Author Archives: admin


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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

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

Back to Top

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

USC scientists use brain organoids to study intellectual disability, with support from the SynGAP Research Fund – USC News

Maybe a child misses a developmental milestone, such as rolling over, or saying that first word. Perhaps the child also falls down due to seizures, or develops symptoms of an autism spectrum disorder. Eventually, a doctor pinpoints the cause: a rare and spontaneous variant in a gene called SYNGAP1, which leads to a variety of debilitating conditions that might include intellectual disability, epilepsy, and autism spectrum disorders. As of now, there is no cure.

To help children with this rare syndrome, Ashley Evans and Mike Graglia founded the SynGAP Research Fund (SRF), which recently donated $46,250 to USC Stem Cell researchers Marcelo Pablo Coba and Giorgia Quadrato. Cobas lab is at the Zilka Neurogenetic Institute, and Quadratos lab is at the Eli and Edythe Broad CIRM Center for Regenerative Medicine and Stem Cell Research at USC.

Im thrilled that SRF is supporting their work, said Graglia, whose son has a SYNGAP1 variant, and who serves as the managing director of SRF. Dr. Coba has spent hours letting us pick his brain and ask questions about his papers related to SYNGAP1. He also told us about his exciting new collaboration with Dr. Quadrato, who uses stem cells from patients with the SYNGAP1 variant to grow 3D networks of human nerve cells, called brain organoids.

The scientists will study these patient-specific brain organoids, mapping the electrophysiological and genetic activities of different types of nerve cells. They will then explore several interventions, such as drug-like molecules, gene editing, and cell transplants, to see if they can modify these activities.

Coba first connected to SRF thanks to the organizations Science & Medicine Lead, Hans P. Schlecht, MD. As a physician, Schlecht responded to his sons diagnosis of Syngap Encephalopathy by learning as much as possible about the variant. In 2017, he was browsing through SYNGAP1 related projects in the NIH RePORTER, an online database that provides details about medical research supported by federal tax dollars. Schlecht found Cobas name and reached out by phone. Not long after this first conversation, Coba offered to meet in person and share even more about his research.

A doctor is assessed on the three As: affable, available, and able, said Schlecht. Coba is all of these. He is easy to talk to, he replies to calls and emails, and hes a capable scientist.

In 2018, Quadrato joined USC as an assistant professor of stem cell biology and regenerative medicine at the Keck School of Medicine of USC. She brought with her an expertise for growing brain organoids over a period of nine months in the laboratoryallowing the nerve cells to mature and diversify in ways that recapitulate some limited aspects of the extraordinary brain development that takes place during a fetus nine months in utero.

In 2019, she was awarded $100,000 by the Donald E. and Delia B. Baxter Foundation to use patient-derived organoids to investigate the role of SYNGAP1 in human brain development.

To further tap into the potential of organoids to enhance our understanding of SYNGAP1 variants, Quadrato began collaborating with Coba, who is an associate professor of psychiatry and the behavioral sciences at the Zilkha Neurogenetic Institute at the Keck School.

Thanks to this support from SRF and the Keck School, we have the means to purchase an incredible new piece of equipment called the MEA Willow System, which allows us to visualize how patient-specific brain organoids function, as well as how they react to potential treatments, said Quadrato. It is a truly groundbreaking technology created by a collaboration between Leaflabs and the Synthetic Neurobiology Group at MIT.

Coba added: Were grateful for this generous funding that allows us to use the latest technology to advance our research collaboration. Im also personally thankful that Ive had the privilege to connect with SRF parents and patient advocates throughout the years. Their dedication and passion reaffirms the critical importance of the work that we do every day in the lab with the goal of finding better treatments for these patients.

See the original post:
USC scientists use brain organoids to study intellectual disability, with support from the SynGAP Research Fund - USC News

Impact Analysis of Covid-19 On Osteonecrosis Treatment Market Growth and Evolution During 2020-2027 | Bone Therapeutics, Enzo Biochem Inc., and…

Global Osteonecrosis Treatment Market has witnessed continuous growth in the past few years and is projected to grow even further during the forecast period (2020-2027). The research presents a complete assessment of the market and contains Future trends, Current Growth Factors, attentive opinions, facts, historical data, and statistically supported and industry-validated market data. This is the latest report, covering the current COVID-19 impact on the market. The pandemic of Coronavirus (COVID-19) has affected every aspect of life globally. It delivers important information to identify and analyze the market need, market growth, and competition.

This all-inclusive Osteonecrosis Treatment Market research report includes a detailed on these trends, share, size that can help the businesses operating in the industry to figure out the market and strategize for their business development accordingly. The research report analyses the growth, market size, key segments, industry share, application, and key drivers.

We Do Offer Sample of this report. Kindly go through the follow information in order to access sample copy.

Note- This report sample includes:

Brief Introduction to the research report.

Table of Contents (Scope covered as a part of the study)

Top players in the market

Research framework (Structure Of The Report)

Research methodology adopted by Coherent Market Insights

Request sample report @ https://www.coherentmarketinsights.com/insight/request-sample/1490

The major manufacturers covered in this report: Bone Therapeutics, Enzo Biochem Inc., and K-Stemcell Co Ltd. Hospitals, clinics, universities,

Key players in the Osteonecrosis Treatment market have been identified through secondary research, and their market shares have been determined through primary and secondary research. All measurement shares split, and breakdowns have been resolute using secondary sources and verified primary sources. The Osteonecrosis Treatment Market report begins with a basic overview of the industry lifecycle, definitions, classifications, applications, and industry chain structure, and all these together will help leading players understand the scope of the Market, what characteristics it offers, and how it will fulfill customers requirements.

