Novartis hunting for sickle cell cure with Precision deal – BioPharma Dive

Dive Brief:

Sickle cell and beta thalassemia, both rare, life-threatening diseases caused by mistakes in DNA, are top targets for biotech and pharma companies working in the genetic medicine field. Several of those efforts have advanced to, or will soon reach, the Food and Drug Administration.

Earlier this month, advisers to the agency recommended approval for a cell-based gene therapy designed by Bluebird bio to treat beta thalassemia. Partners Vertex and CRISPR Therapeutics, meanwhile, plan to later this year submit for approval a CRISPR gene editing treatment that can treat both conditions.

Novartis is already working on a similar treatment through a partnership with the CRISPR biotech Intellia Therapeutics. Early clinical testing of their therapy began in 2020. The Swiss drugmaker also won approval in 2019 for a drug designed to limit the blood vessel blockages that cause severe pain crises in sickle cell patients.

Still, Novartis is exploring other approaches, recently joining forces with the Bill and Melinda Gates Foundation and, on Tuesday, partnering with Precision.

Precision specializes in a type of gene editing technology it calls ARCUS. While the technology shares a similar concept to the better-known CRISPR, it uses a different type of nuclease, or DNA-cutting enzyme. Under the deal with Novartis, Precision will build a custom ARCUS nuclease for use in sickle cell and beta thalassemia. Once thats developed, Novartis will handle R&D, manufacturing and, if research succeeds, commercialization.

In announcing the deal, the companies acknowledged the competition, but noted how their work will focus on a treatment that can be used inside the body, or in vivo.

The in vivo gene editing approach that we are pursuing for sickle cell disease could have a number of significant advantages over other ex vivo gene therapies currently in development, said Derek Jantz, Precisions chief scientific officer, in a statement. Perhaps most importantly, it could open the door to treating patients in geographies where stem cell transplant is not a realistic option.

For Precision, the cash infusion from Novartis will also help extend its operating runway into the second quarter of 2024. Also on Tuesday, the biotech announced it would raise a further $50 million through the sale of nearly 36 million shares.

Go here to read the rest:

Novartis hunting for sickle cell cure with Precision deal - BioPharma Dive

Double Duty: Early Research Reveals how a Single Drug Delivers Twice the Impact in Fragile X – URMC

Like many neurological diseases, theres a lot we dont understand about fragile X syndrome. But, after studying the disorder for several years, Lynne Maquats lab knew two important things: the enzyme AKT, which plays a key role in cell growth and survival, and the quality control pathway known as NMD (nonsense-mediated mRNA decay), are both in overdrive in fragile X.

In a new study in the journal Molecular Cell, the team reveals how these two major players interact, highlighting a complex molecular dance that could inform the development of future treatments for fragile X syndrome.

Two paths to pursue

AKT is a hub for cell signaling, helping cells communicate about important processes like cell growth, proliferation and protein production. When cells are stressed for example, in cancer, diabetes, heart disease and neurological disorders, including fragile X AKT can send too many (or too few) signals or messages as part of a cell survival mechanism.

NMD is like a molecular guide that helps our cells make smart decisions that (in most cases) improve cellular function and contribute to good health. For example, NMD supports gene expression by flagging and destroying mRNAs (messenger RNAs) that are carrying faulty genetic instructions that could lead to disease. It also helps our cells adjust to changes in development and in their environment, andmore rapidly respond to certain stimuli.

Co-lead study authors Hana Cho, Ph.D., and Elizabeth Abshire, Ph.D., discovered how AKT and NMD interact in the context of fragile X:

Drug double whammy

Taking these findings a step further, the team treated the neural stem cells that mimic fragile X syndrome with a drug called Afuresertib, which inhibits AKT and is currently being tested in phase 1 and 2 clinical trials for several types of cancer. They found that blocking AKT in the fragile X cells not only decreased its activity, but decreased NMD, as well. The cells acted more like typical, non-disease cells when AKT was inhibited.

Normalizing two major pathways that contribute to fragile X syndrome is an exciting development, and using a drug that has already been through early clinical trials and that has been shown to be safe in patients puts us a step ahead, as opposed to starting from scratch with a brand new molecule, says Abshire, a postdoctoral fellow in the Maquat lab. There is still a lot we dont know about how AKT and NMD interact, because they are both massive pathways that influence and regulate multiple activities in cells, but this work provides good direction.

Next steps in the research include taking drugs like Afuresertib and testing them in a mouse model of fragile X to determine if what the team found in cells (AKT goes down and NMD goes down) also occurs in a living organism.

Drilling down on disease mechanism

AKT is stimulated or spurred into action by insulin. This study is the first to show that extracellular signaling (something that happens outside the cell, like an increase in insulin) changes the identity of a mark called the exon junction complex or EJC. Discovered by Lynne E. Maquat, Ph.D., founding director of the Center for RNA Biology at the University of Rochester, the EJC promotes NMD when certain conditions are met. Cho and Abshire showed that AKT is unexpectedly a member of the complex of proteins that constitute the EJC, which is important for normal gene expression.

By revealing a new mechanism by which AKT-signaling alters NMD and gene expression, we have a more complete understanding of disease mechanism. The more we know about this important signaling pathway, the more we can think about targets to suppress its hyperactivity, said Maquat, corresponding study author and the J. Lowell Orbison Endowed Chair and Professor ofBiochemistry and Biophysicsat the University of Rochester School of Medicine and Dentistry.This adds another aspect to how we can understand dysregulated pathways in diseases like fragile X and cancer when we are thinking about drugs.

In the study, the team also details a new tool that they developed for screening potential drugs that inhibit NMD, which is hyperactivated in fragile X and a number of cancers.

In addition to Maquat, Cho and Abshire, Maximilian W. Popp and Christoph Prschel, also of the University of Rochester School of Medicine and Dentistry, and Joshua L. Schwartz and Gene W. Yeo, of the University of California-San Diego, contributed to the research. The research was funded by an R01 to Maquat and an R21 to Prschel, both from the National Institutes of Health, and by a University of Rochester Provosts award to Maquat and Prschel.

# # #

Follow this link:

Double Duty: Early Research Reveals how a Single Drug Delivers Twice the Impact in Fragile X - URMC

Ambys Medicines Announces Formation of Clinical and Scientific Advisory Boards with Leading Liver Disease and Cell and Gene Therapy Experts – Business…

SOUTH SAN FRANCISCO, Calif.--(BUSINESS WIRE)--Ambys Medicines, a company pioneering cell-replacement therapies for patients with liver disease, today announced the formation of its clinical and scientific advisory boards comprising leading clinical experts in liver disease and hepatocyte transplantation, and world-class scientists pioneering cell and gene technologies.

The clinical advisory board provides guidance on advancing Ambyss lead program, AMI-918, through the clinic and provides critical input on clinical trial design and patient selection. The scientific advisory board will provide a cross-discipline perspective on applying cutting-edge technology to Ambyss cell therapy platform to progress its discovery pipeline of next generation cell therapies.

Were honored to work with such an esteemed and diverse group of experts in liver disease, hepatocyte transplantation, and cell and gene therapies whose collective experience will be highly valuable as we finalize our clinical development strategy for AMI-918 and progress our genetically engineered hepatocyte follow-on programs, said Ronald Park, M.D., Chief Executive Officer of Ambys Medicines. Were grateful for the engagement and support from our advisors as we work to bring first-in-class hepatocyte replacement therapies to liver failure patients who currently lack treatment options.

Each of our advisors brings incredible knowledge and expertise in their respective fields that will be instrumental to Ambys as we continue to broaden our pipeline and move closer to becoming a clinical-stage company, said Markus Grompe, M.D., Founder and Chief Scientific Officer of Ambys Medicines. Were excited to partner together to realize the potential of our novel replacement cell therapy platform in restoring lost hepatic function to patients with acute or chronic liver failure and genetic liver diseases.

Clinical Advisory Board

Scientific Advisory Board

About Ambys MedicinesAmbys Medicines is focused on pioneering cell replacement therapies for patients with liver failure. Ambyss proprietary platform enables the company to be the first and only company able to develop and manufacture functional human hepatocytes at scale. Our scientific approach has the potential to fundamentally transform the treatment paradigm for patients with acute and chronic liver failure and genetic diseases of the liver. Our lead program, AMI-918, is a hepatocyte replacement cell therapy in development to restore lost hepatic function. Beyond AMI-918, we are building a pipeline of next-generation modified hepatocytes that will rapidly expand the range of treatable patient populations. Learn more at ambys.com and follow us on Twitter, LinkedIn and Instagram.

Read more:

Ambys Medicines Announces Formation of Clinical and Scientific Advisory Boards with Leading Liver Disease and Cell and Gene Therapy Experts - Business...

A Functional Medicine Approach to Detoxing the Body – Magazine of Santa Clarita

Daily our bodies are exposed to toxic chemicals from the environment that can be harmful to our health. Exposure to these toxins comes in many forms, including the air we breathe, the stress we feel, and the food we eat. Most of the harmful toxins are man-made artificial products introduced into the environment due to human activity. Examples of toxins include industrial waste products, pesticides, herbicides, food additives, and preservatives. Several metals (arsenic, lead, mercury, cadmium, and PCBs) are also believed to cause disease.Once those harmful chemicals enter the body they are referred to as xenobiotics. Xenobiotics is a chemical compound (drug, pesticide, or carcinogen) that is foreign to the human body. Several of these xenobiotics are endocrine disruptors, meaning that they mimic or interfere with your bodys hormones. For example, arsenic can poison the beta-cell responsible for producing insulin in the pancreas and damage our DNA. It is suggested that arsenic may cause 15%-20% of diabetes.The body has a natural detoxification system through the work of the liver, kidneys, large intestines, lymphatic system, and sweat glands. The liver is the major player in the bodys detox system, but they all work together to reduce the buildup of toxins. We all have different toxin exposure, and we all have genetic differences in how a persons body is able to produce the enzymes that regulate detoxification. When a persons body burden of toxins has exceeded the bodys ability to eliminate them, symptoms can occur. A few of the most common symptoms include fatigue, headaches, anxiety, depression, brain fog, difficulty losing weight, skin breakouts, and digestive issues.Lowering your toxic load is vital to your health. However, what we eat can either support or hinder the bodys ability to process and eliminate harmful toxins. Several nutrients are needed to push detox pathways in the body and support and regulate detoxification. A deficiency in any of them could cause an increased body burden or buildup of chemicals in the body.In functional medicine, we take a whole-body approach to toxin load by checking for nutritional status, genetic susceptibility to nutritional deficiencies, and environmental exposures. It is important to investigate exposure pathways to identify a source of toxicity elevation.One of the best ways to protect yourself is to identify the toxic exposure and stop it! It is important to support the bodys natural detoxification system by eating organic colorful, plant-based diet rich in antioxidants. Finally, promote glutathione (powerful antioxidant) production in the body by taking vitamin C, eating sulfur-rich foods, and getting restorative sleep as it may increase the excretion of toxins. For more information, call Compassionate Healthcare Associates, 661 295-7777.

Continued here:

A Functional Medicine Approach to Detoxing the Body - Magazine of Santa Clarita

Meet the company trying precision medicine for autism | Spectrum – Spectrum

Ibudilast improved cognition and behavior in men with fragile X syndrome in a small clinical trial. The use of the other drug in STP1, bumetanide, in autistic people is cloudier. A 2020 clinical trial found that only some of the children who received bumetanide for three months showed reduced repetitive behaviors, though studies over the past two years have suggested that patterns of electrical activity in the brain or levels of specific immune molecules in the blood can predict a persons response to the drug. Last year, however, phase 3 studies of bumetanide were ended after researchers found no benefit to treatment.

Bumetanide is an excellent drug for autism, provided you select specific subpopulations of young people, says Yehezkel Ben-Ari, president and co-founder of French biotech company Neurochlore, which owns the patent for bumetanide as an autism treatment. Adults, he adds, may not respond as well. The 2020 trial found that younger children showed more improvement in social communication and responsiveness than did older ones.

Many people have hoped for precision medicine, but generally, the field of autism has moved away to some degree. Catherine Lord

Beyond the questions around bumetanide, others doubt that personalized therapy can be applied to autism at all. Many people have hoped for precision medicine, but generally, the field of autism has moved away to some degree, says Catherine Lord, distinguished professor of psychiatry at the University of California, Los Angeles. Thats because, she says, for something as complex and heterogeneous as autism, theres no clear link between known genetic factors and autism traits. Researchers have not yet had success in finding biomarkers for diagnosis of the condition, let alone predicting who is most likely to respond to treatment, she says.

