

“We are excited to initiate the first-in-human study of SBP-9330 and are grateful for the investment the NIDA has made in the treatment of tobacco use disorder,” says Gonul Velicelebi, PhD, CEO and co-founder of Camino Pharma. “In the future, we also hope to broaden the indication of SBP-9330 to other types of addiction, such as cocaine, opioid, or methamphetamine use disorders. This is supported by preclinical data in other models of substance abuse as well as the mechanism of action of SBP-9330.”

The randomized, placebo-controlled, double-blind, single-ascending and multiple-ascending dose study is being conducted at a single site in the United States under an Investigational New Drug (IND) application recently allowed by the FDA and will enroll up to 80 healthy volunteers through multiple cohorts. The goal of the study is to determine the safety, tolerability and pharmacokinetic profile of SBP-9330 in humans and to determine a safe dose range for further clinical development SBP-9330 for the treatment of people with tobacco use disorder. 

“We are excited about collaborating in the development of SBP-9330 to treat tobacco use disorder. Each year in the United States, roughly half a million people die from tobacco-related diseases. It is critical to have more therapeutic options if we want to reduce the number of deaths and illnesses related to smoking,” says Robert Anthenelli, MD, UC San Diego professor of psychiatry and one of the co-principal investigators on the NIDA project.


Dr. Cosford has an equity interest in Camino Pharma, LLC. Dr. Cosford’s relationship with Camino Pharma, LLC has been reviewed and approved by Sanford Burnham Prebys in accordance with its conflict-of-interest policies.

Institute News

Scientists discover new survival strategy for oxygen-starved pancreatic cancer cells

AuthorMonica May
Date

October 23, 2019

Oxygen is essential to life. When fast-growing tumor cells run out of oxygen, they quickly sprout new blood vessels to keep growing, a process called angiogenesis. 

By blocking pancreatic cancer’s oxygen-sensing machinery—the same field of research studied by the winners of the 2019 Nobel Prize in Medicine—Sanford Burnham Prebys scientists have uncovered a new way that tumors turn on angiogenesis in an animal model. The discovery, published in Cancer Research, could lead to a treatment that is given with an anti-angiogenetic medicine, thereby overcoming drug resistance. 

“Treatment resistance is a major challenge for cancer treatments that block blood vessel growth,” says Garth Powis, D.Phil., professor and director of Sanford Burnham Prebys’ National Cancer Institute (NCI)-designated Cancer Center and senior author of the study. “Our research identifies a new way angiogenesis is activated, opening new opportunities to find medicines that might make existing cancer treatments more effective.” 

Many cancer treatments work by blocking angiogenesis, which rarely occurs in healthy tissues. However, these medicines eventually stop working, and the cancer returns, sometimes in as little as two months. Scientists have been researching why this treatment resistance occurs so it can be stopped.

In this study, the scientists focused on pancreatic cancer, which is notoriously desperate for oxygen and also difficult to treat. Fewer than 10% of people diagnosed with pancreatic cancer are alive five years later. 

To see how a pancreatic tumor responds to a disruption in its oxygen supply, the Sanford Burnham Prebys researchers used a mouse model to block an oxygen-sensing protein called HIF1A—which should cripple the tumor’s growth. Instead of dying, however, after about a month the cells multiplied—indicating they had developed a new way to obtain oxygen. 

Further work revealed that the cancer cells were clear and swollen with the nutrient glycogen (a characteristic also seen in some ovarian and kidney cancers). In response to the excess glycogen, special immune system cells were summoned to the tumor, resulting in blood vessel formation and tumor survival. Each of these responses represents a new way scientists could stop pancreatic tumors from evolving resistance to treatment.

“Our team’s next step is to test tumor samples from people with pancreatic cancer to confirm this escape mechanism occurs in a clinical setting,” says Powis. “One day, perhaps we can create a second medicine that keeps anti-angiogenic drugs working and helps more people survive pancreatic cancer.”    

Research reported in this press release was supported by the U.S. National Institutes of Health (NIH) (5F31CA203286, CA216424 and P30CA030199). The content is solely the responsibility of the authors and does not necessarily represent the official views of the NIH. The study’s DOI is 10.1158/0008-5472.CAN-18-2994. 
 

