Copy number variations linked to autism have diverse but overlapping effects – Spectrum

Mapping outcomes: Some genetic mutations can lead to a wide variety of traits, including those associated with autism.

People with mutations in distant chromosomal regions often share a range of autism traits, even if they do not meet the diagnostic threshold for autism, according to a new study.

Mutations called copy number variations (CNVs) involve duplications or deletions of large stretches of DNA. Having a CNV in the 16p11.2 or 22q11.2 chromosomal region increases a persons likelihood of being diagnosed with autism, but previous studies have found significant variability in the traits associated with mutations in either location.

The new work shows that deletions or duplications in 16p11.2 or 22q11.2 track with distinct profiles of cognitive abilities and autism traits, and that each type of variant is linked to a different probability of being diagnosed with autism.

These profiles overlap, which suggests that the different CNVs have similar impacts on developmental pathways involved with autism, says lead investigator Marianne van den Bree, professor of psychological medicine at Cardiff University in the United Kingdom. The findings also support the idea that other factors such as the environment or other genes shape a persons autism traits.

Van den Bree and her colleagues across eight institutions pooled data from 547 people with a deletion or duplication in 16p11.2 or 22q11.2. They compared the data with similar information from the Autism Genome Project, looking at 2,027 autistic people who do not have these CNVs.

Pulling these datasets together provided an in-depth look at patterns of outcomes. The four groups of people with CNVs a deletion or duplication in either chromosomal region differ the most in motor function, van den Bree and her colleagues found. And people with 22q11.2 deletions are less likely to have an autism diagnosis than those with any of the other CNVs, but they still have a higher autism prevalence than the general population.

People with a duplication in 22q11.2 or 16p11.2 tend to have more severe autism traits than people with deletions, the researchers found. And people with a 16p11.2 duplication or 22q11.2 deletion have greater cognitive impairment than those with one of the other two variants do.

Despite these differences between groups, people within each group show even greater variability, the team found, which suggests that other factors contribute to a persons traits. The work appeared in January in the American Journal of Psychiatry.

These four CNVs have not previously been compared in this way, but the study feels more confirmatory than it feels like its carving out something new, says Elliott Sherr, professor of neurology at University of California, San Francisco, who was not involved in the new work.

Many people, however, including some clinicians, are unaware that these genetic conditions are often linked to autism, says study investigator Samuel Chawner, research fellow in psychology at Cardiff University. He says he hopes that the profiles he and his colleagues identified will inform how genetic conditions are treated. For instance, 54 percent of people carrying one of the CNVs who do not have an autism diagnosis still have significant autism-like difficulties.

Whats missing from the new work is an examination of what else besides the CNVs contributes to the diversity of traits seen in people with these mutations, such as environment and other genes, says David Ledbetter, chief clinical officer at Dascena, a personalized medicine company. Ledbetter was not involved in the study.

For example, people with a 22q11.2 deletion have an increased likelihood of having schizophrenia, but information from the rest of their genome can help to accurately forecast outcomes, according to a study published in November. This same technique could be used to predict traits in people with the other CNVs, Ledbetter says.

A persons environment including their ability to access medical support and early education may also play a role in this variability, Chawner says. Van den Bree, Chawner and their colleagues at the Genes to Mental Health consortium plan to study how these factors in particular contribute to traits in people with CNVs.

See the rest here:

Copy number variations linked to autism have diverse but overlapping effects - Spectrum

Are Gene Therapies the Medicine of the Future? – BioSpace

Over the next 10 years, gene therapies are expected come into their own as a treatment option for a variety of diseases. So far, two such therapies have snagged regulatory approval, Novartis Zolgensma for spinal muscular atrophy, and Sparks Luxturna for a rare form of genetic blindness. More are waiting their turn.

Multiple companies are delving into gene therapy research with hopes of developing a one-time treatment for devastating genetic diseases. Gene therapies offer great reward in the form of treating various devastating diseases, but there are also significant risks. Over the past year, several clinical studies have been halted or scrapped due to safety concerns.

Bay Area-based Audentes Therapeutics had a temporary hold placed on the gene therapy under development for X-linked myotubular myopathy following reports of several patient deaths. That hold has since been lifted by the U.S. Food and Drug Administration. Uniqure also saw a hold placed on its hemophilia B trial after a patient in the study developed liver cancer. The hold was placed weeks after the company announced promising Phase III results at a conference in December.

Despite those risks, hundreds of millions of dollars in research dollars are being invested in gene therapies because of the potential near-curative capabilities the technology could offer. In December, life sciences giant Bayer launched a cell and gene therapy platform within its pharmaceutical division in order to become a leading company within a rapidly emerging and evolving field that offers the potential of life-saving therapies. Eli Lilly also dove into the field in December with the acquisition of Prevail Therapeutics. That deal was expected to extend Eli Lillys research efforts through the creation of a gene therapy program that will be anchored by Prevail's portfolio of clinical-stage and preclinical neuroscience assets.

This week, German scientists reported they were able to use gene therapy to help paralyzed mice run again. The researchers were able to genetically engineer a unique protein dubbed hyper-interleukin-6, which was then able to stimulate the regeneration of nerve cells in the visual system. A few weeks after the treatment, the injured animals were able to walk again.

Scientists in China announced the development of a gene therapy that could potentially reverse the effects of ageing. Initial research was conducted with mice, but if it is proven to be safe, human testing could begin. As Reuters reported, the method involved inactivating a gene called kat7 which the scientists found to be a key contributor to cellular ageing. Researchers used CRISPR/Cas9 to screen thousands of genes for those which were particularly strong drivers of cellular senescence, the term used to describe cellular ageing, Reuters said.

Earlier this month, a public-private partnership in Boston formed to open a new facility to boost advances in cell and gene therapies. This creation of this new facility is being helmed by Harvard University and the Massachusetts Institute of Technology. Those prestigious universities are partnering with industry members such as Fujifilm Diosynth Biotechnologies, Cytivia and Alexandria Real Estate Equities, as well as multiple research hospitals. Part of the goal of this new institute, which is still unnamed at this point, is to boost the supply of materials for research and early clinical studies, provide space for some research and also offer training in equipment used for gene therapies, The Harvard Gazette reported this week.

On Monday, Curadigm, a subsidiary of France-based Nanobiotix, forged a collaboration with Sanofi to assess if that companys Nanoprimer technology is a promising option to significantly improve gene therapy development. The goal of the project is to establish proof-of-concept for the Nanoprimer as a combination product that could improve treatment outcomes for gene therapy product candidates.

Many promising nucleic acid-based therapeutics administered intravenously are limited in their efficacy due to rapid clearance in the liver, which prevents these therapies from reaching the necessary accumulation in target tissues to generate their intended outcomes. Additionally, accumulation in the liver, rather than in the target tissues, can lead to dose-limiting hepatic toxicity, Nanobiotix said in its announcement. The Nanoprimer is designed to precisely and temporarily occupy therapeutic clearance pathways in the liver. Delivered intravenously, immediately prior to the recommended therapy, the technology acts to prevent rapid clearancethereby increasing bioavailability and subsequent accumulation of therapeutics in the targeted tissues.

The Nanoprimer is a combination product candidate that does not alter or modify the therapies it is paired with, which means if the research with Sanofi is successful, Curadigm could seek out other opportunities for its technology.

Most Read Today

Go here to see the original:

Are Gene Therapies the Medicine of the Future? - BioSpace

Press Registration Is Now Open for the 2021 ACMG Annual Clinical Genetics Meeting – A Virtual Experience – PRNewswire

BETHESDA, Md., Jan. 27, 2021 /PRNewswire/ --The ACMG Annual Clinical Genetics Meeting will be a fully virtual meeting in 2021 and continues to provide groundbreaking research and the latest advances in medical genetics, genomics and personalized medicine. To be held April 1316, experience four days of professional growth, education, networking and collaboration with colleagues from around the world and discover what's shaping the future of genetics and genomics, including several sessions on COVID-19. The 2021 ACMG Meeting Virtual Experience is designed to offer a variety of engaging and interactive educational formats and types of sessionsfrom Scientific Sessions and Workshops to TED-Style Talks, Case-based Sessions, Platform Presentations and Short Courses. The 2021 ACMG Meeting Virtual Experience has something for everyone on the genetics healthcare team and will be available to participate in from the convenience of your home or office.

Interview those at the forefront in medical genetics and genomics, connect with new sources, and get story ideas on the clinical practice of genetics and genomics in healthcare today and for the future. Learn how genetics and genomics research is being integrated and applied into medical practice. Topics include COVID-19, gene editing, cancer genetics, molecular genomics, exome sequencing, pre- and perinatal genetics, diversity/equity and inclusion, biochemical/metabolic genetics, genetic counseling, health services and implementation, legal and ethical issues, therapeutics and more.

Credentialed media representatives on assignment are invited to cover the ACMG Annual Meeting A Virtual Experience on a complimentary basis. Contact Kathy Moran, MBA at [emailprotected]for the Press Registration Invitation Code, which will be needed to register at http://www.acmgmeeting.net.

Abstracts of presentations will be available online in February.

A few 2021 ACMG Annual Meeting highlights include:

Program Highlights:

Two Short Courses Available Starting on Tuesday, April 13:

Cutting-Edge Scientific Concurrent Sessions:

Social Media for the 2021 ACMG Meeting Virtual Experience: As the ACMG Annual Meeting approaches, journalists can stay up to date on new sessions and information by following the ACMG social media pages on Facebook,Twitterand Instagramand by usingthe hashtag #ACMGMtg21 for meeting-related tweets and posts.

The ACMG Annual Meeting website has extensive information at http://www.acmgmeeting.net and will be updated as new information becomes available.

About the American College of Medical Genetics and Genomics (ACMG) and the ACMG Foundation for Genetic and Genomic Medicine (ACMGF)

Founded in 1991, the American College of Medical Genetics and Genomics (ACMG) is the only nationally recognized medical society dedicated to improving health through the clinical practice of medical genetics and genomics and the only medical specialty society in the US that represents the full spectrum of medical genetics disciplines in a single organization. The ACMG is the largest membership organization specifically for medical geneticists, providing education, resources and a voice for more than 2,400 clinical and laboratory geneticists, genetic counselors and other healthcare professionals, nearly 80% of whom are board certified in the medical genetics specialties. ACMG's mission is to improve health through the clinical and laboratory practice of medical genetics as well as through advocacy, education and clinical research, and to guide the safe and effective integration of genetics and genomics into all of medicine and healthcare, resulting in improved personal and public health. Four overarching strategies guide ACMG's work: 1) to reinforce and expand ACMG's position as the leader and prominent authority in the field of medical genetics and genomics, including clinical research, while educating the medical community on the significant role that genetics and genomics will continue to play in understanding, preventing, treating and curing disease; 2) to secure and expand the professional workforce for medical genetics and genomics; 3) to advocate for the specialty; and 4) to provide best-in-class education to members and nonmembers. Genetics in Medicine, published monthly, is the official ACMG journal. ACMG's website (www.acmg.net) offers resources including policy statements, practice guidelines, educational programs and a 'Find a Genetic Service' tool. The educational and public health programs of the ACMG are dependent upon charitable gifts from corporations, foundations and individuals through the ACMG Foundation for Genetic and Genomic Medicine.

Kathy Moran, MBA[emailprotected]

SOURCE American College of Medical Genetics and Genomics

http://www.acmg.net

Originally posted here:

Press Registration Is Now Open for the 2021 ACMG Annual Clinical Genetics Meeting - A Virtual Experience - PRNewswire

SMART Study Finds 22q11.2 Microdeletion Prevalence Much Higher than Expected – PRNewswire

SAN CARLOS, Calif., Feb. 1, 2021 /PRNewswire/ --Natera, Inc. (NASDAQ: NTRA), a pioneer and global leader in cfDNA testing, presented key results from its SMART study at the SMFM 41st Annual Pregnancy Meeting.1 The SMART study sets a new standard as the largest prospective NIPT study to date(N = 20,927 enrolled from 21 medical centers), and the only large-scale study to collect genetic outcomes in most of the subjects. The study includes the validation of a new artificial intelligence-based algorithm for Panoramacalled Panorama AI, which utilizes information from over 2 million cfDNA tests performed by Natera.

Key results related to the 22q11.2 microdeletion:

"This is the first prospective NIPT study in which genetic outcomes were confirmed in the vast majority of the patients enrolled, and provides a wealth of data about the real-world performance of NIPT across a diverse group of global centers and patients," said Mary Norton, MD, Professor, UCSF, and one of the Principal Investigators of SMART. "The findings related to high prevalence of 22q11.2 deletion syndrome, the limited ability of ultrasound to detect all cases prenatally, and the performance of NIPT in detection of these cases with high accuracy provide exciting data to inform discussions around testing for a broader set of conditions beyond common aneuploidies."

"The diagnostic odyssey related to 22q11.2 deletion syndrome is well documented, with median time to diagnosis of almost 5 years.6And in the meantime, a window of opportunity might be lost to intervene and impact outcomes. Delivery of a child with 22q11.2 deletion syndrome should be at a tertiary facility well-equipped to deal with short-term complications that are associated with the disorder.7 Depending on the issue at hand (e.g., cardiac, endocrine), appropriate interventions are warranted. For example, timely administration of neonatal calcium has been shown to correlate with preventing the intellectual decline commonly seen in affected children,"8,9 said Pe'er Dar, MD, Albert Einstein College of Medicine, Bronx NY, and one of the Principal Investigators of SMART. "With the ability to detect more accurately in combination with a low false positive rate, I believe that the findings of the SMART study provide professional societies with sufficient evidence to consider including screening for 22q11.2 deletions in routine prenatal genetic screening."

