header logo image


Page 8«..78910..2030..»

Archive for the ‘Genetic Engineering’ Category

ETFs in Focus on Bayer’s Bet on Gene Therapy – Yahoo Finance

Saturday, October 31st, 2020

Merger and acquisition activities have heated up in recent months and the niche spaces are in the limelight. After the telemedicine industry, gene therapy stocks jumped on the bandwagon. Gene therapy is a technique that uses genes to treat or prevent disease.

German drugmaker Bayer has made a big bet on gene therapy by announcing the acquisition of U.S. biotech firm Asklepios BioPharmaceutical for as much as $4 billion. The proposed acquisition will provide Bayer access to the adeno-associated virus (AAV) gene therapy platform and a pipeline led by clinical-phase treatments for Parkinsons, Pompe disease and congestive heart failure. Notably, AAV therapies offer improved efficacy, immune response, and tissue and organ specificity.

Additionally, the transaction will complements Bayers 2019 acquisition of BlueRock Therapeutics and advances its efforts to create platforms with the potential to have an impact on multiple therapeutic areas (read: Genomics ETFs Surge on Nobel Prize for Gene-Editing Pioneers).

Under the terms of the deal, Bayer will pay an upfront consideration of $2 billion and potential success-based milestone payments of up to $ billion. About 75 % of the potential milestone-based contingent payments are expected to be due during the course of the next five years and the remaining amount thereafter.

The deal, pending regulatory approvals, is expected to close during the fourth quarter of 2020. Once the deal closes, Bayer will allow Asklepios, known as AskBio, to operate autonomously as part of a new cell and gene therapy unit in a bid to preserve its entrepreneurial culture. The cell and gene therapy unit will bundle Bayer's activities in this area moving forward in order to establish an innovation ecosystem for the participating partners, the German company said (see: all the Healthcare ETFs here).

The proposed deal will provide a boost to the gene therapy industry. Below, we have highlighted four ETFs that are expected to benefit from Bayers entrance into the gene therapy space:

ARK Genomic Revolution Multi-Sector ETF ARKG

This actively managed ETF is focused on companies that are likely to benefit from extending and enhancing the quality of human and other life by incorporating technological and scientific developments, and advancements in genomics into their business. With AUM of $2.9 billion, the fund holds 47 stocks in its basket and has 0.75% in expense ratio. It trades in an average daily volume of 978,000 shares (read: 4 Sector ETFs That Have Doubled This Year).

Invesco Dynamic Biotechnology & Genome ETF PBE

This fund follows the Dynamic Biotech & Genome Intellidex Index and provides exposure to companies engaged in the research, development, manufacture and marketing and distribution of various biotechnological products, services and processes and companies that benefit significantly from scientific and technological advances in biotechnology and genetic engineering and research. It holds 31 stocks in its basket and charges 57 bps in annual fees. The ETF has managed $229.9 million in its asset base while trades in a light volume of 6,000 shares per day. Expense ratio comes in at 0.57%. The product has a Zacks ETF Rank #3 (Hold) with a High risk outlook.

Global X Genomics & Biotechnology ETF GNOM

This product seeks to invest in companies that potentially stand to benefit from further advances in the field of genomic science, such as companies involved in gene editing, genomic sequencing, genetic medicine/therapy, computational genomics and biotechnology. It follows the Solactive Genomics Index, holding 40 stocks in its basket. This ETF has accumulated $68 million in its asset base and charges 50 bps in annual fees. It trades in average daily volume of 31,000 shares (read: Why You Should Invest in Genomics ETFs).

iShares Genomics Immunology and Healthcare ETF IDNA

This ETF provides access to companies at the forefront of genomics and immunology innovation by tracking the NYSE FactSet Global Genomics and Immuno Biopharma Index. Holding 46 stocks in its basket, the fund has gathered $166.2 million in AUM and trades in moderate average daily volume of 58,000 shares. It charges 47 bps in annual fees.

Story continues

Zacks free Fund Newsletter will brief you on top news and analysis, as well as top-performing ETFs, each week. Get it free >>

Want the latest recommendations from Zacks Investment Research? Today, you can download 7 Best Stocks for the Next 30 Days. Click to get this free reportInvesco Dynamic Biotechnology Genome ETF (PBE): ETF Research ReportsARK Genomic Revolution ETF (ARKG): ETF Research ReportsGlobal X Genomics Biotechnology ETF (GNOM): ETF Research ReportsiShares Genomics Immunology and Healthcare ETF (IDNA): ETF Research ReportsTo read this article on Zacks.com click here.Zacks Investment ResearchWant the latest recommendations from Zacks Investment Research? Today, you can download 7 Best Stocks for the Next 30 Days. Click to get this free report

Read the original here:
ETFs in Focus on Bayer's Bet on Gene Therapy - Yahoo Finance

Read More...

Lesaffre and Recombia Biosciences apply gene editing technology in fermented ingredients production – FoodIngredientsFirst

Saturday, October 31st, 2020

30 Oct 2020 --- French yeast manufacturer Lesaffre and Recombia Biosciences have partnered to advance a gene editing technology for the sustainable production of fermented ingredients.

Using Recombias proprietary gene editing technologies, the partnership aims to generate thousands of new yeast strains while optimizing the production of biosourced ingredients.

We see tremendous potential to leverage our expertise in genome editing and synthetic biology to develop new and innovative fermentation solutions and products, says Dr. Justin Smith, CEO of Recombia Biosciences.

Click to EnlargeThe partnership signals Lesaffres entry into the world of Synthetic Biology, which the company notes is considered to be a major biotechnological opportunity of this decade.Recombia Biosciences was founded by three Stanford University researchers in 2019 as a spin-off from the Stanford Genome Technology Center (SGTC), in the US.

Its technologies are based upon techniques that increase the efficiency of genome editing and enable engineering of yeast at very high throughput.

The technology has broad utility and can be readily applied also to the development of non-genetically modified organisms, adds Carmen Arruda, R&I manager at Lesaffre.

With Recombia, Lesaffre can now explore a larger space of metabolic engineering hypotheses, develop prototype organisms at a faster pace, accelerate the design of appropriate selections and screenings of strains generated by classical breeding methods. Were excited to see what the future holds.

Entry into synthetic biologyThe partnership signals Lesaffres entry into the world of Synthetic Biology, which the company notes is considered to be a major biotechnological opportunity of this decade.

This kind of partnership exemplifies an innovative way that industry can support and foster progress in Biotechnology, says Antoine Baule, CEO of Lesaffre.

Through collaborations with scientists and entrepreneurs, we will be able to find new solutions, which will be beneficial for the future, especially in health or in environment protection.

Recombia is exclusively licensing four genome engineering technologies from Stanford University for their work.

Bridging industry with academiaWhile precision genome editing has certainly advanced recently, there are still challenges, especially in making many genetic changes in parallel, notes Dr. Bob St.Onge, COO and co-founder of Recombia Biosciences.

Recombias technologies enable industrial yeast strain engineering by dramatically increasing the efficiency of high-throughput genome editing, he remarks.

Click to EnlargeWhile precision genome editing has certainly advanced recently, there are still challenges ahead.

St.Onge and Smith co-founded the company with Professor Lars Steinmetz. The team has had a significant working relationship at the Stanford Genome Technology Center (SGTC).

Im very excited to see the technologies we developed in academia applied in the industrial sector, comments Steinmetz.

The Genome Technology Center has a long history of genomics technology development. Im confident Recombia will continue in the tradition of the other successful companies that have spun out of the SGTC.

Unlocking the genome for new ingredientsGenomic research is widely applicable across food-tech applications. For instance, this type of analysis is employed to map the chemical fingerprint of chocolate, using the genes of the tree that cacao pods are harvested from.

Also in this space, seed breeding specialist Equinom is leveraging its advanced breeding techniques to promote agricultural biodiversity. The company upholds its pivotal position in inducing better crop resilience and increased yield.

Meanwhile, a genomic study comparing historic and modern wheat varieties recently revealed an increase in dietary fiber and a decrease in acrylamide, indicating that white bread is not as unhealthy as it has often been portrayed.

Edited by Benjamin Ferrer

To contact our editorial team please email us at editorial@cnsmedia.com

If you found this article valuable, you may wish to receive our newsletters. Subscribe now to receive the latest news directly into your inbox.

Go here to see the original:
Lesaffre and Recombia Biosciences apply gene editing technology in fermented ingredients production - FoodIngredientsFirst

Read More...

Sugar beet industry flips narrative on GMO crops – Idaho State Journal

Saturday, October 31st, 2020

It's only been a few years since U.S. sugar beet farmers faced a potential financial crisis due to negative public perceptions about food products derived from biotechnology.

Nowadays, however, the sugar beet industry is flipping the narrative, capitalizing on what was once its Achilles heel its universal adoption of GMO seed.

Since, 2009, the nation's sugar beet crop has been almost entirely planted in seed genetically modified to resist glyphosate herbicide, which is produced at Bayer's Soda Springs plant. Lately, to strike a chord with an increasingly environmentally conscious consumer base, the sugar beet industry has been touting how biotechnology has made its crop production system far more sustainable.

Story continues below video

Idaho is the No. 2 state in production of sugar beets by ton.

"We have lots of data," said Scott Herndon, vice president and general counsel with the American Sugarbeet Growers Association. "We submitted something to the National Academy of Sciences where we identified 25 environmental gains achieved through biotech seeds related to water, soil and air."

Herndon explained the gene added to confer glyphosate resistance to GMO beets is entirely removed in the processing of sugar. He referenced laboratory testing conducted by his industry proving finished sugar raised by American sugar beet farmers is identical to organic sugar and cane sugar in general.

Yet efforts to pass GMO labeling laws prior to 2016 in states including Vermont, Washington State, Oregon and California resulted in conventional cane sugar enjoying a whopping price advantage of roughly 7 cents over beet sugar, said Luther Markwart, executive vice president of American Sugarbeet Growers Association.

"That's huge," Markwart said.

Furthermore, the large Pennsylvania-based candy manufacturer Hershey Co. removed GMO beet sugar from many of its products in 2015 in favor of conventional cane sugar. In explaining the decision, Hershey affirmed GMO ingredients are safe but emphasized its commitment to openness and transparency.

"People care about the food they eat," Hershey posted on its website. "They want to know what's inside and they want to have choices so that snack options meet their expectations."

Rather than providing the public with transparency, Markwart believes mandatory labeling of beet sugar would be misleading, causing people to falsely believe beet sugar and cane sugar are somehow different.

Under the Obama Congress passed a preemptive law in 2016 exempting beet-derived sugar from state GMO labeling requirements. It took effect in December of 2018, though food companies are still allowed to voluntarily state that a product is derived from a bioengineered crop.

"Once you got the federal preemption, all of a sudden the beet and cane price came back together," Herndon said.

Labeling proponents, however, note that surveys consistently find consumers support GMO labeling by wide margins, believing people who are skeptical of a technology that is found in more than three-quarters of the processed foods in supermarkets should have the ability to avoid it.

"While many in the scientific community assert that GMO foods are not toxic and are safe, a significant number of scientists are sounding the alarm," Mark Fergusson, CEO of Down to Earth Organic & Natural, said in an essay posted on his organization's website. "They say genetic engineering poses risks that scientists simply do not know enough to identify."

Fergusson encouraged the public to choose foods with the organic seal, certifying that GMOs were not used in production.

GMO advocates argue that reputable scientific data evidencing health risks of GMO technology has yet to be produced.

The industry's comments to the National Academy of Sciences on Sept. 9, 2015, for example, cited several studies on the safety of GMO crops, including a 2011 summary report by the European Commission covering a decade of publicly funded research, 130 research projects and 500 research groups concluding "there is no scientific evidence of higher risks of GE crops for food and feed safety, or to the environment."

The environmental benefits of GMO crops, however, are well documented, Markwart said.

When beets were raised conventionally, Markwart said herbicides applied about four times per year to control weeds stymied crop development, essentially taking a month of growth off of the final yield. Glyphosate applications in GMO beets don't set development back whatsoever, enabling farmers to produce more with fewer farming inputs.

Furthermore, GMO beets don't require hand weeding and mechanical cultivation between rows, saving farmers on labor costs and avoiding soil disturbance, which dries out soil and releases greenhouse gases.

The sugar beet industry points to a 2002 study by the National Center for Food and Agriculture Policy evaluating eight of the most commonly used herbicides, finding glyphosate posed the least potential risk. Furthermore, glyphosate binds tightly to the soil and is less likely to contaminate groundwater, according to the industry's literature.

According to USDA data, sugar beet yields increased by 19% with glyphosate-resistant seed from 2008 through 2015 compared with the conventional average. The percentage of sugar in each beet also increased dramatically.

Brad Griff, executive director of the Idaho Sugarbeet Growers Association, said the Gem State's farmers have increased their yields by roughly 10 tons per acre on average since the implementation of GMO sugar beets.

"Before the GE sugar beet we never had reached 18% sugar. Now for three of the last six years we've been at or above 18% sugar," Griff added. "That's all been accomplished while reducing pesticide use by 85% and reducing fuel use by 60%."

In its Agriculture Innovation Agenda, USDA asked farmers of various commodities to plan strategies to increase their output by 40% while cutting their environmental impacts in half. The sugar beet industry submitted comments toward that effort focusing largely on achieving the goals through improved genetics.

Looking ahead, Markwart anticipates sugar beet seed engineered with multiple-trait tolerance to three herbicides glyphosate, dicamba and glufosinate should be released by 2026. He explained the new seed should help farmers avoid the onset of glyphosate-resistant weeds by enabling them to use multiple modes of action.

The industry also sees great promise in gene editing, which allows crop breeders to silence or amplify existing traits in a plant's genetics rather than introducing desirable foreign traits.

Herndon believes sustainability should be viewed as a "three-legged stool" factoring in social, economic and environmental costs. He said farmers are the lifeblood of rural economies and have a social contract to help keep their rural communities afloat. While it's important that they continually push the envelope to reduce their impacts on the environment, Herndon said farmers must also eke out a profit.

Markwart added, "We used to have hand labor. No one is going to go back and do that again. We have made major advances in efficiencies with this technology."

Excerpt from:
Sugar beet industry flips narrative on GMO crops - Idaho State Journal

Read More...

Polymerase Chain Reaction (PCR) Market expanding at a considerable CAGR from 2018 to 2026 – The Think Curiouser

Saturday, October 31st, 2020

Transparency Market Research (TMR)has published a new report titled,Polymerase Chain Reaction (PCR) Market: Global Industry Analysis, Size, Share, Growth, Trends, and Forecast, 20182026.According to the report, theglobal PCR marketis projected to reach over US$ 7.0 Bn by 2026 expanding at a considerable CAGR from 2018 to 2026. North America is expected to dominate the global PCR market during the forecast period, due to high adoption of technologically advanced molecular diagnostic products, growing incidence of infectious diseases, rising prevalence of various types of cancer, growing trend of self-diagnosis of diseases, and affordability of high-cost testing processes for people in the region.

Request Brochure for Report https://www.transparencymarketresearch.com/sample/sample.php?flag=B&rep_id=830

Reagents Segment to Dominate the Global PCR Market

The report offers detailed segmentation of the global PCR market based on product and end-user. In terms of product, the market has been segmented into instruments, reagents, and consumables. The reagents segment held the leading market share in the year 2017 and is expected to expand at the highest CAGR during the forecast period. This is attributable to high consumption of reagents for the PCR technique. Introduction of advanced reagents specific to the type of test is expected to boost the global demand for PCR reagents in the near future. PCR reagents include template DNA, PCR primers and probes, dNTPs, PCR buffers, enzymes, and master mixes. PCR consumables mostly include PCR tubes, plates, and other accessories required to conduct PCR reactions. The instruments segment has been sub-categorized into standard PCR systems, RT PCR systems, and digital PCR systems. Among these, the digital PCR systems sub-segment is expected to witness growth at a significantly rapid pace during the forecast period. This is attributable to advantages of digital PCR systems such as precision, sensitivity, accuracy, reproducibility, direct quantification and multiplexing, and speed of the analysis.

