header logo image


Page 33«..1020..32333435..4050..»

Archive for the ‘Gene therapy’ Category

FDA Official: New "Playbook" Needed for CMC Reviews of Gene Therapy Products – Xconomy

Friday, June 26th, 2020

XconomyNational

A new playbook is needed to ensure consistent chemistry, manufacturing, and controls (CMC) reviews for gene therapy products, the lack of which is hindering the development of these products, asserted a top official at the US Food and Drug Administration .

Now is the time to get things right asserted Peter Marks, director of the FDAs Center for Biologics Evaluation and Research, who spoke at a 15 June virtual Drug Information Association annual meeting session on how innovation can help overcome hurdles for these products.

The sessions moderator, Nancy Myers, president of Catalyst Healthcare Consultants, asked the panelists to describe some of their main CMC constriction points in developing gene therapy products, and to identify potential solutions. The other panelists were Karen Walker, the senior advisor for cell and gene therapy at Genentech, who formerly was at Novartis (NYSE: NVS) and worked on the development of Kymriah, and Michael Paglia, director of CMC for ElevateBio.

Myers said that there are two common types of roadblocks to getting gene therapy products through the development pipeline, and these are logistical and technical challenges. The logistical challenges are having a well-trained workforce, managing global distribution networks and ensuring products are transported in cold temperatures, while the technical challenges are ensuring the quality of the starting materials and scaling up production from the research site to commercial manufacturing.

Another roadblock is the lack of standards and lack of a regulatory framework for these products. Myers said that this is a new and growing field and companies are trying to lay the track as they are trying to drive the train down the track at the same time.

CONSISTENT CMC PLAYBOOK NEEDED

Myers first asked the panelists to discuss what they see as constriction points in manufacturing gene therapy products. In response, Marks said that a lack of consistent reviews is hindering their development.

It has become apparent over the last couple of months that, while we have excellent reviewers, it does happen that people can have differences of opinion. I think we will have to come around and have a clear playbook so that everyone gets the same advice especially as we have grown. I know that someone out there will say, we had two different CMC reviewers and two differences pieces of advice. I am not going to argue with that. That is an issue here. As we come to the post-COVID period we should to try to have more unity in what comes from our CMC reviews. I cannot say the problem is solved but the problem has been identified and is amenable to solutions.

He further noted that the lack of clear regulatory pathway for these products is a major roadblock in accelerating their development. We do not have the preclinical pathways set up and the clinical set up and the regulatory paradigm is yet to be fleshed out. Now is the ripe time to get things right.

Marks also noted some of the manufacturing challenges in the cell and gene therapy space: We are in a place where our current vectors are limiting what we can address in terms of our ability to product them on a very large scale, and what will probably take some years to get there. On the other hand, the piece that really interests me is how do we deal with hundreds and thousands of rare diseases that we cant address right now through the production of gene therapy products where we simply do not have the manufacturing capacity to be able to produce these products in a rapid manner because we just dont have the systems.

MORE ON WHY DEVICE-LIKE REVIEW COULD HELP

Marks expanded on an idea he had suggested in February, that reviews for gene therapies should be more aligned with the device model. (Also see Individualized Gene Therapy: US FDA Considering Device-Like Manufacturing Approval Process Pink Sheet, 28 Feb, 2020.)

It is becoming increasingly clear that for cell and gene therapies, the manufacturing is more like a device paradigm with continued innovations, he said. With a traditional drug you come up with a chemical process to make a small molecule and you are probably using the process similarly across the lifecycle, but you are not constantly finding ways to do things that fundamentally change the yield or quality of a product. Here we have issues that manufacturing changes can potentially change the product for the better.

He added that we have to find some balance here between the traditional drug manufacturing model of once and done to something that is asking you go through multiple cycles of a device every two to three years where you are changing the technology. With device cycles, you may have multiple generations of the device over years. With a device you can measure things nicely, with biologicals you cannot measure easily.

Walker concurred that these are not well-characterized products and so we need to invest heavily in analytics so that we can gain product and process understanding so that we can facilitate rapid changes that we know will not negatively impact the health of the patients.

KYMRIAH TECHNOLOGY ALREADY OUTDATED

Walker said a constriction point for her is not keeping up to date with current technologies. She said that a technology platform developed today may be outdated tomorrow. The rate of change of innovation is now every two or three years, she said.

That mirrors the rate of the device cycles that Marks mentioned, lending further credence to the idea that cell and gene therapies should be reviewed similarly.

Walker said that the technology that Kymriah has been based on has been eclipsed. It took three years for start up to approval and now no one is using the same technology as the basis for their platform. The technology and the state of the art is advancing very rapidly. This is a challenge for regulators. They need to understand we can be early adopters of these technologies without changing the product.

Kymriah was the first gene therapy product approved to treat B-cell acute lymphoblastic leukemia (ALL) and diffuse B-cell lymphoma (DLBCL). The product used spherical beads to isolate, activate and expand T-cells. After the cells are modified, they are infused back into the patient. The FDA approved the drug in August 2017 (Also see FDAs NDA And BLA Approvals: Kymriah, Vabomere, Cyltezo Pink Sheet, 1 Sep, 2017.) and the EU approved it in June 2018. (Also see First CAR T-Cell Therapies OKd In EU: Novartiss Kymriah And Kites Yescarta Pink Sheet, 29 Jun, 2018.)

Walker said there also needs to be flexibility from regulators to allow new technologies. The technology is advancing very rapidly, and that is another challenge for regulators. To understand where they can have flexibility.

AVAILABLE TALENT POOL A MAJOR CHALLENGE

Michael Paglia, senior VP of CMC operations for ElevateBio, said that his main constriction point has to do with staff and talent and supply chain and the cost of goods and quality and access to capacity.

To address the capacity challenges, the company came up with a model of funding multiple start-ups and to utilize the same R&D and manufacturing facility, rather than individual companies whose cell and gene therapy R&D is slowed by the need to build their own lab and production spaces. (Also see ElevateBio Brings Centralized Model To Cell And Gene Therapy Scrip, 13 May, 2019.)

We took the approach to build our own and to build an integrated research to support out cell and gene therapies.

The company in July 2019 announced a partnership with Massachusetts General Hospital. Under the agreement, which runs for 10 years, MGH has access to ElevateBios research, process development and manufacturing facility in Waltham, MA, for development and production of cell and gene therapies.

Paglia said that there are now more skilled employees compared to seven or 10 year ago, but that it is still challenging to find talent. We are fortunate to have experienced staff. It is necessary to put procedures in place to have rigorous training. Training is very important, and we are involved with local universities as well to give them an idea of if you come out of this how do you get into cell and gene therapy.

This article was first published in the Pink Sheet on June 18, 2020.

Image: iStock/IvelinRadkov

Joanne Serpick Eglovitch is a senior editor for Pink Sheet where she writes about manufacturing and quality issues.

See the original post:
FDA Official: New "Playbook" Needed for CMC Reviews of Gene Therapy Products - Xconomy

Read More...

Gene Therapy Market: Predictable To Witness Sustainable Evolution over 2020-2030 – 3rd Watch News

Friday, June 26th, 2020

The Gene Therapy Market Research Report 2020 published by Prophecy Market Insights is an all-inclusive business research study on the current state of the industry which analyzes innovative strategies for business growth and describes significant factors such as top developers/manufacturers, production value, key regions, and growth rate. Impact of Covid-19 pandemic on the market will be completely analyzed in this report and it will also quantify the impact of this pandemic on the market.

The research study encompasses an evaluation of the market, including growth rate, current scenario, and volume inflation prospects, based on DROT and Porters Five Forces analyses. The market study pitches light on the various factors that are projected to impact the overall market dynamics of the Gene Therapy market over the forecast period (2019-2029).

Regional Overview:

The survey report includes a vast investigation of the geographical scene of the Gene Therapy market, which is manifestly arranged into the localities. The report provides an analysis of regional market players operating in the specific market and outcomes related to the target market for more than 20 countries.

Australia, New Zealand, Rest of Asia-Pacific

The facts and data are represented in the Gene Therapy report using graphs, pie charts, tables, figures and graphical representations helping analyze worldwide key trends & statistics on the state of the industry and is a valuable source of guidance and direction for companies and individuals interested in the market.

Get Sample Copy of This Report @ https://www.prophecymarketinsights.com/market_insight/Insight/request-sample/39

The research report also focuses on global major leading industry players of Gene Therapy market report providing information such as company profiles, product picture and specification, R&D developments, distribution & production capacity, distribution channels, price, cost, revenue and contact information. The research report examines, legal policies, and competitive analysis between the leading and emerging and upcoming market trends.

Gene TherapyMarket Key Companies:

GlaxoSmithKline plc, Bluebird Bio, Inc., Adaptimmune Therapeutics plc, Celgene Corporation, Shanghai Sunway Biotech Co. Ltd., Merck KGaA, Transgene SA, and OncoGenex Pharmaceuticals, Inc.

The predictions mentioned in the Gene Therapy market report have been derived using proven research techniques, assumptions and methodologies. This market report states the overview, historical data along with size, share, growth, demand, and revenue of the global industry.

Segmentation Overview:

The report provides an in-depth analysis of the Gene Therapy market segments and highlights the latest trending segment and major innovations in the market. In addition to this, it states the impact of these segments on the growth of the market. Apart from key players analysis provoking business-related decisions that are usually backed by prevalent market conditions, we also do substantial analysis of market based on COVID-19 impact, detailed analysis on economic, health and financial structure.

Request Discount @ https://www.prophecymarketinsights.com/market_insight/Insight/request-discount/39

Key Questions Answered in Report:

Stakeholders Benefit:

Get In-depth TOC @ https://www.prophecymarketinsights.com/market_insight/Global-Gene-Therapy-Market-By-39

About us:

Prophecy Market Insights is specialized market research, analytics, marketing/business strategy, and solutions that offers strategic and tactical support to clients for making well-informed business decisions and to identify and achieve high-value opportunities in the target business area. We also help our clients to address business challenges and provide the best possible solutions to overcome them and transform their business.

Contact Us:

Mr Alex (Sales Manager)

Prophecy Market Insights

Phone: +1 860 531 2701

Email: [emailprotected]

VISIT MY BLOG:- https://medium.com/@varshashirode0000/global-blockchain-in-genomics-market-trends-analysis-and-forecast-till-2029-3f92b8bdc622

Continue reading here:
Gene Therapy Market: Predictable To Witness Sustainable Evolution over 2020-2030 - 3rd Watch News

Read More...

Akouos raises $213m in its IPO, gains nearly 30 percent on first day of trading – BetaBoston

Friday, June 26th, 2020

A Boston biotech trying to develop the first gene therapy to treat hearing loss raised $213 million Friday as it made its stock market debut, 70 percent more than the firm had projected four days ago.

Akouos sold 12.5 million shares at $17, above the original range of $14 to $16, and in line with the upsized share offering and price it filed Thursday morning. On Monday, the company said in a filing with the Securities and Exchange Commission that it hoped to raise $125 million in the initial public offering.

Shares in the company, which is listed on the Nasdaq under the symbol AKUS, closed Friday at $22, up more than 29 percent.

Akouos is the latest biotech to see higher than expected demand in the public markets despite the pandemic-related recession: the 2020 biotech IPO class is averaging a return of 80 percent, according to Renaissance Capital, a pre-IPO research provider for institutional investors.

