header logo image


Page 24«..1020..23242526..3040..»

Archive for the ‘Genetic medicine’ Category

Q&A:Transforming genetic medicine as the medical standard of care (Includes interview) – Digital Journal

Friday, March 20th, 2020

With rare diseases, 72 percent out of the 7000 known are genetic, and 70 percent of those start in childhood. The lack of scientific knowledge and the quality of information often delay diagnosis or lead to misdiagnosed cases, losing precious time that can be vital to find treatment before it's too late.This situation is changing with the advent of genetic medicine. an example is Emedgene's artificial intelligence software, which is the worlds first completely automated genetic interpretation platform using machine learning algorithms.Digital Journal spoke with Einat Metzer, CEO and Co-Founder of Emedgene to talk about the new genetic interpretation software.Digital Journal: How are rare diseases classified?Einat Metzer: Rare diseases defined by the number of people affected. In the U.S., any disease that affects fewer than 200,000 people is defined as rare, in Europe, its any disease affecting fewer than 1 in 2000 people.There are around 6000 known rare diseases, and that number is growing. Whats interesting to know, is that although they are each individually rare, collectively they impact over 300 million people. Those patients have a very difficult time receiving a diagnosis for their disease, and typically go through a diagnostic odyssey lasting on average 5-7 years. Its also worth noting that most rare diseases have a genetic basis, and appear in early childhood. DJ: Is sufficient funding and research invested into rare diseases? What are the factors that influence this?Metzer: There are two challenging aspects to rare diseases, the first is the identification of a rare disease, because obviously, physicians arent familiar with every disease affecting only tens or hundreds of patients worldwide. The second difficult aspect is developing treatments for diseases impacting small numbers of patients. The good news is countries and healthcare systems are increasingly recognizing the need to cover genetic testing for the identification of rare diseases. As of today, over 50% of the US population has insurance coverage for next generation sequencing. However, even insurance coverage for the tests does not entirely solve the problem. Sequencing a patients DNA is easily done, but understanding what variants in a patients genome mean is still quite challenging. Every patient has millions of harmless genetic variants, and only one disease-causing mutation. As a result, geneticists can spend hours manually reviewing hundreds of variants and looking for evidence for the disease in databases and the literature. There are fewer than 5,000 geneticists worldwide available to interpret patients genetic data, resulting in an interpretation bottleneck. Even as more patients become eligible for genetic testing, the workforce capable of diagnosing them is not growing fast enough. We estimate the worldwide capacity of interpretation is capped at roughly 2.4 million tests, less than the predicted rare disease testing volume for 2020. DJ: How can machine learning help?Metzer:Machine learning technologies can reduce the manual labor of interpretation, by offloading both the research and deep analysis tasks from geneticists. Machine learning is a buzzword, widely used, and applied to many types of solutions. Were talking about a unique application of the technology here, where we wont use a single algorithm to solve a single problem. Instead, we need to apply a set of algorithms designed to automate different aspects of the geneticists workflow. On the one hand, the geneticists work is to review thousands of data points for every patients test, and use them to come to a conclusion on the single genetic variant thats causing the disease. We can certainly apply machine learning algorithms to review those data points. But we can go a step further, and collect the data points most likely to impact their decision, and include those in our recommendations. The second labor-intensive task geneticists perform, is looking for the most up-to-date information in databases and the published literature. Thats a task well suited for Natural Language Processing, which can be used to augment existing databases with information curated from the literature. DJ: How does Emedgenes AI software work?Metzer:Emedgenes AI-powered genomic analysis platform tries to do just that, automate the labor-intensive parts of the geneticists workflow, so interpreting a patients genetic test takes less time and effort, and accuracy is not compromised. The goal is to scale the genetic testing interpretation in healthcare systems, so they can offer personalized care to a broader population. Our AI consists of dozens of different algorithms, each solving a different problem, all coming together to automate the genetic testing interpretation workflow. The platform is able to automatically identify the disease-causing variant, compile the evidence, and present it to the geneticist on the case for review. The machine learning algorithms utilize a proprietary knowledge graph that continuously incorporates new knowledge. The knowledge graph contains over 85,000 entities and 340,000 connections today, including unique information curated from the literature that has not yet made its way into public databases.DJ: What were the main challenges when developing the software?Access to large high-quality data sets is a major challenge in developing AI solutions in healthcare in general. For our supervised learning algorithms - those that require labeled data for training the algorithm - once we obtained the data, labeling was a challenge as well. The level of education required to annotate healthcare datasets is quite high.Fortunately, there are good solutions to both problems, both from the scientific and AI perspective. DJ: Are there any case studies you can share, to show the benefits of the approach?Metzer:Weve studied the accuracy of our interpretation algorithms with Baylor Genetics. In the 180-case study, our AI successfully identified the disease-causing mutation in 96% of the cases. Another of our customers, Greenwood Genetic Center, was able to reduce time spent per case by 75%, which was translated directly into shorter turn around times for patients waiting for a genetic diagnosis.

Continued here:
Q&A:Transforming genetic medicine as the medical standard of care (Includes interview) - Digital Journal

Read More...

Studying the African genome could yield new medical treatments for everyone – Genetic Literacy Project

Friday, March 20th, 2020

Dr. Segun Fatumo is a computer scientist with specialization in bioinformatics with keen interest in the genetic impact of non-communicable diseases in Africa and bioinformatics capacity building in Africa. He has been involved in various genetic projects including analysing a large-scale genomic dataset from Ugandan population. During his PhD, he was able to identified twenty-two (22) potential novel drug targets against malaria. Currently, he is an Assistant Professor at the London School of Hygiene and Tropical Medicine (University of London). Genetic Literacy Project (GLP) interviewed him to shed more light on the importance, challenges and future direction of a recent genetic research that he was a leading

As one of the lead authors of your recently published paper, what motivated this research project?

Precision medicine is fundamentally going to change healthcare. Genomic medicines is a key component of precision medicine with enormous potential to inform clinical medicine. One potential limitation to genomic medicine is the underrepresentation of African and other populations in genomics research. Previous studies have warned that a much broader range of populations should be investigated to avoid genomic medicine being of benefit merely to a privileged few. This is especially problematic, as previous studies have shown that Africa studies contribute an outsized number of associations relative to studies of similar sizes in Europeans. To demonstrate the potential of African genomes as a great resource for genomic medicine, we collected and analyzed genome-wide data from 6,407 individuals from Uganda.

What is the value of collecting more genomic data from African populations which are badly underrepresented in genomic databases.

Our findings from even modest side studies highlight the importance and usefulness of examining genetically diverse populations within Africa. Findings from large-scale studies from Africa may foster the development of new treatments that will benefit people living in Africa as well as people of African descent around the world.

What sort of challenges did you face in the study, and how did you overcame them?

So many challenges including community engagement, ethics, recruitment, etc. Globally, genomics research and specifically recruitment of participants regardless of the continent is always challenging. However, 60% of Africans live in rural areas. Prospective participants are more likely to be poor and to have limited access to healthcare and education. This means that the carrying out of research in these settings invariably presents challenges of a different order to those in higher income countries. Researchers should not exploit these challenges.

Is the value of this research project beyond Uganda and why?

Yes. Findings from our study may foster the development of new treatments that will benefit people living in Africa as well as people of African descent around the world.

What were the responses that you have received so far about the findings?

Enormous responses. I find it difficult to attend to all media requests.

What is the future direction of the research?

While there is an urgent need to perform large-scale genomic research in Africa, several ongoing initiatives such as H3Africa and the Nigerian 100K Non-Communicable Diseases Genetic Heritage Study (NCD-GHS) could provide the data to improve the evidence base and make genome medicine useful to diverse populations.

How do you see the future of genetics and bioinformatics in Africa?

I think we are now on the right track. We have established the Nigerian Bioinformatics and Genomics Network (NBGN). There are also other initiatives. We are now focusing on building capacity in Africa.

Olumide Odeyemi is a research scientist with a doctoral degree from the University of Tasmania, Australia. His areas of expertise and interest include food microbiology, microbial food safety and quality, aquaculture microbiology and research communication. Follow him on Twitter @olumide_odeyemi

Dr. Segun Fatumo is an assistant professor of genetic epidemiology and bioinformatics at the London School of Hygiene & Tropical Medicine. Follow him on Twitter @SFatumo

View original post here:
Studying the African genome could yield new medical treatments for everyone - Genetic Literacy Project

Read More...

Genetic Effects of DISC1 and G72 (DAOA) on Visual Learning of Patients | NDT – Dove Medical Press

Friday, March 20th, 2020

Jane Pei-Chen Chang,1,* Kuo-Hao Huang,1,* Chieh-Hsin Lin,2,3 Hsien-Yuan Lane1,3,4

1Department of Psychiatry & Brain Disease Research Center, China Medical University Hospital, Taichung, Taiwan; 2Kaohsiung Chang Gung Memorial Hospital, Chang Gung University College of Medicine, Kaohsiung, Taiwan; 3Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan; 4Department of Psychology, College of Medical and Health Sciences, Asia University, Taichung, Taiwan

*These authors contributed equally to this work

Correspondence: Chieh-Hsin LinDepartment of Psychiatry, Kaohsiung Chang Gung Memorial Hospital, No. 123, Dapi Road, Niaosong District, Kaohsiung 833, TaiwanTel +886-7-7317123 ext. 8753Fax +886-7-7326817Email cyndi36@gmail.com

Hsien-Yuan LaneDepartment of Psychiatry, China Medical University Hospital, No. 2, Yuh-Der Road, Taichung 404, TaiwanTel +886-4-22062121 ext. 1074Fax +886-4-2236-1230Email hylane@gmail.com

Background: Visual learning plays an important role in general populations and patients with schizophrenia. Genetic influences on visual learning remain unknown. Two functional single nucleotide polymorphisms (SNPs), Ser704Cys of the DISC1 gene and M24 (rs1421292) of the G72 gene, are strongly associated with pathogenesis and pathophysiology of schizophrenia. This study examined these two SNPs effects on visual learning in schizophrenia patients.Methods: Two hundred seventy-one patients (mean age, 37.0 years [SD = 9.3]; 159 men) with chronic schizophrenia were genotyped for the DISC1 Ser704Cys and G72 M24 SNPs and assessed for visual learning with Visual Reproduction II (delayed reproduction) of Wechsler Memory Scale III (WMS-III). For comparison, verbal learning (using Wordlist II of WMS-III) and attention (by Continuous Performance Test) were also measured.Results: The DISC1 Ser carriers excelled DISC1 Cys/Cys homozygotes in visual learning (p=0.004, effect size: 0.43), but not in other cognitive functions. G72 M24 A-allele carriers and G72 M24 T/T homozygotes performed similarly (effect size: 0.07). In SNP-SNP interaction analysis, the patients with Ser carrier_T/T had better visual learning than those with Cys/Cys_T/T (p=0.004, effect size: 0.70) and those with Cys/Cys_A-allele carrier (p=0.003, effect size: 0.65). Education had a positive effect (p=0.007), while negative symptoms had a negative effect (p< 0.001) on visual learning.Conclusion: The findings suggest that genetic variations in DISC1 Ser704Cys and G72 M24 affect visual learning in schizophrenia patients. The effect sizes of SNP-SNP interaction surpassed the sum (0.50) of effect sizes from two individual genes, suggesting synergistic DISC1-G72 interaction.

Keywords: attention, DISC1, G72, visual and verbal learning, schizophrenia

This work is published and licensed by Dove Medical Press Limited. The full terms of this license are available at https://www.dovepress.com/terms.php and incorporate the Creative Commons Attribution - Non Commercial (unported, v3.0) License.By accessing the work you hereby accept the Terms. Non-commercial uses of the work are permitted without any further permission from Dove Medical Press Limited, provided the work is properly attributed. For permission for commercial use of this work, please see paragraphs 4.2 and 5 of our Terms.

Continued here:
Genetic Effects of DISC1 and G72 (DAOA) on Visual Learning of Patients | NDT - Dove Medical Press

Read More...

The Lost Family: How genetic info is used to solve crimes – Fast Company

Friday, March 20th, 2020

In her new book The Lost Family: How DNA Testing Is Upending Who We Are, journalist Libby Copeland looks at how home DNA testing has implications for families, for law enforcement, and for how we understand our own privacy and interconnectedness. The mail-in spit kits Copeland writes about are sometimes known as recreational, to distinguish them from the genetic tests ordered by doctors, yet the technologys repercussions can be far more than recreational. This excerpt, from a chapter exploring the use of quasi-public DNA databases like GEDmatch and FamilyTreeDNA to solve violent crimes and identify culprits including the alleged Golden State Killer, explores the tension between privacy and public safety.

The use of genetic information to solve crimes, however worthy, has to be balanced with legitimate concerns on the side of protecting privacy, computational biologist and MyHeritage chief science officer Yaniv Erlich told me. The problem is one of trust. Genetic information is essential if you want to advance precision medicine, he said. We have families, some of them I know personally, that genetics was able to help, to identify the cause of serious conditions. But what if people were to stop trusting researchers to keep their information safe? If we cannot recruit tens of thousands, maybe hundreds of thousands of people for these studies, we cannot help these people, Erlich said. We cannot use the power of the genetic revolution to empower our understanding. And thats a huge missed opportunity.

This erosion of trust may already be happening. As far back as early 2019, 23andMe co-founder and CEO Anne Wojcicki said the market for her product had slowed, speculating that this might have to do with broader privacy concerns stemming from things like the Golden State Killer case and Facebooks dicey data-collection practices. Some observers have suggested that the lack of privacy protections around genetic data may disproportionately affect minorities, and I heard a number of accounts of African Americans quitting GEDmatch after it became clear that police had used it to help identify Golden State Killer suspect Joseph James DeAngelo.

And, of course, concerns about how we protect our genetic information extend beyond the context of recreational testing. Some experts worry about the increasing use by police of Rapid DNA machines, which can process samples in ninety minutes, and which they fear poses the possibility of errors and privacy violations. There are serious concerns about DNA testing on migrants. And all of this is taking place within the broader context of what legal scholar Andelka Phillips calls ever-increasing monitoring, where we are all subject to complex data mining and profiling. The problem, Phillips says, is that its difficult to imagine how data from direct-to-consumer testing may in the future be linked with other data and used in ways we cant anticipate.

It might, in theory, be used by some future totalitarian government to discriminate against its citizens. This may seem implausible, except that the day I spoke with Erlichand asked him if he could imagine a future world in which our ancestral backgrounds could be used to hurt us, and he said he didnt need to project into the future because I can go to the historyon that same day, the New York Times ran a story about how Chinese authorities were using DNA as part of a campaign of surveillance and oppression against the countrys ethnic minority Uighurs.

Journalist Kristen V. Brown has thought a lot about big data and unintended consequences. In 2015, she covered the hack of the website Ashley Madison, an online dating service geared at people who wanted to cheat on their partners. Life is short. Have an affair, the sites slogan went. Brown wanted to know how the hack and subsequent exposure of the sites users personal information had changed their lives, so she interviewed more than one hundred people impacted by it and learned about divorces, blackmail, and suicides. Its easy to assume that if you dont cheat on your spouse, you dont need to worry about this sort of thing, and that if you do cheat, well, you deserve what you get. But Brown thought the incident had broader implications. She was fascinated by a concept from Georgetown Law professor Paul Ohm that we are all at the mercy of the massive troves of data that businesses collect and keep on us, and that somewhere amid all that information, every one of us has a devastating secret. Ohm called this eventual, interconnected treasure trove of information the database of ruin, and he urged in a 2012 Harvard Business Review article, Please dont build this.

This idea stuck with Brown, and when she started covering consumer DNA testing, she saw how this new technology fit the paradigm. I was like, Oh my god, our genomes have now become databases of ruin. Its just another piece of data that can be incriminating, she says. I think were at the beginning of living in a time where you cant really keep secrets anymore. And how does that change how we go about the world and our daily lives?

How does it? Were such bad prognosticators. Historian Melvin Kranzberg once wrote that technology is neither good nor bad, nor is it neutral, by which he meant technologies play out in vastly different ways depending on the context, and that we often lack the ability to anticipate how they will change our lives.

Just over a year after Joseph James DeAngelo was arrested, the rush to what legal scholar Natalie Ram has called a de facto national DNA database abruptly slowed. After a seventy-one-year-old woman in Utah was attacked and choked into unconsciousness as she was practicing the organ in her church, GEDmatch co-founder Curtis Rogers made an exception to the sites policy that law enforcement access the database only for murders and sexual assaults, and allowed police in to help solve a case he described as as close to a homicide as you can get. In the ensuing controversy over the site unilaterally making an exception to its terms of service, GEDmatch decided to expand the definition of violent crimes that law enforcement matching could be used forand to automatically opt all its users information out of being available to law enforcement. If people wanted their genetic information used in this way, Rogers decided, they needed to proactively choose it. Overnight, the database of people that investigative genetic genealogists like CeCe Moore (working for a company called Parabon) and Barbara Rae-Venter (whod helped solve the Golden State Killer case) could access to help police narrow in on suspects went from about a million to zero. Moore called it a setback for justice.