Understanding the more convergent mechanisms of autism, and what is common about people with autism rather than whats different about them, is a more important approach to understanding whats treatable or preventable in the condition, says John Constantino, professor of psychiatry and pediatrics at Washington University in St. Louis, Missouri.

A molecular footprint found only in a subgroup of people with autism may have nothing to do with the condition and instead be related to other factors, such as depression or anxiety, Constantino says. So far, he adds, researchers havent even been able to reliably differentiate people with autism from those without the condition by looking at biological traits. Identifying a biological profile for different autism subtypes would be a significant advance in the field, he says, if Stalicla has actually done it.

Continue reading here:

Meet the company trying precision medicine for autism | Spectrum - Spectrum

CRISPR, 10 Years On: Learning to Rewrite the Code of Life – The New York Times

Ten years ago this week, Jennifer Doudna and her colleagues published the results of a test-tube experiment on bacterial genes. When the study came out in the journal Science on June 28, 2012, it did not make headline news. In fact, over the next few weeks, it did not make any news at all.

Looking back, Dr. Doudna wondered if the oversight had something to do with the wonky title she and her colleagues had chosen for the study: A Programmable Dual RNA-Guided DNA Endonuclease in Adaptive Bacterial Immunity.

I suppose if I were writing the paper today, I would have chosen a different title, Dr. Doudna, a biochemist at the University of California, Berkeley, said in an interview.

Far from an esoteric finding, the discovery pointed to a new method for editing DNA, one that might even make it possible to change human genes.

I remember thinking very clearly, when we publish this paper, its like firing the starting gun at a race, she said.

In just a decade, CRISPR has become one of the most celebrated inventions in modern biology. It is swiftly changing how medical researchers study diseases: Cancer biologists are using the method to discover hidden vulnerabilities of tumor cells. Doctors are using CRISPR to edit genes that cause hereditary diseases.

The era of human gene editing isnt coming, said David Liu, a biologist at Harvard University. Its here.

But CRISPRs influence extends far beyond medicine. Evolutionary biologists are using the technology to study Neanderthal brains and to investigate how our ape ancestors lost their tails. Plant biologists have edited seeds to produce crops with new vitamins or with the ability to withstand diseases. Some of them may reach supermarket shelves in the next few years.

CRISPR has had such a quick impact that Dr. Doudna and her collaborator, Emmanuelle Charpentier of the Max Planck Unit for the Science of Pathogens in Berlin, won the 2020 Nobel Prize for chemistry. The award committee hailed their 2012 study as an epoch-making experiment.

Dr. Doudna recognized early on that CRISPR would pose a number of thorny ethical questions, and after a decade of its development, those questions are more urgent than ever.

Will the coming wave of CRISPR-altered crops feed the world and help poor farmers or only enrich agribusiness giants that invest in the technology? Will CRISPR-based medicine improve health for vulnerable people across the world, or come with a million-dollar price tag?

The most profound ethical question about CRISPR is how future generations might use the technology to alter human embryos. This notion was simply a thought experiment until 2018, when He Jiankui, a biophysicist in China, edited a gene in human embryos to confer resistance to H.I.V. Three of the modified embryos were implanted in women in the Chinese city of Shenzen.

In 2019, a court sentenced Dr. He to prison for illegal medical practices. MIT Technology Review reported in April that he had recently been released. Little is known about the health of the three children, who are now toddlers.

Scientists dont know of anyone else who has followed Dr. Hes example yet. But as CRISPR continues to improve, editing human embryos may eventually become a safe and effective treatment for a variety of diseases.

Will it then become acceptable, or even routine, to repair disease-causing genes in an embryo in the lab? What if parents wanted to insert traits that they found more desirable like those related to height, eye color or intelligence?

Franoise Baylis, a bioethicist at Dalhousie University in Nova Scotia, worries that the public is still not ready to grapple with such questions.

Im skeptical about the depth of understanding about whats at issue there, she said. Theres a difference between making people better and making better people.

Dr. Doudna and Dr. Charpentier did not invent their gene-editing method from scratch. They borrowed their molecular tools from bacteria.

In the 1980s, microbiologists discovered puzzling stretches of DNA in bacteria, later called Clustered Regularly Interspaced Short Palindromic Repeats. Further research revealed that bacteria used these CRISPR sequences as weapons against invading viruses.

The bacteria turned these sequences into genetic material, called RNA, that could stick precisely to a short stretch of an invading viruss genes. These RNA molecules carry proteins with them that act like molecular scissors, slicing the viral genes and halting the infection.

As Dr. Doudna and Dr. Charpentier investigated CRISPR, they realized that the system might allow them to cut a sequence of DNA of their own choosing. All they needed to do was make a matching piece of RNA.

To test this revolutionary idea, they created a batch of identical pieces of DNA. They then crafted another batch of RNA molecules, programming all of them to home in on the same spot on the DNA. Finally, they mixed the DNA, the RNA and molecular scissors together in test tubes. They discovered that many of the DNA molecules had been cut at precisely the right spot.

For months Dr. Doudna oversaw a series of round-the-clock experiments to see if CRISPR might work not only in a test tube, but also in living cells. She pushed her team hard, suspecting that many other scientists were also on the chase. That hunch soon proved correct.

In January 2013, five teams of scientists published studies in which they successfully used CRISPR in living animal or human cells. Dr. Doudna did not win that race; the first two published papers came from two labs in Cambridge, Mass. one at the Broad Institute of M.I.T. and Harvard, and the other at Harvard.

Lukas Dow, a cancer biologist at Weill Cornell Medicine, vividly remembers learning about CRISPRs potential. Reading the papers, it looked amazing, he recalled.

Dr. Dow and his colleagues soon found that the method reliably snipped out pieces of DNA in human cancer cells.

It became a verb to drop, Dr. Dow said. A lot of people would say, Did you CRISPR that?

Cancer biologists began systematically altering every gene in cancer cells to see which ones mattered to the disease. Researchers at KSQ Therapeutics, also in Cambridge, used CRISPR to discover a gene that is essential for the growth of certain tumors, for example, and last year, they began a clinical trial of a drug that blocks the gene.

Caribou Biosciences, co-founded by Dr. Doudna, and CRISPR Therapeutics, co-founded by Dr. Charpentier, are both running clinical trials for CRISPR treatments that fight cancer in another way: by editing immune cells to more aggressively attack tumors.

Those companies and several others are also using CRISPR to try to reverse hereditary diseases. On June 12, researchers from CRISPR Therapeutics and Vertex, a Boston-based biotech firm, presented at a scientific meeting new results from their clinical trial involving 75 volunteers who had sickle-cell anemia or beta thalassemia. These diseases impair hemoglobin, a protein in red blood cells that carries oxygen.

The researchers took advantage of the fact that humans have more than one hemoglobin gene. One copy, called fetal hemoglobin, is typically active only in fetuses, shutting down within a few months after birth.

The researchers extracted immature blood cells from the bone marrow of the volunteers. They then used CRISPR to snip out the switch that would typically turn off the fetal hemoglobin gene. When the edited cells were returned to patients, they could develop into red blood cells rife with hemoglobin.

Speaking at a hematology conference, the researchers reported that out of 44 treated patients with beta thalassemia, 42 no longer needed regular blood transfusions. None of the 31 sickle cell patients experienced painful drops in oxygen that would have normally sent them to the hospital.

CRISPR Therapeutics and Vertex expect to ask government regulators by the end of year to approve the treatment.

Other companies are injecting CRISPR molecules directly into the body. Intellia Therapeutics, based in Cambridge and also co-founded by Dr. Doudna, has teamed up with Regeneron, based in Westchester County, N.Y., to begin a clinical trial to treat transthyretin amyloidosis, a rare disease in which a damaged liver protein becomes lethal as it builds up in the blood.

Doctors injected CRISPR molecules into the volunteers livers to shut down the defective gene. Speaking at a scientific conference last Friday, Intellia researchers reported that a single dose of the treatment produced a significant drop in the protein level in volunteers blood for as long as a year thus far.

The same technology that allows medical researchers to tinker with human cells is letting agricultural scientists alter crop genes. When the first wave of CRISPR studies came out, Catherine Feuillet, an expert on wheat, who was then at the French National Institute for Agricultural Research, immediately saw its potential for her own work.

I said, Oh my God, we have a tool, she said. We can put breeding on steroids.

At Inari Agriculture, a company in Cambridge, Dr. Feuillet is overseeing efforts to use CRISPR to make breeds of soybeans and other crops that use less water and fertilizer. Outside of the United States, British researchers have used CRISPR to breed a tomato that can produce vitamin D.

Kevin Pixley, a plant scientist at the International Maize and Wheat Improvement Center in Mexico City, said that CRISPR is important to plant breeding not only because its powerful, but because its relatively cheap. Even small labs can create disease-resistant cassavas or drought-resistant bananas, which could benefit poor nations but would not interest companies looking for hefty financial returns.

Because of CRISPRs use for so many different industries, its patent has been the subject of a long-running dispute. Groups led by the Broad Institute and the University of California both filed patents for the original version of gene editing based on CRISPR-Cas9 in living cells. The Broad Institute won a patent in 2014, and the University of California responded with a court challenge.

In February of this year, the U.S. Patent Trial and Appeal Board issued what is most likely the final word on this dispute. They ruled in favor of the Broad Institute.

Jacob Sherkow, an expert on biotech patents at the University of Illinois College of Law, predicted that companies that have licensed the CRISPR technology from the University of California will need to honor the Broad Institute patent.

The big-ticket CRISPR companies, the ones that are farthest along in clinical trials, are almost certainly going to need to write the Broad Institute a really big check, he said.

The original CRISPR system, known as CRISPR-Cas9, leaves plenty of room for improvement. The molecules are good at snipping out DNA, but theyre not as good at inserting new pieces in their place. Sometimes CRISPR-Cas9 misses its target, cutting DNA in the wrong place. And even when the molecules do their jobs correctly, cells can make mistakes as they repair the loose ends of DNA left behind.

A number of scientists have invented new versions of CRISPR that overcome some of these shortcomings. At Harvard, for example, Dr. Liu and his colleagues have used CRISPR to make a nick in one of DNAs two strands, rather than breaking them entirely. This process, known as base editing, lets them precisely change a single genetic letter of DNA with much less risk of genetic damage.

Dr. Liu has co-founded a company called Beam Therapeutics to create base-editing drugs. Later this year, the company will test its first drug on people with sickle cell anemia.

Dr. Liu and his colleagues have also attached CRISPR molecules to a protein that viruses use to insert their genes into their hosts DNA. This new method, called prime editing, could enable CRISPR to alter longer stretches of genetic material.

Prime editors are kind of like DNA word processors, Dr. Liu said. They actually perform a search and replace function on DNA.

Rodolphe Barrangou, a CRISPR expert at North Carolina State University and a founder of Intellia Therapeutics, predicted that prime editing would eventually become a part of the standard CRISPR toolbox. But for now, he said, the technique was still too complex to become widely used. Its not quite ready for prime time, pun intended, he said.

Advances like prime editing didnt yet exist in 2018, when Dr. He set out to edit human embryos in Shenzen. He used the standard CRISPR-Cas9 system that Dr. Doudna and others had developed years before.

Dr. He hoped to endow babies with resistance to H.I.V. by snipping a piece of a gene called CCR5 from the DNA of embryos. People who naturally carry the same mutation rarely get infected by H.I.V.

In November 2018, Dr. He announced that a pair of twin girlshad been born with his gene edits. The announcement took many scientists like Dr. Doudna by surprise, and they roundly condemned him for putting the health of the babies in jeopardy with untested procedures.

Dr. Baylis of Dalhousie University criticized Dr. He for the way he reportedly presented the procedure to the parents, downplaying the radical experiment they were about to undertake. You could not get an informed consent, unless you were saying, This is pie in the sky. Nobodys ever done it, she said.

In the nearly four years since Dr. Hes announcement, scientists have continued to use CRISPR on human embryos. But they have studied embryos only when theyre tiny clumps of cells to find clues about the earliest stages of development. These studies could potentially lead to new treatments for infertility.

Bieke Bekaert, a graduate student in reproductive biology at Ghent University in Belgium, said that CRISPR remains challenging to use in human embryos. Breaking DNA in these cells can lead to drastic rearrangements in the chromosomes. Its more difficult than we thought, said Ms. Bekaert, the lead author of a recent review of the subject. We dont really know what is happening.

Still, Ms. Bekaert held out hope that prime editing and other improvements on CRISPR could allow scientists to make reliably precise changes to human embryos. Five years is way too early, but I think in my lifetime it may happen, she said.