Some analysts say COVID-19 has underscored the promise of biotechnology to address deadly health threats, generating investor enthusiasm. Among the biotechs whose market values have soared during the epidemic is Moderna, a Cambridge drug company that was the first to get an experimental coronavirus vaccine into human trials. Moderna, which went public in 2018, has a market value of more than $22 billion, though it has no approved products.

Akouos, founded in 2016, is trying to develop the first gene therapy to treat hearing loss in particular, a form of deafness caused by mutations in a single gene. Gene-based hearing loss afflicts 300,000 people in the United States each year, including more than 4,000 newborns.

Its lead candidate is a treatment for a type of genetic hearing loss that afflicts about 7,000 people. The company hopes to use a small virus called adeno-associated virus, or AAV, as a vector to deliver DNA that encodes a functioning gene in target cells. These viruses dont typically cause disease and can be customized to treat different inherited conditions.

Akouos has partnerships with Massachusetts Eye and Ear and Lonza, a Swiss multinational manufacturer that has contracts with drugmakers.

Jonathan Saltzman can be reached at jonathan.saltzman@globe.com

The rest is here:
Akouos raises $213m in its IPO, gains nearly 30 percent on first day of trading - BetaBoston

Read More...

Cell Therapy For Solid Tumors – Contract Pharma

Friday, June 26th, 2020

Base editing, a new player in the gene editing arena, could have an important role in the development of immune-based cell therapies to treat solid tumors. Using cell therapies, such as CAR-T cells, in solid tumors remains challenging: the current word on the street is that such chimeric antigen receptors (CARs) will need multiple gene modifications to make them efficient and it is in this space that base editing could have a substantial advantage.Immune-cell-based therapy is an exciting cell therapy approach to treat cancer where the natural defenses of a patients immune system are used to target and kill cancer cells. Hopes were high following the initial FDA approvals of the first autologous CAR-T therapies for Novartis KYMRIAH (Aug 2017) and Gilead/Kite Pharmas YESCARTA (Oct 2017), both for blood-based cancers, but translating these successes into solid tumors remains a challenge. This is a consequence of the complexity and heterogeneity of solid tumors together with the immune inhibitory nature of the tumor microenvironment.For T cell-based therapies to work, the patient is treated with modified T cells that are rendered capable of identifying and killing tumor cells and, through this, generating a wider immune response against the tumor. Two key approaches used to modify T cells are through expressing a T cell receptor (TCR) known to target the tumor cell or a CAR. Other approaches include using and/or modifying natural killer cells, gamma delta cells or tumor infiltrating lymphocytes. It is not clear which approach will provide the most effective treatment option and in fact it might be that each tumor type responds better to one approach or to a combination of approaches. Irrespective of the approach, it is clear that the current therapies all face similar challenges; the risk of graft vs host disease (GvHD), a lack of durable remissions, on-target or off-target toxicity and cytokine release syndrome.Another layer of complexity for T cell-based therapies lies with the source of T cellsusing the patients own (autologous T cells), or a donor or iPSC-derived T cells (allogeneic). Each approach has advantages and challenges. Briefly, autologous treatments are attractive because they mitigate the risk of immune rejection and GvHD when infused back into the patient. However, they require a complex manufacturing process that necessitates specialist equipment local to the patient to enable the isolation of their T cells followed by rapid manufacture to transform them into engineered T cells ready to infuse back into the patient. At all points during this manufacturing pipeline, the product must be kept sterile and tracible to ensure the correct cells are transfused back into the correct patient. The allogeneic T cells approach is appealing because of the possibility that these could be engineered to be universal donor cells (suitable for all or most patients). Such cells can be manufactured in bulk and administered to multiple patients all over the world. This bulk manufacturing would attract cost-savings once a critical mass of therapy is reached. It could also allow for engineered T cells to be available on-tap to any patient, a game-changer for particular patients whose own T cell count is either too low for engineering, or that transduce poorly with the engineered construct during manufacture. The challenge for allogeneic T cell therapies is the ability to generate cloaked T cells that do not provoke an immune-response in the patient, as this could kill the engineered T cells after transplantation such that they have no efficacy or potentially lead to the death of the patient as a result of a disseminated cytokine storm. Despite these challenges, in April 2019 the US Food and Drug Administration (FDA) approved the first allogeneic CAR-T for investigational use in patients with multiple myeloma and more are expected to follow.To try and achieve a stealth allogeneic T cell that flies under the radar of the patients immune system, genetic engineering is key. Although T cells can be engineered to express a specific TCR or CAR, additional genetic changes are required to provide a cloak of invisibility, prolong the survival of the cells in the patient and enable them to function in an immune suppressive tumor microenvironment. From a simplistic point of view, one could view the modified CAR or TCR as a sat nav, with the T cell being the engine. You need both to get to your destination and, if youre able to improve your engine, its possible the outcome could be achieved faster and in a more reliable fashion. There are several gene knockouts or gene knockdowns that are seen as a natural first step to improving the properties of engineered allogeneic T cells and these are summarized in the table below:As there are multiple gene knockout options, a gene engineering technology capable of making multiple gene edits with as few off-target effects as possible is needed. Indeed, it is conceivable that an effective allogeneic T cell-based therapy might require ten or more gene edits.This prompts the question What is the best gene editing platform or technology to support multiple gene edits? On the surface of it, and owing to its phenomenal adoption in research labs worldwide over the past decade, most currently use the gene editing approach provided by CRISPR-Cas9. CRISPR was first commercialized in 2012 and quite staggeringly made its debut in a clinical trial for cell therapy in June 2016. Although CRISPR is an efficient gene editing tool, its mode of action of generating double-strand breaks in the DNA could be a source of concern. DNA double-strand breaks, which tend to be repaired by the cells repair machinery in an error prone fashion, can cause unintended changes in the genome of engineered cells. Although there are methods for minimizing these off-target effects when single genes are targeted, targeting multiple genes in one cell all at the same time could lead to genome-altering insertions, deletions and/or chromosomal translocations. The impact of this on a patient could be that the cell therapy is effective but the off-target genetic changes lead to deleterious side-effects, impacting patient recovery and potentially survival. Well-known alternatives to CRISPR include transcription activator-like effector nucleases (TALENS) and zinc finger nucleases (ZFN). These approaches have slightly different safety profiles to CRISPR and while optimized for single gene edits or knockouts, multiple gene knockouts still present a challenge for these technologies. Freedom to operate using these technologies in the therapeutics space can also be challenging, particularly for start-up and biotech companies.The potential deleterious impact of off-target effects, particularly for multiple gene edits has opened the door to a newcomer on the gene engineering scene: base editing. This technology first gained prominence from peer-reviewed papers published by researchers from Harvard University.1 Others, such as Rutgers University, have also developed base editing platforms.2 In brief, base editing uses a deaminase enzyme to make a specific base pair change in the DNA. The base pair alteration can either be an A to G or a C to T depending on which deaminase is used. Importantly, the CRISPR-Cas system is used to guide the deaminase to the base pair that is going to be altered, but in this version of CRISPR-Cas, a DNA double-strand break is not made, meaning that the off-target effects with base editing in terms of insertions, deletions or translocations should be substantially reduced.On the surface, this crucial characteristic makes base editing an excellent choice of gene editor for cell therapyit can be used to specifically knock-out multiple genes through the introduction of stop codons or splice site disruptions with limited capacity to introduce substantial, large-scale chromosomal abnormalities. However, as base editing was first published in May 2016,1 substantial research is required to understand fully the utility of base editing in the therapeutic space and to appreciate its advantages and challenges compared with standard gene editing approaches, such as CRISPR-Cas, TALENs and ZFNs.As is somewhat expected of a fashionable area for scientific research, the gene editing space does not stand-still for very long: Prime editing has followed hot on the heels of base editing. Unlike base editing, which makes changes to specific base pairs in the DNA, prime editing allows changes to be made to a run of base pairs by forcing the cell to use a DNA copying system that exists naturally in cells as part of the DNA repair mechanism. Initial data suggest3 that prime editing has higher off-target effects compared with base editing, in terms of introducing insertions and deletions, and more work is needed to understand the comparison and utility of base editing vs. prime editing. It will be interesting to see how prime editing evolves over the months and years within the cell and gene therapy space.Although cell therapy has demonstrated its potential for driving complete remissions in some patients with hematological cancers, the next big step is to translate these early successes into patients with solid tumors. However, due to the complexity of solid tumors, this is not a simple or straight-forward process and multiple factors need to be considered. While the sat nav in the form of an engineered TCR or CAR is crucial, the T cell engine could be the natural starting point for improving efficacy in patients with solid tumors, as could the use of allogeneic rather than autologous approaches. The multiple edits that will be needed to deliver a stealth, engineered, allogeneic off-the-shelf T cell are only now being investigated in earnest and it could be that the new kid on the block, base editing, provides a compelling route forward.References1. Komor, A.C., Kim, Y.B., Packer, M.S., Zuris, J.A. and Liu, D.R. (2016). Programmable editing of a target base in genomic DNA without double-stranded DNA cleavage. Nature, 533(7603), 420424. Doi:10.1038/nature179462. Horizon Discovery to provide access to novel base editing technology, January 2020;https://horizondiscovery.com/en/news/2020/Horizon-Discovery-to-provide-access-to-novel-base-editing-technology (accessed May 2020)3. Anzalone, A.V., Randolph, P.B., Davis, J.R., et al. (2019). Search-and-replace genome editing without double-strand breaks or donor DNA. Nature, 576(7785), 149157. doi:10.1038/s41586-019-1711-4Dr. Jonathan Frampton is a business development professional who has been working for Horizon Discovery for the past 9 years and currently as their Corporate Development Partner. He is always scouting for exciting novel technology that could complement Horizons already extensive gene engineering toolkit. In addition to this he works closely with Horizons partners to manage out-licensing opportunities.

See the rest here:
Cell Therapy For Solid Tumors - Contract Pharma

Read More...

Sarepta Addressing Gene Therapy Issue With Two Acquisitions – GuruFocus.com

Friday, June 26th, 2020

On the heels of research deals with two small biotechs, shares of Sarepta Therapeutics Inc. (NASDAQ:SRPT) hit their all-time high of more than $172 on June 23.

The share price of the Cambridge, Massachusetts-based biopharmaceutical company has doubled since mid-March. It has a market cap of more than $13 billion.

Sarepta has made a host of deals in the past several years that have made gene therapy a key part of its business, which had been dominated by drugs for muscular dystrophy. One of those pacts, with Roche (RHHBY), could be worth more than $3 billion.

Sarepta currently has a half dozen gene therapies in clinical trials, with another six waiting in the wings, according to BioPharma Dive. One of the companys compounds has shown promise in treating a type of muscular dystrophy, a genetic disease that causes weakness and wasting of the muscles in the arms and legs.

A major challenge of gene therapy is immune system reactions. Sarepta, like other developers, uses a type of virus in its therapy that, while effective, may not be able to given more than once because patients can create antibodies to it.

Thats a problem Sarepta is trying to address in its latest deals with privately held Codiak Biosciences and Selecta Biosciences Inc. (NASDAQ:SELB). In both cases, Sarepta has an option to license the biotechs' technology to develop and commercialize its therapies.

The Selecta deal focuses on gene therapies for Duchene muscular dystrophy and certain limb-girdle muscular dystrophies.

A Zion Market Research report said the global demand for the Duchenne muscular mystrophy therapeutics market was valued at approximately $2.4 billion in 2018 and is expected to grow to more than $20 billion by the end of 2025, a compound annual growth rate of more than 36% between 2019 and 2025.