When I spoke with Rae-Venter a few weeks after GEDmatchs decision, she told me she was hopeful that a growing opt-in movement would eventually make the database a destination for law enforcement matching again. By then tens of thousands of people had logged into GED-match to request that their genetic information be used to solve crimes. In the meantime, she said, she was still able to use the FamilyTreeDNA database for investigations. Some months later, FamilyTreeDNA announced a new investigative genetic genealogy unit to rival the one Moore was heading at Parabon, this one headed by Rae-Venter. And then the New York Times reported that a Florida detective had obtained a warrant giving him permission to search all of GEDmatchs database, including data from the majority of users who hadnt consented to being involved in criminal investigations. The judges decision to grant this order was a game-changer, experts told the paper; it would likely embolden other agencies to seek similar warrants for huge companies like Ancestry and 23andMe, potentially turning all genetic databases into law enforcement databases.

DNA revelations mean that an alleged sadistic serial murderer named Joseph James DeAngelo is behind bars, but they also mean that you could be implicated in the arrest of a relative. It means that an adoptee can find her parents, and it also means that a family can find out that decades before, a man cheated on his wife and produced a childwhich could be good for that child and bad for that wife and a mixed blessing for that man. Perhaps consumer genomics means we are all on guard waiting for the other shoe to drop, or perhaps it means that we are all forced to be more honest with each other. One of the central conundrums of spitting into a tube is the way one persons rights so often collide with anothers after the tube is sealed and sent in.

Excerpt from the new book The Lost Family: How DNA Testing Is Upending Who We Areby Libby Copeland by Abrams Press 2020 Libby Copeland

Read the original here:
The Lost Family: How genetic info is used to solve crimes - Fast Company

Read More...

Staying ahead in the era of precision medicine – PMLiVE

Friday, March 20th, 2020

The potential for precision medicine is evident. It is one of the most exciting things happening in the healthcare sector. Science and technology advances have made precision medicine no longer an obsession or a dream, its become a reality and will become the mainstream in the future of healthcare, according to our advisory board Co-Chair Dr Zhen Su, Head of Merck KGaAs global oncology franchise. This notion that no one size fits all has long been an interest and passion for many healthcare providers.

This potential is reinforced by Dr Martin Murphy, CEO Roundtable on Cancer: Simply put, it means the future of anybody (with cancer) is embodied in the development of more precise means of diagnoses and then properly treating those with the disease. Its revolutionary and its evolutionary.

While it is most advanced in oncology, precision medicine has wider, exciting applications beyond oncology and late-stage disease, such as rheumatology, and rare and genetic diseases. We are also seeing many innovative initiatives and partnerships develop in this field. For example, Lung-MAP, a large-scale umbrella trial in non-small cell lung cancer, was one of the first to involve partnerships between several groups including the FDA, NCI, advocacy groups and the pharmaceutical industry. At

the same time, we are seeing companies like Flatiron Health using real-world data to learn from patients who are not in clinical trials.There are also important advances in the sharing of data, eg Project Data Sphere, which is an important platform that provides researchers with open-access and analytic tools to use when working with data sets from large clinical trials.

Technology is also moving and being applied at pace: wearable devices such as smart watches, computerised pill bottles and other tools are being evaluated as potentially robust sources of patient-reported data. There are also exciting and much-needed opportunities for patients to play an increasing role in the future, eg patients providing valuable insights into the design of clinical trials will lead to secondary endpoints that are most meaningful for patients themselves rather than for the researchers.

...But what are the barriers?

The challenges around precision medicine are illustrated perfectly by looking at lung cancer as an example: treatments such as chemotherapy and radiation were once fairly standard and broadly applied without much distinction between different patients. However, what was once homogeneous has now become heterogeneous.

Different tumour subtypes have been identified, and exciting advances such as next-generation sequencing (NGS) of tumour DNA can more precisely define key characteristics of an individuals cancer.

Suddenly, one treatment option turns into many, and physicians, particularly those at the community level, struggle to keep up and find it difficult to choose or sequence options.

In addition to new technologies such as NGS, we are seeing the advent of novel trial designs (such as basket trials), and many practising physicians may not understand how to apply or contextualise the resulting data. In addition, the ways that clinical trial data and real-world data complement each other may not be clear, not only to physicians but also to regulatory authorities.

As expressed by Professor George Demetri of the Dana- Farber Cancer Institute and Harvard Medical School: Theres a remarkable need for better communications, both about and around the issues in precision medicine. Because the field is moving so fast, physicians who trained two years ago may not know whats happening today. They have to keep up. Communicating this is almost impossible through standard medical education channels.

Alongside these challenges, there are other barriers to contend with. A lack of public awareness and education may hinder trial recruitment, as patients may not understand the role of trials and may view getting the standard therapy as undesirable.

Additional barriers to participation in clinical trials may also include the lack of a centralised system to match patients to trials and geographic disparities (eg urban versus rural). Beyond the trial setting, perceptions about the efficacy of a drug and logistical barriers such as the lack of technical expertise or centralised technology, and the lack of coverage by insurance companies can limit the use of new therapies in clinical practice.

Finally, patient privacy and ethical considerations become more acute as greater volumes of personal genetic data are generated through clinical evaluations.

Call to action: what can healthcare communication specialists do to help?

Our advisors all felt passionately that we will only overcome the barriers by developing and supporting communication and education efforts to help patients (and expert caregivers, including physicians, nurses and others) to understand precision medicine and the role of clinical trials in healthcare.

The top ten specific needs identified by our advisors include:

Whats next for the McCann Health Global Scientific Council in precision medicine?

We are committed to continuing scientific exchange and public dialogue in this important area of medicine. As outlined by our Global Medical Director and Co-Chair of our Advisory Board, Dan Carucci: Precision medicine is the future of medicine and we as professional health communicators have to stay at the cutting edge so we can help our clients navigate this difficult space.

With the ongoing support of our advisors, we will continue to communicate that precision medicine is integral to healthcare reform. Dr Su is clear: There is an enormous need around how we communicate in a clean, clear and trustworthy manner to enable (physicians) to master new treatments and new technology to better select patients and better provide treatment.

Dr Donna Graham, Medical Oncology Consultant at The Christie NHS Foundation Trust is in agreement with this: Communication between oncologists and their patients is absolutely critical. There are so many complex issues for patients to grasp in terms of treatments that are available and how certain genomic abnormalities may affect their cancer, their prognosis and their outcomes. Communications companies can really help to try to bridge that gap.

In line with our mission to make a meaningful difference in healthcare communications and ultimately to patients, we plan to reconveneour advisors in 2020.

Our aim is to ensure that all our internal team members strengthen their precision medicine expertise, while focusing on the development of external and peer-reviewed communications and tangible tools to benefit the wider medical and patient community. Our call to action will continue as we move to improve the understanding, application and opportunities of precision medicine.

Read the original here:
Staying ahead in the era of precision medicine - PMLiVE

Read More...

Is Hydroxychloroquine the Answer to the Coronavirus Pandemic? Inside the Race to Find A COVID-19 Cure – Newsweek

Friday, March 20th, 2020

Long before the coronavirus began to spread beyond China, infectious disease experts around the world knew there was ample reason to fear it. Not only was the pathogen highly contagious and lethal, it was also newscientists had written no medical papers on it, doctors had no vaccines or pills to give their patients. The most effective tools we have, at the moment, are public health measures out of the 19th century such as quarantines and social distancing.

The emergence of Severe Acute Respiratory Syndrome Coronavirus 2019, or SARS CoV-2, has made plain our vulnerability to a novel pathogen. An estimated 160 million to 214 million people in the United States could be infected over the course of the epidemic, by some estimates. Fatalities could run from 200,000 to 1.7 million people, according to the CDC, and into the tens of millions worldwide.

The lack of treatments is a startling contrast to the sophistication of current medical science, which is in something of a golden age of genomics, machine learning and big data. The coronavirus has caught us flat-footed. Yet, at the same time, it has underscored how far the tools of medicine have evolved in recent years. Just days after local infectious disease experts sent virus samples taken from two patients infected with a suspicious form or pneumonia to the Wuhan Institute of Virology, a world-renowned research laboratory, for analysis, scientists had sequenced the newly emergent pathogen's RNA and uploaded its entire 30,000-nucleotide genetic code to the cloud.

Across the globe, scientists downloaded it and began to isolate antibodies. Virologists and computational biologists used machine learning tools to analyze its structure and search for existing drugs that might work against it. Pathologists applied the tools of molecular biology to search for vulnerabilities in the virus' armor of protein. "The pace of the scientific research has been really at a breathtaking speed," says Angela Rasmussen, a virologist and research scientist at Columbia University. "It's unprecedented."

As the caseload continues to rise exponentially in the U.S. and other parts of the world, scientists are racing to find antiviral drugs that are effective in alleviating the worst ravages of the disease, a devastating pneumonia that affects an alarmingly high number of patients. The goal is to give doctors a broader range of weapons in the weeks and months ahead, and save lives.

Rapid response

In recent years, technologies that allow rapid sequencing of genetic material have become standard equipment in most top research laboratories. Because of these tools, scientists were able to state with relative confidence that the current virus is closely related to the SARS coronavirus that hit in 2003, as well as very closely related to a bat coronavirus found in a cave in Yunnan, China, back in 2017. With this knowledge, scientists dusted off the files from that outbreak and picked up where other scientists left off.

Rapid genome sequencing didn't merely allow researchers to publish the full SARS-CoV-2 sequence in days, as opposed to months in the case of the SARS genome in 2003. It also allowed scientists to sequence strains of the virus in Washington State, New York City, Italy and other parts of the world, which they are using to piece together a kind of SARS-CoV-2 ancestry registrya detailed map of how the virus spread and mutated.

Scientists used this information to trace the progress of the virus and estimate how many people have been exposed in any given area, which informed the public health response. "We know from sequencing some of the more recent Seattle viruses, that those viruses were probably derived from the first patient who came to the U.S. with coronavirus in mid-January," says Rasmussen, who noted at the time that the Seattle area had an estimated 6,500 cases.

Tracking the virus in this way helped public health workers conclude early on that the virus was unusually contagious, which informed emergency planning in China, Italy and elsewhere. The most urgent task, of course, is to keep intensive-care wards from being overwhelmed by patients in respiratory distress. For the most critically ill patients, COVID-19 attacks the lungs, triggering the immune system to create a thick soup of white blood cells and other immune agents that flood the lungs. In the most severe cases, this immune response clogs up air cavities critical for transferring oxygen from the air to the body, greatly reducing lung capacity.

To survive, these patients require mechanical ventilators, which can force higher concentrations of oxygen into the parts of the lungs that are still functioning, allowing them to rest, recover and preserve precious energy needed to outlast the viral attack. But ventilators are in dangerously short supply. For instance, fewer than one-tenth of the 925,000 hospital beds in the U.S. are equipped for critically ill patients, who could number between 2.4 million to 21 million people in the United States, say estimates.

Antiviral medication could shorten the time patients need to be on ventilatorsand perhaps prevent many of them from needing one in the first place. One of the most promising ideas is to develop new drugs that can attenuate the immune response to keep the lungs functioning adequately. Doctors in the First Affiliated Hospital of University of Science and Technology of China (Anhui Provincial Hospital) and Anhui Fuyang Second People's Hospital In China used , a drug developed by Chugai, a Japanese company, originally to treat rheumatoid arthritis, on 21 critically ill patients.

Within a few days, the fever returned to normal and all other symptoms improved "remarkably," according to a paper on the study published soon after. Fifteen of the 21 patients had lowered their oxygen intake and one patient needed no oxygen therapy. CT scans revealed that the lung function improved in 19 patients of the 21 patients, and the abnormal percentage of white blood cells found in 17 of the 21 patients before treatment returned to normal in 10 within five days. Nineteen of those treated had been released within two weeks, and the other two were reported to be "recovering well."

Read more

Read more

Regeneron Pharmaceutical, a firm based in Tarrytown, New York, believes its rheumatoid arthritis drug kevzara would be similarly effective in treating critically ill patients. The drug consists of antibodies that bind to and inactivate the tiny protein molecules on the surface of the body's immune cells known as interleukin 6 that play a role in amplifying an immune response.

"People are dying because they are losing the ability to breathebecause their lungs are filling with inflammation," says George D. Yancopoulos, Regeneron's President and Chief Scientific Officer. "That's what's happening. That's a fact. The question is, what's causing the inflammation? If you shut that off, basically the lungs calm down, the cells leave the lung and they are also not making all this bad stuff."

Regeneron is currently talking with the FDA and the U.S. Department of Health and Human Services about fast-tracking clinical trials. It is enrolling 400 patients hospitalized for COVID-19. If all goes well, it could reach a verdict on the treatment in the next month or two. The company already has produced enough of the medicine to treat tens of thousands of severely affected patients, says Yancopoulos. Sanofi, which has the license to distribute the drug outside the U.S., is initiating similar trials in Europe.

Regeneron is also looking into using novel monoclonal antibodies as a potential weapon against COVID-19. These are custom-made proteins designed by the immune system specifically to bind to and neutralize the virus. Regeneron is using mice that have been genetically engineered to produce antibodies that could be used in the human body. The company has already exposed these "VelociMice" to the COVID-19 virus and extracted thousands of antibodies, and is now screening them for potential effectiveness against COVID-19, and identified a few of the most potent antibodies. It will then mass-produce them by growing them inside cell lines incubated in huge "bioreactors," engineered to promote maximum reproduction.

Christos Kyratsous, Regeneron's vice president for research, says it will take about four months to go from picking the most potent antibodies to producing enough cells to provide the tens of thousands of liters of medicine needed to make the drug widely available to those suffering from COVID-19 in the U.S., leading to hopes by some on the front lines that a new custom-made medication could be in place by the end of August.

Other experimental efforts are aimed at helping patients fight off the infection itself. In mid-March, immunologists and medical professionals at Johns Hopkins University submitted plans to the university's institutional review board and the FDA to extract antibodies from the blood of patients who have already recovered from COVID-19, says Arturo Casadevall, an immunologist and infectious disease expert at Johns Hopkins School of Medicine. The idea is to infuse new patients with antibodies filtered out of the blood of patients who have already successfully fought off the infection.

Doctors facing pandemics have used a similar strategy to combat infectious diseases for more than a century, including the 1918 flu. But this time, the approach has a modern twist. Casadevall and his colleagues plan to rely on methods and equipment that hospitals already have in place in blood banks, such as machines that currently remove antibodies from the blood of patients with autoimmune diseases, to prevent their bodies from attacking their own cells. (The blood is usually reinfused into their bodies to prevent anemia). These same machines could be used instead to extract antibodies from COVID-19 survivors. Scientists would test the antibodies to find the most potent ones and then administer them to sick patients or medical personnel in need of protection. This method could be deployed in cities around the nation, or around the globeanywhere where blood baking facilities exist. And Casadevall says he has been in contact with health officials at the Mayo Clinic, in New York City and elsewhere, who are considering taking similar measures.

Although the approach would not immediately yield a drug that could be mass produced, it could serve as a stop-gap treatment, he says, until new drugs, like those being developed by Regeneron, come online. "We can put this in place and we can provide people something more than a respirator to provide oxygen," says Casadevall.

A team that included Hopkins infectious disease experts, blood-banking officials and regulatory personnel has been holding regular conference calls. The team is now testing blood samples and developing a plan to deploy the approach throughout Baltimore. He expects that the first filtered antibodies could be fielded by the beginning of April, in time for a "second wave" of patients to hit the hospitals. The approach, already in use in China, could become widespread in the U.S.

The kitchen sink

Doctors on the front lines of the battle in China, Italy and elsewhere have identified other potential treatments by taking a "kitchen sink" approach that uses every available tool to defeat the virus. Because the outbreak is so recent, solid data isn't available on these kinds of measures, but doctors have given favorable anecdotal reports and have administered scores of ad-hoc trials.

The most promising and widely discussed is remdesivir, a broad-spectrum antiviral drug produced by Gilead. Developed originally to treat Ebola patients, remdesivir works by blocking an enzyme that is crucial for the ability of the viruses to reproduce. The drug did not prove effective for Ebola, but trials demonstrated that it did not have serious side effects. Subsequent studies on non-human primates suggest that the drug is effective against coronavirusesspecifically, Middle East Respiratory Syndrome, or MERswhich has given some public health officials cause for optimism.

"There's only one drug right now that we think may have real efficacy, and that's remdesivir," said Bruce Aylward, a senior advisor and international leader of the World Health Organization's joint mission to China, at a Feb. 24 press conference.

Clinical trials to test the drug are already underway in the U.S. and in China's Hubai province. Preliminary results from the first of those studies are expected as soon as April, says Gilead. Gilead is also in the process of enrolling about 1,000 patients, mostly in counties that have already had high numbers of diagnosed cases, in a trial to evaluate the drug given intravenously.So-called protease inhibitors have also emerged as potential candidates to treat COVID-19. These antiviral drugs, developed during the HIV/AIDS crisis, act on the enzyme protease, which plays a vital role in the ability of HIV to replicate inside the cells that it infects (it chops up big protein molecules into smaller ones). By inhibiting the action of protease, the drugs prevent the progress of an HIV infection, keeping AIDS from developing. Since then, researchers have also developed modified protease inhibitors to fight hepatitis C and other viruses.Coronaviruses like SARS-CoV-2 also use a type of protease during replication, but the virus is different enough that HIV antivirals may not be effective. Research is ongoing to find out.

