Gene-Based Therapy May Thwart a Tough Blood Cancer – Sioux City Journal

MONDAY, June 5, 2017 (HealthDay News) -- Genetically tuning a person's own immune cells to target cancer appears to provide long-lasting protection against a blood cancer called multiple myeloma, an early trial from China shows.

The treatment, called CAR T-cell therapy, caused 33 out of 35 patients with recurring multiple myeloma to either enter full remission or experience a significant reduction in their cancer.

The results are "impressive," said Dr. Len Lichtenfeld, deputy chief medical officer for the American Cancer Society.

"These are patients who have had prior treatment and had their disease return, and 100 percent of the patients are reported to have had some form of meaningful response to these cells that were administered," Lichtenfeld said.

The new therapy is custom-made for each patient. Doctors collect the patient's own T-cells -- one of the immune system's main cell types -- and genetically reprogram them to target and attack abnormal multiple myeloma cells.

Lead researcher Dr. Wanhong Zhao likened the process to fitting immune cells with a GPS that steers them to cancer cells -- making them into professional killers that never miss their target.

Zhao is associate director of hematology at the Second Affiliated Hospital of Xi'an Jiaotong University in Xi'an, China.

CAR T-cell therapy is promising because the genetically altered T-cells are expected to roost in a person's body, multiplying and providing long-term protection, Lichtenfeld said.

"The theory is they should attack the tumor and continue to grow to become a long-term monitoring and treatment system," Lichtenfeld said. "It's not a one-shot deal."

The technology represents the next step forward in immunotherapy for cancer, said Dr. Michael Sabel, chief of surgical oncology at the University of Michigan.

"Immunotherapy is now really providing hope to a lot of patients with cancers that were not really responding to our standard chemotherapies," Sabel said.

CAR T-cell therapy previously has been used to treat lymphoma and lymphocytic leukemia, Lichtenfeld said.

Zhao and his colleagues decided to try the therapy to treat multiple myeloma. They re-engineered the patients' T-cells and then reintroduced them to the body in three infusions performed within one week.

Multiple myeloma is a cancer that occurs in plasma cells, which are mainly found in bone marrow and produce antibodies to fight infections. About 30,300 people will likely be diagnosed with multiple myeloma this year in the United States, researchers said in background notes.

"Multiple myeloma is a disease that historically was fatal in the course of a couple of years," Lichtenfeld said. During the past two decades, new breakthroughs have extended survival out 10 to 15 years in some patients, he noted.

To date, 19 of the first 35 Chinese patients have been followed for more than four months, researchers report.

Fourteen of those 19 patients have reached the highest level of remission, researchers report. There hasn't been a relapse among any of these patients, including five followed for more than a year.

"That's as far as you can go in terms of driving down the amount of tumor that's in the body," Lichtenfeld said.

Out of the remaining five patients, one experienced a partial response and four a very good response, researchers said.

However, about 85 percent of the patients experienced cytokine release syndrome (CRS), a potentially dangerous side effect of CAR T-cell therapy.

Symptoms of cytokine release syndrome can include fever, low blood pressure, difficulty breathing, and impaired organ function, the researchers said. However, most of the patients experienced only transient symptoms, and "now we have drugs to treat it," Lichtenfeld said.

History suggests the therapy will cost a lot if it receives approval, Lichtenfeld said. However, prior to approval, much more research will be needed, he added.

The Chinese research team plans to enroll a total of 100 patients in this clinical trial at four hospitals in China. They also plan a similar clinical trial in the United States by 2018, Zhao said.

The study was funded by Nanjing Legend Biotech Co., the Chinese firm developing the technology.

The findings were presented Monday at the American Society of Clinical Oncology annual meeting, in Chicago. Data and conclusions presented at meetings are usually considered preliminary until published in a peer-reviewed medical journal.

Excerpt from:
Gene-Based Therapy May Thwart a Tough Blood Cancer - Sioux City Journal

Here’s Why Editas Medicine Gained as Much as 17.2% Today – Motley Fool

What happened

Shares of gene-editing pioneer Editas Medicine (NASDAQ:EDIT) rose over 17% today before settling near gains of 5% in the last hour of trading, after the company announced a strategic research-and-development collaboration with Allergan (NYSE:AGN). The pair will team up to advance and develop Editas' lead drug candidate, taking aim at a rare group of eye diseases collectively called Leber Congenital Amaurosis, or LCA. The rare inherited disease is detected at (or within months after) birth and can cause severe loss of vision or blindness.

Allergan, already a leader in treating and developing novel treatments for eye diseases, will also have exclusive access to license up to five of the gene-editing platform's ocular programs. Editas Medicine will receive $90 million up front, plus potential milestones and royalty payments. Of course, it's worth pointing out that even the lead program has yet to enter clinical trials.

Image source: Getty Images.

The partnership announcement specifically mentioned LCA10, which is one of 18 recognized types of LCA. Each type of the disease affects a different single gene, an important consideration for early gene-editing therapeutic candidates. That's because it will be easier to treat diseases with simpler genetic mutations affecting one gene (such as Friedreich's ataxia, sickle-cell anemia, and LCA) than it will be to treat diseases with more complex genetic influences (such as heart disease).

More specifically, there are good reasons for the company to initially focus on diseases affecting vision. CRISPR, the gene-editing technology used by Editas Medicine, has been shown in the past 18 months to restore sight in blind lab animals. Those external studies did not achieve very high efficiency rates and focused on diseases other than LCA, but there are encouraging similarities.

In the short term, given the early-stage nature of the technology and the company's pipeline, investors should focus more on the financial aspects of the deal. The $90 million up-front payment will provide a nice boost to the balance sheet, which showed $185 million in cash at the end of 2016. Plus, investors could expect additional up-front payments should Allergan license programs aside from LCA10.

It's also important to note that Allergan will be responsible for all expenses related to the development and commercialization of each program licensed, unless Editas Medicine exercises its option to co-develop and co-market up to two of the programs licensed by its new partner. That will allow the gene-editing pioneer to avoid significant clinical, regulatory, and marketing expenses while developing and commercializing its platform.

This may not be a blockbuster deal for investors, but there could be more deals on the way, now that the company's technology platform is no longer operating under the fog of uncertainty caused by a recently settled legal dispute. Either way, Allergan is a deep-pocketed and experienced partner that can shield Editas Medicine from development risks as the latter prepares to bring a CRISPR therapeutic into the clinic for the first time.

Maxx Chatsko has no position in any stocks mentioned. The Motley Fool has no position in any of the stocks mentioned. The Motley Fool has a disclosure policy.

See original here:
Here's Why Editas Medicine Gained as Much as 17.2% Today - Motley Fool

Anorexia Nervosa Modeled in a Dish, Risk Gene Uncovered – Genetic Engineering & Biotechnology News

Although anorexia nervosa (AN) is often viewed as a nonbiological disorder, it is strongly suspected of having a genetic component, one that likely involves associations with multiple genes. These associations, however, appear to produce signals that are too faint to catch with the sort of genome-wide studies used thus far to evaluate AN. To enhance our sensitivity to AN's genetic signals, scientists based at the University of California San Diego School (UCSD) of Medicine have created a cellular model of the eating disorder. This model enabled the scientists to identify a gene, TACR1, that seems to contribute to AN pathophysiology.

The new findings appeared March 14 in the journal Translational Psychiatry, in an article entitled "Modeling Anorexia Nervosa: Transcriptional Insights from Human iPSC-Derived Neurons." The article notes that although AN has the highest mortality among psychiatric conditions, it still lacks robust and effective treatment, largely because the cellular and molecular mechanisms underlying the disease remain obscure.

In hopes of bringing these mechanisms to light, the UCSD team, led by Alysson Muotri, Ph.D., reprogrammed skin cells harvested from four females with AN and four healthy controls, induced these skin cells to become induced pluripotent stem cells (iPSCs), and differentiated the iPSCs into neurons.

The resulting neural cultures were "subjected to extensive transcriptome analysis," wrote the article's authors. "Within a small cohort of patients who presented for treatment, we identified a novel gene that appears to contribute to AN pathophysiology, TACR1 (tachykinin 1 receptor)."

The researchers explained that their unbiased comprehensive whole transcriptome and pathway analyses were arranged to determine not just which genes were being expressed or activated in AN neurons, but which genes or transcripts (bits of RNA used in cellular messaging) might be associated with causing or advancing the disease process.

Although the researchers failed to observe predicted differences in neurotransmitter levels, they did succeed in detecting the disruption of the TACR1 gene. This finding led the researchers to speculate that tachykinins might interact with other neurotransmitters to disrupt the tachykinin system and contribute to AN symptoms.

Tachykinins are neuropeptides or proteins expressed throughout the nervous and immune systems. These proteins participate in many cellular and physiological processes and have been linked to multiple diseases, including chronic inflammation, cancer, infection, and affective and addictive disorders.

"Although TACR1 has been associated with psychiatric conditions, especially anxiety disorders, we believe this report is its first association with AN," the authors of the Translational Psychiatry article concluded. "Moreover, our human iPSC approach is a proof-of-concept that AN can be modeled in vitro with a full human genetic complement, and represents a new tool for understanding the elusive molecular and cellular mechanisms underlying the disease."

"Anorexia is a very complicated, multifactorial neurodevelopmental disorder," commented Dr. Muotri. "It has proved to be a very difficult disease to study, let alone treat. We don't actually have good experimental models for eating disorders. In fact, there are no treatments to reverse AN symptoms.

"[Our work is] a novel technological advance in the field of eating disorders, which impacts millions of people. These findings transform our ability to study how genetic variations alter brain molecular pathways and cellular networks to change risk of ANand perhaps our ability to create new therapies."

The rest is here:
Anorexia Nervosa Modeled in a Dish, Risk Gene Uncovered - Genetic Engineering & Biotechnology News

Gene that causes rare disorder, Opitz C syndrome, identified – Science Daily

Gene that causes rare disorder, Opitz C syndrome, identified
Science Daily
A team of researchers has now identified a gene that causes the Opitz C syndrome in the only patient in Catalonia diagnosed with this severe congenital disease. This new scientific advance is a first step to discover the genetic bases of this syndrome ...

Visit link:
Gene that causes rare disorder, Opitz C syndrome, identified - Science Daily

Collected Wisdom: Dr. Val Gene Iven combines love of sports with medicine – NewsOK.com

Dr. Val Gene Iven goes over some medical issues with Marcus Smart, an OSU basketball star from 2012-14. [PHOTO BY BRUCE WATERFIELD, OKLAHOMA STATE UNIVERSITY]

Val Gene Iven grew up in Pond Creek, north of Enid, then graduated from OSU and the OU Health Sciences. In 1993, he became the team doctor for University of Tennessee athletics. In 2007, Iven returned to OSU in the same role. Iven's brother, Van Shea, was the longtime Channel 4 sports reporter who now is on staff with the Oklahoma Secondary School Activities Association.

I was born in Enid. I'd have had to be born at the house if I was born in Pond Creek.

Growing up in Pond Creek, small-town values, to me those are the best days of my life. Just because the community, your work ethic, growing up on a farm, school system, everybody in town knew you. Can't beat that.

I thought at a pretty early age I wanted to be a doctor. Probably somewhere in the junior high years. I loved the farm life but had terrible allergies, just couldn't be around wheat dust. I could be on the tractor, but the wheat dust just ate me up. So I kind of thought, I want to be a doctor. Had a great role model in Enid, my pediatrician, Dr. (Robert) Shuttee. Went to college, and that's the route I went and never wavered.

Got my M.D. from OU Health Sciences Center. Stayed there, did my residency there in family medicine. Then stayed there and did a fellowship in primary care sports medicine. I was the first fellow that they had in primary care sports medicine.

I thought I wanted to go into medicine and probably thought early on, I just liked kids, maybe going into pediatrics. But I loved sports. Grew up around sports. Tried to combine the two worlds.

Right out of my fellowship, '93, there were a couple of openings at Division I, Tennessee and Florida. Interviewed with both. Tennessee, got the call back from them first. Didn't know anybody at Knoxville or anybody affiliated with the university. I remember telling mom and dad, I'm going to go do this for two or three years and I'll be back. Dad reminded me of that when I came back 13 years later.

This job is a lot that you don't learn in med school. There's just so much nowadays, from the NCAA, from the Big 12. It's much more than just being a physician. From all the things we do in regards to training, from rehabilitation, from nutrition, the whole world of drug testing. All of the people that you have to communicate with nowadays, in regards to coaches and administrators and families. So it's grown so much over the years, it's just a full-time job.