Regional Analysis for Osteonecrosis Treatment Market:

For comprehensive understanding of market dynamics, the Global Osteonecrosis Treatment Market is analyzed across key geographies namely North America, Europe, China, Japan, Southeast Asia, India, Central & South America. Each of these regions is analyzed on basis of market findings across major countries in these regions for a macro-level understanding of the market.

What Osteonecrosis Treatment Market report offers:

The complete knowledge of the Osteonecrosis Treatment Market is based on the latest industry news, opportunities, and trends. The Osteonecrosis Treatment Market research report offers a clear insight into the influential factors that are expected to transform the global market in the near future. Both top-down and bottom-up approaches have been used to estimate and validate the market size of the Osteonecrosis Treatment market, to estimate the size of various other dependent submarkets in the overall market.

Remarkable Attributes of Osteonecrosis Treatment Market Report:

The Report Answers Following Questions:

Key Content of Chapters:

Part 1:Terminology Definition, Industry Chain, Industry Dynamics & Regulations and Global Market Overview

Part 2:Upstream (Raw Materials / Components) & Manufacturing (Procurement Methods & Channels and Cost), Major Regional Production Overview and Trade Flow

Part 3:Product Segment Overview and Market Status

Part 4:Application / End-User Segment Overview and Market Status

Part 5:Region Segment Overview and Market Status

Part 6:Product & Application Segment Production & Demand by Region

Part 7:Market Forecast by Product, Application & Region

Part 8:Company information, Products & Services and Business Operation (Sales, Cost, Margin, etc.)

Part 9:Market Competition and Environment for New Entrants

Part 10:Conclusion,In the end, the report makes some important proposals for a new project of Osteonecrosis Treatment Market before evaluating its feasibility. Overall, the report covers the sales volume, price, revenue, gross margin, historical growth, and future perspectives in the Osteonecrosis Treatment market. It offers facts related to the mergers, acquirement, partnerships, and joint venture activities widespread in the market.

All the reports that we list have been tracking the impact of COVID-19 the market. Both upstream and downstream of the entire supplychain has been accounted for while doing this. Also, where possible, we will provide an additional COVID-19 update supplement/report to the report in Q3, please check for with the sales team.

The PDF Research only provides Table of Contents (ToC), scope of the report and research framework of the report.

PDF Brochureof this Report:https://www.coherentmarketinsights.com/insight/request-pdf/1490

**Be Safe and Stay Home**

About Us:

Coherent Market Insights is a prominent market research and consulting firm offering action-ready syndicated research reports, custom market analysis, consulting services, and competitive analysis through various recommendations related to emerging market trends, technologies, and potential absolute dollar opportunity.

Contacts US:

Mr. Shah Email:[emailprotected] Phone: US +1-206-701-6702/UK +44-020 8133 4027

Go here to read the rest:
Impact Analysis of Covid-19 On Osteonecrosis Treatment Market Growth and Evolution During 2020-2027 | Bone Therapeutics, Enzo Biochem Inc., and...

Cell Therapy Market 2020 | Demand and Scope with Outlook, Business Strategies, Challenges and Forecasts to 2025 by Leading Key Players – 3rd Watch…

Cell Therapy Market Size, Share, & Trends Analysis Report By Use (Clinical, Research), By Type (Stem & Non-stem Cells) By Therapy Type (Autologous, Allogenic), By Region, And Segment Forecasts, 2019 2025.The cell therapy market size is expected to grow owing to the addition of cell types that provide extensive opportunities to organizations to strengthen the market situation. Due to this, there are number of businesses working in cell therapy development segment has grown to a large extent in the past years.

Get Sample Report @https://www.adroitmarketresearch.com/contacts/request-sample/611

Cell therapy is an important part of the therapeutic process for various clinical indications. The recent cell therapy market trends indicate that the studies of various use of cells for their therapeutic ability is gaining popularity in the science and healthcare research community. There are various techniques currently in the segment like CAR-t, stem cell and cord blood cell therapy. All of these reasons have led to increased application of cell therapy for clinical conditions like cancer and cardiovascular illness. Owing to this there is a constant growth in the Global Cell Therapy Market.

The most commonly used process of cell therapy aims to use healthy cells from a donor (Allogeneic) which is compatible or autogenic that is from the patient itself along with their alteration to increase their therapeutic ability. There are various complex steps involved in the process like genetic screening of cell, cell harvesting and reinfusion into the patients body. All these steps are complex and important and have therapeutic result on the patient. These advanced usage of cell therapy will result in growth of the cell therapy market size during the forecast period.

Browse Complete Research Report @https://www.adroitmarketresearch.com/industry-reports/cell-therapy-market

Cell therapy market trends indicate growth owing to the various regulations being approved by the government in the desire to provide quick relief to the patients. Furthermore, many healthcare industries are working in collaboration with the government to identify the various processes to ways to improve cell therapy. Furthermore, the cell therapy market size is also influenced by the commercialization of stem cells treatments.

