Activating adult-born neurons through deep brain stimulation alleviates Alzheimer’s symptoms in rodent models

Source: Cell Press
Date: 4/6/2023
Link to original
Image of article

People with Alzheimer’s disease develop defects in cognitive functions like memory as well as problems with noncognitive functions that can lead to anxiety and depression. In a paper published April 6 in the journal Cell Stem Cell, investigators used mice to study a process through which new neurons are generated in adulthood, called adult hippocampus neurogenesis (AHN). The research showed that deep brain stimulation of new neurons helped restore both cognitive and noncognitive functions in mouse models of Alzheimer’s disease.

“We were surprised to find that activating only a small population of adult-born new neurons was enough to make a significant contribution to these brain functions,” says senior author Juan Song, an associate professor at the University of North Carolina at Chapel Hill. The neurons were modified by deep brain stimulation of the suprammamillary nucleus (SuM), which is located in the hypothalamus. “We are eager to find out the mechanisms that underlie these beneficial effects,” Song says.

This research used two distinct mouse models of Alzheimer’s. The investigators used optogenetics to stimulate the SuM and enhance AHN in Alzheimer’s mice. Their earlier research had shown that stimulation of the SuM could increase the production of new neurons and improve their qualities in normal adult mice. In the new study, the investigators showed that this strategy was also effective in the Alzheimer’s mice, leading to the generation of new neurons that made better connections with other parts of the brain.

However, having more improved new neurons is not enough to improve memory and mood. Behavioral improvement in Alzheimer’s mice were seen only when these improved new neurons were activated by chemogenetics. The researchers used memory tests as well as established assessments to look for anxiety-like and depression-like behavior to confirm these improvements. The results suggested that multi-level enhancement of new neurons — enhancement in number, properties, and activity — is required for behavioral restoration in Alzheimer’s brains.

To further understand the mechanism, they also analyzed the protein changes in the hippocampus of Alzheimer’s mice in response to activation of SuM-modified adult-born new neurons. They found several well-known protein pathways activated inside cells, including those known to be important for improved memory performance, as well as those that allow clearance of the plaques related to Alzheimer’s.

“It was striking that multilevel enhancement of such a small number of adult-born new neurons made such a profound functional contribution to the animals’ diseased brains,” Song says. “We were also surprised to find that activation of SuM-enhanced neurons promoted the process that can potentially remove plaques.”

Future efforts of the team will focus on developing potential therapeutics that mimic the beneficial effects mediated by activation of SuM-modified new neurons. “We are hoping these drugs could exert therapeutic effects in patients with low or no hippocampal neurogenesis,” Song says. “Ultimately, the hope is to develop first-in-class, highly targeted therapies to treat Alzheimer’s and related dementia.”

Research in mice identifies neurons that control locomotion

Source: Cell Press
Date: 1/20/2022
Link to original
Image of article

For more than a century, scientists have known that while the commands that initiate movement come from the brain, the neurons that control locomotion once movement is underway reside within the spinal cord. In a study published January 20 in the journal Cell, researchers report that, in mice, they have identified one particular type of neuron that is both necessary and sufficient for regulating this type of movement. These neurons are called ventral spinocerebellar tract neurons (VSCTs).

“We hope that our findings will open up new avenues toward understanding how complex behaviors like locomotion come about and give us new insight into the mechanisms and biological principles that control this essential behavior,” says the paper’s senior author George Mentis, associate professor of pathology and cell biology in the Department of Neurology at Columbia University. “It’s also possible that our findings will lead to new ideas for therapeutic avenues, whether they involve treatments for spinal cord injury or neurodegenerative diseases that affect movement and motor control.”

VSCTs were discovered in the 1940s, but researchers have long believed that their main function was to relay messages about neuronal activity from the spinal cord to the cerebellum. The new study reports that instead they control locomotor behavior both during development and in adulthood.