The opportunity brought me back to Stillwater. I had kept in contact with people. And Dr. (Mark) Pascale, our orthopedist, called and said the team physician, Dr. Ken Smith, who had replaced Dr. (Donald) Cooper, decided he was just going to fulfill a role in the student health center and they were looking for somebody full time. It was just an opportunity I couldn't pass up. Your folks are back in Oklahoma. My grandmother at the time was nearing 100. Kids having the opportunity to be around their grandparents. Being back at your alma mater.

Great opportunity in the SEC, meet those people. Now back at your alma mater for 10 years. I've just been blessed.

I missed most of Coach (Eddie) Sutton. But yeah, we've had unprecedented times now, in regards to the run we've had in football, in particular. When I first got back in '07, we were in the process of building. I remember (growing up) sitting in the end zone, wasn't bowled in. Dad and I would drive over on a Saturday, just for the game, drive back. Just wasn't near the world it is now, game day or facilities. So we've come a million miles.

Van Shea is six years younger. Mom thought she was pretty clever with our names. Dad's name is Gene. So she started with Val Gene. She'd heard there was a Val Gene's restaurant. I think that was part of it. And once she came up with Val Gene, she couldn't go with Frank. So she had to come up with something. And we've both been called each other's names.

I'm completely just Van Shea's brother. Anywhere I go, anybody I'm introduced to, it's all, Oh, your Van Shea's brother. And I'm proud of that.

Pond Creek is our roots. That's your family. That's what you're always going to remember and go back to in life in regards to kind of where you got your values and knowing people. I credit a lot of things I've learned through the years, dating back to my days from grade school and high school in Pond Creek.

More:
Collected Wisdom: Dr. Val Gene Iven combines love of sports with medicine - NewsOK.com

3 Huge Healthcare Trends and How to Invest in Them (Hint: One Is Marijuana) – Motley Fool

A growing and increasingly longer-living global population makes healthcare one of the most attractive sectors for investors, but I thinkthat genetic research, robotic surgery, and marijuana legalization could be the industry's biggest money-making opportunities. If so, then Illumina Corp.(NASDAQ:ILMN), Intuitive Surgical (NASDAQ:ISRG), and GW Pharmaceuticals (NASDAQ:GWPH) could be smart stocks to buy.

Researchers are increasingly finding that disease is caused by genetic abnormalities, and often, those discoveries are being made using machines and disposable supplies sold by gene-sequencing giant Illumina Corp.

IMAGE SOURCE: GETTY IMAGES.

Illumina is the largest manufacturer of systems used to sequence genetic code, and it's launching new machines this year that could make gene sequencing quicker and cheaper.

There are more than 7,500 of Illumina's machines installed at customers already, and increasing spending on DNA-driven research projects globally, such as precision medicine initiatives in China and the United States, should provide significant revenue and profit tailwinds for years, if not decades.

The company's machines can cost $1 million, or more, but the company really benefits from the ongoing sale of consumables necessary for these machines to operate. As more machines are deployed, revenue for consumables is growing, and since consumables offer more attractive profit margins, that's fueling earnings growth. Since 2011, Illumina's sales and profit have grown by compounded annual rates of 18% and 21%, respectively.

Although the boom-and-bust nature of research budgets means there will be some quarters that are better than other quarters, I believe Illumina's unlikely to lose its dominant position in this market, and if I'm right, then a trend over time toward medicine that aims to correct genetic abnormalities will provide significant opportunities for Illumina to reward investors. The company's newest machines could accelerate that trend, because they could eventually help lower the cost of sequencing genomes from $1,000 today to $100. The NovaSeq 6000, which costs about $1 million, began shipping this quarter.

Good news! Surgery is getting increasingly more precise, and that's reducing recovery times and improving patient outcomes.

At the forefront of this trend is robotics, and when it comes to robotic surgery, there's no better pure-play stock to buy than Intuitive Surgical.

Using research pioneered by DARPA for use on the battlefield, Intuitive Surgical pioneered the development of sophisticated machines that allow surgeons to control robotic arms when performing many surgeries, including prostate and gynecological procedures. Advances in these robotic systems should significantly expand their use in more procedures in the coming decades.

Today, there are almost 4,000 of Intuitive Surgical's da Vinci robotic systems installed at hospitals, and similar to Illumina, the high cost of these machines is only part of the reason I think Intuitive Surgical's going to be a big, long-term winner.

A da Vinci system can cost a hospital $1.5 million, but the average amount spent on replacement instruments and accessories used in operations is especially lucrative. According to management, every da Vinci procedure can produce up to $3,500 in instrument and accessory revenue. That's a lot of margin-friendly revenue when you consider that over 4 million procedures have been performed with these systems, including 750,000 last year alone. Instrument and accessory revenue totaled $1.4 billion, or about 70% of sales, in 2016.

SOURCE: INTUITIVE SURGICAL.

As robotic surgery systems improve, surgeons become more comfortable with them, and as use expands into new areas, such as colorectal surgery and hernia repair, it wouldn't surprise me if Intuitive Surgical's sales and profit march considerably higher over the coming decade.

Overwhelmingly, Americans view on medical marijuana has shifted positive, and as a result, over two dozen U.S. states have passed pro-medical marijuana laws that break down barriers to access.

IMAGE SOURCE: GETTY IMAGES.

While no one knows how a new administration in Washington, D.C. may affect marijuana momentum in the short term, the long-term potential for marijuana to gain ground as a viable alternative medicine is big.

GW Pharmaceuticals could be the drugmaker best positioned to profit from a widespread embrace of medical cannabis. The company's been working on marijuana-based medicines since the 1990s, and it could soon launch its first marijuana derived drug in America.

Last year, GW Pharmaceuticals reported trial results from three separate studies showing that a purified formulation of cannabidiol, or CBD, can reduce the number of seizures experienced monthly by patients with tough-to-treat forms of childhood-onset epilepsy. Specifically, GW Pharmaceuticals showed that patients receiving its Epidiolex experienced about 40% fewer seizures than they did before beginning treatment.

The positive efficacy, plus a safety profile that doesn't seem to be raising eyebrows, suggests that Epidiolex could become an important new drug used by doctors to treat patients who don't respond well to existing epilepsy medications. GW Pharmaceuticals estimates that up to one-third of the 2.2 million epilepsy patients living in the U.S. aren't responding adequately to existing medication.

If the FDA green-lights Epidiolex (management plans to submit an application to the regulator soon), then it can be prescribed by doctors nationwide, regardless of whether medical-marijuana laws have been passed in the doctor's state. That's potentially a huge advantage over medical dispensaries, which only market products without the FDA's blessing in states that have passed laws that are friendly to medical marijuana.

GW Pharmaceuticals isn't stopping its marijuana research with epilepsy, either. The company's studying marijuana cannabinoids in other indications, and while results in the past haven't panned out nearly as well as in epilepsy trials, that doesn't mean programs evaluating it in schizophrenia and autism won't bear fruit.

Because I believe that most Americans will continue supporting access to medical marijuana, and that improving perceptions will remove the stigma associated with its use, the future could prove to be very bright for GW Pharmaceuticals shareholders.

Todd Campbell has no position in any stocks mentioned.His clients may have positions in the companies mentioned.The Motley Fool owns shares of and recommends Illumina and Intuitive Surgical. The Motley Fool has a disclosure policy.

Read more here:
3 Huge Healthcare Trends and How to Invest in Them (Hint: One Is Marijuana) - Motley Fool

To maximize a child’s development, genetics provide important insight – Medical Xpress

February 28, 2017 A South African child is evaluated as part of a Stellenbosch University study. Credit: Stellenbosch University

A child's genetic make-up can play a large, hidden role in the success of efforts to maximize his or her development, South African research suggests.

The study, published February 28 in PLoS Medicine and supported by the Government of Canada through Grand Challenges Canada's Saving Brains program, sheds new light on why some children benefit more than others from interventions and raises complex questions about psychosocial intervention programs in future.

In a study led by Professor Mark Tomlinson of Stellenbosch University, the study followed-up an intervention implemented between 1999 and 2003, in which expectant mothers underwent mentoring to improve attachment with their childrenattachment being a measure of a child's psychological security, and predictive of future wellbeing. In the original study, a control group of roughly equal size was composed of expectant mothers who did not receive mentoring.

The original study concluded that the intervention had a small-to-moderate effect on mother-child attachment, evaluated once the children reached 18 months of age.

The follow-up study, conducted thirteen years after the intervention, re-examined the original attachment results and revealed something surprising: the intervention had in fact worked well for toddlers who had a particular genetic characteristic.

Conducted in collaboration with colleagues from the University of Reading, University College London, and Western University, the study re-enrolled and conducted genetic tests on 279 of the original 449 children.

220 children had both genetic and attachment data, enabling the investigators to test whether the original attachment outcomes were influenced by their genes.

The researchers factored in whether the child had the short or long form of gene SLC6A4the serotonin transporter gene, which is involved in nerve signalling, and which other studies have linked to anxiety, depression and other conditions. Serotonin is popularly thought to contribute to feelings of well-being and happiness.

The attachment of children with the short form of the gene, and whose pregnant mothers were mentored, were almost four times more likely to be securely attached to their mothers at 18 months old (84 percent were secure) than children carrying the short form whose mothers did not receive mentoring (58 percent were secure).

Meanwhile, children with the long gene were apparently unaffected by their mother's training or lack thereof: in both cases, the rate of secure attachment was almost identical (70 and 71 percent).

Subject to further validation, says Professor Tomlinson, the insight has "important implications for scientists designing and evaluating interventions to benefit as many people as possible in South Africa and worldwide."

"Without taking genetics into account, it is possible that other studies have under-estimated the impact of their interventions, as we originally did."

Says lead author Dr. Barak Morgan of the University of Cape Town: "The immediate significance of this research is the revelation that in principle, and probably in many cases in practice too, the effectiveness of interventions has been mis-measuredunder-estimated for genetically susceptible individuals and over-estimated for those who are genetically less susceptible. But even more worrying is the implication that the negative consequences of not receiving an intervention also differ by genotype."

"This is an enormously important insight because, in this case, the subgroup with the short form of the SLC6A4 gene is also the one with the most to lose if not helped."

"Individuals with the long form of the gene, on the other hand, appear less sensitive and derived little benefit from the same intervention, and little detriment from not getting it."

Adds Professor Tomlinson: "In the original study, we did not see such a big impact from this intervention because only those with the short gene improved, and this improvement was 'diluted' by the large number of children with the long gene who did not improve."

The researchers caution that, among other limitations, this study involved a relatively small sample and only measured one gene and one outcome (attachment).

Dr. Morgan stressed: "We are certainly not saying that only some people should receive the interventionthose who are 'susceptible' to improving from it. There is little scientific justification for this. For example, many children with the non-susceptible long genotype of the SLC6A4 gene may carry the susceptible form of another gene which renders them much more likely to benefit from the same intervention but for a different but equally important outcome.

"Going forward, the implications are therefore two-fold. Firstly, measuring genetic differences allows for proper assessment of the effectiveness or lack of effectiveness of an intervention for a particular outcome in different individuals. Secondly, this information can then be used to find out how to intervene effectively for allto guide what might be done to improve outcomes for a non-responsive gene-intervention interaction while continuing to optimise outcomes for the responsive one."

Says Dr. Karlee Silver, Vice President Programs of Grand Challenges Canada: "This work is fundamentally about better understanding the impact of interventions which is an important step forward to creating a world where every child can survive and thrive."

Says Dr. Peter A. Singer, Chief Executive Officer of Grand Challenges Canada: "This is a startling finding that changes the way I think about child development. Why is it important? Because child development is the ladder of social mobility used to climb out of the hole of inequity by millions of children around the world."

Explore further: Study explores how to tell children they have HIV

More information: "Serotonin Transporter Gene (SLC6A4) Polymorphism and Susceptibility to a Home-Visiting Maternal-Infant Attachment Intervention Delivered by Community Health Workers in South Africa: Re-analysis of a Randomized Controlled Trial" DOI: 10.1371/journal.pmed.1002237

Journal reference: PLoS Medicine

Provided by: Grand Challenges Canada

For the past two years, Rachel King, PhD, MPH, an academic coordinator at UCSF Global Health Sciences, has been helping Ugandan parents and caregivers find developmentally appropriate ways to tell their children that the ...

A low sense of attachment between an expectant mother and her unborn child could be associated with some infant developmental delays.

A long-term study of mother-child pairs in Pakistan has found that the children turn out pretty much the same, whether or not their mothers received treatment for depression during pregnancy.

A cognitive-behavioral intervention known as problem-solving education (PSE) may help reduce parental stress and depressive symptoms immediately after their child is diagnosed with autism spectrum disorder (ASD), according ...

What social skills does a three-year-old bring to interactions with a new peer partner? If he has strong bonds to his parents, the child is likely to be a positive, responsive playmate, and he'll be able to adapt to a difficult ...