In 2020, Natera performed over 400,000 tests for the 22q11.2 microdeletion. Natera has established a CPT code and favorable pricing for microdeletion testing. Based on high prevalence and excellent performance in the study, Natera looks forward to engaging professional societies for routine testing of pregnancies for the 22q11.2 microdeletion, and will then pursue broader insurance coverage.

About Panorama

Panoramareveals a baby's risk for severe genetic disorders as early as nine weeks into pregnancy. The test uses a unique single-nucleotide polymorphism (SNP)-based technology to analyze fetal/placental DNA obtained through a blood draw from the mother. It is the only commercially available test that differentiates between maternal and fetal DNA to assess the risk of aneuploidies. The test also screens twin pregnancies for zygosity and fetal sex of each baby, and identifies risk for more genetic conditions in twin pregnancies than any other NIPT. Panorama is one of several genetic screening tests from Natera designed to help families on the path to parenthood. Natera has published 23 papers, studying over 1.3 million patients, since the launch of Panorama the largest body of evidence in the space today. Panorama has been developed and its performance characteristics determined by Natera, the CLIA-certified laboratory performing the test. The test has not been cleared or approved by the US Food and Drug Administration (FDA). CAP accredited, ISO 13485 certified, and CLIA certified.

About Natera

Naterais a pioneer and global leader in cell-free DNA testing from a simple blood draw. The mission of the company is to change the management of disease worldwide with a focus on women's health, oncology, and organ health. Natera operates ISO 13485-certified and CAP-accredited laboratories certified under the Clinical Laboratory Improvement Amendments (CLIA) in San Carlos, California and Austin, Texas. It offers proprietary genetic testing services to inform obstetricians, transplant physicians, oncologists, and cancer researchers, including biopharmaceutical companies, and genetic laboratories through its cloud-based software platform. For more information, visitnatera.com. Follow Natera onLinkedIn.

Forward-Looking Statements

All statements other than statements of historical facts contained in this press release are forward-looking statements and are not a representation that Natera's plans, estimates, or expectations will be achieved. These forward-looking statements represent Natera's expectations as of the date of this press release, and Natera disclaims any obligation to update the forward-looking statements. These forward-looking statements are subject to known and unknown risks and uncertainties that may cause actual results to differ materially, including with respect to our efforts to develop and commercialize new product offerings, our ability to successfully increase demand for and grow revenues for our product offerings, whether the results of clinical or other studies will support the use of our product offerings, our expectations of the reliability, accuracy and performance of our tests, or of the benefits of our tests and product offerings to patients, providers and payers. Additional risks and uncertainties are discussed in greater detail in "Risk Factors" in Natera's recent filings on Forms 10-K and 10-Q and in other filings Natera makes with the SEC from time to time. These documents are available atwww.natera.com/investorsandwww.sec.gov.

Contacts

Investor Relations: Mike Brophy, CFO, Natera, Inc., 510-826-2350

Media: Paul Greenland, VP of Corporate Marketing, Natera, Inc., [emailprotected]

References

SOURCE Natera, Inc.

Transforming Management of Genetic Disease

Go here to see the original:

SMART Study Finds 22q11.2 Microdeletion Prevalence Much Higher than Expected - PRNewswire

4 New Life Sciences Licensing Deals and Investments to Watch – BioSpace

Today marked some wheeling and dealing in the life sciences industry as several companies licensed products or invested in other companies. Heres a look.

Eli Lillyand Asahi Kasei Pharma Eli Lilly and Company inked a license agreement with Tokyos Asahi Kasei Pharma Corporation. In it, Lilly picks up exclusive rights to AK1780 from Asahi. The drug is an oral P2X7 receptor antagonist that recently finished a Phase I dosing study. P2X7 receptors are associated with neuroinflammation that drives chronic pain conditions.

Under the terms of the deal, Lilly will handle future global development and regulatory activities. Lilly is paying Asahi Kasei Pharma $20 million up front and the Japanese company is eligible for up to $210 million in development and regulatory milestones. Asahi Kasei will retain the rights to promote the drug in Japan and China, including Hong Kong and Macau. If it makes it to market, Asahi Kesei will also be eligible for up to $180 million in sales milestones and tiered royalties from the mid-single to low-double digits.

Lilly is committed to developing novel medicines that may provide relief for patients suffering with various pain conditions, said Mark Mintun, vice president of pain and neurodegeneration research at Lilly. We are pleased to license this molecule from Asahi Kasei Pharma, and look forward to developing it further as a potential treatment for neuroinflammatory pain conditions.

Artiva Biotherapeutics and Merck San Diego-based Artiva Biotherapeutics announced an exclusive global collaboration and license agreement with Merck to develop novel chimeric antigen receptor (CAR)-NK cell therapies against solid tumor-associated antigens. They will leverage Artivas off-the-shelf allogeneic NK cell manufacturing platform and its proprietary CAR-NK technology. At first, the collaboration will include two CAR-NK programs with an option for a third. None of them are currently part of Artivas current or planned pipeline. Artiva will develop the programs through the first GMP manufacturing campaign and to preparation for the Investigational New Drug (IND) application, where Merck will take over clinical and commercial development.

Merck is paying Artiva $30 million upfront for the first two programs and another $15 million if Merck chooses to go ahead with the third. Artiva will be up for development and commercial milestones up to $612 million per program and royalties on global sales. Merck also is ponying up research funding for each program.

Our NK platform has been developed to be truly off-the-shelf and we believe it will be further validated by this exclusive collaboration with Merck, as we work together to bring cell therapies to all patients who may benefit, said Peter Flynn, chief operating officer of Artiva.

NeuBase Therapeutics and Vera Therapeutics Pittsburgh-based NeuBase Therapeutics announced a binding agreement to acquire infrastructure, programs and intellectual property for several peptide-nucleic acid (PNA) scaffolds from Vera Therapeutics, formerly called TruCode Gene Repair. Vera is based in South San Francisco. On January 19, Vera announced its launch with a $80 million Series C financing led by Abingworth LLP and joined by Sofinnova Investments, Longitude Capital, Fidelity Management & Research Company, Surveyor Capital, Octagon Capital, Kliner Perkins, GV and Alexandria Venture Investments. Veras lead clinical candidate is atacicept, a novel B cell and plasma cell inhibitor being developed for patients with IgA nephropathy (IgAN).

The technology acquired by NeuBase has shown the ability to resolve disease in genetic models of several disease indications. NeuBase is focused on genetic medicine.

With this acquisition, we enhance our PATrOL platform, furthering our unique ability to directly engage and correct malfunctioning genes with exquisite precision to address the root causes of a wide variety of human diseases, said Dietrich A. Stephan, chief executive officer of NeuBase. These assets extend and refine our PATrOL platforms capabilities and accelerates, through our Company, to bring the rapidly growing genetic medicines industry toward a single high-impact focal point. We are committed to advancing our pipeline and candidates to the clinic and to exploiting the full potential of PNA technology to continue creating value for our shareholders and importantly, for patients.

Bio-Techne Corporation and Changzhou Eminence Biotechnology Co Minneapolis-based Bio-Techne Corporation announced an initial minority strategic equity investment in Chinas Changzhou Eminence Biotechnology Co. Eminence plans to use the financing to expand its manufacturing capacity and increase the service capabilities of its China-based GMP media production facility. Eminence, based in Changzhou City, Jiangsu, China, launched in 2016 and initially focused on manufacturing and selling best-in-class media to life science companies, including Chinese Hamster Ovary (CHO) cells and other serum-free media products and services. The company is currently finishing and scaling its GMP production facility, which it plans to complete by the end of this year.

With our protein analysis instruments and expanding GMP protein capabilities, Bio-Techne continues to expand its offering of products and tools critical for bioprocessing, said Chuck Kumeth, president and chief executive officer of Bio-Techne. Investing in Eminence not only gives Bio-Techne a foothold in providing additional products and services to support the critical needs of the rapidly growing Chinese biopharmaceutical industry, but also fits extremely well with our existing high-growth product portfolio in China. We look forward to working with the Eminence team.

Most Read Today

See original here:

4 New Life Sciences Licensing Deals and Investments to Watch - BioSpace

CRISPR Mutants – The Dawn of CRISPR Mutants – SAPIENS – SAPIENS

The Mutant Project: Inside the Global Race to Genetically Modify Humansby Eben Kirksey. St. Martins Press, November 2020. Excerpt previously published by Black Inc.

Surreal artwork in the hotel lobbya gorilla peeking out of a peeled orange, smoking a cigarette; an astronaut riding a cyborg giraffewas the backdrop for bombshell news rocking the world. In November 2018, Hong Kongs Le Mridien Cyberport hotel became the epicenter of controversy about Jiankui He, a Chinese researcher who was staying there when a journalist revealed he had created the worlds first edited babies. Select experts were gathering in the hotel for the Second International Summit on Human Genome Editinga meeting that had been called to deliberate about the future of the human species. As CNN called the experiment monstrous, as heated discussions took place in labs and living rooms around the globe, He sat uncomfortably on a couch in the lobby.

He was trying to explain himself to Jennifer Doudna, the chemist at UC Berkeley, who is one of the pioneers behind CRISPR, a new genetic-engineering tool. Doudna had predicted that CRISPR would be used to direct the evolution of our species,* writing, We possess the ability to edit not only the DNA of every living human but also the DNA of future generations. As He went through his laboratory protocol, describing how he had manipulated the genes of freshly fertilized human eggs with CRISPR, Doudna shook her head. She knew that this moment might be coming someday, but she imagined that it would be in the far future. Amid the bustle of hotel guests, science fiction began to settle into the realm of established fact.

St. Martins Publishing Group

I was checking in to Le Mridien as the story broke and first heard rumors about Hes babies while chatting in the elevator with other summit delegates. We had come to Hong Kong to discuss the science, ethics, and governance of CRISPR and an assortment of lesser-known tools for tinkering with DNA. Struggling to overcome intense jet lagfresh off planes from Europe, the United States, and other parts of Asiawe listened to speculation in the hotels hallways while swimming through reality, caught between waking and dreaming.

Opening the door to my hotel room, a luxury suite courtesy of the U.S. National Academy of Sciences, I hunted for reliable sources of information online. I had been invited to speak on the research ethics panel, after Jiankui He, so I needed to play catch-up, fast. I found YouTube videos posted by Hes lab just hours before, offering details of the experiment. Posing in front of his laboratory equipment, with a broad smile on his face, He announced to the world: Two beautiful little Chinese girls, named Lulu and Nana, came crying into this world as healthy as any other babies a few weeks ago. The experiment aimed to delete a single gene with CRISPR. This new technique of genetic surgery, He claimed, could produce children who were resistant to the HIV virus.

Hunched over the glowing screen of my laptop, I perused the opinions that were just starting to form. Chinese media pundits suggested that a Nobel Prize might be in the making, saying that He was following in the footsteps of scientists who produced the first controversial test-tube baby in 1978. A raucous debate was taking place on WeiboChinas prominent social media platformas 1.9 billion people viewed the hashtag # (#FirstGeneEditedHIVImmuneBabies). Some Chinese influencers were praising Jiankui He as a national scientific hero. Others condemned him, saying that it was shameful to treat children like guinea pigs. Journalists were starting to discover Dr. Hes ties to biotechnology companiesone reportedly worth US$312 millionand alleged that there were serious financial conflicts of interest.

Anyone who follows the news knows the basic story. Over the next few days, Jiankui He experienced a meteoric rise to fame, followed by a dramatic fall from grace. Eventually, he lost his university job and was thrown in jail. A district court in China sentenced him to three years in prison for practicing medicine without a license, denouncing his pursuit of personal fame and profit.

Dr. Hes story is a gateway into a much bigger enterprise: the tale of CRISPR and the emergence of genetic medicine. The gala was quietly abuzz with news of other efforts to genetically modify humans. Experiments were already underway in England, the United States, and many other labs in mainland China. As billionaires and Wall Street investors were getting in on the action, as scientists and doctors were making careers out of CRISPR, I wondered: Who counts as a visionary, and who becomes a pariah?

He spoke about his gene-editing experiment that led to the birth of twin girls while at a summit in Hong Kong in 2018. VOAIris Tong/Wikimedia Commons

He was not alone in the pursuit of fame and fortune. It seemed like none of the scientists at the gala were innocent of financial conflicts of interest. Collectively, these enterprising biologists had already raised hundreds of millionsfrom venture capitalists, big pharma companies, and the stock marketfor genetic engineering experiments in human patients. I overheard excited chatter about new investment opportunities. The first gene therapy, a cancer treatment, had recently been approved in the United Stateswith a US$475,000 price tag. While the scientists gushed about the CRISPR revolution, I was quietly thinking about how genetic medicine is producing other upheavals in society. Profit-driven ventures in research and medicine were producing a new era of dramatic medical inequality.

As market forces propelled CRISPR into the clinic, I set out to answer basic questions about science and justice: Who is gaining access to cutting-edge genetic medicine? Are there creative ways to democratize the field? Panning out, I also explored questions that could have profound implications for the future of our species: Should parents be allowed to choose the genetic makeup of their children? How much can we actually change about the human condition by tinkering with DNA?

As a cultural anthropologist, I have often found myself opposing biologists in debates about human nature. Ever since Margaret Mead wrote her 1928 classicComing of Age in Samoa, anthropologists have argued that a persons life is shaped by the social environment in which each is born and raised rather than genetic heredity alone.Anthropologists have recently joined other progressive thinkers to imagine how science has enabled new experimental possibilities for human beings.Now we are studying how the human social environment has been shaped by synthetic chemistry, smartphones, the internet, and biotechnology.

