Cancer

patient undergoing MRI

Brain tumor team performs first ever LIFU procedure on pediatric DIPG patient

patient undergoing MRI

The ultrasound waves activate the drug selectively within the tumor, causing tumor cell death. Credit: Image provided by Insightec.

A multidisciplinary brain tumor team at Children’s National Hospital successfully performed the first treatment with sonodynamic therapy utilizing low intensity focused ultrasound (LIFU) and 5-aminolevulinic acid (5-ALA) medication on a pediatric patient. The treatment, performed on a 5-year-old child diagnosed with a diffuse intrinsic pontine glioma (DIPG), was done noninvasively through an intact skull. The child was discharged from the hospital one day later.

What happened?

Shortly after announcing the use of LIFU, the brain tumor team at Children’s National treated the patient as part of a cutting-edge trial using LIFU combined with a novel medication.

The ultrasound waves – which are given while the child is asleep through an intact skull and does not require an invasive neurosurgical procedure – activate the drug selectively within the tumor, causing tumor cell death.

“This treatment is currently being trialed in adults diagnosed with recurrent glioblastoma tumors, but has never been attempted in pediatric patients,” said Hasan Syed, M.D., co-director of the Focused Ultrasound Program at Children’s National. “Similar to the adult trial, our protocol involves using a medication that is taken up by tumor cells and then targeting those cells with LIFU to induce tumor cell death, and hopefully leading to tumor control.”

Dr. Syed co-directs the program with Roger Packer, M.D., head of the Brain Tumor Institute, and Lindsay Kilburn, M.D., director of the Experimental Therapeutics Program.

How are we leading the way?

The launch and use of LIFU was possible thanks to the efforts of a multidisciplinary team from various departments that understood if too high a dose of ultrasound was utilized, there could be associated brain swelling and even death.

“Our efforts show great teamwork and a commitment from the hospital and our clinical teams to develop innovative means to treat a tumor that kills 90% of those children afflicted within 18 months of diagnosis,” Dr. Syed said.

The work shows expertise of the brain tumor team, as well as radiology, anesthesiology and intensive care units.

“Despite the risks involved, the use of focused ultrasound is a novel way to try to treat these very deep-seated lesions that have been highly resistant to all forms of therapy and is potentially the greatest breakthrough we’ve had in this disease in the past 50 years,” Dr. Packer said.

What has limited therapy in the past?

DIPGs are deep-seated in critical areas of brain, controlling breathing and heart rate and cannot be removed. The brain has an intrinsic system called a blood brain barrier which blocks drugs from getting to the tumor.

Focused ultrasound is a new way to overcome the brain’s ability to stop the drugs from getting there. It can also be used to activate a drug as it passes through the brain stem.

“We are extremely excited to have taken the first step in developing this novel and non-invasive approach to treating one of our most deadly brain tumors,” Dr. Kilburn said. “This is the first step of numerous steps toward evaluating the many potential uses of LIFU as part of combination therapies to treat children with DIPGs and eventually other pediatric brain tumors.”

Children’s National is partnering with other institutions across the world to perform these studies. But because of the commitment of its team and its expertise, it is the first to use this technique in a child.

“I think we’re in a unique position thanks to the collaborations possible at Children’s National and the expertise of those caring for children with brain tumors,” Dr. Packer added.

Why we’re excited

The Brain Tumor Institute at Children’s National is excited about making this a potential treatment option for DIPG patients, which currently have really no surgical options or alternatives. It’s a way to deliver the ultrasound and therapies in a potentially less toxic way, not requiring surgery.

This trial and subsequently others will give doctors more options for children with DIPGs and other malignant tumors.

Cancer cells

Searching for the key to treating neuroblastoma tumors in kids

Cancer cells

Jianhua Yang, Ph.D., has dedicated his research to finding the molecular mechanism of neuroblastoma development and is working to develop novel therapeutics.

There continues to be an urgent need to identify novel therapies for childhood cancers. Neuroblastoma (NB) is the most common malignant solid tumor in children and contributes to more than 15% of all pediatric cancer-related deaths. Despite strides made in chemotherapy treatment over the past 30 years, NB largely remains an incurable disease. That’s why Jianhua Yang, Ph.D., associate professor and research faculty at the Center for Cancer and Immunology Research at Children’s National Hospital, has dedicated his research to finding the molecular mechanism of NB development and is working to develop novel therapeutics to target molecules he and his team identify in the lab.

Q: What has driven you to do this research?

A: In order to design better and potentially more effective NB treatment approaches, we must further understand the mechanism that activates NB development. We don’t know what that mechanism is yet, and that’s what we’re working to unlock. I felt with my training in cell biology and immunology, I could use that background to help develop novel therapies.

The research is hard and can often times feel frustrating. But I feel I’m working on something that has the potential to make a huge difference. I tell the researchers I work with that you have to really believe in what we’re doing. We’re doing something very different. Before I moved to D.C. to join Children’s National, I sent a text to a former mentor to let him know I was joining the team here to continue my work. His reply said, “I’ve always had confidence in you,” and it’s that type of encouragement that drives me to keep going.

Q: What is your current focus in this area?

A: Specifically, we’re working on two targets right now:

  1. To define the role and regulation of CaM kinase-like vesicle-associated (CAMKV) in NB development and examining the therapeutic potential of CAMKV kinase inhibition for treating NB in pre-clinical models. We’ve found that CAMKV is highly expressed in NB tumor samples and its kinase activity is required for tumor growth. So, if we knock out this gene, tumor cells will die. We’re studying how it is being activated, and if we can find out what causes it, we can find a way to inhibit its activation. Targeting CAMKV is a novel concept for treating NB. CAMKV kinase inhibitors may serve not only as stand-alone therapies but also as effective adjuncts to current chemotherapeutic regimens treating this aggressive pediatric malignancy.
  2. To define the role and regulation of transmembrane protein 108 (TMEM108) in NB development and examine the therapeutic potential of TMEM108 functional blockade for treating NB in pre-clinical models. Evolutionarily, in human genome it has no other family member, it’s a loner. And if you knock it out in NB tumor cells, tumor cells will die. We’re learning how it functions through our basic research, which is quite difficult. But we’re thinking if we can find the antibody to bind to it and block its function, we could stop the tumor from growing or even cause the tumor to die.

Q: What excites you about doing this work within the Center for Cancer and Immunology Research?

A: At Children’s National, I’ll be able to combine my work with the incredible work in immunotherapy that Drs. Catherine Bollard and Muller Fabbri are doing. I’m excited to be here to have that strong collaboration with their labs to develop new therapies.

In the next 5 years, I feel we’ll be able to identify good blocking antibodies that we can then test combinations of to see how it blocks tumor growth. If we can find ways to combine that antibody therapy with traditional chemotherapy options, we can achieve a real cure for NB.

Learn more about the Center for Cancer and Immunology Research.