Children with difficult temperaments are often the most affected by the quality of their relationships with their caregivers. New research suggests that highly irritable children who have secure attachments to their mothers ...

A child's genetic make-up can play a large, hidden role in the success of efforts to maximize his or her development, South African research suggests.

An important learning process is impaired in adolescents who were abused as children, a University of Pittsburgh researcher has found, and this impairment contributes to misbehavior patterns later in life.

Happy memories spring to mind much faster than sad, scary or peaceful ones. Moreover, if you listen to happy or peaceful music, you recall positive memories, whereas if you listen to emotionally scary or sad music, you recall ...

Sending stuffed animals for a sleepover at the library encourages children to read with them, even long after the sleepover took place, say researchers in a new study in Heliyon. For the first time, the study proves stuffed ...

Many musicians are familiar with the phenomenon: Their music sounds much better while performing it live than when they listen back to the recorded version. Scientists at the Max Planck Institute for Human Cognitive and Brain ...

A growing body of research has shown that people's mindsets have measurable physical results.

Please sign in to add a comment. Registration is free, and takes less than a minute. Read more

View post:
To maximize a child's development, genetics provide important insight - Medical Xpress

RNAi.technology – RNAi Medicine, RNAi Developments, Gene …

Optogenetics From Wikipedia, the free encyclopedia Optogenetics (from Greek optiks], meaning seen, visible) is a biological technique which involves the use of light to control cells in living tissue, typically neurons, that have been genetically modified to express light-sensitive ion channels. It is a neuromodulation method employed in neuroscience that uses a combination of techniques from optics and genetics to control and monitor the activities of individual neurons in living tissueeven within freely-moving animalsand to precisely measure the effects of those manipulations in real-time.[1] The key reagents used in optogenetics Read more

GeneQuine develops drugs that are based on gene therapy. The concept of gene therapy is to introduce genetic material into the patients own cells in the body. The cells produce then a therapeutic protein according to the template that the introduced genetic material (DNA) provides

GeneQuine is focused on the development of gene therapy agents for the treatment of osteoarthritis. Osteoarthritis is a degenerative joint disorder characterized by cartilage loss and inflammation. Patients affected by osteoarthritis experience joint pain as well as swelling and stiffness of the joints leading to Read more

Genome Editing with CRISPR-Cas9

Heres a short list of some common diseases that might be curable or preventable with gene editing:

Autism Breast cancer Colon cancer Hemophilia Huntingtons disease Marfan, Parkinsons Prostate cancer Retinitis pigmentosa Sickle cell Skin cancer Tay-Sachs Wilson Duchenne muscular dystrophy Crohns Color blindness Cystic fibrosis Down syndrome Polycystic kidney Turner syndrome.

There are hundreds of other more rare genetic disorders. Read more

Gene therapy is a well-suited approach for the treatment of SMA due to the monogenic nature of the diseasemeaning its caused by the deletion of or mutations in a single gene. AVXS-101 is our clinical-stage, proprietary gene therapy candidate of a one-time, intravenous treatment for SMA Type 1designed to prevent further muscle degeneration caused by SMA through:

Delivery of a fully functional human SMN gene into target motor neuron cells Production of sufficient levels of SMN protein required to improve motor neuron function Rapid onset of effect in addition to Read more

Advantagene Inc. Bluebird Bio Genethon Human Stem Cells Institute Oxford BioMedica Plc Sanofi Shanghai Sunway Biotech Co. Ltd. Sibiono GeneTech Co. Ltd. Spark Therapeutics, LLC UniQure N.V. Vical Inc. ViroMed Co. Ltd. dba VM BioPharma Read more

The rest is here:
RNAi.technology - RNAi Medicine, RNAi Developments, Gene ...

Neanderthal DNA contributes to human gene expression – Science Daily


Science Daily
Neanderthal DNA contributes to human gene expression
Science Daily
"Even 50,000 years after the last human-Neanderthal mating, we can still see measurable impacts on gene expression," says geneticist and study co-author Joshua Akey of the University of Washington School of Medicine. "And those variations in gene ...
Neanderthal DNA Influences Your Biology A Lot More Than You RealizeIFLScience
Neanderthal DNA influences our height, schizophrenia risk: StudyThe Hans India

all 52 news articles »

Read more from the original source:
Neanderthal DNA contributes to human gene expression - Science Daily

Regenerative Medicine Has a Bright Future – Healthline

U.S. Army scientists, working with medical technology companies, have successfully tested and used products and techniques that have enabled Army surgeons to replace the severely burned skin of soldiers as well as transplant new hands and even faces.

At Duke University, researchers are studying zebra fish to learn how science and medicine might someday be able to regenerate severed human spinal cords.

These examples one already in practice and the other in the early research stages illustrate the potential that regenerative medicine offers for the future of medical care.

This research aims to go beyond easing the pain of life-threatening illnesses by changing the way diseases affect the body and then eradicating them.

The vast majority of currently available treatments for chronic and/or life-threatening diseases are palliative, Morrie Ruffin, managing director of the Alliance for Regenerative Medicine (ARM), told Healthline.

ARM, based in Washington, D.C., is considered the preeminent global advocate for regenerative and advanced therapies.

Other treatments delay disease progression and the onset of complications associated with the underlying illness, he said. Very few therapies in use today are capable of curing or significantly changing the course of disease.

Regenerative medicine has the unique ability to alter the fundamental mechanisms of disease, and thereby offer treatment options to patients where there is significant unmet medical need.

And it has the potential to address the underlying causes of disease, Ruffin said, representing a new and growing paradigm in human health.

The field encompasses a number of different technologies, including cell, gene, and tissue-based therapies.

Read more: Re-growing teeth and healing wounds without scars

With the Army breakthroughs, government investment was key.

The U.S. Department of Defense (DOD) has invested more than $250 million in regenerative medicine research over the past decade in an effort to make promising technologies available to wounded service members.

Dr. Wendy Dean is medical officer for the Tissue Injury and Regenerative Medicine Project Management Office at the U.S. Army Medical Materiel Development Activity at Fort Detrick, Md., home to the Armys Medical Research and Materiel Command.

Those investments have yielded a stress-shielding surgical bandage, Embrace, to reduce scarring after surgery, Dean told Healthline. The research has also enabled tremendous progress in burn care, allowing surgeons to improve recovery from severe burns with the use of novel skin replacement strategies, such as ReCell spray-on skin, or skin substitutes such as StrataGraft. These skin replacement methods reduce or eliminate the need for donor sites, a frequent request of burn patients.

These revolutionary products were not developed by the Army, Dean said, but were supported with research funding, initially through the Armed Forces Institute of Regenerative Medicine.

The DOD also has invested in hand and face transplantation efforts for service members and civilians whose injuries are so severe that conventional reconstruction is insufficient, she said.

Dean noted that DOD funding has supported 13 hand transplants to date, including a transplant for retired Sgt. Brendan Marrocco in 2012. He was the first service member to survive quadrilateral amputations sustained in combat. The funding also supported eight face transplants.

The Armys goal is to heal those injured in battle.

Regenerative medicine is still young, but it has shown tremendous progress over the last decade, Dean said. Our mission is to make wounded warriors whole by restoring form, function, and appearance. This field offers the best hope to someday fully restore lost tissue with tissue that is structurally, functionally, and aesthetically a perfect match. It may be years before the vision is a widespread reality, but the field is well on its way.

Read more: Regenerative medicine doctor says forget the pills

At Duke University, Kenneth Poss, professor of cell biology, and director of the Regeneration Next initiative, was the senior investigator for a study of spinal cord regeneration in zebra fish.

Those findings were published in November in the journal ScienceDaily.

In my lab, we are researching genetic factors that enable regeneration of tissues such as heart and spinal in nonmammalian animals like zebra fish, Poss told Healthline. A scientist in my lab, Mayssa Mokalled, led a study finding that a gene called connective tissue growth factor [CTGF] is important for spinal cord regeneration in zebra fish after an injury that completely severs the cord.

CTGF is necessary to stimulate cells called glia to form a tissue bridge across the severed parts of the spinal cord an early step in spinal cord regeneration.

Within eight weeks, the scientists found that zebra fish regenerate a severed spinal cord, including nerve cells, and fully reverse their paralysis.

Developing techniques to treat and reverse spinal cord damage, a paralyzing and often fatal injury, is a pressing need in regenerative medicine, Poss said.

Our findings present a step toward understanding which glial cells can be encouraged to help heal the spinal cord, and how to stimulate this activity, he said. This is just the first step in many before the findings could be applied to humans.

Poss is already planning trials with mice that he hopes to start in the next few months. Mice represent an important stage in applying his latest findings, he said.

Read more: Should you store or donate your childs umbilical cord blood?

So, why is regenerative medicine important?

Regenerative medicine seeks ways to re-grow or engineer healthy tissue without the need for transplants, Poss said. On a global scale, theres a tremendous organ shortage, and transplantation is an expensive and nonpermanent solution.

Imagine the number of lives that could be improved if, for example, we could find ways to use the bodys innate healing mechanisms to regenerate heart muscle in patients that are spiraling toward heart failure after a heart attack.

Imagine how many lives could be improved if we could find interventions that restore functional spinal cord tissue and reverse paralysis.

Ruffin of ARM sees a promising future for regenerative medicine.

We will continue to see the development of additional regenerative medicine therapies for a broad number of acute and chronic, inherited and acquired diseases and disorders, he said. Therapies in this area will continue to advance along the regulatory pathway, many of which are entering phase III clinical trials this year.

In fact, in the next two years, we are anticipating a number of U.S. and E.U. approvals in the cell and gene therapy sector, including therapies that address certain types of cancers, debilitating retinal disorders, rare genetic diseases, and autoimmune conditions. We also expect to see sustained investment, which will help fuel growth and product development within this sector.

A number of cell and gene therapies and technology platforms are demonstrating real potential to address areas of significant unmet medical need, Ruffin said.

These include cell therapies for blood cancers and solid tumors; gene therapies for rare genetic diseases as well as chronic conditions; and gene editing for the precise targeting and modification of genetic material of a patients cells to cure a broad range of diseases with a single treatment.

Poss at Duke talked about the ultimate quest.

Regenerative medicine has been most successful in restoring or replacing the hematopoietic tissue that creates blood, he said.

We still lack successful regenerative therapies for most tissues, Poss said. The future of regenerative medicine the holy grail will be stimulating the regeneration of healthy tissue in patients without adding cells or manufactured tissue.

Working out the details of innate mechanisms of regeneration in animals like salamanders, zebra fish, and mice, can inform this approach, he said. So can improvement in factor delivery and genome editing applications to encourage the regeneration of healthy tissue.

Ultimately, Poss said, regenerative medicine will change the toolbox of physicians and surgeons, with major impact on outcomes of diabetes, spinal cord injuries, neurodegenerative disease, and heart failure.

ARM says the public does not realize how far the field has progressed in recent years.

Currently, there are more than 20 regenerative medicine products on the market, Ruffin said, primarily in the therapeutic areas of oncology, musculoskeletal and cardiovascular repair, and wound healing.

More than 800 clinical trials are now underway to evaluate regenerative advanced therapies in a vast array of therapeutic categories, he said.

Were seeing a significant focus on oncology, cardiovascular disease, and neurodegenerative diseases, with more than 60 percent of trials falling into one of these three categories, he added. Even though the majority of people perceive regenerative medicine as something of the future, its actually here and now.

Read the original post:
Regenerative Medicine Has a Bright Future - Healthline

Gene – Wikipedia

This article is about the heritable unit for transmission of biological traits. For other uses, see Gene (disambiguation).

A gene is a locus (or region) of DNA which is made up of nucleotides and is the molecular unit of heredity.[1][2]:Glossary The transmission of genes to an organism's offspring is the basis of the inheritance of phenotypic traits. Most biological traits are under the influence of polygenes (many different genes) as well as geneenvironment interactions. Some genetic traits are instantly visible, such as eye colour or number of limbs, and some are not, such as blood type, risk for specific diseases, or the thousands of basic biochemical processes that comprise life.

Genes can acquire mutations in their sequence, leading to different variants, known as alleles, in the population. These alleles encode slightly different versions of a protein, which cause different phenotype traits. Colloquial usage of the term "having a gene" (e.g., "good genes," "hair colour gene") typically refers to having a different allele of the gene. Genes evolve due to natural selection or survival of the fittest of the alleles.

