Erythema Extent Predicts Death in Patients With ccGVHD – Medscape

Researchers are calling for the extent of skin erythema to be considered as an outcome measure in patients who develop chronic cutaneous graft-vs-host disease (ccGVHD) after allogeneic stem cell transplants for various blood cancers.

"There is value in collecting erythema serially over time as a continuous variable on a scale of 0%-100%" to identify high-risk patients for prophylactic and preemptive treatment, say investigators led by dermatologist Emily Baumrin, MD, director of the GVHD clinic at the University of Pennsylvania, Philadelphia.

They report a study of more than 300 patients with ccGVHD, which found that the extent of skin erythema strongly predicted the risk for death from GVHD.

Of the 267 patients with cutaneous GVHD at baseline, 103 patients died, the majority without a relapse of their blood cancer.

With additional research, erythema body surface area (BSA) should be "introduced as an outcome measure in clinical practice and trials," they conclude.

At the moment, the NIH Skin Score is commonly used for risk assessment in cutaneous GVHD, but the researchers found that erythema BSA out-predicts this score.

The investigators explain that the NIH Skin Score does incorporate erythema surface area, but it does so as a categorical variable, not a continuous variable. Among other additional factors, it also includes assessments of skin sclerosis, which the investigators found was not associated with GVHD mortality.

Overall, the composite score waters down the weight given to erythema BSA because the score is "driven by stable sclerotic features, and erythema changes are missed," they explain.

The study was published online on March 8 in JAMA Dermatology.

The study included 469 patients with chronic GVHD (cGVHD), of whom 267 (57%) had cutaneous cGVHD at enrollment and 89 (19%) developed skin involvement subsequently.

All of the patients were on systemic immunosuppression for GVHD after allogeneic stem cell transplants for various blood cancers.

They were enrolled from 2007 through 2012 at nine US medical centers all members of the Chronic Graft Versus Host Disease Consortium and followed until 2018.

Erythema BSA and NIH Skin Score were assessed at baseline and then every 3-6 months. Erythema was the first manifestation of skin involvement in the majority of patients, with a median surface area involvement of 11% at baseline.

The study team found that the extent of erythema at first follow-up visit was associated with both nonrelapse mortality (hazard ratio [HR], 1.33 per 10% BSA increase; P < .001) and overall survival (HR 1.28 per 10% BSA increase; P < .001), whereas extent of sclerotic skin involvement was not associated with either.

Participants in the study were predominantly White. The investigators note that "BSA assessments of erythema may be less reliable in patients with darker skin."

The work was funded by the US Department of Veterans Affairs and the National Institutes of Health. Baumrin had no disclosures; one co-author is an employee of CorEvitas, and two others reported grants/advisor fees from several companies, including Janssen, Mallinckrodt, and Pfizer.

JAMA Dermatol. Published online March 8, 2023. Abstract

M. Alexander Otto is a physician assistant with a master's degree in medical science and a journalism degree from Newhouse. He is an award-winning medical journalist who worked for several major news outlets before joining Medscape. Alex is also an MIT Knight Science Journalism fellow. Email: aotto@mdedge.com

For more news, follow Medscape on Facebook, Twitter, Instagram, and YouTube

Visit link:
Erythema Extent Predicts Death in Patients With ccGVHD - Medscape

Diagnostic Imaging Impacting DME and Wet AMD Treatment … – AJMC.com Managed Markets Network

Caesar Luo, MD, FASRS, FACS: What might be the biggest disrupter to our clinical flow is treatment for geographic atrophy and stem cell therapy/complement inhibition. Were at the cusp of geographic atrophy. That conversation is going to be big over the next 6 months to a year. Theres going to be a flood of conversations with payers and managed care and utilization. It is going to be a completely new ball of wax. It will be interesting to hear what the other panelists think, but Im expecting a deluge of new conversations and tricky situations to navigate.

Veeral Sheth, MD, MBA, FACS, FASRS: Thats a great point. One other thing Ill bring up that might be a paradigm shifter is home imaging, like Home OCT [optical coherence tomography]. We havent talked about it. Weve been talking about therapeutics, but diagnostics are equally important. If that means patients can be at home and we can monitor them there because we have to open up space for these patients with geographic atrophy who are going to be coming in. Theres a whole bunch of puzzle pieces that are moving around. Where are they going to end up? Were still going to find out.

Caesar Luo, MD, FASRS, FACS: Thats a great point.

Jim Kenney, RPh, MBA: For our audience, could you describe what OCT is, Dr Sheth?

Veeral Sheth, MD, MBA, FACS, FASRS: OCT is how we scan the retina. Its a pretty quick test. Were able to tell a lot of information from thatin particular, what the anatomy of the macula looks like. Is there fluid? Is there macular degeneration? Is there diabetic retinopathy? As a baseline, thats important. But also as we start to treat that patient and take them through their treatment journey, were going to be able to assess: how well are we treating them? How effectively are we treating them? In all this prior discussion, patients still have fluid. How do we know they have fluid? Its not just the examination, but a lot of the time its the imaging that were doing. If were able to do that imaging remotely at this patients home and get reports, were going to be able to tell if a patient is going in the wrong direction, maybe sooner than we would have if were just bringing them in on a certain interval.

Caesar Luo, MD, FASRS, FACS: Thats a great point. Treatment burden should be broken down into treatmentthe number of times Im sticking that patient with the needleand visits, how many times theyre coming in. Dr Coney, most of us do this treatment and extend regimen, and its the best of both worlds. Youre getting fewer shots and theyre coming into the office less. As-needed dosing didnt work that well. For patients who did better on as-needed dosing, they still have to come in every month. For these sustained drug-delivery systems, like port-delivery systems or potentially gene therapy, are we going to feel comfortable seeing them every 3 months? Probably not without something like Home OCT available to say if there might be some early fluid thats building up.

Joseph Coney, MD, FASRS, FACS: With these alternatives and other modalities of testing with artificial intelligence and Home OCT, we may be revisiting as-needed dosing. This is the best of both worlds if we have some type of feedback, and maybe immediate feedback, that patients can give. If they can just use their iPhone with an app, for example, and get feedback, they could say, You know what? This is what I saw yesterday. This is what Im seeing today. This is a smiley face. This is a sad face. I need to go in and be seen. With this new technology, there may be ways where we revisit as-needed dosing to decrease the number of patient visits to maximize their therapy while increasing the durability of these agents.

