Category Archives: Stem Cell Medicine


Home | Eli and Edythe Broad Center of Regeneration …

In more than 140 labs across UCSF, scientists are carrying out studies in cell culture and animals with the goal of understanding and developing treatment strategies for such conditions as heart disease, diabetes, epilepsy, multiple sclerosis, Parkinsons disease, Lou Gehrigs disease, spinal cord injury and cancer.

The Broad Center is structured around eight research pipelines aimed at driving discoveries from the lab bench to the patient. Each pipeline focuses on a different organ system, including the blood, pancreas, liver, heart, reproductive organs, nervous system, musculoskeletal tissues, skin and eyes. Each of these pipelines is overseen by two leaders of international standing one representing the basic sciences and one representing clinical research. This approach has proven successful in the private sector for driving the development of new therapies.

Like all of UCSF, the Center fosters a highly collaborative culture, encouraging the cross-pollination of ideas between scientists of different disciplines and years of experience. Researchers studying pancreatic beta cells damaged in diabetes collaborate with those studying nervous system diseases, because at the heart of their research are stem cells that undergo similar molecular signaling on the way to becoming both cell types. The opportunity to work in this culture has drawn some of the countrys premier scientists to the center.

UCSF Mourns the Loss of EliBroad(1933-2021)

The UC San Francisco community is deeply saddened to learn of the passing of EliBroad, a renowned entrepreneur and philanthropist whose generosity supported scientific and medical research, the arts, and high-quality educational opportunities for students across the U.S.

The Eli and EdytheBroadCenter of Regeneration Medicine and Stem Cell Research at UCSF will be forever grateful to Mr.Broadfor his extraordinary vision and generosity. His investments in UCSFs stem cell endeavors have enabled our scientists to accelerate our research, by bringing some of the worlds leading stem cell scientists together under one roof and providing them with a setting that promotes collaboration and an exchange of ideas, both key to making clinical advances to improve human health. His legacy will live on through the breakthroughs and improvements in patient care made possible by his support of our work.

In fact, Mr.Broads impact on stem cell science at UCSF and beyond will be felt for generations to come. Along with his wife of over 60 years, Edye, Mr.Broadsupported stem cell research at a time when the country most needed national leadership in this area of scientific inquiry. Eager to leverage his philanthropic dollars for maximum impact, Mr.Broadsaw an opportunity to fund stem cell research when Californians passed a proposition funding $3 billion in bonds to support stem cell research and research facilities in 2004. Shortly after President George W. Bush vetoed a bill that would have supported federal funding of stem cell research, the couples philanthropic organization, The Eli and EdytheBroadFoundation, made an initial investment of $65 million to create three newBroadStem Cell Centers at UCSF, UCLA, and the University of Southern California. The Foundation has since made supplemental gifts bringing their total contribution to these centers to $113 million. The efforts have made California a leading center of stem cell research in the country.

To learn more about the remarkable life of EliBroad, please visitthis link.

In Memoriam: Katja Brueckner, PhD

In Memoriam: Zena Werb, MD, PhD

See the article here:
Home | Eli and Edythe Broad Center of Regeneration ...

Rising Focus on Exploring Potential of Stem Cells as Therapeutic Tools in Drug Targeting and Regenerative Medicine to Fuel Revenue Growth of Stem…

NEW YORK, Jan. 10, 2022 /PRNewswire/ --Reports and Data has published its latest report titled "Stem Cells Market By Product (Adult Stem Cells, Human Embryonic Stem Cells, IPS Cells, and Very Small Embryonic-Like Stem Cells), By Technology (Cell Acquisition, Cell Production, Cryopreservation, and Expansion & Sub-Culture), By Therapies (Allogeneic Stem Cell Therapy and Autologous Stem Cell Therapy), and By Application (Regenerative Medicine and Drug Discovery & Discovery), and By Region Forecast To 2028."

According to the latest report by Reports and Data, the global stem cells market size was USD 10.13 billion in 2020 and is expected to reach USD 19.31 Billion in 2028 and register a revenue CAGR of 8.4% during the forecast period, 2021-2028.

Request a Sample Reporthttps://www.reportsanddata.com/sample-enquiry-form/2981

Drivers, Restraints, & Opportunities

Stem cells are cells that have the potential to differentiate into different types of cells in the body. Stem cells have the ability of self-renewal and differential into specialized adult cell types. Stems cells are being explored for their potential in tissue regeneration and repair and in treatment of chronic diseases. Increasing number of clinical trials are underway to assess and establish safety and efficacy of stem cell therapy for various diseases and disorders. Rapid advancement in stem cell research, rising investment to accelerate stem cell therapy development, and increasing use of stem cells as therapeutic tools for treatment of neurological diseases and malignancies are some key factors expected to drive market revenue growth over the forecast period. in addition, growing incidence of type 1 diabetes, spinal cord injuries, Parkinson's diseases, and Alzheimer's disease, among others have further boosted adoption of stem cell therapies and is expected to fuel revenue growth of the market going ahead.

Stem cells are basic cells in the body from which cells with specialized functions are generated such as heart muscle cells, brain cells, bone cells, or blood cells. Maturation of stem cells into specialized cells have enabled researchers and doctors better understand the pathophysiology of diseases and conditions. Stem cells have great potential to be grown to become new tissues for transplant and in regenerative medicine. Stem cells that are programmed to differentiate into tissue-specific cells are widely being used to test new drugs that target specific diseases, such as nerve cells can be generated to test safety and efficacy of drugs that are being developed for nerve disorders and diseases. Stem cells are of two major types: pluripotent cells that can differentiate into any cells in the adult body and multipotent cells that are restricted to differentiate into limited population of cells. Increasing clinical research is being carried out to advance stem cell therapy to improve cardiac function and to treat muscular dystrophy and heart failure. Recent progress in preclinical and clinical research have expanded application scope of stem cell therapy into treating diseases for which currently available therapies have failed to be effective. This is expected to continue to drive revenue growth of the market going ahead.

However, immunity-related concerns associated with stem cell therapies, increasing incidence of abnormalities in adult stem cells, and rising number of ethical issues associated with stem cell research such as risk of harm during isolation of stem cells, therapeutic misconception, and concerns surrounding safety and efficacy of stem cell therapies are some key factors expected to restrain market growth to a certain extent over the forecast period.

To identify the key trends in the industry, research study at https://www.reportsanddata.com/report-detail/stem-cells-market

COVID-19 Impact Analysis

Rising use of Human Embryonic Stem Cells in Regenerative Medicine to Drive Market Growth:

Human embryonic stem cells (ESCs) segment is expected to register significant revenue growth over the forecast period attributable to increasing use of human embryonic stem cells in regenerative medicine and tissue repair, rising application in drug discovery, and growing importance of embryonic stem cells as in vitro models for drug testing.

Cryopreservation Segment to Account for Largest Revenue Share:

Cryopreservation segment is expected to dominate other technology segments in terms of revenue share over the forecast period. Cryopreservation techniques are widely used in stem cell preservation and transport owing to its ability to provide secure, stable, and extended cell storage for isolated cell preparations. Cryopreservation also provides various benefits to cell banks and have numerous advantages such as secure storage, flexibility and timely delivery, and low cost and low product wastage.

Regenerative Medicine Segment to Lead in Terms of Revenue Growth:

Regenerative medicine segment is expected to register robust revenue CAGR over the forecast period attributable to significant progress in regenerative medicine, increasing research and development activities to expand potential of stem cell therapy in treatment of wide range of diseases such as neurodegenerative diseases, diabetes, and cancers, among others, and rapid advancement in cell-based regenerative medicine.

Buy Premium Research Report @ https://www.reportsanddata.com/checkout-form/2981

North America to Dominate Other Regions in Terms of Revenue Share:

North America is expected to dominate other regional markets in terms of revenue share over the forecast period attributable to increasing adoption of stem cell therapy to treat chronic diseases, rising investment to accelerate stem cell research, approval for clinical trials and research studies, growing R&D activities to develop advanced cell-based therapeutics, and presence of major biotechnology and pharmaceutical companies in the region.