Given the size of the opportunity, its no surprise Sarepta has plenty of competitors vying for a share of the business, including Pfizer Inc. (NYSE:PFE), PTC Therapeutics (NASDAQ:PTCT), FirbroGen Inc. (NASDAQ:FGEN), Roche and Bristol-Myers Squibb Co. (NYSE:BMY).

The two-year deal with Codiak gives Sarepta the right to license its technology for up to five neuromuscular diseases. If Sarepta exercises an option, Codiak will then handle research until right before the candidate goes into human testing. Sarepta will then be responsible for clinical development and commercializing the drug.

In April, the company announced it has resurrected its antiviral program in response to Covid-19. It has a therapy that is meant to block the coronavirus ability to replicate. The treatment will be tested at the U.S. Army Medical Research Institute of Infectious Diseases.

If it works, it will reduce the ability of the virus to replicate and its ability to infect other cells, Sarepta CEO Doug Ingram told Forbes. He cautioned that the drug is at an early stage.

According to CNN Money, the 21 analysts offering 12-month price forecasts for Sarepta have a median target of $192, with a high estimate of $260 and a low $152. The stock is rated a buy.

Disclosure: The author hold positions in Pfizer and Bristol-Myers Squibb.

Read more here:

Not a Premium Member of GuruFocus? Sign up for a free 7-day trial here.

Barry Cohen

He has contributed to a number of financial websites, writing primarily about the stocks of health care companies.

Read more here:
Sarepta Addressing Gene Therapy Issue With Two Acquisitions - GuruFocus.com

Read More...

Out of jobs, a pair of early cell therapy executives went to Seoul, came back with a new company, $70M and a plan to leapfrog natural killer…

Friday, June 26th, 2020

Tom Farrell didnt have much to do after Bellicum announced in January 2017 that they were bringing in a new CEO. He had led the CAR-T company for over a decade, since before Carl Junes New England Journal of Medicinepaper had made cell therapy the hottest thing in cancer research. Now he was facing an 18-month non-compete.

So he worked quickly when, not long after that clock expired in 2018, a banker who helped take Bellicum public told him about a South Korean company called Green Cross LabCell that had built a natural killer cell factory and was looking to develop therapies off it. Farrell hopped a plane to Seoul.

It was hugely impressive, Farrell told Endpoints News.There was nothing [else] I came across that was truly disruptive from a business model perspective.

A year and a half later, Farrell has his new company. Called Artiva, it launches with $78 million in Series A funding and an exclusive deal with Green Cross to push some of their natural killer cell technology into the clinic. Theyll start with a therapy that combines NKs with an approved antibody therapy like rituximab to improve the antibodys effectiveness. Behind that, theyre working on CAR-NK therapy and, longer term, gene-edited CAR-NK cells. RA Capital Management, venBio and 5AM Ventures led the round.

Artiva joins what, after many years, has recently become a booming field. In February, MD Anderson showed that a Takeda-licensed CAR-NK therapy cleared tumors completely in 7 of 11 non-Hodgkins lymphoma patients. Two months later, J&J gave Fate Therapeutics, one of the earliest biotechs in the field, an up-to $3.1 billion deal for their CAR-NK and CAR-T therapies. The Big Pharmas are joined by a slate of recent upstarts, including Celularity, Nkarta, NantKwest, and Cytovia.

Unlike the other newcomers, Artiva makes virtually no claim on having original science. In fact, Farrell said, biotechs emphasis on novel technologies is part of why cell therapy has advanced only incrementally since the approval of the first two CAR-T therapies. Industry hasnt focused enough on addressing the manufacturing issues that have made therapies so costly and difficult to scale, he said.

Lewis Lanier, an immunologist at the University of California, San Francisco and an early pioneer in NK cell research, said Artiva would still face the same questions other drug developers face will some patient reject the cells? Will the natural killer cells actually last a significant amount of time after infusion? but the collaboration could give them an edge.

The Korean Green Cross manufacturing facility is really first rate, thats where the advantage is, Lanier, who is not involved in Artiva, told Endpoints. The science is really routine, theyre not doing anything particularly innovative.

For years, NK cells have been viewed as one of the key potential ways of making off-the-shelf cell therapy. Part of the innate immune system, implanting these cells from donors doesnt lead to the same resistance that donor T cells can. One of the problems, though, is that NKs are finicky, as Lanier puts it, vastly more difficult to grow and manipulate in a lab. Only recently have a couple companies figured out ways to do it consistently. Fate, for instance, uses master lines of iPSC stem cells.

At the Green Cross facility Farrell toured two Novembers ago, the South Korean company had refined a process to derive NK cells from donated umbilical cord blood and cryo-preserve it. A week after his tour, Farrell flew to San Diego for the ASH conference, where he ran into Pete Flynn, another longtime biotech executive out of a job. Flynn had run early development for Fate in its early years before leaving to run R&D for the anti-obesity company Orexigen, which had just gone bankrupt.

Farrell explained what he saw in Seoul and the two debated different approaches to off-the-shelf therapy. They figured the manufacturing base could be a launching pad.

Even though were a Series A company, were looking to become the go-to NK cell, Flynn, now COO, told Endpoints. Basically all the pieces are in place already, whereas for some of those other companies, there might still be some work to do.

Read more here:
Out of jobs, a pair of early cell therapy executives went to Seoul, came back with a new company, $70M and a plan to leapfrog natural killer...

Read More...

Hemophilia Gene Therapy Market Overview on Demanding Applications 2028 – The Cloud Tribune

Friday, June 26th, 2020

Global Hemophilia Gene Therapy market report from Fact.MRs viewpoint

Fact.MR analyzes the Hemophilia Gene Therapy market from a global as well as local perspective in its recent business intelligence study. The Hemophilia Gene Therapy market reached ~US$ xx Mn/Bn in2019, up by xx% from2018. Further, the report suggests that the Hemophilia Gene Therapy market is anticipated to reach ~US$ xx Mn/Bn in2029with a CAGR of xx% over the forecast period2019-2029.

Queries addressed in the Hemophilia Gene Therapy market report:

Request Sample Report @https://www.factmr.co/connectus/sample?flag=S&rep_id=3107

Competitive landscape

Request Methodology On This Report @https://www.factmr.co/connectus/sample?flag=RM&rep_id=3107

The Hemophilia Gene Therapy market report further scrutinizes the regional analysis into important countries alongwith the market share as well as adoption pattern in each country. Key countries include, country 1, country 2, and country 3, among others.

Key findings of the Hemophilia Gene Therapy market study:

The Hemophilia Gene Therapy report considers2018as the base year and20192029as the forecast period to demonstrate the overall market growth.

Ask analyst about this report athttps://www.factmr.com/report/3107/hemophilia-gene-therapy-market

Why choose Fact.MR?

About Fact.MR

Fact.MR is a global market intelligence company providing business information reports and services. The companys exclusive blend of quantitative forecasting and trend analysis provides forward-looking insight for thousands of decision makers. Fact.MRs experienced team of analysts, researchers, and consultants use proprietary data sources and various tools and techniques to gather and analyze information.

Continued here:
Hemophilia Gene Therapy Market Overview on Demanding Applications 2028 - The Cloud Tribune

Read More...

LYSOGENE Releases the Results From June 26, 2020 Ordinary Annual and Extraordinary General Meeting – Business Wire

Friday, June 26th, 2020

PARIS--(BUSINESS WIRE)--Regulatory News:

Lysogene (FR0013233475-LYS) (Paris:LYS) held its ordinary annual and extraordinary general meeting of shareholders on June 26, 2020, in closed session, which was chaired by Karen Aiach, Chairman of the Board of directors and chief executive officer, without the physical presence of the shareholders.

With a quorum of 53.64%, the shareholders have adopted all the resolutions recommended by the Board of Directors, including the financial statements for the 2019 financial year, the compensation policy applicable to the Chairman and Chief Executive Officer and the directors, as well as delegations granted to the Board of Directors related to financial transactions.

Shareholders also approved the renewal of Karen Aiach, Philippe Goupit, Peter Lichtlen, David Schilansky, Mathieu Simon and Carole Deffez as Board members.

Details on the vote results will be available on the companys website.

About LysogeneLysogene is a gene therapy company focused on the treatment of orphan diseases of the central nervous system (CNS). The company has built a unique capability to enable a safe and effective delivery of gene therapies to the CNS to treat lysosomal diseases and other genetic disorders of the CNS. A phase 2/3 clinical trial in MPS IIIA in partnership with Sarepta Therapeutics, Inc. is ongoing and a phase 1/3 clinical trial in GM1 gangliosidosis is in preparation. In accordance with the agreements signed between Lysogene and Sarepta Therapeutics, Inc., Sarepta Therapeutics, Inc. will hold exclusive commercial rights to LYS-SAF302 in the United States and markets outside Europe; and Lysogene will maintain commercial exclusivity of LYS-SAF302 in Europe. Lysogene is also collaborating with an academic partner to define the strategy of development for the treatment of Fragile X syndrome, a genetic disease related to autism. http://www.lysogene.com.

Original post:
LYSOGENE Releases the Results From June 26, 2020 Ordinary Annual and Extraordinary General Meeting - Business Wire

Read More...

Presented with upbeat Alzheimer’s agitation data, FDA sees another ‘breakthrough’ in Axsome’s AXS-05 – Endpoints News

Friday, June 26th, 2020

Axsome Therapeutics surprise win in a late-stage Alzheimers study, unveiled just two months ago, has registered with regulators. The New York-based biotech has notched a breakthrough therapy designation for AXS-05 for the indication, its second after major depressive disorder.

So whats the big deal here? The drug is an oral agent with multimodal activity consisting of two components: dextromethorphan, an NMDA receptor antagonist, and bupropion whose main purpose is to slow down the metabolism of the former.

In the pivotal Phase II/III ADVANCE-1 study, patients treated with AXS-05 saw their Cohen-Mansfield Agitation Inventory (CMAI) total score decrease in 5 weeks from baseline by 15.4 points on average, compared to 11.5 points for placebo (p=0.010). It was also superior to bupropion alone (p<0.001), proving the necessity of a combo.

The CMAI score measures some of the most visible behaviors as reported by caregivers of Alzheimers patients, including episodes of screaming or hitting.

This FDA Breakthrough Therapy designation is an important milestone in the development of AXS-05 for Alzheimers disease agitation, a serious, prevalent, and debilitating condition for which there is currently no approved therapy, CEO Herriot Tabuteau said in a statement.

Analysts tracking Axsome hadnt been paying much attention to Alzheimers agitation, focusing instead on MDD, where the company is lining up a near-term application with the FDA after reporting upbeat data in one of the toughest fields in R&D.

But after consulting with physicians, Cowen analyst Joseph Thome recently noted that the disease has historically been difficult to treat, and that the results were impressive especially given the placebo group actually performed better than expected.

We expect that AXS-05 will be successfully developed for the indication following another Ph. III study and model $750MM in peak U.S. sales, he wrote, adding to the $2 billion opportunity with MDD.

The fact that no other drug has ever been approved for the specific use, though, can be a double-edged sword. Otsukas Avanir has previously scored a Phase III win for AVP-786 deudextromethorphan hydrobromide [d6-DM]/quinidine sulfate) only to be disappointed in the second.

Axsome seems to have the FDA on its side for now. And that has investors stoked, sending shares up 11.33% to $85.29.