The antimalarial drug chloroquine, and its derivative, hydroxychloroquine, are also candidates for COVID-19 treatments. Researchers first began testing their ability to halt the spread of viruses during the battle against AIDS. The drugs are designed to interfere with "endocytosis," the process by which a virus or other microbe enters a cell. They have since been shown to have some success in the lab against a wide range of viral diseases including the common cold and the SARS virus. On March 16, Chinese public health officials announced that a clinical trial at 10 hospitals in Beijing, Guangdong and Hunan Provinces involving more than 100 patients showed a positive effectpatients who took chloroquine were more likely to show a reduction in fever, showed clearer lungs on CT scans and reduced the amount of time to recover.More treatments will emerge as doctors and scientists on the front lines continue to try new drugs. For instance, in March, a Chinese official said that the drug favipiravir, developed by Fujifilm Toyama Chemical as an influenza drug, showed positive results for COVID-19 patients in trials in Wuhan and Shenzen.

Scaling up

There are many obstacles to getting a treatment out of the lab and into the hospital. First, clinical trials must show that the drugs work safely, and many drugs typically fail this test. A cocktail of the HIV drugs lopinavir and ritonavir, which were being tested in China, was reported to have no benefit to patients. The effectiveness of HIV drugs against COVID-19 remains largely anecdotal and unproven. And choloquine in high doses can prove toxic.

Once a drug is proved safe and effective, getting it to millions of patients around the world requires a massive manufacturing capacity. Ramping up can take months, says Prashant Yadav, a visiting fellow at the Center for Global Development and an expert on healthcare supply chains. For instance, he estimates it would likely take six months to a year to sufficiently ramp up production to meet the potential global demand for remdesivir, should it prove effective and safe.Given the urgent need for new drugs around the world, some public health officials have called for new protocols to determine who will decide how to allocate limited supply. There would have to be a way of coordinating the supply of drugs, with clear roles and responsibilities for fast-tracking treatments. This would involve an unprecedented level of coordination among the World Health Organization, organizations that finance global health measures, supply-chain experts in the pharmaceutical companies and governments. Once a country has obtained a drug, the government together with private health care organizations and drug companies have to fast-track distribution of the drugs.

"Can governments and global agencies make extremely fast decisions in the complex and somewhat uncertain environment?" asks Yadav. "How do we run a supply system so that every hospital that orders it can get sufficient supply? It's a capacity rationing problem: someone has to decide how much of demand will we need for existing supply. And as we know, rationing decisions bring out the worst in terms of global coordination and local and national politics. And if a company has never sold much in Africa then they will have to start from scratch."

Long-term fix

Anti-viral treatments can hopefully keep people from dying from COVID-19, but the best long-term hope to control the disease is a vaccine. The typical timeline for vaccine development is 12 to 18 months. The most promising and advanced is mRNA1273, which is being developed by Moderna, a Boston company. In mid-March, Kaiser Permanente Washington Health Research Institute began a safety and dosing trial in which 45 young, healthy volunteers will receive different doses of the vaccine.

Other efforts include INO-4800, a vaccine being developed by Pennsylvania-based Inovio Pharmaceuticals; a vaccine based on previous work against the Avian coronavirus from MIGAL Research Institute in Israel; a company called Heat Biologics, which already has a cancer vaccine in clinical trials, as well as efforts in early stages from Johnson and Johnson, Pfizer and GSK.Few doubt that at least some of these efforts, and many others like them, will eventually result in effective treatments. How long that will take depends on a lot of hard work and some luck. "Against all odds, we figured out to mass produce penicillin, we beat polio and smallpox," Dr. Peter Jay Hotez professor and dean of the National School of Tropical Medicine at Baylor College of Medicine in Houston. "It's unfortunate we have to wait until things got so dire to focus on the needs of the world, but I think we are there now."

Correction, 3/20/2020, 10:34 am: This story was changed to reflect the fact that Regeneron trials for Kevzara are starting with 400 patients, not a few as previously reported.

Correction, 3/20/2020, 1:35 pm: The drug chloroquine works by interfering with endocytosis, the process by which a virus enters a cell, not exocytosis as previously reported; the story has been modified to correct the error.

Correction, 3/20/2020, 2:32 pm: The COVID-19 virus has 30,000 nucleotides of RNA, not 3 billion of DNA as previously reported; this has been corrected in the story.

Read this article:
Is Hydroxychloroquine the Answer to the Coronavirus Pandemic? Inside the Race to Find A COVID-19 Cure - Newsweek

Read More...

Once and for All, the New Coronavirus Was Not Made in a Lab – VICE

Friday, March 20th, 2020

On January 30, Arkansas senator Tom Cotton tweeted that although he didnt know where the novel coronavirus, SARS-CoV-2, originated, he wanted to point out that "Wuhan has Chinas only biosafety level-four super laboratory that works with the worlds most deadly pathogens to include, yes, coronavirus.

He was implying, as other conspiracy theorists have continued to do, that the new virus was intentionally made and released (ignoring the fact there are many "level-four super laboratories" around the world). Politicians, including Donald Trump, continue to call SARS-CoV-2 the Chinese virus or Wuhan Virus.

But the origins of viruses, and how they come to infect humans, are almost always more complicated than a couple of evil geniuses secretly creating infectious killers. Viruses mutate constantly, playing the equivalent of a genetic slot machine until they chance upon the winning genetic sequences to allow them to jump from animals into humans, and be infectious enough to spread their genetic information far and wide.

While theres still so much uncertainty about the COVID-19 pandemichow long it will last, what treatment options might work, when a vaccine will arrive, there are actually a lot of things that we do know about the virus, through genetics and structural biologyincluding where it came from.

In a new paper in Nature Medicine, scientists from the U.S., Australia, and the U.K. analyzed the virus closelyoutlining its likely origins and exactly how and why it seems to be more infectious than previous coronaviruses, like SARS or MERS. Their research also provides a resolution to the made-in-a-lab speculation. Kristian Andersen, an associate professor of immunology and microbiology at Scripps Research, said in a press release: We can firmly determine that SARS-CoV-2 originated through natural processes."

You can tell a lot about a virus from its genomewhat kind of virus it is, and how closely related it is to viruses you already know about. The genetic sequence to SARS-CoV-2 was made available very quickly by Chinese scientists, allowing researchers all over the world to see what made the new virus tick.

Coronaviruses get their name from spikes that cover their surface that look a bit like the suns corona. Those spikes are key to how the virus infects a human cell and two integral parts of the new viruss spikes are slightly different from those of previous coronaviruses. These differences could help us understand how its spreading farther and infecting more people.

The first change is in the receptor-binding domain. This is part of the spike that binds to a human cellfor this virus it attaches to ACE-2, a cell membrane enzyme that regulates blood pressure.

We already knew that the 2003 SARS binding could bind pretty tightly to ACE-2. But in a Science paper this month, researchers using cryo-electron microscopy revealed that the new viruss spike was better. It binds to the ACE-2 receptor 10 times more tightly than a SARS virus does. This difference is made possible by small variations in structure to the receptor binding domain.

The second adaptation in the new coronavirus is a part of the spike called the cleavage site. The spike is a folded up-bundle, and it has to be cleaved, or cut, in a specific place to pop open, like releasing a spring. After being cleaved, the spike is used to grab onto a human cell.

The cleavage site is made up of small amino acid sequences that are recognized by cellular enzymes called proteases, enlisted to do the cutting. These molecular scissors are different for each coronavirus. SARS-CoV-2's cleavage site is made of amino acids that attract an enzyme called furin. Our bodies make furin in a lot of different tissues, but in particular, in the upper respiratory and lower respiratory tract.

When a virus has a cleavage site that attracts furin to do the cutting, it becomes more dangerous. To compare, influenza viruses are often cleaved and activated by enzymes called trypsin, "which are typically restricted to certain tissues and organs," said Jean Millet, a microbiologist at the Molecular Virology and Immunology unit of INRAE, located in France, who wasn't involved in the paper.

Experiments with avian influenza virus have shown that if they evolve a furin cleavage site, they become much more infectious. Having a furin cleavage site means that the virus is able to replicate more, and in different tissues. It can easily go into the lower respiratory tract. It may be one of the reasons that people develop pneumoniathough this hasnt been proven for certain.

Seeing that in the new SARS-CoV-2 when those sequences came out for the first time actually kept me up all night, said Bob Garry, an assistant professor of microbiology and immunology at Tulane University School of Medicine and co-author of the Nature paper.

These changes may be alarming, but they're also how we know this virus wasn't designed in the lab. Simply put, the adaptationsspecifically the binding to ACE2are just too good for a human to have come up with it.

Computer programs that scientists use to model the interactions between a virus's spike and ACE-2 dont predict that the receptor SARS-CoV-2 has would work very well. And yet, it doesas Wrapp found, 10 times better. Its an indication that the alterations in the binding were selected for through natural selection, not genetic engineering.

You couldn't predict that with any computer program, Garry said. Nature usually is better at doing things than we can figure out with a computer these days. That's pretty good evidence that this virus did evolve to bind to human ACE-2 on its own. Nobody helped it. If somebody had designed it, they would have used a different solution.

I asked if there was any possibility some evil-genius person out there, with a different computer algorithm, could have come up with it. Like in the comic books? It doesnt seem likely, Garry said.

"This is a convincing argument," Millet confirmed. "SARS-CoV and SARS-CoV-2 do bind the same receptor but they do so in different ways that is most likely through an evolutionary process whereby each virus has 'figured out' different ways to do so...This goes against the notion that someone or a group would have intentionally used the SARS-CoV sequence to generate a new virus."

Additionally, if someone wanted to make a coronavirus, they would use another virus as a building block, Garry said. But the virus that is closest to SARS-CoV-2 is a bat virus that wasnt discovered until after the outbreak. There's no evidence from the SARS-CoV-2 genome that any other virus was used as a backbone to make something new.

Viruses mutate at a steady rate, and so as they spread, researchers can look at how many adaptations they've acquired and count back in time to figure out when it appeared. Co-author Andrew Rambaut, professor of Molecular Evolution at University of Edinburgh, did this, and found that SARS-CoV-2 sprung up in humans in either late November or early Decemberwhich makes sense given it was December 31 that Chinese authorities told the World Health Organization about the outbreak.

That's the "when," but it doesnt tell us where exactly it came from. In the SARS epidemic from the early 2000s, the virus transferred directly from a civet cat to humans. It didnt have to adapt, Garry said. It was already good to go. With MERS, it was a similar storythe same virus that infected camels got passed to humans.

SARS and MERS didnt transmit between people as well as the new coronavirus does. That could be because SARS-CoV-2 has adapted more to humansmeaning it didnt just jump from an animal, but first adapted to infect us better. I could be proven wrong tomorrow," Garry said. "Somebody could find an animal out there that has a virus that's identical to SARS-CoV-2. I don't think that's going to happen."

The closest virus to the new coronavirus is a bat virus, RaTG13, which is 96 percent similar. Yet its missing one crucial thing: its spike has a different receptor binding domain, not the defining one that SARS-CoV-2 has. Intriguingly, another recently discovered virus, from the pangolin, a scaly anteater, is less like SARS-CoV-2 overall, but does have a strongly similar receptor binding domain.

Garry said that because of this, he and his co-authors think its possible that SARS-CoV-2 is a recombinant virus, meaning its a combination of two different viruses that shared their genetic information. This is like a couple moving in together and combining their kitchen appliances. Suddenly they have access to tools they didnt beforea Vitamix and a food processor. A coronavirus might have been able to gain the enhanced receptor binding and then mutated further until genetic luck brought it the furin cleavage site.

What we dont know is the specifics of where or when this recombination and other mutation occurred. It could have happened while the virus was still in an animalthen, after the furin cleavage adaptation, it was able to jump into humans and spread rapidly afterwards. Another possibility is that a previous non-pathogenic version of the virus was circulating in people for some time before the mutation at the cleavage site occurred and it started spreading rapidly. What we can say is that it's more complicated than just a "Chinese" or "Wuhan" disease. It's a virus that has changed and mutated many times, possibly from different animal sources, or within our own bodies, and with genetic good fortune, happened upon the right adaptations to take hold.

We wont know the virus's origins for sure until we have more data, but the answer could be a predictor of whats to come. If SARS-CoV-2 achieved its adaptations in animals, there's more of a risk for future similar outbreaks. If it adapted while already in humans, it's less likely those same mutations will happen againjust based on probability. Either way, we learn more about the many ways viruses make it into our lives.

The significance is that now we know that there's a new way you can get a pathogenic coronavirus through recombination," Garry said. "Spread or passage doesn't have to be a direct jump from an animal."

Figuring out the origins of SARS-CoV-2 and how it works will be important the next time another new coronavirus emerges.

If we can understand what types of coronaviruseswith what types of featuresare in animals now, it would make it easier to look at a virus's genome sequence and determine where it got its features from, or how its spike might bind to our cells. One way to do this would be to start gathering information about the coronaviruses that are in many kinds of animals now. Bat coronaviruses, for example, are incredibly under-sampled, Garry said. We know that the diversity of coronaviruses in bats is a lot more than what we know about right now. Just figuring that out would be important.

Sign up for our newsletter to get the best of VICE delivered to your inbox daily.

Follow Shayla Love on Twitter.

Go here to read the rest:
Once and for All, the New Coronavirus Was Not Made in a Lab - VICE

Read More...

What the Coronavirus Does to the Body – Discover Magazine

Friday, March 20th, 2020

As the novel coronavirus infection known as COVID-19 continues to spread across the world the number of confirmed cases in the U.S. crossed 15,000 on Friday governments have made incredible efforts to limit the pandemics overall reach.

Yet there is also much uncertainty, and a fair amount of unscientific speculation, about the virus and its effects on peoples bodies. And some of COVID-19s reported symptoms, like fever, cough and shortness of breath, overlap with those of everyday illnesses like strep throat, flu and the common cold.

Carl Fichtenbaum, an infectious disease specialist and professor of clinical medicine at the University of Cincinnati College of Medicine, says theres still much that scientists dont understand about how exactly this virus causes problems. Its very new, and were still trying to unravel it a little bit, he says.

Heres what some researchers and clinicians have learned so far about what the COVID-19 infection does to the body.

The virus that causes COVID-19 is transmitted through tiny, invisible droplets sent into the air when someone already infected coughs or sneezes. Those droplets can then be taken in by people nearby or land on surfaces that others touch, potentially passing viral particles from their hands to their eyes, nose or mouth. Generally, a person will either get it on their hands or occasionally inhale it in their mouth or nose, says Fichtenbaum.

Once inhaled into the back of your throat and nasal passages, the viral particles bind to a type of receptor on the surface of cells. These particles are studded with jagged proteins shaped like spikes, which Fichtenbaum describes as a key capable of opening the locked door of the cell receptor. Those proteins attach to the receptors and the virus is able to begin the process of getting inside and replicating, says Fichtenbaum.

Like any other life form, it just wants to survive, he says.

In order to do that, the virus needs to first copy itself. Once attached to cells, it spills its genetic material, or RNA, inside. Afterward, the virus takes over the cells metabolism to create replication factories to make more copies of its RNA. Its essentially stealing resources from the host cell, says Robert Kirchdoerfer, a biochemist at the University of Wisconsin-Madison who studies coronaviruses.

As the virus multiplies, it prompts an immune response in the body. [The immune system] says, We dont like this thing and we want to get rid of it, says Fichtenbaum. Once that battle occurs, he continues, people start to develop symptoms as previously healthy tissue becomes damaged and inflamed. These symptoms include a sore throat, runny nose, sneezing, coughing and, sometimes, fever. If the virus passes low enough and gets into our lungs, we can develop pneumonia, which leads to shortness of breath and chest pain, adds Fichtenbaum.

For the bulk of the population, explains Fichtenbaum, this period of injury will be followed by a recovery period. Most people will get better from it, he says. At the same time, the Centers for Disease Control and Prevention cautions that older adults and those with serious, preexisting medical conditions like heart disease, diabetes and lung disease are at greater risk of becoming severely ill if they become infected.

Thats not to say that everyone with those problems is going to have a bad time, adds Fichtenbaum. Its just that theyre more likely than an otherwise healthy person to have a worse case.

The lungs arent the only part of the body that can be affected by the virus. Fichtenbaum says that in some people, the infection can cause the heart to beat at irregular intervals and pump less powerfully, potentially leading to heart failure. Sometimes people can have neurologic problems [like] dizziness or weakness in an arm or a leg, he says. And some confusion can occur because our brain is just not functioning as well as it should be.

Because the virus can be swallowed, it can also infect cells in our gut. Since the outbreak of the virus last December, digestive problems have been a common complaint among those infected. The CDC reported that genetic material from the virus has been found in blood and stool samples.

And new research suggests that diarrhea and other gastrointestinal problems could be among COVID-19s first signs. According to a study published Thursday in the American Journal of Gastroenterology, nearly half of the coronavirus patients involved came to the hospital with digestive symptoms as their chief complaint. The study authors looked at data from 204 patients in Chinas Hubei province, where the outbreak originated, and found that 99 of them had symptoms such as diarrhea, vomiting and abdominal pain.

