binary numbers

New datasets predict surgeon performance during complications

binary numbers

In a new study published in JAMA Network Open, experts at Children’s National and allied institutions created and validated the first dataset to depict hemorrhage control for machine learning applications, the simulated outcomes following carotid artery laceration (SOCAL) video dataset.

Computer algorithms, such as machine learning and computer vision, are increasingly able to discover patterns in visual data, powering exciting new technologies worldwide. At Children’s National Hospital, physician-scientists develop and apply these advanced algorithms to make surgery safer by studying surgical video.

The big picture

In a new study published in JAMA Network Open, experts at Children’s National and allied institutions created and validated the first dataset to depict hemorrhage control for machine learning applications, the simulated outcomes following carotid artery laceration (SOCAL) video dataset.

The authors designed SOCAL to serve as a benchmark for data-science applications, including object detection, performance metric development and outcome prediction. Hemorrhage control is a high-stakes adverse event that can pose unique challenges for video analysis. With SOCAL, the authors aim to solve a valuable use case with algorithms.

“As neurosurgeons, we are often called to perform high-risk and high-impact procedures. No one is more passionate about making surgery safer,” said Daniel Donoho, M.D., neurosurgeon at Children’s National Hospital and senior author of the study. “Our team at Children’s National and the Sheikh Zayed Institute is poised to lead this exciting new field of surgical data science.”

The hold-up in the field

These algorithms require raw data for their development, but the field lacks datasets that depict surgeons managing complications.

By creating automated insights from surgical video, these tools may one day improve patient care by detecting complications before patients are harmed, facilitating surgeon development.

Why it matters

“Until very recently, surgeons have not known what may be possible with large quantities of surgical video captured each day in the operating room,” said Gabriel Zada, M.D., M.S., F.A.A.N.S., F.A.C.S., director of the Brain Tumor Center at the University of Southern California (USC) and co-author of the study. “Our team’s research led by Dr. Donoho shows the feasibility and the potential of computer vision analysis in surgical skill assessment, virtual coaching and simulation training of surgeons.”

The lack of videos of adverse events creates a dataset bias which hampers surgical data science. SOCAL was designed to meet this need. After creating a cadaveric simulator of internal carotid artery injury and training hundreds of surgeons on the model at nationwide courses, the authors then developed computational models to measure and improve performance.

“We are currently comparing our algorithms to experts, including those developed using the SOCAL dataset,” Dr. Donoho said. “Human versus machine, and our patients are ultimately the winners in the competition.”

What’s next

The authors are also building a nationwide collective of surgeons and data scientists to share data and improve algorithm performance through exciting partnerships with USC, California Institute of Technology and other institutions.

You can read the full study “Utility of the Simulated Outcomes Following Carotid Artery Laceration Video Data Set for Machine Learning Applications” in JAMA Network Open.

colorful strands of DNA

Paving the way to activate a single gene in Angelman syndrome

colorful strands of DNA

Angelman syndrome (AS) is a rare disorder that causes neurodevelopmental issues such as intellectual disability, impaired speech and motor skills, epilepsy and sleep disruptions. This single gene disorder is caused by mutations or deletions in the maternal copy of the UBE3A gene.

Angelman syndrome (AS) is a rare disorder that causes neurodevelopmental issues such as intellectual disability, impaired speech and motor skills, epilepsy and sleep disruptions. This single gene disorder is caused by mutations or deletions in the maternal copy of the UBE3A gene. To date, there is no treatment for AS.

It is easier to treat this syndrome when the disrupted gene is present but repressed. If experts can figure out how to activate it in clinical trials, they believe patients could receive a treatment that tackles the root of the problem. Children’s National Hospital experts support this vision and the AS community by helping establish appropriate biomarkers for current and future clinical trials.

While the field is trying to figure out the best scientific method to quantify progress in clinical trials for AS, the Sidorov Laboratory found that overnight sleep testing is not necessary for detecting Angelman syndrome electroencephalography (EEG) biomarkers, according to the study published in Autism Research. The data further suggests that while sleep EEGs do not provide additional benefit for detecting delta EEG rhythms, sleep itself represents a valuable AS biomarker.

What this means

“It is encouraging to see that wake EEGs are sufficient, and perhaps ideal, for detecting delta waves in a clinical trial setting,” said Michael S. Sidorov, Ph.D., principal investigator with the Center for Neuroscience Research at Children’s National. “With this biomarker, researchers can measure how AS severity changes in children over the course of a clinical trial. This enables trials to test the efficacy of exciting new treatments.”

The hold-up in the field

In the past decade, the research community has focused on activating the dormant paternal copy of the UBE3A gene in pre-clinical models. Presently, there are three ongoing phase I clinical trials for AS in the U.S. These trials use antisense oligonucleotides (ASOs), which can modify gene expression to treat genetic disorders, and have been FDA approved for other disorders. These new compounds specifically target the gene activation to unleash the existing copy of UBE3A. However, there is a need for better and more accurate ways to know if the drug is working or not. The field has not reached a consensus yet on the appropriate biomarkers that can correctly measure success.

There are also challenges associated with performing overnight EEG studies in children with AS due to the severe sleeping problems, difficulty in tolerating the process and sample recruitment.

The patient benefit

Elizabeth R. Jalazo, M.D., assistant professor of pediatrics at the University of North Carolina in Chapel Hill, chief medical officer at the Angelman Syndrome Foundation, is also the parent of a child with Angelman syndrome. Dr. Jalazo, who was not part of the study, mentioned that her experience with a daughter with a rare disorder had brought challenges to their family over the last seven years. But, alas, she said the joy Evelyn has brought to their lives far outweighs the day-to-day challenges of special needs parenting.

“As a parent I’m thrilled that we can potentially capture as much meaningful EEG data in a short daytime EEG rather than subjecting our children to overnight EEG studies,” said Dr. Jalazo. “As a clinician this is equally exciting from a clinical trial feasibility standpoint.”

One of the greatest challenges facing Angelman syndrome and other neurodevelopmental disorder therapeutic development is the lack of appropriate endpoints to assess the efficacy of our interventions.

“I worry very much that without objective measures specific to Angelman syndrome, potentially beneficial therapeutics may fail to meet the mark and ultimately not reach the community,” she added.

The scientific community has transitioned from the hope of clinical trials to lessen those day-to-day challenges to witnessing first-in-human trials of potentially transformative therapeutics in just the last few years.

“It is a biomarker work like this that is critical as we delve into the exciting landscape of clinical trial design and advance therapeutics for Angelman syndrome,” said Dr. Jalazo.

You can read the full study “Evaluation of electroencephalography biomarkers for Angelman syndrome during overnight sleep” in Autism Research.