See the original post here:
Presented with upbeat Alzheimer's agitation data, FDA sees another 'breakthrough' in Axsome's AXS-05 - Endpoints News

Read More...

One-and-done gene therapy wipes out Parkinson’s symptoms in mouse models – FierceBiotech

Thursday, June 25th, 2020

A researcher at the University of San Diego, California (USCD) made an accidental discovery several years back after he silenced a gene called PTB in mouse fibroblasts, cells in connective tissue. Within weeks, almost all of the fibroblasts were gone, and the rest had transformed into neurons.

Now, researchers in the same lab are applying that discovery to Parkinsons disease in the hopes of creating a one-time gene therapy to replace the dopamine-producing neurons that are lost to the disease.

The UCSD team developed a gene therapy technique that cripplesthe ability of the PTB gene to produce a functioning protein. In mouse models of Parkinsons, the gene therapy turned supportive cells called astrocytes into dopamine-producing neurons, erasing symptoms of the disease, the researchers reported in the journal Nature.

White Paper: Keep Your GI Trials Moving During COVID-19

Clinical Inks intimate knowledge of and experience with GI trials enables a better deployment experience and improved trial conduct. Learn how our GI-specific data capture solutions can support virtual and hybrid trials during COVID-19.

The gene therapy created by the UCSD team consists of a viral vector that carries a piece of DNA called an antisense oligonucleotide into cells. The DNA binds to the RNA that codes for PTB, shutting off the production of the PTB protein and allowing neurons to form.

"Researchers around the world have tried many ways to generate neurons in the lab, using stem cells and other means, said Xiang-Dong Fu, Ph.D., professor of cellular and molecular medicine at UCSD School of Medicine, in a statement. "The fact that we could produce so many neurons in such a relatively easy way came as a big surprise."

RELATED: Mount Sinai researchers uncover new genetic drivers of Parkinson's disease

The UCSD team delivered the gene therapy to the midbrain of the mice and compared them with animals that got a sham procedure. The population of neurons in the brains of mice that received the treatment jumped 30%, and their dopamine levels returned to normal. The control mice saw no improvements, the researchers reported.

Within three months of the gene therapy, the treated mice were able to move their limbs normally, and they remained free of Parkinsons symptoms for the rest of their lives, the team added.

Antisense oligonucleotides are already being used in neurodegenerative diseases. Biogens Spinraza to treat spinal muscular atrophy is an antisense oligonucleotide drug, as are many experimental medicines. Earlier this month, Eli Lilly formed a $20 million deal with Evox Therapeutics, which is using the approach to develop treatments for Duchenne muscular dystrophy and several neurological disorders.

Meanwhile, some research groups are exploring other gene-based approaches to Parkinsons. Last year, a team at Mount Sinai knocked out the gene STMN2 in mice and discovered that nine other genes related to Parkinsons were activateda finding that could lead to new treatment pathways. Startup Prevail Therapeutics is developing a gene therapy to treat Parkinsons thats caused by mutations in the GBA1 gene.

The next step for the UCSD team is to optimize the gene therapy and test it in mouse models of genetic Parkinsons. Theyve patented the technique with plans tofurther optimizeit for testing in people.

See more here:
One-and-done gene therapy wipes out Parkinson's symptoms in mouse models - FierceBiotech

Read More...

With cell and gene therapy boom coming, experts at Novartis, Kite trumpet need for capacity: panel – FiercePharma

Thursday, June 25th, 2020

Cell and gene therapy has evolved into one of the biopharma industry's hottest markets with a major splash of investment and a run of approvals likely in the coming years. To meet what's likely to be massive demand for manufacturing capacity, industry experts are calling for "forward-looking" investmentsbut as one pointed out, those checks aren't easy to write.

During a virtual roundtable Monday hosted by Fierce Pharma, manufacturing experts from Novartis, Gilead's Kite unit, BioMarin and Astellas' Audentes said building capacity and figuring out how to scale production would make the difference in whether the industry will be ready to handle a slate of expected approvals.

According to former FDA Commissioner Scott Gottlieb, the agency could approve between 10 and 20 cell and gene therapies a year through 2025 with as many as 800 such therapies moving through drugmakers' pipelines.

State-of-the-art is not just a state of mind. Its how we operate. AMRI has the next generation in sterile pre-filled syringe technology.

Follow our multi-part blog series on PFS technology to learn more!

Chuck Calderaro, Kite's global head of technical operations, said Kite's primary concern was how to scale production of its cell therapy offerings, including Yescarta, the company's only approved therapy in the space.

VIRTUAL ROUNDTABLE: Up to the Challenge: Manufacturings Central Role in the Cell- and Gene-Therapy Revolution (reg. req'd)

Calderaro noted that Kite has a turnaround time of 16 days from patients having their blood drawn to infusionand the drugmaker is hoping to keep that figure stable as its manufacturing chain grows.

"Access to capacity is always a challenge in a growing area, and especially in cell therapy, which is personalized to order," Calderaro said. "The challenge for us is to be able to scale that excellence as we begin to globalize our cell therapy treatments."

The first challenge for Gilead will come from bringing its newest facility in Amsterdam online after the EU gave it the go-ahead earlier this month.

RELATED: Gilead sees better days ahead for CAR-T therapy Yescarta with Amsterdam manufacturing hub online

Gilead's 117,000-square-foot CAR-T facility at SEGRO Park Amsterdam Airport won a green light after the European Medicines Agency (EMA) approved the plant's end-to-end manufacturing process. The site will house European production fo Yescarta, which won an EMA approval back in August 2018 to treat relapsed or refractory diffuse large B-cell lymphoma and primary mediastinal large B-cell lymphoma.

Calderaro highlighted the Amsterdam facility as the "next step" in Gilead's global manufacturing ramp-up for Yescarta as the site will be able to churn out enough of the pricey therapy for 4,000 patients each year.

Gilead also plans to build a 67,000-square-foot facility at its Oceanside, California, biologics site just for developing viral vectors, the tools needed to deliver genetic material into cells. Calderaro said that facility would give Gilead "a little more control" of its supply chain as it looks to scale up.

RELATED: Pharma's gene and cell therapy ambitions will kick into high gear in 2020despite some major hurdles

Meanwhile, Steffan Lang, Novartis' head of technical operations, pointed out that building capacity for the future will go hand-in-hand with building an experienced team to lead into the future.

"Its about the people and capabilities," Lang said. "You have to have the right team in place to build capacity at the appropriate scale across the globe."

Novartis' gene therapy Kymriah was the first FDA-approved therapy of its kind back in August 2017. Since then, Novartis has expanded Kymriah's reach globally, including opening a new facility in Stein, Switzerland, that cleared up a transatlantic bottleneck for shipments to Novartis' facility in Morris Plains, New Jersey.

Meanwhile, BioMarin and Audentes are both pursuing first FDA approvals for their cell and gene therapy candidates and are looking to scale up manufacturing to make the leap into commercial.

RELATED: Audentes investing $109M in gene therapy manufacturing facility with 200 jobs

In February, Audentes announced it would invest $109 million into a 135,000-square-foot facility in Sanford, North Carolina to flesh out its cell and gene therapy manufacturing needs. The first phase is slated to take 18 months to build and will be operational by 2021. The rest of the investment will play out over two more years.

Donald Wuchterl, Audentes' SVP of technical operations, said building capacity would require a "forward-looking" approach to investingbut the lengthy timelines for these therapies make that foresight difficult.

"These are tough checks to write," Wuchterl said. "Were looking at potentially three years out in a field thats growing rapidlyit takes some constitution, I would say."

BioMarin, which is in the homestretch for an FDA approval for its hemophilia A gene therapy candidate valoctocogene roxaparvovec, or valrox, is in a similar boat looking to make the leap to commercial. For the Robert Baffi, special advisor to the company's CEO, scaling up manufacturing is a big taskbut he hopes that a "biology revolution" could provide a big breakthrough for production in the coming years.

"While I think there's improvements to be had on the manufacturing side today, I think there's a biology revolution still to come in terms of making the vectors more specific, more targeted, more preciseand that would be a big boon for the industry," he said.

More here:
With cell and gene therapy boom coming, experts at Novartis, Kite trumpet need for capacity: panel - FiercePharma

Read More...

Emergent in $75m expansion into viral vectors and gene therapy – BioPharma-Reporter.com

Thursday, June 25th, 2020

Emergent BioSolutions, a contract development and manufacturing organization (CDMO) currently focused on biologics, plans to widen the range of its offerings by adding viral vector and gene therapy services.

According to the company, this will involve a molecule-to-market service, once it has completed the $75m (67m) investment into its Canton, Massachusetts site.

The capital will be put towards buying a property adjacent to its existing live viral drug substance facility, with the expanded area being a multi-suite operation holding capacity up to the 1,000L scale.

Once the work is completed, the CDMO expects to be able to offer advanced therapy drug substance manufacturing services by the beginning of 2023.

Complementing the Canton site are Emergents additional locations in the US, as the company offers development services out of its Gaithersburg location and drug product manufacturing from Rockville, with the latter site also being expanded and with a completion date set for the end of 2021.

Alongside a growing manufacturing footprint, the company has found its services in demand during the novel coronavirus outbreak.

The US government provided the company with a $628m contract to provide CDMO services for COVID-19 programs, with Emergent set to use part of this funding towards expanding drug product fill/finish capacity.

Prior to this, Emergent had already sealed a manufacturing partnership with Johnson & Johnson to develop its lead COVID-19 vaccine candidate as the latter company worked towards securing a supply of one billion doses for its potential vaccine.

Follow this link:
Emergent in $75m expansion into viral vectors and gene therapy - BioPharma-Reporter.com

Read More...

SFARI | SFARI workshop explores challenges and opportunities of gene therapies for autism spectrum disorder – SFARI News

Thursday, June 25th, 2020

On February 67, 2020, the Simons Foundation Autism Research Initiative (SFARI) convened a two-day workshop to explore the possibility of gene therapies for autism spectrum disorder (ASD), a neurodevelopmental condition associated with changes in over 100 genes. Inspired by the recent, stunning successes of gene therapy for the fatal neuromuscular disorder spinal muscular atrophy (SMA)1, and by the accumulation of genes confidently associated with ASD2, SFARI welcomed a diverse collection of researchers to begin to think about whether a similar approach could be taken for ASD. Because gene therapy attempts to fix what is broken at the level of a causative gene, it would offer a more direct and imminent strategy than mitigation of the many and as yet mostly unclear downstream effects of a damaged gene.

The workshop was organized in 20 talks and several discussion panels, which tackled many outstanding issues, including how to choose candidate target genes and predict outcomes; how to optimize vectors for gene delivery; how to decide when to intervene; which animal models to develop; how to find appropriate endpoints for clinical trials and understand the available regulatory pathways. SFARI also raised the question of how its funding might best propel gene therapy efforts amid the emerging, complex ecosystem of academic laboratories, biotech companies, and pharmaceutical industries.

Even the opportunity to have this discussion is very rewarding, said SFARI Investigator Matthew State of the University of California, San Francisco (UCSF), one of the investigators who directed teams of geneticists to analyze the Simons Simplex Collection (SSC).

These efforts have offered up multiple potentially feasible therapeutic targets. Though rare, de novo disruptive mutations in the highest confidence ASD genes often result in severe impairment characterized not only by social difficulties, but also by intellectual disability and seizures. The combination of a single gene mutation of large effect coupled with particularly severe outcomes that include ASD are likely to offer the most immediate targets for gene therapy. For now, this leaves out a large number of individuals with autism for whom genetic causes are not yet known and are likely the result of a combination of many small effect alleles across a large number of genes.

