Search Immortality Topics:

Page 121«..1020..120121122123..130140..»


Category Archives: Genetic Therapy

Tim Kelly Joins AskBio as President of Manufacturing to Lead AAV Vector Production for Gene Therapy – Yahoo Finance

RESEARCH TRIANGLE PARK, N.C., March 12, 2020 (GLOBE NEWSWIRE) -- Asklepios BioPharmaceutical, Inc. (AskBio), a leading, clinical-stage adeno-associated virus (AAV) gene therapy company, today announced the appointment of Tim Kelly, PhD, as President of Manufacturing. He will oversee all manufacturing functions at AskBio and its Viralgen affiliate for the production of clinical- and commercial-scale AAV vectors. Prior to joining AskBio, Dr. Kelly was the President and Chief Executive Officer at KBI Biopharma, a contract services organization that provides drug development and biomanufacturing services to pharmaceutical and biotechnology companies globally.

AskBio currently has clinical studies underway in late-onset Pompe disease and congestive heart failure. To meet the growing demand for AAV gene therapies, the company is investing in manufacturing innovation, talent and capacity that will allow it to effectively and efficiently serve patient populations.

Our goal at AskBio is to continue advancing production technology to drive down costs to make gene therapies accessible to all patients who may benefit from treatment. I am delighted that Tim has joined the company to help us shape the future of manufacturing, said Sheila Mikhail, JD, MBA, Chief Executive Officer and co-founder at AskBio. He brings a wealth of experience successfully leading therapeutic development and manufacturing and fostering the entrepreneurial, patient-focused culture that drives us at AskBio.

In January, Viralgen broke ground on a 300,000 square foot commercial facility in San Sebastin, Spain, with production expected to start in the spring of 2022, complementing the clinical-scale production currently carried out at its existing cGMP facility.

AskBios technology is truly transforming human health, and I am incredibly excited to help translate our innovations into reliable delivery of AAV gene therapy products to patients in need, said Dr. Kelly.

More about Tim KellyDr. Kelly has more than 20 years of experience in the development and manufacture of therapeutic proteins. He has overseen biopharmaceutical services for over 320 molecules at all stages of development and commercialization and supported numerous successful FDA and international regulatory inspections throughout his career. He began his tenure at KBI Biopharma in 2005, initially acting as Vice President of Biopharmaceutical Development, where he led the establishment and growth of KBIs analytical development, formulation development and cGMP laboratory services business. He subsequently served as Executive Vice President of Operations with responsibility for KBIs development and manufacturing functions in North Carolina and Colorado before becoming President and Chief Executive Officer. Prior to KBI, he directed the quality control function for Diosynth Biotechnology, where he supported clinical and commercial biopharmaceutical products. Dr. Kelly earned his PhD in molecular genetics and biochemistry from Georgia State University.

About AskBioFounded in 2001, Asklepios BioPharmaceutical, Inc. (AskBio) is a privately held, clinical-stage gene therapy company dedicated to improving the lives of children and adults with genetic disorders. AskBios gene therapy platform includes an industry-leading proprietary cell line manufacturing process called Pro10 and an extensive AAV capsid and promoter library. Based in Research Triangle Park, North Carolina, the company has generated hundreds of proprietary third-generation AAV capsids and promoters, several of which have entered clinical testing. An early innovator in the space, the company holds more than 500 patents in areas such as AAV production and chimeric and self-complementary capsids. AskBio maintains a portfolio of clinical programs across a range of neurodegenerative and neuromuscular indications with a current clinical pipeline that includes therapeutics for Pompe disease, limb-girdle muscular dystrophy 2i/R9 and congestive heart failure, as well as out-licensed clinical indications for hemophilia (Chatham Therapeutics acquired by Takeda) and Duchenne muscular dystrophy (Bamboo Therapeutics acquired by Pfizer). Learn more at https://www.askbio.com or follow us on LinkedIn.

Story continues

Read more:
Tim Kelly Joins AskBio as President of Manufacturing to Lead AAV Vector Production for Gene Therapy - Yahoo Finance

Posted in Genetic Therapy | Comments Off on Tim Kelly Joins AskBio as President of Manufacturing to Lead AAV Vector Production for Gene Therapy – Yahoo Finance

New drugs are costly and unmet need is growing – The Economist

Mar 12th 2020

BEING ABLE to see all the details of the genome at once necessarily makes medicine personal. It can also make it precise. Examining illness molecule by molecule allows pharmaceutical researchers to understand the pathways through which cells act according to the dictates of genes and environment, thus seeing deep into the mechanisms by which diseases cause harm, and finding new workings to target. The flip side of this deeper understanding is that precision brings complexity. This is seen most clearly in cancer. Once, cancers were identified by cell and tissue type. Now they are increasingly distinguished by their specific genotype that reveals which of the panoply of genes that can make a cell cancerous have gone wrong in this one. As drugs targeted against those different mutations have multiplied, so have the options for oncologists to combine them to fit their patients needs.