 

Hands holding letters that spell autism

Increasing access to autism spectrum disorder services through enhanced training

Hands holding letters that spell autismMany service providers struggle to keep pace with advances in autism-specific knowledge and tend to refer children to autism specialty clinics when the diagnosis of autism spectrum disorder (ASD) is in question. Unfortunately, it is in these settings where children most often wait for months or, worse, experience barriers to accessing any care at all. This has resulted in an access crisis for children and families with ASD concerns contributing to delays in diagnosis and treatment, particularly for children of color and for under-resourced families. Service disruptions and challenges related to the COVID-19 pandemic have only added to delays. As the need for autism-related services continues to grow, innovative models must be used to enhance competence among frontline medical, behavioral health and community-based providers who currently serve these children and families on a regular basis.

Children’s National Hospital has initiated a number of endeavors focused on increasing access to ASD services through enhanced training experiences, mentorship of allied mental health and frontline professionals and utilization of multidisciplinary approaches. These approaches enhance the skills and knowledge of treatment providers, which allows them to accurately address the needs of autistic patients while they await more comprehensive evaluations and sometimes reduce the need for additional evaluation. The following are efforts currently underway.

Virtual ECHO (Extension Community Healthcare Outcomes) Autism Clinics

The Center for Autism Spectrum Disorders (CASD) is hosting virtual ECHO (Extension Community Healthcare Outcomes) Autism Clinics aimed at building autism knowledge and competencies amongst community providers by creating shared learning forums with a multidisciplinary group of autism specialists for dissemination of knowledge and mentorship.

Clinics run in 6-month sessions on a bimonthly basis and target professionals in medical, community and educational/early intervention settings. There is no requirement for prior autism-related knowledge or training. The emphasis in learning stems from case-based discussions primarily, along with targeted autism specific didactics.

We have found good satisfaction with the program overall, as well as self-report of gains in ASD-specific knowledge and care competencies as a result of participation in ECHO. To date, CASD’s ECHO Autism program has reached 290 professionals and trainees serving autistic children and their families.

Integration of autism evaluations into primary care sites

The Community Mental Health (CMH) CORE (Collaboration, Outreach, Research, Equity) within the Children’s National Hospital Child Health Advocacy Institute (CHAI) has been working collaboratively with several other divisions, including CASD, to integrate autism evaluations into primary care sites for young children with high concern about ASD. We aim to increase capacity and access to autism services by training embedded psychologists in primary care settings in autism diagnostics.

By increasing behavioral health provider capacity and integrating in primary care, this clinic has been able to drastically decrease waits for ASD services by months to years. Families served by the program were predominately Black (81%) or Latinx (10%), and most (87%) had public insurance. Nearly one third (32%) were not primary English speakers. An ASD diagnosis was provided in 68% of all cases.

All referring PCPs surveyed indicated that they were “satisfied” or “very satisfied” with the program, that they “strongly like the integrated clinic model,” and that the program “is increasing equitable access to ASD. Currently, CHAI-supported ASD-focused embedded clinics in primary care have served 94 children and their families.

illustration of brain tumor

International initiative aims to find rare brain tumor treatments

illustration of brain tumor

Rare brain tumors are not as well characterized due to the paucity of biological and clinical data available.

Certain brain tumors can be hard to diagnose. And as such, that makes it complicated to find a treatment.

In an effort to identify and tailor treatments to patients with rare brain tumors, Children’s National is launching a rare brain tumor initiative. The hospital is collaborating with other hospitals in North America, South America and Europe to compile a registry of children diagnosed with rare brain tumors. The registry will collect biospecimens, clinical and radiological data from patients diagnosed with certain rare brain tumors.

The goal is to find a correlation between the molecular findings and the clinical findings to categorize them. This will help doctors get different prognosticators or different treatment approaches.

Here, Adriana Fonseca Sheridan, M.D., pediatric neuro-oncologist at Children’s National Hospital, tells us more about this international initiative.

What’s been the hold-up in the field?

The recent incorporation of molecular features as part of the diagnostic criteria and classification of brain tumors highlighted a high biological and molecular heterogeneity within previously histologically defined entities. The improvement in our diagnostic capabilities have been incredibly useful to stratify patients into different disease-specific risk groups and tailor therapeutic approaches accordingly in the most common brain tumors. In contrast, rare brain tumors are not as well characterized due to the paucity of biological and clinical data available. Additionally, newly molecularly defined entities lack specific clinical and therapeutic data and represent a major challenge to patients and doctors alike.

How does this work move the field forward?

The overarching objective of the international rare brain tumor registry is to deepen our understanding of the biological underpinnings of rare brain tumors. The registry also seeks to create infrastructure that allows for development of rational and personalized treatment strategies for children with rare entities.

What are you hoping to discover?

We hope to characterize the clinicopathological features and identify risk factors for survival and optimal therapeutic approaches of:

  • CNS sarcomas
  • BCOR-ITD tumors
  • Astroblastoma/MN1 altered tumors
  • Histologically ambiguous/unclassifiable brain tumors

How unique is this work?

Children’s National will spearhead the development of this initiative and lead an effort to prospectively collect biological specimens, radiological and clinical data that allow us to better understand the biologic mechanisms and therapeutic susceptibilities of these rare diseases.

We know that the best way to lead the advancement of the field in rare diseases is through collaboration. Therefore, we will synchronize efforts and collaborate with our European colleagues. They will be running a similar initiative. Our goal is to generate meaningful and robust data that will allow us to better understand how to successfully treat patients with these rare brain tumors across the globe.

mitochondria

Grant funds study of two maternally inherited mitochondrial diseases

mitochondria

The National Institutes of Health awarded George Washington University and Children’s National Hospital a grant to study two maternally inherited mitochondrial diseases.

The National Institutes of Health awarded George Washington University and Children’s National Hospital a grant to study two maternally inherited mitochondrial diseases. Andrea Gropman, M.D., division chief of Neurodevelopmental Pediatrics and Neurogenetics at Children’s National, along with her co-investigator, Anne Chiaramello, M.D., from the George Washington University School of Medicine, will lead the study.

The proposed studies focus on two ultra-rare maternally inherited mitochondrial diseases:

  • Mitochondrial Encephalopathy, Lactic Acidosis and Stroke-like episodes (MELAS); and
  • Leber’s Hereditary Optic Neuropathy-Plus (LHON-Plus).

Both diseases are among those studied by the Rare Diseases Clinical Research Network.

“We are really pleased to be able to change the landscape for MELAS and LHON, two mitochondrial disorders with relentless progression and no treatment,” Dr. Gropman said. “This grant represents the fruition of an eight-year collaboration with my colleague Dr. Chiaramello and we are fortunate to be able to deliver this at Children’s National and serve our patients and community.”