“These findings were a huge surprise,” Mentis says. “One of the key discoveries in our study was that apart from their connection to the cerebellum, these neurons make connections with other spinal neurons that are also involved in locomotor behavior via their axon collaterals.”

The research involved several novel experimental approaches. One part of the research used optogenetics, employing LED light to regulate certain proteins that were expressed selectively in VSCTs to either activate or suppress the neuronal activity. Another set of experiments used chemogenetics, a process by which a chemical compound is used to activate or suppress synthetic ligands expressed artificially in these neurons, controlling their activity.

Leveraging the ability of intact spinal cords from newborn mice to function in a dish, the researchers showed that activation of VSCTs by light induced locomotor behavior. When VSCT activity was suppressed by light or by drugs, ongoing locomotor behavior was halted. During adulthood, freely moving mice stopped moving when the activity of VSCT was suppressed by injecting an inhibitory drug. Locomotor behavior was also tested by the ability of mice to swim. Mice were unable to swim and simply floated in the water when VSCTs were silenced. In all of these models and experiments, the researchers demonstrated that VSCTs alone were both necessary and sufficient for controlling locomotor activity — activating them was enough to induce activity while suppressing them was enough to stop it.

Mentis acknowledges that there are limitations to conducting this type of research in mice, including the fact that while humans are bipedal, mice are quadrupedal; thus, their locomotion could be regulated in a different way. But he notes that other research on neurodegenerative diseases and processes in mice has led to clinical trials in human patients, suggesting that these findings are also likely to be applicable.

For their next steps, the team plans to identify and map precisely the neuronal circuits that VSCTs make with motor neurons and other spinal neurons. They also would like to identify select genetic markers and uncover potential subpopulations of VSCTs and explore their role in different modes of locomotion. Finally, they plan to explore how the function of VSCTs is altered in the context of pathology and neurodegenerative diseases.

People synchronize heart rates while listening attentively to stories

Source: Cell Press
Date: 9/14/2021
Link to original
Image of article

People often unconsciously synchronize bodily functions like heartbeat and breathing when they share an experience, such as a live performance or have a personal conversation. According to a new study, subjects’ heart rates synchronize even if they are just listening to a story by themselves, and this synchronization only occurs when the subjects are paying attention to the story. The findings from the research are reported September 14 in the journal Cell Reports.

“There’s a lot of literature demonstrating that people synchronize their physiology with each other. But the premise is that somehow you’re interacting and physically present the same place,” says co-senior author Lucas Parra, a professor at City College of New York. “What we have found is that the phenomenon is much broader, and that simply following a story and processing stimulus will cause similar fluctuations in people’s heart rates. It’s the cognitive function that drives your heart rate up or down.”

“What’s important is that the listener is paying attention to the actions in the story,” adds co-senior author Jacobo Sitt (@jdsitt), a researcher at the Paris Brain Institute and Inserm. “It’s not about emotions, but about being engaged and attentive, and thinking about what will happen next. Your heart responds to those signals from the brain.”

The investigators conducted a series of four experiments to explore the role of consciousness and attention in synchronizing participants’ heart rates. In the first, healthy volunteers listened to an audiobook of Jules Verne’s 20,000 Leagues Under the Sea. As they listened, their heart rate changed based on what was happening in the story, as measured by electrocardiogram (EKG). The researchers found that the majority of subjects showed increases and decreases in their heart rate at the same points in the narrative.

In the second experiment, volunteers watched short instructional videos. Because the videos were educational with no underlying emotional variations, this experiment confirmed that emotional engagement in a story was not playing a part. The first time they watched the videos, heart rates across the subjects showed similar fluctuations. The participants then watched the videos a second time while counting backwards in their heads. That time, the lack of attention resulted in a drop in the synchronization of heart rates across subjects, confirming that attention was important.

In the third experiment, the subjects listened to short children’s stories, some while attentive and others while being distracted, and then were asked to recall facts from the stories. The researchers found that the fluctuations seen in the participants’ heart rates were predictive of how well they did at answering questions about the story—more synchronization predicted better test scores. This indicated that changes in heart rate were a signal of conscious processing of the narrative.

