Nanoscope Therapeutics: Gene Therapy Improves Visual Acuity in Patients with Retinitis Pigmentosa – 2 Minute Medicine

The Latest

A recent two-year phase 2b, randomized, double-masked, sham-controlled multicenter clinical trial by Boyer et al. and Nanoscope Therapeutics investigated mutation-agnostic gene therapy for the treatment of permanent and severe vision loss from of retinitis pigmentosa. The results build on an earlier trial that found 89% of patients injected with the gene therapy to experience an improvement of luminance levels across two visual tests compared to control group patients. This newest trial, named RESTORE, demonstrated significant improvement in best-corrected visual acuity after 52 weeks compared to the control arm. The results showed that gene therapy was well tolerated, with no treatment-related serious or severe adverse events reported.

Physicians Perspective

Retinitis Pigmentosa encompasses a group of rare genetic eye disorders in which the retinas photoreceptors degrade over time leading to profound visual field and vision loss in advanced stages. Retinitis pigmentosa affects 1 in every 400 people in the United States and approximately 1 in 5000 worldwide, making it the most common inherited disease of the retina. There are currently no cures available for retinitis pigmentosa. Current gene therapies aim to treat patients with specific gene mutations and are limited in advanced disease with degenerated outer retinal cells. Nanoscopes optogenetic monotherapy targets the intact inner retinal neurons to restore vison loss. This approach has the advantage of restoring vision even in advanced retinitis pigmentosa, regardless of causative gene mutation. Furthermore, the therapy is administered via a single intravitreal injection without any need for external devices.

Molecular Targets

Nanoscope Therapeutics has developed a gene therapy called MCO-010 that uses light sensitive molecules to treat retinal disease. MCO-010 is an injection that transforms bipolar cells that normally do not transmit light (are not sensitizing) to become light sensitizing. The gene therapy works by transfecting the cell layers above the damaged cone layers, such as the bipolar and ganglion cells, into viable light producing cells. MCO-010 is activated by ambient light across the visual spectrum.

Company History

Nanoscope therapeutics is a Texas based late-stage clinical biotechnology company developing gene therapies for inherited retinal diseases and age-related macular degeneration. MCO-010 is the companys lead asset and has recently received FDA fast-track designations. Additionally, the company has recently completed a phase 2 trial of MCO-010 in Stargardt disease.

Further reading: https://www.fiercebiotech.com/biotech/nanoscope-eyes-market-after-gene-therapy-improves-vision-patients-retinal-disease

2024 2 Minute Medicine, Inc. All rights reserved. No works may be reproduced without expressed written consent from 2 Minute Medicine, Inc. Inquire about licensing here. No article should be construed as medical advice and is not intended as such by the authors or by 2 Minute Medicine, Inc.

Continued here:
Nanoscope Therapeutics: Gene Therapy Improves Visual Acuity in Patients with Retinitis Pigmentosa - 2 Minute Medicine

Novel Switch Promises Safer Gene Therapy On-Demand – Mirage News

Just like a doctor adjusts the dose of a medication to the patient's needs, the expression of therapeutic genes, those modified in a person to treat or cure a disease via gene therapy, also needs to be maintained within a therapeutic window. Staying within the therapeutic window is important as too much of the protein could be toxic, and too little could result in a small or no therapeutic effect.

Although the principle of therapeutic window has been known for a long time, there has been no strategy to implement it safely, limiting the potential applications of gene therapy in the clinic. In their current study published in the journal Nature Biotechnology, researchers at Baylor College of Medicine report on a technology to effectively regulate gene expression, a promising solution to fill this gap in gene therapy clinical applications.

"Although there are several gene regulation systems used in mammalian cells, none has been approved by the U.S. Food and Drug Administration for clinical applications, mainly because those systems use a regulatory protein that is foreign to the human body, which triggers an immune response against it," said corresponding author Dr. Laising Yen, associate professor of pathology and immunology and of molecular and cellular biology at Baylor. "This means that the cells that are expressing the therapeutic protein would be attacked, eliminated or neutralized by the patient's immune system, making the therapy ineffective."

For more than a decade, Yen and his colleagues have been working on this technology and now they have found a solution to overcome the main obstacles in its clinical use. "The solution we found does not involve a foreign regulatory protein that will evoke an immune response in patients. Instead, we use small molecules to interact with RNA, which typically do not trigger an immune response," Yen said. "Other groups also have made attempts to resolve this critical issue, but the drug concentrations they used are beyond what the FDA has approved for patients. We were able to engineer our system in such a way that it works at the FDA-approved dosage."

A switch to turn genes on/off on cue

Yen and his colleagues developed a system that turns genes on to different levels on cue using small molecules at FDA-approved doses. The switch is placed in the RNA, the copy of genetic material that is translated into a protein. This approach allows the researchers to control the protein's production a step back by controlling its RNA.

The RNA of interest is first engineered to contain an extra polyA signal, akin to a "stop sign" that genes naturally use to mark the end of a gene. When the machinery of the cell detects a polyA signal in the RNA, it automatically makes a cut and defines the cut point as the end of the RNA. "In our system, we use the added polyA signal, not at the end, but at the beginning of the RNA, so the cut destroys the RNA and therefore the default is no protein production. It is turned off until we turn it on with the small molecule," Yen said.

To turn on the gene at the desired level, the team engineered a switch on the RNA. They modified a section of the RNA near the polyA signal such that it can now bind to a small molecule, FDA-approved tetracycline in this case. "When tetracycline binds to that section that functions as a sensor on the RNA, it masks off the polyA signal, and the RNA will now be translated into protein," Yen said.

Imagine the now possible future situation. A patient has received gene therapy that provides a gene to compensate for a malfunctioning gene that causes a medical condition. The gene the patient received has the switch, which allows the physician to control the production of the therapeutic protein. If the patient only requires a small amount of the therapeutic protein, then he/she will only take a small dose of tetracycline, which will turn on the therapeutic gene only a little. If the patient needs more therapeutic protein, then he/she would take more tetracycline to boost production. To stop production of the therapeutic protein, the patient stops taking tetracycline. In the absence of tetracycline, the switch will be back to its default off position. Some diseases may benefit from the presence of constant low levels of therapeutic protein. In that case, the technology has the flexibility to pre-adjust the default level to specified levels of protein expression while retaining the option of dialing up the expression with tetracycline.

"This strategy allows us to be more precise in the control of gene expression of a therapeutic protein. It enables us to adjust its production according to disease's stages or tune to the patients' specific needs, all using the FDA-approved tetracycline dose," Yen said. "Our approach is not disease-specific, it can theoretically be used for regulating the expression of any protein, and potentially has many therapeutic applications. In addition, this system is more compact and easier to implement than the existing technologies. Therefore, it also can be very useful in the lab to turn a gene of interest on or off to study its function."

Liming Luo, Jocelyn Duen-Ya Jea, Yan Wang and Pei-Wen Chao, all at Baylor College of Medicine, also contributed to this work.

This work was supported by an E&M Foundation Pre-Doctoral Fellowship for Biomedical Research, NIH grants (R01EB013584, UM1HG006348, R01DK114356, R01HL130249, P30 CA125123 and S10 RR024574), Biogen SRA, seed fund from Department of Pathology and Immunology at Baylor College of Medicine and CPRIT Core Facility Support Award CPRIT-RP180672.

Read the original:
Novel Switch Promises Safer Gene Therapy On-Demand - Mirage News

An unprecedented view of gene regulation | MIT News … – MIT News

Much of the human genome is made of regulatory regions that control which genes are expressed at a given time within a cell. Those regulatory elements can be located near a target gene or up to 2 million base pairs away from the target.

To enable those interactions, the genome loops itself in a 3D structure that brings distant regions close together. Using a new technique, MIT researchers have shown that they can map these interactions with 100 times higher resolution than has previously been possible.

Using this method, we generate the highest-resolution maps of the 3D genome that have ever been generated, and what we see are a lot of interactions between enhancers and promoters that haven't been seen previously, says Anders Sejr Hansen, the Underwood-Prescott Career Development Assistant Professor of Biological Engineering at MIT and the senior author of the study. We are excited to be able to reveal a new layer of 3D structure with our high resolution.

The researchers findings suggest that many genes interact with dozens of different regulatory elements, although further study is needed to determine which of those interactions are the most important to the regulation of a given gene.

MIT postdoc Miles Huseyin is also a lead author of the paper, which appears today in Nature Genetics.

High-resolution mapping

Scientists estimate that more than half of the genome consists of regulatory elements that control genes, which make up only about 2 percent of the genome. Genome-wide association studies, which link genetic variants with specific diseases, have identified many variants that appear in these regulatory regions. Determining which genes these regulatory elements interact with could help researchers understand how those diseases arise and, potentially, how to treat them.

Discovering those interactions requires mapping which parts of the genome interact with each other when chromosomes are packed into the nucleus. Chromosomes are organized into structural units called nucleosomes strands of DNA tightly wound around proteins helping the chromosomes fit within the small confines of the nucleus.

To perform Hi-C, researchers use restriction enzymes to chop the genome into many small pieces and biochemically link pieces that are near each other in 3D space within the cells nucleus. They then determine the identities of the interacting pieces by amplifying and sequencing them.

While Hi-C reveals a great deal about the overall 3D organization of the genome, it has limited resolution to pick out specific interactions between genes and regulatory elements such as enhancers. Enhancers are short sequences of DNA that can help to activate the transcription of a gene by binding to the genes promoter the site where transcription begins.

To achieve the resolution necessary to find these interactions, the MIT team built on a more recent technology called Micro-C, which was invented by researchers at the University of Massachusetts Medical School, led by Stanley Hsieh and Oliver Rando. Micro-C was first applied in budding yeast in 2015 and subsequently applied to mammalian cells in three papers in 2019 and 2020 by researchers including Hansen, Hsieh, Rando and others at University of California at Berkeley and at UMass Medical School.

Micro-C achieves higher resolution than Hi-C by using an enzyme known as micrococcal nuclease to chop up the genome. Hi-Cs restriction enzymes cut the genome only at specific DNA sequences that are randomly distributed, resulting in DNA fragments of varying and larger sizes. By contrast, micrococcal nuclease uniformly cuts the genome into nucleosome-sized fragments, each of which contains 150 to 200 DNA base pairs. This uniformity of small fragments grants Micro-C its superior resolution over Hi-C.

However, since Micro-C surveys the entire genome, this approach still doesnt achieve high enough resolution to identify the types of interactions the researchers wanted to see. For example, if you want to look at how 100 different genome sites interact with each other, you need to sequence at least 100 multiplied by 100 times, or 10,000. The human genome is very large and contains around 22 million sites at nucleosome resolution. Therefore, Micro-C mapping of the entire human genome would require at least 22 million multiplied by 22 million sequencing reads, costing more than $1 billion.

To bring that cost down, the team devised a way to perform a more targeted sequencing of the genomes interactions, allowing them to focus on segments of the genome that contain genes of interest. By focusing on regions spanning a few million base pairs, the number of possible genomic sites decreases a thousandfold and the sequencing costs decrease a millionfold, down to about $1,000. The new method, called Region Capture Micro-C (RCMC), is therefore able to inexpensively generate maps 100 times richer in information than other published techniques for a fraction of the cost.

Now we have a method for getting ultra-high-resolution 3D genome structure maps in a very affordable manner. Previously, it was so inaccessible financially because you would need millions, if not billions of dollars, to get high resolution, Hansen says. The one limitation is that you can't get the whole genome, so you need to know approximately what region you're interested in, but you can get very high resolution, very affordably.

Many interactions

In this study, the researchers focused on five regions varying in size from hundreds of thousands to about 2 million base pairs, which they chose due to interesting features revealed by previous studies. Those include a well-characterized gene called Sox2, which plays a key role in tissue formation during embryonic development.

After capturing and sequencing the DNA segments of interest, the researchers found many enhancers that interact with Sox2, as well as interactions between nearby genes and enhancers that were previously unseen. In other regions, especially those full of genes and enhancers, some genes interacted with as many as 50 other DNA segments, and on average each interacting site contacted about 25 others.

People have seen multiple interactions from one bit of DNA before, but it's usually on the order of two or three, so seeing this many of them was quite significant in terms of difference, Huseyin says.

However, the researchers technique doesnt reveal whether all of those interactions occur simultaneously or at different times, or which of those interactions are the most important.