Because patients currently do not have access to effective therapeutic intervention, this results in significant disability, morbidity and premature death. The UG3 phase of the study will focus on translational MELAS and LHON-Plus studies and submission of an IND protocol to the Food and Drug Administration. The UH3 phase will focus on a basket clinical trial with MELAS and LHON-Plus to:

  • Provide proof-of-concept that the basket design can be applied to divergent ultra-rare diseases.
  • Advance the dataset for safety and pharmacokinetics/pharmacodynamics of our lead compound for a larger number of patients than in a conventional clinical trial setting.
  • Gather outcomes and practical information for optimizing the design of future basket clinical trial.

“Dr. Gropman is dedicated to giving children with MELAS the very best care,” said Elizabeth Wells, M.D., vice president of Neuroscience and Behavioral Medicine Center at Children’s National. “This new research funding is exciting and means more patients can benefit from the expertise she has developed at Children’s National.”

brain network illustration

Changing the surgical evaluation of epilepsy

brain network illustrationThe choice between stereoelectroencephalography (SEEG) and subdural evaluation is not mutually exclusive, according to a new opinion piece published in JAMA Neurology.

In their article, Chima Oluigbo, M.D., pediatric epilepsy neurosurgeon, William D. Gaillard, M.D., division chief of Epilepsy and Neurophysiology and Neurology, both at Children’s National Hospital, and Mohamad Z. Koubeissi, M.D., M.A., from The George Washington University Hospital, discuss how the practicing epileptologist requires a profound understanding of the roles of different technologies. It also looks at how to integrate both traditional and emerging paradigms to optimize seizure control. This issue is particularly relevant to choosing the best method of invasive intracranial electroencephalography monitoring in individual cases.

Noting that despite the dramatic increase in SEEG use in recent years, the authors talk about how many patients still benefit from invasive monitoring using subdural grids. Therefore, it is important to define the considerations that should guide decision-making on the choice of SEEG versus subdural monitoring in each patient. The authors expand on their statement explaining that it is critical to define the roles of SEEG vs subdural grid investigation in each patient as subdural grid evaluations are still indicated in specific circumstances.

Additionally combined hybrid deployment of both techniques may be indicated in specific situations. Accommodation should be made to allow customization of the technique chosen to available technical expertise and equipment as well as patient preference.

2021 neurology infographic

2021 at a glance: Neurology and Neurosurgery at Children’s National

2021 neurology infographic

MRI Room

Children’s National uses HIFU to perform first ever non-invasive brain tumor procedure

MRI Room

Children’s National Hospital successfully performed the first-ever high-intensity focused ultrasound (HIFU) procedure on a pediatric patient with neurofibromatosis (NF). This is the youngest patient to undergo HIFU treatment in the world. Image provided by Insightec.

Children’s National Hospital successfully performed the first-ever high-intensity focused ultrasound (HIFU) procedure on a pediatric patient with neurofibromatosis (NF). This is the youngest patient to undergo HIFU treatment in the world. The advancement of children’s medical devices in the U.S. continues to significantly lag behind adult devices. This is why this milestone marks a significant advance in making pediatric surgery more precise and less invasive.

The hospital is offering this treatment to patients under an ongoing research clinical trial. Children’s National is one of the first pediatric hospitals in the nation to use HIFU for neuro-oncology patients. It’s also the first hospital in the world to use it to treat a pediatric patient with NF. NF is a condition that occurs in approximately 1 in 3,500 births and causes tumors to form in the brain, spinal cord and nerves.

“Using HIFU to treat our pediatric patients is a quantum leap towards non-invasive surgery for kids,” said Robert Keating, M.D., division chief of Neurosurgery and co-director of the HIFU program at Children’s National. “It’s exciting because the future is now here and it’s significantly better for our kids, in terms of non-invasive surgery with lower risk of complications and no exposure to radiation.”

Focused ultrasound (FUS) is a non-invasive therapeutic technology with the potential to transform the treatment of many medical disorders by using ultrasonic thermal energy to specifically target tissue deep in the body. The technology can treat without incisions or the need of radiation.

FUS, which has been used for adult clinical trials for many decades, can be delivered through high- or low-intensity focused ultrasound (LIFU). HIFU uses non-invasive therapy that uses focused ultrasound waves to thermally ablate a focal area of tissue. Children’s National will now use HIFU to treat low-grade type tumors located in difficult locations of the brain, such as hypothalamic hamartomas and pilocytic astrocytoma, as well as for movement disorders and epilepsy.

An alternative approach, LIFU uses lower levels of energy to disrupt the blood-brain barrier. Unlike medications, which often have difficulty crossing the blood-brain barrier, LIFU can transiently open the blood-brain barrier to chemotherapy. This may allow more effective treatment of tumors and offer opportunities to treat, for the first time, the entire extent of a malignant brain tumor.

“Having focused ultrasound technology as a tool and conducting clinical trials will allow our neurologists and oncologists to offer a non-invasive treatment option to many patients who suffer from neurological conditions,” said Hasan Syed, M.D., co-director of the HIFU program at Children’s National. “The milestone of performing this first HIFU procedure will lead the way to better understanding of the effect of this technology and provide patients with more options.”

At Children’s National, the HIFU program is being led by Dr. Keating and a multidisciplinary team, including clinicians and investigators from the Sheik Zayed Institute for Pediatric Innovationradiologyoncologysurgery and orthopedics. In an effort to collaborate with the region’s adult hospitals, Children’s National will also treat adult patients on a selective basis who have movement disorders such as essential tremor and Parkinson’s. There is a scarcity of similar resources in the metro region. Many adult patients face one-year wait periods for treatment of their movement disorders, requiring many to travel out of state for treatment.

The LIFU program is scheduled to be operational in 2022. It will likely be the first in the U.S. to treat high-grade pediatric brain tumors with disruption of the blood-brain barrier and provide more effective routes for chemotherapy as well as potential immunotherapy and molecular approaches.

“The use of LIFU with microbubbles to open up the blood-brain barrier is an exciting, potentially game-changing approach for children with these tumors,” said Roger Packer, M.D., senior vice president of the Center for Neurosciences and Behavioral Medicine at Children’s National. “It should safely allow the blood-brain barrier to open and allow delivery of potentially life-saving personalized therapy to the tumor and spare the rest of the brain. It is the most exciting, new development in brain tumor therapy for these malignant midline tumors in the past 50 years.”

Children’s National continues to be a leader in pediatric HIFU use. In 2015, Children’s National doctors became the first in the U.S. to use MR-HIFU to treat pediatric osteoid osteoma – a benign, but painful bone tumor. Successful clinical trial results led to FDA approval in early 2021 for the use of the technology for this treatment. In 2020, the Focused Ultrasound Foundation also designated Children’s National as the first global pediatric Center of Excellence for using this technology to help patients with specific types of childhood tumors.

boy with headache

Kids’ headaches can be disruptive. We need solutions.

Experts leading the Headache Program at Children’s National Hospital recognize how common these disorders are. They also know how disruptive they can be in the day-to-day of children.