Request for Analysis of COVID19 Impact on Polymerase Chain Reaction (PCR) Market

https://www.transparencymarketresearch.com/sample/sample.php?flag=covid19&rep_id=830

Pharmaceutical & Biotechnology Industry Segment Held a Major Share of the Global PCR Market in 2017

The PCR technique has been found to be useful in pharmaceutical and biotechnology research activities as well as microbial quality testing. The technique is also applied in genetic engineering. Genetic engineering is the key driver for the global PCR market. It is used to identify genes related to certain phenotypes includinggenetic disorders. Regular testing of the microbial load of raw materials and finished products is an important process in the pharmaceutical & biotechnology industry. Sophisticated analytical methods such as polymerase chain reaction (PCR) have been widely applied for quality control analysis in the pharmaceutical sector.

Market in Asia Pacific to Expand at a High CAGR

Molecular diagnosis has revolutionized the modern diagnosis technology. PCR has become a method of choice in early and accurate detection of diseases. Expansion by leading manufacturers of PCR products in the Asia Pacific region by strengthening of the distribution network and new product launches in developing countries of Asia Pacific are key factors likely to drive the PCR market in the region during the forecast period. Moreover, rise in the incidence of cancer and infectious diseases has resulted in increase in the demand for use of the PCR technique in clinical diagnosis of these diseases in Asia Pacific. For instance, according to the Korea Central Cancer Registry published in 2016, there were 217,057 cancer cases in South Korea in 2014. Moreover, in 2016, the WHO estimated that the Asia Pacific region has the second-highest number (i.e. 5.1 million) of people living with HIV across the world. Thus, Asia Pacific is expected to be the most lucrative market for PCR by 2026.

Buy Polymerase Chain Reaction (PCR) Market Report https://www.transparencymarketresearch.com/checkout.php?rep_id=830&ltype=S

Competition Landscape

Major players operating in the global PCR market are Bio-Rad Laboratories, Inc., QIAGEN N.V., F. Hoffmann-La Roche AG, Thermo Fisher Scientific, Inc. Becton, Dickinson and Company, Abbott, Siemens Healthcare GmbH (Siemens AG), bioMrieux SA, Danaher Corporation, and Agilent Technologies. Key players are expanding their product portfolio through mergers and acquisitions and partnerships and collaborations with leading pharmaceutical and biotechnology companies and by offering technologically advanced products.

About Us

Transparency Market Research is a global market intelligence company providing global business information reports and services. Our exclusive blend of quantitative forecasting and trends analysis provides forward-looking insight for several decision makers. Our experienced team of analysts, researchers, and consultants use proprietary data sources and various tools and techniques to gather and analyze information.

Our data repository is continuously updated and revised by a team of research experts so that it always reflects latest trends and information. With a broad research and analysis capability, Transparency Market Research employs rigorous primary and secondary research techniques in developing distinctive data sets and research material for business reports.

Contact

Transparency Market Research,

90 State Street, Suite 700,

Albany, NY 12207

Tel: +1-518-618-1030

USA Canada Toll Free: 866-552-3453

Website: https://www.transparencymarketresearch.com/

Read more:
Polymerase Chain Reaction (PCR) Market expanding at a considerable CAGR from 2018 to 2026 - The Think Curiouser

Read More...

Aridis Pharmaceuticals to Present at the ROTH Capital Partners 2020 MedTech Innovation Forum on a COVID-19 Panel – BioSpace

Saturday, October 31st, 2020

SAN JOSE, Calif., Oct. 27, 2020 /PRNewswire/ -- Aridis Pharmaceuticals, Inc. (Nasdaq: ARDS), a biopharmaceutical company focused on the discovery and development of novel anti-infective therapies to treat life-threatening infections, today announced the Company will present at the ROTH Capital Partners 2020 MedTech Innovation Forum on Wednesday, October 28, 2020. Dr. Hasan Jafri, Chief Medical Officer of Aridis Pharmaceuticals, will be a speaker on a panel entitled "Direct Antivirals and Other Agents Against SARS-CoV2 Virus."

Panel: Direct Antivirals and Other Agents Against SARS-CoV2 VirusDate: Wednesday, October 28, 2020Time: 10:30AM-11:50AM ET

Dr. Jafri will present a summary of the Company's recently published preclinical data of its COVID-19 inhaled mAb (AR-711). He will address the preclinical performance of AR-711, the advantages of direct lung delivery using nebulized aerosols, and the COVID-19 clinical program.

About AR-711

AR-711 is a fully human immunoglobulin 1, or IgG1, monoclonal antibody discovered from screening the antibody secreting B-cells of convalescent COVID-19 patients. AR-711 exhibits high affinity for SARS-CoV-2 spike protein, approximately 10-fold or higher than mAb candidates currently in late stage clinical testing. AR-711 was previously shown to be effective in prophylactic as well as therapeutic treatment modes in a SARS-CoV-2 viral challenge study. AR-711 is currently being developed as an inhaled, self-administered treatment for non-hospitalized patients suffering from mild to moderate COVID-19. AR-711 is also one the two mAbs in the company's AR-701 mAb cocktail, which is a separate program being developed as an intravenous treatment of moderate to severe, hospitalized COVID-19 patients.

About Aridis Pharmaceuticals, Inc.

Aridis Pharmaceuticals, Inc. discovers and develops anti-infectives to be used as add-on treatments to standard-of-care antibiotics. The Company is utilizing its proprietary PEXTM and MabIgX technology platforms to rapidly identify rare, potent antibody-producing B-cells from patients who have successfully overcome an infection, and to rapidly manufacture monoclonal antibody (mAbs) for therapeutic treatment of critical infections. These mAbs are already of human origin and functionally optimized for high potency by the donor's immune system; hence, they technically do not require genetic engineering or further optimization to achieve full functionality.

The Company has generated multiple clinical stage mAbs targeting bacteria that cause life-threatening infections such as ventilator associated pneumonia (VAP) and hospital acquired pneumonia (HAP), in addition to preclinical stage antiviral mAbs. The use of mAbs as anti-infective treatments represents an innovative therapeutic approach that harnesses the human immune system to fight infections and is designed to overcome the deficiencies associated with the current standard of care which is broad spectrum antibiotics. Such deficiencies include, but are not limited to, increasing drug resistance, short duration of efficacy, disruption of the normal flora of the human microbiome and lack of differentiation among current treatments. The mAb portfolio is complemented by a non-antibiotic novel mechanism small molecule anti-infective candidate being developed to treat lung infections in cystic fibrosis patients. The Company's pipeline is highlighted below:

Aridis' Pipeline

AR-301 (VAP). AR-301 is a fully human IgG1 mAb currently in Phase 3 clinical development targeting gram-positive Staphylococcus aureus (S. aureus) alpha-toxin in VAP patients.

AR-101 (HAP). AR-101 is a fully human immunoglobulin M, or IgM, mAb in Phase 2 clinical development targeting Pseudomonas aeruginosa (P. aeruginosa) liposaccharides serotype O11, which accounts for approximately 22% of all P. aeruginosa hospital acquired pneumonia cases worldwide.

AR-501 (cystic fibrosis). AR-501 is an inhaled formulation of gallium citrate with broad-spectrum anti-infective activity being developed to treat chronic lung infections in cystic fibrosis patients. This program is currently in a Phase 1/2a clinical study in healthy volunteers and CF patients.

AR-401 (blood stream infections). AR-401 is a fully human mAb preclinical program aimed at treating infections caused by gram-negative Acinetobacter baumannii.

AR-701 (COVID-19). AR-701 is a cocktail of fully human mAbs discovered from convalescent COVID-19 patients that are directed at multiple envelope proteins of the SARS-CoV-2 virus.

AR-711 (COVID-19). AR-711 is an in-licensed mAb that is directed against the receptor binding domain of the SARS-Cov 2 virus. The agent has the potential to be delivered both intravenously and by inhalation using a nebulizer.

AR-201 (RSV infection). AR-201 is a fully human IgG1 mAb out-licensed preclinical program aimed at neutralizing diverse clinical isolates of respiratory syncytial virus (RSV).

For additional information on Aridis Pharmaceuticals, please visit https://aridispharma.com/.

Forward-Looking Statements

Certain statements in this press release are forward-looking statements that involve a number of risks and uncertainties. These statements may be identified by the use of words such as "anticipate," "believe," "forecast," "estimated" and "intend" or other similar terms or expressions that concern Aridis' expectations, strategy, plans or intentions. These forward-looking statements are based on Aridis' current expectations and actual results could differ materially. There are a number of factors that could cause actual events to differ materially from those indicated by such forward-looking statements. These factors include, but are not limited to, the need for additional financing, the timing of regulatory submissions, Aridis' ability to obtain and maintain regulatory approval of its existing product candidates and any other product candidates it may develop, approvals for clinical trials may be delayed or withheld by regulatory agencies, risks relating to the timing and costs of clinical trials, risks associated with obtaining funding from third parties, management and employee operations and execution risks, loss of key personnel, competition, risks related to market acceptance of products, intellectual property risks, risks related to business interruptions, including the outbreak of COVID-19 coronavirus, which could seriously harm our financial condition and increase our costs and expenses, risks associated with the uncertainty of future financial results, Aridis' ability to attract collaborators and partners and risks associated with Aridis' reliance on third party organizations. While the list of factors presented here is considered representative, no such list should be considered to be a complete statement of all potential risks and uncertainties. Unlisted factors may present significant additional obstacles to the realization of forward-looking statements. Actual results could differ materially from those described or implied by such forward-looking statements as a result of various important factors, including, without limitation, market conditions and the factors described under the caption "Risk Factors" in Aridis' 10-K for the year ended December 31, 2019 and Aridis' other filings made with the Securities and Exchange Commission. Forward-looking statements included herein are made as of the date hereof, and Aridis does not undertake any obligation to update publicly such statements to reflect subsequent events or circumstances.

Contact:

Investor RelationsJason WongBlueprint Life Science Groupjwong@bplifescience.com(415) 375-3340 Ext. 4

SOURCE Aridis Pharmaceuticals, Inc.

View original content to download multimedia:http://www.prnewswire.com/news-releases/aridis-pharmaceuticals-to-present-at-the-roth-capital-partners-2020-medtech-innovation-forum-on-a-covid-19-panel-301160211.html

SOURCE Aridis Pharmaceuticals, Inc.

Company Codes: NASDAQ-NMS:ARDS

Read the original post:
Aridis Pharmaceuticals to Present at the ROTH Capital Partners 2020 MedTech Innovation Forum on a COVID-19 Panel - BioSpace

Read More...

Aviceda Therapeutics Announces Formation of Scientific Advisory Board – BioSpace

Saturday, October 31st, 2020

Oct. 27, 2020 12:00 UTC

CAMBRIDGE, Mass.--(BUSINESS WIRE)-- Aviceda Therapeutics, a late-stage, pre-clinical biotech company focused on developing the next generation of immuno-modulators by harnessing the power of glycobiology to manipulate the innate immune system and chronic, non-resolving inflammation, is announcing the members of its Scientific Advisory Board who will help shape ongoing development efforts.

The Aviceda Scientific Advisory Board includes Pamela Stanley, PhD; Ajit Varki, MD; Christopher Scott, PhD; Geert-Jan Boons, PhD; Salem Chouaib, PhD; and Peng Wu, PhD.

Aviceda has assembled an extraordinary multi-disciplinary team of world-class scientists and renowned researchers to join our efforts in developing the next generation of glyco-immune therapeutics for the treatment of immune-dysfunction conditions, said Mohamed A. Genead, MD, Founder, CEO & President of Aviceda Therapeutics. Each individual offers a fresh perspective and unique strategic acumen that complements and strengthens the insights of our in-house leadership development team.

Prof. Scott, Aviceda Scientific Co-Founder, is Director of the Patrick G Johnston Centre for Cancer Research and Cell Biology at Queens University Belfast. He is internationally renowned for his work in development of novel approaches in the field of antibody and nanomedicine-based therapies for the treatment of cancer and other conditions. Prof. Scott has a background in both the pharmaceutical industry and academia and was a founding scientist of Fusion Antibodies Plc. Research in his laboratory is funded by agencies such as Medical Research Council, UK charities and various industrial sources. He also held a Royal Society Industrial Fellowship with GSK from 2012 to 2015 and won the Vice Chancellors Prize for Innovation in 2015 with his groups work on developing a novel Siglec targeting nanomedicine for the treatment of sepsis and other inflammatory conditions.

The novelty of Avicedas platform technology is its potential to affect immune responses associated with a wide range of disease states, many of which are currently unmet or underserved needs. I look forward to the continued development of Avicedas core technology and moving forward to clinical trials that will pave the way for truly disruptive therapeutic strategies to enter the clinic that will significantly impact and improve patients lives in the not-too-distant future, said Prof. Scott.

Avicedas Scientific advisory chairwoman, Prof. Stanley, is the Horace W. Goldsmith Foundation Chair; Professor, Department of Cell Biology; and Associate Director for Laboratory Research of the Albert Einstein Cancer Center, Albert Einstein College of Medicine, New York. She obtained a doctorate degree from the University of Melbourne, Australia, for studies of influenza virus, and was subsequently a postdoctoral fellow of the Medical Research Council of Canada in the laboratory of Louis Siminovitch, University of Toronto, where she studied somatic cell genetics. Prof. Stanleys laboratory is focused on identifying roles for mammalian glycans in development, cancer and Notch signaling. Among her many varied contributions, Prof. Stanleys laboratory has isolated a large panel of Chinese hamster ovary (CHO) glycosylation mutants; characterized them at the biochemical, structural and genetic levels; and used them to identify new aspects of glycan synthesis and functions. She serves on the editorial boards of Scientific Reports, Glycobiology and FASEB Bio Advances; she is an editor of the textbook Essentials of Glycobiology; and her laboratory is the recipient of grants from the National Institutes of Health. Prof. Stanley has received numerous awards, including a MERIT award from the National Institutes of Health, an American Cancer Society Faculty Research Award, the Karl Meyer Award from the Society for Glycobiology (2003) and the International Glycoconjugate Organization (IGO) Award (2003).

Working with Aviceda represents a unique opportunity to contribute to science at the cutting edge. Its pipeline contains a broad range of candidates that represents numerous first-in-class opportunities, said Prof. Stanley.

Prof. Varki is currently a distinguished professor of medicine and cellular and molecular medicine, Co-director of the Glycobiology Research and Training Center and Executive Co-director for the UCSD/Salk Center for Academic Research and Training in Anthropogeny at the University of California, San Diego; and an Adjunct Professor at the Salk Institute for Biological Studies. Dr. Varki is also the executive editor of the textbook Essentials of Glycobiology. He received basic training in physiology, medicine, biology and biochemistry at the Christian Medical College, Vellore, The University of Nebraska, and Washington University in St. Louis, as well as formal training and certification in internal medicine, hematology and oncology. Dr. Varki is the recipient of numerous awards and recognitions, including election to the American Academy of Arts and Sciences and the US National Academy of Medicine, a MERIT award from the National Institutes of Health, an American Cancer Society Faculty Research Award, the Karl Meyer Award from the Society for Glycobiology and the International Glycoconjugate Organization (IGO) Award (2007).

The Aviceda team is already building on the foundational work in the emerging field of glycobiology to develop potential therapeutics and interventional strategies. Their work could be critically important for growing the understanding of how glycobiology and glycochemistry are applicable to immunology, and more broadly, to the field of drug and therapeutic development, said Prof. Varki.

Prof. Boons is a Distinguished Professor in Biochemical Sciences at the Department of Chemistry and the Complex Carbohydrate Research Center (CCRC) of the University of Georgia (USA) and Professor and Chair of the Department of Medicinal and Biological Chemistry of Utrecht University (The Netherlands). Prof. Boons directs a research program focused on the synthesis and biological functions of carbohydrates and glycoconjugates. The diversity of topics to which his group has significantly contributed includes the development of new and better methods for synthesizing exceptionally complex carbohydrates and glycoconjugates. Highlights of his research include contributions to the understanding of immunological properties of complex oligosaccharides and glycoconjugates at the molecular level, which is being used in the development of three-component vaccine candidates for many types of epithelial cancer; development of convergent strategies for complex oligosaccharide assembly, which make it possible to synthesize large collections of compounds with a minimal effort for structure activity relationship studies; and creation of a next generation glycan microarray that can probe the importance of glycan complexity for biological recognition, which in turn led to identification of glycan ligands for various glycan binding proteins that are being further developed as glycomimetics for drug development for various diseases. Among others, Prof. Boons has received the Creativity in Carbohydrate Science Award by the European Carbohydrate Association (2003), the Horace Isbell Award by the American Chemical Society (ACS) (2004), the Roy L. Whistler International Award in Carbohydrate

Chemistry by the International Carbohydrate Organization (2014), the Hudson Award (2015) and the Cope Mid-Career Scholar Award from ACS (2016).

