A Novel Form of Gene Therapy Can Treat Diabetes With Genetically Modified Skin Transplants – Big Think (blog)

A Novel Form of Gene Therapy Can Treat Diabetes With Genetically Modified Skin Transplants

What Psychiatric Wards Teach Us About the Nature of Reality

Had It up to Here with Climate Change? Eat Beans Instead of Beef

Researchers from the University of Chicago have successfully completed a proof-of-concept studywhere they managed to treat obesity in mice using a new type of gene therapy that utilized skin transplants. Human skin transplantation is a well-established clinical approach that has been used for the treatment of burns. However, using it as a vehicle to deliver genetic treatments for non-skin diseases could be revolutionary.

There are several reasons why skin stem cell therapy can be applicable to a broad type of diseases. The skin is the largest human organ, providing an easy access to cells needed for genetic treatments. The skin enables easy monitoring for potential off-target mutations resulting from the CRISPR intervention, as well as easy removal of such mutations, should they occur. Most importantly, proteins that are secreted by epidermal cells can reach the blood circulation and achieve desired therapeutic effects for the entire body.

In the study published this month, titledEngineered Epidermal Progenitor Cells Can Correct Diet-Induced Obesity and Diabetes,the scientists genetically engineered skin cells to be able to deliver GLP1 (glucagon-like peptide 1) - a hormone which regulates blood glucose. Then they developed a surgical procedure which allowed them to successfully engraft the new skin onto a mouse host. Finally, the genetically modified cells had a mechanism for releasing GLP1, which was regulated by a small amount of antibiotic that was fed to the mice. The treated group of obese mice significantly reduced their body weight and insulin resistance, compared to the control group.

Xiaoyang Wu, one of the authors of the study, says in an interview for ResearchGate:

We established a novel mouse to mouse skin transplantation system to test skin gene therapy. [] Our proof-of-concept work demonstrated its possible to use engineered skin grafts to treat many non-skin diseases. Clinical translation of our findings will be relatively easy, as skin transplantation in human patients has been well established and clinically used for treatment of burn wounds for many years.

Skin grafts are an exciting new avenue to explore for genetic treatments of diseases. They are relatively inexpensive compared to other types of gene therapy, the procedure is minimally invasive, and it has already been tested and proven safe.

MOST POPULAR

What the World Will Look Like 4C Warmer

Were the Ancient Egyptians Black or White? Scientists Now Know

Here's When Machines Will Take Your Job, as Predicted by AI Gurus

The Mystery of the Bermuda Triangle May Finally Be Solved

Read the original post:

A Novel Form of Gene Therapy Can Treat Diabetes With Genetically Modified Skin Transplants - Big Think (blog)

Silverstein-backed startup will test gene therapy for Parkinson’s – FierceBiotech

Regenxbio has joined forces with investment firm OrbiMed and a new nonprofit foundation to create Prevail Therapeutics, a startup focused on new biologics and gene therapiesfor Parkinson's disease (PD).

Prevail will draw on the expertise of the Silverstein Foundation for Parkinson's with GBA, which concentrates on a particular form of the disease caused by mutations in the glucocerebrosidase gene.

The foundation was set up this year by OrbiMed's co-head of private equity Jonathan Silverstein, who was diagnosed with GBA-linked PD in February and is mobilizing efforts to discover a cure for the disease. Silverstein backed the foundation with $10 million of his own money, and is intent on accelerating research into PD with GBA as well as other forms of the disease.

Prevail says it will focus initially on research coming out of the lab of its co-founder and CEO Asa Abeliovich, M.D., Ph.D., who is on the faculty of Columbia University as well as being a scientific adviser to the Silverstein Foundation and co-founder of neurodegenerative disease biotech Alector.

By joining forces with Regenxbio, Prevail launches with an exclusive license to the gene therapy specialist's adeno-associated virus (AAV) based vector technology NAV AAV9 for PD and other neurodegenerative disorders.

Silverstein said that the NAV platform and Dr. Abeliovich's "deep expertise in the molecular mechanisms of neurodegeneration provides us with a promising opportunity to develop potential life-changing therapies for patients suffering from Parkinson's disease and other neurodegenerative diseases."

He told CNBC today that Prevail's board will also have some big names, including Leonard Bell, co-founder and former CEO of Alexion, OrbiMed venture partner and Alexion co-founder Steve Squinto and serial entrepreneur Peter Thompson of Silverback Therapeutics and Corvus Pharmaceuticals.

The new company will initially focus on GBA1, the most common of the PD mutations, which is estimated to be present in up to 10% of U.S. PD patients and perhaps 100,000 people worldwide. The disease mechanism linked to the mutationan accumulation of alpha-synuclein in the brainmay have implications for the broader PD population and other neurodegenerative diseases.

"Many of the drugs we are trying for Parkinson's with GBA may work in the broader Parkinson's population," said Silverstein. The aim will be to get drugs approved for use in GBA patients first, and then expand their use into other patient groups.

The work of the foundation is attracting investment from companies who are not even active in PD, with cancer specialist Celgene today pledging a grant of $5 million.

Here is the original post:

Silverstein-backed startup will test gene therapy for Parkinson's - FierceBiotech

Gene Therapy Is Now Available, but Who Will Pay for It? – Scientific American

By Ben Hirschler

LONDON (Reuters) - The science of gene therapy is finally delivering on its potential, and drugmakers are now hoping to produce commercially viable medicines after tiny sales for the first two such treatments in Europe.

Thanks to advances in delivering genes to targeted cells, more treatments based on fixing faulty DNA in patients are coming soon, including the first ones in the United States.

Yet the lack of sales for the two drugs already launched to treat ultra-rare diseases in Europe highlights the hurdles ahead for drugmakers in marketing new, extremely expensive products for genetic diseases.

After decades of frustrations, firms believe there are now major opportunities for gene therapy in treating inherited conditions such as haemophilia. They argue that therapies offering one-off cures for intractable diseases will save health providers large sums in the long term over conventional treatments which each patient may need for years.

In the past five years, European regulators have approved two gene therapies - the first of their kind in the world, outside China - but only three patients have so far been treated commercially.

UniQure's Glybera, for a very rare blood disorder, is now being taken off the market given lack of demand.

The future of GlaxoSmithKline's Strimvelis for ADA-SCID - or "bubble boy" disease, where sufferers are highly vulnerable to infections - is uncertain after the company decided to review and possibly sell its rare diseases unit.

Glybera, costing around $1 million per patient, has been used just once since approval in 2012. Strimvelis, at about $700,000, has seen two sales since its approval in May 2016, with two more patients due to be treated later this year.

"It's disappointing that so few patients have received gene therapy in Europe," said KPMG chief medical adviser Hilary Thomas. "It shows the business challenges and the problems faced by publicly-funded healthcare systems in dealing with a very expensive one-off treatment."

These first two therapies are for exceptionally rare conditions - GSK estimates there are only 15 new cases of ADA-SCID in Europe each year - but both drugs are expected to pave the way for bigger products.

The idea of using engineered viruses to deliver healthy genes has fuelled experiments since the 1990s. Progress was derailed by a patient death and cancer cases, but now scientists have learnt how to make viral delivery safer and more efficient.

Spark Therapeutics hopes to win U.S. approval in January 2018 for a gene therapy to cure a rare inherited form of blindness, while Novartis could get a U.S. go-ahead as early as next month for its gene-modified cell therapy against leukaemia - a variation on standard gene therapy.

At the same time, academic research is advancing by leaps and bounds, with last week's successful use of CRISPR-Cas9 gene editing to correct a defect in a human embryo pointing to more innovative therapies down the line.

Spark Chief Executive Jeffrey Marrazzo thinks there are specific reasons why Europe's first gene therapies have sold poorly, reflecting complex reimbursement systems, Glybera's patchy clinical trials record and the fact Strimvelis is given at only one clinic in Italy.

He expects Spark will do better. It plans to have treatment centers in each country to address a type of blindness affecting about 6,000 people around the world.

Marrazzo admits, however, there are many questions about how his firm should be rewarded for the $400 million it has spent developing the drug, given that healthcare systems are geared to paying for drugs monthly rather than facing a huge upfront bill.

A one-time cure, even at $1 million, could still save money over the long term by reducing the need for expensive care, in much the same way that a kidney transplant can save hundreds of thousands of dollars in dialysis costs.