Jim Kenney, RPh, MBA: The challenge from the health plan side is that theres a phrase they like to use: We dont pay for convenience. We need to be able to drive the message that its not just convenient when you have an extended-dosing interval or something like that. Theres sustained damage to the eye and vision when youre not treating and the patient isnt adherent to therapy. We have to get beyond that initial barrier because there have been a lot of drugs that are brought to market over the years that were strictly for convenience. You didnt get better clinical results, as long as the patient was adherent with therapy. Thats an avenue that were going to have to focus some attention on to convince payers to open up access a little more for these therapies, and not simply view them as a convenience option rather than a true clinical advantage.

Joseph Coney, MD, FASRS, FACS: Jim, if we learned 1 thing during COVID-19, its that decreasing the number of patients in our office was really effective. Going forward, because of the overwhelming number of patients well be seeingparticularly those with geographic atrophywe need to find better ways to limit individuals from coming to the hospital. This isnt just for patients. For the most part, our patients, at least older patients, need caregivers. Individuals are taking off time from work. Thats lost income and lost taxes. These are things that we may not see from a treatment standpoint, but theres also a burden to the family. When these patients are receiving injections, its not just 1 to 2 hours in the office; its half the day. If thats a working population, that individual isnt going back to work. We need to find better ways to limit the burden and to monitor them so they can have the better lifestyles that we all want for ourselves.

Transcript edited for clarity.

See original here:
Diagnostic Imaging Impacting DME and Wet AMD Treatment ... - AJMC.com Managed Markets Network

FDA Accepts Mesoblast’s Resubmission of the Biologic License … – InvestorsObserver

FDA Accepts Mesoblasts Resubmission of the Biologic License Application for Remestemcel-L In Children with Steroid-Refractory Acute Graft Versus Host Disease as a Complete Response and Sets Goal Date of August 2, 2023

NEW YORK, March 07, 2023 (GLOBE NEWSWIRE) -- Mesoblast Limited (Nasdaq:MESO; ASX:MSB), global leader in allogeneic cellular medicines for inflammatory diseases, today announced that the United States Food and Drug Administrations (FDA) Office of Therapeutic Products (OTP) has accepted the Companys Biologics License Application (BLA) resubmission for remestemcel-L in the treatment of children with steroid-refractory acute graft versus host disease (SR-aGVHD). FDA considers the resubmission to be a complete response and has set a Prescription Drug User Fee Act (PDUFA) goal date of August 2, 2023.

Over the last two years we have worked tirelessly to address the issues previously raised by FDA. We look forward to working closely with the Agency over the review period with the aim to make remestemcel-L available as a therapy for children suffering from SR-aGVHD, said Mesoblast Chief Executive Silviu Itescu.

Survival outcomes have not improved over the past two decades for the most severe forms of SR-aGVHD, a life-threatening complication of an allogeneic bone marrow transplant following treatment for blood cancers and other conditions. 1- 3 The lack of any approved treatments for children under 12 means that there is an urgent need for a therapy that improves the dismal survival outcomes. If remestemcel-L receives FDA approval, it will be the first allogeneic off-the-shelf cellular medicine to be approved in the United States, and the first therapy for children under 12 years old with SR-aGVHD.

The resubmission contains the following new information:

FDA granted remestemcel-L Fast Track designation, a process to facilitate the development and expedited review of therapies for serious conditions that fill unmet medical needs, and Priority Review designation, which is given to drugs that treat a serious condition and provide a significant improvement in safety or effectiveness over existing treatments.

About Steroid-Refractory Acute Graft Versus Host Disease Survival outcomes have not improved over the past two decades for children or adults with the most severe forms of SR-aGVHD. 1- 3 The lack of any approved treatments for children under 12 means that there is an urgent need for a therapy that improves the dismal survival outcomes in children.

Acute GVHD occurs in approximately 50% of patients who receive an allogeneic bone marrow transplant (BMT). Over 30,000 patients worldwide undergo an allogeneic BMT annually, primarily during treatment for blood cancers, including about 20% in pediatric patients. 4,5 SR-aGVHD is associated with mortality as high as 90% and significant extended hospital stay costs. 6,7 There are currently no FDA-approved treatments in the US for children under 12 with SR-aGVHD.

About Remestemcel-L Mesoblasts lead product candidate, Remestemcel-L, is an investigational therapy comprising culture expanded mesenchymal stromal cells derived from the bone marrow of an unrelated donor. It is administered to patients in a series of intravenous infusions. Remestemcel-L is believed to have immunomodulatory properties to counteract the inflammatory processes that are implicated in SR-aGVHD by down-regulating the production of pro-inflammatory cytokines, increasing production of anti-inflammatory cytokines, and enabling recruitment of naturally occurring anti-inflammatory cells to involved tissues.

The original BLA submission contained clinical outcomes of 309 children with SR-aGVHD treated with remestemcel-L showing consistent treatment responses and survival across three separate trials. The data were reviewed by the agencys panel of the Oncologic Drugs Advisory Committee (ODAC) which voted in favor 9:1 that the available data support the efficacy of remestemcel-L in pediatric patients with SR-aGVHD.

The BLA resubmission now contains additional clinical and biomarker data, including from a propensity-matched study of children with high-risk disease, based on the validated MAP biomarker score, comparing outcomes in 25 children from Mesoblasts Phase 3 trial and 27 control children treated with various biologics, including ruxolitinib, from the Mount Sinai Acute GvHD International Consortium (MAGIC) database. The study showed that 67% of high-risk children treated with remestemcel responded positively to treatment within 28 days and were alive after 180 days compared to just 10% in both categories in the MAGIC group.

The BLA resubmission also contains results of a 4-year survival study performed by the Center for International Blood and Marrow Transplant Research (CIBMTR) on 51 evaluable patients with SR-aGVHD who were enrolled in the Phase 3 trial. The results demonstrated durability of the early day 180 survival benefits, with 63% survival at 1 year and 51% at 2 years in a group of children with predominantly grade C/D disease (89%) and with expected 2 year survival of just 25-38% using best available therapy. 1 ,8-9

About Mesoblast Mesoblast is a world leader in developing allogeneic (off-the-shelf) cellular medicines for the treatment of severe and life-threatening inflammatory conditions. The Company has leveraged its proprietary mesenchymal lineage cell therapy technology platform to establish a broad portfolio of late-stage product candidates which respond to severe inflammation by releasing anti-inflammatory factors that counter and modulate multiple effector arms of the immune system, resulting in significant reduction of the damaging inflammatory process.

Mesoblast has a strong and extensive global intellectual property portfolio with protection extending through to at least 2041 in all major markets. The Companys proprietary manufacturing processes yield industrial-scale, cryopreserved, off-the-shelf, cellular medicines. These cell therapies, with defined pharmaceutical release criteria, are planned to be readily available to patients worldwide.