Highlights from talks and discussion panel, chaired by Rick Lifton of Rockefeller University

In the first talk of the workshop, State brought the group up to speed on ASD genomics. The most recent tally from exome-sequencing in simplex cases of ASD highlighted 102 genes in which rare mutations confer individually large risks2. In contrast, the task of identifying common variants carrying very small risks remains quite challenging, with less than a half dozen alleles so far identified with confidence3. The rare, disruptive mutations that result in loss of function of one gene copy are an attractive focus for gene therapy because of the tractability of targeting a single spot in the genome per individual and because, in the vast majority of cases, there remains a single unchanged allele. This points to ways to boost gene and/or protein expression back toward the normal state by leveraging the unaffected copy. But both the limited number of cases known so far combined with the possibility that different mutations to the same gene may have different effects complicate thinking about how to prioritize targets for gene therapy.

State made several points that were continually touched on throughout the workshop. Many ASD genes are highly expressed during midfetal development in the cortex, and additional experiments will need to determine whether and how long a window of opportunity may be present for successful gene therapy postnatally. Given the relatively small number of people with these conditions, new clinical trial designs are needed that dont rely on comparisons between large control and intervention groups (see also Bryan Kings talk below).

Beyond the gene-crippling mutations found in the exome, disruptions to transcription may also dramatically raise risk for autism and may be corrected with a type of gene therapy using ASOs. SFARI Investigator Stephan Sanders of UCSF focused on the role of splicing, the process by which an initial transcript is turned into messenger RNA by removal of introns and joining together of exons. Splicing is disrupted in at least 1.5 percent of individuals with ASD4, and possibly many more, as suggested by transcript irregularities found in postmortem autism brain5. Sanders described Illuminas Splice AI project in which machine-learning helps predict noncoding variants that can alter splicing, including those beyond typical splice sites found near a gene6. As a result of incorporating sequence information around and between splice sites, this computational tool detected more mutations with predicted splice-altering consequences in people with ASD and intellectual disability than in those without the condition.

An ASO designed to bind specific portions of RNA could conceivably correct errors in transcription. ASOs have already been approved for use in other disorders in order to skip exons, retain exons or to degrade mRNA. Unlike other forms of gene therapy, ASOs do not permanently alter the genome, making it a kind of gene therapy lite. This reversibility has both disadvantages (having to re-infuse the ASO every few months) and advantages (multiple opportunities to optimize the dose and target; serious adverse effects are not permanent).

Jonathan Weissman of UCSF discussed the available toolbox for controlling gene expression developed by many different laboratories. To turn genes on or off, he has developed a method to combine CRISPR with an enzymatically inactive (dead) Cas9, which can then be coupled with a transcriptional activator (CRISPRa) or repressor (CRISPRi)7 (Figure 2). In the case of loss-of-function mutations, Weissman outlined strategies to make the remaining good allele work harder: increase transcription via CRISPRa, decrease mRNA turnover, increase translation of a good transcript via modification of upstream open reading frames (uORFs) or increase a proteins stability, possibly through small molecules acting on the ubiquitin system8. That said, the effects on a cell may be complicated. Using Perturb-Seq screens, Weissman described genetic interaction manifolds that show nonlinear mapping between genotype and single cell transcriptional phenotypes9. Additionally, Weissman summarized recent work from his laboratory that has identified large numbers of uORFs that result in polypeptides, some of which affect cellular function.

SFARI Investigator Michael Wigler of Cold Spring Harbor Laboratories echoed the idea of a gene-therapy strategy that increases expression of the remaining good copy of a gene, especially given that in his estimate, 45 percent of simplex cases of autism carried a de novo, likely disrupting variant. He also called attention to the uterine environment, especially the challenge posed by expression of paternally derived antigens in the fetus and the impact of a potential maternal immune response, and the need to understand how it interacts with de novo genetic events.

Highlights from talks and discussion panel, chaired by Arnon Rosenthal of Alector

The discussion turned to finding ways of getting genes into the central nervous system. The AAV is the darling of gene therapy, given that it does not replicate and is not known to cause disease in humans. A version that can cross the blood-brain barrier (AAV9) was used to deliver a gene replacement to children with SMA intravenously; though this effectively delivered the genetic cargo to ailing motor neurons in the spinal cord, it does not work that well at delivering genes throughout the brain.

Ben Deverman of the Stanley Center at the Broad Institute of MIT and Harvard detailed his efforts to optimize AAV for efficient transduction of brain cells through a targeted evolution process: his team engineers millions of variants in the capsid of the virus, then screens them for entry into the nervous system and transduction of neurons and glia. This has yielded versions (called AAV-PHP.B and AAV-PHP.eB) that more efficiently enter the brain10,11. One successfully delivered the MECP2 gene to the brain of a Rett syndrome mouse model, resulting in ameliorated symptoms and an extended lifespan12. Unfortunately, these viruses dont work in human cells or in all mouse strains. A quick mouse genome-wide association study (GWAS) revealed that the Ly6a gene mediates efficient blood-brain barrier crossing of AAV-PHP.B and AAV-PHP.eB13. Now his group has identified Ly6a-independent capsids that may translate better to humans. He also noted that the PHP.B vectors have tissue specificity for brain and liver.

With an estimated 87 percent of autism-associated genes raising risk through haploinsufficiency (having only one functional gene copy out of the two), SFARI Investigator Nadav Ahituv of UCSF made the case for approaches that boost expression of the remaining good copy of a gene through endogenous mechanisms a strategy he called cis-regulation therapy. This method also provides a way to work around the small four kb payload of AAV, which strains to contain cDNA of many autism genes. A recent study by his group used CRISPRa targeted at an enhancer or promoter of SIM1 and promoter of MC4R, both obesity genes, in mice. Using one AAV vector for a dCas9 joined to a transcription activator, and another AAV vector having a guide RNA targeting either a promoter or an enhancer, and a guide RNA targeting a promoter, the researchers injected the vectors together into the hypothalamus, which resulted in increased SIM1 or MC4R transcription and reversed the obesity phenotype brought on by loss of these genes14. Targeting regulatory elements had the added benefit of tissue specificity, and there seemed to be a ceiling effect for SIM1 expression, which suggested an endogenous safeguard against overexpression at work. He is now collaborating with SFARI Investigator Kevin Bender, also at UCSF, to apply this approach to the autism gene SCN2A.

Botond Roska of the Institute of Molecular and Clinical Ophthalmology in Basel, Switzerland pointed out that getting genes to the cells where they are needed is crucial when treating eye diseases. Off-target effects there can induce degeneration of healthy cells. For this reason, Roska and his group have created AAVs that target specific cell types in the retina by developing synthetic promoters that efficiently promote expression of the viruss cargo15. The promoters they designed were educated guesses based on four approaches: likely regulatory elements close to genes expressed with cell-type specificity in the retina, conserved elements close to cell typespecific genes, binding sites for cell typespecific transcription factors and open chromatin close to cell typespecific genes. Screening a library of these in mouse, macaque and human retina revealed some with high cell-type specificity (Figure 3). Importantly, macaque data predicted success in human retina much better than did mouse data. In preliminary experiments, and more relevant to gene therapy for ASD, these cell-specific vectors also had some success in mouse cortex, for example lighting up parvalbumin neurons or an apparently new type of astrocyte.

Roska also described new methods for delivery, in which nanoparticles are coated with AAV, then drawn into the brain using magnets16. This magnetophoresis technique allows a library of experimental AAVs to be tested at the same time in one monkey. Steering nanoparticles with magnets gives more control of vector placement and gene delivery. He argued that these in the future could access even deep structures of the brain.

Highlights from talks and discussion panel, chaired by Steven Hyman of the Broad Institute at MIT and Harvard

Kathy High of Spark Therapeutics reviewed the story of gene therapy for spinal muscular atrophy (SMA) type 1. Though she was not directly involved in that research, she is well aware of the regulatory atmosphere surrounding gene therapy, given that Spark Therapeutics developed the first approved AAV-delivered gene for a form of retinal dystrophy. The SMA story is a useful case study in that an ASO-based therapy (nusinersen, marketed as Spinraza), approved in 2016, set the stage for a gene-replacement therapy, marketed as Zolgensma (onasemnogene abeparvovec). Ultimately, the amount of data supporting Zolgensmas approval was modest: a Phase one dose study of 15 infants1, and an ongoing Phase three trial of 21 infants and safety data from 44 individuals. Yet the approval was helped by the dramatic results and clear endpoints: those receiving a single intravenous infusion of an AAV9 vector containing a replacement gene all remained alive at 20 months of age, whereas only 8 percent survived to that age in the natural history data, which compiles the diseases untreated course. High mentioned that maintaining product quality for gene therapeutics may prove trickier than for typical medications.

The attractive, highly customizable nature of gene therapy might have a regulatory downside in that different vector payloads, even when designed to do the same thing, could invite separate approval processes. Though not knowing how regulatory agencies would view this, High said that their perspectives are bound to evolve as more gene therapy trials are completed.

Getting to ASD-related syndromes, Bender talked about SCN2A, which encodes the sodium channel Nav1.2. SCN2A mutations in humans can be gain of function or loss of function; gain-of-function mutations are associated with early onset epilepsy, and loss-of-function mutations with intellectual disability and ASD. In a mouse model missing one copy of SCN2A, Bender and his group have discovered a role for SCN2A in action potential generation in the first week after birth, and in synaptic function and maturation afterward through regulation of dendritic excitability18 (Figure 4). Using AAV containing CRISPRa constructs developed with the Ahituv lab, the researchers successfully increased SCN2A expression, and recovered synapse function and maturity, even when done several weeks postnatally. Getting the appropriate dosage is critical since gain-of-function mutations are linked to epilepsy. However, Bender reported even when SCN2A expression increased to double normal levels, no hints of hyperexcitability appeared. We might be able to overdrive this channel as much as we want and actually may not have risk of producing an epileptic insult, he said. Next steps are to figure out the developmental windows for intervention, evaluate changes in seizure sensitivity and extend this kind of cis-regulatory approach to other ASD genes.

Angelman syndrome is another condition that attracts interest for gene therapy, in part because neurons already harbor an appropriate replacement gene. Angelman syndrome stems from mutations to the maternally inherited UBE3A gene, which is particularly damaging to neurons because they only express the maternal allele, while the paternal allele is silenced by an antisense transcript. SFARI Investigator Mark Zylka of the University of North Carolina and colleagues showed in 2011 that this paternal allele could be unsilenced with a cancer drug in a mouse model of Angelman syndrome19. Since then, three companies have built ASOs to do the same thing, and these are going into clinical trials. To get a more permanent therapeutic, Zylka has been developing CRISPR/Cas9 systems to reactivate paternal UBE3A, and preliminary experiments show that injecting this construct into the brains of embryonic mice, and then again at birth, results in brain-wide expression of paternal UBE3A and is long-lasting (at least 17 months). Zylka is now making human versions of these constructs. He later noted rare cases of mosaicism for the Angelman syndrome mutation people with 10 percent normal cells in blood have a milder phenotype20, which suggests that even inefficient transduction of a gene vector could help.