The researchers also found that DNA appears to coil itself into nested microcompartments that facilitate these interactions, but they werent able to determine how microcompartments form. The researchers hope that further study into the underlying mechanisms could shed light on the fundamental question of how genes are regulated.

Even though we're not currently aware of what may be causing these microcompartments, and we have all these open questions in front of us, we at least have a tool to really stringently ask those questions, Goel says.

In addition to pursuing those questions, the MIT team also plans to work with researchers at Boston Childrens Hospital to apply this type of analysis to genomic regions that have been linked with blood disorders in genome-wide association studies. They are also collaborating with researchers at Harvard Medical School to study variants linked to metabolic disorders.

Christine Eyler, a medical instructor at Duke University School of Medicine, says the new technique will provide a valuable tool for analyzing ultrafine chromatin looping architecture.

I anticipate that pairing the ultraresolved RCMC contact looping data with other assays that define specific regulatory elements will reveal important new insights as to the relationship between nuclear structure and gene regulatory function, says Eyler, who was not involved in this study. Having performed the assay in our own group, we were impressed by the fact that the protocol is easy to follow as written (even for scientists not previously experienced in topology assays), and is economically very efficient given the wealth of information that it provides.

The research was funded by the Koch Institute Support (core) Grant from the National Cancer Institute, the National Institutes of Health, the National Science Foundation, a Solomon Buchsbaum Research Support Committee Award, the Koch Institute Frontier Research Fund, an NIH Fellowship and an EMBO Fellowship.

See more here:
An unprecedented view of gene regulation | MIT News ... - MIT News

Can Europe afford the next generation of medicines? – POLITICO Europe

On the face of it, medical science has delivered at just the right time.

Europes population is growing older: by 2100 nearly a third of people will be aged 65 or over. A major challenge for this century is how to keep people healthy.

At the same time, the last decade has seen personalized medicines come within reach. Cell and gene therapies tailored to individual patients can cure diseases, from fixing misfiring biology to training the immune system to kill cancer cells.

But with price tags of up to 3 million per treatment,healthsystems simply arent designed to absorb these kinds of hits.

The question now is whether Europe will be able to square the circle and take full advantage of the new therapies. Or will budget constraints keep these medical advances out of reach?

To date, the European Commission has authorized 25 cell and gene therapies for use in Europe.

These therapies involve careful manipulation and growth of human cells to be given often as one-off treatments and in some cases cures for debilitating and often deadly rare diseases and certain types of cancer. Novartis Kymriah, a CAR-T therapy that delivers genes into the body to tell the immune system to attack and kill blood cancer cells, is one such example.

These medicines are often tailor-made for a single patient, making them extremely expensive.

Novartis Zolgensma, a gene therapy for children with spinal muscular atrophy, had a U.S. list price of $2.1 million for a one-off dose when it was approved in May 2019 (it was approved in the EU in May 2020). Since then, it has lost the title of the worlds most expensive therapy to CSL Behrings hemophilia B gene therapy Hemgenix, a one-off infusion that costs $3.5 million a dose.

Big upfront costs in building the first manufacturing facilities for these types of therapies play a role in high prices, said Matthew Durdy, chief executive of the Cell and Gene Therapy Catapult, an independent nonprofit that brings together academia, industry, health services and government to deliver these products in the U.K.

But these costs will come down, he said, just as they do with every new technology, from mobile phones to laptops. For that to happen, health systems must start using the therapies. Without a market, competitors will not follow, and without competition, prices wont drop.

But thats easier said than done.

Finding a price that is both affordable to health care payers and profitable for companies is a major and significant challenge, said Tim Hunt, chief executive of the Alliance for Regenerative Medicine (ARM), which represents developers of these therapies.

And its one which has pushed companies to shutter some EU operations and investors to hold back, he said. Seven of the 25 cell and gene therapies approved in Europe have already been withdrawn by drugmakers from EU markets.

Since 2018, the EU cell and gene therapy market has grown just 11 percent, compared with 43 percent in the U.S. and 531 percent in China.

Consequently, EU patients are losing out on the next generation of treatments, Hunt said.

One way to bring things back on track is delivering data that shows a particular therapy works and justifies those initial high costs. But this data can be hard to come by.

Thats because it takes years to prove that a drug prevents a progressive disease, for example. And in rare conditions, there are only a handful of patients to test out new therapies, limiting the evidence.

Therefore, to approve these therapies, Europe has been steadily lowering the bar for clinical evidence, said Lydie Meheus, managing director at the not-for-profit research organization the Anticancer Fund.

While this authorizes more therapies, lack of data creates significant problems for payers asking health systems to take big financial risks when resources are especially stretched, staff are on strike and services are still grappling with pandemic backlogs.

We really need to rebalance the scale when it comes to the evidence generation of these types of products, said Yannis Natsis, chief executive of the European Social Insurance Platform (ESIP). Limited data and high prices are a huge challenge for payers, he said.

One hope lies in Europes 2021 Health Technology Assessment (HTA) regulation.

From 2025, all cell and gene therapies will undergo a single EU assessment of the value they add to patients and health systems, hopefully ending the need for 27 duplicate reviews. This recommendation will be used by payers in each country in their pricing talks with developers.

Companies will also meet jointly with the European Medicines Agency and Europes HTA coordinating group to thrash out the best clinical trial designs that deliver data, not only on safety and efficacy but also on added value.

And the technology is rapidly progressing. While the majority of cell and gene therapies involve taking a patient sample and tailor-making a treatment, in time the aim is to deliver more products that are developed without the need of a patient sample.

A lot of companies are trying to focus more on in vivo gene therapies [where the genetic changes happen inside the body as opposed to in a lab] and off-the-shelf cell therapies that use donor materials, said Stephen Majors, head of communications at ARM.

This will lessen the production costs, curtailing the need for the intensive process of removing a patients cells, sending them elsewhere to be modified, and using cold chain storage infrastructure, he said.

And if the longer-term data bears fruit on curative benefits, payers may be more willing to pay. Some are already adapting payment models to spread the costs over several years.

To Durdy, at the U.K. catapult, these types of therapies will save money in the long run. Spending for example $2 million to $3 million on curing a hemophilia patient will prevent up to $12 million in lifetime care costs for that patient, he said.

He imagined that, in the future, payers would think along the following lines: Its going to blow my budget this year. But if all of these therapeutics come through, Im going to transform the way I do health care.

See original here:
Can Europe afford the next generation of medicines? - POLITICO Europe

Research identifies new cause of heart failure condition in children – EurekAlert

image:A heart muscle cell with mutations in the gene that makes the Rotatin protein (bottom) has disorganized muscle fibers (red) compared to healthy heart muscle cell (top). view more

Credit: Matthew Miyamoto

In an effort to determine the cause behind a rare condition that causes heart failure in children, University of Maryland School of Medicine (UMSOM) researchers have identified new gene mutations responsible for the disorder in an infant patient. They were then able to learn how the mutation works and used a drug to reverse its effects in heart muscle cells derived from stem cells from the patient.

The findings, published in late April inCirculation,suggest that treatments could be developed to manage the condition rather than requiring a heart transplant, which is the standard treatment for this condition in children.

Although much has been studied about heart failure in adults, there is still much to learn about the genetic causes of heart failure in infants,saidCharles Chaz Hong, MD, PhD,Melvin Sharoky, MD Professor of Medicine and Physiology, Director of Cardiology Research, and Co-Chief of Cardiovascular Medicine at UMSOM.Mutations in the gene we identified had been implicated in microcephaly in babies but not yet in human heart disease.

Infantile dilated cardiomyopathy is a common cause of heart failure responsible for about half of pediatric heart failure cases whose cause is most often unknown. Although relatively rare, occurring in about one in 200,000 births, infants with the condition have hearts that fail to contract as effectively, so they are not able to pump as much blood as they should.

This genetic mutation discovered by Dr. Hong and his colleagues was found to normally make a protein found in a cell structure, the centrosome, that functions as a tether for the cells skeleton and is best known for its role during cell division.

Without this protein, muscle cells in the heart were unable to organize themselves neatly and did not contract as well, which in turn affected the hearts pumping, the researchers theorized.

We originally dismissed our findings as artifacts that the cell division machinery would be involved in this kind of heart muscle dysfunction,said Dr. Hong.We thought that once the heart cells matured, this cell division machinery completely disappeared, but it turned out, it moves to a new location in the cell and takes on a new role in heart muscle function.

To identify this gene mutation responsible for infant heart failure, the researchers removed a sample of heart cells from the patients diseased heart after it was removed during a transplant. They then converted this heart tissue to stem cells, so they could grow more cells and study them in the lab. They determined that the patient had two different mutations of a gene, one from each parent, that normally encodes for the Rotatin protein.

When the researchers then conducted an experiment to remove this same protein from zebrafish hearts, these hearts developed with signs of heart failure. The researchers also looked at fruit fly hearts missing Rotatin and saw that the muscle cells in these hearts were disorganized and did not contract as well as they should, similar to what happens in infant hearts with the disorder.

This is the first human disease known to be caused by disrupting the transition in centrosome structure which normally occurs shortly after birth,said Matthew Miyamoto, the first co-author who worked on this project as a rising second-year medical student in Dr. Hongs laboratory. The researchers then used the drug C19 that was known to organize centrosomes in developing heart muscle cells derived from the patient with infantile dilated cardiomyopathy. The drug restored organization of the developing heart muscle cells grown in a dish from the patients stem cells and their ability to contract.

Because centrosomes play such a fundamental role in heart muscle development, specifically cell replication, structure, and function, a better understanding of this tissue-specific programmed process will be highly relevant to future cardiac regenerative therapy efforts,said UMSOM Dean,Mark T. Gladwin, MD, who is also Vice President for Medical Affairs, University of Maryland, Baltimore (UMB), and the John Z. and Akiko K. Bowers Distinguished Professor.

Dr. Hong added,It is only through collaborations between cardiologists, medical student trainees, and laboratory researchers that allowed this biomedical discovery which we hope will one day translate to medical treatments for children with this condition.

Patrice Desvigne-Nickens, MD,a medical officer in the Heart Failure and Arrhythmias Branch in the Division of Cardiovascular Sciences at the National Heart, Lung, and Blood Institute (NHLBI), part of the National Institutes of Health, agreed.This study makes an important contribution toward understanding the biological underpinnings of infantile dilated cardiomyopathy and its relationship to heart failure,she said.We look forward to future studies to clarify and confirm these findings in an effort to improve heart failure outcomes.

This study was funded by grants from the NHLBI (R01HL135129), the Maryland Stem Cell Research Fund (HP-00089001), and an AOA Carolyn L. Kuckein Student Research Fellowship.

The authors have filed a pending patent on using C19 to treat infantile dilated cardiomyopathy. In accordance with UMB policy, the authors have disclosed their interest in the patent, and the university is managing this relationship to ensure objectivity in the research.

DISCLAIMER: The content is solely the responsibility of the authors and does not necessarily represent the official views of the National Institutes of Health.

About the University of Maryland School of Medicine

Now in its third century, the University of Maryland School of Medicine was chartered in 1807 as the first public medical school in the United States.It continues today as one of the fastest growing, top-tier biomedical research enterprises in the world with 46 academic departments, centers, institutes, and programs, and a faculty of more than 3,000 physicians, scientists, and allied health professionals, including members of the National Academy of Medicineand the National Academy of Sciences, and a distinguished two-time winner of the Albert E. Lasker Award in Medical Research.With an operating budget of more than $1.3 billion, the School of Medicine works closely in partnership with the University of Maryland Medical Center and Medical System to provide research-intensive, academic, and clinically based care for nearly 2 million patients each year. The School of Medicine has nearly $600 million in extramural funding, with most of its academic departments highly ranked among all medical schools in the nation in research funding.As one of the seven professional schools that make up the University of Maryland, Baltimore campus, the School of Medicine has a total population of nearly 9,000 faculty and staff, including 2,500 students, trainees, residents, and fellows. The combined School of Medicine and Medical System (University of Maryland Medicine) has an annual budget of over $6 billion and an economic impact of nearly $20 billion on the state and local community. The School of Medicine, which ranks as the8thhighestamong public medical schools in research productivity (according to the Association of American Medical Colleges profile) is an innovator in translational medicine, with 606 active patents and 52 start-up companies.In the latestU.S. News & World Reportranking of the Best Medical Schools, published in 2021, the UM School of Medicine isranked #9among the 92 public medical schoolsin the U.S., and in the top 15 percent(#27) of all 192public and privateU.S. medical schools.The School of Medicine works locally, nationally, and globally, with research and treatment facilities in 36 countries around the world. Visitmedschool.umaryland.edu

Experimental study

Cells

Impaired Reorganization of Centrosome Structure Underlies Human Infantile Dilated Cardiomyopathy

27-Mar-2023

Read the rest here:
Research identifies new cause of heart failure condition in children - EurekAlert

Celebration of Innovation recognizes those making an impact – University Times

The Office of Innovation and Entrepreneurships 2023 Celebration of Innovation on April 25, at the Petersen Events Centers Campus View Club recognizes Pitt innovators and regional businesses who are creating solutions to difficult societal problems and serving unmet needs through the commercialization of their innovations.