My goal has been to map how genetic engineering will transform humanity. Rather than limit my research to a single culture, I followed CRISPR around the globe. I tracked the impact of this gene-editing tool as it traveled from media reports to laboratories, through artificial intelligence algorithms, and into the cells of embryos and the bodies of living people. Using an anthropological lens, I examined new forms of power as scientists, corporate lobbyists, medical doctors, and biotechnology entrepreneurs worked to redesign life itself.

I will offer you a mosaic portrait. This is a story of people and concerns on either side of the dynamics of power that has emerged with CRISPR. I moved among the powerful in their native habitats: conferences, fancy hotels, restaurants, corporate offices, and cluttered labs. To understand how social inequality is changing in this brave new world, I also interviewed chronically ill patients, disabled scholars, and hackers. From the power centers to the margins, I went where I could find answers. Very old conflicts were playing out even as new technologies transformed science and medicine.

An exhibit on reproductive technologies at the China National GeneBank envisions a future where robots rear human embryos. Eben Kirksey

When I set out to meet some of the first genetically modified people, I found activists who were battling insurance agents and biotechnology companies for potentially lifesaving treatments. Nearly a decade before Dr. He stirred up controversy in China, a small group of HIV-positive gay men in the United States quietly participated in a clinical trial dubbed the first-in-man gene-editing experiment. Researchers aimed to delete a gene from these menthe same DNA sequence later targeted by Hein hopes of engineering resistance to the virus and repairing damage to their immune systems from AIDS. One veteran HIV activist who participated in this study, Matt Sharp, convinced me that having his DNA altered wasnt a big deal and that genetic engineering does indeed have real medical promise. Sharp also confirmed my suspicions: Biotech companies are putting profits ahead of human health as they search for lucrative applications of gene editing in the clinic.

Gene editing is not a particularly good metaphor for explaining the science of CRISPR. With a computer, I can easily cut and paste text from one application to another, or make clean deletionsletter by letter, line by line. But CRISPR does not have these precise editorial functions. CRISPR is more like a tiny Reaper drone that can produce targeted damage to DNA. Sometimes it makes a precision missile strike, destroying the target. It can also produce serious collateral damage, like a drone attack that accidentally takes out a wedding party instead of the intended target. Scientists often accidentally blast away big chunks of DNA as they try to improve the code of life. CRISPR can also go astray when the preprogrammed coordinates are ambiguous, like a rogue drone that automatically strikes the friends, neighbors, and relatives of suspected terrorists. CRISPR can persist in cells for weeks, bouncing around the chromosomes, producing damage to DNA over and over again every time it finds a near match to the intended target.

How much can we actually change about the human condition by tinkering with DNA?

It is important to signal a sense of risk or a need for caution in using CRISPR. Other metaphorslike genetic surgery or DNA hackinghave been proposed to replace the idea of editing. The idea of genetic surgery suggests that there can be a slip of the surgeons knife, creating an unintended injury. Each of these imagesthe targeted missile, the surgeons scalpel, the hackers codeoffers a perspective on how CRISPR works, even while concealing messy cellular dynamics. In the absence of a perfect metaphor, ultimately, I think that technical language describes it best: CRISPR is an enzyme that produces targeted mutagenesis.

In other words, CRISPR generates mutants.

Strictly speaking, we are all mutants. At a molecular level, each of us is unique. Each of us starts life with 4080 new mutations that were not found in our parents. From birth, each of us has around 20 inactive genes from loss-of-function mutations. During the course of a normal human life, we also accumulate mutations in our bodies, even in our brains. By the time we reach age 60, a single skin cell will contain between 4,000 and 40,000 mutations, according to a study in theProceedings of the National Academy of Sciences. These genetic changes are the result of mistakes made each time our DNA is copied during cell division or when cells are damaged by radiation, ultraviolet rays, or toxic chemicals. Generally, mutations arent good or bad, just different.

Mutants in popular culture play important roles in our high-tech myths. Some cartoons simply celebrate mutation as whimsical possibility. The pizza-eating Teenage Mutant Ninja Turtles are known for fighting crime in support of established law and order. Darker speculative fiction uses mutants to illustrate the hypocrisy and inhumanity of the scientific establishment. Violent experiments on children who were born with special abilities feature in recent Netflix series likeStranger Things. Horror flicks and video games featuring mindless zombies and flesh-eating mutants have a common theme: Science could create monsters that cannot be controlled.

Reporters who sounded the alarm about Lulu and Nanas birthcalling them freaky CRISPR Frankenbabiesclearly had not done their literary homework. Frankensteins monster is now popularly imagined as a dimwitted giant with electrodes in his neckfollowing imagery from the first black-and-white film, put out by Universal Pictures in 1931. The originalFrankenstein, Mary Shelleys gothic novel from 1818, described a superhuman creature that was driven by the desire to be loved. The highly intelligent, articulate, and high-minded creature only turned violent when he was shunned by human society. Amid the controversy about Dr. Hes experiment, a political theorist and literary scholar named Eileen Hunt Botting defended the rights of genetically modified children to live, love, and flourish. Flipping the mainstream script, she wrote an essay for TheWashington Postsuggesting that Frankenstein is an apt cautionary tale about the possibility of devastating discrimination against a bioengineered child.

Some media reports on Lulu and Nana, the first known gene-edited human babies, referenced the science-fiction character Frankenstein (shown here from the film by that name). Universal Pictures/Wikimedia Commons

During my international adventures in the world of CRISPR research, I kept science fiction classics close at hand. The rich archive of speculative fiction has helped me understand the perils and potential of experiments that are remaking the human species.

Scientists have identified some geneslike those associated with eye and skin colorthat would be relatively easy to manipulate. One Russian American gene-editing expert, Fyodor Urnov, intimated that it should be biologically possible to engineer soldiers or athletes with enhanced endurance, speed, and muscle mass. Genetic enhancements come with serious health risks, but military leaders have a long history of ignoring the health and well-being of their soldiers. Fertility clinics also have a bad track record as profit-driven enterprises, ready to sell couples expensive and scientifically unproven treatments. The New Hope Fertility Center in Manhattan is already advertising a new technique: Couples could soon have the opportunity to create designer babies with CRISPR.As scientists speculate about post-racial futures and nightmare military scenarios, as market forces bring new genetic technologies into the clinic at a dizzying speed, it is time to slow down and establish some clear rules for the road. Misguided attempts to improve the human species have already produced atrocitieslike the Nazi death camps that systematically eliminated homosexuals and Jews from the population. In the wrong hands, CRISPR could have devastating consequences for humanity.

This excerpt has been edited slightly for style and length.

* Clarification: This quote comes from A Crack in Creation: Gene Editing and the Unthinkable Power to Control Evolution, written by Jennifer Doudna and Samuel Sternberg.

Follow this link:

CRISPR Mutants - The Dawn of CRISPR Mutants - SAPIENS - SAPIENS

Genomes, Maps, And How They Affect You – IFLScience

What is a genome

A genome is a collective term for all the genetic material within an organism. In essence,the genome decides exactly what that organism will look and act like at birth one huge, expansive instruction manual that tellscells their duties. Every living thing has a genome, from bacteria to plants to humans, and they are all different in size with various combinations of genes inside.

The human genome packs in 30,000 genes, but this is just 1% of the total genetic material contained within. Quite frankly, its a mess in there much of the genetic material is duplicated DNA that (supposedly) does very little, and the vast majority of DNA simply doesnt code for anything(these sections are calledintrons). That isnt to say it does nothing. In fact,recent studieshave shown us that non-coding DNA is essential to controlling whether our genes get switched on or not. However, most of the time its the actual genes that are the important bit.

Studying the genome of humans and other organisms is vitalfor a number of reasons.Firstly, it helps us characterize each one before genomics, scientists simply grouped animals and plants by what they looked like, but research into their genes now allows for accuratecharacterization oforganismsinto specificgeneraand species.

In humans, genomic research has allowed researchers to understand the underlying causes of many complex diseases and find possible targets for treatment.Currently, the best tool to do thisisgenome-wide association studies (GWAS).

The idea behind GWAS is relatively intuitive simply take a group of people with the disease you wish to study, and compare their genomesfor common genetic variants that could predict the presence of that disease.These studies have illuminated a huge number of variants linked with higher disease prevalence while also helping researchers to understand the role each gene playsin the human body.Although powerful, GWAS studies are purely a starting point. Following a large-scale GWAS, researchers must thenanalyzeany variants that are highlighted in great depth, and many times such research will provide nothing of clinical relevance. However, itsstill our best way of identifying risk variants in genetic disease.

So,we know the genome is packed to the brim with genes that code for proteins, separated by large strings ofnon-coding DNA. However, when cells replicateearly in development they usually go throughchromosomal recombination, in which chromosomes trade regions of their genetic code between each other. This spreads genes to many different positions (called loci)throughout the genome. If we can make a map of these genes, we candiscover their function, how they are inherited, or target them with therapies.

Therefore, we want to create a genome map.There are two types of maps used in genomics: genetic maps and physical maps.

Physical mapsare relatively straightforward, in which genomic loci are mapped based on the physical distance between them, measured in base pairs.The most common way to create a physical map of a human genome is byfirst breaking the DNA sequence into many fragments, before using a variety of different techniques to identify how those pieces fit back together. By understanding which pieces overlapand reconstructing the shattered genome, scientists can gain a decently accurate map of where each gene lies.

Genetic mapsare slightly different,using specific marker regions within the DNA that are used as trackers. These mapsrequiresamples (usually saliva) from family members,which are then compared toidentifyhow much recombination has occurred that includes markers of interest. The principle is thatif two genes are close together on thechromosome, thenthey are more likely to travel together through the genome as it recombines. By using this data,scientists can get a rough idea of where specific genes lie on chromosomes. However, it is not as accurate as physical mapping andrelies heavilyon a decentpopulation size andthe number of genetic markers used.

A genome browser is any available database that allows a user to access and compare genomes in an intuitive way. When you map or sequence a genome, the data is prettymessy.Genomes are usually stored in huge files, calledFASTAfiles, that contain extensive strings of letters that would look foreign to most users. Genome browsers take this data and make it accessibleto scientists around the globe.

Many genome browsers are available online, containing bacterial, model organism, and human reference genomes.

Genomelinkis one of the latest examples of public access and analysis of genomes. The industry took off in recentyears, with the rapid rise of sites that provide ancestry and medical information based on genomic sequencing, includingAncestryand23andMe.These sites work by comparing genetic markers associated with different populations should you share specific regions of DNA that correspond with African populations, for example, you may have some relation to African ancestors. Each site uses its own markers, so information may vary between tests, and some have disputed the true accuracy of these tests, although advances in genomics have significantly improved them in recent years.

Genomelinkgoes further than most sites, claiming to provide information on a huge variety of genetic traits that a user may have. These include metabolism, sports performance, and even personality traits such as loneliness. Each trait isdrawn from genome correlation studies, with each taking a specific trait and comparing the genomes of each carrier of that trait.

However, although bothGenomelinkand other sites use up-to-date reference genomes and are usually relatively accurate, they should never be substituted for medical information. If you believe you carry a pathogenic genevariant, you should seek advice from a genomic counselor.

Continued here:

Genomes, Maps, And How They Affect You - IFLScience

Are Phages Overlooked Mediators of Health and Disease? – The Scientist

When microbiologist Breck Duerkop started his postdoc in 2009, he figured hed be focusing on bacteria. After all, hed joined the lab of microbiome researcher Lora Hooper at the University of Texas Southwestern Medical Center in Dallas to study host-pathogen interactions in the mammalian gut and was particularly interested in what causes some strains of normally harmless commensal bacteria, such as Enterococcusfaecalis, to become dangerous, gut-dominating pathogens. Hed decided to explore the issue by giving germ-free mice a multidrug-resistant strain of E. faecalis that sometimes causes life-threatening infections in hospital patients, and analyzing how these bacteria express their genes in the mouse intestine.

Not long into the project, Duerkop noticed something else going on: some of the genes being expressed in E. faecalis werent from the regular bacterial genome. Rather, they were from bacteriophages, bacteria-infecting viruses that, if they dont immediately hijack and kill the cells they infect, can sometimes incorporate their genetic material into the bacterial chromosome. These stowaway viruses, known as prophages while theyre in the bacterial chromosome, may lie dormant for multiple bacterial generations, until certain environmental or other factors trigger their reactivation, at which point they begin replicating and behaving like infectious agents once again. (See illustration below.) Duerkops data showed that the chromosome of the E. faecalis strain he was using contained seven of these prophages and that the bacteria were churning out virus particles with custom combinations of these prophage sequences during colonization of the mouse gut.

The presence of viruses in Duerkops E. faecalis strain wasnt all that surprising. Natural predators of bacteria, bacteriophages are the most abundant biological entities on the planet, and in many fields, researchers take their presence for granted. Nobody really was thinking about phages in the context of bacterial communities in animal hosts, Duerkop says. It would [have been] very easy to just look at it and say, Oh, there are some phage genes here. . . . Moving on. But he was curious about why E. faecalis would be copying and releasing them, rather than leaving the prophages asleep in its chromosome, while it was trying to establish itself in the mouse intestine.

Predation is just one type of phage-bacteria interaction taking place within the mammalian microbiome; many phages are capable of inserting their genomes into the bacterial chromosome.