But gene therapy companies - which also include Bluebird Bio, BioMarin, Sangamo and GenSight - may need new business models.

One option would be a pay-for-performance system, where governments or insurers would make payments to companies that could be halted if the drug stopped working.

"In an area like haemophilia I think that approach is going to make a ton of sense, since the budget impact there starts to get more significant," Marrazzo said.

Haemophilia, a hereditary condition affecting more than 100,000 people in markets where specialty drugmakers typically operate, promises to be the first really big commercial opportunity. It offers to free patients from regular infusions of blood-clotting factors that can cost up to $400,000 a year.

Significantly, despite its move away from ultra-rare diseases, GSK is still looking to use its gene therapy platform to develop treatments for more common diseases, including cancer and beta-thalassaemia, another inherited blood disorder.

Rivals such as Pfizer and Sanofi are also investing, and overall financing for gene and gene-modified cell therapies reached $1 billion in the first quarter of 2017, according to the Alliance of Regenerative Medicine.

Shire CEO Flemming Ornskov - who has a large conventional haemophilia business and is also chasing Biomarin and Spark in hunting a cure for the bleeding disorder - sees both the opportunities and the difficulties of gene therapy.

"Is it something that I think will take market share mid- to long-term if the data continues to be encouraging? Yes. But I think everybody will have to figure out a business model."

Continue reading here:

Gene Therapy Is Now Available, but Who Will Pay for It? - Scientific American

Timeline: Gene therapy’s long road to market – Reuters

LONDON (Reuters) - Gene therapy, which aims to patch faulty genes with working DNA, has been a long time in development. The following are major milestones:

1972 - Researchers first suggest gene therapy as a treatment for genetic diseases but oppose its use in humans "for the foreseeable future", pending greater understanding of the technology.

1990 - A four-year-old girl with severe immunodeficiency became the first patient to undergo gene therapy in the United States.

1999 - American patient Jesse Gelsinger dies following a gene therapy experiment, setting the field back several years as U.S. regulators put some experiments on hold.

2002-03 - Cases of leukaemia are diagnosed in French children undergoing gene therapy in a further blow to the field.

2003 - The world's first gene therapy is approved in China for the treatment of head and neck cancer.

2007 - Doctors carry out the world's first operation using gene therapy to treat a serious sight disorder caused by a genetic defect.

2012 - Europe approves Glybera, the first gene therapy in a Western market, for an ultra-rare blood disorder.

2016 - Europe approves Strimvelis for a very rare type of immunodeficiency.

2017 or 2018 - The first gene therapy could be approved in United States.

Reporting by Ben Hirschler; editing by David Stamp

Original post:

Timeline: Gene therapy's long road to market - Reuters

Pfizer to invest $100M in Sanford gene therapy operation, add jobs … – WRAL Tech Wire

Updated Aug. 8, 2017 at 7:02 a.m.

Published: 2017-08-07 16:07:00 Updated: 2017-08-08 07:02:05

Sanford, N.C. Pharmaceutical giant Pfizer Inc. plans to invest $100 million in its Sanford operations as part of a push into gene therapy, officials said Monday.

The effort builds on a technology developed at the University of North Carolina at Chapel Hill and will create 40 jobs in Sanford.

"Pfizer is proud to further expand our presence in North Carolina, particularly as we build our leadership in gene therapy," Lynn Bottone, site leader at Pfizer Sanford, said in a statement. "We look forward to the next phase of this expansion as we build a clinical and commercial manufacturing facility."

Preliminary work on the expansion and initial hiring have already begun. The 230-acre campus employs about 450 people, reports the N.C. Biotechnology Center.

Gene therapy is a potentially transformational technology for patients that involves highly specialized, one-time treatments to address the root cause of diseases caused by genetic mutation. The technology involves introducing genetic material into the body to deliver a correct copy of a gene to a patients cells to compensate for a defective or missing gene.

Last year, Pfizer acquired Bamboo Therapeutics Inc., a privately held biotechnology company in Chapel Hill focused on developing gene therapies for the potential treatment of patients with certain rare diseases related to neuromuscular conditions and those affecting the central nervous system. Pfizer also committed $4 million to support postdoctoral fellowships in North Carolina universities for training in gene therapy research.

"We are excited that Carolinas research will improve lives and create jobs for North Carolinians," UNC-Chapel Hill Chancellor Carol Folt said in a statement. "This is a perfect example of how placing innovation at the center of our university creates new opportunities. We are proud to be a part of the technologies, expertise and infrastructure that went into Bamboo Therapeutics and helped make this manufacturing expansion in Sanford possible. Gene therapy is a strength at Carolina, and we look forward to continue to help advance this industry."

Pfizer is also expanding a drug-manufacturing facility in Rocky Mount that it acquired from Hospira in 2015. The $190 million project will add 65,000 square feet of sterile injectable facilities but will not create any new jobs. The plant employs about 300 people.

Gov. Roy Cooper visited Pfizers Sanford facility last week to take a tour and meet with the companys senior leaders.

"North Carolina is one of the few places in the country with the biotech resources to take an idea all the way from the lab to the manufacturing line," Cooper said in a statement. "Pfizers investment in Lee County is a prime example of how North Carolinas world-class universities and cutting-edge industries work together to move our state forward."

Pfizer qualified for a performance-based grant of $250,000 from the One North Carolina Fund, which provides state assistance matched by local governments to help attract economic investment and create jobs. Companies receive no money upfront and must meet job and investment targets to obtain payment.

WRAL TechWire any time: Twitter, Facebook

Original post:

Pfizer to invest $100M in Sanford gene therapy operation, add jobs ... - WRAL Tech Wire

BioMarin Pharmaceutical launches gene therapy drug plant in Novato – North Bay Business Journal

NORTH BAY BUSINESS JOURNAL

August 7, 2017, 1:53PM

08/07/2017

BioMarin Pharmaceutical on Monday dedicated its new Novato manufacturing facility which is expected to be key in its continuing clinical trials on a drug the company believes will potentially genetically repair the cause of hemophilia A.

Before a crowd of 300 to 400 people, the company, which manufactures drugs mostly for rare diseases, called its new production location the largest gene-therapy-manufacturing facility in the world. The project was completed 11 months ahead of schedule, employing 300 people in 200,000 construction hours, according to Robert Baffi, the firms executive vice president, Technical Operations.

Jean-Jacques Bienaim, chairman and CEO of BioMarin, said the drug to be produced at the location, BMN 270 gene therapy for hemophilia A, has the potential to change what future doctors learn about hemophilia.

Because of a genetic flaw, the blood of those who have hemophilia does not clot. The mutation takes places in a single gene that provides instructions to make a protein called Factor VIII, which is essential for blood to clot normally.

According to the company, the drug thus far in investigational clinical trials has shown the ability to genetically correct the problem and allow patients to manufacture and maintain a constant level of Factor VIII. Production of the drug to be used in those continuing trials will begin as soon as possible in Novato.

Among those affected by the hemophilia is the son of Christine Orr a speaker at todays event. Genetic roulette resulted in an older son being born without the problem.

But soon after her younger son was born, it became apparent he had little or no clotting factor. Every other day, home infusions of clotting factor have helped curb the problem, but she said her son experienced the stigma of parents being afraid to invite him to birthday parties or play dates over what might happen if he were to be hurt.

She said a one-shot treatment to potentially genetically treat and cure the disease gives her hope that yes, a cure is on my horizon, and he can choose his path in life and not have hemophilia choose it for him.

On Aug. 2, BioMarin Pharmaceutical reported it reaped $317 million in second-quarter revenue, up 6 percent from the same quarter in 2016.

It operated a loss of $37 million for the second quarter, but far less than the $419 million loss in the same quarter last year. The last quarters losses amounted to 21 cents per diluted share.

BioMarin, which has six main drugs on the market, had two huge contributors to second-quarter revenue: Kuvan, with $102 million, and Vimizim, with $103 million.

Kuvan, sapropterin dihydrochloride, treats a genetic disorder called phenylketonuria. BioMarin bought global rights to Kuvan in 2015 from Merck for 340 million euros, about $405 million. PKU is rare, and causes amino acid phenylalanine to build up in the body. The buildup of the amino acid can cause grave health problems.

Vimizim treats patients with mucopolysaccharidosis type IV-A, also called Morquio A syndrome, which is a metabolic disorder that inhibits the bodys ability to process certain mucopolysaccharides. It is usually inherited.