Mesoblast is developing product candidates for distinct indications based on its remestemcel-L and rexlemestrocel-L allogeneic stromal cell technology platforms. Remestemcel-L is being developed for inflammatory diseases in children and adults including steroid refractory acute graft versus host disease, biologic-resistant inflammatory bowel disease, and acute respiratory distress syndrome. Rexlemestrocel-L is in development for advanced chronic heart failure and chronic low back pain. Two products have been commercialized in Japan and Europe by Mesoblasts licensees, and the Company has established commercial partnerships in Europe and China for certain Phase 3 assets.

Mesoblast has locations in Australia, the United States and Singapore and is listed on the Australian Securities Exchange (MSB) and on the Nasdaq (MESO). For more information, please see http://www.mesoblast.com , LinkedIn: Mesoblast Limited and Twitter: @Mesoblast

References / Footnotes

Forward-Looking Statements This press release includes forward-looking statements that relate to future events or our future financial performance and involve known and unknown risks, uncertainties and other factors that may cause our actual results, levels of activity, performance or achievements to differ materially from any future results, levels of activity, performance or achievements expressed or implied by these forward-looking statements. We make such forward-looking statements pursuant to the safe harbor provisions of the Private Securities Litigation Reform Act of 1995 and other federal securities laws. Forward-looking statements should not be read as a guarantee of future performance or results, and actual results may differ from the results anticipated in these forward-looking statements, and the differences may be material and adverse. Forward-looking statements include, but are not limited to, statements about: the initiation, timing, progress and results of Mesoblasts preclinical and clinical studies, and Mesoblasts research and development programs; Mesoblasts ability to advance product candidates into, enroll and successfully complete, clinical studies, including multi-national clinical trials; Mesoblasts ability to advance its manufacturing capabilities; the timing or likelihood of regulatory filings and approvals, manufacturing activities and product marketing activities, if any; the commercialization of Mesoblasts product candidates, if approved; regulatory or public perceptions and market acceptance surrounding the use of stem-cell based therapies; the potential for Mesoblasts product candidates, if any are approved, to be withdrawn from the market due to patient adverse events or deaths; the potential benefits of strategic collaboration agreements and Mesoblasts ability to enter into and maintain established strategic collaborations; Mesoblasts ability to establish and maintain intellectual property on its product candidates and Mesoblasts ability to successfully defend these in cases of alleged infringement; the scope of protection Mesoblast is able to establish and maintain for intellectual property rights covering its product candidates and technology; estimates of Mesoblasts expenses, future revenues, capital requirements and its needs for additional financing; Mesoblasts financial performance; developments relating to Mesoblasts competitors and industry; and the pricing and reimbursement of Mesoblasts product candidates, if approved. You should read this press release together with our risk factors, in our most recently filed reports with the SEC or on our website. Uncertainties and risks that may cause Mesoblasts actual results, performance or achievements to be materially different from those which may be expressed or implied by such statements, and accordingly, you should not place undue reliance on these forward-looking statements. We do not undertake any obligations to publicly update or revise any forward-looking statements, whether as a result of new information, future developments or otherwise.

Release authorized by the Chief Executive.

For more information, please contact:

Here is the original post:
FDA Accepts Mesoblast's Resubmission of the Biologic License ... - InvestorsObserver

Researchers discover the role of intestinal fibrosis in inflammatory … – Drug Target Review

The scientists used a new in vitro platform, which allowed intestinal organoids to be cultured on an open lumen, planar system that could be manipulated experimentally.

Intestinal fibrosis is a common feature of inflammatory bowel disease (IBD) and the primary cause of end-stage organ failure. Traditionally considered a bystander of inflammation, with negligible involvement in disease pathogenesis, new research published inGastroenterologynow shows that fibrosis has a direct bearing on disease progression in IBD.

The investigation was spearheaded by researchers from Massachusetts General Hospital (MGH) and Harvard Medical School, both US.

The critical question posed by the investigators was how tissue stiffening influences the growth and differentiation of intestinal stem cells, which fuel the regeneration of intestinal epithelium?

This was addressed by developing a newin vitroplatform, which allowed intestinal organoids to be cultured on an open lumen, planar system that could be manipulated experimentally.

The platform permitted the use of soft yet tunable substrates with biophysical properties mimicking native tissue, facilitating the long-term growth and differentiation of intestinal stem cells, like native epithelium.

The team discovered that upon elevating substrate stiffness to a similar range observed in IBD patients, both the number and capacity of stem cells to maintain homeostasis and cellular composition of the epithelium were potently reduced.

Concomitantly, the stem cells preferentially differentiated into goblet cells, leading to epithelial deterioration. Similar phenotypes were also noted in mouse models of IBD as well as in samples from human patients.

The investigators concluded that interfering with the molecular machinery involved in the cellular sensing of stiffness conferred protection against the detrimental effects of fibrosis and stiffening.

These findings demonstrate that intestinal fibrosis and stiffening are critical components of IBD pathogenesisand that targeting mechanosensing and mechanotransduction pathways may offer an attractive therapeutic strategy for IBD, said lead author Dr Nima Saeidi.

The scientists also observed that despite the significant reduction in a specific population of stem cells, stiffening led to the expansion of another stem cell marker (OLFM4) outside the stem cell zone.

Our observations that stiffening increased the expression of OLFM4 may have significant implications for the development of colitis-associated colorectal cancer, said co-first author Dr Shijie He.

A collaborative work between scientists from the Massachusetts General Hospital, MIT, Boston Childrens Hospital, Harvard T.H. Chan School of Public Health, and Boston University, all US.

This research was supported by the US National Institute of Diabetes and Digestive and Kidney Diseases.

See the original post here:
Researchers discover the role of intestinal fibrosis in inflammatory ... - Drug Target Review

Scientists inject stem cells into the brain of Parkinsons patient – Freethink

A new stem cell therapy for Parkinsons disease has just been administered to a person for the first time and if it works as hoped, it could revolutionize how doctors treat the disease.

[W]e maybe have a treatment that we can offer to patientsin the beginning of the disease, like a one time treatment, and that will last for the rest of the patients life and make it possible to reduce the medication that patients otherwise need, said principal investigator Gesine Paul-Visse from Swedens Lund University.

The challenge: An estimated 8.5 million people are living with Parkinsons, a progressive neurodegenerative disorder caused by the loss of the brain neurons that produce the chemical dopamine, which helps coordinate movement.

The shortage of dopamine leads to the hallmark symptoms of Parkinsons, including tremors, stiffness, and impaired coordination. Medications can increase dopamine levels, but they can also cause side effects, interfere with other meds, and become less effective over time.

The use of stem cells will, in theory, enable us to make unlimited amounts of dopamine neurons.