This event included seven special awards; an innovation showcase where nine emerging Pitt innovation teams in the early stages of commercialization were on hand to share their progress on the path from the classroom or lab to market; and recognition of all faculty and students who submitted an invention disclosure, were issued a U.S. patent or had their innovation licensed.

The culture of innovation and entrepreneurship at Pitt is getting stronger all the time, said Evan Facher, vice chancellor for innovation and entrepreneurship and associate dean of commercial translation at the School of Medicine. This years Celebration of Innovation, in-person again for the first time since 2019, is our opportunity to bring our innovation and entrepreneurship community together to celebrate our successes, while recognizing several individuals and companies that are improving lives through their innovations.

The Innovation Award winners are:

Marlin Mickle Outstanding Innovator Award: William Wagner, director of the McGowan Institute for Regenerative Medicine as well as distinguished professor of surgery, chemical engineering and bioengineering

The Marlin Mickle Outstanding Innovator Award is presented to a Pitt faculty member who has achieved a sustained commitment to innovation throughout a distinguished career. Wagners research interests are generally in cardiovascular engineering, with projects that address medical device biocompatibility and design, hypothesis-driven biomaterials development and tissue engineering. His research has generated nearly 50 issued patents and patent filings that have resulted in significant licensing activity and the formation of three startup companies. Under his leadership, the McGowan Institute has grown into the most prolific research institutes at Pitt in terms of commercialization activity.

Emerging Innovator Award: Leah Byrne, assistant professor of ophthalmology at the School of Medicine

The Emerging Innovator Award is presented to an early-to-mid-career Pitt faculty who has demonstrated an extraordinary dedication to innovation commercialization. Byrnes research lab develops gene therapies for retinal disease. Byrnes lab engineers viral vectors with improved capabilities to deliver therapeutic genes to the retina that allow for increased precision of gene delivery and protein expression. In July 2022, the Swiss pharmaceutical company Roche partnered with Avista Therapeuticsa spinout based on Byrnes work and co-founded with Jos-Alain Sahel and Paul Sievingto further develop these therapies.

Student Innovator of the Year Award: Kunal Gandhi, a 2021 graduate of the University of Pittsburgh

Gandhi is CEO of APEX, which empowers clinicians to improve and expand access to musculoskeletal care using 3-D motion capture technology blending computer vision artificial intelligence with neuroscience to bridge in-person and virtual care. He leveraged the programs and resources of the Office of Innovation and Entrepreneurships Big Idea Center in each of his four years as an undergraduate to start on an entrepreneurial path after graduating.

James Chip Hanlon Volunteer Mentor of the Year Award: Jan Berkow, program manager for commercialization at the Pitt Center for Military Medicine Research

The Pitt Innovation Institute relies heavily on volunteer mentors to assist faculty and students interested in exploring the commercial potential of their innovations to navigate the often unfamiliar terrain of innovation commercialization. Berkow is responsible for bringing to market U.S. Department of Defense-funded advanced medical technologies. He previously led InteloMed Inc., a Pitt startup company, as co-founder and chief technology officer. For the past five years, he has served as a volunteer mentor on numerous Pitt innovation teams, helping to guide them through early commercialization exercises in customer discovery and value proposition development as part of the NSF I-Corps First Gear program, as well as assisting teams participating in the Michael G. Wells Student Healthcare Competition.

Startup of the Year: Apollo Neuroscience, Inc.

Apollo Neuroscience is a spinout from the University of Pittsburgh that began in 2018. The company has developed a wearable device and software platform technology that delivers patented vibration patterns, Vibes, that are scientifically shown to increase resilience, helping you to relax, sleep well, focus and stay energized by sending gentle sound waves to any location on the body. The Apollo Technology is based on the research of David Rabin and Greg Siegle from the Department of Psychiatry between 2014-2018. Kathryn Fantauzzi joined the team as an entrepreneurship mentor to help lead it to an award from the Innovation Institutes First Gear commercialization program. The team also received a top prize in the Michael G. Wells Student Healthcare Competition as well as the second-place prize in the inaugural Performance Innovation Tournament. The team placed as a finalist in the Pitt Innovation Challenge (PInCh) in 2016 sponsored by CTSI, only to receive the grand prize at PInCh 2017. Since spinning out in 2018, Apollo Neuroscience has successfully produced and marketed the Apollo wearable, selling more than 100,000 units to date. Apollo Neuroscience continues to be headquartered in Pittsburgh.

Small Business of the Year Award (Less than $1 million in revenue): Stories Like Me

Stories Like Me is an independently owned bookstore and community hub promoting equality, equity and inclusion. Its mission is to be the most comprehensive resource for diverse, accessible and empowered childrens literature and to share the stories of the world with a focus on generating empathy and peace through books. It recently opened a new physical bookstore at 4381 Murray Ave. in Pittsburghs Greenfield neighborhood.

Regional Business of the Year Award ($1 million+ in revenue): 84 Lumber

Founded in 1956 and headquartered in Eighty Four, Pennsylvania, 84 Lumber is the nations leading privately held supplier of building materials, manufactured components, and industry-leading services for single- and multifamily residences and commercial buildings. 84 Lumber is nationally certified through the Womens Business Enterprise National Council as a woman-owned and -operated business. It was named one of Americas largest private companies in 2018 by Forbes and a top workplace in the greater Pittsburgh region in 2018 by the Pittsburgh Post-Gazette.

Link:
Celebration of Innovation recognizes those making an impact - University Times

Gene-edited calf may reduce reliance on antimicrobials against … – EurekAlert

image:Veterinary epidemiologist Brian Vander Ley of the University of Nebraska-Lincoln's School of Veterinary Medicine and Biomedical Sciences, poses with Ginger, the first gene-edited cow resistant to bovine viral diarrheal virus (BVDV). view more

Credit: Craig Chandler|University Communication|University of Nebraska-Lincoln

Cattle worldwide face major health threats from a highly infectious viral disease that decades of vaccinations and other precautions have failed to contain. Federal, private-sector and Husker scientists are collaborating on a new line of defense, by producing a gene-edited calf resistant to the virus.

If follow-up research confirms its efficacy, the gene-editing approach offers long-term potential to reduce antimicrobial and antibiotic use in the cattle industry.

The bovine viral diarrhea virus (BVDV) devastates the bovine immune system and can cause severe respiratory and intestinal harm to infected beef and dairy cattle, said veterinary epidemiologist Brian Vander Ley, an associate professor in the University of Nebraska-Lincolns School of Veterinary Medicine and Biomedical Sciences.

In utero calves are especially vulnerable to infection. If they survive, they can remain infected for life, repeatedly spreading the virus to other cattle.

They show up as normal cattle but really, theyre shedding a tremendous amount of virus. Theyre the Typhoid Marys of BVDV spread, said Vander Ley, assistant director of UNLs Great Plains Veterinary Educational Center in Clay Center.

The cattle industry has vaccinated against the disease since the 1960s, but the highly mutable nature of BVDV and the emergence of highly virulent strains of BVDV contribute to limited success of present control programs, the Academy of Veterinary Consultants has stated.

Scientists identified the specific genetic structure associated with the disease earlier this century. A collaborative project involving scientists with the USDAs Agricultural Research Service and Acceligen, a Minnesota-based private company, used gene editing to change the small number of amino acids that lead to BVDV vulnerability, while keeping the rest of the protein, CD46, unchanged.

Our objective was to use gene-editing technology to slightly alter CD46 so it wouldn't bind the virus yet would retain all its normal bovine functions, said Aspen Workman, a scientist with ARS U.S. Meat Animal Research Center (USMARC) in Clay Center, Nebraska.

A gene-edited calf, named Ginger, was born on July 19, 2021, and was transported to UNL a week later for close monitoring by Vander Ley. Throughout, Ginger has remained a bright, healthy calf, normal both physically and behaviorally, which included a week with a BVDV-infected dairy calf that was shedding the virus in great volume.

The research findings will be published online May 9, by the PNAS Nexus open-access journal, a sibling publication to the Proceedings of the National Academy of Sciences. Workman is lead author.

Ginger is a Gir, a tropically adapted cattle breed used to develop Brahman cattle in North America. Follow-up research will require experimental replication in other cattle breeds. Ginger also will be monitored through pregnancy, if it occurs.

If the gene-editing approach proves viable, it could potentially reduce the cattle sectors use of antimicrobials, Vander Ley said.

The most successful version of the future that I can see is one where we don't have to deal with antimicrobial resistance because we just don't use that many antimicrobials, he said. That's better for everyone. That means that we have eliminated the cause of a lot of the antimicrobial use and we've eliminated that expense for livestock producers.

Michael Heaton, a USMARC researcher for the BVDV project, concurred. This line of research represents another opportunity to lessen the need for antibiotics in agriculture, he said.

In addition to Vander Ley, Workman and Heaton, other study coauthors are Erin E. Jobman (Great Plains Veterinary Educational Center); Gregory P. Harhay (USMARC); private-sector scientists Tad S. Sonstegard, Dennis A. Webster, Luke Sherry, Sabreena Larson, Daniel F. Carlson and Jonathan Bostrom; and Theodore S. Kalbfleisch with the University of Kentucky.

Experimental study

Animals

First gene-edited calf with reduced susceptibility to a major viral pathogen

9-May-2023

Co-authors D.W., J.B. and D.C. are full-time employees of Recombinetics, Inc. S.L. and T.S. are employees of Acceligen, a wholly owned subsidiary of Recombinetics, Inc. Recombinetics, Inc., is a company that commercializes animal gene editing and associated applied technologies for biomedical research, regenerative medicine and animal agriculture. There are no patents to declare, and the interests do not alter the authors' adherence to all the journal's policies on sharing data and materials published herein.

Disclaimer: AAAS and EurekAlert! are not responsible for the accuracy of news releases posted to EurekAlert! by contributing institutions or for the use of any information through the EurekAlert system.

The rest is here:
Gene-edited calf may reduce reliance on antimicrobials against ... - EurekAlert

Researchers discover novel ‘Shanghai APP’ mutation in late-onset Alzheimer’s disease – Medical Xpress

This article has been reviewed according to ScienceX's editorial process and policies. Editors have highlighted the following attributes while ensuring the content's credibility:

by Chongqing Medical University

Credit: Gang Wang from Ruijin Hospital, affiliated to Shanghai Jiao Tong University School of Medicine

Alzheimer's disease (AD) is a progressive neurodegenerative disorder affecting tens of millions of people worldwide, and it is the most common cause of dementia. Early-onset AD is typically associated with mutations in the genes APP, PSEN1, and PSEN2, leading to a more aggressive form of the disease with atypical symptoms. In contrast, the newly discovered "Shanghai APP" mutation has been linked to LOAD, which affects a larger population of AD patients.

In a study published in Genes & Diseases, researchers from Ruijin Hospital, affiliated to Shanghai Jiao Tong University School of Medicine and other three teams detected the Shanghai APP mutation in a Chinese patient who developed memory decline in his mid-70s. Neuroimaging techniques confirmed the presence of widespread amyloid deposition, a key hallmark of AD.

Using molecular dynamics simulation and in vitro experiments, the team found that the E674Q mutation led to increased processing of APP and production of amyloid , a toxic protein linked to AD. Additionally, the biochemical aggregation experiments suggested that the E674Q peptide exhibited higher aggregation than the wild-type peptide, especially the formation of filaments that hinged several fibrils.

To further investigate the mutation's effects in vivo, the researchers introduced the E674Q mutant APP gene into the hippocampi of two-month-old mice using adeno-associated virus (AAV) gene transfer. The study revealed that the E674Q mutation resulted in impaired learning behavior and increased pathological burden in the mouse model, demonstrating its pathogenic role in AD.