Researchers hope to uncover puzzling mechanism of vision loss in kids with $2.7M DOD award

The Department of Defense Neurofibromatosis Research Program awarded Children’s National Hospital $2.7M to better understand a pediatric tumor as a blinding disease. The study design will specifically focus on targeting immune responses during the development of the tumor as a means to prevent or preserve vision before the tumor-associated irreversible neurological damage.

Why it matters

Nearly 20% of individuals with neurofibromatosis type 1 (NF1) develop tumors along the anterior visual pathway, involving optic nerves, optic chiasm and optic tracts, known as NF1-associated optic pathway gliomas (NF1-OPGs). This tumor is mainly diagnosed in children younger than seven years, which could lead to a lifelong disability.

NF1-OPGs often grow extensively along the optic pathway, and surgery is a high-risk treatment option. Consequently, human tumor tissues are rarely available for research.

Why we’re excited

“We are very excited about this research because, if successful, we will provide a strategy to treat patients with NF1-OPGs before visual impairment becomes irreversible,” said Yuan Zhu, Ph.D., scientific director and Gilbert Family Endowed professor at the Gilbert Family Neurofibromatosis Institute and senior investigator at the Center for Cancer and Immunology Research, both part of Children’s National. “We combine the expertise of glioma at the Children’s National and retinal biology at the NIH/NEI.”

The research will combine the synergistic expertise between Zhu on NF1 and OPG using pre-clinical models and Drs. Han-Yu Shih and Wei Li at the National Eye Institute of the National Institutes of Health (NIH/NEI) on retinal biology and immunology.

What’s unique

To shed light on the chemical signaling that occurs in the optical nerve with the presence of gliomas, the research approach will have three aims:

  • Isolate and characterize this abnormally infiltrating inflammatory cells and perform multi-omics experiments, including sophisticated genomic, epigenomic and transcriptomic assays, to study them during OPG initiation and progression.
  • Prevent or alleviate OPG-associated nerve damage, RGC death and vision loss.
  • Develop a novel model using the newly established genetic system to identify signals that induce inflammatory responses.
Drs. Bollard and Hanley

Research into a new way to combat solid tumors earns part of a $25M award

Drs. Bollard and Hanley

Catherine Bollard, M.D., M.B.Ch.B., and Patrick Hanley, Ph.D.

Children’s National Hospital has developed multi-antigen specific T cells that have shown success in pre-clinical models in attacking pediatric solid tumors. Now the promising area of research earned a major boost from the Cancer Grand Challenges — founded in 2020 by the two largest funders of cancer research in the world – Cancer Research UK and the National Cancer Institute in the U.S.

This award supported the foundation of NexTGen, a team of scientists and clinicians with expertise in immunology, proteomics, mathematics and more, across eight institutions in the U.S., U.K. and France. Catherine Bollard, M.B.Ch.B, M.D., director of the Center for Cancer and Immunology Research at Children’s National, and Martin Pule, M.D., clinical associate professor at the University College of London are the co-leads of this effort.

The NexTGen team is one of four Cancer Grand Challenges’ new teams, representing a total investment of $100M to diverse, global teams to take on some of the toughest challenges in cancer research. NexTGen will create a new approach that performs clinical and basic research together to facilitate real-time knowledge exchange from the lab to the clinic and back again.

While the more widely known CAR T-cells have made tremendous progress for patients with B-cell leukemias, lymphomas and other blood cancers, the CAR T-cell field has not made the same impact for adult and pediatric solid tumors.

“A tumor cell is very clever because it tries to hide from the immune system by deleting or down regulating targets that the T cell is directed towards,” said Dr. Bollard.

Dr. Bollard further discusses the importance of having patient voices during the decision-making process in this quest, her hopes for their program and the concept of the combining tumor antigen-specific T cells with CAR-T cells that her team will develop.

Q: Can you explain the NexTGen vision?

A: The overall vision is that we will have developed the next generation of cell therapies to cure children with refractory solid tumors by the end of the five years. It is important to move the field forward, so we wanted to be innovative in our approach to this grand challenge for these children who have no other therapeutic options left.

Q: What are the most three important components of this project?

A: First, science and diplomacy played a significant role in bringing in the right set of investigators from diverse scientific backgrounds. What started as a conversation using the universal language of science, it quickly became an international project to address this complex issue. Second, we worked very hard with our patient advocates during the writing process, and they will be working side by side with the investigators at the bench and clinic. Third, we were the only group to have clinical trials in our proposal starting very early in the grant funding period, which is unprecedented.

Q: Can you describe NexTGen’s research model?

A: From our experience in leukemia, we know that progress is greatly accelerated if discovery occurs hand-in-hand with clinical development. Therefore, unlike classical programs where years of pre-clinical discovery and developmental work is required before the clinical translation, we will take a non-conventional non sequential approach.

Specifically, in the NexTGen Program, clinical development will start early with three cutting-edge clinical studies evaluating engineered T-cell technologies that we have recently developed understanding that there are some questions that can ONLY be answered in the clinic. To that end, clinical and translational data from these clinical trials will be able to feed into and enrich the discovery and pre-clinical science throughout the NexTGen Program in a circular fashion to promote this research program that goes from bedside to bench and back.

Q: How is Children’s National leading the way?

A: Children’s National is leading one of the three clinical trials that combine our non-gene engineered tumor antigen-specific T-cell platform with gene engineered T cells to generate a novel T-cell therapy against relapsed /refractory solid tumors. Combining tumor antigen specific T cells with the CAR T-cell platform represents a novel concept that may have more potency against these hardest to treat tumors in children.

Q: Why is it so important to include the patient voice during the discussion and decision making?

A: Because we are also physicians and scientists, we do not forget the patient and their families. Thus, we have a robust patient advocacy group embedded in this vision. The group will co-develop summaries explaining the challenges NextGen will address, how this will be achieved and how results will be used, with major input in clinical trial design and consent documents as well as key input into how patient tissue samples can be used to facilitate research discoveries. The patient advocacy team will also help find broad representation from multiple geographical locations of advocates with lived experience of different cancer types, including bereaved relatives and cancer survivors. These and many more strategies applied with patient advocacy groups will elevate the call for a broader and accelerated adoption of CAR-T clinical trials to broaden access to all patients.

Q: What excites you most about this?

A: What excites me the most is working with this incredible group of scientists, physicians and patient advocates all with rich and deep expertise who bring together an extensive and diverse knowledge base. The fact that we will be all working together toward a common goal of curing pediatric solid tumors in the next five to 10 years is extraordinarily energizing. This sizeable international collaboration comprises the right talent to get this done. It is also highly exciting to simultaneously have three clinical trials running in parallel with the discovery science and the pre-clinical work. I am extremely optimistic that we will realize NexTGen’s vision to bring next generation engineered T-cell therapies to the routine care of children with solid tumors within a decade.