Link:

CRISPR, 10 Years On: Learning to Rewrite the Code of Life - The New York Times

After 9 Years in Trials, Roche’s Crenezumab Stalls Out – Being Patient

By Simon Spichak, MSc | June 28th, 2022

There were more than 140 experimental Alzheimers drugs in clinical trials as of the start of 2022. On the one hand, this number inspires some optimism that something will break through. But at the same time, each of these drugs faces more than a 99-percent chance of failure. This month, another potential disease-modifying Alzheimers drug, which has been in trials for nearly a decade, is dropping out of the race.

Genentech, a biotech company under Roche, announced that their anti-amyloid drug crenezumab, developed in conjunction with Switzerland-based biotech company AC Immune SA, has ultimately failed to slow or prevent Alzheimers disease in a cohort of cognitively healthy people with early-onset Alzheimers genes.

Were disappointed that the treatment did not demonstrate a statistically significant clinical benefit, Dr. Eric M. Reiman, Banner Alzheimers Institute executive director and one of the study leaders, said in a news release. At the same time, were proud of the impact that this precedent-setting trial has had in shaping a new era in Alzheimers prevention research and were extremely grateful to our research participants and their families.

In the Phase 2 clinical trial, 252 people were randomized to receive crenezumab or placebo over the course of five to eight years. But the drug failed to prevent the decline in cognitive function or episodic memory which occurs as a result of early-onset Alzheimers.

The participants in the clinical trial had autosomal dominant Alzheimers disease which is caused by mutations in the APP, PSEN1 or PSEN2 genes. Even carrying one copy of these genes leads to greater build up of amyloid plaques and symptoms developing in people between their 30s and 60s.

While this attempt at targeting beta-amyloid plaques failed to improve cognitive outcomes, it is unclear whether the drug effectively reduced plaque build-up, highlighting just how little the research community knows for certain about how Alzheimers forms and progresses.

David Knopman, a professor at the Mayo Clinic College of Medicine wrote that crenezumab previously failed Phase 3 clinical trials for non-genetic forms of Alzheimers disease, even though the drug was administered at a higher dose than in this recent study.

Knopman added, The crenezumab trial in the Colombian dominantly inherited [Alzheimers] cohort is disappointing, but not altogether unpredicted based on earlier failures of crenezumab in sporadic Alzheimers, and based on the inability of crenezumab to clear brain amyloid to any extent.

Marc Aurel Busche and Samuel Harris, both physician-scientists at University College London, emphasized that we know very little about the role of amyloid proteins and beta-amyloid plaques in the healthy brain. In one study authored by Busche, anti-amyloid antibodies actually worsened Alzheimers pathology in mice.

We conclude that our understanding of the effect of anti-amyloid antibodies on brain (dys)function in humans with [Alzheimers] is rudimentary, they wrote, adding that therapeutic benefits could be masked by detrimental effects of anti-amyloid antibodies.

The first-ever FDA-approved disease-modifying drug for Alzheimers, Aduhelm (aducanumab) was designed with similar mechanisms. Another monoclonal antibody, Aduhelm targets beta-amyloid, too. But, since its FDA approval, the drug has been mired in controversy over unclear efficacy data. While some researchers and drug developers are exploring other avenues to addressing Alzheimers altogether from the microbiome to tau protein tangles the research community hasnt given up on beta-amyloid as a target, nor on these -mab drugs as an approach.

In the meantime, Roche is focused on advancing its other anti-amyloid drug, gantenerumab. This anti-amyloid received a Breakthrough Therapy designation from the Food and Drug Administration in October 2021.

Gantenerumab failed Phase 3 clinical trial in 2014 but was brought back as an Alzheimers drug candidate in 2017, to test whether it may provide preventative benefits. The ongoing Phase 3 trial is exploring whether the drug can prevent cognitive decline in healthy individuals aged 60 to 80 who have evidence of Alzheimers biomarker beta-amyloid build-up in the brain.

Unlike other anti-amyloid drugs, i.e. Aduhelm, gantenerumab is not administered intravenously. Instead, it would be delivered via subcutaneous injection where the drug is administered under the skin potentially enabling at-home treatment. Results from a Phase 3 clinical trial assessing its efficacy in early Alzheimers is expected by the end of the year.

Go here to read the rest:

After 9 Years in Trials, Roche's Crenezumab Stalls Out - Being Patient

Dr. Stephen Kingsmore to receive Luminary Award at 2022 Precision Medicine World Conference – EurekAlert

image:Stephen Kingsmore, MD, DSc, President and CEO or Rady Children's Institute for Genomic Medicine view more

Credit: Rady Children's Institute for Genomic Medicine

SAN DIEGO, Calif. June 27, 2022 Rady Children's Institute for Genomic Medicine (RCIGM) today announced that Stephen Kingsmore, MD, DSc, President and CEO, will receive the 2022 Luminary Award at the Precision Medicine World Conference (PMWC) to be held from June 28 to 30 in the Silicon Valley region of California.

The Luminary Award recognizes the recent contributions of prominent figures who have accelerated precision medicine into the clinic. Dr. Kingsmore will be recognized for his innovation in rapid neonatal molecular diagnoses using whole-genome sequencing. Additional PMWC 2022 honorees will include Dr. Albert Bourla from Pfizer and Dr. Stephen Hoge from Moderna.

RCIGM at PMWC:

On Wednesday, June 29, at 9 a.m., PST, Dr. Kingsmore will received the Luminary Award in a brief presentation by Martin Reese Ph.D., Co-Founder, CEO, Pres., Fabric Genomics.

Then at 9:15 a.m., PST, Dr. Kingsmore will speak on NICU Current State and Future of Whole Genome Sequencing as part of the Track 4 program, Sequencing in the Neonatal and Pediatric Intensive Care Unit. Dr. Kingsmore will be present to receive his award at this time.

Later at 11:15 a.m. PST, Russell Nofsinger, PhD, Senior Director of Business Development at RCIGM, will present on Partnerships to End the Diagnostic and Therapeutic Odysseys during the Clinical DX Showcase.

RCIGM will also host Booth #C1626 Hall C, Track 3, from June 28-30 during regular conference hours.

Rady Childrens Institute for Genomic Medicine

Rady Childrens Institute for Genomic Medicine is transforming neonatal and pediatric health care by harnessing the power of Rapid Precision Medicine to improve the lives of children and families facing rare genetic disease. Founded by Rady Childrens Hospital and Health Center, the Institute offers the fastest delivery of rapid Whole Genome Sequencing to enable prompt diagnosis and targeted treatment of critically ill newborns and children in intensive care. The Institute now provides clinical genomic diagnostic services for a growing network of more than 70 childrens hospitals. The vision is for this life-changing technology to become standard of care and enable clinicians nationwide to provide rapid, personalized care. Learn more about the non-profit Institute at RadyGenomics.org. Follow us on Twitter and LinkedIn.

###

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:

Dr. Stephen Kingsmore to receive Luminary Award at 2022 Precision Medicine World Conference - EurekAlert

Generation Bio Reports Business Highlights and First Quarter 2022 Financial Results – GuruFocus.com

Company continues to optimize cell-targeted lipid nanoparticle (ctLNP) delivery system for nonviral genetic medicine applications in liver, retina and vaccines

Cash balance of $337.0M expected to fund operations into 2024

CAMBRIDGE, Mass., May 05, 2022 (GLOBE NEWSWIRE) -- Generation Bio Co. ( GBIO), a biotechnology company innovating genetic medicines for people living with rare and prevalent diseases, reported business highlights and first quarter 2022 financial results.

We are making steady advances in optimizing our cell-targeted lipid nanoparticles, or ctLNPs, for use in our liver and retina programs as well as in developing our platform technologies for novel vaccine applications, said Geoff McDonough, M.D., president and chief executive officer of Generation Bio. We believe our approach to genetic medicine has the potential to create highly differentiated therapies capable of fulfilling significant unmet need, and we look forward to providing updates on our progress.

Key priorities across core therapeutic areas:

First Quarter 2022 Financial Results

About Generation Bio

Generation Bio is innovating genetic medicines to provide durable, redosable treatments for people living with rare and prevalent diseases. The companys non-viral genetic medicine platform incorporates a novel DNA construct called closed-ended DNA, or ceDNA; a unique cell-targeted lipid nanoparticle delivery system, or ctLNP; and a highly scalable capsid-free manufacturing process that uses proprietary cell-free rapid enzymatic synthesis, or RES, to produce ceDNA. The platform is designed to enable multi-year durability from a single dose, to deliver large genetic payloads, including multiple genes, to specific cell types, and to allow titration and redosing to adjust or extend expression levels in each patient. RES has the potential to expand Generation Bios manufacturing scale to hundreds of millions of doses to support its mission to extend the reach of genetic medicine to more people, living with more diseases, around the world.

For more information, please visit http://www.generationbio.com.

Forward-Looking Statements

Any statements in this press release about future expectations, plans and prospects for the company, including statements about our strategic plans or objectives, our technology platform, our research and clinical development plans, applications and preclinical data and other statements containing the words believes, anticipates, plans, expects, and similar expressions, constitute forward-looking statements within the meaning of The Private Securities Litigation Reform Act of 1995. Actual results may differ materially from those indicated by such forward-looking statements as a result of various important factors, including: uncertainties inherent in the identification and development of product candidates, including the conduct of research activities, the initiation and completion of preclinical studies and clinical trials and clinical development of the companys product candidates; uncertainties as to the availability and timing of results from preclinical studies and clinical trials; whether results from earlier preclinical studies will be predictive of the results of later preclinical studies and clinical trials; uncertainties regarding the timing and ability to complete the build-out of the companys manufacturing facility and regarding the RES manufacturing process; challenges in the manufacture of genetic medicine products; whether the companys cash resources are sufficient to fund the companys operating expenses and capital expenditure requirements for the period anticipated; the impact of the COVID-19 pandemic on the companys business and operations; expectations for regulatory approvals to conduct trials or to market products; as well as the other risks and uncertainties set forth in the Risk Factors section of our most recent annual report on Form 10-K, which is on file with the Securities and Exchange Commission, and in subsequent filings the company may make with the Securities and Exchange Commission. In addition, the forward-looking statements included in this press release represent the companys views as of the date hereof. The company anticipates that subsequent events and developments will cause the companys views to change. However, while the company may elect to update these forward-looking statements at some point in the future, the company specifically disclaims any obligation to do so. These forward-looking statements should not be relied upon as representing the companys views as of any date subsequent to the date on which they were made.

Investors and Media ContactMaren KillackeyGeneration Bio857-371-4638[emailprotected]

GENERATION BIO CO.CONSOLIDATED BALANCE SHEET DATA (Unaudited)(In thousands)

GENERATION BIO CO.CONSOLIDATED STATEMENTS OF OPERATIONS (Unaudited)(in thousands, except share and per share data)

Read the original:

Generation Bio Reports Business Highlights and First Quarter 2022 Financial Results - GuruFocus.com

Do agri-businesses ‘control’ agriculture? The emerging gene-editing revolution in Latin America is challenging that belief – Genetic Literacy Project

Small actions can create such huge differences. We see that butterfly effect playing out in Latin America. where the development of a new generation of crops using CRISPR and other new breeding techniques (NBTs) is now unfolding and whats smaller than a gene?

Changes in a single gene are revolutionizing science, making a big impact on medicine and agriculture: think COVID vaccines or genes that increase yield or climate resilience or nutritional value of crops. In the food sector, gene-edited tomatoes, wheat and other foods are already being consumed and commercialized, and dozens more are on the way.

The main challenge going forward in the agriculture sector, particularly in Latin America, is how NBTs will be regulated. As GMOs in which genes are moved between species to introduce specific traits? Transgenic techniques are heavily regulated in most jurisdictions because of the presence of so-called foreign genes.

Many of the innovation-blocking regulations were composed in the early 2000s when much less was known about genes, and fear and misunderstanding drove public opinion against the science, and politicians followed suit.

Scientists believe the obsession of distinguishing between what is natural and what is a foreign gene is scientifically silly, as even in nature genes flow from one species to another. This dubious notion has long throttled the biotech ag revolution.

If this is the regulatory model going forward, the CRISPR ag revolution will stall. Gene editingwhich is known as cisgenicscan introduce many new traits but is not a substitute in many cases for transgenicsdoes not involve moving genes among species.

The EU, at least for now, seems intent on sticking to the backward GMO regulations it adopted in 2001 and 2003 in the wake of food scares unrelated to biotechnology. The United States, Canada, China, Japan, and even the United Kingdom are shedding decade-old superstitions and taking steps to greenlight gene editing.