Cancer treatment has been the most obvious beneficiary of the genomic revolution but other diseases, including many in neurology, are set to benefit, too. Some scientists now think there are five different types of diabetes rather than two. There is an active debate about whether Parkinsons is one disease that varies a lot, or four. Understanding this molecular variation is vital when developing treatments. A drug that works well on one subtype of a disease might fail in a trial that includes patients with another subtype against which it does not work at all.

Thus how a doctor treats a disease depends increasingly on which version of the disease the patient has. The Personalised Medicine Coalition, a non-profit advocacy group, examines new drugs approved in America to see whether they require such insights in order to be used. In 2014, it found that so-called personalised medicines made up 21% of the drugs newly approved for use by Americas Food and Drug Administration (FDA). In 2018 the proportion was twice that.

Two of those cited were particularly interesting: Vitrakvi (larotrectinib), developed by Loxo Oncology, a biotech firm, and Onpattro (patisiran), developed by Alnylam Pharmaceuticals. Vitrakvi is the first to be approved from the start as tumour agnostic: it can be used against any cancer that displays the mutant protein it targets. Onpattro, which is used to treat peripheral-nerve damage, is the first of a new class of drugssmall interfering RNAs, or siRNAsto be approved. Like antisense oligonucleotides (ASOs), siRNAs are little stretches of nucleic acid that stop proteins from being made, though they use a different mechanism.

Again like ASOs, siRNAs allow you to target aspects of a disease that are beyond the reach of customary drugs. Until recently, drugs were either small molecules made with industrial chemistry or bigger ones made with biologynormally with genetically engineered cells. If they had any high level of specificity, it was against the actions of a particular protein, or class of proteins. Like other new techniques, including gene therapies and anti-sense drugs, siRNAs allow the problem to be tackled further upstream, before there is any protein to cause a problem.

Take the drugs that target the liver enzyme PCSK9. This has a role in maintaining levels of bad cholesterol in the blood; it is the protein that was discovered through studies of families in which congenitally high cholesterol levels led to lots of heart attacks. The first generation of such drugs were antibodies that stuck to the enzyme and stopped it working. However, the Medicines Company, a biotech firm recently acquired by Novartis, won approval last year for an siRNA called inclisiran that interferes with the expression of the gene PCSK9thus stopping the pesky protein from being made in the first place. Inclisiran needs to be injected only twice a year, rather than once a month, as antibodies do.

New biological insights, new ways of analysing patients and their disease and new forms of drug are thus opening up a wide range of therapeutic possibilities. Unfortunately, that does not equate to a range of new profitable opportunities.

Thanks in part to ever better diagnosis, there are now 7,000 conditions recognised as rare diseases in America, meaning that the number of potential patients is less than 200,000. More than 90% of these diseases have no approved treatment. These are the diseases that personalised, precision medicine most often goes after. Nearly 60% of the personalised medicines approved by the FDA in 2018 were for rare diseases.

Zolgensma is the most expensive drug ever brought to market.

That might be fine, were the number of diseases stable. But precision in diagnosis is increasingly turning what used to be single diseases into sets of similar-looking ones brought about by distinctly different mechanisms, and thus needing different treatment. And new diseases are still being discovered. Medical progress could, in short, produce more new diseases than new drugs, increasing unmet need.

Some of it will, eventually, be met. For one thing, there are government incentives in America and Europe for the development of drugs for rare diseases. And, especially in America, drugs for rare diseases have long been able to command premium prices. Were this not the case, Novartis would not have paid $8.7bn last year to buy AveXis, a small biotech firm, thereby acquiring Zolgensma, a gene therapy for spinal muscular atrophy (SMA). Most people with SMA lack a working copy of a gene, SMN1, which the nerve cells that control the bodys muscles need to survive. Zolgensma uses an empty virus-like particle that recognises nerve cells to deliver working copies of the gene to where it is needed. Priced at $2.1m per patient, it is the most expensive drug ever brought to market. That dubious accolade might not last long. BioMarin, another biotech firm, is considering charging as much as $3m for a forthcoming gene therapy for haemophilia.

Drug firms say such treatments are economically worthwhile over the lifetime of the patient. Four-fifths of children with the worst form of SMA die before they are four. If, as is hoped, Zolgensma is a lasting cure, then its high cost should be set against a half-century or more of life. About 200 patients had been treated in America by the end of 2019.

But if some treatments for rare diseases may turn a profit, not all will. There are some 6,000 children with SMA in America. There are fewer than ten with Jansens disease. When Dr Nizar asked companies to help develop a treatment for it, she says she was told your disease is not impactful. She wrote down the negative responses to motivate herself: Every day I need to remind myself that this is bullshit.