Dr. Bollard and her laboratory

Catherine Bollard, M.D., M.B.Ch.B., selected to lead global Cancer Grand Challenges team

Dr. Bollard and her laboratory

Cancer Grand Challenges NexTGen team members (left to right): Amy Hont, M.D., AeRang Kim, M.D., Nitin Agrawal, Ph.D., Catherine Bollard, M.D., M.B.Ch.B., Conrad Russell Cruz, M.D., Ph.D., Patrick Hanley, Ph.D., and Anqing Zhang.

A world-class team of researchers co-led by Catherine Bollard, M.D., M.B.Ch.B., director of the Center for Cancer and Immunology Research at Children’s National Hospital, has been selected to receive a $25m Cancer Grand Challenges award to tackle solid tumors in children. Cancer Grand Challenges is a global funding platform, co-founded by Cancer Research UK and the National Cancer Institute in the U.S., that supports a community of diverse, global teams to come together, think differently and take on some of cancer’s toughest challenges.

The Cancer Grand Challenges NexTGen team, co-led by University College London’s Martin Pule, M.D., will be working to develop next-generation cell therapies for children with solid cancers. Cancer is a leading cause of death by disease in children worldwide. Although survival has increased for some pediatric cancers, such as blood cancers, survival for some solid tumors has seen little improvement for more than 30 years. The team hopes to build a much deeper understanding of childhood cancers and develop and optimize novel therapies for children with solid tumors, ultimately hoping to improve survival and diminish the lifelong toxicities often experienced by survivors.

“With our Cancer Grand Challenge, we hope to bring next-generation CAR T-cell therapies to children with solid tumors,” said Dr. Bollard. “What excites me most is the energized, passionate group of people we’ve brought together to take this challenge on. Big problems remain to be addressed, but we believe they can be solved, and that we’re the team to solve them.”

“NexTGen represents crucial and overdue work. It has hope written all over,” said Sara Wakeling, patient advocate on the team and CEO and co-founder of Alice’s Arc, a children’s charity for rhabdomyosarcoma. “NexTGen hopes to transform the way these aggressive solid tumors are treated with less toxic side-effects, giving the children a real chance at growing up and realizing their potential. I’m so proud to be part of this exceptional team of scientists, clinicians and advocates who want to change the story for those diagnosed.”

The NexTGen team unites scientists and clinicians with expertise in immunology, proteomics, mathematics and more, across eight institutions throughout the U.S., U.K. and France. The Children’s National investigators that will also join are:

  • Nitin Agrawal, Ph.D., associate professor in the Center for Cancer and Immunology Research at Children’s National.
  • Conrad Russell Cruz, M.D., Ph.D.,principal investigator for the Program for Cell Enhancement and Technologies for Immunotherapies at Children’s National.
  • Patrick Hanley, Ph.D., chief and director of the cellular therapy program at Children’s National and leader of the Good Manufacturing Practices laboratory.
  • Amy Hont, M.D., oncologist in the Center for Cancer and Immunology Research at Children’s National.
  • AeRang Kim, M.D., oncologist in The Center for Cancer and Blood Disorders at Children’s National.
  • Holly Meany, M.D., oncologist in The Center for Cancer and Blood Disorders at Children’s National.
  • Anqing Zhang, biostatistician in the Biostatistics and Study Methodology Department at Children’s National.

The team, co-funded by Cancer Research UK, the National Cancer Institute and The Mark Foundation for Cancer Research, aims to bring much needed new treatments to children with solid cancers.

The NexTGen team is one of four new teams announced today as part of Cancer Grand Challenges, representing a total investment of $100m to diverse, global teams to take on some of the toughest challenges in cancer research.

“Cancer is a global issue that needs to be met with global collaboration. This investment in team science encourages diverse thinking to problems that have long hindered research progress,” said David Scott, Ph.D., director of Cancer Grand Challenges, Cancer Research UK. “Cancer Grand Challenges provides the multidisciplinary teams the time, space and funding to foster innovation and a transformative approach. NexTGen is one of four newly funded teams joining a scientific community addressing unmet clinical needs across cancer research.”

Find out more

Cancer Grand Challenges supports a global community of diverse, world-class research teams with awards of £20m/$25m to come together, think differently and take on cancer’s toughest challenges. These are the obstacles that continue to impede progress and no one scientist, institution or country will be able to solve them alone. Cancer Grand Challenges teams are empowered to rise above the traditional boundaries of geography and discipline.

Founded by the two largest funders of cancer research in the world – Cancer Research UK and the National Cancer Institute* in the U.S. – Cancer Grand Challenges aims to make the progress against cancer we urgently need. Cancer Grand Challenges currently supports more than 700 researchers and advocates across 10 countries, representing 11 teams are supported to take on 10 of the toughest challenges in cancer research.

The Cancer Grand Challenges NexTGen team, announced June 16, 2022, is taking on the initiative’s Solid Tumours in Children challenge. It is led by Dr. Bollard (Children’s National) and Dr. Pule (University College London), along with 23 co-investigators and 7 patient advocates, and is spread across eight institutions across the U.S., U.K. and France: Cardiff University; Children’s Hospital of Philadelphia; Children’s National Hospital; INSERM; the Institute of Cancer Research; Stanford Medicine; Stanford University; University College London. The Cancer Grand Challenges NexTGen team is funded by Cancer Research UK, the National Cancer Institute in the U.S. and The Mark Foundation for Cancer Research.

*The National Cancer Institute is part of the National Institutes of Health.

 

US News Badges

Children’s National named to U.S. News & World Report’s Best Children’s Hospitals Honor Roll

US News BadgesChildren’s National Hospital in Washington, D.C., was ranked No. 5 nationally in the U.S. News & World Report 2022-23 Best Children’s Hospitals annual rankings. This marks the sixth straight year Children’s National has made the list, which ranks the top 10 children’s hospitals nationwide. In addition, its neonatology program, which provides newborn intensive care, ranked No.1 among all children’s hospitals for the sixth year in a row.

For the twelfth straight year, Children’s National also ranked in all 10 specialty services, with seven specialties ranked in the top 10.

“In any year, it would take an incredible team to earn a number 5 in the nation ranking. This year, our team performed at the very highest levels, all while facing incredible challenges, including the ongoing pandemic, national workforce shortages and enormous stress,” said Kurt Newman, M.D., president and chief executive officer of Children’s National. “I could not be prouder of every member of our organization who maintained a commitment to our mission. Through their resilience, Children’s National continued to provide outstanding care families.”

“Choosing the right hospital for a sick child is a critical decision for many parents,” said Ben Harder, chief of health analysis and managing editor at U.S. News. “The Best Children’s Hospitals rankings spotlight hospitals that excel in specialized care.”

The annual rankings are the most comprehensive source of quality-related information on U.S. pediatric hospitals and recognizes the nation’s top 50 pediatric hospitals based on a scoring system developed by U.S. News.