Marc DiSabella, D.O., is the director of the program. He is currently leading five pediatric headache trials. In this Q&A, he tells us about the ongoing trials, offering insight into innovative solutions and how he’s carving a new path to improve the quality of life of his patients.

Q: How has your team advised other neurologists on innovative care for patients with headaches that have been refractory to medicines?

A: We receive referrals from outside institutions when they need additional input for diagnostic and management options. We receive patient consult requests from around the country – and sometimes out of the country – to help improve symptoms. In most instances, these headaches tend to be difficult to control and do not respond to available medications. We really try to take a holistic approach to their care, and use treatments in parallel. For example, diagnostic, lifestyle techniques, medications, pain focused cognitive behavioral therapy and physical therapy. We also use complementary medicine as needed, such as acupuncture, injections and infusions.

Q: It is unusual for neurology divisions to run multiple pediatric trials focused on headaches. You are currently leading five that are open. How does this work move the field forward?

A: The medications we offer through our trials allow us to offer treatments that would otherwise not be available to pediatric patients. We do this in hopes of providing them relief while advancing the field. We are hopeful that these new therapies are as effective in pediatrics as they have shown to be in adults. But it is necessary to complete randomized clinical trials to prove this is the case. Historically, pediatric patients in clinical trials investigating painful conditions like migraines have had a disproportionately high placebo response rate. This means even the patients receiving a benign placebo have a high chance of symptom improvement. The newer medications show much better tolerability to the drugs used historically.

Q: What excites you about this work?

A: Pediatric pain disorders are unbelievably gratifying to treat because we take a mysterious disorder that waxes and wanes with no clear reason and give patients back control of their lives. It is extremely frustrating for a patient and their family to know that their day-to-day life can be abruptly derailed by a pain crisis. We work to provide them with several tools they can use daily to take back their lives.

Q: How is this work unique?

A: Our program was created organically over the years through our experiences with our patients. First, we noticed the disruption to patients’ personal and school performance from having untreated pain and recognized the need for pain psychology. Then, we expanded to have physical therapy to recondition patients and perform desensitization. Finally, we recognized our patients need additional medication options not offered through the standard of care. So, we expanded to open our various clinical trials, including those with pharma and internal protocols. As a result, we incorporated the use of Botox injections, for example, and soon will use a novel remote electro-neuromodulatory device.

girl with down syndrome

Study finds delayed oligodendrocyte progenitor maturation in Down syndrome

girl with down syndrome

People with Down syndrome (DS) can have moderate to severe intellectual disability, which is thought to be associated with changes in early brain development.

People with Down syndrome (DS) can have moderate to severe intellectual disability, which is thought to be associated with changes in early brain development. Children’s National Hospital experts discovered delayed maturation in oligodendrocyte progenitors in DS. Oligodendrocytes produce the white matter which insulates neural pathways and ensures speedy electrical communication in the brain. The researchers identified these delays by measuring gene expression at key steps in cell development, according to a new study published in Frontiers in Cellular Neuroscience.

The findings further suggest that brain and spinal cord oligodendrocytes differ in their developmental trajectories and that “brain-like” oligodendrocyte progenitors were most different from control cells, indicating that oligodendrocytes in the brains of people with DS are not equally affected by the trisomy 21.

“This is one of the critical steps towards identifying the key stages and molecular players in the DS white matter deficits,” said Tarik Haydar, Ph.D., director of the Center for Neuroscience Research. “With this knowledge, and with further work in this direction, we envision future therapies that may improve nerve cell communication in the brains of people with Down syndrome.”

The hold-up in the field

The mechanisms that lead to the reduction of white matter in the brains of people with DS are unknown. To better understand early neural precursors, they used isogenic pluripotent stem cell lines derived from two individuals with Down syndrome to study the brain development and spinal cord oligodendrocytes.

“I was excited that we discovered another example of how important it is not to generalize when studying DS brain development,” said Haydar. “This is one of several papers, from our group and others, that demonstrate how important it is to be very specific about the brain area and the developmental stage when investigating the causes of DS brain dysfunction.”

What’s next

Dysmaturation of oligodendrocyte cells are a relatively new discovery by the Haydar Lab, one of the preeminent labs in DS research. These results isolate specific steps that are affected in human cells with trisomy 21. They are using these results to develop a drug screening platform that may prevent altered generation of oligodendrocytes in the future.

You can read the full study “Sonic Hedgehog Pathway Modulation Normalizes Expression of Olig2 in Rostrally Patterned NPCs With Trisomy 21” in Frontiers in Cellular Neuroscience.

DNA moleucle

Multidisciplinary team seeks to reverse epigenetic changes associated with fetal alcohol syndrome disorder

DNA moleucle

The team hopes to optimize and develop treatments that can reverse epigenetic changes in clinical trials, paving the way to make significant progress in the field — something that is lacking to date.

A clinical team joined forces with a research team at Children’s National Hospital to help advance treatments that can improve a child’s development caused by fetal alcohol syndrome disorder (FASDs), which is a group of conditions that can occur in a person who was exposed to alcohol before birth. This boost in collaboration between the bench and clinical hopes to optimize and develop treatments that can reverse epigenetic changes in clinical trials, paving the way to make significant progress in the field — something that is lacking to date.

So far, Children’s National experts have published various pre-clinical studies that identified epigenetic changes caused by alcohol exposure during pregnancy. These changes observed in the pre-clinical models created neuropsychiatric problems like patients with fetal alcohol syndrome disorder. Now, they want to bring such potential treatments effective in pre-clinical models to the bedside.

“As a first step, we are going to test whether the epigenetic changes that were observed in pre-clinical models of FASD are also true in human patients,” said Kazue Hashimoto-Torii, Ph.D., principal investigator of the Center for Neuroscience Research at Children’s National. “We hope a small amount of blood donated by patients with FASD reveal the changes. Meanwhile, my group has also been optimizing drug candidates that reverse the epigenetic changes toward clinical trials.”

Advances in genetics and genomics have led to discoveries about the timing of exposure and developmental outcomes and genetic and epigenetic signatures that may be protective or harmful in terms of how in utero alcohol exposure affects developmental outcomes.

The hold-up in the field

While the exact number of people with FASDs is unknown, the National Institutes of Health estimates that 1% to 5% of the population have FASDs. FASDs has a spectrum of diagnoses that represent a broad range of effects that can be manifested in an individual whose mother drank alcohol during pregnancy. These conditions can affect everyone in different ways and range from mild to severe. Individuals with mild conditions may go undiagnosed. The more affected individuals have comorbid attention-deficit/hyperactivity disorder (ADHD) and behavioral problems that become the focus of clinical encounters. The individual’s health care provider may not recognize the core features as part of FASD.

“Because there is a stigma associated with drinking while pregnant, many providers fail to get this history, and women may be reluctant to offer this information,” said Andrea Gropman, M.D., division chief of Neurodevelopmental Pediatrics and Neurogenetics at Children’s National. “There are subtle and more obvious facial dysmorphology that may help with suspicion or identification, but many individuals do not have these findings.”