The concept of a gene continues to be refined as new phenomena are discovered.[3] For example, regulatory regions of a gene can be far removed from its coding regions, and coding regions can be split into several exons. Some viruses store their genome in RNA instead of DNA and some gene products are functional non-coding RNAs. Therefore, a broad, modern working definition of a gene is any discrete locus of heritable, genomic sequence which affect an organism's traits by being expressed as a functional product or by regulation of gene expression.[4][5]

The existence of discrete inheritable units was first suggested by Gregor Mendel (18221884).[6] From 1857 to 1864, he studied inheritance patterns in 8000 common edible pea plants, tracking distinct traits from parent to offspring. He described these mathematically as 2ncombinations where n is the number of differing characteristics in the original peas. Although he did not use the term gene, he explained his results in terms of discrete inherited units that give rise to observable physical characteristics. This description prefigured the distinction between genotype (the genetic material of an organism) and phenotype (the visible traits of that organism). Mendel was also the first to demonstrate independent assortment, the distinction between dominant and recessive traits, the distinction between a heterozygote and homozygote, and the phenomenon of discontinuous inheritance.

Prior to Mendel's work, the dominant theory of heredity was one of blending inheritance, which suggested that each parent contributed fluids to the fertilisation process and that the traits of the parents blended and mixed to produce the offspring. Charles Darwin developed a theory of inheritance he termed pangenesis, from Greek pan ("all, whole") and genesis ("birth") / genos ("origin").[7][8] Darwin used the term gemmule to describe hypothetical particles that would mix during reproduction.

Mendel's work went largely unnoticed after its first publication in 1866, but was rediscovered in the late 19th century by Hugo de Vries, Carl Correns, and Erich von Tschermak, who (claimed to have) reached similar conclusions in their own research.[9] Specifically, in 1889, Hugo de Vries published his book Intracellular Pangenesis,[10] in which he postulated that different characters have individual hereditary carriers and that inheritance of specific traits in organisms comes in particles. De Vries called these units "pangenes" (Pangens in German), after Darwin's 1868 pangenesis theory.

Sixteen years later, in 1905, the word genetics was first used by William Bateson,[11] while Eduard Strasburger, amongst others, still used the term pangene for the fundamental physical and functional unit of heredity.[12] In 1909 the Danish botanist Wilhelm Johannsen shortened the name to "gene". [13]

Advances in understanding genes and inheritance continued throughout the 20th century. Deoxyribonucleic acid (DNA) was shown to be the molecular repository of genetic information by experiments in the 1940s to 1950s.[14][15] The structure of DNA was studied by Rosalind Franklin and Maurice Wilkins using X-ray crystallography, which led James D. Watson and Francis Crick to publish a model of the double-stranded DNA molecule whose paired nucleotide bases indicated a compelling hypothesis for the mechanism of genetic replication.[16][17]

In the early 1950s the prevailing view was that the genes in a chromosome acted like discrete entities, indivisible by recombination and arranged like beads on a string. The experiments of Benzer using mutants defective in the rII region of bacteriophage T4 (1955-1959) showed that individual genes have a simple linear structure and are likely to be equivalent to a linear section of DNA.[18][19]

Collectively, this body of research established the central dogma of molecular biology, which states that proteins are translated from RNA, which is transcribed from DNA. This dogma has since been shown to have exceptions, such as reverse transcription in retroviruses. The modern study of genetics at the level of DNA is known as molecular genetics.

In 1972, Walter Fiers and his team at the University of Ghent were the first to determine the sequence of a gene: the gene for Bacteriophage MS2 coat protein.[20] The subsequent development of chain-termination DNA sequencing in 1977 by Frederick Sanger improved the efficiency of sequencing and turned it into a routine laboratory tool.[21] An automated version of the Sanger method was used in early phases of the Human Genome Project.[22]

The theories developed in the 1930s and 1940s to integrate molecular genetics with Darwinian evolution are called the modern evolutionary synthesis, a term introduced by Julian Huxley.[23] Evolutionary biologists subsequently refined this concept, such as George C. Williams' gene-centric view of evolution. He proposed an evolutionary concept of the gene as a unit of natural selection with the definition: "that which segregates and recombines with appreciable frequency."[24]:24 In this view, the molecular gene transcribes as a unit, and the evolutionary gene inherits as a unit. Related ideas emphasizing the centrality of genes in evolution were popularized by Richard Dawkins.[25][26]

The vast majority of living organisms encode their genes in long strands of DNA (deoxyribonucleic acid). DNA consists of a chain made from four types of nucleotide subunits, each composed of: a five-carbon sugar (2'-deoxyribose), a phosphate group, and one of the four bases adenine, cytosine, guanine, and thymine.[2]:2.1

Two chains of DNA twist around each other to form a DNA double helix with the phosphate-sugar backbone spiralling around the outside, and the bases pointing inwards with adenine base pairing to thymine and guanine to cytosine. The specificity of base pairing occurs because adenine and thymine align to form two hydrogen bonds, whereas cytosine and guanine form three hydrogen bonds. The two strands in a double helix must therefore be complementary, with their sequence of bases matching such that the adenines of one strand are paired with the thymines of the other strand, and so on.[2]:4.1

Due to the chemical composition of the pentose residues of the bases, DNA strands have directionality. One end of a DNA polymer contains an exposed hydroxyl group on the deoxyribose; this is known as the 3'end of the molecule. The other end contains an exposed phosphate group; this is the 5'end. The two strands of a double-helix run in opposite directions. Nucleic acid synthesis, including DNA replication and transcription occurs in the 5'3'direction, because new nucleotides are added via a dehydration reaction that uses the exposed 3'hydroxyl as a nucleophile.[27]:27.2

The expression of genes encoded in DNA begins by transcribing the gene into RNA, a second type of nucleic acid that is very similar to DNA, but whose monomers contain the sugar ribose rather than deoxyribose. RNA also contains the base uracil in place of thymine. RNA molecules are less stable than DNA and are typically single-stranded. Genes that encode proteins are composed of a series of three-nucleotide sequences called codons, which serve as the "words" in the genetic "language". The genetic code specifies the correspondence during protein translation between codons and amino acids. The genetic code is nearly the same for all known organisms.[2]:4.1

The total complement of genes in an organism or cell is known as its genome, which may be stored on one or more chromosomes. A chromosome consists of a single, very long DNA helix on which thousands of genes are encoded.[2]:4.2 The region of the chromosome at which a particular gene is located is called its locus. Each locus contains one allele of a gene; however, members of a population may have different alleles at the locus, each with a slightly different gene sequence.

The majority of eukaryotic genes are stored on a set of large, linear chromosomes. The chromosomes are packed within the nucleus in complex with storage proteins called histones to form a unit called a nucleosome. DNA packaged and condensed in this way is called chromatin.[2]:4.2 The manner in which DNA is stored on the histones, as well as chemical modifications of the histone itself, regulate whether a particular region of DNA is accessible for gene expression. In addition to genes, eukaryotic chromosomes contain sequences involved in ensuring that the DNA is copied without degradation of end regions and sorted into daughter cells during cell division: replication origins, telomeres and the centromere.[2]:4.2 Replication origins are the sequence regions where DNA replication is initiated to make two copies of the chromosome. Telomeres are long stretches of repetitive sequence that cap the ends of the linear chromosomes and prevent degradation of coding and regulatory regions during DNA replication. The length of the telomeres decreases each time the genome is replicated and has been implicated in the aging process.[29] The centromere is required for binding spindle fibres to separate sister chromatids into daughter cells during cell division.[2]:18.2

Prokaryotes (bacteria and archaea) typically store their genomes on a single large, circular chromosome. Similarly, some eukaryotic organelles contain a remnant circular chromosome with a small number of genes.[2]:14.4 Prokaryotes sometimes supplement their chromosome with additional small circles of DNA called plasmids, which usually encode only a few genes and are transferable between individuals. For example, the genes for antibiotic resistance are usually encoded on bacterial plasmids and can be passed between individual cells, even those of different species, via horizontal gene transfer.[30]

Whereas the chromosomes of prokaryotes are relatively gene-dense, those of eukaryotes often contain regions of DNA that serve no obvious function. Simple single-celled eukaryotes have relatively small amounts of such DNA, whereas the genomes of complex multicellular organisms, including humans, contain an absolute majority of DNA without an identified function.[31] This DNA has often been referred to as "junk DNA". However, more recent analyses suggest that, although protein-coding DNA makes up barely 2% of the human genome, about 80% of the bases in the genome may be expressed, so the term "junk DNA" may be a misnomer.[5]

The structure of a gene consists of many elements of which the actual protein coding sequence is often only a small part. These include DNA regions that are not transcribed as well as untranslated regions of the RNA.

Firstly, flanking the open reading frame, all genes contain a regulatory sequence that is required for their expression. In order to be expressed, genes require a promoter sequence. The promoter is recognized and bound by transcription factors and RNA polymerase to initiate transcription.[2]:7.1 A gene can have more than one promoter, resulting in messenger RNAs (mRNA) that differ in how far they extend in the 5'end.[32] Promoter regions have a consensus sequence, however highly transcribed genes have "strong" promoter sequences that bind the transcription machinery well, whereas others have "weak" promoters that bind poorly and initiate transcription less frequently.[2]:7.2Eukaryotic promoter regions are much more complex and difficult to identify than prokaryotic promoters.[2]:7.3

Additionally, genes can have regulatory regions many kilobases upstream or downstream of the open reading frame. These act by binding to transcription factors which then cause the DNA to loop so that the regulatory sequence (and bound transcription factor) become close to the RNA polymerase binding site.[33] For example, enhancers increase transcription by binding an activator protein which then helps to recruit the RNA polymerase to the promoter; conversely silencers bind repressor proteins and make the DNA less available for RNA polymerase.[34]

The transcribed pre-mRNA contains untranslated regions at both ends which contain a ribosome binding site, terminator and start and stop codons.[35] In addition, most eukaryotic open reading frames contain untranslated introns which are removed before the exons are translated. The sequences at the ends of the introns, dictate the splice sites to generate the final mature mRNA which encodes the protein or RNA product.[36]

Many prokaryotic genes are organized into operons, with multiple protein-coding sequences that are transcribed as a unit.[37][38] The genes in an operon are transcribed as a continuous messenger RNA, referred to as a polycistronic mRNA. The term cistron in this context is equivalent to gene. The transcription of an operons mRNA is often controlled by a repressor that can occur in an active or inactive state depending on the presence of certain specific metabolites.[39] When active, the repressor binds to a DNA sequence at the beginning of the operon, called the operator region, and represses transcription of the operon; when the repressor is inactive transcription of the operon can occur (see e.g. Lac operon). The products of operon genes typically have related functions and are involved in the same regulatory network.[2]:7.3

Defining exactly what section of a DNA sequence comprises a gene is difficult.[3]Regulatory regions of a gene such as enhancers do not necessarily have to be close to the coding sequence on the linear molecule because the intervening DNA can be looped out to bring the gene and its regulatory region into proximity. Similarly, a gene's introns can be much larger than its exons. Regulatory regions can even be on entirely different chromosomes and operate in trans to allow regulatory regions on one chromosome to come in contact with target genes on another chromosome.[40][41]

Early work in molecular genetics suggested the concept that one gene makes one protein. This concept (originally called the one gene-one enzyme hypothesis) emerged from an influential 1941 paper by George Beadle and Edward Tatum on experiments with mutants of the fungus Neurospora crassa.[42]Norman Horowitz, an early colleague on the Neurospora research, reminisced in 2004 that these experiments founded the science of what Beadle and Tatum called biochemical genetics. In actuality they proved to be the opening gun in what became molecular genetics and all the developments that have followed from that.[43] The one gene-one protein concept has been refined since the discovery of genes that can encode multiple proteins by alternative splicing and coding sequences split in short section across the genome whose mRNAs are concatenated by trans-splicing.[5][44][45]

A broad operational definition is sometimes used to encompass the complexity of these diverse phenomena, where a gene is defined as a union of genomic sequences encoding a coherent set of potentially overlapping functional products.[11] This definition categorizes genes by their functional products (proteins or RNA) rather than their specific DNA loci, with regulatory elements classified as gene-associated regions.[11]

In all organisms, two steps are required to read the information encoded in a gene's DNA and produce the protein it specifies. First, the gene's DNA is transcribed to messenger RNA (mRNA).[2]:6.1 Second, that mRNA is translated to protein.[2]:6.2 RNA-coding genes must still go through the first step, but are not translated into protein.[46] The process of producing a biologically functional molecule of either RNA or protein is called gene expression, and the resulting molecule is called a gene product.

The nucleotide sequence of a gene's DNA specifies the amino acid sequence of a protein through the genetic code. Sets of three nucleotides, known as codons, each correspond to a specific amino acid.[2]:6 The principle that three sequential bases of DNA code for each amino acid was demonstrated in 1961 using frameshift mutations in the rIIB gene of bacteriophage T4[47] (see Crick, Brenner et al. experiment).