Asia Pacific Market Revenue to Expand Significantly:

Asia Pacific is expected to register fastest revenue CAGR over the forecast period attributable to increasing R&D activities to advance stem cell-based therapies owing to rapidly rising prevalence of chronic diseases such as cancer and diabetes, rising investment to accelerate development of state-of-the-art healthcare and research facilities, establishment of a network of cell banks, increasing approval for regenerative medicine clinical trials, and rising awareness about the importance of stem cell therapies in the region.

Major Companies in the Market Include:

Download Report Summary @ https://www.reportsanddata.com/download-summary-form/2981

Market Segmentation:

For the purpose of this report, Reports and Data has segmented the stem cells market based on product, technology, therapies, application, and region:

Product Outlook (Revenue, USD Billion; 2018-2028)

Technology Outlook (Revenue, USD Billion; 2018-2028)

Therapy Outlook (Revenue, USD Billion; 2018-2028)

Application Outlook (Revenue, USD Billion; 2018-2028)

Ask for Customize Research Report @ https://www.reportsanddata.com/request-customization-form/2981

Regional Outlook (Revenue, USD Billion, 2018-2028)

Some points on how the report benefits stakeholders:

Explore Trending Research Reports by Reports and Data:

Genotyping assay marketsize was USD 17.7 Billion in 2020 and is expected to register a robust CAGR of 22.4% during the forecast period. Key factors driving market revenue growth are increasing prevalence of genetic disorders such as Turner syndrome, Alzheimer's, hemophilia, and Parkinson's, and rising demand for genotyping assays globally.

Nurse call system marketsize was USD 1.61 Billion in 2020 and is expected to register a revenue CAGR of 8.8% during the forecast period. Increasing need to improve communication among doctors and nurses, growing focus on reducing patient disturbances, and higher adoption of real-time location systems are key factors expected to drive market revenue growth over the forecast period. Moreover, increasing need to improve patient response time is another factor expected to drive market growth in the near future.

Cellular health screening marketsize was USD 2.84 billion in 2020 and is expected to register a CAGR of 10.3% during the forecast period. Increasing application of cellular health screening in precision medicine, rising emphasis on preventive healthcare, rapidly and growing geriatric population globally are key factors expected to drive market revenue growth over the forecast period.

Live cell encapsulation marketsize was USD 269 million in 2020 and is expected to register a CAGR of 3.6% during the forecast period. Key factors, such as increasing awareness about cell encapsulation techniques to treat several chronic disorders and growing need for clinical potency of cell encapsulation techniques, are escalating global market revenue growth.

Blood preparation marketsize was USD 41.30 billion in 2020 and is expected to register a CAGR of 5.8% during the forecast period. The global market is projected to gain rapid traction in the upcoming years owing to various favorable factors.

About Reports and Data

Reports and Data is a market research and consulting company that provides syndicated research reports, customized research reports, and consulting services. Our solutions purely focus on your purpose to locate, target, and analyze consumer behavior shifts across demographics, across industries, and help clients to make smarter business decisions. We offer market intelligence studies ensuring relevant and fact-based research across multiple industries, including Healthcare, Touch Points, Chemicals, Products, and Energy. We consistently update our research offerings to ensure our clients are aware of the latest trends existent in the market. Reports and Data has a strong base of experienced analysts from varied areas of expertise. Our industry experience and ability to develop a concrete solution to any research problems provides our clients with the ability to secure an edge over their respective competitors.

Contact:

John W Head of Business Development Reports And Data | Web: http://www.reportsanddata.com Direct Line: +1-212-710-1370 E-mail: [emailprotected] LinkedIn | Twitter | Blogs

Read Latest Press Release @ https://www.reportsanddata.com/press-release/global-stem-cells-market

SOURCE Reports And Data

Read more here:
Rising Focus on Exploring Potential of Stem Cells as Therapeutic Tools in Drug Targeting and Regenerative Medicine to Fuel Revenue Growth of Stem...

Wealth of Newer Treatment Options Span the Scope of Multiple Myeloma and Amyloidosis – OncLive

Tomer Mark, MD, discusses the emerging role of minimal residual disease as a surrogate end point in multiple myeloma, treatment selection for patients with newly diagnosed, early relapsed, and late relapsed disease, and the importance of daratumumab being introduced to the treatment paradigm of amyloidosis.

Treatment selection for patients with newly diagnosed, early relapsed, and late relapsed multiple myeloma requires consideration of patient-, disease-, and treatment-related factors, said Tomer Mark, MD, who added that the need for minimal residual disease (MRD) as a surrogate end point for survival is stronger now with optimal sequencing strategies remaining uncertain.

Multiple myeloma is moving very fast right now. There are a lot of new agentsthat are exciting, including oral options. There is an embarrassment of riches in terms of treatment for patients with multiple myeloma, so we have to sift through this treasure to find the best treatment for the right patient at the right time, said Mark in an interview with OncLive during an Institutional Perspectives in Cancer (IPC) webinar on multiple myeloma.

During the interview, Mark, an associate professor of medicine, clinical director of the Plasma Cell Disorders Program, and clinical director of the Autologous Stem Cell Transplant Program, in the Department of Medicine-Hematology, at the University of Colorado (UC) School of Medicine, of UCHealth, discussed the emerging role of MRD as a surrogate end point in multiple myeloma, treatment selection for patients with newly diagnosed, early relapsed, and late relapsed disease, and the importance of daratumumab (Darzalex) being introduced to the treatment paradigm of amyloidosis.

Mark: We are seeing the continued theme of MRD and how important it is as an end point. Although not quite universally accepted as a surrogate marker for survival, it certainly is much closer than what we had before with the IMWG [International Myeloma Working Group] criteria. It is good to see that more studies are using MRD as an end point. Also, its good that more patients are reaching that end point, even in the relapsed/refractory setting. We are seeing that in the CAR T-cell therapy studies with idecabtagene vicleucel [ide-cel; Abecma] and ciltacabtagene autoleucel [cilta-cel] that close to 50% of patients are reaching MRD negativity. Extended results with the use of monoclonal antibodies in the up-front setting, like the GRIFFIN study [NCT02874742], also show continued MRD negativity with continued treatment. Getting these very deep responses is the theme now, which is refreshing. Now our problem returns full circle to sequencing, which used to be a big issue a few years ago.

The message is that incorporation of a monoclonal antibody as part of induction therapy provides a great benefit that is durable to patients. Incorporating the monoclonal antibodies [into frontline treatment] is going to become the standard of care. The data we saw at [the 2021 ASH Annual Meeting and Exposition] simply reinforced those results.

The ICARIA-MM [NCT02990338] and IKEMA [NCT03275285] studies looking at isatuximab-irfc [Sarclisa] in combination with either carfilzomib [Kyprolis] or pomalidomide [Pomalyst] showed outstanding results in terms ofnearly a halving of the risk of progression at any point in time. The hazard ratio was between 0.5 and 0.6 in both studies.

Subgroup analyses were refreshing in that age and cytogenetics didnt seem to have much of an effect [on survival] or at least not as much of an effect as they used to with older agents. The theme continues of using monoclonal antibodies early on in therapy. One point to make about that is that sequential CD38-directed antibodies, no matter the order of daratumumab or isatuximab first followed by the other, does not seem to be an effective strategy. There needs to be a period between those sorts of therapies.

That is the major question [in this setting]. There are clinical trials looking at moving CAR T-cell therapy, as well as bispecific antibodies, closer to the frontline [setting]. We are still learning how to manage toxicities from these therapies, like cytokine release syndrome and immune effector cellassociated neurotoxicity syndrome. As we gather more data, these things are getting more predictable and we are managing them better; however, we are keeping patients in the hospital for 2 weeks to get their CAR T cells because we just dont know what will happen. With bispecific antibodies, we have step-up dosing because we just dont know what will happen. We need more experience with these agents.