Encouraged by Hooper, he put his original project on hold in order to dig deeper. To his surprise, he discovered that the E. faecalis strain, known as V583, seemed to be using its phages to gain a competitive advantage over related strains. Experiments with multiple E. faecalis strains in cell culture and in mice showed that the phage particles produced by the bacteria didnt harm other V583 cells, but infected and killed competing strains. Duerkop and his colleagues realized that, far from being background actors in the bacterial community, the phages are important for colonization behavior for this opportunistic pathogen.

The idea that a phage could play such a significant role in the development of the gut bacterial community was relatively novel when the team published its results in 2012. Since then, its been pretty well established that phages can shape the assembly of microbial communities in the intestine, and that can influence the outcome on the hosteither beneficially or detrimentally, says Duerkop, who now runs his own lab at the University of Colorado School of Medicine in Aurora. Theres evidence that phages help bacteria share genetic material with one another, too, and may even interact directly with the mammalian immune system, an idea that Duerkop says would have had you laughed out of a room of immunologists just a few years ago.

Around the time that Duerkop was working on E. faecalis in Dallas, University of Oxford postdoc Pauline Scanlan was studying Pseudomonas fluorescens, a bacterial species that is abundant in the natural environment and is generally harmless to humans, although its in the same genus as the important human pathogen Pseudomonas aeruginosa. Bacteria in this genus sometimes evolve whats known as a mucoid phenotypethat is, cells secrete large amounts of a compound called alginate, forming a protective goo around themselves. In P. aeruginosa, this goo can help the bacteria evade the mammalian immune system and antibiotics, and when it crops up, its not good news for the patient, Scanlan says. She was curious about what causes a non-mucoid bacterial population to evolve into a mucoid one and had found previous research suggesting that the presence of bacteriophages could play a role. Other studies documented high densities of phages in mucus samples from the lungs of some cystic fibrosis patients with P. aeruginosa infections.

Working in the lab of evolutionary biologist Angus Buckling (now at the University of Exeter), Scanlan grew a strain of P. fluorescenswith a phage called Phi2 that specifically infects and destroys this bacterium. Cells with the gummy mucoid coating, the researchers noted, were more resistant to phage infection than regular cells were. Whats more, over generations, bacterial populations were more likely to evolve the mucoid phenotypes in the presence of Phi2 than they were in its absence, indicating that the phenotype may arise in Pseudomonas as an adaptive response to phage attack. Scanlan, now at University College Cork (UCC) in Ireland, notes that more work is needed to extend the findings to a clinical setting, but the results hint that phages could in some cases be responsible for driving bacteria to adopt more virulent phenotypes.

Such a role for viruses in driving bacterial evolution fits well with phages reputation as the ultimate predators, says Colin Hill, a molecular microbiologist also at UCC who got his introduction to phages studying bacteria used in making fermented foods such as cheese. Hill notes an estimate commonly cited in the context of marine biologya field that explored phage-bacteria interactions long before human biology didthat phages kill up to 50 percent of the bacteria in any environment every 48 hours. The thing that any bacterium has on its mind most, if bacteria had minds, would be phage, Hill says, because its the thing most likely to kill them.

Several in vivo animal studies lend support to the idea that predatory phages help shape bacterial evolution and community composition in the mammalian microbiome. In 2019, for example, researchers at Harvard Medical School reported that phages not only directly affect the bacteria they infect in the mouse gut, but also influence the rest of the microbiome community via cascading effects on the chemical and biological composition of the gut. Observational studies hint at similar processes at work in the human gut. A few years ago, researchers at Washington University Medical School in St. Louis observed patterns of phage and bacterial population dynamics that resembledpredator-prey cycles in the guts of children younger than two years old: low bacterial densities at birth were followed by decreases in phages, after which the bacteria would rebound, and then the phages would follow suit. The team concluded that these cycles were likely a natural part of healthy microbiome development.

Although researchers are only just beginning to appreciate the importance of phages in microbiome dynamics, theyve already begun to explore links to human disease. Authors of one 2015 study reported that Crohns disease and ulcerative colitis patients showed elevated levels of certain phages, particularly within the viral order Caudovirales. They proposed that an altered virome could contribute to pathogenesis through predator-prey interactions between phages and their bacterial hosts. Other studies have explored possible phage-driven changes in the bacterial community in human diseases such as diabetes and certain cancers that are known to be associated with a disrupted microbiome. But the observational nature of human microbiome studies prevents conclusions about what drives whatchanges in virome composition could themselves be the result of disruptions to the bacterial community, for example.

Currently, researchers are exploring the possibility of using predatory phages as weapons against pathogenic bacteria, particularly those that present a serious threat to public health due to the evolution of resistance to multiple antibiotics. Its the principle that the enemy of my enemy is my friend, says Yale University virologist and evolutionary biologist Paul Turner. If we have a pathogen that is in your microbiome, can we go in and remove that bacterial pathogen by introducing a predatory phage, something that is cued to only destroy [that pathogen]? Although the strategy was first proposed more than a century ago, we and others are trying to update it, he adds. (See My Enemys Enemy below.)

Phages can interact with bacteria in two main ways. In the first, phages infect a bacterial cell and hijack that cells protein-making machinery to replicate themselves, after which the newly made virus particles lyse the bacterium and go on to infect more cells. In the second process, known as lysogeny, the viral genome is incorporated into the bacterial chromosome, becoming whats known as a prophage, and lies dormantpotentially for many generationsuntil certain biotic or abiotic factors in the bacterium or the environment induce it to excise itself from the chromosome and resume the cycle of viral replication, lysis, and infection of new cells.

Predation is just one type of phage-bacteria interaction taking place within the mammalian microbiome. Many phages are capable of inserting their genomes into the bacterial chromosome, a trick beyond the bounds of traditional predator-prey relationships in other kingdoms of life that adds complexity to the relationship between phages and bacteria, and consequently, to phages potential influences on human health.

This role for phages has long been of interest to Imperial College Londons Jos Penads. Over the last 15 years or so, he and colleagues have described various ways in which many phages help bacteria swap genetic material among cells. He likens phages to cars that bacteria use to transport cargo around and says that, in his opinion, it almost makes sense to view phages as an extension of bacteria rather than as independent entities. This is part of the bacterium, he says. Without phages, bacteria cannot really evolve. They are absolutely required.

With lateral [transduction] you can move huge parts of the bacterial chromosome.

Jos Penads, Imperial College London

In the simplest case, the genetic material being transported consists of viral genes in the genomes of so-called temperate phages, which spend at least part of their lifecycle stashed away in bacterial chromosomes as prophages. These phages are coming to be appreciated by microbiologists as an important driver of bacterial evolution in the human microbiome, notes Hill. The lack of practical and accurate virus detection methods makes it difficult to precisely characterize a lot of the phages resident in mammalian guts, but microbiologists estimate that up to 50 percent are temperate phages, and, more importantly for human health, that many of them may carry genes relevant to bacterial virulence. Researchers have long known, for example, that many toxins produced by bacteriaincluding Shiga toxin, made by some pathogenic E. coli strains, and cholera toxin, secreted by the cholera-causing bacterium Vibrio choleraeare in fact encoded by viral genes contained in the bacterial chromosome, and that infection by temperate phages that carry these genes may be able to turn a harmless bacterial population into one thats pathogenic.

Evidence from other studies points to phages as capable of transporting not just their own genomes, but bits of bacterial DNA as well. In the best-studied examples of this phenomenon, known as bacterial transduction, tiny chunks of the bacterial genome get packed up into viral particles instead of or alongside the phage genome, and are shuttled to other bacterial cells. In 2018, however, Penads and colleagues presented results showing that very large pieces of bacterial DNA can also be exchanged this way, in a process the team named lateral transduction. Not only does the discovery have implications for how researchers understand viral replication in infected cells, it shines light on a novel way for bacteria to share their genes. With lateral [transduction] you can move huge parts of the bacterial chromosome, says Penads. The team first observed the phenomenon in the important human pathogen Staphylococcus aureus, and is now looking for it in other taxa, he adds. Right now, for us, its important to show that its a general mechanism, with many bugs involved.

Although the research is still in the nascent stages, this mechanism could help explain findings from University of Barcelona microbiologist Maite Muniesa and others who have been studying whether phages transport antibiotic resistance genes between bacterial cells, and whether they can act as reservoirs for these genes in the natural environment. Early studies on this issue had proposed that, like many toxin genes, antibiotic resistance genes might be encoded in viral sequences and thus transported to bacteria with the rest of the viral genome. But the idea wasnt without controversya 2016 analysis of more than 1,100 phage genomes from various environments concluded that phage genomes only rarely include antibiotic resistance genes. That studys authors argued that prior reports of these genes in phage genomes were likely due to contamination, or to the difficulty of distinguishing viral sequences from bacterial ones.

Nevertheless, Muniesas team has published multiple reports of antibiotic resistance sequences in phage particles, including in samples of meat products from a Barcelonan fresh-food retailer, and more recently in seawater samplesnot only from the Mediterranean coastline but even off the coast of Antarctica, far from human populations that use antibiotics. We were pretty surprised that we found these particles in this area with low human influence, Muniesa says. Although her team hasnt determined whether the antibiotic resistance sequences are of phage or bacterial origin, she suspects they might be bacterial genes that ended up in phage particles during lateral transduction or some process like it. Bacteria are using these phage particles in a natural way to move [genes] between their brothers and sisters, lets say, she says. Its happening everywhere.

Duerkop cautions that its not yet clear how often phage-mediated transfer of antibiotic resistance genes occurs or how significant it is in the epidemiology of drug-resistant infections in people. Its not to say that antibiotic resistance cant be mediated through phage, he says. I just dont think its a major driver of antibiotic resistance.

Whatever its natural role, temperate phages ability to insert themselves into bacterial genomes could have applications in new antibacterial therapies. Viruses that insert pathogenicity-reducing genes or disrupt the normal expression of the bacterial chromosome could be used to hobble dangerous bacteria, for examplean approach that proved successful last year in mouse experiments with Bordetella bronchiseptica, a bacterium that often causes respiratory diseases in livestock. Using a phage from the order Siphoviridae, researchers found that infected B. bronchiseptica cells were substantially less virulent in mice than control cells were, likely because the viral genome had inserted itself in the middle of a gene that the bacterium needs to infect its host. Whats more, injecting mice with the phage before exposing them to B. bronchiseptica seemed to completely protect them from infection by the microbe, hinting at the possibility of using temperate phages as vaccines against some bacteria.

Bacteria-infecting viruses, or bacteriophages, may influence microbial communities in the mammalian gut in various ways, some of which are illustrated here. Through predation, phages can influence the abundance of specific bacterial taxa, with indirect effects on the rest of the community, and can drive the evolution of specific bacterial phenotypes. Phages can also incorporate their genomes into bacterial chromosomes, where the viral sequences lie dormant as prophages until reactivated. Researchers have found that phages interact directly with mammalian cells in the gut, too. These cross-kingdom interactions could affect the health of their eukaryotic hosts.

Despite growing interest in phages role in shuttling material among bacteria, some of the biggest recent developments in research on phages in the human gut have turned out not to involve bacteria at all. One of the key pieces of this particular puzzle was fitted by University of Utah microbiologist June Round and her colleagues, who as part of a phage therapy study a few years ago fed several types of Caudovirales phages to mice that were genetically predisposed to certain types of cancer and had been infected with a strain of E. coli known to increase that risk. The premise was pretty simplistic, recalls Round. It was just to identify a cocktail of phage that would target bacteria that we know drive chronic colorectal cancer.

The team was surprised to see that the phages, despite being viewed by most researchers as exclusively bacteria-attacking entities, prompted a substantial response from the mices immune systemsmammalian defenses that should, according to conventional wisdom, be indifferent to the war between bacteria and phages in the gut. Intrigued, the researchers tried adding their phage cocktail to mice that had had their gut bacteria completely wiped out with antibiotics. Still, they saw an immune response. It was then, Round says, that we realized that [the phages] were likely interacting with the immune system.

Exploring further, the team found that the phages were activating both innate and adaptive immune responses in mice. In rodents with colitis, the phages exacerbated inflammation. Turning their attention to people, the researchers isolated phages from ulcerative colitis patients with active disease, as well as from patients with disease in remission and from healthy controls, and showed that only viruses collected from patients with active disease stimulated immune cells in vitro. And when the team studied patients who received fecal microbiota transplantationan experimental treatment for ulcerative colitis that involves giving beneficial gut bacteria to a patient to try to alleviate inflammation and improve symptomsthe researchers found that a lower abundance of Caudovirales in a recipients intestine at the time of transplant correlated with treatment success.

Some of the biggest recent developments in research on phages in the human gut have turned out not to involve bacteria at all.

By the time the team published its results in 2019, a couple of other groups had also documented evidence of direct interactions between phages and host immune systems. Meanwhile, Duerkop, Hooper, and colleagues reported that mice with colitis tended to have specific bacteriophage communities, rich in Caudovirales, that developed in parallel with the disease. Many of the types of phage they identified in the intestines of those diseased mice also turned up in high abundance in samples taken from the guts of people with inflammatory bowel disease, the researchers noted in their paper, supporting a possible role for phages in the development of disease.

Round says that researchers are still unsure about exactly why these trans-kingdom interactions are happeningparticularly when it comes to host adaptive immune responses, which tend to be specific to a particular pathogen. She speculates that mammalian hosts might derive a benefit from destroying certain phages if those phages are carrying genes that could aid a bacterium with the potential to cause disease. Exactly how immune cells would detect what genes a phage is carrying isnt yet clear.