But these are still early days for research on this new coronavirus. Theres still things for us to learn, says Fichtenbaum. And, of course, wed like to understand and learn how to treat it when it is like a more serious case.

See the original post:
What the Coronavirus Does to the Body - Discover Magazine

Read More...

Sequencing.com Launches Free Genetic Analysis To Assess Risk And Severity Of Coronavirus Infection For Each Individual – P&T Community

Friday, March 20th, 2020

LOS ANGELES, March 19, 2020 /PRNewswire/ -- Sequencing.com, the world's largest online marketplace for DNA services, today launched a first-of-its-kind genetic analysis called the Coronavirus DNA Health Report that can quickly assess each user's individual risk of coronavirus infection and the likely severity of reaction to the virus, including the risks of serious illness or death.

Dr. Brandon Colby, Founder of Sequencing.com, said, "The coronavirus outbreak is not the first public health crisis our global society has faced in recent decades, but for the first time ever, through our Coronavirus DNA Health Report, genetic diagnostics and analysis are available that enable each individual to determine his or her likely level of infection risk and the severity of outcomes for the infected. From there, our Coronavirus DNA Health Report provides specific lifestyle and health insights that maximize prevention and preparedness."

The analysis behind the Coronavirus DNA Health Report is based on peer-reviewed, published research on coronavirus. Though preliminary, that body of research has identified specific genetic markers in a portion of the infected population that are associated with susceptibility to coronavirus infection as well as the potential impact upon health if infected.

The Coronavirus DNA Health Report Provides Actionable Guidance

Aside from evaluating users' coronavirus risks, the Coronavirus DNA Health Report delivers personalized, actionable guidance based on each person's DNA. A person who the Coronavirus DNA Health Report indicates may have a greater risk of a severe infection would be armed with vital information to take proactive, preventive measures. Higher risk individuals, for example, could be more aggressive about social distancing and could also ensure any medical conditions, such as high blood pressure and diabetes, are closely monitored and well controlled.

"For anyone who may come in contact with the virus, knowing whether they are at risk for a severe infection can be a crucial piece of information that empowers them to make decisions on how to best protect themselves," Dr. Colby said. "Sometimes even the smallest lifestyle changes can have a significant impact on a person's wellbeing."

While Sequencing.com's Coronavirus DNA Health Report is not a test for the presence of the virus, it can be used in the context of other non-genetic factors to assess a user's risk for infection and the how sick they are likely to get if they catch the virus. Sequencing.com will continuously update the analysis process that generates the report as more is learned about the virus causing the current outbreak.

Free Versions of the Coronavirus DNA Health Report Available

Sequencing.com is offering free versions of the Coronavirus DNA Health Report for users who have already taken a DNA test, either through the company's own sequencing service or through popular, commercially available third-party services such as 23andMe and AncestryDNA. For users who have not yet taken a DNA test, the company offers a $69 DNA test as well as clinical-grade Whole Genome Sequencing for $399.

Once the user's DNA data is stored in the person's account at Sequencing.com, it takes about 10 minutes to generate the Coronavirus DNA Health Report.

The Coronavirus DNA Health Report is one of more than 90 applications and services on Sequencing.com that analyze DNA data and provide insights to empower users to make more informed choices about their health, nutrition and overall wellness. The applications can use data from practically any DNA test including widely available genetic testing services.

Dr. Colby concluded, "Right now, there are more questions about coronavirus than there are answers, including why some infections appear mild while others can require more intensive medical care and hospitalization.Our Coronavirus DNA Health Report uses the information in each individual's DNA to provide actionable answers. We view our Coronavirus DNA Health Report as a reflection of our broader mission of utilizing the latest genetic technologies to help individuals outsmart their genes and live longer, healthier lives."

About Sequencing.com

Sequencing.com was founded in 2014 by Dr. Brandon Colby, an expert, entrepreneur and author in the field of personal genomics. Prior to founding the company, Dr. Colby practiced personalized preventive medicine in Los Angeles. Sequencing.com leverages the accessibility and falling costs of genetic testing to provide analysis and useful, straightforward guidance to users who are seeking to unlock the information in their DNA to live healthier, happier, longer lives.

Sequencing.com's HIPAA and GDPR compliant platform provides safe, confidential storage of a person's genetic data while also providing access to the world's largest marketplace of DNA analysis apps and reports. Sequencing.com's App Market includes more than 90 DNA analysis apps that empower individuals and healthcare professionals to be able to easily tap into genetic data for deep and rich insights. Powered by Universal Genetic Data Compatibility, apps available through Sequencing.com can process genetic data from any genetic test including Ancestry, 23andMe, MyHeritage, exome sequencing and whole genome sequencing.

For those who haven't yet taken a DNA test, Sequencing.com also offers DNA testing for $69 and clinical-grade 30x Whole Genome Sequencing for $399. Sequencing.com is the one-stop-shop that collates and organizes available genetic-based apps and genetic tests into the world's largest App Store for DNA. For more information, please visit https://sequencing.com.

Sequencing.com is backed by Mucker Capital, a leading Los Angeles-based venture capital firm specializing in investments in early-stage software and Internet-based businesses.

About Mucker Capital

Mucker Capital invests in seed and "pre-seed" stage companies building defensible and scalable Internet software and services businesses outside Silicon Valley. The firm partners with exceptional entrepreneurs to provide their earliest institutional funding and works with them side-by-side to help launch and scale their new ventures. It is Mucker's conviction that it must provide more than just capital. As operators and entrepreneurs themselves, Mucker's partners roll up their sleeves and work with entrepreneurs in all parts of their businesses that need additional resources - product development, customer development, business development, operational infrastructure, recruiting and fundraising.

Media Contacts

Joseph Kuo / Andrew WangHaven Tower Group424 317 4851 or 424 317 4859jkuo@haventower.comor awang@haventower.com

View original content:http://www.prnewswire.com/news-releases/sequencingcom-launches-free-genetic-analysis-to-assess-risk-and-severity-of-coronavirus-infection-for-each-individual-301026450.html

SOURCE Sequencing.com

View original post here:
Sequencing.com Launches Free Genetic Analysis To Assess Risk And Severity Of Coronavirus Infection For Each Individual - P&T Community

Read More...

Genomind Launches Express Ship-to-Patient Capability for Genomind Professional PGx Express Test Enabling Access to Critical Mental Health Treatment…

Friday, March 20th, 2020

KING OF PRUSSIA, Pa.--(BUSINESS WIRE)--Genomind, the leading mental health company bringing precision medicine into mainstream mental health treatment, today announced that it is offering an Express Ship-to-Patient option for its flagship product, Genomind Professional PGx Express, the most advanced and comprehensive mental health pharmacogenetic testing service. This test is designed to empower clinicians and pharmacists with genetic guidance to personalize patients treatment plans and reduce the painful process of mental health medication trial and error. Upon a clinician ordering Genomind Professional PGx Express, the new service upgrade enables patients to receive the test at home, with the healthcare provider receiving the results quickly and without requiring the patient to travel to a clinicians office or a lab. Genomind has activated their Express Ship-to-Patient service for all clinicians and is paying for the express shipping to meet the needs of mental health patients during the COVID-19 pandemic.

The travel limitations placed on our lives by the coronavirus pandemic require us to provide innovative solutions to help patients and healthcare providers, particularly during this time of intensified mental health strain, said Shawn Patrick OBrien, Chief Executive Officer at Genomind. I am very proud of my team for being able to bring forward the rollout of this Express Ship-to-Patient solution to address the current reality of the mental health treatment landscape. We look forward to working with our team, our providers, and alongside companies all over the world to do what we can to support those who need it during this global health crisis. Our patients mental health cant wait.

Genominds pharmacogenetic (PGx) testing identifies patient-specific genetic markers that can indicate which treatments have lower risk for side effects or adverse events and may be more likely to be effective. It can also provide guidance for specific dosages based on an individuals pharmacokinetic profile. The FDA has placed gene-specific warnings, precautions, drug interaction guidelines and dosage recommendations on over 270 medications.

Under the new offering, physicians can order the test and Genomind will expedite shipping of the test kit to the address provided by the patient. Patients will follow instructions to provide a sample via a simple cheek swab and return it to Genominds lab for processing. With all required information, the laboratory can turn around results in less than three days the fastest turnaround time in the industry and a results report is then delivered to the physician. Genomind is also supporting this new service with On-Demand test consults with its team of pharmacogenomic experts to provide insights and support to physicians in interpreting test results.

"During this time of uncertainly and instability, our mental health patients need us more than ever, said Gisoo Zarrabi, M.D. Medical Director of Harbor Psychiatry & Mental Health. The Genomind Express direct-to-patient service has already proven itself invaluable to our practice and has allowed us to continue practicing psychiatry to the highest standard possible. We are thankful that our patients will continue to benefit from these essential reports not just today, but for the rest of their lives.

For more information, connect at Genomind.com.

About Genomind Professional PGx Express (PGx Express)

Genomind Professional PGx Express, available by prescription, is the most advanced mental health genetic testing service available, analyzing variants on up to 24 genes selected for inclusion based on guidelines from expert consortia peer-reviewed studies due to their significance in mental health. Test results include information on patients genetic variants that are relevant for the treatment of conditions such as depression, anxiety, autism, schizophrenia, chronic pain, bipolar disorder, obsessive-compulsive disorder (OCD), attention deficit hyperactivity disorder (ADHD), post-traumatic stress disorder (PTSD), and substance abuse.

Genomind Professional PGx Express bundles the test results with a suite of services, including:

About GenomindGenomind is the leading mental health care company delivering the genetic testing tools that empower clinicians to make more informed treatment decisions and create better outcomes for patients with mental illnesses. As the scientific leader in genetic testing, Genominds flagship offering is Genomind Professional PGx Express the most advanced and comprehensive mental health genetic service available. Supported by a world-class genetics lab and unique consultative approach, Genomind is advancing a new paradigm of personalized medicine in mental health care. Learn more at http://www.genomind.com.

Read the original here:
Genomind Launches Express Ship-to-Patient Capability for Genomind Professional PGx Express Test Enabling Access to Critical Mental Health Treatment...

Read More...

How far should genetic engineering go to allow this couple to have a healthy baby? – Brisbane Times

Friday, March 20th, 2020

Normal text sizeLarger text sizeVery large text size

One morning in 2005, Shelley Beverley woke up to find that she had gone deaf. She was 21, and living in Johannesburg with her older brother Neil. I was very scared, she says. It was just so sudden. She struggled through the rest of the day, hoping that her hearing would come back, but it didnt. In one sense, her hearing loss wasnt entirely a surprise: Beverleys grandmother had been deaf, Neil had lost his hearing when he was 13, and her mum, Mary, had lost hers when she was 32. We knew it ran in the family, she says, but I thought Id been lucky and not inherited it.

Beverley, 35, lives in Margate, a semi-rural district south of Hobart, with her husband James. The couple migrated to Australia from South Africa in 2010, looking for space, buying 2 hectares of lush green grass at the foot of a forested ridge near the mouth of the Derwent River. We love the wildlife here, says James, looking out the living room window. Weve seen pademelons, echidnas, quolls, blue-tongue lizards, even a Tassie devil. At dusk, hundreds of kangaroos emerge from the forest to gorge on the grass. Its very peaceful, says James. Its really helped us after everything thats happened.

Apart from their deafness, Beverleys family had largely enjoyed good health. Then, in September 2015, her mother, Mary, then 62, started experiencing fatigue and stomach pain. Doctors in Durban ordered a colonoscopy, but the procedure made her worse. Her feet became swollen and purple. Because of their hearing problems, Shelley and Mary had communicated mainly in text messages. But soon I began noticing that her wording got a bit funny, says Beverley. It didnt always make sense.

Loading

Beverley flew to Durban in February 2016, but by that time her mother could no longer talk or walk. She was so weak that she couldnt move her hands or lift her neck. Two days after Beverley arrived in Durban, her mother caught a virus that caused fluid to build up on her lungs. The doctors tried unsuccessfully to drain it. Shortly afterwards, she died. She weighed just 36 kilograms. It was so fast, Beverley says. And we were still in the dark about what she had.

Shortly before Marys death, Neil had also fallen ill. He developed a number of mysterious symptoms, including facial twitches and seizures. He kept falling over and tripping, and experienced vomiting and headaches so severe he lost his vision for weeks at a time. His behaviour became strange showering with his clothes on, and hallucinating.

One day, Dad was driving him around and Neil started talking to all these little people he thought were around his feet, says Beverley. Doctors in Durban had trouble diagnosing him, so they sent a biopsy to London, where he was found to have a type of mitochondrial cytopathy one of a large family of chronic and progressive diseases that affect the muscles, brain and nervous system. As the family soon learnt, the condition has no cure and no effective therapies. One of the common early symptoms is hearing loss.

Neil died in June 2017, aged 34, by which time Beverley had discovered she also had the condition. It was fear, so much fear, she says. She began experiencing symptoms, including migraines and vision loss. She has since developed diabetes, hypertension, gastro-paresis (when your stomach muscles dont work), and pharyngeal dysphagia (difficulty swallowing). Every time I get sick now, the flu or something, I think, When am I going to need a wheelchair or a feeding tube? When will my legs stop working?

Mito has taken everything from me, she says. If I die, at least James will still have a part of me.

Beverley has bright blue eyes and long, straight, ash-brown hair. Shes got a lazy left eye and uncommonly pale skin, which she attributes to her condition. Oh, and I had bunions out in 2010, she says, laughing wryly.

She doesnt know how long shes got left, but she is determined to make it count. She has joined mito awareness groups, and is an active member of the Mito Foundation, which supports sufferers, and funds research. She has exhaustively researched the condition and takes every opportunity to educate doctors. Youd be surprised by how little they know about it, she says.

But her overriding focus has been on a cutting-edge, and currently illegal, procedure called mitochondrial donation, a form of IVF which would allow those with the condition to have children, safe in the knowledge they would not be passing it on. Mito has taken everything from me, she says. If I die, at least James will still have a part of me. I would like him to look at our child, and say, You have your mums smile or your mums eyes.

An IVF treatment known as mitochondrial donation could potentially save up to 60 Australian children a year from being born with the condition. Credit:

Mitochondrial donation has been labelled immoral and unethical, a slippery slope to designer babies, not to mention potentially unsafe. The only country in the world to have legalised it is the UK. A report by medical experts into the technologys potential application in Australia is due to be delivered to Health Minister Greg Hunt this month.

This fight is really personal to me, Beverley says. Short of a cure, people with mito should at least have the option of having healthy children.

Mitochondria are microscopic structures in human cells that provide the body with energy. For this reason, they are often described as the cells powerhouse. They are crucially important: if your mitochondria fail or mutate, your body will be starved of energy, causing multiple organ failure and premature death.

A stylised representation of a mitochondrion, which provides the body with energy. Malfunction can lead to organ failure and death.Credit:Josh Robenstone

Mito, which is maternally inherited, usually affects the muscles and major organs such as the brain, heart, liver, inner ears, and eyes. But it can cause any symptom in any organ, at any age. Indeed, the term mito includes more than 200 disorders, the symptoms of which are maddeningly varied and seemingly unrelated, leading to delayed diagnoses or incorrect diagnoses or, indeed, no diagnosis.

Many of these people have been fobbed off by doctors or laughed off by people who think they are hypochondriacs, says Dr David Thorburn, a mitochondrial researcher at the Murdoch Childrens Research Institute, in Melbourne, who has diagnosed some 700 cases over the past 28 years. Most people are relieved to finally know what it is, because that is the end of that part of their journey.

Its sometimes said babies produced as a result of mitochondrial donation would have three parents the mother, the father, and the donor.

Up to two million people worldwide have some form of mito. - Others, like Beverley, who have a less severe type of the disease, will get adult onset, and can expect to become ill in their 30s, 40s or 50s.

According to Thorburn, One of the things that most dismays families with mito is the lack of control they have over passing the condition down to future generations of their family.

Remaining childless is one way to stop the condition from being passed down, as is adopting, but as Thorburn acknowledges, There is an innate desire in many individuals to have their own children. For these people, mito donation offers the very real prospect that the condition is eliminated from future generations.

Mitochondrial replacement is a highly specialised procedure, requiring a level of manual dexterity sufficient to manipulate a womans egg, which is roughly the width of a human hair. Within that egg is a nucleus, where a persons genes are located, and the cytoplasm, the jelly-like substance that surrounds it. Mitochondria are found in the cytoplasm.

Mitochondrial replacement involves taking a donor females healthy egg, removing its nucleus and replacing it with the nucleus of the woman affected by mitochondrial disease, but whose nucleus is healthy. The egg is then fertilised using her partners sperm. (Another option is to fertilise the egg first, and then swap the nucleus.) The resulting embryo is then implanted into the mother.

Researcher David Thorburn: "Mito donation offers the very real prospect that the condition is eliminated from future generations."Credit:Josh Robenstone

Since more than 99.9 per cent of our genes are found in the eggs nucleus, which remains unaffected, the procedure will have no impact on the childs height, hair colour or mannerisms. Despite that, its sometimes said that babies produced as a result of mitochondrial donation would have three parents the mother, the father, and the donor.