That said, eventually, CAR T-cell therapy will become part of the standard of care at some point. Just like autologous stem cell transplant, [CAR T-cell therapy] requires some planning, so CAR T-cell therapy may be used in a more elective situation, not as a rescue therapy for patients. Perhaps, the decision will be made based on MRD status after induction. That has yet to be determined.

In emergent situations, we can use bispecific antibodies, which are the off-the-shelf T-cell harnessing therapies. There are practical reasons for using [bispecific antibodies] vs [CAR T-cell therapy].

One nice thing we saw is that we can give sequential antiBCMA-directed therapy, which is the opposite of what we know about [sequential] monoclonal antibodies. That means that we dont have to be as careful in terms of planning whether we are going to give belantamab mafodotin followed by antiBCMA-directed CAR T-cell therapy vs the other way around. Of course, there are some issues with belantamab mafodotin, such as corneal toxicities. The results of belantamab mafodotin in combination with dexamethasone were disappointing, although the later DREAMM trials [evaluating belantamab mafodotin] in combination with bortezomib [Velcade] showed much better results.

It seems that belantamab mafodotin is not destined to be a single-agent therapy and it will be used in combination. It is off-the-shelf, so that is a plus given that we need an ophthalmologic exam before each dose, which is a big minus. In an academic or large hospital where the ophthalmologist is down the hall, that isnt a problem, but in the community clinics where the ophthalmologist is across town, that isnt as practical. These things get baked into the equation.

When we look at comparisons over time, it seems that cilta-cel is a bit more effective than ide-cel. Id like to exercise caution [comparing] apples and oranges. The ide-cel population [comprised] sicker patients in general with higher-risk cytogenetics and higher-risk multiple myeloma. I imagine that if ide-cel were used in a very similar patient population to [the one in which] cilta-cel [was evaluated] this difference might not be so great. We are comparing 2 amazing therapies. For ide-cel, we have seen response rates around 80% with durations of response [DOR] lasting close to 1 year. With cilta-cel, we are looking at 90% to 100% response rates in some studies and a DOR not yet reached.

I get asked this question all the time: Which therapy should I give to give to my patients? The answer is whichever is available. There are manufacturing and supplier problems, as well as slot allocation problems for apheresis and lymphocyte collection. We are grabbing whatever opportunity we get.

It was amazing to perform such a large, randomized trial in AL amyloidosis. I am proud to have participated in this study. It has probably set the world record for the fastest accrual to an amyloidosis trial. That shows the faith that we have in daratumumab working.

The addition of daratumumab to [AL amyloidosis] treatment is probably as significant as the first use of proteasome inhibitors in amyloid treatment. It really is a sea change, and the difference in terms of progression-free survival and organ recovery is so profound that there is no question that daratumumab should be part of frontline care for AL amyloidosis. This is especially [true] for patients who are not transplant eligible.

In transplant-eligible patients, it is more of a question of whether we should transplant patients more up front and then [continue] later with antibody therapy. That is going to be a practical matter in terms of [evaluating] plasma-cell burden, how sick are they, and how many organs are affected. Overall, outcomes for [patients with] amyloidosis are much better now that we have this treatment.

We are working on developing an ex-vivo drug testing platform for patients with multiple myeloma. It is well known that myeloma is not one disease, it is many diseases, which accounts for the variety of outcomes we can see with therapy. This is an effort to tailor the therapy to that patients particular myeloma phenotype, meaning how it behaves in real life. This is opposed to risk-adapted therapy where we make guesses based on high-risk cytogenetics or mutations.

Of course, this only accounts for one aspect of a disease in which there are multiple mutations and clones going on at the same time. Our institution is focused on getting primary patient myeloma samples, real myeloma cells as opposed to cell lines, putting them in cultures with various drugs being tested, and making a prescription for the patient based on this. This is all experimental and more work needs to be done. We are just now doing our translational studies where we are testing this principle out in a clinical trial setting.

Go here to read the rest:
Wealth of Newer Treatment Options Span the Scope of Multiple Myeloma and Amyloidosis - OncLive

Companies In The Regenerative Medicine For Cartilage Market Increase R&D On Gene Therapy Technique As Per – Benzinga

LONDON, Jan. 11, 2022 (GLOBE NEWSWIRE) -- According to The Business Research Company's research report on the regenerative medicine for cartilage market, gene therapies are most promising technique to effectively treat and regenerate the damaged cartilage in case of bone and joint injuries and severe bone deformities. Gene therapy is defined as the treatment and management of disease by the introduction of foreign genes or sequences of encoded proteins into different type of cells by using gene transfer technique. Many manufacturers constantly evaluate the feasibility and safety of this technique in clinical trials.

For instance, in 2017, Kolon TissueGene, Inc received marketing authorization for its gene therapy product Invossa (TissueGene-C) a cell mediated gene therapy that contains non-transduced human chondrocytes (hChonJ) and transduced (hChonJb#7) human allogeneic chondrocytes. The hChonJb#7 cells were transduced with TGF-1 gene by using retroviral vector and irradiated with gamma-ray, in South Korea, based on Phase 1 and Phase 2 clinical studies treating patients with moderate knee OA. In 2020, the US Food and Drug Administration (FDA) allowed the continuation of the Phase 3 clinical trial of Invossa, gene therapy for osteoarthritis, in the US.

The global regenerative medicine for cartilage market reached a value of nearly $4.83 billion in 2021 at a compound annual growth rate (CAGR) of 8.10%. Regenerative medicine for cartilage market growth is mainly due to the companies resuming their operations and adapting to the new normal while recovering from the COVID-19 impact. The market is expected to reach $6.56 billion in 2025 at a CAGR of 7.94%.

LAST WEEK of our huge market research report sale! Grab your deal now.

The global regenerative medicine for cartilage market is fragmented, with a few large players constituting significant market share. The top ten competitors in the market made up to 24.69% of the total market in 2020. The market consolidation can be attributed to the high barriers to entry in terms of high costs associated with the production of regenerative medicine for cartilage. Major players in the regenerative medicine for cartilage market are B. Braun Melsungen AG, Zimmer Biomet Holdings Inc., Vericel Corporation, Stryker Corporation, Smith & Nephew plc, Arthrex Inc., CONMED Corporation, Collagen Solutions PLC, BioTissue Technologies, CellGenix, Osiris Therapeutics Inc., and DePuy Synthes.

Request for a sample of the global regenerative medicine for cartilage market report

TBRC's regenerative medicine for cartilage market analysis report segments the market by treatment modality into cell-based, and non-cell-based; by treatment type into palliative, intrinsic repair stimulus, and others; by site into knee cartilage repair, ribs, and others; by application into hyaline cartilage repair and regeneration, elastic cartilage repair and regeneration, and fibrous cartilage repair and regeneration; by end use into ambulatory surgical centers, hospitals & clinics, and others.

Pharmaceutical companies and federal governments are increasingly working together in partnerships and collaborations to provide funding and implement incentive programs for the research and development (R&D) of regenerative medicine for cartilage. These partnerships provide financial and technical assistance across different clinical development phases to pharmaceutical companies. As per the regenerative medicine for cartilage market trends, in 2021, Government of Canada along with various provincial government have announced multiple investment initiative to setup flexible biomanufacturing capacity and new biotech innovation hubs.

As per TBRC's regenerative medicine for cartilage market research, North America was the largest region in the regenerative medicine for cartilage market, accounting for 52.4% of the total in 2021. It was followed by the Western Europe, Asia Pacific and then the other regions. Going forward, the fastest-growing regions in the regenerative medicine for cartilage market will be Middle East and Africa where growth will be at CAGRs of 31.9% and 30.7% respectively during 2021-2026. These will be followed by Eastern Europe and Asia Pacific.