Originally posted here:

BioMarin Pharmaceutical launches gene therapy drug plant in Novato - North Bay Business Journal

Gene therapy via skin could treat diseases such as obesity – UChicago News

A University of Chicago-based research team has overcome challenges that have limited gene therapy and demonstrated how their novel approach with skin transplantation could enable a wide range of gene-based therapies to treat many human diseases.

In a study inthe journal Cell Stem Cell, the researchers provide proof-of-concept. They describe gene-therapy administered through skin transplants to treat two related and extremely common human ailments: Type 2 diabetes and obesity.

We resolved some technical hurdles and designed a mouse-to-mouse skin transplantation model in animals with intact immune systems, said study author Xiaoyang Wu, assistant professor in the Ben May Department for Cancer Research at the University of Chicago. We think this platform has the potential to lead to safe and durable gene therapy in mice and, we hope, in humans, using selected and modified cells from skin.

Beginning in the 1970s, physicians learned how to harvest skin stem cells from a patient with extensive burn wounds, grow them in the laboratory, then apply the lab-grown tissue to close and protect a patients wounds. This approach is now standard. However, the application of skin transplants is better developed in humans than in mice.

The mouse system is less mature, Wu said. It took us a few years to optimize our 3-D skin organoid culture system.

This study is the first to show that an engineered skin graft can survive long term in wild-type mice with intact immune systems. We have a better than 80 percent success rate with skin transplantation, Wu said. This is exciting for us.

The researchers focused on diabetes because it is a common non-skin disease that can be treated by the strategic delivery of specific proteins.

They inserted the gene for glucagon-like peptide 1 (GLP1), a hormone that stimulates the pancreas to secrete insulin. This extra insulin removes excessive glucose from the bloodstream, preventing the complications of diabetes. GLP1 can also delay gastric emptying and reduce appetite.

Using CRISPR, a tool for precise genetic engineering, they modified the GLP1 gene. They inserted one mutation, designed to extend the hormones half-life in the blood stream, and fused the modified gene to an antibody fragment so that it would circulate in the blood stream longer. They also attached an inducible promoter, which enabled them to turn on the gene to make more GLP1, as needed, by exposing it to the antibiotic doxycycline. Then they inserted the gene into skin cells and grew those cells in culture.

When these cultured cells were exposed to an air/liquid interface in the laboratory, they stratified, generating what the authors referred to as a multi-layered, skin-like organoid. Next, they grafted this lab-grown gene-altered skin onto mice with intact immune systems. There was no significant rejection of the transplanted skin grafts.

When the mice ate food containing minute amounts of doxycycline, they released dose-dependent levels of GLP1 into the blood. This promptly increased blood-insulin levels and reduced blood-glucose levels.

When the researchers fed normal or gene-altered mice a high-fat diet, both groups rapidly gained weight. They became obese. When normal and gene-altered mice got the high-fat diet along with varying levels of doxycycline, to induce GLP1 release, the normal mice grew fat and mice expressing GLP1 showed less weight gain.

Expression of GLP1 also lowered glucose levels and reduced insulin resistance.

Together, our data strongly suggest that cutaneous gene therapy with inducible expression of GLP1 can be used for the treatment and prevention of diet-induced obesity and pathologies, the authors wrote.

When they transplanted gene-altered human cells to mice with a limited immune system, they saw the same effect. These results, the authors wrote, suggest that cutaneous gene therapy for GLP1 secretion could be practical and clinically relevant.

This approach, combining precise genome editing in vitro with effective application of engineered cells in vivo, could provide significant benefits for the treatment of many human diseases, the authors note.

We think this can provide a long-term safe option for the treatment of many diseases, Wu said. It could be used to deliver therapeutic proteins, replacing missing proteins for people with a genetic defect, such as hemophilia. Or it could function as a metabolic sink, removing various toxins.

Skin progenitor cells have several unique advantages that are a perfect fit for gene therapy. Human skin is the largest and most accessible organ in the body. It is easy to monitor. Transplanted skin can be quickly removed if necessary. Skins cells rapidly proliferate in culture and can be easily transplanted. The procedure is safe, minimally invasive and inexpensive.

There is also a need. More than 100 million U.S. adults have either diabetes (30.3 million) or prediabetes (84.1 million), according the Centers for Disease Control and Prevention. More than two out of three adults are overweight. More than one out of three are considered obese.

Additional authors of the study were Japing Yue, Queen Gou, and Cynthia Li from the University of Chicago and Barton Wicksteed from the University of Illinois at Chicago. The National Institutes of Health, the American Cancer Society and the V Foundation funded the study.

Article originally appeared on Science Life.

Read more from the original source:

Gene therapy via skin could treat diseases such as obesity - UChicago News

Bluebird Bio sees Europe as first market for its gene therapies – FierceBiotech

Bluebird Bio plans to bring its gene therapies to market in Europe before the U.S., thanks to a favorable regulatory pathway.

Bluebird's head of Europe, Andrew Obenshain, told the Daily Telegraph that the company is already in negotiations with the EMA and the U.K.'s Medicine and Healthcare products Regulatory Agency (MHRA) on possible regulatory filings.

The EMA's adaptive pathways processwhich allows new therapies to be approved in stages based on stepwise collection of datais a key part of that decision, as is the fact that the agency "works very closely with companies coming forward with new methodologies," said Morgan. And with Brexit looming, it makes sense to discuss these plans with the MHRA separately.

Two years ago, Bluebirdwhich targets severe genetic diseases and cancerwas hit hard when the NorthStar trial of lead therapy LentiGlobin failed to hit the mark in sickle cell disease and beta thalassemia, mainly because of variable patient responses to the treatment.

In a recent SEC filing, the company said that combined data from Northstar and other trials, including a follow-up Northstar-2 study, "could support the filing of a marketing authorization application in the EU" for transfusion-dependent thalassemiaprovided they all meet the primary objective of freeing patients from the need for regular blood transfusions.

So far, no approved gene therapies have been in the U.S., while Europe has seen two approvals, namely for UniGene's Glybera (alipogene tiparvovec) for lipoprotein lipase deficiency and GlaxoSmithKline's Strimvelis for the ultrarare "bubble boy syndrome," or ADA-SCID.

Even getting approval is no guarantee of success, however. Glybera was taken off the market in April due to a lack of demand for the 1 million (around $1.2 million)-per-year therapy, with only one patient receiving it commercially since its launch in 2012.

GSK, meanwhile, has priced Strimvelis at a lower rate (around $650,000 a year) to try to encourage takeup, but hasn't given any updates and said last week it may put its rare disease unit up for sale. Rare disease head Carlo Russo moved to Italian biotech Genenta in January.

See more here:

Bluebird Bio sees Europe as first market for its gene therapies - FierceBiotech

Pioneering gene therapy restores vision to people with rare retinal blindness – Genetic Literacy Project

Three years ago, at a fundraiser near Philadelphia for theCuring Retinal Blindness Foundation, I stood, dumbstruck, as young teen Christian Guardino took the stage and belted outDont Stop Believing. Christian had recently undergone gene therapy that was already beginning to illuminate his darkening world but most in the crowd didnt know that.

Next to me was Dr. Jean Bennett, the physician-scientist who pioneered the gene therapy for what was then called Leber congenital amaurosis type 2. She asked me not to tell anyone about Christians treatment.

The world heard Christian last month when he performed in June on forAmericans Got Talent, singing the Jackson 5s Whos Lovin You.Fox Newsmentioned his blindness in what is perhaps the greatest oversimplification of a biotechnology of all time:When Christian Guardino was young, he learned that he would lose his sight. Fortunately, thanks to some gene therapy, he later regained the gift of sight. In the interim, he turned to music and stuck with it.

A Long Time Coming

Christian didnt just order up gene therapy like a side of fries.

The first clinical trial of gene therapy for a single-gene disease in the U.S. was in 1990. The publication of the phase 3 clinical trial data for the gene therapy for RPE65-mediated inherited retinal dystrophy, the disease that Christian has, bestows the name Luxturna, and FDA acceptance ofSpark TherapeuticsBiologics License Application (BLA) with priority review. These are giant steps forward in achieving the companys mission statement to challenge the inevitability of genetic disease.

Corey Haas owes his vision to the gene therapy he received at age 8.

Corey Haas owes his vision to the gene therapy he received at age 8. He is 14 in the accompanying photo, and will turn 17 in September.