The core features may be nonspecific, such as intellectual disabilities and problems with behavior and learning, difficulties with math, memory, attention, judgment and poor impulse control, which are frequent findings in ADHD, autism, learning disorders and other conditions.

“Unless history is taken and FASD is in the differential diagnosis, the diagnosis may not be made,” said Dr. Gropman. “Individuals with FASD may feel stigmatized and opt not to participate in clinical trials.”

As mentioned by Dr. Gropman, stigma can make a patient family be reluctant to seek treatment, and thus the development of treatment for FASD cannot make significant progress to date, Hashimoto-Torii added.

Children’s National Hospital leads the way in an IRB approved study

Researchers at Children’s National have identified a potential drug candidate that reverse the epigenetic changes and may lead to clinical trials. The team is seeking people to participate in an IRB approved study. The study will involve cognitive testing, filling out surveys about current functioning and cheek swab and blood sample to determine if these changes are seen in patients. To participate, subjects must be

  • Children between the ages 5-12 with prenatal alcohol exposure.
  • Mother of child recruited above.

For participation, please contact Grace Johnson, research coordinator at to screen for eligibility at 202-476-6034 or gjohnson3@childrensnational.org

Meet the multidisciplinary team with different yet complementary skills in different fields, such as basic science, medical, social sciences, neurology and developmental disabilities, and development, who are working tirelessly to address the complex health problem.

Gropman lab:

Andrea Gropman, M.D., received her medical doctorate degree from the University of Massachusetts Medical School and specializes in neurogenetics, with a focus on mitochondrial disorders and Smith Magenis syndrome. Her latest research focuses on atypical patterns of inheritance, childhood mitochondrial disorders and other inborn errors of metabolism presenting with white matter disease.

Meira Meltzer, M.A., M.S., C.G.C., genetic counselor with a demonstrated history of working in the hospital and healthcare industry. Also skilled in molecular biology, clinical research and medical education. Strong healthcare services professional with a M.S. focused on genetic counseling from Brandeis University.

Cathy Scheiner, M.D., developmental behavioral pediatrician with a special interest in attention-deficit / hyperactivity disorder (ADHD), cerebral palsy and premature infant.

Grace Johnson, research assistant.

Hashimoto-Torii Lab:

Kazue Hashimoto-Torii, Ph.D., received her postdoctoral training in the Pasko Rakic laboratory at Yale University. Her research focuses on neurobehavior problems of children that stem from their environment during development, such as prenatal exposure to alcohol, drug and high-level glucose. A few drug candidates that her lab discovered have been patented and her lab is currently working hard to bring those medicines to bedside.

Satoshi Yamashita, M.D., Ph.D., postdoctoral research fellow skilled in developmental neurobiology. He is a pediatrician with Japanese medical license and received his Ph.D. with iPS cell research for STXBP1 encephalopathy in Japan.

Chiho Yamashita, B.N., research assistant passionate about child disease research. She is a nurse with a Japanese nursing license and worked in the pediatric department in Japan.

cancer cells

Advancing immunotherapy for pediatric brain tumors

cancer cells

While immunotherapy has revolutionized cancer treatments, its efficacy remains relatively undefined in pediatric settings for brain tumors.

While immunotherapy has revolutionized cancer treatments, its efficacy remains relatively undefined in pediatric settings for brain tumors. Children’s National Hospital experts and other institutions argue in a review published in Nature Cancer that there is a need for closer collaborations between academia, industry partners, regulatory bodies and funders to progress the field.

Eugene Hwang, M.D., associate division chief of Oncology and neuro-oncologist at Children’s National, led the review that outlines immunotherapeutic hurdles and simultaneously proposes next steps for immunotherapy use in these patients. These considerations will aid pediatric oncologists make better recommendations and advances in this type of treatment.

“The promise of immunotherapy in helping to cure children with brain tumors is exciting,” Dr. Hwang said. “This type of approach has already revolutionized treatments for many different kinds of cancer, and a comprehensive review of this complicated arena, especially by leading voices in the field, can help set the stage for finally moving the needle for these patients.”

The review is especially helpful as children harbor unique elements of immunity and the brain presents distinct obstacles to immune attack that are not present in other cancers. For example, there are challenges in antigen identification, the blood-brain barrier and the tumor microenvironment. For many pediatric cancer doctors as well this novel, complex form of therapy is outside of their historical training.

To overcome these challenges, the authors encourage philanthropic organizations and patient advocacy groups to be part of the process that can help fill funding gaps in patient-focused pre-clinical and clinical research and educate patients and families.

“Multiple stakeholders around pediatric brain cancer immunotherapy must be mobilized in a concerted fashion,” Hawk et al. argue in the piece. “The need for close academic collaboration with industry partners and regulatory bodies is increasingly apparent given the unique pediatric phenotypes and complex outcomes in immunotherapeutic trials, and progress will be made at the interface of the interactions of all these key stakeholders.”

The group of internationally renowned pediatric brain tumor-focused immunotherapy experts comprehensively reviewed the advances in the major modalities of immunotherapy and the landscape of preclinical modeling for these patients to date.

Investigators at Children’s National, for example, are leading several national and international trials involving immunotherapy which have spurred international meetings with a focus in childhood brain tumor immunotherapies.

“The multiple T cell trials led by Children’s National are perfect examples of truly field-leading innovative immunotherapy, as are the other trials that are led by our own investigators,” Dr. Hwang added.

Representative OFF and DIFF spectra

GABA and glutamate in the preterm neonatal brain

Preterm and sick newborns are at high risk of brain injury that can lead to cognitive delays and behavioral disorders including autism and ADHD. Gamma-aminobutyric acid (GABA) and glutamate system disruptions may underlie these neonatal brain injuries and hence it is important to describe their normative profile in the developing neonatal brain.

In a study led by Sudeepta Basu, M.D., neonatologist at Children’s National Hospital and Assistant Professor of Pediatrics at George Washington University School of Medicine and Health Sciences, specialized GABA editing spectroscopy (MEGA-PRESS) was acquired on a 3Tesla MRI scanner. Although MEGA-PRESS has been used in older subjects, there are challenges in the newborn population that have limited investigations with only a few institutions worldwide. Under the leadership of Catherine Limperopoulos, Ph.D., in the Developing Brain Institute (DBI) at Children’s National, a team of scientists (in particular, Dr. Subechhya Pradhan) have diligently overcome the technical challenges to enable use of this cutting-edge technology for research at the institute.

With this unique capability, Dr. Basu’s team prospectively enrolled 58 healthy newborns to describe the normal GABA and glutamate concentrations in different regions of the developing brain. In a recent article published in the American Journal of Neuroradiology, Dr. Basu reports that GABA and glutamate concentrations were highest in the cerebellum, slightly lower in the basal ganglia, but significantly lower in the frontal lobe.