Regenerative Medicine For Cartilage Market Global Report 2022: Market Size, Trends, And Global Forecast 2022 - 2026 is one of a series of new reports from The Business Research Company that provide regenerative medicine for cartilage market overviews, regenerative medicine for cartilage market analyze and forecast market size and growth for the whole market, regenerative medicine for cartilage market segments and geographies, regenerative medicine for cartilage market trends, regenerative medicine for cartilage market drivers, regenerative medicine for cartilage market restraints, regenerative medicine for cartilage market leading competitors' revenues, profiles and market shares in over 1,000 industry reports, covering over 2,500 market segments and 60 geographies.

The report also gives in-depth analysis of the impact of COVID-19 on the market. The reports draw on 150,000 datasets, extensive secondary research, and exclusive insights from interviews with industry leaders. A highly experienced and expert team of analysts and modelers provides market analysis and forecasts. The reports identify top countries and segments for opportunities and strategies based on market trends and leading competitors' approaches.

Not the market you are looking for? Check out some similar market intelligence reports:

Stem Cell Therapy Global Market Report 2021 - By Type (Allogeneic Stem Cell Therapy, Autologous Stem Cell Therapy), By Cell Source (Adult Stem Cells, Induced Pluripotent Stem Cells, Embryonic Stem Cells), By Application (Musculoskeletal Disorders, Wounds And Injuries, Cancer, Autoimmune Disorders), By End-User (Hospitals, Clinics), COVID-19 Growth And Change

Gene Therapy Global Market Report 2021 - By Gene Type (Antigen, Cytokine, Suicide Gene), By Vector (Viral Vector, Non-Viral Vector), By Application (Oncological Disorders, Rare Diseases, Cardiovascular Diseases, Neurological Disorders, Infectious Diseases), By End Users (Hospitals, Homecare, Specialty Clinics), COVID-19 Growth And Change

Orthobiologics Devices And Equipment Global Market Report 2021 - By Product Type (Demineralized Bone Matrix, Synthetic Bone Substitutes, Stem Cell Therapy, Plasma-Rich Protein, Viscosupplementation, Bone Morphogenetic Protein, Synthetic Orthobiologics), By Application (Osteoarthritis And Degenerative Arthritis, Soft-Tissue Injuries, Fracture Recovery, Maxillofacial And Dental Applications, Spinal Fusion, Trauma Repair, Reconstructive Surgery), By End Use (Hospitals, Orthopedic Clinics), COVID-19 Impact And Recovery

Interested to know more about The Business Research Company?

The Business Research Company is a market intelligence firm that excels in company, market, and consumer research. Located globally it has specialist consultants in a wide range of industries including manufacturing, healthcare, financial services, chemicals, and technology.

The World's Most Comprehensive Database

The Business Research Company's flagship product, Global Market Model, is a market intelligence platform covering various macroeconomic indicators and metrics across 60 geographies and 27 industries. The Global Market Model covers multi-layered datasets which help its users assess supply-demand gaps.

See the rest here:
Companies In The Regenerative Medicine For Cartilage Market Increase R&D On Gene Therapy Technique As Per - Benzinga

Regenerative Medicine: The Promise Of Undoing The Ravages Of Time – Hackaday

In many ways, the human body is like any other machine in that it requires constant refueling and maintenance to keep functioning. Much of this happens without our intervention beyond us selecting what to eat that day. There are however times when due to an accident, physical illness or aging the automatic repair mechanisms of our body become overwhelmed, fail to do their task correctly, or outright fall short in repairing damage.

Most of us know that lizards can regrow tails, some starfish regenerate into as many new starfish as the pieces which they were chopped into, and axolotl can regenerate limbs and even parts of their brain. Yet humans too have an amazing regenerating ability, although for us it is mostly contained within the liver, which can regenerate even when three-quarters are removed.

In the field of regenerative medicine, the goal is to either induce regeneration in damaged tissues, or to replace damaged organs and tissues with externally grown ones, using the patients own genetic material. This could offer us a future in which replacement organs are always available at demand, and many types of injuries are no longer permanent, including paralysis.

Our level of understanding of human physiology and that of animals in general has massively expanded since the beginning of the 20th century when technology allowed us to examine the microscopic world in more detail than ever before. Although empirical medical science saw its beginnings as early as the Sumerian civilization of the 3rd millennium BCE, our generalized understanding of the processes and components that underlie the bodys functioning are significantly more recent.

DNA was first isolated in 1869 by Friedrich Miescher, but its structure was not described until 1953. This discovery laid the foundations for the field of molecular biology, which seeks to understand the molecular basis for biological activity. In a sense this moment can be seen as transformative as for example the transition from classical mechanics to quantum mechanics, in that it changed the focus from macroscopic observations to a more fundamental understanding of these observations.

This allowed us to massively increase our understanding of how exactly the body responds to damage, and the molecular basis for regenerative processes, as well as why humans are normally not able to regrow damaged limbs. Eventually in 1999 the term regenerative medicine was coined by William A. Haseltine, who wrote an article in 2001 on what he envisions the term to include. This would be the addressing of not only injuries and trauma from accidents and disease, but also aging-related conditions, which would address the looming demographic crisis as the average age of the worlds populations keeps increasing.

The state of the art in regenerative medicine back in 2015 was covered by Angelo S. Mao et al. (2015). This covers regenerative methods involving either externally grown tissues and organs, or the stimulating of innate regenerative capabilities. Their paper includes the biomedical discipline of tissue engineering due to the broad overlap with the field of regenerative medicine. Despite the very significant time and monetary requirement to bring a regenerative medicine product to market, Mao et al. list the FDA-approved products at that time:

While these were not miracle products by any stretch of the imagination, they do prove the effectiveness of these approaches, displaying similar or better effectiveness as existing products. While getting cells to the affected area where they can induce repair is part of the strategy, another essential part involves the extracellular matrix (ECM). These are essential structures of many tissues and organs in the body which provide not only support, but also play a role in growth and regeneration.

ECM is however non-cellular, and as such is seen as a medical device. They play a role in e.g. the healing of skin to prevent scar tissue formation, but also in the scaffolding of that other tantalizing aspect of regenerative medicine: growing entire replacement organs and body parts in- or outside of the patients body using their own cells. As an example, Mase Jr, et al. (2010) report on a 19-year old US Marine who had part of his right thigh muscle destroyed by an explosion. Four months after an ECM extracted from porcine (pig) intestinal submucossa was implanted in the area, gradual regrowth of muscle tissue was detected.

An important research area here is the development of synthetic ECM-like scaffolding, as this would make the process faster, easier and more versatile. Synthetic scaffolding makes the process of growing larger structures in vitro significantly easier as well, which is what is required to enable growing organs such as kidneys, hearts and so on. These organs would then ideally be grown from induced pluropotent stem cells (iPS), which are a patients own cells that are reverted back to an earlier state of specialization.

It should come as little surprise that as a field which brings together virtually every field that touches upon (human) biology in some fashion, regenerative medicine is not an easy one. While its one thing to study a working system, its a whole different level to get one to grow from scratch. This is why as great as it would be to have an essentially infinite supply of replacement organs by simply growing new ones from iPS cells, the complexity of a functional organ makes this currently beyond our reach.

Essentially the rule is that the less complicated the organ or tissue is, the easier it is to grow it in vitro. Ideally it would just consist out of a single type of cell, and happy develop in some growth medium without the need for an ECM. Attractive targets here are for example the cornea, where the number of people on a waiting list for a corneal transplant outnumber donor corneas significantly.

In a review by Mobaraki et al. (2019), the numerous currently approved corneal replacements as well as new methods being studied are considered. Even though artificial corneas have been in use for years, they suffer from a variety of issues, including biocompatibility issues and others that prevent long-term function. Use of donor corneas comes with shortages as the primary concern. Current regenerative research focuses on the stem cells found in the limbus zone (limbal stem cells, LSC). These seem promising for repairing ocular surface defects, which has been studied since 1977.