A world in which markets shrink, drug development gets costlier and new unmet needs are ceaselessly discovered is a long way from the utopian future envisaged by the governments and charities that paid for the sequencing of all those genomes and the establishment of the worlds biobanks. As Peter Bach, director of the Centre for Health Policy and Outcomes, an academic centre in New York, puts it with a degree of understatement: if the world needs to spend as much to develop a drug for 2,000 people as it used to spend developing one for 100,000, the population-level returns from medical research are sharply diminishing.

And it is not as if the costs of drug development have been constant. They have gone up. What Jack Scannell, a consultant and former pharmaceutical analyst at UBS, a bank, has dubbed Erooms lawEroom being Moore, backwardsshows the number of drugs developed for a given amount of R&D spending has fallen inexorably, even as the amount of biological research skyrocketed. Each generation assumes that advances in science will make drugs easier to discover; each generation duly advances science; each generation learns it was wrong.

For evidence, look at the way the arrival of genomics in the 1990s lowered productivity in drug discovery. A paper in Nature Reviews Drug Discovery by Sarah Duggers from Columbia University and colleagues argues that it brought a wealth of new leads that were difficult to prioritise. Spending rose to accommodate this boom; attrition rates for drugs in development subsequently rose because the candidates were not, in general, all that good.

Today, enthused by their big-science experience with the genome and enabled by new tools, biomedical researchers are working on exhaustive studies of all sorts of other omes, including proteomesall the proteins in a cell or body; microbiomesthe non-pathogenic bacteria living in the mouth, gut, skin and such; metabolomessnapshots of all the small molecules being built up and broken down in the body; and connectomes, which list all the links in a nervous system. The patterns they find will doubtless produce new discoveries. But they will not necessarily, in the short term, produce the sort of clear mechanistic understanding which helps create great new drugs. As Dr Scannell puts it: We have treated the diseases with good experimental models. Whats left are diseases where experiments dont replicate people. Data alone canot solve the problem.

Daphne Koller, boss of Insitro, a biotech company based in San Francisco, shares Dr Scannells scepticism about the way drug discovery has been done. A lot of candidate drugs fail, she says, because they aim for targets that are not actually relevant to the biology of the condition involved. Instead researchers make decisions based on accepted rules of thumb, gut instincts or a ridiculous mouse model that has nothing to do with what is actually going on in the relevant human diseaseeven if it makes a mouse look poorly in a similar sort of way.

But she also thinks that is changing. Among the things precision biology has improved over the past five to 10 years have been the scientists own tools. Gene-editing technologies allow genes to be changed in various ways, including letter by letter; single-cell analysis allows the results to be looked at as they unfold. These edited cells may be much more predictive of the effects of drugs than previous surrogates. Organoidsself-organised, three-dimensional tissue cultures grown from human stem cellsoffer simplified but replicable versions of the brain, pancreas, lung and other parts of the body in which to model diseases and their cures.

Insitro is editing changes into stem cellswhich can grow into any other tissueand tracking the tissues they grow into. By measuring differences in the development of very well characterised cells which differ in precisely known ways the company hopes to build more accurate models of disease in living cells. All this work is automated, and carried out on such a large scale that Dr Koller anticipates collecting many petabytes of data before using machine learning to make sense of it. She hopes to create what Dr Scannell complains biology lacks and what drug designers need: predictive models of how genetic changes drive functional changes.

There are also reasons to hope that the new upstream drugsASOs, siRNAs, perhaps even some gene therapiesmight have advantages over todays therapies when it comes to small-batch manufacture. It may also prove possible to streamline much of the testing that such drugs go through. Virus-based gene-therapy vectors and antisense drugs are basically platforms from which to deliver little bits of sequence data. Within some constraints, a platform already approved for carrying one message might be fast-tracked through various safety tests when it carries another.

One more reason for optimism is that drugs developed around a known molecule that marks out a diseasea molecular markerappear to be more successful in trials. The approval process for cancer therapies aimed at the markers of specific mutations is often much shorter now than it used to be. Tagrisso (osimertinib), an incredibly specialised drug, targets a mutation known to occur only in patients already treated for lung cancer with an older drug. Being able to specify the patients who stand to benefit with this degree of accuracy allows trials to be smaller and quicker. Tagrisso was approved less than two years and nine months after the first dose was given to a patient.

With efforts to improve the validity of models of disease and validate drug targets accurately gaining ground, Dr Scannell says he is sympathetic to the proposal that, this time, scientific innovation might improve productivity. Recent years have seen hints that Erooms law is being bent, if not yet broken.