The Stem cell therapy segment dominates the types of cell therapy and is said to have the maximum success rate. It has a special feature that it differentiates into any category of cell, at the same time ensuring the individual identity is intact. Industry experts state that the stem cell would revolutionize regenerative medicine, owing to its extensive use in treatment of fatal disease like neurodegenerative, cardiovascular and cancer. The growth of cell therapy market size is also factored to the increased research and development about the same. However, at the same time the huge cost involved in the various processes involved might be hinder the market growth.

The cell therapy market size is segmented on various categories like Clinical-use, Research and Therapy type and region. On the basis of region, North America is projected to contribute the maximum share to the market owing to increased development.

Key players in the market are JCR Pharmaceuticals Co., Ltd., Kolon TissueGene, Inc.; and Medipost and many more.

Make an Enquire to buy this Report @https://www.adroitmarketresearch.com/contacts/enquiry-before-buying/611

Segmentation:

The various segments of cell therapy market size are:

By Use & Type Outlook

By Cell Therapy Type

By Therapeutic Area

By Therapy Type

By Region

What to expect from the upcoming report on cell therapy market size:

Future prospects and current trends of the cell therapy market size by the end of forecast period. (2017 2025).

Information regarding technological progressions as well as innovations across the world

Supportive initiatives by government likely to influence the market dynamics.

In-depth analysis of different market segmentations including regional segmentations, applications and types.

Deep analysis about the competitive landscape of the market and the initiatives by them to improve this market.

Trends, drivers, opportunities, restraints, challenges and key developments in the market

Early Buyers will Get 10% Discount on This Premium Research now @https://www.adroitmarketresearch.com/contacts/discount/611

Who should buy this report?

Venture capitalists, Investors, financial institutions, Analysts, Government organizations, regulatory authorities, policymakers ,researchers, strategy managers, and academic institutions looking for insights into the market to determine future strategies.

About Adroit Market Research:

Adroit Market Research provide quantified B2B research on numerous opportunistic markets, and offer customized research reports, consulting services, and syndicate research reports. We assist our clients to strategize business decisions and attain sustainable growth in their respective domain. Additionally, we support them with their revenue planning, marketing strategies, and assist them to make decisions before the competition so that they remain ahead of the curve.

Contact Information:

Ryan Johnson

Account Manager Global

3131 McKinney Ave Ste 600, Dallas,

TX75204, U.S.A.

Phone No.: USA: +1 (214) 884-6068 / +91 9665341414

Read the rest here:
Cell Therapy Market 2020 | Demand and Scope with Outlook, Business Strategies, Challenges and Forecasts to 2025 by Leading Key Players - 3rd Watch...

Cytovia Therapeutics, Inc Adds Two Industry Leaders to Its Board of Directors – BioSpace

NEW YORK, June 24, 2020 (GLOBE NEWSWIRE) -- Cytovia Therapeutics, Inc (Cytovia), an emerging biopharmaceutical company developing Natural Killer (NK) immunotherapies for cancer, today announces the appointment of Dr. Leila Alland and Ms. Jane Wasman to its Board of Directors, effective immediately.

Dr. Leila Alland brings extensive experience in oncology drug development to her role at Cytovia Therapeutics. Dr. Alland is currently Chief Medical Officer at PMV Pharma, a leader in the discovery and development of small molecule therapies targeting p53 mutations, which are prevalent in many cancers. Dr. Alland was previously Chief Medical Officer at Affimed, where she advanced the companys portfolio of clinical-stage immuno-oncology programs. During her career, she has held leadership positions at Tarveda Therapeutics, AstraZeneca, Bristol-Myers Squibb, Novartis and Schering-Plough, and has contributed to numerous successful oncology drug approvals over the course of her career, spanning both molecularly targeted and immuno-oncology therapeutic products. Dr. Alland is a member of the Scientific Advisory Council of Columbia University's Center for Radiological Research. Dr. Alland obtained her medical degree from New York University School of Medicine, completed her residency in pediatrics at The Childrens Hospital of Philadelphia and her fellowship in pediatric hematology/oncology at The New York Hospital and Memorial Sloan-Kettering Cancer Center, and was Assistant Professor of Pediatrics at Albert Einstein College of Medicine. I am very excited to be joining the Board of Directors at Cytovia Therapeutics, said Dr. Alland. Cytovias portfolio of immune therapeutics is steeped in the science of NK cells and the development of powerful new technologies that leverage the ability of NK cells to fight cancer. These novel technologies have the potential to go far beyond current drug development paradigms and make a real difference to individuals living with cancer.

Ms. Jane Wasman is a strategic leader with almost 25 years in the biopharma industry, with extensive U.S. and international experience. Ms. Wasman is Chair of the Board of Directors of Sellas Life Sciences, a board member at Rigel Pharmaceuticals, and serves on the board of NY BIO. Ms. Wasman is Founder and President of JWasman Advisors, (a consulting firm focused on strategic, operational and corporate governance matters for biopharma and life sciences organizations). She previously served as President, International, General Counsel and Chief, Strategic Development at Acorda Therapeutics, where she led long-range planning and development in addition to international expansion and in leadership positions including Vice President at Schering Plough. Ms. Wasman graduated magna cum laude from Princeton University, and earned her J.D. from Harvard Law School. I am thrilled to be able to joinCytovia's team who is implementing an agile partnership strategy and capital-efficient execution approach, said Ms. Wasman. I believe Cytovia Therapeutics is positioned to become a leading NK company with a differentiated product portfolio, leveraging two strong technologies - CAR NK and NK engager antibodies.