Aviceda is leading the field of glycoimmunology in exciting new directions. I look forward to working with the company as it pursues multiple lines of development efforts that will someday transform the way immune-inflammatory conditions are treated in the clinic, said Prof. Boons.

Prof. Chouaib is the Director of Research, Institute Gustave Roussy, Paris, where he is active in research in tumor biology. Previously, Prof. Chouaib worked at the French National Institute of Health and Biomedical Research (INSERM) where he led a research unit focused on the investigation of the functional cross talk between cytotoxic cells and tumor targets in the context of tumor microenvironment complexity and plasticity. His research was directed at the transfer of fundamental concepts in clinical application in the field of cancer vaccines and cancer immunotherapy. Prof. Chouaib is a member of the American Association of Immunologists, New York Academy of Sciences, French Society of Immunologists, International Cytokine Society, American Association for Cancer Research, International Society for Biological Therapy of Cancer and American Association of Biological Chemistry. He was awarded the cancer research prize of the French ligue against cancer in 1992 and in 2004 the presidential prize in biotechnology. He was awarded for translational research and scientific excellency by INSERM. His research has resulted in more than 310 scientific articles and several reviews in the field of human immunology, tumor biology and cancer immunotherapy; he has also been an editor for several textbooks.

Dr. Wu is an Associate Professor in the Department of Molecular Medicine at Scripps Research. The current research in the Wu laboratory integrates synthetic chemistry with glycobiology to explore the relevance of protein glycosylation in human disease and cancer immunotherapy. In 2018, Dr. Wu developed a platform to construct antibody-cell conjugates for cancer immunotherapy, which does not require genetic engineering. Previously, while working as a postdoctoral fellow in the group of Professor Carolyn R. Bertozzi at the University of California, Berkeley, Dr. Wu developed an aldehyde-tag (SMARTag) based technology for site-specific labeling of monoclonal antibodies, which served as the foundation for Redwood Biosciences Inc., a biotech company co-founded by Bertozzi. In 2014, Redwood Bioscience Inc. and the SMARTag Antibody-Drug Conjugate technology platform was acquired by Catalent Pharma Solutions.

About Aviceda Therapeutics

Founded in 2018 and based in Cambridge, Massachusetts, Aviceda Therapeutics is a late-stage, pre-clinical biotechnology company with a mission to develop the next generation of glyco-immune therapeutics (GITs) utilizing a proprietary technology platform to modulate the innate immune system and chronic, non-resolving inflammation. Aviceda has assembled a world-class, cross-disciplinary team of recognized scientists, clinicians and drug developers to tackle devastating ocular and systemic degenerative, fibrotic, oncologic and immuno-inflammatory diseases. At Aviceda, we exploit a unique family of receptors found expressed on all innate immune cells and their associated glycobiological interactions to develop transformative medicines. Combining the power of our biology with our innovative cell-based high-throughput screening platform and proprietary nanoparticle technology, we can modulate the innate immune response specifically and profoundly. Aviceda is developing a pipeline of GITs that are delivered via biodegradable nanoparticles and which safely and effectively target numerous immune-inflammatory conditions. Avicedas lead ophthalmic optimized nanoparticle, as an intravitreal formulation, AVD-104, is being developed to target various immune system responses that contribute to pathology associated with age-related macular degeneration (AMD).

View source version on businesswire.com: https://www.businesswire.com/news/home/20201027005101/en/

Read this article:
Aviceda Therapeutics Announces Formation of Scientific Advisory Board - BioSpace

Read More...

Genetic modification is ready to serve humanity The Miscellany News – Miscellany News

Sunday, October 18th, 2020

On Oct. 7, 2020, Dr. Emmanuelle Charpentier and Dr. Jennifer A. Doudna were awarded the Nobel Prize in Chemistry for their work in the field of gene editing. On top of breaking barriers as the first two women jointly awarded the chemistry prize, Charpentier and Doudnas recognition is a huge step forward for the controversial field of genetic engineering.

Humans have been practicing a form of genetic engineering ever since we started cultivating plants and livestock. Grafting two plants together dates back centuries in both the East and the West, and selective breeding was a staple technique used by even the earliest farmers. These techniques arent using advanced technology to target and change certain genes, but nevertheless the point of these exercises was to eliminate or diminish unwanted characteristics and promote the characteristics that the farmer found most useful. Wild cabbage was bred to create broccoli, brussel sprouts and domesticated cabbage. Cattle were bred to increase their edible volume. This was all uncontroversial, but it was all gene editing.

Today the techniques have changed, but the underlying mission has stayed the same: improve quality of life. Public opinion has shifted, however. Currently, more than half of adults in the U.S. believe that using genetically modified organisms (GMOs) as a food source is worse for your health than using non-modified foods. Of those, 88 percent believe that GMO foods will lead to health problems for the general populace. There is no such thing as non-modified food, but there is a stigma against food modified in a lab.

Part of this bias may be due to the way direct modification was introduced in the 1950s. In order to increase variation in plants so that selective breeding could be done more efficiently, scientists bombarded plants with radiation. This process, known as mutation breeding, was part of an effort to discover a peaceful use for the nuclear knowledge that was proliferating in the aftermath of World War II. Radiation was poorly understood by the general public in the mid-20th century. The possibilities of mutation due to radiation caused imagination to run rampant over reality: 1954s Them! stars giant insects caused by nuclear testing in the area.

The 1957 film Beginning of the End has grasshoppers eat mutated plants and then grow to enormous sizes. Even some of the most famous pop culture characters that exist today were formulated along these lines. In 1961 the Fantastic Four were given their powers by cosmic radiation. Spider-Man has had eight movies over the last 20 years, and he was famously bitten by a radioactive spider. These examples dont insinuate that people really believed that radiation could produce superheroes and skyscraper-sized insects, but they do reflect a general fear of the unknown that the gene modification of radiation could produce.

Radiation is no longer the bugaboo of the modern day, but fear of radiation has been displaced by fear of targeted gene editing, like the Crispr-Cas9 technique pioneered by Charpentier and Doudna. Some of this fear may be well founded: Theres no definite way to know that a gene edited plant or animal wont act similar to an invasive species. Presumably freed from some ailment or deficit that was limiting its growth, it is possible that a plant may grow at a pace that is higher than wanted by its creators. Nature is a delicate balance, and intervening must be done in a reasonable way that weighs the potential costs and benefits.

Mosquito reduction or elimination may not seem to be a worthwhile risk for something with unknown side effects, but that initial intuition would be wrong. Malaria, a disease transmitted mainly through mosquito bites, kills around 400,000 people per year. Zika and West Nile virus, while less deadly, are also transmitted into the human populace via mosquito. No other creature kills humans at the rate of mosquitoes. Despite the environmental damage that may be wreaked by the adjustment of the other flora and fauna to a lack of mosquitoes, gene editing to reduce mosquito population is a clear path to saving hundreds of thousands of lives every year.

With this sort of benefit in mind, the United States Environmental Protection Agency and Florida state government recently came to an agreement that will release over 750 million genetically modified mosquitoes into Florida. This is no small action and could potentially disrupt the entire food web of Florida, and possibly beyond.

The plan in Florida is to introduce a strain of Aedes Aegypti mosquitoes, a spreader of the Zika virus, that are genetically engineered so that their female offspring die off. Mosquitoes bite to extract human blood, and in this exchange mosquitoes can transfer any diseases they are carrying. Mosquitoes only bite so that they can extract iron and proteins in human blood and transfer it to the fertilized eggs that will be the next generation of that mosquitos bloodline. As such, the only mosquitoes that bite, and thus have the chance to transfer diseases, are adult females. The firm Oxitec produced a modified mosquito whose female offspring cant grow out of the larval stage. No adult females means no blood sucking, which means no disease transmission and no new mosquito larvae being produced.

A similar plan was executed in Brazil, where the Aedes Aegypti mosquito population was cut by 89 to 96 percent. With such a large reduction in mosquito population, the benefits move beyond that of just public health. Thousands of tracts of land would become more usable and see an increase in value if mosquitoes died out. Even day-to-day activities like gardening or talking walks could become much more pleasant in the absence of mosquitoes.

2020 has already shown the effects of disease and failures of public health. COVID-19 has killed over a million people; over the last 10 years, malaria has killed over four million. We have to live with COVID-19 for the foreseeable future, but gene editing has given us a tool to end malaria. Genetically modified mosquitoes should not end in Florida or with Aedes Aegypti: they should be of all species, placed all over the globe. For months the world has lived under a new biological terror. Its time we release a new biological salvation.

View original post here:
Genetic modification is ready to serve humanity The Miscellany News - Miscellany News

Read More...

Dyslexia shows the inborn nature of visual imagining and cognition – Genetic Literacy Project

Sunday, October 18th, 2020

Reading is a learned skill; no one is born reading. But learning to read relies on inborn human capacities for language and speech. And dyslexia is ageneticcondition that compromises thesebrain networks.

Yet laypeople are convinced that dyslexia results from troubleswith vision. And these errors matter. A parent who holds these views might fail to recognize her childs difficulties with rhymes and pig Latin (both require phonemic awareness) as warning signs. So why are we so wrong about dyslexia? Why do we mistake dyslexia for word blindness?

At first blush, these misconceptions seem rather innocent; laypeople, by definition, arent reading experts, so perhaps they just dont know better. But aspiringteachers, with ample educational training, make similar mistakes. Moreover, the pattern of mistakes suggests a deeper problem.

While these biases are unconscious, they demonstrably veer off reasoning in numerous areas, from our irrational fascination with the brain to ourfear of artificial intelligence; our troubles with dyslexia, then, are but one of its many victims. To counter these errors, information alone wont sufficea real change requires that we take a hard look within.

Reading, then, rests on decoding in more ways than one. For children to successfully decode printed words, we must all improve our decoding of the human mind.

Read the original post

Originally posted here:
Dyslexia shows the inborn nature of visual imagining and cognition - Genetic Literacy Project

Read More...

AGC Biologics Shifts Leadership Structure at United States and Copenhagen Sites to Support the Continued Development and Growth of the Regions -…

Sunday, October 18th, 2020

SEATTLE, Oct. 15, 2020 /PRNewswire/ -- AGC Biologics, a leading global biopharmaceutical contract development and manufacturing organization (CDMO), has announced a leadership update at the United States and Copenhagen facilities. The changes are being made to strengthen the strategic development and executive oversight of the rapid growing facilities in the US and Copenhagen, and are effective at the date of release, October 15, 2020.

Jeffrey D. Mowery will join the Global Executive Team in the role of Senior Vice President of US Operations, based at company headquarters in Seattle, Washington. Andrea C. Porchia will become the General Manager and Site Head for the Copenhagen Operation.

In his new position, Mr. Mowery will oversee the new Boulder, Colorado facility rollout and ensure that progress is maintained at the expanding Seattle site. Mr. Mowery draws on more than two decades of industry expertise in small molecule, biologic and cell and gene therapy production and technology transfer expertise to deliver quality in his work at AGC Biologics.

"In his most recent role as General Manager of the Copenhagen, Denmark facility, J.D. Mowery achieved a period of strong growth, even with today's challenges from the COVID pandemic. We believe the US sites, and ultimately our customers, will benefit from his leadership skills, results oriented approach and broad operational expertise in the same way that Copenhagen has," said Kasper Moller, CTO of AGC Biologics. He continued, "as part of this transition, Andrea C. Porchia has been promoted to General Manager of the Copenhagen site where her broad and deep biologics experience, and ability to effectively navigate all aspects of biomanufacturing and development will be an indispensable asset for the Copenhagen Site and to our valued customers."

Through more than seven years at AGC Biologics, Ms. Porchia has taken on increasing responsibilities, both at the Copenhagen site and globally as Project Director, Business Development Representative, Global Head of Project Management and now General Manager. She leverages more than two decades of research and process expertise to enhance business operations with a critical focus on project management and customer service.

To learn more about the AGC Biologics global network of facilities, please visit: http://www.agcbio.com/.

About AGC Biologics

AGC Biologics is a leading global biopharmaceutical contract development and manufacturing organization (CDMO) committed to delivering a high standard of service to solve complex customer challenges. The company is driven by innovation and continuously invests in technologies to complement decades of proven expertise in drug development and manufacturing, including working through FDA, PDMA and EMA approvals. A range of customizable bioprocessing services includes development and manufacturing of mammalian and microbial-based therapeutic proteins, protein expression, plasmid DNA (pDNA) support, antibody drug development and conjugation, viral vector production, genetic engineering of cells, cell line development with a proprietary CHEF1 Expression System, cell banking and storage.

AGC Biologics employs more than 1,400 professionals worldwide who are dedicated to supporting customers at all phases of development through to commercialization, with critical expertise in process development, formulation, and analytical testing. The global service network boasts locations in the United States at Seattle, Washington and Boulder, Colorado; across Europe in Copenhagen, Denmark; Heidelberg, Germany; Milan and Bresso, Italy; and in Asia at Chiba, Japan.

Learn more at http://www.agcbiologics.com, or find us on LinkedIn at https://www.linkedin.com/company/agcbiologics/ and Twitter @agcbiologics.

See the original post here:
AGC Biologics Shifts Leadership Structure at United States and Copenhagen Sites to Support the Continued Development and Growth of the Regions -...

Read More...

Scientists Found a New Way to Control the Brain With LightNo Surgery Required – Singularity Hub

Sunday, October 18th, 2020

If I had to place money on a neurotech that will win the Nobel Prize, its optogenetics.

The technology uses light of different frequencies to control the brain. Its a brilliant mind-meld of basic neurobiology and engineering that hijacks the mechanism behind how neurons naturally activateor are silencedin the brain.

Thanks to optogenetics, in just ten years weve been able to artificially incept memories in mice, decipher brain signals that lead to pain, untangle the neural code for addiction, reverse depression, restore rudimentary sight in blinded mice, and overwrite terrible memories with happy ones. Optogenetics is akin to a universal programming language for the brain.

But its got two serious downfalls: it requires gene therapy, and it needs brain surgery to implant optical fibers into the brain.

This week, the original mind behind optogenetics is back with an update that cuts the cord. Dr. Karl Deisseroths team at Stanford University, in collaboration with the University of Minnesota, unveiled an upgraded version of optogenetics that controls behavior without the need for surgery. Rather, the system shines light through the skulls of mice, and it penetrates deep into the brain. With light pulses, the team was able to change how likely a mouse was to have seizures, or reprogram its brain so it preferred social company.

To be clear: were far off from scientists controlling your brain with flashlights. The key to optogenetics is genetic engineeringwithout it, neurons (including yours) dont naturally respond to light.

However, looking ahead, the study is a sure-footed step towards transforming a powerful research technology into a clinical therapy that could potentially help people with neurological problems, such as depression or epilepsy. We are still far from that visionbut the study suggests its science fiction potentially within reach.

To understand optogenetics, we need to dig a little deeper into how brains work.

Essentially, neurons operate on electricity with an additional dash of chemistry. A brain cell is like a living storage container with doorscalled ion channelsthat separate its internal environment from the outside. When a neuron receives input and that input is sufficiently strong, the cells open their doors. This process generates an electrical current, which then gallops down a neurons output brancha biological highway of sorts. At the terminal, the electrical data transforms into dozens of chemical ships, which float across a gap between neurons to deliver the message to its neighbors. This is how neurons in a network communicate, and how that network in turn produces memories, emotions, and behaviors.

Optogenetics hijacks this process.

Using viruses, scientists can add a gene for opsins, a special family of proteins from algae, into living neurons. Opsins are specialized doors that open under certain frequencies of light pulses, something mammalian brain cells cant do. Adding opsins into mouse neurons (or ours) essentially gives them the superpower to respond to light. In classic optogenetics, scientists implant optical fibers near opsin-dotted neurons to deliver the light stimulation. Computer-programmed light pulses can then target these newly light-sensitive neurons in a particular region of the brain and control their activity like puppets on a string.

It gets cooler. Using genetic engineering, scientists can also fine-tune which populations of neurons get that extra powerfor example, only those that encode a recent memory, or those involved in depression or epilepsy. This makes it possible to play with those neural circuits using light, while the rest of the brain hums along.