The E674Q substitution exhibited a strong amyloidogenic effect, and, to the researchers' knowledge, it is the only known pathogenic mutation within the amyloid processing sequence causing LOAD. This finding is significant, as it may open up new avenues for understanding the development of LOAD and lead to more effective treatments for patients suffering from this form of Alzheimer's disease.

The discovery of the novel Shanghai APP mutation provides a unique opportunity to delve deeper into the molecular mechanisms underlying LOAD. Further research into the effects of the E674Q mutation is essential to explore the potential development of targeted therapies or interventions that may slow or halt the progression of AD.

By understanding how this specific mutation contributes to the onset and progression of LOAD, scientists may be able to devise new strategies for preventing or treating this devastating disease, ultimately improving the quality of life for tens of millions of patients and their families.

More information: Yongfang Zhang et al, E674Q (Shanghai APP mutant), a novel amyloid precursor protein mutation, in familial late-onset Alzheimer's disease, Genes & Diseases (2023). DOI: 10.1016/j.gendis.2023.02.051

Provided by Chongqing Medical University

See the original post here:
Researchers discover novel 'Shanghai APP' mutation in late-onset Alzheimer's disease - Medical Xpress

People on the Move: May 12 – Delaware Business Times

People on the Move is a rundown of recent hirings, promotions, appointments and other notable movements by professionals in the state. If youre interested in submitting an entry, please contact news@delawarebusinesstimes.com.

U.S. Senator Tom Carper (D-Del.) was named to the National Advisory Board for President Joe Bidens re-election campaign. The National Advisory Board is a select group of national Democratic leaders who will serve as the primary surrogates for the Democratic National Committee and Biden-Harris campaign in 2023 and 2024.

U.S. Senator Tom Carper (D-Delaware)

Im excited to join President Bidens national advisory board to stand beside my good friend Joe throughout his re-election campaign. Since they took office, President Biden and Vice President Harris have been fighting for everyday Americans and to protect our freedoms,Senator Tom Carper said in a statement. President Biden ran for the presidency in 2020 because he believed that we were in a battle for the soul of this nation and we still are. I encourage all Delawareans to join me in supporting the Biden-Harris campaign so we can all come together to help Joe and Kamala finish the job.

The stakes of this election couldnt be higher, President Joe Biden said. Our freedom and democracy are on the line. Im grateful to this group of diverse and dynamic leaders who will help us win the battle for the soul of America and finish the job for the American people.

Gawthrop Greenwood partner Carl W. Heckert has been elected secretary of the Delaware State Bar Associations Family Law Section during the esteemed organizations 100th anniversary year.

Heckert, who has more than 25 years of experiencepracticing family law in Delaware, has been a member of the Delaware State Bar Association since 1992 and will serve for the 2023-2024 section year.

Carl Heckert | PHOTO COURTESY OF GAWTHROP

Heckert is a member of theFamily Law Departmentin Gawthrop Greenwood, PCs Greater Wilmington office in Greenville, litigating and mediating divorce. In 2019, he became a Certified Family Law Mediator in Delaware following the states inaugural certification program by the Family Court of the State of Delaware as well as the Delaware State Bar Association. A certified Delaware family law mediator is a neutral third party who is charged with preserving confidentiality while helping couples resolve issues more creatively and amicably than they can in court, including custody, visitation, child support, property division and alimony.

Heckert also draws on his more than 30 years of real estate experience to settle real estate disputes during divorce proceedings. In January, he presented a seminar on the topic for Delaware family law judges and practitioners of theMelson-Arsht Inns of Court. He has also led Continuing Legal Education courses in family law matters including custody, protection from abuse and representing an unwilling minor as a guardian-ad-litem. Heckert received his law degree fromWidener University School of Lawand holds a Bachelor of Science degree from theUniversity of Delaware.

Wilmington Trust announced the addition ofSuzanne Lane as the senior relationship executive.

Lane will focus on asset managers and financial institution relationships for the loan market solutions team, employing innovative solutions to help meet individual clients goals. Throughout her 25-year career in corporate trust and banking, she has taken numerous leadership positions on multiple finance and asset management teams.

Lane is a passionate advocate about empowering women in the financial services industry and has served as a mentor for many women as they start or build their careers. Prior to joining Wilmington Trust, Lane was a relationship lead for U.S. Banks Global Platinum Corporate Trust clientele.Previously, she was Chief of Staff for Relationship Management in the Shareowner Services division of BNY Mellon. She also has held roles at KPMG, AIB, and State Street Bank.

Lane has managed large client service teams both domestically and in Europe. She holds a bachelors degree in business administration from Northeastern University, and has earned the CPA, CFA and CAIA designations.

Allen Friedland, M.D., MACP, FAAP, has been recognized with the Dema C. Daley Founders Award for his national impact as an educator, innovator and leader in internal medicine.

Allen Friedland | PHOTO COURTESY OF CHRISTIANACARE

Given by the Alliance for Academic Internal Medicine, the award honors internal medicine doctors who have greatly influenced undergraduate and graduate medical education and the development of training programs in internal medicine.

Dr. Friedland has been a tireless advocate and innovator for internal medicine, medicine-pediatrics, and residency education over two decades, said Vinay Maheshwari, M.D., MHCDS, Hugh R. Sharp Jr., Chair of Medicine and physician executive of the Medical Group at ChristianaCare. He is a mentor to countless physicians across the country and beloved by those who have had the privilege of working alongside him. What is most evident about Dr. Friedland is how much he cares about those he has taught locally and nationally all of them will forever be a part of his family. This award is a testament to a lifetime career dedicated to graduate medical education.

Friedland is ChristianaCares section chief of Medicine-Pediatrics (Med-Peds) and has served as the director of ChristianaCares combined Internal Medicine-Pediatrics residency program for 25 years. He also teaches medical students at ChristianaCare, which is a branch campus of Sidney Kimmel Medical College at Thomas Jefferson University and Philadelphia College of Osteopathic Medicine.

His accomplishments include:

Amanda Hewes, MS, education program manager at ChristianaCares Gene Editing Institute, has been named one of the 2023 Outstanding Delaware Women in STEM by Million Women Mentors, an international movement dedicated to encouraging girls and women to pursue careers in science, technology, engineering and math (STEM).

Amanda Hewes | PHOTO COURTESY OF CHRISTIANACARE

Hewes selection spotlights her dedication to engaging young people in the science of gene editing by introducing the Gene Editing Institutes CRISPR in a BoxTM educational toolkit into classrooms across Delaware and her commitment to bridging disparities in STEM education.

Im overjoyed to be honored among so many amazing women in this state, Hewes said. Its humbling to be considered and to stand alongside them. All of these women foster and lead dynamic communities of young women that inspire me every day. I hope that I can do the same by making young women in this state feel empowered through the work that I do.

Hewes joined ChristianaCares Gene Editing Institute in 2017 with a focus on expanding its CRISPR gene editing system in a cell-free environment. She was first author in a publication in Nature that established the highly innovative gene editing on a chip protocol that allowed CRISPR to edit DNA outside of the cell for the first time. This methodology enables researchers to take fragments of DNA extracted from human cells, place them in a test tube and precisely engineer multiple changes to the genetic code.

This gene editing system eventually led to the creation of the CRISPR in a Box toolkit. This innovative educational resource provides a way for students to learn about this exciting frontier of science through a hands-on exercise in which they use CRISPR gene editing to disrupt a synthetic gene within a plasmid. The simplicity of this experiment allowed for the reaction to be developed into a remarkable teaching tool that can be brought into most school laboratories containing basic laboratory equipment.

Once CRISPR in a Box was developed, Hewes recognized the potential it could have for high school and college students. She took on a new role as education program manager and expanded the Gene Editing 360 platform, which is the Gene Editing Institutes suite of educational tools for engaging students and the public.

Hewes was honored alongside 10 other women by Gov. John Carney, Lt. Gov. Bethany Hall-Long and others at the Delaware State House with the signing of a proclamation to declare March 24, 2023, as Delaware Women and Girls in STEM Day.

After 35 years of wearing Service Unlimited, Inc. (SUI) green, Safety Coordinator David Parag announced his retirement closing out a remarkable career serving the company and the people of New Castle, Delaware, and the surrounding areas.

Originally hired in the mid-1970s as an HVAC mechanic, Parag has served our customers in many different roles over three different terms. In his second term with the SUI family, Parag served as an electrician for our sister company, Electric Unlimited, Inc. (EUI), and ultimately led the company. Parag again returned to SUI as the Director of Construction Quality. He eventually added safety responsibilities to that role. Parag most recently was Safety Coordinator because of his passion for doing things the safest way rather than the fastest or easiest way.

Parag holds a Master License in both HVAC and Electrical. He has conducted countless training sessions, delivered tons of how-to documents, and mentored dozens of team members including Vice President Brian Martinenza. Parag will always be known around SUI for his oversized brain and his oversized heart because he knows virtually everything, and cares deeply about the people in his life. He enjoys being a catalyst for personal development and professional growth.

Continue reading here:
People on the Move: May 12 - Delaware Business Times

Historic $100 Million Gift to Brigham and Womens Hospital to Establish Institute of Immunology and Inflammation of the Brigham, Massachusetts General…

BioLegend Founder Gene Lay makes transformational gift to address steep rise in cancer and other immune-mediated and inflammatory diseases

BOSTON, May 10, 2023--(BUSINESS WIRE)--Brigham and Womens Hospital, a founding member of Mass General Brigham, today announces a historic $100 million gift from eminent biotechnology entrepreneur Gene Lay, MS, DVM, founder and CEO of BioLegend, Inc., through the Laygend Foundation. The landmark giftthe largest in the Brighams historywill establish The Gene Lay Institute of Immunology and Inflammation of Brigham and Womens Hospital, Massachusetts General Hospital (MGH), also a founding member of Mass General Brigham, and Harvard Medical School (HMS). Vijay Kuchroo, DVM, PhD, an immunologist and principal investigator at the Brigham, will serve as inaugural director of the institute, which will be located at and administered by the Brigham. Arlene Sharpe, MD, PhD, chair of the Department of Immunology at HMS, and Ramnik Xavier, MD, PhD, director of the Center for Computational and Integrative Biology at MGH, will serve as the institutes vice directors.

"I have always been fascinated by the immune systems ability to heal the body," says Lay. "And Ive had a longstanding relationship with and respect for the scientists in the Harvard medical community, who played an instrumental role in my career. With this gift, I am bringing together the best scientific minds I know to translate research discoveries into therapies for immune-mediated diseases rooted in chronic inflammation."

With a philanthropic investment of $100 million, the new institute will leverage the collective strengths of Boston and HMS-affiliated immunology and biomedical experts and their collaborations with some of the worlds most distinguished scientists. Kuchroo will lead day-to-day operations, convening core and affiliated faculty and trainees around three central platforms: basic science, translational science, and technology. The primary areas of research will include basic understanding of immune-mediated diseases, aging, and cancer and translation of this knowledge to the development of new immunotherapies. In addition, the Gene Lay Institute will provide substantial training opportunities for students and fellows to support immunology innovators of the future.

Story continues

"We are honored to receive this historic and visionary gift from Dr. Lay, who is deeply committed to solving an emerging trend in medicinethe rise of chronic inflammation associated with increased levels of disease and suffering," says Robert S.D. Higgins, MD, MSHA, president of the Brigham and executive vice president of Mass General Brigham. "This will amplify our ability to improve the quality of patients lives across the country and around the world. Were thrilled to be part of this pivotal moment in the field of medicine."

"During the past several decades, the astounding rise in inflammatory diseases has made it increasingly clear that chronic inflammation is a root cause of many diseasesnot only allergies and autoimmune diseases, but also cancer and neurodegenerative, cardiovascular, and metabolic diseases," says Kuchroo. "This transformative gift and investment will allow some of the best immunologists of our time to address this epidemic head-on and build new knowledge to promote basic understanding for preventing and treating immune-mediated diseases."

Adds George Q. Daley, MD, PhD, dean of HMS, "As weve seen with the COVID-19 pandemic, collaboration across institutions, disciplines, and modes of scientific inquiry is key to addressing our most confounding scientific and medical challenges. Were deeply grateful for this opportunity to convene the best immunologists in the world to bring solutions to patients and families."

Lay is the founder of San Diego-based BioLegend, now part of Revvity, Inc., which is a global life sciences and diagnostics company. BioLegend focuses on the development and production of high-quality antibodies, proteins, and assays for cellular immunity, inflammation, cancer, stem cells, and other reagents required for research and diagnosis. Since its founding in 2002, BioLegend has expanded its reach across the globe, with research and development facilities in Taiwan and Japan, as well as subsidiaries in Taiwan, Japan, China, Germany, the United Kingdom, the Netherlands, and France. Lay continues to give back to the scientific community while pursuing his dream of cultivating more expertise in Taiwan, the country where he was born.