The idea: Rather than relying on meds, Paul-Visse and her collaborators in Sweden and the UK hope to actually replace dopamine-producing nerve cells in the brains of Parkinsons patients using embryonic stem cells, which can develop into almost any type of cell in the body.

For their therapy, STEM-PD, the researchers programmed stem cells sourced from donated embryos to turn into dopamine nerve cells. When transplanted into the brains of rodent models of Parkinsons, the cells developed as hoped, and the animals motor symptoms were reversed.

The researchers have now administered the treatment to a person for the first time, and by the end of their newly launched STEM-PD trial, eight people with moderate Parkinsons will undergo the therapy.

Looking ahead: The trials primary goal is to assess the safety of STEM-PD, but the researchers will also be looking to see if the therapy improves symptoms, reduces the need for medication, or leads to the development of new dopamine-producing neurons in the brain.

The efficacy of the treatment wont be apparent right away, though.

These cells that we are transplanting are actually immature, so they need some time to mature in the adult brain, and that will take at least a year, maybe even longer, said Paul-Visse. So we wont expect to see any changes before in one years time.

The big picture: Treatments that work in animals often dont translate to people, but if STEM-PD proves safe and effective, the impact could be huge, given that stem cells can be duplicated an unlimited number of times.

The use of stem cells will, in theory, enable us to make unlimited amounts of dopamine neurons and thus opens the prospect of producing this therapy to a wide patient population, said clinical lead Roger Barker from the University of Cambridge. This could transform the way we treat Parkinsons disease.

Wed love to hear from you! If you have a comment about this article or if you have a tip for a future Freethink story, please email us at tips@freethink.com.

Read the rest here:
Scientists inject stem cells into the brain of Parkinsons patient - Freethink

How organoids are advancing the understanding of chronic kidney … – Nature.com

Kidney-like structures called organoids can be grown from stem cells.Credit: Xia Lab

Ryuichi Nishinakamuras quest to build a transplantable kidney began in the 1990s, when the nephrologist found he had little to offer his patients. At times he was ridiculed for setting such an unrealistic goal, but I was very naive and young, so I just went forward, recalls Nishinakamura, who is now a stem-cell biologist at Kumamoto University in Japan.

But the discoveries of human embryonic stem cells in 1998 and of a way to create induced pluripotent stem (iPS) cells in 2006 made the task of growing fresh kidneys seem more achievable. Many investigators differentiated various types of kidney cell from iPS cells and grew kidney organoids tiny, organ-like structures with multiple types of kidney cell that partly mimic kidney structure and function. In 2013, Nishinakamuras lab achieved one of the early milestones, demonstrating both mouse and human kidney organoids1. Many labs are now producing ever-more functional organoids that are proving useful in modelling kidney development and disease.

Part of Nature Outlook: Chronic kidney disease

But Nishinakamuras goal of a transplantable human kidney is still many years away. We do have kidney in a dish, says Melissa Little, a developmental biologist at the Murdoch Childrens Research Institute in Melbourne, Australia. But will it be useful if we transplant it? Thats a much bigger question.

We are at a bottleneck, says Yun Xia, a stem-cell biologist at Nanyang Technological University in Singapore, who sees an enormous gap between todays research and what people need. Like many of her colleagues, Xia worries that to keep its credibility with funders and the public, organoid research needs to show progress towards treatments. Some groups are working towards auxiliary kidneys, which would be a fraction of the size of a normal kidney but could still stabilize a persons health.

The kidney is an exceptionally tricky organ to replicate in a lab. Youve got 2530 distinct cell types with functional roles that have to be anatomically placed in the right position for the organ to work, says Little. By contrast, the heart is thought to have only nine major cell types2.

The kidneys functional unit for removing waste from the blood, the nephron, is an intricate and precisely organized structure. The first step in filtering the blood takes place in networks of small blood vessels called glomeruli. The resulting filtrate then passes through a series of tubes, in which various solutes are exchanged with blood vessels, before ending up in a branching tree of collecting ducts that funnels the waste to the ureter and out towards the bladder. For a kidney to function, it is not enough to simply have the right cells they must also be arranged correctly.

A number of daunting obstacles remain on the road to a transplantable kidney. One of the biggest is immaturity of the cells, which typically resemble progenitor cells from the first or second trimester of human development, limiting their functionality.

There has been steady progress on this front. In 2022, for instance, Little and her colleagues demonstrated more functional human proximal tubule cells, which she calls the powerhouses of the kidney3. The lab of nephrologist Joseph Bonventre at Brigham and Womens Hospital in Boston, Massachusetts, did the same that year for the collecting ducts two main functional cell types4.

Researchers have also worked out how to boost their ability to create kidney organoids in volume, another key requirement for potential treatments. For example, researchers in the Netherlands have grown sheets of iPS-cell-derived nephrons at a large scale5.

Like other forms of tissue derived from pluripotent stem cells, kidney organoids can include undesired off-target cells such as muscle neurons, and researchers need to follow precise protocols to guard against the appearance of tumour cells. General advances in the stem-cell field are minimizing these challenges.

Forming a vasculature, however, is a much greater hurdle. A fully developed and precisely structured blood system is needed to keep the flows of blood and urine exchanging correctly throughout the nephron. This has not been achieved in experimental systems, says Jamie Davies, a developmental biologist and tissue engineer at the University of Edinburgh, UK. Instead, the vasculature generally remains in a primitive state and soon dies out.

Organoids transplanted into immunodeficient mice do attract blood vessels from the host animal, allowing nephrons to start filtering the blood and generating urine, says Nishinakamura. However, the urine has nowhere to go, so transplants typically fail at that stage, he says.

The push to build better organoids based on iPS cells has vastly increased researchers understanding of kidney development and disease. Compared with cell cultures, organoids already offer enhanced models of kidney disease particularly for genetic illnesses in children. For example, the crucial cells that wrap around glomerulus capillaries and begin the filtering process, called podocytes, are rubbish in 2D cell cultures but much better representations in 3D organoids, Little says.

Organoid models also readily display the characteristic cysts of autosomal dominant polycystic kidney disease the most prevalent genetic kidney disease and one subject to intense research. One 2022 study reported a scalable human kidney organoid platform that enabled the testing of hundreds of small-molecule drugs against this condition6.

Researchers are now able to grow kidney organoids with more-functional cells.Credit: J. M. Vanslambrouck et al. Nature Commun. 13, 5943 (2022).

Diabetes is the largest driver of chronic disease in adults but a formidable task to model, because the condition impairs the blood vessels that are difficult to reproduce in organoids. Moreover, says Xia, kidney organoids, like many cell cultures, are generally bathed in high levels of glucose, making it hard to pick out the effects of the raised blood glucose levels generated in diabetes.