Meanwhile, hints of collaboration between eukaryotic cells and phages have cropped up in the work of several other labs. One recent study of a phage therapy against P. aeruginosa found that phages and immune cells seem to act in synergy to clear infections in mice. Other work has indicated that phages bind to glycoproteins presented by cells along the mucosal surfaces of the mammalian gut and may provide a protective barrier against bacterial pathogensa relationship that some microbiologists have argued represents an example of phage-animal symbiosis. Duerkop adds that theres evidence emerging to support the idea that phages in the mammalian intestine not only can be engulfed by certain eukaryotic cells, but also might slip out of the gut and into the bloodstream to make their way to other parts of the body, with as yet undiscovered consequences.

Whether these mechanisms can be exploited for therapeutic purposes remains to be seen, but Round notes that they do raise the possibility of unintended effects in some circumstances if researchers try to use phages to influence human health via the gut microbiome. At least in the type of chronic inflammatory diseases she and her team have been studying, we might just be making it worse by using phages to target disease-causing bacteria, she says, adding that all research groups studying such approaches should take into account potential knock-on effects. Considering phages multiple interactions, with both bacteria and animal cells, she says, its a lot more complex than what wed appreciated.

Bacteriophages ability to selectively target and kill specific bacterial strains has long been recognized as a possible basis for antimicrobial therapies. Proposed by researchers in Europe as early as 1919, phage therapy went on to be widely promoted in Germany, the USSR, and elsewhere before being overtaken worldwide by the soaring popularity of antibiotics in the 1940s. But the strategy has come back into fashion among many microbiologists, thanks to the growing public health problem of antibiotic resistance in bacterial pathogens and to the rapidly improving scientific understanding of phage-bacteria interactions.

Some of the latest approaches aim not only to target specific bacteria with phages, but also to avoid (or exploit) the seemingly inevitable evolution of phage resistance in those bacteria. One way researchers try to do this is by taking advantage of an evolutionary trade-off: bacterial strains that evolve adaptations to one therapy will often suffer reduced fitness when confronted with a second therapy, perhaps one that targets the same or similar pathways in a different way.

Yale University virologist and evolutionary biologist Paul Turner, for example, has studied how phages in the Myoviridae (a family in the order Caudovirales) can promote antibiotic sensitivity in the important human pathogen Pseudomonas aeruginosa. Turner and colleagues showed a few years ago that one such phage binds to a protein called OprM in the bacterial cell membrane, and that bacterial populations under attack from these phages will often evolve reduced production of OprM proteins as a way of avoiding infection. However, OprM also happens to be important for pumping antibiotics out of the cell, such that abnormal OprM levels can reduce bacterias abilityto survive antibiotic treatment in vitro.

A handful of groups have published case studies using this kind of approach, known as phage steering, in humans. A couple years ago, for example, Turner and colleagues reported that a post-surgery patients chronicP. aeruginosa infection cleared up after treatment with the OprM-binding phage and the antibiotic ceftazidime. Researchers at the University of California, San Diego, in partnership with California-based biotech AmpliPhi Biosciences (now Armata Pharmaceuticals), reportedsimilar successin a cystic fibrosis patient with a P. aeruginosa infection who was treated with a mixture of phages and with antibiotics. A Phase 1/2 trial for that therapy was greenlighted by the US Food and Drug Administration last October.

The complexity of the relationship between phages and bacteria, not to mention recently discovered interactions between phages and eukaryotic cells, has many researchers tempering optimism about phage therapy with caution. There might be off-target effects to this that we hadnt really thought about, says University of Colorado School of Medicine microbiologist Breck Duerkop. That said, thanks to research in the last few years, the black veil on phage therapy is, I believe, being lifted, he adds, which Im really excited about because I think they have a ton of potential to be used in biomedicine.

Read this article:

Are Phages Overlooked Mediators of Health and Disease? - The Scientist

Two Gene Therapies Fix Fault in Sickle Cell Disease and -thalassemia – MD Magazine

Two different gene therapies have been used to mitigate a mechanism underlying development of sickle cell disease (SCD) and transfusion-dependent -thalassemia (TDT), and both have demonstrated clinical success in separate, concurrently published trials.

The hemoglobinopathies manifest after fetal hemoglobin synthesis is replaced by adult hemoglobin in individuals who have inherited a mutation in the hemoglobin subunit gene (HBB).Identifying factors in the conversion from fetal to adult hemoglobin synthesis, however, has provided potential targets for therapeutic intervention.

Gene therapy that can safely arrest or reduce the conversion offers the potential for a one-time treatment to obviate the need for lifetime transfusions and iron chelation for patients with TDT, and the pain management, transfusions and hydroxyurea administration for those with SCD.

Two groups of investigators have now reported in The New England Journal of Medicine that, using different gene therapy techniques that target the transcription factor, BCL11a, involved in the globin switching, they have improved clinical outcomes in patients with TDT and with SCD.

In an editorial in the issue featuring the 2 studies, Mark Walters, MD, Blood and Marrow Transplant Program, University of California, San Francisco-Benioff Children's Hospital, welcomed the breakthroughs.

"These trials herald a new generation of broadly applicable curative treatments for hemoglobinopathies," Walters wrote.

In one clinical trial with 2 patients, one with TDT and the other with SCD, Haydar Frangoul, MD, MS, Medical Director, Pediatric Hematology/Oncology, Sarah Cannon Center for Blood Cancer at the Children's Hospital at Tristar Centennial, and colleagues administered CRISPR-Cas9 gene edited hematopoietic stem and progenitor cells (HSPCs) with reduced BCL11A expression in the erythroid lineage.

The product, CTX001, had been shown in preclinical study to restore -globulin synthesis and reactivate production of fetal hemoglobin. Both patients underwent busulfan-induced myeloablation prior to receiving the treatment.

The investigators suggested that the CRISPR-Cas9-based gene-edited product could change the paradigm for patients with these conditions, if it was found to successfully and durably graft, produce no "off-target" editing products, and, importantly, improve clinical course.

"Recently approved therapies, including luspatercept and crizanlizumab, have reduced transfusion requirements in patients with TDT and the incidence of vaso-occlusive episodes in those with SCD, respectively, but neither treatment addressed the underlying cause of the disease nor fully ameliorates disease manifestations," Frangoul and colleagues wrote.

The investigators reported that both patients had "early, substantial, and sustained increases" in pancellularly distributed fetal hemoglobin levels during the 12-month study period. Further, the patients no longer required transfusions, and the patient with SCD no longer experienced vaso-occlusive episodes after the treatment.

In commentary accompanying the report, Harry Malech, MD, Genetic Immunotherapy Section, Laboratory of Clinical Immunology and Microbiology, National Institute of Allergy and Infectious Disease (NIAID), National Institutes of Health (NIH), Bethesda, MD, described the investigators' application of the gene-editing technology as a "remarkable level of functional correction of the disease phenotype."

"With tangible results for their patients, Frangoul et al have provided a proof of principle of the emerging clinical potential for gene-editing treatments to ameliorate the burden of human disease," Malech pronounced.

In the other published trial, with 6 patients with SCD, Erica Esrick MD, Dana-Farber/Boston Children's Cancer and Blood Disorders Center, Harvard Medical School, and colleagues described results with infusion of gene-modified cells derived from lentivirus insertion of a gene that knocks down BCL11a by encoding an erythroid-specific, inhibitory short-hairpin RNA (shRNA).

The severity of SCD that qualified patients for enrollment included history of stroke (n = 3), frequent vaso-occlusive events (n = 2) and frequent episodes of priapism (1).Patients were followed for 2 years, and offered enrollment in a 13-year long-term follow-up study.The infusion of the experimental drug BCH-BB694, from the short hairpin RNA embedded within an endogeonous micro RNA scaffold (termed a shmiR vector), was initiated after myeloablation with busulfan.

Esrick and colleagues reported that, at median follow-up of 18 months (range, 7-29), all patients had engraftment and a robust and stable HbF induction broadly distributed in red cells.Clinical manifestations of SCD were reduced or absent during the follow-up period; with no patient having a vaso-occlusive crisis, acute chest syndrome, or stoke subsequent to the gene therapy infusion.Adverse events were consistent with effects of the preparative chemotherapy.

"The field of autologous gene therapies for hemoglobinopathies is advancing rapidly," Esrick and colleagues reported, "including lentiviral trials of gene addition in which the nonsickling hemoglobin is formed from an exogenous -globin or modified -globin gene."

Walters agreed that gene therapy is rapidly progressing, but expressed concern about the large gap that looms between laboratory bench and clinical bedside, particularly for this affected population.

"Access to and delivery of these highly technical therapies in patients with sickle cell disease will be challenging and probably limited to resource-rich nations, at least in the short term," Walters commented.

The studies, CRISPR-Cas9 Gene Editing for Sickle Cell Disease and -Thalassemia, as well as, Post-Transcriptional Genetic Silencing of BCL11A to Treat Sickle Cell Disease, were published online in The New England Journal of Medicine.

Continue reading here:

Two Gene Therapies Fix Fault in Sickle Cell Disease and -thalassemia - MD Magazine

The First Targeted Therapy For Lung Cancer Patients With The KRAS Gene MutationExtraordinary Results With Sotorasib – SurvivorNet

First Targeted Therapy For Lung Cancer With KRAS

For the first time, there may be an effective treatment option for people with lung cancer that contains a genetic mutation called KRAS. The results of a groundbreaking using a drug calledSotorasib have just been published in the highly-respected New England Journal of Medicine.

Dr. Roy Herbst, Chief of Medical Oncology at Yale tells SurvivorNet We are excited we have a drug that could work in these patients. The fact that tumors respond to this therapy is a big deal.

Lung cancer remains the leading cause of cancer death in the united states. The most common form of lung cancer, non-small cell lung cancer (NSCLC), has recently seen major advancements with new treatments such as immunotherapy and targeted therapies extending the lives of thousands of patients. However, despite these recent advancements little has been available to help patients who have lung cancer with a KRAS mutation. This mutation is found in approximately 10-12% of patients with NSCLC and any drug that can improve the outlook for these patients would be a game-changer for lung cancer.

Now we finally have targeted therapy options for these patients.

In patients who have advanced stage or metastatic NSCLC most patients will have their tumor tested for genetic abnormalities or biomarkers to help their doctors select what treatments are best. Some common biomarkers such as EGFR and PDL1 have medications that doctors can use to target the lung cancer and improve a patients survival and quality of life. Despite this, one biomarker that has never had a treatment is KRAS. KRAS is a mutation that occurs in some patients with NSCLC and is generally associated with poor outcomes. One reason this mutation is considered a bad risk factor is that unlike other mutations such as EGFR there has never been a drug approved to treat this type of lung cancer.

Fortunately, for patients, this may be changing soon. A new drug called Sotorasib that specifically targets the KRAS mutation recently showed positive results in the early phase CODEBREAK 100 study. Based on the results from the early phase study Sotorasib was granted Break Through Therapy Designation and the drug has been accepted into the Real-Time Oncology Pilot Review Program by the U.S. Food and Drug Administration (FDA). When discussing the trial, Dr. Velcheti, Director of the Thoracic Medical Oncology Program at NYU Langone says The CODEBREAK 100 trial represents the clinical validation of significant research efforts spanning decades. Now we finally have targeted therapy options for these patients.

Overall I am impressed with this drug. It is hard for the public to understand just how far drug development has come.

So what does this mean for patients? This means that the new drug targeting KRAS may soon be available for patients whose tumors harbor this mutation and who have not responded to other treatments.

Lung specialists from across the country were eager to speak with SurvivorNet regarding the exciting news. Dr. Brendon Stiles, Associate Professor of Cardiothoracic Surgery at Weill Cornell Medical Center tells SurvivorNet Overall I am impressed with this drug. It is hard for the public to understand just how far drug development has come. The KRAS mutation has long been considered undruggable, meaning if you have this mutation, there was not a medicine designed specifically to treat this type of cancer. The chance of responding to the new therapy is around 40%. Although, researches would prefer to see this percent be higher the results of the study give hope that future therapies may have even better outcomes. Dr. Herbst is also optimistic about the future of drugs targeting KRAS and thinks the results of this study opens up a whole new world for lung cancer. If you or a loved one have NSCLC with a KRAS mutation ask your doctor about what treatment options are best for you.

Learn more about SurvivorNet's rigorous medical review process.

Dr. Roy Herbst, Chief of Medical Oncology at Yale tells SurvivorNet We are excited we have a drug that could work in these patients. The fact that tumors respond to this therapy is a big deal.

Now we finally have targeted therapy options for these patients.

In patients who have advanced stage or metastatic NSCLC most patients will have their tumor tested for genetic abnormalities or biomarkers to help their doctors select what treatments are best. Some common biomarkers such as EGFR and PDL1 have medications that doctors can use to target the lung cancer and improve a patients survival and quality of life. Despite this, one biomarker that has never had a treatment is KRAS. KRAS is a mutation that occurs in some patients with NSCLC and is generally associated with poor outcomes. One reason this mutation is considered a bad risk factor is that unlike other mutations such as EGFR there has never been a drug approved to treat this type of lung cancer.

Fortunately, for patients, this may be changing soon. A new drug called Sotorasib that specifically targets the KRAS mutation recently showed positive results in the early phase CODEBREAK 100 study. Based on the results from the early phase study Sotorasib was granted Break Through Therapy Designation and the drug has been accepted into the Real-Time Oncology Pilot Review Program by the U.S. Food and Drug Administration (FDA). When discussing the trial, Dr. Velcheti, Director of the Thoracic Medical Oncology Program at NYU Langone says The CODEBREAK 100 trial represents the clinical validation of significant research efforts spanning decades. Now we finally have targeted therapy options for these patients.