The technology has been tested in mice for more than 30 years, but only since 2009 has research been done on human embryos, mainly in the UK. Almost from the start, the research was subject to sensational headlines about scientists playing God, and the possibility of genetic engineering, with much of the hysteria being fuelled by anti-abortion groups. The Catholic Church described it as a further step in commodification of the human embryo and a failure to respect new individual human lives.

In 2012, the Human Genetics Alert, an independent watchdog group in London, wrote a paper comparing any baby produced with mitochondrial replacement to Frankensteins creation, since they would be produced by sticking together bits from many different bodies. According to the Conservative British MP Jacob Rees-Mogg, the procedure was not a cure for disease, it is the creating of a different person.

Regulators subjected the technology to four separate scientific reviews, together with rounds of ethical debate and community consultation. In 2015, the UK Parliament voted to legalise the technology for use in humans, on the proviso that it only be available to those women at high risk of passing on the disease. Since then, 13 couples in the UK have received the go-ahead to undergo the procedure.

Its unclear how many children, if any, have been born: the parents have asked that details not be published. Meanwhile, scientists like Thorburn wait eagerly for news of any developments. I know the UK researchers well and have asked several of them, and they are keeping completely quiet about it in respecting the families wishes, he says.

Loading

If there have been babies born in the UK using the procedure, they arent the first. In April 2016, a child was born using the technique in Mexico, to a Jordanian mother who carried a fatal mitochondrial condition known as Leigh syndrome. The doctor in charge, an American fertility specialist called Dr John Zhang, later admitted that he had gone to Mexico because the procedure is illegal in America. In Mexico, he admitted, There are no rules.

Even those who want mitochondrial donation legalised in Australia concede that much remains unknown about the procedure. Its long-term risks can only be understood through lifelong health check-ups, but this is impossible until any children conceived via this procedure become adults. Implications for subsequent generations also remain unclear.

No medical procedure is 100 per cent safe, says Sean Murray, CEO of the Mito Foundation. But we think we are at the stage now where the benefits of the technology are greater than the risks.

One of the issues around safety concerns the compatibility of the donors mitochondria with the recipients nuclear genes. A 2016 study in mice suggested that mismatched mitochondria affected their metabolism and shortened their lives. Another concern is known as carryover, whereby a tiny amount of mutant mitochondria is inevitably transferred from the affected mothers egg into the donor egg during the procedure.

Instead of it being wiped out, the mutation might then reappear in the descendants of any girls born as a result. For this reason, some people have proposed that the procedure be restricted to male embryos only, but this raises all kinds of ethical issues around selective breeding and sex selection.

Loading

Indeed, it often seems as if the term ethical minefield was coined especially with mitochondrial donation in mind.

My primary ethical concern has to do with the sanctity of human life, says Father Kevin McGovern, a Catholic priest and member of the National Health and Medical Research Councils Mitochondrial Donation Expert Working Committee.

If mitochondrial donation is permitted here, the technique most likely to be used is pronuclear transfer, which requires that both the donors egg and the affected mothers egg be fertilised. [This is to ensure that both eggs are at the same developmental stage.] But once the nucleus is removed from the donors fertilised egg, it is discarded. For people who believe that life begins at conception, this is akin to murder. You are creating two lives and destroying one for spare parts.

The Catholic Church has consistently opposed mitochondrial donation. In a Senate inquiry into the technology in 2018, Dr Bernadette Tobin, director of the Plunkett Centre for Ethics at the Australian Catholic University, suggested the process was intrinsically evil.

The inquiry also heard from Father Anthony Fisher, Catholic Archbishop of Sydney, who raised concerns about the moral right of the child to know how he or she was conceived the problem of what he called genealogical bewilderment and the donors right to remain anonymous. He also worried that women might effectively become egg vending machines: The availability of human ova is often assumed when people talk about reproductive technology as if they were somehow there in a cupboard to be used. In fact, it means women have to be used to obtain these eggs. They are extracted by invasive procedures that do carry some risk.

A report by medical experts into mitochondrial donation and its potential application in Australia is due to be delivered to Health Minister Greg Hunt this month. Credit:Alex Ellinghausen

Equally troubling for the Australian Catholic Bishops Conference, the peak national body for the churchs bishops, was the fact that mitochondrial donation involved conceiving babies not by marital intercourse [but by] a technical procedure.

Most of these concerns are redundant, argues the Mito Foundations Sean Murray. We already have a well defined regulatory framework for dealing with all this, he says. As far as the donors right to remain anonymous, we would defer to the appropriate federal or state and territory regulations that apply for sperm or egg donations. In regard to a kids right to know they had a mitochondrial donor, societally there seems to be a preference to inform kids. Its important for them to understand their genetic lineage.

Then theres the matter of consent. The parents can wrestle with the ethical issues and weigh up all the risks, but the only person who cant consent to the procedure is the unborn child. Well, says Murray, they cant consent to being born with mito, either.

The Mito Foundations Sean Murray: "In regard to a kids right to know they had a mitochondrial donor, societally there seems to be a preference to inform kids."Credit:Joshua Morris

Murray, 47, is one of the founding directors of the Mito Foundation, which was established in Sydney in 2009. Mito runs in my family, he says. My older brother, Peter, died of it in 2009 at 45, and my mum passed away in 2011, at 70. What people often dont understand is that even in families that have mito, each member can have different mutational loads basically, different amounts of bad mitochondria. Peter got a high load, but I didnt. Thats why Im still here.

A computer scientist by training, Murray now works full-time on the foundation. Much of his job involves travelling around the country, explaining mito to politicians, journalists and philanthropists, raising funds for research and, most crucially, advocating for a change to the laws.

Mitochondrial donation falls foul of two pieces of legislation: the Research Involving Human Embryos Act 2002, and the Prohibition of Human Cloning for Reproduction Act 2002. The laws prohibit the implantation of a human embryo that contains more than two peoples genetic material. The laws were subject to a mandatory review in 2010, but the then Labor government recommended they remain the same.

In 2013, the Mito Foundation urged the government to revisit its decision. Two years later, it began lobbying in earnest. What we tried to get across was that the science around mito donation has come a long way since 2010, says Murray. Also, the process that the UK went through to legalise it really reassured us that the procedure is safe and effective.

Loading

In the past five years, Murray and his colleagues have consulted with more than 100 MPs and senators. Only one of them, according to Murray, said I dont like this. They have also talked to dozens of industry experts, including academics and medical and research bodies, about the benefits of mitochondrial donation. Most of them get it straight away, he says. We are talking about a technique that will prevent the chance of having a morbidly ill child.

Now, a breakthrough appears imminent. In February 2019, Health Minister Greg Hunt asked the National Health and Medical Research Council to look into the matter, review the science and conduct public consultation. The NHMRC is due to hand its report to Hunt this month. The expectation among the mito community is that he will recommend the laws be changed. Any proposals would then need to be debated in Parliament, where issues around reproductive medicine have, in the past, been hotly contested.

Murray expects some opposition from more conservative MPs, but nothing like the rancour seen in the NSW Parliament during last years debate over legalising abortion. Shadow health minister Chris Bowen has, for his part, said that Labor will support changing the laws.

Mitochondrial sufferer Shelley Beverley at home in Tasmania. This fight is really personal to me. Credit:Peter Mathew

Whether this will help people like Shelley Beverley is unclear. If Hunt gives it the green light, it will take two years at least for mitochondrial donation to become available to prospective parents, given the time involved in drafting and passing legislation, establishing a regulatory regime and getting doctors up to speed with the technology.

This will probably be too late for Beverley. I really only have about a year left to give it a go, she tells me. After that, my symptoms may progress and biologically things get worse after 35. She says she would consider going to the UK for the treatment, but that at present they are not accepting international patients.

Loading

In the meantime, she watches TV, and reads a little, but not too much. (It puts me to sleep.) She gardens: she has a bed of huge white and pink roses out the back of her house, as a memorial to her mother and brother. And she eats. James cooks for me. He lets me choose the best meat and potatoes! Ive put on weight since I met him. She describes James as something close to an angel. He will listen to every problem I have or feeling I experience. He will always put me first.

Beverley started going out with James when she was 21, right around the time she first went deaf. I was so scared that he wouldnt like me as much. I remember calling him and saying I was scared he would leave me. But James is still here. Im very lucky to have him, she says. If I go, I want him to have a part of me.

To read more from Good Weekend magazine, visit our page at The Sydney Morning Herald, The Age and Brisbane Times.

Tim Elliott is a senior writer with Good Weekend.

Read the rest here:
How far should genetic engineering go to allow this couple to have a healthy baby? - Brisbane Times

Read More...

When will a coronavirus vaccine be ready? – The Guardian

Friday, March 20th, 2020

Even at their most effective and draconian containment strategies have only slowed the spread of the respiratory disease Covid-19. With the World Health Organization finally declaring a pandemic, all eyes have turned to the prospect of a vaccine, because only a vaccine can prevent people from getting sick.

About 35 companies and academic institutions are racing to create such a vaccine, at least four of which already have candidates they have been testing in animals. The first of these produced by Boston-based biotech firm Moderna will enter human trials imminently.

This unprecedented speed is thanks in large part to early Chinese efforts to sequence the genetic material of Sars-CoV-2, the virus that causes Covid-19. China shared that sequence in early January, allowing research groups around the world to grow the live virus and study how it invades human cells and makes people sick.

But there is another reason for the head start. Though nobody could have predicted that the next infectious disease to threaten the globe would be caused by a coronavirus flu is generally considered to pose the greatest pandemic risk vaccinologists had hedged their bets by working on prototype pathogens. The speed with which we have [produced these candidates] builds very much on the investment in understanding how to develop vaccines for other coronaviruses, says Richard Hatchett, CEO of the Oslo-based nonprofit the Coalition for Epidemic Preparedness Innovations (Cepi), which is leading efforts to finance and coordinate Covid-19 vaccine development.

Coronaviruses have caused two other recent epidemics severe acute respiratory syndrome (Sars) in China in 2002-04, and Middle East respiratory syndrome (Mers), which started in Saudi Arabia in 2012. In both cases, work began on vaccines that were later shelved when the outbreaks were contained. One company, Maryland-based Novavax, has now repurposed those vaccines for Sars-CoV-2, and says it has several candidates ready to enter human trials this spring. Moderna, meanwhile, built on earlier work on the Mers virus conducted at the US National Institute of Allergy and Infectious Diseases in Bethesda, Maryland.

Sars-CoV-2 shares between 80% and 90% of its genetic material with the virus that caused Sars hence its name. Both consist of a strip of ribonucleic acid (RNA) inside a spherical protein capsule that is covered in spikes. The spikes lock on to receptors on the surface of cells lining the human lung the same type of receptor in both cases allowing the virus to break into the cell. Once inside, it hijacks the cells reproductive machinery to produce more copies of itself, before breaking out of the cell again and killing it in the process.

All vaccines work according to the same basic principle. They present part or all of the pathogen to the human immune system, usually in the form of an injection and at a low dose, to prompt the system to produce antibodies to the pathogen. Antibodies are a kind of immune memory which, having been elicited once, can be quickly mobilised again if the person is exposed to the virus in its natural form.

Traditionally, immunisation has been achieved using live, weakened forms of the virus, or part or whole of the virus once it has been inactivated by heat or chemicals. These methods have drawbacks. The live form can continue to evolve in the host, for example, potentially recapturing some of its virulence and making the recipient sick, while higher or repeat doses of the inactivated virus are required to achieve the necessary degree of protection. Some of the Covid-19 vaccine projects are using these tried-and-tested approaches, but others are using newer technology. One more recent strategy the one that Novavax is using, for example constructs a recombinant vaccine. This involves extracting the genetic code for the protein spike on the surface of Sars-CoV-2, which is the part of the virus most likely to provoke an immune reaction in humans, and pasting it into the genome of a bacterium or yeast forcing these microorganisms to churn out large quantities of the protein. Other approaches, even newer, bypass the protein and build vaccines from the genetic instruction itself. This is the case for Moderna and another Boston company, CureVac, both of which are building Covid-19 vaccines out of messenger RNA.

Cepis original portfolio of four funded Covid-19 vaccine projects was heavily skewed towards these more innovative technologies, and last week it announced $4.4m (3.4m) of partnership funding with Novavax and with a University of Oxford vectored vaccine project. Our experience with vaccine development is that you cant anticipate where youre going to stumble, says Hatchett, meaning that diversity is key. And the stage where any approach is most likely to stumble is clinical or human trials, which, for some of the candidates, are about to get under way.

Clinical trials, an essential precursor to regulatory approval, usually take place in three phases. The first, involving a few dozen healthy volunteers, tests the vaccine for safety, monitoring for adverse effects. The second, involving several hundred people, usually in a part of the world affected by the disease, looks at how effective the vaccine is, and the third does the same in several thousand people. But theres a high level of attrition as experimental vaccines pass through these phases. Not all horses that leave the starting gate will finish the race, says Bruce Gellin, who runs the global immunisation programme for the Washington DC-based nonprofit, the Sabin Vaccine Institute.

There are good reasons for that. Either the candidates are unsafe, or theyre ineffective, or both. Screening out duds is essential, which is why clinical trials cant be skipped or hurried. Approval can be accelerated if regulators have approved similar products before. The annual flu vaccine, for example, is the product of a well-honed assembly line in which only one or a few modules have to be updated each year. In contrast, Sars-CoV-2 is a novel pathogen in humans, and many of the technologies being used to build vaccines are relatively untested too. No vaccine made from genetic material RNA or DNA has been approved to date, for example. So the Covid-19 vaccine candidates have to be treated as brand new vaccines, and as Gellin says: While there is a push to do things as fast as possible, its really important not to take shortcuts.

An illustration of that is a vaccine that was produced in the 1960s against respiratory syncytial virus, a common virus that causes cold-like symptoms in children. In clinical trials, this vaccine was found to aggravate those symptoms in infants who went on to catch the virus. A similar effect was observed in animals given an early experimental Sars vaccine. It was later modified to eliminate that problem but, now that it has been repurposed for Sars-CoV-2, it will need to be put through especially stringent safety testing to rule out the risk of enhanced disease.

Its for these reasons that taking a vaccine candidate all the way to regulatory approval typically takes a decade or more, and why President Trump sowed confusion when, at a meeting at the White House on 2 March, he pressed for a vaccine to be ready by the US elections in November an impossible deadline. Like most vaccinologists, I dont think this vaccine will be ready before 18 months, says Annelies Wilder-Smith, professor of emerging infectious diseases at the London School of Hygiene and Tropical Medicine. Thats already extremely fast, and it assumes there will be no hitches.

In the meantime, there is another potential problem. As soon as a vaccine is approved, its going to be needed in vast quantities and many of the organisations in the Covid-19 vaccine race simply dont have the necessary production capacity. Vaccine development is already a risky affair, in business terms, because so few candidates get anywhere near the clinic. Production facilities tend to be tailored to specific vaccines, and scaling these up when you dont yet know if your product will succeed is not commercially feasible. Cepi and similar organisations exist to shoulder some of the risk, keeping companies incentivised to develop much-needed vaccines. Cepi plans to invest in developing a Covid-19 vaccine and boosting manufacturing capacity in parallel, and earlier this month it put out a call for $2bn to allow it to do so.

Once a Covid-19 vaccine has been approved, a further set of challenges will present itself. Getting a vaccine thats proven to be safe and effective in humans takes one at best about a third of the way to whats needed for a global immunisation programme, says global health expert Jonathan Quick of Duke University in North Carolina, author of The End of Epidemics (2018). Virus biology and vaccines technology could be the limiting factors, but politics and economics are far more likely to be the barrier to immunisation.

The problem is making sure the vaccine gets to all those who need it. This is a challenge even within countries, and some have worked out guidelines. In the scenario of a flu pandemic, for example, the UK would prioritise vaccinating healthcare and social care workers, along with those considered at highest medical risk including children and pregnant women with the overall goal of keeping sickness and death ra tes as low as possible. But in a pandemic, countries also have to compete with each other for medicines.

Because pandemics tend to hit hardest those countries that have the most fragile and underfunded healthcare systems, there is an inherent imbalance between need and purchasing power when it comes to vaccines. During the 2009 H1N1 flu pandemic, for example, vaccine supplies were snapped up by nations that could afford them, leaving poorer ones short. But you could also imagine a scenario where, say, India a major supplier of vaccines to the developing world not unreasonably decides to use its vaccine production to protect its own 1.3 billion-strong population first, before exporting any.

Outside of pandemics, the WHO brings governments, charitable foundations and vaccine-makers together to agree an equitable global distribution strategy, and organisations like Gavi, the vaccine alliance, have come up with innovative funding mechanisms to raise money on the markets for ensuring supply to poorer countries. But each pandemic is different, and no country is bound by any arrangement the WHO proposes leaving many unknowns. As Seth Berkley, CEO of Gavi, points out: The question is, what will happen in a situation where youve got national emergencies going on?