When the researchers looked at changes in breathing rates, they didn’t see the same synchronization among the subjects. This was surprising, since breathing is known to affect heart rate.

The fourth experiment was similar to the first, but it included both healthy volunteers and patients with disorders of consciousness—such as those in comas or persistent vegetative states. All subjects were presented with an audiobook of a children’s story. As expected, the patients had lower rates of heart synchronization than did healthy controls. When the patients were examined six months later, some of them with higher synchronization had regained some consciousness.

“This study is still very preliminary, but you can imagine this being an easy test that could be implemented to measure brain function,” Sitt says. “It doesn’t require a lot of equipment. It even could be performed in an ambulance on the way to the hospital.” He notes that much more validation is needed with larger numbers of patients as well as comparisons to accepted tests of brain function like EEGs and fMRIs. This is something his group is continuing to study.

Parra says such research is also important for understanding mindfulness and the brain-body connection. “Neuroscience is opening up in terms of thinking of the brain as part of an actual anatomical, physical body,” he says. “This research is a step in the direction of looking at the brain-body connection more broadly, in terms of how the brain affects the body.”

Researchers identify a psychedelic-like drug without the hallucinogenic side effects

Source: Cell Press
Date: 4/28/2021
Link to original
Image of article

Psychedelic drugs have shown promise for treating neuropsychiatric disorders such as depression and posttraumatic stress disorder. However, due to their hallucinatory side effects, some researchers are trying to identify drugs that could offer the benefits of psychedelics without causing hallucinations. In the journal Cell on April 28, researchers report they have identified one such drug through the development of a genetically encoded fluorescent sensor–called psychLight–that can screen for hallucinogenic potential by indicating when a compound activates the serotonin 2A receptor.

“Serotonin reuptake inhibitors have long been used for treating depression, but we don’t know much about their mechanism. It’s like a black box,” says senior author Lin Tian (@LinTianLab), an associate professor in the Department of Biochemistry and Molecular Medicine in the School of Medicine at the University of California, Davis. “This sensor allows us to image serotonin dynamics in real time when animals learn or are stressed and visualize the interaction between the compound of interest and the receptor in real time.”

Tian’s lab joined forces with the lab of David E. Olson, an assistant professor in the Department of Chemistry at UC Davis, whose lab is focused on drug discovery. “This paper was an exceptionally collaborative effort,” says Olson, a co-author on the study. “My lab is really interested in the serotonin 2A receptor, which is the target of both psychedelic drugs and classic antipsychotics. Lin’s lab is a leader in developing sensors for neuromodulators like serotonin. It just made perfect sense for us to tackle this problem together.”

Experts believe that one of the benefits of using psychedelic drugs over existing drugs is that they appear to promote neural plasticity–essentially allowing the brain to rewire itself. If proven effective, this approach could lead to a drug that works in a single dose or a small number of doses, rather than having to be taken indefinitely. But one thing that researchers don’t know is whether patients would be able to gain the full benefit of neural plasticity without undergoing the “psychedelic trip” part of the treatment.

In the paper, the investigators report that they used psychLight to identify a compound called AAZ-A-154, a previously unstudied molecule that has the potential to act on beneficial pathways in the brain without hallucinogenic effects. “One of the problems with psychedelic therapies is that they require close guidance and supervision from a medical team,” Olson says. “A drug that doesn’t cause hallucinations could be taken at home.”

The serotonin 2A receptor, also known as 5-HT2AR, belongs to a class of receptors called G protein-coupled receptors (GPCRs). “More than one-third of all FDA-approved drugs target GPCRs, so this sensor technology has broad implications for drug development,” Tian says. “The special funding mechanisms of BRAIN Initiative from the National Institutes of Health allowed us to take a risky and radical approach to developing this technology, which could open the door to discovering better drugs without side effects and studying neurochemical signaling in the brain.”