About Brigham and Women's Hospital

Brigham and Womens Hospital is a founding member of Mass General Brigham and a teaching affiliate of Harvard Medical School. With nearly 1,000 inpatient beds, approximately 50,000 inpatient stays, and over 2.6 million outpatient encounters annually, clinicians across the Brigham provide compassionate, high-quality care in virtually every medical and surgical specialty to patients locally, regionally, nationally and around the world. An international leader in basic, clinical, and translational research, Brigham and Womens Hospital has nearly 5,000 scientists, including physician-investigators, renowned biomedical researchers and faculty supported by nearly $750 million in funding. The Brighams medical preeminence and service to the community dates to 1832, with the opening of the Boston Lying In, one of the nation's first maternity hospitals designed to care for women unable to afford in-home medical care. Its merger with the Free Hospital for Women resulted in the Boston Hospital for Women in 1966. In 1980, the Boston Hospital for Women, the Peter Bent Brigham Hospital and the Robert Breck Brigham Hospital officially merged to become Brigham and Womens Hospital. With nearly 21,000 employees across the Brigham family including the Brigham and Womens Physicians Organization and Brigham and Womens Faulkner Hospital that rich history is the foundation for our commitment to providing superb care for some of the most complex cases, pursuing breakthroughs in biomedical research, training the next generation of health care providers, and serving the local and global community.

View source version on businesswire.com: https://www.businesswire.com/news/home/20230510005276/en/

Contacts

Jessica Pastore 617-874-6346

Excerpt from:
Historic $100 Million Gift to Brigham and Womens Hospital to Establish Institute of Immunology and Inflammation of the Brigham, Massachusetts General...

Prevalence of BRCA homopolymeric indels in an ION Torrent-based … – Nature.com

Patients cohort

Among consecutive patients who underwent BRCA tumour testing through ION Torrent-based sequencing between August 2017 and February 2022, we retrospectively selected 222 high-grade ovarian cancer (HGOC) patients with the following histological subtypes: 203 serous (HGSOC), seven endometrioid, five clear-cell and seven with mixed histotypes.

Since NGS BRCA1/2 tumour testing was not available before 2017 in our Institution, 19 of 222 subjects underwent germline testing before tumour sequencing based on personal (very early age at diagnosis/previous breast cancer) or family history. According to the workflow used by our Molecular Tumour Board (MTB), in 73 out of 203 patients with upfront tumour testing subsequently received genetic counselling, either for targeted germline sequencing of a tumour-detected PV or for large genomic rearrangement analysis14.

The Ethics committee of Fondazione IRCCS Istituto Nazionale dei Tumori of Milan approved the use of both clinical and molecular data collected by the MTB for clinical studies and granted exemption from requiring written consent for tumour genetic testing from the patients, as these analyses were carried out in the context of a diagnostic and care setting (Approval Number INT 227/20). All the probands who underwent germline testing were aged over 18 and provided signed informed consent for the use of their biological samples and data for both diagnostic and research purposes. All methods were carried out in accordance with relevant guidelines and with the ethical principles of the Declaration of Helsinki.

The BRCA1 and BRCA2 genes were assessed by in-house NGS testing using the Oncomine BRCA Research Assay (Thermo Fisher Scientific, Inc). This assay provided a 100% coverage of all BRCA1 and BRCA2 exons, with an average of 64 bases of intronic flanking sequences upstream and downstream of each exon. Five m sections from formalinfixed paraffin-embedded (FFPE) samples were manually microdissected to isolate the highest percentage of neoplastic cells. Genomic DNA was extracted with protease K (incubation ON at 55C) and quantified with Qubit dsDNA BR kit (Thermo Fisher Scientific, Inc). The libraries were prepared with the IonAmpliSeq Library kit 2.0 (Thermo Fisher Scientific, Inc) and quantified with Qubit dsDNA HS kit (Thermo Fisher Scientific, Inc) following the manufacturers instructions. The libraries are diluted to 25pm, pooled and loaded on the Ion Chef to perform emulsion PCR and chip loading on 318 v2 chips. Sequencing was performed on ION PGM, using the HI-Q view Chef kit, according to the manufacturers instructions. Data were processed using the Torrent Suite 5.12.3 (TS). The quality of sequencing output was first evaluated through the plugin Coverage Analysis on the TS. Only samples whose librarys uniformity and on-target values were at least 80% and with a medium coverage of 1500X were considered valid. SNV analysis was performed in duplicate: the first variant calling was generated by the Variant Caller plugin from the TS and the resulting VCF file was loaded in the Variant Effect Predictor Tool (Ensembl, Version GRCh37) for the variants annotation. To eliminate erroneous base calling, we set each variant coverage>40X, a variant frequency on each sample>2% and a quality value>30. Variants within homopolymer (HP) longer than eight bases and with strand bias80% were not reported. In the second analysis, the BAM files were automatically uploaded from the TS to the Ion Reporter Software (IR, version 5.6 to 5.16) and the variant calling was integrated into the analysis pipeline Oncomine BRCA Research Somatic318. The results of both analyses were manually compared. Each variant was displayed on IGV (ver. 2.3.97). Synonymous variants were filtered out, while the remaining variants were classified into pathogenicity classes according to the Evidence-based Network for the Interpretation of Mutant Alleles (ENIGMA) consortium guidelines (https://enigmaconsortium.org/). Our assay could not reliably detect large intragenic rearrangements.

Two EDTA tubes of peripheral blood samples were collected from each patient who performed genetic counselling and was eligible for germline testing, either for targeted sequencing of tumour-detected pathogenic/likely pathogenic variants or for the analysis of large genomic rearrangements in patients with no actionable variants detected at tumour testing. Whole blood DNA was isolated through the MagCore Super automatic workstation with the MagCore Genomic DNA Whole Blood Kit (Diatech LabLine SRL, Jesi, Italy). Targeted Sanger sequencing of tumour-detected BRCA1/2 PVs was performed on purified PCR products by using BigDye Terminator v.3.1 Cycle Sequencing kit (Thermo Fisher Scientific, Inc.) and run on 3730Xl DNA Analyzer (Applied Biosystems; Thermo Fisher Scientific, Inc.), after purification with Agencourt CleanSeq-Beckman Coulter. Sequences were analysed by Mutation Surveyor Software (v5.0.1; SoftGenetics, LLC., State College, PA, USA). Targeted sequencing results were confirmed on both blood aliquots collected from each patient. Variants of uncertain clinical significance identified at tumour testing were not systematically investigated at the germline level. Eligible probands, who resulted negative at tumour testing with the Oncomine BRCA assay, were analysed for large deletions and duplications of BRCA1 and BRCA2 on blood DNA with the SALSA MLPA kits P045 BRCA2/CHEK2 and P002 BRCA1 probe mix (MRC-Holland, Amsterdam, the Netherlands), following the manufacturers instructions. MLPA products were run on the 3730Xl DNA Analyzer (Applied Biosystems; Thermo Fisher Scientific, Inc.) with the Gene Mapper Module (Applied Biosystems; Thermo Fisher Scientific, Inc.). The results were analysed through the Gene Marker Software v2.7.0 (SoftGenetics, LLC, State College, PA, USA).

Based on previous observations on the performance at homopolymers of ion semiconductor sequencing techniques, we focussed our analysis on stretches of six or more identical bases since the calling accuracy has been consistently shown to dramatically drop beyond this length15,17,25,28,30. We thus selected all 29 homopolymeric regions exceeding five repetitions to be analysed within the coding regions of both genes, including five in BRCA1 and 24 in BRCA2. Since truncating variants beyond codon 3326 of BRCA2 are not classified as high-risk variants, homopolymers downstream of the residue c.9976 of BRCA2 were not included in the analysis (Table 2).

To overcome the limitations of the ION reporter software, which filters out most indels at homopolymeric regions, we manually visualised the BAM alignment files of the 222 patients at the 29 regions with the IGV software (ver. 2.3.97). The median depth of coverage of the regions of interest ranged from 1045 to 6989X. Each sample showed a variable frequency of sequence alterations (both insertions and deletions) at each region. We estimated the variant allele frequency (VAF) of insertions and deletions (indels) by calculating the ratios of the maximum inserted or deleted reads over the total reads at each homopolymer (Suppl. Table 1).

Since the VAF of indels at homopolymeric regions has, in general, a left-skewed distribution, we employed a modified version of the Cancer Outlier Profile Analysis (COPA) approach31, which consists in scaling the above-cited to a normal distribution and subsequently calculating the outliers that exceeded the mean+3 median-adjusted deviations (+3) threshold.

To validate the outliers, for each of the 29 regions, we further defined a threshold based on the normalized distributions (either for percentage of insertion or deletion) of a control population. Since both in ovarian and other BRCA-associated cancers the predominant second hit is most often represented by loss of heterozygosity (LOH), while a second point mutation is an extremely rare event32,33,34, we used as control population a cohort of 46 patients in which a non-homopolymeric PV (either somatic or germline) had been already identified.

To avoid potential selection bias, which would affect the estimated frequency of pathogenic variants occurring at homopolymeric regions in our cohort, we excluded from the analysis the 19 patients who underwent germline testing before tumour testing. This group also included two patients with germline-confirmed homopolymeric PVs who resulted negative at tumour testing.

Therefore, we applied the (+3) thresholds estimated on the control population to the normalized distributions of the study cohort, composed of 157 individuals with no evidence of pathogenic variants at tumour testing with ION Torrent. In addition, according with filtering criteria used in a previous study, which focused on germline variants29, we considered only homopolymeric indels with an absolute VAF above 15% and in any case higher than the maximum value of the control population at each homopolymeric region. Lastly, regions with a total read count of less than 100 were excluded from the analysis.

Targeted Sanger sequencing was performed on tumour DNA to confirm the occurrence of outlier homopolymeric indels selected by using the defined thresholds.

More:
Prevalence of BRCA homopolymeric indels in an ION Torrent-based ... - Nature.com

Thanks to UVA Research, You Might Live to 120. But Can Society … – UVA Today

Its perhaps ironic then that ORourke, in sharing the story behind the research, is among those currently thinking aloud about the pros and cons of engineering dramatically longer lives.

The researcher said firmly, Theres a discussion around anti-aging therapeutics that society needs to have.

Scientists have known for more than a decade now that several genetic and biochemical pathways can either extend or shorten a persons life. The long answer to why the genome evolved this way is complex, ORourke said. The short answer resides in reproduction.

Nature cares about organisms becoming healthy reproductive adults, she said. But once we have produced as many babies as we are capable of, nature doesnt care about our health.

Should our muscles atrophy, or remain firm and tight? We can obviously improve our muscle tone by exercise, for example. But at some juncture, instructions meant to optimize development and survival earlier in life begin to tell the body to slow down. Obsolescence is in our genetic programming.

In that regard, our bodies are complex communication networks. The inputs and middle links in aging communication were identified prior to the UVA research. And acting on that accumulated knowledge, previous scientists indeed figured out ways to slow down, and even reverse, aging in animals such as lab mice.

But one reason you may not have heard about all the developments has been the ugly tradeoffs: compromised immunity, cancer.

The problem with playing around with many of the input and middle-link genes is that as they are such important players in the cell and control so many things it is very hard or even impossible to find a condition in which you can only get the good effects of changing their activities, ORourke explained.

In her attempt to solve this problem, ORourke assembled a Hoos-who of UVA scientists.

The team that yielded the revelatory research was led by biology graduate student Abbas Ghaddar and postdoctoral fellow Vinod Mony, with the contributions of graduate students Swarup Mishra, Elisa Enriquez-Hesles and Mary Kate Horak; undergraduate students Samuel Berhanu, Emma Harrison, James C. Johnson and Aaroh Patel; and aging expert Jeffrey S. Smith, a professor of biochemistry and molecular genetics.

Traditionally, worms have been associated with death. In science, however, worms in particular, roundworms have been responsible for some major health breakthroughs, winning Nobel Prizes in physiology and medicine (along with their scientists).

The type of roundworm ORourke and company used wasnt the parasitic type sometimes found in our pets, but rather C. elegans, which grows to about a millimeter long and is clear-bodied. As the first multicellular organism to have had its entire genome sequenced, the roundworm is transparent in more than one way. The creepy-crawly may seem far removed from us humans, but its chemical pathways are remarkably analogous.