“Our ability to reliably describe the normal metabolic-neurotransmitter milieu of the developing newborn brain is the first step in filling a critical gap in knowledge,” says Dr. Basu. “We hope to identify early bio-markers of brain injury of cognitive delays and autism and ADHD risk which remains a major challenge until clinical symptoms manifest later in childhood.”

Under the direction of Dr. Limperopoulos, advanced multi-modal high precision MRI protocols have been developed for use in research studies at Children’s National that allows the scientists to identify subtle signs of delayed growth and development of the newborn brain. With the optimization of MEGA-PRESS for newborns, Children’s National is one of a few institutions worldwide capable of investigating the newborn brain neurotransmitters in future research studies.

Read the full article in American Journal of Neuroradiology.

Representative OFF and DIFF spectra

Representative OFF and DIFF spectra.

x-ray of child with dislocated hip

Hip surveillance helps identify dislocations in children with cerebral palsy

x-ray of child with dislocated hip

Hip surveillance is a process used to monitor the hips closely and frequently, identifying the problems earlier.

Children with cerebral palsy (CP) have an increased risk for hip displacement. Hip displacement in children with CP can happen slowly over time and can be painful, but a hip surveillance program can prevent this. Hip surveillance is a process used to monitor the hips closely and frequently, identifying the problems earlier. It is an ongoing process that continues for every child until skeletal maturity.

“Every child with cerebral palsy should be referred for hip surveillance regardless of determination by the Gross Motor Function Classification System,” said Sean Tabaie, M.D., orthopaedic surgeon at Children’s National Hospital.

Dr. Tabaie created a hip surveillance manual for primary care providers who care for this patient population. In most cases, these patients are monitored and followed closely by their primary care team. Education material regarding hip surveillance, including the background knowledge, is often not available to those practitioners in a concise format. To successfully initiate a hip surveillance program, it is important to promote education and provide the appropriate materials to that group of practitioners.

“Our goal is to improve the care of children with cerebral palsy by decreasing the overall presentation of dislocated hips in our clinic settings and promote the appropriate timing of referrals for evaluation of hip subluxation secondary to cerebral palsy or neuromuscular conditions,” said Tabaie.

Download the Surveillance Guidelines for Children with Cerebral Palsy here.

Neuronal network with electrical activity

Neonatal hypoxia-ischemia causes damage to the cholinergic system

Neuronal network with electrical activity

Study suggests permanent injury to the cholinergic system after neonatal hypoxia-ischemia is responsible for the poor executive functions and difficulties in learning and memory.

Newborn babies who go through periods of low oxygen — also known as hypoxic-ischemic encephalopathy — during their first hours of life often experience difficulties in learning, memory and executive functions later on. Even when treated with therapeutic hypothermia, memory deficits and executive functions remain severely affected. These functions are linked to a neurotransmitter network called the cholinergic system.

“Complications from hypoxic-ischemic brain injury contribute to one-quarter of neonatal deaths worldwide and cause significant long-term neurological morbidity,” explains Panagiotis Kratimenos, M.D., Ph.D., neonatologist at Children’s National Hospital and Assistant Professor of Pediatrics at the George Washington University School of Medicine and Health Sciences.

In a study published in the Journal of Comparative Neurology led by Frances Northington, M.D., co-director of Neurosciences Intensive Care Nursery at Johns Hopkins and Professor of Pediatrics at Johns Hopkins University School of Medicine, with contributions from Dr. Kratimenos, the authors found significant injury to the neurons of the cholinergic systems in specific parts of the brain after exposure to low oxygen and restricted blood flow. These areas included the ipsilateral medial septal nucleus (MSN), the ipsilateral nucleus basalis of Meynert (nbM) and striatum. Within the injured part of the cortex at the site of injury, acetylcholine — the neurotransmitter found in cholinergic systems — was abnormally overactivated.

The authors hypothesize that permanent injury to the cholinergic system after neonatal hypoxia-ischemia is responsible for the poor executive functions and difficulties in learning and memory.

“Because cholinergic systems can easily be manipulated pharmacologically with already established treatments that have been used in other areas of medicine, they could be a good a target for therapeutic interventions for neonates with hypoxic-ischemic encephalopathy,” says Dr. Kratimenos.

Read the full article in the Journal of Comparative Neurology.

Tommie Robinson

Tommie L. Robinson, Jr., Ph.D., receives ASHA award

Tommie Robinson

Tommie L. Robinson, Jr., Ph.D., CCC-SLP, division chief of Hearing and Speech at Children’s National Hospital.

Recognized for his transformational and innovative leadership and his impactful work in the areas of diversity and inclusion, Tommie L. Robinson, Jr., Ph.D., CCC-SLP, division chief of Hearing and Speech at Children’s National Hospital, received the 2021 American Speech-Language-Hearing Association (ASHA) Honors award.

ASHA recognizes members for their distinguished contributions to the discipline of communication sciences and disorders and is the highest honor that the more than 218,000 member organization bestows. The Honors of the Association award recognizes the contributions of individuals of such excellence that have enhanced or altered the course of the profession.

Dr. Robinson was nominated for his transformational and innovative leadership, his work in the areas of diversity and inclusion, and his mentorship over his career. Dr. Robinson is one of the earliest researchers to examine the characteristics of fluency and disfluencies in Blacks. His unique work laid the foundation for his later efforts in the application of that knowledge to intervention in Black children who stutter. Dr. Robinson’s work continues to serve as the primary contribution to the science of the discipline in this area.

Throughout his career, Dr. Robinson has served many leadership roles including: being elected and serving as the first Black male president of ASHA; leadership roles with the District of Columbia Speech-Language-Hearing Association (DCSHA) and his work with the National Black Association for Speech Language and Hearing (NBASLH); the American Speech-Language-Hearing Foundation (ASH Foundation); and the International Association of Logopedics and Phoniatrics (IALP).  Throughout his leadership roles, Dr. Robinson has always been an innovative leader on issues of diversity and empowerment and he noted he is “honored to be recognized by my peers.”

In commenting about Dr. Robinson’s leadership, Patricia Prelock, Ph.D., former ASHA President and Provost/Senior Vice President at the University of Vermont, commented, “He initiated discussions on the importance of and value in developing new leaders.” She further recognized his commitment to ensuring the ASHFoundation sparks innovation while investing in researchers and clinicians.

Dr. Robinson is also respected for his leadership and his mentorship within Children’s National Hospital, where he has been since 1987.  Beginning as a speech-language pathologist, he was later promoted to coordinator of speech-language pathology and then as director of the Scottish Rite Center for communication disorders. Denice Cora-Bramble, M.D., chief diversity officer at Children’s National, noted, “I have been most impressed by his strong leadership skills and his ability to overcome obstacles. His affable and supportive interpersonal skills have been a pillar of strength for other colleagues, particularly to minorities in academia.”