If pharmaceutical companies do not make good on the promise of these new approaches then charities are likely to step in, as they have with various ASO treatments for inherited diseases. And they will not be shackled to business models that see the purpose of medicine as making drugs. The Gates Foundation and Americas National Institutes of Health are investing $200m towards developing treatments based on rewriting genes that could be used to tackle sickle-cell disease and HIVtreatments that have to meet the proviso of being useful in poor-country clinics. Therapies in which cells are taken out of the body, treated in some way and returned might be the basis of a new sort of business, one based around the ability to make small machines that treat individuals by the bedside rather than factories which produce drugs in bulk.

There is room in all this for individuals with vision; there is also room for luck: Dr Nizar has both. Her problem lies in PTH1R, a hormone receptor; her PTH1R gene makes a form of it which is jammed in the on position. This means her cells are constantly doing what they would normally do only if told to by the relevant hormone. A few years ago she learned that a drug which might turn the mutant receptor off (or at least down a bit) had already been characterisedbut had not seemed worth developing.

The rabbit, it is said, outruns the fox because the fox is merely running for its dinner, while the rabbit is running for its life. Dr Nizars incentives outstrip those of drug companies in a similar way. By working with the FDA, the NIH and Massachusetts General Hospital, Dr Nizar helped get a grant to make enough of the drug for toxicology studies. She will take it herself, in the first human trial, in about a years time. After that, if things go well, her childrens pain may finally be eased.

This article appeared in the Technology Quarterly section of the print edition under the headline "Kill or cure?"

Read the original here:
New drugs are costly and unmet need is growing - The Economist

Posted in Genetic Therapy | Comments Off on New drugs are costly and unmet need is growing – The Economist

Genetic testing hopes to speed up drug development in rare eye diseases – – pharmaphorum

When it comes to fighting rare diseases, the biggest barrier to overcome is often the lack of knowledge around the condition. A free genetic test is hoping to combat this and speed up drug development in inherited retinal diseases.

The small patient populations in rare diseases and resulting lack of understanding about the conditions natural histories has many implications for researchers trying to develop orphan drugs.

Clinical trials become difficult to set up due to the limited experience in conducting studies for these diseases, including the lack of understanding of what endpoints will be most sensitive, says Daniel de Boer, chief executive officer of ProQR Therapeutics, which is developing RNA therapies to treat inherited retinal diseases (IRD).

Standard trial designs are not optimised to obtain adequate safety and efficacy data from small numbers of patients, and trials are frequently conducted on an international scale with various regulatory agency oversights.

Like in many rare diseases, there are some dire statistics in IRDs. There are as many as 300 inherited retinal diseases known today, affecting over two million people but only a fraction of these patients have access to a therapy.

IRDs also require precise and targeted treatments to deliver an accurate genetic diagnosis.

The programme is taking a crowdsourcing approach to gathering the data researches need when working on orphan drugs

From talking to retina specialists, we know that barriers to genetic testing remain a challenge for patients, says de Boer. For instance, at ProQR we are developing RNA therapies for specific mutations causing Lebers congenital amaurosis 10 (LCA10), Usher syndrome type 2 and autosomal dominant retinitis pigmentosa (adRP). However, there are hundreds of genetic mutations that can cause inherited retinal diseases.

Without knowing the exact mutations and the prevalence in which they occur, it can be difficult to design therapies and execute clinical trials for these rare and ultra-rare diseases.

In an effort to give researchers the best possible start when developing treatments for eye diseases, The Foundation Fighting Blindness has founded the My Retina Tracker programme, in partnership with Blueprint Genetics and InformedDNA.

The programme is a global, patient-driven registry for inherited retinal diseases (IRD). It offers free, open access genetic testing and genetic counseling for individuals living in the United States with a clinical diagnosis of IRD.

The genetic counseling consists of reviewing a patients medical and family history, interpretation of genetic variants, correlation with the disease, and medical management recommendations.

The programme also gives individuals the opportunity to contribute to focus groups, patient journey analyses, research studies, and the opportunity to be enrolled in relevant natural history studies and clinical trials.

My Retina Tracker also records subjective information from patients regarding how their disease affects their day to day activities. This recording option is available to doctors, allowing them to enter any additional information during appointments with patients.

In other words, the programme is taking a crowdsourcing approach to gathering the data researches need when working on drugs for IRDs.

Since its establishment in 2017 over 6,000 people with an IRD have participated, with the Foundation FIghting Blindness aiming to test over 20,000 patients within the next few years.

This has created a powerful genotype/phenotype database, says de Boer.

ProQR has recently become the first industry partner for the programme, giving it access to expert physicians and de-identified data from specific participating IRD patients.

This data has the potential to expedite the advancement of new treatments by identifying the mutations causing inherited rare diseases, de Boer explains.