First look at how hallucinogens bind structurally to serotonin receptors

Source: Cell Press
Date: 9/17/2020
Link to original
Image of article

Although hallucinogenic drugs have been studied for decades, little is known about the underlying mechanisms in the brain by which they induce their hallucinogenic–and (for disorders like depression and anxiety) potential therapeutic–effects. A paper publishing September 17 in the journal Cell reveals the first X-ray crystallography structure of LSD bound to its target in the brain, the serotonin receptor. The paper also includes the first cryo-electron microscopy (cryo-EM) structure of a prototypical hallucinogen coupled with the entire serotonin receptor complex.

“Millions of people have taken these drugs recreationally, and now they’re emerging as therapeutic agents,” says senior author Bryan Roth, a professor in the Department of Pharmacology at the University of North Carolina School of Medicine. “Gaining this first glimpse of how they act at the molecular level is really, really important, and it’s key to understanding how they work.”

Scientists believe that activation of the 5-HT2A serotonin receptor (HTR2A) in the brain is essential to the effects of hallucinogenic drugs. “These receptors are expressed at very high levels in the human cerebral cortex,” Roth says. “When they’re activated, they cause neurons to fire in an asynchronous and disorganized fashion, putting noise into the system. We think that’s one of the reasons they cause a psychedelic experience. However, it’s not at all clear how they exert their therapeutic actions.”

In this study, Roth collaborated with Georgios Skiniotis, a structural biologist at the Stanford University School of Medicine. “A combination of several different advances allowed us to do this research,” Skiniotis says. “One of these is better, more homogeneous preparations of the receptor proteins. Another is the evolution of cryo-EM technologies, which allows us to obtain high-resolution structures of complexes without having to crystalize them.”

In the paper, the investigators revealed the first X-ray crystallography structure of LSD bound to HTR2A. In addition, cryo-EM was used to uncover images of a prototypical hallucinogen, called 25-CN-NBOH, bound together with the entire receptor complex, including the effector protein Gαq. In the brain, this complex modulates the release of neurotransmitters and influences many biological and neurological processes.

Roth, a psychiatrist and biochemist, leads the Psychoactive Drug Screening Program, funded by the National Institute of Mental Health. This gives him access to hallucinogenic drugs for research purposes. Normally, these compounds are difficult to study in the lab because they’re regulated by the Drug Enforcement Agency as Schedule 1 drugs.

“One of the main goals of my work is to understand how hallucinogens exert their actions on the brain,” Roth says. “If we can understand how they work at the molecular level, this will ultimately give us clues into human consciousness, perception, and awareness.”

Going forward, the investigators plan to apply their findings to structure-based drug discovery for new therapeutics. One of the goals is to discover potential candidates that may be able offer therapeutic benefit without the psychedelic effects.

“The more we understand about how these drugs engage and activate the receptors, the better we’ll understand their signaling properties,” Skiniotis says. “This work doesn’t give us the whole picture yet, but it’s an important piece of the puzzle.”

COVID-19 patients who experience cytokine storms may make few memory B cells

Source: Cell Press
Date: 8/19/2020
Link to original
Image of article

Newswise — The release of massive amounts of proteins called cytokines can lead to some of the most severe symptoms of COVID-19. When large numbers of immune cells release cytokines, this increases inflammation and creates a feedback loop in which more immune cells are activated and this is sometimes called a cytokine storm. An August 19 study in the journal Cell now suggests that high levels of some cytokines may also prevent people who are infected from developing long-term immunity as affected patients were observed to make very few of the type of B cells needed to develop a durable immune response.

“We’ve seen a lot of studies suggesting that immunity to COVID-19 is not durable because the antibodies decline over time,” says co-senior author Shiv Pillai, a professor at Harvard Medical School and member of the Ragon Institute of Massachusetts General Hospital, MIT, and Harvard. “This study provides a mechanism that explains this lower-quality immune response.”