LSCs play a role in the regular regenerative abilities of the cornea, and provide a starting point for either growing a replacement cornea, or to repair a damaged cornea, along with the addition of an ECM as necessary. This can be done in combination with the inhibiting of the local immune response, which promotes natural wound healing. Even so, there is still a lot more research that needs to be performed before viable treatments for either repairing the cornea in situ, or growing a replacement in vitro can be approved the FDA or national equivalent.

A similar scenario can be seen with the development of artificial skin, where fortunately due to the large availability of skin on a patients body grafts (autografts) are usually possible. Even so, the application of engineered skin substitutes (ESS) would seem to be superior. This approach does not require the removal of skin (epidermis) elsewhere, and limits the amount of scar formation. It involves placing a collagen-based ECM on the wound, which is optionally seeded with keritanocytes (skin precursor cells), which accelerates wound closure.

Here the scaffolding proved to be essential in the regeneration of the skin, as reported by Tzeranis et al. (2015). This supports the evidence from other studies that show the cell adhesion to the ECM to be essential in cell regulation and development. With recent changes, it would seem that both the formation of hair follicles and nerve innervation may be solved problems.

It will likely still be a long time before we can have something like a replacement heart grown from a patients own iPS cells. Recent research has focused mostly on decellularization (leaving only the ECM) of an existing heart, and repopulating it with native cells (e.g. Glvez-Montn et al., 2012). By for example creating a synthetic scaffold and populating it with cells derived from a patients iPS cells, a viable treatment could be devised.

Possibly easier to translate into a standard treatment is the regrowth of nerves in the spinal cord after trauma, with a recent article by lvarez et al, (2021) (press release) covering recent advances in the use of artificial scaffolds that promotes nerve regeneration, reduces scarring and promotes blood vessel formation. This offers hope that one day spinal cord injures may be fully repairable.

If we were to return to the body as a machine comparison, then the human body is less of a car or piece of heavy machinery, and more of a glued-together gadget with complex circuitry and components inside. With this jump in complexity comes the need for a deeper level of understanding, and increasingly more advanced tools so that repairs can be made efficiently and with good outcomes.

Even so, regenerative medicine is already saving the lives of for example burn victims today, and improving the lives of countless others. As further advances in research continue to translate into treatments, we should see a gradual change from youll have to learn to live with that, to a more optimistic give it some time to grow back, as in the case of an injured veteran, or the victim of an accident.

See the original post:
Regenerative Medicine: The Promise Of Undoing The Ravages Of Time - Hackaday

Global Induced Pluripotent Stem Cell (iPSC) Market Report 2021-2028 – Increasing Demand for Body Reconstruction Procedures and Tissue Engineering -…

DUBLIN--(BUSINESS WIRE)--The "Induced Pluripotent Stem Cell (iPSC) Market Share, Size, Trends, Industry Analysis Report By Application (Manufacturing, Academic Research, Drug Development & Discovery, Toxicity Screening, Regenerative Medicine); By Derived Cell; By Region, Segment & Forecast, 2021 - 2028" report has been added to ResearchAndMarkets.com's offering.

The global Induced Pluripotent Stem Cell (iPSC) market size is expected to reach $2,893.3 million by 2028

The ability to model human diseases in vitro as well as high-throughput screening has greatly propelled market growth. Companies have effectively overcome market hurdles faced in the recent past such as proper culturing and differentiation of derived cells at a commercial scale and have developed state-of-the-art manufacturing processes that can achieve scalability and can achieve stringent quality parameters. Such trends are propelling the overall industry growth.

Companies have also developed advanced platforms for Induced pluripotent stem cells that guarantee close connection with a host of in-house technologies that are useful in the proper definition of disease signatures as well as relationships between genetic mutations as well as that properly describe perturbation of specific molecular pathways. This has resulted in the creation of human translational models that are aiding better target identification of diseases that have high unmet medical needs.

Many companies have developed transfection kits, reprogramming vectors, differentiation media, live staining kits, immunocytochemistry, among others to aid the smooth workflow of iPSC production.

However, it has been observed in the recent past that the demand for cells for screening and other purposes is significant and that there are significant challenges that pose a significant hurdle in large-scale iPSC production and differentiation.

Heavy investment in R&D activities pertaining to the development and optimization of iPSC reprogramming process in order to achieve sufficient production is a key industry trend. In the recent past, companies focused more on hepatic, cardiac, pancreatic cells, among others.

However, with the advent of a number of new participants as well as advancements and breakthroughs achieved, it is anticipated that the application portfolio will further increase in the near future.

Industry participants operating in the industry are:

Key Topics Covered:

1. Introduction

2. Executive Summary

3. Research Methodology

4. iPSC Market Insights

4.1. iPSC - Industry Snapshot

4.2. iPSC Market Dynamics

4.2.1. Drivers and Opportunities

4.2.1.1. Increasing demand for body reconstruction procedures and tissue engineering

4.2.1.2. Rising Investments across the globe

4.2.2. Restraints and Challenges

4.2.2.1. Scalability Issues

4.3. Porter's Five Forces Analysis

4.4. PESTLE Analysis

4.5. iPSC Market Industry trends

4.6. COVID-19 Impact Analysis

5. Global iPSC Market, by Derived Cell

5.1. Key Findings

5.2. Introduction

5.3. Hepatocytes

5.4. Fibroblasts

5.5. Amniotic Cells

5.6. Cardiomyocytes

6. Global iPSC Market, by Application

6.1. Key Findings

6.2. Introduction

6.2.1. Global iPSC Market, by Application, 2017 - 2028 (USD Million)

6.3. Manufacturing

6.4. Academic Research

6.5. Drug Development & Discovery

6.6. Toxicity Screening

6.7. Regenerative Medicine

7. Global iPSC Market, by Geography

7.1. Key findings

7.2. Introduction

7.2.1. iPSC Market Assessment, By Geography, 2017 - 2028 (USD Million)

8. Competitive Landscape

8.1. Expansion and Acquisition Analysis

8.1.1. Expansion

8.1.2. Acquisitions

8.2. Partnerships/Collaborations/Agreements/Exhibitions

9. Company Profiles

9.1. Company Overview

9.2. Financial Performance

9.3. Product Benchmarking

9.4. Recent Development

For more information about this report visit https://www.researchandmarkets.com/r/ykewbe

Excerpt from:
Global Induced Pluripotent Stem Cell (iPSC) Market Report 2021-2028 - Increasing Demand for Body Reconstruction Procedures and Tissue Engineering -...

CRISPR Therapeutics and ViaCyte, Inc. to Start Clinical Trial of the First Gene-Edited Cell Replacement Therapy for Treatment of Type 1 Diabetes -…

-Initiation of patient enrollment expected by year-end-

ZUG, Switzerland and CAMBRIDGE, Mass. and SAN DIEGO, Nov. 16, 2021 (GLOBE NEWSWIRE) -- CRISPR Therapeutics (NASDAQ: CRSP), a biopharmaceutical company focused on developing transformative gene-based medicines for serious diseases, and ViaCyte, Inc., a clinical-stage regenerative medicine company developing novel cell replacement therapies to address diseases with significant unmet needs, today announced that Health Canada has approved the companies Clinical Trial Application (CTA) for VCTX210, an allogeneic, gene-edited, immune-evasive, stem cell-derived therapy for the treatment of type 1 diabetes (T1D). Initiation of patient enrollment is expected by year-end.

With the approval of our CTA, we are excited to bring a first-in-class CRISPR-edited cell therapy for the treatment of type 1 diabetes to the clinic, an important milestone in enabling a whole new class of gene-edited stem cell-derived medicines, said Samarth Kulkarni, Ph.D., Chief Executive Officer of CRISPR Therapeutics. The combination of ViaCytes leading stem cell capabilities and CRISPR Therapeutics pre-eminent gene-editing platform has the potential to meaningfully impact the lives of patients living with type 1 diabetes.