Additionally, a "start codon", and three "stop codons" indicate the beginning and end of the protein coding region. There are 64possible codons (four possible nucleotides at each of three positions, hence 43possible codons) and only 20standard amino acids; hence the code is redundant and multiple codons can specify the same amino acid. The correspondence between codons and amino acids is nearly universal among all known living organisms.[48]

Transcription produces a single-stranded RNA molecule known as messenger RNA, whose nucleotide sequence is complementary to the DNA from which it was transcribed.[2]:6.1 The mRNA acts as an intermediate between the DNA gene and its final protein product. The gene's DNA is used as a template to generate a complementary mRNA. The mRNA matches the sequence of the gene's DNA coding strand because it is synthesised as the complement of the template strand. Transcription is performed by an enzyme called an RNA polymerase, which reads the template strand in the 3' to 5'direction and synthesizes the RNA from 5' to 3'. To initiate transcription, the polymerase first recognizes and binds a promoter region of the gene. Thus, a major mechanism of gene regulation is the blocking or sequestering the promoter region, either by tight binding by repressor molecules that physically block the polymerase, or by organizing the DNA so that the promoter region is not accessible.[2]:7

In prokaryotes, transcription occurs in the cytoplasm; for very long transcripts, translation may begin at the 5'end of the RNA while the 3'end is still being transcribed. In eukaryotes, transcription occurs in the nucleus, where the cell's DNA is stored. The RNA molecule produced by the polymerase is known as the primary transcript and undergoes post-transcriptional modifications before being exported to the cytoplasm for translation. One of the modifications performed is the splicing of introns which are sequences in the transcribed region that do not encode protein. Alternative splicing mechanisms can result in mature transcripts from the same gene having different sequences and thus coding for different proteins. This is a major form of regulation in eukaryotic cells and also occurs in some prokaryotes.[2]:7.5[49]

Translation is the process by which a mature mRNA molecule is used as a template for synthesizing a new protein.[2]:6.2 Translation is carried out by ribosomes, large complexes of RNA and protein responsible for carrying out the chemical reactions to add new amino acids to a growing polypeptide chain by the formation of peptide bonds. The genetic code is read three nucleotides at a time, in units called codons, via interactions with specialized RNA molecules called transfer RNA (tRNA). Each tRNA has three unpaired bases known as the anticodon that are complementary to the codon it reads on the mRNA. The tRNA is also covalently attached to the amino acid specified by the complementary codon. When the tRNA binds to its complementary codon in an mRNA strand, the ribosome attaches its amino acid cargo to the new polypeptide chain, which is synthesized from amino terminus to carboxyl terminus. During and after synthesis, most new proteins must fold to their active three-dimensional structure before they can carry out their cellular functions.[2]:3

Genes are regulated so that they are expressed only when the product is needed, since expression draws on limited resources.[2]:7 A cell regulates its gene expression depending on its external environment (e.g. available nutrients, temperature and other stresses), its internal environment (e.g. cell division cycle, metabolism, infection status), and its specific role if in a multicellular organism. Gene expression can be regulated at any step: from transcriptional initiation, to RNA processing, to post-translational modification of the protein. The regulation of lactose metabolism genes in E. coli (lac operon) was the first such mechanism to be described in 1961.[50]

A typical protein-coding gene is first copied into RNA as an intermediate in the manufacture of the final protein product.[2]:6.1 In other cases, the RNA molecules are the actual functional products, as in the synthesis of ribosomal RNA and transfer RNA. Some RNAs known as ribozymes are capable of enzymatic function, and microRNA has a regulatory role. The DNA sequences from which such RNAs are transcribed are known as non-coding RNA genes.[46]

Some viruses store their entire genomes in the form of RNA, and contain no DNA at all.[51][52] Because they use RNA to store genes, their cellular hosts may synthesize their proteins as soon as they are infected and without the delay in waiting for transcription.[53] On the other hand, RNA retroviruses, such as HIV, require the reverse transcription of their genome from RNA into DNA before their proteins can be synthesized. RNA-mediated epigenetic inheritance has also been observed in plants and very rarely in animals.[54]

Organisms inherit their genes from their parents. Asexual organisms simply inherit a complete copy of their parent's genome. Sexual organisms have two copies of each chromosome because they inherit one complete set from each parent.[2]:1

According to Mendelian inheritance, variations in an organism's phenotype (observable physical and behavioral characteristics) are due in part to variations in its genotype (particular set of genes). Each gene specifies a particular trait with different sequence of a gene (alleles) giving rise to different phenotypes. Most eukaryotic organisms (such as the pea plants Mendel worked on) have two alleles for each trait, one inherited from each parent.[2]:20

Alleles at a locus may be dominant or recessive; dominant alleles give rise to their corresponding phenotypes when paired with any other allele for the same trait, whereas recessive alleles give rise to their corresponding phenotype only when paired with another copy of the same allele. For example, if the allele specifying tall stems in pea plants is dominant over the allele specifying short stems, then pea plants that inherit one tall allele from one parent and one short allele from the other parent will also have tall stems. Mendel's work demonstrated that alleles assort independently in the production of gametes, or germ cells, ensuring variation in the next generation. Although Mendelian inheritance remains a good model for many traits determined by single genes (including a number of well-known genetic disorders) it does not include the physical processes of DNA replication and cell division.[55][56]

The growth, development, and reproduction of organisms relies on cell division, or the process by which a single cell divides into two usually identical daughter cells. This requires first making a duplicate copy of every gene in the genome in a process called DNA replication.[2]:5.2 The copies are made by specialized enzymes known as DNA polymerases, which "read" one strand of the double-helical DNA, known as the template strand, and synthesize a new complementary strand. Because the DNA double helix is held together by base pairing, the sequence of one strand completely specifies the sequence of its complement; hence only one strand needs to be read by the enzyme to produce a faithful copy. The process of DNA replication is semiconservative; that is, the copy of the genome inherited by each daughter cell contains one original and one newly synthesized strand of DNA.[2]:5.2

The rate of DNA replication in living cells was first measured as the rate of phage T4 DNA elongation in phage-infected E. coli and found to be impressively rapid.[57] During the period of exponential DNA increase at 37 C, the rate of elongation was 749 nucleotides per second.

After DNA replication is complete, the cell must physically separate the two copies of the genome and divide into two distinct membrane-bound cells.[2]:18.2 In prokaryotes(bacteria and archaea) this usually occurs via a relatively simple process called binary fission, in which each circular genome attaches to the cell membrane and is separated into the daughter cells as the membrane invaginates to split the cytoplasm into two membrane-bound portions. Binary fission is extremely fast compared to the rates of cell division in eukaryotes. Eukaryotic cell division is a more complex process known as the cell cycle; DNA replication occurs during a phase of this cycle known as S phase, whereas the process of segregating chromosomes and splitting the cytoplasm occurs during M phase.[2]:18.1

The duplication and transmission of genetic material from one generation of cells to the next is the basis for molecular inheritance, and the link between the classical and molecular pictures of genes. Organisms inherit the characteristics of their parents because the cells of the offspring contain copies of the genes in their parents' cells. In asexually reproducing organisms, the offspring will be a genetic copy or clone of the parent organism. In sexually reproducing organisms, a specialized form of cell division called meiosis produces cells called gametes or germ cells that are haploid, or contain only one copy of each gene.[2]:20.2 The gametes produced by females are called eggs or ova, and those produced by males are called sperm. Two gametes fuse to form a diploid fertilized egg, a single cell that has two sets of genes, with one copy of each gene from the mother and one from the father.[2]:20

During the process of meiotic cell division, an event called genetic recombination or crossing-over can sometimes occur, in which a length of DNA on one chromatid is swapped with a length of DNA on the corresponding homologous non-sister chromatid. This can result in reassortment of otherwise linked alleles.[2]:5.5 The Mendelian principle of independent assortment asserts that each of a parent's two genes for each trait will sort independently into gametes; which allele an organism inherits for one trait is unrelated to which allele it inherits for another trait. This is in fact only true for genes that do not reside on the same chromosome, or are located very far from one another on the same chromosome. The closer two genes lie on the same chromosome, the more closely they will be associated in gametes and the more often they will appear together; genes that are very close are essentially never separated because it is extremely unlikely that a crossover point will occur between them. This is known as genetic linkage.[58]

DNA replication is for the most part extremely accurate, however errors (mutations) do occur.[2]:7.6 The error rate in eukaryotic cells can be as low as 108 per nucleotide per replication,[59][60] whereas for some RNA viruses it can be as high as 103.[61] This means that each generation, each human genome accumulates 12 new mutations.[61] Small mutations can be caused by DNA replication and the aftermath of DNA damage and include point mutations in which a single base is altered and frameshift mutations in which a single base is inserted or deleted. Either of these mutations can change the gene by missense (change a codon to encode a different amino acid) or nonsense (a premature stop codon).[62] Larger mutations can be caused by errors in recombination to cause chromosomal abnormalities including the duplication, deletion, rearrangement or inversion of large sections of a chromosome. Additionally, DNA repair mechanisms can introduce mutational errors when repairing physical damage to the molecule. The repair, even with mutation, is more important to survival than restoring an exact copy, for example when repairing double-strand breaks.[2]:5.4

When multiple different alleles for a gene are present in a species's population it is called polymorphic. Most different alleles are functionally equivalent, however some alleles can give rise to different phenotypic traits. A gene's most common allele is called the wild type, and rare alleles are called mutants. The genetic variation in relative frequencies of different alleles in a population is due to both natural selection and genetic drift.[63] The wild-type allele is not necessarily the ancestor of less common alleles, nor is it necessarily fitter.

Most mutations within genes are neutral, having no effect on the organism's phenotype (silent mutations). Some mutations do not change the amino acid sequence because multiple codons encode the same amino acid (synonymous mutations). Other mutations can be neutral if they lead to amino acid sequence changes, but the protein still functions similarly with the new amino acid (e.g. conservative mutations). Many mutations, however, are deleterious or even lethal, and are removed from populations by natural selection. Genetic disorders are the result of deleterious mutations and can be due to spontaneous mutation in the affected individual, or can be inherited. Finally, a small fraction of mutations are beneficial, improving the organism's fitness and are extremely important for evolution, since their directional selection leads to adaptive evolution.[2]:7.6

Genes with a most recent common ancestor, and thus a shared evolutionary ancestry, are known as homologs.[64] These genes appear either from gene duplication within an organism's genome, where they are known as paralogous genes, or are the result of divergence of the genes after a speciation event, where they are known as orthologous genes,[2]:7.6 and often perform the same or similar functions in related organisms. It is often assumed that the functions of orthologous genes are more similar than those of paralogous genes, although the difference is minimal.[65][66]

The relationship between genes can be measured by comparing the sequence alignment of their DNA.[2]:7.6 The degree of sequence similarity between homologous genes is called conserved sequence. Most changes to a gene's sequence do not affect its function and so genes accumulate mutations over time by neutral molecular evolution. Additionally, any selection on a gene will cause its sequence to diverge at a different rate. Genes under stabilizing selection are constrained and so change more slowly whereas genes under directional selection change sequence more rapidly.[67] The sequence differences between genes can be used for phylogenetic analyses to study how those genes have evolved and how the organisms they come from are related.[68][69]

The most common source of new genes in eukaryotic lineages is gene duplication, which creates copy number variation of an existing gene in the genome.[70][71] The resulting genes (paralogs) may then diverge in sequence and in function. Sets of genes formed in this way comprise a gene family. Gene duplications and losses within a family are common and represent a major source of evolutionary biodiversity.[72] Sometimes, gene duplication may result in a nonfunctional copy of a gene, or a functional copy may be subject to mutations that result in loss of function; such nonfunctional genes are called pseudogenes.[2]:7.6

"Orphan" genes, whose sequence shows no similarity to existing genes, are less common than gene duplicates. Estimates of the number of genes with no homologs outside humans range from 18[73] to 60.[74] Two primary sources of orphan protein-coding genes are gene duplication followed by extremely rapid sequence change, such that the original relationship is undetectable by sequence comparisons, and de novo conversion of a previously non-coding sequence into a protein-coding gene.[75] De novo genes are typically shorter and simpler in structure than most eukaryotic genes, with few if any introns.[70] Over long evolutionary time periods, de novo gene birth may be responsible for a significant fraction of taxonomically-restricted gene families.[76]

Horizontal gene transfer refers to the transfer of genetic material through a mechanism other than reproduction. This mechanism is a common source of new genes in prokaryotes, sometimes thought to contribute more to genetic variation than gene duplication.[77] It is a common means of spreading antibiotic resistance, virulence, and adaptive metabolic functions.[30][78] Although horizontal gene transfer is rare in eukaryotes, likely examples have been identified of protist and alga genomes containing genes of bacterial origin.[79][80]

The genome is the total genetic material of an organism and includes both the genes and non-coding sequences.[81]

The genome size, and the number of genes it encodes varies widely between organisms. The smallest genomes occur in viruses (which can have as few as 2 protein-coding genes),[90] and viroids (which act as a single non-coding RNA gene).[91] Conversely, plants can have extremely large genomes,[92] with rice containing >46,000 protein-coding genes.[93] The total number of protein-coding genes (the Earth's proteome) is estimated to be 5million sequences.[94]

Although the number of base-pairs of DNA in the human genome has been known since the 1960s, the estimated number of genes has changed over time as definitions of genes, and methods of detecting them have been refined. Initial theoretical predictions of the number of human genes were as high as 2,000,000.[95] Early experimental measures indicated there to be 50,000100,000 transcribed genes (expressed sequence tags).[96] Subsequently, the sequencing in the Human Genome Project indicated that many of these transcripts were alternative variants of the same genes, and the total number of protein-coding genes was revised down to ~20,000[89] with 13 genes encoded on the mitochondrial genome.[87] Of the human genome, only 12% consists of protein-coding genes,[97] with the remainder being 'noncoding' DNA such as introns, retrotransposons, and noncoding RNAs.[97][98] Every multicellular organism has all its genes in each cell of its body but not every gene functions in every cell .