The investigators focused on germinal centers–the areas within the lymph nodes and spleens where B cells, the immune cells that produce antibodies, differentiate. Differentiation and changes in antibody genes are required to build immunity to an infectious agent.

“When we looked at the lymph nodes and spleens of patients who died from COVID-19, including some who died very soon after getting the disease, we saw that these germinal center structures had not formed,” says co-senior author Robert Padera, a pathology professor at Harvard. “We decided to determine why that’s the case.”

Because the disease was so new, animal models for studying COVID-19 infection were not yet available at the time they began their study. The researchers instead gained insights from previous studies involving mouse models of other infections that induce cytokine storm syndrome–a malaria model and one of bacterial infection in which germinal centers were lost.

In people with severe COVID-19, one of most abundant cytokines released is called TNF. In the infected mice, TNF appeared to block the formation of germinal centers. In previous cytokine storm models, when the mice were given antibodies to block TNF or had their TNF gene deleted, the germinal centers were able to form. When the researchers studied the lymph nodes of patients who had died of the disease, they found high levels of TNF in these organs. This led them to conclude that TNF may be preventing the germinal centers from forming in people with COVID-19 as well.

“Studies have suggested this lack of germinal centers happens with SARS infections,” Pillai says. “We even think this phenomenon occurs in some patients with Ebola, so it was not surprising to us.”

The researchers also studied blood and lymphoid tissue from people with active infections who were in different stages of COVID-19. They found that although germinal centers were not formed, B cells were still activated and appeared in the blood, which would allow the patients to produce some neutralizing antibodies. “There is an immune response,” Padera says. “It’s just not coming from a germinal center.”

“Without the germinal centers, there is no long-term memory to the antigens,” Pillai adds. He notes that studies of other coronaviruses that cause colds have suggested that someone can get infected with the same coronavirus three or four times in the same year.

The authors say despite their findings, they still believe a successful COVID-19 vaccine can be developed as it should not cause high levels of cytokines to be released.

New approach allows blind, sighted to “see” shapes

Source: Cell Press
Date: 5/15/2020
Link to original
Image of article

For most adults who lose their vision, blindness results from damage to the eyes or optic nerve while the brain remains intact. For decades, researchers have proposed developing a device that could restore sight by bypassing damaged eyes and delivering visual information from a camera directly to the brain. In study published in the journal Cell, researchers from Baylor College of Medicine report that they are one step closer to this goal.

They describe an approach in which implanted electrodes are stimulated in a dynamic sequence, essentially “tracing” shapes on the surface of the visual cortex that participants were able to “see.”

“When we used electrical stimulation to dynamically trace letters directly on patients’ brains, they were able to ‘see’ the intended letter shapes and could correctly identify different letters,” said Dr. Daniel Yoshor, professor and chair of neurosurgery and senior author on the paper. “They described seeing glowing spots or lines forming the letters, like skywriting.”

Previous attempts to stimulate the visual cortex have been less successful. Earlier methods treated each electrode like a pixel in a visual display, stimulating many of them at the same time. Participants could detect spots of light but found it hard to discern visual objects or forms. “Rather than trying to build shapes from multiple spots of light, we traced outlines,” said Dr. Michael Beauchamp, professor and in neurosurgery, director of the Core for Advanced MRI and first author on the paper. “Our inspiration for this was the idea of tracing a letter in the palm of someone’s hand.”

The investigators tested the approach in four sighted people who had electrodes implanted in their brains to monitor epilepsy and two blind people who had electrodes implanted over their visual cortex as part of a study of a visual cortical prosthetic device. Stimulation of multiple electrodes in sequences produced perceptions of shapes that subjects were able to correctly identify as specific letters.

The approach, the researchers say, demonstrates that it could be possible for blind people to regain the ability to detect and recognize visual forms by using technology that inputs visual information directly into the brain, should they wish to. The researchers note, however, that several obstacles must be overcome before this technology could be implemented in clinical practice.