The companys RNA therapies aim to edit RNA, the blueprint for proteins. Targeting RNA does not affect a persons DNA, the source of the genetic code, but instead reduces the risk of permanent side effects.

For inherited retinal diseases, RNA therapies can be administered through intravitreal injections, which is a far less invasive procedure than the retinal surgery required for gene therapies, de Boer says. RNA therapies do not need a vector to enter cells rather, intravitreal injection allows the drug to be distributed to the entire retina, which is where the therapy is needed.

The data can have additional benefits in clinical trial recruitment by identifying patients with these rare diseases.

Data like this is instrumental when recruiting participants for a trial. Treatments for inherited retinal diseases, like our RNA therapies, are specifically designed for a certain genetic mutation. Identifying the exact mutation that causes the disease encourages patients to participate in trials that target their mutation.

There are also benefits in diagnosis a confirmed genetic diagnosis may help patients and doctors better understand prognosis and help guide medical management.

With this information, physicians can determine the correct diagnosis and potential treatment for a patient, says de Boer. The data may match the patients mutation to an existing therapy or an ongoing clinical trial.

In terms of research and the development of therapeutics, it is crucial to increase our knowledge on the prevalence of the specific gene variants that cause an IRD.

Read this article:
Genetic testing hopes to speed up drug development in rare eye diseases - - pharmaphorum

Posted in Genetic Therapy | Comments Off on Genetic testing hopes to speed up drug development in rare eye diseases – – pharmaphorum

HORAMA Strengthens Its Management Team With the Appointment of Rodolphe Clerval as Chief Business Officer (CBO) – Yahoo Finance

HORAMA, a clinical stage gene therapy company in ophthalmology, today announced that Rodolphe Clerval has been appointed as Chief Business Officer (CBO). Rodolphe has a strong track record of executing strategic partnerships, business development deals and financing efforts. He will be responsible for business and corporate development, supporting HORAMA in operating a portfolio of gene therapy programs in inherited retinal dystrophies.

We are delighted to welcome Rodolphe, who brings us solid international experience in business development and corporate development strategy, acquired in biotech and pharmaceutical companies. His precious skills will allow us to prepare the next steps and to optimize the potential of our gene therapy products in ophthalmic diseases, for which there is a strong and unmet medical need , says Christine Placet, CEO, HORAMA.

Rodolphe Clerval (44) has over 18 years international experience in the pharma and biotech industry. Prior to joining HORAMA, Mr. Clerval was co-founder and Chief Business Officer at Enterome. During his tenure at Enterome, he has executed over 15 transactions, including major industrial partnerships with Takeda, J&J, BMS, Abbvie and Nestle Health Sciences, totalizing over 100m in upfront and R&D payments and in equity investments. He was also actively involved in fundraising rounds. Rodolphe previously worked with TcLand Expression and Genzyme in business development, and with Natixis as sell-side equity analyst. He started his career at Aventis Animal Nutrition as a scientist.

Rodolphe graduated with a degree in Biochemical Engineering from Polytech Marseille and he is a Certified European Financial Analyst from EFFAS/SFAF.

"HORAMA is a remarkable company, combining top gene therapy research, clinical stage programs with a strong team of industry leaders. I am thrilled to join HORAMA and to support the company in delivering best-in-class therapies in inherited retinal dystrophies", said Rodolphe Clerval, CBO, Horama.

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

Gene therapy is being developed with an aim to treat rare conditions with limited or no treatment options.Genetic disorders occur due to gene mutations, which can result in incorrect protein synthesis. Gene therapy is used to introduce a healthy gene into cells to allow the synthesis of a functional protein. Growing awareness and acceptance of gene therapy for various disease treatments are favouring market growth.The global gene therapy market is estimated to reach $5.5 billion by 2026, while the global ophthalmology market is projected to grow to $43 billion by 2026 (April 2019 report issued by Grand View Research, Inc.).

About HORAMA

At HORAMA, we believe in gene therapy to treat a broad range of inherited disorders.Our focus is on Inherited Retinal Dystrophies with our lead clinical program targeting patients with PDE6B gene mutations, a condition which leads to progressive vision loss in children and adults leading to legal blindness.Our team is pushing the boundaries of gene therapy by advancing next generation delivery platforms that will improve effectiveness and coverage of gene transfer to address multiple diseases. For more information, please go to: http://www.horama.fr.