The bulk of the score for each specialty service is based on quality and outcomes data. The process includes a survey of relevant specialists across the country, who are asked to list hospitals they believe provide the best care for patients with the most complex conditions.

The seven Children’s National specialty services that U.S. News ranked in the top 10 nationally are:

The other three specialties ranked among the top 50 were cardiology and heart surgerygastroenterology and gastro-intestinal surgery, and urology.

illustration of the brain

LIFU successfully delivers targeted therapies past the blood-brain barrier

illustration of the brain

LIFU offers doctors the first opportunity to open the blood-brain barrier and treat the entire malignant brain tumor.

Children’s National Hospital will leverage low-intensity focused ultrasound (LIFU) to deliver therapy directly to a child’s high-grade glioma. The approach offers doctors the first opportunity to open the blood-brain barrier and treat the entire malignant brain tumor.

Children’s National will be the first hospital in the U.S. to treat high-grade pediatric brain tumors with LIFU to disrupt the blood-brain barrier. Crossing it has been a major hurdle for effective therapy. The barrier, a network of blood vessels and tissue, prevents harmful substances from reaching the brain but also stops molecular targeted therapy and immunotherapy from getting into the tumor site and staying there.

“LIFU gives us a way to potentially transiently open up the barrier, so we can deliver novel therapy directly to the tumor and improve the likelihood of survival,” said Roger Packer, M.D., senior vice president of the Center for Neurosciences and Behavioral Medicine at Children’s National. “It is the greatest breakthrough we’ve potentially had in the past 50 years or more for the management of these tumors. We made great strides in our understanding of molecular genetics and the molecular drivers of tumors, but we have not yet translated that knowledge into better therapies; this may be our most effective mechanism to overcome the barrier.”

In 2020, Children’s National was recognized as the first worldwide Center of Excellence by the Focused Ultrasound Foundation.

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.

How it works

Doctors at Children’s National will be using LIFU in two different types of procedures:

  • 5-ALA: Doctors will give the patient 5-aminolevulinic acid (5-ALA) with the LIFU treatment. 5-ALA enters rapidly dividing cells and is activated by the ultrasound to a state where it kills the dividing cells of the tumor. The surrounding normal brain cells around the tumor are not dividing, so they do not take up the 5-ALA and are left unharmed after ultrasound therapy.
  • Microbubbles: While receiving different doses of LIFU over a one- to two-hour period, the patient is given “microbubbles,” which are widely used in medical imaging and as carriers for targeted drug delivery. These microbubbles bounce around against the walls like seltzer, opening the blood vessels and transiently opening that space.

Both studies are the first in the world for pediatric gliomas of the brain stem, allowing experts to treat patients 4-6 weeks after radiotherapy. The patient then receives medication orally or intravenously as it passes through the bloodstream. It does not go at high levels anywhere within the brain except where the blood-brain-barrier was opened, allowing oral medication or immune therapies to rush into the tumor.

The launch of this program comes a few months after the hospital successfully performed the first-ever high-intensity focused ultrasound surgery on a pediatric patient with neurofibromatosis.

Watch this video to learn more.

mother and daughter embracing

Understanding end-of-life treatment preferences for adolescents

mother and daughter embracing

FACE-TC effectively increases communication between adolescents with cancer and their families about the patients’ preferences.

Talking about death and dying is taboo. Some families believe it is their role alone to make end-of-life healthcare decisions or they may believe pediatric advance care planning is against their religion.

In a recent trial, Maureen Lyon, Ph.D., a clinical health psychologist at Children’s National Hospital and lead author of the study, analyzed the value of high-quality pediatric advance care planning and how this enabled families to know their adolescents’ end-of-life treatment preferences.

This is the first fully powered randomized controlled trial to focus on adolescents with cancer and their engagement with their families in pediatric advance care planning conversations.

What this means

Some physicians believe it is not their role to discuss the “what ifs.” Others report that they do not have the training or time to do so. As a result, in clinical practice, adolescents living with a serious illness rarely have documented advance care plans. The default is to provide intensive treatments that potentially increase suffering.

“Despite cancer being the leading cause of disease-related death in adolescents, conversations about goals of care and documentation of end-of-life care and treatment preferences for adolescents with cancer are not a routine and standard part of care,” Dr. Lyon said.

Why it matters

Family-centered advance care planning for teens with cancer (FACE-TC) effectively increases communication between adolescents with cancer and their families about the patients’ end-of-life preferences. This meets the first challenge of pediatric advance care planning – knowledge of patient’s preferences.

This low-tech intervention commits to more deeply respecting adolescents with cancer, integrating them into health care decision-making and giving them some control in a low control situation.

The patient and family benefits

“FACE-TC strengthens communication between adolescents with cancer and their families about adolescents’ understanding of their illness, their hopes and fears, their goals of care and their end-of-life treatment preferences,” Dr. Lyon added. “With increased access to palliative care services and pediatric advance care planning, families may better understand that stopping intensive medical interventions when their child is dying is not giving up, but rather choosing how best to spend the final days of one’s life.”

Dr. Lyon and the team at Children’s National have pioneered this effort to give seriously ill adolescents a voice and help families break the ice so they know what their child would want if the worst were to happen. The team also aims to provide an extra level of support for busy clinicians so the first conversation about goals of end-of-life care does not happen in the intensive care unit.

You can read the full trial, An Intervention in Congruence for End-of-Life Treatment Preference: A Randomized Trial, in Pediatrics.

You can also read the last manuscript from this clinical trial, Effect of the Family-Centered Advance Care Planning for Teens with Cancer Intervention on Sustainability of Congruence About End-of-Life Treatment Preferences, in JAMA Network.

girl hugging stuffed animal

Developing next-generation T cells to fight cancer

girl hugging stuffed animal

In the last decade, researchers have witnessed significant advances in the immunotherapy field. Most recently, a study in Nature claimed a novel CAR T-cell therapy “cured” a patient.

In the last decade, researchers have witnessed significant advances in the immunotherapy field. Most recently, a study in Nature claimed a novel CAR T-cell therapy “cured” a patient. Given the landmark scientific achievement for patients with different types of leukemia and lymphoma, Children’s National Hospital experts chimed in on the technology they have developed beyond CAR T cells.

Catherine Bollard, M.D., M.B.Ch.B., director of the Center for Cancer and Immunology Research at Children’s National Hospital, discusses the implications of this research, how it relates to the work she’s doing at Children’s National and the future of T-cell therapies.

Q: What did the research published in Nature find?

A: It reported a decade-long experience with this novel T-cell therapy called CD19 CAR T cells. These were used to treat patients with a type of leukemia or lymphoma that expresses the CD19 on its surface. While the article reported the experience of Children’s Hospital of Philadelphia and the University of Pennsylvania, multiple groups throughout the country did similar trials that have used these unique CD19 CAR T cells to treat children and adults with these refractory blood cancers.

Q: What are your thoughts on the implications of this research?