Latin American nations are also taking global leadership in the research and development of gene-edited crops. Argentina, Brazil, and other South American countries are being proactive, creating regulatory frameworks that will make these novel crops accessible to farmers and consumers in just a few years time. They will be evaluated as conventional crops, making the research and development process needed for environmental release and testing affordable to developers something that does not happen with transgenic GMOs in which only big companies can afford to run the expensive, time-consuming regulatory gauntlet.

The relatively minuscule costs necessary to bring new products to the evaluation stage could help unleash an entrepreneurial revolution in the food sector, ironically undercutting the influence of multinational agribusinesses. This regulatory revolution could reshape the publics perception of genetically modified crops and knee-cap aggressive anti-biotech activist groups that have spent decades spreading alarmist, unfounded fears that multinational corporations are out to control the global food supply.

In Latin America, agricultural biotechnology public research centers are popping up, allocating resources to develop novel crop varieties. New and small companies are being founded and funded. Together they are revolutionizing the future of food production in Latin America, with crops and foods being developed from a farmers and consumers perspective and by creating novel crop varieties with regional interest.

In Argentina, BioHeuris, an expert in weed management, is leading the synthetic biology revolution, working with CRISPR tools to develop next-generation herbicide-resistant soybean, sorghum, alfalfa, rice, and cotton. Created in 2016, BioHeuris accelerates plant breeding to evolve crops faster than weeds. Together with integrated pest management techniques, they expect to develop a sustainable system for numerous crops with varieties available for commercialization by 2026 or 2027.

With our platforms fully operational, time to market will be reduced by half and development costs cut 100 times compared to current GMO crops developed by the big seed companies, said co-founder and Director of Strategy Carlos Perez.

CONABIA, Argentinas biosafety commission, has already ruled that these gene-edited plants should not be regulated as GMOs. In January, APHIS in the United States and Brazils CTNBio (National Technical Biosafety Commission) also gave the green light for the introduction of BioHeuris CRISPR solution.

In Brazil, the government-funded company Embrapa is emerging as an innovation leader in exploiting. It developed the first gene-edited sugarcane in the world. It has two varieties: one offers higher cell wall digestibility and a higher concentration of sucrose in plant tissues. CTNBio has determined that neither variety will be regulated as a transgenic GMO.

According to the deputy head of Research and Development at Embrapa, the development of new cultivars through CRISPR is at the frontier of knowledge.

These cultivars are only the beginning, and they pave the way for the development and delivery of other cultivars for the production sector with characteristics that will directly impact the productivity and reduce production costs.

Colombia is following a different path. CIAT (International Center for Tropical Agriculture), a public research center, has been identified as a pioneer in genome editing in Colombia and is leading the national research and development of gene-edited crops. CIATs researchers have worked on several crops including cassava, rice, and beans. The modified rice variety is resistant to the hoja blanca virus, a disease that bleaches the leaves off and eventually kills rice plants. It is prevalent in Latin America. CIATs team is also exploring the use of genome editing to make it easier for people to digest beans.

Chiles research team is based in the Department of Biology of Sciences Faculty at the University of Chile. It is working on creating gene-edited crops, focusing on tomatoes and kiwi that can tolerate drought and soil salinity. In addition, they are using CRISPR to modify apples to increase their nutritional profile, adding a higher content of carotenoids, and resisting the oxidation that causes browning after they are cut.

In addition to the efforts made by the University of Chile, NeoCrop, an emerging company, is joining them in the domestic research landscape of gene-edited crops. They are developing gene-edited wheat that produces higher levels of fiber and is resistant to drought and extreme heat. NeoCrop expects to have this new gene-edited wheat variety ready for field trials this year or next.

This emerging dynamic offers a preview of the future in which small and entrepreneurial start-ups will lead in the research and development of novel crops with a focus on farmers and consumers. Increasingly, NGO criticism that crop biotechnology in Latin America is being driven by monopolistic corporations is obsolete and misguided.

Luis Ventura is a trained biologist and biosafety expert. Luis is a member of Mexican Scientist Allies for Knowledge in Agriculture and a 2016 Alliance for Science Global Leadership Fellow and trainer. View Luis LinkedIn pagehere. Find Luis on Twitter@Iuisventura

See the original post:

Do agri-businesses 'control' agriculture? The emerging gene-editing revolution in Latin America is challenging that belief - Genetic Literacy Project

New hope for IVF patients as global study published in Human Reproduction shows AI can effectively assess genetic integrity of embryos USA – English -…

SAN FRANCISCO, June 28, 2022 /PRNewswire/ -- Human Reproduction journal has published the ground-breaking results of an international clinical study, where a novel AI algorithm called Life Whisperer Genetics was developed by AI healthcare company Presegen to assess the genetic integrity of embryos using only images. The assessment is non-invasive, low-cost, and provides results instantly. This is in stark contrast with PGT-A, the standard method used in IVF today, which requires an invasive and potentially risky biopsy to remove a portion of the embryo, followed by an expensive and time-consuming genetic testing procedure.

The study was conducted with IVF clinics globally, including Ovation Fertility (USA), IVF-Life (Europe), Alpha IVF & Women's Specialists (SE Asia), and Wings IVF (India). Results showed that the AI algorithm could identify whether an embryo was genetically normal, or 'euploid'. Identification of euploid embryos can result in improved clinical outcomes, such as a better chance at pregnancy success.

Presagen's Chief Medical Science Officer Dr Sonya Diakiw explained "Because this assessment is based on images alone, it is not as accurate as PGT-A itself, which involves actual DNA sequencing. But we are finding that PGT-A results themselves can be variable, as they depend on the embryo sample being tested. PGT-A only tests 5 cells from a total of around 200, so it is not always representative of the entire embryo. Life Whisperer Genetics is a whole-embryo assessment of genetic integrity that does not require any invasive procedures, which can be used to prioritize embryos for use in IVF procedures."

The technology was evaluated prospectively on patients in Europe in collaboration with the IVF-Life Group. Dr Jon Aizpurua from IVF-Life said "Life Whisperer Genetics can be used for patients as a pre-screen, to ensure we only genetically test embryos that are likely to be normal, saving patients time and money. For patients who are not comfortable with invasive genetic tests, or in countries like Germany where invasive genetic tests are not permitted, Life Whisperer Genetics is a viable alternative to help select embryos that are most likely to be euploid."

Prospective studies were also performed in collaboration with Alpha IVF & Women's Specialists in Malaysia. Chief Embryologist Adelle Yun Xin Lim said "Computer vision with AI may revolutionise IVF treatment and this new technique is another milestone of AI in IVF. The technique will help doctors and embryologists around the world to predict the chromosome status of embryos in a rapid and non-invasive manner enabling the prioritization of embryos that are likely to be euploid for transfer or for further confirmatory PGT testing, leading to a faster time to pregnancy and reducing the cost of the treatment."

Ovation Fertility's VP of Scientific Advancement, Dr Matthew (Tex) VerMilyea said "This new product is very exciting. In some ways it is like a 'Rapid Antigen Test (RAT)' for embryo assessment, providing a non-invasive, instantaneous evaluation of genetic integrity, which will have massive potential for the US market when it receives FDA approval."

Life Whisperer Genetics is already available for IVF clinics and their patients in over 40 countries globally. It can be used in combination with Life Whisperer Viability, which assesses if an embryo is likely to lead to a pregnancy. International clinical studies have shown that Life Whisperer Viability can perform better than embryologists' current manual embryo assessment methods. Together, Life Whisperer Viability and Life Whisperer Genetics provide a comprehensive assessment of embryo quality.

Paper Title

Development of an artificial intelligence model for predicting the likelihood of human embryo euploidy based on blastocyst images from multiple imaging systems during IVF

https://academic.oup.com/humrep/advance-article/doi/10.1093/humrep/deac131/6604228

Authors

S. M. Diakiw1, J. M. M. Hall1,2,3, M. D. VerMilyea4,5, J. Amin6, J. Aizpurua7, L. Giardini7, Y. G. Briones7, A. Y. X. Lim8, M. A. Dakka1, T. V. Nguyen1, D. Perugini1, M. Perugini1,9

Paper Abstract

STUDY QUESTION

Can an artificial intelligence (AI) model predict human embryo ploidy status using static images captured by optical light microscopy?

SUMMARY ANSWER

Results demonstrated predictive accuracy for embryo euploidy, and showed a significant correlation between AI score and euploidy rate, based on assessment of images of blastocysts at Day 5 after IVF.

MAIN RESULTS AND THE ROLE OF CHANCE

Overall accuracy for prediction of euploidy on a blind test dataset was 65.3%, with a sensitivity of 74.6%. When the blind test dataset was cleansed of poor quality and mislabeled images, overall accuracy increased to 77.4%. This performance may be relevant to clinical situations where confounding factors, such as variability in PGT-A testing, have been accounted for. There was a significant positive correlation between AI score and the proportion of euploid embryos, with very high scoring embryos (9.0-10.0) twice as likely to be euploid than the lowest scoring embryos (0.0-2.4). When using the genetics AI model to rank embryos in a cohort, the probability of the top-ranked embryo being euploid was 82.4%, which was 26.4% more effective than using random ranking, and ~13-19% more effective that using the Gardner score. The probability increased to 97.0% when considering the likelihood of one of the top two ranked embryos being euploid, and the probability of both top two ranked embryos being euploid was 66.4%. Additional analyses showed that the AI model generalized well to different patient demographics and could also be used for evaluation of Day 6 embryos and for images taken using multiple time-lapse systems. Results suggested that the AI model could potentially be used to differentiate mosaic embryos based on the level of mosaicism.

WIDER IMPLICATIONS OF THE FINDINGS

These findings collectively support the use of this genetics AI model for evaluation of embryo ploidy status in a clinical setting. Results can be used to aid in prioritizing and enriching for embryos that are likely to be euploid for multiple clinical purposes, including selection for transfer in the absence of alternative genetic testing methods, selection for cryopreservation for future use, or selection for further confirmatory PGT-A testing, as required.

About Presagen and Life Whisperer

Presagen is an AI healthcare company that is changing the way clinics, patients, and medical data from around the world are connected through AI. Its platform, The Social Network for Healthcare, connects clinics and patients globally, and enables collaboration and data sharing to create scalable AI healthcare products that are affordable and accessible for all. The decentralized network democratizes the creation of AI products, promotes collaboration through incentives, and protects data privacy and ownership. With a focus on improving Women's Health outcomes globally, Presagen's first product, Life Whisperer, is being used by IVF clinics globally to improve pregnancy outcomes for couples struggling with fertility. With a vision of creating the largest network of clinics, patients, and medical data from around the world, Presagen is driving the future of AI Enhanced Healthcare.

About Ovation Fertility

Ovation Fertility is a national network of reproductive endocrinologists and scientific thought leaders focused on reducing the cost of having a family through more efficient and effective fertility care. Ovation's IVF and genetics laboratories, along with affiliated physician practices, work collaboratively to raise the bar for IVF treatment, with state-of-the-art, evidence-based fertility services that give hopeful parents the best chance for a successful pregnancy. Physicians partner with Ovation to offer their patients advanced preconception carrier screening; preimplantation genetic testing; donor egg and surrogacy services; and secure storage for their frozen eggs, embryos and sperm. Ovation also helps IVF labs across America improve their quality and performance with expert off-site lab direction and consultation. Learn more about Ovation's vision of a world without infertility at http://www.OvationFertility.com.

About IVF-Life

IVF-Life is a group of fertility clinics specialized in complex cases. Centres located inSpain and the UK have the latest advances in Reproductive Medicine and outstanding professionals in this field. The constant innovation and a firm commitment to technology keep IVF-Life at the forefront in the assisted reproduction field treating patients from all over the world with ahigh degree of success. IVF-Life perform a comprehensive range of treatments and diagnostic tests with the aim to provide effective solutions to a wide variety of fertility problems.

About Alpha IVF & Women's Specialists

Alpha IVF group comprises IVF centres in Kuala Lumpur, Penang and Singapore. Alpha IVF is a world-class fertility treatment provider bringing the most advanced fertility technologies and excellent success rates in achieving the goal of having a baby. Alpha IVF consists of a team of highly qualified and skilled doctors, scientists and nurses that deliver international standards of patient care. As the name Alpha suggests, the team have pioneered numerous innovative fertility treatments. Alpha IVF offers its patients access to a network of fertility experts and facilities fully equipped with a full range of cutting-edge laboratories, innovative technologies such as Artificial Intelligence, Next Generation Sequencing (NGS), 100% post-warm survival rate for embryo cryopreservation, time-lapse embryo monitoring, PIEZO-ICSI, sperm separation technologies and many others. Continuous R&D have led Alpha IVF to achieve numerous world firsts and innovative fertility treatment protocols both regionally and globally.