This is being debated, but it will be a while before we see how it plays out. The pandemic, says Wilder-Smith, will probably have peaked and declined before a vaccine is available. A vaccine could still save many lives, especially if the virus becomes endemic or perennially circulating like flu and there are further, possibly seasonal, outbreaks. But until then, our best hope is to contain the disease as far as possible. To repeat the sage advice: wash your hands.

This article was amended on 19 March 2020. An earlier version incorrectly stated that the Sabin Vaccine Institute was collaborating with the Coalition for Epidemic Preparedness Innovations (Cepi) on a Covid-19 vaccine.

Go here to read the rest:
When will a coronavirus vaccine be ready? - The Guardian

Read More...

Coronavirus outbreak revives dangerous race myths and pseudoscience – NBC News

Friday, March 20th, 2020

The news last week that NBA player Rudy Gobert, a Frenchman of Caribbean heritage, had tested positive for the coronavirus shattered a myth that some of the world's more conspiracy-minded had circulated online through jokes, news stories and social media posts.

Black people are not, in fact, immune to the coronavirus.

On Tuesday, the Afro-British actor Idris Elba, who lives part time in the United States and tested positive for COVID-19 this week, posted on social media about his early lack of symptoms and subsequent changes, how he managed to be tested, the dangers of the disease and the myth of black immunity.

"Something that is scaring me, when I read the comments and some of the reactions, my people, black people, please, please understand that coronavirus is ... you can get it," Elba said. "There are so many stupid, ridiculous conspiracy theories about black people not being able to get it. ...That is the quickest way to get more black people killed. And I'm talking about the whole world, wherever we are. ... Just know you have to be just as vigilant as every other race."

Variations on the immunity myth claims that black worshipers can't be infected at church where a pastor refused to cancel in-person services and false assertions that there are zero COVID-19 infections in Africa to name a few remain on the internet along with other fantastical ideas. The myth of group immunity may, public health, disease control and bioethicists say, provide some people with a bit of levity or sense of control in a seemingly dire time. But the risk of false information circulating in any form far outweighs the value of a few chuckles or nerve-calming denial.

What's more, fictional claims about black immunity from a potentially deadly viral infection are connected with a long history of contradictory but uniformly racist ideas serving the social or political needs of the moment, experts say.

"I want to be very clear there is, despite many claims to the contrary, no truth, no fact at all in claims of genetic differences, immunity or susceptibility, to disease based on race," said Otis Brawley, a professor of epidemiology and oncology at the Johns Hopkins University Bloomberg School of Public Health.

Full coverage of the coronavirus outbreak

The Morning Rundown

Get a head start on the morning's top stories.

The list of health conditions linked to genetic variations found more often in specific geographic regions is a short one. Even health conditions that appear disproportionately in some populations, such as sickle cell disease, fall in this category, Brawley said. As a result there are people classified in the United States as both white and black who have sickle cell disease or the genetic trait. But, this pattern is, too often, wrongly conflated with race, he said.

"Race medicine is almost always bad medicine," said Brawley. "So I do not have and, to my knowledge, no one else has, any data demonstrating either racial or geographic immunity from coronaviruses."

While the false notion of black immunity to coronavirus has, to some degree, faded in the days since the Gobert news which was followed by several other black NBA players testing positive as well other absurd notions, conspiracy theories and lies have rippled through many social media feeds.

"There are a whole range of crazy notions gaining traction," said Gail Christopher, executive director of the National Collaborative for Health Equity. "We all would like to have hope right now to counter the anxiety that the bulk of the news is giving us. So anybody is possibly vulnerable to misinformation. We do, all have, naturally, a psychological propensity to cling to hope, but that is also part of what makes this so dangerous."

Jokes may help some deal with the fear of a disease creating massive, disempowering social upheaval, Christopher said. Conspiracy theories may do the same. And the effort to rebrand the virus as the fault of the Chinese (or vice versa), is nothing more than a flagrant attempt to shift blame and deflect attention from a disjointed and inadequate official response, she said. What's more, black Americans are overrepresented among those living with some of the underlying health conditions asthma, diabetes, high blood pressure, heart disease that can put a person at risk of becoming seriously ill or dying due to the coronavirus, Christopher said. Global differences in health care access and quality compound this problem.

Representatives from the World Health Organization have met with many of the nation's major technology companies, and the organization is working with Google to curb the spread of misinformation online.

"The COVID-19 outbreak and response has been accompanied by a massive 'infodemic,'" the organization said in a statement to NBC News last week.

The WHO is also working with social media companies and influencers to detect misinformation and limit its spread, the agency added

"These myths have a track record not just of shaping attitudes but of shaping policy and practice in public and private spaces, in hospitals and in schools, in workplaces, too," said Dorothy Roberts, a University of Pennsylvania bioethicist, lawyer and sociologist who researches race in medicine. "It's not farfetched to fear that now."

Over the course of 100 years, the false belief spread that people of African descent had inferior or weak lungs ideas championed by no less than Thomas Jefferson and Samuel Cartwright, a respected doctor and professor at the school that would become Tulane University, according to Roberts, author of "Fatal Invention: How Science, Politics and Big Business Re-Create Race in the Twenty-First Century." The lungs of black people were so in need of constant exertion, the men argued, that forced unpaid labor slavery was a form of treatment.

Download the NBC News app for full coverage and alerts about the coronavirus outbreak

In the 1790s, nearly two decades after he signed the Declaration of Independence, Dr. Benjamin Rush, a respected white Philadelphia physician, and others advanced the idea that black people were somehow biologically immune to yellow fever. Rush did so during a massive outbreak in that city. Black people, Rush and others said, were uniquely positioned to care for the sick, dig graves, and cart away and bury bodies. In the end, 4,000 to 5,000 people died, including an estimated 240 black residents.

Given that history, this month, The Philadelphia Inquirer published a column refuting false claims that black Americans are immune to coronavirus. The city is more than 42 percent black. But the problem of racial myths in medicine remains a part of modern thinking well beyond Philadelphia, Roberts said.

In 2016, a survey of 222 medical students and residents found that half believed at least one of several myths about biological differences between black and white people that shaped their approach to treating pain. As a result, the medical professionals were overwhelmingly less willing to regard or treat the pain of black patients the same as white patients.

And the problems do not end there. To this day, most of the nation's medical facilities use different standards for measuring lung and kidney function for black patients versus others, Roberts said. This, in turn, means that black patients must register more significant breathing difficulties or kidney dysfunction before the most serious medical interventions are offered.

"Any way you look at it, I see no benefit to any of the myths about black physical peculiarity," Roberts said. "They have all been dangerous to black people's health and welfare in America, supportive of white supremacy and promoted low levels of care and concern about black people's health.

"So I think it's a mistake to even joke about it. I just don't find it funny at all."

Visit link:
Coronavirus outbreak revives dangerous race myths and pseudoscience - NBC News

Read More...

HORAMA Signs Exclusive License Agreement with Leiden University Medical Center Targeting CRB1 Gene Mutations to Treat Inherited Retinal Dystrophies -…

Friday, March 20th, 2020

PARIS & LEIDEN, Netherlands--(BUSINESS WIRE)--HORAMA SA, a French biotechnology company focusing on gene therapy for the treatment of rare genetic diseases in ophthalmology, announced today an exclusive licensing agreement with the Leiden University Medical Center (LUMC) for global rights to a gene therapy program to treat the Inherited Retinal Dystrophy associated with pathogenic CRB1 gene mutations, a rare but devastating ophthalmic condition leading to blindness

"We are excited to enter into this agreement with the LUMC, a leading academic institution with highly recognised scientific leaders in the field of gene therapy such as Jan Wijnholds, to expand our leadership in gene therapy. This collaboration enables us to expand our pipeline of gene therapy treatments for ophthalmic conditions for which there is a high unmet medical need, commented Christine Placet, CEO of HORAMA.

Our studies in the last 20 years resulted in the development of a platform for candidate gene therapy medicines for children with pathogenic CRB1 mutations. The main obstacle to test our novel innovative medicine gene therapy products in clinical studies was the high costs of the clinical development phase. We are, therefore, excited about this research agreement with HORAMA team, which is a global expert in this field, commented Jan Wijnholds, LUMC.

Under the agreement, HORAMA will receive an exclusive worldwide license to certain patent rights and know-how for the drug candidate (referenced as HORA-001). In return for these rights, LUMC will receive an undisclosed upfront payment, milestone payments and royalties on net sales of products. HORAMA shall be responsible to bring the gene therapy to market with completion of the non-clinical and clinical studies. Based on current timelines, and subject to regulatory review, HORAMA expects initiating a Phase I/II clinical study with HORA-001 in 2023.

Per the agreement, the parties have entered into a non-clinical development agreement with Leiden University Medical Center (LUMC), led by Dr. Jan Wijnholds, Team Leader and permanent staff member at the LUMC Department of Ophthalmology.

About HORAMA

At HORAMA, we believe in gene therapy to treat a broad range of inherited disorders.

Our focus is on Inherited Retinal Dystrophies with our lead clinical program targeting patients with PDE6B gene mutations, a condition which leads to progressive vision loss in children and adults ultimately leading to legal blindness.

Our team is pushing the boundaries of gene therapy by advancing next generation delivery platforms that will improve effectiveness and coverage of gene transfer to address multiple diseases. For more information, please go to: http://www.horama.fr.

Gene therapy market (source: FiorMarkets and Grand View Research, Inc)

Gene therapy is being developed with an aim to treat rare conditions with limited or no treatment options.

Genetic disorders occur due to gene mutations, which can result in incorrect protein synthesis. Gene therapy is used to introduce a healthy gene into cells to allow the synthesis of a functional protein. Growing awareness and acceptance of gene therapy for various disease treatments are favouring market growth.

The global gene therapy market is estimated to reach $5.5 billion by 2026, while the global ophthalmology market is projected to grow to $43 billion by 2026 (April 2019 report issued by Grand View Research, Inc.).

Inherited Retinal Dystrophies

Inherited Retinal Dystrophies (IRD) represent a diverse group of progressive visually debilitating diseases that can lead to blindness. In patients with an IRD, mutations in genes, which are critical to retinal function, lead to progressive, direct or indirect photoreceptor cell death and associated visual function losses.

IRDs are a genetically heterogeneous group of diseases, with over 260 genes identified to date, IRDs associated with pathogenic CRB1 gene mutations are among this heterogeneous group, similar to the autosomal recessive IRD associated with pathogenic PDE6B gene mutations.

About CRB1

CRB1 gene mutations are a major cause of early onset and delayed onset IRD. Proteins such as CRB1 and CRB2 are essential in the retina to maintain adhesion between photoreceptors and Mller glial cells. Loss of CRB function results in loss of photoreceptors and causes blindness.

Go here to see the original:
HORAMA Signs Exclusive License Agreement with Leiden University Medical Center Targeting CRB1 Gene Mutations to Treat Inherited Retinal Dystrophies -...

Read More...

An updated guide to the coronavirus drugs and vaccines in development – STAT

Friday, March 20th, 2020

In the months since the novel coronavirus rose from a regional crisis to a global threat, drug makers large and small have scrambled to advance their best ideas for thwarting a pandemic.

Some are taking a cue from older antivirals. Some are tapping tried-and-true technologies, and others are pressing forward with futuristic approaches to human medicine.

Heres a guide to some of the most talked-about efforts to treat or prevent coronavirus infection, with details on the science, history, and timeline for each endeavor.Were looking at novel medicines, not repurposed drugs. (For more on some of the efforts to repurpose drugs, read this.) The below therapies and vaccines are sorted in order of how close they could be to approval, starting with a treatment in Phase 3 trials, followed by others in Phase 1 studies and then preclinical development. Approval, of course, would only come if they are proven safe and effective.

advertisement

Gilead SciencesApproach: TreatmentStage: Phase 3

Gileads remdesivir is being studied in five clinical trials around the world. In China, Gilead is recruiting about 1,000 patients diagnosed with the coronavirus to determine whether multiple doses of remdesivir can reverse the infection. The primary goals are reducing fever and helping patients get out of the hospital within two weeks. The drug, which previously failed in a study on Ebola virus, is administered intravenously. More on the drug here.

advertisement

Ascletis PharmaApproach: TreatmentStage: Phase 1

Chinese drug maker Ascletis Pharma is testing a combination of antivirals, one approved for HIV and one approved for hepatitis C, that might treat coronavirus infection. Last month, the company enrolled 11 patients with coronavirus-caused pneumonia and administered a cocktail of danoprevir and ritonavir. All 11 were eventually discharged, according to Ascletis. The company hasnt disclosed plans for a larger study.

Moderna TherapeuticsApproach: VaccineStage: Phase 1Moderna set a drug industry record with mRNA-1273, a vaccine candidate identified just 42 days after the novel coronavirus was sequenced. The company is working with the National Institutes of Health on a healthy-volunteer study that began earlier this month. If mRNA-1273 proves itself to be safe, Moderna will enroll hundreds more patients to determine whether the vaccine protects against infection. Modernas product is a synthetic strand of messenger RNA, or mRNA, designed to convince bodily cells to produce antibodies against the virus. The company, founded in 2010, is yet to win Food and Drug Administration approval for any of its mRNA medicines. More on the vaccine candidate here.

CanSino BiologicsApproach: VaccineStage: Phase 1

CanSino Biologics, headquartered in Tianjin, is close to testing its novel coronavirus vaccine in a clinical trial in China. CanSinos approach involves taking a snippet of coronavirus genetic code and entwining it with a harmless virus, thereby exposing healthy volunteers to the novel infection and spurring the production of antibodies. The company said this week that Chinese authorities approved its planned trial, which will begin as soon as possible. CanSino markets a vaccine for Ebola virus in China.

Arcturus TherapeuticsApproach: VaccineStage: Preclinical

Arcturus Therapeutics is pressing forward with a vaccine that relies on engineering RNA. The company plans to take an RNA virus that has been edited to encode for proteins that will protect against infection and load it into a liquid nanoparticle. The resulting vaccine, being developed in partnership with Duke University, promises a better immune response at a lower dose than competing mRNA approaches, according to the company. The vaccine remains in preclinical development, and Arcturus has promised to start a human trial as quickly as possible.

BioNTechApproach: VaccineStage: PreclinicalGermanys BioNTech is working on an mRNA vaccine for the novel coronavirus with plans to enter clinical testing in April. Like its competitors, the company uses strands of mRNA to spur the production of protective antibodies. Earlier this month, Shanghais Fosun Pharma signed a deal to market BioNTechs vaccine in China if its eventually approved. Pfizer has agreed to co-develop the vaccine in the rest of the world.

CureVacApproach: VaccineStage: Preclinical

Like Moderna, CureVac uses man-made mRNA to spur the production of proteins. And, like Moderna, it got a grant from the nonprofit Coalition for Epidemic Preparedness Innovations to apply its technology to coronavirus. CureVac has said it expects to have a candidate ready for animal testing by April, aiming to start a clinical study this summer. The company is also working with CEPI on a mobile mRNA manufacturing technology, one that would theoretically allow health care workers to rapidly produce vaccines to respond at the site of an outbreak.

Eli LillyApproach: TreatmentStage: Preclinical

Eli Lilly has partnered with a Canadian firm called AbCellera to develop antibody treatments for coronavirus infection. Using a blood sample from a coronavirus survivor, AbCellera identified more than 500 antibodies that might protect against the virus. Now its working with Lilly to identify which are most potent. The two companies aim to have a treatment ready for human trials within the next four months.

GlaxoSmithKlineApproach: VaccineStage: Preclinical

GlaxoSmithKline, one of the worlds largest vaccine manufacturers, is lending its technology to a Chinese biotech firm at work on a coronavirus vaccine. Under an agreement signed last month, GSK is providing its proprietary adjuvants compounds that enhance the effectiveness of vaccines to Clover Biopharmaceuticals, a privately held company based in Chengdu. Clovers approach involves injecting proteins that spur an immune response, thereby priming the body to resist infection. GSK struck a similar deal with the University of Queensland in Australia, which is also working on a protein vaccine.The company has not said when it expects to advance either into human testing. GSK is also lending its scientific expertise to CEPI.

Inovio PharmaceuticalsApproach: VaccineStage: Preclinical

Inovio has spent the last four decades working to turn DNA into medicine, and the company believes its technology could quickly generate a vaccine for the novel coronavirus. Working with CEPI grant money, Inovio has come up with a DNA vaccine it believes can generate protective antibodies and keep patients from infection. The company has partnered with a Chinese manufacturer, Beijing Advaccine Biotechnology, and is working through preclinical development with a candidate called INO-4800. The company expects to progress into clinical trials in April and has promised to manufacture 1 million doses of its candidate this year.

Johnson & JohnsonApproach: Vaccine and treatmentStage: Preclinical

Johnson & Johnson, which has in the past responded to outbreaks of the Ebola and Zika viruses, is taking a multipronged approach to the coronavirus. The company is in the early days of developing a vaccine that would introduce patients to a deactivated version of the virus, triggering an immune response without causing infection. Human trials could begin by November. At the same time, J&J is working with the federal Biomedical Advanced Research and Development Authority on potential treatments for patients who are already infected, a process that includes investigating whether any of its older medicines might work against the coronavirus.