“The primary visual cortex, where the electrodes were implanted, contains half a billion neurons. In this study we stimulated only a small fraction of these neurons with a handful of electrodes,” Beauchamp said. “An important next step will be to work with neuroengineers to develop electrode arrays with thousands of electrodes, allowing us to stimulate more precisely. Together with new hardware, improved stimulation algorithms will help realize the dream of delivering useful visual information to blind people.

Researchers restore injured man’s sense of touch using brain-computer interface technology

Source: Cell Press
Date: 4/23/2020
Link to original
Image of article

While we might often take our sense of touch for granted, for researchers developing technologies to restore limb function in people paralyzed due to spinal cord injury or disease, re-establishing the sense of touch is an essential part of the process. And on April 23 in the journal Cell, a team of researchers at Battelle and the Ohio State University Wexner Medical Center report that they have been able to restore sensation to the hand of a research participant with a severe spinal cord injury using a brain-computer interface (BCI) system. The technology harnesses neural signals that are so miniscule they can’t be perceived and enhances them via artificial sensory feedback sent back to the participant, resulting in greatly enriched motor function.

“We’re taking subperceptual touch events and boosting them into conscious perception,” says first author Patrick Ganzer, a principal research scientist at Battelle. “When we did this, we saw several functional improvements. It was a big eureka moment when we first restored the participant’s sense of touch.”

The participant in this study is Ian Burkhart, a 28-year-old man who suffered a spinal cord injury during a diving accident in 2010. Since 2014, Burkhart has been working with investigators on a project called NeuroLife that aims to restore function to his right arm. The device they have developed works through a system of electrodes on his skin and a small computer chip implanted in his motor cortex. This setup, which uses wires to route movement signals from the brain to the muscles, bypassing his spinal cord injury, gives Burkhart enough control over his arm and hand to lift a coffee mug, swipe a credit card, and play Guitar Hero.

“Until now, at times Ian has felt like his hand was foreign due to lack of sensory feedback,” Ganzer says. “He also has trouble with controlling his hand unless he is watching his movements closely. This requires a lot of concentration and makes simple multitasking like drinking a soda while watching TV almost impossible.”

The investigators found that although Burkhart had almost no sensation in his hand, when they stimulated his skin, a neural signal — so small it was his brain was unable to perceive it — was still getting to his brain. Ganzer explains that even in people like Burkhart who have what is considered a “clinically complete” spinal cord injury, there are almost always a few wisps of nerve fiber that remain intact. The Cell paper explains how they were able to boost these signals to the level where the brain would respond.

The subperceptual touch signals were artificially sent back to Burkhart using haptic feedback. Common examples of haptic feedback are the vibration from a mobile phone or game controller that lets the user feel that something is working. The new system allows the subperceptual touch signals coming from Burkhart’s skin to travel back to his brain through artificial haptic feedback that he can perceive.

The advances in the BCI system led to three important improvements. They enable Burkhart to reliably detect something by touch alone: in the future, this may be used to find and pick up an object without being able to see it. The system also is the first BCI that allows for restoration of movement and touch at once, and this ability to experience enhanced touch during movement gives him a greater sense of control and lets him to do things more quickly. Finally, these improvements allow the BCI system to sense how much pressure to use when handling an object or picking something up — for example, using a light touch when picking up a fragile object like a Styrofoam cup but a firmer grip when picking up something heavy.

The investigators’ long-term goal is to develop a BCI system that works as well in the home as it does in the laboratory. They are working on creating a next-generation sleeve containing the required electrodes and sensors that could be easily put on and taken off. They also aim to develop a system that can be controlled with a tablet rather than a computer, making it smaller and more portable.

“It has been amazing to see the possibilities of sensory information coming from a device that was originally created to only allow me to control my hand in a one-way direction,” Burkhart says.