About Wings

Wings IVF comprises a chain of leading fertility clinics across India, with more than 12,000 live births through IVF. Wings hospitals are state of art specialty hospitals & clinics providing all infertility treatments and IVF. They provide top quality, comprehensive, holistic care to women of India at a reasonable cost. An interdisciplinary team of expert and caring professionals is committed to meeting the physical as well as emotional and spiritual needs for each woman and her family. The Wings Hospitals have been designed and furnished to provide a high level of fertility care with comfort and privacy.

SOURCE Presagen

Go here to see the original:

New hope for IVF patients as global study published in Human Reproduction shows AI can effectively assess genetic integrity of embryos USA - English -...

Akouos Presents Nonclinical Data Supporting the Planned Clinical Development of AK-OTOF and Strategies for Regulated Gene Expression in the Inner Ear…

- Nonclinical data demonstrate that a single intracochlear administration of an AAVAnc80 vector led to durable restoration of auditory function and was well tolerated, supporting planned clinical development of AK-OTOF for the treatment of OTOF-mediated hearing loss

- MicroRNA target site (miR-TS)-incorporation in AAV vectors is shown to have potential benefits for de-targeting transgene expression in the inner ear, supporting future development of gene therapies targeting a broad range of inner ear conditions

BOSTON, May 19, 2022 (GLOBE NEWSWIRE) -- Akouos, Inc. ( AKUS), a precision genetic medicine company dedicated to developing potential gene therapies for individuals living with disabling hearing loss worldwide, presented nonclinical data at the American Society of Gene and Cell Therapy (ASGCT) 25th Annual Meeting. The company gave two nonclinical presentations at the meeting: one that supports the planned clinical development of AK-OTOF, a gene therapy intended for the treatment of OTOF-mediated hearing loss; and another that supports the potential use of microRNA target site (miR-TS) in adeno-associated viral (AAV) vectors for regulated gene expression in the inner ear.

We are excited to present these nonclinical data, which highlight our precision genetic medicine platform and the potential of genetic medicines to address a broad range of inner ear conditions, to the gene and cell therapy community. The AK-OTOF nonclinical data demonstrate durable restoration of auditory function and show that the product candidate was systemically and locally well tolerated in two translationally relevant animal species, said Manny Simons, Ph.D., founder, president, and chief executive officer of Akouos. As we continue to progress toward planned IND submissions for AK-OTOF in the first half of 2022 and AK-antiVEGF in 2022, we are encouraged by the growing body of evidence supporting these filings, as well as by our efforts to advance preclinical development of other potential gene therapies for inner ear conditions, such as GJB2-mediated hearing loss, and to develop platform capabilities that can be applied to regenerative medicine approaches in the inner ear.

Nonclinical In Vivo Expression, Durability of Effect, Biodistribution/Shedding, and Safety Evaluations Support Clinical Development of AK-OTOF (AAVAnc80-hOTOF Vector) for OTOF-mediated Hearing Loss Presenting Author: Ann E. Hickox, Ph.D.Session Title and Room: Ophthalmic and Auditory Diseases; Salon G

AK-OTOF is an AAV vector-based gene therapy intended for the treatment of patients with otoferlin gene (OTOF)-mediated hearing loss by delivering transgenes encoding OTOF to inner hair cells (IHCs). Following intracochlear delivery, and subsequent co-transduction of IHCs by each component vector, the two transgene products recombine to generate a full-length otoferlin mRNA transcript and subsequently a full-length otoferlin protein. Results from this presentation show:

Together, these nonclinical studies further support the planned clinical development of AK-OTOF for the treatment of OTOF-mediated hearing loss.

The digital presentation is located at https://akouos.com/gene-therapy-resources/.

Evaluating miR-Target Sites as a Strategy to Allow AAV Vector-based De-targeting of Gene Expression in the Inner EarPresenting Author: Richard M. Churchill Jr.Poster Board Number: Tu-37

In the development of AAV gene therapy vectors, a goal is to generate safe and effective product candidates that deliver targeted transgene expression. Ubiquitous promoters can drive strong widespread expression in the inner ear in mice and NHPs. This expression can be well tolerated across the inner ear, as is the case for Akouoss first two programs, AK-OTOF and AK-antiVEGF. Addition of selective cis-regulatory elements may be needed for some transgenes, such as GJB2, where expression in a portion of nontarget cells is not well tolerated. This nonclinical study explored the potential use of miR-TS incorporation in AAV vectors for de-targeting transgene expression in different cell types of the cochlea. Using an in vitro model, expression of transgene mRNA and protein in the presence or absence of the target sites was evaluated. Akouos identified multiple microRNA target sites to drive various differential expression patterns demonstrating that a combination of AAVAnc80 and miR-TS can drive expression in supporting cells, while limiting expression in hair cells in cochlear explants. Future work will focus on evaluating miR-TS regulation in vivo and identifying combinations of different miR-TSs to enhance de-targeting in specific cell types where, for example, expression driven by ubiquitous promoters is not well tolerated.

The digital presentation is located at https://akouos.com/gene-therapy-resources/.

About AkouosAkouos is a precision genetic medicine company dedicated to developing gene therapies with the potential to restore, improve, and preserve high-acuity physiologic hearing for individuals living with disabling hearing loss worldwide. Leveraging its precision genetic medicine platform that incorporates a proprietary adeno-associated viral (AAV) vector library and a novel delivery approach, Akouos is focused on developing precision therapies for forms of sensorineural hearing loss. Headquartered in Boston, Akouos was founded in 2016 by leaders in the fields of neurotology, genetics, inner ear drug delivery, and AAV gene therapy.

Forward-Looking StatementsStatements in this press release about future expectations, plans and prospects, as well as any other statements regarding matters that are not historical facts, may constitute forward-looking statements within the meaning of The Private Securities Litigation Reform Act of 1995. These statements include, but are not limited to, statements relating to the initiation, plans, and timing of our future clinical trials and our research and development programs, and the timing of our IND submissions for AK-OTOF and AK-antiVEGF. The words anticipate, believe, continue, could, estimate, expect, intend, may, plan, potential, predict, project, should, target, will, would, and similar expressions are intended to identify forward-looking statements, although not all forward-looking statements contain these identifying words. Actual results may differ materially from those indicated by such forward-looking statements as a result of various important factors, including: our limited operating history; uncertainties inherent in the development of product candidates, including the initiation and completion of nonclinical studies and clinical trials; whether results from nonclinical studies will be predictive of results or success of clinical trials; the timing of and our ability to submit applications for, and obtain and maintain regulatory approvals for, our product candidates; our expectations regarding our regulatory strategy; our ability to fund our operating expenses and capital expenditure requirements with our cash, cash equivalents, and marketable securities; the potential advantages of our product candidates; the rate and degree of market acceptance and clinical utility of our product candidates; our estimates regarding the potential addressable patient population for our product candidates; our commercialization, marketing, and manufacturing capabilities and strategy; our ability to obtain and maintain intellectual property protection for our product candidates; our ability to identify additional products, product candidates, or technologies with significant commercial potential that are consistent with our commercial objectives; the impact of government laws and regulations and any changes in such laws and regulations; risks related to competitive programs; the potential that our internal manufacturing capabilities and/or external manufacturing supply may experience delays; the impact of the COVID-19 pandemic on our business, results of operations, and financial condition; our ability to maintain and establish collaborations or obtain additional funding; and other factors discussed in the Risk Factors section of our Quarterly Report on Form 10-Q for the quarter ended March 31, 2022, which is on file with the Securities and Exchange Commission, and in other filings that Akouos may make with the Securities and Exchange Commission. 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 statement, whether as a result of new information, future events or otherwise.

Contacts

Media:Katie Engleman, 1AB[emailprotected]

Investors:Courtney Turiano, Stern Investor Relations [emailprotected]

Excerpt from:

Akouos Presents Nonclinical Data Supporting the Planned Clinical Development of AK-OTOF and Strategies for Regulated Gene Expression in the Inner Ear...

Realizing Gene Therapy and Cell Regeneration in Heart Disease – BioSpace

Only a few companies are using gene therapy to treat heart disease. One of them, Tenaya Therapeutics, is using adeno-associated virus 9 (AAV9) to deliver healthy copies of select genes to target cardiac tissue and express the protein of interest at high levels. Whats particularly interesting is that it also has a cell regeneration program that at least in animal models shows the potential toregrow cardiomyocytes (which die after heart attacks).

Were heart disease specialists with multiple approaches. We saw early opportunities for gene therapy to address genetic forms of heart disease, so we had good reason to go in that direction, Tenaya CEO Faraz Ali told BioSpace, emphasizing that Tenaya is modality agnostic. Its approaches include programs from three platforms, including cellular regeneration, gene therapy and precision medicine.

With three of its gene therapy programs in preclinical development, Ali said he expects TN-201 to be the first to enter the clinic, and Tenaya anticipates filing an IND for that program in 2022. It treats the leading genetic cause of hypertrophic cardiomyopathy (gHCM). For many, this gHCM program is particularly exciting. It would be the first example of a disease-modifying therapy for that indication, Ali said.

TN-201 delivers a functional MYBPC3 gene via AAV9 to address gene mutations that lead to thickening heart walls, arrhythmia, dysfunction, heart failure and sudden cardiac death. There are approximately 115 thousand people in the U.S. with this disease. Disease symptoms can occur with only a single mutation in a single allele, but when two mutations occur, it is life-threatening within the first few months of life.

Weve shown, in animal models, that we can reverse the declining heart function, Ali said, as well as the arrhythmia and the hypertrophy (thickness of the heart), returning it to normal size after a single dose. In animals, prevention or reversal of symptoms leads to extended survival. To be clear, whether (such symptoms) can be returned to normal in humans is yet to be determined, but we think some symptoms could be significantly improved, he stressed. TN-201 was granted Orphan Drug Designation by the U.S. Food and Drug Administration (FDA).

This, and Tenayas other gene therapies are possible because of the revelation by multiple gene therapy pioneers that AAV9 vectors have a propensity to go to the heart.

They showed that AAV9 does a good job with broad distribution and transduction of the cells, Ali said. So, although not every target cell receives a healthy copy of the gene, the vector copy number, on average, is sufficiently high that each cell has a shot at being functionally corrected.

The other factor is the growing body of knowledge regarding the genetic forms of heart disease. We asked several leading gene therapy experts about the most attractive genetic forms of heart diseases, but couldnt get a good answer, Ali recalled. There was a gap in either their knowledge or their imagination.

Now, we have much better insight into the genetic forms of heart disease, such as hypertrophic cardiomyopathy (gHCM) and arrhythmogenic right ventricular cardiomyopathy (gARVC). Once we started to ask the right questions, we found several opportunities. In Tenayas case, the technology to answer those questions coincided with advancements in delivery and manufacturing, which also created new opportunities.

Gene therapy isnt Tenayas only approach, though.

Some forms of heart disease are caused by the loss of cardiomyocytes, for example, after a heart attack, Ali pointed out. Once they are lost, no therapy exists today to bring those cells back. Our approach is to create new cells in vivo, using AAV to deliver proprietary combinations of genes.

That program is based on work by the company co-founders Deepak Srivastava, M.D., president of the Gladstone Institutes, and Eric Olson, Ph.D., at the University of Texas Southwestern Medical Center. Dr. Srivastava took Shinya Yamanakas work on induced pluripotent stem cells (for which he won the 2012 Nobel Prize) a step further, finding, with Dr. Olson, a combination of factors that could turn cardiac fibroblasts into new cardiomyocytes. By altering those factors, they found they also could induce existing cardiomyocytes to divide and create new cells.

Both of these approaches have achieved proof of concept in large animal models with human-sized hearts, Ali said. This means, potentially, that hearts may possibly be returned to normal function after heart attacks.

Another Tenaya program dubbed TYA-11631 uses a small molecule to treat heart failure with preserved ejection fraction (HFpEF) and Tenaya also expects to file an IND for that program in 2022. HFpEF disease involves a thickening and stiffening of the heart's ventricles (the major pumping chambers) that restricts the heart's ability to fill up with blood in between heartbeats. There are approximately 3 million people in the U.S. with this disease.

Each of our programs is built on strong scientific foundations. Were not a me-too company, Ali said.

The future of heart disease is changing dramatically with the steady advance of precision medicine approaches, doing for cardiac patients what has been done so successfully for oncology patients. After decades oftreating the big categories of heart disease hypertension, for example the ability to address the genetics of heart disease is within sight.