View source version on businesswire.com: https://www.businesswire.com/news/home/20200311005023/en/

Contacts

HORAMA Christine Placet c.placet@horama.fr

Press: ALIZE RP Caroline Carmagnol Tel: +33 (0)6 64 18 99 59caroline@alizerp.com

See the original post:
HORAMA Strengthens Its Management Team With the Appointment of Rodolphe Clerval as Chief Business Officer (CBO) - Yahoo Finance

Posted in Genetic Therapy | Comments Off on HORAMA Strengthens Its Management Team With the Appointment of Rodolphe Clerval as Chief Business Officer (CBO) – Yahoo Finance

Solid Biosciences Reports Fourth Quarter and Full Year 2019 Financial Results and Provides Business Update – Yahoo Finance

Biopsy results from the third patient dosed at 2E14 vg/kg in the SGT-001 IGNITE DMD clinical trial provide further support for continued development

Solid continues to make progress to address the IGNITE DMD clinical hold and advance the next steps for the SGT-001 program

CAMBRIDGE, Mass., March 12, 2020 (GLOBE NEWSWIRE) -- Solid Biosciences Inc. (SLDB) today reported financial results for the fourth quarter and full year ending December 31, 2019 and provided a business update. We are working to advance our lead program, SGT-001, a gene therapy candidate for Duchenne muscular dystrophy. We are pleased that biomarker data from all three patients dosed in the 2E14 vg/kg cohort of IGNITE DMD showed SGT-001 microdystrophin protein expression and associated neuronal nitric oxide synthase (nNOS) function, providing further evidence of the therapeutic potential of SGT-001. Our priority is to address the IGNITE DMD clinical hold so we can continue to evaluate the ability of SGT-001 to help patients with Duchenne, said Ilan Ganot, Chief Executive Officer, President and Co-Founder of Solid Biosciences.

Recent Developments

Financial Highlights

Research and development expenses for the fourth quarter of 2019 were $27.1 million, compared to $17.8 million for the fourth quarter of 2018. Research and development expenses for the year ended December 31, 2019 were $94.7 million, compared to $58.0 million for the year ended December 31, 2018. The increase was primarily attributable to compensation and other costs associated with additional headcount, as well as facility costs and increased expenses related to the clinical development and manufacturing activities for SGT-001.

General and administrative expenses for the fourth quarter of 2019 were $5.3 million, compared to $4.6 million for the fourth quarter of 2018. General and administrative expenses for the year ended December 31, 2019 were $24.6 million, compared to $17.7 million for the year ended December 31, 2018. The increase was primarily attributable to increased personnel costs.

Net loss for the fourth quarter of 2019 was $31.9 million, compared to $21.9 million for the fourth quarter of 2018. Net loss for the year ended December 31, 2019 was $117.2 million, compared to $74.8 million for the year ended December 31, 2018.

Solid had $83.5 million in cash, cash equivalents and available-for-sale securities as of December 31, 2019. Solid expects that it has sufficient capital to fund its operations into 2021.

In January 2020, Solid announceda reduction in workforce of approximately one third was implemented as part of a strategic plan designed to create a leaner company focused on advancing SGT-001. In connection with that, Solid curtailed its research and development activities supporting the company's complementary disease modifying and assistive device programs.

About SGT-001

Solids SGT-001 is a novel adeno-associated viral (AAV) vector-mediated gene transfer therapy under investigation for its ability to address the underlying genetic cause of Duchenne muscular dystrophy (Duchenne). Duchenne is caused by mutations in the dystrophin gene that result in the absence or near absence of dystrophin protein. SGT-001 is a systemically administered candidate that delivers a synthetic dystrophin gene, called microdystrophin, to the body. This microdystrophin encodes for a functional protein surrogate that is expressed in muscles and stabilizes essential associated proteins, including neuronal nitric oxide synthase (nNOS). Data from Solids preclinical program suggests that SGT-001 has the potential to slow or stop the progression of Duchenne, regardless of genetic mutation or disease stage.

SGT-001 is based on pioneering research in dystrophin biology by Dr. Jeffrey Chamberlain of the University of Washington and Dr. Dongsheng Duan of the University of Missouri. SGT-001 has been granted Rare Pediatric Disease Designation, or RPDD, in the United States and Orphan Drug Designations in both the United States and European Union.

About Solid Biosciences

Solid Biosciences is a life science company focused solely on finding meaningful therapies for Duchenne muscular dystrophy (Duchenne). Founded by those touched by the disease, Solid is a center of excellence for Duchenne, bringing together experts in science, technology and care to bring forward meaningful therapies that have life-changing potential. For more information, please visit http://www.solidbio.com.