Dr. Daniel Teper, co-founder, Chairman and CEO of Cytovia Therapeutics, Inc stated:"We are delighted to welcome two exceptional industry leaders to our boardof directors. Cytovia Therapeutics is at a growth inflexion point where it can fully benefit from Dr. Alland's solid track record in oncology drug development and Ms. Wasman's expertise in corporate and financial transactions."

Media Snippets accompanying this announcement are available by clicking on the images or links below:

ABOUT CYTOVIA THERAPEUTICS, INC Cytovia Therapeutics Inc is an emerging biotechnology company that aims to accelerate patient access to transformational immunotherapies, addressing several of the most challenging unmet medical needs in cancer and severe acute infectious diseases. Cytovia focuses on Natural Killer (NK) cell biology and is leveraging multiple advanced patented technologies, including an induced pluripotent stem cell (iPSC) platform for CAR (Chimeric Antigen Receptors) NK cell therapy, next-generation precision gene-editing to enhance targeting of NK cells, and NK engager multi-functional antibodies. Our initial product portfolio focuses on both hematological malignancies such as multiple myeloma and solid tumors including hepatocellular carcinoma and glioblastoma. The company partners with the University of California San Francisco (UCSF), the New York Stem Cell Foundation (NYSCF), the Hebrew University of Jerusalem and Macromoltek.

Learn more atwww.cytoviatx.com

Contact for media enquiries at Cytovia Therapeutics, Inc Sophie Badr Vice President, Corporate Affairs Sophie.badre@cytoviatx.com 1(929) 317 1565

The rest is here:
Cytovia Therapeutics, Inc Adds Two Industry Leaders to Its Board of Directors - BioSpace

Cell Therapy Industry Production, Sales and Consumption Status and Prospects Professional Market Research Report 2020-2025 – 3rd Watch News

The Cell Therapy Market report covers extensive study about the market players operating in the global market. The report offers a complete analysis of the strategies implemented by leading service providers. Additionally, the report contains qualitative and quantitative market evaluation depending on the market segmentation. This report offers an in-depth analysis of the market trends that are influencing the market growth. The report comprises the comprehensive study of geographical regions which may include North America, Europe, Asia Pacific, and the MEA. The report on the Cell Therapy Market is specially designed to provide cutting-edge market intelligence as well as aid investors to take investment estimate decisions.

Leading Companies Reviewed in the Report are:

JCR Pharmaceuticals Co., Ltd., Kolon TissueGene, Inc.; and Medipost and many more.

Get Exclusive Sample of Report on Cell Therapy market is available @ https://www.adroitmarketresearch.com/contacts/request-sample/611

Furthermore, the report covers the PESTLE as well as Porters Five Forces analysis for in-depth comparisons and other significant factors for market analysis. In addition to this, every section of the Cell Therapy research report has offered significant information to provide for service providers to increase their revenue margin, marketing strategy and sales, as well as profit margin. In addition, the Cell Therapy research report offered a comprehensive qualitative as well as quantitative analysis with the several opportunities assessment across the world. Besides, the research report used as a tool for getting extensive market analysis, service providers can recognize the required changes into their operation and gain their position across the global market. In addition, the number of business tactics aids the Cell Therapy Market players to give competition to the other players in the market while recognizing the significant growth prospects.

Likewise, the Cell Therapy research report provides market insights from the huge number of statistics which are collected from robust Cell Therapy Market data such as channel partners, manufacturers, regulatory bodies, as well as decision makers. Similarly, the research report evaluates the market growth rate as well the current market value according to the market dynamics as well as the growth prospects. The market analysis offered in this report is assessed on the basis of market data, market trends, and the number of growth potentials. In addition, it includes an extensive investigation of the market scenario with the in-depth analysis of their major service providers. In addition to this, on the basis of several clients conditions, the Cell Therapy report produces highly customized data that will help regional as well as global service providers to increase their market position over the globe.

Quick Read Table of Contents of this Report @ https://www.adroitmarketresearch.com/industry-reports/cell-therapy-market

Global Cell Therapy market is segmented based by type, application and region.

Based on Type, the market has been segmented into:

By Use & Type Outlook, (Clinical-use,By Cell Therapy Type,,Non-stem Cell Therapies,Stem Cell Therapies,BM, Blood, & Umbilical Cord-derived Stem Cells,Adipose derived cells,Others), By Therapeutic Area, (Malignancies,Muscoskeletal Disorders,Autoimmune Disorders,Dermatology,Others,Research-use), By Therapy Type, (Allogenic Therapies,Autologous Therapies)

The report offers a complete data analysis about the current trends which have developed and are expected to become one of the strongest Cell Therapy Market forces into coming future. Moreover, the research report offers a holistic overview of the Cell Therapy Market, several factors driving the market growth, as well as the companies involved in the Cell Therapy Market. In addition to this, the Cell Therapy report provides the extensive analysis of the market restraints that are responsible for hampering the Cell Therapy Market growth along with the report offers a comprehensive description of each and every aspects and its influence on the market. Furthermore, the Cell Therapy report provides a detailed value chain analysis of the Cell Therapy Market across the world.