Institute News

Sanford Burnham Prebys welcomes U.S. Congressman Mike Levin

AuthorMonica May
Date

October 22, 2019

On October 1, 2019, U.S. Representative Mike Levin (D-CA) toured Sanford Burnham Prebys and met with several faculty members to learn more about the innovative biomedical research taking place in his backyard. Levin represents California’s 49th Congressional District, which includes North County San Diego, South Orange County and neighbors our La Jolla campus. 

The visit kicked off with a visit to a lab working to find medicines for a heart arrhythmia condition called atrial fibrillation (AFib), a disorder that hits home for Levin: His grandmother struggled with the disease. Levin peered into a microscope to view beating heart cells and learned how a team of experts from Sanford Burnham Prebys and Scripps Clinic are working to develop personalized treatments for the condition, which affects nearly six million Americans (meet the A-team.)

“Sanford Burnham Prebys is a great example of the vibrant biomedical research taking place in San Diego that has the potential to improve the quality of life for families across the country,” says Levin. “Seeing the Institute’s critical research up close and hearing firsthand how National Institutes of Health (NIH) funding has accelerated medical discovery only strengthens my commitment to supporting biomedical science. Following my visit to Sanford Burnham Prebys, I was proud to introduce legislation that would invest $10 billion in the NIH to support biomedical research, and I will continue to fight for this much-needed funding.”

Following the lab tour, Levin met with faculty members who—thanks to federally funded research—are working to find treatments for Alzheimer’s disease and addiction, and study the aging process to address age-related diseases such as cancer. The visit wrapped up in the lab of Hudson Freeze, PhD, the director of our Human Genetics Program, who studies a rare childhood disease called congenital disorders of glycosylation, or CDG. 

“Americans today are living longer and healthier lives because of federally funded medical research,” says Chris Larson, PhD, the adjunct associate professor of Development, Aging and Regeneration at the Institute who arranged the visit. “We are grateful that Mike took the time to sit down with us to learn about our NIH-funded work and how he can help support us on our mission to find cures for human disease.”

Editor’s note: Shortly after his visit Levin introduced legislation that calls for a $10 billion investment in biomedical research. 

Institute News

Sanford Burnham Prebys scientist joins historic effort to help children with rare disease

AuthorMonica May
Date

October 3, 2019

Hudson Freeze, PhD, professor of Human Genetics at Sanford Burnham Prebys, has joined a historic effort that establishes—for the first time—a nationwide network of 10 regional academic centers, Sanford Burnham Prebys researchers and patient advocacy groups to address decades of unresolved questions surrounding congenital disorders of glycosylation, or CDG, a rare disease that affects children. The consortium is funded by a $5 million, five-year grant from the National Institutes of Health (NIH). 

“We are extremely pleased that the NIH is investing in an initiative that will improve the lives of people affected by CDG,” says Freeze, who leads efforts to develop and validate disease biomarkers that will aid in diagnoses, and measuring treatment benefits during clinical trials. “Although globally the number of people living with CDG is relatively small, the impact on the lives of these individuals and their families can be profound. We look forward to working with the patients, families, physicians, scientists and other stakeholders focused on this important study.”

CDG is caused by genetic mutations that disrupt how the body’s sugar chains attach to proteins. First described in the 1990s, today scientists have discovered more than 140 types of mutations that lead to CDG. Symptoms are wide-ranging, but can include developmental delays, movement problems and impaired organ function. Some children may benefit from a sugar-based therapy; however, developing treatments for those who need alternative treatment options has been hindered by a lack of natural history data—tracking the course of the condition over time—comprehensive patient registry, and reliable methods to establish the CDG type.

Working together, the consortium will overcome these hurdles by: 

  • Defining the natural history of CDG through a patient study, validating patient-reported outcomes and sharing CDG knowledge 
  • Developing and validating new biochemical diagnostic techniques and therapeutic biomarkers to use in clinical trials 
  • Evaluating whether dietary treatments restore glycosylation to improve clinical symptoms and quality of life

Freeze will lead the efforts to develop and validate biomarkers for CDG, working alongside the Children’s Hospital of Philadelphia and the Mayo Clinic. The principal investigator of the CDG Consortium Project is Eva Morava, MD, PhD, professor of Medical Genetics at the Mayo Clinic. The patient advocacy groups involved are CDG CARE and NGLY1.org. 

Sanford Burnham Prebys and CDG Care will host the 2020 Rare Disease Day Symposium and CDG Family Conference from February 28 to March 1 in San Diego, which welcomes researchers, clinicians, children with CDG and their families, and additional CDG community members. Register to attend.