Story continues

Forward-Looking Statements

This press release contains forward-looking statements within the meaning of the Private Securities Litigation Reform Act of 1995, including statements regarding our expectations regarding the IGNITE DMD clinical trial, the safety or potential efficacy of SGT-001, the sufficiency of our cash, cash equivalents and investments to fund our operations and other statements containing the words anticipate, believe, continue, could, estimate, expect, intend, may, plan, potential, predict, project, should, target, would, and similar expressions. Any forward-looking statements are based on managements current expectations of future events and are subject to a number of risks and uncertainties that could cause actual results to differ materially and adversely from those set forth in, or implied by, such forward-looking statements. These risks and uncertainties include, but are not limited to, risks associated with Solids ability to satisfactorily respond to requests from the FDA for further information and data regarding IGNITE DMD; successfully resolve the clinical hold with regard to IGNITE DMD; obtain and maintain necessary approvals from the FDA and other regulatory authorities and investigational review boards at clinical trial sites; enroll patients in its clinical trials; continue to advance SGT-001 in clinical trials; replicate in clinical trials positive results found in preclinical studies and earlier stages of clinical development; advance the development of its product candidates under the timelines it anticipates in current and future clinical trials; successfully scale its manufacturing process; obtain, maintain or protect intellectual property rights related to its product candidates; compete successfully with other companies that are seeking to develop Duchenne treatments and gene therapies; manage expenses; and raise the substantial additional capital needed to achieve its business objectives. For a discussion of other risks and uncertainties, and other important factors, any of which could cause the Companys actual results to differ from those contained in the forward-looking statements, see the Risk Factors section, as well as discussions of potential risks, uncertainties and other important factors, in the Companys most recent filings with the Securities and Exchange Commission. In addition, the forward-looking statements included in this press release represent the Companys views as of the date hereof and should not be relied upon as representing the Companys views as of any date subsequent to the date hereof. The Company anticipates that subsequent events and developments will cause the Company's views to change. However, while the Company may elect to update these forward-looking statements at some point in the future, the Company specifically disclaims any obligation to do so.

Investor Contact:Carlo Tanzi, Ph.D.Kendall Investor Relations617-337-4680investors@solidbio.com

Media Contact:Courtney HeathScient Public Relations617-872-2462media@solidbio.com

Read more here:
Solid Biosciences Reports Fourth Quarter and Full Year 2019 Financial Results and Provides Business Update - Yahoo Finance

Posted in Genetic Therapy | Comments Off on Solid Biosciences Reports Fourth Quarter and Full Year 2019 Financial Results and Provides Business Update – Yahoo Finance

Reviewing Evidence on the Screening, Diagnosis, and Care of Familial Hypercholesterolemia – The Cardiology Advisor

Familialhypercholesterolemia (FH) is one of the most clinically relevant monogenicdisorders contributing to the development of atherosclerotic cardiovasculardisease (ASCVD). The prevalence of FH was estimated to be 1 in 200 to 1 in 250 individualsin studies in which genetic testing was conducted on large community populationsamples.1 However, the disease often remains undetected and thusuntreated, with only 10% of individuals with FH receiving adequate diagnosisand treatment.2

Notingthe recent accumulation of studies on FH, the authors of a Nature ReviewsCardiology article sought tosummarize the key elements of a model of care for the condition that canbe adapted as new evidence emerges.1 Selected points are highlightedbelow.

Screening and detection. A combination of selective, opportunistic (eg, genetic screening of blood donors), systematic, and universal screening approaches is recommended to improve the detection of FH. Universal screening of children and childparent (reverse) cascade testing is potentially a highly effective method for detecting patients with FH at a young age, before they develop ASCVD32 [and] might be particularly relevant to communities with gene founder effects, noted the review authors. All children with FH should ideally be detected from the age of 5 years or earlier if homozygous FH (hoFH) is suspected.

Diagnosis. In the United States, elevated levels of low-density lipoprotein cholesterol (LDL-C) and a family history of FH are the main phenotypic criteria for FH diagnosis in children. Patients with hoFH, heterozygous FH (heFH), and polygenic hypercholesterolemia may also present with overlapping LDL-C levels, posing a challenge for the development of a standardized diagnostic tool for FH.

Genetic testing. Aninternational expert panel recently endorsed genetic testing in the care ofpatients with FH as it would [allow] a definitive diagnosis, improve[e] riskstratification, address the increasing need for more potent therapies, improve[e]adherence to treatments, and increase[e] the precision and cost- effectivenessof cascade testing.1,3 However, genetic testing remains underuseddue to issues such as cost, low access to genetic counseling, and lack ofclinician knowledge in this area.

Clinical risk assessment.Cumulative lifetime exposure to elevated LDL-C is the key factor driving ASCVDrisk in asymptomatic patients with FH, further underscoring the need for timelydiagnosis and risk stratification. In addition to phenotypic and geneticfactors, imaging of subclinical atherosclerosis, might be the most usefulclinical tool for assessing risk in FH.1 For example, imaging ofcoronary artery calcium can be used to predict coronary events in asymptomaticmiddle-aged patients with FH taking statins, and computed tomography coronaryangiography can be used to assess plaque burden and to intensify therapy.