Using the worm as a key model for this research, the team sought to decipher what happens at the end of those previously mentioned communication chains that control aging (as opposed to the inputs or middle links).

Specifically, ORourke wanted to find the molecular players most responsible for aging, which are those that break or repair cells, and by extension, tissues and organs. The thought was that by being at the end of the communication chain, playing around with the genes might mean fewer unwanted effects.

They set out by looking at biologys natural process of cellular cleanup and repair, called autophagy. The command to renew cells has long been thought to underlie longevity.

Autophagy is a process that clears the unwanted and recycles parts of the cells, ORourke said. When cell components go bad, they need to be disposed of. To this end, autophagy breaks them down to use the parts to make new cell components.

So autophagy was the main anti-aging candidate, in particular because we had already defined that the mid-link gene we were studying acted as a switch to turn autophagy on when animals were fasting, a dietary intervention that extends lifespan.

If their hypothesis was correct and autophagy was promoting longevity, then by stopping it, animals would not live longer.

Read the original post:
Thanks to UVA Research, You Might Live to 120. But Can Society ... - UVA Today

Article from Cloud-based Medicine Studio A national authoritative medical organization for rare diseases will be established, and the second list of…

Article from Cloud-based Medicine Studio A national authoritative medical organization for rare diseases will be established, and the second list of rare diseases will be updated  Marketscreener.com

See original here:
Article from Cloud-based Medicine Studio A national authoritative medical organization for rare diseases will be established, and the second list of...

‘The luckiest of the unlucky’: A Duchenne gene therapy brings hope … – BioPharma Dive

The diagnosis was news no parent ever wants to receive. Susan and Chris Finazzo heard it twice in two months.

Both of their children, Chase and Dylan, have a genetic disease called Duchenne muscular dystrophy. The progressive condition slowly and unstoppably lays waste to muscles, first stealing away the ability to walk, then weakening the lungs and heart.

The hopes they held for their childrens future were darkened by fears of a shortened life, and dread of when Chase and Dylan would no longer be able to climb a playground slide, dress independently or swallow food.

Youre just sitting there like, this is not what was supposed to happen, Susan said. Youre mourning the death of the life that you thought your child was going to have.

The Finazzos didnt accept the outlook doctors gave their sons, though. They trawled through the internet looking for answers to their many questions. They met with patient advocates to learn about experimental medicines being developed, and enrolled Chase and Dylan in a clinical trial of a cutting-edge gene therapy.

We always say were the luckiest of the unlucky, she said. This gives us hope.

The Finazzos are part of a large network of parents and patient advocates who believe the gene therapy, developed by biotechnology company Sarepta Therapeutics, represents a medical breakthrough for Duchenne. The treatment is an infusion meant to change the course of the disease for years, if not permanently. Available medicines, by comparison, are thought to only slow Duchennes relentless advance, not halt it, as a gene therapy might.

There isnt a parent, family or person with this diagnosis that doesn't go to bed every night and say, Stop the disease right here. Let it be stopped, said Pat Furlong, president and CEO of Parent Project Muscular Dystrophy, a prominent patient advocacy group. The community believes this is a very important step forward.

Sarepta has asked the Food and Drug Administration for an accelerated approval, a type of clearance thats used to speed treatments for serious diseases like Duchenne to market. The companys case is supported by data showing its drug produces large amounts of a potentially helpful protein, as well as signs some study participants are doing better than medical history suggests they should.

I am not, as I sit here today, aware of an approved therapy under the accelerated approval pathway with more compelling evidence than we have, said Sarepta CEO Doug Ingram.

Yet the treatment failed an important part of its only placebo-controlled test to date. The effects of the protein it helps produce, a diminutive molecule called microdystrophin, arent fully understood. Nor is how long any benefit might last. An approval, if granted, would set a precedent for other gene therapy developers to follow, raising the stakes of the agencys decision.

The FDA is expected to issue its verdict by the end of May, about six months before a Phase 3 study will deliver results that either confirm or refute the benefit of a treatment likely to cost more than $1 million. The regulator is convening a group of outside experts on Friday to discuss it a meeting reportedly scheduled because of intra-agency disagreement about the strength of Sareptas application.

This is a very, between a rock and a hard place situation for the FDA, said Dae Gon Ha, an analyst at the investment bank Stifel, who covers gene therapy companies.

Tim Revell embraces his then 8-year-old son Timothy after running the Austin Marathon.

Permission granted by Tim Revell

Seventeen years ago, Tim Revells son Timothy was having trouble walking.

A whirlwind of brain and blood tests followed, confirming that Duchenne was why Timothy, then 2, had fallen behind his peers. The Revells doctor told Tim that all he could do was go home and love his son.

Timothy is one of an estimated 300,000 people worldwide, almost exclusively boys, who have the condition, which is caused by a genetic mutation that stops the body from making a muscle-protecting protein called dystrophin. A few years later, the Revells learned Timothys younger brother had it too.

Parents of children with Duchenne have long received the same advice as Revell heard in 2006. Theres zero hope, he recalls being told.

Revell became an advocate for drug research and got his sons into earlier clinical trials of Duchenne medicines that ultimately proved unhelpful. In the meantime, he's watched the disease slowly take hold. Timothy lost the ability to walk three years ago.

Duchenne is like death by 1,000 cuts, he said. After years of decline, death often occurs in young adulthood, when the muscles of the heart or lungs fail.

There are a few treatments available. Some patients with particular mutations can get drugs known as exon-skippers, which help the body produce a shortened form of dystrophin thats thought but not proven to modestly slow progression. Others, like Revells sons, arent eligible.

However, most rely on steroids, which can slow muscle damage but cause other problems like weight gain, weak bones and behavioral changes.

It's not a great choice, and it's not an easy one for parents to make, said Jennifer Handt, whose 5-year-old son Charlie has Duchenne. The expectation is to hopefully buy a little bit of time until science can give us something more tangible.

For decades, patient advocates have put their hopes in gene therapy, a way of shuttling replacements for missing or damaged genes into the body to restore needed proteins like dystrophin. Until recently, it had always seemed just out of reach.

I remember in my first meeting 30 years ago, it was raised, At some point we'll have gene therapy, said PPMDs Furlong, and it sounded way back then like, Oh my gosh, what will it take to get there?

Necrotic muscle fiber is associated with Duchenne.

Jose Luis Calvo Martin, Jose Enrique Garcia-Maurino Muzquiz via Getty Images

Jerry Mendell saw his first Duchenne patient in 1969, during a postdoctoral fellowship at the National Institutes of Health.

We didn't know anything about the disease then, he said. Just what it looked like under a microscope.

What started as a fellowship became a lifelong mission for Mendell, who in the following decades emerged as a top researcher in the field of neuromuscular disease.

In 1989, he and his team at Ohio State University published research that established the steroid prednisone as the standard of care for Duchenne. In the late 1990s, he was the first to test a gene therapy for another neuromuscular condition in humans and later conducted an early gene therapy experiment in Duchenne. More than a decade later, after Mendell became director of gene therapy research at Nationwide Childrens Hospital, the team he assembled invented Zolgensma, a dramatically beneficial medicine for infants born with the rare condition spinal muscular atrophy.

I understood the potential for gene therapy if we could make it work for Duchenne, he said. There were many scientists and clinicians who doubted it, but to me it was the best approach we had.

Duchenne presents several vexing scientific problems, however.

Jerry Mendell

Permission granted by Nationwide Children's Hospital

The disease has been seen as a target for gene therapy since the 1980s, when its genetic roots were first identified. But the gene that encodes for dystrophin is too large to be packaged into the microscopic viruses researchers use to deliver corrective gene therapy. And because Duchenne affects muscle, researchers need to use very high doses to shuttle in enough genetic material to hope for a benefit.

This disease is a significant adversary, said John Brandsema, a pediatric neurologist at the Childrens Hospital of Philadelphia. We have been hammering away at it for decades.

Scientists have spent years searching for workarounds, dissecting the gene and trying to figure out how to make it small enough to fit into the virus, Mendell said.

The solution was found in people with a milder form of the condition known as Becker muscular dystrophy. Patients with Becker have a large deletion in the middle of their dystrophin gene, but are still able to make a shorter version of the protein, explained Timothy Lotze, a pediatric neurology professor and director of a muscular dystrophy care center at Texas Children's Hospital. Published research has described a Becker patient who, while missing nearly half of that gene, was still walking at 61 years old.

Those insights sparked a rush among rival research groups to develop gene therapies built around micro or mini forms of the dystrophin gene, many of which were later licensed to or acquired by biopharmaceutical companies.

One program from the laboratory of Jude Samulski at the University of North Carolina was picked up by Pfizer. Another, from Jeff Chamberlain at the University of Washington and Dongsheng Duan at the University of Missouri, ended up in the hands of a biotech called Solid Biosciences.

Nationwides work, co-invented by Mendell and one of his recruits, Louise Rodino-Klapac, was acquired by Sarepta. It was a decision made by former CEO Ed Kaye, who had unsuccessfully tried to license Zolgensma years earlier.

I knew Jerry would be first in the clinic and get it done quickly, he said in a 2019 interview with Xconomy. In this business, its who gets there first thats important.

Charlie Handt, now 5, smiles at his home in Darien, Connecticut.

Permission granted by Jennifer Handt

Jennifer Handt wrestled with whether to enroll her son Charlie in one of Sareptas trials.

She met with other parents, visited different trial sites and did her homework, including vetting the doctor who would treat Charlie.

Still, it wasnt a decision she made lightly.

I had that moment of, Is this the right thing to do? Handt said.

Early study results from Sarepta had shown the companys gene therapy could produce levels of miniature dystrophin markedly higher than previously reported in trials of other Duchenne medicines well beyond the amount that researchers think will alter the disease.

Yet data have been more mixed as to whether those protein levels translate to functional benefits. In some cases, boys who would have been expected to decline on tests evaluating their ability to walk, stand and balance, havent yet, which some experts point to as evidence the treatment is working.

What we're seeing is stabilization of the disease that we've never been able to stabilize before, said CHOPs Brandsema, who is an investigator in multiple Duchenne gene therapy trials, including Sareptas. That is a tremendous achievement.

While Handt couldnt be sure Sareptas treatment would help, Charlies disease was certain to progress. What is our alternative? she said. The idea of doing nothing, when we had something, was not an option.

Still, the gene therapies being developed for Duchenne, including Sareptas, use trillions of copies of the viral shells that deliver dystrophin DNA into the body. Though the virus in question has been safely used in scores of gene therapy experiments, prominent researchers have warned higher doses administered intravenously might not be as benign.

Those alarms have some merit. Four children died in a study of a gene therapy for a different type of neuromuscular disease. And while most of the side effects so far associated with Duchenne gene therapy appear manageable with close monitoring and a short course of immune-suppressing drugs, some rare but serious events have caused concern.

One patient died in a trial of Pfizers Duchenne gene therapy in 2021. Solids research was stalled multiple times due to safety worries. Data to date suggests Sareptas gene therapy to be generally safe, but it, too, was linked to one case of serious muscle weakness that researchers believe to be a shared effect among microdystrophin gene therapies. The company has excluded patients with certain mutations from testing in response.

We have to be very aware that this is an irreversible decision when we do this. It's like transplanting an organ or doing surgery, said Brandsema. We cannot take it back once weve given it, and the reaction can be significant.

Susan and Chris Finazzo carry their children around Jungle Island in Miami, Florida, on Oct. 23, 2022.

Permission granted by Susan and Chris Finazzo

The Handt family in Weed Beach in Darien, Connecticut, when Charlie (far right) was 3 years old.

Permission granted by Jennifer Handt

Seven years ago, hundreds of patients and family members traveled to a Washington, D.C., suburb to testify in support of an emerging treatment for Duchenne.

The drug, known then as eteplirsen and also developed by Sarepta, was up for an accelerated approval. The FDA had gathered a panel of experts to review the evidence.

The data primarily came from a clinical trial of just 12 boys with Duchenne, and reviewers were skeptical. The treatment produced a tiny amount of dystrophin less than 1% of normal levels and it was very difficult to discern whether it had an effect.

However, parents, patients and doctors were convinced the drug worked. At the meeting, some chided FDA officials, drawing cheers from the audience. Although the panel ruled narrowly against eteplirsen that day, the agencys top drug evaluator, Janet Woodcock, overruled her own review team, concluding the dystrophin levels were reasonably likely to result in a benefit. Her decision was supported by Robert Califf, the FDA commissioner then and now.