Kidney organoids show great promise in drug testing. Many drug candidates fail testing because they cause kidney damage, but this is not picked up in 2D cell cultures because they often lose their sensitivity, says Bonventre. For example, he says, the protein KIM-1 is a strong biomarker for damage to proximal tubule cells in vivo but not in 2D cell culture. If kidney organoids can display the same KIM-1 gene-expression patterns that are seen in vivo, they will provide excellent toxicity models, he says. His lab is studying such organoid-based models.

Using organoids based on iPS cells as a treatment for kidney disease is far from the first cell-based therapy to be proposed. Many clinical trials have tested the effect of mesenchymal stem cells (multipotent stem cells found in tissue, such as bone marrow), with mixed results. Most researchers agree that although these cells might secrete factors that help with kidney repair, they dont structurally improve the kidney. One long-studied alternative technique that selects, enhances and reinserts kidney cells from people with kidney disease is being examined in a phase III clinical trial sponsored by the biotechnology company ProKidney in Winston-Salem, North Carolina.

But injecting iPS-cell-based organoid-derived cells alone into kidneys doesnt seem to be a promising strategy, says Nishinakamura. Such cells might secrete factors that improve kidney function, much as mesenchymal stem cells are thought to do, he says. But these progenitor cells are unlikely to stay and play happily within the kidney; its not clear where the cells might go, or if and how they then mature.

Organoid-derived cells might help when it comes to improving transplants of donated kidneys, says Nria Montserrat, a stem-cell biologist at the Institute for Bioengineering of Catalonia in Barcelona, Spain. She is testing that hypothesis in collaboration with Cyril Moers, a transplant surgeon at the University of Groningen in the Netherlands. Donated organs are often maintained before transplant by being perfused in a liquid bath rather than frozen. Moers hopes that adding organoid-derived cells to these baths will allow these organs to be preserved better, evaluated more accurately and (eventually) made healthier before transplant. Montserrats lab is running pilot experiments with human organoid cells released into perfused pig kidneys.

More broadly, a number of groups are studying the potential for transplanting a more substantial set of organoid tissues into people with kidney disease. Little wants to create what she calls an auxiliary kidney, designed to connect to a persons failing kidney.

In her labs unpublished experiments, human kidney organoids transplanted into immunocompromised mice successfully gather blood vessels and start filtering urine. Getting all those nephrons to connect to the underlying kidney will be the challenge, she says. If the nephrons connect into the existing kidney itself, then the urine will go out the way all of the urine goes. Youre essentially freeloading on the anatomy of the existing patient kidney, even though that patients kidney is pretty sick.

More from Nature Outlooks

Biotechnology company Trestle Biotherapeutics in San Diego, California, is also developing a transplantable auxiliary kidney. Co-founder and developmental biologist Alice Chen says that this tissue might end up in another location, such as below the existing kidney near the bladder. Trestle is growing organoids about 100 times the size of those commonly reported in the scientific literature, she says, and is seeing encouraging progress in how these tissues engraft in mice, connect to the host circulation and continue to mature.

The start-up was launched with the view that a bioengineered kidney will demand industrial-scale expertise in many fast-evolving disciplines, including stem-cell science and 3D bioprinting. We had to pull all of that brainpower, those technologies and those ideas together, says Chen.

Most people with kidney disease are hoping for treatments that let them live their lives replacing or minimizing dialysis, or postponing the immediate need for a donated kidney rather than for a complete bioengineered organ. Were not creating an entire organ, we need to create some sort of tissue that can return 1020% function to these patients, Chen says. And that is achievable.

Some research groups are using organoids as potential sources of cells for hybrid external devices with bioengineered 3D scaffolds, designed to act like improved dialysis systems.

In one such effort, Bonventre hopes to make a device with a sub-population of just two types of cell: proximal tubule cells and collecting duct cells. Other types of kidney cells remain important, he says, but achieving every function of a normal kidney seems like a distant goal. Lets not shoot for a galaxy thats three billion light years away, he says. Lets try to get to the Moon first, and maybe Mars.

But Nishinakamura remains fixed on his original dream of a complete kidney, which he thinks is needed more than ever for the millions of people whose chronic kidney disease steadily progresses towards end-stage renal disease. Im always telling my graduate students, Dont say it is impossible.

Read more from the original source:
How organoids are advancing the understanding of chronic kidney ... - Nature.com

Why CRISPR babies are still too risky embryo studies highlight … – Nature.com

A human embryo at the 16-cell stage sits on the tip of a pin. Researchers say genome-editing techniques are still not safe enough to be used in embryos destined for reproduction.Credit: Dr Yorgos Nikas/Science Photo Library

More than four years after the first children with edited genomes were born, genome-editing techniques are still not safe enough to be used in human embryos that are destined for reproduction, organizers of the Third International Summit on Human Genome Editing announced at the conclusion of the meeting.

Heritable human genome editing remains unacceptable at this time, they said in a statement issued on 8 March. Preclinical evidence for the safety and efficacy of heritable human genome editing has not been established, nor has societal discussion and policy debate been concluded.

Beyond CRISPR babies: how human genome editing is moving on after scandal

The statement came at the end of a day of discussion and debate in London about the potential of altering the genomes of either embryos or reproductive cells, called gametes, in ways that would be passed down to future generations. Many of the talks at the meeting were focused on technical and scientific challenges, such as the uncertain consequences of breaking both strands of the DNA double helix a necessary step in some forms of genome editing in embryos.

In addition to those challenges, society must grapple with questions about whether the technology should be deployed, organisers said: Governance frameworks and ethical principles for the responsible use of heritable human genome editing are not in place.

Some researchers have argued that heritable genome editing could help people who carry genetic diseases to avoid passing those conditions on to their children. In many cases, this can already be done by combining in vitro fertilization (IVF) with testing of the resulting embryos for a given genetic disorder. But that is not always an option, such as when all a couples embryos will inevitably inherit the genetic disorder, or when all available embryos happen to carry the responsible genes.

In addition to broader concerns about ethics and social justice, editing embryos would require a safe and effective genome-editing platform to minimize the chances of harming the embryo, the resulting child, and that childs descendants. Most research on genome editing in embryos, however, has been done using animal models such as mice, which might not accurately reflect what happens in human embryos. And although potential genome-editing therapies have been widely studied in adult human cells, embryos might respond differently than adult cells to the DNA damage caused by some genome editing tools.

Only a handful of laboratories have directly tried to edit the genomes of human embryos using the popular editing system CRISPR-Cas9, and several of these presented concerning results at the summit.