Essential genes are the set of genes thought to be critical for an organism's survival.[100] This definition assumes the abundant availability of all relevant nutrients and the absence of environmental stress. Only a small portion of an organism's genes are essential. In bacteria, an estimated 250400 genes are essential for Escherichia coli and Bacillus subtilis, which is less than 10% of their genes.[101][102][103] Half of these genes are orthologs in both organisms and are largely involved in protein synthesis.[103] In the budding yeast Saccharomyces cerevisiae the number of essential genes is slightly higher, at 1000 genes (~20% of their genes).[104] Although the number is more difficult to measure in higher eukaryotes, mice and humans are estimated to have around 2000 essential genes (~10% of their genes).[105] The synthetic organism, Syn 3, has a minimal genome of 473 essential genes and quasi-essential genes (necessary for fast growth), although 149 have unknown function.[99]

Essential genes include Housekeeping genes (critical for basic cell functions)[106] as well as genes that are expressed at different times in the organisms development or life cycle.[107] Housekeeping genes are used as experimental controls when analysing gene expression, since they are constitutively expressed at a relatively constant level.

Gene nomenclature has been established by the HUGO Gene Nomenclature Committee (HGNC) for each known human gene in the form of an approved gene name and symbol (short-form abbreviation), which can be accessed through a database maintained by HGNC. Symbols are chosen to be unique, and each gene has only one symbol (although approved symbols sometimes change). Symbols are preferably kept consistent with other members of a gene family and with homologs in other species, particularly the mouse due to its role as a common model organism.[108]

Genetic engineering is the modification of an organism's genome through biotechnology. Since the 1970s, a variety of techniques have been developed to specifically add, remove and edit genes in an organism.[109] Recently developed genome engineering techniques use engineered nuclease enzymes to create targeted DNA repair in a chromosome to either disrupt or edit a gene when the break is repaired.[110][111][112][113] The related term synthetic biology is sometimes used to refer to extensive genetic engineering of an organism.[114]

Genetic engineering is now a routine research tool with model organisms. For example, genes are easily added to bacteria[115] and lineages of knockout mice with a specific gene's function disrupted are used to investigate that gene's function.[116][117] Many organisms have been genetically modified for applications in agriculture, industrial biotechnology, and medicine.

For multicellular organisms, typically the embryo is engineered which grows into the adult genetically modified organism.[118] However, the genomes of cells in an adult organism can be edited using gene therapy techniques to treat genetic diseases.

Alberts B, Johnson A, Lewis J, Raff M, Roberts K, Walter P (2002). Molecular Biology of the Cell (Fourth ed.). New York: Garland Science. ISBN978-0-8153-3218-3. A molecular biology textbook available free online through NCBI Bookshelf.

Go here to read the rest:
Gene - Wikipedia

Alnylam Receives Fast Track Designation for Vutrisiran for the Treatment of the Polyneuropathy of hATTR Amyloidosis – BioSpace

CAMBRIDGE, Mass.--(BUSINESS WIRE)-- Alnylam Pharmaceuticals, Inc. (Nasdaq: ALNY), the leading RNAi therapeutics company, announced today that the U.S. Food and Drug Administration (FDA) has granted Fast Track designation to vutrisiran, an investigational therapeutic for the treatment of the polyneuropathy of hereditary transthyretin-mediated (hATTR) amyloidosis in adults. According to the FDA, Fast Track designation is designed to facilitate the development and expedite the review of drugs that treat serious conditions and fill an unmet medical need. With this designation, Alnylam will be eligible to submit a rolling New Drug Application for vutrisiran.

Vutrisiran has demonstrated an encouraging safety profile in the Phase 1 study, with infrequent quarterly dosing with low-volume, subcutaneous administration which potentially reduces the burden of care for this progressive, life-threatening and multisystem disease. We are therefore pleased that the FDA has granted vutrisiran Fast Track designation, said Rena Denoncourt, Vutrisiran Program Leader at Alnylam. After completing enrollment earlier this year, we look forward to sharing topline results of the HELIOS-A Phase 3 study of vutrisiran in early 2021. More broadly, we remain committed to developing additional therapeutic options for the treatment of ATTR amyloidosis to augment the market-leading position of ONPATTRO (patisiran), approved for the treatment of the polyneuropathy of hATTR amyloidosis in adults.

In addition to Fast Track designation, vutrisiran has been granted Orphan Drug designation in the United States and the European Union for the treatment of ATTR amyloidosis. The safety and efficacy of vutrisiran are being evaluated in the ongoing HELIOS-A and HELIOS-B Phase 3 clinical trials. Together, these studies comprise a comprehensive clinical development program intended to demonstrate the broad impact of vutrisiran across the multisystem manifestations of disease and the full spectrum of patients with ATTR amyloidosis.

About Vutrisiran Vutrisiran is an investigational, subcutaneously-administered RNAi therapeutic in development for the treatment of ATTR amyloidosis, which encompasses both hereditary (hATTR) and wild-type (wtATTR) amyloidosis. It is designed to target and silence specific messenger RNA, blocking the production of wild-type and mutant transthyretin (TTR) protein before it is made. Quarterly administration of vutrisiran may help to reduce deposition and facilitate the clearance of TTR amyloid deposits in tissues and potentially restore function to these tissues. Vutrisiran utilizes Alnylams next-generation delivery platform known as the Enhanced Stabilization Chemistry (ESC)-GalNAc-conjugate delivery platform. The safety and efficacy of vutrisiran have not been evaluated by the U.S. Food and Drug Administration, European Medicines Agency or any other health authority.

About HELIOS-A Phase 3 Study HELIOS-A is a Phase 3 global, randomized, open-label study to evaluate the efficacy and safety of vutrisiran in patients with hATTR amyloidosis with polyneuropathy. The trial randomized patients 3:1 to receive either 25mg of vutrisiran subcutaneously once every 12 weeks or 0.3 mg/kg of patisiran intravenously once every three weeks. For most endpoints, results from the vutrisiran arm will be compared to results from the placebo arm of the landmark APOLLO Phase 3 study, which evaluated the efficacy and safety of patisiran in people with hATTR amyloidosis with polyneuropathy. The co-primary endpoints of HELIOS-A are the change from baseline in the modified Neurologic Impairment Score +7 (mNIS+7) and in the Norfolk Quality of Life-Diabetic Neuropathy (Norfolk QoL-DN) score, at 9 months. Secondary endpoints include the change from baseline in key clinical evaluations including the timed 10-meter walk test (10-MWT), modified body mass index (mBMI), and Rasch-built Overall Disability Scale (R-ODS). The percent reduction in serum transthyretin (TTR) levels in the vutrisiran arm will be compared to the within-study patisiran arm. Additional exploratory endpoints will be assessed to determine the effect of vutrisiran on other aspects of the multisystem nature of this disease, including manifestations of cardiac amyloid involvement.

About HELIOS-B Phase 3 Study HELIOS-B will evaluate the efficacy of vutrisiran versus placebo toward the composite outcome of all-cause mortality and recurrent cardiovascular hospitalizations at 30 months, the primary study endpoint. The study protocol includes an optional interim analysis to be conducted at the Companys discretion. HELIOS-B complements the ongoing HELIOS-A Phase 3 study in patients with hereditary ATTR amyloidosis with polyneuropathy, creating a comprehensive clinical development program to evaluate the safety and efficacy of vutrisiran across the entire disease spectrum of ATTR amyloidosis.

ONPATTRO Important Safety Information Infusion-Related Reactions Infusion-related reactions (IRRs) have been observed in patients treated with ONPATTRO (patisiran). In a controlled clinical study, 19% of ONPATTRO-treated patients experienced IRRs, compared to 9% of placebo-treated patients. The most common symptoms of IRRs with ONPATTRO were flushing, back pain, nausea, abdominal pain, dyspnea, and headache.

To reduce the risk of IRRs, patients should receive premedication with a corticosteroid, acetaminophen, and antihistamines (H1 and H2 blockers) at least 60 minutes prior to ONPATTRO infusion. Monitor patients during the infusion for signs and symptoms of IRRs. If an IRR occurs, consider slowing or interrupting the infusion and instituting medical management as clinically indicated. If the infusion is interrupted, consider resuming at a slower infusion rate only if symptoms have resolved. In the case of a serious or life-threatening IRR, the infusion should be discontinued and not resumed.

Reduced Serum Vitamin A Levels and Recommended Supplementation ONPATTRO treatment leads to a decrease in serum vitamin A levels. Supplementation at the recommended daily allowance (RDA) of vitamin A is advised for patients taking ONPATTRO. Higher doses than the RDA should not be given to try to achieve normal serum vitamin A levels during treatment with ONPATTRO, as serum levels do not reflect the total vitamin A in the body.

Patients should be referred to an ophthalmologist if they develop ocular symptoms suggestive of vitamin A deficiency (e.g. night blindness).

Adverse Reactions The most common adverse reactions that occurred in patients treated with ONPATTRO were upper respiratory tract infections (29%) and infusion-related reactions (19%).

Indication ONPATTRO is indicated for the treatment of the polyneuropathy of hereditary transthyretin-mediated amyloidosis in adults.

For additional information about ONPATTRO, please see the full Prescribing Information.

About Transthyretin (ATTR) Amyloidosis Transthyretin (ATTR) amyloidosis is a rare, progressively debilitating, and fatal disease caused by misfolded TTR proteins that accumulate as amyloid deposits in multiple tissues including the nerves, heart and gastrointestinal (GI) tract. There are two types of ATTR amyloidosis: hereditary ATTR (hATTR) amyloidosis and wild-type (wtATTR) amyloidosis. hATTR amyloidosis is an inherited disease resulting in intractable peripheral sensory-motor neuropathy, autonomic neuropathy, and/or cardiomyopathy. It is estimated to affect 50,000 people worldwide. The condition can have a debilitating impact on a patients life and may lead to premature death within 4.7 years of diagnosis. wtATTR amyloidosis is a nonhereditary, progressive type of the disease with undefined etiology. It affects an estimated 200,000-300,000 people worldwide. It primarily manifests as cardiomyopathy, which leads to heart failure and mortality within 2 to 6 years.

About RNAi RNAi (RNA interference) is a natural cellular process of gene silencing that represents one of the most promising and rapidly advancing frontiers in biology and drug development today. Its discovery has been heralded as a major scientific breakthrough that happens once every decade or so, and was recognized with the award of the 2006 Nobel Prize for Physiology or Medicine. By harnessing the natural biological process of RNAi occurring in our cells, a new class of medicines, known as RNAi therapeutics, is now a reality. Small interfering RNA (siRNA), the molecules that mediate RNAi and comprise Alnylam's RNAi therapeutic platform, function upstream of todays medicines by potently silencing messenger RNA (mRNA) the genetic precursors that encode for disease-causing proteins, thus preventing them from being made. This is a revolutionary approach with the potential to transform the care of patients with genetic and other diseases.