Zylka also made a case for prenatal interventions in Angelman syndrome: studies of mouse models indicate that early reinstatement of UBE3A expression in mouse embryos rescues multiple Angelman syndrome-related phenotypes, whereas later postnatal interventions rescue fewer of these21; for humans, a diagnostic, cell-based, noninvasive prenatal test will be available soon22; ultrasound-guided injections into fetal brain of nonhuman primates have been developed23; prenatal surgeries are now standard of care for spinal bifida; and intervening prenatally decreases the risk of an immunogenic response to an AAV vector or its cargo. During the discussion, it was noted that another benefit of acting early was that less AAV would be needed to transduce a much smaller brain; however, a drawback is the lack of data on Angelman syndrome development from birth to one year of age. This natural history would be necessary for understanding whether a prenatal therapy is more effective than treatment of neonates.

SFARI Investigator Guoping Feng of the Massachusetts Institute of Technology has been investigating SHANK3, a high-confidence autism risk gene linked to a severe neurodevelopmental condition called Phelan-McDermid syndrome, which is marked by intellectual disability, speech impairments, as well as ASD. SHANK3 is a scaffold protein important for organizing post-synaptic machinery in neurons. Mouse studies by Feng have shown that SHANK3 re-expression in adult mice that have developed without it can remedy some, but not all, of their phenotypes, including dendritic spine densities, neural function in the striatum and social interaction24. Furthermore, early postnatal re-expression rescued most phenotypes. This makes SHANK3 a potential candidate for gene therapy; however, it is a very large gene 5.2kb as a cDNA that is difficult to fit into a viral vector. To get around this, Fengs group has designed a smaller SHANK3 mini-gene as a substitute for the full-sized version. Preliminary experiments show that AAV delivery of the mini-gene can rescue phenotypes like anxiety, social behavior and corticostriatal synapse function in SHANK3 knockout mice. Feng also discussed his success in editing the genome in marmosets and macaques using CRISPR/Cas9 technology and showed data from a macaque model of SHANK3 dysfunction25. These models may help test gene therapy approaches and identify biomarkers of brain development closely related to the human disorder.

For people with rare conditions brought on by even rarer mutations, individualized gene therapies can provide a pathway for treatment. SFARI Investigator Timothy Yu of Boston Childrens Hospital/Harvard described his N-of-1 study in treating a girl with Batten disease, a recessive disorder in which a child progressively loses vision, speech and motor control while developing seizures. In a little over a year, an ASO that targeted her unusual splice-site mutation in the CLN7 gene was designed, developed and given intrathecally to the girl26. The lift was in negotiating with the FDA and working with private organizations, not just in the science, Yu said. After a year of treatment with the ASO (dubbed milasen after the girl, Mila), there were no serious adverse events; seizure frequency and duration had decreased (Figure 5); and possibly her decline had slowed. Though she remains blind, without intelligible speech and unable to walk on her own, she was still attentive and could respond happily to her familys voices. The highly personalized framework for this drugs approval is completely different from how medications meant for populations are approved, and it opens a regulatory can of worms, Yu said, though he added that the regulators were willing to countenance drug approval for an individuals clinical benefit.

Rett syndrome is a neurodevelopmental condition caused by mutations to the MECP2 gene that has a substantial research base in mouse models. Over 10 years ago, mouse models highlighted the possibility for therapeutics in this condition when Rett-associated phenotypes were rescued by adding back MECP2, even in adulthood27. This reversibility has spurred interest in gene therapy for Rett syndrome, but getting the MECP2 dose right is critical, said Stuart Cobb of the University of Edinburgh and Neurogene: just as too little MECP2 leads to Rett syndrome, too much also results in severe phenotypes. For this reason, it would be nice to package a replacement MECP2 gene with other regulatory elements to control its expression, but this results in constructs that do not fit into viral vectors. To make more room, Cobb and his colleagues have been able to chop away two-thirds of the MECP2, reserving two domains that interact to make a complex on DNA (Figure 6). Mice with this mini-gene are viable and have near normal phenotypes; likewise, injecting this mini-gene into MECP2-deficient mice extended their survival28. Doubling the dose, however, substantially lowered survival. Putting in safety valves to prevent overexpression is going to be quite important, he said. One idea is to add back a construct containing only the last two exons of MECP2, which is where most Rett mutations land. These would then be spliced into native transcripts (called trans-splicing), and thus their expression controlled by endogenous regulatory elements.

Underscoring the double-edged sword of MECP2 dosage, Yingyao Shao from Huda Zoghbis lab at Baylor described an MECP2 duplication syndrome (MDS) in humans, which features hypotonia, intellectual disability, epilepsy and autism. Experiments in an MDS mouse model, which carries one mouse version and one human version of MECP2, recapitulates some of the phenotypes of the human condition and can be rescued by an ASO targeting the human allele29. Shao described work to optimize the ASO for translation into humans, which involved developing a more humanized MDS model that carries two human MECP2 alleles. An acute injection of the ASO was able to knock down MECP2 expression in a dose-dependent manner in these mice, and RNA levels dropped a week after injection, with protein levels falling a week later. MECP2 target genes also normalized their expression level, and one maintained this for at least 16 weeks post-injection. The ASO also rescued behavioral phenotypes of motor coordination and fear conditioning, but not of anxiety; these corrections followed the molecular effects, and these timelines would be important to keep in mind while designing clinical trials. Shao also noted that overtreatment with the ASO resulted in Rett-associated phenotypes, but that this was reversible, which suggests that some fine-tuning of dosing in humans might be possible.

To avoid overtreatment and toxicity of any MDS-directed therapy, Mirjana Maletic-Savatic, also at Baylor, is leaving no stone unturned in a hunt for MDS biomarkers that can predict, in each individual, the safety of a particular dose and regimen. Such biomarkers would also help monitor individuals during treatment, give information about target engagement and identify candidates for a particular treatment. Anything found to be sensitive to expression levels of MECP2 could also be useful for Rett, though she noted that MECP2 levels measured in blood do not track linearly with gene copy number. Thus, because of interindividual variability, her approach is to collect a kitchen sink of data deriving composite biomarkers that accurately reflect the stage and severity of disease in a given case. She and her colleagues are collecting clinical, genetic, neurocircuitry (such as EEG and sleep waves), immunology and molecular data detected in blood, urine and CSF. These measures are also being explored in induced neurons derived from skin samples of people with MDS. She highlighted two interrelated potential biomarkers in the blood of those with this condition; both measures are downstream targets of MECP2 and are responsive to ASO treatment.

Highlights from Early detection and clinical trial issues talks and panel discussion, chaired by Paul Wang of SFARI

Coming up with objective measures of a persons status either their eligibility for a treatment, or whether the treatment has engaged with its target or even whether the treatment is effective is a real necessity in autism-related conditions, which comprise multiple interrelated behaviors. Eye-tracking methodology may provide such a marker, argued SFARI Investigator Ami Klin of Emory University. Focusing on the core social challenges of autism, Klin, Warren Jones and colleagues have been studying children as they view naturalistic social scenes to quantify their social attention patterns. This has revealed how remarkably early in development social visual learning begins and that this process is disrupted in infants later diagnosed with ASD prior to features associated with the condition appearing. By missing social cues, autism in many ways creates itself, moment by moment, Klin said. In considering gene therapy, it may be useful to know that eye looking (how much a subject looks at a persons eyes, an index of social visual engagement) in particular and social visual engagement in general are under genetic control30; that eye-tracking differences emerge as early as 26 months of age; and that homologies in social visual engagement exist between human babies and nonhuman infant primates.

In getting to a point to test gene therapies, identifying those who need them is essential. Wendy Chung of Columbia University and the Simons Foundation illustrated how diagnosis is yoked closely to therapy. To illustrate this, she described her pilot study of newborn blood spots to screen for SMA; at the start, no treatment was available, but the screen identified newborns for a clinical trial of nusinersin. Notably, the screen only cost an additional 11 cents per baby. In the three years since her pilot screen began, the FDA approved two gene therapies for SMA and the SMA screen was adopted for nationwide newborn screening. Currently she is piloting a screen for Duchenne muscular dystrophy and plans to develop a platform that will allow researchers to add other conditions. In prioritizing genetic conditions for gene therapy, she outlined some ideas for focus, such as genes resulting in phenotypes that would not be identified early without screening, those that are relatively frequent, those that are lethal or neurodegenerative, those with a treatment in clinical trials or with FDA-approved medications, and those conditions that are reversible.

In the meantime, Chung also outlined SFARIs involvement in establishing well-characterized cohorts of individuals with autism, which can help lay a groundwork for gene therapy. People with an ASD diagnosis can join SPARK (Simons Foundation Powering Autism Research for Knowledge), which collects medical, behavioral and genetic information (through analysis of DNA from saliva, at no cost to the participant). If a de novo genetic variant is found in one of ~150 genes, that person is referred to Simons Searchlight, which fosters rare conditions communities and which is also compiling natural history data on people with these mutations.

Bryan King of UCSF discussed how current trial designs for ASD were inadequate for gene therapy trials. As ASD prevalence has grown, parallel design trials with one group receiving an experimental medicine and the other a placebo are the standard, but these wont be possible for the rare conditions that are candidates for gene therapy. Also, change is hard to capture, given the malleable nature of ASD: with no intervention, diagnosis can shift between ASD and pervasive developmental disorder-not otherwise specified (PDD-NOS) in 1284 months (as defined by the DSM-IV). Current scales are subjective and may miss specific items of clinical significance. (Last year, SFARI funded four efforts to develop more sensitive outcome measures.) King outlined other pitfalls in ASD clinical trials, including significant placebo responses, inadequate sample sizes and not being specific enough when asking about adverse effects. King also mentioned improvements that may arise from just enrolling in a study, which could prompt previously housebound families to venture out with their child, which could kick off a cascade of positive effects. He reiterated how, for gene therapy, a natural history comparison group may be more appropriate, combined with solid outcome measures.

SFARI Investigator James McPartland of Yale University then underlined the need for objective biomarkers for clinical trials, for which there are currently none that are FDA qualified for ASD. As the director of the Autism Biomarkers Consortium for Clinical Trials (ABC-CT), he works with other scientists to develop reliable biomarkers that can be scaled for use in large samples across different sites. McPartland noted a biomarker studied in the ABC-CT: an event-related potential (N170) to human faces, which is on average slower in ASD than in typically developing children. He is working on ways to make it easier for people with ASD and intellectual disabilities to participate in biomarker studies and to make them more socially naturalistic. In discussion, he mentioned he thought it would be possible to look for these kinds of biomarkers in younger children.

SFARI Investigator Shafali Jeste of the University of California, Los Angeles recounted her experience in working with children with genetic syndromes associated with neurodevelopmental conditions. Though she is asked to participate in clinical trials for these conditions, she senses the field has some work to do to be ready for these trials, particularly in those with additional challenges such as epilepsy and intellectual disability. Meaningful and measurable clinical endpoints are still insufficient, and there needs to be more ways to improve accessibility of these trials for these rare conditions. This means developing new measures, such as gait-mat technology that senses walking coordination, or EEG measures in waking and sleep, which have been applied to people with chromosome 15q11.2-13.1 duplication (dup15q) syndrome, who have severe intellectual disability and motor impairments. Jeste also emphasized that increasing remote access to some measures can make a big difference for a trial; for example, a trial of a behavioral intervention for tuberous sclerosis complex that required weekly lab visits was disappointingly under-enrolled until researchers revamped it so most of the intervention could be done remotely31.