Finally, the researchers throw light on different ways to discover the strengths, weaknesses, opportunities, and threats affecting the growth of the Global Cell Therapy Market. The feasibility of the new report is measured in this research report.

Do You Have Any Query Or Specific Requirement? Ask to Our Industry [emailprotected] https://www.adroitmarketresearch.com/contacts/enquiry-before-buying/611

About Us :

Adroit Market Research is an India-based business analytics and consulting company. Our target audience is a wide range of corporations, manufacturing companies, product/technology development institutions and industry associations that require understanding of a markets size, key trends, participants and future outlook of an industry. We intend to become our clients knowledge partner and provide them with valuable market insights to help create opportunities that increase their revenues. We follow a code Explore, Learn and Transform. At our core, we are curious people who love to identify and understand industry patterns, create an insightful study around our findings and churn out money-making roadmaps.

Contact Us :

Ryan Johnson Account Manager Global 3131 McKinney Ave Ste 600, Dallas, TX 75204, U.S.A Phone No.: USA: +1 972-362 -8199 / +91 9665341414

Read the rest here:
Cell Therapy Industry Production, Sales and Consumption Status and Prospects Professional Market Research Report 2020-2025 - 3rd Watch News

Update on FDAs Comprehensive Regenerative Medicine Framework: Looming November 2020 Deadline Preceded by a Flurry of Letters from CBER and a New JAMA…

As we discussed in our last update on the Food and Drug Administrations Comprehensive Regenerative Medicine Policy Framework back in December 2019 (during the much simpler, pre-COVID-19 world), this coming November will conclude the three-year period of enforcement discretion announced by the agency when it first articulated the policies and goals of this comprehensive framework. In particular, under the dual-track program announced in 2017, the Food and Drug Administration (FDA) has been focused on: (1) clarifying the regulatory criteria for product marketing through guidance and providing support to legitimate product developers through formal and informal interactions; and (2) removing unapproved, unproven, and potentially unsafe products from the U.S. market.

None of the COVID-19-related operational updates provided by the FDA generally or by the Center for Biologics Evaluation and Research (CBER) in recent months has suggested that the November 2020 deadline will be extended or otherwise altered as a result of the ongoing public health emergency, even as certain other enforcement discretion policies have been put into place. Additionally, a recent editorial published by agency leadership and a noticeable increase in Warning/Untitled Letters to persons offering unapproved cellular therapy products, taken together, strongly suggest that folks in this industry that are currently operating outside of the applicable regulatory framework should not expect to be given any additional time to come into compliance.

June 2020 JAMA Editorial Strong Language and No Sign of a Deadline Extension

Multiple statements on the topic of regenerative medicine have been issued by the governing FDA Commissioner as well as CBER Director Peter Marks over the past several years, which indicates how important this area is to the agencys broader public health priorities at the start of the 21st century. The most recent salvo from agency leadership came in the form of an editorial published online by JAMA on June 17, 2020, authored by Dr. Marks and Commissioner Stephen Hahn, who has been in his new job for about six months. Their editorial includes some of the strongest language we have seen to date on the topic of unapproved regenerative medicine products. For example, Dr. Marks and Dr. Hahn state that [d]espite assertions by some individuals to the contrary, these products, whether autologous or allogeneic, are not inherently safe and may be associated with serious adverse consequences. They assert that [t]he increasing number of adverse events being reported following the widespread use of unapproved regenerative medicine therapies at hundreds of clinics across the country make it necessary for the FDA to act to prevent harm to individuals receiving them.

Drs. Marks and Hahn briefly highlight some of the enforcement that the agency has undertaken in this space since 2017 and ask for engagement from both clinicians and patients to help to ensure that instead of remaining unintentionally or intentionally hidden, potentially harmful unapproved regenerative medicine therapies are identified and removed from the market. They then provide basic guidelines for patients and caregivers to use when assessing whether a cellular therapy product is being offered in compliance with applicable laws and FDA regulations. Specifically, they recommend the following key considerations for anyone considering treatment with a cellular product:

Nothing in this newly-published editorial suggests that FDA/CBER will be taking its proverbial foot off the pedal to slow down its efforts towards further oversight of the private stem cell clinic industry after November 2020. To the contrary, the piece could represent one of the last informal warnings those businesses get from the agency before they receive a customized Warning or Untitled Letter or become subject to whatever increased enforcement activity the federal government initiates in this area in 2021 and beyond.

Relatively Large Number of Warning Letters Sent Since January 2020

We previously noted that FDA/CBER appeared to have increased the pace of issuing Warning and Untitled Letters to sellers of unapproved stem cell products during the second half of 2019, with many of those letters involving companies that processed and marketed unapproved umbilical cord blood-derived cellular products. We also reported that the agency had issued a Public Safety Notification on Exosome Products on December 6, 2019, informing the public of multiple reports of serious adverse events experienced by patients in Nebraska who were treated with unapproved products marketed as containing exosomes. That safety alert also described the unscrupulous conduct of sellers of such products in forceful and direct language, similar to the language used by Dr. Marks and Commissioner Hahn in this months editorial piece.