Brigham Leads Development of Surgical Techniques for Treating AVM

Source: Brigham and Women's Hospital
Date: 1/9/2023
Link to original
Image of article

The treatment for brain arteriovenous malformation (AVM) consists of observation, surgery, stereotactic radiosurgery, or endovascular embolization—either on its own or before surgery to remove the AVM. But these techniques don’t always yield a safe cure for patients, most of whom are young and experiencing debilitating symptoms.

Brigham and Women’s Hospital created the AVM Program, directed by neurosurgeon Nirav J. Patel, MD, to address this challenge and look for better solutions. The program is made up of cerebrovascular neurosurgeons, neuro-interventional radiologists, and vascular neurologists who collaborate on the development of interdisciplinary approaches. The team’s goal is to provide the best possible nonsurgical and surgical treatment options for people with AVMs.

“We at the Brigham started this program to help cure patients,” Dr. Patel says. “They often show up on our doorstep years after their initial diagnosis with a lot of misinformation. When someone in their teens or 20s is told they have a ticking time bomb in their brains, it’s very hard for them to hear. When we tell them we can cure them, they are shocked.”

Providing the Best Outcomes for Patients

Dr. Patel’s primary focus is a surgical technique that involves sealing off the feeding arteries to the AVM with clips before surgical resection (in other words, a surgical embolization), rather than blocking the arteries by endovascular embolization. “By using this AVM technique, we avoid the risks and costs of endovascular embolization,” he says. “The procedure takes longer and requires much more practice and surgical skill, but it often delivers better patient outcomes.”

After being trained in the technique by neurosurgeon Michael Morgan, AO, in Australia, Dr. Patel brought this approach to the Brigham. He’s made it his mission to offer the procedure to anyone who could benefit from it and train others to perform this delicate operation.

Toward that end, he sees the training of residents and fellows as an especially important role. “Everyone is expected to improve their microsurgical techniques,” he says. “It’s nothing special, just a lot of hard work that gets them there.”

Dr. Patel has built a 3D skills lab where trainees can practice working with brain blood vessels safely and perform bypass and repair. Training is done initially under a microscope with plastic tubes that simulate blood vessels. Dr. Patel also leads a preclinical rodent lab where residents and fellows can further hone their surgical techniques.

Creating Programs to Help AVM Patients Internationally

Many of Dr. Patel’s former trainees have taken the techniques with them upon moving on to faculty positions as far away as Japan, as well as to Puerto Rico and other parts of the United States.

Dr. Patel and his team have also traveled internationally to train local surgeons and provide care for patients unable to travel to the Brigham for surgery. In the summer of 2022, he journeyed to Paraguay, where he performed surgery on three patients. The trip was made in collaboration with the charity Solidarity Bridge—including the group’s program director, Lindsey Douchette, MPA, and neurosurgeon Richard Moser, MD—and neurosurgeon José Kuzli, MD, at the Hospital Nacional de Itaugua in Asuncion. Brigham anesthesiologist Grace Youngsook Kim, MD, and scrub nurse Carlos Vazquez accompanied Dr. Patel, with Vazquez also serving as a translator for Spanish-speaking patients.

“In addition to helping these patients, I shared my techniques with surgeons from all over the country who came to learn,” Dr. Patel says. “For countries like Paraguay that don’t have the instrumentation and resources to perform presurgical embolizations, teaching this procedure can make a profound difference in the lives of many patients with AVM. Our goal is to create a program there that is self-sustaining.” Dr. Patel had previously traveled to Puerto Rico, where he performed two AVM procedures and helped set up a similar program.

In the United States, Dr. Patel works with insurance companies to help patients out of network obtain coverage for treatment at the Brigham. He recently treated a 29-year-old mother of four from Chicago who experienced debilitating side effects after several failed embolizations.

A Continued Focus on Research

Dr. Patel notes that the role of the AVM Program is not only to develop new surgical techniques but also to refine all treatments that can benefit patients. This includes research in animal models to understand cerebral physiology and neuronal regeneration.