By grappling with the challenges to gene therapy for ASD, the workshop marked out a faint road map of a way forward. As the scientific questions are answered, the regulatory and clinical trial infrastructure will need to develop apace, and coordination between private, academic and advocacy sectors will be essential. But as gene therapy for diverse human conditions continues to be explored and gene discovery in ASD continues, there is reason to believe that some forms of ASD can eventually benefit from this strategy.This workshop provided a terrific discussion about the challenges in developing targeted gene interventions and their potentially transformative effects as therapies, said John Spiro, Deputy Scientific Director of SFARI. We are grateful to all theparticipants, and SFARI looks forward to translating these discussions into focused funding decisions in the near future.

Back to Top

Go here to see the original:
SFARI | SFARI workshop explores challenges and opportunities of gene therapies for autism spectrum disorder - SFARI News

Read More...

Sarepta tries to fine-tune its gene therapy approach – BioPharma Dive

Thursday, June 25th, 2020

Dive Brief:

Known for its muscular dystrophy drugs Exondys 51 and Vydonys 53, Sarepta has quickly become a leader in gene therapy as well.

The last couple of years have seen the Cambridge, Massachusetts-based biotech ink gene therapy manufacturing and development deals, including a partnership with Roche that could be worth more than $3 billion.

Sarepta now has six gene therapies in clinical testing and another half dozen in preclinical stages. Most recently, the company announced positive, albeit early, results from a study of its experimental treatment for limb-girdle muscular dystrophy, a potentially deadly genetic disease.

With gene therapy set to become a cornerstone of its business, Sarepta is trying to avoid some the challenges presented by current technologies.

For example, when the company teamed up with North Carolina-based StrideBio late last year, one of the main goals was to use the partner's technology to "address re-dosing challenges in patients who have received AAV-delivered gene therapy." The deal came just days after a study testing an experimental gene therapy from Solid Biosciences, one of Sarepta's rivals, was paused due to a patient experiencing an immune response and organ complications.

"If successful, the ability to re-dose will be an enormous leap forward in the science of gene therapy and provide invaluable benefits to patients beyond those we anticipate with one-time dosing," said Doug Ingram, Sarepta's CEO, in a June 18 statement announcing the Selecta deal.

Under terms of that deal, Sarepta will pay Selecta an initial, undisclosed amount. Selecta is eligible to receive pre-clinical milestone payments, and could take home additional development, regulatory and commercial milestones should Sarepta exercise its options to enter a licensing agreement.

Specifically, the deal with Selecta centers on gene therapies for Duchenne muscular dystrophy and certain limb-girdle muscular dystrophies.

With Codiac, Sarepta has offered up $72.5 million in upfront and near-term license payments plus research funding. In addition, Codiak is eligible for "significant" milestone payments, according to Sarepta.

The two-year deal gives Sarepta the option to license Codiak's technology for up to five neuromuscular targets. The companies said they will collaborate on the design of exosomes that can deliver and release gene therapy, gene editing or RNA payloads. If Sarepta exercises an option, Codiak will then be in charge of research until right before the candidate goes into in-human testing. Sarepta is responsible for clinical development and commercial activities.

Here is the original post:
Sarepta tries to fine-tune its gene therapy approach - BioPharma Dive

Read More...

UniQure gets out the gate first in race for Huntington’s gene therapy – BioPharma Dive

Thursday, June 25th, 2020

Dive Brief:

Huntington's disease is marked by physical and cognitive decline caused by cellular production of an abnormal version of a protein called huntingtin, which is essential for nerve cell health. About 30,000 people in the U.S. have the condition.

No treatments exist for the underlying disease, but Lundbeck's Xenazine and Teva's Austedo can help control the abnormal movements associated with Huntington's.

AMT-130, by contrast, uses a common virus to deliver strips of nucleic acid that can block the mutated genes that cause production of abnormal huntingtin. It is the first gene therapy to be tested in Huntington's disease patients.

UniQure plans to enroll 26 patients in this trial, with two groups receiving different doses of AMT-130 and a placebo arm that involves a partial surgery to make it appear patients have received the injection deep in the brain.

The main goal of the trial will be to determine if the therapy is safe, but as a secondary goal researchers will want to see how persistent AMT-130 is in the brain. Researchers will also be looking for signs that patients receiving the gene therapy decline more slowly than those who didn't, but the small trial may not be able to definitively answer that question.

UniQure likely won't be alone in this chase for long. Voyager, which has already advanced a Parkinson's disease gene therapy into the clinic in partnership with Neurocrine Biosciences, is nearing a decision on when to advance VY-HTT01 into the clinic.

At its first quarter corporate update, Voyager said it is "engaged in the ongoing conduct and review of preclinical studies" and would provide an update at mid-year.

View post:
UniQure gets out the gate first in race for Huntington's gene therapy - BioPharma Dive

Read More...

Timing Is an Essential Element of Cell & Gene Therapy Product Development – Xconomy

Thursday, June 25th, 2020

XconomyPhiladelphia

The science underpinning the latest investigational cell and gene therapies is complex enough. But the rapid advance of technologies that support development of these kinds of drugs presents logistical considerations, too.

Drug development is an interesting process because it takes five to 10 years to get a drug to market and invariably, somewhere along that line, the technologys a little outdated before it even is approved, and theres new things out there that are better, said Jeffrey Castelli, chief portfolio officer and head of gene therapy at Amicus Therapeutics (NASDAQ: FOLD). You look at some of the gene therapies that are now just ready coming to market and you think, wow, thats a pretty outdated gene therapy approachbut you really get locked in as youre moving your product toward development.

Castelli was among the executives who spoke as part of a panel at Xconomys online Xcelerating Life Sciences Philadelphia forum last week.

Amicus, headquartered in Cranbury Township, NJ, operates a gene therapy R&D center in Philadelphia.To keep its therapies from becoming stale prior to commercialization, the company keeps an eye on new technologies that arise, Castelli said. But some innovations invariably surface too late to incorporate.

We really try to implement them early on, and then make sure that when we have our second wave of programs and products weve incorporated some of the innovation, he said. But you do get locked into your approach along the way, and theres more and more switching costs as you move along that pathway.

To guide that process Tom Wilton, chief business officer at Philadelphia-based cell therapy company Carisma Therapeutics, says its essential to collect and analyze data early on to guide the path of drug development.

As a relatively small company youve really got to focus the majority of your resource on pushing forward that lead program and getting it into the clinic, he said. Later this year well get a lot of data back from that first-in-human clinical study, the first time anyones ever taken an engineered macrophage into the clinic. What that should give us then is a set of criteria and priorities around what we need to bring forward in a next-generator program.

To date the companys consideration of new technologies has been perhaps a bit more opportunisticeverything from gene editing the macrophages to novel binders to different combination strategiesbut once Carisma has the initial clinical data in hand, that will narrow its focus, Wilton said.

Theres always a tendency [with a first-generation product] to say we could do this to this, we could do this to this, but you have to lock it down, you have to get into the clinic and get that data set to really understand what your priorities are and where you need to focus, and thats what were planning to do.

As experimental products move into later stages of development, another consideration arises, the panelists said: Producing them at scale.

For cell and gene therapy companies, figuring out when to lock in the space needed to make these drugs so it is available when needed for clinical trials or commercialization is difficult to determine precisely, especially given the shortage of manufacturers versed in some of the advanced technologies needed to do so.

There is a capacity shortage right now in cell and gene therapy manufacturing, said Audrey Greenberg, co-founder and executive managing director of Discovery Labs, an MLP Ventures-backed biotech coworking and incubator space in King of Prussia, PA. Theres estimates that its five times [current cumulative bioreactor volumetric capacity] now and will be 50 times in five years given the FDA pipeline and the dollars flowing into these companies.

Discovery Labs is building out a $1.1 billion gene and cell treatment manufacturing operation plus developing a contract development manufacturing organization to provide services to companies in the sector.

Image: iStock/f11photo

Sarah de Crescenzo is an Xconomy editor based in San Diego. You can reach her at sdecrescenzo@xconomy.com.

More here:
Timing Is an Essential Element of Cell & Gene Therapy Product Development - Xconomy

Read More...

Sarepta Addressing Gene Therapy Issue With Two Acquisitions – Yahoo Finance

Thursday, June 25th, 2020

On the heels of research deals with two small biotechs, shares of Sarepta Therapeutics Inc. (NASDAQ:SRPT) hit their all-time high of more than $172 on June 23.

The share price of the Cambridge, Massachusetts-based biopharmaceutical company has doubled since mid-March. It has a market cap of more than $13 billion.

Sarepta has made several deals in the past several years that have made gene therapy a key part of its business, which had been dominated by drugs for muscular dystrophy. One of those pacts, with Roche (RHHBY), could be worth more than $3 billion.

Sarepta currently has a half dozen gene therapies in clinical trials, with another six waiting in the wings, according to BioPharma Dive. One of the company's compounds has shown promise in treating a type of muscular dystrophy, a genetic disease that causes weakness and wasting of the muscles in the arms and legs.

A major challenge of gene therapy is immune system reactions. Sarepta, like other developers, uses a type of virus in its therapy that, while effective, may not be able to given more than once because patients can create antibodies to it.

That's a problem Sarepta is trying to address in its latest deals with privately held Codiak Biosciences and Selecta Biosciences Inc. (NASDAQ:SELB). In both cases, Sarepta has an option to license the biotechs' technology to develop and commercialize its therapies.

The Selecta deal focuses on gene therapies for Duchene muscular dystrophy and certain limb-girdle muscular dystrophies.

A Zion Market Research report said the global demand for the Duchenne muscular mystrophy therapeutics market was valued at approximately $2.4 billion in 2018 and is expected to grow to more than $20 billion by the end of 2025, a compound annual growth rate of more than 36% between 2019 and 2025.

Given the size of the opportunity, it's no surprise Sarepta has plenty of competitors vying for a share of the business, including Pfizer Inc. (NYSE:PFE), PTC Therapeutics (NASDAQ:PTCT), FirbroGen Inc. (NASDAQ:FGEN), Roche and Bristol-Myers Squibb Co. (NYSE:BMY).

14b4c0913859c7da3fa2339160bde942.png

The two-year deal with Codiak gives Sarepta the right to license its technology for up to five neuromuscular diseases. If Sarepta exercises an option, Codiak will then handle research until right before the candidate goes into human testing. Sarepta will then be responsible for clinical development and commercializing the drug.

In April, the company announced it has resurrected its antiviral program in response to Covid-19. It has a therapy that is meant to block the coronavirus' ability to replicate. The treatment will be tested at the U.S. Army Medical Research Institute of Infectious Diseases.

"If it works, it will reduce the ability of the virus to replicate" and its ability to infect other cells, Sarepta CEO Doug Ingram told Forbes. He cautioned that the drug is at an "early stage."

According to CNN Money, the 21 analysts offering 12-month price forecasts for Sarepta have a median target of $192, with a high estimate of $260 and a low $152. The stock is rated a buy.

Disclosure: The author hold positions in Pfizer and Bristol-Myers Squibb.

Read more here:

Not a Premium Member of GuruFocus? Sign up for a free 7-day trial here.

This article first appeared on GuruFocus.

Read the original post:
Sarepta Addressing Gene Therapy Issue With Two Acquisitions - Yahoo Finance

Read More...

$14M Federal Grant to Research CAR-T Gene Therapy to Cure HIV – POZ

Thursday, June 25th, 2020

A group of collaborating scientists received a $13.65 million federal grant to study and develop a CAR-T therapy that will genetically modify immune cells and potentially cure HIV, according to a press release from the University of California, Los Angeles (UCLA).