Over the first half of this year, as we get yet closer to the November 2020 deadline for stem cell clinics and medical practitioners to come into compliance with federal law, there has been a more noticeable increase in the Warning/Untitled Letters issues regarding the marketing of unapproved products that put patients at risk. These include at least nine Untitled Letters issued since January 2020 (which can each be accessed from this CBER webpage) and at least two Warning Letters, one from March and one from June. The Warning Letters in particular include charges that the firms in question were violating current good manufacturing practices (CGMPs) and current good tissue practices (CGTPs) for human cells and tissue products, putting patient safety at risk.

Interestingly, the most recent FDA Warning Letter issued on June 4, 2020 not only cites the recipient for marketing unapproved stem cell products and an unapproved exosome product, but it also states that the unapproved exosome product was being marketed for the treatment and prevention of COVID-19 something the June 17 Marks/Hahn JAMA editorial alluded to generally as well. Given that there are currently no FDA-approved products to prevent or treat COVID-19, any such claims will automatically heighten the enforcement risk to a company or physician engaged in the sale of products for those intended uses.

In addition to the work being done by FDA, moreover, the Federal Trade Commission (FTC) has also been monitoring the commercial marketplace closely and taking various actions to protect consumers from fraudulent COVID-19 products, including a few marketed by stem cell clinics. So far this month, FTC announced on June 4, 2020 that it had issued a 35 warning letters and an additional 30 warning letters on June 18, 2020. The first batch of these FTC warning letters notably included one to a stem cell clinic that, among other things, had claimed that stem cells can be administered intravenously and by inhalation through a nebulizer to treat lung damage caused by COVID-19 without scientific evidence to support the efficacy claim, while the second batch included two letters addressed to marketers of stem cell products.

FDA and the FTC coordinate quite closely on consumer protection matters that implicate both agencies primary missions, as is apparent from the large number of COVID-19 Warning Letters that have been jointly issued by the two agencies since March 2020. So they may very well be coordinating more actively now on the monitoring of stem cell clinics and individual physicians offering unapproved cellular therapies to the general public, as the focus shifts to the next phase of the Comprehensive Regenerative Medicine Policy Framework. The next five or six months should offer everyone more insight into what the enforcement landscape is likely to evolve into once the FDAs enforcement discretion period ends in November. As always, well keep our readers apprised of any notable developments.

[View source.]

Read the original post:
Update on FDAs Comprehensive Regenerative Medicine Framework: Looming November 2020 Deadline Preceded by a Flurry of Letters from CBER and a New JAMA...

Global Canine Stem Cell Therapy Market with (Covid-19) Impact Analysis: Future Development, Business Growth and Applications to 2025 – Cole of Duty

Researchstore.biz has released a new research report titled Global Canine Stem Cell Therapy Market 2020 by Company, Type and Application, Forecast to 2025 which offers a strategic evaluation of the market. The report presents prime parameters such as market size, revenue, sales analysis, and key drivers, and projection of the market by product, area, and use. The report delivers an in-depth analysis of the market with current and future trends. The report recognizes overall growth opportunities, industry drivers, obstacles, latest discoveries, and openings available for newcomers in the global Canine Stem Cell Therapy market.

Competitive Outlook:

The report further contains a competitive scenario of the major market players focusing on their sales revenue, customer demands, company profile, import/export scenario, business strategies that will help the emerging market segments in making major business decisions. In addition, their product portfolio, respective product applications, and product features have been emphasized in the report. The report also provides company shares and distribution shares data for the Canine Stem Cell Therapy market category and global corporate-level profiles of the key market participants.

DOWNLOAD FREE SAMPLE REPORT: https://www.researchstore.biz/sample-request/48453

NOTE: This report takes into account the current and future impacts of COVID-19 on this industry and offers you an in-dept analysis of Canine Stem Cell Therapy market.

The study profiles and examines leading companies and other prominent companies operating in the global Canine Stem Cell Therapy industry, covering: VETSTEM BIOPHARMA, VetMatrix, Aratana Therapeutics, Cell Therapy Sciences, Vetbiologics, Regeneus, ANIMAL CELL THERAPIES, Okyanos, Medivet Biologics, Magellan Stem Cells, Stemcellvet,

The far-reaching market study enlists a focused assessment of this business space and the regional landscape of this vertical. As per the report, the Canine Stem Cell Therapy market has established its presence across regions such as: North America (United States, Canada and Mexico), Europe (Germany, France, United Kingdom, Russia and Italy), Asia-Pacific (China, Japan, Korea, India, Southeast Asia and Australia), South America (Brazil, Argentina), Middle East & Africa (Saudi Arabia, UAE, Egypt and South Africa)

On the basis of product, this report displays the production, revenue, price, market share, and growth rate of each type, primarily split into: Allogeneic Stem Cells, Autologous Stem cells

On the basis of the end users/applications, this report focuses on the status and outlook for major applications/end users, consumption (sales), market share market growth rate for each application, including: Veterinary Hospitals, Veterinary Clinics, Veterinary Research Institutes

This search provides an analysis of indirect and direct sales channels, helps you to plan the right distribution strategy. The study also predicts the influence of different industry aspects on the global Canine Stem Cell Therapy market segments and regions. It delivers analysis on consumption volume, region-wise import/export analysis, and forecast Canine Stem Cell Therapy market from 2020-2025. At last, the report exhibits an analysis of leading marketing players, product specification, company profiles along with the contact details, production cost.