As Ali said, We have better tools now, and better delivery methods, and can begin to apply them to the leading cause of death throughout the world to address heart disease in a more precise way, thus turning large categories into ever-smaller categories of disease. As in oncology, the desired result with this approach is more precise and less expensive clinical trials, faster approvals and ultimately more hope and better therapies for patients.

Excerpt from:

Realizing Gene Therapy and Cell Regeneration in Heart Disease - BioSpace

Small children getting less sick from Omicron; Genetic mutation protects against severe COVID – Reuters

A child is seen near a syringe containing a dose of the Pfizer-BioNTech coronavirus disease (COVID-19) vaccine at Smoketown Family Wellness Center in Louisville, Kentucky, U.S., November 8, 2021. REUTERS/Jon Cherry

Register

Jan 21 (Reuters) - The following is a summary of some recent studies on COVID-19. They include research that warrants further study to corroborate the findings and that has yet to be certified by peer review.

Small children are getting less sick from Omicron

In very young children, the Omicron variant of the coronavirus causes less severe disease than the Delta variant, according to a new study.

Register

Researchers reviewed data on nearly 80,000 U.S. children under age 5 with a first infection, including 7,201 infected in late December or early January when Omicron was causing more than 90% of cases. After accounting for other risk factors, including medical conditions and socioeconomic circumstances, researchers found children infected during the Omicron surge had a 29% lower risk of emergency department visits, a 67% lower risk of hospitalization, a 68% lower risk of needing intensive care, and a 71% lower risk of needing machines to breathe, compared to children infected with Delta. However, "because of Omicron's increased transmissibility, the overall number of emergency department visits, hospitalizations, ICU admissions, and mechanical ventilator use in children may still be greater" with Omicron than with Delta, according to a report posted on medRxiv ahead of peer review.

The investigators have also observed that infection rates were disproportionately higher in Black and Hispanic children for both Omicron and Delta for this age group, and the gap widened for infections with Omicron, said study leader Rong Xu of Case Western Reserve University School of Medicine. Not yet published data shows that "children under 5 had the highest infection rate with Omicron" compared to older children and adults in all age groups, she said.

Genetic mutation protects against severe COVID-19

New findings add to evidence that people with a certain version of a gene are less likely to develop severe COVID-19.

Earlier research had identified a specific group of genes, called the OAS1/2/3 gene cluster, as being involved in the risk for severe COVID-19. One version of a gene in that cluster - passed down from Neanderthals - appeared to protect against severe disease, reducing the risk by about 23%. The earlier research was done mainly in people of European ancestry. According to a report published in Nature Genetics, researchers now see the same association of this genetic variant with less severe COVID-19 in people of African ancestry.

"The fact that individuals of African descent had the same protection allowed us to identify the unique variant in the DNA that actually protects from COVID-19 infection," coauthor Dr. Jennifer Huffman of said in a statement. OAS genes are involved in a cascade of effects that help cells fight viruses, the researchers said. Understanding of these genes and their effect on COVID-19 risks could aid development of future drugs, they added.

Fewer Delta breakthroughs with Moderna vs Pfizer/BioNTech

When the Delta variant of the coronavirus was prevalent in the United States, recipients of two doses of Moderna's (MRNA.O) mRNA vaccine were less likely to experience a breakthrough infection - and if they did, were slightly less likely to be hospitalized - than recipients of two doses of the mRNA vaccine from Pfizer (PFE.N) and BioNTech , a large study found.

Researchers analyzed health records of more than 637,000 vaccine recipients who were not previously infected with the virus and had not gotten a booster shot. As reported on Thursday in JAMA, breakthrough infections steadily increased every month from July to November 2021, with higher rates in the Pfizer/BioNTech group. In November, there were 2.8 cases among every thousand people vaccinated with the Pfizer/BioNTech shots, compared to 1.6 cases per thousand recipients of the Moderna vaccines. The vaccines protected equally well against death, but the hospitalization rate was 12.7% for infected Moderna recipients and 13.3% for Pfizer/BioNTech recipients. When the researchers compared 62,584 Moderna recipients to a closely-matched equal-sized group of Pfizer/BioNTech recipients, the risk for breakthrough infection was 15% lower in the Moderna group.

"Although there is a difference in breakthrough infections, both vaccines are highly protective against SARS-COV2 infection and especially against the most severe consequences of infection," said coauthor Pamela Davis of Case Western Reserve University School of Medicine in a statement.

Click for a Reuters graphic on vaccines in development.

Register

Reporting by Nancy Lapid; Editing by Bill Berkrot

Our Standards: The Thomson Reuters Trust Principles.

The rest is here:

Small children getting less sick from Omicron; Genetic mutation protects against severe COVID - Reuters

Bionano Genomics Announces its Support for the AGMG – GlobeNewswire

SAN DIEGO, Jan. 19, 2022 (GLOBE NEWSWIRE) -- Bionano Genomics, Inc. (BNGO), pioneer of optical genome mapping (OGM) solutions on the Saphyr system and provider of NxClinical, the leading software solutions for visualization, interpretation and reporting of genomic data, today announced that it will financially support the ACMG Foundation for Genetic and Genomic Medicine and its Next Generation Fellowship & Residency Training Awards Program in genetics and genomics.

Patients with genetic and genomic disorders require access to medical professionals who are qualified to analyze and treat their often extremely rare conditions. Due to shortages in medical professionals in specialty areas of clinical genomics, it can be difficult for a patient to connect with board-certified PhDs or MDs in medical genetics and genomics. The Next Generation Fellowship & Residency Training Awards Program has been helping fill that gap for the past twenty years.

As the chief executive officer for the ACMG Foundation for Genetic and Genomic Medicine (ACMGF) and in support of the Next Generation Fellowship & Residency Training Awards Program, I want to personally thank and recognize Bionano Genomics. Their generous commitment to three years of support for our Laboratory Genetics and Genomics (LGG) fellowships will be critical to bringing new talent to our specialty field. We believe Bionanos support will contribute to the future of clinical genomics, and Im glad that Bionano Genomics recognizes the importance of supporting ACMGF and the program in laboratory genetic medicine, said Dr. Max Muenke, FACMG.

The Next Generation Fellowship & Residency Training Awards Program is focused on expanding the workforce pipeline by attracting excellent physicians and PhD laboratorians to genetic and genomic medicine. Bionanos three years of financial support in this program is expected to play a significant role in the ACMGFs work to add more experts to the field and expand the reach of genetic and genomic medicine.

Bionano is honored and proud to support theACMGF and the laboratory geneticists who will be supported by these fellowships, commented Erik Holmlin, PhD, President and Chief Executive Officer of Bionano. Bionanos mission is to transform the way the world sees the genome so we can elevate the health and wellness of all people. Supporting the development of the next generation of leaders in the practice of laboratory genetic medicine through theACMGF Next Generation Fellowship & Residency Awards Program is important in achieving that objective and we are thankful to theACMGF for the opportunity to be a part of their efforts.

About Bionano Genomics

Bionano Genomics is a provider of genome analysis solutions that can enable researchers and clinicians to reveal answers to challenging questions in biology and medicine. The Companys mission is to transform the way the world sees the genome through OGM solutions, diagnostic services and software. The Company offers OGM solutions for applications across basic, translational and clinical research. Through its Lineagen business, the Company also provides diagnostic testing for patients with clinical presentations consistent with autism spectrum disorder and other neurodevelopmental disabilities. Through its BioDiscovery business, the Company also offers an industry-leading, platform-agnostic software solution, which integrates next-generation sequencing and microarray data designed to provide analysis, visualization, interpretation and reporting of copy number variants, single-nucleotide variants and absence of heterozygosity across the genome in one consolidated view. For more information, visit http://www.bionanogenomics.com, http://www.lineagen.comor http://www.biodiscovery.com.

Forward-Looking Statements of Bionano Genomics

This press release contains forward-looking statements within the meaning of the Private Securities Litigation Reform Act of 1995. Words such as may, will, expect, plan, anticipate, estimate, intend and similar expressions (as well as other words or expressions referencing future events, conditions or circumstances) convey uncertainty of future events or outcomes and are intended to identify these forward-looking statements. Forward-looking statements include statements regarding our intentions, beliefs, projections, outlook, analyses or current expectations concerning, among other things, the benefits of Bionanos support of ACMGF and its Next Generation Fellowship & Residency Awards Program, Bionanos anticipated role in advancing ACMGFs work and building the future of clinical genomics, and the contributions of Bionanos support toward bringing talent to the clinical genomics field. Each of these forward-looking statements involves risks and uncertainties. Actual results or developments may differ materially from those projected or implied in these forward-looking statements. Factors that may cause such a difference include the risks and uncertainties associated with: the impact of the COVID-19 pandemic on our business and the global economy; general market conditions; changes in the competitive landscape, including the introduction of competitive technologies or improvements in existing technologies; failure of ACMGF or its Next Generation Fellowship & Residency Awards Program to meet expectations; changes in our strategic plans; our ability to obtain sufficient financing to fund our strategic plans; and the risks and uncertainties associated with our business and financial condition in general, including the risks and uncertainties described in our filings with the Securities and Exchange Commission, including, without limitation, our Annual Report on Form 10-K for the year ended December 31, 2020 and in other filings subsequently made by us with the Securities and Exchange Commission. All forward-looking statements contained in this press release speak only as of the date on which they were made and are based on managements assumptions and estimates as of such date. We do not undertake any obligation to publicly update any forward-looking statements, whether as a result of the receipt of new information, the occurrence of future events or otherwise.

CONTACTSCompany Contact:Erik Holmlin, CEOBionano Genomics, Inc.+1 (858) 888-7610eholmlin@bionanogenomics.com

Investor Relations:Amy ConradJuniper Point+1 (858) 366-3243amy@juniper-point.com

Media Relations:Michael SullivanSeismic+1 (503) 799-7520michael@teamseismic.com

More here:

Bionano Genomics Announces its Support for the AGMG - GlobeNewswire

Discovery of hundreds of genes potentially associated with ALS may steer scientists toward treatments – Stanford Medical Center Report

In addition to finding many genes that could contribute to the ALS, the researchers believe the study has settled a few important questions about the disease.

Theres a long-standing debate about where ALS originates in the cell, said Johnathan Cooper-Knock, a Stanford visiting scholar and lecturer at the University of Sheffield in the United Kingdom. This new technique has surfaced genetic evidence that really pins down the axon of motor neurons as the place of disease origin. (The axon of the neuron is a long cord that helps transmit electrical signals from one neuron to another.)

A paper describing the study was published Jan. 18 in Neuron. Snyder, the Stanford W. Ascherman, MD, FACS, Professor of Genetics, is the senior author. Zhang and Cooper-Knock are co-lead authors.

Typically, ALS researchers investigate one gene at a time, performing in-depth analyses to tease out if and how that gene might contribute to the onset of the disease. The Stanford teams approach was to cast a net far and wide for genes that may play a role in ALS. Zhang trained the algorithm to sift through millions of data points from studies known as genome-wide associated screens, which contain anonymized genetic information from thousands of patients with and without ALS. The strategy was to look for genetic mutations that often occur in people who have ALS.

The team narrowed the search further: Sorting through ALS patients data, the algorithm looked for mutations only in genes that support motor neuron function. Searching only in motor neurons allowed our approach to discover more risk genes compared with previous methods, Zhang said. The analysis spit out 690 candidate genes, some that were already known to be implicated in ALS.

We can use this information to learn more about how and why motor neurons fail in ALS, Cooper-Knock said. As an example, he added, Many of the genes we uncovered pointed to the disease originating in the axon of the cell, rather than the cell body.

Previously it was not clear if axon defects were an effect of the disease, but our results indicate these defects are likely causative, added Snyder.

One gene, which repeatedly showed up in the data analysis, caught the researchers attention: KANK1, which is involved in functions at the very end of the axon. Through a series of experiments using stem cells and gene editing, the team showed that mutations in this gene lead to loss of a protein called TDP-43 from the nucleus of motor neurons, a hallmark of ALS.

If you were to look in the brains of 100 people with ALS and analyze the motor neurons, youd see this loss of TDP in something like 98, Cooper-Knock said. Its almost the definition of ALS. If this phenomenon isnt occurring, you probably dont have ALS.

The finding is an exciting discovery, but its too early to consider KANK1 a drug target. Zhang said. More research will be needed to determine if reversing the effects of a mutated KANK1 gene can help treat the disease.

The team also plans to do some experimental work on other hits from their dataset to determine whether any of the other hundreds of genes identified in the analysis could lead to ALS pathology.