Roger Packer, M.D., senior vice president of the Center for Neuroscience and Behavioral Medicine at Children’s National, stated that in Dr. Robinson’s “long and stellar career, he has worked tirelessly to promote the need for early and comprehensive speech-language-hearing services for children across the Washington, D.C., region, including those from the most impoverished and financially challenged areas. He has been a champion for diversity and inclusion and has mentored many minority trainees, including Black students. He has continued to act as a role model for these trainees as they proceed in their careers.”

The 2021 ASHA Award recipients will be honored at the ASHA 2021 annual meeting held from November 18-20, 2021 in Washington, D.C. To learn about all the award recipients, visit the ASHA web site here.

Dr. Javad Nazarian

Q&A with Dr. Javad Nazarian on his upcoming work on low-grade gliomas

Dr. Javad Nazarian

Supported by the Gilbert Family Foundation, Dr. Nazarian’s return is part of a special research program within the Gilbert Family Neurofibromatosis Institute that focuses on NF1 research.

Javad Nazarian, Ph.D., M.Sc., associate professor of Pediatrics at George Washington University and professor at the University of Zurich, has expanded his research group at Children’s National to focus on Neurofibromatosis type 1 (NF1) transformed low-grade gliomas (LGGs). Dr. Nazarian will apply his expertise from establishing a successful DIPG (diffuse intrinsic pontine glioma) and DMG (diffuse midline glioma) program in Zurich Switzerland and previously at Children’s National.

In addition to his continued research in Zurich, as a principal investigator at the Department of Genomics and Precision Medicine at Children’s National Dr. Nazarian plans on aggregating his knowledge to the new research and work spearheaded at Children’s National. As one of the first research teams to move to the Children’s National Research & Innovation Campus, Dr. Nazarian’s group is excited to use the opportunity to establish cutting-edge and clinically translational platforms.

Supported by the Gilbert Family Foundation, Dr. Nazarian’s return is part of a special research program within the Gilbert Family Neurofibromatosis Institute that focuses on NF1 research. This research includes associated gliomas with a special emphasis on NF1-associated transformed anaplastic LGGs. His team will develop new avenues of research into childhood and young adult NF-associated LGGs with a special emphasis on transformed high-grade gliomas.

Dr. Nazarian is excited for what’s to come and his goals are clear and set. Here, Dr. Nazarian tells us more about his main objectives and what it means for the future of pediatric neuro-oncology care at Children’s National.

  1. What excites you most about being back at Children’s National?

I have received most of my training at Children’s National, so this is home for me. Being one of the nation’s top children’s hospitals gives a unique advantage and ability to advocate for childhood diseases and cancers. It is always exciting to play a part in the vision of Children’s National.

  1. What are some of the lessons learned during your time working in Zurich? And how do you think these will compliment your work at Children’s National?

We developed a focused group with basic research activities intertwined with clinical needs.  The result was the launch of two clinical trials. I also helped in developing the Diffuse Midline Glioma-Adaptive Combinatory Trial (DMG-ACT) working group that spans across the world with over 18-member institutions that will help to design the next generation clinical trials. I will continue leading the research component of these efforts, which will have a positive impact on our research activities at Children’s National.

  1. How does your work focusing on low-grade gliomas formulating into high-grade gliomas expand and place Children’s National as a leader in the field?

Scientifically speaking, transformed LLGs are very intriguing. I became interested in the field because these tumors share molecular signatures similar to high-grade gliomas (HGGs). Our team has done a great job at Children’s National to develop tools – including biorepositories, avatar models, drug screening platforms, focused working groups, etc. – for HGGs. We will apply the same model to transformed LGGs with the goal of developing biology-derived clinical therapeutics for this patient population.

  1. How will this work support families and patients seeking specific neuro-oncology care?

We will develop new and high thruput tools so that we can better study cancer formation or transformation. These tools and platforms will allow us to screen candidate drugs that will be clinically effective. The main focus is to accelerate discovery, push drugs to the clinic, feed information back to the lab from clinical and subsequently design better therapies.

  1. You are one of the first scientists to move to the Children’s National Research & Innovation Campus. What are some of the valuable changes or advancements you hope to see as a result of the move?

The campus will provide high-end facilities, including cutting-edge preclinical space, and allow for team expansion. The close proximity to Virginia Tech will also provide an environment for cross-discipline interactions.

  1. Anything else you think peers in your field should know about you, the field or our program?

The team at Children’s National includes Drs. Roger Packer and Miriam Bornhorst. Both have provided constant clinical support, innovation and clinical translation of our findings. I look forward to working with them.

brain network illustration

Cardiopulmonary bypass may cause significant changes to developing brain and nerve cells

brain network illustration

Cardiopulmonary bypass, more commonly known as heart-and-lung bypass, has some unique impacts on the creation and growth of brain cells in the area of a child’s brain called the subventricular zone (SVZ), according to a study in the Annals of Neurology. The SVZ is a critical area for the growth and migration of neurons and nerve cells called neuroblasts, both of which ultimately contribute to the proper development of key brain structures and functions during the early years of life.

The findings, from a study conducted in the Cardiac Surgery Research Laboratory at Children’s National Hospital, provide new insight into the cellular impacts of the cardiopulmonary bypass machine on brain growth and development for newborn infants with congenital heart disease. They will have an important role in the refinement of strategies to help protect the fragile brains of children who require lifesaving cardiac surgery with cardiopulmonary bypass immediately after birth.

Specifically, the research team found that during cardiopulmonary bypass:

  • Creation of neurons (neurogenesis) in the neonatal and infant subventricular zone is altered.
  • Migration of nerve cells, called neuroblasts, to the frontal lobe is potentially disrupted.
  • Changes to the growth and movement of neurons in the SVZ are prolonged.
  • Cortical development and expansion is impaired.
  • Specific types of neurons found only in the brain and spinal cord, called interneurons, are also affected.

The study uses an innovative pre-clinical model of the developing brain that is more anatomically and physiologically similar to human neonates and infants than those used in prior studies and in most neurological laboratory-based research.

Cardiopulmonary bypass is one of several key factors thought to cause children with congenital heart disease to sometimes demonstrate delays in the development of cognitive and motor skills. These disabilities often persist into adolescence and adulthood and can ultimately represent long-term neurocognitive disabilities. It is also believed that genetic factors, abnormal blood flow to the brain while in utero or low cardiac output after surgical procedures on the heart may contribute to these challenges.

“Unraveling cellular and molecular events during surgery using this preclinical model will allow us to design therapeutic approaches that can be restorative or reparative to the neurogenic potential of the neuronal stem precursor cells found in the subventricular zone of the neonatal or infant brain,” says Nobuyuki Ishibashi. M.D., Foglia-Hills Professor of Pediatric Cardiac Research, director of the Cardiac Surgery Research Laboratory at Children’s National and senior author on the study. “In particular, previous studies in our laboratory have shown improvement in the neurogenic activities of these precursor cells when they are treated with mesenchymal stromal cells (MSCs).”