Overall I am impressed with this drug. It is hard for the public to understand just how far drug development has come.

So what does this mean for patients? This means that the new drug targeting KRAS may soon be available for patients whose tumors harbor this mutation and who have not responded to other treatments.

Lung specialists from across the country were eager to speak with SurvivorNet regarding the exciting news. Dr. Brendon Stiles, Associate Professor of Cardiothoracic Surgery at Weill Cornell Medical Center tells SurvivorNet Overall I am impressed with this drug. It is hard for the public to understand just how far drug development has come. The KRAS mutation has long been considered undruggable, meaning if you have this mutation, there was not a medicine designed specifically to treat this type of cancer. The chance of responding to the new therapy is around 40%. Although, researches would prefer to see this percent be higher the results of the study give hope that future therapies may have even better outcomes. Dr. Herbst is also optimistic about the future of drugs targeting KRAS and thinks the results of this study opens up a whole new world for lung cancer. If you or a loved one have NSCLC with a KRAS mutation ask your doctor about what treatment options are best for you.

Learn more about SurvivorNet's rigorous medical review process.

Read this article:

The First Targeted Therapy For Lung Cancer Patients With The KRAS Gene MutationExtraordinary Results With Sotorasib - SurvivorNet

Atsena Therapeutics Raises $55 Million Series A Financing to Advance LCA1 Gene Therapy Clinical Program, Two Preclinical Assets, and Novel Capsid…

DURHAM, N.C. and BOSTON, Dec. 16, 2020 (GLOBE NEWSWIRE) -- Atsena Therapeutics, a clinical-stage gene therapy company focused on bringing the life-changing power of genetic medicine to reverse or prevent blindness, today announced it has closed an oversubscribed $55 million Series A financing led by Sofinnova Investments with participation from additional new investors Abingworth and Lightstone Ventures. Founding investors Hatteras Venture Partners and the Foundation Fighting Blindness RD Fund, along with existing investors Osage University Partners, University of Florida, and Manning Family Foundation, also participated in the round. Sarah Bhagat, PhD, Partner at Sofinnova, Jackie Grant, PhD, Principal at Abingworth, and Jason Lettmann, General Partner at Lightstone, will join Atsenas board of directors.

Proceeds will be used to advance Atsenas ongoing Phase I/II clinical trial evaluating a gene therapy for patients with GUCY2D-associated Leber congenital amaurosis (LCA1), one of the most common causes of blindness in children, as well as complete manufacturing development for Phase 3. In addition, the funds will enable Atsena to expand its team to support the research and development of novel gene therapies, including the progression of two existing preclinical programs in inherited retinal diseases toward the clinic and advancement of the companys innovative adeno-associated virus (AAV) technology platform.

We are grateful for the support of our new and existing investors and are encouraged by their enthusiasm for the potential of our technology to overcome the unique hurdles of inherited retinal diseases to prevent or reverse blindness, said Patrick Ritschel, MBA, Chief Executive Officer of Atsena. The Series A financing provides financial runway to reach the key inflection point of reading out efficacy data from our LCA1 clinical trial. While we continue expeditiously advancing this trial and our preclinical programs, we are excited to accelerate our growth as a leading ophthalmic gene therapy company.

The Phase I/II LCA1 clinical trial is currently enrolling patients in the second dosing cohort. Atsena exclusively licensed the rights to the gene therapy from Sanofi, which originally licensed it from University of Florida. LCA is the most common cause of blindness in children. LCA1 is caused by mutations in the GUCY2D gene and results in early and severe vision impairment or blindness. GUCY2D-LCA1 is one of the most common forms of LCA, affecting roughly 20 percent of patients who live with this inherited retinal disease.

We believe Atsenas foundation in ocular gene therapy and potentially game-changing novel AAV vectors position the company to become a partner of choice, said Dr. Bhagat. Sofinnova is delighted to support Atsena and we look forward to helping the team further its mission to develop life-changing gene therapies for patients with inherited retinal diseases.

About Atsena TherapeuticsAtsena Therapeutics is a clinical-stage gene therapy company developing novel treatments for inherited forms of blindness. The companys ongoing Phase I/II clinical trial is evaluating a potential therapy for one of the most common causes of blindness in children. Its additional pipeline of leading preclinical assets is powered by an adeno-associated virus (AAV) technology platform tailored to overcome significant hurdles presented by inherited retinal disease, and its unique approach is guided by the specific needs of each patient condition to optimize treatment. Founded by ocular gene therapy pioneers Dr. Shannon Boye and Sanford Boye, Atsena has a licensing, research and manufacturing collaboration with the University of Florida and has offices in Boston, MA and North Carolinas Research Triangle, environments rich in gene therapy expertise. For more information, please visit atsenatx.com.

About Sofinnova InvestmentsSince our founding in 1974, Sofinnova has been active in life science investing. We are a clinical-stage biopharmaceutical investment firm with approximately $2.3B in assets under management and committed capital. We invest in both private and public equity of therapeutics-focused companies. Our goal is to actively partner with entrepreneurs in both the U.S. and Europe, across all stages of company formation. From drug development and navigating the regulatory process to company building and IPO, we strive to be collaborative, meaningful board members, and excellent partners at every level. We seek to build world class companies that aspire to dramatically improve the current state of medical care today and ultimately, the lives of patients. Sofinnova has expertise investing in gene therapy companies, including investments in Spark, which developed the first approved gene therapy, Akouos, and Audentes, and Xylocor. For more information, please visit http://www.sofinnova.com.

About Abingworth Abingworth is a leading transatlantic life sciences investment firm. Abingworth helps transform cutting-edge science into novel medicines by providing capital and expertise to top caliber management teams building world-class companies. Since 1973, Abingworth has invested in approximately 168 life science companies, leading to more than 44 M&A/exits and close to 70 IPOs. Our therapeutic focused investments fall into 3 categories: seed and early-stage, development stage, and clinical co-development. Abingworth supports its portfolio companies with a team of experienced professionals at offices in London, Menlo Park (California) and Boston. For more information, visit abingworth.com.

About Lightstone VenturesLightstone Ventures is a leading venture capital firm investing in therapeutic-oriented companies across the life science spectrum, from breakthrough medical devices to novel drugs and biopharmaceuticals. Founded in 2012, Lightstone has been part of many successful new ventures from inception through commercialization and plays a critical role guiding and building successful healthcare companies. With a proven strategy and global footprint, the Lightstone team has been involved in several of the largest venture-backed life science exits over the last decade including: ALX Oncology, Acceleron, Ardian, Calithera, Claret Medical, Disarm, MicroVention, Nimbus, Plexxikon, Portola, Promedior, Proteolix, Ra Pharma, Tizona, Twelve and Zeltiq. For more information, visithttps://www.lightstonevc.com.

Read more:

Atsena Therapeutics Raises $55 Million Series A Financing to Advance LCA1 Gene Therapy Clinical Program, Two Preclinical Assets, and Novel Capsid...

Locanabio Announces $100 Million Series B Financing to Advance Portfolio of Novel RNA-Targeted Gene Therapies for Neurodegenerative, Neuromuscular and…

SAN DIEGO, Dec. 14, 2020 /PRNewswire/ --Locanabio, Inc., an RNA-targeting gene therapy company focused on developing life-changing therapies for patients with severe neurodegenerative, neuromuscular and retinal diseases, today announced a $100 million Series B financing. The financing was led by Vida Ventures with participation from new investors including RA Capital Management, Invus, Acuta Capital Partners and an investment fund associated with SVB Leerink and prior Locanabio investors ARCH Venture Partners, Temasek, Lightstone Ventures, UCB Ventures and GV.

"This financing positions us to accelerate our efforts to advance multiple promising programs into IND-enabling studies in 2021 and to further develop our novel RNA-targeting platform, which has the potential to be a major new advance in medicine that can bring hope to patients with many devastating genetic diseases," said Jim Burns, Ph.D., chief executive officer at Locanabio. "We are pleased that a team of highly sophisticated investors led by Vida Ventures has joined in this financing round, further validating our progress in research and the significant potential of our unique RNA-targeting platform."

The financing will advance Locanabio's portfolio of RNA-targeted gene therapies and expand the technology platform to pursue a broad range of therapeutic indications. Locanabio is currently developing therapies to treat multiple genetic diseases with no approved therapeutic alternatives, including Huntington's disease, spinocerebellar ataxia type 1, myotonic dystrophy type 1, genetic forms of amyotrophic lateral sclerosis and retinal diseases.

The Company also announced that Rajul Jain, M.D., director of Vida Ventures, will join Locanabio's board of directors. Prior to joining Vida, Dr. Jain, a physician-scientist by background, was on the executive team and headed development for Kite Pharma, a Gilead company, and previously was global development lead for Amgen.

"The unique approach in RNA targeting using gene therapy to deliver RNA binding proteins developed by Locanabio represents the next frontier of genetic medicine with the ability to target the root cause of a range of genetic diseases," said Dr. Jain. "They have built a strong management team to execute this bold vision and we are proud to support them."

About Locanabio, Inc.

Locanabio is the global leader in developing a new class of genetic medicines. Our unique and multi-dimensional approach uses gene therapy to deliver RNA binding protein-based systems to correct the message of disease-causing RNA and thereby change the lives of patients with devastating genetic diseases. These broad capabilities delivered via gene therapy enable Locanabio to potentially address a wide range of severe diseases with a single administration. The company is currently advancing programs in neurodegenerative, neuromuscular and retinal diseases. For more information, visit http://www.locanabio.com.

Contacts

Company ContactMicah MackisonChief Business OfficerLocanabio, Inc. [emailprotected]

Media ContactJenna UrbanBerry & Company[emailprotected]+1-212-253-8881

SOURCE Locanabio, Inc.

Home page

More here:

Locanabio Announces $100 Million Series B Financing to Advance Portfolio of Novel RNA-Targeted Gene Therapies for Neurodegenerative, Neuromuscular and...

NeuBase Therapeutics Announces Positive Preclinical In Vivo Data for PATrOL-enabled Anti-gene for the Treatment of Myotonic Dystrophy Type 1 -…

In vivo data after single-dose IV administration demonstrate engagement with DMPK mRNA and broad rescue of mis-splicing across key transcripts

Findings provide support for hypothesized mechanism of action of anti-gene, which is designed to not degrade the DMPK transcript

Data further validate the potential of the PATrOL platform to develop highly targeted therapies that increase, decrease or change causal protein function

NeuBase management to hold conference call and webcast today, December 16, at 8:00 a.m. EST

PITTSBURGH, Pa., Dec. 16, 2020 (GLOBE NEWSWIRE) -- NeuBase Therapeutics, Inc. (Nasdaq: NBSE) ("NeuBase" or the "Company"), a biotechnology company accelerating the genetic revolution using a new class of synthetic medicines, today announced positive in vitro and in vivo preclinical data for its PATrOL-enabled anti-gene therapies for the treatment of myotonic dystrophy type 1 (DM1). These new data show that PATrOL-enabled Compound A can rapidly resolve mis-splicing without negatively impacting DMPK protein levels. They also support the potential of NeuBases anti-gene approach to comprehensively treat the underlying cause of DM1.

Despite the fact that the genetic basis of DM1 is well understood today, there is still an urgent need to find the first genetically-targeted, disease-modifying treatment option for affected patients, said Curt Bradshaw, Ph.D., Chief Scientific Officer of NeuBase. DM1 is caused by a genetic mutation in the DMPK gene leading to mis-splicing of a broad spectrum of genes and DMPK protein insufficiency. A treatment option that addresses mis-splicing while retaining functional DMPK protein levels may be key to treating all aspects of DM1.

Dietrich A. Stephan, Ph.D., Chief Executive Officer of NeuBase, added, Using our proprietary PATrOL platform, we have designed a first-in-class anti-gene candidate that selectively binds mutant DMPK mRNA and opens its hairpin secondary structure, as opposed to a mechanism of action that explicitly degrades the mutant and wild-type transcripts indiscriminately, making it a unique option for the treatment of DM1. These in vitro and in vivo data both support our hypothesized mechanism of action and demonstrate rapid and broad resolution of the mis-splicing that is the primary cause of DM1.

This is the second set of positive data that weve announced in 2020 for our PATrOL-enabled therapies, which we believe serves as proof of concept that further validates our technologic foundation. With a single unified platform, we believe we can increase, decrease or change protein function of potentially any nucleic acid target, unique among genetic medicine approaches. We are excited by the progress we have made and look forward to providing additional updates on our platform and pipeline of programs at an R&D day in the first half of 2021.

In vitro data highlights in DM1 patient-derived fibroblasts:

In vivo data highlights in the HSALR transgenic mouse model of DM1 that expresses high levels of mutant CUG-repeat-containing mRNA (HSA) in skeletal muscle:

DM1 is a rare, autosomal dominant repeat expansion disorder characterized by progressive muscle wasting and weakness. It also affects the central nervous system (CNS) and heart. DM1 is caused by CTG nucleic acid repeats in the DMPK gene that produce a hairpin structure in the transcribed DMPK mRNA. The hairpin structure sequesters critical splice regulators and results in the mis-splicing of multiple gene transcripts. Furthermore, the binding of splice regulators traps the mutant DMPK mRNA in the nucleus, resulting in DMPK protein haploinsufficiency, or half the level of protein that is needed for normal function, which is thought to exacerbate the CNS and cardiac symptoms that are characteristic of DM1 (as knock-out mice for Dmpk show both severe cardiac conduction defects as well as issues with neuronal cytoskeletal remodeling manifesting in aberrant long-term potentiation). The prevalence of DM1 is >5/100,000 in the general population. There are currently no approved treatments for DM1.