PfizerApproach: Vaccine and treatmentStage: Preclinical

Outside of its vaccine work with BioNTech, Pfizer has put out a five-point plan to address the outbreak, which includes making its technology, scientists, expertise, and manufacturing available to outside institutions. The company has also promised to create a rapid-response program to make it easier to respond to future pandemics.

Regeneron PharmaceuticalsApproach: TreatmentStage: Preclinical

Regeneron has grown into a $50 billion business based on its ability to craft human antibodies out of genetically engineered mice. Now its tapping that technology in hopes of treating coronavirus. The company immunized its proprietary antibody-generating mice with a harmless analog of the novel coronavirus, generating potential treatments for the infection. Regeneron plans to select the two most potent antibodies and advance the cocktail into human studies by early summer. The last time Regeneron embarked on this process, during the Ebola outbreak of 2015, it came up with an antibody cocktail that roughly doubled survival rates for treated patients. More on Regenerons treatment here.

SanofiApproach: Vaccine and treatmentStage: Preclinical

Sanofi, which has successfully developed vaccines for yellow fever and diphtheria, is working with BARDA on an answer to the coronavirus. Sanofis approach involves taking some of the coronaviruss DNA and mixing it with genetic material from a harmless virus, creating a chimera that can prime the immune system without making patients sick. Sanofi expects to have a vaccine candidate to test in the lab within six months and could be ready to test a vaccine in people within a year to 18 months. Approval would likely be at least three years away, the company said. Outside of vaccines, Sanofi and Regeneron have started a clinical trial to test whether Kevzara, an approved anti-inflammatory drug, can help with the symptoms of Covid-19.

TakedaApproach: TreatmentStage: Preclinical

Japanese pharma giant Takeda is at work on a treatment derived from the blood of people who have already been infected by the coronavirus. The company is drawing blood from coronavirus survivors, harvesting the plasma, and then isolating the protective antibodies that kept those patients alive. Its not a new idea. Blood transfusions have been used to combat viral outbreaks since at least the Spanish Flu pandemic of 1918. But Takedas take on it could prove to be faster in development than other therapeutic approaches. According to the company, the therapy could be available to patients in 12 to 18 months.

Vir BiotechnologyApproach: TreatmentStage: Preclinical

Vir Biotechnology, a company focused on infectious disease, has isolated antibodies from people who survived SARS, a viral relative of the novel coronavirus, and is working to determine whether they might treat the infection. Teaming up with Chinese pharma contractor WuXi Biologics, the San Francisco-based Vir is in the early stages of development and hasnt specified when it expects to have products ready for human testing. The company has also aligned with Alnylam Pharmaceuticals to work on treatments that might halt viral replication by interfering with RNA signaling. Virs CEO, Biogen veteran George Scangos, is also coordinating the trade group BIOs response to the coronavirus outbreak.

See the rest here:
An updated guide to the coronavirus drugs and vaccines in development - STAT

Read More...

‘The genie is out of the bottle’: How ‘mind reading’ will transform medical care – Genetic Literacy Project

Friday, March 20th, 2020

The ability to detect electrical activity in the brain through the scalp, and to control it, will soon transform medicine and change society in profound ways. Patterns of electrical activity in the brain can reveal a persons cognitionnormal and abnormal. New methods to stimulate specific brain circuits can treat neurological and mental illnesses and control behavior. In crossing this threshold of great promise, difficult ethical quandaries confront us.

Mind reading

The ability to interrogate and manipulate electrical activity in the human brain promises to do for the brain what biochemistry did for the body. Likewise, in experimental research destined to soon enter medical practice, just a few minutes of monitoring electrical activity in your brain using EEG and other methods can reveal not only neurological illness but also mental conditions like ADHD and schizophrenia.

Against the historical backdrop of ethical lapses and concerns that curtailed brain stimulation research for mental illnesses decades ago, we are reaching a point where it will become unethical to deny people suffering from severe mental or neurological illness treatments by optogenetic or electrical stimulation of their brain, or to withhold diagnosing their conditions objectively by reading their brains electrical activity. But the genie is out of the bottle. We better get to know her.

Read the original post

See the original post:
'The genie is out of the bottle': How 'mind reading' will transform medical care - Genetic Literacy Project

Read More...

Medicine is getting to grips with individuality – The Economist

Friday, March 13th, 2020

Mar 12th 2020

NEENA NIZAR is 42 years old, a professor of business studies and just 122cm tall. The ends of her bones are soft and pliable: on an x-ray they look frayed, like old paintbrushes. During her childhood and adolescence in Dubai she was operated on 30 times. The source of her problem remained a mystery. In 2010, after three decades of wondering, she finally received a diagnosis: Jansens Metaphyseal Chondrodysplasia, a condition first recognised in the 1930s. Her problems stem from a broken copy of just one of her 20,000 genes.

Dr Nizar is in some ways very unusual. Fewer than one in 200m people have the mutation to the PTH1R gene that causes Jansens disease. In other ways she is like everyone else. Although few people have a defect as debilitating, everyones health, and ill-health, is tied to the contents of their genomes. All genomes contain arrangements of genes that make psychological disorders, cancers, dementias or circulatory diseases either more of a problem or less of one. Everyone has genes that make them better or worse at metabolising drugs, more or less likely to benefit from specific forms of exercise, better able to digest some foods than others.

The same arrangement will never be seen twice. Though for identical twins the differences are the height of subtlety, each of the 7.5bn human genomes sharing the planet is unique. That irreducible diversity represents a challenge to many of the 20th centurys greatest medical advances, which were based on a one-size-fits-all approach. Personalising medicine is an enticing opportunity for improvement.

Good doctors have always treated their patients as individuals. In the 20th century blood tests, X-rays, body scans and other diagnostic tools made the specifics of each patients particular problems ever more visible. A spectacular reduction in the cost of reading, or sequencing, the DNA bases that make up human genetic information is adding a new level of individuality. It is now possible to inspect genetic differences with an ease previously unimaginable, and thus to know something about propensities to disease well before any symptoms show up.

Nobody knows exactly how many human genomes have been fully sequenced, and different sequencing procedures read the genome to different degreesthere are quick skims and painstaking philological studies. But the number is in the millions (see chart). By the 2030s genome sequencing is likely to be as routine in some places as taking a pin-prick of blood from a babys heel is todayit may even be part of the same procedure. Genome science is becoming a matter of practical medicine. New therapies that make it possible to adjust or edit this genetic inheritance are coming to market.

This flood of data is allowing medicine to become more precise and more personalin many ways, the p-words are two sides of the same coin. Previously recognised genetic diseases, such as Jansens, have been traced to specific genes and can be connected to defects in the proteins they create (almost all genes describe proteins, and proteins do almost all the bodys chemical work). Most of these diseases are rare, in that they typically affect no more than one person in 2,000 in the general population. But with over 6,000 such rare diseases now recognised, this means they are common in the aggregate. In Britain one in 17 people can expect to suffer from a rare disease at some point.

Studies of genetic diseases are not just a worthwhile end in themselves. Understanding what goes wrong when a specific protein is out of whack can reveal basic information about the bodys workings that may be helpful for treating other ailments. And the growing understanding of how large sets of genes may contribute to disease is making it possible to pick out the patients most at risk from common diseases like diabetes, heart conditions and cancer. That will help doctors personalise their interventions. In theory, the rise in access to personal genetic information allows individuals to better calculate these risks and to take pre-emptive action. In practice, so far, few people seem to do so.

Genomics is not the only source of new personal-health data. Just as all genomes are unique, so are the lives that all those genome-carriers lead. The increase in other forms of data about individuals, whether in other molecular information from medical tests, electronic health records, or digital data recorded by cheap, ubiquitous sensors, makes what goes on in those lives ever easier to capture. The rise of artificial intelligence and cloud computing is making it possible to analyse this torrent of data.

Almost 4bn people carry smartphones that can monitor physical activity. It is estimated that by 2022, 1bn people may be wearing a device such as a smart watch that can monitor their heart rate. The data-driven giants and startups of Silicon Valley are eager to help. Consumers no longer need to go to a doctor for a genome scan or to engage with a wide range of opinion about what ails them, or will ail them. The pharmaceutical companies used to dominating medicine are working hard to keep up. So are doctors, hospitals and health systems.

These possibilities are not without their risks, drawbacks and potential for disappointment. The ability to pinpoint what has gone wrong in a genome does not make it easy to fix. Moreover, as technology helps people monitor themselves in more ways, the number of the worried well will swell and unnecessary care will grow. Many could be done real harm by an algorithmic mirage.

Beyond this, the move fast and break things attitude common in tech companies sits uneasily with first, do no harm. And the untrammelled, unsupervised and unaccountable means of data accrual seen in other industries which have undergone digital transformations sits uneasily with concerns over medical privacy.

The very nature of medicine, though, means that the future will not just be a matter of business goals, research cultures, technological prowess, wise practice and well-crafted regulations. It will also be subject to the driving interests of particular individuals in ways never seen before. The development of gene-based medical research in Britain was deeply affected by the short, difficult life of Ivan Cameron, whose father, David Cameron, did much to build up genomics when he was prime minister. Many of those working in this field are impelled by personal loss.

And then there are those whose interests stem from the way in which their own genes shape their lives. People like Dr Nizar, who is now crafting a new research agenda for Jansens disease. There may only be 30 people in the world who suffer from it. But two of them are her children, and they are in ceaseless pain. Science knows why; medicine cannot yet help. We believe in miracles, she says. She is also working to make one happen.

This article appeared in the Technology Quarterly section of the print edition under the headline "Populations of one"

Excerpt from:
Medicine is getting to grips with individuality - The Economist

Read More...

Understanding SARS-CoV-2 and the drugs that might lessen its power – The Economist

Friday, March 13th, 2020

Mar 12th 2020

THE INTERCONNECTEDNESS of the modern world has been a boon for SARS-CoV-2. Without planes, trains and automobiles the virus would never have got this far, this fast. Just a few months ago it took its first steps into a human host somewhere in or around Wuhan, in the Chinese province of Hubei. As of this week it had caused over 120,000 diagnosed cases of covid-19, from Troms to Buenos Aires, Alberta to Auckland, with most infections continuing to go undiagnosed (see article).

But interconnectedness may be its downfall, too. Scientists around the world are focusing their attention on its genome and the 27 proteins that it is known to produce, seeking to deepen their understanding and find ways to stop it in its tracks. The resulting plethora of activity has resulted in the posting of over 300 papers on MedRXiv, a repository for medical-research work that has not yet been formally peer-reviewed and published, since February 1st, and the depositing of hundreds of genome sequences in public databases. (For more coverage of covid-19 see our coronavirus hub.)

The assault on the vaccine is not just taking place in the lab. As of February 28th Chinas Clinical Trial Registry listed 105 trials of drugs and vaccines intended to combat SARS-CoV-2 either already recruiting patients or proposing to do so. As of March 11th its American equivalent, the National Library of Medicine, listed 84. This might seem premature, considering how recently the virus became known to science; is not drug development notoriously slow? But the reasonably well-understood basic biology of the virus makes it possible to work out which existing drugs have some chance of success, and that provides the basis for at least a little hope.

Even if a drug were only able to reduce mortality or sickness by a modest amount, it could make a great difference to the course of the disease. As Wuhan learned, and parts of Italy are now learning, treating the severely ill in numbers for which no hospitals were designed puts an unbearable burden on health systems. As Jeremy Farrar, the director of the Wellcome Trust, which funds research, puts it: If you had a drug which reduced your time in hospital from 20 days to 15 days, thats huge.

Little noticed by doctors, let alone the public, until the outbreak of SARS (severe acute respiratory syndrome) that began in Guangdong in 2002, the coronavirus family was first recognised by science in the 1960s. Its members got their name because, under the early electron microscopes of the period, their shape seemed reminiscent of a monarchs crown. (It is actually, modern methods show, more like that of an old-fashioned naval mine.) There are now more than 40 recognised members of the family, infecting a range of mammals and birds, including blackbirds, bats and cats. Veterinary virologists know them well because of the diseases they cause in pigs, cattle and poultry.

Virologists who concentrate on human disease used to pay less attention. Although two long-established coronaviruses cause between 15% and 30% of the symptoms referred to as the common cold, they did not cause serious diseases in people. Then, in 2002, the virus now known as SARS-CoV jumped from a horseshoe bat to a person (possibly by way of some intermediary). The subsequent outbreak went on to kill almost 800 people around the world.

Some of the studies which followed that outbreak highlighted the fact that related coronaviruses could easily follow SARS-CoV across the species barrier into humans. Unfortunately, this risk did not lead to the development of specific drugs aimed at such viruses. When SARS-CoV-2similarly named because of its very similar genomeduly arrived, there were no dedicated anti-coronavirus drugs around to meet it.

A SARS-CoV-2 virus particle, known technically as a virion, is about 90 nanometres (billionths of a metre) acrossaround a millionth the volume of the sort of cells it infects in the human lung. It contains four different proteins and a strand of RNAa molecule which, like DNA, can store genetic information as a sequence of chemical letters called nucleotides. In this case, that information includes how to make all the other proteins that the virus needs in order to make copies of itself, but which it does not carry along from cell to cell.

The outer proteins sit athwart a membrane provided by the cell in which the virion was created. This membrane, made of lipids, breaks up when it encounters soap and water, which is why hand-washing is such a valuable barrier to infection.

The most prominent protein, the one which gives the virions their crown- or mine-like appearance by standing proud of the membrane, is called spike. Two other proteins, envelope protein and membrane protein, sit in the membrane between these spikes, providing structural integrity. Inside the membrane a fourth protein, nucleocapsid, acts as a scaffold around which the virus wraps the 29,900nucleotides of RNA which make up its genome.

Though they store their genes in DNA, living cells use RNA for a range of other activities, such as taking the instructions written in the cells genome to the machinery which turns those instructions into proteins. Various sorts of virus, though, store their genes on RNA. Viruses like HIV, which causes AIDS, make DNA copies of their RNA genome once they get into a cell. This allows them to get into the nucleus and stay around for years. Coronaviruses take a simpler approach. Their RNA is formatted to look like the messenger RNA which tells cells what proteins to make. As soon as that RNA gets into the cell, flummoxed protein-making machinery starts reading the viral genes and making the proteins they describe.

First contact between a virion and a cell is made by the spike protein. There is a region on this protein that fits hand-in-glove with ACE2, a protein found on the surface of some human cells, particularly those in the respiratory tract.

ACE2 has a role in controlling blood pressure, and preliminary data from a hospital in Wuhan suggest that high blood pressure increases the risks of someone who has contracted the illness dying of it (so do diabetes and heart disease). Whether this has anything to do with the fact that the viruss entry point is linked to blood-pressure regulation remains to be seen.

Once a virion has attached itself to an ACE2 molecule, it bends a second protein on the exterior of the cell to its will. This is TMPRSS2, a protease. Proteases exist to cleave other proteins asunder, and the virus depends on TMPRSS2 obligingly cutting open the spike protein, exposing a stump called a fusion peptide. This lets the virion into the cell, where it is soon able to open up and release its RNA (see diagram).

Coronaviruses have genomes bigger than those seen in any other RNA virusesabout three times longer than HIVs, twice as long as the influenza viruss, and half as long again as the Ebola viruss. At one end are the genes for the four structural proteins and eight genes for small accessory proteins that seem to inhibit the hosts defences (see diagram). Together these account for just a third of the genome. The rest is the province of a complex gene called replicase. Cells have no interest in making RNA copies of RNA molecules, and so they have no machinery for the task that the virus can hijack. This means the virus has to bring the genes with which to make its own. The replicase gene creates two big polyproteins that cut themselves up into 15, or just possibly 16, short non-structural proteins (NSPs). These make up the machinery for copying and proofreading the genomethough some of them may have other roles, too.

Once the cell is making both structural proteins and RNA, it is time to start churning out new virions. Some of the RNA molecules get wrapped up with copies of the nucleocapsid proteins. They are then provided with bits of membrane which are rich in the three outer proteins. The envelope and membrane proteins play a large role in this assembly process, which takes place in a cellular workshop called the Golgi apparatus. A cell may make between 100 and 1,000 virions in this way, according to Stanley Perlman of the University of Iowa. Most of them are capable of taking over a new celleither nearby or in another bodyand starting the process off again.

Not all the RNA that has been created ends up packed into virions; leftovers escape into wider circulation. The coronavirus tests now in use pick up and amplify SARS-CoV-2-specific RNA sequences found in the sputum of infected patients.

Because a viral genome has no room for free riders, it is a fair bet that all of the proteins that SARS-CoV-2 makes when it gets into a cell are of vital importance. That makes each of them a potential target for drug designers. In the grip of a pandemic, though, the emphasis is on the targets that might be hit by drugs already at hand.

The obvious target is the replicase system. Because uninfected cells do not make RNA copies of RNA molecules, drugs which mess that process up can be lethal to the virus while not necessarily interfering with the normal functioning of the body. Similar thinking led to the first generation of anti-HIV drugs, which targeted the process that the virus uses to transcribe its RNA genome into DNAanother thing that healthy cells just do not do.

Like those first HIV drugs, some of the most promising SARS-CoV-2 treatments are molecules known as nucleotide analogues. They look like the letters of which RNA or DNA sequences are made up; but when a virus tries to use them for that purpose they mess things up in various ways.