Being first into the clinic with a gene-edited, immune-evasive cell therapy to treat patients with type 1 diabetes is breaking new ground as it sets a path to potentially broadening the treatable population by eliminating the need for immunosuppression with implanted cell therapies, said Michael Yang, President and Chief Executive Officer of ViaCyte. This approach builds on previous accomplishments by both companies and represents a major step forward for the field as we strive to provide a functional cure for this devastating disease.

The Phase 1 clinical trial of VCTX210 is designed to assess its safety, tolerability, and immune evasion in patients with T1D. This program is being advanced by CRISPR Therapeutics and ViaCyte as part of a strategic collaboration for the discovery, development, and commercialization of gene-edited stem cell therapies for the treatment of diabetes. VCTX210 is an allogeneic, gene-edited, stem cell-derived product developed by applying CRISPR Therapeutics gene-editing technology to ViaCytes proprietary stem cell capabilities and has the potential to enable a beta-cell replacement product that may deliver durable benefit to patients without requiring concurrent immune suppression.

About CRISPR Therapeutics CRISPR Therapeutics is a leading gene editing company focused on developing transformative gene-based medicines for serious diseases using its proprietary CRISPR/Cas9 platform. CRISPR/Cas9 is a revolutionary gene editing technology that allows for precise, directed changes to genomic DNA. CRISPR Therapeutics has established a portfolio of therapeutic programs across a broad range of disease areas including hemoglobinopathies, oncology, regenerative medicine and rare diseases. To accelerate and expand its efforts, CRISPR Therapeutics has established strategic collaborations with leading companies including Bayer, Vertex Pharmaceuticals and ViaCyte, Inc. CRISPR Therapeutics AG is headquartered in Zug, Switzerland, with its wholly-owned U.S. subsidiary, CRISPR Therapeutics, Inc., and R&D operations based in Cambridge, Massachusetts, and business offices in San Francisco, California and London, United Kingdom. For more information, please visit http://www.crisprtx.com.

About ViaCyte ViaCyte is a privately held clinical-stage regenerative medicine company developing novel cell replacement therapies based on two major technological advances: cell replacement therapies derived from pluripotent stem cells and medical device systems for cell encapsulation and implantation. ViaCyte has the opportunity to use these technologies to address critical human diseases and disorders that can potentially be treated by replacing lost or malfunctioning cells or proteins. ViaCytes first product candidates are being developed as potential long-term treatments for patients with type 1 diabetes to achieve glucose control targets and reduce the risk of hypoglycemia and diabetes-related complications. To accelerate and expand ViaCytes efforts, it has established collaborative partnerships with leading companies, including CRISPR Therapeutics and W.L. Gore & Associates. ViaCyte is headquartered in San Diego, California. For more information, please visitwww.viacyte.comand connect with ViaCyte onTwitter,Facebook, andLinkedIn.

CRISPR Therapeutics Forward-Looking Statement This press release may contain a number of forward-looking statements within the meaning of the Private Securities Litigation Reform Act of 1995, as amended, including statements made by Dr. Kulkarni and Mr. Yang in this press release, as well as regarding CRISPR Therapeutics expectations about any or all of the following: (i) the safety, efficacy and clinical progress of our various clinical programs including our VCTX210 program; (ii) the status of clinical trials (including, without limitation, activities at clinical trial sites) and expectations regarding data from clinical trials; (iii) the data that will be generated by ongoing and planned clinical trials, and the ability to use that data for the design and initiation of further clinical trials; and (iv) the therapeutic value, development, and commercial potential of CRISPR/Cas9 gene editing technologies and therapies, including as compared to other therapies. Without limiting the foregoing, the words believes, anticipates, plans, expects and similar expressions are intended to identify forward-looking statements. You are cautioned that forward-looking statements are inherently uncertain. Although CRISPR Therapeutics believes that such statements are based on reasonable assumptions within the bounds of its knowledge of its business and operations, forward-looking statements are neither promises nor guarantees and they are necessarily subject to a high degree of uncertainty and risk. Actual performance and results may differ materially from those projected or suggested in the forward-looking statements due to various risks and uncertainties. These risks and uncertainties include, among others: the potential for initial and preliminary data from any clinical trial and initial data from a limited number of patients not to be indicative of final trial results; the potential that clinical trial results may not be favorable; potential impacts due to the coronavirus pandemic, such as the timing and progress of clinical trials; that future competitive or other market factors may adversely affect the commercial potential for CRISPR Therapeutics product candidates; uncertainties regarding the intellectual property protection for CRISPR Therapeutics technology and intellectual property belonging to third parties, and the outcome of proceedings (such as an interference, an opposition or a similar proceeding) involving all or any portion of such intellectual property; and those risks and uncertainties described under the heading "Risk Factors" in CRISPR Therapeutics most recent annual report on Form 10-K, quarterly report on Form 10-Q and in any other subsequent filings made by CRISPR Therapeutics with the U.S. Securities and Exchange Commission, which are available on the SEC's website at http://www.sec.gov. Existing and prospective investors are cautioned not to place undue reliance on these forward-looking statements, which speak only as of the date they are made. CRISPR Therapeutics disclaims any obligation or undertaking to update or revise any forward-looking statements contained in this press release, other than to the extent required by law.

CRISPR Therapeutics Investor Contact: Susan Kim +1-617-307-7503 susan.kim@crisprtx.com

CRISPR Therapeutics Media Contact: Rachel Eides +1-617-315-4493 rachel.eides@crisprtx.com

ViaCyte Investor Contact: David Carey, Lazar-FINN Partners +1-212-867-1768 david.carey@finnpartners.com

ViaCyte Media Contact: Glenn Silver, Lazar-FINN Partners +1-973-818-8198 glenn.silver@finnpartners.com

Read more:
CRISPR Therapeutics and ViaCyte, Inc. to Start Clinical Trial of the First Gene-Edited Cell Replacement Therapy for Treatment of Type 1 Diabetes -...

Researchers use model of hypothalamus to implicate genes associated with sleep, BMI, puberty, and more – EurekAlert

Stem cells help researchers create more complete genomic picture of difficult-to-study part of the brain involved in many key functions

Philadelphia, November 19, 2021 A study led by researchers at Childrens Hospital of Philadelphia (CHOP) has implicated several genes involved in a variety of bodily functions associated with the hypothalamus, a notoriously difficult-to-study region of the brain. The findings could help clinicians identify potential causes of dysfunction for many important traits regulated by the hypothalamus, such as sleep, stress, and reproduction.

The findings were published today in the journal Nature Communications.

The hypothalamus helps maintain health and stable metabolism by influencing a variety of vital functions, including appetite and thirst, puberty and reproductive timing, sleep cycles, and body temperature. However, the hypothalamus is located in the center of the brain, making it extremely difficult to study the gene regulation associated with these traits.

To overcome that hurdle, the researchers used an embryonic stem cell (ESC) model to study gene expression during development of the hypothalamus. This model allowed them to study the genetic architecture first in hypothalamic progenitor cells cells prior to their full development into a hypothalamus and then in arcuate nucleus-like hypothalamic neurons. The hypothalamus contains several different subtypes of neurons, and the researchers integrated results from various genome-wide association studies (GWAS) to implicate genes driving particular traits regulated by the hypothalamus.

By studying the three-dimensional genomic architecture of these cell models, we can see the dynamic process of how the hypothalamus is formed over different stages of development, said senior study author Struan F.A. Grant, PhD, Director of the Center for Spatial and Functional Genomics and the Daniel B. Burke Endowed Chair for Diabetes Research at CHOP. The information we yielded in this study provides us with more concrete information about diseases that are relevant to hypothalamic function.

Grant and his collaborators assessed variants associated with puberty, body mass index, height, bipolar disorder, sleep, and major depressive disorder, among others. They identified both known and novel genes associated with these traits. For example, their data confirmed the role of the BDNF of gene in influencing body mass index and obesity risk. Another gene of interest identified in the study was PER2, which was implicated in sleep regulation.