“We’re hoping to find a molecular marker for AVM that we can target and treat through medical means,” he says. “We’re working on sensitization of the endothelium inside the arteries. Eventually, we may be able to give patients a pill that makes their arteries selectively sensitive to radiation.” This might eventually provide another good treatment option for patients who cannot have surgeries like the ones that Dr. Patel performs.

Making Headway in Targeting Glioblastoma Multiforme

Source: Brigham and Women's Hospital
Date: 6/14/2022
Link to original
Image of article

The past two decades have seen considerable progress in developing targeted therapies for cancer. Many of these drugs are successful in treating both primary tumors and metastases. Additionally, when patients develop resistance to certain therapies, often other targeted treatments and new options are available.

Unfortunately, advances in precision oncology for glioblastoma multiforme (GBM) have lagged behind many other cancers. A multicenter, philanthropically funded effort now aims to address that deficit. Break Through Cancer is a foundation that brings together several elite cancer research centers to accelerate research through collaboration, conduct clinical trials, and ultimately develop cures for the deadliest cancers. One of those cancers is GBM.

Along with colleagues at the Dana-Farber Cancer Institute (DFCI), as well as investigators at the MD Anderson Cancer Center, Memorial Sloan Kettering Cancer Center, Johns Hopkins Medicine, and Massachusetts Institute of Technology, a team from Brigham and Women’s Hospital has been assembled to work on these efforts.

Conducting Serial Biopsies of the Brain

One reason for the disparity in available targeted therapies for GBM is the difficulty in obtaining regular biopsies of these tumors due to their location in the brain. Biopsies are a crucial part of targeted therapy.

“For decades we have run multiple trials for GBM, and none of them have been very successful,” says Nathalie Y. R. Agar, PhD, founding director of the Surgical Molecular Imaging Laboratory in the Brigham’s Department of Neurosurgery. “What many of us in the field have realized is that the main difference in treating other cancers is that, as part of clinical trials, patients undergo biopsies to assess the response of the tumor to the therapeutic agent under investigation.”

“Patients routinely receive multiple biopsies throughout the course of treatment for other cancers or other diseases to see whether drug X or drug Y actually is doing what it’s supposed to do,” adds E. Antonio Chiocca, MD, PhD, chair of the Department of Neurosurgery. “But we’ve never done that for GBM. This is what we’re proposing to do.”

Treatment With a Modified Oncolytic Virus

Because of the logistical and ethical issues surrounding the repeated collection of biopsies that require drilling into the skull, this research is being conducted in conjunction with a clinical trial that also requires direct access to the brain—delivery of an oncolytic virus that aims to destroy GBM tumors.

This treatment, in which a modified herpes simplex virus 1 is injected directly into the tumor, has already been tested in a phase 1 clinical trial at the Brigham and DFCI that enrolled 50 patients between 2017 and 2022. The treatment appeared to be safe and provided benefit to some patients.

The new research, an extension of the earlier trial, will enroll 12 more patients (three at each of the four Break Through Cancer-funded clinical institutions) to receive up to six doses over a four-month period. At the same time the patients receive the treatment, a biopsy of their tumor will be collected to assess how the cancer is responding.

“This treatment requires only conscious sedation and is guided by a GPS-like robotic system,” Dr. Chiocca says. “It’s very well tolerated, and most patients go home the next day.”

The hope is that by regularly collecting tumor tissue, researchers will be able to learn more about the mechanisms that underlie the cancer’s response to treatment, potentially leading to more personalized approaches as well as new, more effective therapies.

Looking at Combination Treatments

Eventually, the investigators plan to conduct additional trials in which other agents that require intratumoral injection are used. This next phase is likely to include a biologic agent that triggers the immune system to fight cancer.

Dr. Agar noted that this research has a strong translational science component, including the development of new model systems that will help investigators select new drugs or combinations of drugs that may also be effective.

“This work has been possible not only because of Break Through Cancer, but thanks to strong leadership at both the Brigham and DFCI,” she says. “We hope that it eventually will revolutionize the way that GBM is treated.”