The decade-long story of the clinical trial for the inherited blindness frames my history of gene therapy, published in 2012.The Forever Fix: Gene Therapy and the Boy Who Saved It(St. Martins Press, 2012) follows Corey Haas, the same age as Christian, who had his first eye treated in 2008 and sees so well that he goes fishing and turkey hunting. Without gene therapy, his world would be dark. Since the book came out Ive followed families whose children are having gene therapy for a variety of conditions right here atDNA Science.

The phase 3 findings for the blindness, published inThe Lancet, evaluate 21 patients in the treatment group and 10 others randomized to the control group who continued to have their vision evaluated the same way that the treated patients did (because sham surgery isnt ethical). A year later, all of the controls had the procedure too. And all have done great as have others in the earlier clinical trials.

The crux of the research, and part of what took so long, was the invention of a mobility course to evaluate a persons ability to navigate obstacles on a winding pathway under low light conditions. A test of real life, its a crucial complement to standard assessments of visual field, visual acuity, and imaging the layers of the retina.

Ill never forget watching Corey at his two-year check-up at Childrens Hospital of Philadelphia (CHOP) tackle the mobility course after just one eye had been treated. Covering that eye, the boy wobbled and careened, Dr. Jean offering her hand when he teetered too far of course. Yet covering his still-blind eye, he zipped through the maze.

Q&A withDr. Katherine High

The media largely ignored the blindness gene therapy news last week, perhaps because a few days earlier came news of an FDA advisory committees thumbs up for chimeric antigen receptor (CAR) technology to treat a form of leukemia. Two genetics news stories in one week? Nah.

I was frustrated. CAR, although widely described asgene therapybecause it sticks DNA into viruses, isnt really. Instead, it creates a double-whammy drone-like not-seen-in-nature immune response; it doesnt bolster a deficient enzyme. And a cancer is not at all like a single-gene disease.

So I believe that Luxturna to treatRPE65blindness will still be the first actual gene therapy approval.Katherine High, MD, president, chief scientific officer and a founder ofSpark Therapeutics, agrees. I spoke with her earlier this week about the recent progress. An excellent interview about her background ishere.

Ricki Lewis:How will patients be selected to receive Luxturna? Is newborn screening plausible?

Katherine High:Its for symptoms of vision loss in people with two autosomal recessive mutations in the geneRPE65. Initial licensing will be for children 3 years of age through adults. All of our work was done in children aged 3 or older, because treating younger children would require innovations in surgical technique.

RL:Can you describe the protocol?

KH:The first eye is done and within 6 to 18 days, the second eye is done. So it takes about 2 weeks. The protocol provides a chance to make sure the first eye is healing and that the person has the opportunity to get the patch off.

RL:What will Luxturna cost? Will savings on no-longer-needed treatments and aids be considered?

KH:Cost is under discussion. Part of the organization that Spark is building is to make sure that anybody who is a candidate for the therapy will have access to it. Were talking to key stakeholders the patient community, payers, health care providers, and policy makers. Its different in Europe, where all social services and educational services and so forth required for people with impaired vision comes out of the same pot, whereas in the US those kinds of services are separate from the payers. So cost, and cost savings, will involve a more complicated algorithm.

RL:Spark is also developing gene therapy forhemophilia B, with interim findings recently presented at the International Society for Thrombosis and Hemostasis meeting in Berlin. Youve been working on that condition a long time! Update?

KH: Its hard to imagine what its like to take clotting factor once or twice a week for your whole life and then not to have to. Individuals with hemophilia are always planning around when they last had an infusion of factor IX. Do the infusion, the level of clotting factor peaks, and then it slowly comes down until the next infusion. As its coming down, the patient is at risk for bleeds. So people always have to think about it.Id like to go to the gym, and the last infusion was 40 hours ago. Can I? I want to take the kids down to the park. When was my last infusion?

RL:Another hemophilia B clinical trial uses the oldest form of genome editing,zinc finger nucleases. Are the two approaches in competition? (Gene therapy adds a gene; gene editing can swap a functioning gene for a mutant one.)

KH:They both could work. Clinical experience with gene editing is much earlier and it can take time in clinical development to uncover challenges and work through them. With these complex biologics, only a certain amount can be learned from the animal models. Then in clinical investigation you have to look at results and understand what they mean and possibly modify a protocol or institute a course correction. That was true for monoclonal antibodies, for bone marrow transplants, and for straight gene therapy. Will gene editing have a shorter path? Perhaps.

RL:Where will gene therapy be in five years?

KH:Clinical development timelines will become shorter. We started the clinical work for voretigene (aka Luxturna) in 2007, but I began working on it, with Jean Bennett and Al Maguire, in 2005. We initiated the trial in 2007. We paused along the way to discuss with regulators the best way to proceed. We had to do a natural history study, for example, and we had to evaluate the mobility test. We had to build the infrastructure for studying this ultrarare form of inherited retinal dystrophy.

We will continue to see proof-of-concept of gene therapy demonstrated in different target tissues. Were seeing promising results in gene delivery to the liver and the central nervous system. I hope we will see continued accumulation of successful clinical results in a range of target tissues and continued progress in bringing gene therapy products to licensing. One of the challenges in my career has been interesting physicians in learning more about gene therapy. When gene therapy products are licensed, there will be increased interest in the medical community, and that will help to expand opportunities.

RL:Your attempt to catalyze interest in gene transfer goes back pretty far, as does gene therapy itself. And of course the field had to overcome the setbacks of the turn of the century Jesse Gelsingers death and the boys who developed leukemia.

KH:Back in the 1990s when I worked with a gene therapy company in northern California, I kept telling them they should work on this gene,RPE65. They declined because of the small patient population, and I said its a great proof-of-concept idea and its probably going to work. (Its a straightforward enzyme replacement via genetic instructions to an accessible tissue.)

After the high profile adverse events and everyone pulling out of gene therapy, Childrens Hospital of Philadelphia gave us the resources to set up vector production in-house. Dr. Steven Altschuler, then CHOP CEO and now chairman of the board at Spark, said well help, but you cant just commit the resources all to hemophilia. You have to work on a genetic disease that affects children. I said I know exactly what Im going to do, and I went and talked to Jean Bennett.

CHOP gave us the luxury of working without worrying how big the market was. I will always be grateful to Dr. Altschuler for having had the courage to make that decision when things didnt look very good for gene therapy. The other important decision he made was to spin out Spark. We were discussing many possibilities, including partnering with biotech or pharma, but most of them knew very little about gene therapy. And I was concerned about what would happen to the program for gene therapy for this ultrarare disease if put in the hands of a big company and they have a bad year.

(Spark Therapeutics formed in March 2013, the month my book came out in paperback.)

RL:So when will ophthalmologists be able to save young people like Corey Haas and Christian Guardino from living in darkness?

KH:FDA has accepted the Biologics License Application for priority review. That means that they have 6 months to decide. So there will be much back and forth with requests for additional information, and then typically for a novel therapy like this they would arrange an advisory committee meeting.

RL:So 2018 will likely be the year for Luxturna to challenge the inevitability of this one genetic disease. Its a giant first step to achieving Sparks vision:A world where no life is limited by genetic disease.

A version of this article was originally published on the PLOSwebsite as Luxturna: A Giant Step Forward For Blindness Gene Therapy A Conversation with Dr. Kathy High and has been republished here with permission.

Ricki Lewis has a PhD in genetics and is a genetics counselor, science writer and author of Human Genetics: The Basics. Follow her at her website or Twitter @rickilewis.

Read more:

Pioneering gene therapy restores vision to people with rare retinal blindness - Genetic Literacy Project

Agilis Biotherapeutics, Gene Therapy Research Institution Enter Strategic Partnership – Drug Discovery & Development

Agilis Biotherapeutics, Inc. (Agilis), a biotechnology company advancing innovative DNA therapeutics for rare genetic diseases that affect the central nervous system (CNS), and Gene Therapy Research Institution Co, Ltd. (GTRI), a corporation with the mission of developing and delivering of the safest and most efficient gene therapies, announced that the companies have completed a manufacturing and collaboration partnership joint venture (JV) to advance adeno-associated virus (AAV) gene therapies. The JV was initiated earlier this year in connection with a grant from the Japanese Ministry of Trade, Economics and Industry (METI) and Japan External Trade Organization (JETRO) for the development of a state-of-the-art AAV manufacturing facility in Japan. GTRI was co-founded by Professor Shin-ichi Muramatsu, M.D., a leading pioneer in gene therapy who has performed basic science and clinical research in the field for over two decades.