Conference Call and Webcast Details

NeuBase Therapeutics, Inc. will discuss these data during a webcasted conference call with slides today, December 16, 2020, at 8:00 a.m. EST. To access the webcast, please click here. An archived recording of this presentation will be available following the call through the IR Calendar page on the Investors section of the Companys website, http://www.neubasetherapeutics.com.

About NeuBase Therapeutics, Inc.NeuBase is accelerating the genetic revolution using a new class of synthetic medicines. NeuBase's designer PATrOL therapies are centered around its proprietary drug scaffold to address genetic diseases at the source by combining the highly targeted approach of traditional genetic therapies with the broad organ distribution capabilities of small molecules. With an initial focus on silencing disease-causing mutations in debilitating neurological, neuromuscular and oncologic disorders, NeuBase is committed to redefining medicine for the millions of patients with both common and rare conditions. To learn more, visit http://www.neubasetherapeutics.com.

Use of Forward-Looking StatementsThis press release contains "forward-looking statements" within the meaning of the Private Securities Litigation Reform Act. These forward-looking statements are distinguished by use of words such as "will," "would," "anticipate," "expect," "believe," "designed," "plan," or "intend," the negative of these terms, and similar references to future periods. These forward-looking statements include, among others, those related to the potential significance and implications of the Companys positive in vitro and in vivo preclinical data for its PATrOL-enabled anti-gene therapies for the treatment of myotonic dystrophy. These views involve risks and uncertainties that are difficult to predict and, accordingly, our actual results may differ materially from the results discussed in our forward-looking statements. Our forward-looking statements contained herein speak only as of the date of this press release. Factors or events that we cannot predict, including those risk factors contained in our filings with the U.S. Securities and Exchange Commission, may cause our actual results to differ from those expressed in forward-looking statements. The Company may not actually achieve the plans, carry out the intentions or meet the expectations or projections disclosed in the forward-looking statements, and you should not place undue reliance on these forward-looking statements. Because such statements deal with future events and are based on the Company's current expectations, they are subject to various risks and uncertainties, and actual results, performance or achievements of the Company could differ materially from those described in or implied by the statements in this press release, including: the Company's plans to develop and commercialize its product candidates; the timing of initiation of the Company's planned clinical trials; the risks that prior data will not be replicated in future studies; the timing of any planned investigational new drug application or new drug application; the Company's plans to research, develop and commercialize its current and future product candidates; the clinical utility, potential benefits and market acceptance of the Company's product candidates; the Company's commercialization, marketing and manufacturing capabilities and strategy; global health conditions, including the impact of COVID-19; the Company's ability to protect its intellectual property position; and the requirement for additional capital to continue to advance these product candidates, which may not be available on favorable terms or at all, as well as those risk factors contained in our filings with the U.S. Securities and Exchange Commission. Except as otherwise required by law, the Company disclaims any intention or obligation to update or revise any forward-looking statements, which speak only as of the date hereof, whether as a result of new information, future events or circumstances or otherwise.

NeuBase Investor Contact:Dan FerryManaging DirectorLifeSci Advisors, LLCdaniel@lifesciadvisors.com OP: (617) 430-7576

NeuBase Media Contact:Cait Williamson, Ph.D.LifeSci Communicationscait@lifescicomms.com OP: (646) 751-4366

Excerpt from:

NeuBase Therapeutics Announces Positive Preclinical In Vivo Data for PATrOL-enabled Anti-gene for the Treatment of Myotonic Dystrophy Type 1 -...

Genetic Analysis Services Market: Uptake of Next-generation Sequencing and Multi-gene Tests to Drive Market – BioSpace

Genetic Testing to Establish Strong Foothold in Current and Future Healthcare System

The notable rise in the demand for hereditary genetic testing over the past few years is one of the major factors that is expected to fuel the growth of the global genetic analysis services market in the upcoming decade. Technological advancements coupled with the drive to discover new and innovative genetic analysis techniques are set to shape the overall growth trajectory of the global genetic analysis services market during the forecast period. Over the past decade, the genome testing sector has witnessed consistent developments due to which, the global genetic analysis services market is anticipated to expand at an impressive rate during the assessment period.

Read Report Overview - https://www.transparencymarketresearch.com/genetic-analysis-services-market.html

Hereditary genetic testing has emerged as ideal, and a rapidly evolving technology within the genetic analysis services market. This is likely to continue, owing to advancements in technology and findings of research activities. The increasing demand for improved and cutting-edge prediction and diagnostic tools and services coupled with surge in demand for disease monitoring is anticipated to play a key role in the overall growth of the global genetic analysis services market during the assessment period.

Request Brochure of Report - https://www.transparencymarketresearch.com/sample/sample.php?flag=B&rep_id=2555

Healthcare experts and credible researchers around the world are of the opinion that genetic testing is expected to be the future of the healthcare ecosystem. Advancements in the biomedical field coupled with the notable rise in the number of companies that are developing new genetic-testing kits are expected to augment the global genetic analysis services market during the forecast period. Moreover, as interest levels for precision medicine continues to witness sizeable growth around the world, as a result of which the demand for genetic analysis services is projected to grow at an impressive pace.

Request for Analysis of COVID-19 Impact on Genetic Analysis Services Market- https://www.transparencymarketresearch.com/sample/sample.php?flag=covid19&rep_id=2555

Uptake of Next-generation Sequencing and Multi-gene Tests to Drive Market

Advancements in the genetic technology are likely to play an instrumental role in shaping the growth trajectory of the global genetic analysis services market during the forecast period. Furthermore, due to advancements in technology, the scope of genetic testing has widened by a considerable margin due to which, the demand for genetic analysis services is increasing. While genetic analysis services in the past were largely time-consuming and cumbersome, at present, increasing speed and availability of genomic testing are anticipated to present a plethora of opportunities to the players involved in the current market landscape for genetic analysis services.

Request for Custom Research - https://www.transparencymarketresearch.com/sample/sample.php?flag=CR&rep_id=2555

In addition, the gradual shift in the point of access to testing is evolving, as more number of consumers can avail genetic analysis services outside the healthcare setting. Advancements in genetic medicine at the back of advancements in technology are likely to bolster the growth of the global genetic analysis services market during the assessment period.

Pre Book Genetic Analysis Services Market Report - https://www.transparencymarketresearch.com/checkout.php?rep_id=2555&ltype=S

Research and Development Activities in Full Swing amid COVID-19 Pandemic

Research and development activities are expected to continue in full swing amid the ongoing COVID-19 pandemic. The significant rise in the demand for genetic counseling services during the ongoing COVID-19 crisis is anticipated to generate consistent revenue for the players involved in the genetic analysis services market. Furthermore, researchers and scientists are increasingly focusing on discovering genetic mechanisms that are required to prevent the spread and transmission of the novel coronavirus disease. Genetic research is estimated to unlock various intricate details of the novel coronavirus, thereby opening up new opportunities for mitigation. The ongoing research pertaining to genetics and its correlation with the ongoing pandemic is expected to provide a detailed and microscopic understanding of the overall cellular mechanisms of the virus.

About Us

Transparency Market Research is a next-generation market intelligence provider, offering fact-based solutions to business leaders, consultants, and strategy professionals.

Our reports are single-point solutions for businesses to grow, evolve, and mature. Our real-time data collection methods along with ability to track more than one million high growth niche products are aligned with your aims. The detailed and proprietary statistical models used by our analysts offer insights for making right decision in the shortest span of time. For organizations that require specific but comprehensive information we offer customized solutions through ad hoc reports. These requests are delivered with the perfect combination of right sense of fact-oriented problem solving methodologies and leveraging existing data repositories.

TMR believes that unison of solutions for clients-specific problems with right methodology of research is the key to help enterprises reach right decision.

Contact

Mr. Rohit BhiseyTransparency Market Research

State Tower,

90 State Street,

Suite 700,

Albany NY - 12207

United States

USA - Canada Toll Free: 866-552-3453

Email: sales@transparencymarketresearch.com

Website: https://www.transparencymarketresearch.com/

See the rest here:

Genetic Analysis Services Market: Uptake of Next-generation Sequencing and Multi-gene Tests to Drive Market - BioSpace

FDA Clears Genetic Modification in Pigs for Biomedicine and Food – The Scientist

The US Food and Drug Administration granted approval yesterday (December 14) for a genetically modified line of pigs that marks the first time a GM animal has been given the regulatory greenlight for both therapeutic development and food consumption, the agency says in a statement. The alteration knocks out alpha-gal, a sugar molecule on the surface of cells, and could help minimize allergic reactions to pork and reduce the risk of organ rejection in transplant patients.

The move represents a a tremendous milestone for scientific innovation, FDA Commissioner Stephen Hahn says in the statement. The FDA strongly supports advancing innovative animal biotechnology products that are safe for animals, safe for people, and achieve their intended results.

Pigs with the genetic modification are known as GalSafe pigs and are made by Revivicor Inc, a subsidiary of the US biotech United Therapeutics. Research in the mid-2010s indicated that the knockout made transplants from the pigs less likely to be rejected by primate recipients.

The director of the FDAs Center for Veterinary Medicine, Steven Solomon, told reporters in a conference call yesterday that developers who want to use GalSafe pigs for therapeutic purposes will still have to seek approval for their applications. I think that people need to be careful, Solomon said, STATreports. Thats why in part, its going to require further evaluation for xenotransplantation, xenograft, or the other activities by the medical products centers and FDA.

As far as food production is concerned, the statement notes that the meat is safe for consumption by the general population, adding that Revivicor intends to sell meat from GalSafe pigs by mail order, rather than in supermarkets.

The agencys evaluation also concluded that GalSafe pigs presented low risk to the environment, with an impact that is no greater than from conventional pigs. It adds that no animal safety concerns were noted for GalSafe pigs beyond those that would be expected in well-managed, commercial swine populations.

Multiple other efforts to develop genetically engineered pigs are underway around the world, including some alterations designed to make pigs grow faster, and others aimed at making the animals more resistant to lethal viruses such as porcine reproductive and respiratory syndrome virus (PRRSV).

Excerpt from:

FDA Clears Genetic Modification in Pigs for Biomedicine and Food - The Scientist

Key Genes Related to Severe COVID-19 Infection Identified – The Scientist

More than a year after the first cases of COVID-19 were detected, researchers are still trying to understand why some people infected with the virus become critically ill, while others have little to no symptoms. Scientists have turnedto look at the genes of patients with severe COVID-19 to understand if their bodies mount an immune defense differently than healthy patients do. A genome-wide association study published in Natureon December 11 finds that variants of five key genes responsible for antiviral immunity and lung inflammation are associated with severe COVID-19.

The findings offer potential therapeutic targets to create an effective COVID-19 treatment. Our results immediately highlight which drugs should be at the top of the list for clinical testing, Kenneth Baillie, a consultant in critical care medicine and a senior research fellow at the University of Edinburgh, tells Reuters.

Baillie and his colleagues analyzed the DNA of 2,244 patients across 208 UK intensive care units and compared it to matched controls who did not have a positive COVID-19 PCR test on file. They identified eight loci where variants were more common among the intensive care patients, of which five were in genes linked to the immune systemIFNAR2, TYK2, OAS1, DPP9, and CCR2.

An analysis looking into the relationship between the activity of certain genes and severe COVID-19 pointed to a lower expression of IFNAR2, a gene that encodes a building block of a receptor for interferons, which act as emergency flares to warn the immune system of an intruder and have been a target for researchers hoping to develop a COVID-19 treatment. However, administering interferon to hospitalized COVID-19 patients did not reduce mortality, according to a large clinical trial published earlier this month.

The researchers also homed in on a possible link between severe COVID-19 and higher expression levels of TYK2and CCR2, which encode proteins used in cytokine signaling, which drives inflammation and can lead to lung injury. The anti-inflammatory drug baricitinib, typically used to treat rheumatoid arthritis, inhibits the protein encoded by the TYK2gene and has shown promising results in treating COVID-19 when paired with remdesivir, a broad spectrum antiviral that has shown limited effectiveness against the virus by itself, according to a study published December 11 in The New England Journal of Medicine.

While the Naturestudy may offer clues for the genetic underpinnings of severe COVID-19 infection, finding an effective treatment in humans may still be years away. There is no guarantee that when a gene is found, targeting that gene will result in therapeutic efficacy, Tom Hemming Karlsen, a physician at the University of Oslo who did not participate in the new work, tells The Washington Post. What genetics studies like this then do is they help us find very specific starting points for further investigation.

The authors' investigation indicated that there is a causal role for IFNAR2 and TYK2. Sara Clohisey, a research fellow at the University of Edinburgh and a coauthor of the Nature study, notes that there are likely many other factors beyond these gene variants that contribute to COVID-19 disease severity.

A chunk of the answer is in our genes, but its unlikely that a single element is fully responsible for the development of severe COVID-19, she tells the Post. Its more likely to be a combination of factors, which may include genetics as well as age, obesity, gender, and other characteristics.

Correction (December 15): A previous version of this article misinterpreted Sara Clohisey's comments about the causality of her findings.The Scientistregrets the error.