All the data ascertained from this study will be made publicly available. Many of the disorders studied can be caused by other factors, so the findings will help researchers distinguish which genes play a more central role in this tissue and in turn inform clinical practice. For example, body mass index can be affected by variants in genes conferring their effects in hypothalamus or fat tissue cells, so being able to distinguish the context in which genes and subsequent tissues or hormones operate can lead to more personalized treatment options.

The data set we derived from this study allows other researchers to determine which diseases or conditions are relevant when doing a genetic workup of the patient, Grant said. As more information about the hypothalamus is known, that information can be queried against this data set and potentially identify therapeutic targets for multiple disorders.

This research was supported by the National Center for Research Resources (UL1RR024134) and the National Center for Advancing Translational Sciences (UL1TR000003). This research was also supported in part by the Institute for Translational Medicine and Therapeutics (ITMAT) Transdisciplinary Program in Translational Medicine and Therapeutics, the Eunice Kennedy Shriver National Institute of Child Health (NIH1K99HD099330-01), and National Institutes of Health grants DK52431-23, P30DK026687-41, R01 HD056465, R01 HG010067, R01 HL143790, and the Daniel B. Burke Endowed Chair for Diabetes Research.

Pahl et al, Cis-regulatory architecture of human ESC-derived hypothalamic neuron differentiation aids in variant-to-gene mapping of relevant complex traits. Nat Comm. Online November 19, 2021. DOI: 10.1038/s41467-021-27001-4.

About Childrens Hospital of Philadelphia: Childrens Hospital of Philadelphia was founded in 1855 as the nations first pediatric hospital. Through its long-standing commitment to providing exceptional patient care, training new generations of pediatric healthcare professionals, and pioneering major research initiatives, Childrens Hospital has fostered many discoveries that have benefited children worldwide. Its pediatric research program is among the largest in the country. In addition, its unique family-centered care and public service programs have brought the 564-bed hospital recognition as a leading advocate for children and adolescents. For more information, visit http://www.chop.edu

Nature Communications

Experimental study

Lab-produced tissue samples

Cis-regulatory architecture of human ESC-derived hypothalamic neuron differentiation aids in variant-to-gene mapping of relevant complex traits

19-Nov-2021

Disclaimer: AAAS and EurekAlert! are not responsible for the accuracy of news releases posted to EurekAlert! by contributing institutions or for the use of any information through the EurekAlert system.

More:
Researchers use model of hypothalamus to implicate genes associated with sleep, BMI, puberty, and more - EurekAlert

COVID-19: Researchers warn against overhyping early-stage therapies – Medical News Today

The COVID-19 pandemic has created a sense of urgency to generate new drugs and vaccines. In many cases, this urgency became a regulatory opportunity to bypass established regulatory pathways for new drugs.

While this has led to the fast emergence of many useful drugs and vaccines for COVID-19, it has also led to a general reduction in the quality of medical research from which to derive conclusions.

For example, according to Janet Woodcock, former director of the Food and Drug Administrations (FDA) Center for Drug Evaluation and Research, an FDA analysis found that 6% of clinical trials are yielding results the agency deems actionable.

The lack of regulation coupled with a sense of urgency has also led to overhype and rushed development of certain treatments, including cell-based therapies often sold as stem cell treatments.

While some of these products have undergone well-designed, adequately controlled trials, most are in the early stages. Some clinics are nevertheless offering these unproven and unlicensed treatments to people, promising to boost their immune system or overall health to protect against COVID-19.

Promoting and selling unproven and unlicensed treatments can harm public health and could lead many to undergo untested and potentially harmful treatments.

Recently, a group of researchers from the University of California, Irvine, the Georgia Institute of Technology, the University at Buffalo, NY, and the University of Melbourne in Australia, published a report outlining misinformation around cell-based treatments for COVID-19, calling for their stronger regulation.

Efforts to rapidly develop therapeutic interventions should never occur at the expense of the ethical and scientific standards that are at the heart of responsible clinical research and innovation, said Dr. Laertis Ikonomou, assistant professor of Oral Biology at the University at Buffalo, and co-author of the study.

Scientists, regulators, and policymakers must guard against the proliferation of poorly designed, underpowered, and duplicative studies that are launched with undue haste because of the pandemic, but are unlikely to provide convincing, clinically meaningful safety and efficacy data, said co-author Dr. Leigh Turner, professor of Health, Society and Behavior at the University of California, Irvine.

The researchers published their report in Stem Cell Reports.

Researchers conducted a study in August 2020 of 70 clinical trials involving cell-based treatments for COVID-19. They found that most were small, with an average of 51.8 participants, and only 22.8% were randomized, double-blinded, and controlled experiments.

The authors concluded that the cell-based interventions for COVID-19 were likely to have a relatively small collective clinical impact.

Cell-based treatments for COVID-19 are still at an experimental stage, Dr. Ikonomou told Medical News Today. There are tens of clinical trials, of varied complexity and rigor, that evaluate various cell types, such as mesenchymal stromal cells, for COVID-19 treatment.

Expanded or compassionate use of cell-based interventions has also been reported, but these individual cases are unlikely to tell us whether and how cell therapies could help with COVID-19 and do not substitute for the systematic clinical evaluation of cell-based products, he added.

A few completed phase 1/2 trials have shown a favorable safety profile, but larger size trials are required. Eventually, properly-powered, controlled, randomized, double-blinded clinical trials will help determine whether cell-based treatments are a viable therapeutic option for COVID-19 and its complications, he explained.

The urgency of the pandemic has made it easy to exaggerate early-stage research. The scientists highlight this is especially the case in press releases, where media professionals can over-hype findings and understate or omit limitations to gain more media coverage.

The researchers also say that even when online media include limitations and key aspects of studies, other communication channels can strip these away easily. What is left then gets amplified, as the public is desperate to see positive news.

To address this, the researchers say science communicators should ensure they have an accurate understanding of the information they report and highlight the required steps for the science to advance without exaggerating its speed.

The researchers also say that simply feeding the public more information in what is known as the information deficit model alone is insufficient. They also suggest science communicators should strive for an engaged or dialogue-based communication approach.

Over-hyping of promising treatments and in particular cell-based treatments has been a longstanding problem, and it did not first emerge with the COVID-19 pandemic, said Dr. Ikonomou. It has become a salient issue during these times due to the global nature of this health emergency and the resulting devastation and health toll.

Therefore, it is even more important to communicate promising developments in COVID-19-related science and clinical management [responsibly]. Key features of good communication are an accurate understanding of new findings, including study limitations and avoidance of sensationalist language, he explained.

Realistic timeframes for clinical translation are equally important as is the realization that promising interventions at preliminary stages may not always translate to proven treatments following rigorous testing, he added.

The researchers say that commercial investments by biotechnology companies to develop cell-based therapies for COVID-19 have led to well-designed and rigorous clinical trials.

However, some other businesses have overlooked the demanding process of pre-marketing authorization of their products. Instead, they made unsubstantiated and inaccurate claims about their stem cell products for COVID-19 based on hyperbolic reporting of cell-based therapies in early testing.

Some clinics advertise unproven and unlicensed mesenchymal stem cell treatments or exosome therapies as immune boosters that prevent COVID-19 and repair and regenerate lungs.

Often, these businesses make their treatments available via infusion or injection. However, one anti-aging clinic in California shipped its kits to clients, where they were to self-administer with a nebulizer and mask.

Such companies often market stem cell treatments via online and social media. In an initial review of many of these brands, the researchers could not find published findings from preclinical studies and clinical trials to support their commercial activities.

Instead, they found that these companies drew from uncritical news media reports, preliminary clinical studies, or case reports in which those diagnosed with COVID-19 received stem cell interventions.

Promoting such therapies that have not undergone proper tests for safety and efficacy have the potential for significant physical and financial harm.

Health experts have documented adverse events due to unlicensed stem cell products, including vision loss and autoimmune, infectious, neurological, and cardiovascular complications.