The JV, headquartered in Japan, will initially focus on developing and manufacturing AAV gene therapy vectors using Sf9 baculovirus and HEK293 mammalian cell systems and operate a process development and production facility located in the Tokyo area designed to meet international manufacturing standards, including cGMP, GCTP and PIC/S GMP requirements. Agilis and GTRI will also collaborate to expedite the development, approval and commercialization of select gene therapies in specific CNS diseases. Terms of the joint venture were not disclosed.

We are pleased to collaborate with Agilis to leverage each organizations capabilities and know-how, advance the manufacturing state-of-the art for gene therapy, and develop novel gene therapies, commented Katsuhito Asai, Chief Executive Officer of GTRI and a Director of the joint venture. Our partnership will seek to capitalize on the strong recent progress in the field of gene therapy and expedite the development of innovative gene therapies for patients in need, with a major emphasis on the quality production of safe, effective therapeutics.

We are thrilled to partner with GTRI, said Mark Pykett, Agilis CEO and a Director of the joint venture. We believe that our partnership will enhance the efforts of both organizations, build important shared production capabilities, and accelerate development and commercialization of important gene therapies. We look forward to working with GTRI on a range of initiatives.

See the article here:

Agilis Biotherapeutics, Gene Therapy Research Institution Enter Strategic Partnership - Drug Discovery & Development

Agilis forms joint venture to advance gene therapy vectors … – FierceBiotech

Agilis Biotherapeutics has formed a joint venture with Japans Gene Therapy Research Institution (GTRI). The alliance gives Agilis a base in Japan and a partnership with a fellow CNS specialist to support its development of adeno-associated virus (AAV) vectors and gene therapies.

Cambridge, Massachusetts-based Agilis set up the joint venture using a grant from the Japanese government. The agreement will establish an AAV manufacturing facility in Japan, from where Agilis and GTRI will work on vectors using Sf9 baculovirus and HEK293 mammalian cell systems. Agilis and GTRI plan to develop and manufacture AAV gene therapy vectors through the joint venture.

Agilis and GTRI also plan is to collaborate on the development and commercialization of certain CNS gene therapies.

GTRIs background suggests it is well-equipped to contribute to the project. The Japanese company grew out of the work of Shin-ichi Muramatsu, M.D., a scientist who sequenced AAV3 in the 1990s before going on to create AAVs designed to cross the blood-brain barrier. GTRI is working on gene therapies against diseases including Alzheimers, amyotrophic lateral sclerosis and Parkinsons that build on this research into AAVs.

Both biotechs are developing gene therapies to treat aromatic l-amino acid decarboxylase (AADC) deficiency. GTRI aims to get its candidate into the clinic in 2019. Agilispicked up its candidate from a university in Taiwan, which enrolled 18 patients in two clinical trials of the gene therapy. Those trials have taken the candidate toward a pivotal trial.

These programs may benefit from the joint venture. Working out of the Life Science Innovation Center of Kawasaki City, the joint venture intends to develop and produce AAVs for use in gene therapies against AADC deficiency and Parkinson's.

The joint venture marks the second time Agilis has looked outside of its walls for help with AAV vectors. Late in 2013, Agilis struck a deal with Intrexon that gave it access to the latters vector platform. Agilis is using the vectors to develop a treatment for Friedreichs ataxia.

Read more:

Agilis forms joint venture to advance gene therapy vectors ... - FierceBiotech

Man’s best mend Gene therapy reverses muscular dystrophy … – Digital Trends

Why it matters to you

Duchenne muscular dystrophy is a horrible disease which slowly attacks every muscle in the body. Gene therapy could help treat it -- whether you're a dog or a human.

As difficult as it is when our beloved pet dogs get old, its a whole lot worse if their decline is the result of a horrible disease like Duchenne muscular dystrophy, a genetic disorder characterized by progressive muscle degeneration and weakness. Fortunately, a team of gene-editing researchers from France and the U.K. have been working to develop gene therapy as an answer and its one that could help humans, too.

Their solution involves using gene therapy to restore muscle strength and stabilize clinical symptoms. This is achieved by way of a shortened version of the dystrophin gene, containing just 4,000 base pairs, which is combined with a viral vector and injected into patients.

Duchenne muscular dystrophy is a debilitating muscle-wasting disease affecting young boys and male animals, which is caused by inheritance of a damaged gene, George Dickson, professor of molecular cell biology at University of London, told Digital Trends. There is currently no very effective cure. Our work has involved producing a healthy functioning copy of the damaged gene in the lab, and then using a harmless virus to carry the gene into the affected muscles, a so-called gene therapy.

Please enable Javascript to watch this video

The gene treatment has so far been tested on twelve golden retrievers affected by Duchenne muscular dystrophy. After a single dose, the dogs dystrophin production the protein responsible for maintaining the integrity and strength of muscles returned to its normal levels. Best of all, the trial was carried out a few years back, and all signs point to this being a lasting treatment. A paper describing the work was recently published in the journal Nature Communications.

We now have treated a number of dogs affected by this condition with a single round of gene therapy, Dickson continued. At the correct dose, the results have been very encouraging with dogs looking very active and healthy over 3 years after the treatment. The treatment involves a simple intravenous infusion, a one-off treatment, and we have been very pleased and quite surprised at how effective the gene therapy has been given that muscle is a major tissue spread all over the body.

Next up, the researchers hope to extrapolate the findings to human children, since they they are roughly the same weight and display similar clinical symptoms to canines.

The goal is to expand this gene therapy treatment into human patients, boys suffering from the Duchenne muscular dystrophy disease, Dickson said. We have to scale up production of the gene therapy medicine for human use, complete more safety tests, and then embark on recruiting patients into full-scale clinical trials.

Should all go according to plan, itll be another reminder of why gene therapy is the future of medicine as we know it.

View original post here:

Man's best mend Gene therapy reverses muscular dystrophy ... - Digital Trends

Philly gene therapy company reports early promising hemophilia A results – Philly.com

Shares of Spark Therapeuticssurged nearly 20 percentWednesday after the Philadelphia gene therapy company revealed promisingresults from a study of its potential one-time therapy for hemophilia A.

Preliminary data from a Phase 1/2 dose-escalation clinical trial of SPK-8011showed human proof-of-concept in three participants, the drug maker said.

The encouraging start for hemophilia A reinforces the strength of our gene-therapy platform and positions us well to potentially transform the current treatment approach for this life-altering disease with a onetime intervention, said Katherine A. High, Sparks president and chief scientific officer.

Hemophilia is a genetic disorder caused by missing or defective factor VIII, a clotting protein. About 20,000 Americans live with hemophilia. The way the medical community has addressed the disorder is to ensure that patients have continuous injections of blood-clotting factors. Patients infuse themselves two to three times a week for the rest of their lives.

In the study, three patientsreceived infusions of vector genomes and no serious adverse events were reported, Spark said. One person has been followed for 23 weeks and another for 12 weeks. The initial dose created stable factor VIII levels with no spontaneous bleeds, the company said.

For a third patient, the genome dose was doubled and that persons factor VIII activity level is tracking proportionally higher, consistent with the dose escalation. So far, the drug has been safe and well tolerated, with no reports of serious adverse events, no thrombotic events, no immune responses, and no elevations of liver enzymes, the company said.

The data must be considered preliminary and one must be careful not to overinterpret them, said Cowen & Co. analyst Phil Nadeau in a client update. That being said, we find the results quite encouraging.

Despite a low starting dose, the gene therapy produced stable and clinically meaningful factor levels sufficient to prevent spontaneous bleeds in patients, Nadeau said. Moreover, the safety profile is clean thus far. The results suggest the company may be able to achieve greater factor levels at higher doses. We find SPK-8011s early data encouraging, and think they suggest that Spark has a viable and competitive hemophilia A program.

Spark will present full data at a medical conference in December.

The hemophilia A results, though early, along with previously reported data for the companys hemophilia B candidate, confirm Sparks thought leadership in hemophilia gene therapy, and the likelihood of achieving a leading position in the overall hemophilia market (currently $7 billion, growing to $14 billion in 2030), Chardan Capital Markets analyst Gbola Amusa said in a client note. Chardan raised its peak earnings forecast for Sparks hemophilia A therapy to $1.3 billion, up from $397 million.