Other Stanford co-authors of the study are life science researchers Minyi Shi, PhD, and Annika Weimer, PhD.

Researchers from the University of Sheffield; UC San Francisco; the Montreal Neurological Institute; Lund University; the Weizmann Institute of Science; and the University Medical Center in Utrecht, the Netherlands, contributed to this study.

This study was funded by the European Research Council, Health~Holland, the ALS Foundation Netherlands, the National Lottery of Belgium, the KU Leuven Opening the Future Fund, the Kingsland Fellowship, the My Name5 DoddieFoundation, the Wellcome Trust and the National Institute for Health Research.

Original post:

Discovery of hundreds of genes potentially associated with ALS may steer scientists toward treatments - Stanford Medical Center Report

Ceptur Therapeutics Launches with $75M Series A Financing to Advance RNA Therapeutics Based on Proprietary U1 Adaptor Technology – Business Wire

PHILADELPHIA--(BUSINESS WIRE)--Ceptur Therapeutics, Inc. (Ceptur), a biotechnology company focused on developing targeted oligonucleotide therapeutics based on U1 Adaptor technology, today announced the completion of a $75M Series A financing. The round was co-led by venBio Partners and Qiming Venture Partners USA with participation by new investors Perceptive Xontogeny Venture (PXV) Fund, Bristol Myers Squibb and Janus Henderson Investors along with existing Seed investors Affinity Asset Advisors, Boxer Capital and LifeSci Venture Partners.

We are extremely grateful for the support of our new and existing investors, said Ceptur Therapeutics co-founder, President and CEO P. Peter Ghoroghchian, MD, PhD. In 2021, we in-licensed and internally expanded our foundational IP portfolio on U1 Adaptor technology; we further recruited a world-class scientific advisory board, comprising academic and industry leaders in oligonucleotide therapeutics. Moving forward, Ceptur will use the proceeds of this financing round to advance our broad discovery pipeline of differentiated genetic medicines.

U1 Adaptors are bivalent oligonucleotides that engage sequence-specific mRNA and the U1 small nuclear ribonuclear protein (U1 snRNP), which is a ubiquitous intracellular machine that regulates transcription and splicing. U1 Adaptor therapeutics control gene expression at the pre-mRNA level within the nucleus, affording advantageous properties for drugging difficult targets.

Therapeutic approaches that target RNA have become an essential treatment modality for patients with genetic diseases and a priority for many biopharma companies; we believe that the U1 Adaptor technology is a differentiated approach to RNA regulation that has multiple potential advantages over current technologies, said Aaron Royston, M.D., M.B.A., Managing Partner at venBio Partners. We are excited to further build out Cepturs team and capabilities, to demonstrate these unique applications, and, ultimately, to advance novel therapeutics for patients with genetic diseases.

Colin Walsh, Ph.D., Partner at Qiming Venture Partners USA, added, RNA-based drugs have already become an essential tool in our therapeutic arsenal; and, we strongly believe that this modality will continue to deliver meaningful new therapies for patients. Cepturs use of synthetic oligonucleotides that engage U1 snRNP offers the ability to co-opt this master regulator of the transcriptome to regulate mRNA in a highly targeted fashion. We are thrilled to support Cepturs next phase of growth as they apply this disruptive approach for novel therapeutic applications.

With this financing, Aaron Royston, M.D., M.B.A., and Colin Walsh, Ph.D., join Cepturs Board of Directors.

Daniel Heller, M.S., M.B.A., General Partner and Chief Investment Officer at Affinity Asset Advisors, continued, In leading the Series Seed round, we identified early the potential of U1 Adaptor technology. Over the past year, we have worked closely with Peter and the Ceptur team and are delighted at the progress that has been made towards establishing the platform. In this financing round, we have significantly expanded upon our initial commitment and are inspired to partner with our new investor syndicate to advance U1 Adaptors for unmet patient needs.

To realize the revolutionary potential of the U1 Adaptor technology, several new members join Samuel Gunderson, Ph.D., co-founder of Ceptur, Professor of Molecular Biology at Rutgers University, and a leading expert on U1 snRNP biology, on Cepturs Scientific Advisory Board:

Thomas Andresen, Ph.D.Dr. Thomas L. Andresen is the CEO of T-Cypher Bio and the former CSO of Torque Therapeutics, now Repertoire Immune Medicines. While at Torque, he led the companys cellular immunotherapy programs from early-stage discovery to CMC scaling and through to clinical development. Dr. Andresen is a serial entrepreneur, having founded several US and EU life-science companies that further include Nanovi A/S and Monta Biosciences. His company creation track record spans early discovery to commercial and maps across multiple immunotherapy approaches for oncology. Dr. Andresen sits on several boards/advisory boards, including for Tidal Therapeutics (acquired by Sanofi), Monta Biosciences, and Nanovi; in academia, hes further founded the Institute of Health Technology at the Technical University of Denmark, where he maintains a professorial position. Dr. Andresen has co-authored over >200 research articles, has been listed as an inventor on >45 patent applications, and has received multiple research prizes, including the Elite Research Price from the Danish Ministry of Science.

Dennis Benjamin, Ph.D.Dr. Dennis Benjamin is the former SVP of Research at Seagen where he was a key developer of the companys ADC technology and clinical pipeline. Prior, he worked at Praecis Pharmaceuticals and Genetics Institute, advancing DNA encoded libraries and working in protein and small molecule discovery. Over his career, he has led teams that have discovered 25 biologics and small molecules that entered clinical trials and has contributed to 4 drug approvals. He is currently an advisor and SAB member at several start-up biotechnology companies.

Steven Dowdy, Ph.D.Dr. Steven F. Dowdy is a Professor of Cellular & Molecular Medicine at the UCSD School of Medicine and a cancer biologist, specializing in the development and delivery of RNA therapeutics as well as in G1 cell cycle control in cancer. The Dowdy lab is focused on the molecular details of delivery of RNA therapeutics across the endosomal lipid bilayer as well as the synthesis of endosomal escape domains to overcome this rate-limiting and billion year-old delivery challenge; its members were the first to synthesize bioreversible, charge neutralizing phosphotriester backbone RNAi prodrug triggers that increase metabolic stability, that augment pharmacokinetics and that enhance endosomal escape. Dr. Dowdy currently serves on five Science Advisory Boards for biotech companies and is an elected member of the Oligonucleotide Therapeutics Society (OTS) Board of Directors.

Sridhar Ganesan, M.D., Ph.D.Dr. Shridar Ganesan is the Associate Director for Translational Science, Chief of the Section of Molecular Oncology, and the co-Leader of the Clinical Investigations and Precision Therapeutics Program at the Rutgers Cancer Institute of New Jersey; he is also the Omar Boraie Chair in Genomic Science and Professor of Medicine at the Rutgers Robert Wood Johnson Medical School. Dr. Ganesan is a medical oncologist with clinical expertise in triple-negative breast cancer, hereditary breast cancer and rare cancer. His research interests include the characterization of DNA repair abnormalities in cancer with a focus on the BRCA1 tumor suppressor gene, the multi-modal molecular characterizations of different cancers, and the identification of biomarkers of response and resistance in early phase clinical trials. He has authored or co-authored over 120 publications, serves on multiple national and international grant review committees and is an Associate Editor of JCO-Precision Oncology.

Adrian Krainer, Ph.D.Dr. Adrian Krainer is the St Giles Professor at Cold Spring Harbor Laboratory (CSHL) and Deputy Director of Research of the CSHL Cancer Center. A world-renowned biochemist recognized for his basic work on RNA splicing and the development of its mechanism-based therapeutic applications, his seminal work with antisense oligonucleotides in mouse models of spinal muscular atrophy led to the development of nusinersen (Spinraza), which is the first FDA-approved drug for this disease; he is also a co-founder and a member of the Board of Directors at Stoke Therapeutics (NASDAQ: STOK). Dr. Krainer is the recipient of the Life Sciences Breakthrough Prize, the RNA Societys Lifetime Achievement Award, the Reemtsma Foundation International Prize in Translational Neuroscience, the Speiser Award in Pharmaceutical Sciences, and the Ross Prize in Molecular Medicine, amongst others. He previously served as the President of the RNA Society and is a member of the National Academy of Sciences, the National Academy of Medicine, the National Academy of Inventors, and the American Academy of Arts & Sciences.

Iain Mattaj, Ph.D.Dr. Iain Mattaj is the founding Director of Fondazione Human Technopole in Milan, Italy. Dr. Mattaj has made seminal contributions to the fields of transcription, RNA metabolism, nucleocytoplasmic transport and cell division. His prominent standings in these fields are underlined by his election as the past President of the RNA Society, Fellow of the Royal Society (London), Fellow of the Royal Society of Edinburgh, elected Member of the German Academy of Sciences Leopoldina, Member of Academia Europea, Foreign Honorary Member of the American Academy of Arts and Science, Fellow of the Academy of Medical Sciences (London) and Foreign Associate of the National Academy of Sciences (US). Dr. Mattaj was previously awarded the prestigious Louis-Jeantet Prize for Medicine in 2001. He is further a member of the European Molecular Biology Organization (EMBO) and helped to make The EMBO Journal a highly successful international publication, acting as Executive Editor from 1990 to 2004. Prior to his current appointment, Dr. Mattaj was a member of EMBL Heidelberg, Germany, serving as Group Leader (1985-1990), Coordinator of the Gene Expression Unit (1990-1999), and, subsequently, as the institutes Scientific Director (1999-2005) and Director General (2005-2018).

Henrik Oerum, Ph.D.Dr. Henrik Oerum the co-founder and CSO of Civi BioPharma and has previously founded 3 other oligonucleotide companies. Dr. Oerum has over 25 years of experience in the development and commercialization of oligonucleotide therapeutics, has authored >70 peer reviewed publications, and has been listed as an inventor on numerous patents in the field. In 1993, he founded PNA Diagnostics A/S, where he was also the CSO until 1999. During his tenure at PNA, the company was sold to Boehringer Mannheim (1994) and later to Hoffman-La Roche (1997). In 1996, Dr. Oerum cofounded Exiqon A/S, a nucleic acid diagnostics company that was floated on the Copenhagen Stock Exchange in 2007 (CSE:EXQ). In 2000, he co-founded the LNA-oligotherapeutics company Santaris Pharma A/S, where he served as CSO and VP of Business Development until its acquisition by Roche in 2014. Thereafter, he worked for Roche Pharma as Global Head of RNA therapeutics until March 2016, where he left to pursue new opportunities, leading to his founding of CiVi.

Thomas Tuschl, Ph.D.Dr. Thomas Tuschl is a Professor of RNA Molecular Biology at Rockefeller University. Dr. Tuschl is world renown for his research on the regulation of RNA and has discovered small interfering RNAs (siRNAs), microRNAs (miRNAs) and piwi-interacting RNAs (piRNAs). He is a member of the German National Academy of Sciences and the recipient of numerous awards, including the NIH Directors Transformative Research Project Award, the Ernst Jung Prize, the Max Delbrck Medal, the Molecular Bioanalytics Prize, the Meyenburg Prize, the Wiley Prize and the AAAS Newcomb Cleveland Prize. He is also the co-founder and a former Director of Alnylam Pharmaceuticals (NASDAQ: ALNY).

About Ceptur Therapeutics, Inc.Headquartered in Hillsborough New Jersey, Ceptur Therapeutics is a pre-clinical stage biotechnology company focused on developing targeted oligonucleotide therapeutics based on U1 Adaptor technology. For more information about Ceptur Therapeutics, please visit http://www.cepturtx.com or follow http://www.linkedin.com/company/ceptur-therapeutics/ on Linkedin.

Read the rest here:

Ceptur Therapeutics Launches with $75M Series A Financing to Advance RNA Therapeutics Based on Proprietary U1 Adaptor Technology - Business Wire

EdiGene Enters Strategic R&D Collaboration with Haihe Laboratory of Cell Ecosystem to Develop Hematopoietic Stem Cell Regenerative Therapies and…

BEIJING & CAMBRIDGE, Mass., January 24, 2022--(BUSINESS WIRE)--EdiGene, Inc., a global biotechnology company focused on translating gene-editing technologies into transformative therapies for patients with serious genetic diseases and cancer, announced a research and development collaboration with Haihe Laboratory of Cell Ecosystem to develop hematopoietic stem cell regenerative therapies and platform technology by combining resources and expertise from both sides.

The Haihe Laboratory of Cell Ecosystem, run by the Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, is focused on conducting fundamental research, innovation, and translation in the cell ecosystem.