Care of adults.Emerging evidence continues to support aggressive cholesterol-lowering therapyand lifestyle management in patients with FH from as young as 8 years tomaximally mitigate the cumulative cholesterol burden of risk. The review authorsemphasize the importance of patient-centered care and shared decision making,although health literacy is a challenge that may need to be addressed with somepatients.

Whilethere is insufficient evidence to develop strictly defined LDL-C treatmenttargets, current evidence-based recommendations stipulate that in adultpatients with FH, statin therapy and diet should initially be targeted toachieve a 50% reduction in LDL-cholesterol level and an LDL-cholesterol level<1.8 mmol/l (70 mg/dl) or <2.6 mmol/l (100 mg/dl) for primaryprevention, and <1.4 mmol/l (55 mg/dl) or <1.8 mmol/l (70 mg/dl) forsecondary prevention or for patients at very high risk.1

The addition of ezetimibe is indicated in patients who do not achieve the recommended LDL-C levels with statins alone. The use of a proprotein convertase subtilisin/kexin type 9 (PCSK9) inhibitor as a third-line therapy is recommended in those patients or in patients who are intolerant to statins. The addition of a PCSK9 inhibitor in patients with heFH can further reduce LDL-C levels by approximately 60% and lead to recommended treatment targets in more than 80% of patients. However, these agents should not be used during pregnancy, as they cross the placenta and their impact on fetal development has not yet been determined.

Care of children. Extensive evidence supports the treatment of FH starting in childhood, as [m]odest and sustained reductions in LDL- cholesterol levels from early life can have a major effect on reducing mortality associated with ASCVD. Initial therapy is based on lifestyle management in early childhood, with the addition of statins by age 10 years in children with HeFH and upon diagnosis in children with hoFH. Ongoing research is investigating the efficacy and safety of PCSK9 inhibitors in children with heFH or hoFH.4,5

Radical therapies and novel approaches. Lipoprotein apheresis may be required insevere cases of FH, including in pregnant women, and liver transplantationremains the only curative therapy for patients with severe hoFH.

In ongoing studies, an array of novel treatment approaches are being examined, including functional LDL receptor gene transfer therapy in patients with hoFH and targeted RNA-based therapies to lower elevated lipoporotein(a) levels.6-8

Reviewauthors also emphasized the importance of clinical registries, patient supportgroups and networks, and the need for structured research programs that areunderpinned by actionable dissemination and implementation strategies,research skills and training among service providers, and sustainable fundingmodels. They stated that a major challenge is translating new evidence intohealth policy and routine care. Systems approaches for supporting healthorganizations and providers in addressing these gaps in care and serviceprovision are essential.

We spoke with Seth Shay Martin, MD, MHS, associate professor ofmedicine at the Johns Hopkins University School of Medicine in Baltimore,Maryland, and director of the Advanced Lipid Disorders Program of the Ciccarone Center atJohns Hopkins.

Cardiology Advisor: What are examplesof the latest advances in knowledge or practice pertaining to FH?

Dr Martin: A big advance inpractice has been the introduction of PCSK9 inhibitors. When added to statinsand ezetimibe, this class of medications can lower LDL-C by 60% sometimes the reduction can be lower, but inmy experience the effect is commonly approximately 60%. This leads to patientscoming back to clinic really satisfied.

Cardiology Advisor: What is the optimalapproach for the treatment of these patients, and what are some of the toptreatment challenges?

Dr Martin: The optimal approach is to follow the 2018 American Heart Association/American College of Cardiology multi-society guidelines, which recommend a combination approach of lifestyle modification with first-line maximal statin therapy, followed by the addition of ezetimibe and PCSK9 inhibitors. The LDL-C threshold at which additional therapy should be considered is70 mg/dL in high-riskpatients with ASCVD and FH. In patients with isolated FH (termed severe hypercholesterolemia by the guidelines,based on LDL-C levels 190 mg/dL), the LDL-C threshold is 100 mg/dL.

Cardiology Advisor: What are otherrelevant treatment implications for clinicians who treat these patients?

Dr Martin: One of the joys intaking care of a patient with FH is taking care of a family. It is a geneticdisorder with a 50% chance of being passed from parent to child. It is key toperform cascade testing to identify other members of the family; family visitsto the clinic can be beneficial for all.

Cardiology Advisor: What are remaining needs in thisarea?

Dr Martin: There is a great need for increasing awareness and diagnosis rates for FH. This is what our center is working to do as partners of the FH Foundation and as a CASCADE FH Registry site.

References

Read more from the original source:
Reviewing Evidence on the Screening, Diagnosis, and Care of Familial Hypercholesterolemia - The Cardiology Advisor

Posted in Genetic Therapy | Comments Off on Reviewing Evidence on the Screening, Diagnosis, and Care of Familial Hypercholesterolemia – The Cardiology Advisor