Early in the pandemic, scientific and professional societies, including the Alliance for Regenerative Medicine and the International Society for Stem Cell Research, have warned the public against businesses engaged in the marketing of cell-based treatments that have not undergone adequate testing.

The researchers highlight that it is unclear whether these warnings reached individuals and their loved ones or significantly affected public understanding of the risks of receiving unlicensed and unproven stem cell treatments for COVID-19.

They also indicate that it is unclear whether these societies and organizations have an important role in convincing regulatory bodies to increase enforcement in this space. Nevertheless, at the time of writing, the FDA and Federal Trade Commission have issued 22 letters to businesses selling unproven and unlicensed cell-based therapies.

And while many of these companies have ceased market activity, the presence of other companies continuing to pedal the same claims makes it clear that regulatory bodies must increase their enforcement.

Additionally, the researchers question whether warning letters are sufficient to disincentivize clinicians and others to sell unlicensed products. They write:

If companies and affiliated clinicians are not fined, forced to return to patients whatever profits they have made, confronted with criminal charges, subject to revocation of medical licensure, or otherwise subject to serious legal and financial consequences, it is possible that more businesses will be drawn to this space because of the profits that can be generated from selling unlicensed and unproven cell-based products in the midst of a pandemic.

The researchers conclude that regulators should increase enforcement against unproven and unlicensed therapies for COVID-19.

They also say that science communicators should report on scientific claims more realistically and include the public in more discourse.

In the U.S. and elsewhere, there are regulations and enforcement mechanisms that deal with harms caused by unproven and unlicensed cell-based interventions and false advertising claims, said Dr. Ikonomou. It may be preferable to implement existing regulations more vigorously than introduce new ones.

Stakeholders, such as scientific, professional, and medical associations, can contribute towards this goal with reporting and monitoring of cell therapy misinformation. There is a shared responsibility to combat cell-therapy related misinformation and disinformation that undercuts legitimate research and clinical efforts and portrays unproven interventions as silver bullets for COVID-19, he concluded.

For live updates on the latest developments regarding the novel coronavirus and COVID-19, click here.

Original post:
COVID-19: Researchers warn against overhyping early-stage therapies - Medical News Today

Albert Einstein Cancer Center researcher receives NCI Outstanding Investigator Award to study two deadly blood diseases – EurekAlert

image:Dr. Ulrich G. Steidl view more

Credit: Albert Einstein College of Medicine

October 27, 2021(BRONX, NY)Ulrich G. Steidl, M.D., Ph.D., co-director of the Blood Cancer Institute and associate director of basic science at the Albert Einstein Cancer Center (AECC), has received a prestigious Outstanding Investigator Award from the National Cancer Institute (NCI). This award is accompanied by a seven-year, $7 million grant to study the molecular and cellular mechanisms that lead to two related blood diseases, myelodysplastic syndromes (MDS) and acute myeloid leukemia (AML). Dr. Steidl is one of only 17 recipients of this award in 2021, which is given to accomplished leaders in cancer research who provide significant contributions in their field. The ultimate goal of this research is to develop new treatments and cures for these usually fatal disorders.

Clinical outcomes in MDS and AML have not significantly improved over the past half-century, and cure rates remain below 15% for most patients, said Dr. Steidl, who is also professor of cell biology and of medicine and the Diane and Arthur B. Belfer Faculty Scholar in Cancer Research at Einstein. There is an urgent need to improve our understanding of how these diseases develop and to devise more effective therapies.

MDS and AML Explained

MDS occurs when blood-forming (hematopoietic) stem cells in the bone marrow acquire genetic and non-genetic irregularities, leading to the production of abnormal, dysfunctional blood cells, which out-compete healthy cells. Common symptoms include anemia, infections, and bleeding.

The incidence of MDS in the United States is unclear, with estimates ranging from 10,000 to 40,000 new cases annually; about one-third of MDS patients will go on to develop AML. Treatment for MDS is generally limited to preventing or reducing complications, particularly severe anemia. The only cure is a bone-marrow transplanta therapy not easily tolerated and therefore often reserved for the youngest, most resilient patients. However, most people diagnosed with MDS are elderly.

AML, like MDS, begins with abnormal bone marrow stem cells. But in AML, those cells, after becoming cancerous, proliferate rapidly and quickly spread to the blood and other hematopoietic organs, such as the bone marrow and spleen, and sometimes to other tissues, causing many of the same symptoms seen in MDS, plus others. AML is often fatal within just a few months and afflicts about 21,000 Americans each year. It is usually treated with chemotherapy. Bone-marrow transplantation can cure AML in some patients.

From Stem Cells to Cancer

Recent studies led by Dr. Steidl and his research team have shown that both MDS and AML arise from pre-leukemic stems cells (pre-LSCs), a subpopulation of blood-forming stem cells that have genetic and non-genetic aberrations. Certain varieties (clones) of these pre-LSCs go on to develop into leukemic stem cells (LSCs)cancer cells that are capable of self-renewal. These LSCs lead to sustained leukemia growth and are particularly resistant to drugs. We now know that the considerable diversity of pre-LSC clones affects the development, progression, and treatment resistance of both MDS and AML, said Dr. Steidl, one of the nations leading authorities on both diseases.

What causes some pre-LSCs but not others to become leukemic is not clear, but transcription factors are thought to play a key role. Transcription factors are proteins that turn specific genes on or off, determining a cells function by regulating the activity of genes. In the case of stem cells, transcription factors guide their differentiation into mature cells. Our recent work has shown that the actions of key transcription factors are dysregulated in pre-LSCs and LSCs, meaning that the transcription factors and the molecular programs they govern behave abnormally, he added.

Thanks to his new NCI grant, Dr. Steidl hopes to:

To accomplish these goals, Dr. Steidls research team will employ novel tools for analyzing stem cell clones in patients, as well as newly developed mouse models of pre-LSC progression to MDS and AML.

Developing New Cancer Therapies

The knowledge we gain from this research should enable us to develop drugs that target pre-LSCs and their aberrant transcription factors, said Dr. Steidl. Such an approach holds the promise of achieving lasting remissions and, ultimately, even cures. Hopefully, our understanding of the early events in the progression of MDS and AML may even allow us in the future to prevent these diseases by interrupting the transformation of pre-LSCs to LSCs before overt leukemia can occur.

The grant (R35CA253127) is titled Molecular and Cellular Regulation of Pre-Leukemic Stem Cells and their Therapeutic Targeting.

***

About Albert Einstein College of Medicine

Albert Einstein College of Medicine is one of the nations premier centers for research, medical education and clinical investigation. During the 2020-21 academic year, Einstein is home to 721 M.D. students, 178 Ph.D. students, 109 students in the combined M.D./Ph.D. program, and 265 postdoctoral research fellows. The College of Medicine has more than 1,900 full-time faculty members located on the main campus and at its clinical affiliates. In 2020, Einstein received more than $197 million in awards from the National Institutes of Health (NIH). This includes the funding of major research centers at Einstein in aging, intellectual development disorders, diabetes, cancer, clinical and translational research, liver disease, and AIDS. Other areas where the College of Medicine is concentrating its efforts include developmental brain research, neuroscience, cardiac disease, and initiatives to reduce and eliminate ethnic and racial health disparities. Its partnership with Montefiore, the University Hospital and academic medical center for Einstein, advances clinical and translational research to accelerate the pace at which new discoveries become the treatments and therapies that benefit patients. Einstein runs one of the largest residency and fellowship training programs in the medical and dental professions in the United States through Montefiore and an affiliation network involving hospitals and medical centers in the Bronx, Brooklyn and on Long Island. For more information, please visit einsteinmed.org, read our blog, followus on Twitter, like us on Facebook, and view us on YouTube.

Disclaimer: AAAS and EurekAlert! are not responsible for the accuracy of news releases posted to EurekAlert! by contributing institutions or for the use of any information through the EurekAlert system.

See the article here:
Albert Einstein Cancer Center researcher receives NCI Outstanding Investigator Award to study two deadly blood diseases - EurekAlert