This selectivity is partially why optogenetics is so powerful. But its not all ponies and rainbows. As you can imagine, mice dont particularly enjoy being tethered by optical fibers sprouting from their brains. Humans dont either, hence the hiccup in adopting the tool for clinical use. Since its introduction, a main goal for next-generation optogenetics has been to cut the cord.

In the new study, the Deisseroth team started with a main goal: lets ditch the need for surgical implants altogether. Immediately, this presents a tough problem. It means that bioengineered neurons, inside a brain, need to have a sensitive and powerful enough opsin door that responds to lighteven when light pulses are diffused by the skull and brain tissue. Its like a game of telephone where one person yells a message from ten blocks away, through multiple walls and city noise, yet you still have to be able to decipher it and pass it on.

Luckily, the team already had a candidate, one so good its a ChRmine (bad joke cringe). Developed last year, ChRmine stands out in its shockingly fast reaction times to light and its ability to generate a large electrical current in neuronsabout a 100-fold improvement over any of its predecessors. Because its so sensitive, it means that even a spark of light, at its preferred wavelength, can cause it to open its doors and in turn control neural activity. Whats more, ChRmine rapidly shuts down after it opens, meaning that it doesnt overstimulate neurons but rather follows their natural activation trajectory.

As a first test, the team used viruses to add ChRmine to an area deep inside the brainthe ventral tegmental area (VTA), which is critical to how we process reward and addiction, and is also implicated in depression. As of now, the only way to reach the area in a clinical setting is with an implanted electrode. With ChRmine, however, the team found that a light source, placed right outside the mices scalp, was able to reliably spark neural activity in the region.

Randomly activating neurons with light, while impressive, may not be all that useful. The next test is whether its possible to control a mouses behavior using light from outside the brain. Here, the team added ChRmine to dopamine neurons in a mouse, which in this case provides a feeling of pleasure. Compared to their peers, the light-enhanced mice were far more eager to press a lever to deliver light to their scalpsmeaning that the light is stimulating the neurons enough for the mice to feel pleasure and work for it.

As a more complicated test, the team then used light to control a population of brain cells, called serotonergic cells, in the base of the brain, called the brainstem. These cells are known to influence social behaviorthat is, how much an individual enjoys social interaction. It gets slightly disturbing: mice with ChRmine-enhanced cells, specifically in the brainstem, preferred spending time in their test chambers social zone versus their siblings who didnt have ChRmine. In other words, without any open-brain surgery and just a few light beams, the team was able to change a socially ambivalent mouse into a friendship-craving social butterfly.

If youre thinking creepy, youre not alone. The study suggests that with an injection of a virus carrying the ChRmine geneeither through the eye socket or through veinsits potentially possible to control something as integral to a personality as sociability with nothing but light.

To stress my point: this is only possible in mice for now. Our brains are far larger, which means light scattering through the skull and penetrating sufficiently deep becomes far more complicated. And again, our brain cells dont normally respond to light. Youd have to volunteer for what amounts to gene therapywhich comes with its own slew of problemsbefore this could potentially work. So keep those tin-foil hats off; scientists cant yet change an introvert (like me) into an extrovert with lasers.

But for unraveling the inner workings of the brain, its an amazing leap into the future. So far, efforts at cutting the optical cord for optogenetics have come with the knee-capped ability to go deep into the brain, limiting control to only surface brain regions such as the cortex. Other methods overheat sensitive brain tissue and culminate in damage. Yet others act as 1990s DOS systems, with significant delay between a command (activate!) and the neurons response.

This brain-control OS, though not yet perfect, resolves those problems. Unlike Neuralink and other neural implants, the study suggests its possible to control the brain without surgery or implants. All you need is light.

Image Credit: othebo from Pixabay

More here:
Scientists Found a New Way to Control the Brain With LightNo Surgery Required - Singularity Hub

Read More...

No evidence that coronavirus genetic sequences were fabricated, contrary to preprint by Li-Meng Yan and colleagues – Health Feedback

Sunday, October 18th, 2020

CLAIM

Fabricated genetic sequences were used to support the hypothesis that the virus arose naturally

DETAILS

Inadequate support: The preprint by Yan et al. offers no evidence to support their claim that the genetic sequences of other coronavirus strains were fabricated to support the hypothesis that SARS-CoV-2 arose naturally.Incorrect: The fact that multiple coronavirus strains share highly similar or identical genetic or protein sequences is not evidence that those viruses were fabricated. Shared genetic or protein sequences is common among viruses that belong to the same family and indicates their evolutionary relatedness.

KEY TAKE AWAY

There is no evidence supporting the claim by Yan et al. that genetic sequences of several coronaviruses were fabricated to support the hypothesis that SARS-CoV-2 arose naturally. The presence of highly similar or identical gene and protein sequences are common among organisms that are evolutionarily related to each other. Therefore, it is expected that members of the coronavirus family share similar or identical genetic or protein features. Scientific evidence supports the hypothesis that the virus arose naturally in wildlife before it crossed over to humans.

REVIEW Uncertainty surrounding the origin of the novel coronavirus has provided fertile ground for breeding conspiracy theories, some of which Health Feedback previously found to be inaccurate and unsubstantiated (see here and here). The recent claim by virologist Li-Meng Yan that the SARS-CoV-2 virus is manmade is the latest in a long series of conspiracy theories stretching back to the beginning of the coronavirus pandemic.

On 14 September 2020, Yan and her colleagues published a preprint on the online repository Zenodo claiming that the SARS-CoV-2 virus is a product of genetic engineering. A preprint is a research paper that has not been peer-reviewed by other scientists yet. Experts who examined the preprint found it was highly flawed and provided no supporting evidence for their claims, as detailed in this Health Feedback review.

Yan et al. published a second preprint on 8 October 2020 claiming that the virus is an unrestricted bioweapon and alleging that the genetic sequences of ten other coronaviruses are fabricated and do not exist in nature. Contrary to this claim, these ten coronaviruses, including RaTG13which is the closest known relative to SARS-CoV-2 and has about 96% genome sequence identity to SARS-CoV-2[1]and some pangolin coronaviruses, were analyzed by other scientists and found to support the natural origin hypothesis for SARS-CoV-2[2-7]. The second preprint from Yan et al. received more than 130,000 views on Zenodo since it was published, and was promoted by outlets known for publishing misinformation, such as Zero Hedge and National Pulse.

The alleged motivation for fabricating genetic sequences is related to one of the primary claims by Yan et al., specifically that the bat coronaviruses ZC45 and ZXC21 provided the genetic backbone for SARS-CoV-2. In support of this claim, Yan et al. point to the 100% identity in the envelope (E) protein sequence that exists between these three viruses. The E protein is a small protein on the surface of the membrane that encloses the viral genome and is important for producing virus particles that can efficiently infect cells[8].

Firstly, the claim that the bat coronaviruses ZC45 and ZXC21 provided the genetic backbone to artificially create SARS-CoV-2 was presented in the first preprint by Yan et al. This claim was debunked by scientists, who pointed out that the genetic sequences of ZC45 and ZXC21 are very different to that of SARS-CoV-2. In fact, the virus ZC45 is only 89% related to SARS-CoV-2, said Stanley Perlman, a professor at the University of Iowa who studies coronaviruses, in this FactCheck.org article:

Perlman said it would be nearly impossible to make the reverse genetics system needed to manipulate the virus and changing its sequence to arrive at SARS-CoV-2 would be virtually impossible since it would not be known how to manipulate the virus.

Kristian Andersen, a professor at Scripps Research who studies the evolution of viruses including SARS-CoV-2, also pointed out the incongruency of the claim on Twitter: This simply cant be true there are more than 3,500 nucleotide differences between SARS-CoV-2 and these viruses.

Marvin Reitz, a virologist at the University of Maryland, put it more bluntly in his review of the first preprint: [I]t still would require more than 3,000 nucleotide substitutions [for ZC45] to become SARS-CoV-2. This is not even slightly credible; it beggars reason.

A response by scientists at the Johns Hopkins University Center for Health Security also provides a detailed rebuttal of the claims made by Yan et al. in their first preprint. It also highlights the implausible use of ZC45 and ZXC21 as the genetic backbone for SARS-CoV-2.

In short, ZC45 and ZXC21 are very different from SARS-CoV-2 in terms of genome identity. Altering a backbone from either of the two to transform it into the genome of SARS-CoV-2 would require a feat of genetic engineering that is extremely difficult, if not impossible, to accomplish with current technology.

Based on their spurious initial assumption that ZC45 and ZXC21 provided the genetic backbone for SARS-CoV-2, Yan et al. claim that the genetic sequences of RaTG13 and the other coronaviruses were fabricated to obscure the link between SARS-CoV-2 and ZC45/ZXC21, and that RaTG13 and the other coronaviruses do not exist. To support this claim, they point to the observation that all these viruses also have an E protein sequence that is 100% identical to that of ZC45 and ZXC21.

The argument by Yan et al. that the genetic sequences of some coronaviruses were fabricated to support the hypothesis that SARS-CoV-2 arose naturally does not hold up to scrutiny. In a Business Insider interview, Emma Hodcroft, a postdoctoral fellow at the University of Basel and co-developer of the Nextstrain project that studies the evolution of pathogens, including SARS-CoV-2, pointed out that most of the samples that Yans group says are fake predate the start of the pandemic. Hodcroft also explained:

This accusation implies there were years of coordination and fake sequence generation, Hodcroft said, adding: This is an incredible claim, and would require a significant evidence burden to back it up, which is missing from the paper.

Virologists have also analyzed the genome sequence of RaTG13 and found it to be authentic and supported by good-quality data.

Although some coronaviruses share certain identical genetic sequences with SARS-CoV-2, this is not evidence that the other coronaviruses were fabricated. Instead, similar or identical genetic and protein sequences of coronaviruses are evidence of their evolutionary relatedness, which is expected since these viruses all belong to the coronavirus family. Specifically, the E protein sequence of SARS-CoV-2, RaTG13, and the other coronaviruses analyzed in the preprint by Yan et al. are indeed identical to that of ZC45 and ZXC21, but this in itself does not indicate that the RaTG13 and the other coronaviruses were fabricated to mimic the E protein sequence of ZC45 and ZXC21.

Lastly, one feature of concern in both preprints by Yan and her co-authors is the listing of their affiliations as the Rule of Law Society and the Rule of Law Foundation. These two organizations have no prior experience in conducting biological research and are linked to Stephen Bannon and Wengui Guo, both of whom have published COVID-19 misinformation in the past.

Overall, the claims in the second preprint by Yan and her colleagues are as ill-founded as the claims made in their first preprint. Evidence supporting claims that the virus was engineered is lacking. In contrast, scientific analyses support the hypothesis that SARS-CoV-2 arose naturally in wildlife before crossing over to humans during a zoonotic infection (transmission of pathogens from animals/insects to humans). There are numerous examples of emerging zoonotic pathogens causing disease outbreaks throughout human history and across the world[9].

Original post:
No evidence that coronavirus genetic sequences were fabricated, contrary to preprint by Li-Meng Yan and colleagues - Health Feedback

Read More...

Nobel Prize in Chemistry 2020 Winners from a Patent Perspective – Lexology

Sunday, October 18th, 2020

Further to my recent article about the 2020 Nobel Prize announcements, the winners of the 2020 Chemistry Nobel Prize were announced as Emmanuelle Charpentier and Jennifer Doudna on 7 October 2020 "for the development of a method for genome editing". This, of course, refers to the revolutionary genetic scissors technology, CRISPR/Cas9. For this article, Ive teamed up with my colleague Jamie Atkins, whose specialisms include prosecution of CRISPR-related patent applications at the EPO, to get into the details of the winning technology.

In this article, we explore:

Diversity in the Chemistry Nobel Prize;

The winning technology;

What is CRISPR and how does it work?;

How can CRISPR be used?;

CRISPR Patents; and

The future could CRISPR be used to combat COVID -19?

Diversity in the Chemistry Nobel Prize

Before looking at the patent side of things, it is worth noting that this is the first Nobel Prize awarded to two women. Charpentier commented shortly after the prize announcement that:

My wish is that this will provide a positive message to the young girls who would like to follow the path of science, and to show them that women in science can also have an impact through the research that they are performing This is not just for women, but we see a clear lack of interest in following a scientific path, which is very worrying.

While there is still a significant gender gap in the Laureates of the Chemistry Nobel Prize, it is encouraging that there are now an additional two female winners to add to the previous five: Marie Curie (1911), Irne Joliot-Curie (1935), Dorothy Crowfoot Hodgkin (1964), Ada Yonath (2009) and Frances H. Arnold (2018). We hope this figure continues to increase each year, along with wider recognition of other under-represented groups, for example in terms of BAME and LGBTQ+ representation.

The winning technology

At face value, CRISPR/Cas9 (CRISPR) seems to be more biological than chemical, but this only serves to highlight the breadth of chemistry as a field. As described in Nobels will, the Nobel Prize in Chemistry is to be awarded to the person who shall have made the most important chemical discovery or improvement, and this requirement is surely met by CRISPR.

What is CRISPR and how does it work?

The CRISPR/Cas9 editing tool developed by the Nobel Prize winners is based on the discovery of a naturally occurring system used by bacteria to defend against viral infection. When a virus is detected, the bacteria produce short RNA sequences that guide a DNA cutting enzyme (Cas9) to viral DNA matching the RNA sequence. Cas9 cuts the viral DNA, thereby disabling the virus. Doudna and Charpentier made several important discoveries leading to a better understanding of this bacterial system, developed a simplified version of the system and crucially showed that it could be programmed to target almost any DNA sequence of interest, as reported in the seminal Jinek et al. 2012 paper:

''Our study reveals a family of endonucleases that use dual-RNAs for site-specific DNA cleavage and highlights the potential to exploit the system for RNAprogrammable genome editing.

The basic components of the CRISPR/Cas9 system are the DNA cutting enzyme (Cas9) and the guide RNA based on the target DNA sequence, which directs the nuclease to the desired cutting location. Once the DNA in a cell is cut, the cell tries to fix the break using its own repair mechanisms. Due to the error-prone nature of such mechanisms, this fix can actually disable a gene. Alternatively, by supplying a template sequence together with the CRISPR machinery, the cells DNA repair mechanisms can be exploited to replace a section of DNA with the template sequence of choice.

Building on this foundational work of the Nobel Laureates, many complementary genome editing tools based on the CRISPR principle have been developed. For example, in a technique known as base editing, a deactivated Cas9 is fused to a cytidine deaminase enzyme, which allows targeted conversion of the cytidine base (C) to thymine (T) without cleaving the DNA (Komor et al, 2016). Another example is prime editing, in which deactivated Cas9 is fused to reverse transcriptase. This is coupled with a guide RNA that specifies the target site and encodes a desired replacement sequence, allowing new genetic information to be written into a specified DNA target site (Anzalone et al, 2019).

How can CRISPR be used?

There are innumerable exciting possibilities that stem from the ability to edit the genome of any living cell in a targeted manner using these basic principles. For example, CRISPR is already revolutionising genetic research by providing a quicker and easier way for researchers to knock out specific genes in order to investigate the function of those genes and their role in cellular pathways. There also important applications in agriculture, where it is being used to speed up the generation of improved crop varieties and could play an important role in food security.

Another key application is in the filed of diagnostics (more on that below), but perhaps one of the most exciting and lucrative CRISPR applications is in the field of medicine. Doudna is a co-founder of Intellia and Charpentier is a co-founder of CRISPR Therapeutics, both of which are developing CRISPR-based therapies, some of which are already in early stage clinical trials, e.g. for the treatment of sickle cell anaemia. Many current trials involve editing the genome of cells extracted from the body (e.g. hematopoietic stem cells) before reinserting the modified cells back into the patient. An alternative approach also being explored is delivering the CRISPR machinery directly into the body, for example to disable faulty disease-causing genes. This year saw the first delivery of CRISPR machinery to a patient in an attempt to treat an inherited form of blindness called Leber congenital amaurosis 10 (LCA10).

CRISPR patents

Any new technology with such a great potential for commercial application is an ideal candidate for patent protection. The European Patent Office (EPO) has published 32 European patent applications naming Doudna as an inventor and 7 European patent applications naming Charpentier as an inventor. Patent applications are published 18 months after their effective filing date, so there may be many more unpublished patent applications that have already been filed naming these Laureates as inventors.