ACCESS FULL REPORT: https://www.researchstore.biz/report/global-canine-stem-cell-therapy-market-2020-48453

Important Aspects of Canine Stem Cell Therapy Market Report:

Customization of the Report: This report can be customized to meet the clients requirements. Please connect with our sales team ([emailprotected]), who will ensure that you get a report that suits your needs. You can also get in touch with our executives on +1-201-465-4211 to share your research requirements.

About Us

Researchstore.biz is a fully dedicated global market research agency providing thorough quantitative and qualitative analysis of extensive market research.Our corporate is identified by recognition and enthusiasm for what it offers, which unites its staff across the world.We are desired market researchers proving a reliable source of extensive market analysis on which readers can rely on. Our research team consist of some of the best market researchers, sector and analysis executives in the nation, because of which Researchstore.biz is considered as one of the most vigorous market research enterprises. Researchstore.biz finds perfect solutions according to the requirements of research with considerations of content and methods. Unique and out of the box technologies, techniques and solutions are implemented all through the research reports.

Contact Us Mark Stone Head of Business Development Phone: +1-201-465-4211 Email: [emailprotected] Web: http://www.researchstore.biz

The rest is here:
Global Canine Stem Cell Therapy Market with (Covid-19) Impact Analysis: Future Development, Business Growth and Applications to 2025 - Cole of Duty

Graham Company and Regenexx Announce Innovative Partnership to Reduce Orthopedic Surgical Spend for Employers – Stockhouse

PHILADELPHIA, June 23, 2020 /PRNewswire/ -- Graham Company, one of the country's largest insurance brokerages, today announced a new partnership with Regenexx, a network of leading interventional orthopedics clinics. The partnership gives Graham Company's self-insured clients the option to offer alternative non-surgical orthopedic treatment methods, provided by Regenexx through its expansive network of physicians, as a benefit enhancement for employees. In addition, the partnership will help Graham Company's clients control and reduce overall costs of orthopedic surgery procedures for employees.

According to data from Regenexx, its procedures have effectively intervened on 70% of elective orthopedic injuries and conditions where surgery was previously the only solution.* Regenexx offers a unique approach to treating orthopedic injuries non-surgically through stem cell therapy and other regenerative medicine. Procedures performed by Regenexx's specially trained physician network involve injections of custom orthobiologics, derived from the patient's own cells and natural healing agents, to treat approximately 40 orthopedic conditions. This interventional approach enables employers to save up to 70% of orthopedic costs by reducing the number of unnecessary surgeries performed annually.

"At Graham Company, we are committed to identifying solutions to help our clients control escalating health care costs while also enhancing their benefits offerings for employees," said Debra Montella, Graham Company's Vice President of the Employee Benefits Division. "Through this partnership with Regenexx, we are now able to present our self-insured clients with a unique approach that is transforming orthopedic care and driving down costs associated with surgical procedures."

This partnership also benefits the employees of clients with self-funded plans, providing them with alternative non-surgical treatment options to address orthopedic injuries or conditions. Regenexx company data found that when employees are given the choice between an invasive orthopedic surgery or its non-invasive procedure to treat orthopedic conditions, employees chose Regenexx's procedure a majority of the time. Compared to traditional orthopedic surgeries, Regenexx procedures require less downtime for patients and are considered less costly, less invasive and less risky.

"Regenexx is looking forward to expanding our mission of producing the best possible patient outcomes for Graham Company and their clients through our innovative alternative to orthopedic surgery," said Regenexx CEO Jason Hellickson. "Our patented regenerative approach to treating common orthopedic conditions is transforming the care pathway of an orthopedic patient who can now avoid a surgical encounter with less risk and less downtime."

*This data applies only to elective orthopedic surgery without fracture-related care and acute care trauma.

About Graham Company Graham Company is one of the largest insurance and employee benefits brokers in the country, committed to enhancing employee safety and business viability through an action-oriented approach to risk management. In business for nearly 60 years, Graham Company designs customized and effective property and casualty, surety and employee benefits programs for its clients to protect employees and prevent losses. With offices in Philadelphia, New York City and Washington, D.C., Graham Company became 100 percent employee-owned through an employee stock ownership plan in 2017. Through its innovative insurance and safety training programs, Graham Company is redefining what it means to be an insurance broker. To learn more, visit http://www.grahamco.com.

GRAHAM COMPANY IS AN INSURANCE BROKER AND NOT A MEDICAL CARE PROVIDER. ANY QUESTIONS CONCERNING THIS PROCEDURE SHOULD BE REFERRED TO REGENEXX OR YOUR MEDICAL CARE PROVIDER.

About Regenexx Regenexx is a nationwide network of physicians who practice Interventional Orthopedics, a new specialty that focuses on using the most advanced regenerative protocols available as an alternative to many orthopedic surgeries. Regenexx has published roughly half of the research worldwide on the use of orthobiologics for treating orthopedic injuries, and our patented treatment lab-processing and treatment protocols allow us to achieve unmatched results. Our procedures use your body's natural healing agents including blood platelets and bone marrow concentrate to repair damaged bone, muscle, cartilage, tendons and ligaments. We believe in educating patients, offering options and encouraging people to take an active role in their own treatment. For more information on Regenexx Corporate, visit http://www.regenexxcorporate.com or call 888-547-6667. For more information on the Regenexx procedures, visit http://www.regenexx.com.