Under the agreement, both parties will jointly develop hematopoietic stem cell regenerative therapies, including the development of innovative genetically-modified hematopoietic stem cell therapies and the exploration of novel biomarkers to optimize quality control for stem cell production.

"With top-notch resources and industry-university-research cooperation, well facilitate the development of cell-based medicine and therapies," said Professor Tao Cheng, Deputy Director of Haihe Laboratory of Cell Ecosystem and President of the Institute of Hematology and Blood Diseases Hospital at the Chinese Academy of Medical Sciences and Peking Union Medical College, a leading hematology researcher who has made a series of discoveries relating to the regulatory and regenerative mechanisms of hematopoietic stem cells. "Hematopoietic stem cells (HSCs) have the potential for long-term self-renewal and can differentiate into various types of mature blood cells. These stem cells can be harnessed to provide treatment for a broad range of diseases such as hematological tumors, autoimmune diseases, and hereditary blood disorders. We believe that this collaboration with EdiGene will accelerate the innovation and translation in the field of HSCs, thus enabling healthier patients with new therapies."

Story continues

Professor Cheng was awarded the second prize of the National Natural Science Award 2020 as the first author of work on basic and translational research that advanced the development of adult hematopoietic stem cells for therapeutic applications.

EdiGene is scaling up clinical translation and development of the first gene-editing hematopoietic stem cell therapy in China following the 2021 approval by the China National Medical Products Administration its IND for its investigational therapy ET-01. "Our team has extensive experience in the development and translation of cutting-edge technologies including hematopoietic stem cell and gene editing," said Dong Wei, Ph.D., CEO of EdiGene. "This collaboration with Haihe Laboratory of Cell Ecosystem will further our exploration in the field of hematopoietic stem cells. The partnership with this leading academic institute and our translational know-how enable us to move forward in bringing more innovative treatment options to patients in China and around the world."

In 2021, EdiGene initiated a Phase I multicenter clinical trial of ET-01, its gene-editing hematopoietic stem cell therapy for transfusion-dependent -thalassemia. EdiGene has enrolled the first patient at the Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College. Currently, the clinical trial is being conducted in Tianjin and Guangdong-Hong Kong-Macao Greater Bay Area (Greater Bay Area). EdiGene also presented its latest research on new surface markers and migration of hematopoietic stem cells at the 63rd Annual Meeting of the American Society of Hematology (ASH) in 2021.

About Haihe Laboratory of Cell Ecosystem

The Haihe Laboratory of Cell Ecosystem ("the Laboratory"), run by the Institute of Hematology and Blood Diseases Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, is one of the five registered Haihe Laboratories approved by Tianjin Municipal People's Government. With the goal of promoting population health with cell ecosystem, the Laboratory adheres to developing technological frontier, enhancing peoples health, and promoting research, innovation, and development of cell ecosystem in five key areas: cellular ecosystem, cellular ecology and immunity, cellular ecological imbalance and major diseases, cellular ecological reconstruction and frontier technology of cellular ecological research.

About Institute of Hematology and Blood Diseases Hospital (IH), Chinese Academy of Medical Sciences & Peking Union Medical College (CAMS/PUMC)

Founded in 1957, IH is a tertiary specialty hospital under the National Health Commission of China and is the supporting unit of the National Clinical Research Center of Hematologic Diseases and the State Key Laboratory of Experimental Hematology. It is also the main founding unit of Tianjin Base, the core base of the Chinese medical science and technology innovation system with the goal of becoming "the innovation hub of hematology in China." IH mainly engages in basic research, applied research, clinical diagnosis and treatment of hematological diseases, standard-setting, new technology research, new drug evaluation, and translation in hematology and related fields. IH is leading in the diagnosis and treatment of hematological diseases in China and a global scale and has made original achievements. Since 2010, IH has been awarded first place in the Hospital Specialty Reputation Ranking (Hematology) for 12 consecutive years. It has won first place in the Hematology Specialty Ranking for ten consecutive years since 2010 and ranked the first in hematology by the Scientific and Technological Evaluation Metrics (STEM) for Chinese hospitals for eight consecutive years since 2014.

About EdiGene, Inc

EdiGene is a global, clinical-stage biotechnology company focused on translating gene editing technologies into transformative therapies for patients with serious genetic diseases and cancer. The company has established its proprietary ex vivo genome-editing platforms for hematopoietic stem cells and T cells, in vivo therapeutic platform based on RNA base editing, and high-throughput genome-editing screening to discover novel targeted therapies. Founded in 2015, EdiGene is headquartered in Beijing, with offices in Guangzhou and Shanghai, China and Cambridge, Massachusetts, USA. More information can be found at http://www.EdiGene.com.

View source version on businesswire.com: https://www.businesswire.com/news/home/20220124005428/en/

Contacts

Xiaomeng ZhangEdiGene, Inc.+86 10-80733899media@edigene.com

See more here:

EdiGene Enters Strategic R&D Collaboration with Haihe Laboratory of Cell Ecosystem to Develop Hematopoietic Stem Cell Regenerative Therapies and...

Roche revives a closely watched Huntington’s disease drug – BioPharma Dive

Dive Brief:

As 2021 began, Huntington's drug research appeared to be on the verge of turning a corner.

Both Roche and Wave Life Sciences were advancing drugs that were similarly designed to block production of a protein implicated in disease progression. For Roche, the stakes were particularly high: A successful result could've led to an approval application for the first drug that might slow the march of the deadly, inherited disease.

By the end of the first quarter, however, both companies reported negative data, raising questions about their drugs as well as researchers' understanding of Huntington's. It also dialed up pressure on companies with earlier stage projects, such as UniQure and Passage Bio.

But it turns out Roche isn't done with tominersen after all, a decision that could have implications for others. While the new clinical trial will be a small, Phase 2 test that will require follow-up studies, tominersen remains one of the most advanced disease-modifying drug in clinical development for Huntington's.

The Swiss drugmaker is in "the early stages" of designing the new trial, Ionis said, which will evaluate different doses of tominersen in younger patients with less severe disease. Roche will share the design of the trial with Huntington's disease specialists in medical meetings later this year, according to Ionis.

Ahead of the trial design presentations, Roche later this week will begin a series of webinars to discuss with Huntington's specialists the after-the-fact analysis of GENERATION-HD1 that hinted at a benefit for the younger, less severe patients.

Roche's Huntington's collaboration with Ionis dates back to 2013, when the big drugmaker acquired initial licensing rights for $30 million and promised up to $362 million in future payouts. Following positive signs in early testing, Roche paid $45 million to license tominersen and cover clinical development as well as commercial costs.

As a disease caused by an identifiable genetic defect, Huntington's seems to be a good target for a medicine that can block the mutation, which results in a flawed version of a protein called huntingtin. Tominersen is a type of medicine known as antisense oligonucleotide and works by going after the RNA that encodes for the protein.

Gene therapies are also aimed at the disease's genetic cause, but work by replacing the defective gene with what could in theory be a one-time treatment. Tominersen, by comparison, was given to patients once every eight or 16 weeks in GENERATION-HD1.

An estimated 41,000 people in the U.S. have Huntington's disease, although many are undiagnosed.

Read the original here:

Roche revives a closely watched Huntington's disease drug - BioPharma Dive

WashU part of $65 million NIH study of schizophrenia in young people Washington University School of Medicine in St. Louis – Washington University…

Visit the News Hub

Teens, young adults needed for study aimed at improving early diagnosis

Daniel Mamah, MD, of Washington University School of Medicine in St. Louis, has received a grant as part of an international study focused on young people at high risk of schizophrenia. Mamah and his colleagues plan to characterize symptoms that can help diagnose schizophrenia early, as well as identify biomarkers in the blood and brain that may help predict risk.

Washington University School of Medicine in St. Louis is part of a major international study aimed at identifying causes and effects of the early stages of schizophrenia in young people, with the goal of improving early diagnosis and treatment.

The mental illness is characterized by alterations in thoughts, feelings and behaviors, which may include psychosis, a loss of contact with reality.

By studying young people at high risk of schizophrenia, the researchers plan to characterize the variety of symptoms that can help diagnose schizophrenia early, as well as identify biomarkers in the blood and brain that may help predict risk. Such information could help identify drug targets that have potential for treating schizophrenia early or even preventing problems, such as disordered thinking, that are associated with the disease.

For the study, the researchers are seeking adolescent and young adult volunteers, ages 12 to 30, who have experienced symptoms of psychosis such as hallucinations, delusions or intrusive disturbing thoughts suggesting they may be at risk for developing schizophrenia.

About 100,000 young people in the United States experience a first episode of psychosis every year, and over 1 million children and adolescents experience other problems in perception, thinking, mood and social functioning that put them at risk for schizophrenia, said Daniel Mamah, MD, an associate professor of psychiatry and the lead investigator at the Washington University clinical site. Those problems tend to get worse over time, especially when untreated, so the goal here is to identify problems at the earliest possible stage.

The Psychosis-Risk Outcomes Network (ProNET) study is funded by a grant of more than $65 million from the National Institute of Mental Health of the National Institutes of Health (NIH). Overall, the study will recruit 1,040 young people at high risk of schizophrenia at 26 sites around the world. There are 18 U.S. sites, with other sites in Canada, the United Kingdom, Italy, Spain, Germany, China and South Korea. About 50 patients will be enrolled in the study at the Washington University site.

The grant is a component of an NIH public-private partnership designed to meet the urgent need for early therapeutic interventions for people at high risk of developing schizophrenia. The effort brings together the NIH, the Food and Drug Administration, and a number of nonprofit and private universities and other organizations, including Washington University.

The groups involved are working toward the shared mission of discovering promising biological markers to help identify those at high risk for schizophrenia as early as possible, track the progression of their symptoms and other outcomes, and identify targets for treatment.

Schizophrenia is one of the leading causes of disability worldwide, but it often goes undiagnosed until symptoms have become disruptive in a persons life. Treatment delays can be associated with long-term problems, such as alcohol and drug abuse, difficulty holding a job and homelessness.

Just being at high risk of schizophrenia is increasingly recognized as a public health problem that affects adolescents and young adults, Mamah said. To develop more effective therapies, we must characterize the substantial variations of symptoms among those at risk, as well as untangle the roots of those differences.

Many experts think that by detecting schizophrenia earlier and starting treatment sooner, it may be possible to relieve, postpone or even prevent some of the long-term difficulties associated with the disorder.

Often, when doctors first see young patients who may have experienced a psychotic episode, its not possible to know whether they are on the path to more serious problems, partly because the early symptoms of schizophrenia can vary so much between individuals, Mamah said. By studying brain structure and function, psychopathology and cognition, genetics, behavior and other factors, this project is designed to identify patients at high risk so that when we have available treatments, they can begin receiving those treatments more quickly.

Mamah is director of the Washington Early Recognition Center, a Washington University clinic that treats young people experiencing the earliest signs of mental illness involving psychosis and those in early stages of psychotic disorders, such as schizophrenia and some forms of bipolar disorder.

The study is enrolling young people who may be at risk for schizophrenia after experiencing an episode of psychosis or other symptoms for example, a young person who previously was outgoing but suddenly becomes more introverted and withdrawn, who stops doing as well in school as in the past, or who begins to behave in a suspicious or paranoid manner or seems to respond to voices that no one else hears.

Young people experiencing such symptoms can be referred by doctors, parents or teachers who worry that the adolescent or young adult may be developing problems. Those under 18 must have a parents or guardians consent to participate in the study. Young men and women over age 18 who think they may qualify can refer themselves to the study. A short screening on the groups website gives young people the option to provide information if they might be interested in clinical services or in participating in the study.

Researchers will follow study volunteers for two years, assessing genetic and clinical biomarkers that may be linked to hallucinations or intrusive thoughts. The researchers also will conduct imaging studies of brain structure and function and collect blood and saliva samples. In addition, subjects will be assessed for psychopathology, language, speech and cognitive ability.

For more information about the study, call Carli Ryan at 314-362-5216, e-mail carli.ryan@wustl.edu, or visit the clinics website.

This work is supported by the National Institute of Mental Health of the National Institutes of Health (NIH). Grant number U01 MH124639.

Washington University School of Medicines 1,700 faculty physicians also are the medical staff of Barnes-Jewish and St. Louis Childrens hospitals. The School of Medicine is a leader in medical research, teaching and patient care, and is among the top recipients of research funding from the National Institutes of Health (NIH). Through its affiliations with Barnes-Jewish and St. Louis Childrens hospitals, the School of Medicine is linked to BJC HealthCare.

Read more from the original source:

WashU part of $65 million NIH study of schizophrenia in young people Washington University School of Medicine in St. Louis - Washington University...