Eteplirsens approval caused a rift within the agency. Multiple reviewers left afterwards. In emails made public following the decision, Woodcock was accused by other staff members of flouting agency norms, keeping unusually close ties with Sarepta and patient advocates, and deciding to approve before reviewers had completed their evaluation.

Outside of the FDA, the approval was viewed as an example of the growing pressure patient groups many of which receive some degree of funding from drug companies were putting on regulators.

That advisory committee meeting harmed relationships within the [Duchenne] community, with the FDA, and the perception of our community outside, PPMDs Furlong said.

Sarepta still hasnt completed a required trial to confirm whether eteplirsen, which is now sold as Exondys 51, actually changes the diseases course.

According to Ingram, the companys CEO, the post-marketing study requested by the FDA wont directly answer that question, only whether higher doses might be more beneficial. He pointed instead to real-world evidence presented at a recent medical meeting, as well as a study showing dystrophin levels as low as 0.5% of normal are associated with milder disease. Neither type of data is as conclusive as results from a placebo-controlled trial.

In the meantime, Sarepta has become one of biotechs most valuable companies, currently worth about $12 billion. It followed a similar blueprint as Exondys 51 to bring two more drugs to market for different subsets of Duchenne patients, and began investing in gene therapy.

In my view, we wouldnt have [the gene therapy] today without Exondys 51s approval, he said. Its clear the FDA did the right thing.

Peter Marks, director of the Food and Drug Administrations Center for Biologics Evaluation and Research, testifies during a hearing in Washington, D.C., on March 18, 2021.

Susan Walsh/Pool/AP

In March, Peter Marks, head of an FDA office that reviews new drugs, stood before more than 1,000 researchers, patient advocates and doctors to give a speech on gene therapy.

Marks spoke of the agencys urgency to speed development of gene therapies for rare and life-threatening diseases, and the tools the FDA had to help. Among those was the accelerated approval pathway, which has been criticized in the years following Exondys clearance and only used once before for a gene therapy.

Marks acknowledged the criticism. Some see speedy approvals as a shortcut, he said. But we cant be so careful about our approvals under accelerated approval that we prevent potentially life-saving therapies from getting to market in a timely manner, he added.

The comments were notable given the venue. Marks was speaking at the yearly meeting of the Muscular Dystrophy Association, a large nonprofit group that supports research into neuromuscular diseases like Duchenne.

The talk was days after the FDA had scheduled an advisory meeting to review Sareptas accelerated approval application a meeting, Stat News subsequently reported, that Marks called after agency staff appeared ready to reject the treatment. Sarepta had previously said a meeting wasnt expected.

The conflicting news jarred the patient community, as advisory committees add an element of risk to the review process.

It made people step back a little bit, said Debra Miller, head of the advocacy group CureDuchenne, adding that theres a real feeling people need to band together to show the FDA we're behind this.

We all feel there's a really good chance that the therapy is going to be approved, Miller said.

Still, the tension highlights the risk Sarepta took by seeking an accelerated approval in the first place.

Sareptas headquarters are seen in Cambridge, Massachusetts in this undated photo.

Permission granted by Sarepta Therapeutics

The decision to file early, rather than wait for the results of the companys ongoing Phase 3 trial, was made after a lot of contemplation within the company, according to Ingram.

One factor was time. If Sarepta waited, at least one more year would pass before the gene therapy could possibly become available. In the life of a Duchenne kid, thats a monumentally long time, he said.

Ingram also pointed to the totality of the evidence Sarepta has already compiled. The treatments design is based on decades of research on the dystrophin genes of Becker patients, he said. It has been tested in about 150 boys and is supported by biological data suggesting its working. Children in the studies are doing better than normally would be expected and the therapys benefit may grow with time, he added.

Other experts interviewed by BioPharma Dive were supportive of the data as well.

This looks like a highly beneficial drug with relative risks that can be managed, said Lotze, of Texas Childrens, who isnt involved in Sarepta's trials. I would hope, and somewhat expect, that a treatment like this might halt further progression of disease or markedly slow it.

Still, Sareptas closest competitor, Pfizer, chose to wait for results from a Phase 3 study before deciding whether to file an application for its gene therapy.

The reason, according to its development head of rare neurologic diseases, Dan Levy, is that its challenging to tease apart a drugs effect by comparing the performance of Duchenne patients to historical data.

The patients who join a clinical trial and the ones involved in the natural history studies that document disease trajectory could be different. There are also biases and confounding factors that can skew results of the primary test used to measure Duchennes progression.

More here:
'The luckiest of the unlucky': A Duchenne gene therapy brings hope ... - BioPharma Dive

Researchers solve mystery of how statins improve blood vessel health – Stanford Medical Center Report

ATAC-seq revealed that simvastatin-treated cells had closed chromatin structures that reduced the expression of genes that cause the endothelial-to-mesenchymal transition. Working backward, the researchers found that simvastatin prevents a protein known as YAP from entering the nucleus and opening chromatin.

The YAP protein is known to play important roles in development, such as regulating the size of our organs, but also has been implicated in the abnormal cell growth seen in cancer.

To see the drug in context, the researchers tested simvastatin on diabetic mice. Diabetes causes subtle changes to blood vessels that mimic the damage commonly seen in people who are prescribed statins older patients who do not have a cardiovascular condition, Liu said.

They found that after eight weeks on simvastatin, the diabetic mice had significantly improved vascular function, with arteries that more easily relaxed and contracted.

If we can understand the mechanism, we can fine-tune this drug to be more specific to rescuing vascular function, Liu said.

The findings also provide a more detailed picture of the vascular disease process, which could help doctors identify and treat early signs of vascular damage.

Ive been taking statins for the past 10 years to keep my cholesterol down. I also knew it has good vascular effects. I just didnt know how it does it, said Wu, the Simon H. Stertzer, MD, Professor who is also the director of the Stanford Cardiovascular Institute. This study explains how.

Researchers from the University of North Texas and the Ohio State University College of Medicine contributed to this study.

The study was supported by funding from the National Institutes of Health (grants R01 HL130020, R01 HL150693, R01 HL163680, R01 HL145676, P01 HL141084, R01 HL141371, R01 HL126527, R01 HL15864, R01 HL161002, R01 HL155282 and 18CDA34110293), an American Heart Association SFRN grant, an AHA Career Development Award and the Tobacco-Related Disease Research Program.

Read the rest here:
Researchers solve mystery of how statins improve blood vessel health - Stanford Medical Center Report

Brain-belly connection: gut health may influence likelihood of … – EurekAlert

image:A UNLV researcher holds a human brain model. view more

Credit: Josh Hawkins/UNLV

Could changing your diet play a role in slowing or even preventing the development of dementia? Were one step closer to finding out, thanks to a new UNLV study that bolsters the long-suspected link between gut health and Alzheimers disease.

The analysis led by a team of researchers with the Nevada Institute of Personalized Medicine (NIPM) at UNLV and published this spring in the Nature journalScientific Reports examined data from dozens of past studies into the belly-brain connection. The results? Theres a strong link between particular kinds of gut bacteria and Alzheimers disease.

Between 500 and 1,000 species of bacteria exist in the human gut at any one time, and the amount and diversity of these microorganisms can be influenced by genetics and diet.

The UNLV teams analysis found a significant correlation between 10 specific types of gut bacteria and the likelihood of developing Alzheimers disease. Six categories of bacteria Adlercreutzia, Eubacterium nodatum group, Eisenbergiella, Eubacterium fissicatena group, Gordonibacter,andPrevotella9 were identified as protective, and four types of bacteria Collinsella, Bacteroides, Lachnospira,andVeillonella were identified as a risk factor for Alzheimers disease.

Certain bacteria in humans guts can secrete acids and toxins that thin and seep through the intestinal lining, interact with theAPOE(a gene identified as a major risk factor for Alzheimers disease), and trigger a neuroinflammatory response affecting brain health and numerous immune functions, and potentially promoting development of the neurodegenerative disorder.

Researchers said their novel discovery of the distinct bacterial groups associated with Alzheimers disease provides new insights into the relationship between gut microbiota and the worlds most common form of dementia. The findings also advance scientists understanding of how an imbalance of that bacteria may play a role in the disorders development.

Most of the microorganisms in our intestines are considered good bacteria that promote health, but an imbalance of those bacteria can be toxic to a persons immune system and linked to various diseases, such as depression, heart disease, cancer, and Alzheimers disease, said UNLV research professorJingchun Chen. The take-home message here is that your genes not only determine whether you have a risk for a disease, but they can also influence the abundance of bacteria in your gut.

While their analysis established overarching categories of bacteria typically associated with Alzheimers disease, the UNLV team said further research is needed to drill down into the specific bacterial species that influence risk or protection.

The hope is to one day develop treatments that are customized for an individual patient and their genetic makeup, such as medications or lifestyle change. Studies have shown that changes in gut microbiome through probiotic use and dietary adjustments can positively impact the immune system, inflammation, and even brain function.

With more research it would be possible to identify a genetic trajectory that could point to a gut microbiome that would be more or less prone to developing diseases such as Alzheimers, said study lead author and UNLV graduate student Davis Cammann, but we also have to remember that the gut biome is influenced by many factors including lifestyle and diet.

Genetic correlations between Alzheimers disease and gut microbiome generawas published this spring inScientific Reports.

In addition to faculty, undergraduate, and graduate student researchers from NIPM, scientists from the UNLV College of Sciences, UNLV School of Dental Medicine, UNLV School of Integrated Health Sciences Department of Brain Health, Kirk Kerkorian School of Medicine at UNLV, Columbia University, and University of Texas Health Science Center at Houston contributed to the study.

Scientific Reports

Meta-analysis

Cells

Genetic correlations between Alzheimers disease and gut microbiome genera

31-Mar-2023

The authors declare no competing interests.

Disclaimer: AAAS and EurekAlert! are not responsible for the accuracy of news releases posted to EurekAlert! by contributing institutions or for the use of any information through the EurekAlert system.

Read the original here:
Brain-belly connection: gut health may influence likelihood of ... - EurekAlert

Endovascular Thrombectomy Shows Better Functional Outcomes … – Neurology Live

Recently published in The New England Journal of Medicine, new findings from the SELECT2 trial (NCT03876457), a phase 3 international, randomized, open-label clinical trial, showed endovascular thrombectomy (EVT) resulted in better functional outcomes than medical care among patients with large ischemic strokes 24 hours after onset.1 These results provide evidence of the efficacy and safety of endovascular thrombectomy in patients with large ischemic strokes, which has been carried out in limited populations to date.

In favor of EVT, the generalized odds ratio for a shift in the distribution of modified Rankin scale (mRS) scores toward better outcomes was 1.51 (95% CI, 1.20-1.89; P <.001). Notably, a total of 20% of the patients in the EVT group and 7% in the medical-care group had functional independence (RR, 2.97; 95% CI, 1.60-5.51).

These findings were also presented as an oral presentation in the clinical trials plenary session at the 2023 American Academy of Neurology (AAN) Annual Meeting, April 22-27, in Boston, Massachusetts, by lead author Amrou Sarraj, MD, director of the Cerebrovascular Center and Comprehensive Stroke Center at University Hospitals Cleveland Medical Center. The session covered the latest clinical trial results that impact the landscape of patient care in all the neurology subspecialities.2

Between September 2019 and September 2022, at the time the trial was stopped, 958 patients had been screened, and among those, 352 were eligible and enrolled. The trial included patients with stroke because of occlusion of the internal carotid artery or the first segment of the middle cerebral artery, and assessed EVT in the time span of 24 hours after onset. The patients enrolled had a large ischemic-core volume, defined by the Alberta Stroke Program Early Computed Tomography Score of 3 to 5 (range, 0- 10, with lower scores indicating larger infarction) or at least 50 mL of a core volume on computed tomography perfusion or diffusion-weighted MRI.

Patients were assigned on a 1:1 ratio to either the EVT plus medical care group (n = 178) or the medical care alone group (n = 174). The mRS score at 90 days (range, 0-6, with higher scores indicating greater disability) was the primary outcome, and functional independence was the secondary outcome.

Among both groups, mortality was similar. Arterial access-site complications occurred in 5 patients, dissection in 10 patients, cerebral-vessel perforation in 7 patients, and transient vasospasm in 11 patients, all of whom were in the EVT group. One patient in the EVT group and 2 patients in the medical-care group experienced symptomatic intracranial hemorrhage.