CRISPR patents have also been at the centre of attention in both Europe and the US over recent years. In Europe, weve seen the high-profile CRISPR priority appeal, in which one of the Broad Institutes fundamental CRISPR patents (EP2771468, claiming an earliest priority date of 12 December 2012) was revoked for lack of novelty over some of the seminal CRISPR papers. These papers became prior art because the patent was found not to be entitled to its claimed priority dates (see our articles here, here and here from earlier this year for the details). Doudna and Charpentiers patents have also come under attack in Europe; their 2013 patent EP2800811 was opposed by seven parties, and maintained in amended form in May 2020.

In the US, high profile interference proceedings between University of California and others and the Broad Institute and others (Broad) before the US Patent Trial and Appeal Board culminated in a decision in favour of Broad, which was upheld in 2018 by the Court of Appeals for the Federal Circuit. Further such proceedings are currently in progress.

Stay tuned for a more in-depth discussion of the ongoing challenges relating to CRISPR patents.

The future could CRISPR be used to combat COVID -19?

Shortly after the Nobel Prize was announced, Charpentier was asked whether CRISPR could be used to make a vaccine for COVID-19. She indicated this was unlikely in a direct way, but that it could be useful indirectly by allowing researchers to understand the virus in ways that help them develop a vaccine (e.g. understanding what is important for the virus to replicate). The full Q&A with Charpentier is available here.

It has in fact already been deployed in a fast and accurate diagnostic test for Covid-19. This test, referred to as SARS-CoV-2 DNA Endonuclease-Targeted CRISPR Trans Reporter (DETECTR), harnesses the targeting function of the guide RNAs of the CRISPR system to bind to coronavirus sequences, and the cutting function of Cas12 (a nuclease related to Cas9) to cleave a reporter molecule, to confirm detection of the virus. Fittingly, Doudnas own lab recently announced its own CRISPR-based diagnostic test that can detect SARS-CoV-2 in just 5 minutes. This high processing speed is achieved by avoiding the need to amplify the viral genome (as required by earlier assay formats). Instead, the new test uses combinations of CRISPR RNA which target different parts of the virus RNA and activate multiple Cas nucleases (Cas13a) per piece of viral RNA, boosting the fluorescent signal generated when a reporter molecule is cut. Moreover, the researchers showed that the fluorescence could be measured with a mobile phone camera, demonstrating the simplicity and portability of the assay.

As mentioned above, clinical trials involving CRISPR-based approaches are already underway, and we look forward to seeing more success stories in the coming years. While these are no doubt exciting times, it is clear that extreme caution must be exercised to fully understand and mitigate the risk of CRISPR acting off-target. There are also ethical debates to be had about how far to take gene editing. Should scientists be permitted to introduce heritable changes into the genome even if this can be done safely and efficiently?

One thing is for sure, the work conducted by Doudna and Charpentier has revolutionised the field of genetic engineering, and for that work these inspirational inventors should be celebrated.

Link:
Nobel Prize in Chemistry 2020 Winners from a Patent Perspective - Lexology

Read More...

We’ve learned much in this crisis including what we don’t know, by John McGauley – The Keene Sentinel

Sunday, October 18th, 2020

In the ninth month of the COVID scourge which covered the land, the people lamented their fate and looked to the heavens for relief and succor.

That is, I imagine, how the Bible might recount the pandemic weve been in since February, if the Good Book were being written today.

Around here, at least, weve been spared the worst from this thing. But if you recall when it all started, and for several months hence, the hospital geared up for scores of deathly ill patients and local doctors and nurses were told to expect reporting for emergency duty. A mobile refrigerated morgue was brought in. They even retrofitted parts of Keene State College to handle the overflow. Main Street downtown was bereft of traffic; for a couple of months, it was always 6 a.m. Sunday morning down there. Planes, trains and buses stopped; there were no pedestrians. There was a mad rush for PPE (we all learned what that acronym meant), and ventilators.

No one knows for sure where this pandemic is headed, and those up here in the northern climes are wondering if it will get worse as the winter winds drive us all inside until April and May. What will our Thanksgiving and Christmas holidays look like this year?

Assessing the past nine months, Ive had a number of observations, and you may have had similar thoughts.

For starters, I think that society came unglued. And, I think I know why. We collided with something that all of our sophisticated science and technology couldnt handle, and we freaked out. Our new Apple phones, PET scans, 5G networks, genetic engineering all the kings horses and all the kings men were helpless in the face of one of the worlds oldest adversaries: pestilence, which is one of the Four Horsemen of the Apocalypse in the Book of Revelation, to use another biblical reference. While astronauts aboard the International Space Station circle the earth, ensconced in the most amazing science, they look down upon an earth stymied and suffering from a primordial disease.

Secondly, we learned that government has severe limitations. Like our belief in technology, we have come to think in the past century that the state and feds can come up with solutions and come to our rescue. Thats just not true, and never was. You can blame whoever you want, but governments response was and is severely flawed. Its not a failure of the Republicans or Democrats or career bureaucrats at the NIH or CDC its just the fact that government cannot solve our problems when the you-know-what hits the fan. Never could. If youve ever been through the aftermath of a severe hurricane or a strong earthquake, you know that there comes a time when youre just on your own. When a crisis hits, you learn immediately that the emperor lives very far away.

Thirdly, most people behaved, but enough didnt that it showed the sour side of humanity. Panic buying without cause, denying others of necessities. A refusal to wear masks in public. We can sometimes be a stupid bunch.

Fourth, our economy is very fragile, despite being the richest country in the world. That showed itself right away. Our weakest link proved to be our distribution network for goods and services. One of the correct responses by the federal government was the rapid carpet-bombing of cash to companies and individuals. Well have to pay that big bill later, but it saved us from a collapsed system.

Fifth, we have, in fact, dodged the bullet. Despite the number of hospitalizations and deaths, COVID-19 could have been worse, with a much higher mortality rate. If we didnt already possess a sophisticated medical system, the death rate might have been off the charts.

Sixth, instant communication and 24/7 media coverage is half good, half terrible. It disseminates at the speed of light erroneous theories, rumors, dubious statistics, malevolent gossip, wild statements from wrong-headed people parading as scientists, shoddy studies and surveys, and inflammatory rhetoric from politicians and bureaucrats. Cant change that; information and misinformation spread at the same rate, and people believe weird things. Well see that when a vaccine is finally developed, and millions refuse to take it.

Lastly, if there is a silver lining to all this, its that well be prepared for the next disaster, or disease, that comes our way. That is, if we remember what we did right and what we did wrong this time. Unfortunately, humans have a propensity to reinvent the wheel over and over.

John McGauley, an author and local radio talk-show host, writes from Keene. He can be contacted at mcgauleyink@gmail.com

Excerpt from:
We've learned much in this crisis including what we don't know, by John McGauley - The Keene Sentinel

Read More...

Viewpoint: Greenpeace-funded study backfires, undermining case to treat gene-edited crops as GMOs – Genetic Literacy Project

Sunday, October 18th, 2020

The anti-GMO movement dominated the public discourse about crop biotechnology for decades. Led by committed activists who knew how to manipulate the media, they effectively steamrolled a scientific community that wasnt ready for the PR war that Greenpeace and other NGOs launched against frankenfoods in the mid-1990s. Thirty years later, were beginning to see that dynamic shift as plant breeding technology improves and experts successfully defend it against activist attacks.

In August, John Fagan, organic food champion, biologist and Raja of World Peace in the Maharishi organization, led a Greenpeace-funded study claiming that gene-edited crops developed with new breeding techniques (NBTs) like CRISPR can be detected.

This may seem unimpressive to most people, but the result is a big deal to anti-GMO activists like Fagan. Gene-edited crops may be essentially identical to conventionally bred plants; the only difference is that gene editing dramatically speeds up the breeding process, saving time, money and getting enhanced seeds into farmers fields much more quickly than was previously possibleall without inserting foreign DNA into the crops genome. This is the primary distinction between gene editing and transgenesis (GMO in the vernacular).

These facts aside, European law treats gene-edited and GMO crops the same. Commercial cultivation of both is effectively banned in the EU (farmers have access to only one transgenic corn variety), though political pressure is building to reform Europes strict regulations. Anti-biotech groups have been on a campaign to block these reforms since July 2018, just after the European Court of Justice first ruled on crop gene editing. Fagans paper was the latest contribution to this effort. If gene-edited and conventional crops can be distinguished, the argument goes, then the former should be regulated as GMOs.

But theres a problema big one in fact: Fagans study actually demonstrated that its not possible to detect most gene-edited plants, therefore destroying the EUs justification for regulating them as GMOs. Its the perfect example of what we fondly call an own goal. And it illustrates how anti-science groups flog disinformation with the help of gullible journalists who stenograph their questionable claims for wide distribution.

Given the onslaught of disinformation we face in a post-COVID world, Fagans paper offers us the perfect opportunity to review the activist playbook and immunize ourselves against bad science and its harmful consequences.

As gene editing becomes an increasingly effective tool for improving agricultural production and reducing its environmental impacts, many countries (the US, Canada, Brazil, Argentina among dozens more) have split from the European Union (EU) on NBTs, exempting them from the expensive and exhausting regulations that govern GMO crops.

This is not only scientifically sound, its pragmatic. There is no way to detect gene edits in most cases. These changes look just like natural mutations found in wild plants, or the genetic changes induced by old-fashioned and EU-approved practices like bathing seeds in mutagens or irradiating them, or changes that occur in plants produced via tissue culture. The tests at our disposal cannotI repeat, cannotdistinguish mutations caused by any of these techniques.

This creates a problem for Food Purity Rajas and other opponents of biotechnology. If these gene-edited crops gain public acceptance and dont count as GMO, the organic industry will be at a competitive disadvantage. How could they justify a premium price for Non-GMO Project-certified corn flakes in those circumstances? Naturally, they have to challenge the efficacy and safety of gene editing to prevent such an outcome. Fear is their go-to currency in this effort, as John Fagan explained almost six years ago in a mailing list for anti-GMO campaigners, run by Claire Robinson of the activist website GM Watch. Its part of a long-term plan to make people fear all engineered food.

Fagan recently gave credit for Americas rejection of GMOs to transcendental meditation (TM), which caused a sharp increase in coherence in U.S. collective consciousness, when a large permanent group of TM practitioners was assembled in Iowa, USA. I havent seen the evidence for that, though Id be happy to take a look at the research if anyone can locate it. What is more likely, and supported by data, is that a long-term misinformation campaign made up of bad science and shock marketing scared parents everywhere into buying organic fruit snacks to avoid scary GMOs.

Down to the last detail, this tried and true activism strategy was deployed to influence the current discourse around gene editing. Besides his enthusiasm for TM, John Fagan also has training in molecular biology and operates a non-profit lab with the necessary testing capacity (incidentally, he also started a company that certifies products for the Non-GMO Project). Greenpeace, meanwhile, has an effective, stunt-based PR machine that can churn out multimedia presentations and widely read press releases, which sympathetic NGOs can dutifully amplify. But things didnt turn out as intended this time.

Its impossible to know which came first: the idea for a campaign to attack gene-edited crops, which needed supporting science, or a study in need of the PR muscle Greenpeace could leverage. Someday an intrepid investigative journalist might be able to work this out. But in any case, the outcome on September 7, 2020 was the release of a paper in the peer-reviewed journal Foods, which claimed to reveal a test that could uniquely and specifically detect the first commercial gene-edited crop, a variety of herbicide-resistant canola developed by the seed company Cibus. With a press release, media blitz, and slickly produced website, Greenpeace and other funders launched the #NowhereToHide campaign to promote Fagans paper and encourage EU regulators to treat this herbicide-tolerant canola as a GMO.

Similar to the guy who claimed he invented email, Fagans team implied they developed a new test to identify this crop. Thats not the case; the qPCR (polymerase chain reaction) method used in the study is well established for canola. Fagans novelty claim is therefore quite erroneous, as one scientist noted on Twitter. Nobody disputes that you can find point mutations, changes to a single DNA base pair, with qPCR. The key is that its impossible to determine if a variation is naturally occurring or purposefully induced. Many experts have pointed this out in response to the paper. The test would likewise detect herbicide-resistant plants that have been known to scientists and regulators since in 2002, from wild populations with that same mutation. Etienne Bucher, a plant geneticist based in Switzerland, tried to help Greenpeace grasp this:

But heres the kicker: this canola is not gene edited. It is a somaclonal mutation that was found in the screenings for an herbicide-tolerant variety, one of those changes that occurs in tissue culture. So what Fagans team has, in fact, definitively proved: they cannot detect edited canola this way. Additionally, it appears this rapeseed could be classified as non-GMO in Europe, since it was developed with one of the grandfathered techniques not subject to the onerous EU GMO approval process.

Own. Goal. Reminds me of the time anti-vaxxers commissioned research that confirmed vaccines do not cause autism.

Let the goal-post moving commence!

After the scientific community made quick work of Fagans study, Greenpeace and activists like Claire Robinson at GMWatch began furiously backpedaling. Maharishi TM trainer and geneticist Michael Antoniou told the anti-GMO website that the method they have developed reliably detects a single DNA base unit change, regardless of how it came about. But he went on to assert that EU regulators should still rely on this test to detect the canola variety. This makes absolutely no sense, as the German Central Committee for Biosafety experts observed [automated Google translation]:

The publication by Chhalliyil et al does not add any new knowledge to the current state of science and technology. Rather, it proves that it is not possible to distinguish genome-edited plants from plants with spontaneously occurring mutations. Without prior knowledge of the manufacturing process [my emphasis], no statement can be made as to whether or not it is a GMO within the meaning of the ECJ [European Court of Justice] ruling.

GM Watch agreed, though it fell back on a legal argument to excuse the studys weakness:

What the test cannot do is detect the technique by which a mutation was brought about but under EU law it doesnt need to. The way that the law deals with proof of origin for all GMOs products of gene editing included is to require the developer to declare that their product is a GMO and provide a test method and reference material.

In other words, if Cibus tells regulators its canola is gene edited, then regulators can determine if the canola is gene edited.

The media blitz around Fagans study was designed to promote organic food, but ironically enough, all this talk about identifying the source of mutations dredged up a potentially serious problem for the organic industry. In 2014, the USDAs National Organic Standards Board investigated what kinds of genetic modifications led to many of the key organic crops, only to realize how difficult it would be to classify breeding and laboratory mutations:

Exploring this issue has brought to the attention of the subcommittee that engineered genetic manipulation of plant breeding materials has already occurred in many of the crop varieties that are currently being used in organic farming. A partial list:

Many of these techniques that were used in initial crosses that have now passed down through many generations may not be traceable any longer

The board realized that many of the crops in organic production right now were the result of laboratory processes (genetic engineering, one could say) that are undetectable with molecular testing. Why is this significant? Well, a cynical scientist could use Fagans test to detect mutations in organic tangerines just as well as Cibus canola, demonstrating the inanity of labeling the tangerine non-GMO and the canola genetically engineered.

Bungled though it was, this parallel science PR stunt helpfully illustrated how disinformation can sow confusion and lead to nonsensical policy. For example, Greenpeace celebrated when an Austrian health minister declared that Fagans test should be used to enforce the EUs GMO rules. Well-known anti-crop biotech German politicians also eagerly embraced the results of the study. If people with so much influence over food safety rules in their countries can be fooled, you can see why junk science poses the risk it does.

Still, Greenpeace clearly lost this round. The activist-media juggernaut went down in flames before it could do too much damageand GM Watch spent most of September explaining away Fagans study in the face of intense expert scrutiny. Expect the anti-science crusaders to fall back on these same tactics in the future, because its all they know how to do. But look forward to the fact that there are now scientists, battle hardened by years in the social media trenches, ready to blow air horns the next time an NGO launches a scheme like this.

Mary Mangan holds a PhD in cell, molecular, and developmental biology from the University of Rochester. She co-founded OpenHelix, a company that provides awareness and training on open source genomics software tools. Follow her on Twitter @mem_somerville

See original here:
Viewpoint: Greenpeace-funded study backfires, undermining case to treat gene-edited crops as GMOs - Genetic Literacy Project

Read More...

CAR T-cell Therapies for the Treatment of Patients with Acute Lymphoblastic Leukemia – OncoZine

Sunday, October 18th, 2020

Acute Lymphoblastic Leukemia (AML), also called acute myeloblastic leukemia, acute myelogenous leukemia, acute myeloid leukemia, or acute nonlymphocytic leukemia, is an aggressive, fast-growing, heterogenous group of blood cancers that arise as a result of clonal expansion of myeloid hematopoietic precursors in the bone marrow. Not only are circulating leukemia (blast) cells seen in the peripheral blood, but granulocytopenia, anemia, and thrombocytopenia are also common as proliferating leukemia cells interfere with normal hematopoiesis.