CRISPR gene editing in human embryos wreaks chromosomal mayhem

The Cas9 enzyme works by breaking both strands of DNA at a site designated by a guiding piece of RNA. The cell then repairs that DNA break, either by using an error-prone mechanism that stitches the two ends together but sometimes deletes or inserts a few DNA letters in the process, or by replacing the missing DNA with a sequence copied from a template provided by the researcher. DNA breaks created by Cas9 in embryos are usually repaired using the error-prone pathway, rather than using the template DNA, said Deitrich Egli, a stem cell biologist at Columbia University in New York City, at the conference.

Egli and other researchers also reported on the consequences of the double-strand breaks made by Cas9. Developmental biologist Kathy Niakan at the University of Cambridge, UK, recounted her labs experience with the apparent loss of large regions of chromosomes that occurred when using CRISPR-Cas9 to edit human embryos1. Shoukhrat Mitalipov, a reproductive biologist at Oregon Health & Science University in Portland, also said that his laboratory had found large DNA deletions at the editing site in human embryos, and that these deletions might not be detected using standard tests2.

Can human embryos at this stage really tolerate this kind of intervention? asked Dagan Wells, a reproductive geneticist at the University of Oxford, UK, who also reported concerning responses to DNA breaks in human embryos. About 40% of the embryos in one of his genome-editing studies failed to repair broken DNA. Over one-third of those embryos continued to develop, he said, resulting in the loss or gain of pieces of chromosomes in some cells. That could risk the health of offspring if such embryos were allowed to develop further. These results are really a warning, he said.

There are newer variations on CRISPR-Cas9 editing that do not break both strands of the DNA helix. Base editing, for example, can convert one single DNA letter to another, and a technique called prime editing allows researchers to insert DNA sequences more predictably than CRISPR-Cas9 editing. Neither of these methods cause double-strand breaks, but they have not been as thoroughly studied and optimized as CRISPR-Cas9. At the summit, developmental biologist Yuyu Niu at the Kunming University of Science and Technology in China reported that one kind of base editor did not cause off-target DNA mutations in rhesus macaque (Macaca mulatta) embryos, but it did cause unwanted RNA mutations3.

Super-precise CRISPR tool enhanced by enzyme engineering

An alternative to editing embryos would be to instead edit gametes, such as eggs and sperm, or the stem cells that give rise to them. This would also sidestep concerns that efforts to edit embryos might not succeed in all cells of the embryo, resulting in offspring that contain a mixture of edited and unedited cells. Several researchers at the summit reported progress towards generating gametes in the laboratory, but doing this with human cells destined for reproductive uses still poses challenges.

The summit organizers urged that researchers continue to explore each of these options, even as policy makers and the public grapple with what restrictions should be placed on heritable genome editing.. We are still keen that the research goes ahead, said developmental biologist Robin Lovell-Badge of the Francis Crick Institute in London, who chaired the organizing committee for the summit. In parallel, there has to be more debate about whether the technique is ever used.

Read this article:
Why CRISPR babies are still too risky embryo studies highlight ... - Nature.com

Study reveals limitations in evaluating gene editing technology in … – OHSU News

Gene editing technologies hold promise in preventing and treating debilitating inherited diseases, however new research from Oregon Health & Science University reveals limitations that must be overcome before gene-editing to establish a pregnancy can be deemed safe or effective. (OHSU/Sara Hottman)

A commonly used scientific method to analyze a tiny amount of DNA in early human embryos fails to accurately reflect gene edits, according to new research led by scientists at Oregon Health & Science University.

The study, published today in the journal Nature Communications, involved sequencing the genomes of early human embryos that had undergone genome editing using the gene-editing tool CRISPR. The work calls into question the accuracy of a DNA-reading procedure that relies on amplifying a small amount of DNA for purposes of genetic testing.

In addition, the study reveals that gene editing to correct disease-causing mutations in early human embryos can also lead to unintended and potentially harmful changes in the genome.

Together, the findings raise a new scientific basis for caution for any scientist who may be poised to use genetically edited embryos to establish pregnancies. Although gene editing technologies hold promise in preventing and treating debilitating inherited diseases, the new study reveals limitations that must be overcome before gene-editing to establish a pregnancy can be deemed safe or effective.

Shoukhrat Mitalipov, Ph.D. (OHSU)

It tells you how little we know about editing the genome, and particularly how cells respond to the DNA damage that CRISPR induces, said senior author Shoukhrat Mitalipov, Ph.D., director of the OHSU Center for Embryonic Cell and Gene Therapyand, professor of obstetrics and gynecology, molecular and cellular biosciences, OHSU School of Medicine, OHSU Oregon National Primate Research Center. Gene repair has great potential, but these new results show that we have a lot of work to do.

The findings come during the Third International Summit on Human Genome Editing in London. On the eve of the last international summit, held in Hong Kong in November 2018, a Chinese scientist revealed the birth of the worlds first babies resulting from gene-edited embryos through an experiment that generated global condemnation.

Interview with Shoukhrat Mitalipov, Ph.D., in the Mitalipovlab at OHSU. (OHSU/Sara Hottman)

Before an edited embryo can be transferred to establish a pregnancy, it is important to make sure the procedure worked as intended.

Because early human embryos consist of just a few cells, its not possible to collect enough genetic material to effectively analyze them. Instead, scientists interpret data from a small sample of DNA taken from a few or even a single cell, which then must be multiplied millions of times during a process known as whole genome amplification.

The same process known as preimplantation genetic testing, or PGT is often used to screen human embryos for various genetic conditions in patients undergoing in vitro fertilization.

Paula Amato, M.D.,(OHSU)

Whole genome amplification has limitations that reduce the accuracy of genetic testing, said senior co-author Paula Amato, M.D., professor of obstetrics and gynecology in the OHSU School of Medicine.

The concern is that we might be misdiagnosing embryos, Amato said.

Amato, who uses in vitro fertilization to treat patients struggling with infertility as well as to prevent the transmission of inherited diseases, said PGT using more advanced technology is still clinically useful for detecting chromosomal abnormalities and genetic disorders caused by a single gene mutation transmitted from parent to child.

The study highlights the challenges of establishing the safety of gene-editing techniques.

We may not be able to reliably predict that this embryo will result in a healthy baby, Mitalipov said. Thats a major problem.

To overcome these issues, OHSU researchers, along with collaborators with research institutions in South Korea and China, established embryonic stem cell lines from gene-edited embryos. Embryonic stem cells grow indefinitely and provide ample DNA material that does not require whole genome amplification to analyze.

Researchers say the discovery highlights the error-prone nature of whole genome amplification and the need to verify edits in embryos by establishing embryonic stem cell lines.