The National Institutes of Health (NIH) funds the five-year grant as part of its effort to support HIV cure research. Participating researchers are affiliated with UCLA, the University of WashingtonFred Hutchinson Cancer Research Center and CSL-Behring, a biotech company based in the United States and Australia.

The overarching goal of our proposed studies is to identify a newgene therapy strategy to safely and effectively modify a patients own stem cells to resist HIV infection andsimultaneously enhance their ability to recognize and destroy infected cells in the body in hopes of curing HIV infection, said UCLAs Scott Kitchen, PhD, an associate professor of medicine in the division of hematology and oncology, in the press release. Kitchen will colead the research with Irvin Chen, PhD, director of the UCLA AIDS Institute at the David Geffen School of Medicine.

Transplantation ofHIV-resistant stem cells is the only approach that has ever led to a known cure for HIV(andlikely a second such cure). But stem cell transplants are risky and can only be done in people with HIVwho need them for cancer treatment. Using gene therapy tomodify an individuals own stem cells might be a safer way toachieve the same result.

The Food and Drug Administration first approved CAR-T therapywhich stands for chimeric antigen receptor T-cell therapyin 2017. Its used to treat some forms of cancer, but as POZs sister publication Cancer Health has reported, it hasnt been commonly used because it is expensive and must be custom made for each patient.

In the case of cancer treatment, CAR-T therapy involves taking a patients T cells and sending them to alab where they are genetically modified to recognize and attack the cancer. The resulting cells are then infused back into the individual after the person has received strong chemotherapy to kill off some of their existing immune cells to make room for the new ones.

In CAR-T therapy for HIV, blood-forming stem cells would be genetically engineered togive rise to T cells that would seek out and destroy cells infected with HIV.

In a recent early study of the approach, the UCLA scientists found that engineered CAR T cells destroyed HIV-infected cellsand lived for more than two years.

Our work under the NIH grant will provide a great deal of insight into ways the immune response can be modified to better fight HIV infection, said Chen, a professor of medicine and of microbiology, immunology and molecular genetics at the Geffen School of Medicine. The development of this unique strategy that allows the body to develop multiple ways to attack HIV could have an impact on other diseases as well, including the development of similar approaches targeting other types of chronic viral infections and cancers.

View post:
$14M Federal Grant to Research CAR-T Gene Therapy to Cure HIV - POZ

Read More...

Catalyst Biosciences Presents Preclinical FIX Gene Therapy Data in an Oral Presentation at the World Federation of Hemophilia Virtual Summit 2020 -…

Thursday, June 25th, 2020

SOUTH SAN FRANCISCO, Calif., June 19, 2020 (GLOBE NEWSWIRE) -- Catalyst Biosciences, Inc. (NASDAQ: CBIO), today presented data from preclinical studies of its hemophilia B gene therapy CB 2679d-GT at the WorldFederation of Hemophilia Virtual Summit, taking place from June 14 -19, 2020.

The oral presentation, entitled: Combination of a Novel Chimeric AAV Capsid and Potency Enhanced FIX Variant for Hemophilia B Gene Therapy, given by Dr. Grant Blouse, senior vice president of translational research, provided preclinical results of CB 2679d-GT, the companys novel FIX gene therapy. CB 2679d-GT was designed to achieve clinically relevant FIX levels at a reduced viral load by combining engineered AAV capsids with Catalysts novel high potency FIX transgene.

The preclinical data from our constructs demonstrated a strong dose response and improved reduction in bleeding relative to the Padua variant, said Nassim Usman, Ph.D., president and chief executive officer of Catalyst. The enhanced FIX activity and reduced viral dose may offer advantages over current AAV-based gene therapies in clinical development.

Studies of CB 2679d-GT in hemophilia B mice have demonstrated a 4-fold reduction in blood loss and an 8-fold reduction in bleeding time when compared with the same dose of the Padua variant of FIX. Furthermore, when packaged in a proprietary chimeric AAV capsid, CB 2679d-GT demonstrated a clear dose response of high stable FIX levels across the three dose levels in hemophilia B mice.

A pilot non-human primate study compared the expression and tolerability of CB 2679d-GT in the novel chimeric capsid KP1 with the LK03 capsid. The study demonstrated that CB 2679d-GT was well tolerated with high FIX expression that stabilized to approximately 25% to 50% FIX above baseline levels at the 6-week interim data cutoff. The novel chimeric capsid had differentiated and superior response to anti-capsid neutralizing antibodies than that observed for the LK03 comparator during the screening of non-human primates for the study.

A copy of the presentation slides can be accessed on the Events and Presentations section of the Catalyst website.

About Catalyst BiosciencesCatalyst is a research and clinical development biopharmaceutical company focused on addressing unmet needs in rare hematologic and systemic complement-mediated disorders. Our protease engineering platform includes development programs in hemophilia, a research program on subcutaneous (SQ) systemic complement inhibitors and a partnered preclinical development program with Biogen for dry age-related macular degeneration (AMD). One of our key competitive advantages is that the product candidates generated by our protease engineering platform have improved functionality and potency. These characteristics allow for improved dosing of our candidates including SQ systemic administration of recombinant coagulation factors and complement inhibitors, low-dose, high activity gene therapy constructs, and less frequently dosed intravitreal therapeutics. Our most advanced asset, SQ MarzAA has successfully completed Phase 2 development in prophylaxis, significantly reducing the annualized bleed rate (ABR) in individuals with Hemophilia A or B with inhibitors. Following regulatory guidance from the U.S. Food and Drug Administration and European Medicines Agency, we recently announced the design of a Phase 3 registration study that is planned for late 2020. Subcutaneous dalcinonacog alfa (DalcA) is being developed for the treatment of Hemophilia B and has demonstrated efficacy and safety in a Phase 2b clinical trial. We have a discovery stage Factor IX gene therapy construct - CB 2679d-GT - for Hemophilia B, that has demonstrated superiority compared with the Padua variant in preclinical models. Finally, we have a global license and collaboration agreement with Biogen for the development and commercialization of anti-complement Factor 3 (C3) pegylated CB 2782 for the potential treatment of geographic atrophy-associated dry AMD.

Forward-Looking StatementsThis press release contains forward-looking statements that involve substantial risks and uncertainties. Forward-looking statements include statements about the superiority of CB 2679d-GT over the Padua variant, enhanced FIX activity of CB 2679d-GT, which may reduce viral dose and maintain high FIX activity levels while potentially decreasing liver toxicity, the chimeric capsid, which may have lower neutralization by pre-existing AAV antibodies, as well as plans for a Phase 3 trial of MarzAA in late 2020 and the Companys collaboration with Biogen for the development and commercialization of pegylated CB 2782 for the potential treatment of geographic atrophy-associated dry age-related macular degeneration. Actual results or events could differ materially from the plans, intentions, expectations and projections disclosed in the forward-looking statements. Various important factors could cause actual results or events to differ materially, including, but not limited to, the risk that trials and studies may be delayed as a result of the COVID-19 virus and other factors, that trials may not have satisfactory outcomes, that additional human trials will not replicate the results from animal trials or earlier human trials, that potential adverse effects may arise from the testing or use of DalcA or MarzAA, including the generation of neutralizing antibodies, which has been observed in patients treated with DalcA, the risk that costs required to develop or manufacture the Companys products will be higher than anticipated, including as a result of delays in development and manufacturing resulting from COVID-19 and other factors, the risk that Biogen will terminate Catalysts agreement, competition and other risks described in the Risk Factors section of the Companys quarterly report filed with the Securities and Exchange Commission on May 11, 2020, and in other filings with the Securities and Exchange Commission. The Company does not assume any obligation to update any forward-looking statements, except as required by law.

Contact:Ana KaporCatalyst Biosciences, Inc.investors@catbio.com

Read more from the original source:
Catalyst Biosciences Presents Preclinical FIX Gene Therapy Data in an Oral Presentation at the World Federation of Hemophilia Virtual Summit 2020 -...

Read More...

The Alliance for Regenerative Medicine Announces Its 2020 Cell & Gene Meeting on the Mesa Goes Virtual – GlobeNewswire

Thursday, June 25th, 2020

Washington, DC, June 24, 2020 (GLOBE NEWSWIRE) -- via NEWMEDIAWIRE -- The Alliance for Regenerative Medicine (ARM), an international multi-stakeholder advocacy organization representing the regenerative medicine and advanced therapy sector, today announced a new virtual format for its annual Cell & Gene Meeting on the Mesa, which will take place October 12-16, 2020.

ARM's leadership and Board of Directors arrived at the decision to host the 2020 Cell & Gene Meeting on the Mesa virtually after consulting with numerous ARM members and gathering information from various health authorities. Ultimately, hosting the meeting virtually will be the safest and most inclusive solution, allowing attendees to connect with potential partners globally while avoiding any risk to safety.

This year's Cell & Gene Meeting on the Mesa will bring together senior executives from leading cell therapy, gene therapy, and tissue engineering companies worldwide, large pharma and biotech, institutional investors, academic research institutions, patient foundations and disease philanthropies, life science media, and more.

During this unprecedented period of social distancing, ARM continues to provide members with avenues to engage and connect, said ARM CEO Janet Lambert. Our virtual Meeting on the Mesa is an invaluable opportunity for stakeholders from across the sector to convene, to network, and to continue to work to ensure innovative cell and gene therapies reach patients in need.

The conference, which will now take place over five days, includes a virtual form of the meetings signature partnering system, expected to facilitate more than 3,000 one-to-one meetings between industry leaders. The program will include 15+ digital panels and workshops featuring key industry leaders discussing issues and trends in the regenerative medicine and advanced therapy sector, from market access to the latest discoveries in gene editing. Representatives from more than 80 prominent public and private companies will deliver on-demand presentations highlighting their clinical and commercial progress to interested partners and investors.

Additional event details will be updated regularly on the event website http://www.meetingonthemesa.com.

Registration is currently open, with discounted early-bird rates available through July 24. Registration is complimentary for investors and credentialed members of the media. To learn more and to register, please visitwww.meetingonthemesa.com. For members of the media interested in attending, please contact Kaitlyn (Donaldson) Dupont atkdonaldson@alliancerm.org.

For interested organizations looking to increase exposure to this fields top decision-makers via sponsorship, please contact Laura Stringham at lparsons@alliancerm.org for additional information.

About the Alliance for Regenerative Medicine

The Alliance for Regenerative Medicine (ARM) is an international multi-stakeholder advocacy organization that promotes legislative, regulatory, and reimbursement initiatives necessary to facilitate access to life-giving advances in regenerative medicine worldwide. ARM also works to increase public understanding of the field and its potential to transform human healthcare, providing business development and investor outreach services to support the growth of its member companies and research organizations. Prior to the formation of ARM in 2009, there was no advocacy organization operating in Washington, D.C. to specifically represent the interests of the companies, research institutions, investors, and patient groups that comprise the entire regenerative medicine community. Today, ARM has more than 350 members and is the leading global advocacy organization in this field. To learn more about ARM or to become a member, visithttp://www.alliancerm.org.

Kaitlyn (Donaldson) Dupont

803-727-8346

kdonaldson@alliancerm.org

Continue reading here:
The Alliance for Regenerative Medicine Announces Its 2020 Cell & Gene Meeting on the Mesa Goes Virtual - GlobeNewswire

Read More...

Page 33«..1020..32333435..4050..»


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