Approximately 40-45% of younger and 10-20% of older adults diagnosed with AML are cured with current standard chemotherapy. However, the outlook for patients with relapsed and/or refractory disease is gloomy. Relapse following conventional chemotherapy remains is a major cause of death.

The process of manufacturing chimeric antigen receptor (CAR) T-cell therapies. [1] T-cells (represented by objects labeled as t) are removed from the patients blood. [2] Then in a lab setting the gene that encodes for the specific antigen receptors is incorporated into the T-cells. [3] Thus producing the CAR receptors (labeled as c) on the surface of the cells. [4] The newly modified T-cells are then further harvested and grown in the lab. [5]. After a certain time period, the engineered T-cells are infused back into the patient. This file is licensed by Reyasingh56 under the Creative Commons Attribution-Share Alike 4.0 International license.Today, the only curative treatment option for patients with AML is allogeneic hematopoietic stem cell transplantation or allo-HSCT, which through its graft-vs.-leukemia effects has the ability to eliminate residual leukemia cells. But it is an ption for only a minority. And despite a long history of success, relapse following allo-HSCT is still a major challenge and is associated with poor prognosis.

In recent years, rresearchers learned a lot about the genomic and epigenomic landscapes of AML. This understanding has paved the way for rational drug development as new drugable targets, resulting in treatments including the antibody-drug conjugate (ADC) gemtuzumab ozogamycin (Mylotarg; Pfizer/Wyeth-Ayerst Laboratories).

CAR T-cell TherapiesChimeric antigen receptor (CAR) T-cells therapies, using a patients own genetically modified T-cells to find and kill cancer, are one of the most exciting recent developments in cancer research and treatment.

Traditional CAR T-cell therapies are an autologous, highly personalised, approach in which T-cells are collected from the patient by leukopheresis and engineered in the laboratory to express a receptor directed at a cancer antigen such as CD19. The cells are then infused back into the patient after administration of a lymphodepletion regimen, most commonly a combination of fludarabine and cyclophosphamide. Durable remissions have been observed in pediatric patients with B-ALL and adults with NHL.

CD19-targeted CAR T-cell therapies, have, over the last decade, yielded remarkable clinical success in certain types of B-cell malignancies, and researchers have made substantial efforts aimed at translating this success to myeloid malignancies.

While complete ablation of CD19-expressing B cells, both cancerous and healthy, is clinically tolerated, the primary challenge limiting the use of CAR T-cells in myeloid malignancies is the absence of a dispensable antigen, as myeloid antigens are often co-expressed on normal hematopoietic stem/progenitor cells (HSPCs), depletion of which would lead to intolerable myeloablation.

A different approachBecause autologous CAR T-cell therapies are patient-specific, each treatment can only be used for that one patient. Furthermore, because CAR T-cells are derived from a single disease-specific antibody, they are, by design, only recognized by one specific antigen. As a consequence, only a small subset of patients with any given cancer may be suited for the treatment.

This specificity means that following leukopheresis, a lot of work needs to be done to create this hyper personalised treatment option, resulting in 3 5 weeks of manufacturing time.

The manufacturing process of CAR T-cell therapies, from a single academic center to a large-scale multi-site manufacturing center further creates challenges. Scaling out production means developing processes consistent across many collection, manufacturing, and treatment sites. This complexity results in a the realitively high cost of currently available CAR T-cell therapies.

To solve some of the concerns with currently available CAR T-cell therapies, researchers are investigating the option to develop allogenic, off-the-shelf Universal CAR T-cell (UCARTs) treatments that can be mass manufactured and be used for multiple patients.

Allogeneic CAR T-cell therapy are generally created from T-cells from healthy donors, not patients. Similar to the autologous approach, donor-derived cells are shipped to a manufacturing facility to be genetically engineered to express the antibody or CAR, however, in contrast to autologous CAR T-cells, allogeneic CAR T-cells are also engineered with an additional technology used to limit the potential for a graft versus host reaction when administered to patients different from the donor.

One unique benefit ofn this approach is that because these therapies hey are premade and available for infusion, there is no requirement to leukopheresis or a need to wait for the CAR T-cells to be manufactured. This strategy also will benefit patients who are cytopenic (which is not an uncommon scenario for leukemia patients) and from whom autologous T-cell collection is not possible.

PioneersAmong the pioneers of developing allogeneic CAR-T therapies are companies including Celyad Oncology, Cellectis, Allogene Therapeutics, and researchers at University of California, Los Angeles (UCLA) in colaboration with Kite/Gilead.

Researchers at UCLA were, for example, able to turn pluripotent stem cells into T-cells through structures called artificial thymic organoids. These organoids mimic the thymus, the organ where T-cells are made from blood stem cells in the body.

Celyad OncologyBelgium-based Celyad Oncology is advancing a number of both autologous and allogeneic CAR T-cell therapies, including proprietary, non-gene edited allogeneic CAR T-cell candidates underpinned by the companys shRNA technology platform. The shRNA platform coupled with Celyads all-in-one vector approach provides flexibility, versatility, and efficiency to the design of novel, off-the-shelf CAR T-cell candidates through a single step engineering process.

In July 2020, the company announced the start of Phase I trials with CYAD-211, Celyads first-in-class short hairpin RNA (shRNA)-based allogeneic CAR T candidate and second non-gene edited off-the-shelf program. CYAD-211 targets B-cell maturation antigen (BCMA) for the treatment of relapsed/refractory multiple myeloma and is engineered to co-express a BCMA-targeting chimeric antigen receptor and a single shRNA, which interferes with the expression of the CD3 component of the T-cell receptor (TCR) complex.

During the 2020 American Society of Clinical Oncology (ASCO) Virtual Scientific Program in May 2020, the company presented updates from its allogeneic programs, including additional data from the alloSHRINK study, an open-label, dose-escalation Phase I trial assessing the safety and clinical activity of three consecutive administrations of CYAD-101, an investigational, non-gene edited, allogeneic CAR T-cell candidate engineered to co-express a chimeric antigen receptor based on NKG2D (a receptor expressed on natural killer (NK) cells that binds to eight stress-induced ligands and the novel inhibitory peptide TIM TCR Inhibitory Molecule), for the treatment of metastatic colorectal cancer (mCRC).

The expression of TIM reduces signalling of the TCR complex, which is responsible for graft-versus host disease.every two weeks administered concurrently with FOLFOX (combination of 5-fluorouracil, leucovorin and oxaliplatin) in patients with refractory metastatic colorectal cancer (mCRC).

The safety and clinical activity data from the alloSHRINK trial in patients with mCRC demonstrated CYAD-101s differentiated profile as an allogeneic CAR T-cell candidate. Furthermore, the absence of clinical evidence of graft-versus-host-disease (GvHD) for CYAD-101 confirms the potential of non-gene edited approaches for the development of allogeneic CAR-T candidates.

Interim data from the alloSHRINK trial showed encouraging anti-tumor activity, with two patients achieving a confirmed partial response (cPR) according to RECIST 1.1 criteria, including one patient with a KRAS-mutation, the most common oncogenic alteration found in all human cancers. In addition, nine patients achieved stable disease (SD), with seven patients demonstrating disease stabilization lasting more than or equal to three months of duration.

Based on these results, clinical trials were broadened to include evaluating CYAD-101 following FOLFIRI (combination of 5-fluorouracil, leucovorin and irinotecan) preconditioning chemotherapy in refractory mCRC patients, at the recommended dose of one billion cells per infusion as an expansion cohort of the alloSHRINK trial. Enrollment in the expansion cohort of the trial is expected to begin during the fourth quarter of 2020.

CellectisCellectis is developping a universal CAR T-cell (UCART) platform in an attempy to create off-the-shelf CAR T-cell therapies. The companys pipeline includes UCART123, a CAR T-cell therapy designed to targets CD123+ leukemic cells in acute myeloid leukemia (AML). The investigational agent is being studied in two open-label Phase I trials: AML123 studying the therapys safety and efficacy in an estimated 156 AML patients, and ABC123 studying the therapys safety and activity in an estimated 72 patients with blastic plasmacytoid dendritic cell neoplasm (BPDCN).

UCART22Another investigational agent in clinical trials is UCART22 which is designed to treat both CD22+ B-cell acute lymphoblastic leukemia (B-ALL) and CD22+ B-cell non-Hodgkin lymphoma (NHL). Cellectis reported that UCART22 is included in an open-label, dose-escalating Phase I trial to study its safety and activity in relapsed or refractory CD22+ B-ALL patients.

UCART22 harbors a surface expression of an anti-CD22 CAR (CD22 scFv-41BB-CD3z) and the RQR8 ligand, a safety feature rendering the T-cells sensitive to the antibody rituximab. Further, to reduce the potential for alloreactivity, the cell surface expression of the T-cell receptor is abrogated through the inactivation of the TCR constant (TRAC) gene using Cellectis TALEN gene-editing technology.[1]

Preclinical data supporting the development of UCART22 was presented by Marina Konopleva, M.D., Ph.D. and her vteam during the 2017 annual meeting of the American Society of Hematology (ASH) meeting. [1]

Cellectis is also developing UCARTCS1 which is developed to treat CS1-expressing hematologic malignancies, such as multiple myeloma (MM). UCARTCLL1 is in preclinical development for treating CLL1-expressing hematologic malignancies, such as AML.

Cellectis and Allogene Therapeutics, another biotech company involved in the developmen t of CAR T-cell therapies, are developing ALLO-501, another CAR T-cell therapy which targets CD19 and is being developed for the the treatment of patients with relapsed or refractory NHL. Allogene Therapeutics is also developing ALLO-715, an investigational CAR T-cell therapy targeting the B-cell maturation antigen (BCMA) for treating relapsed or refractory multiple myeloma and ALLO-819, which targets CD135 (also called FLT3), for treating relapsed or refractory AML.

Allogene, in collaboration with both Cellectis, Pfizer (which has a 25% stake in Allogene) and Servier have numerous active open-label, single-arm Phase I trials for an off-the-shelf allogeneic CAR-T therapy UCART19* in patients with relapsed or refractory CD19+ B-ALL. Participating patients receive lymphodepletion with fludarabine and cyclophosphamide with alemtuzumab, followed by UCART19 infusion. Adults patients with R/R B-ALL are eligible.

The PALL aims to evaluate the safety and feasibility of UCART19 to induce molecular remission in pediatric patients with relapsed or refractory CD19-positive B-cell acute lymphoblastic leukemia (B-ALL) in 18 pediatric patients.

The CALM trial is a dose-escalating study evaluating the therapys safety and tolerability in 40 adult patients; and a long-term safety and efficacy follow-up study in 200 patients with advanced lymphoid malignancies.

Allogene reported preliminary proof-of-concept results during the annual meeting of the American Society of Hematology (ASH) in December 2018.

Data from the first 21 patients from both the PALL (n=7) and CALM (n=14) Phase I studies were pooled. The median age of the participating patients was 22 years (range, 0.8-62 years) and the median number of prior therapies was 4 (range, 1-6). Sixty-two percent of the patients (13/21) had a prior allogeneic stem cell transplant.

Of the 17 patients who received treatment with UCART19 and who received lymphodepletion with fludarabine, cyclophosphamide and alemtuzumab, an anti-CD52 monoclonal antibody, 14 patients (82%) achieved CR/CRi, and 59% of them (10/17) achieved MRD-negative remission.

In stark contrast, the four patients who only received UCART19 and fludarabine and cyclophosphamide without alemtuzumab did not see a response and minimal UCART19 expansion.

Based on these results, researchers noted that apparent importance of an anti-CD52 antibody for the efficacy of allogeneic CAR-T therapies. In addition, safety data also looked promising. The trial results did not include grade 3 or 4 neurotoxicity and only 2 cases of grade 1 graft-versus-host disease (10%), 3 cases of grade 3 or 4 cytokine release syndrome which were considered manageable (14%), 5 cases of grade 3 or 4 viral infections (24%), and 6 cases of grade 4 prolonged cytopenia (29%).

Precision BiosciencesPrecision Biosciences is developing PBCAR0191, an off-the-shelf investigational allogeneic CAR T-cell candidate targeting CD19. The drug candidate is being investigated in a Phase I/IIa multicenter, nonrandomized, open-label, parallel assignment, dose-escalation, and dose-expansion study for the treatment of patients with relapsed or refractory (R/R) non-Hodgkin lymphoma (NHL) or R/R B-cell precursor acute lymphoblastic leukemia (B-ALL).

The NHL cohort includes patients with mantle cell lymphoma (MCL), an aggressive subtype of NHL, for which Precision has received both Orphan Drug and Fast Track Designations from the U.S. Food and Drug Administration (FDA).

A clinical trial with PBCAR0191 Precision Biosciences is exploring some novel lymphodepletion strategies in addition to fludarabine and cyclophosphamide. Patients with R/R ALL, R/R CLL, R/R Richter transformation, and R/R NHL are eligible. Patients with MRD+ B-ALL are eligible as well. This trial is enrolling patients.

In late September 2020, Precision BioSciences, a clinical stage biotechnology amd Servier, announced the companies have added two additional hematological cancer targets beyond CD19 and two solid tumor targets to its CAR T-cell development and commercial license agreement.

PBCAR20APBCAR20A is an investigational allogeneic anti-CD20 CAR T-cell therapy being developed by Precision Biosciences for the treartment of patients with relapsed/refractory (R/R) non-Hodgkin lymphoma (NHL) and patients with R/R chronic lymphocytic leukemia (CLL) or R/R small lymphocytic lymphoma (SLL). The NHL cohort will include patients with mantle cell lymphoma (MCL), an aggressive subtype of NHL, for which Precision BioSciences has received orphan drug designation from the United States Food and Drug Administration (FDA).

PBCAR20A is being evaluated in a Phase I/IIa multicenter, nonrandomized, open-label, dose-escalation and dose-expansion clinical trial in adult NHL and CLL/SLL patients. The trial will be conducted at multiple U.S. sites.

PBCAR269APrecision Biosciences is, in collaboration with Springworks Therapeutics, also developing PBCAR269A, an allogeneic BCMA-targeted CAR T-cell therapy candidate being evaluated for the safety and preliminary clinical activity in a Phase I/IIa multicenter, nonrandomized, open-label, parallel assignment, single-dose, dose-escalation, and dose-expansion study of adults with relapsed or refractory multiple myeloma. In this trial, the starting dose of PBCAR269A is 6 x 105 CAR T cells/kg body weight with subsequent cohorts receiving escalating doses to a maximum dose of 6 x 106 CAR T cells/kg body weight.

PBCAR269A is Precision Biosciencess third CAR T-cell candidate to advance to the clinic and is part of a pipeline of cell-phenotype optimized allogeneic CAR T-cell therapies derived from healthy donors and then modified via a simultaneous TCR knock-out and CAR T-cell knock-in step with the =companys proprietary ARCUS genome editing technology.

The FDA recently granted Fast Track Designation to PBCAR269A for the treatment of relapsed or refractory multiple myeloma for which the FDA previously granted Orphan Drug Designation.

TCR2 TherapeuticsTCR2 Therapeutics is developing a proprietary TRuC (TCR Fusion Construct) T-cells designed to harness the natural T cell receptor complex to recognize and kill cancer cells using the full power of T-cell signaling pathways independent of the human leukocyte antigen (HLA).

While succesful in hematological malignancies, CAR T-cells therapies have generally struggled to show efficacy against solid tumors. Researchers at TCR2 Therapeutics believe this is is caused by the fact that CAR T-cell therapies only utilize a single TCR subunit, and, as a result, do not benefit from all of the activation and regulatory elements of the natural TCR complex. By engineering TCR T-cells, which are designed to utilize the complete TCR, they have demonstrated clinical activity in solid tumors. However, this approach has also shown major limitations. TCR T-cells require tumors to express HLA to bind tumor antigens. HLA is often downregulated in cancers, preventing T-cell detection. In addition, each specific TCR-T cell therapy can only be used in patients with one of several specific HLA subtypes, limiting universal applicability of this approach and increasing the time and cost of patient enrollment in clinical trials.