Sparks lead drug, a treatment for rare inherited blindness, is under priority review with the U.S. Food and Drug Administration, with a possible approval date of Jan. 12, 2018. If approved, it would be the first gene therapy for a genetic disease in the United States.

Spark, which was spun out of Childrens Hospital of Philadelphia, reported a second-quarter financial loss of $74.4 million in the quarter ended June 30, or $2.40 per share, on revenue of $1.5 million from itscollaboration with Pfizer Inc. for hemophilia B.

Sparks shares have risen 58 percent since Jan. 1 and 37 percent in the last 12 months. The stock closed up 19.72 percent, or $13.13, to $79.72.

Published: August 2, 2017 1:12 PM EDT

We recently asked you to support our journalism. The response, in a word, is heartening. You have encouraged us in our mission to provide quality news and watchdog journalism. Some of you have even followed through with subscriptions, which is especially gratifying. Our role as an independent, fact-based news organization has never been clearer. And our promise to you is that we will always strive to provide indispensable journalism to our community. Subscriptions are available for home delivery of the print edition and for a digital replica viewable on your mobile device or computer. Subscriptions start as low as 25 per day. We're thankful for your support in every way.

Go here to see the original:

Philly gene therapy company reports early promising hemophilia A results - Philly.com

In Breakthrough, Scientists Edit a Dangerous Mutation From Genes in Human Embryos – New York Times

Weve always said in the past gene editing shouldnt be done, mostly because it couldnt be done safely, said Richard Hynes, a cancer researcher at the Massachusetts Institute of Technology who co-led the committee. Thats still true, but now it looks like its going to be done safely soon, he said, adding that the research is a big breakthrough.

What our report said was, once the technical hurdles are cleared, then there will be societal issues that have to be considered and discussions that are going to have to happen. Nows the time.

Scientists at Oregon Health and Science University, with colleagues in California, China and South Korea, reported that they repaired dozens of embryos, fixing a mutation that causes a common heart condition that can lead to sudden death later in life.

If embryos with the repaired mutation were allowed to develop into babies, they would not only be disease-free but also would not transmit the disease to descendants.

The researchers averted two important safety problems: They produced embryos in which all cells not just some were mutation-free, and they avoided creating unwanted extra mutations.

It feels a bit like a one small step for (hu)mans, one giant leap for (hu)mankind moment, Jennifer Doudna, a biochemist who helped discover the gene-editing method used, called CRISPR-Cas9, said in an email.

Scientists tried two techniques to remove a dangerous mutation. In the first, genetic scissors were inserted into fertilized eggs. The mutation was repaired in some of the resulting embryos but not always in every cell. The second method worked better: By injecting the scissors along with the sperm into the egg, more embryos emerged with repaired genes in every cell.

When gene-editing components were introduced into a fertilized egg, some embryos contained a patchwork of repaired and unrepaired cells.

Gene-editing

components inserted

after fertilization

Cell with

unrepaired

gene

Mosaicism in

later-stage embryo

When gene-editing components were introduced with sperm to the egg before fertilization, more embryos had repaired mutations in every cell.

Gene-editing components

inserted together with sperm,

before fertilization

In 42 of 58

embryos

tested, all

cells were

repaired

Uniform

later-stage embryo

When gene-editing components were introduced into a fertilized egg, some embryos contained a patchwork of repaired and unrepaired cells.

Gene-editing

components inserted

after fertilization

Cell with

unrepaired

gene

Mosaicism in

later-stage embryo

When gene-editing components were introduced with sperm to the egg before fertilization, more embryos had repaired mutations in every cell.

Gene-editing

components inserted

together with sperm,

before fertilization

In 42 of 58

embryos

tested, all

cells were

repaired

Uniform

later-stage embryo

I expect these results will be encouraging to those who hope to use human embryo editing for either research or eventual clinical purposes, said Dr. Doudna, who was not involved in the study.

Much more research is needed before the method could be tested in clinical trials, currently impermissible under federal law. But if the technique is found to work safely with this and other mutations, it might help some couples who could not otherwise have healthy children.

Potentially, it could apply to any of more than 10,000 conditions caused by specific inherited mutations. Researchers and experts said those might include breast and ovarian cancer linked to BRCA mutations, as well as diseases like Huntingtons, Tay-Sachs, beta thalassemia, and even sickle cell anemia, cystic fibrosis or some cases of early-onset Alzheimers.

You could certainly help families who have been blighted by a horrible genetic disease, said Robin Lovell-Badge, a professor of genetics and embryology at the Francis Crick Institute in London, who was not involved in the study.

You could quite imagine that in the future the demand would increase. Maybe it will still be small, but for those individuals it will be very important.

The researchers also discovered something unexpected: a previously unknown way that embryos repair themselves.

In other cells in the body, the editing process is carried out by genes that copy a DNA template introduced by scientists. In these embryos, the sperm cells mutant gene ignored that template and instead copied the healthy DNA sequence from the egg cell.

We were so surprised that we just couldnt get this template that we made to be used, said Shoukhrat Mitalipov, director of the Center for Embryonic Cell and Gene Therapy at Oregon Health and Science University and senior author of the study. It was very new and unusual.

The research significantly improves upon previous efforts. In three sets of experiments in China since 2015, researchers seldom managed to get the intended change into embryonic genes.

And some embryos had cells that did not get repaired a phenomenon called mosaicism that could result in the mutation being passed on as well as unplanned mutations that could cause other health problems.

In February, a National Academy of Sciences, Engineering and Medicine committee endorsed modifying embryos, but only to correct mutations that cause a serious disease or condition and when no reasonable alternatives exist.

Sheldon Krimsky, a bioethicist at Tufts University, said the main uncertainty about the new technique was whether reasonable alternatives to gene editing already exist.

As the authors themselves noted, many couples use pre-implantation genetic diagnosis to screen embryos at fertility clinics, allowing only healthy ones to be implanted. For these parents, gene editing could help by repairing mutant embryos so that more disease-free embryos would be available for implantation.

Hank Greely, director of the Center for Law and the Biosciences at Stanford, said creating fewer defective embryos also would reduce the number discarded by fertility clinics, which some people oppose.

The larger issue is so-called germline engineering, which refers to changes made to embryo that are inheritable.

If youre in one camp, its a horror to be avoided, and if youre in the other camp, its desirable, Dr. Greely said. Thats going to continue to be the fight, whether its a feature or a bug.

For now, the fight is theoretical. Congress has barred the Food and Drug Administration from considering clinical trials involving germline engineering. And the National Institutes of Health is prohibited from funding gene-editing research in human embryos. (The new study was funded by Oregon Health and Science University, the Institute for Basic Science in South Korea, and several foundations.)

The authors say they hope that once the method is optimized and studied with other mutations, officials in the United States or another country will allow regulated clinical trials.

I think it could be widely used, if its proven safe, said Dr. Paula Amato, a co-author of the study and reproductive endocrinologist at O.H.S.U. Besides creating more healthy embryos for in vitro fertilization, she said, it could be used when screening embryos is not an option or to reduce arduous IVF cycles for women.

Dr. Mitalipov has pushed the scientific envelope before, generating ethical controversy with a so-called three-parent baby procedure that would place the nucleus of the egg of a woman with defective cellular mitochondria into the egg from a healthy woman. The F.D.A. has not approved trials of the method, but Britain may begin one soon.

The new study involves hypertrophic cardiomyopathy, a disease affecting about one in 500 people, which can cause sudden heart failure, often in young athletes.

It is caused by a mutation in a gene called MYBPC3. If one parent has a mutated copy, there is a 50 percent chance of passing the disease to children.

Using sperm from a man with hypertrophic cardiomyopathy and eggs from 12 healthy women, the researchers created fertilized eggs. Injecting CRISPR-Cas9, which works as a genetic scissors, they snipped out the mutated DNA sequence on the male MYBPC3 gene.

They injected a synthetic healthy DNA sequence into the fertilized egg, expecting that the male genome would copy that sequence into the cut portion. That is how this gene-editing process works in other cells in the body, and in mouse embryos, Dr. Mitalipov said.

Instead, the male gene copied the healthy sequence from the female gene. The authors dont know why it happened.

Maybe human sex cells or gametes evolved to repair themselves because they are the only cells that transmit genes to offspring and need special protection, said Juan Carlos Izpisua Belmonte, a co-author and geneticist at the Salk Institute.