The findings from this study further support the work already underway in the NIH-funded MeDCaP clinical trial for neonates and infants undergoing cardiac surgery using the cardiopulmonary bypass machine. That trial uses the heart and lung machine itself to deliver MSCs directly into the main arteries that carry blood to the brain.

microglia cells damage the myelin sheath of neuron axons

Katrina Adams, Ph.D., awarded fellowship to help restore functions in MS patients

microglia cells damage the myelin sheath of neuron axons

Multiple sclerosis is a demyelinating disease in which the insulating covers of nerve cells are damaged. Microglia cells (orange) attack the oligodendrocytes that form the insulating myelin sheath around neuron axons, leading to the destruction of the myelin sheath and to the loss of nerve function.

For her contributions to Multiple Sclerosis (MS) research, Katrina Adams, Ph.D., postdoctoral researcher at Children’s National Hospital, received the career transition fellowship from The National Multiple Sclerosis Society. The $600,000 fellowship will support a two-year period of advanced postdoctoral training in MS research and the first three years of research support in a new faculty appointment.

MS symptoms, including vision loss, pain, fatigue and reduced motor coordination, result from the demyelination of neuronal axons that transport critical information across the brain and spinal cord. Demyelination is the loss of myelin protein, which is normally produced by oligodendrocyte cells.

In the healthy brain, oligodendrocytes repair demyelinated areas by replacing damaged or lost myelin in a process called remyelination. Recent evidence has shown that oligodendrocytes display differences in their molecular and functional properties. One source of new oligodendrocytes in the adult brain is neural stem cells, which have been shown to generate oligodendrocytes that contribute to remyelination.

“The goal of this project is to determine whether neural stem cell-derived oligodendrocytes are distinct from other oligodendrocytes, both in the healthy brain and in MS,” said Adams. “I aim to understand the molecular mechanisms that regulate generation of oligodendrocytes from neural stem cells, with the goal of identifying signals that could be targeted in MS patients to promote remyelination.”

Remyelination is very limited in MS patients and current therapies for MS have very little impact on promoting remyelination.

This study will take advantage of the state-of-the-art facilities for single-cell analysis, transcriptomics, microscopy, and animal research in Children’s Research Institute at Children’s National. Adams also added that her postdoctoral mentor, Vittorio Gallo, Ph.D., interim chief academic officer and interim director of the Children’s National Research Institute, and principal investigator for the DC-IDDRC, has renowned expertise in glial biology, animal models of MS and white matter injury.

“This research will be the first to directly compare neural stem cell-derived oligodendrocytes with other resident oligodendrocytes in MS brain samples,” said Adams. “The results of this study will provide critical insight into the role that neural stem cells play in repair of MS demyelinated lesions.”

Adams received her doctorate in molecular biology from the University of California, Los Angeles where she used pluripotent stem cells to study motor neuron development. She currently investigates signaling pathways that regulate neural stem and progenitor cell maintenance and differentiation in the developing postnatal and adult brain, with a focus on the Endothelin-1 pathway. She is interested in understanding how stem and progenitor cells respond to disease or injury, such as in Multiple Sclerosis, with the hope of identifying new therapeutic targets.

doctor looking at brain MRIs

NINDS awards $10 million for pediatric concussion research

doctor looking at brain MRIs

Researchers will use advanced brain imaging and blood tests to explore biological markers—changes in blood pressure, heart rate and pupil reactivity—that could predict which children will develop persistent symptoms after concussion.

The National Institute of Neurological Disorders and Stroke has awarded a $10-million grant to the Four Corners Youth Consortium, a group of academic medical centers studying concussions in school-aged children. Led in part by the Safe Concussion Outcome Recovery and Education (SCORE) program at Children’s National Hospital, the project is named Concussion Assessment, Research and Education for Kids, or CARE4Kids.

Researchers will use advanced brain imaging and blood tests to explore biological markers—changes in blood pressure, heart rate and pupil reactivity—that could predict which children will develop persistent symptoms after concussion. The five-year CARE4Kids study will enroll more than 1,300 children ages 11-18 nationwide.

The five-year study will be led by Gerard Gioia, Ph.D., division chief of Neuropsychology at Children’s National Hospital, Frederick Rivara, M.D., M.P.H., at Seattle Children’s Center for Child Health, Behavior and Development and University of Washington’s Medicine’s Department of Pediatrics, and Dr. Chris Giza at University of California, Los Angeles (UCLA).

“We will be gathering innovative data to help answer the critical question asked by every patient: ‘When can I expect to recover from this concussion?’” said Dr. Gioia. “We have a great team and are excited to have been selected to study this important issue.”

Christopher G. Vaughan, Psy.D., neuropsychologist, and Raquel Langdon, M.D., neurologist, both at Children’s National, will join Dr. Gioia as principal investigators of the study at this site.

Every year, more than 3 million Americans are diagnosed with concussions. Symptoms continue to plague 30 percent of patients three months after injury—adolescents face an even higher risk of delayed recovery. Chronic migraine headaches, learning and memory problems, exercise intolerance, sleep disturbances, anxiety and depressed mood are common.

“Providing individualized symptom-specific treatments for youth with a concussion has been a longstanding aim of the SCORE program,”Dr. Vaughan said. “This project will lead to a better understanding of the specific markers for which children may have a longer recovery. With this knowledge, we can start individualized treatments earlier in the process and ultimately help to reduce the number of children who experienced prolonged effects after concussion.”

The grant was announced on September 9, 2021.

In Washington, D.C., an estimated 240 children ages 11 to 18, will participate in the study.

The study will unfold in two phases. The first part will evaluate children with concussion to identify a set of biomarkers predictive of persistent post-concussion symptoms. To validate the findings, the next stage will confirm that these biomarkers accurately predict prolonged symptoms in a second group of children who have been diagnosed with concussion. The goal is to develop a practical algorithm for use in general clinical practice for doctors and other health professionals caring for pediatric patients.

Institutions currently recruiting patients for the study include Children’s National Hospital, UCLA Mattel Children’s Hospital, Seattle Children’s, the University of Washington, University of Rochester, University of Texas Southwestern Medical Center and Wake Forest School of Medicine. Indiana University, the National Institute of Nursing Research, University of Arkansas, University of Southern California and the data coordinating center at the University of Utah are also involved in the project.

Earlier research conducted by the Four Corners Youth Consortium that led to this project was funded by private donations from Stan and Patti Silver, the UCLA Steve Tisch BrainSPORT Program and the UCLA Easton Clinic for Brain Health; Children’s National Research Institute; as well as from the Satterberg Foundation to Seattle Children’s Research Institute; and an investment from the Sports Institute at UW Medicine.