A: We now have three FDA-approved commercial CD19 CAR T-cell products developed by several academic institutions. This is revolutionary for our patients who have B-cell leukemias and lymphomas. It’s incredibly exciting for our T-cell therapy field in general because this was the first time the FDA approved a T-cell therapy. What it means now is the field is extremely excited to develop similar effective therapies for other patients with cancer.

Q: How does this relate to your work at Children’s National?

A: While CAR T cells have made tremendous progress for patients with B-cell leukemias, lymphomas and other blood cancers, the CAR T-cell field has not made the same impact for adult and pediatric solid tumors. We think the field is going to expand the type of T-cell therapies we’re generating beyond just CAR T cells. That’s where the work we’re doing comes in – not only by developing new T cells that don’t need gene engineering but also T cells that can be used as a platform for next-generation engineering approaches. We think the technology we’ve developed at Children’s National will help make an impact, especially in the solid tumor space. I hope in the next 10 years, we’ll be having a conversation not just about CAR T cells, but about other types of T cells that are now making an impact for solid tumors.

Q: How are the CAR T cells you develop different than those in the Nature article?

A: We think our multi-antigen specific T cells are complimentary and could have more potency than conventional CAR T cells for solid tumors especially when used in combination. This is in part because they can identify multiple targets on a tumor cell. Tumor cells are very clever and try to hide from T-cell therapies by down regulating the target that the T cell is directed towards. However, our novel T-cell therapies get around that escape by targeting multiple targets in a single product, making it much harder for the cancer cell to hide from the immune attack by the T cells.

Additionally, we’re excited by our approach because not all of our products require gene engineering, unlike CAR T cells. We have effectively used our T cells to target viruses in the “off-the-shelf” setting and we’re now about to start a first human clinical trial at Children’s National using an off the shelf T-cell product for children with solid tumors. It makes the T-cell therapy more like an “off-the-shelf” drug therapy that will allow us to treat many more children and adults nationally, as well as we hope, internationally.

doctor and cancer patient smiling

Manufactured leukemia-specific T cells may help increase survival rates

doctor and cancer patient smiling

Infusion of a novel, multi-targeted donor-derived T-cell therapy is safe and well-tolerated in patients with high-risk or relapsed leukemia after a donated bone marrow transplant, according to a new study published in Blood Advances.

Infusion of a novel, multi-targeted donor-derived T-cell therapy is safe and well-tolerated in patients with high-risk or relapsed leukemia after a donated bone marrow transplant, according to a new study published in Blood Advances. The findings suggest that this strategy may make a difference in these patients, as will be evaluated in later phase trials.

“A tumor cell is very clever because it tries to hide from T-cell therapies by deleting or down regulating targets that the T cell is directed towards,” said Catherine Bollard, M.D., M.B.Ch.B., director of the Center for Cancer and Immunology Research at Children’s National Hospital and co-senior author. “This novel cell therapy has the potential to get around that escape by targeting multiple proteins in a single product, making it much harder for the cancer cell to hide from the immune attack by the T cells.”

The tumor-associated antigen-specific T cell (TAA-T) product targets WT1, survivin and PRAME, which are proteins that play a role in cancer cell proliferation and survival. They are overexpressed in leukemia and many other human malignancies. The researchers chose to expand the T cells to target many malignancies through at least one expressed antigen. The manufactured TAA-T products are derived from peripheral blood mononuclear cells (PBMCs) obtained from the patient’s own BMT donor.

The hold-up in the field

Conventional therapies for patients with high-risk or relapsed malignancies often fail due to toxicity associated with additional chemotherapy and second transplant, particularly in those who relapse early after transplant. This novel cellular immunotherapy approach is shown to be safe and targets antigens that are found in CD19 positive and negative blood cancers, which may broaden the applicability to other cancer types, such as acute myeloid leukemia, that are currently lacking effective T cell therapy options.

What’s next

“Evaluation and tracking of unique T cell receptor clonotypes in patients following TAA-T cell infusion demonstrated expansion and persistence of some clonotypes up to 6 months to one-year post-infusion,” said Hannah Kinoshita, M.D., oncology fellow at Children’s National and co-lead author. “In future studies, we are hoping to identify and track unique target antigen-specific clonotypes from the T cell product infused to better understand the immunobiological effect of the infused T cells and how that can be translated into improved clinical outcomes.”

Children’s National Hospital leads the way

The Cell Enhancement and Technologies for Immunotherapy (CETI) program at Children’s National specializes in developing and analyzing novel cellular therapeutics such as this one.

You can read the full study “Outcome of Donor-derived TAA-T cell therapy in Patients with High-risk or Relapsed Acute Leukemia Post Allogeneic BMT,” in Blood Advances. Children’s National researchers worked in partnership with Rick Jones, M.D., co-senior author and Kenneth Cooke, M.D., Ph.D., co-lead author, both at Johns Hopkins Medicine.

Illustration of white blood cells attacking a cancer cell

Alpha/beta T cell depletion lifts barriers to transplantation

Illustration of white blood cells attacking a cancer cell

Removal of A/B T cells from the infused cell product significantly minimizes the risk of GvHD and eliminates the need for immunosuppressive medications after transplant.

Alpha/beta T cell depletion (A/B TCD) is a cutting-edge hematopoietic stem cell transplant (HSCT) technique by which donor derived immune cells, called A/B T cells, can be removed by selectively using magnetic beads before the donor cells are infused into the recipient’s body. A/B T cells have the potential to cause life threatening inflammation in the recipient’s body, called graft-versus-host disease (GVHD). GVHD is a major complication after transplant, especially when the donor is not fully matched. Therefore, removal of A/B T cells from the infused cell product (graft) significantly minimizes the risk of GVHD and eliminates the need for immunosuppressive medications after transplant.

Unlike previous methodologies that completely remove all immune cells, the novel A/B TCD approach preserves beneficial immune cells (like gamma delta T cells, natural killer cells, monocytes and dendritic cells) in the graft to preserve the capability to fight viral infections and residual cancer. Therefore, this innovative transplant approach can cure leukemia while decreasing the risk of life threatening infections and relapse after transplant.

In this Q&A, Anant Vatsayan, M.D., blood and marrow transplant specialist at Children’s National Hospital, tells us more about this new exciting technique.

Q: What is the specific research question that you are hoping to answer?

A: Children’s National Hospital is participating in the largest multicenter pediatric trial of A/B TCD hematopoietic stem cell transplant in the United States. The primary objective of this research is to assess whether disease-free survival at one-year after-HCT for children with high-risk leukemia and myelodysplastic syndrome can be improved with A/B TCD hematopoietic stem cell transplant.

Patients with other types of blood disorders may also be eligible to undergo A/B TCD hematopoietic stem cell transplant in this study based on the discretion of the principal investigator. The study will also assess the overall survival and rates of acute and chronic GVHD. Another objective is to compare the cost of transplantation using half-matched (haploidentical) donors versus other stem cell sources (for example, matched unrelated adult donors or cord blood donors) at participating centers.