In an attempt to solve this problem, researchers at TCR2 Therapeutics have developped a proprieatarry TRuC-T Cells which are designed to incorporate the best features of CAR-T and TCR-T cell therapies and overcome the limitations. The TRuC platform is a novel T cell therapy platform, which uses the complete TCR complex without the need for HLA matching.

By conjugating the tumor antigen binder to the TCR complex, the TRuC construct recognizes highly expressed surface antigens on tumor cells without the need for HLA and engage the complete TCR machinery to drive the totality of T-cell functions required for potent, modulated and durable tumor killing.

In preclinical studies, TCR2 Therapeutics TRuC T-cells technology has demonstrated superior anti-tumor activity in vivo compared to CAR T-cells therapies, while, at the same time, releasing lower levels of cytokines. These data are encouraging for the treatment of solid tumors where CAR T-cells have not shown significant clinical activity due to very short persistence and for hematologic tumors where a high incidence of severe cytokine release syndrome remains a major concern.

TCR2 Therapeutics product candidates include TC-210 and TC-110.

TC-210 is designed to targets mesothelin-positive solid tumors. While its expression in normal tissues is low, mesothelin is highly expressed in many solid tumors. Mesothelin overexpression has also been correlated with poorer prognosis in certain cancer types and plays a role in tumorigenesis. TC-210 is being developed for the treatment of non-small cell lung cancer, ovarian cancer, malignant pleural/peritoneal mesothelioma and cholangiocarcinoma.

The companys TRuC-T cell targeting CD19-positive B-cell hematological malignancies, TC-110, is being developed to improve upon and address the unmet needs of current CD19-directed CAR T-cell therapies. The clinical development TC-110 focus on the treatment of adult acute lymphoblastic leukemia (ALL), diffuse large B-cell lymphoma (DLBCL) and follicular lymphoma (FL). Preclinical data demonstrates that TC-110 is superior to CD19-CAR-T cells (carrying either 4-1BB or CD28 co-stimulatory domains) both in anti-tumor activity as well as the level of cytokine release which may translate into lower rates of adverse events. The development of TC-110 starts with autologous T-cells collection by leukopheresis. These T-cells undergo genetic engineering to create TRuC-T cells targeting CD19.

This strategy combines the best features of CAR T-cells and the native T-cell receptor. It is open for R/R NHL and R/R B-ALL.

AUTO1Auto1 is an autologous CD19 CAR T-cell investigational therapyis being developped by Autolus Therapeutics. The investigational drug uses a single-chain variable fragment (scFv) called CAT with a lower affinity for CD19 and a faster off-rate compared to the FMC63 scFv used in other approved CD19 CAR T-cell therapies. The investigational therapy is designed to overcome the limitations in safety while maintaining similar levels of efficacy compared to current CD19 CAR T-cell therapies.

Designed to have a fast target binding off-rate to minimize excessive activation of the programmed T-cells, AUTO1 may reduce toxicity and be less prone to T-cell exhaustion, which could enhance persistence and improve the T-cells abilities to engage in serial killing of target cancer cells.

In 2018, Autolus signed a license agreement UCL Business plc (UCLB), the technology-transfer company of UCL, to develop and commercialize AUTO1 for the treatment of B cell malignancies. AUTO1 is currently being evaluated in two Phase I studies, one in pediatric ALL and one in adult ALL.

CARPALL trialInitial results from the ongoing Phase I CARPALL trial of AUTO1 were presented during European Hematology Association 1st European CAR T Cell Meeting held in Paris, France, February 14-16, 2019.

Enrolled patients had a median age of 9 years with a median of 4 lines of prior treatment. Seventeen patients were enrolled, and 14 patients received an infusion of CAR T cells. Ten of 14 patients had relapsed post allogeneic stem cell transplant. Eight patients were treated in second relapse, 5 in > second relapse and 3 had relapsed after prior blinatumomab or inotuzumab therapy. Two patients had ongoing CNS disease at enrollment.

This data confirmed that AUTO1 did not induces severe cytokine release syndrome (CRS) (Grade 3-5). Nine patients experienced Grade 1 CRS, and 4 patients experienced Grade 2 CRS. No patients required tociluzumab or steroids. As previously reported, one patient experienced Grade 4 neurotoxicity; there were no other reports of severe neurotoxicity (Grade 3-5). The mean cumulative exposure to AUTO1 CAR T-cells in the first 28 days as assessed by AUC was 1,721,355 copies/g DNA. Eleven patients experienced cytopenia that was not resolved by day 28 or recurring after day 28: 3 patients Grades 1-3 and 8 patients Grade 4. Two patients developed significant infections, and 1 patient died from sepsis while in molecular complete response (CR).

With a single dose of CAR T cells at 1 million cells/kg dose, 12/14 (86%) achieved molecular CR. Five patients relapsed with CD19 negative disease. Event free survival (EFS) based on morphological relapse was 67% (CI 34-86%) and 46% (CI 16-72%) and overall survival (OS) was 84% (CI 50-96%) and 63% (CI 27-85%) at 6 and 12 months, respectively.

CAR T cell expansion was observed in all responding patients (N=12), with CAR T cells comprising up to 84% of circulating T cells at the point of maximal expansion. The median persistence of CAR T-cells was 215 days.

The median duration of remission in responding patients was 7.3 months with a median follow-up of 14 months. Five of 14 patients (37%) remain in CR with ongoing persistence of CAR T-cells and associated B cell aplasia.

Fate TherapeuticsFT819 is an off-the-shelf CAR T-cell therapy targeting CD19 being developed by Fate Therapeutics. The T-cells are derived from a clonal engineered master induced pluripotent stem cell line (iPSCs) with a novel 1XX CAR targeting CD19 inserted into the T-cell receptor alpha constant (TRAC) locus and edited for elimination of T-cell receptor (TCR) expression.

Patients participating in the companys clinbical trial will receive lymphodepletion with fludarabine and cyclophosphamide. Some patients will also receive IL-2. Patients with R/R ALL, R/R CLL, R/R Richter transformation, and R/R NHL are eligible. Patients with MRD+ B-ALL are eligible as well.

At the Annual Meeting of the American Societ of Hematology held in December 2019, researchers from Fate Therapeutics presented new in vivo preclinical data demonstrating that FT819 exhibits durable tumor control and extended survival. In a stringent xenograft model of disseminated lymphoblastic leukemia, FT819 demonstrated enhanced tumor clearance and control of leukemia as compared to primary CAR19 T-cells. At Day 35 following administration, a bone marrow assessment showed that FT819 persisted and continued to demonstrate tumor clearance, whereas primary CAR T cells, while persisting, were not able to control tumor growth. [2]

CAR-NK CD19Allogeneic cord blood-derived Natural Killer (NK) cells are another off-the-shelf product that does not require the collection of cells from each patient.

Unlike T-cells, NK-cells do not cause GVHD and can be given safely in the allogeneic setting. At MD Anderson Cancer Center, Katy Rezvani, M.D., Ph.D, Professor, Stem Cell Transplantation and Cellular Therapy, and her team broadly focuses their research on the role of natural killer (NK) cells in mediating protection against hematologic malignancies and solid tumors and strategies to enhance killing function against various cancer.

As part of their research, the team has developed a novel cord blood-derived NK-CAR product that expresses a CAR against CD19; ectopically produces IL-15 to support NK-cell proliferation and persistence in vivo; and expresses a suicide gene, inducible caspase 9, to address any potential safety concerns.

In this phase I and II trial researchers administered HLA-mismatched anti-CD19 CAR-NK cells derived from cord blood to 11 patients with relapsed or refractory CD19-positive cancers (non-Hodgkins lymphoma or chronic lymphocytic leukemia [CLL]). NK cells were transduced with a retroviral vector expressing genes that encode anti-CD19 CAR, interleukin-15, and inducible caspase 9 as a safety switch. The cells were expanded ex vivo and administered in a single infusion at one of three doses (1105, 1106, or 1107 CAR-NK cells per kilogram of body weight) after lymphodepleting chemotherapy. The preliminarry resilts of the trials confirmed that administration of CAR-NK cells was not associated with the development of cytokine release syndrome, neurotoxicity, or graft-versus-host disease, and there was no increase in the levels of inflammatory cytokines, including interleukin-6, over baseline.

The study results also demonstrated that of the 11 patients who were treated, 8 patients (73%) had a response. Of these patients, 7 (4 with lymphoma and 3 with CLL) had a complete remission ICR), and 1 had remission of the Richters transformation component but had persistent CLL. Noteworthy was that responses were rapid and seen within 30 days after infusion at all dose levels. The infused CAR-NK cells expanded and persisted at low levels for at least 12 months. The researchers also noted that a majority of the 11 participating patients with relapsed or refractory CD19-positive cancers had a response to treatment with CAR-NK cells without the development of major toxic effects.[3]

Note* Servier will hold ex-US commercial rights. Servier is the sponsor of the UCART19 trials.

Clinical trialsalloSHRINK Standard cHemotherapy Regimen and Immunotherapy With Allogeneic NKG2D-based CYAD-101 Chimeric Antigen Receptor T-cells NCT03692429Study Evaluating Safety and Efficacy of UCART123 in Patients With Relapsed/ Refractory Acute Myeloid Leukemia (AMELI-01) NCT03190278Study to Evaluate the Safety and Clinical Activity of UCART123 in Patients With BPDCN (ABC123) NCT03203369Study of UCART19 in Pediatric Patients With Relapsed/Refractory B Acute Lymphoblastic Leukemia (PALL) NCT02808442Dose Escalation Study of UCART19 in Adult Patients With Relapsed / Refractory B-cell Acute Lymphoblastic Leukaemia (CALM) NCT02746952Dose-escalation Study of Safety of PBCAR0191 in Patients With r/r NHL and r/r B-cell ALL NCT03666000.Dose-escalation Study of Safety of PBCAR20A in Subjects With r/r NHL or r/r CLL/SLL NCT04030195A Dose-escalation Study to Evaluate the Safety and Clinical Activity of PBCAR269A in Study Participants With Relapsed/Refractory Multiple Myeloma NCT04171843TC-110 T Cells in Adults With Relapsed or Refractory Non-Hodgkin Lymphoma or Acute Lymphoblastic Leukemia NCT04323657Phase 1/2 Trial of TC-210 T Cells in Patients With Advanced Mesothelin-Expressing Cancer NCT03907852CARPALL: Immunotherapy With CD19 CAR T-cells for CD19+ Haematological Malignancies NCT02443831Umbilical & Cord Blood (CB) Derived CAR-Engineered NK Cells for B Lymphoid Malignancies NCT03056339

Reference[1] Petti F. Broadening the Applicability of CAR-T Immunotherapy to Treat the Untreatable. OncoZine. October 24, 2019 [Article][2] Wells J, Cai T, Schiffer-Manniou C, Filipe S, Gouble A, Galetto R, Jain N, Jabbour EJ, Smith J, Konopleva M. Pre-Clinical Activity of Allogeneic Anti-CD22 CAR-T Cells for the Treatment of B-Cell Acute Lymphoblastic Leukemia Blood (2017) 130 (Supplement 1): 808. https://doi.org/10.1182/blood.V130.Suppl_1.808.808%5B3%5D Chang C, Van Der Stegen S, Mili M, Clarke R, Lai YS, Witty A, Lindenbergh P, Yang BH, et al. FT819: Translation of Off-the-Shelf TCR-Less Trac-1XX CAR-T Cells in Support of First-of-Kind Phase I Clinical Trial. Blood (2019) 134 (Supplement_1): 4434.https://doi.org/10.1182/blood-2019-130584%5B4%5D Liu E, Marin D, Banerjee P, Macapinlac HA, Thompson P, Basar R, Nassif Kerbauy L, Overman B, Thall P, Kaplan M, Nandivada V, Kaur I, Nunez Cortes A, Cao K, Daher M, Hosing C, Cohen EN, Kebriaei P, Mehta R, Neelapu S, Nieto Y, Wang M, Wierda W, Keating M, Champlin R, Shpall EJ, Rezvani K. Use of CAR-Transduced Natural Killer Cells in CD19-Positive Lymphoid Tumors. N Engl J Med. 2020 Feb 6;382(6):545-553. doi: 10.1056/NEJMoa1910607. PMID: 32023374; PMCID: PMC7101242.

Featured image: T-cells attacking a cancer cell. Photo courtesy: Fotolia/Adobe 2016 2020. Used with permission.

Read more:
CAR T-cell Therapies for the Treatment of Patients with Acute Lymphoblastic Leukemia - OncoZine

Read More...

Was the MERS virus a model for the creation of COVID-19? – WION

Sunday, October 18th, 2020

First reported in 2012 in Saudi Arabia, Middle East Respiratory Syndrome (MERS) is a respiratory illness caused by a coronavirus with symptoms similar to the COVID-19 coronavirus, namely, fever, cough and shortness of breath with a range from none, to mild, to severe.

As of January 2020, about 2,500 cases of MERS have been reported worldwide. Human-to-human transmission typically requires close contact with an infected person, the spread being uncommon outside of hospitals.

In contrast to COVID-19, the death rate from MERS is about 35%.

MERS is believed to have originated in bats, was transmitted to camels as an intermediate host, then infecting humans, who had contact with the infected animals.

Although the COVID-19 virus has structural similarities to bat coronaviruses, its precise origin has yet to be identified.

The most distinguishing and unique structural feature of the COVID-19 virus is the furin polybasic cleavage site, a sequence of amino acids that interacts with human cell enzymes, which cut or cleave parts of the viral structure, thus contributing to the life cycle of the virus.

In the case of COVID-19, that sequence of amino acids is usually identified as proline-arginine-arginine-alanine or, in scientific notation, PRRA, which precedes an arginine-serine cleavage point, R-S.

It is unknown from where the PRRA sequence originated because it does not exist in any of the bat coronaviruses identified as close relatives of the COVID-19 virus.

A model for such a structure, however, does exist in the MERS coronavirus, which has a proline-arginine-serine-valine or PRSV sequence preceding the R-S cleavage point and having the following alignment:

COVID-19 PRRAR-S

MERS PRSVR-S

Both sequences begin with proline (P), both are polybasic having more than one arginine (R) and both have a non-polar amino acid in the fourth position, alanine (A) and valine (V), respectively, prior to the cleavage point, R-S.

It is important to note that COVID-19 and MERS are from two completely different families of coronaviruses, so one could not have evolved from the other.

According to the scientific article Structures and dynamics of the novel S1/S2 protease cleavage site loop of the SARS-CoV-2 spike glycoprotein," the presence of proline (P) is highly unusual.

Unlike other amino acids, proline produces structural rigidity in proteins and is found in only 5 out of 132 identified furin cleavage site sequences.

Likewise, alanine (A) located just prior to the R-S cleavage point exists in only 5 out of 132 furin cleavage site sequences.

In an early June scientific article, A novel bat coronavirus closely related to SARS-CoV-2 contains natural insertions at the S1/S2 cleavage site of the spike protein, the authors claimed to have identified a bat coronavirus, called RmYN02, that appears to have a precursor of the COVID-19 furin polybasic cleavage site.

RmYN02 has a proline-alanine-alanine (PAA) insertion roughly in a similar position to the COVID-19 virus, but PAA is chemically neutral, lacks any basic amino acids and has no R-S point to be cleaved.

RmYN02's PAA sequence, therefore, cannot be considered a precursor of the COVID-19 furin polybasic cleavage site.

So, the question remains, if no yet identified close relative of COVID-19 has a similar furin polybasic cleavage site, from where did such a unique structural feature with amino acids in unusual positions arise?

Furin polybasic cleavage sites are known to increase viral infectivity and pathogenicity. Genetic engineering techniques for inserting such cleavage sites have existed for at least fifteen years.

At present, no natural evolutionary pathway has been identified to explain the presence of COVID-19s furin polybasic cleavage site.

Those who may have manufactured the COVID-19 virus, could have been trying to mimic the cleavage site found in MERS.

Furthermore, the high rate of human-to-human transmission found for COVID-19, may have resulted from "pre-adapting" the virus for human infection by serial infection or passaging of the virus using animal models genetically-engineered to express the human coronavirus receptor.

There is now a preponderance of evidence that the COVID-19 virus was the product of laboratory experimentation rather than a natural infectious "jump" from bats to humans.

China still has a lot of explaining to do.

(Lawrence Sellin, Ph.D. is a retired U.S. Army Reserve colonel, who previously worked at the U.S. Army Medical Research Institute of Infectious Diseases and conducted basic and clinical research in the pharmaceutical industry. His email address is lawrence.sellin@gmail.com)

(Disclaimer: The opinions expressed above are the personal views of the author and do not reflect the views of ZMCL.)