Limitations of the trial include its early termination and the smaller than anticipated sample size. Although treatment was open label, the outcome assessment was conducted by assessors who were unaware of trial-group assignments. In addition, some patients enrolled had lower ischemic-core volumes than intended for enrollment and only approximately 20% of patients received intravenous thrombolytic agents before randomization.

Previous coverage of SELECT2 identified no safety concerns during a review of 90-day outcomes for the first 200 included participants.3 Findings from that analysis were presented at the 2022 International Stroke Conference (ISC) February 9-11, in New Orleans, Louisiana, by Sarraj. In the analysis, 200 patients were enrolled in the phase 3 study, 100 patients randomized received EVT and 100 were randomized to medical management (MM).

In the data presented at ISC 2023, participants in the EVT group had a median age of 66.5 years (IQR, 58.5-75) and those in the MM group had a median age of 67.5 (IQR, 59-76). Presentation National Institutes of Health Stroke Scale (NIHSS) scores were similar, averaging 19 (IQR, 15-23) for the EVT group and 19 (IQR, 15-23) in the MM group, as was time from stroke onset to randomization, averaging 9.3 hours (IQR, 5.0-15.25) for the EVT group and 9.9 hours (IQR, 5.95-15.9) for the MM group, and physician read CV Alberta Stroke Program Early CT Scores (ASPECTS), at 4 (IQR, 4-5) for the EVT group and 4 (IQR, 4-5) for the MM group. Overall, 176 patients had NCCT ASPECTS between 3-5, with a median ischemic core of 72 mL (IQR, 38.5-108.5). A total of 145 patients had an ischemic core of 50 mL, with a median ischemic core of 94 (IQR, 72-138).

Click here for more coverage of AAN 2023.

See original here:
Endovascular Thrombectomy Shows Better Functional Outcomes ... - Neurology Live

Autism-related genes converge on microglia and dopamine in … – Spectrum – Autism Research News

Mutations in any of 10 genes strongly linked to autism have several converging effects on brain size, activity and behavior in zebrafish via pathways that involve the proliferation of dopamine neurons and microglia, according to a new study.

None of the mutations cause identical changes. But their varied effects on brain size occur largely in the forebrain and cerebellum, and their influence on activity comes mainly from the thalamus and dopamine neurons, the study shows. And all 10 genes cluster into three groups based on how they affect the sleep and sensory-processing behaviors of zebrafish.

We were able to identify subgroups of autism genes that share related behavioral features, says lead investigator Ellen Hoffman, associate professor in the Child Study Center at Yale University. Were hoping in future studies to leverage these subgroups to identify potential pharmacological targets using a precision medicine approach.

Hoffman and her colleagues used gene-editing tools to introduce damaging mutations into autism-linked genes that are known to have diverse functions: CHD8, CUL3, KDM5B, POGZ and TBR1 regulate gene expression; CNTNAP2, SCN1LAB (the zebrafish version of SCN1A and SCN2A) and GRIN2B facilitate communication among neurons; and DYRK1A and KATNAL2 are involved with cell-structure components, including microtubules and cytoskeletons.

The team uncovered the points of convergence by analyzing more than 7,500 larval zebrafish in a battery of tests: They imaged each animals brain structure and volume, stained a protein called phosphorylated ERK as a proxy of brain activity, and used automated motion-tracking cameras to monitor the larvas sleep-wake cycles and observe its responses to sudden light or dark exposures. The findings were published in Cell Reports in March.

The approach of this work is exactly where we need to be these days, says Helen Willsey, assistant professor of psychiatry and behavioral sciences at the University of California, San Francisco, who was not involved in the new study. Were starting to understand how these seemingly disparate genes actually do similar things during brain development, and then being able to leverage that to figure out how to reverse it.

Hoffmans team studied the DYRK1A and SCN1LAB models in further depth, because those two showed the greatest changes across all the assays.

Both models significantly downregulated genes associated with neurogenesis and dopamine signaling, RNA sequencing showed. They also have a decrease in the number of dopamine neurons in the forebrain.

High throughput: Hoffman and her team examined thousands of zebrafish larvae in 96-well plates, including this one.

The fish also showed upregulation of microglia-associated genes and greater numbers of microglia throughout the entire brain. Increased microglia expression can boost synaptic pruning and may explain the reduced brain volume seen in the two models.

This convergence suggests that there could be common druggable targets for autism subtypes that were not previously known to be related, says Holly Stessman, assistant professor of pharmacology and neuroscience at Creighton University in Omaha, Nebraska, who was not involved in the study. Taking individual genes and going down the rabbit hole is going to be the way that we are now going to move the field a big pace forward, Stessman says.

The study is thorough and high quality, but it also reflects some limitations of the methods used in the autism genetics field, says Hazel Sive, dean of the College of Science at Northeastern University in Boston, Massachusetts, who was not involved in the work. For instance, all the animals in this study had two nonfunctional copies of the gene in question, but people with the same variant tend to only have one, with the other remaining intact.

Hoffman and her colleagues are now screening drugs on the subgroups with similar behavioral profiles to see if any molecules can address multiple genetic models. They are also imaging the brains of live fish to observe dopamine circuit development and investigating the mechanisms that contribute to the global increases in microglia.

Cite this article: https://doi.org/10.53053/THAB6590

See the original post here:
Autism-related genes converge on microglia and dopamine in ... - Spectrum - Autism Research News

Editas Medicine Presents Preclinical Data on EDIT-103 for Rhodopsin-associated Autosomal Dominant Retinitis Pigmentosa at the European Society of Gene…

Studies in non-human primates demonstrated nearly 100% gene editing and knockout of endogenous RHO gene and more than 30% replacement protein levels using a dual vector AAV approach

Treated eyes showed morphological and functional photoreceptor preservation

EDIT-103 advancing towards IND-enabling studies

CAMBRIDGE, Mass., Oct. 13, 2022 (GLOBE NEWSWIRE) -- Editas Medicine, Inc. (Nasdaq: EDIT), a leading genome editing company, today announced ex vivo and in vivo preclinical data supporting its experimental medicine EDIT-103 for the treatment of rhodopsin-associated autosomal dominant retinitis pigmentosa (RHO-adRP). The Company reported these data in an oral presentation today at the European Society of Gene and Cell Therapy 29th Annual Meeting in Edinburgh, Scotland, UK.

EDIT-103 is a mutation-independent CRISPR/Cas9-based, dual AAV5 vectors knockout and replace (KO&R) therapy to treat RHO-adRP. This approach has the potential to treat any of over 150 dominant gain-of-function rhodopsin mutations that cause RHO-adRP with a one-time subretinal administration.

These promising preclinical data demonstrate the potential of EDIT-103 to efficiently remove the defective RHO gene responsible for RHO-adRP while replacing it with an RHO gene capable of producing sufficient levels of RHO to preserve photoreceptor structure and functions. The program is progressing towards the clinic, said Mark S. Shearman, Ph.D., Executive Vice President and Chief Scientific Officer, Editas Medicine. EDIT-103 uses a dual AAV gene editing approach, and also provides initial proof of concept for the treatment of other autosomal dominant disease indications where a gain of negative function needs to be corrected.

Key findings include:

Full details of the Editas Medicine presentations can be accessed in the Posters & Presentations section on the Companys website.

About EDIT-103EDIT-103 is a CRISPR/Cas9-based experimental medicine in preclinical development for the treatment of rhodopsin-associated autosomal dominant retinitis pigmentosa (RHO-adRP), a progressive form of retinal degeneration. EDIT-103 is administered via subretinal injection and uses two adeno-associated virus (AAV) vectors to knockout and replace mutations in the rhodopsin gene to preserve photoreceptor function. This approach can potentially address more than 150 gene mutations that cause RHO-adRP.

AboutEditas MedicineAs a leading genome editing company, Editas Medicine is focused on translating the power and potential of the CRISPR/Cas9 and CRISPR/Cas12a genome editing systems into a robust pipeline of treatments for people living with serious diseases around the world. Editas Medicine aims to discover, develop, manufacture, and commercialize transformative, durable, precision genomic medicines for a broad class of diseases. Editas Medicine is the exclusive licensee of Harvard and Broad Institutes Cas9 patent estates and Broad Institutes Cas12a patent estate for human medicines. For the latest information and scientific presentations, please visit http://www.editasmedicine.com.

Forward-Looking StatementsThis press release contains forward-looking statements and information within the meaning of The Private Securities Litigation Reform Act of 1995. The words "anticipate," "believe," "continue," "could," "estimate," "expect," "intend," "may," "plan," "potential," "predict," "project," "target," "should," "would," and similar expressions are intended to identify forward-looking statements, although not all forward-looking statements contain these identifying words. The Company may not actually achieve the plans, intentions, or expectations disclosed in these forward-looking statements, and you should not place undue reliance on these forward-looking statements. Actual results or events could differ materially from the plans, intentions and expectations disclosed in these forward-looking statements as a result of various factors, including: uncertainties inherent in the initiation and completion of preclinical studies and clinical trials and clinical development of the Companys product candidates; availability and timing of results from preclinical studies and clinical trials; whether interim results from a clinical trial will be predictive of the final results of the trial or the results of future trials; expectations for regulatory approvals to conduct trials or to market products and availability of funding sufficient for the Companys foreseeable and unforeseeable operating expenses and capital expenditure requirements. These and other risks are described in greater detail under the caption Risk Factors included in the Companys most recent Annual Report on Form 10-K, which is on file with theSecurities and Exchange Commission, as updated by the Companys subsequent filings with theSecurities and Exchange Commission, and in other filings that the Company may make with theSecurities and Exchange Commissionin the future. Any forward-looking statements contained in this press release speak only as of the date hereof, and the Company expressly disclaims any obligation to update any forward-looking statements, whether because of new information, future events or otherwise.

Read the original here:
Editas Medicine Presents Preclinical Data on EDIT-103 for Rhodopsin-associated Autosomal Dominant Retinitis Pigmentosa at the European Society of Gene...

USC researcher uses mammal DNA to zoom into the human … – EurekAlert

Why do humans have disease if they went through millions of years of evolution? Its a question Steven Gazal, PhD, assistant professor of population and public health sciences at the Keck School of Medicine of USC, hopes to answer.

Gazal is part of an international team of researchers who have become the first to precisely identify base pairs of the human genome that remained consistent over millions of years of mammalian evolution, and which play a crucial role in human disease. Thefindings werepublishedin a specialZoonomiaedition ofScience.

Gazal and his team analyzed the genomes of 240 mammals, including humans, zooming in with unprecedented resolution to compare DNA. They were able to identify base pairs that were constrained meaning they remained generally consistent across mammal species over the course of evolution. Individuals born with mutations on these genes may not have been as successful within their species or were otherwise not likely to pass down the genetic variation. We were able to identify where gene mutations are not tolerated in evolution, and we demonstrated that these mutations are significant when it comes to disease, explains Gazal.

The team found that3.3% of bases in the human genome are"significantly constrained,"including 57.6% of the coding bases that determine amino acid position, meaningthese bases had unusually few variants across species in the dataset. The most constrained base pairs in mammals were over seven times more likely to be causal forhuman disease and complex trait, and over 11 times more likely when researchers looked at the most constrained base pairs in primates alone.

Thedataset wasprovided bytheZoonomiaconsortium, whichaccording to the project website, "is applying advances inDNA sequencingtechnologies to understand how genomes generate the tremendous wealth of animaldiversity.Gazalgives credit to Zoonomia for making this type of data available to researchers and anticipates it will be widely used by human geneticists. Its a cheap resource togenerate, as opposed to datasets generated in human genetic studies, says Gazal.

His teams findings are a significant step forward, as Gazal notes, "we do not understand 99% of the human genome, so it is fundamental to understand which part has been constrained by evolution and is likely to have an impact on human phenotypes. Their discoveries and methods could become crucial tools for further research.

The next step for Gazal and his team is to repeat the process with a primate-only dataset. By restricting the subjects, they hope to focus on functions of DNA that appeared more recently in human evolution. We expect this to be even more useful in determining information on human disease, says Gazal.

Meta-analysis

People

Leveraging base-pair mammalian constraint to understand genetic variation and human disease

28-Apr-2023

Disclaimer: AAAS and EurekAlert! are not responsible for the accuracy of news releases posted to EurekAlert! by contributing institutions or for the use of any information through the EurekAlert system.

Read more here:
USC researcher uses mammal DNA to zoom into the human ... - EurekAlert