Q: Why is this work exciting?

A: A/B TCD hematopoietic stem cell transplant has several benefits:

  • One of the remarkable benefits of this technique is the possibility of using haploidentical related donors for transplant if a fully matched related or unrelated donor is not available. This is a common scenario for patients of certain races (African American) and ethnicities (Hispanic) where it is difficult to identify a fully matched unrelated donor. Therefore, A/B TCD hematopoietic stem cell transplant expands the pool of donor options and ensures more equitable donor availability across every race and ethnicity.
  • A/B TCD significantly decreases the risk of severe GVHD and post-transplant infections. It eliminates the need for post-transplant immunosuppressive medications (like cyclosporine, tacrolimus or sirolimus) that can have numerous side effects and require frequent monitoring of drug levels in the blood.
  • The A/B TCD technique also promotes faster recovery of blood counts (engraftment) after transplant. Therefore, patients take fewer medications, have shorter durations of hospitalization for transplant and need less frequent blood tests and clinic visits after transplant. Hence, this patient friendly and family centric transplantation strategy will ensure that patients can spend more time with their family and have a better quality of life.

Q: How do you hope this will benefit patients?

A: Alpha/beta T cell depleted HSCT using half matched (haploidentical) donors will ensure donor availability for almost every patient regardless of race/ethnicity and probability of finding a matched related/unrelated donor. This methodology has tremendous prospects for wider applications, including the use of matched related and unrelated donors with the intent to eliminate the need for post-transplant immunosuppressive medications. This could be especially beneficial for patients with Fanconi anemia or other patients who are at risk of developing severe side effects from the use of immunosuppressive medications.

Q: How unique is this work?

A: The Shirley and William Howard Cellular Therapy Laboratory Stem Cell Processing program processes stem cells and performs cutting edge clinical trials while providing innovative care for patients. This work benefits from access to CliniMACS Plus Cell Selection Device, along with a multidisciplinary team with laboratory and clinical expertise to perform A/B TCD hematopoietic stem cell transplant. Access to our state of the art Cellular Therapy Laboratory allows us to further complement this transplantation strategy with other cellular therapies after transplant, such as virus specific and leukemia targeting T cells, which further mitigate the risk of post-transplant viral infections and leukemia relapse.

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.

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.

Meghan Delaney

Meghan Delaney, D.O., M.P.H., receives 2021 James Blundell Award

Meghan Delaney

The British Blood Transfusion Society (BBTS) recognized Meghan Delaney, D.O., M.P.H., division chief of Pathology and Lab Medicine and director of Transfusion Medicine at Children’s National Hospital, with the James Blundell Award.

The James Blundell Award is the most prestigious award given by the BBTS. Introduced in 1984, this award is given to recipients for original research resulting in an important and significant contribution to the body of medical and or scientific knowledge within the field of blood transfusion.

“I am honored to receive this award from the British Blood Transfusion Society,” says Dr. Delaney. “The field of transfusion medicine is dynamic and provides an important part of our healthcare infrastructure.  It’s wonderful to have this recognition and I am grateful to the society for the award.”

She is professor of Pathology and Pediatrics at the George Washington University with over 100 manuscripts, 25 book chapters and over 60 presented abstracts. Dr. Delaney serves as a member of the Board of Directors for the Association for the Advancement of Blood & Biotherapies (AABB), is a Scientific Member of the BEST Collaborative and is a member of the American Board of Pathology Test Development and Advisory Committee. She serves as the chair of the National Institutes of Health’s BLOODSAFE Program that aims to improve access to safe blood in Sub Saharan Africa.

Dr. Delaney received the award on September 14 at the 2021 BBTS Annual Conference, where she presented the lecture titled: “Access to safe blood transfusion in low- and middle-income nations: From ‘big data’ to mosquitos.”

Hyundai Hope on Wheels Logo

Oncologists receive Hyundai Hope on Wheels grants

Hyundai Hope on Wheels Logo

Keri Toner, M.D., and Hannah Kinoshita, M.D., both oncology researchers at Children’s National Hospital, were recently awarded Hyundai Hope on Wheels cancer research grants.

Dr. Toner, who is an attending physician in the Center for Cancer and Blood Disorders and the Center for Cancer and Immunology Research at Children’s National, received a $300,000 Hyundai Scholar Hope Grant that she will use to develop and functionally evaluate a novel T cell therapy which can be translated to the clinic for treatment of pediatric patients with acute myeloid leukemia (AML).

Currently, patients with relapsed AML have very poor outcomes and the success that T cell therapy has had in treating B-cell malignancies has not yet been achieved for AML. Dr. Toner’s goal is to try to overcome some of these barriers with a novel T cell therapy which combines both native and chimeric T cell receptors to target AML.

“There are currently critical barriers to the success of T cell therapies for the treatment of AML,” Dr. Toner explains. “Successful completion of this research would allow for translation of a novel CAR-TAA-T therapy to the clinic for the treatment of relapsed/refractory AML, which has very poor prognosis.”

Meanwhile, Dr. Kinoshita, a pediatric hematology oncology fellow at Children’s National, received a $200,000 Hyundai Young Investigator Grant. She will use the funds to evaluate the immunobiology of multi-antigen specific T cell therapy infused to patients to reduce the two most common causes of morbidity and mortality following hematopoietic stem cell transplant (HSCT) for malignant disease: relapse and infection.

The administration of multiantigen specific T cells targeting tumor and viral-associated antigens following stem cell transplant may serve to prolong remission of malignant disease and prevent and treat viral infections that can cause devastating disease in children. Dr. Kinoshita’s study will evaluate the anti-viral and anti-leukemia immune response in vivo following targeted T cell therapy.

“There have been incredible advancements in the field of pediatric oncology and bone marrow transplant over the past 20-30 years but there are still many areas in which we need to continue to improve,” Dr. Kinoshita says. “Our patients and their families go through so much to get into remission and it is devastating if they relapse or develop severe infectious complications. Adoptive immunotherapy is a promising tool in aiding to treat and prevent these complications, particularly for patients with high-risk hematologic malignancies.”

The Hyundai Scholar Hope Grants and the Hyundai Young Investigator Grants are competitive research grants that are peer-reviewed by the Hyundai Hope on Wheels Medical Advisory Committee, which is comprised of leading pediatric oncologists from children’s hospitals and research institutions nationwide. The grants are open to U.S.-based Children’s Oncology Group member institutions.

Epstein-Barr virus

Study with largest cohort in the Western world sheds light on Epstein-Barr virus

Epstein-Barr virus

Epstein-Barr virus is a member of the herpes family and it spreads primarily through saliva.

Children’s National Hospital experts provided a contemporary description of the epidemiology, clinical presentation and management of chronic active Epstein-Barr virus (CAEBV), shedding light on this very rare disease. The paper, published in Blood Advances, assessed 57 patients outside of Asia — the biggest international retrospective cohort study published in the Western world.