About Alnylam Alnylam (Nasdaq: ALNY) is leading the translation of RNA interference (RNAi) into a whole new class of innovative medicines with the potential to transform the lives of people afflicted with rare genetic, cardio-metabolic, hepatic infectious, and central nervous system (CNS)/ocular diseases. Based on Nobel Prize-winning science, RNAi therapeutics represent a powerful, clinically validated approach for the treatment of a wide range of severe and debilitating diseases. Founded in 2002, Alnylam is delivering on a bold vision to turn scientific possibility into reality, with a robust RNAi therapeutics platform. Alnylams commercial RNAi therapeutic products are ONPATTRO (patisiran), approved in the U.S., EU, Canada, Japan, Switzerland and Brazil, and GIVLAARI (givosiran), approved in the U.S. and EU. Alnylam has a deep pipeline of investigational medicines, including five product candidates that are in late-stage development. Alnylam is executing on its "Alnylam 2020" strategy of building a multi-product, commercial-stage biopharmaceutical company with a sustainable pipeline of RNAi-based medicines to address the needs of patients who have limited or inadequate treatment options. Alnylam is headquartered in Cambridge, MA. For more information about our people, science and pipeline, please visit http://www.alnylam.com and engage with us on Twitter at @Alnylam or on LinkedIn.

Forward Looking Statements Various statements in this release, including, without limitation, Alnylam's views and plans with respect to the potential for RNAi therapeutics, including vutrisiran, its expectations with respect to the encouraging safety profile of vutrisiran in the Phase 1 study, timing for reporting topline results from its HELIOS-A Phase 3 study, its commitment to developing multiple therapeutic options for the treatment of ATTR amyloidosis, the intended goals of the HELIOS-A and -B studies to demonstrate the broad impact of vutrisiran across the multisystem manifestations of disease and the full spectrum of patients with ATTR amyloidosis, and expectations regarding the continued execution on its Alnylam 2020 guidance for the advancement and commercialization of RNAi therapeutics, constitute forward-looking statements for the purposes of the safe harbor provisions under The Private Securities Litigation Reform Act of 1995. Actual results and future plans may differ materially from those indicated by these forward-looking statements as a result of various important risks, uncertainties and other factors, including, without limitation: potential risks to Alnylams business, activities and prospects as a result of the COVID-19 pandemic, or delays or interruptions resulting therefrom; Alnylam's ability to discover and develop novel drug candidates and delivery approaches and successfully demonstrate the efficacy and safety of its product candidates, including vutrisiran; the pre-clinical and clinical results for its product candidates, which may not be replicated or continue to occur in other subjects or in additional studies or otherwise support further development of product candidates for a specified indication or at all; actions or advice of regulatory agencies, which may affect the design, initiation, timing, continuation and/or progress of clinical trials or result in the need for additional pre-clinical and/or clinical testing; delays, interruptions or failures in the manufacture and supply of its product candidates or its marketed products; obtaining, maintaining and protecting intellectual property; intellectual property matters including potential patent litigation relating to its platform, products or product candidates; obtaining regulatory approval for its product candidates, including inclisiran and lumasiran, and maintaining regulatory approval and obtaining pricing and reimbursement for its products, including ONPATTRO and GIVLAARI; progress in continuing to establish a commercial and ex-United States infrastructure; successfully launching, marketing and selling its approved products globally, including ONPATTRO and GIVLAARI, and achieve net product revenues for ONPATTRO within our expected range during 2020; Alnylams ability to successfully expand the indication for ONPATTRO in the future; competition from others using technology similar to Alnylam's and others developing products for similar uses; Alnylam's ability to manage its growth and operating expenses within the ranges of our expected guidance and achieve a self-sustainable financial profile in the future without the need for future equity financing; Alnylams ability to establish and maintain strategic business alliances and new business initiatives; Alnylam's dependence on third parties, including Regeneron, for development, manufacture and distribution of certain products, including eye and CNS products, and Ironwood, for assistance with the education about and promotion of GIVLAARI; the outcome of litigation; the risk of government investigations; and unexpected expenditures, as well as those risks more fully discussed in the "Risk Factors" filed with Alnylam's most recent Annual Report on Form 10-K filed with the Securities and Exchange Commission (SEC) and in other filings that Alnylam makes with the SEC. In addition, any forward-looking statements represent Alnylam's views only as of today and should not be relied upon as representing its views as of any subsequent date. Alnylam explicitly disclaims any obligation, except to the extent required by law, to update any forward-looking statements.

View source version on businesswire.com: https://www.businesswire.com/news/home/20200414005125/en/

Go here to read the rest:
Alnylam Receives Fast Track Designation for Vutrisiran for the Treatment of the Polyneuropathy of hATTR Amyloidosis - BioSpace

Gene H. Kim, MD – The University of Chicago Medicine

Gene Kim, MD, provides skilled medical care to adults with cardiovascular disease. He focuses on heart transplantation and patients with advanced heart failure.

Dr. Kim is investigating microRNA regulation of cardiovascular development and function. He is also currently researching the use of high-frequency ultrasonic imaging in laboratory models to detect a wide range of cardiac disorders, including aortic and vascular disorders, hypertension, hypertrophy, cardiomyopathy, and right ventricular dysfunction.

The University of Chicago Medicine 5841 S. Maryland Avenue Chicago, IL 60637

2008

Internal Medicine Cardiovascular Diseases

The University of Chicago Pritzker School of Medicine

The University of Chicago Medicine

American Heart Association

English

gkim1@medicine.bsd.uchicago.edu

(773) 702-3936

(773) 834-1764

Gene H. Kim, MD The University of Chicago Medicine 5841 S. Maryland Avenue, MC 6080 Chicago, IL 60637

Request an appointment online or call UCM Connect at 1-888-824-0200

Physicians, contact the Referring Physician Access Line at 1-877-DOM-2730

Additionally, Dr. Kim provides cardiology care through the Urban Health Initiative, a partnership designed to improve access to health care for residents on the South Side of Chicago.

View a partial list of Dr. Kim's publications through the National Library of Medicine's PubMed online database.

Read more:
Gene H. Kim, MD - The University of Chicago Medicine

New ammunition uncovered by U of T researchers to develop colorectal cancer treatment – Varsity

One in 14 Ontarians can expect to be diagnosed with colorectal cancer in their lifetime. COURTESY OF ED UTHMAN/FLICKR

University of Toronto scientists have identified a key protein as a common factor in the growth of many different types of colorectal cancer tumours, according to research published in the Journal of Cell Biology. Colorectal cancer develops in the colon or rectum. In Ontario, it is also the second most fatal cancer, and one in 14 Ontarians can expect to be diagnosed with this form of cancer in their lifetime.

In past research, scientists have linked the excessive accumulation of beta-catenin, a protein with crucial functions in cell development, to the expression of genes that drive tumour proliferation. Research has associated 80 per cent of colorectal cancers with gene mutations that greatly increase the production of beta-catenin.

The co-authors of the study have identified another protein, Importin-11, as the compound that enables beta-catenin transportation to the nucleus of the human cell. Cancer therapies that inhibit this transport could be a promising way to treat colorectal cancer.

Fundamental research provides new knowledge for cancer therapies

The Varsity spoke to Dr. Stephane Angers, a co-author of the study and an associate professor at U of Ts Department of Biochemistry. Angers lab has spent a considerable amount of time studying biological pathways the series of chemical changes during cellular development that give cells their final functions.

Angers noted that Monika Mis, the lead author of the study and a PhD student, uncovered the role of Importin-11 in colorectal cancer in Angers lab. Mis used the gene-editing CRISPR-Cas9 technology to screen genes in colorectal cancer calls to identify a novel gene, IPO11, which encodes for the protein Importin-11.

Current treatment options for colorectal cancer include surgery, chemotherapy, and other radiation therapy. Although this discovery is still in its fundamental stages, blocking the transport of beta-catenin holds great promise for developing new therapies.

As Angers put it, It provides new ammunition, new possibilities, and new knowledge that could lead in the future to new therapies, but it is very much at the discovery level at this point.

More research required to develop therapies

Further research could involve drug discovery and widen the scope of Importin-11 function in various cells. Researchers may also find it valuable to analyze existing data about colorectal cancer. The goal is to understand how the mutations affect tumour formation and develop therapies that harness this knowledge.

Angers lab is also investigating other potential applications of the Wnt pathway, a specific biological pathway associated with beta-catenin. A particularly interesting aspect is its role in regenerative medicine, which is the study of restoring human cells, tissues, and organs.

We think that with new molecules that we have developed we can now activate the pathway in order to promote the regenerative abilities of tissues, noted Angers.

Tags: biology, cancer, medicine, oncology

Excerpt from:
New ammunition uncovered by U of T researchers to develop colorectal cancer treatment - Varsity

Akouos Announces New Data at the Association for Research in Otolaryngology Midwinter Meeting – Financialbuzz.com

Akouos, a precision genetic medicine company developing gene therapies to potentially improve and preserve hearing, announced today that data from its inner ear gene therapy platform will be presented during the 43rd Annual Midwinter Meeting of the Association for Research in Otolaryngology (ARO), being held January 25 to January 29, 2020 in San Jose, CA.

Akouos continues to advance our platform for inner ear disorders, and we are excited to share our progress with the scientific community, said Greg Robinson, Ph.D., chief scientific officer of Akouos. The data presented at ARO further substantiates Akouoss use of AAVAnc80 vector technology and its potential to address many forms of hearing loss.

SYMPOSIUM

Title: The Adeno-associated Viral Anc80 (AAVAnc80) Vector Precision Genetic Medicines to Address Hearing Loss

Presenter: Michelle Valero, Ph.D., Director, Anatomy & Physiology, Akouos

Session: Symposium 11

Date and Time: Saturday, January 25, 3 p.m. (PST)

POSTER PRESENTATIONS

Title: The Adeno-associated Viral Anc80 Vector Efficiently Transduces Inner Ear Cells in Olive Baboons (Papio anubis)

Day and Time: Monday, January 27, 1 p.m. (PST)

Title: The Adeno-associated Viral Anc80 Vector Efficiently Transduces Inner Ear Cells in Cynomolgus Macaques (Macaca fascicularis)

Day and Time: Monday, January 27, 1 p.m. (PST)

Title: Dual Adeno-associated Viral Anc80 Vector Efficiently Transduces Inner Ear Cells in Non-human Primates

Day and Time: Monday, January 27, 1 p.m. (PST)

About Akouos

Akouos is a precision genetic medicine company dedicated to developing gene therapies with the potential to improve and preserve hearing. Leveraging its adeno-associated viral (AAV) vector-based gene therapy platform, Akouos is focused on developing precision therapies for forms of sensorineural hearing loss. Headquartered in Boston, the Company was founded in 2016 by world leaders in the fields of neurotology, genetics, inner ear drug delivery, and AAV gene therapy. Akouos has strategic partnerships with Massachusetts Eye and Ear and Lonza, Inc. For more information, please visit http://www.akouos.com.

About AAVAnc Technology

Ancestral AAV (AAVAnc) technology was developed in the laboratory of Luk Vandenberghe, Ph.D., Director of the Grousbeck Gene Therapy Center at Harvard Medical School. AAVAnc technology uses computational and evolutionary methods to predict novel conformations of the adeno-associated viral particle. AAVAnc80, one of 40,000 AAVAnc vectors, has demonstrated preliminary safety and effective gene delivery in both mice and non-human primates in numerous preclinical studies.

View source version on businesswire.com: https://www.businesswire.com/news/home/20200124005380/en/

See original here:
Akouos Announces New Data at the Association for Research in Otolaryngology Midwinter Meeting - Financialbuzz.com

The Roivant-Sumitomo Deal: $3 Billion, 5 Companies, $550 Million in Loans and an Option on 6 More Companies – BioSpace

In September, Roivant Sciences, Vivek Ramaswamys umbrella biotech company, agreed to sell ownership of five of its Vant companies to Japans Sumitomo Dainippon Pharma for $3 billion. Sumitomo Dainippon also was buying an equity stake of more than 10% of Roivant shares.

The companies have released more details of the deal, which includes an option on six more Vant companies until 2024. The agreement was formally signed yesterday. Sumitomo will create an as-yet unnamed company to shift the five companies into. They will be run by Myrtle Potter, a former Genentech executive who has been the operating chair of Roivant since July 2018.

There is a $3 billion upfront fee. In addition, Sumitomo will offer a $350 million loan facility to Myovant to fund the launch of relugolix, its Phase III candidate for uterine fibroids if it is approved. The Japanese firm will also loan Urovant $200 million for its vibegron for overactive bladder.

If Sumitomo options the other six companies, will have acquired a pipeline of more than 25 clinical programs with multiple possible launches from 2020 to 2022. In addition to buying the five initial companies, the deal gives Sumitomo access to Roivants proprietary technology platforms, DrugOme and Digital Innovation. It will also enter separate strategic client relationships with Datavant and Alyvant.

The five Vant companies are Myovant Sciences, Urovant Sciences, Enzyvant Therapeutics, Altavant Sciences, and Spirovant Sciences. Spirovant is a new Vant that focuses on developing gene therapies for cystic fibrosis.