Using embryonic stem cells, the new study verifies the process of gene repair that Mitalipovs lab developed; the findings were published in the journal Naturein 2017 and verifiedin 2018.

In that study, scientists cut a specific target sequence on a mutant gene known to be carried by a sperm donor.

Researchers found that human embryos repair these breaks, using the normal copy of the gene from the other parent as a template. Mitalipov and co-authors confirmed that this process, known as gene conversion, occurs regularly in early human embryos following a double-strand break in their DNA. Such a repair, if used to establish a pregnancy through in vitro fertilization and embryo transfer, could theoretically prevent a known familial disease from being passed on to the child, as well as all future generations of the family.

In the study published in 2017, the OHSU researchers targeted a gene known to cause a deadly heart disease.

In this new publication, researchers targeted other discrete mutations using donated sperm and eggs, including one mutation known to cause hypertrophic cardiomyopathy, a condition in which the heart muscle becomes abnormally thick, and a different one associated with high cholesterol. In each case an enzyme known as Cas9, used in tandem with CRISPR, induced a double-strand break in DNA at the precise site of the mutation.

In addition to replicating and confirming the gene-repair mechanism reported in 2017, the new study examines what happens in the genome beyond the specific site where the mutant gene is repaired. And thats where a problem can occur.

In this paper we asked, how extensive is that gene conversion repair mechanism? Amato said. It turns out that it can be very lengthy.

Extensive copying of the genome, from one parent to the other, creates a scenario known as loss of heterozygosity.

Every human being shares two versions, or alleles, of every gene on the human genome one contributed from each parent. Most of the time, the alleles are identical, given 99.9% of any individuals DNA sequence is shared with the rest of humanity. In some cases, however, one parent will carry a recessive disease-causing mutation thats normally canceled out by the other parents dominant healthy version of the same gene.

These polymorphisms in the genetic code can be critically important. For example, a gene may encode a protein that protects against specific types of cancer.

If you have one abnormal copy of a recessive mutation, that may pose no risk, Amato said. But if you have loss of heterozygosity leading to two mutant copies of the same tumor suppressor gene, now youre at significantly increased risk for cancer.

The more genetic code thats copied, the greater the risk of dangerous genetic changes. In the new study, scientists measured gene conversion tracts ranging from a relatively small segment to as large as 18,600 base pairs of DNA.

In effect, the repair of one known mutation may create more problems than it solves.

If youre cutting in the middle of a chromosome, there could be 2,000 genes there, Mitalipov said. Youre fixing one tiny spot, but all these thousands of genes upstream and downstream may be affected.

The finding suggests that much more research is needed to understand the mechanism at work in gene-editing before using it clinically to establish a pregnancy.

Studies conducted at the OHSU Center for Embryonic Cell and Gene Therapy were supported by OHSU institutional funds and a grant from the Burroughs Wellcome Fund.

Original post:
Study reveals limitations in evaluating gene editing technology in ... - OHSU News

Stem Cell Assay Market :How the Market will perform in Upcoming … – Digital Journal

The Stem Cell Assay Market Industry research projection to 2023-2030 offers thorough industry data to help companies create growth plans and make smarter business choices based on predictions and market trends. The dynamic market structure, the product offerings of major players, their challenges, technical innovation, impediments and barriers, data on communication and sales, sales by country, risk, prospects, the competitive environment, growth strategy, and others are among the marketing variables covered in the study. It goes into great detail regarding the present and future conditions of the market. The report examines a range of elements, such as technology advancements, technological levels, and the various business models employed by the markets leading competitors at the moment.

Stem cells are the most fundamental type of biological cells. They have the ability to develop into multiple cell types and can multiply into more of the same type of stem cell. Adult stem cells and embryonic stem cells are the two different kinds of stem cells. These cells can be found in the bone marrow, adipose tissue, and blood, among other body parts. Also harvested from umbilical cord blood are stem cells. Two processesobligatory asymmetric replication and stochastic differentiationare used to maintain the bodys stem cell population. The introduction of stem cells has demonstrated promising outcomes in the treatment of numerous disorders, including cancer. Stem cells play a significant role in the bodys natural healing process.

Get Sample Report With Covid-19 Impact Analysis: https://www.coherentmarketinsights.com/insight/request-sample/1632

List of Key players in the global Stem Cell Assay Market: Merck & Co., Thermo Fisher Scientific, GE Healthcare, Agilent Technologies, Bio-Rad Laboratories, Promega Corporation, Cell Biolabs, PerkinElmer, Miltenyi Biotec, HemoGenix, Bio-Techne Corporation, STEMCELL Technologies, and Cellular Dynamics International.

SWOT Analysis of Global Stem Cell Assay Market

The report focuses on revenue analysis and SWOT analysis in addition to market share analysis of businesses, detailed profiles, product/service overviews, and business overviews to better correlate market competitiveness.

Information source and Research Methodology:

Using both primary (surveys and interviews) and secondary (industry body databases, dependable paid sources, and trade periodicals) data collection techniques, our researchers put the study together. A complete qualitative and quantitative analysis is included in the report. The research covers growth trends, micro- and macroeconomic indicators, as well as legislation and governmental programmes.

Buy This Premium Report With Amazing Offer : https://www.coherentmarketinsights.com/insight/buy-now/1632

Purchasing the Stem Cell Assay Market for the Following Reasons:

The study looks at new market trends as well as the chance that different trends will have an impact on growth.The report also covers the elements, difficulties, and chances that will significantly affect the worldwide Stem Cell Assay market.Standards and technological instruments that take the Stem Cell Assay industrys predicted expansion into account.To forecast future growth rates, the research comprises a thorough analysis of market information as well as past and present growth trends.In order to produce forecasts for the future growth, the research comprises a thorough review of market statistics as well as historical and present growth conditions.

What are the goals of the report?

This market report displays the estimated market size for the Stem Cell Assay Market Industry at the end of the projected period.The research also examines historical and contemporary market sizes.The graphs display the compound annual growth rate (CAGR) and year-over-year growth (percent) for the specified forecast time.The study includes a market overview, geographic scope, segmentation, and financial results of the major rivals.The research evaluates the current situation of the industry inNorth America, Asia Pacific, Europe, Latin America, the Middle East, and Africa,as well as future growth opportunities.The study examines the future periods growth rate, market size, and market worth.