Epstein-Barr virus is a member of the herpes family and it spreads primarily through saliva. Once a person is infected with Epstein-Barr virus, the immune system will control the infections, but the virus lies in a dormant state in the patient’s B Cells. However, in some patients, there is a failure of the body to control the infection, and the virus is found inside the patient’s T and/or NK cells. These rare patients are diagnosed with CAEBV. The hallmark of the disease is proliferation of Epstein-Barr virus-infected T or NK cells that infiltrate tissues, leading to end-organ damage. Patients most often experience fevers, hepatosplenomegaly, liver inflammation, cytopenias and lymphoproliferation that may progress to lymphoma.

Given it is most prevalent in Asia, little is known about the disease in the Western world. There has only been one published paper regarding the outcomes of patients in the U.S., which included 19 patients amassed over 28 years, and was published a decade ago.

Multiple treatments have been attempted to control the disease, but none have resulted in consistent remission. Historically, the consensus is to use steroids and/or antiviral drug in combination with proteasome inhibitor agents. In some cases, clinicians also use cytotoxic chemotherapy to reduce disease burden and improve the patient’s condition before HSCT. Still, this approach is limited because most patients die due to the progression of their disease despite these interventions.

Ultimately, most of these patients are referred for allogeneic hematopoietic stem cell transplantation (HSCT), which is the only known curative therapy for CAEBV. However, the best approach to control disease prior to HSCT, as well as the optimal conditioning regimen, are unknown.

“For the first time in many years, we provide insight on contemporary treatment options to consider for patients with CAEBV, as well as identifying risk factors for worse outcomes,” said Blachy Dávila Saldaña, M.D., blood and marrow transplant specialist at Children’s National and lead author of the study. “HSCT is curative, but patients need to be considered prior to the evolution of more advanced disease, particularly lymphoma. We also provide a new platform that will inform research on new interventions and therapies for this population.”

“CAEBV remains a challenging disorder to treat, especially once severe complications develop,” said Catherine Bollard, M.D., M.B.Ch.B., director of the Center for Cancer and Immunology Research at Children’s National. “However, our data suggests that T cell modulating therapies may enhance disease control, and future studies should address this question in a controlled setting.”

Future steps also include performing genetic studies to identify those at risk of developing the disease, and developing new platforms for treatment, including checkpoint inhibitors and cytotoxic lymphocyte therapies (CTL’s), which is a form of adoptive immunotherapy that employs virus-specific T cells.

The cohort includes patients treated in CNH and multiple institutions around the world, including Texas Children’s and the National Institutes of Health. “This work was only possible through our collaborative research in anti-EBV cellular therapies,” said Dr. Dávila.

t cells fighting cancer cell

Personalized T cell immunotherapy for brain tumors closer to becoming reality

t cells fighting cancer cell

Children’s National Hospital experts developed a new approach that discovered unique proteins in an individual tumor’s cells, which then helped scientists generate personalized T cells to target and kill tumors.

Children’s National Hospital experts developed a new approach that discovered unique proteins in an individual tumor’s cells, which then helped scientists generate personalized T cells to target and kill tumors, according to a pre-clinical study published in Nature Communications.

This effort is the first to create a new workflow for neoantigen identification that incorporates both genetic sequencing and protein identification to create a personalized treatment for medulloblastoma in children, a common malignant brain tumor. Given these promising findings, the researchers are now designing a phase I clinical trial slated to open in 12-18 months.

“This work is an incredibly exciting advancement in personalized medicine. It will allow us to treat patients with a novel T cell therapy that is developed for each individual patient to specifically attack and kill their tumor,” said Catherine Bollard, M.D., M.B.Ch.B., director of the Center for Cancer and Immunology Research at Children’s National and co-author on the paper. “This treatment will offer a potential option for children with hard-to-treat brain tumors for which all other therapeutic options have been exhausted.”

Catherine Bollard

Catherine Bollard, M.D., M.B.Ch.B., director of the Center for Cancer and Immunology Research at Children’s National and co-senior author on the paper.

First, the researchers sequenced the DNA of small tissue samples while studying its complete set of proteins that influence cancer biology — also named a “low-input proteogenomic approach” by the authors. After analyzing the empirical data, which shies away from the commonly used predictive models, the researchers developed a T cell immunotherapy that targets the tumor’s unique proteins and allows the T cells to distinguish between healthy cells and tumor cells. This means that Rivero-Hinojosa et al. managed to merge two research fields, proteogenomics and immunotherapy, and lay the groundwork for personalized, targeted T cell therapies to treat children with brain tumors.

“Neoantigen discovery techniques have either been dependent upon in silico prediction algorithms or have required a significant amount of tumor tissue, making them inappropriate for most brain tumors,” said Brian Rood, M.D., medical director of Neuro-oncology and the Brain Tumor Institute at Children’s National. “This neoantigen identification pipeline creates a new opportunity to expand the repertoire of T cell-based immunotherapies.”

Tumor cells have damaged DNA that create mutations during the repair process because they do not do a good job at maintaining their DNA fidelity. The repairs therefore create aberrant DNA that codes for proteins that were never intended by the genetic code and, consequently, they are unique to the individual’s tumor cells.

Brian Rood

Brian Rood, M.D., medical director of Neuro-oncology and the Brain Tumor Institute at Children’s National and co-senior author on the paper.

“We developed a new filtering pipeline to remove non-annotated normal peptides. Targeting antigens that are completely specific to the tumor, and expressed nowhere else in the body, will potentially increase the strength of tumor antigen-specific T cell products while decreasing the toxicity,” said Samuel Rivero-Hinojosa, Ph.D., staff scientist at Children’s National and first author of the study.

Once the experts identified these unique peptides, they used them to select and expand T cells, which showed specificity for the tumor specific neoantigens and the ability to kill tumor cells. The next step is to conduct a clinical trial in which a patient’s own T cells are trained to recognize their tumor’s unique neoantigens and then reinfused back into the patient.

From an immunotherapy standpoint, tumor specificity is important because when clinicians treat patients with T cell therapies, they want to make sure that the T cells directly target and kill the tumor and will not cause devastating harm to healthy cells. This paper demonstrated that it may be possible to create a better efficacy and safety margin with this new approach.

In the past five years, under the leadership of Dr. Bollard, the Center for Cancer and Immunology Research at Children’s National has advanced the scientific knowledge in preclinical and clinical settings. The center discovered a signaling pathway that can be hijacked to prevent brain tumor development, and further advanced translational research with several key first-in-human studies that utilized novel cell therapies to treat cancer and life-threatening viral infections.

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.