Out of 54 embryos, 36 emerged mutation-free, a significant improvement over natural circumstances in which about half would not have the mutation. Another 13 embryos also emerged without the mutation, but not in every cell.

The researchers tried to eliminate the problem by acting at an earlier stage, injecting the egg with the sperm and CRISPR-Cas9 simultaneously, instead of waiting to inject CRISPR-Cas9 into the already fertilized egg.

That resulted in 42 of 58 embryos, 72 percent, with two mutation-free copies of the gene in every cell. They also found no unwanted mutations in the embryos, which were destroyed after about three days.

The method was not perfect. The remaining 16 embryos had unwanted additions or deletions of DNA. Dr. Mitalipov said he believed fine-tuning the process would make at least 90 percent of embryos mutation-free.

And for disease-causing mutations on maternal genes, the same process should occur, with the fathers healthy genetic sequence being copied, he said.

But the technique will not work if both parents have two defective copies. Then, scientists would have to determine how to coax one gene to copy a synthetic DNA sequence, Dr. Mitalipov said.

Otherwise, he said, it should work with many diseases, a variety of different heritable mutations.

R. Alta Charo, a bioethicist at University of Wisconsin at Madison, who led the committee with Dr. Hynes, said the new discovery could also yield more information about causes of infertility and miscarriages.

She doubts a flood of couples will have edited children.

Nobodys going to do this for trivial reasons, Dr. Charo said. Sex is cheaper and its more fun than IVF, so unless youve got a real need, youre not going to use it.

A version of this article appears in print on August 3, 2017, on Page A1 of the New York edition with the headline: Scientists Repair A Risky Mutation In Human Embryo.

See the original post here:

In Breakthrough, Scientists Edit a Dangerous Mutation From Genes in Human Embryos - New York Times

Gene Therapy Could Cure Muscular Dystrophy for Dogs and Humans – Edgy Labs (blog)

There is new hope for muscular dystrophy patients as researchers, using gene therapy, successfully reversed the disease in dogs.

Gene therapy aims to replace missing or defective genes in the DNA of a given cell.

The technique has evolved over the years to become a viable therapy thats safe and effective, opening new paths in the management of many difficult diseases.

Not only can gene editing be used to treat pathologies, but it can also to prevent them. Only, were not there just yet.

Muscular dystrophy is the term used for a group of diseases in which musculature weakens and progressively degenerates until the patient loses most, ifnot all of their mobility.

Symptoms of muscular dystrophy often include general muscle weakness and degeneration, stiff joints, coordination and mobility troubles, and frequent falls.

In most cases a congenital condition, muscular dystrophy disorders are rare. Each disorder of muscular dystrophy is associated with distinct genetic mutations. The nature and location of the genetic mutation define the form of muscular dystrophy.

Although they can occur at any age, the onset of most MD disorders starts during childhood, and usually, affected persons dont live past 30 years of age, especially with particularly aggressive forms of the disease like Duchenne muscular dystrophy.

The most common and most studied form of muscular dystrophy is Duchenne muscular dystrophy (DMD), which affects 1 in 5,000 children at birth, and especially boys (1/3500).

Theres hope for children and other DMD patients, as a scientific experiment suggests that the disease could be reversed and a cure might be on the way.

An international research team, comprised of scientists from Genethon and Insermin France and the Royal Hollowayat the University of London UK, announced theyd managed to treat Duchenne muscular dystrophy (DMD) with gene therapy in dogs.

Their findings were published in the journal Nature Communications.

The team has shown the efficacy of gene therapy in restoring normal muscle function in 12 dogs (Golden Retrievers) affected by canine DMD, with a stabilization of clinical symptoms.

A video of these dogs before and after treatment can be found here.

Researchers injected highly functional micro-dystrophin genes (a short version of the dystrophin gene) through a drug vector (harmless virus) so that the repaired gene could produce the protein involved in muscle function.

2 years after the injection of the drug, researchers observed that all dogs demonstrated signs of significant restoration of their muscles and regained their motor skills. Not to mention that the same dogs werent expected to live past the age of 6 months.

Now, with the method has been shown to be safe and efficient in animals, the next logical step would human trials.

For the many people affected by this debilitating disease, this is a miraculous development.

More:

Gene Therapy Could Cure Muscular Dystrophy for Dogs and Humans - Edgy Labs (blog)

Chiesi hands back gene therapy to uniQure | BioPharma Dive – BioPharma Dive

Dive Brief:

Even as gene therapies are being touted as the next wave of innovation that could offer cures for certain genetic conditions, it remains to be seen whether these products are actually commercially viable. There has yet to be a gene therapy approved in the U.S. (although Spark Therapeutics' application is pending), but two of the transformative drugs have been on the market in Europe.

Yet neither of those commercially available gene therapies have found much success. GlaxoSmithKline plc. said just last week it is looking to move away from its rare disease portfolio, including the gene therapy Strimvelis. Meanwhile, uniQureannounced back in April it would not renew the marketing authorization application in Europe for its already-approved gene therapy Glybera.

This latest move by Chiesifurther exemplifies the challenges gene therapy producers face. The announcement ends a deal which has been in place since 2013. Chiesisaid in a statement that the decision was "driven by recent changes in our strategic priorities."

uniQuretried to put brave face on the news, but partnership exits are rarely good news for a biotech.

"By regaining unencumbered, global rights to a late-stage program that has demonstrated significant clinical benefit for patients with hemophilia B, we believe uniQure is better positioned to accelerate the global clinical development plan, maximize shareholder return on our pipeline and take advantage of new potential opportunities related to the program," said CEO Matthew Kapusta.

The company recently announced positive developments in a Phase 1/2 trial of AMT-060, which supported further expansion of the eligibility of the adeno-associated virus 5 (AAV5) gene therapy to nearly all patients with hemophilia B. Meanwhile, investors are paying close attention to Spark's gene therapy for hemophilia B, which is also in early-to mid-stage development.

Read more from the original source:

Chiesi hands back gene therapy to uniQure | BioPharma Dive - BioPharma Dive

New Gene Therapy to Fix Dystrophin Deficiency in DMD Shows Promise in Mice, Study Shows – Muscular Dystrophy News

Researchers at the University of Missouri have developed a new method to efficiently deliver the correct form of dystrophin gene to muscles as a way to correct the faulty gene that characterizes Duchenne muscular dystrophy (DMD), a mouse study shows.

Their study, A Five-Repeat Micro-Dystrophin Gene Ameliorated Dystrophic Phenotype in the Severe DBA/2J-mdx Model of Duchenne Muscular Dystrophy, appearedin the journal Molecular TherapyMethods & Clinical Development.

DMD is caused by a modification of the gene that encodes the dystrophin protein, which is essential for normal muscle activity. Such mutations interfere withproduction of the functional protein, severely affecting muscle fiber structure and strength.

Correcting the faulty gene could potentially treatthis disease. Several attempts atgene therapyhave been tried, but all have failed to efficiently reverse all DMD symptoms.

Gene therapy commonly uses vectors basedviral genetic sequences to achieve the desired gene transfer capacity. The therapeutic potential of these techniques rely not only on the delivery system, but also on the sequence of the gene of interest that is used. In this case, smaller versions of dystrophin known asmicrodystrophinmust be used, since its natural form is just too big to be useful in gene therapy.

There have been other gene-transfer vectors attempted in the past (such as adenoviral vector, herpes simplex virus and plasmid), but they have largely been unsuccessful due to the complexity of the disease, challenges associated with delivery, and the large size of the native dystrophin gene, the studys senior author, Dongsheng Duan, said in a news release.

Duans team used an engineered form of the adeno-associated virus (AAV) vector to replace the damaged gene specifically in the muscles.

Researchers alsoused a version of the dystrophin gene that can potentiallyminimize the toxicity signs commonly associated with such methods, such as inadequate blood supply and fatigue during muscle contraction. This AAV viral vector has also been used in the past, but this is the first time researchers have combined it with such a version of dystrophin.

This strategy boostedlevels of dystrophin protein in the muscles of mice models of DMD, and significantly reduced some disease symptoms. Yet researchers could not accurately measure the impact of this new potential therapy to correct DMD-associated effects on the hearts of the animals.