View original content:http://www.prnewswire.com/news-releases/graham-company-and-regenexx-announce-innovative-partnership-to-reduce-orthopedic-surgical-spend-for-employers-301081460.html

SOURCE Regenexx

Originally posted here:
Graham Company and Regenexx Announce Innovative Partnership to Reduce Orthopedic Surgical Spend for Employers - Stockhouse

Stem Cell Cartilage Regeneration Market Overview By Growing Demands, Trends And Business Opportunities 2020 To 2027 – 3rd Watch News

Trusted Business Insights answers what are the scenarios for growth and recovery and whether there will be any lasting structural impact from the unfolding crisis for the Stem Cell Cartilage Regeneration market.

Trusted Business Insights presents an updated and Latest Study on Stem Cell Cartilage Regeneration Market 2019-2026. The report contains market predictions related to market size, revenue, production, CAGR, Consumption, gross margin, price, and other substantial factors. While emphasizing the key driving and restraining forces for this market, the report also offers a complete study of the future trends and developments of the market.The report further elaborates on the micro and macroeconomic aspects including the socio-political landscape that is anticipated to shape the demand of the Stem Cell Cartilage Regeneration market during the forecast period (2019-2029). It also examines the role of the leading market players involved in the industry including their corporate overview, financial summary, and SWOT analysis.

Get Sample Copy of this Report @ Global Stem Cell Cartilage Regeneration Market 2020 (Includes Business Impact of COVID-19)

Global Stem Cell Cartilage Regeneration Market Analysis Trends, Applications, Analysis, Growth, and Forecast to 2028 is a recent report generated by Trusted Business Insights. The global stem cell cartilage regeneration market report has been segmented on the basis of stem cell type, treatment, end user, and region.

Request Covid 19 Impact

Global Stem Cell Cartilage Regeneration Market: Overview

Stem cell-based therapy is important factor in functional replacement of cartilage regeneration. Stem cells are categorized into three kind of cells namely: hematopoietic stem cell, mesenchymal stem cells, and pluripotent stem cells.

Global Stem Cell Cartilage Regeneration Market: Dynamics

Rising adoption of minimally invasive surgery procedures, owing to its benefits such as lower risk and shorter duration for surgery is a key factor expected to drive growth of stem cell cartilage regeneration market over the forecast period.

In addition, increasing incidence of osteoarthritis among population across the globe, which in turn expected to support rising adoption of stem cell cartilage regeneration therapy. This factor is expected to boost growth of the target market over the forecast period.

Moreover, government initiatives such as government agencies which focusing on some policies in order to increase adoption of stem cell therapies is another factor expected to propel growth of target market over the forecast period.

However, higher treatment cost is a key factor expected to restrain growth of the global stem cell cartilage regeneration market over the forecast period. In addition, lack of skilled professionals to access stem cell sourcing, processing, or delivering these technologies is another factor expected to hamper growth of the target market over the forecast period.

Ongoing trend observed in the target market is increasing prevalence of osteoarthritis among population and rising adoption of minimally invasive surgery procedures which is turn expected to support growth of the global stem cell based cartilage regeneration market over the forecast period.

Increasing development of innovative technologies, products, and research and development (R&D) activities by key players are major factors expected to create potential revenue opportunities for the target market over the forecast period. In addition, increasing strategic partnership, merger, and acquisition activities by manufacturers are some other factors expected to create lucrative opportunities for player operating in the global market.

Global Stem Cell Cartilage Regeneration Market: Segment Analysis

Among the stem cell type segments, the mesenchymal stem cells segment is expected to register highest CAGR in the target market, owing to increasing research and development activities by various research organizations across the globe.

Global Stem Cell Cartilage Regeneration Market: Regional Analysis

The market in North America is expected to dominate in the global stem cell cartilage regeneration market in terms of revenue over the forecast period, owing to presence of key players operating in the target market such as Vericel Corporation, Osiris Therapeutics, among others and innovative stem cell based cartilage regeneration product launches, and development of new technologies across various countries in the region. In addition, rising geriatric population, which is more prone to degenerative disorders such as osteoarthritis across US is another factor expected to boost growth of the target market in this region.

The market in Asia Pacific is expected to register significant growth in terms of revenue over the forecast period, owing to stem cell based cartilage regeneration procedures are conducted by various hospital, specialty care, and clinics across India.

Global Stem Cell Cartilage Regeneration Market Segmentation:

Segmentation by stem cell type:

Hematopoietic Stem Cells (HSCs) Pluripotent Stem Cells (iPSC/ESCs) Mesenchymal Stem Cells (MSCs)

Segmentation by treatment:

Microfracture Osteochondral Transplant Autologous Chondrocyte Implantation Stem Cell Injections

Segmentation by end user:

Hospitals & Clinics Ambulatory Surgical Centers

Quick Read Table of Contents of this Report @ Global Stem Cell Cartilage Regeneration Market 2020 (Includes Business Impact of COVID-19)

Trusted Business Insights Shelly Arnold Media & Marketing Executive Email Me For Any Clarifications Connect on LinkedIn Click to follow Trusted Business Insights LinkedIn for Market Data and Updates. US: +1 646 568 9797 UK: +44 330 808 0580

More:
Stem Cell Cartilage Regeneration Market Overview By Growing Demands, Trends And Business Opportunities 2020 To 2027 - 3rd Watch News