Some of the Key Questions Answered in this Report:

Table of Content

Chapter 1 Industry Overview1.1 Definition1.2 Assumptions1.3 Research Scope1.4 Market Analysis by Regions1.5 Market Size Analysis from 2023 to 203011.6 COVID-19 Outbreak: Stem Cell Assay Industry Impact

Chapter 2 Stem Cell Assay Competition by Types, Applications, and Top Regions and Countries2.1Market (Volume and Value) by Type2.3Market (Volume and Value) by Regions

Chapter 3 Production Market Analysis3.1 Global Production Market Analysis3.2 Regional Production Market Analysis

Chapter 4 Stem Cell Assay Sales, Consumption, Export, Import by Regions (2016-2022)Chapter 5 North AmericaIndustry Market AnalysisChapter 6 East Asia Stem Cell Assay Market AnalysisChapter 7 EuropeIndustry Market AnalysisChapter 8 South Asia Stem Cell Assay Market AnalysisChapter 9 Southeast Asia Market AnalysisChapter 10 Middle East Stem Cell Assay Market AnalysisChapter 11 Africa Market AnalysisChapter 12 Oceania Market AnalysisChapter 13 South America Stem Cell Assay Market AnalysisChapter 14 Company Profiles and Key Figures in Stem Cell Assay BusinessChapter 15 Stem Cell Assay Market Forecast (2023-2030)Chapter 16 ConclusionsResearch MethodologyContinued.

Buy This Premium Report With Amazing Offer : https://www.coherentmarketinsights.com/insight/buy-now/1632

Finally, the report majorly enlightens the key growth and limiting factors which majorly targets at the center of the market affecting the growth and its development in either positive or negative extent. The report also specifies the impact of regulations and policies implemented by the administration over the current growth and upcoming opportunities that may lead to the market development escalation. The Stem Cell Assay Market report offers a superior vision of the global market, which will help clients to manage the business precisely with better growth and expansion compared to its contenders in the market.

Read Our More Report:

Cell surface markers detection market

Car t cell therapy market

Bovine plasma market

Gmp protein e coli contract manufacturing market

About Coherent Market Insights

Coherent Market Insights is a global market intelligence and consulting organization that provides syndicated research reports, customized research reports, and consulting services. We are known for our actionable insights and authentic reports in various domains including aerospace and defense, agriculture, food and beverages, automotive, chemicals and materials, and virtually all domains and an exhaustive list of sub-domains under the sun. We create value for clients through our highly reliable and accurate reports. We are also committed in playing a leading role in offering insights in various sectors post-COVID-19 and continue to deliver measurable, sustainable results for our clients.

Contact:

Mr Shah

Coherent Market Insights533 Airport Boulevard, Suite 400,Burlingame, CA 94010, United States.Email: [emailprotected]United States of America: +1-206-701-6702United Kingdom: +44-020-8133-4027Japan: +050-5539-1737India: +91-848-285-0837

Go here to see the original:
Stem Cell Assay Market :How the Market will perform in Upcoming ... - Digital Journal

Leading innovators in cell therapy for ocular disorders for the … – Pharmaceutical Technology

The pharmaceutical industry continues to be a hotbed of innovation, with activity driven by the evolution of new treatment paradigms, and the gravity of unmet needs, as well as the growing importance of technologies such as pharmacogenomics, digital therapeutics, and artificial intelligence. In the last three years alone, there have been over 633,000 patents filed and granted in the pharmaceutical industry, according to GlobalDatas report on Innovation in Pharmaceuticals: Cell therapy for ocular disorders.

However, not all innovations are equal and nor do they follow a constant upward trend. Instead, their evolution takes the form of an S-shaped curve that reflects their typical lifecycle from early emergence to accelerating adoption, before finally stabilising and reaching maturity.

Identifying where a particular innovation is on this journey, especially those that are in the emerging and accelerating stages, is essential for understanding their current level of adoption and the likely future trajectory and impact they will have.

110 innovations will shape the pharmaceutical industry

According to GlobalDatas Technology Foresights, which plots the S-curve for the pharmaceutical industry using innovation intensity models built on over 756,000 patents, there are 110 innovation areas that will shape the future of the industry.

Within the emerging innovation stage, cell therapy for ocular disorders, coronavirus vaccine components, and DNA polymerase compositions are disruptive technologies that are in the early stages of application and should be tracked closely. Adeno-associated virus vectors, alcohol dehydrogenase compositions, and antibody serum stabilisers are some of the accelerating innovation areas, where adoption has been steadily increasing. Among maturing innovation areas are anti-influenza antibody compositions and anti-interleukin-1, which are now well established in the industry.

Innovation S-curve for the pharmaceutical industry

Cell therapy for ocular disorders is a key innovation area in pharmaceuticals

Stem cells have the capacity to revive degenerated cells or replace cells. Various cell types have been used as the source of therapeutic cells, including human embryonic stem cells (hESCs), induced pluripotent stem cells (iPSCs), and human umbilical tissue-derived cells (hUTCs). The regeneration, proliferation and differentiation potential of stem cells result in therapeutic intervention in different kinds of eye disease, including age-related macular degeneration (AMD), inherited retinal diseases (IRDs), glaucoma, and corneal diseases.

GlobalDatas analysis also uncovers the companies at the forefront of each innovation area and assesses the potential reach and impact of their patenting activity across different applications and geographies. According to GlobalData, there are 30+ companies, spanning technology vendors, established pharmaceutical companies, and up-and-coming start-ups engaged in the development and application of cell therapy for ocular disorders.

Key players in cell therapy for ocular disorders a disruptive innovation in the pharmaceutical industry

Application diversity measures the number of different applications identified for each relevant patent and broadly splits companies into either niche or diversified innovators.

Geographic reach refers to the number of different countries each relevant patent is registered in and reflects the breadth of geographic application intended, ranging from global to local.

Senju Pharmaceutical is the leading patent holder of cell therapies for ocular disorders. The company has filed a number of patents covering various cell therapies for the treatment of ophthalmic disorders. One notable patent is a miR-203 inhibitor for corneal epithelial disorder.

In terms of application diversity, Acro Biomedical is the top company, followed by Daiichi Sankyo, and Mayo Clinic. By means of geographic reach, Intellia Therapeutics holds the top position, while HLB Co Ltd, and Mayo Clinic stand in second and third positions, respectively.

To further understand the key themes and technologies disrupting the pharmaceutical industry, access GlobalDatas latest thematic research report on Pharmaceutical.

Get industry leading news, data and analysis delivered to your inbox

GlobalData, the leading provider of industry intelligence, provided the underlying data, research, and analysis used to produce this article.

GlobalDatas Patent Analytics tracks patent filings and grants from official offices around the world. Textual analysis and official patent classifications are used to group patents into key thematic areas and link them to specific companies across the worlds largest industries.

Originally posted here:
Leading innovators in cell therapy for ocular disorders for the ... - Pharmaceutical Technology