Human studies have shown that one-time intramuscular injection of an AAV vector can result in the expression of a therapeutic protein for many years. For example, a study showed Factor IX expression for 10 years in a hemophilia patient, Duan said. In preclinical studies in murine and canine models, we have also observed persistent multiyear microdystrophin expression from AAV vectors. In the case of mice, a single injection can lead to microdystrophin expression throughout the lifespan.

Read the original post:

New Gene Therapy to Fix Dystrophin Deficiency in DMD Shows Promise in Mice, Study Shows - Muscular Dystrophy News

Philly drug maker seeks approval in EU for gene therapy – Philly.com

Philadelphia gene therapy company Spark Therapeutics has applied to the European Medicines Agency for approval to sell its treatment of rare inherited blindness in the European Union.

The experimental therapy, Luxturna, or voretigene neparvovec, is under priority review with the U.S. Food and Drug Administration, with a possible approval date of Jan. 12, 2018.

Spark was spun out of Childrens Hospital of Philadelphia, based on research led by Katherine A. High, Sparks cofounder, president, and chief scientific officer. If approved, it would be the first gene therapy for a genetic disease in the United States.

With Luxturna now in regulatory review on both sides of the Atlantic, we are building out our medical and commercial infrastructure to bring the drug to patients, said John Furey, Sparks chief operating officer. For the first time, adults and children, who otherwise would progress to complete blindness, have hope for a potential treatment option that may restore their vision, he said.

About 3,500 people in the United States and Europe live with the disease.

The review period will begin in Europe once the agency validates the application, Spark said.

Published: August 1, 2017 3:01 AM EDT | Updated: August 1, 2017 11:40 AM EDT

We recently asked you to support our journalism. The response, in a word, is heartening. You have encouraged us in our mission to provide quality news and watchdog journalism. Some of you have even followed through with subscriptions, which is especially gratifying. Our role as an independent, fact-based news organization has never been clearer. And our promise to you is that we will always strive to provide indispensable journalism to our community. Subscriptions are available for home delivery of the print edition and for a digital replica viewable on your mobile device or computer. Subscriptions start as low as 25 per day. We're thankful for your support in every way.

See original here:

Philly drug maker seeks approval in EU for gene therapy - Philly.com

Agilis Biotherapeutics and Gene Therapy Research Institution Enter into Strategic Partnership – Business Wire (press release)

CAMBRIDGE, Mass. & TOKYO--(BUSINESS WIRE)--Agilis Biotherapeutics, Inc. (Agilis), a biotechnology company advancing innovative DNA therapeutics for rare genetic diseases that affect the central nervous system (CNS), and Gene Therapy Research Institution Co, Ltd. (GTRI), a corporation with the mission of developing and delivering of the safest and most efficient gene therapies, today announced that the companies have completed a manufacturing and collaboration partnership joint venture (JV) to advance adeno-associated virus (AAV) gene therapies. The JV was initiated earlier this year in connection with a grant from the Japanese Ministry of Trade, Economics and Industry (METI) and Japan External Trade Organization (JETRO) for the development of a state-of-the-art AAV manufacturing facility in Japan. GTRI was co-founded by Professor Shin-ichi Muramatsu, M.D., a leading pioneer in gene therapy who has performed basic science and clinical research in the field for over two decades.

The JV, headquartered in Japan, will initially focus on developing and manufacturing AAV gene therapy vectors using Sf9 baculovirus and HEK293 mammalian cell systems and operate a process development and production facility located in the Tokyo area designed to meet international manufacturing standards, including cGMP, GCTP and PIC/S GMP requirements. Agilis and GTRI will also collaborate to expedite the development, approval and commercialization of select gene therapies in specific CNS diseases. Terms of the joint venture were not disclosed.

We are pleased to collaborate with Agilis to leverage each organizations capabilities and know-how, advance the manufacturing state-of-the art for gene therapy, and develop novel gene therapies, commented Katsuhito Asai, Chief Executive Officer of GTRI and a Director of the joint venture. Our partnership will seek to capitalize on the strong recent progress in the field of gene therapy and expedite the development of innovative gene therapies for patients in need, with a major emphasis on the quality production of safe, effective therapeutics.

We are thrilled to partner with GTRI, said Mark Pykett, Agilis CEO and a Director of the joint venture. We believe that our partnership will enhance the efforts of both organizations, build important shared production capabilities, and accelerate development and commercialization of important gene therapies. We look forward to working with GTRI on a range of initiatives.

Agilis Biotherapeutics

Agilis is advancing innovative gene therapies designed to provide long-term efficacy for patients with debilitating, often fatal, rare genetic diseases that affect the central nervous system. Agilis gene therapies are engineered to impart sustainable clinical benefits by inducing persistent expression of a therapeutic gene through precise targeting and restoration of lost gene function to achieve long-term efficacy. Agilis rare disease programs are focused on gene therapy for AADC deficiency, Friedreichs ataxia, and Angelman syndrome, all rare genetic diseases that include neurological deficits and result in physically debilitating conditions.

We invite you to visit our website at http://www.agilisbio.com

About GTRI

GTRI, a bio-tech venture in Japan, was founded in May 2014 based on the pioneering research of Dr. Shin-ichi Muramatsu, focusing on gene therapy using AAV vector as the leading company in Japan in this field. Its pipeline includes more than 20 diseases, targeting CNS diseases and monogenic disorders, such as Parkinsons disease, AADC deficiency, ALS, Alzheimers disease, spinocerebellar degeneration, Tay-Sachs disease, GLUT1 deficiency, and others.

Dr. Muramatsu, PhD, MD, of Jichi Medical University, is one of the top researchers of AAV vectors and AAV-mediated gene therapy in the world. He originated AAV3 in 1995 during his research at the NIH, USA, and afterwards developed his original modified AAV3/9 in Japan, which enables to deliver the gene of interest effectively in CNS through the blood-brain barrier.

Safe Harbor Statement

Some of the statements made in this press release are forward-looking statements. These forward-looking statements are based upon our current expectations and projections about future events and generally relate to our plans, objectives and expectations for the development of our business. Although management believes that the plans and objectives reflected in or suggested by these forward-looking statements are reasonable, all forward-looking statements involve risks and uncertainties and actual future results may be materially different from the plans, objectives and expectations expressed in this press release.

Go here to read the rest:

Agilis Biotherapeutics and Gene Therapy Research Institution Enter into Strategic Partnership - Business Wire (press release)

Chiesi dumps uniQure’s hemophilia B gene therapy – FierceBiotech

Chiesi has cut its ties to uniQures hemophilia B gene therapy. The split gives uniQure full rights to AMT-060 but leaves it without a partner to cofund R&D as it closes in on the start of a pivotal trial.

Italian drugmaker Chiesi picked up the rights to commercialize AMT-060 in certain markets in 2013 as part of a deal that also gave it a piece of Glybera, the gene therapy that made history by coming to market in Europe only to flop commercially. Chiesi backed out of the Glybera agreement earlier this year and has now completed its split from uniQure by terminating the hemophilia B pact.

Amsterdam, the Netherlands-based uniQure framed the termination as it reacquiring the rights to AMT-060, rather than Chiesi dumping the program. But as the deal will see money transfer from Chiesi to uniQure and the former stated a shift in priorities prompted it to sever ties to AMT-060, it seems clear the Italian drugmaker wanted to exit the agreement.

That leaves uniQure facing the prospect of taking AMT-060 into a pivotal trial without the financial support of a partner. Chiesi and uniQure have evenly shared R&D costs since 2013. The loss of the support of Chiesi will add $3 million to uniQures outlay this year, although the Dutch biotech still thinks it has enough cash to take it into 2019.

After a trying time on public markets dotted with stock drops following unfavorable comparisons to Spark Therapeutics rival hemophilia B program, uniQure is less well equipped to raise more money than in the past. But uniQure CEO Matthew Kapusta spun the regaining of full rights to the gene therapy as a boost for the company.

We believe uniQure is better positioned to accelerate the global clinical development plan, maximize shareholder return on our pipeline and take advantage of new potential opportunities related to the program, Kapusta said in a statement.

If the potential opportunities are to include a deal covering AMT-060, uniQure must persuade a potential partner of the merits of its asset. UniQure has sought to focus attention on the durable clinical benefits associated with AMT-060 but investors have fixated on Sparks clear advantage in terms of Factor IX activity.

Go here to read the rest:

Chiesi dumps uniQure's hemophilia B gene therapy - FierceBiotech