The rest is here:
Was the MERS virus a model for the creation of COVID-19? - WION

Read More...

Study shows a molecular dance that keeps your heart beating – WSU News

Sunday, October 18th, 2020

A microscope photograph of a heart muscle cell. The regular green patterns show stained actin filaments.

By Tina Hilding, Voiland College of Engineering and Architecture

It might look like a little game at the molecular scale.

Filament-like proteins in heart muscle cells have to be exactly the same length so that they can coordinate perfectly to make the heart beat.

Another protein decides when the filament is the right size and puts a wee little cap on it. But, if that protein makes a mistake and puts the cap on too early, another protein, leiomodin, comes along and knocks the cap out of the way.

This little dance at the molecular scale might sound insignificant, but it plays a critical role in the development of healthy heart and other muscles. Reporting in the journal, Plos Biology,a WSU research team has proven for the first time how the mechanism works.

The finding could someday lead to improved diagnostics and medical treatments for serious and sometimes devastating hereditary heart conditions that come about from genetic mutations in the proteins. One of these conditions, cardiomyopathy, affects as many as one in 500 people around the world and can often be fatal or have lifetime health consequences. A similar condition called nemaline myopathy affects skeletal muscles throughout the body with often devastating consequences.

Mutations in these proteins are found in patients with myopathy, saidAlla Kostyukova, associate professor in the Gene and LindaVoiland School of Chemical Engineering and Bioengineeringand leader of the project. Our work is to prove that these mutations cause these problems and to propose strategies for treatment.

Heart muscle is made of tiny thick and thin filaments of proteins. With the help of electrical signals, the rope-like filaments bind and unbind in an intricate and precise architecture, allowing heart muscle to contract and beat.

The thin filaments are made of actin, the most abundant protein in the human body. Tropomysin, another protein, wraps itself around the actin filaments. Tropomyosin together with two other proteins, tropomodulin and leiomodin, at the end of the actin filaments act as a sort of cap and determine the filament length.

Its beautifully designed, said Kostyukova, whose research is focused on understanding protein structures.

And, tightly regulated.

To keep heart muscle healthy, the actin filaments, which are about a micron long, all have to be the exact same length. In families with cardiomyopathy, genetic mutations result in formation of filaments that are either too short or too long. Those affected can have significant heart problems that cause disability, illness and death.

In a project that spanned seven years, the researchers proved that leiomodin attaches to the end of the actin filament and kicks out the other protein, tropomodulin, to assure the actin filaments proper length.

This is the first time that this has been shown with the atomic-level precision, said Dmitri Tolkatchev, research assistant professor in the Voiland School and lead author on the paper. Previously, several laboratories attempted to solve this problem with very little success. With our data we finally have a direct proof.

The researchers used state-of-the-art approaches to make the key proteins and study them at the molecular and cellular level. The work entailed designing the molecules, constructing them at the gene level in a plasmid, and then producing them into bacterial or cardiac cells. The researchers used nuclear magnetic resonance, which works on the same physical principle as Magnetic Resonance Imaging (MRIs), to understand the proteins binding at the atomic level. They also used molecular dynamic simulation to model them.

The probability of being able to show this mechanism was not high, but the impact of the discovery is, said Tolkatchev, an expert in nuclear magnetic resonance. This was a very important problem to study and could have a significant impact in the field of muscle mechanics.

The researchers hope to continue the work, identifying additional components and molecular mechanisms that regulate thin filament architecture, whether diseased or healthy.

The multidisciplinary group included researchers from the University of Arizona led by Carol Gregorio, director of the Cellular and Molecular Medicine Department. WSUs group has expertise in protein structure, structural biochemistry, and properties of actin filaments and regulatory proteins, and UAs group has expertise in molecular, cellular and developmental biology of muscle assembly. The collaborative work was funded by the National Institutes of Health.

More:
Study shows a molecular dance that keeps your heart beating - WSU News

Read More...

Genetic Engineering Drug Market 2020 | What Is The Estimated Market Size In The Upcoming Years? – The Daily Chronicle

Wednesday, September 30th, 2020

The Global Marketers provides you regional research analysis on Genetic Engineering Drug Market and forecast to 2026. The global Genetic Engineering Drug Market report comprises a valuable bunch of information that enlightens the most imperative sectors of the Genetic Engineering Drug market. The global Genetic Engineering Drug market report provides information regarding all the aspects associated with the market, which includes reviews of the final product, and the key factors influencing or hampering the market growth.

Click Here To Access The Sample Report:

https://www.globalmarketers.biz/report/life-sciences/global-genetic-engineering-drug-market-report-2020-by-key-players,-types,-applications,-countries,-market-size,-forecast-to-2026-(based-on-2020-covid-19-worldwide-spread)/155824#request_sample

Main players in the Genetic Engineering Drug Market:

GeneScience Pharmaceuticals Co., LtdBeijing SL Pharmaceutical Co., LtdBiotech Pharmaceutical Co., LtdShenzhen Neptunus Interlong Bio-Technique Co., LtdJiangsu Sihuan Bioengineering Co., LtdTonghua Dongbao Pharmaceutical Co., LtdAnhui Anke Biotechnology (Group) Co., Ltd3SBio Inc.Shanghai Lansheng Guojian Pharmaceutical Co., Ltd

Some of the geographic regions examined in the overall Genetic Engineering Drug Market are:

In addition, the global Genetic Engineering Drug market report delivers brief information about federal regulations and policies that may ultimately affect market growth as well as the financial state. The situation of the global market at the global and regional levels is also described in the global Genetic Engineering Drug market report through geographical segmentation. The Genetic Engineering Drug report introduces speculation attainability evaluation, a task SWOT investigation, and venture yield evaluation.

Any Questions? Speak to Analyst @

https://www.globalmarketers.biz/report/life-sciences/global-genetic-engineering-drug-market-report-2020-by-key-players,-types,-applications,-countries,-market-size,-forecast-to-2026-(based-on-2020-covid-19-worldwide-spread)/155824#inquiry_before_buying

Global Genetic Engineering Drug Market Segmentation:

On the Basis of The Application:

30 Years Old30 Years Old-60 Years Old60 Years Old

On the Basis of Type:

Monoclonal AntibodyRecombinant Human ErythropoietinRecombinant Human InterferonRecombinant Human Growth HormoneRecombinant Human Insulin

Moreover, the report comprises the main developments made in the Genetic Engineering Drug market. Porters five force analysis is used to conclude the competition in the Genetic Engineering Drug market along with new entrants and their strategies & tactics. The report involves the value chain analysis which denotes workflow in the Genetic Engineering Drug market. Also, the market has been classified on the basis of category, processes, end-use industry, and region. On the basis of geography, the report Genetic Engineering Drug the market.

Ask for Discount:

https://www.globalmarketers.biz/discount_inquiry/discount/155824

The Genetic Engineering Drug Market research report presents a comprehensive analysis of the market and contains attentive insights, facts, past data, and statistical support, and industry-validated market data. It furthermore contains projections applying a suitable set of assumptions and methodologies. The research Genetic Engineering Drug report provides examination and information according to market segments such as geographies, applications, and industry by considering major players.

Key questions answered in this report

Highlights of the TOC of the Genetic Engineering Drug Report:

Get Complete Details with TOC of Genetic Engineering Drug Report for Free :

https://www.globalmarketers.biz/report/life-sciences/global-genetic-engineering-drug-market-report-2020-by-key-players,-types,-applications,-countries,-market-size,-forecast-to-2026-(based-on-2020-covid-19-worldwide-spread)/155824#table_of_contents

The rest is here:
Genetic Engineering Drug Market 2020 | What Is The Estimated Market Size In The Upcoming Years? - The Daily Chronicle

Read More...

Orphan Drug Exclusivity for CRISPR/Cas-Based Therapeutics – JD Supra

Wednesday, September 30th, 2020

The prospect of genetic engineering using CRISPR (clustered regularly interspaced short palindromic repeats) and CRISPR-associated nucleases (Cas) has long been hailed as a revolutionary development in medicine.

This technology is rapidly advancing, and several CRISPR/Cas-based drugs have entered clinical trials over the past several years. One kind of product in clinical trials is CRISPR-modified cells, such as CTX001 (CRISPR-Cas9-modified autologous hematopoietic stem cells), currently under study for the treatment of b-thalassemia and severe sickle cell anemia. Another CRISPR-based product, AGN-151587, is injected into the eye with the goal of eliminating a genetic mutation in patients with Leber congenital amaurosis 10, a leading cause of childhood blindness. In parallel, others are working to harness the CRISPR/Cas system to develop drugs for rare diseases, including bespoke therapies tailored to an individual patients needs.

Given CRISPR/Cas-based drugs potential to treat rare diseases, issues relating to orphan drug exclusivity will arise as these products are developed. In May 2020, for example, CTX001 received an orphan drug designation for transfusion-dependent b-thalassemia.

In January 2020, the FDA provided draft guidance regarding orphan drug exclusivity for gene therapy products, which includes CRISPR/Cas gene editing (Draft Guidance). This guidance focuses on the analysis of whether two gene therapy products are the same under the Orphan Drug Act. Although informative, the limited scope of the Draft Guidance invites more questions than it answers.

Same Drugs Under the Orphan Drug Act

Obtaining orphan drug exclusivity involves a two-step process. First, a sponsor requests designation of a drug for a particular rare disease or condition. See 21 C.F.R. 316.20. If this drug is the same drug as a drug already approved to treat the same rare disease or condition, the sponsor must provide a plausible hypothesis that the new drug is clinically superior to the previously-approved drug. Id. Whether two drugs are the same depends on consideration of structural features relevant to that type of drug. See id. 316.3(b)(14).

If the new drug later obtains marketing approval for a use or indication within the rare disease or condition for which it received orphan drug designation, the FDA will determine if the drug is eligible for orphan drug exclusivity. See 21 C.F.R. 316.31(a). In this situation, to receive exclusivity, the sponsor of the new drug must show that its drug is clinically superior to the same previously-approved drug for the same rare disease or condition. See id. 316.34(c). A clinical superiority determination is based on the new drugs greater efficacy, greater safety, or a major contribution to patient care. See id. 316.3(b)(3).

Highlights from Draft FDA Guidance

To determine whether one gene therapy product is the same as another, per 316.3(b)(14)(ii), the FDA will evaluate the principal molecular structural features of the two products, particularly transgenes (e.g., transgenes that encode different enzymes for treatment of the same rare disease) and vectors. For example:

Additionally, [w]hen applicable, the FDA generally intends to consider additional features of the final gene therapy product, such as regulatory elements or, in the case of genetically-modified cells, the type of cell that is transduced. It generally intends to consider requests for designation and exclusivity of gene therapy products to evaluate whether such additional features may also be considered to be principal molecular structural features.

Implications for CRISPR/Cas Therapy Exclusivity

The Draft Guidance helps answer certain high-level questions relating to whether two gene therapy products would be considered the same under the Orphan Drug Act. As various stakeholders have recognized, however, it is short on the details that meaningfully aid the process of drug research and development.

It is clear from the Draft Guidance that a new product can be considered the same as a previously-approved product even if the two products are not perfectly identical, but the guidance does not explain what would constitute a minor difference between such products, or what the scope of additional features would be.

For example, the Draft Guidance does not clarify what makes two transgenes the same. Nor does it cite to prior guidance or regulations that may answer this question. The question is significant because Cas nucleases and other parts of the CRISPR/Cas system may be modified in various ways. To address whether these modifications bar a finding of same-ness, the FDA could potentially import the kinds of considerations that govern same-ness of other kinds of large-molecule products, such as polynucleotide drugs or closely related, complex partly definable drugs with similar therapeutic intent (e.g., viral vaccines). See 21 C.F.R. 316.3(b)(14)(ii)(C), (D). However, this is not clear from the Draft Guidance.

The Draft Guidance also does not explain what will factor into the case-by-case basis assessment of whether viral vectors from the same viral class are the same. In the case of AAV2 and AAV5the two viruses identified in the guidanceresearchers have found that these viruses differ with respect to sequence analysis, tissue tropism, and heparin sensitivity. It is not clear from the guidance, however, whether a plausible hypothesis of clinical superiority will be required to seek orphan drug designation for a drug based on AAV2 if the previously-approved drug expresses the same transgene(s) but is based on AAV5.

It would be beneficial to sponsors and other stakeholders if these aspects of gene therapy drugs sameness are clarified further before they invest significant resources into the design and development of these therapeutics.

[View source.]

See the original post:
Orphan Drug Exclusivity for CRISPR/Cas-Based Therapeutics - JD Supra

Read More...

Inventing the future for humankind | Community Perspectives – Fairbanks Daily News-Miner

Wednesday, September 30th, 2020

Back in the halcyon days, when I somehow got paid for messing with the minds of the impressionable youth of UAF, I liked to ask said minds to project themselves back in time 400 years, to take a look around and report back what, if anything, they noticed different between those times (counting back from now, for example, to the Lords Year 1620. (James I was King, if that helps) and our own times: changes in musical tastes, ethics, physics, theology or attitudes regarding leprosy, for instance.

1620 CE was earlier than heart transplants, genetic engineering and baseball. It was before George Washington and water-seal toilets. Oxygen wouldnt be invented until the mid-1700s (Really: no Periodic Table of Elements, no radioactivity). The Holy Inquisition was in practice: pious religious officials were still torturing heretics and burning witches. It was before abortion rights. Autochthonous peoples in many parts of the world had not been introduced to the blessings of European economics, religion and warfare. It was before Facebook.

Things had changed in the last 400 years. Bigly. My students always got that answer right.

Then Id ask them to project themselves 400 years into the future, to the early 25th Century, say, to look around, to report back. I asked them to pay particular attention to the way our descendants in 2420 look back on our (presumably long-obsolete) ways of doing things: our medicine, say, or our governmental systems, or our responses to global hunger, overpopulation, pollution.

This was a harder task. The problem with prognostication is that we normal people are not particularly good at it, being annoyingly set in our ways. This is not to say that we cant make predictions, but even deeply considered and finely calibrated events such as space launches, brain surgery, or steering an oil tanker around Bligh Reef occasionally go awry. Some events, like nuclear meltdowns or worldwide pandemics, can present unanticipated difficulties.

I asked my students to avoid fantasies like self-aware computers, two-way wrist radios or honest politicians. I was hoping for revolutionary ways of perceiving the world, something on the order of the atalatl, General Relativity or Akira Kurosawa. I was angling for new stuff: examples of true scientific, artistic or musical invention.

My students always protested. Were on to you, old man, you you English teacher! Youve been harping all semester about how we mortals really cant see into the future, that we make up the future with our words. Now you want us to think something no one has ever thought before!

Thats exactly what I wanted them to do, of course. To be fair, really new ideas are not particularly common. It took humans millennia to come up with the atlatl (c.20,000 BCE), even longer to invent the calculus (c.1665 CE) or germ theory (c.1840). But without inventive ways of looking at the world, humanity might still believe that malaria is caused by bad air, that light travels across a medium called luminiferous ether, or that things burn because they contain phlogiston.

Theres been much talk lately of returning to normal, but I wonder if thats really what we want. I wonder if normal isnt what got us into our present public health and economic crises. I think for a lot of people in our community normal is worrying about buying groceries, paying the rent, health care, personal safety.

In this Year of Our Trump and the Corona pandemic (known also to certain elderly cynics as the beer virus or the sniffles) the question for my students would be, Given that we really cant see into the future and given that our current pandemic is unlikely to be our last, whats our best strategy for the survival of Our People (defined however you like) for the next seven generations or so?

Id hope for some inventive thinking along the lines of how to take care of every person on Earth in honest and practical ways. Emphasizing that we have plenty to be humble be about when predicting the future, Id ask them to come up with ideas never tried before. Id suggest that food, shelter and health care need never to be money-dependent, for example. Id ask our youth for creative ways of feeding people, sheltering people, caring for people all people on this, our planetary spaceship.

Id invite them to approach the task with an honest and generous spirit.

Lynn Basham lives in Fairbanks. He taught atthe University of Alaska Fairbanks as an instructor, mostly in the English Department, for about 20 years and retired about10 years ago.

Read more:
Inventing the future for humankind | Community Perspectives - Fairbanks Daily News-Miner

Read More...

Page 8«..78910..2030..»


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