Read more:

Key Genes Related to Severe COVID-19 Infection Identified - The Scientist

UNLV Researcher on the Curious Case of COVID-19 Reinfection – UNLV NewsCenter

Once youve had COVID-19, youre safe from getting it again, right?A northern Nevada man learned the hard way earlier this year that getting re-infected with COVID-19 soon after surviving it just might be possible. After testing positive in April, the man subsequently tested negative multiple times until becoming ill just 48 days later and testing positive again in June.To figure out what was going on, scientists from the Nevada State Public Health Laboratory at the University of Nevada, Reno, along withUNLV researcher Richard Tillett with the Nevada Institute of Personalized Medicine, looked deeper into the genetic blueprint of the mans virus samples and found differences between the two cases.In other words, he was infected twice.Thankfully, reinfection looks to be pretty uncommon, says Tillett. And I hope that it remains that way.Still, the work conducted by Tillett and colleagues uncovered the first confirmed case of a person reinfected with the virus in North America. Their research was published in the prestigious medical journal The Lancet InfectiousDiseases in October.

The scientific community in Nevada and beyond has rallied together like never before to uncover the many mysteries of COVID-19.Tillett, a biostatistician, joined UNLV in August after working with the Nevada Center for Bioinformatics in Reno. He studies the genome or building blocks of disease, and his work with the Nevada Institute of Personalized Medicine is now squarely focused on finding ways to conquer COVID-19.We spoke with Tillett to learn more about the likelihood of COVID-19 reinfection, the teams landmark study, whats next, and his advice for what we can all do to keep ourselves and our loved ones safe.

My role in this study was to analyze the virus genomes collected from this patient from his earlier and later positive tests, compare the genomes to one another and to genomes in circulation, and finally, either find evidence that the two viruses differed or rule it out.

We identified the man, a Washoe County resident, as a part of our effort to survey SARS-CoV-2 genomes across Nevada. With both public health labs in the state providing samples, corresponding author Dr. Mark Pandori, director of the Nevada State Public Health Lab with the University of Nevada, Reno, was able to identify the man.

We started with a basic set of facts in the testing history. We knew that the patient had tested positive via the PCR test, which is used to diagnose active infection. He had symptoms, then improved. He subsequently had two negative tests, indicating his infection was over. Then six or seven weeks after his first positive test, he felt sick again, got tested again, and was again positive.Now, PCR test results like these are only suggestive of re-infection. In order to confirm reinfection, we needed to sequence the viral genomes from both positive tests. When we obtained those genome sequences, we found six differences between the virus in the samples, by which we were able to rule out alternative scenarios such as it being one long infection.

Honestly, I felt fright. We had to be sure we were right if we were going to claim proof that the possibility of reinfection was more than possible, but had actually happened for this person.

Currently, reinfection looks to be pretty uncommon. And I hope that it remains that way. Though we don't yet know how long an average person may have lasting immunity to SARS-CoV-2, less than a year into this pandemic, it currently looks like reinfection is not the norm.

My advice is to stay vigilant. Wear masks. Social distance. Avoid gatherings. Wash your hands. And if you feel like you maybe haven't been doing a great job at enacting these suggestions, this is too important to beat yourself up over. Remember that its not too late to start or restart lowering your exposure risk.

The research community here in Nevada, the U.S., and globe are doing great work together in response to the pandemic and have embraced collaboration and knowledge-sharing to help tackle this threat. Our hope is that continued study will help us understand this virus, track it, monitor unfolding outbreaks, and keep guard in the future, even after vaccines become widely available.

At the Nevada Institute of Personalized Medicine, my colleagues including Professor Edwin Oh and I are continuing to track and study COVID-19 genomes from Nevada patient samples.Weve begun a major effort to detect SARS-CoV-2 in wastewater, and once detected, identify the mixture of mutations found in the viruses in that wastewater. This work will give us a tremendous amount of new information on the burden and spread of this virus in the community complementary to and beyond patient samples.

Read more from the original source:

UNLV Researcher on the Curious Case of COVID-19 Reinfection - UNLV NewsCenter

Genomics and medicine it’s complicated | Health | willistonherald.com – Williston Daily Herald

In a truly grand feat of modern science, The Human Genome Project, an international collaborative effort, set out in 1990 to map our genetic makeup using diverse human genetic samples. In 2003 the project was completed, ultimately showing about 20,000 human genes. Since that projects completion, with ever-improving DNA sequencing technology, genomics researchers continue to gather more and more information about human DNA.

A single human cell contains a mind-numbing 6 billion base pairs (each base pair is one of four types of nucleic acid molecules) in its DNA, organized into 23 pairs of chromosomes. Fascinatingly, only about 1.5 percent of our DNA actually codes for proteins, while the vast remainder is noncoding DNA, serving a regulatory function or, at least as far as we understand, no function at all.

In 2007 the first individual human genome was sequenced and published. In 2008, James Watson (as in, the 1962 Nobel Prize winning Watson and Crick model of the DNA double helix) poetically had his genome sequenced and published. The ability to sequence an individual humans genome held much promise, we hoped, in regard to predicting illness and personalizing medical interventions. But in 2020, this promise remains very much unfulfilled. In most cases primary care physicians dont yet utilize genomics information in our daily practice. Why is this?

The short answer: Its complicated. In some specific instances, genetic information can clearly convey an increased risk for disease. One example of this might be the BRCA gene mutation and associated risk of future breast or ovarian cancer. Because this specific gene mutation is so tightly linked with elevated risk, testing and finding the mutation in an individual (based on their family history or known relative with a mutation) can have direct practical implications on strategies for cancer screening or even consideration of surgery to remove the at-risk tissue.

Scenarios like BRCA mutation are outliers, however. When we look to common diseases, such as cardiovascular disease or diabetes, finding genetic information useful gets, well, complicated. In these cases, what we have found is that many genes are involved, and it is extremely difficult to estimate how much a mutation in one of those genes affects overall risk. Thats not to mention all the environmental factors which may affect risk as much or more than the genetic profile.

Genomics remains a vast, new, and thus-far difficult to access specialty of medicine. At its current rate of growth, however, I am confident my previous statement will not remain true during my career in medicine.

Kelly Evans-Hullinger, M.D. is part of The Prairie Doc team of physicians and currently practices internal medicine in Brookings, South Dakota. For free and easy access to the entire Prairie Doc library, visit http://www.prairiedoc.org and follow Prairie Doc on Facebook, featuring On Call with the Prairie Doc a medical Q&A show streamed most Thursdays at 7 p.m. central.

Excerpt from:

Genomics and medicine it's complicated | Health | willistonherald.com - Williston Daily Herald

Emedgene collaborates with Illumina to scale the interpretation of genomic data for rare diseases – PRNewswire

PALO ALTO, California, Dec. 16, 2020 /PRNewswire/ --Emedgene, a leading precision medicine intelligence company, announces a non-exclusive partnership with Illumina Inc. and the integration of automated interpretation into Illumina's TruSight Software Suite for rare genetic diseases. Emedgene's Clinical Rare Disease application is one of a portfolio of artificial intelligence (AI)-powered applications for high-throughput genomic interpretation across clinical and research use cases.

Healthcare is in the midst of a leap towards precision medicine.Continued improvements in the cost of next-generation sequencing (NGS) technologies, such as those developed by Illumina, have dramatically accelerated the adoption of genetic testing. Consequently, data generated from genomics doubles every year and is expected to reach 20 exabytes by 2025. The genomic interpretation market transforms data into meaningful insights that improve patient care and impact the many different medical disciplines that incorporate genomics into routine care.

As next generation sequencing has become readily accessible throughout the globe, data interpretation of sequencing outputs has struggled to keep pace. Gaining insights can require hours of manual review. Machine learning offers the opportunity to automate and speed the process, uncover new insights and provide additional context to researchers in a shorter time.

Emedgene's Cognitive Genomics IntelligenceTM solution automatically produces insights from genomic data, reducing the time and cost of its interpretation. High-value features within Emedgene's AI solution are now integrated into Illumina's TruSight Software Suite platform. With AI-enhanced interrogation of medical literature and phenotypes associated with rare disease, researchers are positioned to make new and unexpected discoveries.

"In the future, patients will have access to the $100 genome, making it as ubiquitous as a blood test. This genomic data will be attached to an electronic medical record, and inform clinical decision making throughout our lives. Billions of patients across disease areas will benefit from faster diagnosis and better treatment. Illumina has initiated the precision medicine revolution by unlocking the power of the genome. Emedgene is on a mission to equip healthcare and lifesciences organizations challenged to practice precision medicine, by making genomic insights accessible to all. We're tremendously enthusiastic about the scale our new partnership with Illumina will enable, and the number of patients that will benefit from a genetic diagnosis," said Einat Metzer, Emedgene's Co-Founder and CEO.

"It's essential that we deliver best-in-class tertiary analysis capabilities that complement our platforms which is why we have chosen to work with Emedgene on the integration of their AI tools into TruSight Software Suite," says Ryan Taft, VP, Scientific Research at Illumina. "The combination of Illumina's comprehensive analysis of the genome with Emedgene's AI-driven insights will allow our customers to identify disease-causing variants at-scale, reducing interpretation time and benefiting individuals with rare genetic disorders across the globe."

AboutEmedgene:Emedgene is on a mission to unlock genomic insights for the world's health organizations by providing data-driven AI solutions for genomic diagnostics and discovery at scale. We work with leading healthcare and lifesciences organizations to enable precision medicine programs, enable high throughput diagnostics and accelerate the pace of research. To learn more, visit http://www.emedgene.com

Read more at http://www.emedgene.com.

SOURCE Emedgene

https://emedgene.com/

Read the original post:

Emedgene collaborates with Illumina to scale the interpretation of genomic data for rare diseases - PRNewswire

Polymerase Chain Reaction Market | Increased Outbreak of Infectious Diseases to Accentuate Demand in the Market – BioSpace

The growth of the global polymerase chain reaction market is likely to be driven by its growing utilization in various clinical diagnostics, increasing demand for precision and personalized medicine. In addition, growing application in the development of drugs and technological progress is likely to work in favor of the global polymerase chain reaction market over the timeframe of analysis, from 2019 to 2029.

In the last few years, personalized medicine has gained increasing popularity due to its ability to offer tailor-made therapies to patients. Personalized medicines come with a high margin of safety and promise of offering improved patient care and lower the overall cost of healthcare. The increasing demand for personalized medicines is likely to generate immense opportunities for the genetic medicines. There is a growing need for the development of novel technologies, which can be utilized for the purpose of expansion of the global polymerase chain reaction market. In an attempt to attain competitive edge in the rising field of personalized medicine, pharmaceutical and biotechnological companies are exploring the grounds of direct-to-consumer genetic testing and genomic medicine. These factors are likely to foster growth of the global polymerase chain reaction market over the period of assessment, from 2019 to 2029.

Get Brochure of the Report @ https://www.tmrresearch.com/sample/sample?flag=B&rep_id=7029

Product, end user, and region are the three significant parameters based on which the global polymerase chain reaction market has been categorized. The main purpose of such segmentation is to offer a lucid and detailed view of the said market.

Global Polymerase Chain Reaction Market: Notable Developments

The global polymerase chain reaction market is marked with presence of quite a few prominent players and it is considered moderately competitive market for both the existing players and new entrants. These companies are entering into partnerships, collaborations, and mergers in an effort to gain larger revenue and share of the market.

Some of the key players in the global polymerase chain reaction market comprise the below-mentioned:

Global Polymerase Chain Reaction Market: Key Trends

The below-mentioned market dynamics are estimated to characterize the global polymerase chain reaction market over the assessment period, from 2019 to 2029.

The development of the global polymerase chain reaction market is likely to gather momentum with the outbreak of Covid-19 across the globe. In an effort to facilitate growth of personalized medicine, it is essential to come up with reproducible and precise tool of gaining information about molecule about underlying conditions. Such developments are likely to guide correctly the clinical decision making abilities with a large number of tests making use of PCR techniques. All these factors are likely to pave way for growth of the global polymerase chain reaction market over the timeframe of analysis, from 2019 to 2029.

In addition to Covid-19, outbreak of infectious diseases, such as Zika has generated increased demand for PCR-based molecular assays. This technique is gaining increased importance for the purpose of identification of pathogens of these infectious diseases, which is why the demand for polymerase chain reaction is likely to rise in the years to come.

Global Polymerase Chain Reaction Market: Geographical Analysis

North America is expected to dominate the growth of the global polymerase chain reaction market over the timeframe of analysis, from 2019 to 2027. Growth of the North America is ascribed to the presence of flexible regulatory guidelines and favorable government initiatives, Asia Pacific is likely to come up as rapidly growing region in the global polymerase chain reaction market. Improvement in the healthcare infrastructure and increased expenditure on healthcare industry is expected to propel growth of the Asia Pacific market in the years to come.

Get Table of Content of the Report @ https://www.tmrresearch.com/sample/sample?flag=T&rep_id=7029

The global polymerase chain reaction market is segmented as:

Product

Application

About TMR Research

TMR Research is a premier provider of customized market research and consulting services to business entities keen on succeeding in todays supercharged economic climate. Armed with an experienced, dedicated, and dynamic team of analysts, we are redefining the way our clients conduct business by providing them with authoritative and trusted research studies in tune with the latest methodologies and market trends.

Contact:

Rohit Bhisey

TMR Research,

3739 Balboa St # 1097,

San Francisco, CA 94121

United States

Tel: +1-415-520-1050

Visit Site: https://www.tmrresearch.com/

Read the original here:

Polymerase Chain Reaction Market | Increased Outbreak of Infectious Diseases to Accentuate Demand in the Market - BioSpace