In addition to Potter, other Roivant team members will move to the new entity. They include Adele Gulfo, Roivants chief of Commercial Development, Sam Azoulay, Roivants chief medical officer, and Dan Rothman, Roivants chief information officer.

I am happy to announce that we have reached an agreement on the Strategic Alliance with Roivant, one of the strategic investments that we are making to address our challenges laid out in the Mid-Term Business Plan 2022, said Hiroshi Nomura, representative director, president and chief executive officer of Sumitomo Dainippon Pharma. This Strategic Alliance allows us to not only acquire potential blockbusters and innovative health technology platforms developed by Roivant, but it will also enable us to deepen our relationship with Roivant, a company that possesses an innovative business model and underlying culture. We expect this relationship will contribute significantly to the establishment of a position as a Global Specialized Player which we aspire to be in by 2033.

Spirovant is working to develop two therapies for cystic fibrosis. SPIRO-2101 uses an adeno-associated virus vector (AAV), like most gene therapies, to deliver a functional l CFTR gene to airway epithelial cells. SPIRO-2102 uses a proprietary lentiviral vector to deliver the gene. In animal models, both have showed restoration of CFTR function. The companys aerosolization technology is designed to maximize uptake in the lungs.

The two therapeutics leverage technology out of the University of Iowa Center for Gene Therapy at the Carver College of Medicine. Its collaboration with Childrens Hospital of Philadelphia (CHOP) was involved with the manufacture of the preclinical supply of the AAV products.

We are proud to enter this unique Alliance with Sumitomo Dainippon Pharma, said Ramaswamy. We hope that our contributions to this Alliance will enable Sumitomo Dainippon Pharma to realize its vision to be a global leader in the biopharmaceutical industry. In addition, we believe that this Alliance will increase the long-term value of each Vant in the Alliance through a combination of strong financial backing and other capabilities.

Read more from the original source:
The Roivant-Sumitomo Deal: $3 Billion, 5 Companies, $550 Million in Loans and an Option on 6 More Companies - BioSpace

Scientists track the arms race thats playing out between bacteria in your gut – GeekWire

The mixture of bacteria shown in this photomicrograph contains five different species of the genus Bacteroides. (UW Medicine Photo / Mougous Lab / Kevin Cutler)

The balance of bacteria in your gut can make the difference between sickness and health and now scientists report that different species of bacteria share immunity genes to protect themselves against each others toxins and maintain their balance of power.

In effect, closely related species of bacteria acquire each others defense systems to fend off threats from alien invaders.

The findings appear in a paper published today in the journal Nature. The senior authors are Joseph Mougous, a microbiology professor at the University of Washington School of Medicine; and Elhanan Borenstein, a former UW Medicine geneticist who now works at Tel Aviv University.

The researchers focused on a particular species of gut bacteria known as Bacteroides fragilis, which produces toxic proteins to kill off other types of bacteria that get too close. They identified a cluster of genes that gives B. fragilis immunity to its own toxins but they also found the immunity factors in samples that didnt contain any B. fragilis bacterla.

This finding strongly suggests that these anti-B. fragilis immunity elements were encoded by other bacteria in the gut, the team reported in the Nature paper.

A statistical analysis led the researchers to suspect that the immunity genes were shared by at least four other Bacteroides species: B. ovatus, B. vulgatus, B. helcogenes and B. copracola. Gene sequencing confirmed their suspicion. The acquired shielding effect was observed in lab-dish conditions, as well as in the guts of living lab mice.

As a follow-up experiment, the researchers mixed one type of bacteria that had a specific group of immunity genes with another type that didnt have those genes. They found that the genes were transferred from the haves to the have-nots, giving them immunity as well.

Such transfers took place in other cases as well for example, when the target genes helped one species of Bacteroides fend off attacks by non-Bacteroides bacteria.

Gene-swapping thus appears to be a crucial element of a molecular arms race by which groups of bacteria maintain their status in the gut microbiome, the researchers said. So if scientists hope to treat diseases by tweaking the microbiome, they might have to account for that strategy in their medical playbook.

Two former UW postdoctoral researchers Benjamin Ross, whos now on Dartmouth Colleges faculty; and Adrian Verster, a bioinformatician at Health Canada are the principal authors.of the Nature paper, titled Human Gut Bacteria Contain Acquired Interbacterial Defence Systems. In addition to Mougous and Borenstein, the other study authors include Matthew Radey, Danica Schmidtke, Christopher Pope, Lucas Hoffman, Adeline Hajjar and S. Brook Peterson.

View post:
Scientists track the arms race thats playing out between bacteria in your gut - GeekWire

MUTYH gene – Genetics Home Reference – NIH

Cheadle JP, Sampson JR. Exposing the MYtH about base excision repair and human inherited disease. Hum Mol Genet. 2003 Oct 15;12 Spec No 2:R159-65. Epub 2003 Aug 5. Review.

Croitoru ME, Cleary SP, Di Nicola N, Manno M, Selander T, Aronson M, Redston M, Cotterchio M, Knight J, Gryfe R, Gallinger S. Association between biallelic and monoallelic germline MYH gene mutations and colorectal cancer risk. J Natl Cancer Inst. 2004 Nov 3;96(21):1631-4.

Farrington SM, Tenesa A, Barnetson R, Wiltshire A, Prendergast J, Porteous M, Campbell H, Dunlop MG. Germline susceptibility to colorectal cancer due to base-excision repair gene defects. Am J Hum Genet. 2005 Jul;77(1):112-9. Epub 2005 May 3.

Fleischmann C, Peto J, Cheadle J, Shah B, Sampson J, Houlston RS. Comprehensive analysis of the contribution of germline MYH variation to early-onset colorectal cancer. Int J Cancer. 2004 Apr 20;109(4):554-8.

Jones S, Emmerson P, Maynard J, Best JM, Jordan S, Williams GT, Sampson JR, Cheadle JP. Biallelic germline mutations in MYH predispose to multiple colorectal adenoma and somatic G:C-->T:A mutations. Hum Mol Genet. 2002 Nov 1;11(23):2961-7.

Jones S, Lambert S, Williams GT, Best JM, Sampson JR, Cheadle JP. Increased frequency of the k-ras G12C mutation in MYH polyposis colorectal adenomas. Br J Cancer. 2004 Apr 19;90(8):1591-3.

Kambara T, Whitehall VL, Spring KJ, Barker MA, Arnold S, Wynter CV, Matsubara N, Tanaka N, Young JP, Leggett BA, Jass JR. Role of inherited defects of MYH in the development of sporadic colorectal cancer. Genes Chromosomes Cancer. 2004 May;40(1):1-9.

Lipton L, Halford SE, Johnson V, Novelli MR, Jones A, Cummings C, Barclay E, Sieber O, Sadat A, Bisgaard ML, Hodgson SV, Aaltonen LA, Thomas HJ, Tomlinson IP. Carcinogenesis in MYH-associated polyposis follows a distinct genetic pathway. Cancer Res. 2003 Nov 15;63(22):7595-9.

Lipton L, Tomlinson I. The multiple colorectal adenoma phenotype and MYH, a base excision repair gene. Clin Gastroenterol Hepatol. 2004 Aug;2(8):633-8. Review.

Russell AM, Zhang J, Luz J, Hutter P, Chappuis PO, Berthod CR, Maillet P, Mueller H, Heinimann K. Prevalence of MYH germline mutations in Swiss APC mutation-negative polyposis patients. Int J Cancer. 2006 Apr 15;118(8):1937-40.

Sampson JR, Dolwani S, Jones S, Eccles D, Ellis A, Evans DG, Frayling I, Jordan S, Maher ER, Mak T, Maynard J, Pigatto F, Shaw J, Cheadle JP. Autosomal recessive colorectal adenomatous polyposis due to inherited mutations of MYH. Lancet. 2003 Jul 5;362(9377):39-41.

Sampson JR, Jones S, Dolwani S, Cheadle JP. MutYH (MYH) and colorectal cancer. Biochem Soc Trans. 2005 Aug;33(Pt 4):679-83. Review.

Sieber OM, Lipton L, Crabtree M, Heinimann K, Fidalgo P, Phillips RK, Bisgaard ML, Orntoft TF, Aaltonen LA, Hodgson SV, Thomas HJ, Tomlinson IP. Multiple colorectal adenomas, classic adenomatous polyposis, and germ-line mutations in MYH. N Engl J Med. 2003 Feb 27;348(9):791-9.

Venesio T, Molatore S, Cattaneo F, Arrigoni A, Risio M, Ranzani GN. High frequency of MYH gene mutations in a subset of patients with familial adenomatous polyposis. Gastroenterology. 2004 Jun;126(7):1681-5.

Wang L, Baudhuin LM, Boardman LA, Steenblock KJ, Petersen GM, Halling KC, French AJ, Johnson RA, Burgart LJ, Rabe K, Lindor NM, Thibodeau SN. MYH mutations in patients with attenuated and classic polyposis and with young-onset colorectal cancer without polyps. Gastroenterology. 2004 Jul;127(1):9-16. Erratum in: Gastroenterology. 2004 Nov;127(5):1651.

See more here:
MUTYH gene - Genetics Home Reference - NIH

Here’s Why Editas Medicine Fell as Much as 15.7% Today – Madison.com

What happened

Shares of gene editing pioneer Editas Medicine (NASDAQ: EDIT) dropped nearly 16% today after a new study published in Nature Methods drew attention to unintended effects of using the highly touted genetic engineering tool known as CRISPR. Shares of genome-editing peers CRISPR Therapeutics (NASDAQ: CRSP) and Intellia Therapeutics (NASDAQ: NTLA) were down as much as 6.9% and 14.9%, respectively, on the news.

The study, conducted by a team from Columbia University Medical Center, provided data showing that the technology can "introduce hundreds of unintended mutations into the genome," according to Genetic Engineering & Biotechnology News. That contradicts one of the better-known characteristics of CRISPR: precision.

Simply put, it's not sitting well with investors, who are (in knee-jerk fashion) adjusting the value placed on early-stage platforms, especially Editas Medicine, which will be the first of the group to enter clinical trials. As of 3:31 p.m. EDT, the stock had settled to a 11.3% loss.

Image source: Getty Images.

The study is among the first to quantify the specificity of CRISPR tools, which work by delivering gene editing enzymes to specific parts of the genome through the use of synthetic guide RNAs. Or that's how they're supposed to work. The authors of the study show that although intended edits can be made with respectable efficiency, such as correcting a mutation in a gene that causes blindness in mice, there are also unintended secondary edits made to the genome.

This may seem like a bombshell report, but it's a matter of optics. Researchers have never shied away from the reality that CRISPR gene editing tools can stray off target and make unintended edits to genomes in mammalian cells (i.e., humans). Many labs -- including Editas Medicine, CRISPR Therapeutics, and Intellia Therapeutics -- are working on increasing the efficiency and specificity of the technology. This is how science works. By quantifying these off-target mutations, which the paper attempted to do, researchers can begin to better understand how to improve the technology.

Investors and traders did not take the same cool-headed approach to the news, instead giving into a knee-jerk reaction to adjust the value of each pre-clinical technology platform. While off-target edits could prove troublesome for a CRISPR therapeutic used in humans, it's important to remember that there are currently no clinical trials underway in the United States. Editas Medicine will become the first to initiate a clinical trial later this year.

The sharp contrasts in reactions from researchers and investors is likely driven by how CRISPR is perceived by the media. Unfortunately, there is a generous amount of hyped-up science journalism that sticks to simple narratives -- "CRISPR has arrived and will cure all diseases!" -- instead of more nuanced takes that give equal weight to each current obstacles and future potential facing an emerging technology. Just remember: Biology is never quite so simple.

The results from the study don't really change anything, except for bringing more attention to the already existent clinical risk inherent to the development of early-stage CRISPR therapeutics. There is still plenty of work and new technology left to be developed before gene editing fulfills its promise in treating and curing human diseases. Hopefully, this can be a long-term positive for investors in CRISPR stocks by forcing them to listen to the fundamental hurdles for the technology. Hopefully.

10 stocks we like better than Editas Medicine

When investing geniuses David and Tom Gardner have a stock tip, it can pay to listen. After all, the newsletter they have run for over a decade, Motley Fool Stock Advisor, has tripled the market.*

David and Tom just revealed what they believe are the 10 best stocks for investors to buy right now... and Editas Medicine wasn't one of them! That's right -- they think these 10 stocks are even better buys.

*Stock Advisor returns as of May 1, 2017

Maxx Chatsko has no position in any stocks mentioned. The Motley Fool has no position in any of the stocks mentioned. The Motley Fool has a disclosure policy.

View original post here:
Here's Why Editas Medicine Fell as Much as 15.7% Today - Madison.com