Jeffrey Dome

Q&A with Dr. Jeffrey Dome on his new role as Continental President of SIOP-North America

Jeffrey Dome

In March 2021, Jeffrey Dome, M.D., Ph.D., senior vice president of the Center for Cancer and Blood Disorders at Children’s National Hospital, was elected as the International Society of Paediatric Oncology’s (SIOP) Continental President of North America.

In March 2021, Jeffrey Dome, M.D., Ph.D., senior vice president of the Center for Cancer and Blood Disorders at Children’s National Hospital, was elected as the International Society of Paediatric Oncology’s (SIOP) Continental President of North America.

On October 21-24, the society will hold its 53rd SIOP Annual Congress virtually. During the congress, Dr. Dome will begin his 3-year term as SIOP continental president of North America and will also chair and speak at an educational symposium on current approaches to the treatment of recurrent Wilms tumor.

Dr. Dome attended his very first SIOP meeting in 2005 and was captivated by how regional context influences pediatric cancer treatment. In 2017, he was chair of the local organizing committee for the 49th annual congress in Washington, D.C., and served on the SIOP Board of Directors.  After 15 years of involvement and attending many of the annual meetings, Dr. Dome shares what he looks forward to while serving as continental president of SIOP North America and the legacy he hopes to leave behind.

  1. What does it mean to you to be elected SIOP continental president of North America?

I’m very excited about this role. There are several important societies and organizations in North America that have made a mark on the field of pediatric oncology, but SIOP is unique in its sole focus on childhood cancer and global approach to improving outcomes, as encapsulated by its vision statement: No child should die of cancer: cure for more, care for all.

  1. What excites you most about this role?

In an eye-opening statistic, North America has only about 10% of the global burden of childhood cancer and less than 2% of worldwide childhood cancer deaths.  Although we relentlessly strive to improve childhood cancer outcomes in the United States, what we experience here is just the tip of the iceberg of the worldwide problem. SIOP seeks to make a difference on the international level by improving education, research and access to care for children with cancer around the world. And I’m excited to have a platform to lead North American ambassadors to do that.

Even though North America has a relatively small fraction of the overall childhood cancer cases, we are one of the most well-resourced continents. The question is, how can we use our knowledge, technology and resources to help the rest of the world.  A big part of this role is to make connections and liaisons to move the needle on improving outcomes.

The other thing we’ve learned from a research standpoint is that pediatric cancers are relatively uncommon and are becoming even rarer through molecular classification, which divides cancers into small genetically defined subgroups.  While these advances are tremendously exciting, they require international collaboration to amass a sufficient number of patients to evaluate novel treatment strategies.  My vision for SIOP North America is to be a convener of researchers and connect people around the world to facilitate that work.

  1. What are some of your goals while serving as continental president?

We recently sent a survey to more than 450 SIOP North America members and had a nearly 45% response rate, which I’m told is superb.  This speaks to an excellent level of engagement in SIOP’s mission, with many members volunteering to participate in committees related to research, advocacy and global health. The majority of the respondents to the survey were physicians but improving childhood cancer treatment takes a holistic approach.  One of my main goals is to increase SIOP North American membership to grow the number of nurses, pharmacists, scientists, psychologists, other behavioral health specialists and clinical research coordinators onboard.

I’d like to also identify two to three very specific projects that will impact pediatric cancer care on a global level. There are different ways to do that. We could improve education in different areas around the world (nursing education that we provide to areas that are lacking nursing support, for example). It could be research education and database education for regions of the world that would like to develop more robust research programs. It can also be medical support and developing medical guidelines for oncologists around the world that are adjusted to different levels of resources that are available.

The other goal would be to enhance supportive care and education for cancer care delivery on the global level.

  1. Why is this work important for you?

One of my mentors from when I was a junior faculty member advised me that to be a well-rounded oncologist, one must be familiar with how childhood cancer is treated around the world because different regions have different approaches. There is something to be learned from everyone.  I took that advice to heart and have tried to look beyond the North American approaches.  I think it’s very important to have a global exchange of ideas and serving as continental president of SIOP-North America will enable more to facilitate this dialogue.

  1. What’s the legacy or impact you hope to leave behind?

SIOP North America has a strong and devoted membership but has largely functioned at the level of the individual members.  I hope to bring more structure to the organization to tackle the global challenges of childhood cancer treatment.

Once this structure is in place, I hope to complete two or three SIOP-North America initiatives that have a measurable impact on improving childhood cancer care delivery or outcomes.  The specific projects have yet to be defined but will likely be in the categories of enhancing education, supportive care and facilitating research infrastructure. There’s so much to tackle that if you just look at the overall problem of childhood cancer, it’s overwhelming.  We’re not going to be able to solve everything in three years, but if we could have a few victories and be able to move the needle in some areas, I think that would be a huge success.

close-up of two people holding hands

Effective palliative end-of-life care for Spanish-speaking teens with cancer

close-up of two people holding hands

Despite research showing how vital advance care planning conversations are between adolescents with cancer and their families, the efficacy of pediatric advance care planning has not been studied in Hispanic adolescents living with cancer.

Pediatric advance care planning has positively impacted English-speaking adolescents with cancer and HIV, but it has not been studied in other populations — exacerbating health disparities. In a new study published in Clinical Practice in Pediatric Psychology, Maureen E. Lyon, Ph.D., lead author and clinical health psychologist at Children’s National Hospital, and other experts look to adapt and refine the evidence-based Family-Centered Advance Care Planning for Teens with Cancer (FACE®-TC) for Spanish-speaking adolescents with cancer. Using a community-based participatory approach and key informant interviews with patients and families, the experts identified important themes and outcomes.

Despite research showing how vital advance care planning conversations are between adolescents with cancer and their families, the efficacy of pediatric advance care planning has not been studied in Hispanic adolescents living with cancer. According to the authors, this creates a health disparity as Hispanic adolescents with cancer and their families do not have access to or provision of this potentially beneficial service.

“If successfully adapted, FACE-TC Spanish would benefit patient’s communication with their families about their end-of-life treatment preferences if the worse were to happen and they could not communicate,” Dr. Lyon said. “It could also increase families’ positive appraisal of their caregiving and increase communication about goals of care with treating physicians, so the first conversation about goals of care is not in the intensive care unit.”

The study’s findings showed that first-generation Spanish-speaking individuals living in the Washington D.C., Maryland and Virginia area wanted community education about advance care planning for Spanish-speaking adults, many of whom were unfamiliar with the concept.

These findings, in turn, showed the need for future research to include informational messages on Hispanic radio stations, educational workshops or radionovelas.

“We learned that fear of deportation meant that potential participants only felt safe to participate while at the hospital,” Dr. Lyon added. “Because of COVID-19, this was not feasible during the study period. There was consensus that families should be involved in the conversations and that the goals of care conversations and advance directives should be communicated to the physician.”

While this is the first study to adapt a family-centered approach to pediatric advance care planning for Spanish-speaking teens with cancer and their families, it is consistent with cultural values of ‘familismo’ (family) and ‘respecto’ (respect).