The nucleotide-analogue drug that has gained the most attention for fighting SARS-CoV-2 is remdesivir. It was originally developed by Gilead Sciences, an American biotechnology firm, for use against Ebola fever. That work got as far as indicating that the drug was safe in humans, but because antibody therapy proved a better way of treating Ebola, remdesivir was put to one side. Laboratory tests, though, showed that it worked against a range of other RNA-based viruses, including SARS-CoV, and the same tests now show that it can block the replication of SARS-CoV-2, too.

There are now various trials of remdesivirs efficacy in covid-19 patients. Gilead is organising two in Asia that will, together, involve 1,000 infected people. They are expected to yield results in mid- to late-April. Other nucleotide analogues are also under investigation. When they screened seven drugs approved for other purposes for evidence of activity against SARS-CoV-2, a group of researchers at the State Key Laboratory of Virology in Wuhan saw some potential in ribavirin, an antiviral drug used in the treatment of, among other things, hepatitis C, that is already on the list of essential medicines promulgated by the World Health Organisation (WHO).

Nucleotide analogues are not the only antiviral drugs. The second generation of anti-HIV drugs were the protease inhibitors which, used along with the original nucleotide analogues, revolutionised the treatment of the disease. They targeted an enzyme with which HIV cuts big proteins into smaller ones, rather as one of SARS-CoV-2s NSPs cuts its big polyproteins into more little NSPs. Though the two viral enzymes do a similar job, they are not remotely relatedHIV and SARS-CoV-2 have about as much in common as a human and a satsuma. Nevertheless, when Kaletra, a mixture of two protease inhibitors, ritonavir and lopinavir, was tried in SARS patients in 2003 it seemed to offer some benefit.

Another drug which was developed to deal with other RNA-based virusesin particular, influenzais Favipiravir (favilavir). It appears to interfere with one of the NSPs involved in making new RNA. But existing drugs that might have an effect on SARS-CoV-2 are not limited to those originally designed as antivirals. Chloroquine, a drug mostly used against malaria, was shown in the 2000s to have some effect on SARS-CoV; in cell-culture studies it both reduces the viruss ability to get into cells and its ability to reproduce once inside them, possibly by altering the acidity of the Golgi apparatus. Camostat mesylate, which is used in cancer treatment, blocks the action of proteases similar to TMPRSS2, the protein in the cell membrane that activates the spike protein.

Not all drugs need to target the virus. Some could work by helping the immune system. Interferons promote a widespread antiviral reaction in infected cells which includes shutting down protein production and switching on RNA-destroying enzymes, both of which stop viral replication. Studies on the original SARS virus suggested that interferons might be a useful tool for stopping its progress, probably best used in conjunction with other drugs

Conversely, parts of the immune system are too active in covid-19. The virus kills not by destroying cells until none are left, but by overstimulating the immune systems inflammatory response. Part of that response is mediated by a molecule called interleukin-6one of a number of immune-system modulators that biotechnology has targeted because of their roles in autoimmune disease.

Actemra (tocilizumab) is an antibody that targets the interleukin-6 receptors on cell surfaces, gumming them up so that the interleukin-6 can no longer get to them. It was developed for use in rheumatoid arthritis. China has just approved it for use against covid-19. There are anecdotal reports of it being associated with clinical improvements in Italy.

While many trials are under way in China, the decline in the case rate there means that setting up new trials is now difficult. In Italy, where the epidemic is raging, organising trials is a luxury the health system cannot afford. So scientists are dashing to set up protocols for further clinical trials in countries expecting a rush of new cases. Dr Farrar said on March 9th that Britain must have its trials programme agreed within the week.

International trials are also a high priority. Soumya Swaminathan, chief scientist at the WHO, says that it is trying to finalise a master protocol for trials to which many countries could contribute. By pooling patients from around the world, using standardised criteria such as whom to include and how to measure outcomes, it should be possible to create trials of thousands of patients. Working on such a large scale makes it possible to pick up small, but still significant, benefits. Some treatments, for example, might help younger patients but not older ones; since younger patients are less common, such an effect could easily be missed in a small trial.

The caseload of the pandemic is hard to predict, and it might be that even a useful drug is not suitable in all cases. But there are already concerns that, should one of the promising drugs prove to be useful, supplies will not be adequate. To address these, the WHO has had discussions with manufacturers about whether they would be able to produce drugs in large enough quantities. Generic drug makers have assured the organisation that they can scale up to millions of doses of ritonavir and lopinavir while still supplying the HIV-positive patients who rely on the drugs. Gilead, meanwhile, has enough remdesivir to support clinical trials and, thus far, compassionate use. The firm says it is working to make more available as rapidly as possible, even in the absence of evidence that it works safely.

In the lab, SARS-CoV-2 will continue being dissected and mulled over. Details of its tricksiness will be puzzled out, and the best bits of proteins to turn into vaccines argued over. But that is all for tomorrow. For today doctors can only hope that a combination of new understanding and not-so-new drugs will do some good.

Dig deeper:

This article appeared in the Briefing section of the print edition under the headline "Anatomy of a killer"

Read more here:
Understanding SARS-CoV-2 and the drugs that might lessen its power - The Economist

Read More...

GW Pharmaceuticals submits Type II Variation Application to the European Medicines Agency (EMA) to expand the use of EPIDYOLEX, (cannabidiol) oral…

Friday, March 13th, 2020

LONDON, March 13, 2020 (GLOBE NEWSWIRE) -- GW Pharmaceuticals plc (NASDAQ:GWPH) ("GW", "the Company" or "the Group"), a world leader in discovering, developing and commercialising cannabinoid prescription medicines, today announces the submission of a Type II Variation Application to the European Medicines Agency (EMA) seeking approval of EPIDYOLEX, (cannabidiol) oral solution, for the treatment of seizures associated with Tuberous Sclerosis Complex (TSC), a rare genetic condition and a leading cause of genetic epilepsy. If approved, this will be the third licensed indication for GW's cannabidiol oral solution in Europe.

"This submission to the EMA is an important step for GW and furthers GW's mission to bring innovative cannabinoid medicines to patients with high unmet need," said Chris Tovey, GW's Chief Operating Officer. "We look forward to working with the EMA to demonstrate GW's cannabidiol oral solution's potential in this new indication and hope to make this rigorously tested cannabis-based medicine available to a new group of patients through a potential approval in due course."

TSC is a condition that causes mostly benign tumours to grow in vital organs of the body including the brain, skin, heart, eyes, kidneys and lungs, and in which epilepsy is the most common neurological feature. TSC is typically diagnosed in childhood.1

The Type II Variation Application is based on data from a positive Phase 3 safety and efficacy study. The study met its primary endpoint with patients treated with GW's cannabidiol oral solution 25 mg/kg/day experiencing a significantly greater reduction from baseline in TSC-associated seizures compared to placebo (49% vs 27%; p=0.0009). Results for the 50 mg/kg/day dose group were similar, with seizure reductions of 48% from baseline vs 26.5% for placebo (p=0.0018). All key secondary endpoints were supportive of the effects on the primary endpoint. The safety profile observed was consistent with findings from previous studies, with no new safety risks identified.

ADDITIONAL INFORMATION

About Tuberous Sclerosis Complex (TSC)Tuberous Sclerosis Complex (TSC) is a rare genetic condition that has an estimated prevalence in the EU of 10 in 100,000.2 The condition causes mostly benign tumours to grow in vital organs of the body including the brain, skin, heart, eyes, kidneys and lungs and is a leading cause of genetic epilepsy.1,3 TSC often occurs in the first year of life with patients suffering from either focal seizures or infantile spasms. It is associated with an increased risk of autism and intellectual disability.1 The severity of the condition can vary widely. In some children the disease is very mild, while others may experience life-threatening complications.4

About EPIDIOLEX/EPIDYOLEX (cannabidiol) oral solutionEPIDIOLEX/EPIDYOLEX (cannabidiol) oral solution, the first prescription, plant-derived cannabis-based medicine approved by the U.S. Food and Drug Administration (FDA) for use in the U.S. and the European Medicines Agency's (EMA) for use in Europe, is an oral solution which contains highly purified cannabidiol (CBD). EPIDYOLEX received approval in Europe in September 2019 for the treatment of seizures associated with Lennox-Gastaut syndrome (LGS) or Dravet syndrome in patients two years of age or older in conjunction with clobazam. In the U.S., EPIDIOLEX was approved in June 2018 by the FDA and is indicated for the treatment of seizures associated with LGS or Dravet syndrome in patients two years of age or older. A supplemental New Drug Application (sNDA) was submitted to the FDA in early 2020 for the treatment of seizures associated with Tuberous Sclerosis Complex (TSC). GW's cannabidiol oral solution has received Orphan Drug Designation from the FDA and the EMA for the treatment of seizures associated with Dravet syndrome, LGS and TSC, each of which are severe childhood-onset, drug-resistant syndromes.

About GW Pharmaceuticals plc Founded in 1998, GW is a biopharmaceutical company focused on discovering, developing and commercialising novel therapeutics from its proprietary cannabinoid product platform in a broad range of disease areas. The Company's lead product, EPIDIOLEX/EPIDYOLEX (cannabidiol) oral solution is commercialised in Europe by GW, and in the U.S. by the Company's subsidiary, Greenwich Biosciences. The Company has a strong pipeline of additional cannabinoid product candidates, with late-stage clinical trials in autism, schizophrenia, post-traumatic stress disorder (PTSD) and spasticity associated with multiple sclerosis (MS) and spinal cord injury. For further information, please visit http://www.gwpharm.com.

1 NIH Tuberous Sclerosis Fact Sheet. https://www.ninds.nih.gov/Disorders/Patient-Caregiver-Education/Fact-Sheets/Tuberous-Sclerosis-Fact-Sheet. 2 Prevalence and incidence or rare diseases: Bibliographic data.https://www.orpha.net/orphacom/cahiers/docs/GB/Prevalence_of_rare_diseases_by_alphabetical_list.pdf3 TS Alliance Website. https://www.tsalliance.org/. Accessed November 19, 2019.4 de Vries PJ, Belousova E, Benedik MP, et al. TSC-associated neuropsychiatric disorders (TAND): findings from the TOSCA natural history study. Orphanet J Rare Dis. 2018;13(1):157.5 Kwan P., Brodie M.J. Early identification of refractory epilepsy. N. Engl. J. Med. 2000;342(5):314319.6 French JA. Refractory epilepsy: clinical overview. Epilepsia. 2007;48 Suppl 1:3-7.

See more here:
GW Pharmaceuticals submits Type II Variation Application to the European Medicines Agency (EMA) to expand the use of EPIDYOLEX, (cannabidiol) oral...

Read More...

Stem cells to help the heart – Science Magazine

Friday, March 13th, 2020

Shinya Yamanaka's 2006 discovery of induced pluripotent stem cells (iPSCs) ignited a revolution in the field of stem cell biology (1). For the first time, nearly all human somatic tissues could be produced from iPSCs reprogrammed from blood or skin cells, in a process that took only weeks. This advance was particularly crucial for obtaining surrogate tissues from cell types that are otherwise difficult to procure and do not readily expand in vitro, such as cardiac or neural cells. Additionally, many ethical concerns are avoided, because this technology uses a patient's own genetic material to create iPSCs rather than relying on embryonic stem cells. In the aftermath of Yamanaka's discovery, entire biomedical industries have developed around the promise of using human iPSCs (hiPSCs) and their derivatives for in vitro disease modeling, drug screening, and cell therapy (2).

The hiPSC technology has had a particularly notable impact in cardiac regenerative medicine, a field where scientists and clinicians have been working to devise new methods to better understand how cardiovascular disease manifests and how to restore cardiovascular function after disease strikes (3). The heart is limited in its ability to regenerate lost cardiomyocytes (beating heart muscle cells), following an adverse event such as a heart attack (4). Cardiomyocytes derived from hiPSCs (hiPSC-CMs) may represent a potential replacement option for dead cells in such a scenario. However, certain issues remain to be addressed, such as whether hiPSC-CMs can integrate with host myocardial tissue in the long term (5).

While using hiPSC-CMs for in vivo cell therapy may become practical in the future, employing hiPSC-CMs for high-throughput drug discovery and screening is becoming a reality in the present (6). Cardiovascular diseases can be recapitulated in a dish with patient-specific hiPSC-CMs. For example, if a patient exhibits a cardiac arrhythmia caused by a genetic abnormality in a sarcomeric protein or ion channel, that same rhythm problem can be recapitulated in vitro (7). Thanks to advances in hiPSC differentiation protocols, hiPSC-CMs can now be mass-produced to study cardiovascular disease mechanisms in vitro (8).

My graduate thesis in the laboratories of Joseph Wu and Sean Wu at Stanford University focused on in vitro applications of hiPSC-CMs for cardiovascular disease modeling and for high-throughput screening of chemotherapeutic compounds to predict cardiotoxicity. I initially embarked on a project using hiPSC-CMs to model viral myocarditis, a viral infection of the heart, caused by the B3 strain of coxsackievirus (9). I began by demonstrating that hiPSC-CMs express the receptors necessary for viral internalization and subsequently found that hiPSC-CMs were highly susceptible to coxsackievirus infection, exhibiting viral cytopathic effect within hours of infection. I also identified compounds that could alleviate coxsackievirus infection on hiPSC-CMs, a translationally relevant finding, as there remains a shortage of treatments for viral myocarditis.

Using a genetically modified variant of coxsackievirus B3 expressing luciferase, I developed a screening platform for assessing the efficacy of antiviral compounds. Pretreatment with interferon-, ribavirin, or pyrrolidine dithiocarbamate markedly suppressed viral replication on hiPSC-CMs by activating intracellular antiviral response and viral protein clearance pathways. These compounds alleviated viral replication in a dose-dependent fashion at low concentrations without causing cellular toxicity.

I next sought to use hiPSC-CMs to screen anticancer chemotherapeutic compounds for their off-target cardiovascular toxicities (10). Cardiotoxicity represents a major cause of drug withdrawal from the pharmaceutical market, and several chemotherapeutic agents can cause unintended cardiovascular damage (11). Using cultured hiPSC-CMs, I evaluated 21 U.S. Food and Drug Administrationapproved tyrosine kinase inhibitors (TKIs), commonly prescribed anticancer compounds, for their cardiotoxic potential. HiPSC-CMs express the major tyrosine kinase receptor proteins such as the insulin, insulin-like growth factor (IGF), vascular endothelial growth factor (VEGF), and platelet-derived growth factor (PDGF) receptors, lending validity to this cellular model.

Initially, human induced pluripotent stem cells (hiPSCs) can be produced by reprogramming skin or blood cells by nonviral or viral reprogramming methods. Cardiac differentiation protocols allow for the creation of cardiomyocytes derived from hiPSCs (hiPSC-CMs) for downstream applications, including in vitro disease modeling, drug screening, and regenerative cell therapy.

With data from a battery of cellular apoptosis, contractility, electrophysiology, and signaling assays, I generated a cardiac safety index to help align in vitro toxicity data to clinical drug safety guidelines (12). From the safety index, I determined that a subclass of VEGF receptor 2/PDGF receptorinhibiting tyrosine kinase inhibitors, some of which exhibit toxicity clinically, also elicited cardiotoxicities in hiPSC-CMs. These manifested as substantial alterations in cellular electrophysiology, contractility, and viability when administered at clinically relevant concentrations. I also discovered that cotreatment with either IGF or insulin partially rescued TKI-induced toxicity by up-regulating antiapoptotic signaling pathways. This work could prove useful for groups aiming to develop effective screening platforms to assess new chemotherapeutic compounds for cardiotoxic side effects.

I also collaborated with the Center for the Advancement of Science in Space (CASIS) to send a sample of hiPSC-CMs to the International Space Station. As humankind ventures beyond our home planet, it is imperative that we better understand how the heart functions for long periods of time in microgravity. Analysis of these hiPSC-CMs revealed microgravity-induced alterations in metabolic gene expression and calcium handling (13).

In recent years, the stem cell field has experienced an explosion of studies using hiPSC-CMs as a model cellular system to study cardiovascular biology. As improvements in hiPSC-CM mass production continue, we will see a rise in studies using these cells for disease modeling and drug screening. Thus, although hiPSC-CM technology is in its infancy, it holds great potential to improve cardiovascular health.

PHOTO: COURTESY OF A. SHARMA

FINALIST

Arun Sharma

Arun Sharma received his undergraduate degree from Duke University and a Ph.D. from Stanford University. Having completed a postdoctoral fellowship at the Harvard Medical School, Sharma is now a senior research fellow jointly appointed at the Smidt Heart Institute and Board of Governors Regenerative Medicine Institute at the Cedars-Sinai Medical Center in Los Angeles. His research seeks to develop in vitro platforms for cardiovascular disease modeling and drug cardiotoxicity assessment. http://www.sciencemag.org/content/367/6483/1206.1

See the original post here:
Stem cells to help the heart - Science Magazine

Read More...

Page 24«..1020..23242526..3040..»


2024 © StemCell Therapy is proudly powered by WordPress
Entries (RSS) Comments (RSS) | Violinesth by Patrick